[go: up one dir, main page]

WO2010021516A9 - Novel use of lipocalin 2 for treating brain damage - Google Patents

Novel use of lipocalin 2 for treating brain damage Download PDF

Info

Publication number
WO2010021516A9
WO2010021516A9 PCT/KR2009/004676 KR2009004676W WO2010021516A9 WO 2010021516 A9 WO2010021516 A9 WO 2010021516A9 KR 2009004676 W KR2009004676 W KR 2009004676W WO 2010021516 A9 WO2010021516 A9 WO 2010021516A9
Authority
WO
WIPO (PCT)
Prior art keywords
lcn2
astrocytes
cells
protein
cell
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Ceased
Application number
PCT/KR2009/004676
Other languages
French (fr)
Korean (ko)
Other versions
WO2010021516A3 (en
WO2010021516A2 (en
Inventor
석경호
이신려
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Industry Academic Cooperation Foundation of KNU
Original Assignee
Industry Academic Cooperation Foundation of KNU
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 Industry Academic Cooperation Foundation of KNU filed Critical Industry Academic Cooperation Foundation of KNU
Publication of WO2010021516A2 publication Critical patent/WO2010021516A2/en
Publication of WO2010021516A3 publication Critical patent/WO2010021516A3/en
Publication of WO2010021516A9 publication Critical patent/WO2010021516A9/en
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia

Definitions

  • the present invention relates to a novel use of lipocalin 2 for the treatment of brain injury, and more particularly, a composition for treating brain injury containing a lipocalin 2 (Lipocalin 2, LCN2) inhibitor and a method for treating brain injury by administering the same. It is about.
  • a lipocalin 2 Lipocalin 2, LCN2
  • Astrocytes are the most abundant glia cell type in the brain (Barres, BA et al. 2000, Curr Opin Neurobiol 10: 642-648 Aschner, M. 1998, Neurotoxicology 19: 269-281). Astrocytes provide neuronal metabolism and nutritional support and modulate synaptic activity. They are involved in the formation and maintenance of blood-brain barriers for neuroprotection, antioxidant defense, ion and pH homeostasis, and neuronal-glial networks. In addition to the physiological role of astrocytes in this intact central nervous system (CNS), astrocytes are key responders to damage to the CNS under various physiological conditions such as injury, ischemia, infection and neurodegeneration. (Farina, C. et al.
  • Astrocytes become reactive in response to all forms of CNS damage and undergo a process called reactive astrocytosis (Correa-Cerro, LS et al. 2007, J Neuropathol Exp Neurol 66: 169-176; Sofroniew , MV 2005, Neuroscientist 11: 400-407; Pekny, M. et al. 2005, Glia 50: 427-434).
  • astrocytes proliferate to fill gaps, increase the amount of cytoplasm, protuberance and branching, glial fibrillary acidic protein (GFAP), nestin and nonmethine ( conventional morphological changes such as increased expression of intermediate fibers such as vimetin).
  • GFAP glial fibrillary acidic protein
  • reactive astrocytosis is beneficial or detrimental.
  • Reactive astrocytosis is associated with the protection of nerve damage and the promotion of vascular brain barrier repair.
  • reactive astrocytosis is also associated with potentially harmful effects such as inhibiting the regeneration of axons that act as barriers and increasing the secretion of pro-inflammatory and neurotoxic mediators (Silver, J. et. al. 2004, Nat Rev Neurosci 5: 146-156).
  • lcn2 lipocalin 2
  • MET mesenchymal-epithelial transition
  • Lcn2 is an inducer of endogenous epithelial cells (Yang, J. et al, 2002, Mol Cell 10: 1045-1056) and stimulates transformed cells to regulate invasion and metastasis (Hanai, J. et al. 2005 J Biol Chem 280: 13641-13647; Lee, HJ et al.
  • lcn2 promotes tubulogenesis by regulating morphogenesis of epithelial cells (Lee, HJ et al. 2006, Int J Cancer 118: 2490-2497).
  • Lcn2 is a member of the lipocalin family and binds to or carries lipids and other hydrophobic molecules (Flower, DR et al. 2000, Biochim Biophys Acta 1482: 9-24; Kjeldsen, L. et al. 2000, Biochim Biophys Acta) 1482: 272-283. Lcn2 also contains 24p3 (Flower, DR, et al. 1991. Biochem Biophys Res Commun 180: 69-74), SIP24 (24 kDa superinducible protein) (Hamilton, RT et al.
  • Lcn2 neurotrophil gelatinase-associated lipocalin, a human homologue of lcn2
  • NGAL neurotrophil gelatinase-associated lipocalin, a human homologue of lcn2
  • lcn2 has been suggested as an indicator of active kidney injury (Mori, K. et al. 2007, Kidney Int 71: 967-970). In various forms of gastrointestinal damage, lcn2 facilitates cell regeneration by promoting cell migration (Playford, RJ et al. 2006, Gastroenterology 131: 809-817). However, in vivo studies in mice lacking lcn2 refute the role of lcn2 in protecting kidneys (Berger, T. et al. 2006, Proc Natl Acad Sci USA 103: 1834-1839). In addition, mice lacking lcn2 did not have iron sequestration, which increased the susceptibility to bacterial infection.
  • lcn2 prevents degradation of NACP (Yan, L. et al. 2001, J Biol Chem 276: 37258-37265), and acute phase protein (Liu, Q. et al. 1995, J Biol Chem 270: 22565 -22570) and has been suggested to act as adipokine associated with insulin resistance (Yan, QW et al. 2007, Diabetes 56: 2533-2540; Zhang, J. et al. 2008, Mol Endocrino l 22: 1416-1426). Recently two cellular receptors for lcn2 have been identified.
  • LCN2 is proactively involved in astrocytosis, which inhibits the regeneration of neurons, and thus, by inhibiting LCN2, it is possible to promote the regeneration of neurons after brain injury and to complete the present invention. Reached.
  • the present invention provides a composition for treating brain injury containing an LCN2 inhibitor.
  • the present invention relates to a method of treating brain injury by administering a composition for treating brain injury containing an LCN2 inhibitor.
  • Astrocytes one of the brain's most abundant glial cells, provide neuronal metabolic and nutritional support and modulate synaptic activity.
  • astrocytes amplify, increase the expression of intermediate filament proteins, and become hypertrophic. This process is called (reactive) astrocytosis.
  • Lipocalin 2 is a member of the lipocalin family that binds to small hydrophobic molecules and is involved in various physiological processes and can be highly induced in inflammatory and neoplastic diseases.
  • LCN2 is considered to be an autocrine mediator of reactive astrocytosis. This conclusion is based on the results confirming that LCN2 is involved in the regulation of apoptosis, morphology and migration of astrocytes as follows.
  • LCN2 increases the apoptosis sensitivity of astrocytes to cytotoxic substances. That is, it accelerates the cell death of astrocytes by cytotoxic substances. LCN2 accelerates astrocytic cell death induced by NO, as well as necrosis cell death induced by H 2 O 2 or paraquat.
  • lcn2 is strongly induced by lipopolysaccharide (LPS) and TNF- ⁇ and weakly by a mixture of serum withdrawal, PMA, INF- ⁇ and gangliosides. This means that the expression and secretion of lcn2 in astrocytes increases under inflammatory conditions in the CNS.
  • LPS lipopolysaccharide
  • LCN2 glial fibrillary acidic protein
  • NO amplifies the expression of GFAP in astrocytes (Brahmachari, S. et al. 2006. I J Neurosci 26: 4930-4939). Since the present invention confirmed that LCN2 can also induce the expression of GFAP, it was investigated whether NO is involved in LCN2 action in astrocytes. LCN2 induces an increase in NO production in astrocytes, similar to that of LPS. Expression of GFAP induced by LCN2 by the NOS inhibitor NMMA is blocked, and the sensitivity to cell death induced by LCN2 is attenuated. This means that NO plays an important role in cell death, GFAP expression and morphological changes of astrocytes mediated by LCN2.
  • Rho subfamily of small G proteins is involved in the regulation of astrocyte morphology (Boran, MS et al. 2007. J Neurochem 102: 216-230; Chen, CJ et al. 2006. stellation. Eur J Neurosci 23: 1977-1987; Suidan, HS et al. 1997. Glia 21: 244-252; John, GR et al. 2004. J Neurosci 24: 2837-2845; Ramakers, GJ et al. 1998. Exp Cell Res 245: 252- 262; Holtje, M. et al. 2005. J Neurochem 95: 1237-1248; Hall, A. 2005.
  • Rho protein's relevance in morphological changes of astrocytes induced by LCN2 was investigated.
  • LCN2 induces activation of Rho, while ROCK inhibitors block morphological changes induced by LCN2 and inhibit the expression of GFAP. This means that the Rho / ROCK pathway is involved in morphological regulation of astrocytes.
  • LCN2 a secretory protein of astrocytes, performs two functions that determine the pathway for the shape and function of activated astrocytes (FIG. 36).
  • LCN2 is a protein that leads to the process of astrocytes in astrocytes, so it can be controlled by controlling this protein.
  • Astrocytes are a major cause of inhibition of regeneration of CNS neurons. Therefore, it is possible to suppress astrocytosis by inhibiting the expression of the lcn2 gene or inhibiting the activity of the LCN2 protein, and promote regeneration of CNS neurons as astrocytosis is suppressed.
  • neural cell regeneration ie brain injury treatment is required for all types of brain injury, it is not particularly limited, but degenerative nerves, including, for example, Alzheimer's disease, Parkinson's disease, Lou Gehrig's disease, Huntington's disease and multiple sclerosis Diseases, stroke, trauma, brain infections, Creutzfeldt-Jakob disease, brain inflammatory diseases, and the like.
  • degenerative nerves including, for example, Alzheimer's disease, Parkinson's disease, Lou Gehrig's disease, Huntington's disease and multiple sclerosis Diseases, stroke, trauma, brain infections, Creutzfeldt-Jakob disease, brain inflammatory diseases, and the like.
  • the present invention provides a composition for treating brain injury containing an inhibitor of LCN2.
  • the inhibitor of LCN2 may be an activity inhibitor of LCN2 or an expression inhibitor of LCN2.
  • the activity inhibitor of LCN2 may be, for example, but not limited to, peptides, polypeptides, proteins, peptide replicas, compounds, and biologics.
  • it may be an anti-LCN2 antibody capable of neutralizing the activity of LCN2.
  • the anti-LCN2 antibody may be a polyclonal antibody or a monoclonal antibody.
  • Antibodies of the invention can be prepared by conventional methods well known in the art of immunology using LCN2 protein as an antigen.
  • Polyclonal antibodies can be used in one of ordinary skill in the art from several warm-blooded animals such as horses, cows, goats, sheep, dogs, chickens, turkeys, rabbits, mice or rats. That is, the antigen is immunized to the animal via intraperitoneal, intramuscular, intraocular or subcutaneous injection. Immunity to the antigen can be increased using an adjuvant, for example Freund's complete adjuvant or incomplete adjuvant. Following booster immunization, a small sample of serum is collected and tested for reactivity to the desired antigen. Once the titer of the animal reaches a steady state in terms of its reactivity to the antigen, large amounts of polyclonal immune serum can be obtained by bleeding weekly or by bleeding the animal.
  • an adjuvant for example Freund's complete adjuvant or incomplete adjuvant.
  • Monoclonal antibodies can also be generated using known techniques (Kennettm McKearn and Bechtol (eds.), Monoclonal Antibodies, Hybridomas; A New Dimension in Biological Analyses , Plenum Press, 1980). Monoclonal antibodies immunize animals with LCN2 protein as an immunogen, fusion of splenocytes of the immunized animal with myeloma cells to produce hybridomas, select hybridomas that selectively recognize LCN2 protein, and select high It can be prepared by culturing the bridoma and separating the antibody from the culture medium of the hybridoma.
  • the monoclonal antibody of the present invention can be prepared by injecting the above-mentioned hybridomas producing an anti-LCN2 antibody selectively recognizing LCN2 protein into an animal and separating it from the ascites of the recovered animal after a certain period of time after the injection. Can be.
  • inhibitortion of gene expression includes inhibition of gene transcription and translation into protein. In addition, not only the gene expression is completely stopped, but also the expression is reduced.
  • Antisense molecules are most commonly used as a method of inhibiting gene expression. Antisense molecules inhibit the expression of target genes by inhibiting transcriptional initiation by triple chain formation, transcriptional inhibition by hybridization at sites where local open loop structure is formed by RNA polymerase, and in RNA where synthesis is in progress. Inhibition of transcription by hybrid formation, inhibition of splicing by hybridization at the junction of introns and exons, inhibition of splicing by hybridization at the site of splicosomal formation, transition from nucleus to cytoplasm by hybridization with mRNA Inhibition of translation initiation by hybridization at the site of translation initiation factor binding. They inhibit the expression of target genes by inhibiting transcription, splicing or translation processes.
  • the antisense molecule used in the present invention may inhibit the expression of the target gene by any of the above actions.
  • Representative antisense molecules include triplets, ribozymes, RNAi, or antisense nucleic acids.
  • the triple agent allows the initiation of transcription to be suppressed by winding around double helix DNA to form a three-stranded helix (Maher et al., Antisense Res. And Dev ., 1 (3): 227, 1991; Helene, C., Anticancer Drug Design , 6 (6): 569, 1991).
  • Ribozymes are RNA enzymes that possess the ability to specifically cleave single-stranded RNA. Ribozymes inhibit protein expression of target genes by recognizing and site-specific cleavage of specific nucleotide sequences in target RNA molecules (Cech, J. Amer. Med. Assn ., 260: 3030, 1998; Sarver et al., Science 247: 1222-1225, 1990).
  • RNAi RNA interference
  • RNA interference is a method of inhibiting gene expression at the transcriptional level or at the post-transcriptional level by using RNA of hairpin type small molecule that acts in sequence (Mette et al., EMBO J. , 19: 5194-). 5201, 2000).
  • the small molecule RNA used in the RNAi method is a double-stranded RNA molecule having homology with the target gene.
  • RNA molecules As a method of preparing the RNA molecule in the above, known chemical synthesis methods and enzymatic methods can be used.
  • the chemical synthesis of RNA molecules can use the methods described in the literature (Verma and Eckstein, Annu. Rev. Biochem. 67, 99-134, 1999), and the enzymatic synthesis of RNA molecules is T7, T3.
  • phage RNA polymerases such as SP6 RNA polymerase are disclosed in the literature (Milligan and Uhlenbeck, Methods Enzymol . 180: 51-62, 1989).
  • Antisense nucleic acids refer to DNA or RNA molecules that are at least partially complementary to a target mRNA molecule (Weintraub, Scientific American , 262: 40, 1990). In cells, antisense nucleic acids hybridize with their corresponding mRNAs to form double-stranded molecules that inhibit protein translation by inhibiting mRNA translation of target genes (Marcus-Sakura, Anal. Biochem ., 172: 289, 1988). The antisense nucleic acid may be prepared by any suitable method known in the art, preferably in the form of oligonucleotides.
  • the antisense oligonucleotides may be used in chemical synthesis, for example, such as phosphoramidite chemistry, which is sulfided with tetraethylthiuram disulfide in acetonitrile as described in Tetrahedron Lett ., 1991, 32, 30005-30008. It can manufacture very easily.
  • the LCN2 inhibitor may be administered by any route as long as the LCN2 inhibitor can be directed to the lesion, i.
  • the compositions of the present invention may be used in various forms including topical (including buccal, sublingual, skin and intraocular), parenteral (including subcutaneous, intradermal, intravascular and intraarticular) or transdermal administration. Routes may also be administered, preferably parenterally, most preferably directly at the site where the beta amyloid has been deposited.
  • the LCN2 inhibitor can be administered to a subject by suspending in a suitable diluent, which diluent is used for the purpose of protecting and maintaining the cells and facilitating use when infused into the desired brain tissue.
  • a suitable diluent which diluent is used for the purpose of protecting and maintaining the cells and facilitating use when infused into the desired brain tissue.
  • the diluent may include physiological saline, PBS, HBSS buffer solution, plasma, cerebrospinal fluid or blood components.
  • the LCN2 inhibitor may be used in admixture with a pharmaceutically acceptable carrier according to conventional methods.
  • suitable carriers include lactose, dextrose, sucrose, sorbitol, mannitol, xylitol, maltitol, starch, acacia rubber, alginate, gelatin, calcium phosphate, calcium silicate, cellulose, methyl cellulose, microcrystalline cellulose, Polyvinylpyrrolidone, water, methylhydroxybenzoate, propylhydroxybenzoate, talc, magnesium stearate and mineral oil.
  • the composition may further include a filler, an anticoagulant, a lubricant, a humectant, a perfume, an emulsifier, a preservative, and the like.
  • compositions of the invention can be formulated using methods well known in the art to provide rapid or delayed release of the active ingredient after administration to a subject.
  • the formulations may be in the form of tablets, powders, pills, emulsions, solutions, syrups, aerosols, soft or hard gelatin capsules, sterile injectable solutions, sterile powders and the like.
  • the formulation will be convenient in a single dosage form.
  • composition of the present invention is administered in a pharmaceutically effective amount.
  • pharmaceutically effective amount means an amount sufficient to treat a disease, and an effective dose level may include the severity of the disease; The age, body weight, health and sex of the patient; Sensitivity to the drug of the patient; Time of administration, route of administration, and rate of excretion; Duration of treatment; It may be determined according to factors including drugs used in combination or coincidental with the composition of the present invention and other factors known in the medical field.
  • the pharmaceutical composition of the present invention may vary the effective amount depending on the extent of the disease, preferably 1 to 10,000 g / weight kg / day, more preferably 10 to 1000 mg / kg / day The effective amount may be repeated several times a day.
  • LCN2 inhibitors can inhibit astrocytosis, which inhibits the regeneration of neurons, they can promote neuronal regeneration.
  • LCN2 inhibitors can be used to treat a variety of diseases that require neuronal regeneration, ie, any disease associated with brain damage.
  • LCN2 is involved in the upper stages in the process of astrocytosis, so LCN2 inhibitors can effectively inhibit astrocytosis and promote neuronal regeneration.
  • Figure 1 shows the results of analysis of protein expression and changes in lcn2 mRNA in C6 cells transfected with sense or antisense lcn2 cDNA (S3: sense lcn2 transfectants; AS7: antisense lcn2 transfectants).
  • Figure 2 shows the change in cell viability by SNP, H 2 O 2 and paraquat in C6 cells transfected with sense or antisense lcn2 cDNA (S3: sense lcn2 transfectant; AS7: antisense lcn2 transfectant).
  • Figure 3 shows the results of the analysis of lcn2 expression in C6 cells transfected with adenovirus vectors containing lcn2 DNA fused with GFP.
  • Figure 4 shows the change in cell viability by NO donors SNP, H 2 O 2 and paraquat in C6 cells transfected with adenovirus vectors comprising lcn2 cDNA fused with GFP.
  • 5 shows recombinant mouse LCN2 protein.
  • FIG. 6 shows changes in cell viability when stellate cells were treated with LCN2 alone and when treated with SNP, H 2 O 2 or paraquat.
  • Figure 7 shows the changes in necrosis and apoptosis cell death rate of astrocytes when LCN2 was treated with astrocytes alone and when treated with SNP, H 2 O 2 or paraquat.
  • Figure 9 shows the results confirming whether the LCN2 protein affects the cell cycle distribution of C6 cells and astrocytes.
  • FIG. 10 shows changes in cell viability when astrocytes and C6 cells were treated with iron chelator (DFO) alone and when treated with NO donor SNAP.
  • DFO iron chelator
  • FIG. 11 shows changes in cell viability when treated with iron donors (FC) alone and when treated with SNAP in astrocytes and C6 cells.
  • FIG. 12 shows the change in cell viability when C6 cells were treated with LCN2 and SNAP and co-treated with the cyder pores and iron complexes.
  • FIG. 13 shows changes in Bim RNA and protein amounts in culture of C6 cells treated with LCN2.
  • Figure 14 shows the changes in the amount of Bim RNA and protein in the culture of astrocytes treated with LCN2.
  • Figure 15 shows the results of analyzing the change in the amount of LCN2 expression by the mixture of SNP, LPS, serum clearance (SW), PMA, IFN- ⁇ , TNF- ⁇ and gangliosides.
  • Figure 16 shows the results of analyzing the change in the secretion amount of LCN2 by LPS.
  • Figure 17 shows the results of analyzing the change in the expression amount of LCN2 receptor lcn2R / 24p3R in C6 cells and astrocytes.
  • Figure 18 shows the results of analyzing the changes in morphological changes, protuberance length and cell viability when astrocytes were exposed to LCN2, Forskolin or dbcGMP.
  • Figure 19 shows the morphological changes of astrocytes with the dose of LCN2 and the time of exposure to LCN2.
  • FIG. 20 shows the results of analyzing changes in GFAP mRNA and protein expression levels of astrocytes with the dose of LCN2 and time exposed to LCN2.
  • Figure 21 shows the change in cell viability when co-treatment with SNP and LCN2, Forskolin or dbcGMP in astrocytes.
  • Figure 22 shows the results of analyzing the changes in the LCN2 protein or GFAP expression when treated with Forskolin or dbcGMP in astrocytes.
  • Figure 23 shows the results of analyzing the changes in GFAP mRNA and protein levels by SNP.
  • Figure 24 shows the effect of LCN2 on the NO production of astrocytes.
  • Figure 25 shows the effect of polymyxin B (PB) on the NO production of astrocytes induced by LCN2.
  • PB polymyxin B
  • Figure 26 shows the effect of the NOS inhibitor NMMA on GFAP expression in astrocytes induced by LCN2.
  • 29 shows the effect of Y27632 on the expression of GFAP induced by LCN2.
  • Figure 31 shows the results confirmed the effect of LCN2, Forskolin or dbcGMP on the migration of astrocytes using an in vitro wound treatment assay.
  • Fig. 34 shows the result of confirming the effect on the generation of radial glial cells by injecting LCN2 mRNA into the zebrafish embryo.
  • Figure 35 shows the change in the projection thickness, length and number of radial glial cells by injecting the mRNA of LCN2 into the embryo of zebrafish.
  • FIG. 36 is a schematic showing that LCN2 is involved at higher stages in sensitization and morphological changes of astrocytes to cell death.
  • lipocalin 2 antibody inhibits astrocytosis of astrocytes.
  • Lipopolysaccharide (LPS) obtained from E. coli 0111: B4 was prepared by phenol extraction and gel filtration chromatography.
  • Rho Kinase (ROCK) inhibitor Y27632 was purchased from Calbiochem (La Jolla, Calif.). Recombinant human TNF-a and mouse IFN-g proteins were purchased from R & D Systems (Minneapolis, MN). Iron-saturated enterochelin (0.7 kDa) was purchased from EMC Microcollections GmbH (Tuebingen, Germany). All other compounds were purchased from Sigma Chemical Co. unless otherwise noted.
  • C6 rat glioma cells were maintained in DMEM (Dulbecco's modified Eagle medium) supplemented with 5% heat inactivated fetal bovine serum (FBS) (GibcoBRL Gaithersburg, MD), gentamicin (50 mg / ml).
  • Astrocyte cultures were prepared from the brains of three-day-old ICR mice (Samtako Co .; Osan, Korea) by the method of McCarthy and de Vellis. Whole brains were disassociated in DMEM supplemented with 10% FBS, 100 U / ml penicillin and 100 mg / ml streptomycin (Gibco-BRL).
  • Cells were seeded in 75 cm 2 tissue culture flasks coated with poly D-lysine (Falcon, Becton Dickinson and Company Franklin Lakes, NJ). Cells were incubated at 37 ° C. in 5% CO 2 humid air. The culture medium was changed after every 3 days thereafter. Secondary pure cultures of astrocytes were obtained by shaking the culture of mixed glial cells at 250 rpm overnight and the culture medium was removed. Astrocytes were harvested using trypsin-EDTA and then centrifuged at 1,000 rpm for 10 minutes.
  • astrocytes were inoculated at 1 x 10 5 cells per sphere on sterile cover slips on a 24 sphere plate, then fixed for 4 minutes with 4% formaldehyde and washed twice with PBS. Samples were blocked with 1% BSA dissolved in PBS-Tween 20 and placed in PBS containing 3% BSA and mouse anti-GFAP antibody (1:30 dilution) (Biogenex; San Ramon, CA).
  • the spheres were washed twice with PBS-Tween 20 followed by 2.5 mg / ml of Hoechst 33342 fluorochrome (Molecular Probes Eugene, OR) and anti-mouse IgG-fluorescein isothiocyanate (FITC) -conjugated secondary antibody (BD Biosciences; San Jose , CA). Samples were observed using a fluorescence microscope (Olympus BX50 Tokyo, Japan). Microscopic images were processed with MetaMorph Imaging System (Molecular Devices; Sunnyvale, Calif.). Treatment of astrocytes was performed with a slight modification of known methods (Wilhelmsson, U. et al. 2004. J Neurosci 24: 5016-5021; Boran, MS et al. 2007. J Neurochem 102: 216-230). The average projection length is based on the longest projection for each cell from five microscopic regions containing at least 100 cells optionally selected.
  • Example 5 Flow cytometry of apoptosis and cell cycle
  • Astrocytes were detached using trypsin-EDTA and washed twice with cold PBS. The cells were then resuspended in 250 ml of binding buffer (10 mM HEPES, 140 mM NaCl, 2.5 mM CaCl 2 pH 7.4) and then placed in 3 ⁇ l of FITC-conjugated annexin V (Molecular Probes) according to the manufacturer's instructions. It was. The cells were then vortexed lightly and left under dark conditions for 15 minutes at room temperature. After adding Propidium iodide (20 mg / ml), flow cytometry was performed for 1 hour with FACSAria (BD Biosciences).
  • binding buffer 10 mM HEPES, 140 mM NaCl, 2.5 mM CaCl 2 pH 7.4
  • FITC-conjugated annexin V Molecular Probes
  • Cells were suspended by PBS-5 mM EDTA and analyzed by adding 100% ethanol drops for analysis of cell cycle distribution.
  • RNase A 40 mg / ml
  • Propidium iodide 100 mg / ml was added and left to stand for 30 minutes.
  • the percentage of cells at each stage of the cell cycle was determined by flow cytometry using FACSCalibur (BD Biosciences).
  • Cells in 6-spheres were lysed in three lysis solutions (50 mM Tris-HCl, pH 8.0, 150 mM NaCl, 0.02% sodium azide, 0.1% SDS, 1% NP-40, 0.5% sodium deoxycholate, 1 mM phenylmethylsulfonyl fluoride). Protein concentration in cell lysate was determined using Bio-Rad Protein Assay Kit (Bio-Rad; Hercules, CA). For each sample, the same amount of protein was separated by 12% SDS-PAGE and transferred to a Hybond ECL nitrocellulose membrane (Amersham Biosciences Piscataway, NJ).
  • the cell lysate containing a similar amount of Rho was left standing with GST-Rhotekin immobilized on agarose, and the coprecipitate was subjected to anti-Rho Western blot assay to evaluate the amount of Rho protein bound to GTP. It is known that the anti-Rho antibodies used herein can recognize RhoA, RhoB and RhoC.
  • RT-PCR was performed using total RNA isolated from C6 rat glioma cells.
  • Lcn2 cDNA sequences of rats with sense or antisense orientation were PCR amplified from pooled cDNA using target sequence specific primers by Gateway cloning (Invitrogen).
  • the sequence of the PCR primer for gateway cloning is as follows: sense lcn2 forward, 5'-GGGG ACA AGT TTG TAC AAA AAA GCA GGCT CCA CC ATG GGC CTG GGT GTC CTG TGT -3 '(SEQ ID NO: 1) sense lcn2 reverse , 5'-GGGG ACC ACT TTG TAC AAG AAA GCT GGG TTG TT GTC AAT GCA TTG GTC GGT -3 '(SEQ ID NO: 2) antisense lcn2 forward, 5'-GGGG ACA AGT TTG TAC AAA AAA GCA GGCT CCA CC ATG TCA GTT GTC AAT GCA TTG GTC -3 '(SEQ ID NO: 3) Antisense lcn2 Reverse, 5'-GGGG ACC ACT TTG TAC AAG AAA GCT GGG TTG TT ATG GGC CTG GGT GTC CTG TG T-3' (SEQ ID NO: 4) .
  • the forward primer was used by introducing the attB1 sequence (underlined) followed by the Kozak sequence (CCACC) and the gene specific sequence (bold).
  • reverse primers were used by introducing an attB2 sequence (underlined) followed by a gene specific sequence (bold).
  • PCR products were cloned into the pDONR207 dornor vector (Invitrogen), and the sequence was reconfirmed with (Macrogen Inc. Seoul, Korea) and then converted to the pDS-GFP-XB destination vector (Invitrogen).
  • C6 cells were transformed with lipofectAMINE reagent (Invitrogen) with sense or antisense rat lcn2 cDNA with 4 ⁇ g of GFP (Green fluorescent protein) tag.
  • An empty pEGFP vector was used as a control for stable expression of lcn2.
  • Stable transformants were selected in the presence of G418 (800 ⁇ g / ml) 2 days after transformation. Up or down regulation of lcn2 mRNA or protein in stable transformants was confirmed by RT-PCR or Western blot.
  • Recombinant adenovirus (Ad-lcn2-GFP) expressing rat lcn2-GFP was developed by Newgex Inc. Produced by (Seoul, Korea). That is, cDNA encoding rat lcn2-GFP was inserted into pShuttle-cytomegalovirus vector (Clontech Palo Alto, Calif.). The pShuttle-cytomegalovirus vector containing rat lcn2-GFP was linearized with Pme I and cotransformed with BJ5183 E. coli with a vector of pAdEasy-1 (Clontech) adenovirus.
  • Transformed cells were placed on agarose plates containing kanamycin and each colony was checked for the presence of a suitable transformant. After sequence identification, stocks of recombinant adenovirus were expanded by infection and extraction with HEK-293A cells. Adenovirus was semi-purified from high titer supernatants of infected HEK-293A cells. Supernatants were clarified by centrifugation to remove cell debris and stored at -80 ° C. C6 cells were infected with adenovirus expressing GFP or rat lcn2-GFP for 2 days and observed by fluorescence microscopy. More than 100 cells were identified from several randomly selected regions. Adenoviruses expressing GFP were used as controls.
  • Recombinant mouse LCN2 protein was prepared as known (Yang, J. et al. 2002, Mol cell 10: 1045-1056). In other words, the recombinant LCN2 protein does not synthesize siderophores.
  • E. coli From the BL21 strain It was expressed as glutathione S-transferase (GST) fusion protein. Proteins were purified by using glutathione-Sepharose 4B beads (Amersham Biosciences) and then eluting with thrombin or glutathione.
  • enterokeline EMC Microcollections GmbH
  • EMC Microcollections GmbH saturated with 5-fold molar excess of ions was mixed with recombinant LCN2 protein.
  • zymosan A S. cerevisiae BioParticles, Alexa Fluor 594 conjugate (Molecular Probes)
  • zymosan A S. cerevisiae BioParticles, Alexa Fluor 594 conjugate (Molecular Probes)
  • Molecular Probes Alexa Fluor 594 conjugate
  • Cell migration was measured using a 48-ball Boyden chamber (Neuro Probe, Inc. Gaithersburg, MD). Various concentrations of LCN2 protein or compound in DMEM were placed on a lower wall separated from the upper wall by a polycarbonate filter (8-mm pore size, 25 x 80 mm Neuro Probe, Inc.) without polyvinylpyrrolidone. Located. Cells were recovered by treatment with trypsin, resuspended in DMEM and added to the upper chamber at 1 ⁇ 10 4 cells / sphere. Cells were left at 37 ° C. under 5% CO 2 . Cells were then fixed with methanol for 10 minutes and stained with modified Giemsa stain (Sigma) for 1 hour.
  • scratch wounds were made using a 10 ⁇ l pipette tip in a confluent cell monolayer on a 24-neck culture plate and 10% FBS containing 100 U / ml penicillin and 100 ⁇ g / ml streptomycin. Refilled with DMEM including. Cells were left at 37 ° C. under 5% CO 2 while the monolayer moved to the wound area. The wound area was observed under a microscope (Olympus CK2) (magnification, x 100). Relative cell migration distance was determined by measuring the width of the wound and subtracting it from the initial value (Bassi, R. et al. 2008, J Neurooncol 87: 23-33). A total of three regions were randomly selected and calculated for each sphere. T results are expressed as multiples of the increase in travel distance.
  • Embryos were recovered from crosses and raised in egg water at 28.5 ° C. and developed according to days after fertilization (Park, HC et al. J Neurosci 25: 6836-6844; Kucenas, S. et al. 2008. Nat Neurosci 11: 143 -151).
  • AB and Tg (GFAP: egfp) fish Bosset, RL et al. 2006, Gene Expr Patterns 6: 1007-1013
  • Rat lcn2 cDNA including full-length open reading frame, was subcloned into the pCSDest vector for mRNA injection (Villefranc, JA et al. 2007, Dev Dyn 236: 3077-3087.) mRNA was expressed in the Message Machine Kit (Ambion Austin, TX). Produced as. The production of lcn2 mRNA was confirmed by gel electrophoresis of transcriptional reactants (data not shown). 100 pg of lcn2 mRNA was injected into egg yolk at one to two cell stages. Expression of LCN2 protein in embryos injected with mRNA was confirmed by immunocytochemistry (data not shown).
  • the embryos were fixed at 4 ° C. overnight with AB Fix (4% paraformaldehyde, 8% sucrose, 1 ⁇ PBS), embedded in 1.5% agarose / 30% sucrose and immersed in liquid nitrogen and frozen in frozen 2-methyl butane.
  • a 10 ⁇ m transreverse section was recovered using a cryostat microtome.
  • the following primary antibodies were used for immunocytochemistry: mouse antibody against Zrf-1 (1: 400 dilution, University of Oregon Monoclonal Antibody Facility), rabbit polyclonal anti-LCN2 / NGAL antibody (1: 100 dilution, Santa Cruz Biotech).
  • Example 15 Increases Sensitivity of Astrocytes to Cytotoxic Stimulators
  • lcn2 is involved in the survival and apoptosis of various types of cells
  • the following three methods were used to investigate how lcn2 is involved in the apoptosis of activated astrocytes: 1) Transfection of C6 glial cells with either sense or antisense lcn2 cDNA Stable overexpression or knockdown of lcn2 by; 2) transient expression mediated by adenovirus of lcn2 in C6 cells; And 3) treatment of primary astrocytic cultures or C6 with recombinant LCN2 protein.
  • Transient overexpression of lcn2 was achieved by using an adenovirus vector containing lcn2 cDNA fused with GFP (FIG. 3). Expression of lcn2 increased the sensitivity of C6 glial cells to NO donors SNP, H 2 O 2 and paraquat (FIG. 4).
  • LCN2 protein was prepared and tested for potential cytotoxic effects (FIG. 5). LCN2 protein sensitized primary astrocytic cultures to cell death, whereas LCN2 protein alone did not affect astrocytic survival (FIG. 6). Similar results were obtained from C6 glial cells (data not shown). In order to confirm the properties of cell death, the effect of increasing the cell death of LCN2 protein was evaluated by staining with propidium iodide (PI) and annexin V and flow cytometry (FIG. 7).
  • PI propidium iodide
  • LCN2 Treatment of LCN2 protein increased the sensitivity of apoptosis (PI ⁇ / annexin V + or PI + / annexin V + ) as well as necrosis (PI + / annexin V ⁇ ) of astrocytes.
  • the effect of increasing cell death of LCN2 protein was observed from a dose dependent and statistically significant effect of 0.1 ng / ml of LCN2 (FIG. 8). However, there was no significant effect on cell cycle distribution of C6 glial or primary astrocytic cells (FIG. 9).
  • the iron supply to the cells reduces the amount of transferrin receptor (TfR1) expression and increases the amount of ferritin.
  • TfR1 transferrin receptor
  • the influx of iron into the cell reduces the amount of expression of Bim protein, a proapoptosis protein.
  • Bim protein transferrin receptor 1
  • intracellular mammalian ciderpore iron complex binds to lcn2 and is released out of the cell by exocytosis. Depletion of iron in cells leads to upregulation of Bim, a proapoptotic molecule.
  • Example 17 Expression and Control of lcn2 and lcn2 Receptors (lcn2R / 24p3R) in Astrocytes
  • lcn2 has been proposed as an acute phase protein (Liu, Q. et al. 1995. J Biol Chem 270: 22565-22570) and expression of lcn2 was regulated by immune stimulators in macrophages (Liu, Q. et al. 1995 J Biol Chem 270: 22565-22570; Meheus, LA et al. 1993. J Immunol 151: 1535-1547; Cowland, JB et al. 2003. J Immunol 171: 6630-6639. Therefore, it was confirmed whether the expression of lcn2 is regulated by immunity or other stimulator in the phase.
  • LCN2 induced morphological changes in astrocytes in addition to apoptosis sensitization effects.
  • astrocytes were exposed to LCN2 protein, the number of cell processes increased without affecting cell viability (FIG. 18).
  • cyclic AMP (cAMP) and cyclic GMP (cGMP) are known to induce similar morphological changes (Boran, MS et al. 2007. J Neurochem 102: 216-230; Hu, W. et al. 2008.
  • Forskolin and dibutyryl cyclic GMP (dbcGMP) were used for comparison ( Cell Mol Neurobiol 28: 519-528) (FIG. 18).
  • Morphological changes in astrocytes induced by LCN2 were dose and time dependent (FIG. 19). Significant changes occurred at 12 ng / ml of LCN2 and 12 hours after exposure thereto. They have been shown to increase the expression of lcn2 in astrocytes (FIGS. 15 and 16) and to induce activation of glial cells (Jou, I. et al. 2006. Am J Pathol 168: 1619-1630; Yoon, HJ et al. 2008. Mol Cells 25: 99-104).
  • LCN2 Changes in the cell processes induced by LCN2 are made by upregulation of the expression of GFAP mRNA and protein, which is dependent on the dose and time of LCN2 (FIG. 20). Morphological changes in this type of astrocytes are similar to those occurring in in vivo reactive astrocytes (Sofroniew, MV 2005. Neuroscientist 11: 400-407).
  • Hypertrophy and increased GFAP expression are two features of reactive astrocytic cells that appear after all forms of neuronal damage in vivo (Wilhelmsson, U. et al. 2004. J Neurosci 24: 5016-5021). Changes in astrocyte morphology induced by LCN2 were confirmed to be associated with phenotypes sensitive to cell death. Forskolin and dbcGMP, which induce changes in astrocytic morphology similar to those induced by LCN2, also led to phenotypes sensitive to cell death (FIG. 21). dbcGMP increased LCN2 expression while forskolin did not (FIG. 22).
  • the amount of NO produced induced by LCN2 was similar to that of LPS and was not offset by polymyxin B treatment to rule out the possibility of LPS contamination in recombinant LCN2 preparations (FIG. 25).
  • Recombinant GST protein prepared in the same manner as LCN2 was also used as a control to exclude the possibility of contamination of LPS (FIG. 25).
  • NO production induced by LPS / IFN- ⁇ was completely offset by polymyxin B treatment.
  • Expression of GFAP induced by LCN2 was blocked by the NOS inhibitor NMMA (FIG. 26).
  • the increase in cell death sensitivity induced by LCN2 was attenuated by NMMA (FIG. 27). The above results indicate that NO plays an important role in astrocytic cell death, GFAP expression and morphological changes mediated by LCN2.
  • Rho subfamily of small G proteins is known to be involved in the regulation of astrocyte morphology (Boran, MS et al. 2007. J Neurochem 102: 216-230; Chen, CJ et al. 2006. stellation. Eur J Neurosci 23 : 1977-1987; Suidan, HS et al. 1997. Glia 21: 244-252; John, GR et al. 2004. J Neurosci 24: 2837-2845; Ramakers, GJ et al. 1998. Exp Cell Res 245: 252 Hol262, M. et al. 2005. J Neurochem 95: 1237-1248; Hall, A. 2005. Biochem Soc Trans 33: 891-895).
  • Rho protein in the morphological changes of astrocytes induced by LCN2 was investigated.
  • the Rho / ROCK pathway appears to play an important role in the action of LCN2 on astrocyte morphology according to the following results: 1) ROCK inhibitor Y27632 partially blocked morphological changes induced by LCN2 (FIG. 28); 2) Y27632 also inhibited the expression of GFAP induced by LCN2 (FIG. 29) and 3) LCN2 induced the activation of Rho (FIG. 30).
  • Y27632 had no effect on cell viability at the concentrations used in this example (data not shown).
  • Activation of Rho induced by LCN2 was initiated 1 hour after LCN2 stimulation and continued up to 24 hours (FIG. 30).
  • a zebrafish model was used to confirm the expression of lcn2 and its functional role in in vivo reactive astrocytes.
  • the expression of lcn2 in zebrafish CNS was examined. Labeling of zebrafish embryos with anti-LCN2 polyclonal antibodies showed no signal on day 2 after fertilization (data not shown), but LCN2 + cells were identified in the whole brain and spine on day 3 (A, B in FIG. 33). ). LCN2 antibodies in whole brain sections labeled microglia-like cells in the form of microglia (arrows in A of FIG. 33).
  • Tg (gfap-egfp) embryos which were anti-Zrf-1 markers of radial glial protuberances. It was labeled with an antibody.
  • Tg (gfap-egfp) embryos express EGFP in radial glial cells under the control of the gfap promoter (Bernardos, RL et al. 2006. Gene Expr Patterns 6: 1007-1013). By labeling the processes (Trevarrow, B. et al. 1990.
  • compositions of the present invention can be used to treat brain damage.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Immunology (AREA)
  • Biomedical Technology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Psychiatry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Hospice & Palliative Care (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Epidemiology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present invention relates to a composition containing a lipocalin 2 (LCN2) inhibitor for treating brain damage and a method for treating brain damage by administering the composition. When the central nervous system (CNS) is injured, astrocytes are activated and astrocytosis is induced. Astrocytosis is a main factor causing nerve cell damage and suppressing regeneration of damaged nerve cells. Since the LCN2 mainly induces astrocytosis, additional damage of nerve cells after brain damage is prevented using the LCN2 inhibitor. Also, the regeneration of damaged nerve cells can be promoted.

Description

뇌 손상 치료를 위한 리포칼린 2의 신규한 용도New Uses of Lipocalin 2 to Treat Brain Injury

본 발명은 뇌 손상 치료를 위한 리포칼린 2의 신규한 용도에 관한 것으로, 보다 자세하게는 리포칼린 2(Lipocalin 2, LCN2) 억제제를 함유한 뇌손상 치료용 조성물 및 이를 투여하여 뇌 손상을 치료하는 방법에 관한 것이다.The present invention relates to a novel use of lipocalin 2 for the treatment of brain injury, and more particularly, a composition for treating brain injury containing a lipocalin 2 (Lipocalin 2, LCN2) inhibitor and a method for treating brain injury by administering the same. It is about.

성상세포(astrocyte)는 뇌에서 가장 풍부한 아교세포(glia cell) 유형이다(Barres, B. A. et al. 2000, Curr Opin Neurobiol 10: 642-648 Aschner, M. 1998, Neurotoxicology 19: 269-281). 성상세포는 신경세포에 대사 및 영양적 지원을 제공하고, 시냅스의 활성을 조정한다. 이들은 신경보호를 위한 혈액뇌장벽(blood-brain barrier)의 형성과 유지, 항산화제 방어, 이온과 pH 항상성 및 신경세포-아교세포의 네트워크에 관여한다. 이와 같은 손상되지 않은 중추신경계(central nervous system, CNS)에서 성상세포의 생리적인 역할에 더하여, 성상세포는 손상, 허혈, 감염 및 신경퇴행과 같은 다양한 생리학적 조건하에 있는 CNS의 손상에 대한 주요 반응자이다(Farina, C. et al. 2007, Trends Immunol 28: 138-145). 성상세포는 모든 형태의 CNS 손상에 반응하여 반응성이 되고 반응성 성상세포증(reactive astrocytosis)이라고 하는 과정을 진행하게 된다(Correa-Cerro, L. S. et al. 2007, J Neuropathol Exp Neurol 66: 169-176; Sofroniew, M. V. 2005, Neuroscientist 11: 400-407; Pekny, M. et al. 2005, Glia 50: 427-434). 이 과정에서 성상세포는 증식하여 갭(gap)을 메우고, 세포질의 양이 커지고, 돌기가 길어지고 분지되며, 신경교섬유산단백질(glial fibrillary acidic protein, GFAP), 네스틴(nestin) 및 비메틴(vimetin)과 같은 중간미세섬유의 발현이 증가되는 것과 같은 통상의 형태학적 변화를 수행한다. 반응성 성상세포증의 기본적 과정은 진화적으로 보존되고, 신경외상과 신경퇴행과 같은 모든 형태의 신경 손상에 동반됨에도 불구하고, 반응성 성상세포증이 이로운지 또는 유해한지에 대해서는 잘 알려져 있지 않다. 반응성 성상세포증은 신경 손상에 대한 보호와 혈관뇌장벽 복구의 촉진과 관련이 있다. 이러한 이로운 역할과는 반대로, 반응성 성상세포증은 장벽으로서 작용하는 축삭의 재생성을 억제하고, 전염증 및 신경독성 매개자의 분비를 증가시키는 것과 같은 잠재적으로 해로운 효과와도 연관이 있다(Silver, J. et al. 2004, Nat Rev Neurosci 5: 146-156). 반응성 성상세포의 반대 역할이 뇌의 외상적 손상 후에도 확인되었다(Laird, M. D. et al. 2008, Neurosignals 16: 154-164). CNS 생리에서 반응성 성상세포증의 기능적 역할에 대한 이해는 잠재적 치료술을 위한 새로운 접근으로 이어질 것이다. 이를 위하여, 반응성 성상세포증에 대한 정확한 분자 메커니즘을 이해할 필요가 있다.Astrocytes are the most abundant glia cell type in the brain (Barres, BA et al. 2000, Curr Opin Neurobiol 10: 642-648 Aschner, M. 1998, Neurotoxicology 19: 269-281). Astrocytes provide neuronal metabolism and nutritional support and modulate synaptic activity. They are involved in the formation and maintenance of blood-brain barriers for neuroprotection, antioxidant defense, ion and pH homeostasis, and neuronal-glial networks. In addition to the physiological role of astrocytes in this intact central nervous system (CNS), astrocytes are key responders to damage to the CNS under various physiological conditions such as injury, ischemia, infection and neurodegeneration. (Farina, C. et al. 2007, Trends Immunol 28: 138-145). Astrocytes become reactive in response to all forms of CNS damage and undergo a process called reactive astrocytosis (Correa-Cerro, LS et al. 2007, J Neuropathol Exp Neurol 66: 169-176; Sofroniew , MV 2005, Neuroscientist 11: 400-407; Pekny, M. et al. 2005, Glia 50: 427-434). In this process, astrocytes proliferate to fill gaps, increase the amount of cytoplasm, protuberance and branching, glial fibrillary acidic protein (GFAP), nestin and nonmethine ( conventional morphological changes such as increased expression of intermediate fibers such as vimetin). Although the basic process of reactive astrocytosis is evolutionarily conserved and accompanied by all forms of nerve damage, such as neurotrauma and neurodegeneration, it is not known whether reactive astrocytosis is beneficial or detrimental. Reactive astrocytosis is associated with the protection of nerve damage and the promotion of vascular brain barrier repair. In contrast to this beneficial role, reactive astrocytosis is also associated with potentially harmful effects such as inhibiting the regeneration of axons that act as barriers and increasing the secretion of pro-inflammatory and neurotoxic mediators (Silver, J. et. al. 2004, Nat Rev Neurosci 5: 146-156). The opposite role of reactive astrocytic cells has been identified after traumatic injury of the brain (Laird, MD et al. 2008, Neurosignals 16: 154-164). Understanding the functional role of reactive astrocytosis in CNS physiology will lead to new approaches for potential therapies. To this end, it is necessary to understand the exact molecular mechanisms for reactive astrocytosis.

최근의 연구들은 CNS에서 염증 세포는 림프구의 활성화에 의해 유도되는 세포 사멸(activation-induced cell death, AICD)과 유사한 방식으로 활성화되자마자 아폽토시스 된다는 것을 암시하고 있다(Yang, M. S. et al. 2002, Glia 38: 273-280; Falsig, J. et al. 2004, J Neurochem 88: 181-193; Liu, B., et al. 2001, J Neurochem 77: 182-189; Mabuchi, T. et al. 2000, Stroke 31: 1735-1743; Suk, K. et al. 2001, Brain Res 900: 342-347; Lee, P. et al. 2001, Brain Res 892: 380-385; Suk, K. 2005, Curr Enzyme Inhibition 1: 43-50). 성상세포 뿐만 아니라 소교세포도 강력한 염증 활성화가 일어나자마자 아폽토시스 되는 것으로 확인되었다(Ryu, J. K. et al. 2003, Neurobiol Dis 12: 121-132; Ferrari, D., et al. 1997, Neuropharmacology 36: 1295-1301; Hu, J. et al. 1996, Biochim Biophys Acta 1313: 239-245; Kingham, P. J. et al. 2000, J Neurochem 74: 1452-1462; Takuma, K. et al. 2004, Prog Neurobiol 72: 111-127; Suk, K. et al. 2002, J Neurochem 80: 230-238; Lee, J. et al. 2001, J Biol Chem 276: 32956-32965). 방대한 증폭 후 아교세포의 아폽토시스는 군집화(population)를 제어하는 수단일 수 있다(Jones, L. L. et al. 1997, J Neurocytol 26: 755-770). 그러나, 아교세포의 자동 조절적인 아폽토시스 및 신경염증의 종료에 대한 분자 메커니즘에 대하여 알려진 것이 거의 없다. 산화 질소(N)(Lee, J. et al. 2001, J Biol Chem 276: 32956-32965), BTG1 (Lee, H. et al. 2003, J Immunol 171: 5802-5811), N-myc (Jung, D. Y. et al. 2005, J Neurochem 94: 249-256), TLR4 (Jung, D. Y. et al. 2005, J Immunol 174: 6467-6476), IL-4 (Yang, M. S. et al. 2002, Glia 38: 273-280; Park, K. W. et al. 2005, J Neurosci Res 81: 397-402), IL-13 (Shin, W. H. et al. 2004, 46: 142-152), 사이클로옥시제나제-2(Yang, M. S. et al. 2006, J Immunol 177: 1323-1329) 및 항산화 효소(Yang, M. S. et al. 2007, J Neurosci Res 85: 2298-2305)가 활성화된 소교세포의 사멸과 관련이 있는 것으로 알려져 있다. 본 발명자는 최근에 리포칼린 2(lcn2)가 활성화된 소교세포의 아폽토시스에 의한 제거를 용이하게 한다는 것을 밝혀냈다(Lee, S. et al. 2007, J Immunol 179: 3231-3241). 또한, lcn2는 중간엽세포의 상피세포로의 전이(mesenchymal-epithelial transition, MET)에 관여한다. Lcn2는 내인성 상피세포의 유도자이고(Yang, J. et al, 2002, Mol Cell 10: 1045-1056), 형질전환된 세포를 자극하게 되면 침입 및 전이를 조절한다(Hanai, J. et al. 2005, J Biol Chem 280: 13641-13647; Lee, H. J. et al. 2006, Int J Cancer 118: 2490-249). 또한, lcn2는 상피세포의 형태발생(morphogenesis)를 조절함에 의해 관발생(tubulogenesis)를 촉진한다(Lee, H. J. et al. 2006, Int J Cancer 118: 2490-2497).Recent studies suggest that inflammatory cells in the CNS become apoptized upon activation in a manner similar to activation-induced cell death (AICD) induced by lymphocyte activation (Yang, MS et al. 2002, Glia). 38: 273-280; Falsig, J. et al. 2004, J Neurochem 88: 181-193; Liu, B., et al. 2001, J Neurochem 77: 182-189; Mabuchi, T. et al. 2000, Stroke 31: 1735-1743; Suk, K. et al. 2001, Brain Res 900: 342-347; Lee, P. et al. 2001, Brain Res 892: 380-385; Suk, K. 2005, Curr Enzyme Inhibition 1: 43-50). Astrocytes as well as microglia were found to be apoptized as soon as potent inflammatory activation occurs (Ryu, JK et al. 2003, Neurobiol Dis 12: 121-132; Ferrari, D., et al. 1997, Neuropharmacology 36: 1295-). 1301; Hu, J. et al. 1996, Biochim Biophys Acta 1313: 239-245; Kingham, PJ et al. 2000, J Neurochem 74: 1452-1462; Takuma, K. et al. 2004, Prog Neurobiol 72: 111 Suk, K. et al. 2002, J Neurochem 80: 230-238; Lee, J. et al. 2001, J Biol Chem 276: 32956-32965). Apoptosis of glial cells after extensive amplification may be a means of controlling population (Jones, LL et al. 1997, J Neurocytol 26: 755-770). However, little is known about the molecular mechanisms for the autonomous apoptosis of glial cells and the termination of neuritis. Nitric oxide (N) (Lee, J. et al. 2001, J Biol Chem 276: 32956-32965), BTG1 (Lee, H. et al. 2003, J Immunol 171: 5802-5811), N-myc (Jung , DY et al. 2005, J Neurochem 94: 249-256), TLR4 (Jung, DY et al. 2005, J Immunol 174: 6467-6476), IL-4 (Yang, MS et al. 2002, Glia 38: Park, KW et al. 2005, J Neurosci Res 81: 397-402), IL-13 (Shin, WH et al. 2004, 46: 142-152), cyclooxygenase-2 (Yang, MS et al. 2006, J Immunol 177: 1323-1329) and antioxidant enzymes (Yang, MS et al. 2007, J Neurosci Res 85: 2298-2305) are known to be involved in the death of activated microglia. We have recently discovered that lipocalin 2 (lcn2) facilitates apoptosis removal of activated microglia (Lee, S. et al. 2007, J Immunol 179: 3231-3241). In addition, lcn2 is involved in mesenchymal-epithelial transition (MET) of mesenchymal cells to epithelial cells. Lcn2 is an inducer of endogenous epithelial cells (Yang, J. et al, 2002, Mol Cell 10: 1045-1056) and stimulates transformed cells to regulate invasion and metastasis (Hanai, J. et al. 2005 J Biol Chem 280: 13641-13647; Lee, HJ et al. 2006, Int J Cancer 118: 2490-249). In addition, lcn2 promotes tubulogenesis by regulating morphogenesis of epithelial cells (Lee, HJ et al. 2006, Int J Cancer 118: 2490-2497).

Lcn2는 리포칼린 패밀리의 일원으로, 지질 및 여타 소수성 분자와 결합하거나 그들을 운반한다(Flower, D. R. et al. 2000, Biochim Biophys Acta 1482: 9-24; Kjeldsen, L. et al. 2000, Biochim Biophys Acta 1482: 272-283). 또한, Lcn2는 24p3 (Flower, D. R., et al. 1991. Biochem Biophys Res Commun 180: 69-74), SIP24(24 kDa superinducible protein)(Hamilton, R. T. et al. 1985, J Cell Physiol 123: 201-208) 및 NGAL(neutrophil gelatinase-associated lipocalin, a human homologue of lcn2)(Borregaard, N. et al. 2006, Biometals 19: 211-215; Kjeldsen, L. et al. 1993, J Biol Chem 268: 10425-10432)로서 알려져 있다. Lcn2는 다양한 기능을 가지고 있다. in vitro 연구는 lcn2가 세포의 아폽토시스 및 생존 모두에서 중요하다는 것을 보여주었다(Nelson, A. M. et al. 2008, J Clin Invest 118: 1468-1478; Yousefi, S. et al. 2002, Cell Death Differ 9: 595-597; Devireddy, L. R. et al. 2005, Cell 123: 1293-1305; Devireddy, L. R. et al. 2001, Science 293: 829-834; Tong, Z. et al. 2003, Biochem J 372: 203-210; Tong, Z. et al. 2005, Biochem J 391: 441-448). 배발생(embryogenesis) 중에 신장에서 세포 분화를 유도하는데 중심적인 역할을 하고(Yang, J. et al. 2002, Mol Cell 10: 1045-1056), 신장을 허혈성 손상으로부터 보호한다(Mishra, J. et al. 2004, J Am Soc Nephrol 15: 3073-3082; Mori, K. et al. 2005, J Clin Invest 115: 610-621). lcn2는 활성 신장 손상의 표시자로 제시되어 왔다(Mori, K. et al. 2007, Kidney Int 71: 967-970). 다양한 형태의 위장관 손상에서 lcn2는 세포 이동을 촉진함으로써 점막의 재생을 용이하게 한다(Playford, R. J. et al. 2006, Gastroenterology 131: 809-817). 그러나 lcn2가 결핍된 마우스를 대상으로 한 in vivo 연구는 신장을 보호한다는 lcn2의 역할을 반박하고 있다(Berger, T. et al. 2006, Proc Natl Acad Sci U S A 103: 1834-1839). 또한, lcn2가 결핍된 마우스는 철 분리(iron sequestration)가 되지 않아 박테리아의 감염에 대한 민감성이 증가하였다. 이는 바이러스 감염에 대한 보호에서 lcn2의 중요한 역할을 암시한다(Flo, T. H. et al. 2004, Nature 432: 917-921). 이전의 연구들은 lcn2가 NACP의 분해를 방지하고(Yan, L. et al. 2001, J Biol Chem 276: 37258-37265), 급성기 단백질(Liu, Q. et al. 1995, J Biol Chem 270: 22565-22570) 및 인슐린 저항성과 관련된 아디포카인(adipokine)으로서 작용할 것으로 제안되어 왔다(Yan, Q. W. et al. 2007, Diabetes 56: 2533-2540; Zhang, J. et al. 2008, Mol Endocrino l 22: 1416-1426). 최근에 lcn2에 대한 두 개의 세포 수용체가 동정되었다. Megalin은 저밀도 지질단백질 수용체 패밀리의 인원으로서, 사람의 lcn2과 결합하고 이의 세포로의 유입을 매개하는 것으로 확인되었다(Hvidberg, V. et al. 2005, FEBS Lett 579: 773-777). 마우스 lcn2에 대한 또 다른 세포 표면 수용체이고, 이는 선택적으로 아폽토시스를 매개하는 것으로 확인되었다(Devireddy, L. R. et al. 2001, Science 293: 829-834). 수용체 동정에도 불구하고, 세포 생존 및 사멸에서 lcn2의 정확한 역할은 확인되지 못하고 있다.Lcn2 is a member of the lipocalin family and binds to or carries lipids and other hydrophobic molecules (Flower, DR et al. 2000, Biochim Biophys Acta 1482: 9-24; Kjeldsen, L. et al. 2000, Biochim Biophys Acta) 1482: 272-283. Lcn2 also contains 24p3 (Flower, DR, et al. 1991. Biochem Biophys Res Commun 180: 69-74), SIP24 (24 kDa superinducible protein) (Hamilton, RT et al. 1985, J Cell Physiol 123: 201-208 ) And NGAL (neutrophil gelatinase-associated lipocalin, a human homologue of lcn2) (Borregaard, N. et al. 2006, Biometals 19: 211-215 ; Kjeldsen, L. et al. 1993, J Biol Chem 268: 10425-10432 Is known as Lcn2 has a variety of features. In vitro studies have shown that lcn2 is important in both apoptosis and survival of cells (Nelson, AM et al. 2008, J Clin Invest 118: 1468-1478; Yousefi, S. et al. 2002, Cell Death Differ 9: 595-597; Devireddy, LR et al. 2005, Cell 123: 1293-1305; Devireddy, LR et al. 2001, Science 293: 829-834; Tong, Z. et al. 2003, Biochem J 372: 203-210 Tong, Z. et al. 2005, Biochem J 391: 441-448). Plays a central role in inducing cell differentiation in the kidney during embryogenesis (Yang, J. et al. 2002, Mol Cell 10: 1045-1056) and protects the kidney from ischemic damage (Mishra, J. et. al. 2004, J Am Soc Nephrol 15: 3073-3082; Mori, K. et al. 2005, J Clin Invest 115: 610-621). lcn2 has been suggested as an indicator of active kidney injury (Mori, K. et al. 2007, Kidney Int 71: 967-970). In various forms of gastrointestinal damage, lcn2 facilitates cell regeneration by promoting cell migration (Playford, RJ et al. 2006, Gastroenterology 131: 809-817). However, in vivo studies in mice lacking lcn2 refute the role of lcn2 in protecting kidneys (Berger, T. et al. 2006, Proc Natl Acad Sci USA 103: 1834-1839). In addition, mice lacking lcn2 did not have iron sequestration, which increased the susceptibility to bacterial infection. This suggests an important role of lcn2 in protection against viral infections (Flo, TH et al. 2004, Nature 432: 917-921). Previous studies have shown that lcn2 prevents degradation of NACP (Yan, L. et al. 2001, J Biol Chem 276: 37258-37265), and acute phase protein (Liu, Q. et al. 1995, J Biol Chem 270: 22565 -22570) and has been suggested to act as adipokine associated with insulin resistance (Yan, QW et al. 2007, Diabetes 56: 2533-2540; Zhang, J. et al. 2008, Mol Endocrino l 22: 1416-1426). Recently two cellular receptors for lcn2 have been identified. Megalin, a member of the low density lipoprotein receptor family, has been shown to bind to human lcn2 and mediate its influx into cells (Hvidberg, V. et al. 2005, FEBS Lett 579: 773-777). Another cell surface receptor for mouse lcn2, which has been shown to selectively mediate apoptosis (Devireddy, LR et al. 2001, Science 293: 829-834). Despite receptor identification, the exact role of lcn2 in cell survival and death has not been identified.

본 발명자들은 LCN2가 신경세포의 재생을 저해하는 성상세포증에 주도적으로 관여하는 것을 확인하고, 그에 따라 LCN2를 억제함으로써 뇌 손상 후 신경세포의 재생을 촉진할 수 있다는 것을 확인하고 본 발명을 완성하기에 이르렀다.The present inventors have confirmed that LCN2 is proactively involved in astrocytosis, which inhibits the regeneration of neurons, and thus, by inhibiting LCN2, it is possible to promote the regeneration of neurons after brain injury and to complete the present invention. Reached.

본 발명은 한 관점으로서, LCN2 억제제를 함유한 뇌 손상 치료용 조성물을 제공한다.In one aspect, the present invention provides a composition for treating brain injury containing an LCN2 inhibitor.

다른 관점으로서, LCN2 억제제를 함유한 뇌 손상 치료용 조성물을 투여하여 뇌 손상을 치료하는 방법에 관한 것이다.In another aspect, the present invention relates to a method of treating brain injury by administering a composition for treating brain injury containing an LCN2 inhibitor.

뇌에서 가장 풍부한 아교세포(glial cell)의 한 유형인 성상세포(astrocyte)는 신경세포에 대사적 및 영양적 지지를 제공하고 시냅스의 활성을 조정한다. 뇌 손상에 반응하여 성상세포는 증폭하고 중간미세섬유 단백질(intermediate filament protein)의 발현을 증가시키며 비대(hypertrophic)해진다. 이러한 과정을 (반응성) 성상세포증(astrocytosis)이라고 한다.Astrocytes, one of the brain's most abundant glial cells, provide neuronal metabolic and nutritional support and modulate synaptic activity. In response to brain damage, astrocytes amplify, increase the expression of intermediate filament proteins, and become hypertrophic. This process is called (reactive) astrocytosis.

리포칼린 2(lipocalin 2, LCN2)는 작은 소수성 분자에 결합하는 리포칼린 패밀리의 일원으로서, 다양한 생리적 과정에 관여하고, 염증 및 신생물 질환에서 높게 유도될 수 있다. 본 발명에 따르면 LCN2가 반응성 성상세포증의 자가조절 매개자(autocrine mediator)로 여겨진다. 이러한 결론은 본 발명에서 다음과 같이 LCN2가 성상세포의 세포 사멸, 형태 및 이동(migration)의 조절에 관여함을 확인한 결과를 기반으로 한 것이다.Lipocalin 2 (LCN2) is a member of the lipocalin family that binds to small hydrophobic molecules and is involved in various physiological processes and can be highly induced in inflammatory and neoplastic diseases. According to the present invention LCN2 is considered to be an autocrine mediator of reactive astrocytosis. This conclusion is based on the results confirming that LCN2 is involved in the regulation of apoptosis, morphology and migration of astrocytes as follows.

LCN2는 세포독성 물질에 대한 성상세포의 세포 사멸 민감도를 증가시킨다. 즉, 세포독성 물질에 의한 성상세포의 세포 사멸을 가속화한다. LCN2는 성상세포를 H2O2 또는 파라콰트(paraquat)에 의해 유도되는 네크로시스 세포 사멸 뿐만 아니라 NO에 의해 유도되는 아폽토시스 세포 사멸을 가속화한다.LCN2 increases the apoptosis sensitivity of astrocytes to cytotoxic substances. That is, it accelerates the cell death of astrocytes by cytotoxic substances. LCN2 accelerates astrocytic cell death induced by NO, as well as necrosis cell death induced by H 2 O 2 or paraquat.

한편, 세포로 철의 유입은 전아폽토시스(proapoptosis) 단백질인 Bim 단백질의 발현양을 감소시킨다. 반대로 세포내에 철이 고갈되면 Bim의 상향조절(up-regulation)로 이어진다. 본 발명의 실시예를 통해 lcn2에 의해 유도된 성상세포의 세포 사멸로의 민감화가 사이더로포어(siderophore)-철 복합체의 첨가에 의해(즉, 철의 공급에 의해) 상쇄되는 것을 확인하였다. 이는 lcn2에 의해 유도되는 성상세포의 세포 사멸로의 민감화가 철의 운반 및 대사에 관여하는 Bim 단백질과 관련이 있다는 것을 의미한다.On the other hand, the influx of iron into the cell reduces the amount of expression of Bim protein, a proapoptosis protein. Conversely, iron depletion in cells leads to up-regulation of Bim. Examples of the present invention confirmed that the sensitization of astrocytic cells induced by lcn2 to apoptosis was counteracted by addition of a siderophore-iron complex (ie, by the supply of iron). This means that the sensitization of astrocytes to apoptosis induced by lcn2 is associated with the Bim protein involved in iron transport and metabolism.

또한, lcn2의 발현은 리포폴리사카라이드(LPS) 및 TNF-α에 의해 강하게 유도되고, 혈청 제거(Serum Withdrawal), PMA, INF-γ 및 강글리오사이드의 혼합물에 의해 약하게 유도된다. 이는 성상세포에서 lcn2의 발현 및 분비는 CNS에서 염증 조건하에서 증가한다는 것을 의미한다.In addition, the expression of lcn2 is strongly induced by lipopolysaccharide (LPS) and TNF-α and weakly by a mixture of serum withdrawal, PMA, INF-γ and gangliosides. This means that the expression and secretion of lcn2 in astrocytes increases under inflammatory conditions in the CNS.

또한, LCN2가 성상세포에 노출되게 되면 성상세포의 세포 돌기의 개수를 증가시킨다. 이와 같이 LCN2에 의해 유도되는 성상세포의 형태학적 변화는 용량 및 시간에 의존적이다. LCN2에 의해 유도된 세포 돌기에서의 변화는 GFAP(glial fibrillary acidic protein) 발현의 상향조절에 의해 이루어지고, 이 또한 LCN2의 용량 및 시간에 의존적이다. In addition, exposure of LCN2 to astrocytes increases the number of cell processes of astrocytes. As such, the morphological changes of astrocytes induced by LCN2 are dose and time dependent. Changes in the cell processes induced by LCN2 are made by upregulation of GFAP (glial fibrillary acidic protein) expression, which is also dependent on the dose and time of LCN2.

한편, 본 발명의 실시예를 통해 LCN2에 의해 유도되는 것과 유사한 성상세포의 형태학적 변화를 유도하는 Forskolin 및 dbcGMP도 성상세포의 세포 사멸로의 민감성을 증가시키는 것이 확인되었다. 이는 성상세포의 형태학적 변화와 세포 사멸로의 민감성이 서로 밀접한 연관성이 있다는 것을 의미한다.On the other hand, it was confirmed through the embodiments of the present invention that Forskolin and dbcGMP, which induce morphological changes of astrocytes similar to those induced by LCN2, also increase the sensitivity to apoptosis. This means that morphological changes of astrocytes and susceptibility to cell death are closely related to each other.

또한, NO가 성상세포에서 GFAP의 발현을 증폭시킨다(Brahmachari, S. et al. 2006. IJ Neurosci 26: 4930-4939). 본 발명에서 LCN2가 또한 GFAP의 발현을 유도할 수 있다는 것을 확인하였기 때문에 NO가 성상세포에서 LCN2 작용에 관여하는지 조사하였다. LCN2는 성상세포에서 NO 생산양의 증가를 유도하고, 이는 LPS의 그것과 유사하다. NOS 억제제인 NMMA에 의해 LCN2에 의해 유도된 GFAP의 발현은 차단되고, LCN2에 의해 유도된 세포 사멸로의 민감성은 약화된다. 이는 NO가 LCN2에 의해 매개되는 성상세포의 세포 사멸, GFAP 발현 및 형태 변화에서 중요한 역할을 한다는 것을 의미한다.In addition, NO amplifies the expression of GFAP in astrocytes (Brahmachari, S. et al. 2006. I J Neurosci 26: 4930-4939). Since the present invention confirmed that LCN2 can also induce the expression of GFAP, it was investigated whether NO is involved in LCN2 action in astrocytes. LCN2 induces an increase in NO production in astrocytes, similar to that of LPS. Expression of GFAP induced by LCN2 by the NOS inhibitor NMMA is blocked, and the sensitivity to cell death induced by LCN2 is attenuated. This means that NO plays an important role in cell death, GFAP expression and morphological changes of astrocytes mediated by LCN2.

한편, 스몰 G 단백질의 Rho 서브패밀리가 성상세포 형태의 조절에 관여하므로(Boran, M. S. et al. 2007. J Neurochem 102: 216-230; Chen, C. J. et al. 2006. stellation. Eur J Neurosci 23: 1977-1987; Suidan, H. S. et al. 1997. Glia 21: 244-252; John, G. R. et al. 2004. J Neurosci 24: 2837-2845; Ramakers, G. J. et al. 1998. Exp Cell Res 245: 252-262; Holtje, M. et al. 2005. J Neurochem 95: 1237-1248; Hall, A. 2005. Biochem Soc Trans 33: 891-895) LCN2에 의해 유도되는 성상세포의 형태학적 변화에서 Rho 단백질의 관련성을 조사하였다. LCN2는 Rho의 활성화를 유도하는 한편, ROCK 억제제는 LCN2에 의해 유도된 형태학적 변화를 블로킹하고, GFAP의 발현을 억제한다. 이는 성상세포의 형태 조절에 Rho/ROCK 경로가 관련되어 있다는 것을 의미한다.On the other hand, the Rho subfamily of small G proteins is involved in the regulation of astrocyte morphology (Boran, MS et al. 2007. J Neurochem 102: 216-230; Chen, CJ et al. 2006. stellation. Eur J Neurosci 23: 1977-1987; Suidan, HS et al. 1997. Glia 21: 244-252; John, GR et al. 2004. J Neurosci 24: 2837-2845; Ramakers, GJ et al. 1998. Exp Cell Res 245: 252- 262; Holtje, M. et al. 2005. J Neurochem 95: 1237-1248; Hall, A. 2005. Biochem Soc Trans 33: 891-895) Rho protein's relevance in morphological changes of astrocytes induced by LCN2 Was investigated. LCN2 induces activation of Rho, while ROCK inhibitors block morphological changes induced by LCN2 and inhibit the expression of GFAP. This means that the Rho / ROCK pathway is involved in morphological regulation of astrocytes.

또한, 세포의 이동은 세포의 형태와 밀접하게 연관된 세포의 표현형이기 때문에 성상세포의 이동에 대한 LCN2의 영향을 조사하였다. in vitro 상처 치료 에세이 및 Boyden 챔버 에세이를 통하여 LCN2가 성상세포의 이동성을 증가시킨다는 것을 확인하였다.In addition, the effect of LCN2 on astrocyte migration was investigated because cell migration is a phenotype of cells that is closely related to cell morphology. In vitro wound treatment assays and Boyden chamber assays confirmed that LCN2 increased stellate cell mobility.

이로써, 성상세포의 분비 단백질인 LCN2는 활성화된 성상세포의 형태 및 기능에 대한 경로를 결정하는 두 가지 기능을 수행한다는 것을 알 수 있다(도 36).As a result, it can be seen that LCN2, a secretory protein of astrocytes, performs two functions that determine the pathway for the shape and function of activated astrocytes (FIG. 36).

결론적으로, LCN2는 성상세포의 성상세포증 과정을 주도하는 단백질이므로 이 단백질을 조절함으로써 성상세포증을 제어할 수 있다. 성상세포증은 CNS 신경세포의 재생을 저해하는 주요 원인이다. 따라서, lcn2 유전자의 발현을 억제하거나, LCN2 단백질의 활성을 억제함으로써 성상세포증을 억제할 수 있으며, 성상세포증이 억제됨에 따라 CNS 신경세포의 재생을 촉진할 수 있다.In conclusion, LCN2 is a protein that leads to the process of astrocytes in astrocytes, so it can be controlled by controlling this protein. Astrocytes are a major cause of inhibition of regeneration of CNS neurons. Therefore, it is possible to suppress astrocytosis by inhibiting the expression of the lcn2 gene or inhibiting the activity of the LCN2 protein, and promote regeneration of CNS neurons as astrocytosis is suppressed.

신경세포 재생이 필요한 경우 즉, 뇌 손상 치료가 요구되는 경우는 모든 종류의 뇌 손상 시이므로 특별히 제한적이지 않지만, 예를 들면 알츠하이머 질환, 파킨슨 질환, 루게릭 질환, 헌팅톤 질환 및 다발성 경화증을 포함한 퇴행성 신경질환, 뇌졸중, 외상, 뇌 감염 질환, 크로이츠펠트 야곱병, 뇌 염증 질환 등이 있다.If neural cell regeneration is required, ie brain injury treatment is required for all types of brain injury, it is not particularly limited, but degenerative nerves, including, for example, Alzheimer's disease, Parkinson's disease, Lou Gehrig's disease, Huntington's disease and multiple sclerosis Diseases, stroke, trauma, brain infections, Creutzfeldt-Jakob disease, brain inflammatory diseases, and the like.

따라서, 본 발명은 LCN2의 억제제를 함유한 뇌 손상 치료용 조성물을 제공한다. 여기서, LCN2의 억제제는 LCN2의 활성 억제제 또는 LCN2의 발현 억제제일 수 있다.Accordingly, the present invention provides a composition for treating brain injury containing an inhibitor of LCN2. Herein, the inhibitor of LCN2 may be an activity inhibitor of LCN2 or an expression inhibitor of LCN2.

본 발명에서 LCN2의 활성 억제제로는 예를 들면, 이에 한정되지는 않으나 펩타이드, 폴리펩타이드, 단백질, 펩타이드 모조물, 화합물 및 생물제제일 수 있다. 바람직하게는 LCN2의 활성을 중화시킬 수 있는 항-LCN2 항체일 수 있다.In the present invention, the activity inhibitor of LCN2 may be, for example, but not limited to, peptides, polypeptides, proteins, peptide replicas, compounds, and biologics. Preferably it may be an anti-LCN2 antibody capable of neutralizing the activity of LCN2.

본 발명에서 항-LCN2 항체는 다클론 항체 또는 단클론 항체일 수 있다. 본 발명의 항체는 LCN2 단백질을 항원으로 하여 면역학 분야에서 널리 알려져 있는 통상의 방법으로 제조할 수 있다.In the present invention, the anti-LCN2 antibody may be a polyclonal antibody or a monoclonal antibody. Antibodies of the invention can be prepared by conventional methods well known in the art of immunology using LCN2 protein as an antigen.

다클론 항체는 말, 소, 염소, 양, 개, 닭, 칠면조, 토끼, 마우스 또는 랫트와 같은 여러 온혈 동물로부터 당해 분야의 통상적인 기술 중의 하나를 사용할 수 있다. 즉, 항원을 복막 내, 근육 내, 안내 또는 피하 주사를 통해 동물을 면역시킨다. 상기 항원에 대한 면역성은 보조제, 예를 들어 프로인트(Freund)의 완전 보조제 또는 불완전 보조제를 사용하여 증가시킬 수 있다. 부스터(booster) 면역처리에 따른 다음, 혈청의 소형 샘플을 수집하고 목적하는 항원에 대한 반응성을 시험한다. 동물의 역가가 일단 항원에 대한 이의 반응성의 관점으로 정체 상태에 도달하면, 다량의 다클론 면역혈청을 1주 마다의 출혈 또는 동물을 방혈시킴으로써 수득할 수 있다.Polyclonal antibodies can be used in one of ordinary skill in the art from several warm-blooded animals such as horses, cows, goats, sheep, dogs, chickens, turkeys, rabbits, mice or rats. That is, the antigen is immunized to the animal via intraperitoneal, intramuscular, intraocular or subcutaneous injection. Immunity to the antigen can be increased using an adjuvant, for example Freund's complete adjuvant or incomplete adjuvant. Following booster immunization, a small sample of serum is collected and tested for reactivity to the desired antigen. Once the titer of the animal reaches a steady state in terms of its reactivity to the antigen, large amounts of polyclonal immune serum can be obtained by bleeding weekly or by bleeding the animal.

단클론 항체도 공지된 기술을 사용하여 생성시킬 수 있다(Kennettm McKearn and Bechtol(eds.), Monoclonal Antibodies, Hybridomas; A New Dimension in Biological Analyses, Plenum Press, 1980). 단클론 항체는 LCN2 단백질을 면역원으로 하여 동물을 면역화시키고, 면역화된 동물의 비장세포를 골수종 세포와 융합하여 하이브리도마를 생성하고, LCN2 단백질을 선택적으로 인식하는 하이브리도마를 선별하며, 선별한 하이브리도마를 배양하고, 하이브리도마의 배양액으로부터 항체를 분리함으로써 제조할 수 있다. 또한, 본 발명의 단클론 항체는 LCN2 단백질을 선택적으로 인식하는 항-LCN2 항체를 생산하는 상기의 하이브리도마를 동물에 주입하고, 주입 후 일정기간이 지난 다음 회수한 동물의 복수로부터 분리함으로써 제조할 수 있다.Monoclonal antibodies can also be generated using known techniques (Kennettm McKearn and Bechtol (eds.), Monoclonal Antibodies, Hybridomas; A New Dimension in Biological Analyses , Plenum Press, 1980). Monoclonal antibodies immunize animals with LCN2 protein as an immunogen, fusion of splenocytes of the immunized animal with myeloma cells to produce hybridomas, select hybridomas that selectively recognize LCN2 protein, and select high It can be prepared by culturing the bridoma and separating the antibody from the culture medium of the hybridoma. In addition, the monoclonal antibody of the present invention can be prepared by injecting the above-mentioned hybridomas producing an anti-LCN2 antibody selectively recognizing LCN2 protein into an animal and separating it from the ascites of the recovered animal after a certain period of time after the injection. Can be.

본 발명에서 "유전자의 발현 억제"에는 유전자 전사의 억제 및 단백질로의 번역 억제가 포함된다. 또한, 유전자 발현이 완전히 정지된 것 뿐 만 아니라 발현이 감소된 것도 포함된다.In the present invention, "inhibition of gene expression" includes inhibition of gene transcription and translation into protein. In addition, not only the gene expression is completely stopped, but also the expression is reduced.

유전자의 발현을 억제하는 방법으로는 안티센스 분자를 이용하는 것이 가장 보편적이다. 안티센스 분자가 표적 유전자의 발현을 억제하는 작용으로는 삼중쇄 형성에 의한 전사개시 저해, RNA 폴리머라제에 의해 국부적인 개상 루프 구조가 만들어진 부위에서 하이브리드 형성에 의한 전사 억제, 합성이 진행되고 있는 RNA에서 하이브리드 형성에 의한 전사 저해, 인트론과 엑손과의 접합점에서 하이브리드 형성에 의한 스플라이싱 억제, 스플라이소좀 형성 부위에서 하이브리드 형성에 의한 스플라이싱 억제, mRNA와의 하이브리드 형성에 의한 핵으로부터 세포질로의 이행 억제, 번역 개시인자 결합 부위에서 하이브리드 형성에 의한 번역개시 억제 등이 있다. 이들은 전사, 스플라이싱 또는 번역 과정을 저해하여 표적 유전자의 발현을 억제한다.Antisense molecules are most commonly used as a method of inhibiting gene expression. Antisense molecules inhibit the expression of target genes by inhibiting transcriptional initiation by triple chain formation, transcriptional inhibition by hybridization at sites where local open loop structure is formed by RNA polymerase, and in RNA where synthesis is in progress. Inhibition of transcription by hybrid formation, inhibition of splicing by hybridization at the junction of introns and exons, inhibition of splicing by hybridization at the site of splicosomal formation, transition from nucleus to cytoplasm by hybridization with mRNA Inhibition of translation initiation by hybridization at the site of translation initiation factor binding. They inhibit the expression of target genes by inhibiting transcription, splicing or translation processes.

본 발명에 사용되는 안티센스 분자는 상기의 어느 작용으로든지 표적 유전자의 발현을 억제해도 좋다. 대표적인 안티센스 분자로는 3중제, 리보자임(ribozyme), RNAi, 또는 안티센스 핵산 등이 포함된다. 3중제는 이중 나선 DNA 주변을 감아 3 본쇄 나선을 형성하도록 함으로써 전사 개시가 억제되도록 한다(Maher et al., Antisense Res. and Dev., 1(3):227, 1991; Helene, C., Anticancer Drug Design, 6(6):569, 1991).The antisense molecule used in the present invention may inhibit the expression of the target gene by any of the above actions. Representative antisense molecules include triplets, ribozymes, RNAi, or antisense nucleic acids. The triple agent allows the initiation of transcription to be suppressed by winding around double helix DNA to form a three-stranded helix (Maher et al., Antisense Res. And Dev ., 1 (3): 227, 1991; Helene, C., Anticancer Drug Design , 6 (6): 569, 1991).

리보자임은 1 본쇄 RNA를 특이적으로 절단하는 능력을 보유한 RNA 효소이다. 리보자임은 표적 RNA 분자 내 특정 뉴클레오티드 서열을 인식하여 부위 특이적으로 절단시킴으로써 표적 유전자의 단백질 발현을 억제한다(Cech, J. Amer. Med. Assn., 260:3030, 1998; Sarver et al., Science 247:1222-1225, 1990). Ribozymes are RNA enzymes that possess the ability to specifically cleave single-stranded RNA. Ribozymes inhibit protein expression of target genes by recognizing and site-specific cleavage of specific nucleotide sequences in target RNA molecules (Cech, J. Amer. Med. Assn ., 260: 3030, 1998; Sarver et al., Science 247: 1222-1225, 1990).

RNAi(RNA interference)는 염기서열 특이적으로 작용하는 헤어핀 형태의 소분자의 RNA를 사용하여 전사 수준 혹은 전사 후 수준에서 유전자 발현을 억제시키는 방법이다(Mette et al., EMBO J., 19: 5194-5201, 2000). 상기 RNAi 방법에 사용되는 소분자 RNA는 표적 유전자와 상동성을 가지는 이중가닥 RNA 분자이다. RNAi (RNA interference) is a method of inhibiting gene expression at the transcriptional level or at the post-transcriptional level by using RNA of hairpin type small molecule that acts in sequence (Mette et al., EMBO J. , 19: 5194-). 5201, 2000). The small molecule RNA used in the RNAi method is a double-stranded RNA molecule having homology with the target gene.

상기에서 RNA 분자를 제조하는 방법으로는 공지된 화학적 합성 방법 및 효소적 방법을 사용할 수 있다. 예를 들면, RNA 분자의 화학적 합성은 문헌에 개시되어 있는 방법을 사용할 수 있으며(Verma and Eckstein, Annu. Rev. Biochem. 67, 99-134, 1999), RNA 분자의 효소적 합성은 T7, T3 및 SP6 RNA 폴리머라제와 같은 파아지 RNA 폴리머라제를 이용하는 방법이 문헌에 개시되어 있다(Milligan and Uhlenbeck, Methods Enzymol. 180:51-62, 1989).As a method of preparing the RNA molecule in the above, known chemical synthesis methods and enzymatic methods can be used. For example, the chemical synthesis of RNA molecules can use the methods described in the literature (Verma and Eckstein, Annu. Rev. Biochem. 67, 99-134, 1999), and the enzymatic synthesis of RNA molecules is T7, T3. And phage RNA polymerases such as SP6 RNA polymerase are disclosed in the literature (Milligan and Uhlenbeck, Methods Enzymol . 180: 51-62, 1989).

안티센스 핵산은 표적 mRNA 분자와 적어도 일부분이 상보적인 DNA 또는 RNA 분자를 말한다(Weintraub, Scientific American, 262:40, 1990). 세포 내에서, 안티센스 핵산은 이에 상응하는 mRNA와 혼성화되어 2본쇄 분자를 형성함으로써 표적 유전자의 mRNA 해독을 저해하여 단백질 발현을 억제한다(Marcus-Sakura, Anal. Biochem., 172:289, 1988). 상기 안티센스 핵산은 바람직하게는 올리고뉴클레오타이드 형태로서 당업계에 공지된 임의의 적합한 방법에 의해 제조할 수 있다. 상기 안티센스 올리고뉴클레오타이드는 화학 합성법, 예를 들어 문헌(Tetrahedron Lett., 1991, 32, 30005-30008)에 기재된 바와 같이 아세토니트릴 중에서 테트라에틸티우람 디술파이드로 황화시키는 포스포아미다이트 화학과 같은 방법에 의해 매우 용이하게 제조할 수 있다.Antisense nucleic acids refer to DNA or RNA molecules that are at least partially complementary to a target mRNA molecule (Weintraub, Scientific American , 262: 40, 1990). In cells, antisense nucleic acids hybridize with their corresponding mRNAs to form double-stranded molecules that inhibit protein translation by inhibiting mRNA translation of target genes (Marcus-Sakura, Anal. Biochem ., 172: 289, 1988). The antisense nucleic acid may be prepared by any suitable method known in the art, preferably in the form of oligonucleotides. The antisense oligonucleotides may be used in chemical synthesis, for example, such as phosphoramidite chemistry, which is sulfided with tetraethylthiuram disulfide in acetonitrile as described in Tetrahedron Lett ., 1991, 32, 30005-30008. It can manufacture very easily.

본 발명에서는 LCN2 억제제를 병변, 즉 뇌 손상 치료가 필요한 부분으로 유도할 수 있다면 어떠한 경로를 통해서든지 투여 가능하다. 따라서, 본 발명의 조성물은 국소 (협측(buccal), 설하, 피부 및 안내의 투여를 포함), 비경구(피하, 피내 근육내, 혈관내 및 관절내의 투여를 포함) 또는 경피성 투여를 포함한 여러 경로를 통해 투여할 수도 있으며, 바람직하게는 비경구로, 가장 바람직하게는 베타 아밀로이드가 침착된 부위에 직접 투여한다.In the present invention, the LCN2 inhibitor may be administered by any route as long as the LCN2 inhibitor can be directed to the lesion, i. Accordingly, the compositions of the present invention may be used in various forms including topical (including buccal, sublingual, skin and intraocular), parenteral (including subcutaneous, intradermal, intravascular and intraarticular) or transdermal administration. Routes may also be administered, preferably parenterally, most preferably directly at the site where the beta amyloid has been deposited.

한 양태로서, LCN2 억제제는 적합한 희석제에 현탁시켜 개체에 투여할 수 있는데, 이 희석제는 세포를 보호 및 유지하고, 목적하는 뇌 조직에 주입시 사용에 용이하도록 하는 용도로서 사용된다. 상기 희석제는 생리식염수, PBS, HBSS 등의 완충용액, 혈장, 뇌척수액 또는 혈액성분 등이 있을 수 있다.In one embodiment, the LCN2 inhibitor can be administered to a subject by suspending in a suitable diluent, which diluent is used for the purpose of protecting and maintaining the cells and facilitating use when infused into the desired brain tissue. The diluent may include physiological saline, PBS, HBSS buffer solution, plasma, cerebrospinal fluid or blood components.

다른 양태로서, LCN2 억제제는 통상적인 방법에 따라 약제학적으로 허용되는 담체와 혼합하여 사용할 수 있다. 적합한 담체의 예로는, 락토오스, 덱스트로오스, 수크로오스, 솔비톨, 만니톨, 자일리톨, 말티톨, 전분, 아카시아 고무, 알지네이트, 젤라틴, 칼슘 포스페이트, 칼슘 실리케이트, 셀룰로오스, 메틸 셀루로오스, 미정질 셀루로오스, 폴리비닐피롤리돈, 물, 메틸하이드록시벤조에이트, 프로필하이드록시벤조에이트, 탈크, 마그네슘 스테아레이트 및 광물유를 들 수 있다. 필요에 따라 상기 조성물에 충진제, 항응집제, 윤활제, 습윤제, 향료, 유화제, 방부제 등을 추가로 포함할 수 있다.In another embodiment, the LCN2 inhibitor may be used in admixture with a pharmaceutically acceptable carrier according to conventional methods. Examples of suitable carriers include lactose, dextrose, sucrose, sorbitol, mannitol, xylitol, maltitol, starch, acacia rubber, alginate, gelatin, calcium phosphate, calcium silicate, cellulose, methyl cellulose, microcrystalline cellulose, Polyvinylpyrrolidone, water, methylhydroxybenzoate, propylhydroxybenzoate, talc, magnesium stearate and mineral oil. If necessary, the composition may further include a filler, an anticoagulant, a lubricant, a humectant, a perfume, an emulsifier, a preservative, and the like.

본 발명의 조성물은 개체에 투여된 후 유효 성분의 신속 또는 지연된 방출을 제공할 수 있도록 당업계에 잘 알려진 방법을 사용하여 제형화될 수 있다. 제형은 정제, 분말, 환제, 에멀전, 용액, 시럽, 에어로졸, 연질 또는 경질 젤라틴 캅셀, 멸균 주사용액, 멸균 분말 등의 형태일 수 있다. 상기 제형은 1회용 투약 형태가 편리할 것이다.Compositions of the invention can be formulated using methods well known in the art to provide rapid or delayed release of the active ingredient after administration to a subject. The formulations may be in the form of tablets, powders, pills, emulsions, solutions, syrups, aerosols, soft or hard gelatin capsules, sterile injectable solutions, sterile powders and the like. The formulation will be convenient in a single dosage form.

본 발명의 조성물은 약제학적으로 유효한 양으로 투여한다. 본 발명에서 “약제학적으로 유효한 양”은 질환을 치료하기에 충분한 양을 의미하며, 유효 용량 수준은 질환의 중증도; 환자의 연령, 체중, 건강, 성별; 환자의 약물에 대한 민감도; 투여 시간, 투여 경로 및 배출 비율; 치료 기간; 사용된 본 발명의 조성물과 배합 또는 동시 사용되는 약물을 포함한 요소 및 기타 의학 분야에 알려진 요소에 따라 결정될 수 있다. 바람직하게는, 본 발명의 약학적 조성물은 질환의 정도에 따라 유효량을 달리할 수 있으나, 바람직하게는 1~10000㎍/체중kg/day, 더욱 바람직하게는 10~1000㎎/체중kg/day의 유효량으로 하루에 수회 반복 투여될 수 있다. The composition of the present invention is administered in a pharmaceutically effective amount. In the present invention, “pharmaceutically effective amount” means an amount sufficient to treat a disease, and an effective dose level may include the severity of the disease; The age, body weight, health and sex of the patient; Sensitivity to the drug of the patient; Time of administration, route of administration, and rate of excretion; Duration of treatment; It may be determined according to factors including drugs used in combination or coincidental with the composition of the present invention and other factors known in the medical field. Preferably, the pharmaceutical composition of the present invention may vary the effective amount depending on the extent of the disease, preferably 1 to 10,000 g / weight kg / day, more preferably 10 to 1000 mg / kg / day The effective amount may be repeated several times a day.

LCN2 억제제에 의해 신경세포의 재생을 저해하는 성상세포증을 억제할 수 있으므로 신경세포의 재생을 촉진시킬 수 있다.Because LCN2 inhibitors can inhibit astrocytosis, which inhibits the regeneration of neurons, they can promote neuronal regeneration.

이로써, LCN2 억제제는 신경세포의 재생이 요구되는 다양한 질병, 즉, 뇌 손상과 관련된 임의의 질병을 치료하는데 이용될 수 있다.As such, LCN2 inhibitors can be used to treat a variety of diseases that require neuronal regeneration, ie, any disease associated with brain damage.

특히, LCN2는 성상세포증의 과정에서 상위 단계에서 주도적으로 관여하므로 LCN2 억제제를 이용하면 효과적으로 성상세포증을 억제하여 신경세포의 재생을 촉진시킬 수 있다. In particular, LCN2 is involved in the upper stages in the process of astrocytosis, so LCN2 inhibitors can effectively inhibit astrocytosis and promote neuronal regeneration.

도 1는 센스 또는 안티센스 lcn2 cDNA로 형질감염된 C6 세포에서 단백질 발현과 lcn2 mRNA에서의 변화를 분석한 결과를 나타낸 것이다(S3: 센스 lcn2 형질감염체; AS7: 안티센스 lcn2 형질감염체). Figure 1 shows the results of analysis of protein expression and changes in lcn2 mRNA in C6 cells transfected with sense or antisense lcn2 cDNA (S3: sense lcn2 transfectants; AS7: antisense lcn2 transfectants).

도 2는 센스 또는 안티센스 lcn2 cDNA로 형질감염된 C6세포에서 SNP, H2O2 및 파라콰트에 의한 세포 생존율에서의 변화를 나타낸 것이다(S3: 센스 lcn2 형질감염체; AS7: 안티센스 lcn2 형질감염체).Figure 2 shows the change in cell viability by SNP, H 2 O 2 and paraquat in C6 cells transfected with sense or antisense lcn2 cDNA (S3: sense lcn2 transfectant; AS7: antisense lcn2 transfectant).

도 3는 GFP와 융합된 lcn2 DNA를 포함하는 아데노바이러스 벡터로 형질감염된 C6 세포에서 lcn2 발현 여부를 분석한 결과를 나타낸 것이다.Figure 3 shows the results of the analysis of lcn2 expression in C6 cells transfected with adenovirus vectors containing lcn2 DNA fused with GFP.

도 4는 GFP와 융합된 lcn2 cDNA를 포함하는 아데노바이러스 벡터로 형질감염된 C6 세포에서 NO 공여자인 SNP, H2O2 및 파라콰트에 의한 세포 생존율에서의 변화를 나타낸 것이다.Figure 4 shows the change in cell viability by NO donors SNP, H 2 O 2 and paraquat in C6 cells transfected with adenovirus vectors comprising lcn2 cDNA fused with GFP.

도 5는 재조합 마우스 LCN2 단백질을 나타낸 것이다. 5 shows recombinant mouse LCN2 protein.

도 6는 성상세포에 LCN2를 단독 처리한 경우 및 SNP, H2O2 또는 파라콰트와 함께 처리한 경우에 세포 생존율에서의 변화를 나타낸 것이다.FIG. 6 shows changes in cell viability when stellate cells were treated with LCN2 alone and when treated with SNP, H 2 O 2 or paraquat.

도 7는 성상세포에 LCN2를 단독 처리한 경우 및 SNP, H2O2 또는 파라콰트와 함께 처리한 경우에 성상세포의 네크로시스 및 아폽토시스 세포 사멸율에서의 변화를 나타낸 것이다.Figure 7 shows the changes in necrosis and apoptosis cell death rate of astrocytes when LCN2 was treated with astrocytes alone and when treated with SNP, H 2 O 2 or paraquat.

도 8는 LCN2 단백질의 용량에 따른 성상세포 사멸율에서의 변화를 나타낸 것이다.8 shows the change in astrocyte death rate according to the dose of LCN2 protein.

도 9는 LCN2 단백질이 C6 세포 및 성상세포의 세포 주기 분포에 영향을 미치는지 여부를 확인한 결과를 나타낸 것이다.Figure 9 shows the results confirming whether the LCN2 protein affects the cell cycle distribution of C6 cells and astrocytes.

도 10는 성상세포 및 C6 세포에 철 킬레이터(DFO)를 단독으로 처리한 경우와 NO 공여자인 SNAP와 함께 처리한 경우에 세포 생존율에서의 변화를 나타낸 것이다.FIG. 10 shows changes in cell viability when astrocytes and C6 cells were treated with iron chelator (DFO) alone and when treated with NO donor SNAP.

도 11는 성상세포 및 C6 세포에 철 공여자(FC)를 단독으로 처리한 경우와 SNAP와 함께 처리한 경우에 세포 생존율에서의 변화를 나타낸 것이다.11 shows changes in cell viability when treated with iron donors (FC) alone and when treated with SNAP in astrocytes and C6 cells.

도 12는 C6 세포에 LCN2와 SNAP를 처리한 경우 및 사이더포어와 철의 복합체를 동시 처리한 경우에 세포 생존율에서의 변화를 나타낸 것이다.FIG. 12 shows the change in cell viability when C6 cells were treated with LCN2 and SNAP and co-treated with the cyder pores and iron complexes.

도 13는 LCN2로 처리된 C6 세포의 배양물에서 Bim RNA 및 단백질 양의 변화를 나타낸 것이다.FIG. 13 shows changes in Bim RNA and protein amounts in culture of C6 cells treated with LCN2.

도 14는 LCN2로 처리된 성상세포의 배양물에서 Bim RNA 및 단백질 양의 변화를 나타낸 것이다.Figure 14 shows the changes in the amount of Bim RNA and protein in the culture of astrocytes treated with LCN2.

도 15는 SNP, LPS, 혈청 제거(SW), PMA, IFN-γ, TNF-α 및 강글리오사이드의 혼합물에 의한 LCN2 발현양에서의 변화를 분석한 결과를 나타낸 것이다.Figure 15 shows the results of analyzing the change in the amount of LCN2 expression by the mixture of SNP, LPS, serum clearance (SW), PMA, IFN-γ, TNF-α and gangliosides.

도 16는 LPS에 의한 LCN2의 분비양의 변화를 분석한 결과를 나타낸 것이다.Figure 16 shows the results of analyzing the change in the secretion amount of LCN2 by LPS.

도 17는 C6 세포 및 성상세포에서 LCN2의 수용체인 lcn2R/24p3R의 발현양의 변화를 분석한 결과를 나타낸 것이다.Figure 17 shows the results of analyzing the change in the expression amount of LCN2 receptor lcn2R / 24p3R in C6 cells and astrocytes.

도 18은 성상세포가 LCN2, Forskolin 또는 dbcGMP에 노출된 경우에 형태학적 변화, 돌기의 길이 및 세포 생존율에서의 변화를 분석한 결과를 나타낸 것이다.Figure 18 shows the results of analyzing the changes in morphological changes, protuberance length and cell viability when astrocytes were exposed to LCN2, Forskolin or dbcGMP.

도 19은 LCN2의 용량 및 LCN2에 노출된 시간에 따른 성상세포의 형태학적 변화를 나타낸 것이다.Figure 19 shows the morphological changes of astrocytes with the dose of LCN2 and the time of exposure to LCN2.

도 20은 LCN2의 용량 및 LCN2에 노출된 시간에 따른 성상세포의 GFAP mRNA 및 단백질 발현양에서의 변화를 분석한 결과를 나타낸 것이다.FIG. 20 shows the results of analyzing changes in GFAP mRNA and protein expression levels of astrocytes with the dose of LCN2 and time exposed to LCN2.

도 21는 성상세포에 SNP와 LCN2, Forskolin 또는 dbcGMP를 동시 처리한 경우에 세포 생존율에서의 변화를 나타낸 것이다.Figure 21 shows the change in cell viability when co-treatment with SNP and LCN2, Forskolin or dbcGMP in astrocytes.

도 22는 성상세포에 Forskolin 또는 dbcGMP를 처리한 경우에 LCN2 단백질 또는 GFAP 발현양에서의 변화를 분석한 결과를 나타낸 것이다.Figure 22 shows the results of analyzing the changes in the LCN2 protein or GFAP expression when treated with Forskolin or dbcGMP in astrocytes.

도 23는 SNP에 의한 GFAP mRNA 및 단백질 수준에서의 변화를 분석한 결과를 나타낸 것이다.Figure 23 shows the results of analyzing the changes in GFAP mRNA and protein levels by SNP.

도 24는 LCN2가 성상세포의 NO 생산에 미치는 영향을 나타낸 것이다.Figure 24 shows the effect of LCN2 on the NO production of astrocytes.

도 25는 polymyxin B(PB)가 LCN2에 의해 유도된 성상세포의 NO 생산에 미치는 영향을 나타낸 것이다.Figure 25 shows the effect of polymyxin B (PB) on the NO production of astrocytes induced by LCN2.

도 26는 NOS 억제제인 NMMA가 LCN2에 의해 유도된 성상세포에서의 GFAP 발현에 미치는 영향을 나타낸 것이다.Figure 26 shows the effect of the NOS inhibitor NMMA on GFAP expression in astrocytes induced by LCN2.

도 27는 NMMA가 LCN2에 의해 유도된 성상세포의 세포 사멸에 미치는 영향을 나타낸 것이다.27 shows the effect of NMMA on cell death of astrocytic cells induced by LCN2.

도 28는 ROCK 억제제인 Y27632가 LCN2에 의해 유도되는 성상세포의 형태학적 변화에 미치는 영향을 나타낸 것이다.28 shows the effect of ROCK inhibitor Y27632 on the morphological changes of astrocytes induced by LCN2.

도 29는 Y27632는 LCN2에 의해 유도되는 GFAP의 발현에 미치는 영향을 나타낸 것이다.29 shows the effect of Y27632 on the expression of GFAP induced by LCN2.

도 30는 LCN2가 Rho의 활성화에 미치는 영향을 나타낸 것이다.30 shows the effect of LCN2 on the activation of Rho.

도 31는 in vitro 상처 치료 에세이를 이용하여 LCN2, Forskolin 또는 dbcGMP가 성상세포의 이동에 미치는 영향을 확인한 결과이다.Figure 31 shows the results confirmed the effect of LCN2, Forskolin or dbcGMP on the migration of astrocytes using an in vitro wound treatment assay.

도 32는 Boyden 챔버 에세이를 이용하여 LCN2, Forskolin 또는 dbcGMP가 성상세포의 이동에 미치는 영향을 확인한 결과이다.32 is a result confirming the effect of LCN2, Forskolin or dbcGMP on the migration of astrocytes using the Boyden chamber assay.

도 33은 수정 후 3일째 제프라피쉬 배의 뇌와 척추에서 LCN2의 발현 여부를 확인한 결과이다.33 shows the results of LCN2 expression in the brain and spine of zebrafish embryos at 3 days after fertilization.

도 34는 제브라피쉬의 배에 LCN2의 mRNA를 주입함으로써 방사형 아교세포의 발생에 미치는 영향을 확인한 결과이다.Fig. 34 shows the result of confirming the effect on the generation of radial glial cells by injecting LCN2 mRNA into the zebrafish embryo.

도 35는 제브라피쉬의 배에 LCN2의 mRNA를 주입함으로써 방사형 아교세포의 돌기 두께, 길이 및 개수에서의 변화를 나타낸 것이다.Figure 35 shows the change in the projection thickness, length and number of radial glial cells by injecting the mRNA of LCN2 into the embryo of zebrafish.

도 36는 LCN2가 성상세포의 세포 사멸로의 민감화 및 형태학적 변화에 상위 단계에서 관여한다는 것을 보여주는 모식도이다.36 is a schematic showing that LCN2 is involved at higher stages in sensitization and morphological changes of astrocytes to cell death.

도 37은 리포칼린 2 항체가 성상세포의 성상세포증을 억제함을 보여주는 것이다.37 shows that lipocalin 2 antibody inhibits astrocytosis of astrocytes.

이하, 실시예는 오로지 본 발명을 보다 구체적으로 설명하기 위한 것으로서, 본 발명의 요지에 따라 본 발명의 범위가 이들 실시예에 의해 제한되지 않는다는 것은 본 발명이 속하는 기술 분야에서 통상의 지식을 가진 자에게 있어서 자명할 것이다.Hereinafter, the examples are only for illustrating the present invention in more detail, and the scope of the present invention is not limited by these examples in accordance with the gist of the present invention, those skilled in the art to which the present invention pertains. Will be self-evident.

실시예 1: 시약 및 세포의 입수Example 1: Obtaining Reagents and Cells

E. coli 0111:B4로부터 얻은 리포폴리사카라이드(Lipopolysaccharide (LPS))는 페놀 추출과 겔 여과 크로마토그래피에 의해 제조하였다. SNP(sodium nitroprusside dihydrate), SNAP (S-nitroso-N-acetylpenicillamine), PMA(phorbol 12-myristate 13-acetate), 파라콰트 디클로라이드(paraquat dichloride), 강글리오사이드 혼합물(ganglioside mixture), H2O2, NMMA (NG-Methyl-L-arginine), forskolin, dbcGMP(dibutyryl cyclic GMP), deferoxamine mesylate, ferric citrate, and polymyxin B는 모두 Sigma Chemical Co. (St Louis, MO)로부터 입수하였다. Rho Kinase (ROCK) 억제제인 Y27632는 Calbiochem (La Jolla, CA)로부터 구입하였다. 재조합 사람 TNF-a 및 마우스 IFN-g 단백질은 R&D Systems (Minneapolis, MN)로부터 구입하였다. 철로 포화된 엔테로켈린 (Iron-saturated enterochelin,0.7 kDa) EMC Microcollections GmbH (Tuebingen, Germany)로부터 구입하였다. 다른 모든 화합물들은 달리 언급하지 않는 한 Sigma Chemical Co.로부터 구입한 것이다.Lipopolysaccharide (LPS) obtained from E. coli 0111: B4 was prepared by phenol extraction and gel filtration chromatography. Sodium nitroprusside dihydrate (SNP), S-nitroso- N -acetylpenicillamine (SNAP), phorbol 12-myristate 13-acetate (PMA), paraquat dichloride, ganglioside mixture, H 2 O 2 , NMMA (N G -Methyl- L -arginine), forskolin, dibutyryl cyclic GMP (dbcGMP), deferoxamine mesylate, ferric citrate, and polymyxin B are all Sigma Chemical Co. (St Louis, MO). Rho Kinase (ROCK) inhibitor Y27632 was purchased from Calbiochem (La Jolla, Calif.). Recombinant human TNF-a and mouse IFN-g proteins were purchased from R & D Systems (Minneapolis, MN). Iron-saturated enterochelin (0.7 kDa) was purchased from EMC Microcollections GmbH (Tuebingen, Germany). All other compounds were purchased from Sigma Chemical Co. unless otherwise noted.

C6 래트 신경아교종 세포는 5% 열로 비활성화된 소태아혈청 (FBS) (GibcoBRL Gaithersburg, MD), 겐타마이신(50 mg/ml)으로 보충된 DMEM (Dulbecco’s modified Eagle medium)에 유지시켰다. 성상세포 배양물은 3일령의 ICR 마우스 (Samtako Co.; Osan, Korea)의 뇌로부터 McCarthy 및 de Vellis의 방법을 통해 제조하였다. 전체 뇌를 10% FBS, 100 U/ml 페니실린 및 100 mg/ml 스트렙토마이신(Gibco-BRL)으로 보충된 DMEM에서 분리(diassociation)하였다. 세포들을 poly D-lysine (Falcon, Becton Dickinson and Company Franklin Lakes, NJ)으로 코팅된 75 cm2 조직 배양 플라스크에 접종하였다. 세포들을 5% CO2 습한 공기 중에서 37℃에서 배양하였다. 배양 배지는 일 후에 그 후에는 매 3일마다 교환하였다. 성상세포의 이차 순수한 배양물은 혼합된 아교세포의 배양물을 250rpm에서 하룻밤동안 진탕하여 수득하고 배양 배지는 제거하였다. 성상세포는 트립신-EDTA를 이용하여 분리하고 이어서 1,000rpm으로 10분 동안 원심분리하여 수득하였다. 세포는 10% FBS, 100 U/ml 페니실린 및 100 mg/ml 스트렙토마이신으로 보충된 DMEM으로 재현탁하고, poly D-lysine으로 코팅된 6-구판에 1 x 105 세포/ml로 접종한 후 4일 동안 배양하였다. 성상세포 배양물의 순도는 GFAP 면역세포화학법에 의해 결정된 바와 같이 95% 이상이었다(데이터 미도시). 본 실시예에서 이용된 동물은 실험동물의 관리 및 이용에 대한 NIH 가이드에서 공개된 가이드라인에 따라 획득하고 관리하였다.C6 rat glioma cells were maintained in DMEM (Dulbecco's modified Eagle medium) supplemented with 5% heat inactivated fetal bovine serum (FBS) (GibcoBRL Gaithersburg, MD), gentamicin (50 mg / ml). Astrocyte cultures were prepared from the brains of three-day-old ICR mice (Samtako Co .; Osan, Korea) by the method of McCarthy and de Vellis. Whole brains were disassociated in DMEM supplemented with 10% FBS, 100 U / ml penicillin and 100 mg / ml streptomycin (Gibco-BRL). Cells were seeded in 75 cm 2 tissue culture flasks coated with poly D-lysine (Falcon, Becton Dickinson and Company Franklin Lakes, NJ). Cells were incubated at 37 ° C. in 5% CO 2 humid air. The culture medium was changed after every 3 days thereafter. Secondary pure cultures of astrocytes were obtained by shaking the culture of mixed glial cells at 250 rpm overnight and the culture medium was removed. Astrocytes were harvested using trypsin-EDTA and then centrifuged at 1,000 rpm for 10 minutes. Cells were resuspended in DMEM supplemented with 10% FBS, 100 U / ml penicillin and 100 mg / ml streptomycin, inoculated at 1 x 10 5 cells / ml in 6-spheres coated with poly D-lysine, and then 4 Incubated for days. Purity of astrocytic cultures was greater than 95% as determined by GFAP immunocytochemistry (data not shown). Animals used in this example were obtained and managed according to the guidelines published in the NIH Guide for the Management and Use of Laboratory Animals.

실시예 2: MTT 에세이에 의한 세포독성 평가Example 2: Cytotoxicity Assessment by MTT Assay

세포를 96-구판에 200 ml/구로 5 x 104 세포를 접종하고 지정된 시간 간격 동안 다양한 자극원으로 처리하였다. 처리 후에 배지를 제거하고 3-[4,5-Dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide (MTT; 0.5 mg/ml)를 추가한 다음 CO2 배양기를 이용하여 37℃에서 2시간 동안 정치시켰다. 불용성 결정은 DMSO로 완전히 용해시킨 후에 마이크로플레이트 리더 (Anthos Labtec Instruments GmbH Salzburg, Austria)를 이용하여 570nm에서의 흡광도를 측정하였다.Cells were inoculated with 5 × 10 4 cells at 200 ml / sphere in 96-spheres and treated with various stimulants for a designated time interval. After treatment, the medium was removed, 3- [4,5-Dimethylthiazol-2-yl] -2,5-diphenyltetrazolium bromide (MTT; 0.5 mg / ml) was added, followed by CO 2 incubator at 37 ° C. for 2 hours. Let it stand. Insoluble crystals were completely dissolved in DMSO and then absorbed at 570 nm using a microplate reader (Anthos Labtec Instruments GmbH Salzburg, Austria).

실시예 3: 질산 정량화Example 3: Nitric Acid Quantification

세포는 96구판에서 자극원으로 처리된 다음 배양물의 상층액내 NO2 - 를 측정함으로써 NO 생산을 평가하였다. 96구판에서 50㎕의 시료 방울을 그리스 시약 (Griess reagent; 1% sulfanilamide/0.1% naphthylethylene diamine dihydrochloride/2% phosphoric acid)과 혼합하고 25℃에서 10분 동안 정치하였다. 마이크로플레이트 리더 (Anthos Labtec Instruments GmbH)로 540nm에서의 흡광도를 측정하였다. NaNO2가 NO2 - 농도를 계산하기 위한 기준으로 이용되었다.Cells were treated with a stimulus on 96 platelets and then NO in supernatant of the culture.2                 -                  NO production was evaluated by measuring. 50 μl of the sample drop in 96 spheres was mixed with Greases reagent (1% sulfanilamide / 0.1% naphthylethylene diamine dihydrochloride / 2% phosphoric acid) and left at 25 ° C. for 10 minutes. Absorbance at 540 nm was measured with a microplate reader (Anthos Labtec Instruments GmbH). NaNO2NO2                 -                  It was used as a reference for calculating the concentration.

실시예 4: 면역형광에 의한 성상세포의 형태 분석Example 4 Morphological Analysis of Astrocytes by Immunofluorescence

면역형광 분석을 공지된 바에 따라 수행하였다(Seo, M. C. et al. 2008. J Neurosci Res 86: 1087-1095.).Immunofluorescence assays were performed as known (Seo, MC et al. 2008. J Neurosci Res 86: 1087-1095.).

즉, 성상세포를 24구판에 있는 멸균된 커버 슬립 상에서 구당 1 x 105 세포로 접종하여 배양하고, 이어서 4% 포름알데히드로 30분 동안 고정시킨 후 PBS로 두 차례 세척하였다. 시료를 PBS-Tween 20에 용해된 1% BSA로 블로킹하고 3%의 BSA 및 마우스 항-GFAP 항체 (1:30 희석) (Biogenex; San Ramon, CA)를 함유한 PBS로 정치하였다. 구를 PBS-Tween 20으로 두 차례 세척한 후 2.5 mg/ml의 Hoechst 33342 fluorochrome (Molecular Probes Eugene, OR) 및 항-마우스 IgG-fluorescein isothiocyanate (FITC)-콘쥬게이티드 이차 항체 (BD Biosciences; San Jose, CA)로 정치하였다. 시료는 형광 현미경 (Olympus BX50 Tokyo, Japan)을 이용하여 관찰하였다. 현미경의 이미지는 MetaMorphImaging System (Molecular Devices; Sunnyvale, CA)으로 처리하였다. 성상세포의 처리는 공지된 방법을 약간 수정하여 수행하였다(Wilhelmsson, U. et al. 2004. J Neurosci 24: 5016-5021; Boran, M. S. et al. 2007. J Neurochem 102: 216-230). 평균 돌기 길이는 임의로 선택된 적어도 100개의 세포를 포함하는 5개의 현미경 영역으로부터 각 세포에 대하여 가장 긴 돌기를 기반으로 한 것이다.That is, astrocytes were inoculated at 1 x 10 5 cells per sphere on sterile cover slips on a 24 sphere plate, then fixed for 4 minutes with 4% formaldehyde and washed twice with PBS. Samples were blocked with 1% BSA dissolved in PBS-Tween 20 and placed in PBS containing 3% BSA and mouse anti-GFAP antibody (1:30 dilution) (Biogenex; San Ramon, CA). The spheres were washed twice with PBS-Tween 20 followed by 2.5 mg / ml of Hoechst 33342 fluorochrome (Molecular Probes Eugene, OR) and anti-mouse IgG-fluorescein isothiocyanate (FITC) -conjugated secondary antibody (BD Biosciences; San Jose , CA). Samples were observed using a fluorescence microscope (Olympus BX50 Tokyo, Japan). Microscopic images were processed with MetaMorph Imaging System (Molecular Devices; Sunnyvale, Calif.). Treatment of astrocytes was performed with a slight modification of known methods (Wilhelmsson, U. et al. 2004. J Neurosci 24: 5016-5021; Boran, MS et al. 2007. J Neurochem 102: 216-230). The average projection length is based on the longest projection for each cell from five microscopic regions containing at least 100 cells optionally selected.

실시예 5: 아폽토시스 및 세포주기의 유속세포분석Example 5: Flow cytometry of apoptosis and cell cycle

성상세포는 트립신-EDTA를 이용하여 떼어낸 후 차가운 PBS를 이용하여 두 차례 세척하였다. 이어서 세포는 250 ml의 결합 완충용액 (10 mM HEPES, 140 mM NaCl, 2.5 mM CaCl2 pH 7.4)에 재현탁한 후 제조업자의 설명문에 따라 3 ㎕의 FITC-콘쥬게이티드 annexin V (Molecular Probes)로 정치하였다. 이어서, 세포는 가볍게 볼텍싱(vortexing)되었고 실온에서 15분 동안 어두운 조건 하에서 정치하였다. 프로피디움 아이오다이드 Propidium iodide (20 mg/ml)를 첨가한 후 유속세포분석을 FACSAria (BD Biosciences)로 1시간 동안 수행하였다.Astrocytes were detached using trypsin-EDTA and washed twice with cold PBS. The cells were then resuspended in 250 ml of binding buffer (10 mM HEPES, 140 mM NaCl, 2.5 mM CaCl 2 pH 7.4) and then placed in 3 μl of FITC-conjugated annexin V (Molecular Probes) according to the manufacturer's instructions. It was. The cells were then vortexed lightly and left under dark conditions for 15 minutes at room temperature. After adding Propidium iodide (20 mg / ml), flow cytometry was performed for 1 hour with FACSAria (BD Biosciences).

세포 주기 분포의 분석을 위하여 세포를 PBS-5 mM EDTA로 현탁한 후 100% 에탄올 방울을 첨가함으로써 고정하였다. RNase A (40 mg/ml)를 재현탁된 세포에 첨가한 후 실온에서 30분 동안 정치하였다. 프로피디움 아이오다이드 (100 mg/ml)를 첨가하고 30분 동안 정치하였다. 세포 주기의 각 단계에 있는 세포의 백분율을 FACSCalibur (BD Biosciences)을 사용하여 유속세포분석에 의하여 결정하였다.Cells were suspended by PBS-5 mM EDTA and analyzed by adding 100% ethanol drops for analysis of cell cycle distribution. RNase A (40 mg / ml) was added to the resuspended cells and then left at room temperature for 30 minutes. Propidium iodide (100 mg / ml) was added and left to stand for 30 minutes. The percentage of cells at each stage of the cell cycle was determined by flow cytometry using FACSCalibur (BD Biosciences).

실시예 6: 역전사 중합효소반응 (RT-PCR)Example 6: Reverse Transcription Polymerase Reaction (RT-PCR)

총 RNA는 6구판에 있는 C6 세포 또는 일차 성상세포로부터 TRIzol 시약 (Invitrogen Carlsbad, CA)에 의해 제조업자의 프로토콜에 따라 추출하였다. 역전사는 Superscript (Gibco-BRL) 및 올리고(dT) 프라이머를 사용하여 수행하였다. 특정 프라이머 세트를 사용한 PCR 증폭은 60℃의 어닐링 온도에서 25회 수행하였다. 프라이머의 뉴클레오타이드 서열은 공지된 cDNA 서열을 참조하여 작제하였다(표 1). PCR 반응은 DNA Engine Tetrad Peltier Thermal Cycler (MJ Research Waltham, MA)를 사용하여 수행하였다. PCR 산물의 분석을 위하여 PCR 반응물의 10 ㎕를 1% 아가로스 겔 상에서 전기영동하고 UV 광하에서 검출하였다. β-액틴이 내부 대조군으로서 이용되었다. Total RNA was extracted according to the manufacturer's protocol by TRIzol reagent (Invitrogen Carlsbad, Calif.) From C6 cells or primary astrocytic cells on the sixth plaque. Reverse transcription was performed using Superscript (Gibco-BRL) and oligo (dT) primers. PCR amplification using specific primer sets was performed 25 times at annealing temperatures of 60 ° C. The nucleotide sequence of the primers was constructed with reference to known cDNA sequences (Table 1). PCR reactions were performed using a DNA Engine Tetrad Peltier Thermal Cycler (MJ Research Waltham, Mass.). For the analysis of PCR products 10 μl of PCR reactions were electrophoresed on 1% agarose gel and detected under UV light. β-actin was used as internal control.

실시예 7: 웨스턴 블롯 에세이Example 7: Western Blot Essay

6-구판에 있는 세포를 3개의 세정제를 함유한 용균 완충용액(50 mM Tris-HCl, pH 8.0, 150 mM NaCl, 0.02% sodium azide, 0.1% SDS, 1% NP-40, 0.5% sodium deoxycholate, 1 mM phenylmethylsulfonyl fluoride)으로 용균하였다. 세포 용균액에서 단백질 농도는 Bio-Rad 단백질 에세이 키트 (Bio-Rad; Hercules, CA)를 이용하여 결정하였다. 각 시료에 대하여 동량의 단백질은 12% SDS-PAGE에 의해 분리되었고 Hybond ECL 니트로셀룰로오스 멤브레인 (Amersham Biosciences Piscataway, NJ)으로 옮겨졌다. 멤브레인은 5% 스킴 밀크로 블로킹하고 이어서 일차 항체 [goat polyclonal anti-LCN2 antibody (R&D systems) rabbit polyclonal anti-LCN2/NGAL antibody (Santa Cruz Biotech) rat monoclonal anti-BIM antibody (Calbiochem) mouse monoclonal anti-GFAP antibody (Biogenex) monoclonal anti-a-tubulin clone B-5-1-2 mouse ascites fluid (Sigma)] 및 HRP-콘주게이티드 이차 항체 (anti-goat, anti-rabbit, anti-rat, and anti-mouse IgG; Amersham Biosciences)와 정치한 후 ECL 검출 (Amersham Biosciences)을 수행하였다.Cells in 6-spheres were lysed in three lysis solutions (50 mM Tris-HCl, pH 8.0, 150 mM NaCl, 0.02% sodium azide, 0.1% SDS, 1% NP-40, 0.5% sodium deoxycholate, 1 mM phenylmethylsulfonyl fluoride). Protein concentration in cell lysate was determined using Bio-Rad Protein Assay Kit (Bio-Rad; Hercules, CA). For each sample, the same amount of protein was separated by 12% SDS-PAGE and transferred to a Hybond ECL nitrocellulose membrane (Amersham Biosciences Piscataway, NJ). Membrane blocked with 5% scheme milk followed by primary polyclonal anti-LCN2 antibody (R & D systems) rabbit polyclonal anti-LCN2 / NGAL antibody (Santa Cruz Biotech) rat monoclonal anti-BIM antibody (Calbiochem) mouse monoclonal anti-GFAP antibody (Biogenex) monoclonal anti-a-tubulin clone B-5-1-2 mouse ascites fluid (Sigma)] and HRP-conjugated secondary antibody (anti-goat, anti-rabbit, anti-rat, and anti-mouse) ECL detection (Amersham Biosciences) was performed after standing with IgG; Amersham Biosciences).

분비된 LCN2 단백질을 검출하기 위하여 배양 배지에 대한 웨스턴 블롯 분석을 수행하였다. 즉, 먼저 100mm 배양 접시에서 배양시킨 성상세포 배양물을 PBS로 5차례 세척하였다. 이어서, 세포를 최소 양의 배양 배지로 커버하고 37℃에서 자극제로 처리하였다. 조건화 배지를 회수하고 2,000 x g (5 분) 및 15,600 x g (10 분)에서 원심분리하여 비접착 세포와 세포 데브리스(debris)를 제거하였다. 시료는 -20℃에서 하룻밤 동안 트리클로로아세트산(TCA)과 아세톤의 혼합물(10% TCA and 10 mM DTT in acetone)로 침전시켰다. 침전된 단백질은 SDS-PAGE 및 ECL 검출을 수행하였다.Western blot analysis was performed on the culture medium to detect secreted LCN2 protein. That is, astrocyte cultures cultured in a 100 mm culture dish were first washed five times with PBS. Cells were then covered with minimal amount of culture medium and treated with stimulant at 37 ° C. The conditioned medium was recovered and centrifuged at 2,000 x g (5 minutes) and 15,600 x g (10 minutes) to remove non-adherent cells and cell debris. Samples were precipitated with a mixture of trichloroacetic acid (TCA) and acetone (10% TCA and 10 mM DTT in acetone) overnight at -20 ° C. Precipitated protein was subjected to SDS-PAGE and ECL detection.

실시예 8: Rho GTPase 활성 에세이Example 8: Rho GTPase Activity Assay

100mm 배양 접시에서 세포를 자극원으로 처리하고 차가운 PBS로 세척한 후 용균 완충용액 [20 mM Tris-HCl (pH 7.6), 100 mM NaCl, 10 mM MgCl2, 1% Nonidet P-40, 10% glycerol, and 1 x protease inhibitor mixture (Roche Molecular Biochemicals Mannheim, Germany)]에서 용균하였다. 용균물을 정화하고 단백질 농도를 표준화한 후 용균물 중 GTP와 결합된 Rho를 EZ-DetectTMRho activation kit (Pierce Rockford, IL)로 effector pull-down assay에 의해 측정하였다. 즉, 유사한 양의 Rho를 포함하는 세포 용균물을 아가로스에 고정된 GST-Rhotekin과 정치하고 공침전물을 항-Rho 웨스턴 블롯 에세이를 수행하여 GTP와 결합된 Rho 단백질의 양을 평가하였다. 여기서 이용된 항-Rho 항체는 RhoA, RhoB 및 RhoC를 인식할 수 있는 것으로 알려져 있다.Cells were treated with stimulants in a 100 mm petri dish and washed with cold PBS, followed by lysis buffer [20 mM Tris-HCl (pH 7.6), 100 mM NaCl, 10 mM MgCl 2 , 1% Nonidet P-40, 10% glycerol , and 1 x protease inhibitor mixture (Roche Molecular Biochemicals Mannheim, Germany). After lysing the lysates and normalizing the protein concentration, Rho bound to GTP in the lysates was measured by effector pull-down assay with EZ-Detect Rho activation kit (Pierce Rockford, IL). That is, the cell lysate containing a similar amount of Rho was left standing with GST-Rhotekin immobilized on agarose, and the coprecipitate was subjected to anti-Rho Western blot assay to evaluate the amount of Rho protein bound to GTP. It is known that the anti-Rho antibodies used herein can recognize RhoA, RhoB and RhoC.

실시예 9: lcn2 cDNA의 Gateway 클로닝 및 안정적인 형질감염Example 9 Gateway Cloning and Stable Transfection of lcn2 cDNA

RT-PCR은 C6 래트 신경아교종 세포로부터 분리한 총 RNA를 이용하여 수행하였다. 센스 또는 안티센스 방향성을 갖는 래트의 lcn2 cDNA 서열은 Gateway 클로닝 (Invitrogen)에 의한 표적 서열 특이적인 프라이머를 사용하여 pooled cDNA로부터 PCR 증폭되었다. Gateway 클로닝을 위한 PCR 프라이머의 서열은 다음과 같다: 센스 lcn2 포워드, 5’-GGGG ACA AGT TTG TAC AAA AAA GCA GGCT CCA CC ATG GGC CTG GGT GTC CTG TGT-3’(서열번호: 1) 센스 lcn2 리버스, 5’-GGGG ACC ACT TTG TAC AAG AAA GCT GGG TTG TT GTC AAT GCA TTG GTC GGT-3’(서열번호: 2) 안티센스 lcn2 포워드, 5’-GGGG ACA AGT TTG TAC AAA AAA GCA GGCT CCA CC ATG TCA GTT GTC AAT GCA TTG GTC- 3’(서열번호: 3) 안티센스 lcn2 리버스, 5’-GGGG ACC ACT TTG TAC AAG AAA GCT GGG TTG TT ATG GGC CTG GGT GTC CTG TGT-3’(서열번호: 4). 포워드 프라이머는 attB1 서열 (밑줄) 이어서 Kozak 서열 (CCACC) 및 유전자 특이적 서열 (굵은체)을 도입하여 사용하였다. 유사하게 리버스 프라이머는 attB2 서열(밑줄) 이어서 유전자 특이적 서열 (굵은체)를 도입하여 사용하였다. PCR 산물은 pDONR207 dornor 벡터 (Invitrogen)로 클로닝하고, 서열을 (Macrogen Inc. Seoul, Korea)로 재확인한 후 pDS-GFP-XB destination 벡터 (Invitrogen)로 전환하였다. 6구판에서 C6 세포는 4 ㎍ 의 GFP (Green fluorescent protein) 태그를 갖는 센스 또는 안티센스 래트 lcn2 cDNA로 lipofectAMINE reagent (Invitrogen)를 사용하여 형질전환하였다. empty pEGFP 벡터가 lcn2의 안정적인 발현을 위한 대조군으로서 이용되었다. 안정적인 형질전환체는 형질전환 후 2일째에 G418 (800 ㎍/ml)의 존재 하에서 선별되었다. 안정적 형질전환체에서 lcn2 mRNA 또는 단백질의 상향 또는 하향 조정은 RT-PCR 또는 웨스턴 블롯에 의해 확인하였다.RT-PCR was performed using total RNA isolated from C6 rat glioma cells. Lcn2 cDNA sequences of rats with sense or antisense orientation were PCR amplified from pooled cDNA using target sequence specific primers by Gateway cloning (Invitrogen). The sequence of the PCR primer for gateway cloning is as follows: sense lcn2 forward, 5'-GGGG ACA AGT TTG TAC AAA AAA GCA GGCT CCA CC ATG GGC CTG GGT GTC CTG TGT -3 '(SEQ ID NO: 1) sense lcn2 reverse , 5'-GGGG ACC ACT TTG TAC AAG AAA GCT GGG TTG TT GTC AAT GCA TTG GTC GGT -3 '(SEQ ID NO: 2) antisense lcn2 forward, 5'-GGGG ACA AGT TTG TAC AAA AAA GCA GGCT CCA CC ATG TCA GTT GTC AAT GCA TTG GTC -3 '(SEQ ID NO: 3) Antisense lcn2 Reverse, 5'-GGGG ACC ACT TTG TAC AAG AAA GCT GGG TTG TT ATG GGC CTG GGT GTC CTG TG T-3' (SEQ ID NO: 4) . The forward primer was used by introducing the attB1 sequence (underlined) followed by the Kozak sequence (CCACC) and the gene specific sequence (bold). Similarly, reverse primers were used by introducing an attB2 sequence (underlined) followed by a gene specific sequence (bold). PCR products were cloned into the pDONR207 dornor vector (Invitrogen), and the sequence was reconfirmed with (Macrogen Inc. Seoul, Korea) and then converted to the pDS-GFP-XB destination vector (Invitrogen). In 6-plates, C6 cells were transformed with lipofectAMINE reagent (Invitrogen) with sense or antisense rat lcn2 cDNA with 4 μg of GFP (Green fluorescent protein) tag. An empty pEGFP vector was used as a control for stable expression of lcn2. Stable transformants were selected in the presence of G418 (800 μg / ml) 2 days after transformation. Up or down regulation of lcn2 mRNA or protein in stable transformants was confirmed by RT-PCR or Western blot.

실시예 10: 바이러스 및 감염Example 10 Viruses and Infections

래트 lcn2-GFP를 발현하는 재조합 아데노바이러스(Ad-lcn2-GFP)를 Newgex Inc. (Seoul, Korea)에 의해 생산하였다. 즉, 래트 lcn2-GFP를 코딩하는 cDNA를 pShuttle-사이토메갈로바이러스 벡터 (Clontech Palo Alto, CA)에 삽입하였다. 래트 lcn2-GFP를 포함하는 pShuttle-사이토메갈로바이러스 벡터를 PmeI로 선형화하고 pAdEasy-1 (Clontech) 아데노바이러스의 벡터와 함께 BJ5183 E. coli로 공형질전환시켰다. 형질전환된 세포를 가나마이신을 함유한 아가로오스 판 상에 올려놓고 각 콜로니를 적합한 형질전환체의 유무에 대하여 체크하였다. 서열 확인 후에 재조합 아데노바이러스의 stock은 HEK-293A 세포로의 감염과 추출에 의해 확장하였다. 아데노바이러스는 감염된 HEK-293A 세포의 고역가 상층액으로부터 반정제하였다. 상층액은 원심분리하여 정화하여 세포 데브리스를 제거하고 -80℃에서 보관하였다. C6 세포를 GFP 또는 래트 lcn2-GFP를 발현하는 아데노바이러스로 2일간 감염시키고, 형광 현미경으로 관찰하였다. 100개 이상의 세포가 몇 개의 임의로 선택된 영역으로부터 확인되었다. GFP를 발현하는 아데노바이러스가 대조군으로서 이용되었다. Recombinant adenovirus (Ad-lcn2-GFP) expressing rat lcn2-GFP was developed by Newgex Inc. Produced by (Seoul, Korea). That is, cDNA encoding rat lcn2-GFP was inserted into pShuttle-cytomegalovirus vector (Clontech Palo Alto, Calif.). The pShuttle-cytomegalovirus vector containing rat lcn2-GFP was linearized with Pme I and cotransformed with BJ5183 E. coli with a vector of pAdEasy-1 (Clontech) adenovirus. Transformed cells were placed on agarose plates containing kanamycin and each colony was checked for the presence of a suitable transformant. After sequence identification, stocks of recombinant adenovirus were expanded by infection and extraction with HEK-293A cells. Adenovirus was semi-purified from high titer supernatants of infected HEK-293A cells. Supernatants were clarified by centrifugation to remove cell debris and stored at -80 ° C. C6 cells were infected with adenovirus expressing GFP or rat lcn2-GFP for 2 days and observed by fluorescence microscopy. More than 100 cells were identified from several randomly selected regions. Adenoviruses expressing GFP were used as controls.

실시예 11: 재조합 리포칼린 2 단백질의 정제Example 11: Purification of Recombinant Lipocalin 2 Protein

재조합 마우스 LCN2 단백질은 공지된 바에 따라 제조하였다(Yang, J. et al. 2002, Mol Cell 10: 1045-1056). 즉, 재조합 LCN2 단백질은시데로포어(siderophore)를 합성하지 않는 E. coli의 BL21 균주에서 glutathione S-transferase (GST) 융합 단백질로서 발현하였다. 단백질은 glutathione-Sepharose 4B beads (Amersham Biosciences)를 사용하고 이어서 트롬빈 또는 글루타치온으로 용출함으로써 정제하였다. 철과 엔테로켈린(enterochelin)의 로딩을 위하여, 5배 과량의 몰의 이온으로 포화된 엔테로켈린(EMC Microcollections GmbH)을 재조합 LCN2 단백질과 혼합하였다.Recombinant mouse LCN2 protein was prepared as known (Yang, J. et al. 2002,Mol cell10: 1045-1056). In other words, the recombinant LCN2 protein does not synthesize siderophores.E. coliFrom the BL21 strain It was expressed as glutathione S-transferase (GST) fusion protein. Proteins were purified by using glutathione-Sepharose 4B beads (Amersham Biosciences) and then eluting with thrombin or glutathione. For loading of iron and enterochelin, enterokeline (EMC Microcollections GmbH) saturated with 5-fold molar excess of ions was mixed with recombinant LCN2 protein.

실시예 12: 식세포작용(phagocytosis) 에세이Example 12 Phagocytosis Essay

일차 성상세포를 6구판에 구당 1 x 106 세포의 농도로 접종하였다. 배지를 제거한 후 20 mg/ml의 fluorescent zymosan particle [zymosan A (S. cerevisiae) BioParticles, Alexa Fluor 594 conjugate (Molecular Probes)]을 각 구에 첨가하고, 중력에 의해 가라앉히고 1-3시간 동안 두었다. 결합되지 않은 입자는 차가운 PBS로 3차례 세척하여 전체적으로 제거한 후 배지를 교체하였다. 그 시료는 형광 현미경(Olympus BX50)에 의해 관찰하거나 FACSAria (BD Biosciences)를 사용하여 유속세포분석에 의해 분석하였다.Primary astrocytes were seeded at 6 x platelets at a concentration of 1 x 10 6 cells per sphere. After removing the medium, 20 mg / ml of fluorescent zymosan particles [zymosan A (S. cerevisiae) BioParticles, Alexa Fluor 594 conjugate (Molecular Probes)] were added to each sphere, allowed to settle by gravity and left for 1-3 hours. Unbound particles were washed three times with cold PBS to remove the whole and the medium was replaced. The samples were observed by fluorescence microscopy (Olympus BX50) or analyzed by flow cytometry using FACSAria (BD Biosciences).

실시예 13: 세포 이동 에세이Example 13: Cell Migration Assay

세포 이동은 48구 Boyden 챔버 (Neuro Probe, Inc. Gaithersburg, MD)를 사용하여 측정하였다. DMEM에 있는 다양한 농도의 LCN2 단백질 또는 화합물을 폴리비닐피롤리돈이 없는 폴리카르보네이트 필터 (8-mm pore size, 25 x 80 mm Neuro Probe, Inc.)에 의해 상단 벽과 분리된 하단 벽에 위치시켰다. 세포는 트립신으로 처리하여 회수하고 DMEM으로 재현탁한 후 상부 챔버에 1 x 104 세포/구로 첨가하였다. 세포는 5% CO2하 37℃에서 정치하였다. 그 후 세포를 메탄올로 10분 동안 고정시키고 1시간 동안 modified Giemsa stain (Sigma)로 염색하였다. 막의 상측에 있는 세포를 면 탈지면을 사용하여 제거하였다. 이동된 세포는 광 현미경(Olympus CK2;Tokyo, Japan) (magnifications, x 100) 하에서 계수하였다. 이동된 모든 세포를 계수하고 결과는 3반복의 평균± SD (이동된 총 세포수)로서 나타내었다.Cell migration was measured using a 48-ball Boyden chamber (Neuro Probe, Inc. Gaithersburg, MD). Various concentrations of LCN2 protein or compound in DMEM were placed on a lower wall separated from the upper wall by a polycarbonate filter (8-mm pore size, 25 x 80 mm Neuro Probe, Inc.) without polyvinylpyrrolidone. Located. Cells were recovered by treatment with trypsin, resuspended in DMEM and added to the upper chamber at 1 × 10 4 cells / sphere. Cells were left at 37 ° C. under 5% CO 2 . Cells were then fixed with methanol for 10 minutes and stained with modified Giemsa stain (Sigma) for 1 hour. Cells on top of the membrane were removed using cotton wool. The migrated cells were counted under a light microscope (Olympus CK2; Tokyo, Japan) (magnifications, x100). All migrated cells were counted and the results were expressed as the mean ± SD of 3 replicates (total number of cells moved).

In vitro 상처 치유 에세이를 위하여 스크래치 상처를 24구 배양 판 상에 있는 콘플루언트 세포 단층에 10 ㎕ 피펫 팁을 사용하여 만들고 100 U/ml 페니실린 및 100 ㎍/ml 스트렙트마이신을 함유한 10% FBS를 포함한 DMEM으로 다시 채워주었다. 단층이 상처 영역으로 이동하는 중에 세포들을 5% CO2하 37℃에서 정치하였다. 상처 영역은 현미경(Olympus CK2)(배율, × 100)으로 관찰하였다. 상대적인 세포 이동 거리는 상처의 폭을 측정하고, 이를 초기 값으로부터 뺄셈하여 결정하였다(Bassi, R. et al. 2008, J Neurooncol 87: 23-33). 총 3개의 영역을 임의로 선택하고 각 구에서 계산하였다. T결과는 이동 거리의 증가의 배수로서 나타내었다.For in vitro wound healing assays, scratch wounds were made using a 10 μl pipette tip in a confluent cell monolayer on a 24-neck culture plate and 10% FBS containing 100 U / ml penicillin and 100 μg / ml streptomycin. Refilled with DMEM including. Cells were left at 37 ° C. under 5% CO 2 while the monolayer moved to the wound area. The wound area was observed under a microscope (Olympus CK2) (magnification, x 100). Relative cell migration distance was determined by measuring the width of the wound and subtracting it from the initial value (Bassi, R. et al. 2008, J Neurooncol 87: 23-33). A total of three regions were randomly selected and calculated for each sphere. T results are expressed as multiples of the increase in travel distance.

실시예 14: 제브라피쉬 분석Example 14 Zebrafish Analysis

14-1: 제브라피쉬 번식 및 관리14-1: Zebrafish Breeding and Management

배는 교배로부터 회수하여 에그 워터에 28.5℃에서 기르고, 수정 후 일수에 따라 발생하였다(Park, H. C. et al. J Neurosci 25: 6836-6844; Kucenas, S. et al. 2008. Nat Neurosci 11: 143-151). 본 실시예에서는 GFAP 프로모터의 제어하에서 EGFP를 발현하는 AB 및 Tg(GFAP:egfp) 피쉬 (Bernardos, R. L. et al. 2006, Gene Expr Patterns 6: 1007-1013)가 이용되었다.Embryos were recovered from crosses and raised in egg water at 28.5 ° C. and developed according to days after fertilization (Park, HC et al. J Neurosci 25: 6836-6844; Kucenas, S. et al. 2008. Nat Neurosci 11: 143 -151). In this example, AB and Tg (GFAP: egfp) fish (Bernardos, RL et al. 2006, Gene Expr Patterns 6: 1007-1013) expressing EGFP under the control of the GFAP promoter were used.

14-2: RNA 주입14-2: RNA injection

mRNA 주입을 위하여 전장의 오픈 리딩 프레임을 포함한 래트 lcn2 cDNA를 pCSDest 벡터로 서브클로닝하였고(Villefranc, J. A. et al. 2007, Dev Dyn 236: 3077-3087.) mRNA는 Message Machine Kit (Ambion Austin, TX)로 생산하였다. lcn2 mRNA의 생성은 전사 반응물의 겔 전기영동에 의하여 확인하였다(데이터 미도시). 100pg의 lcn2 mRNA가 일 내지 이세포 단계에 있는 배의 난황으로 주입되었다. mRNA가 주입된 배에서 LCN2 단백질의 발현은 면역세포화학법에 의하여 확인하였다(데이터 미도시).Rat lcn2 cDNA, including full-length open reading frame, was subcloned into the pCSDest vector for mRNA injection (Villefranc, JA et al. 2007, Dev Dyn 236: 3077-3087.) mRNA was expressed in the Message Machine Kit (Ambion Austin, TX). Produced as. The production of lcn2 mRNA was confirmed by gel electrophoresis of transcriptional reactants (data not shown). 100 pg of lcn2 mRNA was injected into egg yolk at one to two cell stages. Expression of LCN2 protein in embryos injected with mRNA was confirmed by immunocytochemistry (data not shown).

14-3: 면역세포화학법14-3: Immunocytochemistry

배는 AB Fix (4% paraformaldehyde, 8% sucrose, 1x PBS)로 하룻밤 동안 4℃에서 고정하고 1.5% 아가로오스/30% 수크로오스로 포매하고 액체 질소에 침지하여 얼린 2-메틸 부탄으로 동결시켰다. 냉동조직 절편기(cryostat microtome)를 이용하여 10㎛의 트랜스리버스 섹션을 회수하였다. 면역세포화합법을 위해 다음 1차 항체를 이용하였다: Zrf-1에 대한 마우스 항체 (1:400 희석, University of Oregon Monoclonal Antibody Facility), rabbit polyclonal anti-LCN2/NGAL 항체 (1:100 희석, Santa Cruz Biotech). 제브라피쉬의 방사형 아교세포의 돌기의 길이, 두께 및 개수는 다음과 같이 정량화하였다: 각 섹션별로 5개의 가장 길고 두꺼운 세포의 돌기를 측정하였다. 돌기의 총 수는 각 섹션별로 계수하였다. 각 처리별로 7개의 제브라피쉬 섹션을 정량화하였다. 결과는 척추의 직경[(가장 긴 수직 직경 + 가장 긴 수평 직경)/2)]으로 표준화하고 평균 ± SEM (n = 7)으로 나타내었다.The embryos were fixed at 4 ° C. overnight with AB Fix (4% paraformaldehyde, 8% sucrose, 1 × PBS), embedded in 1.5% agarose / 30% sucrose and immersed in liquid nitrogen and frozen in frozen 2-methyl butane. A 10 μm transreverse section was recovered using a cryostat microtome. The following primary antibodies were used for immunocytochemistry: mouse antibody against Zrf-1 (1: 400 dilution, University of Oregon Monoclonal Antibody Facility), rabbit polyclonal anti-LCN2 / NGAL antibody (1: 100 dilution, Santa Cruz Biotech). The length, thickness and number of projections of the zebrafish radial glial cells were quantified as follows: The projections of the five longest and thickest cells for each section were measured. The total number of protrusions was counted for each section. Seven zebrafish sections were quantified for each treatment. Results were normalized to the diameter of the spine [(longest vertical diameter + longest horizontal diameter) / 2) and expressed as mean ± SEM (n = 7).

하기 실시예는 전술된 실시예 1 내지 14에 기재된 실험 방법 중 적어도 하나 이상에 의하여 분석한 결과이다.The following examples are the results analyzed by at least one of the experimental methods described in Examples 1-14 described above.

실시예 15: LCN2는 세포독성 자극원에 대한 성상세포의 민감성을 증가시킴 Example 15 LCN2 Increases Sensitivity of Astrocytes to Cytotoxic Stimulators

배양된 소교세포와 성상세포의 면역에 의한 활성화는 자신의 아폽토시스를 유도하는 것으로 알려져 있다(Suk, K. et al. 2002. J Neurochem 80: 230-238; Lee, J. et al. 2001. J Biol Chem 276: 32956-32965). lcn2는 다양한유형의 세포의 생존 및 사멸에 관여하므로 다음 3가지 방식을 도입하여 활성화된 성상세포의 세포사멸에서 lcn2가 어떻게 관여하는지 조사하였다: 1) C6 아교세포에 센스 또는 안티센스 lcn2 cDNA로 형질감염에 의한 lcn2의 안정적인 과발현 또는 녹다운; 2) C6 세포에서 lcn2의 아데노바이러스에 의해 매개된 일시적인 발현; 및 3) 재조합 LCN2 단백질로 일차 성상세포 배양물 또는 C6의 처리.Immune activation of cultured microglia and astrocytes is known to induce their apoptosis (Suk, K. et al. 2002. J Neurochem 80: 230-238; Lee, J. et al. 2001. J Biol Chem 276: 32956-32965. Since lcn2 is involved in the survival and apoptosis of various types of cells, the following three methods were used to investigate how lcn2 is involved in the apoptosis of activated astrocytes: 1) Transfection of C6 glial cells with either sense or antisense lcn2 cDNA Stable overexpression or knockdown of lcn2 by; 2) transient expression mediated by adenovirus of lcn2 in C6 cells; And 3) treatment of primary astrocytic cultures or C6 with recombinant LCN2 protein.

센스 또는 안티센스 lcn2 cDNA의 안정적인 형질감염에 의해 lcn2가 증가 또는 감소 발현되는 C6 세포를 수득하였다. 안정적 형질감염체(lcn2 센스 형질감염체, S3; lcn2 안티센스 형질감염체, AS7)에서 단백질 발현과 lcn2 mRNA에서의 변화를 웨스턴 블롯 분석과 RT-PCR에 의해 각각 확인하였다(도 1). lcn2의 안정적 과발현은 C6 아교세포의 NO 공여자 SNP, H2O2 및 파라콰트에 대한 민감성을 증가시켰는데(도 2), 이들은 성상세포의 세포 사멸을 유도하는 것으로 공지되어 있다(Suk, K. et al. 2001. Brain Res 900: 342-347; Son, E. et al. 2005. Biochem Pharmacol 70: 590-597; Kim, S. et al. 2008. J Neurosci Res 86: 2062-2070). 반대로, 안티센스 lcn2 작제물의 형질감염에 의한 lcn2의 안정적 녹다운은 세포독성물질에 대한 아교세포의 민감성을 감소시켰다(도 2).By stable transfection of the sense or antisense lcn2 cDNA, C6 cells with increased or decreased expression of lcn2 were obtained. Protein expression and changes in lcn2 mRNA in stable transfectants (lcn2 sense transfectants, S3; lcn2 antisense transfectants, AS7) were confirmed by Western blot analysis and RT-PCR, respectively (FIG. 1). Stable overexpression of lcn2 increased the sensitivity of C6 glial cells to NO donors SNP, H 2 O 2 and paraquat (FIG. 2), which are known to induce apoptosis of astrocytes (Suk, K. et. 2001. Brain Res 900: 342-347; Son, E. et al. 2005. Biochem Pharmacol 70: 590-597; Kim, S. et al. 2008. J Neurosci Res 86: 2062-2070). In contrast, stable knockdown of lcn2 by transfection of antisense lcn2 constructs reduced the sensitivity of glial cells to cytotoxic substances (FIG. 2).

lcn2의 일시적인 과발현은 GFP와 융합된 lcn2 cDNA를 포함하는 아데노바이러스 벡터를 사용함으로써 달성되었다(도 3). lcn2의 발현은 C6 아교세포의 NO 공여자 SNP, H2O2 및 파라콰트에 대한 민감성을 증가시켰다(도 4).Transient overexpression of lcn2 was achieved by using an adenovirus vector containing lcn2 cDNA fused with GFP (FIG. 3). Expression of lcn2 increased the sensitivity of C6 glial cells to NO donors SNP, H 2 O 2 and paraquat (FIG. 4).

재조합 마우스 LCN2 단백질을 제조하고 잠재적 세포독성 효과를 검사하였다(도 5). LCN2 단백질은 일차 성상세포 배양물을 세포 사멸로 민감화시키는데 반하여 LCN2 단백질 단독으로는 성상세포의 생존율에 영향을 미치지 않았다(도 6). 유사한 결과가 C6 아교세포로부터 얻어졌다 (데이터 미도시). 세포 사멸의 성질을 확인하기 위하여 LCN2 단백질의 세포 사멸 증가 효과는 PI (propidium iodide)와 annexin V로의 염색 및 유속세포분석법에 의해 평가하였다(도 7). LCN2 단백질의 처리는 성상세포의 네크로시스(PI+/annexin V-) 뿐만 아니라 아폽토시스(PI-/annexin V+ 또는 PI+/annexin V+)에 대한 민감성을 증가시켰다. LCN2는 성상세포를 H2O2 또는 파라콰트에 의해 유도되는 네크로시스 세포 사멸 뿐만 아니라 NO에 의해 유도되는 아폽토시스 세포 사멸로 민감화시켰다. LCN2 단백질의 세포 사멸 증가 효과는 용량 의존적이고 통계적으로 유의한 효과가 0.1 ng/ml의 LCN2로부터 관찰되었다(도 8). 그러나, C6 아교세포 또는 일차 성상세포의 세포 주기 분포에는 유의적인 영향을 미치지 않았다(도 9).Recombinant mouse LCN2 protein was prepared and tested for potential cytotoxic effects (FIG. 5). LCN2 protein sensitized primary astrocytic cultures to cell death, whereas LCN2 protein alone did not affect astrocytic survival (FIG. 6). Similar results were obtained from C6 glial cells (data not shown). In order to confirm the properties of cell death, the effect of increasing the cell death of LCN2 protein was evaluated by staining with propidium iodide (PI) and annexin V and flow cytometry (FIG. 7). Treatment of LCN2 protein increased the sensitivity of apoptosis (PI / annexin V + or PI + / annexin V + ) as well as necrosis (PI + / annexin V ) of astrocytes. LCN2 sensitized astrocytes with necrosis cell death induced by H 2 O 2 or paraquat as well as apoptosis cell death induced by NO. The effect of increasing cell death of LCN2 protein was observed from a dose dependent and statistically significant effect of 0.1 ng / ml of LCN2 (FIG. 8). However, there was no significant effect on cell cycle distribution of C6 glial or primary astrocytic cells (FIG. 9).

실시예 16: LCN2의 세포 사멸 민감화에서 철과 Bim의 역할 규명 Example 16: Characterization of Iron and Bim in Apoptosis Sensitization of LCN2

lcn2의 전아폽토시스의 효과가 철 대사와 관련이 있다는 것이 공지되어 있으므로(Devireddy, L. R. et al. 2005. Cell 123: 1293-1305) 성상세포의 생존에 대한 철 킬레이터 DFO(deferoxamine) 또는 철 공여자 FC(ferric citrate)의 영향을 조사하였다. 박테리아의 사이더포어의 철 복합체를 포함한 lcn2가 lcn2의 수용체(lcn2R/24p3R)를 통하여 세포로 철을 공급한다는 것이 알려져 있다(Devireddy, L. R. et al. 2005. Cell 123: 1293-1305). lcn2 및 이의 수용체의 내재화(internalization)는 사이더포어(siderophore)-철 복합체로부터 철의 유입으로 이어진다. 세포로 철의 공급은 트랜스페린 수용체(TfR1)의 발현양을 감소시키고 페리틴(ferritin)의 양을 증가시킨다. 또한, 세포로 철의 유입은 전아폽토시스(proapoptosis) 단백질인 Bim 단백질의 발현양을 감소시킨다. 반대로 lcn2가 철 복합체 없이 lcn2R/24p3R에 결합하고 세포내로 내재화되면 세포내 포유동물의 사이더포어 철 복합체가 lcn2에 결합하게 되고 세포외유출(exocytosis)에 의해 세포 밖으로 방출된다. 세포내에 철이 고갈되면 전아폽토시스 분자인 Bim의 상향조절로 이어진다.It is known that the effect of preapoptosis of lcn2 is associated with iron metabolism (Devireddy, LR et al. 2005. Cell 123: 1293-1305), the iron chelator deferoxamine (DFO) or iron donor FC on survival of astrocytes. The effect of ferric citrate was investigated. It is known that lcn2, including the iron complex of bacterial cyderpore, supplies iron to cells through the lcn2 receptor (lcn2R / 24p3R) (Devireddy, LR et al. 2005. Cell 123: 1293-1305). Internalization of lcn2 and its receptors leads to the influx of iron from the siderophore-iron complex. The iron supply to the cells reduces the amount of transferrin receptor (TfR1) expression and increases the amount of ferritin. In addition, the influx of iron into the cell reduces the amount of expression of Bim protein, a proapoptosis protein. Conversely, when lcn2 binds to lcn2R / 24p3R without iron complex and internalizes intracellularly, intracellular mammalian ciderpore iron complex binds to lcn2 and is released out of the cell by exocytosis. Depletion of iron in cells leads to upregulation of Bim, a proapoptotic molecule.

lcn2의 세포 사멸 민감화 영향에서 철과 BIM 단백질의 역할을 성상세포에서 조사하였다. 철 킬레이터 (deferoxamine; DFO) 단독으로는 성상세포 및 C6 아교세포에 대하여 약하게 독성이지만, 추가 효과로서 NO 공여자 (SNAP)에 의해 유도되는 성상세포 및 C6 아교세포의 사멸을 증가시켰다(도 10). 반대로 철 공여자(ferric citrate; FC)는 부분적으로 NO 수여자에 의해 유도되는 세포 사멸을 억제하였다(도 11). 추가로, lcn2에 의해 유도된 세포 사멸 민감화는 사이더포어-철 복합체의 동시 첨가에 의해 상쇄되었다(도 12). C6 아교세포 뿐만 아니라 일차 성상세포 배양물에서 Bim mRNA 및 단백질 양은 LCN2 단백질 처리에 의하여 현저하게 증가하였다(도 13, 14).The role of iron and BIM proteins in the apoptosis sensitization effect of lcn2 was investigated in astrocytes. Iron deferoxamine (DFO) alone is weakly toxic to astrocytes and C6 glial cells, but as a further effect increased the killing of astrocytes and C6 glial cells induced by NO donors (SNAP) (FIG. 10). . In contrast, ferric citrate (FC) partially inhibited cell death induced by NO donors (FIG. 11). In addition, cell death sensitization induced by lcn2 was canceled by the simultaneous addition of the cider pore-iron complex (FIG. 12). The amount of Bim mRNA and protein in primary astrocytic cultures as well as C6 glial cells was significantly increased by LCN2 protein treatment (FIGS. 13 and 14).

실시예 17: 성상세포에서 lcn2, lcn2 수용체(lcn2R/24p3R)의 발현 및 제어 규명 Example 17: Expression and Control of lcn2 and lcn2 Receptors (lcn2R / 24p3R) in Astrocytes

lcn2는 급성기 단백질로서 제안되어 왔고(Liu, Q. et al. 1995. J Biol Chem 270: 22565-22570) lcn2의 발현은 대식세포에서 면역 자극원에 의해 조절되었다(Liu, Q. et al. 1995.J Biol Chem 270: 22565-22570; Meheus, L. A. et al. 1993. J Immunol 151: 1535-1547; Cowland, J. B. et al. 2003. J Immunol 171: 6630-6639). 이에 lcn2의 발현이 성상에서 면역 또는 다른 자극원에 의해 조절되는지 확인하였다.lcn2 has been proposed as an acute phase protein (Liu, Q. et al. 1995. J Biol Chem 270: 22565-22570) and expression of lcn2 was regulated by immune stimulators in macrophages (Liu, Q. et al. 1995 J Biol Chem 270: 22565-22570; Meheus, LA et al. 1993. J Immunol 151: 1535-1547; Cowland, JB et al. 2003. J Immunol 171: 6630-6639. Therefore, it was confirmed whether the expression of lcn2 is regulated by immunity or other stimulator in the phase.

lcn2의 발현은 리포폴리사카라이드(LPS) 및 TNF-α에 의해 강하게 증가되었다(도 15). 혈청 제거(SW), PMA, IFN-γ 및 강글리오사이드의 혼합물은 lcn2 발현에서의 약한 증가를 유도하였다(도 15). lcn2의 분비는 성상세포 배양물의 웨스턴 블롯 분석에 의해 결정된 바와 같이 LPS에 의해 증가하였다(도 16). 이러한 결과는 성상세포의 lcn2의 발현 및 분비는 CNS에서 염증 조건하에서 증가한다는 것을 의미한다. lcn2 유도 세포 사멸을 매개할 것으로 보이는(Devireddy, L. R. et al. 2005, Cell 123: 1293-1305) lcn2 수용체(lcn2R/24p3R)의 발현이 일차 성상세포 및 C6 아교세포에서 검출되었다(도 17).Expression of lcn2 was strongly increased by lipopolysaccharide (LPS) and TNF-α (FIG. 15). A mixture of serum clearance (SW), PMA, IFN-γ and gangliosides induced a slight increase in lcn2 expression (FIG. 15). Secretion of lcn2 was increased by LPS as determined by Western blot analysis of astrocytic cultures (FIG. 16). These results indicate that the expression and secretion of lcn2 in astrocytes increases under inflammatory conditions in the CNS. Expression of lcn2 receptor (lcn2R / 24p3R), which appears to mediate lcn2 induced cell death (Devireddy, LR et al. 2005, Cell 123: 1293-1305), was detected in primary astrocytes and C6 glial cells (FIG. 17).

실시예 18: LCN2에 의해 유도된 성상세포의 형태학적 변화 규명 Example 18: Identification of Morphological Changes of Astrocytes Induced by LCN2

LCN2는 세포 사멸 민감화 효과에 더하여, 성상세포의 형태학적 변화를 유도하였다. 성상세포가 LCN2 단백질에 노출되었을 때 세포 생존율에는 영향을 미치지 않으면서 세포 돌기의 개수가 증가되었다(도 18). 사이클릭 AMP(cAMP) 및 사이클릭 GMP(cGMP)가 유사한 형태학적 변화를 유도한다고 알려져 있기 때문에(Boran, M. S. et al. 2007. J Neurochem 102: 216-230; Hu, W. et al. 2008. Cell Mol Neurobiol 28: 519-528) 비교를 위해 forskolin 및 dibutyryl cyclic GMP (dbcGMP)가 이용되었다(도 18).LCN2 induced morphological changes in astrocytes in addition to apoptosis sensitization effects. When astrocytes were exposed to LCN2 protein, the number of cell processes increased without affecting cell viability (FIG. 18). Because cyclic AMP (cAMP) and cyclic GMP (cGMP) are known to induce similar morphological changes (Boran, MS et al. 2007. J Neurochem 102: 216-230; Hu, W. et al. 2008. Forskolin and dibutyryl cyclic GMP (dbcGMP) were used for comparison ( Cell Mol Neurobiol 28: 519-528) (FIG. 18).

LCN2에 의해 유도되는 성상세포의 형태학적 변화는 용량 및 시간 의존적이었다(도 19). 유의적인 변화가 100 ng/ml의 LCN2 및 이에 노출된 후 12시간째부터 발생하였다. 이들은 성상세포에서 lcn2의 발현을 증가시키고(도 15 및 16) 아교세포의 활성화를 유도하는 것으로 확인되었다(Jou, I. et al. 2006. Am J Pathol 168: 1619-1630; Yoon, H. J. et al. 2008. Mol Cells 25: 99-104).Morphological changes in astrocytes induced by LCN2 were dose and time dependent (FIG. 19). Significant changes occurred at 12 ng / ml of LCN2 and 12 hours after exposure thereto. They have been shown to increase the expression of lcn2 in astrocytes (FIGS. 15 and 16) and to induce activation of glial cells (Jou, I. et al. 2006. Am J Pathol 168: 1619-1630; Yoon, HJ et al. 2008. Mol Cells 25: 99-104).

LCN2에 의해 유도된 세포 돌기에서의 변화는 GFAP mRNA 및 단백질의 발현의 상향조정에 의해 이루어지고, 이는 LCN2의 용량 및 시간에 의존적이다(도 20). 이러한 유형의 성상세포에서의 형태학적 변화는 in vivo 반응성의 성상세포에서 일어나는 것과 유사하다(Sofroniew, M. V. 2005. Neuroscientist 11: 400-407).Changes in the cell processes induced by LCN2 are made by upregulation of the expression of GFAP mRNA and protein, which is dependent on the dose and time of LCN2 (FIG. 20). Morphological changes in this type of astrocytes are similar to those occurring in in vivo reactive astrocytes (Sofroniew, MV 2005. Neuroscientist 11: 400-407).

세포 돌기의 비대증(hypertrophy) 및 증가된 GFAP 발현은 in vivo 모든 형태의 신경 손상 이후 나타나는 반응성 성상세포의 두 가지 특징이다(Wilhelmsson, U. et al. 2004. J Neurosci 24: 5016-5021). LCN2에 의해 유도된 성상세포 형태에서의 변화가 세포 사멸에 민감한 표현형과 관련이 있는지 확인하였다. LCN2에 의해 유도되는 것과 유사한 성상세포 형태에서의 변화를 유도하는 Forskolin 및 dbcGMP 또한 세포 사멸에 민감한 표현형으로 이어졌다(도 21). dbcGMP는 LCN2 발현을 증가시키는 한편, forskolin은 그렇지 않았다(도 22). 그러나, forskolin 및 dbcGMP 모두 GFAP의 발현을 정도의 차이가 있기는 하나 증가시켰다(도 22). GFAP 발현에 대한 dbcGMP의 영향은 약함에도 불구하고, 반복된 실험에서 재현성있게 관찰되었다(데이터 미도시). dbcGMP 및 forskolin은 LCN2에 의존적 및 비의존적 방식으로 형태학적 변화를 유도할 수 있다는 것을 의미한다. 이러한 결과는 또한 성상세포의 형태학적 변화와 세포 사멸에 민감한 표현형은 서로 밀접하게 연관되어 있고, LCN2는 성상세포의 세포 사멸 및 형태의 조절에서 이중 기능을 수행한다는 것을 의미한다.Hypertrophy and increased GFAP expression are two features of reactive astrocytic cells that appear after all forms of neuronal damage in vivo (Wilhelmsson, U. et al. 2004. J Neurosci 24: 5016-5021). Changes in astrocyte morphology induced by LCN2 were confirmed to be associated with phenotypes sensitive to cell death. Forskolin and dbcGMP, which induce changes in astrocytic morphology similar to those induced by LCN2, also led to phenotypes sensitive to cell death (FIG. 21). dbcGMP increased LCN2 expression while forskolin did not (FIG. 22). However, both forskolin and dbcGMP increased the expression of GFAP to some extent (FIG. 22). Although the effect of dbcGMP on GFAP expression was weak, it was reproducibly observed in repeated experiments (data not shown). dbcGMP and forskolin mean that morphological changes can be induced in LCN2 dependent and independent manner. These results also indicate that morphological changes in astrocytes and phenotypes sensitive to cell death are closely related to each other, and that LCN2 plays a dual role in the regulation of apoptosis and morphology of astrocytes.

실시예 19: LCN2의 효과에서 NO의 역할 규명 Example 19: Role of NO in the Effects of LCN2

최근, NO가 성상세포에서 GFAP의 발현을 증폭시킨다는 것이 밝혀졌다(Brahmachari, S. et al. 2006. IJ Neurosci 26: 4930-4939). 앞서 LCN2가 또한 GFAP의 발현을 유도할 수 있다는 것을 확인하였기 때문에 (도 20) NO가 성상세포에서 LCN2 작용에 관여하는지 확인하였다. NO에 의해 유도된 GFAP 발현은 NO 공여자 SNP를 사용하여 mRNA 및 단백질 수준에서 재확인하였다(도 23). 비교를 위해 이용된 비메틴의 발현은 영향을 받지 않았다. 성상세포의 NO 생산에 대한 LCN2의 영향을 확인하였다. LCN2는 성상세포에서 NO 생산의 유의적인 증가를 유도하였다(도 24). LCN2에 의해 유도되는 NO 생산양은 LPS의 그것과 유사하고, 재조합 LCN2 제조물에서 LPS의 오염 가능성을 배제하기 위하여 polymyxin B 처리에 의해서도 상쇄되지 않았다(도 25). LCN2와 같은 방식으로 제조된 재조합 GST 단백질은 LPS의 오염 가능성을 배제하기 위한 대조군으로서 또한 사용되었다(도 25). LPS/IFN-γ에 의해 유도된 NO 생산은 polymyxin B 처리에 의해 완전히 상쇄되었다. LCN2에 의해 유도된 GFAP의 발현은 NOS 억제제 NMMA에 의해 차단되었다 (도 26). LCN2에 의해 유도된 세포 사멸 민감성에서의 증가가 NMMA에 의해 약화되었다(도 27). 이상의 결과는 NO가 LCN2에 의해 매개되는 성상세포 세포 사멸, GFAP 발현 및 형태학적 변화에 중요한 역할을 한다는 것을 의미한다.Recently, NO has been found to amplify the expression of GFAP in astrocytes (Brahmachari, S. et al. 2006. I J Neurosci 26: 4930-4939). Since it was previously confirmed that LCN2 can also induce the expression of GFAP (FIG. 20), it was confirmed whether NO is involved in LCN2 action in astrocytes. GFAP expression induced by NO was reconfirmed at the mRNA and protein levels using NO donor SNP (FIG. 23). The expression of nonmethine used for comparison was not affected. The effect of LCN2 on NO production of astrocytes was confirmed. LCN2 induced a significant increase in NO production in astrocytes (FIG. 24). The amount of NO produced induced by LCN2 was similar to that of LPS and was not offset by polymyxin B treatment to rule out the possibility of LPS contamination in recombinant LCN2 preparations (FIG. 25). Recombinant GST protein prepared in the same manner as LCN2 was also used as a control to exclude the possibility of contamination of LPS (FIG. 25). NO production induced by LPS / IFN-γ was completely offset by polymyxin B treatment. Expression of GFAP induced by LCN2 was blocked by the NOS inhibitor NMMA (FIG. 26). The increase in cell death sensitivity induced by LCN2 was attenuated by NMMA (FIG. 27). The above results indicate that NO plays an important role in astrocytic cell death, GFAP expression and morphological changes mediated by LCN2.

실시예 20: LCN2에 의해 유도된 성상세포의 변화에서 Rho/ROCK 경로의 역할 규명 Example 20: Characterization of Rho / ROCK Pathway in Changes in Astrocytes Induced by LCN2

스몰 G 단백질의 Rho 서브패밀리가 성상세포 형태의 조절에 관여한다는 것이 알려져 있으므로(Boran, M. S. et al. 2007. J Neurochem 102: 216-230; Chen, C. J. et al. 2006. stellation. Eur J Neurosci 23: 1977-1987; Suidan, H. S. et al. 1997. Glia 21: 244-252; John, G. R. et al. 2004. J Neurosci 24: 2837-2845; Ramakers, G. J. et al. 1998. Exp Cell Res 245: 252-262; Holtje, M. et al. 2005. J Neurochem 95: 1237-1248; Hall, A. 2005. Biochem Soc Trans 33: 891-895). LCN2에 의해 유도되는 성상세포의 형태학적 변화에서 Rho 단백질의 관련성을 조사하였다. Rho/ROCK 경로는 다음 결과를 보면 성상세포 형태에 대한 LCN2의 작용에서 중요한 역할을 할 것으로 보인다: 1) ROCK 억제제인 Y27632는 부분적으로 LCN2에 의해 유도된 형태학적 변화를 블로킹하였다(도 28); 2) Y27632는 또한 LCN2에 의해 유도된 GFAP의 발현을 억제하였다(도 29) 및 3) LCN2는 Rho의 활성화를 유도하였다(도 30). Y27632는 본 실시예에서 이용된 농도에서는 세포 생존율에 영향을 미치지 않았다(데이터 미도시). LCN2에 의해 유도된 Rho의 활성화는 LCN2 자극 후 1시간째에 개시되어 24시간까지 지속되었다(도 30). Since the Rho subfamily of small G proteins is known to be involved in the regulation of astrocyte morphology (Boran, MS et al. 2007. J Neurochem 102: 216-230; Chen, CJ et al. 2006. stellation. Eur J Neurosci 23 : 1977-1987; Suidan, HS et al. 1997. Glia 21: 244-252; John, GR et al. 2004. J Neurosci 24: 2837-2845; Ramakers, GJ et al. 1998. Exp Cell Res 245: 252 Hol262, M. et al. 2005. J Neurochem 95: 1237-1248; Hall, A. 2005. Biochem Soc Trans 33: 891-895). The association of Rho protein in the morphological changes of astrocytes induced by LCN2 was investigated. The Rho / ROCK pathway appears to play an important role in the action of LCN2 on astrocyte morphology according to the following results: 1) ROCK inhibitor Y27632 partially blocked morphological changes induced by LCN2 (FIG. 28); 2) Y27632 also inhibited the expression of GFAP induced by LCN2 (FIG. 29) and 3) LCN2 induced the activation of Rho (FIG. 30). Y27632 had no effect on cell viability at the concentrations used in this example (data not shown). Activation of Rho induced by LCN2 was initiated 1 hour after LCN2 stimulation and continued up to 24 hours (FIG. 30).

세포의 이동은 세포의 형태와 밀접하게 연관된 세포의 표현형이기 때문에 성상세포의 이동에 대한 LCN2의 영향을 조사하였다. 예상된 바와 같이 in vitro 상처 치료 에세이 및 Boyden 챔버 에세이에서 성상세포의 이동성을 증가시켰다(도 31)(도 32). LCN2는 상처 봉합(도 31) 및 막을 통한 이동(도 32)을 가속화하였다.Because cell migration is a phenotype of cells that is closely related to cell morphology, the effects of LCN2 on astrocytic migration were investigated. As expected, the stellate cell mobility was increased in the in vitro wound treatment assay and in the Boyden chamber assay (FIG. 31) (FIG. 32). LCN2 accelerated wound closure (FIG. 31) and migration through the membrane (FIG. 32).

실시예 21: 제브라피쉬에서 lcn2의 발현 및 기능적 분석 Example 21 Expression and Functional Analysis of lcn2 in Zebrafish

In vivo 반응성 성상세포에서 lcn2의 발현 및 그의 기능적 역할을 확인하기 위하여 제브라피쉬 모델이 이용되었다. 먼저 제브라피쉬 CNS에서 lcn2의 발현을 조사하였다. 제브라피쉬 배아의 항-LCN2 폴리클로날 항체로의 표지는 수정 후 2일째에는 신호가 보이지 않았으나(데이터 미도시), LCN2+세포가 3일째에 전뇌 및 척추에서 확인되었다(도 33의 A, B). 전뇌 절편에서 LCN2 항체는 소교세포의 형태를 갖는 소교 유사 세포를 표지하였다(도 33의 A에서 화살표). 이는 3일째에 제브라피쉬 배의 뇌에서 모든 대식세포는 초기 소교세포로의 특정한 표현형으로의 형질전환을 수행한다는 이전의 보고와 일치한다(Herbomel, P. et al. 2001. Dev Biol 238: 274-288). 흥미롭게도 수정 후 3일째 제브라피쉬 배의 척추에서 LCN2 항체는 방사형 아교세포의 특징, 즉 척추의 중심 관으로부터 연막 표면으로의 긴 방사형 돌기를 갖는 세포를 표지하였다. LCN2 항체에 의해 표지된 세포 유형을 분석하기 위하여 LCN2 항체와 방사형 아교세포의 표지인(Inagaki, M., Y. Nakamura, M. Takeda, T. Nishimura, and N. Inagaki. 1994. Glial fibrillary acidic protein: dynamic property and regulation by phosphorylation. Brain Pathol 4: 239-243) 항-GFAP 항체로 척추 절편을 표지하였다. 모든 LCN2+ 돌기가 GFAP를 발현하였고, 이는 모든 LCN2+ 세포가 제브라피쉬 배의 척추에 있는 방사형 아교세포라는 것을 의미한다(도 33의 C, D).A zebrafish model was used to confirm the expression of lcn2 and its functional role in in vivo reactive astrocytes. First, the expression of lcn2 in zebrafish CNS was examined. Labeling of zebrafish embryos with anti-LCN2 polyclonal antibodies showed no signal on day 2 after fertilization (data not shown), but LCN2 + cells were identified in the whole brain and spine on day 3 (A, B in FIG. 33). ). LCN2 antibodies in whole brain sections labeled microglia-like cells in the form of microglia (arrows in A of FIG. 33). This is consistent with previous reports that on day 3 all macrophages in the zebrafish embryo's brain undergo transformation with specific phenotypes to early microglia (Herbomel, P. et al. 2001. Dev Biol 238: 274-). 288). Interestingly, LCN2 antibodies in the spinal column of zebrafish embryos on day 3 after fertilization labeled cells with features of radial glial cells, ie long radial projections from the central duct of the spine to the surface of the smoke. In order to analyze cell types labeled by LCN2 antibodies, markers of LCN2 antibodies and radial glial cells (Inagaki, M., Y. Nakamura, M. Takeda, T. Nishimura, and N. Inagaki. 1994. Glial fibrillary acidic protein Brain Pathol 4: 239-243) Spinal sections were labeled with anti-GFAP antibodies. All LCN2 + processes expressed GFAP, meaning that all LCN2 + cells were radial glial cells in the spine of the zebrafish embryo (FIG. 33C, D).

방사형 아교세포의 발생에서 lcn2의 역할을 조사하기 위하여 Tg(gfap-egfp) 배의 일 세포 단계에서 래트 LCN2 단백질을 코딩하는 합성 mNRA를 주입하였고, 이들은 방사형 아교세포 돌기의 마커인 항-Zrf-1 항체로 표지되었다. Tg(gfap-egfp) 배는 gfap 프로모터의 조절 하에서 방사형 아교세포에서 EGFP를 발현하고(Bernardos, R. L. et al. 2006. Gene Expr Patterns 6: 1007-1013) 항-Zrf-1 항체가 방사형 아교세포의 돌기를 표지하므로(Trevarrow, B. et al. 1990. Neuron 4: 669-679) 동시에 EGFP+ 세포체 및 방사형 아교세포의 Zrf-1+ 돌기를 관찰할 수 있었다 (도 34의 A 및 C). 방사 아교의 돌기의 개수와 굵기는 in vitro 실험과 일치하게 lcn2 mRNA로 주입된 대부분의 배에서 증가하였고, 반면에 돌기의 길이는 영향을 받지 않았다(도 34의 B 및 D, 도 35). 대부분의 아교세포 돌기가 척추의 중심 관으로부터 연막 표면까지 연장되었으므로 돌기의 길이는 방사형 아교세포의 표현형의 변화를 측정하기 위한 적합한 수단이 될 수 없다.To investigate the role of lcn2 in the generation of radial glial cells, synthetic mNRAs encoding rat LCN2 proteins were injected at one cell level of Tg (gfap-egfp) embryos, which were anti-Zrf-1 markers of radial glial protuberances. It was labeled with an antibody. Tg (gfap-egfp) embryos express EGFP in radial glial cells under the control of the gfap promoter (Bernardos, RL et al. 2006. Gene Expr Patterns 6: 1007-1013). By labeling the processes (Trevarrow, B. et al. 1990. Neuron 4: 669-679), the Zrf-1 + processes of EGFP + cell bodies and radial glial cells could be observed simultaneously (A and C in FIG. 34). The number and thickness of the protrusions of the spine increased in most of the embryos injected with lcn2 mRNA, consistent with in vitro experiments, while the length of the protrusions was unaffected (FIGS. 34B and 35, 35). Since most glial processes extend from the central tube of the spine to the surface of the smoke, the length of the processes cannot be a suitable means for measuring the change in the phenotype of radial glial cells.

실시예 22: LCN 2 항체에 의한 성상세포증 억제Example 22 Inhibition of Astrocytes by LCN 2 Antibodies

In vitro 성상세포에 LCN 2를 단독으로 처리하는 한편, 성상세포에 LCN 2와 이에 대한 항체(Ab)를 함께 처리한 후 성상세포의 세포 돌기의 개수 및 형태 변화를 관찰하였다.In vitro astrocytes were treated with LCN 2 alone, while astrocytes were treated with LCN 2 and antibodies against them (Ab), and then the number and shape of cell processes of astrocytes were observed.

그 결과, 성상세포가 LCN2 단백질에 노출되었을 때 세포 돌기의 개수가 증가되며, LCN2 항체를 처리하였을 때 세포 돌기의 개수가 감소되는 것을 확인할 수 있었다. 즉 LCN2 항체에 의해 성상세포증이 억제되는 것을 알 수 있었다(도 37).As a result, when the astrocytes were exposed to the LCN2 protein, the number of cell protrusions was increased, and when the LCN2 antibody was treated, the number of cell protrusions was confirmed to decrease. In other words, it was found that astrocytosis is suppressed by the LCN2 antibody (FIG. 37).

본 발명의 조성물은 뇌 손상을 치료하는데 이용 가능하다.The compositions of the present invention can be used to treat brain damage.

Claims (7)

리포칼린 2의 억제제를 함유한 뇌 손상 치료용 조성물.A composition for treating brain damage containing an inhibitor of lipocalin 2. 제1항에 있어서,The method of claim 1, 리포칼린 2의 억제제는 성상세포의 성상세포증을 억제하는 것을 특징으로 하는 뇌 손상 치료용 조성물.Inhibitor of lipocalin 2 is a composition for treating brain damage, characterized in that it inhibits astrocytosis of astrocytes. 제 1항 또는 제 2항에 있어서,The method according to claim 1 or 2, 리포칼린 2의 억제제는 리포칼린 2에 대한 항체임을 특징으로 하는 뇌 손상 치료용 조성물.Inhibitor of lipocalin 2 is a composition for treating brain damage, characterized in that the antibody against lipocalin 2. 제 1항 또는 제 2항에 있어서,The method according to claim 1 or 2, 리포칼린 2의 억제제는 리포칼린 2의 유전자에 대한 안티센스 분자임을 특징으로 하는 뇌 손상 치료용 조성물.Inhibitor of lipocalin 2 is a composition for treating brain damage, characterized in that the antisense molecule for the gene of lipocalin 2. 제 1항 또는 제 2항에 있어서,The method according to claim 1 or 2, 뇌 손상은 퇴행성 신경질환, 뇌졸중, 외상, 뇌 감염 질환, 크로이츠펠트 야곱병, 뇌 염증 질환으로 이루어진 그룹으로부터 선택되는 것을 특징으로 하는 뇌 손상 치료용 조성물.Brain damage is a composition for treating brain damage, characterized in that selected from the group consisting of neurodegenerative diseases, stroke, trauma, brain infection diseases, Creutzfeldt Jacob disease, brain inflammatory diseases. 제 5항에 있어서,The method of claim 5, 상기 퇴행성 신경질환은 알츠하이머 질환, 파킨슨 질환, 루게릭 질환, 헌팅톤 질환 및 다발성 경화증으로 이루어진 그룹으로부터 선택되는 것을 특징으로 하는 뇌 손상 치료용 조성물.The neurodegenerative disease is a composition for treating brain damage, characterized in that selected from the group consisting of Alzheimer's disease, Parkinson's disease, Lou Gehrig's disease, Huntington's disease and multiple sclerosis. 제 1항에 따른 뇌 손상 치료용 조성물을 투여하여 뇌 손상을 치료하는 방법.A method of treating brain injury by administering a composition for treating brain damage according to claim 1.
PCT/KR2009/004676 2008-08-21 2009-08-21 Novel use of lipocalin 2 for treating brain damage Ceased WO2010021516A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
KR1020080081712A KR20100023117A (en) 2008-08-21 2008-08-21 Novel use of lipocalin 2 for treatment of brain injury
KR10-2008-0081712 2008-08-21

Publications (3)

Publication Number Publication Date
WO2010021516A2 WO2010021516A2 (en) 2010-02-25
WO2010021516A3 WO2010021516A3 (en) 2010-06-17
WO2010021516A9 true WO2010021516A9 (en) 2010-08-12

Family

ID=41707574

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2009/004676 Ceased WO2010021516A2 (en) 2008-08-21 2009-08-21 Novel use of lipocalin 2 for treating brain damage

Country Status (2)

Country Link
KR (1) KR20100023117A (en)
WO (1) WO2010021516A2 (en)

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR101295019B1 (en) * 2011-04-22 2013-08-09 경북대학교 산학협력단 Diagnosing composition for MCI, KIT by measurement lipocalin 2 and providing method of information about diagnosis MCI
CA3029178C (en) * 2016-06-30 2022-07-19 Biois Co.,Ltd Double-stranded nucleic acid signal probe and method for detecting target molecule using same
WO2020091301A1 (en) * 2018-10-31 2020-05-07 고려대학교 산학협력단 Inflammation-forming transgenic animal model through selective overexpression of lipocalin-2 and method for manufacturing same
KR102156089B1 (en) * 2018-10-31 2020-09-15 고려대학교 산학협력단 Inflammation-forming transgenic animal model through selective over-expression of lipocalin-2 and a method for producing the same
CN119548489A (en) * 2024-12-05 2025-03-04 新乡医学院 Use of LCN2 inhibitors in the treatment of infantile neuronal ceroid lipidosis

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR100681763B1 (en) * 2005-02-28 2007-02-15 재단법인 목암생명공학연구소 Cancer metastasis inhibiting pharmaceutical composition comprising human lipocalin 2 as an active ingredient, cancer metastasis inhibiting method using same
KR100806914B1 (en) * 2007-02-14 2008-02-22 경북대학교 산학협력단 Novel Use of Lipocalin 2 for the Prevention and Treatment of Degenerative Neurological Diseases
KR20080076717A (en) * 2008-01-16 2008-08-20 경북대학교 산학협력단 Novel Use of Lipocalin 2 for the Diagnosis of Degenerative Neurological Diseases

Also Published As

Publication number Publication date
WO2010021516A3 (en) 2010-06-17
WO2010021516A2 (en) 2010-02-25
KR20100023117A (en) 2010-03-04

Similar Documents

Publication Publication Date Title
Langley et al. Myostatin inhibits myoblast differentiation by down-regulating MyoD expression
Gupta et al. β-Cell mass restoration by α7 nicotinic acetylcholine receptor activation
JP2006515163A (en) Methods and compositions for modulating XBP-1 activity
WO2010021516A9 (en) Novel use of lipocalin 2 for treating brain damage
WO2019039700A1 (en) Composition including melittin for removing m2-type tumor-associated macrophage
WO2014178653A1 (en) Use of human small leucine zipper protein in adipocyte differentiation procedure
KR101321284B1 (en) Pharmaceutical composition for treating aging-related diseases comprising inhibitor of progerin and screening method thereof
Huang et al. Cbx4 governs HIF-1α to involve in Th9 cell differentiation promoting asthma by its SUMO E3 ligase activity
WO2018212503A1 (en) Cotl1 protein involved in maintaining homeostasis of hematopoietic stem cell, and use thereof
KR20100080769A (en) Modulators of hypersensitivity reactions
WO2016159575A2 (en) Composition for inhibiting growth or proliferation of chronic myelogenous leukemia cancer stem cells, and screening method therefor
WO2021246755A1 (en) Composition, for preventing, alleviating or treating metabolic disease, comprising phospholipase d2 inhibitor
Cui et al. Galanin protects against intracellular amyloid toxicity in human primary neurons
WO2013176503A1 (en) Tau protein mediated neurodegenerative disease therapeutic agent
Xu et al. NELF is a nuclear protein involved in hypothalamic GnRH neuronal migration
WO2020213982A1 (en) Composition for prevention or treatment of neurodegenerative diseases using cd9 and method for screening therapeutic agent for neurodegenerative diseases
WO2019235876A1 (en) Pharmaceutical composition comprising expression or activity inhibitor of c-src for preventing or treating synucleinopathy
WO2011087343A2 (en) Composition for treating cancer related to an hpv infection
WO2020263011A1 (en) Olfactory receptor as microglia marker and use thereof
WO2020242279A2 (en) Composition and method for inhibiting tau protein accumulation, aggregation, and tangle formation
Zhang et al. Overexpression of apolipoprotein E4 increases kainic-acid-induced hippocampal neurodegeneration
WO2020263012A1 (en) Composition for treating degenerative brain diseases, containing 2-pentylfuran as active ingredient
WO2015111971A1 (en) Pharmaceutical composition containing gpr119 ligand as active ingredient for preventing or treating non-alcoholic fatty liver disease
EP1366773A1 (en) Cell proliferation inhibitors comprising ets transcription factor or gene encoding the same
WO2014178649A1 (en) Use of human small leucine zipper protein in osteogenesis procedure

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: 09808420

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: 09808420

Country of ref document: EP

Kind code of ref document: A2