WO2009136752A9 - Novel peptide for augmenting brain-derived neutrotrophic factor (bdnf) protein expression in hippocampal neurons, hippocampal tissue and cerebral-cortex tissue - Google Patents

Novel peptide for augmenting brain-derived neutrotrophic factor (bdnf) protein expression in hippocampal neurons, hippocampal tissue and cerebral-cortex tissue Download PDF

Info

Publication number
WO2009136752A9
WO2009136752A9 PCT/KR2009/002421 KR2009002421W WO2009136752A9 WO 2009136752 A9 WO2009136752 A9 WO 2009136752A9 KR 2009002421 W KR2009002421 W KR 2009002421W WO 2009136752 A9 WO2009136752 A9 WO 2009136752A9
Authority
WO
WIPO (PCT)
Prior art keywords
bdnf
peptide
disease
mvg
hippocampal
Prior art date
Application number
PCT/KR2009/002421
Other languages
French (fr)
Korean (ko)
Other versions
WO2009136752A2 (en
WO2009136752A3 (en
Inventor
김길룡
문성아
김홍기
정우람
이병주
신민규
Original Assignee
성균관대학교 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 성균관대학교 산학협력단 filed Critical 성균관대학교 산학협력단
Publication of WO2009136752A2 publication Critical patent/WO2009136752A2/en
Publication of WO2009136752A9 publication Critical patent/WO2009136752A9/en
Publication of WO2009136752A3 publication Critical patent/WO2009136752A3/en

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/08Tripeptides
    • C07K5/0802Tripeptides with the first amino acid being neutral
    • C07K5/0804Tripeptides with the first amino acid being neutral and aliphatic
    • C07K5/081Tripeptides with the first amino acid being neutral and aliphatic the side chain containing O or S as heteroatoms, e.g. Cys, Ser
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof

Definitions

  • the present invention relates to novel peptides that increase protein expression of BDNF with various effects such as neuronal growth and differentiation and learning, memory, antidepression and the like.
  • Neurotrophic factor regulates the survival and differentiation of neurons during development (Davies, 1994), and maintains neural structure and activity of ion channels, neurotransmitter release, and axon pathways during the life of the individual (Davies, 1994). axon path-finding) (Schnell et al., 1994; Song and Poo, 1999; Schinder and Poo, 2000).
  • Examples of such neurotrophic factors include nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), neurotrophin-3 (NT-3), and neurotrophs.
  • NNF nerve growth factor
  • BDNF brain-derived neurotrophic factor
  • NT-3 neurotrophin-3
  • Pin-4 / 5 Neurotrophin-4 / 5, NT-4 / 5 is present.
  • BDNF is expressed in the hippocampus, cortex and basal brain which are responsible for learning, memory, and higher thinking in the central nervous system (CNS).
  • BDNF expressed in this position is a major regulator of synaptic plasticity and recognition processes, which is the neurobiochemical basis for synaptic transmission and learning and memory (Lu B, 2003). Not only does it promote long-term potentiation (LTP), a cellular phenomenon in learning and memory processes, but it also inhibits long-term depression, LTD (opposite) (Ikegaya, Y. et al., 2002; Huber, K. et al., 1998).
  • LTD opposite
  • this unique role is consistent with the fact that BDNF and its receptor, TrkB (tropomyosin-related kinase B), are widely distributed in glutamate synapses.
  • BDNF an important regulator of synaptic transmission and synaptic plasticity, is associated with a variety of diseases. Typical examples include degenerative brain diseases such as Alzheimer's disease (AD), Parkinson's disease (PD), depression from stress, stroke, Huntington's chorea, cerebral ischemic disease, neurodegenerative diseases or diabetic neuropathy Etc. can be mentioned.
  • AD Alzheimer's disease
  • PD Parkinson's disease
  • depression from stress stroke
  • Huntington's chorea cerebral ischemic disease
  • neurodegenerative diseases or diabetic neuropathy Etc can be mentioned.
  • Alzheimer's disease causes deterioration of cognitive ability by the degeneration of cholinergic neurons in the whole brain and a decrease in the release of the neurotransmitter acetylcholine (Murer MG et al., 2001).
  • BDNF is known to promote the survival and differentiation of basal cholinergic neurons as well as to stimulate the release of acetylcholine in these neurons (Knipper M et al., 1994). Therefore, it is suggested that deterioration of homeostasis of cells due to lack of synthesis of BDNF will cause Alzheimer's disease.
  • neuroplastic deficiency ie reduction in synaptic contact, presents neuropathological and clinical signs (Mesulam MM., 1999).
  • Parkinson's disease results in a decrease in the number of substantia nigral dopaminergic neurons, leading to depletion of striatal dopamine in the striatum, leading to symptoms such as motor dysfunction.
  • Another characteristic of Parkinson's disease patients is that the cognitive function is deteriorated.
  • neurotrophic factors are expected to play an important role in the protection of dopaminergic neurons (Siegel GJ and Chauhan NB., 2000).
  • the interaction of BDNF with dopaminergic neurons is well known.
  • BDNF mRNA is significantly increased in the hippocampus, anterior cerebral cortex or both regions (Duman, R.S. and Monteggia, L.M. 2006). These results suggest the hypothesis of 'neurotrophin hypothesis of depression'. In other words, the treatment of mental disorders such as depression by restoring neuronal breakdown and cell number reduction by re-increasing the expression of reduced BDNF in the hippocampus and the anterior cerebral cortex by stress (Duman, R.S. and Monteggia, L.M. 2006).
  • BDNF exhibits various functions such as neuronal cell growth and differentiation, learning and memory, and antidepressant as well as maintenance of neural structure, activation of ion channels, and release of neurotransmitters.
  • BDNF can be used for Alzheimer's disease, Parkinson's disease, depression, stroke (Schabitz et al., Stroke, 38: 2165-2172, 2007), and Huntington's chorea (Zuccato C et al., Science 293, 20). , July 2001), cerebral ischemic disease (Han BH et al., The Journal of neuroscience, 2000, 20 (15): 5775-5781, August 1, 2000), neurodegenerative diseases (Tsuzaka K et al., Muscle Nerve.
  • BDNF itself is a 14kDa macromolecular protein that does not cross the blood-brain barrier (BBB) and has problems such as administration method. Therefore, it is easy to cross the blood brain barrier and has very low cytotoxicity, and by discovering a substance that increases the expression of BDNF by administering a small amount, it is possible to detect neurological diseases such as Alzheimer's disease, Parkinson's disease, depression, stroke, which are chronic stress diseases. It can be used for prevention and treatment.
  • BBB blood-brain barrier
  • the present inventors prepared an MVG peptide consisting of three amino acids by selecting the amino acids with the highest expression of BDNF at each position through a peptide library technology called PS-SPCL (Positional Scanning-Synthetic Peptide Combinatorial Library).
  • PS-SPCL Porous Scanning-Synthetic Peptide Combinatorial Library
  • the present invention was completed by confirming that BDNF mRNA and protein expression is increased in hippocampal neurons, hippocampal tissues and cerebral cortical tissues by MVG peptide.
  • BDNF Brain-derived neurotrophic factor
  • Another object of the present invention is to provide a peptide for increasing protein expression of brain-derived neurotrophic factor (BDNF) in hippocampal neurons, hippocampal tissues and cerebral cortical tissues.
  • BDNF brain-derived neurotrophic factor
  • Another object of the present invention to provide a pharmaceutical composition for the prevention and treatment of neurological diseases comprising a therapeutically effective amount of the peptide.
  • the present invention provides a peptide consisting of the amino acid (MVG; Methionine-Valine-Glycine) described in SEQ ID NO: 1 (hereinafter referred to as 'MVG peptide').
  • the peptide of the present invention increases the expression of Brain-derived neurotrophic factor (BDNF) mRNA and / or the protein expression of Brain-derived neurotrophic factor (BDNF) in hippocampal neurons, hippocampus and cerebral cortex, and expresses protein expression of TrkB and May increase phosphorylation.
  • BDNF Brain-derived neurotrophic factor
  • BDNF Brain-derived neurotrophic factor
  • the present invention also provides a polynucleotide consisting of a DNA sequence encoding the peptide.
  • the polynucleotide comprises an equivalent nucleic acid sequence, that is, a codon degeneracy sequence that encodes MVG peptides of different but identical sequences.
  • the present inventors selected the amino acid with the highest expression of BDNF at each position through a peptide library technology called PS-SPCL (Positional Scanning-Synthetic Peptide Combinatorial Library) (MVG; Methionine-Valine-Glycine) Afterwards, it was again confirmed by RT-PCR and Western blotting that expression increased substantially at the cellular level. In addition, MTT (3- (4,5-Dimethylthiazol-2-yl) -2,5-diphenyltetrazolium bromide) test was performed to determine whether MVG peptide is toxic at the cellular level. To confirm the expression of BDNF at the actual animal level, 0.1 ⁇ g / kg of peptide was injected through the tail vein, and the hippocampus and cerebral cortex were removed and Western blotting was performed.
  • PS-SPCL Pural Scanning-Synthetic Peptide Combinatorial Library
  • MTT 3- (4,5-Dimethylthiazol-2-yl) -2,
  • the present inventors confirmed that the MVG peptide consisting of three amino acids induced an increase in BDNF expression not only in hippocampal neuronal cell line but also in hippocampal tissue and cerebral cortical tissue, and showed no cytotoxicity up to 1 mM at the cellular level. It was.
  • the present invention will be described in more detail.
  • the present inventors incubated HT22 cells, a mouse hippocampal neuronal cell line, in a 24 well plate at 5 ⁇ 10 4 per well for 12 hours, and then prepared PS-SPCL stock (stock). Was treated to each well. After 12 hours, the protein was obtained using RIPA (Radio-immunoprecipitation Assay) buffer to which the protease inhibitor was added, and the amount of BDNF was measured through Enzyme-Linked ImmunoSorbent Assay (ELISA). As a result, as shown in FIG. 1, M (Methionine) at the first position, V (Valine) at the second position, and G (Glycine) at the third position expressed the most BDNF.
  • RIPA Radio-immunoprecipitation Assay
  • the results show that the peptide composed of the amino acid (MVG) having the highest expression of BDNF at each position is resynthesized and treated to H19-7 cells of rat hippocampal neuronal cell line to increase the expression of BDNF mRNA. It was confirmed by RT-PCR. As a result, BDNF mRNA expression was increased in a dose-dependent manner in the experimental group treated with 0.001, 0.01, 0.1 ⁇ M MVG peptide as shown in FIG.
  • MVG amino acid
  • the BDNF protein expression was confirmed by Western blotting by treating the hippocampal neuronal H19-7 cells with MVG peptide.
  • MVG peptide As a result, as shown in FIG. 3, BDNF protein expression was increased in a dose-dependent manner in the experimental group treated with 0.001, 0.01, and 0.1 ⁇ M MVG peptide as compared to the control group.
  • male SD (Sprague-Dawely) rats (4 weeks of age) are purchased and subjected to stabilization for 1 week, after which 1X PBS (control) and the MVG peptides determined above are 0.1 ⁇ g / Kg injections and hippocampal and cerebral cortical tissues were performed and Western blotting was performed.
  • 1X PBS control
  • MVG peptides determined above are 0.1 ⁇ g / Kg injections and hippocampal and cerebral cortical tissues were performed and Western blotting was performed.
  • H19-7 cells were treated with MVG peptides from 10nM to 1mM to confirm cytotoxicity of the MVG peptides, and then the survival rate of the cells was confirmed by MTT solution. It was confirmed (FIG. 6).
  • one of the spatial memory tests was performed to determine whether the MVG peptides promote spatial learning and memory at the experimental animal level. Enhancement of memory was confirmed through animal experiments (FIG. 8, FIG. 10), and increased BDNF expression by MVG peptides in hippocampus (FIG. 11A) and cerebral cortex (FIG. 11B).
  • TrkB phosphorylation and TrkB expression were increased in the group injected with MVG peptides in the hippocampus and cerebral cortex, so that spatial learning and memory enhancement by MVG peptide increased the expression of its receptor (TrkB) as the hippocampus increased BDNF expression. It can be seen that the activation and the expression of the receptor is enhanced (Fig. 12a, 12b).
  • the present invention provides a pharmaceutical composition for the prevention and treatment of neurological diseases comprising a therapeutically effective amount of the MVG peptide of the present invention.
  • the present invention also provides a gene carrier comprising a polynucleotide consisting of a DNA sequence encoding the MVG peptide of the present invention, the gene carrier of the present invention can act as a pharmaceutical composition for the prevention and treatment of neurological diseases.
  • the pharmaceutical composition is prepared in various forms by mixing a pharmaceutically acceptable non-toxic carrier that is commercially available according to the method of administration with the MVG peptide of the present invention, the prepared pharmaceutical composition is a neurological disease described above It can be administered as a prophylactic and therapeutic agent of.
  • MVG peptide was treated with 0.001, 0.01, 0.1, 1 nM for 24 hours in SH-SY5Y cell line, a human dopaminergic cell line, and BDNF.
  • Western blot expression of the anti-apoptotic protein, Bcl-2 was found to increase BDNF and Bcl-2 expression in the presence of 0.1 nM MVG peptide for 24h.
  • MVG peptide was pretreated with 0.1 nM for 24 h in a SH-SY5Y cell line and treated with 2.5 mM MPP + for 24 h, and MVG peptide was measured by MTT assay 12 h and 24 h after MPP + treatment.
  • BDNF brain-derived neurotrophic factor
  • AD Alzheimer's disease
  • Arancibia S Silhol M, Mouli re F, Meffre J, H llinger I, Maurice T, Tapia-Arancibia L.
  • the brain-derived neurotrophic factor gene as a possible susceptibility candidate for Alzheimer's disease in a chinese population.
  • Brain-derived neurotrophic factor, stress and depression a minireview.
  • Brain-derived neurotrophic factor and antidepressant action another piece of evidence from pharmacogenetics.
  • Acute hippocampal brain-derived neurotrophic factor restores motivational and forced swim performance after corticosterone.
  • TrkB neurotrophin receptor Targeting TrkB neurotrophin receptor to treat depression.
  • Rasagiline promotes regeneration of substantia nigra dopaminergic neurons in post-MPTP-induced Parkinsonism via activation of tyrosine kinase receptor signaling pathway.
  • Valproate protects dopaminergic neurons in midbrain neuron / glia cultures by stimulating the release of neurotrophic factors from astrocytes.
  • Neurotrophic factors stabilize microtubules and protect against rotenone toxicity on dopaminergic neurons.
  • Brain-derived growth factor and glial cell line-derived growth factor use distinct intracellular signaling pathways to protect PD cybrids from H2O2-induced neuronal death.
  • Brain-derived neurotrophic factor over-expression in the forebrain ameliorates Huntington's disease phenotypes in mice.
  • Pharmaceutically acceptable carriers compatible with various formulations include all forms of diluents or solvents, fillers, fillers, binders, dispersants, disintegrants, surfactants, lubricants, excipients, and wetting agents. Furthermore, if necessary, commercially available dissolution aids, buffers, preservatives, colorants, flavors, sweeteners, and the like may also be added to the drug.
  • the dosage unit form of the pharmaceutical composition according to the present invention is not particularly limited and may be widely selected according to various therapeutic purposes, for example, tablets, capsules, granules, pills, syrups, solutions, emulsions, suspensions.
  • Oral dosages such as and the like, injection (subcutaneous injection, intravenous injection, intramuscular injection, intraperitoneal injection, etc.) and parenteral administration such as suppositories.
  • oral dosage formulations are preferred.
  • microinjection electroporation using electroshock (electorporation)
  • delivery using cations or liposomes intracellular delivery methods of peptides known in the art, such as PTD (protein transduction domain) can be used.
  • PTD protein transduction domain
  • the various types of medicaments described above can be prepared by conventional methods, for example in the preparation of oral dosage forms such as tablets, capsules, granules and pills, they are known as sucrose, lactose, glucose, starch, mannitol Excipients; Binders such as syrup, gum arabic, sorbitol, trigacanth rubber, methylcellulose, polyvinylpyrrolidone and the like; Disintegrants such as starch, carboxymethyl cellulose and its calcium salts, microcrystalline cellulose, polyethylene glycol and the like; Lubricants such as talc, magnesium stearate, calcium stearate, silica and the like; It may be prepared by conventional methods using wetting agents such as sodium laurate, glycerol and the like.
  • injectables solutions, emulsions, suspensions and syrups
  • they are prepared by conventional methods, such as ethyl alcohol, isopropyl alcohol, propylene glycol, 1,3-butylene glycol, polyethylene glycol, castor oil and the like.
  • Solvent for component dissolution Surfactants such as sorbitol fatty acid ester, polyoxyethylene sorbitol fatty acid ester, polyoxyethylene ester, polyoxyethylene ether of hydrogenated castor oil, lecithin and the like; Manufactured using appropriate preservatives such as cellulose derivatives such as sodium carboxymethyl cellulose, methyl cellulose, suspending agents such as natural rubbers such as trigacanth rubber, gum arabic, esters of paraoxybenzoic acid, benzalkonium chloride, sorbitan fatty acid salts, etc. Can be.
  • Surfactants such as sorbitol fatty acid ester, polyoxyethylene sorbitol fatty acid ester, polyoxyethylene ester, polyoxyethylene ether of hydrogenated castor oil, lecithin and the like
  • Manufactured using appropriate preservatives such as cellulose derivatives such as sodium carboxymethyl cellulose, methyl cellulose, suspending agents such as natural rubbers such as trigacanth
  • suppositories they can be prepared using excipients such as polyethylene glycol, lanolin, coconut oil and the like using conventional methods.
  • the dose of the pharmaceutical composition for the prevention and treatment of neurological diseases depends on the method of administration, the type of preparation, the age of the patient, the weight of the patient, the sensitivity of the patient, and the condition of the disease. It may be appropriately selected and is not specifically limited.
  • the therapeutically effective amount of the MVG peptide included in the pharmaceutical composition of the present invention may be administered at 0.1 ⁇ g / kg / day to 10 ⁇ g / kg / day.
  • the therapeutically effective amount may be in an amount outside the ranges described above.
  • the pharmaceutical composition of the present invention can be administered to mammals such as rats, mice, livestock, humans, etc. by various routes. All modes of administration can be envisaged, for example, by oral, subcutaneous, intravenous, intramuscular, nasal, intraperitoneal, rectal, intrauterine dural or intracerebroventricular injection.
  • the MVG peptide of the present invention has little toxicity and side effects, and therefore can be used with confidence even for prolonged administration for prophylactic purposes.
  • the pharmaceutical composition of the present invention comprises a polynucleotide or equivalent nucleic acid sequence consisting of a DNA sequence encoding an MVG peptide, that is, a codon degeneracy sequence encoding a MVG peptide having a different sequence but the same DNA sequence.
  • a polynucleotide or equivalent nucleic acid sequence consisting of a DNA sequence encoding an MVG peptide, that is, a codon degeneracy sequence encoding a MVG peptide having a different sequence but the same DNA sequence.
  • It may be a gene carrier, and the gene delivery method of the nucleotides is also included in the technical scope of the present invention.
  • the oligonucleotides can be delivered intracellularly in a known form. For example, retroviral vectors, adenovirus vectors, and adeno-associated viral vectors that have been used as conventional gene carriers.
  • Viral vector comprising a rule, liposomes, polylysine, polyethylenimine (PEI), protamine, histone, polyesteramine and their respective variants
  • non-viral vectors such as micelles, emulsions, nanoparticles, and the like can be used.
  • it can be efficiently delivered intracellularly using known intracellular substance transfer peptides.
  • MVG peptide consisting of three amino acids according to the present invention not only increases the expression of BDNF without cytotoxicity in the hippocampal neuronal cell line but also increases the expression of BDNF in hippocampal and cortical tissues. Therefore, the small MVG peptides, such as the present invention, have a low molecular weight, easily cross the Blood Brain Barrier (BBB), and do not have cytotoxicity. Therefore, Alzheimer's disease, Parkinson's disease, depression, stroke, and Huntington's chorea It is very effective as a preventive and therapeutic agent for neurological diseases such as cerebral ischemic disease, neurodegenerative disease and diabetic neuropathy.
  • BBB Blood Brain Barrier
  • Figure 1 shows the results of finding the amino acid with the highest BDNF expression at each position of the peptide consisting of three amino acids (Methionine-Valine-Glycine) through the PS-SPCL (Positional Scanning-Synthetic Peptide Combinatorial Library) .
  • Figure 2 shows the results of mRNA expression of BDNF after treatment of MVG synthetic peptide of the present invention in rats (Hat), a hippocampal nerve cell line of rat (Rat) by concentration.
  • Figure 3 shows the results of protein expression of BDNF after treatment of the MVG synthetic peptide of the present invention to the rat hippocampal neuronal cell line H19-7 by concentration.
  • Figure 4 shows the results of BDNF protein expression by extracting the hippocampal tissue site after the rat intravenous injection of 0.1g / kg MVG synthetic peptide of the present invention.
  • Figure 5 shows the results of BDNF protein expression by extracting the cerebral cortical tissue site after the tail vein injection of 0.1V / kg MVG synthetic peptide of the present invention.
  • Figure 6 shows the cytotoxicity results of MVG synthetic peptides by MTT (3- (4,5-Dimethylthiazol-2-yl) -2,5-diphenyltetrazolium bromide) test.
  • FIG. 7 illustrates a method of performing a Y-maze test
  • FIG. 8 illustrates the number of times of entering the novel arm and the moving distance from the novel arm.
  • FIG. 9 illustrates a method of performing a Morris water maze test.
  • Figure 10 shows the time spent on finding the platform and the test stayed on the platform.
  • FIG. 11 shows BDNF expression in the hippocampus (FIG. 11 (a)) and the cerebral cortex (FIG. 11 (b)) of mice that underwent the Y-maze test and Morris water maze test.
  • FIG. 12 shows phosphorylation and TrkB expression of TrkB in hippocampus (FIG. 12 (a)) and cortex (FIG. 12 (b)) of mice that underwent Y-maze test and Morris water maze test. It is.
  • FIG. 13 shows the results of in vitro experiments confirming the anti-parkinson effect of MVG peptides that regulate BDNF expression.
  • PS-SPCL trimmer package stock was supported by the Peptide library support facility at Pohang University of Science and Technology for the discovery of peptides that increase BDNF expression.
  • PS-SPCL Pural Scanning-Synthetic Peptide Combinatorial Library
  • a peptide library technology used in the present invention is a peptide consisting of three amino acids, the first position of which is substituted with tyrosine in alanine except cysteine. And the remaining second and third positions are fixed in such a way as to identify the most effective amino acid sequence at each position, thereby finding the amino acid with the highest BNDF expression at each position.
  • HT22 cells 57 mouse hippocampal neuronal cell lines, HT22 cells, were placed in a 24-well plate (SPL) at a total of 57 by 5 ⁇ 10 4 per well for 12 hours at 37 ° C. in a 5% CO 2 incubator (Incubator, VISION). Incubated in the. After spin down of the PS-SPCL trimmer package stock, the tube was opened, tapped with 15 ⁇ l of distilled water (DW) for cell culture, and then spun down again. 15 ⁇ l of PS-SPCL trimmer package stock prepared on a 24-well plate in which HT22 cells are incubated is placed in each well and incubated in an incubator for 12 hours.
  • SPL 24-well plate
  • DW distilled water
  • ELISA Enzyme-linked immunosorbent assay, BDNF ELISA Kit, Promega
  • M Methionine
  • V Valine
  • G Glycine
  • an MVG synthetic peptide consisting of three amino acids was purchased from Peptron Co., Ltd. H19-7 cells, the hippocampal neuronal cell lines of rats purchased from the American Type Culture Collection (ATCC), were placed in 6 well plates (NUNC) at 10 6 per well and incubated in a 34 ° C., 5% CO 2 incubator for 12 hours. , MVG synthetic peptides were treated with 0.001, 0.01, 0.1, 1 ⁇ M and further incubated for 12 hours.
  • RNA was isolated from each sample according to the protocol of the Easy-blue Total RNA extraction kit (Intron), and then 1 ug total RNA, 1 ol oligo (dT) 18 , and the rest were processed using DW treated with DEPC (Diethylpyrocarbonate). The volume was added to the tube for PCR to 20 mu l and heated at 70 °C for 5 minutes.
  • Complementary DNA cDNA was prepared by transferring 20 ⁇ l of the tube heated with RT premix (Reverse transcriptase premix, Bioneer) tube and performing reverse transcription at 42 ° C. for 1 hour.
  • an MVG synthetic peptide consisting of three amino acids was purchased from Peptron Co., Ltd. H19-7 cells, rat hippocampal neuronal cell lines purchased from the American Type Culture Collection (ATCC), were placed in a 6 well plate (NUNC) at 10 6 per well and incubated in a 34 ° C., 5% CO 2 incubator for 12 hours. , MVG synthetic peptides were treated with 0.001, 0.01, 0.1, 1 ⁇ M and further incubated for 12 hours.
  • Example 2 Obtain protein in the same manner as in Example 1, make 15% acrylamide gel through 1.5 mm Western blotting gel caster (Gel caster, Bio-Rad) and load 30 ⁇ g sample 2 hours 30 minutes at 100V with an electrophoresis device (Electrophoresis Power supply, EPS 601, Amersham). Polyvinylidene Difluoride (PVDF) was used to transfer the gel (Mighty small transphor, Amersham) using an electrophoresis power supply (EP 301, GE healthcare) at 400 mA for 2 hours and 30 minutes.
  • PVDF Polyvinylidene Difluoride
  • mice Four-week-old male SD (Sprague-Dawley) rats were purchased from Orient Co., Ltd. and separated into three groups of experimental groups (MVG-administered group) and control group (1X PBS-administered group). Adaptation and stabilization were made. After one week, the experimental group was tail vein injection of 0.1 ⁇ g / kg of MVG and the control group of 0.1 ⁇ g / kg of 1 ⁇ PBS. After 12 hours, anesthesia was temporarily used in ether, and blood was collected from the heart after laparotomy. The brains of the rats were quickly extracted, rapidly cooled in liquid nitrogen, and stored at -80 ° C.
  • the RIPA buffer and protease inhibitor were prepared in the same ratio as in Example 1, and the hippocampal tissue was completely ground using a Homogenizer (RZR2020M, Heidolph). Centrifuge at 4 ° C and 2,000rpm for 30 minutes (Centrifuge, UNION 32R, Hanil Science Industrial Co., Ltd), transfer only the supernatant to 1.5ml tube, and then centrifuge for 30 minutes at 4 ° C, 9,000rpm ( Centrifuge, MICRO 17TR, Hanil Science Industrial Co., Ltd) was used to quantify the protein by Bradford assay once again transfer only the supernatant to 1.5ml tube.
  • a Homogenizer RZR2020M, Heidolph
  • a 15% acrylamide gel was made through a 1.5 mm Western blotting gel caster (Gel caster, Bio-Rad) and 30 ⁇ g of sample was loaded to obtain an electrophoresis power supply.
  • EPS 601, Amersham for 2 hours 30 minutes at 100V.
  • PVDF Polyvinylidene Difluoride
  • EP 301, GE healthcare electrophoresis power supply
  • TBST Tris-Buffered Saline Tween 20
  • Goat anti-rabbit IgG Horseradish Peroxidase Conjugate, SAB-300, Bioreagents Incubate for 50 minutes at a ratio of 1: 3000.
  • detection was performed with a chemiluminescent HRP substrate kit (Millipore).
  • the amount of BDNF and its precursor (precursor) form proBDNF in the experimental group compared to the control group was confirmed to increase compared to ⁇ -actin.
  • mice Four-week-old male SD (Sprague-Dawley) rats were purchased from Orient Co., Ltd. and separated into three groups of experimental groups (MVG-administered group) and control group (1X PBS-administered group). Adaptation and stabilization were made. After one week, the experimental group was tail vein injection of 0.1 ⁇ g / kg of MVG and the control group of 0.1 ⁇ g / kg of 1 ⁇ PBS. After 12 hours, anesthesia was temporarily used in ether, and blood was collected from the heart after laparotomy. The brains of the rats were quickly extracted, rapidly cooled in liquid nitrogen, and stored at -80 ° C.
  • RIPA buffer and protease inhibitor were prepared in the same ratio as in Example 1, and the cortical tissue was completely ground using a Homogenizer (RZR2020M, Heidolph). Centrifuge at 4 ° C and 2,000rpm for 30 minutes (Centrifuge, UNION 32R, Hanil Science Industrial Co., Ltd), transfer only the supernatant to 1.5ml tube, and then centrifuge for 30 minutes at 4 ° C, 9,000rpm ( Centrifuge, MICRO 17TR, Hanil Science Industrial Co., Ltd) was used to quantify the protein by Bradford assay once again transfer only the supernatant to 1.5ml tube.
  • a Homogenizer RZR2020M, Heidolph
  • a 15% acrylamide gel was made through a 1.5 mm Western blotting gel caster (Gel caster, Bio-Rad) and 30 ⁇ g of sample was loaded to obtain an electrophoresis power supply.
  • EPS 601, Amersham for 2 hours 30 minutes at 100V.
  • PVDF Polyvinylidene Difluoride
  • EP 301, GE healthcare electrophoresis power supply
  • TBST Tris-Buffered Saline Tween 20
  • Goat anti-rabbit IgG Horseradish Peroxidase Conjugate, SAB-300, Bioreagents Incubate for 50 minutes at a ratio of 1: 3000.
  • detection was performed with a chemiluminescent HRP substrate kit (Millipore).
  • the amount of BDNF and its precursor (precursor) form proBDNF in the experimental group was higher than that of the control group, compared to ⁇ -actin.
  • H19-7 cells were incubated in 96 well plates (FALCON) at 2 ⁇ 10 3/200 ⁇ l and incubated in 34 ° C., 5% CO 2 incubator for 12 hours, and then treated with MVG synthetic peptide from 10 nM to 1 mM and 3 days Incubated for After 3 days, 20 ⁇ l of MTT (50 mg / ml) solution was treated, wrapped in aluminum foil, and incubated again in an incubator. After 4 hours, the medium and the MTT solution were removed, treated with 200 ⁇ l of DMSO (Dimetyl sulfoxide) solution, wrapped in foil, and placed at room temperature for 4 hours.
  • MTT 50 mg / ml
  • DMSO Dimetyl sulfoxide
  • MVG synthetic peptide according to the present invention increases the protein expression of BDNF in hippocampal neuronal cell line, hippocampal tissue and cerebral cortical tissue, and because there is no cytotoxicity, Alzheimer's disease, Parkinson's disease, chronic stress depression, stroke, Huntington's chorea, It can be effectively used as a pharmaceutical composition for the prevention and treatment of cerebral ischemic diseases, neurodegenerative diseases, diabetic neuropathy and the like.
  • Y-maze TEST (FIG. 7A) shows that the first arm blocked the novel arm and started the rat for 15 minutes to explore the other arm, and then moved to the novel arm for 5 minutes and moved on the novel arm in the second trial. This was done by examining the distance to see if memory was improved.
  • the Morris water maze test (Fig. 9a), which measures spatial learning and memory, was performed.
  • the mice were dropped in a tank containing water, and the visual cues installed outside the tank were recognized and the location of the platform was known.
  • mice subjected to the Y-maze and Morris underwater maze test were extracted, and BDNF expression was compared using Western blotting. Experimental methods were the same as in Examples 4 and 5. As a result, it was confirmed that the expression of BDNF increased in the group injected with the MVG peptide compared to the control group, therefore, the learning and memory enhancement confirmed in the Y-maze and Morris underwater maze TEST was found in the hippocampus (Fig. 11a) and the cerebral cortex (Fig. It can be seen that the increase in BDNF expression by the MVG peptide in 11b).
  • a 15% acrylamide gel was made through a 1.5 mm Western blotting gel caster (Gel caster, Bio-Rad) and 30 ⁇ g of sample was loaded to obtain an electrophoresis power supply.
  • EPS 601, Amersham for 2 hours 30 minutes at 100V.
  • PVDF Polyvinylidene Difluoride
  • EP 301, GE healthcare electrophoresis power supply
  • TBST Tris-Buffered Saline Tween 20
  • TrkB phosphorylation and TrkB expression were increased in the group injected with MVG peptide in the hippocampus and cerebral cortex (FIG. 12A and FIG. 12B, respectively) and enhance spatial learning and memory by MVG peptide.
  • the increase in BDNF expression in the hippocampus may be attributed to the activation of the receptor and enhanced expression of the receptor.
  • MVG peptides that regulate BDNF expression.
  • MVG peptides were treated in SH-SY5Y cell line, a human dopaminergic cell line, at concentrations of 0.001, 0.01, 0.1, and 1 nM for 24 hours, and the expression of BDNF, an anti-apoptotic protein, Bcl-2, was confirmed by Western blotting.
  • Blocking PVDF for 1 hour with 7% skimmed milk (Skim milk, Difco TM Skim milk, BD), rabbit anti BDNF polyclonal antibody (AB1534, chemicon), mouse anti Bcl-2 Monoclonal antibody (Mouse anti Bcl-2 antibody, sc-7382, Santa Cruz) was reacted overnight at a ratio of 1: 1000.
  • Goat anti-rabbit IgG Horseradish Peroxidase Conjugate, SAB-300, Bioreagents, after washing for 1 hour with 0.05% TBST (Tris-Buffered Saline Tween 20) (3 times 20 min)
  • Goat anti-mouse IgG Horseradish Peroxidase Conjugate, sc-2005, Santa Cruz was incubated at a ratio of 1: 3000, 1: 1000 for 50 minutes. After washing for 1 hour with 0.05% TBST (3 times for 20 minutes), detection was performed with a chemiluminescent HRP substrate kit (Millipore).
  • the medium and the MTT solution were removed, treated with 200 ⁇ l of DMSO (Dimetyl sulfoxide) solution, wrapped in foil, and placed at room temperature for 4 hours.
  • the absorbance was measured at 570 nm using a microplate reader (Molecular device) to convert the control to 100% to calculate the value.
  • DMSO Dimetyl sulfoxide
  • the MVG peptide according to the present invention has an anti-parkinson effect, and thus may function as a preventive and therapeutic agent for Parkinson's disease.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • General Health & Medical Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)

Abstract

The present invention relates to a novel peptide for augmenting brain-derived neutrotrophic factor (BDNF) protein expression. More specifically, the present invention provides a novel peptide which can be used to prevent or treat neurological disorders such as Alzheimer's disease, Parkinson's disease, depression, stroke, Huntington's chorea, ischaemic disorders of the cerebrum, degenerative neurological disorders or diabetic neural disorders, by augmenting BDNF mRNA and protein expression in hippocampal and cerebral cortical tissue. A Methionine-Valine-Glycine (MVG) peptide comprising three different amino acids of the present invention augmented BDNF mRNA and protein expression in hippocampal neurons, hippocampal tissue and cerebral-cortex tissue; BDNF being an important functional substance in learning and memory, dopamine neuron protection and depression prevention.

Description

해마 신경 세포, 해마 조직 및 대뇌 피질 조직에서 BDNF의 단백질 발현을 증가시키는 신규 펩타이드Novel peptides increase protein expression of WNDF in hippocampal neurons, hippocampal tissues, and cerebral cortical tissues
본 발명은 신경세포의 성장과 분화 및 학습, 기억, 항우울증 등의 다양한 효과를 가진 BDNF의 단백질 발현을 증가시키는 신규 펩타이드에 관한 것이다. The present invention relates to novel peptides that increase protein expression of BDNF with various effects such as neuronal growth and differentiation and learning, memory, antidepression and the like.
신경 영양성 인자 (Neurotrophic factor)는 발생 과정 동안 신경 세포의 생존과 분화를 조절하며(Davies, 1994), 개체의 일생 동안 신경 구조의 유지 및 이온 채널의 활성, 신경 전달 물질 방출, 그리고 액손 경로 탐색 (axon path-finding) 등의 기능에 영향을 끼친다 (Schnell et al., 1994; Song and Poo, 1999; Schinder and Poo, 2000). 이러한 신경영양성 인자의 예로는 신경 성장 인자 (Nerve growth factor, NGF), 뇌 유래 신경영양 인자 (Brain-derived neurotrophic factor, BDNF), 뉴로트로핀-3 (Neurotrophin-3, NT-3) 그리고 뉴로트로핀-4/5 (Neurotrophin-4/5, NT-4/5)가 존재한다.Neurotrophic factor regulates the survival and differentiation of neurons during development (Davies, 1994), and maintains neural structure and activity of ion channels, neurotransmitter release, and axon pathways during the life of the individual (Davies, 1994). axon path-finding) (Schnell et al., 1994; Song and Poo, 1999; Schinder and Poo, 2000). Examples of such neurotrophic factors include nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), neurotrophin-3 (NT-3), and neurotrophs. Pin-4 / 5 (Neurotrophin-4 / 5, NT-4 / 5) is present.
신경 영양성 인자 중 BDNF는 중추신경계 (Central Nervous System, CNS)에서 학습 및 기억, 고등 사고 능력을 담당하는 해마, 피질 그리고 전뇌 기저부에 많이 발현되고 있다. 이러한 위치에서 발현된 BDNF는 시냅스 전달 및 학습과 기억 능력의 신경생화학적 근거가 되는 시냅스 가소성, 인식 과정의 주요한 조절자로 그 기능을 담당하고 있다 (Lu B, 2003). 특히 학습 및 기억 과정의 세포 현상인 장기 강화 작용 (Long-term potentiation, LTP)을 촉진시킬 뿐 아니라 이에 반대되는 현상인 장기 약화 작용 (Long-term depression, LTD)을 억제하는 역할을 한다 (Ikegaya, Y. et al., 2002; Huber, K. et al., 1998). 신경 영양성 인자 중에서도 이러한 유일무이한 역할은 BDNF와 그것의 수용체인 TrkB (tropomyosin-related kinase B)가 광범위하게 글루타메이트 시냅스에 분포하고 있다는 사실과 일치한다.Among the neurotrophic factors, BDNF is expressed in the hippocampus, cortex and basal brain which are responsible for learning, memory, and higher thinking in the central nervous system (CNS). BDNF expressed in this position is a major regulator of synaptic plasticity and recognition processes, which is the neurobiochemical basis for synaptic transmission and learning and memory (Lu B, 2003). Not only does it promote long-term potentiation (LTP), a cellular phenomenon in learning and memory processes, but it also inhibits long-term depression, LTD (opposite) (Ikegaya, Y. et al., 2002; Huber, K. et al., 1998). Among neurotrophic factors, this unique role is consistent with the fact that BDNF and its receptor, TrkB (tropomyosin-related kinase B), are widely distributed in glutamate synapses.
시냅스 전달 및 시냅스 가소성의 중요한 조절자인 BDNF는 다양한 질병과 연관성을 가지고 있다. 대표적인 예로 퇴행성 뇌질환인 알츠하이머 병 (Alzheimer's disease, AD), 파킨슨병 (Parkinson's disease, PD), 스트레스에 의한 우울증 (Depression), 뇌졸중, 헌팅톤무도병, 대뇌허혈성질환, 신경퇴화질환 또는 당뇨병성 신경병증 등을 들 수 있다.BDNF, an important regulator of synaptic transmission and synaptic plasticity, is associated with a variety of diseases. Typical examples include degenerative brain diseases such as Alzheimer's disease (AD), Parkinson's disease (PD), depression from stress, stroke, Huntington's chorea, cerebral ischemic disease, neurodegenerative diseases or diabetic neuropathy Etc. can be mentioned.
알츠하이머병은 전뇌의 콜린성 뉴런이 퇴화하여 신경 전달 물질인 아세틸콜린의 방출이 감소함으로써 인식 능력의 저하를 일으킨다 (Murer MG et al., 2001). BDNF의 경우, 전뇌기저부 콜린성 뉴런의 생존과 분화를 촉진시킬 뿐 아니라, 이러한 신경 세포에서 아세틸콜린의 방출을 자극하는 것으로 알려져 있다 (Knipper M et al., 1994). 따라서 BDNF의 합성 부족에 의한 세포의 항상성 악화가 알츠하이머병을 야기할 것이라고 제안되고 있다. 또한 신경가소성의 결핍 즉, 시냅스 접촉의 감소가 신경병리학적, 임상적 징후를 나타낸다고 알려져 있다 (Mesulam MM., 1999). 임상적인 결과에 의하면 알츠하이머병 환자의 사후 뇌 부검 결과, BDNF와 그것의 수용체인 TrkB의 발현이 학습 및 기억을 담당하는 해마, 피질 부위에서 유의하게 감소되었다 (Phillips HS et al., 1991; Holsinger RM et al., 2000; Allen SJ et al., 1999). BDNF는 이러한 부위에서 시냅스 가소성을 통한 학습 및 기억의 세포 과정의 현상인 LTP (Long-term potentiation)를 조절한다고 알려져 있다 (Figurov, A. et al., 1996). 따라서 BDNF의 발현 감소함에 의한 인식 및 기억 과정의 기능 저하가 알츠하이머병을 일으킬 것이라고도 생각되고 있다. Alzheimer's disease causes deterioration of cognitive ability by the degeneration of cholinergic neurons in the whole brain and a decrease in the release of the neurotransmitter acetylcholine (Murer MG et al., 2001). BDNF is known to promote the survival and differentiation of basal cholinergic neurons as well as to stimulate the release of acetylcholine in these neurons (Knipper M et al., 1994). Therefore, it is suggested that deterioration of homeostasis of cells due to lack of synthesis of BDNF will cause Alzheimer's disease. It is also known that neuroplastic deficiency, ie reduction in synaptic contact, presents neuropathological and clinical signs (Mesulam MM., 1999). Clinical results showed that post-mortem brain necropsy in Alzheimer's disease significantly reduced the expression of BDNF and its receptor, TrkB, in the hippocampus and cortex, which are responsible for learning and memory (Phillips HS et al., 1991; Holsinger RM). et al., 2000; Allen SJ et al., 1999). BDNF is known to regulate long-term potentiation (LTP), a phenomenon in cellular processes of learning and memory through synaptic plasticity at these sites (Figurov, A. et al., 1996). Therefore, it is also thought that a decrease in the function of the recognition and memory process by reducing the expression of BDNF will cause Alzheimer's disease.
파킨슨병은 흑질 도파민성 (substantia nigral dopaminergic) 신경세포의 수가 감소되게 되고 이는 선조체의 도파민 (striatal dopamine)의 고갈을 초래하여 운동 기능 장애와 같은 징후를 초래하게 된다. 이러한 파킨슨병 환자의 또 다른 특징으로 인식 기능이 퇴보한다는 것이다. 파킨슨병의 원인과 연관되고 있는 단백질로써, 신경 영양성 인자가 도파민성 뉴런의 보호에 중요한 역할을 할 것이라고 예상되고 있다 (Siegel GJ and Chauhan NB., 2000). 신경 영양성 인자 중에서도 BDNF와 도파민성 뉴런과의 상호작용에 대해서 잘 알려져 있다. 복측 중뇌 (Ventral midbrain), 흑질 그리고 배쪽 피개부 (ventral tegmental) 부위의 도파민성 뉴런은 BDNF을 발현하는데 (Seroogy KB et al., 1994), 흑질 부위에서 BDNF 발현을 감소시킨 결과, 흑질 도파민성 뉴런의 수가 현저하게 줄어들었다 (Porritt MJ et al., 2005). 또한 흑질의 도파민 뉴런의 수를 적정수준으로 유지하는데 BDNF가 필요하다는 보고 (Baquet ZC et al., 2005)와 실제 파킨슨 환자의 경우, 흑질선조체 도파민 뉴런의 BDNF 발현이 현저하게 감소된 결과 (MogiM et al., 1999; Howells DW et al., 2000) 등을 통하여 파킨슨병의 도파민성 뉴런의 감소가 BDNF 생합성의 결핍과 관계있다고 보고 되어지고 있다. Parkinson's disease results in a decrease in the number of substantia nigral dopaminergic neurons, leading to depletion of striatal dopamine in the striatum, leading to symptoms such as motor dysfunction. Another characteristic of Parkinson's disease patients is that the cognitive function is deteriorated. As a protein associated with the cause of Parkinson's disease, neurotrophic factors are expected to play an important role in the protection of dopaminergic neurons (Siegel GJ and Chauhan NB., 2000). Among neurotrophic factors, the interaction of BDNF with dopaminergic neurons is well known. Dopaminergic neurons in the ventral midbrain, melanoma and ventral tegmental areas express BDNF (Seroogy KB et al., 1994), resulting in decreased BDNF expression in the melanoma region. The number of is significantly reduced (Porritt MJ et al., 2005). In addition, we report that BDNF is necessary to maintain the appropriate number of dopamine neurons in the vaginal cells (Baquet ZC et al., 2005) and that in the case of Parkinson's patients, the BDNF expression of the stratified dopamine neurons is significantly reduced (MogiM et. al., 1999; Howells DW et al., 2000) reported that the reduction of dopaminergic neurons in Parkinson's disease is associated with a deficiency of BDNF biosynthesis.
많은 스트레스를 받으며 살아가는 현대 사회에 우울증 환자의 수는 급격하게 증가하고 있다. 스트레스는 신경 쇠약과 해마를 포함한 다양한 뇌 지역의 부피를 감소시킬 뿐 아니라 (Duman, R.S. and Monteggia, L.M. 2006) 해마의 BDNF mRNA 발현을 감소시키는 것으로 알려져 있다 (Duman, R.S. and Monteggia, L.M. 2006 Smith, M.A. et al., 1995). 실제 우울증 환자의 사후 뇌 부검 결과에서도 BDNF의 발현이 정상인에 비해 현저하게 감소되었고, 뇌 영상 (imaging) 결과에서도 해마의 부피가 유의하게 감소되었다 (Sheline, Y.I et al., 2003). 이러한 우울증을 치료하기 위해서 다양한 화학적 항울제들이 사용되고 있는데 공통적으로 해마, 전전뇌 피질 또는 두 지역에서 BDNF mRNA의 발현을 현저하게 증가시켰다 (Duman, R.S. and Monteggia, L.M. 2006). 이러한 결과들에 의해서 'neurotrophin hypothesis of depression'이라는 가설이 대두되어지고 있다. 즉, 스트레스에 의하여 해마나 전전뇌 피질의 감소된 BDNF의 발현을 다시 증가시킴으로써 신경쇠약과 세포수 감소를 회복시킴으로 우울증과 같은 정신질환을 치료하자는 것이다 (Duman, R.S. and Monteggia, L.M. 2006). The number of depressed patients is increasing rapidly in the modern society, which is heavily stressed. Stress is known to reduce the volume of various brain regions, including nervous breakdown and hippocampus (Duman, RS and Monteggia, LM 2006), as well as reducing BDNF mRNA expression in the hippocampus (Duman, RS and Monteggia, LM 2006 Smith, MA et al., 1995). In postmortem brain necropsy of depressed patients, the expression of BDNF was significantly reduced compared to normal subjects, and the volume of hippocampus was significantly reduced in brain imaging (Sheline, Y.I et al., 2003). Various chemical antidepressants have been used to treat these depressions. Commonly, the expression of BDNF mRNA is significantly increased in the hippocampus, anterior cerebral cortex or both regions (Duman, R.S. and Monteggia, L.M. 2006). These results suggest the hypothesis of 'neurotrophin hypothesis of depression'. In other words, the treatment of mental disorders such as depression by restoring neuronal breakdown and cell number reduction by re-increasing the expression of reduced BDNF in the hippocampus and the anterior cerebral cortex by stress (Duman, R.S. and Monteggia, L.M. 2006).
전술한 바와 같이 BDNF는 신경 구조의 유지 및 이온 채널의 활성, 신경전달물질의 방출뿐만 아니라 신경세포의 성장과 분화, 학습 및 기억, 항우울 등의 다양한 기능을 나타내고 있다. 이러한 다양한 기능과 더불어 BDNF는 알츠하이머병, 파킨슨병, 만성 스트레스 질환인 우울증, 뇌졸중 (Schabitz et al., Stroke, 38:2165-2172, 2007), 헌팅턴무도병 (Zuccato C et al., Science 293, 20, July 2001), 대뇌허혈성질환 (Han BH et al., the Journal of neuroscience, 2000, 20(15):5775-5781, August 1, 2000), 신경퇴화질환 (Tsuzaka K et al., Muscle Nerve. 24(4):474-80, April 2001) 또는 당뇨병성 신경병증 (Nitta A et al., Neurotoxicology and Teratology, 24:695-701, 2002) 등 주요 질병의 원인으로 밝혀지고 있다. 하지만 BDNF 그 자체는 14kDa의 거대분자 단백질로써 혈뇌장벽 (Blood-brain barrier, BBB)을 통과하지 못할 뿐 아니라 투여방법 등의 문제점을 지니고 있다. 따라서 혈뇌장벽을 쉽게 통과할 수 있을 뿐만 아니라 세포독성이 매우 적고, 적은 양을 투여하여 BDNF의 발현을 증가시키는 물질을 발굴함으로써 알츠하이머병, 파킨슨병, 만성스트레스질환인 우울증, 뇌졸중 등의 신경질환의 예방 및 치료에 활용 가능하다.As described above, BDNF exhibits various functions such as neuronal cell growth and differentiation, learning and memory, and antidepressant as well as maintenance of neural structure, activation of ion channels, and release of neurotransmitters. Along with these diverse functions, BDNF can be used for Alzheimer's disease, Parkinson's disease, depression, stroke (Schabitz et al., Stroke, 38: 2165-2172, 2007), and Huntington's chorea (Zuccato C et al., Science 293, 20). , July 2001), cerebral ischemic disease (Han BH et al., The Journal of neuroscience, 2000, 20 (15): 5775-5781, August 1, 2000), neurodegenerative diseases (Tsuzaka K et al., Muscle Nerve. 24 (4): 474-80, April 2001) or diabetic neuropathy (Nitta A et al., Neurotoxicology and Teratology, 24: 695-701, 2002). However, BDNF itself is a 14kDa macromolecular protein that does not cross the blood-brain barrier (BBB) and has problems such as administration method. Therefore, it is easy to cross the blood brain barrier and has very low cytotoxicity, and by discovering a substance that increases the expression of BDNF by administering a small amount, it is possible to detect neurological diseases such as Alzheimer's disease, Parkinson's disease, depression, stroke, which are chronic stress diseases. It can be used for prevention and treatment.
본 발명자들은 PS-SPCL (Positional Scanning-Synthetic Peptide Combinatorial Library)이라는 펩타이드 라이브러리 기술을 통해 각 위치에서 BDNF의 발현이 가장 많은 아미노산을 선정하여 이를 재합성한 세 개의 아미노산으로 이루어진 MVG 펩타이드를 제조하였고, 이 MVG 펩타이드에 의하여 해마신경세포, 해마조직 및 대뇌 피질조직에서 BDNF mRNA 및 단백질 발현이 증가됨을 확인함으로써 본 발명을 완성하였다. The present inventors prepared an MVG peptide consisting of three amino acids by selecting the amino acids with the highest expression of BDNF at each position through a peptide library technology called PS-SPCL (Positional Scanning-Synthetic Peptide Combinatorial Library). The present invention was completed by confirming that BDNF mRNA and protein expression is increased in hippocampal neurons, hippocampal tissues and cerebral cortical tissues by MVG peptide.
본 발명의 목적은 해마신경세포, 해마조직 및 대뇌 피질조직에서 BDNF (Brain-derived neurotrophic factor)의 mRNA 발현을 증가시키는 펩타이드를 제공하는 것이다. It is an object of the present invention to provide peptides that increase mRNA expression of Brain-derived neurotrophic factor (BDNF) in hippocampal neurons, hippocampal tissues and cerebral cortical tissues.
본 발명의 다른 목적은 해마신경세포, 해마조직 및 대뇌 피질조직에서 BDNF (Brain-derived neurotrophic factor)의 단백질 발현을 증가시키는 펩타이드를 제공하는 것이다. Another object of the present invention is to provide a peptide for increasing protein expression of brain-derived neurotrophic factor (BDNF) in hippocampal neurons, hippocampal tissues and cerebral cortical tissues.
본 발명의 또 다른 목적은 상기 펩타이드의 치료학적 유효량을 포함하는 신경질환의 예방 및 치료용 약학 조성물을 제공하는 것이다. Another object of the present invention to provide a pharmaceutical composition for the prevention and treatment of neurological diseases comprising a therapeutically effective amount of the peptide.
상기와 같은 목적을 달성하기 위해, 본 발명은 서열번호 1에 기재된 아미노산 (MVG; Methionine-Valine-Glycine)으로 구성되는 펩타이드(이하, 'MVG 펩타이드'라 함)를 제공한다. In order to achieve the above object, the present invention provides a peptide consisting of the amino acid (MVG; Methionine-Valine-Glycine) described in SEQ ID NO: 1 (hereinafter referred to as 'MVG peptide').
본 발명의 펩타이드는 해마신경세포, 해마조직 및 대뇌 피질조직에서 BDNF (Brain-derived neurotrophic factor) mRNA의 발현 및/또는 BDNF (Brain-derived neurotrophic factor)의 단백질 발현을 증가시키며, TrkB의 단백질 발현 및 인산화를 증가시킬 수 있다. The peptide of the present invention increases the expression of Brain-derived neurotrophic factor (BDNF) mRNA and / or the protein expression of Brain-derived neurotrophic factor (BDNF) in hippocampal neurons, hippocampus and cerebral cortex, and expresses protein expression of TrkB and May increase phosphorylation.
또한, 본 발명은 상기 펩타이드를 코딩하는 DNA 서열로 구성된 폴리뉴클레오티드를 제공한다. 상기 폴리뉴클레오티드는 등가의 핵산서열, 즉 DNA 서열은 상이하나 동일한 서열의 MVG 펩타이드를 암호화하는 코돈 축퇴성 (codon degeneracy) 서열을 포함한다. The present invention also provides a polynucleotide consisting of a DNA sequence encoding the peptide. The polynucleotide comprises an equivalent nucleic acid sequence, that is, a codon degeneracy sequence that encodes MVG peptides of different but identical sequences.
구체적으로, 본 발명자들은 PS-SPCL (Positional Scanning-Synthetic Peptide Combinatorial Library)이라는 펩타이드 라이브러리 기술을 통해 각 위치에서 BDNF의 발현이 가장 많은 아미노산을 선정하여 (MVG; Methionine-Valine-Glycine) 이를 재합성한 후, 세포 수준에서 실질적으로 발현이 증가하는지 RT-PCR과 웨스턴 블라팅 (Western blotting)으로 다시 확인하였다. 또한 MVG 펩타이드가 세포 수준에서 독성을 나타내는지 알아보기 위해서 MTT (3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) 검사를 수행하였다. 실질적인 동물 수준에서 BDNF의 발현이 증가하는지 확인하기 위해 0.1㎍/㎏의 펩타이드를 꼬리 정맥을 통하여 주사 한 후, 해마와 대뇌 피질 부위를 적출하여 웨스턴 블라팅 (Western blotting)을 시행하였다.  Specifically, the present inventors selected the amino acid with the highest expression of BDNF at each position through a peptide library technology called PS-SPCL (Positional Scanning-Synthetic Peptide Combinatorial Library) (MVG; Methionine-Valine-Glycine) Afterwards, it was again confirmed by RT-PCR and Western blotting that expression increased substantially at the cellular level. In addition, MTT (3- (4,5-Dimethylthiazol-2-yl) -2,5-diphenyltetrazolium bromide) test was performed to determine whether MVG peptide is toxic at the cellular level. To confirm the expression of BDNF at the actual animal level, 0.1 μg / kg of peptide was injected through the tail vein, and the hippocampus and cerebral cortex were removed and Western blotting was performed.
그 결과, 본 발명자들은 세 개의 아미노산으로 이루어진 MVG 펩타이드가 해마 신경세포주 뿐만 아니라 해마 조직 및 대뇌 피질 조직에서도 BDNF 발현의 증가를 유도한다는 사실을 확인하였고, 세포 수준에서 1mM까지는 세포 독성이 나타나지 않음을 확인하였다. 이하, 본 발명을 더욱 상세히 설명하면 다음과 같다. As a result, the present inventors confirmed that the MVG peptide consisting of three amino acids induced an increase in BDNF expression not only in hippocampal neuronal cell line but also in hippocampal tissue and cerebral cortical tissue, and showed no cytotoxicity up to 1 mM at the cellular level. It was. Hereinafter, the present invention will be described in more detail.
한 구체예에서, 본 발명자들은 마우스 (Mouse)의 해마신경세포주인 HT22 세포를 24 웰 플레이트 (well plate)에 웰당 5x104씩 깔고 12시간 인큐베이션 (incubation) 한 후, 준비된 PS-SPCL 스톡 (stock)을 각 웰에 처리하였다. 12시간 후 단백질 분해효소 저해제가 첨가된 RIPA (Radio-immunoprecipitation Assay) 완충액 (buffer)을 이용하여 단백질을 얻어서 ELISA (Enzyme-Linked ImmunoSorbent Assay)을 통해서 BDNF양을 측정하였다. 그 결과, 도 1에 나타난 바와 같이 첫 번째 위치에서는 M (Methionine), 두 번째 위치에서는 V (Valine), 세 번째 위치에서는 G (Glycine)가 BDNF을 가장 많이 발현시켰다.In one embodiment, the present inventors incubated HT22 cells, a mouse hippocampal neuronal cell line, in a 24 well plate at 5 × 10 4 per well for 12 hours, and then prepared PS-SPCL stock (stock). Was treated to each well. After 12 hours, the protein was obtained using RIPA (Radio-immunoprecipitation Assay) buffer to which the protease inhibitor was added, and the amount of BDNF was measured through Enzyme-Linked ImmunoSorbent Assay (ELISA). As a result, as shown in FIG. 1, M (Methionine) at the first position, V (Valine) at the second position, and G (Glycine) at the third position expressed the most BDNF.
또 다른 구체예에서, 상기 결과를 통해 각각의 위치에서 BDNF의 발현이 가장 많은 아미노산 (MVG)으로 구성된 펩타이드를 재합성하여 래트의 해마신경세포주인 H19-7 세포에 처리하여 BDNF mRNA의 발현 증가를 RT-PCR로 확인하였다. 그 결과, 도 2에 나타난 바와 같이 대조군에 비해서 0.001, 0.01, 0.1μM의 MVG 펩타이드를 처리한 실험군에서 BDNF mRNA 발현이 농도 의존적 (Dose-dependent)으로 증가하였다. In another embodiment, the results show that the peptide composed of the amino acid (MVG) having the highest expression of BDNF at each position is resynthesized and treated to H19-7 cells of rat hippocampal neuronal cell line to increase the expression of BDNF mRNA. It was confirmed by RT-PCR. As a result, BDNF mRNA expression was increased in a dose-dependent manner in the experimental group treated with 0.001, 0.01, 0.1 μM MVG peptide as shown in FIG.
또 다른 구체예에서, 해마신경세포주인 H19-7 세포에 MVG 펩타이드를 처리하여 BDNF 단백질 발현을 웨스턴 블라팅으로 확인하였다. 그 결과 도 3에 나타난 바와 같이 대조군에 비해서 0.001, 0.01, 0.1μM의 MVG 펩타이드를 처리한 실험군에서 BDNF 단백질 발현이 농도 의존적 (Dose-dependent)으로 증가하였다. In another embodiment, the BDNF protein expression was confirmed by Western blotting by treating the hippocampal neuronal H19-7 cells with MVG peptide. As a result, as shown in FIG. 3, BDNF protein expression was increased in a dose-dependent manner in the experimental group treated with 0.001, 0.01, and 0.1 μM MVG peptide as compared to the control group.
또 다른 구체예에서, 수컷 SD (Sprague-Dawely) 래트 (Rat) 4주령을 구입하여, 1주일간 안정화 과정을 거친 후, 1X PBS (대조군)와 상기에서 결정된 MVG 펩타이드를 꼬리 정맥을 통하여 0.1㎍/㎏씩 주사하고 해마 조직과 대뇌 피질조직을 적출하여 웨스턴 블라팅을 시행하였다. 그 결과, 도 4, 5에 나타난 바와 같이 대조군에 비해 실험군에서 BDNF와 proBDNF 단백질의 발현이 증가한 것을 확인하였다.  In another embodiment, male SD (Sprague-Dawely) rats (4 weeks of age) are purchased and subjected to stabilization for 1 week, after which 1X PBS (control) and the MVG peptides determined above are 0.1 μg / Kg injections and hippocampal and cerebral cortical tissues were performed and Western blotting was performed. As a result, it was confirmed that the expression of BDNF and proBDNF protein in the experimental group increased as shown in Figure 4, 5 compared with the control group.
또 다른 구체예에서, MVG 펩타이드의 세포 독성을 확인하기 위해 H19-7 세포에 10nM부터 1mM까지 MVG 펩타이드를 처리한 후 MTT 용액을 통하여 세포의 생존율을 확인한 결과 대조군과 유의한 차이가 나타나지 않았음을 확인하였다 (도6).  In another embodiment, H19-7 cells were treated with MVG peptides from 10nM to 1mM to confirm cytotoxicity of the MVG peptides, and then the survival rate of the cells was confirmed by MTT solution. It was confirmed (FIG. 6).
또 다른 구체예에서, MVG 펩타이드가 실험동물수준에서 공간 학습 및 기억력을 증진시키는지를 확인하기 위해서 공간 기억 테스트 중 하나인 Y-maze test 및 Morris water maze test 를 수행한 결과, MVG펩타이드가 공간 학습 및 기억을 증진시킴을 동물실험을 통하여 확인할 수 있었으며(도 8, 도 10), 해마(도 11a) 및 대뇌피질(도 11b)에서의 MVG 펩타이드에 의한 BDNF 발현 증가를 확인하였다. 또한 해마 및 대뇌피질에서 MVG 펩타이드를 주사한 그룹에서 TrkB의 인산화 및 TrkB의 발현이 증가함을 확인함으로써, MVG 펩타이드에 의한 공간 학습 및 기억 증진이 해마의 BDNF 발현 증가에 따라 그 수용체(TrkB)의 활성화 및 수용체의 발현 증진에 의한 것임을 알 수 있다.(도 12a,도 12b)In another embodiment, one of the spatial memory tests, the Y-maze test and the Morris water maze test, was performed to determine whether the MVG peptides promote spatial learning and memory at the experimental animal level. Enhancement of memory was confirmed through animal experiments (FIG. 8, FIG. 10), and increased BDNF expression by MVG peptides in hippocampus (FIG. 11A) and cerebral cortex (FIG. 11B). In addition, TrkB phosphorylation and TrkB expression were increased in the group injected with MVG peptides in the hippocampus and cerebral cortex, so that spatial learning and memory enhancement by MVG peptide increased the expression of its receptor (TrkB) as the hippocampus increased BDNF expression. It can be seen that the activation and the expression of the receptor is enhanced (Fig. 12a, 12b).
본 발명은 본 발명의 MVG 펩타이드의 치료학적 유효량을 포함하는 신경질환의 예방 및 치료용 약학 조성물을 제공한다. The present invention provides a pharmaceutical composition for the prevention and treatment of neurological diseases comprising a therapeutically effective amount of the MVG peptide of the present invention.
또한 본 발명은 본 발명의 MVG 펩타이드를 코딩하는 DNA 서열로 구성된 폴리뉴클레오티드를 포함하는 유전자 전달체를 제공하며, 본 발명의 유전자 전달체의 경우 신경질환의 예방 및 치료용 약학 조성물로서 작용할 수 있다.The present invention also provides a gene carrier comprising a polynucleotide consisting of a DNA sequence encoding the MVG peptide of the present invention, the gene carrier of the present invention can act as a pharmaceutical composition for the prevention and treatment of neurological diseases.
상기 신경질환의 예로는 알츠하이머병, 파킨슨병, 만성스트레스성 우울증, 뇌졸중, 헌팅톤무도병, 대뇌허혈성질환, 신경퇴화질환 또는 당뇨병성 신경병증 등이 있으며, 이것에 한정되는 것은 아니다. 일반적으로, 상기 약학 조성물은 투여방법에 따라 상용되는 약학적으로 허용되는 무독성의 담체를 본 발명의 MVG 펩타이드와 혼합하여 여러 가지 형태의 약제형으로 제조되어지며, 제조된 약학 조성물은 전술한 신경질환의 예방제 및 치료제로서 투여될 수 있다.Examples of the neurological diseases include Alzheimer's disease, Parkinson's disease, chronic stress depression, stroke, Huntington's chorea, cerebral ischemic disease, neurodegenerative disease or diabetic neuropathy, and the like. In general, the pharmaceutical composition is prepared in various forms by mixing a pharmaceutically acceptable non-toxic carrier that is commercially available according to the method of administration with the MVG peptide of the present invention, the prepared pharmaceutical composition is a neurological disease described above It can be administered as a prophylactic and therapeutic agent of.
본 발명의 일 구체예에서, 본 발명의 MVG 펩타이드가 anti-parkinson 효과를 가지는지 확인하기 위하여 MVG 펩타이드를 human dopaminergic cell line인 SH-SY5Y 세포주에 24시간 동안 0.001, 0.01, 0.1, 1nM 처리하고 BDNF, anti-apoptotic protein인 Bcl-2의 발현을 웨스턴 블라팅한 결과, 24h 동안 0.1nM의 MVG 펩타이드를 처리한 상태에서 BDNF, Bcl-2의 발현이 대조군에 비해 증가함을 확인하였다. 또한, SH-SY5Y 세포주에 MVG 펩타이드를 0.1nM로 24h 동안 선 처리후 (pretreatment) 2.5mM의 MPP+ 을 처리하여, MPP+ 처리 12h, 24h 후에 MTT assay을 통해 Cell viability를 측정한 결과, MVG 펩타이드를 선처리한 그룹에서 유의하게 cell viability가 높음을 확인함으로써 본 발명의 MVG 펩타이드는 파킨슨 병의 예방 및 치료용 조성물로 유용하게 활용될 수 있음을 입증하였다.(도 13)In one embodiment of the present invention, to determine whether the MVG peptide of the present invention has an anti-parkinson effect, MVG peptide was treated with 0.001, 0.01, 0.1, 1 nM for 24 hours in SH-SY5Y cell line, a human dopaminergic cell line, and BDNF. Western blot expression of the anti-apoptotic protein, Bcl-2, was found to increase BDNF and Bcl-2 expression in the presence of 0.1 nM MVG peptide for 24h. In addition, MVG peptide was pretreated with 0.1 nM for 24 h in a SH-SY5Y cell line and treated with 2.5 mM MPP + for 24 h, and MVG peptide was measured by MTT assay 12 h and 24 h after MPP + treatment. By confirming that the cell viability was significantly high in the group pretreated, it was proved that the MVG peptide of the present invention can be usefully used as a composition for preventing and treating Parkinson's disease (FIG. 13).
BDNF와 알츠하이머병, 파킨슨병, 만성스트레스성 우울증, 뇌졸중, 헌팅톤무도병, 대뇌허혈성질환, 신경퇴화질환 또는 당뇨병성 신경병증의 관계는 하기 나열된 문헌에 충분히 개시되어 있고 하기 문헌들은 본 발명에 참조되고 통합되어, 따라서, 본 발명의 BDNF (Brain-derived neurotrophic factor)의 mRNA 및 단백질 발현을 증가시키는 MVG 펩타이드의 상기 질병의 예방 및 치료용 약학 조성물로서의 용도는 본 발명에서의 일 기술적 사상이다.The relationship between BDNF and Alzheimer's disease, Parkinson's disease, chronic stress depression, stroke, Huntington's chorea, cerebral ischemic disease, neurodegenerative disease or diabetic neuropathy is fully disclosed in the documents listed below and the following references are incorporated herein by reference. Integrated, therefore, the use of MVG peptides to increase mRNA and protein expression of the brain-derived neurotrophic factor (BDNF) of the present invention as a pharmaceutical composition for the prevention and treatment of the disease is one technical idea in the present invention.
BDNF와 Alzheimer s disease (AD)BDNF and Alzheimer's disease (AD)
(1) Neuroprotective effects of brain-derived neurotrophic factor in rodent and primate models of Alzheimer s disease. (1) Neuroprotective effects of brain-derived neurotrophic factor in rodent and primate models of Alzheimer s disease.
Alan H Nagahara, David A Merrill, Giovanni Coppola, Shingo Tsukada, Brock E Schroeder, Gideon M Shaked, Ling Wang, Armin Blesch, Albert Kim, James M Conner, Edward Rockenstein, Moses V Chao, Edward H Koo, Daniel Geschwind, Eliezer Masliah, Andrea A Chiba,Alan H Nagahara, David A Merrill, Giovanni Coppola, Shingo Tsukada, Brock E Schroeder, Gideon M Shaked, Ling Wang, Armin Blesch, Albert Kim, James M Conner, Edward Rockenstein, Moses V Chao, Edward H Koo, Daniel Geschwind, Eliezer Masliah, Andrea A Chiba,
Mark H Tuszynski. NATURE MEDICINE LETTERS. 2009 Mar;15(3):331-7.Mark H Tuszynski. NATURE MEDICINE LETTERS. 2009 Mar; 15 (3): 331-7.
(2) The expanding role of BDNF: a therapeutic target for Alzheimer's disease? Fumagalli, F., Racagni, G. and Riva, M. A. (2006) Pharmacogenomics J, 6, 8-15. (2) The expanding role of BDNF: a therapeutic target for Alzheimer's disease? Fumagalli, F., Racagni, G. and Riva, MA (2006) Pharmacogenomics J, 6, 8-15.
(3)(3) New insights into brain BDNF function in normal aging and Alzheimer disease.New insights into brain BDNF function in normal aging and Alzheimer disease.
Tapia-Arancibia L, Aliaga E, Silhol M, Arancibia S.Brain Res Rev. 2008 Nov;59(1):201-20. Epub 2008 Aug 3. Review.Tapia-Arancibia L, Aliaga E, Silhol M, Arancibia S. Brain Res Rev. 2008 Nov; 59 (1): 201-20. Epub 2008 Aug 3. Review.
(4)(4) Protective effect of BDNF against beta-amyloid induced neurotoxicity in vitro and in vivo in rats.Protective effect of BDNF against beta-amyloid induced neurotoxicity in vitro and in vivo in rats.
Arancibia S, Silhol M, Mouli re F, Meffre J, H llinger I, Maurice T, Tapia-Arancibia L.Arancibia S, Silhol M, Mouli re F, Meffre J, H llinger I, Maurice T, Tapia-Arancibia L.
Neurobiol Dis. 2008 Sep;31(3):316-26. Epub 2008 Jul 15.Neurobiol Dis. 2008 Sep; 31 (3): 316-26. Epub 2008 Jul 15.
(5)(5) Association study of the decreased serum BDNF concentrations in amnestic mild cognitive impairment and the Val66Met polymorphism in Chinese Han.Association study of the decreased serum BDNF concentrations in amnestic mild cognitive impairment and the Val66Met polymorphism in Chinese Han.
Yu H, Zhang Z, Shi Y, Bai F, Xie C, Qian Y, Yuan Y, Deng L.Yu H, Zhang Z, Shi Y, Bai F, Xie C, Qian Y, Yuan Y, Deng L.
J Clin Psychiatry. 2008 Jul;69(7):1104-11.J Clin Psychiatry. 2008 Jul; 69 (7): 1104-11.
(6)(6) Increase of BDNF serum concentration during donepezil treatment of patients with early Alzheimer's disease.Increase of BDNF serum concentration during donepezil treatment of patients with early Alzheimer's disease.
Leyhe T, Stransky E, Eschweiler GW, Buchkremer G, Laske C.Leyhe T, Stransky E, Eschweiler GW, Buchkremer G, Laske C.
Eur Arch Psychiatry Clin Neurosci. 2008 Mar;258(2):124-8. Epub 2007 Nov 7.Eur Arch Psychiatry Clin Neurosci. 2008 Mar; 258 (2): 124-8. Epub 2007 Nov 7.
(7) (7) [Protective effect of adeno-associated viral vector-mediated expression of human brain-derived neurotrophic factor in rat neurons against beta-amyloid-induced Alzheimer's disease in vitro][Protective effect of adeno-associated viral vector-mediated expression of human brain-derived neurotrophic factor in rat neurons against beta-amyloid-induced Alzheimer's disease in vitro]
Liu ZH, Ma DL, Jin H, Ma YB, Hu HT.Liu ZH, Ma DL, Jin H, Ma YB, Hu HT.
Nan Fang Yi Ke Da Xue Xue Bao. 2006 Oct;26(10):1388-93. Chinese. Nan Fang Yi Ke Da Xue Xue Bao. 2006 Oct; 26 (10): 1388-93. Chinese.
(8) (8) The brain-derived neurotrophic factor gene as a possible susceptibility candidate for Alzheimer's disease in a chinese population.The brain-derived neurotrophic factor gene as a possible susceptibility candidate for Alzheimer's disease in a chinese population.
Tsai SJ, Hong CJ, Liu HC, Liu TY, Liou YJ.Tsai SJ, Hong CJ, Liu HC, Liu TY, Liou YJ.
Dement Geriatr Cogn Disord. 2006;21(3):139-43. Epub 2006 Jan 2.Dement Geriatr Cogn Disord. 2006; 21 (3): 139-43. Epub 2006 Jan 2.
(9) (9) Precursor form of brain-derived neurotrophic factor and mature brain-derived neurotrophic factor are decreased in the pre-clinical stages of Alzheimer's disease.Precursor form of brain-derived neurotrophic factor and mature brain-derived neurotrophic factor are decreased in the pre-clinical stages of Alzheimer's disease.
Peng S, Wuu J, Mufson EJ, Fahnestock M.Peng S, Wuu J, Mufson EJ, Fahnestock M.
J Neurochem. 2005 Jun;93(6):1412-21.J Neurochem. 2005 Jun; 93 (6): 1412-21.
(10) 10 Decreased levels of BDNF protein in Alzheimer temporal cortex are independent of BDNF polymorphisms.Decreased levels of BDNF protein in Alzheimer temporal cortex are independent of BDNF polymorphisms.
Lee J, Fukumoto H, Orne J, Klucken J, Raju S, Vanderburg CR, Irizarry MC, Hyman BT, Ingelsson M.Lee J, Fukumoto H, Orne J, Klucken J, Raju S, Vanderburg CR, Irizarry MC, Hyman BT, Ingelsson M.
Exp Neurol. 2005 Jul;194(1):91-6.Exp Neurol. 2005 Jul; 194 (1): 91-6.
BDNF와 Depression and anxiety BDNF and Depression and anxiety
(1) New insights into BDNF function in depression and anxiety. (1) New insights into BDNF function in depression and anxiety.
Keri Martinowich, Husseini Manji, Bai Lu. NATURE NEUROSCIENCE. VOLUME 10 NUMBER 9 SEPTEMBER 2007.Keri Martinowich, Husseini Manji, Bai Lu. NATURE NEUROSCIENCE. VOLUME 10 NUMBER 9 SEPTEMBER 2007.
(2)(2) Brain-derived neurotrophic factor, stress and depression: a minireview.Brain-derived neurotrophic factor, stress and depression: a minireview.
Yulu B, Ozan E, Gonul AS, Kilic E.Yulu B, Ozan E, Gonul AS, Kilic E.
Brain Res Bull. 2009 Mar 30;78(6):267-9. Epub 2008 Dec 26. Review.Brain Res Bull. 2009 Mar 30; 78 (6): 267-9. Epub 2008 Dec 26. Review.
(3)(3) Brain-derived neurotrophic factor and antidepressant action: another piece of evidence from pharmacogenetics.Brain-derived neurotrophic factor and antidepressant action: another piece of evidence from pharmacogenetics.
Tsai SJ, Hong CJ, Liou YJ.Tsai SJ, Hong CJ, Liou YJ.
Pharmacogenomics. 2008 Sep;9(9):1353-8. Review.Pharmacogenomics. 2008 Sep; 9 (9): 1353-8. Review.
(4) (4) Acute hippocampal brain-derived neurotrophic factor restores motivational and forced swim performance after corticosterone.Acute hippocampal brain-derived neurotrophic factor restores motivational and forced swim performance after corticosterone.
Gourley SL, Kiraly DD, Howell JL, Olausson P, Taylor JR.Gourley SL, Kiraly DD, Howell JL, Olausson P, Taylor JR.
Biol Psychiatry. 2008 Nov 15;64(10):884-90. Epub 2008 Aug 3.Biol Psychiatry. 2008 Nov 15; 64 (10): 884-90. Epub 2008 Aug 3.
(5)(5) Targeting TrkB neurotrophin receptor to treat depression.Targeting TrkB neurotrophin receptor to treat depression.
Rantam ki T, Castr n E.Rantam ki T, Castr n E.
Expert Opin Ther Targets. 2008 Jun;12(6):705-15. Review.Expert Opin Ther Targets. 2008 Jun; 12 (6): 705-15. Review.
BDNF와 Parkinson s disease (PD) BDNF and Parkinson's disease (PD)
(1)Shedding light into the role of BDNF in the pharmacotherapy of Parkinson's disease.Fumagalli, F., Racagni, G. and Riva, M. A. (2006) Pharmacogenomics J, 6, 95-104. (1) Shedding light into the role of BDNF in the pharmacotherapy of Parkinson's disease. Fumagalli, F., Racagni, G. and Riva, MA (2006) Pharmacogenomics J, 6, 95-104.
(2) (2) Neurotrophic factors as a therapeutic target for Parkinson's disease.Neurotrophic factors as a therapeutic target for Parkinson's disease.
Evans JR, Barker RA.Evans JR, Barker RA.
Expert Opin Ther Targets. 2008 Apr;12(4):437-47. Review.Expert Opin Ther Targets. 2008 Apr; 12 (4): 437-47. Review.
(3)(3) Rasagiline promotes regeneration of substantia nigra dopaminergic neurons in post-MPTP-induced Parkinsonism via activation of tyrosine kinase receptor signaling pathway.Rasagiline promotes regeneration of substantia nigra dopaminergic neurons in post-MPTP-induced Parkinsonism via activation of tyrosine kinase receptor signaling pathway.
Mandel SA, Sagi Y, Amit T.Mandel SA, Sagi Y, Amit T.
Neurochem Res. 2007 Oct;32(10):1694-9. Epub 2007 Aug 16. Review.Neurochem Res. 2007 Oct; 32 (10): 1694-9. Epub 2007 Aug 16. Review.
(4) (4) Neuroprotective effect of BDNF in young and aged 6-OHDA treated rat model of Parkinson disease.Neuroprotective effect of BDNF in young and aged 6-OHDA treated rat model of Parkinson disease.
Singh S, Ahmad R, Mathur D, Sagar RK, Krishana B.Singh S, Ahmad R, Mathur D, Sagar RK, Krishana B.
Indian J Exp Biol. 2006 Sep;44(9):699-704.Indian J Exp Biol. 2006 Sep; 44 (9): 699-704.
(5) (5) Valproate protects dopaminergic neurons in midbrain neuron/glia cultures by stimulating the release of neurotrophic factors from astrocytes.Valproate protects dopaminergic neurons in midbrain neuron / glia cultures by stimulating the release of neurotrophic factors from astrocytes.
Chen PS, Peng GS, Li G, Yang S, Wu X, Wang CC, Wilson B, Lu RB, Gean PW, Chuang DM, Hong JS.Chen PS, Peng GS, Li G, Yang S, Wu X, Wang CC, Wilson B, Lu RB, Gean PW, Chuang DM, Hong JS.
Mol Psychiatry. 2006 Dec;11(12):1116-25. Epub 2006 Sep 12.Mol Psychiatry. 2006 Dec; 11 (12): 1116-25. Epub 2006 Sep 12.
(6) (6) Neurotrophic factors stabilize microtubules and protect against rotenone toxicity on dopaminergic neurons.Neurotrophic factors stabilize microtubules and protect against rotenone toxicity on dopaminergic neurons.
Jiang Q, Yan Z, Feng J.Jiang Q, Yan Z, Feng J.
J Biol Chem. 2006 Sep 29;281(39):29391-400. Epub 2006 Aug 3.J Biol Chem. 2006 Sep 29; 281 (39): 29391-400. Epub 2006 Aug 3.
(7) (7) Repeated l-DOPA treatment increases c-fos and BDNF mRNAs in the subthalamic nucleus in the 6-OHDA rat model of Parkinson's disease.Repeated l-DOPA treatment increases c-fos and BDNF mRNAs in the subthalamic nucleus in the 6-OHDA rat model of Parkinson's disease.
Zhang X, Andren PE, Svenningsson P.Zhang X, Andren PE, Svenningsson P.
Brain Res. 2006 Jun 20;1095(1):207-10. Epub 2006 May 24.Brain Res. 2006 Jun 20; 1095 (1): 207-10. Epub 2006 May 24.
(8)(8) Brain-derived growth factor and glial cell line-derived growth factor use distinct intracellular signaling pathways to protect PD cybrids from H2O2-induced neuronal death.Brain-derived growth factor and glial cell line-derived growth factor use distinct intracellular signaling pathways to protect PD cybrids from H2O2-induced neuronal death.
Onyango IG, Tuttle JB, Bennett JP Jr.Onyango IG, Tuttle JB, Bennett JP Jr.
Neurobiol Dis. 2005 Oct;20(1):141-54.Neurobiol Dis. 2005 Oct; 20 (1): 141-54.
BDNF와 Hungtington s disease BDNF and Hungtington's disease
(1) Expression profiling of Huntington's disease models suggests that brain-derived neurotrophic factor depletion plays a major role in striatal degeneration. (1 ) Expression profiling of Huntington's disease models suggests that brain-derived neurotrophic factor depletion plays a major role in striatal degeneration.
Strand AD, Baquet ZC, Aragaki AK, et al (2007) J Neurosci 27 (43): 11758 68Strand AD, Baquet ZC, Aragaki AK, et al (2007)J Neurosci                     27 (43): 11758 68
(2)(2) Up-regulating BDNF with an ampakine rescues synaptic plasticity and memory in Huntington's disease knockin mice.Up-regulating BDNF with an ampakine rescues synaptic plasticity and memory in Huntington's disease knockin mice.
Simmons DA, Rex CS, Palmer L, Pandyarajan V, Fedulov V, Gall CM, Lynch G.Simmons DA, Rex CS, Palmer L, Pandyarajan V, Fedulov V, Gall CM, Lynch G.
Proc Natl Acad Sci U S A. 2009 Mar 24;106(12):4906-11. Epub 2009 Mar 5.Proc Natl Acad Sci U S A. 2009 Mar 24; 106 (12): 4906-11. Epub 2009 Mar 5.
(3)(3) Decreased BDNF levels are a major contributor to the embryonic phenotype of huntingtin knockdown zebrafish.Decreased BDNF levels are a major contributor to the embryonic phenotype of huntingtin knockdown zebrafish.
Diekmann H, Anichtchik O, Fleming A, Futter M, Goldsmith P, Roach A, Rubinsztein DC.Diekmann H, Anichtchik O, Fleming A, Futter M, Goldsmith P, Roach A, Rubinsztein DC.
J Neurosci. 2009 Feb 4;29(5):1343-9.J Neurosci. 2009 Feb 4; 29 (5): 1343-9.
(4) (4) Blood level of brain-derived neurotrophic factor mRNA is progressively reduced in rodent models of Huntington's disease: restoration by the neuroprotective compound CEP-1347.Blood level of brain-derived neurotrophic factor mRNA is progressively reduced in rodent models of Huntington's disease: restoration by the neuroprotective compound CEP-1347.
Conforti P, Ramos C, Apostol BL, Simmons DA, Nguyen HP, Riess O, Thompson LM, Zuccato C, Cattaneo E.Conforti P, Ramos C, Apostol BL, Simmons DA, Nguyen HP, Riess O, Thompson LM, Zuccato C, Cattaneo E.
Mol Cell Neurosci. 2008 Sep;39(1):1-7. Epub 2008 May 10.Mol Cell Neurosci. 2008 Sep; 39 (1): 1-7. Epub 2008 May 10.
(5)(5) Brain-derived neurotrophic factor over-expression in the forebrain ameliorates Huntington's disease phenotypes in mice.Brain-derived neurotrophic factor over-expression in the forebrain ameliorates Huntington's disease phenotypes in mice.
Gharami K, Xie Y, An JJ, Tonegawa S, Xu B.Gharami K, Xie Y, An JJ, Tonegawa S, Xu B.
J Neurochem. 2008 Apr;105(2):369-79. Epub 2007 Dec 12.J Neurochem. 2008 Apr; 105 (2): 369-79. Epub 2007 Dec 12.
여러 가지 제형에 따라 상용되는 약학적으로 허용되는 담체로서, 모든 형태의 희석제 또는 용매, 충진제, 비산제, 결합제, 분산제, 붕해제, 계면활성제, 윤활제, 부형제 및 습윤제가 있다. 더우기, 필요시, 상용되는 용해 보조제, 완충제, 보존제, 착색제, 향료, 감미제 등도 상기 약제에 가할 수 있다. Pharmaceutically acceptable carriers compatible with various formulations include all forms of diluents or solvents, fillers, fillers, binders, dispersants, disintegrants, surfactants, lubricants, excipients, and wetting agents. Furthermore, if necessary, commercially available dissolution aids, buffers, preservatives, colorants, flavors, sweeteners, and the like may also be added to the drug.
본 발명에 따른 약학 조성물의 투여 단위 형태는 구체적으로 제한되지 않으며, 여러 가지 치료 목적에 따라 널리 선택되어질 수 있으며, 예를 들어, 정제, 캡슐제, 과립제, 환제, 시럽제, 액제, 유제, 현탁제 등과 같은 경구 투여제, 주사제(피하주사, 정맥주사, 근육주사, 복강주사 등) 및 좌제와 같은 비경구 투여제가 있다. 이들 제제 중에서, 경구 투여 제제가 바람직하다. The dosage unit form of the pharmaceutical composition according to the present invention is not particularly limited and may be widely selected according to various therapeutic purposes, for example, tablets, capsules, granules, pills, syrups, solutions, emulsions, suspensions. Oral dosages such as and the like, injection (subcutaneous injection, intravenous injection, intramuscular injection, intraperitoneal injection, etc.) and parenteral administration such as suppositories. Among these formulations, oral dosage formulations are preferred.
또한, 미세주입법 (microinjection), 전기충격을 이용한 전기천공법 (electorporation), 양이온이나 리포좀을 이용한 전달법, PTD(protein transduction domain) 등 당업계에 공지된 펩타이드의 세포내 전달방법을 활용할 수 있다.In addition, microinjection, electroporation using electroshock (electorporation), delivery using cations or liposomes, intracellular delivery methods of peptides known in the art, such as PTD (protein transduction domain) can be used.
전술된 여러 가지 형태의 약제는 통상의 방법으로 제조될 수 있다 예를들어, 정제, 캡슐제, 과립제 및 환제와 같은 경구 투여 제제를 제조함에 있어서, 이들은 백당, 락토오스, 글루코오스, 전분, 만니톨과 같은 부형제; 시럽, 아라비아 고무, 솔비톨, 트리가칸트 고무, 메틸셀룰로오스, 폴리비닐피롤리돈 등과 같은 결합제; 전분, 카르복시메틸 셀룰로오스 및 그의 칼슘염, 미결정 셀룰로오스, 폴리에틸렌글리콜 등과 같은 붕해제; 탈크, 마그네슘 스테아레이트, 칼슘 스테아레이트, 실리카 등과 같은 윤활제; 소듐라우레이트, 글리세롤 등과 같은 습윤제를 이용하여 통상의 방법으로 제조될 수 있다. The various types of medicaments described above can be prepared by conventional methods, for example in the preparation of oral dosage forms such as tablets, capsules, granules and pills, they are known as sucrose, lactose, glucose, starch, mannitol Excipients; Binders such as syrup, gum arabic, sorbitol, trigacanth rubber, methylcellulose, polyvinylpyrrolidone and the like; Disintegrants such as starch, carboxymethyl cellulose and its calcium salts, microcrystalline cellulose, polyethylene glycol and the like; Lubricants such as talc, magnesium stearate, calcium stearate, silica and the like; It may be prepared by conventional methods using wetting agents such as sodium laurate, glycerol and the like.
주사제, 액제, 유제, 현탁제 및 시럽제를 제조함에 있어서, 이들은, 통상의 방법에 의하여, 에틸 알코올, 이소프로필 알코올, 프로필렌 글리콜, 1,3-부틸렌 글리콜, 폴리에틸렌 글리콜, 카스터오일 등과 같은 활성성분 용해용의 용매; 솔비톨 지방산 에스테르, 폴리옥시에틸렌 솔비톨 지방산 에스테르, 폴리옥시에틸렌에스테르, 수소화된 카스터 오일의 폴리옥시에틸렌 에테르, 레시틴 등과 같은 계면활성제; 소듐 카르복시메틸 셀룰로오스, 메틸 셀룰로오스 등과 같은 셀룰로오스 유도체, 트리가칸트 고무, 아라비아 고무 등과 같은 천연 고무 등의 현탁화제, 파라옥시벤조산의 에스테르, 벤잘코늄 클로라이드, 솔비탄 지방산염 등과 같은 보존제를 적절히 사용하여 제조될 수 있다. In the preparation of injectables, solutions, emulsions, suspensions and syrups, they are prepared by conventional methods, such as ethyl alcohol, isopropyl alcohol, propylene glycol, 1,3-butylene glycol, polyethylene glycol, castor oil and the like. Solvent for component dissolution; Surfactants such as sorbitol fatty acid ester, polyoxyethylene sorbitol fatty acid ester, polyoxyethylene ester, polyoxyethylene ether of hydrogenated castor oil, lecithin and the like; Manufactured using appropriate preservatives such as cellulose derivatives such as sodium carboxymethyl cellulose, methyl cellulose, suspending agents such as natural rubbers such as trigacanth rubber, gum arabic, esters of paraoxybenzoic acid, benzalkonium chloride, sorbitan fatty acid salts, etc. Can be.
좌제를 제조함에 있어서, 이들은 통상의 방법을 사용하여 폴리에틸렌 글리콜, 라놀린, 코코넛 오일 등과 같은 부형제를 사용하여 제조될 수 있다. In preparing suppositories, they can be prepared using excipients such as polyethylene glycol, lanolin, coconut oil and the like using conventional methods.
본 발명에 따른 MVG 펩타이드의 치료학적 유효량을 포함하는 신경질환의 예방 및 치료용 목적의 약학 조성물의 용량은 투여방법, 제제 형태, 환자의 나이, 환자의 체중, 환자의 감수성, 질환의 상태에 따라 적절하게 선택될 수 있으며 구체적으로 제한된 것은 아니다.  The dose of the pharmaceutical composition for the prevention and treatment of neurological diseases, including the therapeutically effective amount of the MVG peptide according to the present invention, depends on the method of administration, the type of preparation, the age of the patient, the weight of the patient, the sensitivity of the patient, and the condition of the disease. It may be appropriately selected and is not specifically limited.
발명의 약학 조성물 중에 포함된 MVG 펩타이드의 치료학적 유효량은 0.1 ㎍/㎏/일 내지 10 ㎍/㎏/일로 투여될 수 있다. 물론, 상기 치료학적 유효량은 전술된 범위를 벗어난 양일 수도 있다.  The therapeutically effective amount of the MVG peptide included in the pharmaceutical composition of the present invention may be administered at 0.1 μg / kg / day to 10 μg / kg / day. Of course, the therapeutically effective amount may be in an amount outside the ranges described above.
본 발명의 약학 조성물은 래트, 마우스, 가축, 인간 등의 포유동물에 다양한 경로로 투여될 수 있다. 투여의 모든 방식이 예상될 수 있는데, 예를 들면, 경구, 피하, 정맥 내, 근육 내, 비강 내, 복강 내, 직장 내, 자궁 내 경막 또는 뇌실내 (intracerebroventricular) 주사에 의해 투여될 수 있다. 본 발명의 MVG 펩타이드는 독성 및 부작용이 거의 없으므로 예방 목적으로 장기간 복용시에도 안심하고 사용할 수 있다. The pharmaceutical composition of the present invention can be administered to mammals such as rats, mice, livestock, humans, etc. by various routes. All modes of administration can be envisaged, for example, by oral, subcutaneous, intravenous, intramuscular, nasal, intraperitoneal, rectal, intrauterine dural or intracerebroventricular injection. The MVG peptide of the present invention has little toxicity and side effects, and therefore can be used with confidence even for prolonged administration for prophylactic purposes.
또한, 본 발명의 약학조성물은 MVG 펩타이드를 코딩하는 DNA 서열로 구성된 폴리뉴클레오티드 또는 등가의 핵산서열, 즉 DNA 서열은 상이하나 동일한 서열의 MVG 펩타이드를 암호화하는 코돈 축퇴성 (codon degeneracy) 서열을 포함하는 유전자 전달체 일 수 있으며, 상기 뉴클레오티드들의 세포 내 전달(gene delivery)방식 역시 본 발명의 기술적 범위에 포함된다. 상기 올리고 뉴클레오타이드는 공지된 형태로 세포 내 전달될 수 있으며, 일 예로 종래의 유전자 운반체로 이용되어온 레트로바이러스 벡터(retroviral vector), 아데노바이러스 벡터(adenoviral vector), 아데노 연관 바이러스 벡터(adeno-associated viral vector) 룰 포함하는 바이러스 성 벡터, 리포좀, 폴리리신 (polylysine), 폴리에틸렌이민 (polyethylenimine (PEI)), 프로타민(protamine), 히스톤(histone), 폴리에스테르아민(Polyester amine) 및 이들 각각의 변형체를 포함하는 양이온성 고분자를 비롯하여, 미셀, 에멀젼, 나노입자 등의 비바이러스성 벡터를 사용할 수 있으며, 벡터 시스템 외에도, 공지된 세포내 물질전달 펩타이드 등을 활용하여 효율적으로 세포내 전달될 수 있다.In addition, the pharmaceutical composition of the present invention comprises a polynucleotide or equivalent nucleic acid sequence consisting of a DNA sequence encoding an MVG peptide, that is, a codon degeneracy sequence encoding a MVG peptide having a different sequence but the same DNA sequence. It may be a gene carrier, and the gene delivery method of the nucleotides is also included in the technical scope of the present invention. The oligonucleotides can be delivered intracellularly in a known form. For example, retroviral vectors, adenovirus vectors, and adeno-associated viral vectors that have been used as conventional gene carriers. Viral vector comprising a rule, liposomes, polylysine, polyethylenimine (PEI), protamine, histone, polyesteramine and their respective variants In addition to cationic polymers, non-viral vectors such as micelles, emulsions, nanoparticles, and the like can be used. In addition to the vector system, it can be efficiently delivered intracellularly using known intracellular substance transfer peptides.
본 발명에 따른 세 개의 아미노산으로 구성된 MVG 펩타이드는 해마신경세포주에서 세포독성 없이 BDNF의 발현을 증가시킬 뿐 아니라 실제 해마조직과 피질조직에서도 BDNF의 발현을 증가시켰다. 따라서, 본 발명과 같이 크기가 작은 MVG 펩타이드는 분자량도 작고 혈뇌장벽 (Blood Brain Barrier, BBB)을 쉽게 통과하며 세포독성도 없으므로 알츠하이머병, 파킨슨병, 만성스트레스성 질환인 우울증, 뇌졸중, 헌팅톤무도병, 대뇌허혈성질환, 신경퇴화질환, 당뇨병성 신경병증 등과 같은 신경질환의 예방 및 치료제로써 매우 유효하다.  MVG peptide consisting of three amino acids according to the present invention not only increases the expression of BDNF without cytotoxicity in the hippocampal neuronal cell line but also increases the expression of BDNF in hippocampal and cortical tissues. Therefore, the small MVG peptides, such as the present invention, have a low molecular weight, easily cross the Blood Brain Barrier (BBB), and do not have cytotoxicity. Therefore, Alzheimer's disease, Parkinson's disease, depression, stroke, and Huntington's chorea It is very effective as a preventive and therapeutic agent for neurological diseases such as cerebral ischemic disease, neurodegenerative disease and diabetic neuropathy.
도 1은 펩타이드 라이브러리 기술인 PS-SPCL (Positional Scanning-Synthetic Peptide Combinatorial Library)을 통하여서 세 개의 아미노산 (Methionine-Valine-Glycine)으로 이루어진 펩타이드의 각 위치에서 BDNF 발현이 가장 많은 아미노산을 찾은 결과를 도시한 것이다. Figure 1 shows the results of finding the amino acid with the highest BDNF expression at each position of the peptide consisting of three amino acids (Methionine-Valine-Glycine) through the PS-SPCL (Positional Scanning-Synthetic Peptide Combinatorial Library) .
도 2는 본 발명의 MVG 합성 펩타이드를 래트 (Rat)의 해마신경세포주인 H19-7에 농도별로 처리를 한 후, BDNF의 mRNA 발현 결과를 도시한 것이다. Figure 2 shows the results of mRNA expression of BDNF after treatment of MVG synthetic peptide of the present invention in rats (Hat), a hippocampal nerve cell line of rat (Rat) by concentration.
도 3 은 본 발명의 MVG 합성 펩타이드를 래트의 해마신경세포주인 H19-7에 농도별로 처리를 한 후, BDNF의 단백질 발현 결과를 도시한 것이다.Figure 3 shows the results of protein expression of BDNF after treatment of the MVG synthetic peptide of the present invention to the rat hippocampal neuronal cell line H19-7 by concentration.
도 4는 본 발명의 MVG 합성 펩타이드 0.1㎍/㎏을 래트의 꼬리 정맥 주사를 한 후 해마 조직 부위를 적출하여 BDNF 단백질 발현 결과를 도시한 것이다.Figure 4 shows the results of BDNF protein expression by extracting the hippocampal tissue site after the rat intravenous injection of 0.1g / kg MVG synthetic peptide of the present invention.
도 5는 본 발명의 MVG 합성 펩타이드 0.1㎍/㎏을 래트의 꼬리 정맥 주사를 한 후 대뇌 피질 조직 부위를 적출하여 BDNF 단백질 발현 결과를 도시한 것이다. Figure 5 shows the results of BDNF protein expression by extracting the cerebral cortical tissue site after the tail vein injection of 0.1V / kg MVG synthetic peptide of the present invention.
도 6은 MTT (3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) 검사를 통한 MVG 합성 펩타이드의 세포 독성 결과를 도시한 것이다. Figure 6 shows the cytotoxicity results of MVG synthetic peptides by MTT (3- (4,5-Dimethylthiazol-2-yl) -2,5-diphenyltetrazolium bromide) test.
도 7은 Y-maze test 수행방법을 설명한 것이며, 도 8은 실험결과 novel arm으로 들어가는 횟수와 novel arm에서의 이동거리를 도시한 것이다. FIG. 7 illustrates a method of performing a Y-maze test, and FIG. 8 illustrates the number of times of entering the novel arm and the moving distance from the novel arm.
도 9는 모리스 수중 미로 (Morris water maze) test 수행방법을 설명한 것이다. 9 illustrates a method of performing a Morris water maze test.
도 10은 실험결과 platform을 찾는데 소요된 시간 및 platform에 머무른 도시한 것이다. Figure 10 shows the time spent on finding the platform and the test stayed on the platform.
도 11은 Y-maze test 및 모리스 수중 미로 (Morris water maze) test를 수행한 쥐들의 해마(도 11(a)) 및 대뇌피질(도 11(b))에서의 BDNF 발현을 도시한 것이다.FIG. 11 shows BDNF expression in the hippocampus (FIG. 11 (a)) and the cerebral cortex (FIG. 11 (b)) of mice that underwent the Y-maze test and Morris water maze test.
도 12는 Y-maze test 및 모리스 수중 미로 (Morris water maze) test를 수행한 쥐들의 해마(도 12(a)) 및 대뇌피질(도 12(b))에서의 TrkB의 인산화 및 TrkB 발현을 도시한 것이다.FIG. 12 shows phosphorylation and TrkB expression of TrkB in hippocampus (FIG. 12 (a)) and cortex (FIG. 12 (b)) of mice that underwent Y-maze test and Morris water maze test. It is.
도 13은 BDNF 발현을 조절하는 MVG 펩타이드의 anti-parkinson 효과를 확인하는 in vitro 실험을 수행한 결과를 도시한 것이다. FIG. 13 shows the results of in vitro experiments confirming the anti-parkinson effect of MVG peptides that regulate BDNF expression.
실시예 1: PS-SPCL을 통한 초기 스크리닝 (Initial screening)Example 1 Initial Screening with PS-SPCL
BDNF 발현을 증가시키는 펩타이드 발굴을 위해서 PS-SPCL 트리머 페키지 스톡 (trimer package stock)을 포항공과대학교 생명공학연구센터 펩타이드 라이브러리 (peptide library) 지원 시설로부터 지원받았다. 본 발명에 사용된 펩타이드 라이브러리 기술인 PS-SPCL (Positional Scanning-Synthetic Peptide Combinatorial Library)은 세 개의 아미노산으로 이루어진 펩타이드로 첫 번째 위치는 시스테인 (Cysteine)을 제외한 알라닌 (Alanine)에서 타이로신 (Tyrosine)으로 치환되어져 있고 나머지 두 번째, 세 번째 위치는 고정되어져 있는 방식으로 각각의 위치에서 가장 효과적인 아미노산 서열을 밝힐 수 있고, 이를 통하여 각 위치에서 BNDF 발현이 가장 많은 아미노산을 찾을 수 있다.  PS-SPCL trimmer package stock was supported by the Peptide library support facility at Pohang University of Science and Technology for the discovery of peptides that increase BDNF expression. PS-SPCL (Positional Scanning-Synthetic Peptide Combinatorial Library), a peptide library technology used in the present invention, is a peptide consisting of three amino acids, the first position of which is substituted with tyrosine in alanine except cysteine. And the remaining second and third positions are fixed in such a way as to identify the most effective amino acid sequence at each position, thereby finding the amino acid with the highest BNDF expression at each position.
먼저, 마우스 (Mouse) 해마신경세포주인 HT22 세포를 24 웰 플래이트 (24 well plate, SPL)에 웰 당 5X104씩 총 57개를 깔고 12시간 동안 37℃, 5% CO2 인큐베이터 (Incubator, VISION)에 배양하였다. PS-SPCL 트리머 패키지 스톡 (trimer package stock)을 스핀다운 (spin down)한 후 튜브를 열고 15㎕의 세포배양용 D.W. (Distilled Water)를 넣어서 탭핑 (tapping)한 후, 다시 스핀다운하였다. HT22 세포가 배양되고 있는 24 웰 플래이트에 준비된 PS-SPCL 트리머 패키지 스톡 15㎕를 각 웰에 넣고 12시간 인큐베이터에 배양한다. 12시간 후 웰에 들어있는 배지를 버리고 1X PBS (Potassium persulfate) 1㎖로 2번 세척하였다. RIPA 1㎖당 단백질분해효소 저해제 (Protease inhibitor, Amersham) 10㎕ 비율로 준비하여 각각의 웰에 넣고 세포 긁개 (Cell Lifter, Corning)을 이용하여 세포 내용물을 1.5㎖ 튜브에 담아두었다. 이를 4℃, 9000rpm에서 5분 동안 원심분리 (Centrifuge, MICRO 17TR, Hanil Science Industrial Co., Ltd)를 통해 세포막을 펠렛 (pellet)화시키고 단백질이 포함된 상층액을 새로운 1.5㎖ 튜브에 옮기고, 단백질의 농도를 브래드포드법 (Bradford assay)을 이용하여 정량하였다. First, 57 mouse hippocampal neuronal cell lines, HT22 cells, were placed in a 24-well plate (SPL) at a total of 57 by 5 × 10 4 per well for 12 hours at 37 ° C. in a 5% CO 2 incubator (Incubator, VISION). Incubated in the. After spin down of the PS-SPCL trimmer package stock, the tube was opened, tapped with 15 μl of distilled water (DW) for cell culture, and then spun down again. 15 μl of PS-SPCL trimmer package stock prepared on a 24-well plate in which HT22 cells are incubated is placed in each well and incubated in an incubator for 12 hours. After 12 hours, the medium in the well was discarded and washed twice with 1 ml of 1X PBS (Potassium persulfate). 10 μl of protease inhibitor (Amersham) per 1 mL of RIPA was prepared, placed in each well, and the cell contents were placed in a 1.5 mL tube using a cell lifter (Cell Lifter, Corning). Pelletize the cell membrane by centrifugation (Centrifuge, MICRO 17TR, Hanil Science Industrial Co., Ltd) for 5 minutes at 4 ° C., 9000 rpm, and transfer the supernatant containing protein to a new 1.5 ml tube. The concentration of was quantified using the Bradford assay.
BDNF 발현양을 측정하기 위해서 ELISA (Enzyme-linked immunosorbent assay, BDNF ELISA Kit, Promega)를 프로토콜 (Protocol)에 따라서 실시하였다. 도 1에서 보는 바와 같이 첫 번째 위치에서는 M (Methionine), 두 번째 위치에서는 V (Valine), 세 번째 위치에서는 G (Glycine)가 BDNF 발현이 가장 많았다. In order to measure the amount of BDNF expression, ELISA (Enzyme-linked immunosorbent assay, BDNF ELISA Kit, Promega) was performed according to the protocol. As shown in FIG. 1, M (Methionine) in the first position, V (Valine) in the second position, and G (Glycine) in the third position showed the highest BDNF expression.
실시예 2: 해마신경세포주 H19-7에서 MVG 펩타이드에 의한 BDNF mRNA 발현의 증가Example 2: Increase of BDNF mRNA Expression by MVG Peptide in Hippocampal Neuronal Cell Line H19-7
상기 실시예 1을 통한 결과로써 세 개의 아미노산으로 구성된 MVG 합성 펩타이드를 (주) 펩트론으로부터 구입하였다. ATCC (American Type Culture Collection)로부터 구입한 래트의 해마신경세포주인 H19-7세포를 6 웰 플래이트 (Well plate, NUNC)에 웰당 106씩 깔고 12시간 동안 34℃, 5% CO2 인큐베이터에 배양 후, MVG 합성 펩타이드를 0.001, 0.01, 0.1, 1μM로 처리하여 12시간 추가 배양하였다. Easy-blue Total RNA extraction kit (Intron)의 프로토콜에 따라서 각각의 샘플로부터 RNA를 분리한 후 1㎍ total RNA, 1㎕ oligo (dT)18, 나머지는 DEPC (Diethylpyrocarbonate)를 처리한 D.W.을 이용하여 총 부피가 20㎕가 되게 PCR용 튜브에 넣고, 70℃에서 5분 동안 가열하였다. RT premix (Reverse transcriptase premix, Bioneer) 튜브로 가열한 20㎕을 옮겨 담고 42℃에서 1시간 동안 역전사 (Reverse transcription) 시켜서 상보 DNA (complementary DNA, cDNA)을 만들었다. 만들어진 cDNA 5㎕와 BDNF 프라이머 2㎕, DDW (Double Distilled Water) PCR 13㎕를 PCR premix (Bioneer) 튜브에 담아서 94℃에서 5분, 94℃에서 30초, 60℃에서 30초, 72℃에서 45초, 72℃에서 5분 29 사이클 돌렸다. 그 결과 도 2에 나타난 바와 같이 0.001, 0.01, 0.1μM에서 농도별로 BDNF mRNA의 발현 양이 증가하는 것을 확인할 수 있다. 이를 통해서 MVG 합성 펩타이드가 래트의 해마신경세포주에서 BDNF mRNA 발현을 증가시킨다는 것과 실시예 1을 통한 펩타이드 선정이 제대로 이루어졌음을 입증하였다.As a result of Example 1, an MVG synthetic peptide consisting of three amino acids was purchased from Peptron Co., Ltd. H19-7 cells, the hippocampal neuronal cell lines of rats purchased from the American Type Culture Collection (ATCC), were placed in 6 well plates (NUNC) at 10 6 per well and incubated in a 34 ° C., 5% CO 2 incubator for 12 hours. , MVG synthetic peptides were treated with 0.001, 0.01, 0.1, 1μM and further incubated for 12 hours. RNA was isolated from each sample according to the protocol of the Easy-blue Total RNA extraction kit (Intron), and then 1 ug total RNA, 1 ol oligo (dT) 18 , and the rest were processed using DW treated with DEPC (Diethylpyrocarbonate). The volume was added to the tube for PCR to 20 mu l and heated at 70 ℃ for 5 minutes. Complementary DNA (cDNA) was prepared by transferring 20 μl of the tube heated with RT premix (Reverse transcriptase premix, Bioneer) tube and performing reverse transcription at 42 ° C. for 1 hour. 5 μl of the prepared cDNA, 2 μl of BDNF primer and 13 μl of DDW (Double Distilled Water) PCR were placed in a PCR premix (Bioneer) tube for 5 minutes at 94 ° C., 30 seconds at 94 ° C., 30 seconds at 60 ° C., 45 at 72 ° C. The cycle was turned 5 minutes and 29 cycles at 72 ° C. As a result, as shown in Figure 2 it can be seen that the amount of BDNF mRNA expression increases by concentration at 0.001, 0.01, 0.1μM. This demonstrates that MVG synthetic peptides increase BDNF mRNA expression in hippocampal neuronal cell lines of rats, and that peptide selection through Example 1 was achieved.
실시예 3: 해마신경세포주 H19-7에서 MVG 펩타이드에 의한 BDNF 단백질 발현의 증가Example 3: Increase of BDNF Protein Expression by MVG Peptide in Hippocampal Neuronal Cell Line H19-7
상기 실시예 1을 통한 결과로써 세 개의 아미노산으로 구성된 MVG 합성 펩타이드를 (주) 펩트론으로부터 구입하였다. ATCC (American Type Culture Collection)로부터 구입한 래트의 해마신경세포주인 H19-7세포를 6 웰 플래이트(Well plate, NUNC)에 웰당 106씩 깔고 12시간 동안 34℃, 5% CO2 인큐베이터에 배양 후, MVG 합성 펩타이드를 0.001, 0.01, 0.1, 1μM로 처리하여 12시간 추가 배양하였다. 실시예 1과 동일한 방법으로 단백질을 얻고 1.5mm 웨스턴 블라팅 (Western blotting) 겔 케스터 (Gel caster, Bio-Rad)을 통하여 15% 아크릴아미드 겔 (acrylamide gel)을 만들고 30㎍의 샘플을 로딩 (Loading)하여 전기영동 장치 (Electrophoresis Power supply, EPS 601, Amersham)로 100V에서 2시간 30분 동안 러닝하였다. PVDF (Polyvinylidene Difluoride, Millipore)을 이용하여 400mA에서 2시간 30분 전기영동장치 (Electrophoresis Power supply, EPS 301, GE healthcare)를 통해 겔을 트랜스퍼 (Mighty small transphor, Amersham)하였다. 7% 스킴 밀크 (Skim milk, DifcoTM Skim milk, BD)로 PVDF를 1시간 동안 블로킹 (blocking)하고 래빗 안티 BNDF 폴리클로널 항체 (Rabbit anti BDNF polyclonal antibody, AB1534, chemicon)를 1:1000의 비율로 밤새 (overnight) 반응시켰다. 0.05% TBST (Tris-Buffered Saline Tween 20)로 1시간 동안 세척 (20분씩 3번) 한 후, 고트 안티-래빗 IgG:HRP (Goat anti-rabbit IgG:Horseradish Peroxidase Conjugate, SAB-300, Bioreagents)을 1:3000의 비율로 50분 동안 인큐베이션하였다. 0.05% TBST로 1시간 동안 세척(20분씩 3번)한 후, chemiluminescent HRP substrate kit (Millipore)로 검출(detection)하였다. 그 결과, 도 3에서 나타난 바와 같이 0.001, 0.01, 0.1μM에서 농도별로 BDNF 단백질의 발현 양이 증가하는 것을 확인할 수 있다. 또한 이는 도 2에서 나타난 BDNF mRNA발현 결과와 동일한 증가 양상을 보인다.As a result of Example 1, an MVG synthetic peptide consisting of three amino acids was purchased from Peptron Co., Ltd. H19-7 cells, rat hippocampal neuronal cell lines purchased from the American Type Culture Collection (ATCC), were placed in a 6 well plate (NUNC) at 10 6 per well and incubated in a 34 ° C., 5% CO 2 incubator for 12 hours. , MVG synthetic peptides were treated with 0.001, 0.01, 0.1, 1μM and further incubated for 12 hours. Obtain protein in the same manner as in Example 1, make 15% acrylamide gel through 1.5 mm Western blotting gel caster (Gel caster, Bio-Rad) and load 30 μg sample 2 hours 30 minutes at 100V with an electrophoresis device (Electrophoresis Power supply, EPS 601, Amersham). Polyvinylidene Difluoride (PVDF) was used to transfer the gel (Mighty small transphor, Amersham) using an electrophoresis power supply (EP 301, GE healthcare) at 400 mA for 2 hours and 30 minutes. Blocking PVDF for 1 hour with 7% skimmed milk (Skim milk, Difco Skim milk, BD) and a rabbit anti BDNF polyclonal antibody (AB1534, chemicon) in a ratio of 1: 1000 React overnight. After washing for 1 hour with 0.05% TBST (Tris-Buffered Saline Tween 20) (3 times for 20 minutes), Goat anti-rabbit IgG: Horseradish Peroxidase Conjugate, SAB-300, Bioreagents Incubate for 50 minutes at a ratio of 1: 3000. After washing for 1 hour with 0.05% TBST (3 times for 20 minutes), detection was performed with a chemiluminescent HRP substrate kit (Millipore). As a result, as shown in Figure 3 it can be seen that the amount of BDNF protein expression increased by concentration at 0.001, 0.01, 0.1μM. In addition, this shows the same increase pattern as the BDNF mRNA expression results shown in FIG.
실시예 4: 해마조직에서 MVG 펩타이드에 의한 BDNF 단백질 발현의 증가Example 4 Increasing BDNF Protein Expression by MVG Peptides in Hippocampus
4 주령의 수컷 SD (Sprague-Dawley) 래트를 (주) 오리엔트 사로부터 구입하여 실험군 (MVG 투여 그룹)과 대조군 (1X PBS 투여 그룹)으로 3 마리씩 분리한 후, 환경에 적응시키기 위해서 1 주일간 사육실에 적응 및 안정화를 시켰다. 1 주일 뒤 실험군은 0.1㎍/㎏의 MVG를, 대조군은 0.1㎍/㎏의 1X PBS을 꼬리 정맥 주사 (Tail vein injection)를 하였다. 12시간 후에 에테르 (Ether)를 이용하여 일시 마취시키고, 개복한 후에 심장에서 혈액을 채취하고, 재빨리 쥐의 뇌를 적출한 후 액체 질소에서 급속히 냉각시켜 -80℃에 보관하였다. 적출된 뇌에서 해마부분만을 분리한 후, RIPA 완충액과 단백질 분해효소 저해제를 상기의 실시예 1과 동일한 비율로 준비하여 분쇄기 (Homogenizer) (RZR2020M, Heidolph)를 이용하여 해마 조직을 완전히 분쇄하였다. 이를 4℃, 2,000rpm에서 30분 동안 원심분리 (Centrifuge, UNION 32R, Hanil Science Industrial Co.,Ltd)하여 상층액만 1.5㎖ 튜브로 옮긴 후 이를 다시 4℃, 9,000rpm에서 30분 동안 원심분리 (Centrifuge, MICRO 17TR, Hanil Science Industrial Co., Ltd) 하여 다시 한번 상층액만을 1.5㎖ 튜브로 옮겨 브래드포드 (Bradford assay)를 통해서 단백질을 정량하였다.  Four-week-old male SD (Sprague-Dawley) rats were purchased from Orient Co., Ltd. and separated into three groups of experimental groups (MVG-administered group) and control group (1X PBS-administered group). Adaptation and stabilization were made. After one week, the experimental group was tail vein injection of 0.1 μg / kg of MVG and the control group of 0.1 μg / kg of 1 × PBS. After 12 hours, anesthesia was temporarily used in ether, and blood was collected from the heart after laparotomy. The brains of the rats were quickly extracted, rapidly cooled in liquid nitrogen, and stored at -80 ° C. After separating only the hippocampus from the brain, the RIPA buffer and protease inhibitor were prepared in the same ratio as in Example 1, and the hippocampal tissue was completely ground using a Homogenizer (RZR2020M, Heidolph). Centrifuge at 4 ° C and 2,000rpm for 30 minutes (Centrifuge, UNION 32R, Hanil Science Industrial Co., Ltd), transfer only the supernatant to 1.5ml tube, and then centrifuge for 30 minutes at 4 ° C, 9,000rpm ( Centrifuge, MICRO 17TR, Hanil Science Industrial Co., Ltd) was used to quantify the protein by Bradford assay once again transfer only the supernatant to 1.5ml tube.
1.5mm 웨스턴 블라팅 (Western blotting) 겔 케스터 (Gel caster, Bio-Rad)을 통하여 15% 아크릴아미드 겔 (acrylamide gel)을 만들고 30㎍의 샘플을 로딩 (Loading)하여 전기영동 장치 (Electrophoresis Power supply, EPS 601, Amersham)로 100V에서 2시간 30분 동안 러닝하였다. PVDF (Polyvinylidene Difluoride, Millipore)을 이용하여 400mA에서 2시간 30분 전기영동장치 (Electrophoresis Power supply, EPS 301, GE healthcare)를 통해 겔을 트랜스퍼 (Mighty small transphor, Amersham)하였다. 7% 스킴 밀크 (Skim milk, DifcoTM Skim milk, BD)로 PVDF를 1시간 동안 블로킹 (blocking)하고 래빗 안티 BNDF 폴리클로널 항체 (Rabbit anti BDNF polyclonal antibody, AB1534, chemicon)를 1:1000의 비율로 밤새 (overnight) 반응시켰다. 0.05% TBST (Tris-Buffered Saline Tween 20)로 1시간 동안 세척 (20분씩 3번) 한 후, 고트 안티-래빗 IgG:HRP (Goat anti-rabbit IgG:Horseradish Peroxidase Conjugate, SAB-300, Bioreagents)을 1:3000의 비율로 50분 동안 인큐베이션하였다. 0.05% TBST로 1시간 동안 세척 (20분씩 3번)한 후, chemiluminescent HRP substrate kit (Millipore)로 검출 (detection)하였다. 그 결과, 도 4에 나타난 바와 같이 대조군에 비해서 실험군에서 BDNF와 그것의 전구체(precursor) 형태인 proBDNF의 양이 β-actin 대비 증가한 것을 확인할 수 있었다.A 15% acrylamide gel was made through a 1.5 mm Western blotting gel caster (Gel caster, Bio-Rad) and 30 μg of sample was loaded to obtain an electrophoresis power supply. EPS 601, Amersham) for 2 hours 30 minutes at 100V. Polyvinylidene Difluoride (PVDF) was used to transfer the gel (Mighty small transphor, Amersham) using an electrophoresis power supply (EP 301, GE healthcare) at 400 mA for 2 hours and 30 minutes. Blocking PVDF for 1 hour with 7% skimmed milk (Skim milk, Difco Skim milk, BD) and a rabbit anti BDNF polyclonal antibody (AB1534, chemicon) in a ratio of 1: 1000 React overnight. After washing for 1 hour with 0.05% TBST (Tris-Buffered Saline Tween 20) (3 times for 20 minutes), Goat anti-rabbit IgG: Horseradish Peroxidase Conjugate, SAB-300, Bioreagents Incubate for 50 minutes at a ratio of 1: 3000. After washing for 1 hour with 0.05% TBST (3 times for 20 minutes), detection was performed with a chemiluminescent HRP substrate kit (Millipore). As a result, as shown in FIG. 4, the amount of BDNF and its precursor (precursor) form proBDNF in the experimental group compared to the control group was confirmed to increase compared to β-actin.
실시예 5: 대뇌 피질 조직에서 MVG 펩타이드에 의한 BDNF 단백질 발현의 증 Example 5: Increased BDNF Protein Expression by MVG Peptides in Cerebral Cortical Tissues
4 주령의 수컷 SD (Sprague-Dawley) 래트를 (주) 오리엔트 사로부터 구입하여 실험군 (MVG 투여 그룹)과 대조군 (1X PBS 투여 그룹)으로 3 마리씩 분리한 후, 환경에 적응시키기 위해서 1 주일간 사육실에 적응 및 안정화를 시켰다. 1 주일 뒤 실험군은 0.1㎍/㎏의 MVG를, 대조군은 0.1㎍/㎏의 1X PBS을 꼬리 정맥 주사 (Tail vein injection)를 하였다. 12시간 후에 에테르 (Ether)를 이용하여 일시 마취시키고, 개복한 후에 심장에서 혈액을 채취하고, 재빨리 쥐의 뇌를 적출한 후 액체 질소에서 급속히 냉각시켜 -80℃에 보관하였다. 적출된 뇌에서 대뇌 피질부분만을 분리한 후, RIPA 완충액과 단백질 분해효소 저해제를 상기의 실시예 1과 동일한 비율로 준비하여 분쇄기 (Homogenizer) (RZR2020M, Heidolph)를 이용하여 피질 조직을 완전히 분쇄하였다. 이를 4℃, 2,000rpm에서 30분 동안 원심분리 (Centrifuge, UNION 32R, Hanil Science Industrial Co.,Ltd)하여 상층액만 1.5㎖ 튜브로 옮긴 후 이를 다시 4℃, 9,000rpm에서 30분 동안 원심분리 (Centrifuge, MICRO 17TR, Hanil Science Industrial Co., Ltd) 하여 다시 한번 상층액만을 1.5㎖ 튜브로 옮겨 브래드포드 (Bradford assay)를 통해서 단백질을 정량하였다.  Four-week-old male SD (Sprague-Dawley) rats were purchased from Orient Co., Ltd. and separated into three groups of experimental groups (MVG-administered group) and control group (1X PBS-administered group). Adaptation and stabilization were made. After one week, the experimental group was tail vein injection of 0.1 μg / kg of MVG and the control group of 0.1 μg / kg of 1 × PBS. After 12 hours, anesthesia was temporarily used in ether, and blood was collected from the heart after laparotomy. The brains of the rats were quickly extracted, rapidly cooled in liquid nitrogen, and stored at -80 ° C. After only the cerebral cortex was separated from the extracted brain, RIPA buffer and protease inhibitor were prepared in the same ratio as in Example 1, and the cortical tissue was completely ground using a Homogenizer (RZR2020M, Heidolph). Centrifuge at 4 ° C and 2,000rpm for 30 minutes (Centrifuge, UNION 32R, Hanil Science Industrial Co., Ltd), transfer only the supernatant to 1.5ml tube, and then centrifuge for 30 minutes at 4 ° C, 9,000rpm ( Centrifuge, MICRO 17TR, Hanil Science Industrial Co., Ltd) was used to quantify the protein by Bradford assay once again transfer only the supernatant to 1.5ml tube.
1.5mm 웨스턴 블라팅 (Western blotting) 겔 케스터 (Gel caster, Bio-Rad)을 통하여 15% 아크릴아미드 겔 (acrylamide gel)을 만들고 30㎍의 샘플을 로딩 (Loading)하여 전기영동 장치 (Electrophoresis Power supply, EPS 601, Amersham)로 100V에서 2시간 30분 동안 러닝하였다. PVDF (Polyvinylidene Difluoride, Millipore)을 이용하여 400mA에서 2시간 30분 전기영동장치 (Electrophoresis Power supply, EPS 301, GE healthcare)를 통해 겔을 트랜스퍼 (Mighty small transphor, Amersham)하였다. 7% 스킴 밀크 (Skim milk, DifcoTM Skim milk, BD)로 PVDF를 1시간 동안 블로킹 (blocking)하고 래빗 안티 BNDF 폴리클로널 항체 (Rabbit anti BDNF polyclonal antibody, AB1534, chemicon)를 1:1000의 비율로 밤새 (overnight) 반응시켰다. 0.05% TBST (Tris-Buffered Saline Tween 20)로 1시간 동안 세척 (20분씩 3번) 한 후, 고트 안티-래빗 IgG:HRP (Goat anti-rabbit IgG:Horseradish Peroxidase Conjugate, SAB-300, Bioreagents)을 1:3000의 비율로 50분 동안 인큐베이션하였다. 0.05% TBST로 1시간 동안 세척 (20분씩 3번)한 후, chemiluminescent HRP substrate kit (Millipore)로 검출 (detection)하였다. 그 결과, 도 5에 나타난 바와 같이 대조군에 비해서 실험군에서 BDNF와 그것의 전구체 (precursor) 형태인 proBDNF의 양이 β-actin 대비 증가한 것을 확인할 수 있었다. A 15% acrylamide gel was made through a 1.5 mm Western blotting gel caster (Gel caster, Bio-Rad) and 30 μg of sample was loaded to obtain an electrophoresis power supply. EPS 601, Amersham) for 2 hours 30 minutes at 100V. Polyvinylidene Difluoride (PVDF) was used to transfer gels (Mighty small transphor, Amersham) using an electrophoresis power supply (EP 301, GE healthcare) at 400 mA for 2 hours and 30 minutes. Blocking PVDF for 1 hour with 7% skimmed milk (Skim milk, Difco Skim milk, BD) and a rabbit anti BDNF polyclonal antibody (AB1534, chemicon) in a ratio of 1: 1000 React overnight. After washing for 1 hour with 0.05% TBST (Tris-Buffered Saline Tween 20) (3 times for 20 minutes), Goat anti-rabbit IgG: Horseradish Peroxidase Conjugate, SAB-300, Bioreagents Incubate for 50 minutes at a ratio of 1: 3000. After washing for 1 hour with 0.05% TBST (3 times for 20 minutes), detection was performed with a chemiluminescent HRP substrate kit (Millipore). As a result, as shown in FIG. 5, the amount of BDNF and its precursor (precursor) form proBDNF in the experimental group was higher than that of the control group, compared to β-actin.
실시예 6: MVG 펩타이드의 세포 독성 효과 측정Example 6: Determination of Cytotoxic Effect of MVG Peptides
H19-7 세포를 96 웰 플래이트 (Well plate, FALCON)에 2X103/200㎕씩 깔고 12시간 동안 34℃, 5% CO2 인큐베이터에서 배양한 후, MVG 합성 펩타이드를 10nM에서 1mM까지 처리하고 3일 동안 배양하였다. 3일 후에 MTT (50mg/ml) 용액을 20㎕씩 처리하고 알루미늄 호일로 싼 후 다시 인큐베이터에서 배양하였다. 4시간 후에 배지와 MTT용액을 제거하고 DMSO (Dimetyl sulfoxide) 용액을 200㎕씩 처리하고 호일로 싼 후 실온에 4시간 동안 두었다. 마이크로플래이트 리더 (Microplate reader, Molecular device)를 이용하여 570nm에서 흡광도를 측정하여 대조군을 100%로 환산하여 그 값을 계산하였다. 그 결과, 도 6에 나타난 바와 같이 대조군과 비교하였을 때 10nM에서 1mM까지의 농도에서 세포 독성을 나타내지 않았다. 따라서, 본 발명에 따른 MVG 합성 펩타이드는 해마신경세포주, 해마조직 및 대뇌 피질조직에서 BDNF의 단백질 발현을 증가시키며, 세포독성이 없으므로 알츠하이머병, 파킨슨병, 만성스트레스성 우울증, 뇌졸중, 헌팅톤무도병, 대뇌허혈성질환, 신경퇴화질환, 당뇨병성 신경병증 등의 예방 및 치료용 약학 조성물로서 유효하게 사용될 수 있다. H19-7 cells were incubated in 96 well plates (FALCON) at 2 × 10 3/200 μl and incubated in 34 ° C., 5% CO 2 incubator for 12 hours, and then treated with MVG synthetic peptide from 10 nM to 1 mM and 3 days Incubated for After 3 days, 20 μl of MTT (50 mg / ml) solution was treated, wrapped in aluminum foil, and incubated again in an incubator. After 4 hours, the medium and the MTT solution were removed, treated with 200 μl of DMSO (Dimetyl sulfoxide) solution, wrapped in foil, and placed at room temperature for 4 hours. The absorbance was measured at 570 nm using a microplate reader (Molecular device) to convert the control to 100% to calculate the value. As a result, as shown in Figure 6 it did not show cytotoxicity at a concentration from 10nM to 1mM compared to the control. Therefore, MVG synthetic peptide according to the present invention increases the protein expression of BDNF in hippocampal neuronal cell line, hippocampal tissue and cerebral cortical tissue, and because there is no cytotoxicity, Alzheimer's disease, Parkinson's disease, chronic stress depression, stroke, Huntington's chorea, It can be effectively used as a pharmaceutical composition for the prevention and treatment of cerebral ischemic diseases, neurodegenerative diseases, diabetic neuropathy and the like.
실시예 7: MVG 펩타이드의 학습 및 기억 증진 효과 측정Example 7 Measurement of Learning and Memory Enhancing Effects of MVG Peptides
7-1.7-1. Y-maze TESTY-maze TEST
MVG 펩타이드가 실험동물수준에서 공간 학습 및 기억력을 증진시키는지 보기 위해서 공간 기억 테스트 중 하나인 Y-maze test을 수행하였다. Y-maze TEST (도 7a)는 첫번째 Trial에서는 novel arm을 막아 놓고 쥐를 15분 동안 출발시켜, 다른 arm을 탐색하도록 한 후, 두번째 Trial에서 5분동안 novel arm으로 들어가는 횟수와 novel arm에서의 이동거리를 조사하여 기억력이 증진되었는지 확인하는 방법으로 수행되었다.The Y-maze test, one of the spatial memory tests, was performed to see if MVG peptides enhance spatial learning and memory at the experimental animal level. Y-maze TEST (FIG. 7A) shows that the first arm blocked the novel arm and started the rat for 15 minutes to explore the other arm, and then moved to the novel arm for 5 minutes and moved on the novel arm in the second trial. This was done by examining the distance to see if memory was improved.
㈜ 오리엔트사로 부터 4주령 male 쥐를 구입하여 1주일간 안정화 시킨 후 꼬리 정맥을 통하여 MVG 펩타이드를 0.1μg/kg을 주사한 후 6시간 후에 첫 번째 Trial을 하고 그 이후 6시간이 지난 후 두 번째 Trial을 하였다. (도 7b)Four weeks old male rats were purchased from Orient Co., Ltd. and stabilized for one week. After injection of 0.1 μg / kg of MVG peptide through the tail vein, the first trial was performed 6 hours later, and the second trial was passed 6 hours later. It was. (FIG. 7B)
두번째 Trial에서 novel arm으로 들어가는 횟수 및 novel arm에서 이동한 거리를 대조군과 비교한 결과 MVG 펩타이드를 주사한 그룹에서 novel arm으로 보다 유의하게 많이 들어가며, 보다 유의하게 이동거리가 증가한 것을 확인하였으며, 이는 MVG 펩타이드를 주사한 그룹이 학습 및 기억 부분에서 증진된 활성을 가짐을 나타낸다(도 8) As a result of comparing the number of entry into the novel arm from the second trial and the distance from the novel arm to the control group, the MVG peptide-injected group entered the novel arm more significantly and the movement distance increased more significantly. Groups injected with peptides show enhanced activity in the learning and memory portions (FIG. 8)
7-2. 모리스 수중 미로 TEST7-2. Morris Underwater Maze TEST
공간 학습 및 기억력을 측정하는 모리스 수중 미로 (Morris water maze) 테스트(도 9a)를 수행하였으며, 물이 들어있는 수조에 쥐를 떨어뜨린 후 수조 밖에 설치된 visual cues 인식하고 그것을 바탕으로 platform에 위치를 알게 얼마나 빨리 platform에 도달하는지 시간을 측정하여 공간 학습 및 기억력을 측정하였다The Morris water maze test (Fig. 9a), which measures spatial learning and memory, was performed. The mice were dropped in a tank containing water, and the visual cues installed outside the tank were recognized and the location of the platform was known. We measured spatial learning and memory by measuring how quickly we arrived at the platform.
㈜ 오리엔트사로부터 4주령 male 쥐를 구입한 후 1주일간 안정화 거친 후, 하루 4번씩 총 3일간 모리스 수중 미로 테스트를 수행하였다. MVG 펩타이드를 0.1μg/kg을 꼬리 정맥을 통하여 주사한 후 6시간 후에 4번의 테스트를 수행하였고, 3일간의 모리스 수중 미로 테스트를 거친 후 48시간 후에 platform을 제거하고 platform에 있던 곳에 머물렀던 시간을 측정하였다. (도 9b)Four weeks old male rats were purchased from Orient Co., Ltd., and then stabilized for one week, followed by a four-day Morris underwater maze test. Four tests were performed 6 hours after the injection of 0.1 μg / kg MVG peptide through the tail vein, and after 48 hours of Morris water maze testing, the platform was removed and the time spent on the platform was measured 48 hours later. It was. (FIG. 9B)
실험결과, 모리스 수중 미로 테스트 첫째날 4번의 Trial에서는 두 그룹간에 platform을 찾아가는데 유의한 차이가 없었으나, 둘째날 첫 번째 및 세번째, 네번째 trial에서는 MVG 펩타이드를 주사한 그룹이 대조군에 비해 유의하게 빨리 platform에 찾아가는 것을 확인하였으며, 셋째날 첫 번째 trial에서 MVG 그룹이 대조군에 비해 유의하게 빨리 찾아감을 발견하였다. 또한, 셋째날 마지막 trial 후 48시간이 지난 다음 probe test (얼마나 오랫동안 platform이 있었던 곳에 존재하는지 시간 측정)를 수행 한 결과, 대조군에 비해 MVG 펩타이드를 주사한 그룹이 platform이 있었던 target quadrant에 유의하게 오랫동안 머무르고, Platform이 있었던 곳을 제외한 다른 quadrant에 존재하는 시간이 MVG펩타이드를 주사한 그룹에서 유의하게 감소함을 확인하였다.The experimental results showed that there was no significant difference between the two groups in the trial of four trials on the first day of the Morris Maze test, but in the first, third and fourth trials on the second day, the group injected with MVG peptide was significantly faster than the control group. In the first trial on the third day, the MVG group was found to be significantly faster than the control group. In addition, after 48 hours after the last trial on the third day, a probe test (measuring how long the platform was present) showed that the group injected with the MVG peptide was significantly longer in the target quadrant where the platform was compared to the control group. The time remaining in the quadrant other than where the platform resided was significantly reduced in the group injected with the MVG peptide.
라서, MVG펩타이드가 공간 학습 및 기억을 증진시킴을 동물실험을 통하여 확인할 수 있었다. (도 10) Thus, animal experiments confirmed that MVG peptides enhance spatial learning and memory. (Figure 10)
7-3. Y-maze 및 모리스 수중 미로 TEST 수행 쥐의 해마 및 대뇌피질에서 MVG 펩타이드에 의한 BDNF 단백질 발현의 증가7-3. Increase of BDNF Protein Expression by MVG Peptide in Hippocampus and Cerebral Cortex of Rats Performing Y-maze and Morris Underwater Maze TEST
Y-maze 및 모리스 수중 미로 테스트를 수행한 쥐의 해마 및 대뇌피질을 적출한 후 웨스턴 블라팅을 이용하여 BDNF 발현을 비교하였으며, 실험방법은 실시예 4 및 5와 동일하다. 실험결과, 대조군에 비해서 MVG 펩타이드를 주사한 그룹에서 BDNF 발현이 증가함을 확인하였으며, 따라서, Y-maze 및 모리스 수중 미로 TEST에서 확인된 학습 및 기억 증진이 해마(도 11a) 및 대뇌피질(도 11b)에서의 MVG 펩타이드에 의한 BDNF 발현 증가에 의한 것임을 알 수 있다.  The hippocampus and cerebral cortex of mice subjected to the Y-maze and Morris underwater maze test were extracted, and BDNF expression was compared using Western blotting. Experimental methods were the same as in Examples 4 and 5. As a result, it was confirmed that the expression of BDNF increased in the group injected with the MVG peptide compared to the control group, therefore, the learning and memory enhancement confirmed in the Y-maze and Morris underwater maze TEST was found in the hippocampus (Fig. 11a) and the cerebral cortex (Fig. It can be seen that the increase in BDNF expression by the MVG peptide in 11b).
실시예 8: MVG 펩타이드의 TrkB 발현 증진 효과 측정Example 8: Determination of TrkB Expression Enhancement Effect of MVG Peptides
MVG 펩타이드에 의하여 BDNF의 수용체인 TrkB의 인산화 및 TrkB의 발현을 비교하기 위하여, 상기 실시예 7의 Y-maze 및 모리스 수중 미로 TEST 수행 쥐들의 적출된 뇌에서 해마 및 대뇌피질 부분만을 각각 분리한 후, RIPA 완충액과 단백질 분해효소 저해제를 상기의 실시예 1과 동일한 비율로 준비하여 분쇄기 (Homogenizer) (RZR2020M, Heidolph)를 이용하여 해마 및 대뇌피질 부분만을 각각을 완전히 분쇄하였다. 이를 4℃, 2,000rpm에서 30분 동안 원심분리 (Centrifuge, UNION 32R, Hanil Science Industrial Co.,Ltd)하여 상층액만 1.5㎖ 튜브로 옮긴 후 이를 다시 4℃, 9,000rpm에서 30분 동안 원심분리 (Centrifuge, MICRO 17TR, Hanil Science Industrial Co., Ltd) 하여 다시 한번 상층액만을 1.5㎖ 튜브로 옮겨 브래드포드 (Bradford assay)를 통해서 단백질을 정량하였다. In order to compare the phosphorylation of TrkB and TrkB expression of BDNF receptor by MVG peptide, only the hippocampus and cerebral cortex sections were isolated from the brains of the Y-maze and Morris water maze TEST mice of Example 7, respectively. RIPA buffer and protease inhibitors were prepared in the same ratio as in Example 1, and only the hippocampus and the cerebral cortex portion were completely ground using a Homogenizer (RZR2020M, Heidolph). Centrifuge at 4 ° C and 2,000rpm for 30 minutes (Centrifuge, UNION 32R, Hanil Science Industrial Co., Ltd), transfer only the supernatant to 1.5ml tube, and then centrifuge for 30 minutes at 4 ° C, 9,000rpm ( Centrifuge, MICRO 17TR, Hanil Science Industrial Co., Ltd) was used to quantify the protein by Bradford assay once again transfer only the supernatant to 1.5ml tube.
1.5mm 웨스턴 블라팅 (Western blotting) 겔 케스터 (Gel caster, Bio-Rad)을 통하여 15% 아크릴아미드 겔 (acrylamide gel)을 만들고 30㎍의 샘플을 로딩 (Loading)하여 전기영동 장치 (Electrophoresis Power supply, EPS 601, Amersham)로 100V에서 2시간 30분 동안 러닝하였다. PVDF (Polyvinylidene Difluoride, Millipore)을 이용하여 400mA에서 2시간 30분 전기영동장치 (Electrophoresis Power supply, EPS 301, GE healthcare)를 통해 겔을 트랜스퍼 (Mighty small transphor, Amersham)하였다. 7% 스킴 밀크 (Skim milk, DifcoTM Skim milk, BD)로 PVDF를 1시간 동안 블로킹 (blocking)하고 래빗 안티 TrkB 모노클로널 항체 (Rabbit anti TrkB monoclonal antibody, Cell signaling)를 1:1000의 비율로 밤새 (overnight) 반응시켰다. 0.05% TBST (Tris-Buffered Saline Tween 20)로 1시간 동안 세척 (20분씩 3번) 한 후, 고트 안티-래빗 IgG:HRP (Goat anti-rabbit IgG:Horseradish Peroxidase Conjugate, SAB-300, Bioreagents)을 1:3000의 비율로 50분 동안 인큐베이션하였다. 0.05% TBST로 1시간 동안 세척 (20분씩 3번)한 후, chemiluminescent HRP substrate kit (Millipore)로 검출 (detection)하였다. 그 결과, 해마 및 대뇌피질에서 대조군에 비해서 MVG 펩타이드를 주사한 그룹에서 TrkB의 인산화 및 TrkB의 발현이 증가함을 확인할 수 있었으며, (각각 도 12a,도 12b) MVG 펩타이드에 의한 공간 학습 및 기억 증진이 해마의 BDNF 발현 증가에 따라 그 수용체의 활성화 및 수용체의 발현 증진에 의한 것임을 알 수 있다.A 15% acrylamide gel was made through a 1.5 mm Western blotting gel caster (Gel caster, Bio-Rad) and 30 μg of sample was loaded to obtain an electrophoresis power supply. EPS 601, Amersham) for 2 hours 30 minutes at 100V. Polyvinylidene Difluoride (PVDF) was used to transfer the gel (Mighty small transphor, Amersham) using an electrophoresis power supply (EP 301, GE healthcare) at 400 mA for 2 hours and 30 minutes. Blocking PVDF for 1 hour with 7% skimmed milk (Skim milk, Difco Skim milk, BD) and rabbit anti TrkB monoclonal antibody (Cell signaling) at a ratio of 1: 1000 Reacted overnight. After washing for 1 hour with 0.05% TBST (Tris-Buffered Saline Tween 20) (3 times for 20 minutes), Goat anti-rabbit IgG: Horseradish Peroxidase Conjugate, SAB-300, Bioreagents Incubate for 50 minutes at a ratio of 1: 3000. After washing for 1 hour with 0.05% TBST (3 times for 20 minutes), detection was performed with a chemiluminescent HRP substrate kit (Millipore). As a result, it was confirmed that TrkB phosphorylation and TrkB expression were increased in the group injected with MVG peptide in the hippocampus and cerebral cortex (FIG. 12A and FIG. 12B, respectively) and enhance spatial learning and memory by MVG peptide. The increase in BDNF expression in the hippocampus may be attributed to the activation of the receptor and enhanced expression of the receptor.
실시예 9: MVG 펩타이드의 anti-parkinson 효과 측정Example 9: Determination of anti-parkinson effect of MVG peptide
BDNF 발현을 조절하는 MVG 펩타이드의 anti-parkinson 효과를 확인하는 in vitro 실험을 수행하였다. MVG 펩타이드를 human dopaminergic cell line인 SH-SY5Y 세포주에 24시간동안 0.001, 0.01, 0.1, 1nM 농도로 처리하고 BDNF, anti-apoptotic protein인 Bcl-2의 발현을 웨스턴 블라팅으로 확인하였다. 실시예 1과 동일한 방법으로 단백질을 얻고 1.5mm 웨스턴 블라팅 (Western blotting) 겔 케스터 (Gel caster, Bio-Rad)을 통하여 15% 아크릴아미드 겔 (acrylamide gel)을 만들고 20㎍의 샘플을 로딩 (Loading)하여 전기영동 장치 (Electrophoresis Power supply, EPS 601, Amersham)로 100V에서 2시간 30분 동안 러닝하였다. PVDF (Polyvinylidene Difluoride, Millipore)을 이용하여 400mA에서 2시간 30분 전기영동장치 (Electrophoresis Power supply, EPS 301, GE healthcare)를 통해 겔을 트랜스퍼 (Mighty small transphor, Amersham)하였다. 7% 스킴 밀크 (Skim milk, DifcoTM Skim milk, BD)로 PVDF를 1시간 동안 블로킹 (blocking)하고 래빗 안티 BNDF 폴리클로널 항체 (Rabbit anti BDNF polyclonal antibody, AB1534, chemicon), 마우스 안티 Bcl-2 모노클로널 항체 (Mouse anti Bcl-2 antibody, sc-7382, Santa Cruz)를 1:1000의 비율로 밤새 (overnight) 반응시켰다. 0.05% TBST (Tris-Buffered Saline Tween 20)로 1시간 동안 세척 (20분씩 3번) 한 후, 고트 안티-래빗 IgG:HRP (Goat anti-rabbit IgG:Horseradish Peroxidase Conjugate, SAB-300, Bioreagents), 고트 안티-마우스 IgG:HRP (Goat anti-mouse IgG:Horseradish Peroxidase Conjugate, sc-2005, Santa Cruz)을 1:3000, 1:1000의 비율로 50분 동안 인큐베이션하였다. 0.05% TBST로 1시간 동안 세척(20분씩 3번)한 후, chemiluminescent HRP substrate kit (Millipore)로 검출(detection)하였다.In vitro experiments were conducted to determine the anti-parkinson effect of MVG peptides that regulate BDNF expression. MVG peptides were treated in SH-SY5Y cell line, a human dopaminergic cell line, at concentrations of 0.001, 0.01, 0.1, and 1 nM for 24 hours, and the expression of BDNF, an anti-apoptotic protein, Bcl-2, was confirmed by Western blotting. Obtain protein in the same manner as in Example 1, make 15% acrylamide gel through 1.5 mm Western blotting gel caster (Gel caster, Bio-Rad) and load 20 μg sample ) Was run for 2 hours and 30 minutes at 100V with an electrophoresis device (Electrophoresis Power supply, EPS 601, Amersham). Polyvinylidene Difluoride (PVDF) was used to transfer the gel (Mighty small transphor, Amersham) using an electrophoresis power supply (EP 301, GE healthcare) at 400 mA for 2 hours and 30 minutes. Blocking PVDF for 1 hour with 7% skimmed milk (Skim milk, Difco Skim milk, BD), rabbit anti BDNF polyclonal antibody (AB1534, chemicon), mouse anti Bcl-2 Monoclonal antibody (Mouse anti Bcl-2 antibody, sc-7382, Santa Cruz) was reacted overnight at a ratio of 1: 1000. Goat anti-rabbit IgG: Horseradish Peroxidase Conjugate, SAB-300, Bioreagents, after washing for 1 hour with 0.05% TBST (Tris-Buffered Saline Tween 20) (3 times 20 min) Goat anti-mouse IgG: Horseradish Peroxidase Conjugate, sc-2005, Santa Cruz was incubated at a ratio of 1: 3000, 1: 1000 for 50 minutes. After washing for 1 hour with 0.05% TBST (3 times for 20 minutes), detection was performed with a chemiluminescent HRP substrate kit (Millipore).
실험 결과, 24h 동안 0.1nM의 MVG 펩타이드를 처리한 상태에서 BDNF, Bcl-2의 발현이 대조군에 비해 증가함을 확인하였다.(도 13a)As a result, it was confirmed that the expression of BDNF and Bcl-2 was increased compared to the control group in the state treated with 0.1 nM MVG peptide for 24 h (FIG. 13A).
또한, MPP+을 이용하여 BDNF 조절 펩타이드의 anti-parkinson 효과를 확인하기 위하여, SH-SY5Y 세포주에 MVG 펩타이드를 0.1nM로 24h 동안 선 처리후 (pretreatment) 2.5mM의 MPP+ (D048, Sigma) 을 처리하고, MPP+ 처리 12시간, 24시간 후에 MTT assay을 실시예 6과 동일한 방법으로 Cell viability를 측정하였다. MPP+ 처리 12시간, 24시간 후에 MTT (50mg/ml) 용액을 20㎕씩 처리하고 알루미늄 호일로 싼 후 다시 인큐베이터에서 배양하였다. 4시간 후에 배지와 MTT용액을 제거하고 DMSO (Dimetyl sulfoxide) 용액을 200㎕씩 처리하고 호일로 싼 후 실온에 4시간 동안 두었다. 마이크로플래이트 리더 (Microplate reader, Molecular device)를 이용하여 570nm에서 흡광도를 측정하여 대조군을 100%로 환산하여 그 값을 계산하였다. 그 결과, 펩타이드를 선처리한 그룹에서는 12 시간 후에 MPP+만을 처리한 그룹에 비해 9.39%, 24시간 후에는 25.6%의 cell viability가 증가하였다.In addition, in order to confirm the anti-parkinson effect of the BDNF regulatory peptide using MPP + , 2.5 mM MPP + (D048, Sigma) was pretreated after 0.1 h MMGG peptide in SH-SY5Y cell line for 24 h. Cell viability was measured by MTT assay in the same manner as in Example 6 after 12 hours and 24 hours after MPP + treatment. After 12 hours and 24 hours of MPP + treatment, 20 μl of MTT (50 mg / ml) solution was treated, wrapped in aluminum foil, and incubated again in an incubator. After 4 hours, the medium and the MTT solution were removed, treated with 200 μl of DMSO (Dimetyl sulfoxide) solution, wrapped in foil, and placed at room temperature for 4 hours. The absorbance was measured at 570 nm using a microplate reader (Molecular device) to convert the control to 100% to calculate the value. As a result, in the group pretreated with peptide, cell viability of 9.39% and 25.6% increased after 24 hours compared to the group treated with MPP + only after 12 hours.
MVG 펩타이드를 선처리한 그룹에서 유의하게 cell viability가 높음을 확인함으로써 (도 13b) 본 발명에 따른 MVG 펩타이드가 anti-parkinson 효과를 가져, 파킨슨병의 예방 및 치료제로서 기능할 수 있음을 알 수 있다. By confirming that the cell viability is significantly high in the group pretreated with the MVG peptide (FIG. 13B), it can be seen that the MVG peptide according to the present invention has an anti-parkinson effect, and thus may function as a preventive and therapeutic agent for Parkinson's disease.
이상에서 본 발명을 특정 실시예들을 중심으로 하여 설명하였지만, 본 발명의 취지 및 첨부된 특허청구범위 내에서 다양한 변형, 변경 또는 수정이 당해 기술 분야에서 가능하며, 따라서 전술한 설명 및 도면은 본 발명의 기술사상을 한정하는 것이 아니라 본 발명을 예시하는 것으로 해석되어져야 한다. 본 발명에서 인용된 모든 문헌은 본 발명의 명세서에 참조로서 통합된다.While the invention has been described above with reference to specific embodiments, various modifications, changes or modifications are possible in the art within the spirit and scope of the appended claims, and thus the foregoing description and drawings illustrate the invention. It should not be construed as limiting the technical spirit of the present invention but as illustrating the present invention. All documents cited in the present invention are incorporated herein by reference.

Claims (11)

  1. 서열번호 1에 기재된 아미노산으로 구성된 펩타이드Peptides consisting of the amino acids set forth in SEQ ID NO: 1
  2. 제 1항에 있어서, 상기 펩타이드는 BDNF (Brain-derived neurotrophic factor)의 mRNA 발현 또는 단백질 발현을 증가시키는 것을 특징으로 하는 펩타이드.The peptide of claim 1, wherein the peptide increases mRNA expression or protein expression of a brain-derived neurotrophic factor (BDNF).
  3. 제 2항에 있어서, 해마신경세포, 해마조직 및 대뇌 피질조직에서 BDNF (Brain-derived neurotrophic factor)의 mRNA 발현 또는 단백질 발현을 증가시키는 펩타이드. The peptide according to claim 2, wherein the peptide increases the mRNA expression or protein expression of brain-derived neurotrophic factor (BDNF) in hippocampal neurons, hippocampal tissue and cerebral cortex.
  4. 제 2항 또는 제 3항에 있어서, 해마신경세포는 마우스의 HT22 세포주 또는 래트의 H19-7 세포주임을 특징으로 하는 펩타이드. The peptide according to claim 2 or 3, wherein the hippocampal neurons are HT22 cell lines of mice or H19-7 cell lines of rats.
  5. 제 1항에 있어서, 상기 펩타이드는 TrkB의 단백질 발현을 증가시키는 것을 특징으로 하는 펩타이드.  The method of claim 1, wherein the peptide is TrkB Peptides characterized by increasing protein expression.
  6. 제 1항의 펩타이드를 코딩하는 DNA 서열로 구성된 폴리뉴클레오티드. A polynucleotide consisting of a DNA sequence encoding the peptide of claim 1.
  7. 제 6항의 폴리뉴클레오티드를 포함하는 유전자 전달체. Gene delivery system comprising the polynucleotide of claim 6.
  8. 제 1항의 펩타이드 또는 제 7항의 유전자 전달체의 치료학적 유효량을 포함하는, 알츠하이머병, 파킨슨병, 만성스트레스성 우울증, 뇌졸중, 헌팅톤무도병, 대뇌허혈성질환, 신경퇴화질환, 또는 당뇨병성 신경병증의 예방 및 치료용 약학 조성물. Prevention of Alzheimer's disease, Parkinson's disease, chronic stress depression, stroke, Huntington's chorea, cerebral ischemic disease, neurodegenerative disease, or diabetic neuropathy comprising a therapeutically effective amount of the peptide of claim 1 or the gene carrier of claim 7 And therapeutic pharmaceutical compositions.
  9. 제 8항에 있어서, 펩타이드의 치료학적 유효량은 0.1 ㎍/㎏/일 내지 10 ㎍/㎏/일임을 특징으로 하는 약학 조성물. The pharmaceutical composition of claim 8, wherein the therapeutically effective amount of the peptide is 0.1 μg / kg / day to 10 μg / kg / day.
  10. 제 8항에 있어서, 경구, 피하, 정맥 내, 근육 내, 비강 내, 복강 내, 직장 내, 자궁 내 또는 뇌실내로 투여함을 특징으로 하는 약학 조성물. The pharmaceutical composition of claim 8, which is administered orally, subcutaneously, intravenously, intramuscularly, intranasally, intraperitoneally, rectally, intrauterinely or intraventricularly.
  11. 제 8항에 있어서, 약학 조성물은 정제, 캡슐제, 과립제, 환제, 시럽제, 액제, 유제, 현탁제, 주사제 또는 좌제의 형태로 제조됨을 특징으로 하는 약학 조성물.The pharmaceutical composition according to claim 8, wherein the pharmaceutical composition is prepared in the form of tablets, capsules, granules, pills, syrups, solutions, emulsions, suspensions, injections or suppositories.
PCT/KR2009/002421 2008-05-09 2009-05-08 Novel peptide for augmenting brain-derived neutrotrophic factor (bdnf) protein expression in hippocampal neurons, hippocampal tissue and cerebral-cortex tissue WO2009136752A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
KR10-2008-0043266 2008-05-09
KR1020080043266A KR100973641B1 (en) 2008-05-09 2008-05-09 A novel peptide for increasing the protein expression of Brain-derived neurotrophic factor in the hippocampal neuronal cell, the hippocampal tissue and the cerebral cortex tissue

Publications (3)

Publication Number Publication Date
WO2009136752A2 WO2009136752A2 (en) 2009-11-12
WO2009136752A9 true WO2009136752A9 (en) 2010-01-28
WO2009136752A3 WO2009136752A3 (en) 2010-03-18

Family

ID=41265173

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2009/002421 WO2009136752A2 (en) 2008-05-09 2009-05-08 Novel peptide for augmenting brain-derived neutrotrophic factor (bdnf) protein expression in hippocampal neurons, hippocampal tissue and cerebral-cortex tissue

Country Status (2)

Country Link
KR (1) KR100973641B1 (en)
WO (1) WO2009136752A2 (en)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR101138048B1 (en) * 2009-11-06 2012-04-23 성균관대학교산학협력단 Novel peptides upregulating BDNF expression and pharmaceutical composition for protection and therapy against Alzheimer's disease and Parkinson's disease comprising the same
KR102123021B1 (en) 2018-07-26 2020-06-16 재단법인 대구경북과학기술원 Serotonin reuptake inhibitor-based antidepressant responsiveness modulating peptides and uses thereof

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5189020A (en) * 1989-11-22 1993-02-23 Neurex Corporation Method of reducing neuronal damage using omega conotoxin peptides
EP0971956A2 (en) 1997-03-24 2000-01-19 Myelos Corporation Synthetic saposin c-derived neurotrophic peptides
US7259146B2 (en) * 2000-05-26 2007-08-21 Ortho-Mcneil Pharmaceutical, Inc. Neuroprotective peptides
JP2005240653A (en) * 2004-02-26 2005-09-08 Denso Corp Fuel supply device

Also Published As

Publication number Publication date
KR100973641B1 (en) 2010-08-02
WO2009136752A2 (en) 2009-11-12
WO2009136752A3 (en) 2010-03-18
KR20090117286A (en) 2009-11-12

Similar Documents

Publication Publication Date Title
KR101138048B1 (en) Novel peptides upregulating BDNF expression and pharmaceutical composition for protection and therapy against Alzheimer's disease and Parkinson's disease comprising the same
WO2016108586A1 (en) Glucagon derivative having improved stability
WO2014182051A1 (en) Composition comprising asm inhibitor as active ingredient for preventing or treating degenerative neurological disorders
WO2012050402A2 (en) Cell-permeable recombinant parkin protein and a pharmaceutical composition for treating degenerative brain diseases containing the same
WO2016190697A1 (en) Pharmaceutical composition for treating sarcopenia comprising glucagon-like peptide-1 receptor agonist
WO2009136752A9 (en) Novel peptide for augmenting brain-derived neutrotrophic factor (bdnf) protein expression in hippocampal neurons, hippocampal tissue and cerebral-cortex tissue
WO2022131756A1 (en) Composition for treating or preventing myopathy, obesity or diabetes
WO2009093864A2 (en) Composition for preventing or treating brain diseases
WO2020091463A1 (en) Pharmaceutical composition comprising isolated mitochondria for preventing or treating tendinopathy
WO2020138674A1 (en) Composition for muscle relaxation
WO2020106048A1 (en) Pharmaceutical composition for prevention or treatment of neurodegenerative disease
WO2022050778A1 (en) Improved cell-permeable modified parkin recombinant protein for treatment of neurodegenerative diseases and use thereof
WO2013022139A1 (en) Method for treatment of diseases caused by accumulation of beta-amyloids in brain parenchyma with low concentration thiazolidinedione derivatives
WO2023055007A1 (en) Peptide having anti-aging activity, and use thereof
WO2020213982A1 (en) Composition for prevention or treatment of neurodegenerative diseases using cd9 and method for screening therapeutic agent for neurodegenerative diseases
WO2020122392A1 (en) Composition for preventing or treating cellular senescence-related diseases comprising zotarolimus as active ingredient
WO2015108372A1 (en) Composition for preventing or treating neurological disorders caused by excitotoxicity or synaptic dysfunction, containing osmotin, and method for preventing or treating neurological disorders by using same
WO2024063223A1 (en) Pharmaceutical composition containing mitochondria-targeting compound as active ingredient for treating macular degeneration
WO2023234703A1 (en) Novel d-amino acid derivative and pharmaceutical composition comprising same
WO2022065923A1 (en) Use of hdac6 as preventive or therapeutic agent for tdp-43 proteinopathy
WO2024019603A1 (en) Protein for preventing or treating brain-nervous system diseases and pharmaceutical composition comprising same
WO2012067305A1 (en) COMPOSITIONS FOR REDUCING BETA-AMYLOID-INDUCED NEUROTOXICITY COMPRISING β-SECRETASE INHIBITOR
WO2022145710A1 (en) Composition for preventing or treating neurodevelopmental, neurological or psychiatric diseases, comprising extracellular vesicles derived from micrococcus luteus
WO2021162460A1 (en) Novel pharmaceutical composition for treating non-alcoholic liver disease
WO2023171982A1 (en) Composition for preventing or treating obesity, diabetes, or nash comprising shlp2 as active ingredient

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: 09742858

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 09742858

Country of ref document: EP

Kind code of ref document: A2