US20120093796A1 - Novel use of ec-sod and method for preparing thereof - Google Patents

Novel use of ec-sod and method for preparing thereof Download PDF

Info

Publication number
US20120093796A1
US20120093796A1 US12/532,539 US53253908A US2012093796A1 US 20120093796 A1 US20120093796 A1 US 20120093796A1 US 53253908 A US53253908 A US 53253908A US 2012093796 A1 US2012093796 A1 US 2012093796A1
Authority
US
United States
Prior art keywords
sod
protein
sod protein
diseases
cells
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.)
Abandoned
Application number
US12/532,539
Inventor
Dong Heon Shin
Tae Yoon Kim
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 Catholic University of Korea
Original Assignee
Industry Academic Cooperation Foundation of Catholic University of Korea
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
Priority claimed from KR1020070028327A external-priority patent/KR101019470B1/en
Priority claimed from KR1020070056973A external-priority patent/KR20080108876A/en
Application filed by Industry Academic Cooperation Foundation of Catholic University of Korea filed Critical Industry Academic Cooperation Foundation of Catholic University of Korea
Assigned to INDUSTRY ACADEMIC COOPERATION FOUNDATION, THE CATHOLIC UNIVERSITY OF KOREA reassignment INDUSTRY ACADEMIC COOPERATION FOUNDATION, THE CATHOLIC UNIVERSITY OF KOREA ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: KIM, TAE YOON, SHIN, DONG HEON
Publication of US20120093796A1 publication Critical patent/US20120093796A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/44Oxidoreductases (1)
    • A61K38/446Superoxide dismutase (1.15)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/0004Oxidoreductases (1.)
    • C12N9/0089Oxidoreductases (1.) acting on superoxide as acceptor (1.15)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/02Nasal agents, e.g. decongestants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/04Antipruritics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/10Anti-acne agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/06Antiglaucoma agents or miotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/14Decongestants or antiallergics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/16Otologicals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y115/00Oxidoreductases acting on superoxide as acceptor (1.15)
    • C12Y115/01Oxidoreductases acting on superoxide as acceptor (1.15) with NAD or NADP as acceptor (1.15.1)
    • C12Y115/01001Superoxide dismutase (1.15.1.1)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • the present invention relates to a novel use of an EC-SOD protein and a method for preparing thereof. More particularly, the present invention relates to compositions for preventing or treating angiogenesis-mediated disease or allergic diseases which contain, an EC-SOD protein or a vector having a polynucleotide encoding said EC-SOD proteins an active ingredient.
  • SODs superoxide dismutases
  • SODs function to protect cells by removing reactive oxygen species and enabling other antioxidant enzymes
  • SODs known to date include Cu/Zn SOD (SOD 1) containing copper and zinc atoms, Mn SOD (SOD 2) containing a manganese atom, and extracellular superoxide dismutase (EC-SOD) present on the cell surface or in the extracellular fluid.
  • SOD 1 Cu/Zn SOD
  • SOD 2 Mn SOD
  • EC-SOD extracellular superoxide dismutase
  • EC-SOD contains copper and zinc atoms as in Cu/Zn SOD, but is characterized in that a heparin binding domain is present in the C-terminal end. Since EC-SOD has the heparin-binding domain, it is assumed that EC-SOD will function to protect cell membranes by binding to the cell membranes. According to literatures, it was known that EC-SOD plays a role in the body's defense mechanism in serums and extracellular matrices (Marklund et al, Biochem. J. 266, 213-219, 1990; Su et al., Am J Respir Cell Mol Biol ., February 16(2), 162-70, 1997; et al., Thromb. Vasc. Bio.
  • EC-′SOD acts as a nuclear localization signal, so that it is located within the nuclei of thymus and testis cells so as to protect genomic DNA from oxidative stress and to regulate the DNA transcription sensitive to oxidation-reduction reaction (Ookawara T et al., BBRC, 296, 54-61, 2002).
  • the present inventors reported in Korean Patent Registration No. 10-0676502 that EC-SOD has the effect of treating skin diseases such as psoriasis by removing reactive oxygen species in skin cells and inhibiting the over-proliferation of epidermal cells.
  • angiogenesis is the process by which new capillary blood vessels are formed from preexisting vessels.
  • Angiogenesis normally occurs only in some specific situations, including embryonic development, wound healing and the female reproductive cycle, and does not substantially occur in normal conditions.
  • diseases such as cancer may be induced.
  • the angiogenic process consists of several steps that include stimulation of endothelial cell growth by tumor cytokine, vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF), degradation of extracellular matrix proteins by matrix metalloprotease (MMP), and migration of endothelial cells, which is mediated by membrane adhesion molecules, endothelial cell differentiation and tube formation (Bussolino, F. et al., Trends Biochem. Sci. 22:251-256, 1997; Kuwano, M. et al., Intern. Med. 40:565-572, 2001; Risau, W. Angiogenesis and endothelial cell function. Arzneistoffforschung 44:416-417, 1994).
  • angiogenesis inhibitors which are natural or synthetic, include protease inhibitors, tyrosine kinase inhibitors, chemokines, interleukins, and proteolytic fragments of matrix proteins (Abedi, H. et. al., J. Biol. Chem. 272:15442-15451, 1997; Cao, Y., Int. J. Biochem. Cell Biol. 33:357-369, 2001; Fong, T. A et al., Cancer Res. 59:99-106, 1999; Kwon, H.
  • Reactive oxygen species have been reported to be major regulators of angiogenesis (Jolanta Grzenkowicz-Wydra, et. al., Mol Cell Biochem., 264(1-2):169-81, 2004), but the relationship between SODs, which remove reactive oxygen species, and angiogenesis, has not yet been sufficiently examined. It was recently reported that the overexpression of Cu/Zn SOD (SOD 1), having copper and zinc atoms, among SODs, leads to the stimulation of angiogenesis, and thus the inhibition of SOD1 leads to the inhibition of angiogenesis (Jolanta Grzenkowicz-Wydra, et. al., Mol Cell Biochem., 264(1-2):169-81, 2004).
  • SOD 1 Cu/Zn SOD
  • ATN-224 which is an inhibitor of SOD1
  • SOD1 inhibits angiogenesis
  • allergy-causing factors have increased due to environmental pollution, an increase in new synthetic substances and a change in residential environments, and thus the number of people suffering from asthma and allergies has gradually increased.
  • the function of the body to show specific resistance against foreign matter, which has invaded the body, is referred to as immunity.
  • An allergy is a hypersensitive immune reaction, and typical allergic diseases include atopic dermatitis, bronchial asthma and pollenosis.
  • Clinical symptoms resulting from allergic reactions are broadly classified into specific immune reactions at an early stage, and inflammatory reactions at a late stage.
  • mast cells which are known to be widely distributed in the systemic organs, including the skin, the respiratory organ, the gastrointestinal mucosa, the brain, around lymphatic vessels and around blood vessels and to cause allergic reactions.
  • Mediators known to activate mast cells include an IgE antibody (Fc ⁇ RI) bound to a high-affinity immunoglobulin E (IgE) receptor (Fc ⁇ RI) located on the cellular membrane, compound 48/80 and the like.
  • IgE antibody Fc ⁇ RI
  • IgE immunoglobulin E
  • an IgE antibody bound to an IgE receptor forms a bridge with an antigen
  • substances such as histamine, chondroitin sulfate, heparin and protease, stored in the mast cell granules, are released through the action of phospholipase C, protein phosphatase C and calcium ions, thus mediating the early reaction stage.
  • substances including histamine, cause clinical symptoms occurring in the early allergic reaction, histamine accounts for the largest amount. In other words, in the early allergic reaction, the expression of histamine is predominant, and clinical symptoms resulting therefrom include vasodilation, edema and the like.
  • the present inventors have conducted studies on the EC-SOD protein, and as a result, have found that the EC-SOD shows an activity of inhibiting diseases caused by angiogenesis or allergic diseases. On the basis of this finding, the present inventors have developed a method for preparing the EC-SOD protein having the said activity, thereby completing the present invention.
  • the present invention provides a composition for preventing or treating angiogenesis-mediated diseases comprising an EC-SOD protein, as an active ingredient.
  • the present invention provides a composition for preventing or treating angiogenesis-mediated diseases comprising a vector having a polynucleotide encoding an EC-SOD protein, as an active ingredient.
  • the present invention provides a composition for preventing or treating allergic diseases comprising an EC-SOD protein, as an active ingredient.
  • the present invention provides a composition for preventing or treating allergic diseases comprising a vector having a polynucleotide encoding an EC-SOD protein, as an active ingredient.
  • the present invention provides a method of preparing an EC-SOD protein.
  • the present invention provides a composition for preventing or treating angiogenesis-mediated diseases comprising an EC-SOD protein, as an active ingredient.
  • the present invention provides a composition for preventing or treating angiogenesis-mediated diseases comprising a vector having a polynucleotide encoding an EC-SOD protein, as an active ingredient.
  • the present invention provides a composition for preventing or treating allergic diseases comprising an EC-SOD protein, as an active ingredient.
  • the present invention provides a composition for preventing or treating allergic diseases comprising a vector having a polynucleotide encoding an EC-SOD protein, as an active ingredient.
  • the present invention provides a method for preparing an EC-SOD protein, the method comprising the steps of: (a) cloning into an expression vector a polynucleotide encoding said EC-SOD protein (b) introducing the expression vector into host cells to transform the host cells (c) culturing the transformed host cells to express the EC-SOD protein in the cells (d) collecting the inclusion body of the expressed EC-SOD; and (e) dissolving the inclusion body and inducing the refolding of the inclusion body.
  • expression vector describes a vector capable of expressing a target protein or target RNA in a suitable host cell, and refers to a genetic construct comprising essential regulatory elements to which a gene insert is operably linked.
  • operably linked it is meant that there is a functional linkage between a nucleotide expression control sequence and a nucleotide sequence encoding a target protein, in such a manner as to perform general functions.
  • a promoter is linked with a nucleotide sequence coding for a protein or RNA, such that it can affect the expression of the coding nucleotide sequence.
  • the operable linkage to a recombinant vector may be prepared using a genetic recombinant technique that is well known in the art, and site-specific DNA cleavage and ligation may be carried out using enzymes that are generally known in the art.
  • the present invention provides a composition for preventing or treating angiogenic diseases, which contains an EC-SOD protein as an active ingredient.
  • the EC-SOD protein preferably refers to, but is not limited to, a natural or recombinant EC-SOD protein.
  • the natural EC-SOD protein is preferably derived from, but is not limited to, mice or humans, and more preferably from humans.
  • the natural EC-SOD protein preferably has, but is not limited to, amino acid sequences of SEQ ID NO: 3 or SEQ ID NO: 5, and most preferably an amino acid sequence of SEQ ID NO: 3.
  • the recombinant EC-SOD protein is not specifically limited, as long as it is prepared from the natural EC-SOD protein so as to maintain the activity thereof.
  • the recombinant EC-SOD protein has an amino acid sequence.
  • the present invention provides a composition for preventing or treating angiogenesis-mediated diseases, which contains, as an active ingredient, a vector having a polynucleotide encoding the EC-SOD protein.
  • the polynucleotide encoding the EC-SOD protein means a polynucleotide capable of encoding the natural EC-SOD protein or the recombinant EC-SOD protein and preferably has a base sequence selected from the group consisting of, but is not limited to, SEQ ID NO: 2, SEQ ID NO: 4 and SEQ ID NO: 6.
  • mice overexpressing mouse EC-SOD were prepared according to a known method (see Test Example 1), and were irradiated with UVB, and then the morphological change in the tissue thereof was compared with that of wild-type mice.
  • test Example 1 mice overexpressing mouse EC-SOD was prepared according to a known method (see Test Example 1), and were irradiated with UVB, and then the morphological change in the tissue thereof was compared with that of wild-type mice.
  • vascular endothelial growth factor (VEGF) and MMP-9 in the EC-SOD-overexpressing mice by UV light irradiation were examined using Western blot (Example 2-2), an immunohistochemical method (Example 2-3) and a zymography method (Example 2-4).
  • VEGF vascular endothelial growth factor
  • Example 2-3 an immunohistochemical method
  • Example 2-3 an immunohistochemical method
  • a zymography method Example zymography method
  • recombinant EC-SOD from which a heparin-binding domain has been removed, was expressed in E. coli , and then isolated from the E. coli strain, thus producing a refolded and purified EC-SOD protein (see Example 1), and whether the produced protein would inhibit angiogenesis was examined (see Example 3).
  • Example 1 recombinant EC-SOD protein
  • Example 3 recombinant EC-SOD
  • the EC-SOD protein or the vector having inserted therein the polynucleotide encoding the EC-SOD protein can be effectively used to prevent and treat angiogenesis-mediated diseases.
  • the angiogenesis-mediated diseases include all diseases induced by the expression of VEGF (vascular endothelial growth factor) and the expression of MMP (matrix metalloproteinase)-9, and preferred examples thereof may include, are not limited to, cancer, diabetes, rheumatoid arthritis, arteriosclerosis, hemangioma, angiofibroma, diabetic retinopathy, retinopathy of prematurity, neovascular glaucoma, corneal diseases caused by angiogenesis, degenerative spots, macular degeneration, pterygium, retinal degeneration, retrolental fibroplasia, Granular conjunctivitis, telangiectasia, pyogenic granuloma and acne ( Ophthalmol 102, 1261-1262, 1995 ;
  • the present invention provides a composition for preventing or treating allergic diseases, which contains the EC-SOD protein as an active ingredient.
  • the EC-SOD protein refers to, but is not limited to, a natural or recombinant EC-SOD protein.
  • the natural EC-SOD protein is preferably derived from, but is not limited to, mice or humans, and more preferably from humans.
  • the natural EC-SOD protein preferably has an amino acid sequence of, but is not limited to, SEQ ID NO: 3 or SEQ ID NO: 5, and more preferably SEQ ID NO: 3.
  • the recombinant EC-SOD protein is not specifically limited, as long as it is prepared from the natural EC-SOD protein using recombinant technology and maintains the activity thereof.
  • the recombinant EC-SOD protein has an amino acid sequence of SEQ ID NO: 1.
  • the present invention provides a composition for preventing or treating allergic diseases, which contains, as an active ingredient, a vector having inserted therein a polynucleotide encoding the EC-SOD protein.
  • the polynucleotide encoding the EC-SOD protein means a polynucleotide capable of encoding the natural EC-SOD protein or the recombinant EC-SOD protein and has preferably a base sequence selected from, but is not limited to, the group consisting of SEQ ID NO: 2, SEQ ID NO: 4 and SEQ ID NO: 6.
  • the effect of the recombinant EC-SOD protein on the proliferation and differentiation of T-cells was analyzed.
  • the recombinant EC-SOD protein induced the formation of the Th1 cytokine IFN- ⁇ in T cells and inhibited the formation of the Th2 cytokine IL-4.
  • the recombinant EC-SOD protein has an effect on the differentiation of T cells, and that it blocks the over-differentiation of Th2 cells causing allergic diseases and inducing the differentiation of Th2 cells into Th1 cells, and thus it can be advantageously used to prevent or treat allergic diseases (see Example 4).
  • Example 5 the effect of the natural human protein EC-SOD on the differentiation of T cells was examined. As a result, it could be seen that the EC-SOD protein also blocked the over-differentiation of Th2 cells causing allergic diseases and induced the differentiation into Th1 cells (see Example 5).
  • Example 7 whether the recombinant EC-SOD protein has the effect of inhibiting transcriptional factor activity was examined. As a result, it could be seen that the recombinant EC-SOD protein could inhibit the abnormal activity of NF- ⁇ B (see Example 6) and had the effect of reducing the degranulation of human mast cells. Accordingly, in view of the fact that mediators, such as histamine, stored in mast cells, are released due to the degranulation of the mast cells to cause inflammatory reactions such as allergy, it can be seen that the recombinant EC-SOD protein of the present invention can be advantageously used to prevent or treat allergies (see Example 7).
  • the EC-SOD protein or the vector having inserted therein the polynucleotide encoding the protein will be useful in a composition for preventing or treating allergic disease.
  • the allergy is preferably selected from the group consisting of allergic asthma, allergic rhinitis, allergic otitis media, anaphylatic shock, allergic skin disease, atopic dermatitis, psoriasis, contact allergic dermatitis and urticaria.
  • the said EC-SOD proteins which are comprised in the inventive composition may include proteins having equivalent physiological activities.
  • the said proteins with equivalent physiological activities may include functional equivalents of EC-SOD proteins as well as functional derivatives.
  • equivalent physiological activities refer to the activities of inhibiting angiogenesis or allergy.
  • the said functional equivalents may polypeptides which show at least 70% amino acid sequence homology with the amino acid sequence of SEQ ID NO: 3, preferably at least 80%, and more preferably 90%.
  • the term functional equivalents refer to peptides which have at least 70% amino acid sequence homology (i.e., identity) with the peptides produced by as a result of addition, substitution or deletion of SEQ ID NO: 3, preferably at least 80%, and more preferably at least 90%, for example, 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 9.0%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% and 100% and exhibit substantially identical physiological activity to the polypeptide of SEQ ID NO: 3.
  • Sequence identity or homology is defined herein as the percentage of amino acid residues in the candidate sequence that are identical with amino acid sequence of SEQ ID NO: 3, after aligning the sequences and introducing gaps. If necessary, to achieve the maximum percent sequence identity, any conservative substitutions is not considered as part of the sequence identity. None of N-terminal, C-terminal, or internal extensions, deletions, or insertions into the amino acid sequence of SEQ ID NO: 3 shall be constructed as affecting sequence identity or homology. Thus, sequence identity can be determined by standard methods that are commonly used to compare the similarity, in position of the amino acids of two polypeptides.
  • two polypeptides are aligned for optimal matching of their respective amino acids (either along the full length of one or both sequences or along a predetermined portion of one or both sequences).
  • the programs provide a default opening penalty and a default gap penalty, and a scoring matrix such as PAM 250 (a standard scoring matrix; Dayhoff et al., in Atlas of Protein Sequence and Structure , vol 5, supp. 3, 1978).
  • PAM 250 a standard scoring matrix; Dayhoff et al., in Atlas of Protein Sequence and Structure , vol 5, supp. 3, 1978.
  • the percent identity can be calculated as: the total number of identical matches multiplied by 100 and then divided by sum of the length of a longer sequence within the matched span and the number of gaps introduced into longer sequences in order to align the two sequences.
  • polypeptide derivatives obtained by modifying a part of the chemical structure of the polypeptide while maintaining the basic framework and physiological activity of the polypeptide. For example, this includes structural modifications for altering the stability, storage, volatility or solubility of the polypeptide.
  • polynucleotide sequences of the inventive compositions may comprise DNA, cDNA, and RNA sequences.
  • the said polynucleotide may comprise the sequence selected from the group consisting of SEQ ID NO: 2, 4 and 6.
  • the inventive compositions may comprise, as an active ingredient, plasmid having the said polynucleotide sequence thereto, or expression vectors such as viral vectors.
  • the inventive expression vector may comprise expression control sequence by which EC-SOD protein and cDNA of the EC-SOD are expressed.
  • the said polynucleotides are isolated from nature or manufactured by the methods which are known in the art.
  • the said expression control sequence refer to a DNA sequence essential to expression of coding sequence which is operatively linked within a certain host cell.
  • the said expression control sequence may comprise promoters for transcription, certain operator sequences for controlling of the said transcription, the sequences encoding relevant mRNA ribosomal binding site, and the sequences terminating transcription and translation.
  • the control sequences for prokaryotes may comprise promoter, operator, and ribosomal binding site.
  • the control sequences may comprise promoter, polyadenylated signal and enhancer.
  • an expression vector After an expression vector was inserted, it could be introduced into a target cell as a phenotype by infection, transfection and transduction which are well known method in the art.
  • Gene introducing method by using plasmid expression vector is the method which introduce plasmid DNA directly into a mammalian cell, and FDA has approved to use for human (Nabel, E. G., et al., Science, 249:1285-1288, 1990). Unlike viral vector, a plasmid DNA has advantage in respect of even purification.
  • the acceptable expression plasmids of the invention may comprise mammalian expression plasmids which are used in the art. For example, but not limited thereto, pRK5(European Patent No. 307,247), pSV16B (PCT Publication No. WO91/08291) and pVL1392(PharMingen).
  • the plasmid expression vector which comprise the said nucleic acid could be introduced to a target cell by, but not limited thereto, transient transfection, microinjection, transduction, cell fusion, calcium phosphate precipitation, liposome-mediated transfection, DEAE Dextran-mediated transfection, polybrene-mediated transfection, electroporation, gene gun, and other methods which are well known in the art (Wu et al., J. Bio. Chem., 267:963-967, 1992; Wu and Wu, J. Bio. Chem., 263:14621-14624, 1988).
  • the viral vectors which contain the said polynucleotide sequence may comprise, but not limited thereto, retrovirus, adenovirus, herpes virus, avipox virus, and lentivirus and the like. All of the viral genes of the said retroviral vectors were deleted or modified, and consequently non-viral proteins of the said vectors were produced by the infected cells.
  • the main advantages of the retroviral vectors for gene therapy are to transfer large amount of genes into cloned cells, to integrate genes specifically which are transferred to cellular DNA, and to prevent additional infection after gene transformation (Miller, A. D., Nature, 357:455-460, 1992).
  • the retroviral vectors which are approved by the FDA is manufactured by using PA317 amphotrophic retroviral packaging cell (Miller, A. D. and Buttimore, C., Molec. Cell Biol., 6:2895-2902, 1986).
  • PA317 amphotrophic retroviral packaging cell Miller, A. D. and Buttimore, C., Molec. Cell Biol., 6:2895-2902, 1986.
  • the non-retroviral vectors there is the said adenovirus (Rosenfeld et al., Cell, 68:143-155, 1992; Jaffe et al., Nature Genetics, 1:372-378, 1992; Lemarchand et al., Proc. Natl. Acad. Sci. USA, 89:6482-6486, 1992).
  • the main advantages of the adenovirus are to transfer large molecular DNA fragment (36 kb), and to transfect non-cloned cells with very high titer.
  • herpes viruses could be used in gene therapy for human (Wolfe, J. H., et al., Nature Genetics, 1:379-384, 1992).
  • the lentivirus is a kind of retrovirus, and it is developed from modification of HIV backbone since late 1990's. Unlike to existing retrovirus carriers, the lentivirus is more effective in non-divisional cells as well as divisional cells since its activity does not influenced by cell cycle.
  • the lentivirus is more effective than other viral carriers, and therefore the studies of the lentivirus as a gene transfering carrier are progressed in the field of gene therapy by using hematopoietic stem cells, and keratinocytes.
  • relevant viral vector of well known in the art may be used in the invention.
  • the term “pharmaceutically acceptable” means what is physiologically acceptable and, when administered to human, generally does not cause allergic reactions, such as gastrointestinal disorder and dizziness, or similar reactions thereto.
  • a pharmaceutically acceptable carrier for example carriers for the parenteral or oral preparations may be included.
  • the carriers for the parenteral preparations may comprise lactose, starch, cellulose derivatives, magnesium stearate, stearic acid.
  • the carriers for the parenteral preparations may comprise water, oil, saline, aqueous glucose and glycol, and stabilizers and preservatives.
  • the examples of the stabilizers may be antioxidant such as sodium hydrogen sulfite, sodium sulfite, and ascorbic acid.
  • the examples of the preservatives may be benzalkonium chloride, methyl- or prophyl-paraben, and chlorobutanol.
  • the inventive protein or a vector having inserted therein a polynucleotide encoding said protein may be prepared by mixing with the said pharmaceutically acceptable carriers into various oral or parenteral preparations according to any method known in the art.
  • the inventive peptide may be administered by various routes according to any method known in the art. Namely, it may be administered by oral or parenteral routes.
  • the injectable preparations such as isotonic solution or suspension and ointments are desirable for the parenteral preparations.
  • the injectable preparations may be prepared by using relevant dispersants, wetting agents, or suspending agents according to any method known in the art.
  • the compositions may be formulated into injectable compositions by dissolving components into saline or buffers.
  • the inventive composition can be formulated in the form of powder, granule, tablets, pills and capsules, and the like.
  • These preparations may also comprise diluents (e.g., lactose, dextrose, sucrose, mannitol, sorbitol, cellulose and/or glycine), lubricants (e.g., silica, talc, stearic acid and a magnesium or calcium salt thereof, and/or polyethylene glycol) in addition to the active ingredient.
  • diluents e.g., lactose, dextrose, sucrose, mannitol, sorbitol, cellulose and/or glycine
  • lubricants e.g., silica, talc, stearic acid and a magnesium or calcium salt thereof, and/or polyethylene glycol
  • tablets may also comprise binders, such as magnesium aluminum silicate, starch pastes, gelatin, tragacanth, methylcellulose, sodium carboxymethylcellulose and/or polyvinylpyrrolidone, and, if desired, may further comprise disintegrating agents, such as starches, agar or alginic acid or a sodium salt thereof, heterogenous mixtures and/or absorbents, colorants, flavors and sweeteners.
  • binders such as magnesium aluminum silicate, starch pastes, gelatin, tragacanth, methylcellulose, sodium carboxymethylcellulose and/or polyvinylpyrrolidone
  • disintegrating agents such as starches, agar or alginic acid or a sodium salt thereof, heterogenous mixtures and/or absorbents, colorants, flavors and sweeteners.
  • the said composition may be prepared by mixing, granulization or coating.
  • inventive pharmaceutical composition may be administered by various routes, but not limited thereto, according to any biotechnological method, any method known in the art, namely, it may be administered by oral, intravenous, intramuscular, intraarterial, intramarrow, subdural, intracardiac, intracUtaneous, subcutaneous, intraperitoneal, intranasal, gastrointestinal tracts, parenteral, sublingual or rectum.
  • an injectable form of the inventive pharmaceutical composition may be administered with a given amount of the formulation by injecting into a subcutaneous layer with a 4-6 mm syringe or lightly pricking the skin with 30-gauge injection needle, namely, the Messo therapy.
  • an ointment form of the inventive pharmaceutical composition may be administered by a method of applying directly to the skin.
  • the said ‘intracutaneous’ refers to transfer effective amount of an active ingredient which is comprised in the inventive composition by administered into the skin.
  • the composition whose active ingredient is the EC-SOD protein of the invention should be administered directly to the skin.
  • the inventive composition may be administered by biotechnological method related to protein transfer.
  • the inventive compositions may be administered to patients with the amount which is effective for preventing disease.
  • the effective amount of the inventive composition is about 0.0001 to 100 mg/kg body weight/day. Preferably 0.01 to 1 mg/kg body weight/day. It may be suitably determined by considering various factors, such as age, body weight, health condition, sex, disease severity, diet and excretion of a subject in need of treatment, as well as administration time and administration route.
  • the present invention provides the method for preparing an EC-SOD protein, which comprises the steps of:
  • the step (a) of cloning the polynucleotide encoding the EC-SOD protein into the expression vector can be carried out using a conventional recombinant DNA technique known in the art.
  • the polynucleotide encoding the EC-SOD means a polynucleotide capable of encoding the natural EC-SOD protein or the recombinant EC-SOD protein and preferably has a base sequence selected from, but is not limited to, the group consisting of SEQ ID NO: 2, SEQ ID NO: 4 and SEQ ID NO: 6.
  • the polynucleotide can be constructed by performing PCR amplification using suitable primers.
  • the DNA sequence may also be synthesized by a standard method known in the art, for example, using an automatic DNA synthesizer (commercially available from Biosearch or Applied Biosystems).
  • the expression vector may be a conventional expression vector used in the art and can be suitably selected depending on host cells. For example, when the host cells are E. coli cells, a pET28a vector (Novagene, USA) may be used. Because the preparation method of the present invention aims to express and produce EC-SOD in large amounts, any expression vector may be suitably selected and used, as long as it can achieve this purpose.
  • inventive vectors may comprise, but not limited thereto, plasmid vector, cosmid vector, bacteriophasge vector, and viral vector and the like.
  • the suitable expression vectors may comprise expression control sequences such as promoter, operator, initiation codon, stop codon, polyadenyated signal and enhancer (stimulating gene) as well as membrane targeting, signal sequence for secretion, leader sequences, and may be manufactured according to a purpose.
  • a promoter of the inventive promoter may be constitutive or inducible.
  • the inventive expression vector may comprise selective markers to select the host cells which contain the vector, and if the expression vector is replicable, it may comprise origin of replication.
  • the step (b) transforming a host cells by introducing the expression vector to the host cells may comprise any method of introducing nucleic acid to the host cells, and may be performed by transformation techniques which are well known in the art.
  • the techniques may comprise, but not limited thereto, microprojectile bombardment, electroporation, CaPO 4 precipitation, CaCl 2 precipitation, PEG-mediated fusion, microinjection, liposome-mediated method an the like.
  • the host cells may be prokaryote or eukaryote.
  • the host cells may comprise, but not limited thereto, prokaryotic host cells such as Escherichia coli, Bacillus subtilis, Streptomyces, Pseudomonas, Proteus mirabilis , and Staphylococcus , and the lower eukaryotes such as the fungi (for example, Aspergillus ), yeasts (for example, Pichia pastoris, Saccharomyces cerevisiae, chizosaccharomyces, Neurospora crassa ), higher eukaryotic cells originated from insect cells, plant cells, mammalian cells. More preferably, this invention may use Escherichia coli as a host cell.
  • the step (c) of expressing the EC-SOD protein by culturing the transformed host cells comprises culturing the host cells in a suitable medium and conditions so as to express the EC-SOD protein in the host cells.
  • the method of expressing the transformed cells to express the protein in the cells is known in the art.
  • the expression of the protein can be induced by inoculating the transformed cells in a suitable medium allowing the cells to grow, preculturing the cells, inoculating the precultured cells in a medium, and then culturing the cells in suitable conditions.
  • Collecting the inclusion body in the step (d) can be carried out using a known method for isolating and collecting insoluble inclusion bodies.
  • the inclusion body can be collected from the cell precipitate by centrifugation.
  • the step (e) of dissolving the inclusion body and inducing the refolding of the inclusion body can be carried out using any conventional method known in the art. For example, it can be carried out by dissolving the inclusion body in a buffer solution containing a protein denaturant, adsorbing the solution onto a metal affinity resin column, and inducing the refolding of the inclusion body while reducing the concentration of the denaturant.
  • dissolving the inclusion body and inducing the refolding of the inclusion body can be performed by dissolving the inclusion body in a Tris buffer solution (8 M urea, 50 mM Tris-Cl, 100 mM NaCl) containing 8M urea as a denaturant, adsorbing the solution onto a metal affinity resin-packed column, and slowly removing the urea while applying a concentration gradient to the column using a Tris buffer containing 1 M urea, thus inducing the refolding of the inclusion body.
  • a Tris buffer solution 8 M urea, 50 mM Tris-Cl, 100 mM NaCl
  • a pure refolded protein can be obtained by allowing either an elution buffer solution showing strong adsorption onto the column, or an elution buffer solution having a low pH value (generally less than 4), to flow from the column top downward so as to flow out through the column bottom.
  • the pure refolded protein can be obtained by allowing an imidazole (generally 1M)-containing elution buffer solution having strong adsorption onto a metal affinity chromatography column, to flow from the column top downward so as to flow out through the column bottom.
  • the inventive method for preparing the EC-SOD protein may additionally comprise a step (f) of separating and purifying the EC-SOD by gel-filtration column. Then, various isolation and purification processes, including additional purification, concentration and solvent exchange, may be carried out depending on the intended use.
  • the resulting EC-SOD protein can be subjected to techniques, such as salting-out (e.g., ammonium sulfate precipitation and sodium phosphate precipitation), solvent precipitation (a protein fractionation precipitation method using acetone, ethanol, or the like), dialysis, gel filtration, ion exchange, ion exchange chromatography, reverse phase chromatography and affinity chromatography, alone or in combination, to purify the EC-SOD protein.
  • salting-out e.g., ammonium sulfate precipitation and sodium phosphate precipitation
  • solvent precipitation a protein fractionation precipitation method using acetone, ethanol, or the like
  • dialysis gel filtration, i
  • step of (f) separating and purifying the EC-SOD by gel-filtration column.
  • a vector having inserted therein a polynucleotide encoding said EC-SOD protein could be administered to a required individual with effective amount, and prevent or treat diseases caused by angiogenesis or allergic diseases.
  • the said vector may be used for preparation a therapeutic agent for angiogenesis-mediated diseases or allergic diseases.
  • an EC-SOD protein or a vector having a polynucleotide encoding the EC-SOD protein has the effect on inhibiting angiogenesis by inhibiting the expression of VEGF and MMP-9 which induce angiogenesis. Therefore, the EC-SOD protein or the vector may be useful for preventing or treating angiogenesis-mediated diseases. And the EC-SOD protein or the vector having a polynucleotide encoding said EC-SOD protein also has the effect on inhibiting over-differentiation of Th2 cells which cause allergic diseases, inhibiting transcription factor (NF- ⁇ B), and reducing degranulation of human mast cells. Accordingly, the EC-SOD protein or the vector may be useful for preventing or treating allergy diseases. In addition, the inventive method for preparing an EC-SOD protein may be used in preparing an EC-SOD protein having the activity on a large scale. Therefore, the inventive method may be useful industrially.
  • FIG. 1 is a graph showing the result of Fast Protein Purification Liquid Chromatography of the inventive recombinant EC-SOD protein linear equilibrium region
  • B sample loading region
  • C column washing region
  • D refolding region
  • E refolding equilibrium region
  • F elusion region
  • G column washing region
  • X axis time and volume (hr:min:sec, mL)
  • Y axis UV density (Unit)).
  • FIG. 2 is a photograph of Coomassie-blue protein staining showing the SDS-PAGE result of the inventive recombinant EC-SOD protein which is refolded and purified.
  • M molecular size marker
  • A inclusion body which is diluted with 8M urea buffer (before refolding purification)
  • B inclusion body impurity which is not attached in the column and eluted
  • C eluted EC-SOD after refolding purification
  • D concentrated EC-SOD by ultrafiltration after desalting purification).
  • FIG. 3 is a western blot result of recombinant EC-SOD protein by using anti-EC-SOD protein antibody (SM: size marker, 1: control (HaCaT human EC-SOD), 2: inventive recombinant EC-SOD protein).
  • SM size marker
  • 1 control (HaCaT human EC-SOD)
  • 2 inventive recombinant EC-SOD protein
  • FIG. 4 is a graph of EC-SOD activity during purification step (209 FT: eluted but not attached sample during purification 209 E1: primary purified recombinant EC-SOD protein refolded by metal affinity chromatography, 209 E2: isolated and purified recombinant EC-SOD protein by gel-filtration column).
  • FIG. 5 is a photograph showing inhibition activity of recombinant EC-SOD protein from ROS caused by UVB in the HaCaT cell (UVB: UVB irradiation only, EC-SOD+UVB: UVB irradiation and EC-SOD treatment).
  • FIG. 6 is a photograph showing morphological changes in skins of mice by UV irradiation (A: wild type mouse, B: EC-SOD overexpressed mouse, C: UV irradiated wild type mouse, D:UV irradiated EC-SOD overexpressed mouse).
  • FIG. 7 is a western blot result of VEGF expression in UV irradiated mouse tissue. Actin was used as loading control (WT: wild type mouse, TG: EC-SOD overexpressed mouse).
  • FIG. 8 is a photograph showing VEGF expression of UV irradiated mouse by using immunohistological method (A: wild type mouse, B: EC-SOD overexpressed mouse, C:UV irradiated wild type mouse, D: UV irradiated EC-SOD overexpressed mouse).
  • FIG. 9 is a zymography of MMP-9 expression of UV irradiated mouse (Lane 1: wild type mouse, Lane 2: EC-SOD overexpressed mouse, Lane 3:UV irradiated wild type mouse, Lane 4: UV irradiated EC-SOD overexpressed mouse).
  • FIG. 10 is a photograph showing VEGF expression in UV irradiated keratinocyte HaCaT cells depending on treating the inventive recombinant EC-SOD using immunohistological method (A: control, B: UVB irradiated, C: recombinant EC-SOD treatment+UVB irradiation, D: recombinant EC-SOD treatment).
  • FIG. 11 is a result confirming whether treatment of recombinant EC-SOD protein has the effect on T-cell proliferation (Neg: negative control)
  • FIG. 12 is a result confirming by identifying the secretion level of IL-4 whether treatment of recombinant EC-SOD protein has the effect on T-cell differentiation (neg.: negative control, ⁇ : treated with antigen (Ova peptide) only, p: treated with antigen (Ova peptide) and PBS containing EC-SOD together, NB: treated with antigen (Ova peptide) and Sodium borate containing EC-SOD together).
  • FIG. 13 is a result confirming by identifying the secretion level of IL-4 and IFN- ⁇ whether treatment of recombinant EC-SOD protein has the effect on T-cell differentiation (neg.: negative control, ⁇ : treated with antigen (Ova peptide) only).
  • FIG. 14 is a result showing the effect of EC-SOD on cell proliferation and cytokine generation when an Ova protein and an immune adjuvant CFA (Compleat Freund's Adjuvant) was administered into human EC-SOD protein-overexpressed mice.
  • CFA Comppleat Freund's Adjuvant
  • FIG. 15 is a result showing the effect of EC-SOD on cell proliferation and cytokine generation when an Ova protein and an immune adjuvant Alum (Aluminum Hydroxide) was administered into human EC-SOD protein-overexpressed mice.
  • Alum Aluminum Hydroxide
  • FIG. 16 is a result showing the effect of recombinant EC-SOD protein on NF- ⁇ B inhibition
  • Control EC-SOD transformed HaCaT cell without UVB irradiation, UVB:HaCaT cells with UVB irradiation (100 J/m 2 ), EC-SOD/UVB:HaCaT cells treated with recombinant EC-SOD protein and UVB irradiation (100 J/m 2 ).
  • FIG. 17 is photographs showing mast cell degranulation inhibition activity of recombinant EC-SOD protein (A) and its microscopic picture (B) (Con: human mast cells group without treatment, EC-SOD: EC-SOD treated group, A+P: group having high cellular concentration of calcium, A+P+EC+SOD: group pretreated with EC-SOD and having high cellular concentration of calcium).
  • mice overexpressing mouse EC-SOD were prepared in the following manner. Meanwhile, the base sequence and amino acid sequence of the mouse EC-SOD are set forth in SEQ ID NO: 5 and SEQ ID NO: 6, respectively.
  • mice 8-10-week-old BDF1 mice (Central Laboratory Animal Inc., Korea) were used.
  • the EC-SOD-overexpressing mice were prepared according to a known method using the 8-10-week-old BDF1 mice (H. Y. Ha. et. al., Biochem Biophys Res Commun, 348: 450-458, 2006).
  • the animals were used under the approval of the clergy Institute, The clergy University of Korea.
  • hybrid mice C57BL/6 ⁇ CBA F1 were purchased from the Korea Research Institute of Bioscience and Biotechnology.
  • PMSG pregnant mare's serum gonadotropin
  • hCG human chorionic gonadotropin
  • 5 IU of each of pregnant mare's serum gonadotropin (PMSG) and human chorionic gonadotropin (hCG) were injected intraperitoneally into the female mice at an interval of 48 hours, and then mated with the same line of male mice at 1:1.
  • PMSG pregnant mare's serum gonadotropin
  • hCG human chorionic gonadotropin
  • 0.4% BSA-containing M16 culture medium was used as a culture medium.
  • mice where the skin tissue-overexpression of EC-SOD had been induced the mouse EC-SOD cDNA in a pCR11TOPO vector (provided by professor Suh Jun-Gyo, College of Medicine, Hallym University) containing mouse EC-SOD cDNA (SEQ ID NO: 6) was digested with restriction enzymes and inserted into a pBS KS vector containing a human keratin K14 promoter. Also, the vector was inserted with chicken ovalbumin poly (A) to facilitate expression in vitro. In order to inject DNA into the embryos, the expression vector was cut with restriction enzymes Hind and XhoI.
  • the DNA cut with the restriction enzymes was electrophoresed on 1% agarose gel, recovered by dialysis, and then isolated and purified with phenol-chloroform. It was dialyzed in 10 mM Tris (pH 7.4)/0.2 mM EDTA) to a final concentration of 4 ng/l.
  • the foreign gene-containing DNA solution was injected into the pronuclei of one-cell-stage embryos. The embryos injected with the foreign gene were cultured to the two-cell stage, and healthy embryos were selected from the cultured embryos. Female ICR mice in heat were mated with male mice, and next day, the presence or absence of a vaginal plug was observed, and a mouse having the vaginal plug was used as a surrogate mother.
  • the PCR amplification was performed in the following conditions: predenaturation at 94° C., 30 cycles of 30 sec at 94° C., 30 sec at 51° C., and 45 sec at 72.5° C., and then final extension at 72.5° C. for 5 min.
  • Sense primer (SEQ ID NO: 7) 5′- TTG TCT CTA ATA GAG GGT C-3′
  • Anti-sense primer (SEQ ID NO: 8) 5′-TCA AGC CTG TCT ATC TTC T-3′
  • mice overexpressing human EC-SOD were obtained from the National Jewish Medical & Research Center (Colorado, USA; bury et al. JBC 268 (21): 15394, 1993).
  • recombinant EC-SOD consisting of 209 amino acids and containing deletion of the signal peptide at the N-terminal end of human EC-SOD and deletion of 13 amino acids at the C-terminal end, was cloned in the following manner.
  • the base sequence and amino acid sequence of the recombinant EC-SOD are set forth in SEQ ID NO: 2 and SEQ ID NO: 1, respectively, and the base sequence and amino acid sequence of the human EC-SOD are set forth in SEQ ID NO: 4 and SEQ ID NO: 3, respectively.
  • a pUC 18-hEC-SOD vector (provided by Professor Marklund, Clinical Chemistry, Sweden) containing human EC-SOD cDNA (SEQ ID NO: 4), asa template, was amplified by PCR using the following primers, thus preparing human EC-SOD cDNA:
  • Sense primer (SEQ ID NO: 9) 5′-tagattctggacgggcgagga-3′
  • Anti-sense primer (SEQ ID NO: 10) 5′-tactcgagtcactctgagtgct-3′
  • the PCR amplification was performed using PfuI polymerase in the following conditions: predenaturation at 95° C. for 5 min, and then 30 cycles of 30 sec at 95° C., sec at 55° C. and 1 min at 72° C., followed by final extension at 72° C. for 5 min.
  • the amplified gene product was digested with restriction enzymes EcoRI and XhoI, and then ligated into a pET28a, vector (NovaGene, USA), digested with the same restriction enzymes, using T4 ligase at 4° C. for 12 hours.
  • the vector was transfected into E. coli DH5a, and then the transformants were cultured in LB solid medium containing 25 mg/mL of kanamycin, and the cultured transformants were screened. Whether the EC-SOD gene had been correctly inserted into the screened transformants was examined by gene sequence analysis. From the transformants determined to have the recombinant EC-SOD gene inserted therein, a vector was isolated. The vector was inserted into host cell E. coli BL21 (NovaGene, USA) for overexpression, and the cells were cultured in LB solid medium containing 25 mg/mL of kanamycin, while a strain overexpressing the EC-SOD protein was screened.
  • Example 1-1 The strain screened in Example 1-1 was cultured in LB liquid medium, containing 25 mg/mL of kanamycin, at 37° C. When the cells reached an OD (optical density) of 0.6 at 600 nm, IPTG (1 mM) was added thereto to induce the expression of the EC-SOD protein, and the cells were cultured at 37° C. for 6 hours, thus expressing the recombinant EC-SOD protein in the cells.
  • OD optical density
  • the cells were collected from the culture medium by centrifugation, and the collected cells were suspended in 50 mM Tris buffer (pH 7.5) containing 100 mM calcium chloride and were disrupted with an ultrasonic disruptor.
  • the cell suspension was centrifuged, and the precipitate layer was isolated and washed three times with 50 mM Tris buffer containing 100 mM calcium chloride.
  • the washed cell precipitate contained an insoluble protein inclusion body. 10 mg of the washed inclusion body (a fusion protein content of 50%) was added to Tris buffer solution (50 mM Tris-Cl, 100 mM NaCl, pH 7.5), and then dissolved by the addition of 8M urea. After the inclusion body was completely suspended at room temperature for 2 hours, it was subjected to phase separation by centrifugation. The upper aqueous layer was collected because it contained the dissolved recombinant EC-SOD protein inclusion body (inclusion body solution).
  • the refolding and purification of the recombinant EC-SOD protein were performed in the following manner using fast protein purification liquid chromatography (FPLC).
  • the buffer solution (8 M urea, 50 mM Tris-Cl, 100 mM NaCl, pH 7.5) was introduced into a metal affinity resin-packed chromatography column (5 ml His Trap FF crude, GE Healthcare, USA) at a flow rate of 1 ml/min to equilibrate the column, and then 5 ml of the above-obtained inclusion body solution was injected downward into the top of the metal affinity resin-packed chromatography column (3 ml) at a flow rate of 1 mL/min, so that the folded protein was adsorbed onto the solid matrix in the column.
  • FPLC fast protein purification liquid chromatography
  • a Tris buffer solution (50 mM Tris buffer, 1 M urea, 100 mM NaCl, 100 mM ZnCl 2 , 50 mM CuSO 4 , pH 7.5), containing 1 M urea and comprising Zn and Cu atoms having an effect on the activity of EC-SOD, was injected downward into the top of the column at a flow rate of 0.5 mL/min while applying a linear concentration gradient, thus gradually reducing the concentration of the denaturant in the column. Accordingly, the metal ions (Zn and Cu) were completely substituted into the target protein with the natural three-dimensional structure, and thus the folded protein adsorbed onto the solid matrix was refolded to have activity.
  • Tris buffer solution containing 2M imidazole and 1M urea, was fed downward into the top of the column with a concentration gradient.
  • the refolded fusion protein was eluded when an elution buffer solution having an imidazole concentration of 0.5M was fed into the column (see FIG. 1 ).
  • a fast protein purification liquid chromatography (FPLC) column and a gel filtration chromatography column were used.
  • FPLC fast protein purification liquid chromatography
  • a gel filtration chromatography column Sepharose 12 column
  • a Tris buffer solution 50 mM Tris-Cl, 100 mM NaCl, pH 7.5
  • a phosphate buffered saline pH 7.5
  • the recombinant EC-SOD protein of the present invention could be obtained. Also, the sample in each purification step was analyzed by SDS-PAGE and, as a result, as can be seen in FIG. 2 , the recombinant EC-SOD protein was well separated.
  • Example 1-4 Western blot analysis was performed.
  • human EC-SOD expressed by human keratinocyte HaCaT cells (provided by Professor N. Fusenig, Cancer Research, Germany) was used.
  • the polyvinylidene fluoride membrane was blocked with 5% skimmed milk and cultured with an anti-EC-SOD polyclonal antibody (1: 500, Santa Cruz, USA).
  • the membrane was washed with water, incubated with peroxidase-conjugated secondary antibody IgG (Santa Cruz, USA), washed again with water, and then color-developed using an ECL detection kit (GE Healthcare, USA) according to the manufacturer's instruction.
  • the sample (209FT) eluted without being adsorbed in the purification process showed no activity
  • the primarily purified recombinant EC-SOD protein (209 E1) refolded using the metal affinity chromatography column showed an activity of 250 unit/mg protein
  • the recombinant EC-SOD protein (209 E2) separated and purified using the gel-filtration column showed an activity of 360 unit/mg protein.
  • Example 1-4 In order to examine the ROS (reactive oxygen species) removal effect of the recombinant EC-SOD protein in HaCaT cells, the recombinant EC-SOD protein, separated and purified in Example 1-4, was treated in the following manner.
  • ROS reactive oxygen species
  • Skin keratinocyte HaCaT cells were plated on 18-mm cover slips at a concentration of 1 ⁇ 10 4 cells/cover slip, and then cultured in an IMDM medium (Isocove's modified Dulbecco's medium, GIBCO), containing 10% fetal bovine serum (FBS, GIBCO) and 1% of 100 units penicillin and 100 g streptomycin/ml of medium in 5% CO 2 at 37° C.
  • FBS fetal bovine serum
  • FBS fetal bovine serum
  • the cells were irradiated with UVB at a dose of 100 J/m 2 for 3 hours, the cells were allowed to react with 10 mM DCFH-DA at 37° C. for 30 minutes and observed with a fluorescent microscope.
  • the fluorescent micrograph was obtained using a Zeiss digital camera.
  • 6 UVB lamps F24T12/UVB/HO, 290-320 nm, Voltare, Fairfield, Conn., USA
  • the height of the lamps was controlled such that the surface of the back skin of each of the anesthetized mice was irradiated with a dose of 0.3 mW/cm 2 .
  • VEGF vascular endothelial growth factor
  • an extraction solution 50 mM Tris HCl pH 8.0, 5 mM EDTA, 150 mM NaCl, 0.5% sodium deoxycholate, 1% Nonidet P-40, 0.1% SDS, 1 mM PMSF, 1 mM NaF, 1 mM NaVO 4
  • the protein extracts were separated on 10% polyacrylamide gel, and then transferred to a polyvinylidene fluoride membrane (Gelman Laboratory, MI, USA). Then, the polyvinylidene fluoride membrane was blocked with 5% skimmed milk, and then incubated with each of an anti-VEGF polyclonal antibody (1: 500, Santa Cruz, USA) and an anti-beta-actin antibody. The membrane was washed with water, incubated with peroxidase-conjugated secondary antibody IgG (SantaCruz, USA), washed again with water, and then color-developed with an ECL detection kit.
  • the fixed tissue was washed with running water, dehydrated with ethanol, embedded in paraffin, sectioned to a thickness of 5 mm and attached to glass slides.
  • the sections were deparaffinized with xylene solution, hydrated with ethanol, and then stained with hematoxylin and eosin.
  • the deparaffinized tissue segments were blocked by treatment with 3% hydrogen peroxide solution. To reduce nonspecific binding and background staining, the sections were treated with 10% goat serum.
  • Each of the tissue slides was treated with an anti-VEGF polyclonal antibody (1: 500, Santa Cruz, USA) and visualized by the avidin-biotin-immunoperoxidase complex method using a streptavidin-biotin kit (LSAB, Dako, Calif., USA).
  • the stained tissue was washed with water, and then color-developed with diaminobenzidine (DAB), and the tissue sections were counterstained with 0.2% Mayer's hematoxylin (Sigma, Mo., USA). The staining results are shown in FIG. 8 .
  • VEGF As shown in FIG. 8 , in the wild-type mice, VEGF was highly expressed by UVB irradiation, but in the EC-SOD-overexpressed mice, the expression of VEGF was significantly inhibited compared to that in the wild-type mice. These results were consistent with the results of Example 1-2. These results suggest that natural EC-SOD has the effect of inhibiting the expression of angiogenesis-inducing VEGF expression in vivo to inhibit angiogenesis.
  • MMP9 is known to be a factor regulating the tumor angiogenesis process through the VEGF-VEGFR system (Bergers et al., Nat. Cell Biol., 2: 737-744, 2000).
  • the present inventors examined the expression level of MMP-9 in EC-SOD-overexpressing mice by UVB irradiation.
  • the natural EC-SOD protein inhibits the expression of VEGF in vivo, and in addition, inhibits the expression of MMP-9, which is involved in angiogenesis-related signaling through the VEGF-VEGFR system, thus effectively inhibiting angiogenesis.
  • Keratinocyte HaCaT cells were plated on 18-mm cover slips at a concentration of 1 ⁇ 10 4 cells/cover slip. The cells were cultured in DME medium (Dulbecco's minimum essential medium, GIBCO), containing 10% fetal bovine serum (FBS, GIBCO) and 100 units penicillin and 100 g streptomycin/ml of medium, in 5% CO 2 at 37° C. When the adhered cells reached a confluence of 30-40%, the fetal bovine serum was removed, and then cultured for 24 hours.
  • DME medium Dulbecco's minimum essential medium, GIBCO
  • FBS fetal bovine serum
  • the cells When the cells reached a confluence of 50-60%, the cells were pretreated with 10 ⁇ g of EC-SOD. After the cells were treated with the purified recombinant EC-SOD protein for 30 minutes, the cells were treated with UV light at a dose of 100 J, and after 24 hours, the skin keratinocyte cells were fixed with paraformaldehyde and immunostained. The fixed cells were blocked with 10% normal goat serum and washed three times with phosphate buffered saline (PBS). For immunostaining for VEGF (vascular endothelial growth factor), the cells were treated with a rabbit anti-VEGF antibody (Santacruz, USA), and then incubated at 4° C. for at least 16 hours. Then, the cells were fluorescence-stained with a FITC-conjugated anti-mouse-IgG secondary antibody (Zymed, USA), and then observed with a fluorescent microscope (Carl Zeiss, Germany).
  • VEGF vascular end
  • EC-SOD Although recombinant EC-SOD has no effect on the proliferation of T cells, it can have an effect on the differentiation of T cells. For this reason, T cell cytokines were measured to examine the differentiation of T cells.
  • Spleen cells were cultured with the antigen ova peptide for 48 hours, and then IL-4 (interleukin-4) produced in the culture medium was measured.
  • the measurement was carried out using an ELISA-kit (Becton, Dickinson and Company, USA) according to the manufacturer's instruction.
  • EC-SOD reduced the formation of IL-4. This suggests that EC-SOD has an effect on the differentiation of T cells.
  • neg. a test group not treated with an antigen
  • a test group treated only with an antigen (Ova peptide)
  • p a test group treated with an antigen (Ova peptide) together with EC-SOD in PBS NB: a test group treated with an antigen (Ova peptide) together with EC-SOD in sodium borate. All the test groups were treated in culture media for 48 hours.
  • the recombinant EC-SOD protein was administered into the T-cells of mice, and the amounts of production of cytokines, such as IL-4 and IFN- ⁇ , were compared.
  • CD4 T cells were isolated using MACS (Miltenyi Biotech).
  • MACS Miltenyi Biotech
  • Ova peptide 1 ⁇ g/ml, Ova323-339, Peptron
  • CD4 T cells isolated using MACS in RPMI1640 medium supplemented with 10% FBS.
  • the recombinant EC-SOD protein separated and purified in Example 1, was added to the culture medium. After 72 hours, the culture medium was collected, and the amounts of IL-4 and IFN- ⁇ in the culture medium were measured using ELISA (antibody: Becton Dickinson).
  • the recombinant EC-SOD protein As a result, as can be seen in FIG. 13 , the recombinant EC-SOD protein, separated and purified in Example 1, induced the formation of Th1 cytokine IFN- ⁇ in T cells and inhibited the formation of Th2 cytokine IL-4. This suggests that the recombinant EC-SOD protein had an effect on immune cells, particularly on the differentiation of T cells.
  • the recombinant EC-SOD protein of the present invention blocks the over-differentiation of Th2 cells inducing allergic diseases and induces differentiation into Th1 cells, and thus it can be advantageously used to treat or prevent allergic diseases.
  • An Ova protein and an immune adjuvant CFA (Complete Freund's Adjuvant, Sigma) inducing Th1 immunity were mixed with alum (aluminum hydroxide, Pierce) inducing Th2 immunity, and 25 ⁇ g of the mixture was immunized into the foot sole and tail of the human EC-SOD-overexpressed mice, prepared in Test Example 1. After 11 days, from lymphocytes having inflammation occurred therein, cells were isolated and observed to examine whether EC-SOD had effects on cell proliferation and cytokine production.
  • CFA Complete Freund's Adjuvant
  • the natural EC-SOD protein blocks the over-differentiation of Th2 cells inducing allergic diseases and induces differentiation into Th1 cells, and thus it can be advantageously used to treat or prevent allergic diseases.
  • HaCaT cells were plated on a 14-mm culture slide, and then cultured in 10% fetal bovine serum-containing DMEM (Dulbecco's modified Eagle's medium) for 24 hours. After 24 hours of the culture, the cells were starved in 1% fetal bovine serum-containing DMEM medium for 24 hours. After the starvation, the cells were pretreated with 10 ⁇ g of EC-SOD for 30 minutes, and then washed three times with phosphate buffered saline (PBS).
  • PBS phosphate buffered saline
  • the cells were then irradiated with UV light at a dose of 100 J/m 2 .
  • the cells were washed again with phosphate buffered saline, treated with 10 ⁇ g of EC-SOD, and then further cultured in 1% fetal bovine serum-containing DMEM medium for 4 hours. Then, the cells were fixed with methanol and immunostained. The fixed cells were blocked with 10% normal goat serum and washed three times with phosphate buffered saline (PBS).
  • PBS phosphate buffered saline
  • the cells were treated with each of a mouse anti-EC-SOD antibody (Lab Frontier, Korea) and a rabbit anti-NF-kB antibody (Santa Cruz, USA) and incubated at 4° C. for at least 16 hours. Then, the cells were fluorescence-stained with TRITC (tetramethyl rhodamine isothiocyanate)-conjugated anti-rabbit IgG secondary antibody, Serotec, GB) and a FITC-fused anti-mouse IgG secondary antibody (Zymed, USA), and then counterstained with Hoechst (Sigma, USA).
  • TRITC tetramethyl rhodamine isothiocyanate
  • FIG. 16 200 ⁇ magnification
  • NF- ⁇ B was not immunostained in the cytoplasm and the nucleus, suggesting that NF- ⁇ B was inactivated
  • FIGS. 16A , D, G and J It was also observed that, in the UVB group (where the HaCaT cells were irradiated with UVB at a dose of 100 J/m 2 ), NF- ⁇ B was rapidly activated at the nuclear site and the perinuclear site ( FIGS. 16B , E, H and K).
  • the recombinant EC-SOD protein of the present invention inhibits the abnormal activation of NF- ⁇ B, and thus it can be used to prevent or treat autoimmune diseases, such as allergies.
  • mast cells were stained with toluidine blue, and then the number of degranulated cells among 100 stained cells was counted.
  • Human mast cells (semi-adhesion, Human Mast Cell-1 (HMC-1) provided by professor Hyung-Min Kim, Kyunghee University) were plated on 18 ⁇ 18 mm cover slips at a concentration of 1 ⁇ 10 4 cells/cover slip.
  • the cells were cultured in IMDM medium (Isocove's modified Dulbecco's medium, GIBCO), containing 10% fetal bovine serum (FBS, GIBCO) and 100 units penicillin and 100 g streptomycin/ml of medium, in 5% CO 2 at 37° C. Because the cells were semi-adhesion cells, the ratio of adhered cells to non-adhered cells was about 4:6.
  • the adhered cells reached a confluence of about 30-40%, the cells were pretreated with 5 ⁇ g of EC-SOD for 1 hour, and then treated with 1 ⁇ M of calcium channel A23187 and 50 nM of phorbol 12-myristate 13-acetate (PMA) to increase the concentration of calcium in the cells.
  • PMA phorbol 12-myristate 13-acetate
  • the human mast cells were washed with phosphate buffered saline (PBS), and then fixed with 4% paraformaldehyde for 10 minutes. After the cells were washed with phosphate buffered saline, they were stained with a solution of 0.1% toluidine blue in 1% acidic saline (pH 2.3) for 2-3 minutes. After the cells were washed again with water, the cells were dehydrated with 95% and 100% alcohol and fixed with xylene, and the number of degranulated cells among 100 stained cells was counted.
  • PBS phosphate buffered saline
  • FIG. 17A the number of degranulated cells in the human mast cell group (control) not treated with anything was 10, and the number of degranulated cells in the group (EC-SOD) treated with EC-SOD was 16.
  • the number of degranulated cells was increased five-fold to 55, but in the group (A+P+EC-SOD) where the cells were pretreated with EC-SOD and treated to increase the concentration of calcium in the cells, the number of degranulated cells was reduced again to 25. This indicates that the reduction of mast cell degranulation occurred due to the inhibition of reactive oxygen species caused by the action of EC-SOD.
  • a micrograph showing the degranulation of the mast is shown in FIG. 17B .
  • the recombinant EC-SOD protein of the present invention can be advantageously used to prevent or treat allergies.
  • an EC-SOD protein or a vector having a polynucleotide encoding the EC-SOD protein has the effect on inhibiting angiogenesis by inhibiting the expression of VEGF and MMP-9 which induce angiogenesis. Therefore, the EC-SOD protein or the vector may be useful for preventing or treating angiogenesis-mediated diseases. And the EC-SOD protein or the vector having a polynucleotide encoding said EC-SOD protein also has the effect on inhibiting over-differentiation of Th2 cells which cause allergic diseases, inhibiting transcription factor (NF- ⁇ B), and reducing degranulation of human mast cells. Accordingly, the EC-SOD protein or the vector may be useful for preventing or treating allergy diseases. In addition, the inventive method for preparing an EC-SOD protein may be used in preparing an EC-SOD protein having the activity on a large scale. Therefore, the inventive method may be useful industrially.

Abstract

The present invention relates to compositions for preventing or treating angiogenesis-mediated diseases or allergic diseases which contain, as an active ingredient, an EC-SOD protein or a vector having a polynucleotide encoding thereof. The EC-SOD protein or the vector having a polynucleotide encoding the EC-SOD protein has the effect on inhibiting angiogenesis by inhibiting the expression of VEGF and MMP-9 which induce angiogenesis. Therefore, the EC-SOD protein or the vector may be useful for preventing or treating angiogenesis-mediated diseases. And the EC-SOD protein or the vector having a polynucleotide encoding said EC-SOD protein also has the effect on inhibiting over-differentiation of Th2 cells which cause allergic diseases, inhibiting transcription factor (NF-κ B), and reducing degranulation of human mast cells. Accordingly, the EC-SOD protein or the vector may be useful for preventing or treating allergy diseases. In addition, the inventive method for preparing an EC-SOD protein may be used in preparing an EC-SOD protein having the activity on a large scale. Therefore, the inventive method may be useful industrially.

Description

    TECHNICAL FIELD
  • The present invention relates to a novel use of an EC-SOD protein and a method for preparing thereof. More particularly, the present invention relates to compositions for preventing or treating angiogenesis-mediated disease or allergic diseases which contain, an EC-SOD protein or a vector having a polynucleotide encoding said EC-SOD proteins an active ingredient.
  • BACKGROUND ART
  • Superoxide dismutases (SODs) function to protect cells by removing reactive oxygen species and enabling other antioxidant enzymes, and SODs known to date include Cu/Zn SOD (SOD 1) containing copper and zinc atoms, Mn SOD (SOD 2) containing a manganese atom, and extracellular superoxide dismutase (EC-SOD) present on the cell surface or in the extracellular fluid.
  • Particularly, EC-SOD contains copper and zinc atoms as in Cu/Zn SOD, but is characterized in that a heparin binding domain is present in the C-terminal end. Since EC-SOD has the heparin-binding domain, it is assumed that EC-SOD will function to protect cell membranes by binding to the cell membranes. According to literatures, it was known that EC-SOD plays a role in the body's defense mechanism in serums and extracellular matrices (Marklund et al, Biochem. J. 266, 213-219, 1990; Su et al., Am J Respir Cell Mol Biol., February 16(2), 162-70, 1997; et al., Thromb. Vasc. Bio. 18, 157-167, 1998). In addition, it was reported that the heparin-binding domain of EC-′SOD acts as a nuclear localization signal, so that it is located within the nuclei of thymus and testis cells so as to protect genomic DNA from oxidative stress and to regulate the DNA transcription sensitive to oxidation-reduction reaction (Ookawara T et al., BBRC, 296, 54-61, 2002). The present inventors reported in Korean Patent Registration No. 10-0676502 that EC-SOD has the effect of treating skin diseases such as psoriasis by removing reactive oxygen species in skin cells and inhibiting the over-proliferation of epidermal cells.
  • Meanwhile, angiogenesis is the process by which new capillary blood vessels are formed from preexisting vessels. Angiogenesis normally occurs only in some specific situations, including embryonic development, wound healing and the female reproductive cycle, and does not substantially occur in normal conditions. However, when angiogenesis is not regulated correctly, diseases such as cancer may be induced.
  • The angiogenic process consists of several steps that include stimulation of endothelial cell growth by tumor cytokine, vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF), degradation of extracellular matrix proteins by matrix metalloprotease (MMP), and migration of endothelial cells, which is mediated by membrane adhesion molecules, endothelial cell differentiation and tube formation (Bussolino, F. et al., Trends Biochem. Sci. 22:251-256, 1997; Kuwano, M. et al., Intern. Med. 40:565-572, 2001; Risau, W. Angiogenesis and endothelial cell function. Arzneimittelforschung 44:416-417, 1994). Thus, inhibition of these above processes has been proposed as a new therapeutic strategy for the treatment of angiogenesis-mediated diseases, including cancer and other human diseases. For this purpose, a variety of angiogenesis inhibitors have been developed. The inhibitors, which are natural or synthetic, include protease inhibitors, tyrosine kinase inhibitors, chemokines, interleukins, and proteolytic fragments of matrix proteins (Abedi, H. et. al., J. Biol. Chem. 272:15442-15451, 1997; Cao, Y., Int. J. Biochem. Cell Biol. 33:357-369, 2001; Fong, T. A et al., Cancer Res. 59:99-106, 1999; Kwon, H. J. et al., Acalycigorgia inermis. J. Microbiol. Biotechnol. 11:656-662, 2001). These antiangiogenic molecules affect various processes, including the inhibition of endothelial cell proliferation, migration, protease activity, and tubule formation, as well as the induction of apoptosis (Folkman, J. et. al., Semin. Cancer Biol. 3:89-96, 1992; Kishi, K. et al., Nippon Rinsho 58:1747-1762, 2000; Marme, D., Onkologie 1:1-5, 2001). The antiangiogenic function of many of these molecules has been well studied in vitro and in vivo, and some antiangiogenic drugs are currently being tested in clinical trials (Deplanque, G. et. al., Eur. J. Cancer 36:1713-1724, 2000; Liekens, et. al., Biochem. Pharmaco1.61:253-270, 2001; Mross, K., Drug Resist. Updat. 3: 223-235, 2000).
  • Reactive oxygen species have been reported to be major regulators of angiogenesis (Jolanta Grzenkowicz-Wydra, et. al., Mol Cell Biochem., 264(1-2):169-81, 2004), but the relationship between SODs, which remove reactive oxygen species, and angiogenesis, has not yet been sufficiently examined. It was recently reported that the overexpression of Cu/Zn SOD (SOD 1), having copper and zinc atoms, among SODs, leads to the stimulation of angiogenesis, and thus the inhibition of SOD1 leads to the inhibition of angiogenesis (Jolanta Grzenkowicz-Wydra, et. al., Mol Cell Biochem., 264(1-2):169-81, 2004). In addition, it was reported that ATN-224, which is an inhibitor of SOD1, inhibits angiogenesis, and thus can be used as an anticancer agent, and it is in a phase II trial (Juarez et al. Clin Cancer Res., 12: 4974-4982, 2006). However, there is no report on the relationship between EC-SOD and angiogenesis.
  • Meanwhile, with the development of industry, allergy-causing factors have increased due to environmental pollution, an increase in new synthetic substances and a change in residential environments, and thus the number of people suffering from asthma and allergies has gradually increased. The function of the body to show specific resistance against foreign matter, which has invaded the body, is referred to as immunity. An allergy is a hypersensitive immune reaction, and typical allergic diseases include atopic dermatitis, bronchial asthma and pollenosis. Clinical symptoms resulting from allergic reactions are broadly classified into specific immune reactions at an early stage, and inflammatory reactions at a late stage. These immune reactions are mostly mediated by mast cells, which are known to be widely distributed in the systemic organs, including the skin, the respiratory organ, the gastrointestinal mucosa, the brain, around lymphatic vessels and around blood vessels and to cause allergic reactions. Mediators known to activate mast cells include an IgE antibody (Fcε RI) bound to a high-affinity immunoglobulin E (IgE) receptor (Fcε RI) located on the cellular membrane, compound 48/80 and the like. The activation mechanism of mast cells by IgE antibody is as follows.
  • When an IgE antibody bound to an IgE receptor forms a bridge with an antigen, substances, such as histamine, chondroitin sulfate, heparin and protease, stored in the mast cell granules, are released through the action of phospholipase C, protein phosphatase C and calcium ions, thus mediating the early reaction stage. Among the chemical substances, including histamine, cause clinical symptoms occurring in the early allergic reaction, histamine accounts for the largest amount. In other words, in the early allergic reaction, the expression of histamine is predominant, and clinical symptoms resulting therefrom include vasodilation, edema and the like.
  • Currently, antihistamine drugs or steroidal drugs are frequently used to treat allergies. However, these drugs mostly have temporary effects, and in many cases, cause serious side effects. Accordingly, there is an urgent need to develop a novel substance, which has the effect of preventing and treating rapidly increasing allergic diseases, and at the same time, has reduced side effects and lasting effects. However, whether EC-SOD has the effect of treating allergic diseases is not yet known.
  • Also, even if the EC-SOD has various activities, when a method for producing the EC-SOD in large amounts is not developed, there is a problem in that the EC-SOD cannot be efficiently used in industry. However, mass-production methods for proteins, developed to date, comprise transforming host cells, such as E. coli cells, with expression vectors, and inducing the expression of proteins in the transformed cells, and the mass production of proteins has problems in that it often causes the formation of inclusion bodies, and thus the original activities of the proteins are eliminated or reduced.
  • The present inventors have conducted studies on the EC-SOD protein, and as a result, have found that the EC-SOD shows an activity of inhibiting diseases caused by angiogenesis or allergic diseases. On the basis of this finding, the present inventors have developed a method for preparing the EC-SOD protein having the said activity, thereby completing the present invention.
  • Disclosure Technical Problem
  • Therefore, in order to achieve the object, the present invention provides a composition for preventing or treating angiogenesis-mediated diseases comprising an EC-SOD protein, as an active ingredient.
  • In another aspect, the present invention provides a composition for preventing or treating angiogenesis-mediated diseases comprising a vector having a polynucleotide encoding an EC-SOD protein, as an active ingredient.
  • In still another aspect, the present invention provides a composition for preventing or treating allergic diseases comprising an EC-SOD protein, as an active ingredient.
  • In still another aspect, the present invention provides a composition for preventing or treating allergic diseases comprising a vector having a polynucleotide encoding an EC-SOD protein, as an active ingredient.
  • In still another aspect, the present invention provides a method of preparing an EC-SOD protein.
  • Technical Solution
  • To achieve the above objects, in one aspect, the present invention provides a composition for preventing or treating angiogenesis-mediated diseases comprising an EC-SOD protein, as an active ingredient.
  • In another aspect, the present invention provides a composition for preventing or treating angiogenesis-mediated diseases comprising a vector having a polynucleotide encoding an EC-SOD protein, as an active ingredient.
  • In still another aspect, the present invention provides a composition for preventing or treating allergic diseases comprising an EC-SOD protein, as an active ingredient.
  • In still another aspect, the present invention provides a composition for preventing or treating allergic diseases comprising a vector having a polynucleotide encoding an EC-SOD protein, as an active ingredient.
  • In yet another aspect, the present invention provides a method for preparing an EC-SOD protein, the method comprising the steps of: (a) cloning into an expression vector a polynucleotide encoding said EC-SOD protein (b) introducing the expression vector into host cells to transform the host cells (c) culturing the transformed host cells to express the EC-SOD protein in the cells (d) collecting the inclusion body of the expressed EC-SOD; and (e) dissolving the inclusion body and inducing the refolding of the inclusion body. Hereinafter, the present invention will be described in further detail.
  • As used herein, the term “expression vector” describes a vector capable of expressing a target protein or target RNA in a suitable host cell, and refers to a genetic construct comprising essential regulatory elements to which a gene insert is operably linked.
  • By the term “operably linked”, as used herein, it is meant that there is a functional linkage between a nucleotide expression control sequence and a nucleotide sequence encoding a target protein, in such a manner as to perform general functions. For example, a promoter is linked with a nucleotide sequence coding for a protein or RNA, such that it can affect the expression of the coding nucleotide sequence. The operable linkage to a recombinant vector may be prepared using a genetic recombinant technique that is well known in the art, and site-specific DNA cleavage and ligation may be carried out using enzymes that are generally known in the art.
  • The present invention provides a composition for preventing or treating angiogenic diseases, which contains an EC-SOD protein as an active ingredient.
  • The EC-SOD protein preferably refers to, but is not limited to, a natural or recombinant EC-SOD protein. The natural EC-SOD protein is preferably derived from, but is not limited to, mice or humans, and more preferably from humans. The natural EC-SOD protein preferably has, but is not limited to, amino acid sequences of SEQ ID NO: 3 or SEQ ID NO: 5, and most preferably an amino acid sequence of SEQ ID NO: 3. Meanwhile, the recombinant EC-SOD protein is not specifically limited, as long as it is prepared from the natural EC-SOD protein so as to maintain the activity thereof. Preferably the recombinant EC-SOD protein has an amino acid sequence.
  • In another aspect, the present invention provides a composition for preventing or treating angiogenesis-mediated diseases, which contains, as an active ingredient, a vector having a polynucleotide encoding the EC-SOD protein.
  • The polynucleotide encoding the EC-SOD protein means a polynucleotide capable of encoding the natural EC-SOD protein or the recombinant EC-SOD protein and preferably has a base sequence selected from the group consisting of, but is not limited to, SEQ ID NO: 2, SEQ ID NO: 4 and SEQ ID NO: 6.
  • In one Example of the present invention, mice overexpressing mouse EC-SOD were prepared according to a known method (see Test Example 1), and were irradiated with UVB, and then the morphological change in the tissue thereof was compared with that of wild-type mice. As a result, it was observed that, in the wild-type mice, angiogenesis was induced by UVB irradiation, but in the EC-SOD-overexpressed mice, angiogenesis was significantly inhibited by UVB irradiation (see Example 2-1).
  • In other Examples of the present invention, the expression levels of vascular endothelial growth factor (VEGF) and MMP-9 in the EC-SOD-overexpressing mice by UV light irradiation were examined using Western blot (Example 2-2), an immunohistochemical method (Example 2-3) and a zymography method (Example 2-4). As a result, it could be observed that, in the wild-type mice, the expressions of VEGF and MMP-9 were increased due to UV light irradiation, but in the EC-SOD-overexpressed mice, the expressions of VEGF and MMP-9 were significantly inhibited. The above experimental results suggest that EC-SOD inhibits the expression of VEGF inducing angiogenesis is and inhibits the expression of MMP-9 regulating angiogenesis, thus inhibiting angiogenesis.
  • In still other Examples of the present invention, recombinant EC-SOD, from which a heparin-binding domain has been removed, was expressed in E. coli, and then isolated from the E. coli strain, thus producing a refolded and purified EC-SOD protein (see Example 1), and whether the produced protein would inhibit angiogenesis was examined (see Example 3). As a result, it could be seen that the expression of VEGF was induced by UV light irradiation, but treatment with the recombinant EC-SOD protein resulted in inhibition of the expression of VEGF. Accordingly, the EC-SOD protein or the vector having inserted therein the polynucleotide encoding the EC-SOD protein can be effectively used to prevent and treat angiogenesis-mediated diseases. The angiogenesis-mediated diseases include all diseases induced by the expression of VEGF (vascular endothelial growth factor) and the expression of MMP (matrix metalloproteinase)-9, and preferred examples thereof may include, are not limited to, cancer, diabetes, rheumatoid arthritis, arteriosclerosis, hemangioma, angiofibroma, diabetic retinopathy, retinopathy of prematurity, neovascular glaucoma, corneal diseases caused by angiogenesis, degenerative spots, macular degeneration, pterygium, retinal degeneration, retrolental fibroplasia, Granular conjunctivitis, telangiectasia, pyogenic granuloma and acne (Ophthalmol 102, 1261-1262, 1995; J Am Acad Derm 34(3):486-497, 1996; Circultion 93(4):632-682, 1996; Cell 86: 353-364, 1996).
  • In another aspect, the present invention provides a composition for preventing or treating allergic diseases, which contains the EC-SOD protein as an active ingredient.
  • As described above, the EC-SOD protein refers to, but is not limited to, a natural or recombinant EC-SOD protein. The natural EC-SOD protein is preferably derived from, but is not limited to, mice or humans, and more preferably from humans. The natural EC-SOD protein preferably has an amino acid sequence of, but is not limited to, SEQ ID NO: 3 or SEQ ID NO: 5, and more preferably SEQ ID NO: 3. Meanwhile, the recombinant EC-SOD protein is not specifically limited, as long as it is prepared from the natural EC-SOD protein using recombinant technology and maintains the activity thereof. Preferably, the recombinant EC-SOD protein has an amino acid sequence of SEQ ID NO: 1.
  • In still another aspect, the present invention provides a composition for preventing or treating allergic diseases, which contains, as an active ingredient, a vector having inserted therein a polynucleotide encoding the EC-SOD protein.
  • As described above, the polynucleotide encoding the EC-SOD protein means a polynucleotide capable of encoding the natural EC-SOD protein or the recombinant EC-SOD protein and has preferably a base sequence selected from, but is not limited to, the group consisting of SEQ ID NO: 2, SEQ ID NO: 4 and SEQ ID NO: 6.
  • In one Example of the present invention, the effect of the recombinant EC-SOD protein on the proliferation and differentiation of T-cells was analyzed. As a result, it could be seen that the recombinant EC-SOD protein induced the formation of the Th1 cytokine IFN-γ in T cells and inhibited the formation of the Th2 cytokine IL-4. This suggests that the recombinant EC-SOD protein has an effect on the differentiation of T cells, and that it blocks the over-differentiation of Th2 cells causing allergic diseases and inducing the differentiation of Th2 cells into Th1 cells, and thus it can be advantageously used to prevent or treat allergic diseases (see Example 4).
  • In another Example of the present invention, the effect of the natural human protein EC-SOD on the differentiation of T cells was examined. As a result, it could be seen that the EC-SOD protein also blocked the over-differentiation of Th2 cells causing allergic diseases and induced the differentiation into Th1 cells (see Example 5).
  • In still another Example of the present invention, whether the recombinant EC-SOD protein has the effect of inhibiting transcriptional factor activity was examined. As a result, it could be seen that the recombinant EC-SOD protein could inhibit the abnormal activity of NF-κ B (see Example 6) and had the effect of reducing the degranulation of human mast cells. Accordingly, in view of the fact that mediators, such as histamine, stored in mast cells, are released due to the degranulation of the mast cells to cause inflammatory reactions such as allergy, it can be seen that the recombinant EC-SOD protein of the present invention can be advantageously used to prevent or treat allergies (see Example 7).
  • Thus, the EC-SOD protein or the vector having inserted therein the polynucleotide encoding the protein will be useful in a composition for preventing or treating allergic disease. The allergy is preferably selected from the group consisting of allergic asthma, allergic rhinitis, allergic otitis media, anaphylatic shock, allergic skin disease, atopic dermatitis, psoriasis, contact allergic dermatitis and urticaria.
  • The said EC-SOD proteins which are comprised in the inventive composition may include proteins having equivalent physiological activities. The said proteins with equivalent physiological activities may include functional equivalents of EC-SOD proteins as well as functional derivatives.
  • The term “functional equivalents” in the invention refer to polypeptides which have equivalent physiological activities with the EC-SOD of SEQ ID NO: 3.
  • The term “equivalent physiological activities” refer to the activities of inhibiting angiogenesis or allergy. The said functional equivalents may polypeptides which show at least 70% amino acid sequence homology with the amino acid sequence of SEQ ID NO: 3, preferably at least 80%, and more preferably 90%. The term functional equivalents refer to peptides which have at least 70% amino acid sequence homology (i.e., identity) with the peptides produced by as a result of addition, substitution or deletion of SEQ ID NO: 3, preferably at least 80%, and more preferably at least 90%, for example, 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 9.0%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% and 100% and exhibit substantially identical physiological activity to the polypeptide of SEQ ID NO: 3. Sequence identity or homology, is defined herein as the percentage of amino acid residues in the candidate sequence that are identical with amino acid sequence of SEQ ID NO: 3, after aligning the sequences and introducing gaps. If necessary, to achieve the maximum percent sequence identity, any conservative substitutions is not considered as part of the sequence identity. None of N-terminal, C-terminal, or internal extensions, deletions, or insertions into the amino acid sequence of SEQ ID NO: 3 shall be constructed as affecting sequence identity or homology. Thus, sequence identity can be determined by standard methods that are commonly used to compare the similarity, in position of the amino acids of two polypeptides. Using a computer program such as BLAST or FASTA, two polypeptides are aligned for optimal matching of their respective amino acids (either along the full length of one or both sequences or along a predetermined portion of one or both sequences). The programs provide a default opening penalty and a default gap penalty, and a scoring matrix such as PAM 250 (a standard scoring matrix; Dayhoff et al., in Atlas of Protein Sequence and Structure, vol 5, supp. 3, 1978). For example, the percent identity can be calculated as: the total number of identical matches multiplied by 100 and then divided by sum of the length of a longer sequence within the matched span and the number of gaps introduced into longer sequences in order to align the two sequences. The scope of the functional equivalents as used herein encompasses polypeptide derivatives obtained by modifying a part of the chemical structure of the polypeptide while maintaining the basic framework and physiological activity of the polypeptide. For example, this includes structural modifications for altering the stability, storage, volatility or solubility of the polypeptide.
  • In addition, polynucleotide sequences of the inventive compositions may comprise DNA, cDNA, and RNA sequences. Preferably, the said polynucleotide may comprise the sequence selected from the group consisting of SEQ ID NO: 2, 4 and 6. The inventive compositions may comprise, as an active ingredient, plasmid having the said polynucleotide sequence thereto, or expression vectors such as viral vectors. The inventive expression vector may comprise expression control sequence by which EC-SOD protein and cDNA of the EC-SOD are expressed. The said polynucleotides are isolated from nature or manufactured by the methods which are known in the art.
  • The said expression control sequence refer to a DNA sequence essential to expression of coding sequence which is operatively linked within a certain host cell. The said expression control sequence may comprise promoters for transcription, certain operator sequences for controlling of the said transcription, the sequences encoding relevant mRNA ribosomal binding site, and the sequences terminating transcription and translation. For example, the control sequences for prokaryotes may comprise promoter, operator, and ribosomal binding site. The control sequences may comprise promoter, polyadenylated signal and enhancer.
  • After an expression vector was inserted, it could be introduced into a target cell as a phenotype by infection, transfection and transduction which are well known method in the art.
  • Gene introducing method by using plasmid expression vector is the method which introduce plasmid DNA directly into a mammalian cell, and FDA has approved to use for human (Nabel, E. G., et al., Science, 249:1285-1288, 1990). Unlike viral vector, a plasmid DNA has advantage in respect of even purification. The acceptable expression plasmids of the invention may comprise mammalian expression plasmids which are used in the art. For example, but not limited thereto, pRK5(European Patent No. 307,247), pSV16B (PCT Publication No. WO91/08291) and pVL1392(PharMingen). The plasmid expression vector which comprise the said nucleic acid could be introduced to a target cell by, but not limited thereto, transient transfection, microinjection, transduction, cell fusion, calcium phosphate precipitation, liposome-mediated transfection, DEAE Dextran-mediated transfection, polybrene-mediated transfection, electroporation, gene gun, and other methods which are well known in the art (Wu et al., J. Bio. Chem., 267:963-967, 1992; Wu and Wu, J. Bio. Chem., 263:14621-14624, 1988).
  • In addition, the viral vectors which contain the said polynucleotide sequence, may comprise, but not limited thereto, retrovirus, adenovirus, herpes virus, avipox virus, and lentivirus and the like. All of the viral genes of the said retroviral vectors were deleted or modified, and consequently non-viral proteins of the said vectors were produced by the infected cells. The main advantages of the retroviral vectors for gene therapy are to transfer large amount of genes into cloned cells, to integrate genes specifically which are transferred to cellular DNA, and to prevent additional infection after gene transformation (Miller, A. D., Nature, 357:455-460, 1992). The retroviral vectors which are approved by the FDA is manufactured by using PA317 amphotrophic retroviral packaging cell (Miller, A. D. and Buttimore, C., Molec. Cell Biol., 6:2895-2902, 1986). For the non-retroviral vectors, there is the said adenovirus (Rosenfeld et al., Cell, 68:143-155, 1992; Jaffe et al., Nature Genetics, 1:372-378, 1992; Lemarchand et al., Proc. Natl. Acad. Sci. USA, 89:6482-6486, 1992). The main advantages of the adenovirus are to transfer large molecular DNA fragment (36 kb), and to transfect non-cloned cells with very high titer. In addition, herpes viruses could be used in gene therapy for human (Wolfe, J. H., et al., Nature Genetics, 1:379-384, 1992). The lentivirus is a kind of retrovirus, and it is developed from modification of HIV backbone since late 1990's. Unlike to existing retrovirus carriers, the lentivirus is more effective in non-divisional cells as well as divisional cells since its activity does not influenced by cell cycle. Accordingly, even in the cells which divide slowly, such as hematopoietic stem cells, the lentivirus is more effective than other viral carriers, and therefore the studies of the lentivirus as a gene transfering carrier are progressed in the field of gene therapy by using hematopoietic stem cells, and keratinocytes. In addition, relevant viral vector of well known in the art may be used in the invention.
  • As used herein, the term “pharmaceutically acceptable” means what is physiologically acceptable and, when administered to human, generally does not cause allergic reactions, such as gastrointestinal disorder and dizziness, or similar reactions thereto.
  • A pharmaceutically acceptable carrier, for example carriers for the parenteral or oral preparations may be included. The carriers for the parenteral preparations may comprise lactose, starch, cellulose derivatives, magnesium stearate, stearic acid. In addition, the carriers for the parenteral preparations may comprise water, oil, saline, aqueous glucose and glycol, and stabilizers and preservatives. The examples of the stabilizers may be antioxidant such as sodium hydrogen sulfite, sodium sulfite, and ascorbic acid. The examples of the preservatives may be benzalkonium chloride, methyl- or prophyl-paraben, and chlorobutanol.
  • The list of pharmaceutically acceptable carriers are disclosed in Remington's Pharmaceutical Sciences, 19th ed., Mack Publishing Company, Easton, Pa., 1995.
  • The inventive protein or a vector having inserted therein a polynucleotide encoding said protein may be prepared by mixing with the said pharmaceutically acceptable carriers into various oral or parenteral preparations according to any method known in the art. Also, the inventive peptide may be administered by various routes according to any method known in the art. Namely, it may be administered by oral or parenteral routes. The injectable preparations such as isotonic solution or suspension and ointments are desirable for the parenteral preparations. The injectable preparations may be prepared by using relevant dispersants, wetting agents, or suspending agents according to any method known in the art. For example, the compositions may be formulated into injectable compositions by dissolving components into saline or buffers. Furthermore, for oral administration, which are not limited thereto, the inventive composition can be formulated in the form of powder, granule, tablets, pills and capsules, and the like. These preparations may also comprise diluents (e.g., lactose, dextrose, sucrose, mannitol, sorbitol, cellulose and/or glycine), lubricants (e.g., silica, talc, stearic acid and a magnesium or calcium salt thereof, and/or polyethylene glycol) in addition to the active ingredient. Among various preparations, tablets may also comprise binders, such as magnesium aluminum silicate, starch pastes, gelatin, tragacanth, methylcellulose, sodium carboxymethylcellulose and/or polyvinylpyrrolidone, and, if desired, may further comprise disintegrating agents, such as starches, agar or alginic acid or a sodium salt thereof, heterogenous mixtures and/or absorbents, colorants, flavors and sweeteners. The said composition may be prepared by mixing, granulization or coating.
  • The inventive pharmaceutical composition may be administered by various routes, but not limited thereto, according to any biotechnological method, any method known in the art, namely, it may be administered by oral, intravenous, intramuscular, intraarterial, intramarrow, subdural, intracardiac, intracUtaneous, subcutaneous, intraperitoneal, intranasal, gastrointestinal tracts, parenteral, sublingual or rectum.
  • Preferably, it may be administered parenterally, by subcutaneous, intravenous, intramuscular, instra joint, intrabursal, intrasternum, subdural, intralesional, and intracranium, with injection or infusion. For example, an injectable form of the inventive pharmaceutical composition may be administered with a given amount of the formulation by injecting into a subcutaneous layer with a 4-6 mm syringe or lightly pricking the skin with 30-gauge injection needle, namely, the Messo therapy. In addition, an ointment form of the inventive pharmaceutical composition may be administered by a method of applying directly to the skin. The said ‘intracutaneous’ refers to transfer effective amount of an active ingredient which is comprised in the inventive composition by administered into the skin. In particular, It is preferred that the composition whose active ingredient is the EC-SOD protein of the invention should be administered directly to the skin. In addition, the inventive composition may be administered by biotechnological method related to protein transfer.
  • The inventive compositions may be administered to patients with the amount which is effective for preventing disease. Generally, the effective amount of the inventive composition is about 0.0001 to 100 mg/kg body weight/day. Preferably 0.01 to 1 mg/kg body weight/day. It may be suitably determined by considering various factors, such as age, body weight, health condition, sex, disease severity, diet and excretion of a subject in need of treatment, as well as administration time and administration route.
  • In addition, the present invention provides the method for preparing an EC-SOD protein, which comprises the steps of:
  • (a) cloning the polynucleotide encoding the EC-SOD protein into the expression vector;
  • (b) transforming a host cells by introducing the expression vector to the host cells;
  • (c) expressing the EC-SOD protein by culturing the transformed host cells;
  • (d) collecting the inclusion body of the expressed EC-SOD protein; and
  • (e) dissolving the inclusion body, and inducing refolding of the EC-SOD protein.
  • The step (a) of cloning the polynucleotide encoding the EC-SOD protein into the expression vector can be carried out using a conventional recombinant DNA technique known in the art. The polynucleotide encoding the EC-SOD means a polynucleotide capable of encoding the natural EC-SOD protein or the recombinant EC-SOD protein and preferably has a base sequence selected from, but is not limited to, the group consisting of SEQ ID NO: 2, SEQ ID NO: 4 and SEQ ID NO: 6. The polynucleotide can be constructed by performing PCR amplification using suitable primers. Alternatively, the DNA sequence may also be synthesized by a standard method known in the art, for example, using an automatic DNA synthesizer (commercially available from Biosearch or Applied Biosystems). The expression vector may be a conventional expression vector used in the art and can be suitably selected depending on host cells. For example, when the host cells are E. coli cells, a pET28a vector (Novagene, USA) may be used. Because the preparation method of the present invention aims to express and produce EC-SOD in large amounts, any expression vector may be suitably selected and used, as long as it can achieve this purpose.
  • The inventive vectors may comprise, but not limited thereto, plasmid vector, cosmid vector, bacteriophasge vector, and viral vector and the like.
  • The suitable expression vectors may comprise expression control sequences such as promoter, operator, initiation codon, stop codon, polyadenyated signal and enhancer (stimulating gene) as well as membrane targeting, signal sequence for secretion, leader sequences, and may be manufactured according to a purpose. A promoter of the inventive promoter may be constitutive or inducible. In addition, the inventive expression vector may comprise selective markers to select the host cells which contain the vector, and if the expression vector is replicable, it may comprise origin of replication.
  • The step (b) transforming a host cells by introducing the expression vector to the host cells may comprise any method of introducing nucleic acid to the host cells, and may be performed by transformation techniques which are well known in the art. Preferably, the techniques may comprise, but not limited thereto, microprojectile bombardment, electroporation, CaPO4 precipitation, CaCl2 precipitation, PEG-mediated fusion, microinjection, liposome-mediated method an the like.
  • In the above cases, the host cells may be prokaryote or eukaryote. However, preferably, the host cells may comprise, but not limited thereto, prokaryotic host cells such as Escherichia coli, Bacillus subtilis, Streptomyces, Pseudomonas, Proteus mirabilis, and Staphylococcus, and the lower eukaryotes such as the fungi (for example, Aspergillus), yeasts (for example, Pichia pastoris, Saccharomyces cerevisiae, chizosaccharomyces, Neurospora crassa), higher eukaryotic cells originated from insect cells, plant cells, mammalian cells. More preferably, this invention may use Escherichia coli as a host cell.
  • The step (c) of expressing the EC-SOD protein by culturing the transformed host cells comprises culturing the host cells in a suitable medium and conditions so as to express the EC-SOD protein in the host cells. The method of expressing the transformed cells to express the protein in the cells is known in the art. For example, the expression of the protein can be induced by inoculating the transformed cells in a suitable medium allowing the cells to grow, preculturing the cells, inoculating the precultured cells in a medium, and then culturing the cells in suitable conditions.
  • Collecting the inclusion body in the step (d) can be carried out using a known method for isolating and collecting insoluble inclusion bodies. For example, the inclusion body can be collected from the cell precipitate by centrifugation.
  • The step (e) of dissolving the inclusion body and inducing the refolding of the inclusion body can be carried out using any conventional method known in the art. For example, it can be carried out by dissolving the inclusion body in a buffer solution containing a protein denaturant, adsorbing the solution onto a metal affinity resin column, and inducing the refolding of the inclusion body while reducing the concentration of the denaturant.
  • For example, dissolving the inclusion body and inducing the refolding of the inclusion body can be performed by dissolving the inclusion body in a Tris buffer solution (8 M urea, 50 mM Tris-Cl, 100 mM NaCl) containing 8M urea as a denaturant, adsorbing the solution onto a metal affinity resin-packed column, and slowly removing the urea while applying a concentration gradient to the column using a Tris buffer containing 1 M urea, thus inducing the refolding of the inclusion body. Then, a pure refolded protein can be obtained by allowing either an elution buffer solution showing strong adsorption onto the column, or an elution buffer solution having a low pH value (generally less than 4), to flow from the column top downward so as to flow out through the column bottom. For example, the pure refolded protein can be obtained by allowing an imidazole (generally 1M)-containing elution buffer solution having strong adsorption onto a metal affinity chromatography column, to flow from the column top downward so as to flow out through the column bottom. In order to increase the yield of the protein, it is preferable to feed the elution buffer solution into the column in such a manner that it has a concentration gradually increasing with the passage of time, that is, it has a concentration gradient.
  • The inventive method for preparing the EC-SOD protein may additionally comprise a step (f) of separating and purifying the EC-SOD by gel-filtration column. Then, various isolation and purification processes, including additional purification, concentration and solvent exchange, may be carried out depending on the intended use. For example, the resulting EC-SOD protein can be subjected to techniques, such as salting-out (e.g., ammonium sulfate precipitation and sodium phosphate precipitation), solvent precipitation (a protein fractionation precipitation method using acetone, ethanol, or the like), dialysis, gel filtration, ion exchange, ion exchange chromatography, reverse phase chromatography and affinity chromatography, alone or in combination, to purify the EC-SOD protein.
  • the step of (f) separating and purifying the EC-SOD by gel-filtration column.
  • The inventive standard recombinant DNA and molecular cloning techniques are well known in the art and described in the following references (Sambrook, J., Fritsch, E. F. and Maniatis, T., Molecular Cloning: A Laboratory Manual, 2nd ed., Cold Spring Harbor Laboratory: Cold Spring Harbor, N.Y. (1989); by Silhavy, T. J., Bennan, M. L. and Enquist, L. W., Experiments with Gene Fusions, Cold Spring Harbor Laboratory: Cold Spring Harbor, N.Y. (1984); and by Ausubel, F. M. et al., Current Protocols in Molecular Biology, published by Greene Publishing Assoc. and Wiley-Interscience (1987)).
  • As described above, a vector having inserted therein a polynucleotide encoding said EC-SOD protein could be administered to a required individual with effective amount, and prevent or treat diseases caused by angiogenesis or allergic diseases. In addition, the said vector may be used for preparation a therapeutic agent for angiogenesis-mediated diseases or allergic diseases.
  • Advantageous Effects
  • As mentioned above, an EC-SOD protein or a vector having a polynucleotide encoding the EC-SOD protein has the effect on inhibiting angiogenesis by inhibiting the expression of VEGF and MMP-9 which induce angiogenesis. Therefore, the EC-SOD protein or the vector may be useful for preventing or treating angiogenesis-mediated diseases. And the EC-SOD protein or the vector having a polynucleotide encoding said EC-SOD protein also has the effect on inhibiting over-differentiation of Th2 cells which cause allergic diseases, inhibiting transcription factor (NF-κ B), and reducing degranulation of human mast cells. Accordingly, the EC-SOD protein or the vector may be useful for preventing or treating allergy diseases. In addition, the inventive method for preparing an EC-SOD protein may be used in preparing an EC-SOD protein having the activity on a large scale. Therefore, the inventive method may be useful industrially.
  • DESCRIPTION OF DRAWINGS
  • FIG. 1 is a graph showing the result of Fast Protein Purification Liquid Chromatography of the inventive recombinant EC-SOD protein linear equilibrium region, B: sample loading region, C: column washing region, D: refolding region, E: refolding equilibrium region F: elusion region, G: column washing region, X axis: time and volume (hr:min:sec, mL), Y axis: UV density (Unit)).
  • FIG. 2 is a photograph of Coomassie-blue protein staining showing the SDS-PAGE result of the inventive recombinant EC-SOD protein which is refolded and purified. (M: molecular size marker, A: inclusion body which is diluted with 8M urea buffer (before refolding purification), B: inclusion body impurity which is not attached in the column and eluted, C: eluted EC-SOD after refolding purification, D: concentrated EC-SOD by ultrafiltration after desalting purification).
  • FIG. 3 is a western blot result of recombinant EC-SOD protein by using anti-EC-SOD protein antibody (SM: size marker, 1: control (HaCaT human EC-SOD), 2: inventive recombinant EC-SOD protein).
  • FIG. 4 is a graph of EC-SOD activity during purification step (209 FT: eluted but not attached sample during purification 209 E1: primary purified recombinant EC-SOD protein refolded by metal affinity chromatography, 209 E2: isolated and purified recombinant EC-SOD protein by gel-filtration column).
  • FIG. 5 is a photograph showing inhibition activity of recombinant EC-SOD protein from ROS caused by UVB in the HaCaT cell (UVB: UVB irradiation only, EC-SOD+UVB: UVB irradiation and EC-SOD treatment).
  • FIG. 6 is a photograph showing morphological changes in skins of mice by UV irradiation (A: wild type mouse, B: EC-SOD overexpressed mouse, C: UV irradiated wild type mouse, D:UV irradiated EC-SOD overexpressed mouse).
  • FIG. 7 is a western blot result of VEGF expression in UV irradiated mouse tissue. Actin was used as loading control (WT: wild type mouse, TG: EC-SOD overexpressed mouse).
  • FIG. 8 is a photograph showing VEGF expression of UV irradiated mouse by using immunohistological method (A: wild type mouse, B: EC-SOD overexpressed mouse, C:UV irradiated wild type mouse, D: UV irradiated EC-SOD overexpressed mouse).
  • FIG. 9 is a zymography of MMP-9 expression of UV irradiated mouse (Lane 1: wild type mouse, Lane 2: EC-SOD overexpressed mouse, Lane 3:UV irradiated wild type mouse, Lane 4: UV irradiated EC-SOD overexpressed mouse).
  • FIG. 10 is a photograph showing VEGF expression in UV irradiated keratinocyte HaCaT cells depending on treating the inventive recombinant EC-SOD using immunohistological method (A: control, B: UVB irradiated, C: recombinant EC-SOD treatment+UVB irradiation, D: recombinant EC-SOD treatment).
  • FIG. 11 is a result confirming whether treatment of recombinant EC-SOD protein has the effect on T-cell proliferation (Neg: negative control)
  • FIG. 12 is a result confirming by identifying the secretion level of IL-4 whether treatment of recombinant EC-SOD protein has the effect on T-cell differentiation (neg.: negative control, −: treated with antigen (Ova peptide) only, p: treated with antigen (Ova peptide) and PBS containing EC-SOD together, NB: treated with antigen (Ova peptide) and Sodium borate containing EC-SOD together).
  • FIG. 13 is a result confirming by identifying the secretion level of IL-4 and IFN-γ whether treatment of recombinant EC-SOD protein has the effect on T-cell differentiation (neg.: negative control, −: treated with antigen (Ova peptide) only).
  • FIG. 14 is a result showing the effect of EC-SOD on cell proliferation and cytokine generation when an Ova protein and an immune adjuvant CFA (Compleat Freund's Adjuvant) was administered into human EC-SOD protein-overexpressed mice.
  • FIG. 15 is a result showing the effect of EC-SOD on cell proliferation and cytokine generation when an Ova protein and an immune adjuvant Alum (Aluminum Hydroxide) was administered into human EC-SOD protein-overexpressed mice.
  • FIG. 16 is a result showing the effect of recombinant EC-SOD protein on NF-κ B inhibition (Control: EC-SOD transformed HaCaT cell without UVB irradiation, UVB:HaCaT cells with UVB irradiation (100 J/m2), EC-SOD/UVB:HaCaT cells treated with recombinant EC-SOD protein and UVB irradiation (100 J/m2).
  • FIG. 17 is photographs showing mast cell degranulation inhibition activity of recombinant EC-SOD protein (A) and its microscopic picture (B) (Con: human mast cells group without treatment, EC-SOD: EC-SOD treated group, A+P: group having high cellular concentration of calcium, A+P+EC+SOD: group pretreated with EC-SOD and having high cellular concentration of calcium).
  • MODE FOR INVENTION
  • Hereinafter, the present invention will be described in detail by examples. It is to be understood, however, that these examples are for illustrative purpose only and are not constructed to limit the scope of the present invention.
  • Test Example 1 Preparation of Mice Overexpressing EC-SOD
  • First, mice overexpressing mouse EC-SOD were prepared in the following manner. Meanwhile, the base sequence and amino acid sequence of the mouse EC-SOD are set forth in SEQ ID NO: 5 and SEQ ID NO: 6, respectively.
  • As wild-type BDF1 mice, 8-10-week-old BDF1 mice (Central Laboratory Animal Inc., Korea) were used. The EC-SOD-overexpressing mice were prepared according to a known method using the 8-10-week-old BDF1 mice (H. Y. Ha. et. al., Biochem Biophys Res Commun, 348: 450-458, 2006). In the experiment, the animals were used under the approval of the Catholic Institute, The Catholic University of Korea.
  • Specifically, hybrid mice (C57BL/6×CBA F1) were purchased from the Korea Research Institute of Bioscience and Biotechnology. To induce superovulation, 5 IU of each of pregnant mare's serum gonadotropin (PMSG) and human chorionic gonadotropin (hCG) were injected intraperitoneally into the female mice at an interval of 48 hours, and then mated with the same line of male mice at 1:1. Next day morning, the presence or absence of a vaginal plug in the animals was observed, and only an individual having the vaginal plug was butchered, from which an oviduct was taken. From the oviduct ampulla, embryos were collected. As a culture medium, 0.4% BSA-containing M16 culture medium was used.
  • In order to obtain mice where the skin tissue-overexpression of EC-SOD had been induced, the mouse EC-SOD cDNA in a pCR11TOPO vector (provided by professor Suh Jun-Gyo, College of Medicine, Hallym University) containing mouse EC-SOD cDNA (SEQ ID NO: 6) was digested with restriction enzymes and inserted into a pBS KS vector containing a human keratin K14 promoter. Also, the vector was inserted with chicken ovalbumin poly (A) to facilitate expression in vitro. In order to inject DNA into the embryos, the expression vector was cut with restriction enzymes Hind and XhoI. The DNA cut with the restriction enzymes was electrophoresed on 1% agarose gel, recovered by dialysis, and then isolated and purified with phenol-chloroform. It was dialyzed in 10 mM Tris (pH 7.4)/0.2 mM EDTA) to a final concentration of 4 ng/l. The foreign gene-containing DNA solution was injected into the pronuclei of one-cell-stage embryos. The embryos injected with the foreign gene were cultured to the two-cell stage, and healthy embryos were selected from the cultured embryos. Female ICR mice in heat were mated with male mice, and next day, the presence or absence of a vaginal plug was observed, and a mouse having the vaginal plug was used as a surrogate mother. About 15-20 embryos well grown to the two-cell stage were collected and transplanted into the oviduct ampulla of the pseudopregnant recipients, after which a muscular layer and an epidermal layer were sutured to each other. After the transplantation, born animals were grown for about 3 weeks. Next, their tails were cut about 1 cm and treated with lysis buffer at 55° C., and phenol was added thereto to extract genomic DNA. The genomic DNA was PCR-amplified with the following primer set so that whether it had been transformed or not was examined. The PCR amplification was performed in the following conditions: predenaturation at 94° C., 30 cycles of 30 sec at 94° C., 30 sec at 51° C., and 45 sec at 72.5° C., and then final extension at 72.5° C. for 5 min.
  • Sense primer
    (SEQ ID NO: 7)
    5′- TTG TCT CTA ATA GAG GGT C-3′
    Anti-sense primer
    (SEQ ID NO: 8)
    5′-TCA AGC CTG TCT ATC TTC T-3′
  • Meanwhile, mice overexpressing human EC-SOD were obtained from the National Jewish Medical & Research Center (Colorado, USA; bury et al. JBC 268 (21): 15394, 1993).
  • Example 1 Preparation of Recombinant EC-SOD Protein Having Biological Activity <1-1> Cloning of Fusion EC-SOD Gene for Expression of Recombinant EC-SOD Protein
  • In order to prepare a recombinant EC-SOD protein having biological activity, recombinant EC-SOD, consisting of 209 amino acids and containing deletion of the signal peptide at the N-terminal end of human EC-SOD and deletion of 13 amino acids at the C-terminal end, was cloned in the following manner. The base sequence and amino acid sequence of the recombinant EC-SOD are set forth in SEQ ID NO: 2 and SEQ ID NO: 1, respectively, and the base sequence and amino acid sequence of the human EC-SOD are set forth in SEQ ID NO: 4 and SEQ ID NO: 3, respectively.
  • A pUC 18-hEC-SOD vector (provided by Professor Marklund, Clinical Chemistry, Sweden) containing human EC-SOD cDNA (SEQ ID NO: 4), asa template, was amplified by PCR using the following primers, thus preparing human EC-SOD cDNA:
  • Sense primer
    (SEQ ID NO: 9)
    5′-tagattctggacgggcgagga-3′
    Anti-sense primer
    (SEQ ID NO: 10)
    5′-tactcgagtcactctgagtgct-3′
  • The PCR amplification was performed using PfuI polymerase in the following conditions: predenaturation at 95° C. for 5 min, and then 30 cycles of 30 sec at 95° C., sec at 55° C. and 1 min at 72° C., followed by final extension at 72° C. for 5 min. The amplified gene product was digested with restriction enzymes EcoRI and XhoI, and then ligated into a pET28a, vector (NovaGene, USA), digested with the same restriction enzymes, using T4 ligase at 4° C. for 12 hours.
  • The vector was transfected into E. coli DH5a, and then the transformants were cultured in LB solid medium containing 25 mg/mL of kanamycin, and the cultured transformants were screened. Whether the EC-SOD gene had been correctly inserted into the screened transformants was examined by gene sequence analysis. From the transformants determined to have the recombinant EC-SOD gene inserted therein, a vector was isolated. The vector was inserted into host cell E. coli BL21 (NovaGene, USA) for overexpression, and the cells were cultured in LB solid medium containing 25 mg/mL of kanamycin, while a strain overexpressing the EC-SOD protein was screened.
  • <1-2> Expression of Recombinant EC-SOD Protein
  • The strain screened in Example 1-1 was cultured in LB liquid medium, containing 25 mg/mL of kanamycin, at 37° C. When the cells reached an OD (optical density) of 0.6 at 600 nm, IPTG (1 mM) was added thereto to induce the expression of the EC-SOD protein, and the cells were cultured at 37° C. for 6 hours, thus expressing the recombinant EC-SOD protein in the cells.
  • <1-3> Induction of Refolding and Purification of Recombinant EC-SOD Protein
  • After the completion of the expression, the cells were collected from the culture medium by centrifugation, and the collected cells were suspended in 50 mM Tris buffer (pH 7.5) containing 100 mM calcium chloride and were disrupted with an ultrasonic disruptor. The cell suspension was centrifuged, and the precipitate layer was isolated and washed three times with 50 mM Tris buffer containing 100 mM calcium chloride. The washed cell precipitate contained an insoluble protein inclusion body. 10 mg of the washed inclusion body (a fusion protein content of 50%) was added to Tris buffer solution (50 mM Tris-Cl, 100 mM NaCl, pH 7.5), and then dissolved by the addition of 8M urea. After the inclusion body was completely suspended at room temperature for 2 hours, it was subjected to phase separation by centrifugation. The upper aqueous layer was collected because it contained the dissolved recombinant EC-SOD protein inclusion body (inclusion body solution).
  • The refolding and purification of the recombinant EC-SOD protein were performed in the following manner using fast protein purification liquid chromatography (FPLC). The buffer solution (8 M urea, 50 mM Tris-Cl, 100 mM NaCl, pH 7.5) was introduced into a metal affinity resin-packed chromatography column (5 ml His Trap FF crude, GE Healthcare, USA) at a flow rate of 1 ml/min to equilibrate the column, and then 5 ml of the above-obtained inclusion body solution was injected downward into the top of the metal affinity resin-packed chromatography column (3 ml) at a flow rate of 1 mL/min, so that the folded protein was adsorbed onto the solid matrix in the column.
  • After the completion of the injection, 50 ml of the buffer solution was injected downward into the top of the column at a flow rate of 1 mL/min in order to remove non-absorbed solids. After cell debris was completely removed from the column as described above, a Tris buffer solution (50 mM Tris buffer, 1 M urea, 100 mM NaCl, 100 mM ZnCl2, 50 mM CuSO4, pH 7.5), containing 1 M urea and comprising Zn and Cu atoms having an effect on the activity of EC-SOD, was injected downward into the top of the column at a flow rate of 0.5 mL/min while applying a linear concentration gradient, thus gradually reducing the concentration of the denaturant in the column. Accordingly, the metal ions (Zn and Cu) were completely substituted into the target protein with the natural three-dimensional structure, and thus the folded protein adsorbed onto the solid matrix was refolded to have activity.
  • Then, a Tris buffer solution, containing 2M imidazole and 1M urea, was fed downward into the top of the column with a concentration gradient. The refolded fusion protein was eluded when an elution buffer solution having an imidazole concentration of 0.5M was fed into the column (see FIG. 1).
  • <1-4> Separation and Purification of Recombinant EC-SOD Protein
  • In order to remove a large amount of imidazole from the eluted protein obtained in Example 1-3, completely refold the protein and separate the protein according to size, a fast protein purification liquid chromatography (FPLC) column and a gel filtration chromatography column (Sepharose 12 column) were used. In this process, a Tris buffer solution (50 mM Tris-Cl, 100 mM NaCl, pH 7.5) or a phosphate buffered saline (pH 7.5) was injected downward into the top of the column at a flow rate of 0.5 mL/min to separate and purify the protein. The separated target protein was concentrated by ultrafiltration.
  • As a result, the recombinant EC-SOD protein of the present invention could be obtained. Also, the sample in each purification step was analyzed by SDS-PAGE and, as a result, as can be seen in FIG. 2, the recombinant EC-SOD protein was well separated.
  • <1-5> Identification of EC-SOD Protein by Western Blot
  • In order to examine whether the recombinant EC-SOD protein separated and purified in Example 1-4 was rightly EC-SOD, Western blot analysis was performed. As the standard protein of EC-SOD, human EC-SOD expressed by human keratinocyte HaCaT cells (provided by Professor N. Fusenig, Cancer Research, Germany) was used. The human EC-SOD, expressed by HaCaT cells, and the EC-SOD proteins, separated and purified in Example 1-4, were separated by electrophoresis in 10% polyacrylamide gel, and then transferred to a polyvinylidene fluoride membrane (Gelman Laboratory, MI, USA). Then, the polyvinylidene fluoride membrane was blocked with 5% skimmed milk and cultured with an anti-EC-SOD polyclonal antibody (1: 500, Santa Cruz, USA). The membrane was washed with water, incubated with peroxidase-conjugated secondary antibody IgG (Santa Cruz, USA), washed again with water, and then color-developed using an ECL detection kit (GE Healthcare, USA) according to the manufacturer's instruction.
  • As a result, as can be seen in FIG. 3, a positive response was shown in the human EC-SOD (lane 1) and also detected in the separated and purified recombinant EC-SOD protein (lane 2). This suggests that the purified protein was human EC-SOD.
  • <1-6> Examination of Activity of Recombinant EC-SOD Protein
  • In order to examine whether the separated and purified recombinant EC-SOD protein has the biological activity of SOD, the activity of the recombinant EC-SOD protein was tested using an SOD assay kit-WST (Dojindo Molecular Technology, USA) according to the manufacturer's instruction.
  • As a result, as can be seen in FIG. 4, the sample (209FT) eluted without being adsorbed in the purification process showed no activity, the primarily purified recombinant EC-SOD protein (209 E1) refolded using the metal affinity chromatography column showed an activity of 250 unit/mg protein; and the recombinant EC-SOD protein (209 E2) separated and purified using the gel-filtration column showed an activity of 360 unit/mg protein.
  • <1-7> Examination of ROS Removal Effect of Separated and Purified Recombinant EC-SOD Protein
  • In order to examine the ROS (reactive oxygen species) removal effect of the recombinant EC-SOD protein in HaCaT cells, the recombinant EC-SOD protein, separated and purified in Example 1-4, was treated in the following manner.
  • Skin keratinocyte HaCaT cells were plated on 18-mm cover slips at a concentration of 1×104 cells/cover slip, and then cultured in an IMDM medium (Isocove's modified Dulbecco's medium, GIBCO), containing 10% fetal bovine serum (FBS, GIBCO) and 1% of 100 units penicillin and 100 g streptomycin/ml of medium in 5% CO2 at 37° C. When the adhered cells reached a confluence of about 30-40%, the fetal bovine serum was removed, and then the cells were cultured. When the cells reached a confluence of 50-60%, the cells were pretreated with 10 μg of the EC-SOD protein. After the cells were irradiated with UVB at a dose of 100 J/m2 for 3 hours, the cells were allowed to react with 10 mM DCFH-DA at 37° C. for 30 minutes and observed with a fluorescent microscope. The fluorescent micrograph was obtained using a Zeiss digital camera.
  • As a result, as can be seen in FIG. 5, in the UVB group where the cells were irradiated only with UVB, ROS was not removed, but in the EC-SOD+UVB group where the cells were irradiated with UVB and treated with the inventive recombinant EC-SOD protein, ROS was effectively removed.
  • Example 2 Angiogenesis Inhibitory Effect of Natural EC-SOD Protein
  • <2-1> Morphological Comparison of EC-SOD-Expressing Mice with Wild-Type Mice
  • The back skin of each of the mouse EC-SOD-overexpressing mice, prepared in Test Example 1, and wild-type BDF1 mice, was irradiated four times with UVB at a dose of 2 kJ/m2 at 24-hr intervals. Specifically, the back skin of each mouse was shaved at 2 days before UVB irradiation, and twenty-four 8-week-old mice were irradiated with UVB at a dose of 200 mJ/cm2 using 6 UVB lamps (FS24T12/UVB/HO, 290-320 nm, Voltare, Fairfield, Conn., USA) having the highest energy at 305 nm in a wavelength range from 280 μm to 340 nm. During the UVB irradiation, the height of the lamps was controlled such that the surface of the back skin of each of the anesthetized mice was irradiated with a dose of 0.3 mW/cm2. The mice of each group (n=6) were irradiated four times with UVB at 24-hr intervals, and at 1 hour after the 4th UVB irradiation, the mice were sacrificed for experiments. The back skin of each of the sacrificed mice was incised and visually observed, and the observation results are shown in FIG. 6.
  • As can be seen in FIG. 6, in the case where the wild-type mice were irradiated with UVB (FIG. 6C), angiogenesis, a skin inflammatory reaction and erythema were significantly induced compared to the case where the mice were not irradiated with UVB (FIG. 6A). However, in the EC-SOD-overexpressing mice, there was no statistically significant difference between before UVB irradiation (FIG. 6B) and after UVB irradiation (FIG. 6D). In other words, it could be observed that angiogenesis induced by UV light (UVB) irradiation in the wild-type mice (FIG. 6B) was significantly inhibited in the EC-SOD-overexpressing mice (FIG. 6D). These results suggest that the EC-SOD protein functions to inhibit angiogenesis.
  • <2-2> Western Blot
  • The expression level of angiogenesis-inducing vascular endothelial growth factor (VEGF) in the mouse EC-SOD-overexpressing mice prepared in Test Example 1 was analyzed by Western blot.
  • The back skin tissues or cells of each of the EC-SOD-overexpressing mice, sacrificed in Example 2-1, and the wild-type mice, were homogenized and extracted with an extraction solution (50 mM Tris HCl pH 8.0, 5 mM EDTA, 150 mM NaCl, 0.5% sodium deoxycholate, 1% Nonidet P-40, 0.1% SDS, 1 mM PMSF, 1 mM NaF, 1 mM NaVO4, and a protease inhibitor cocktail (Roche, Germany)). To determine protein concentration, a bio-rad protein kit (Bio-Rad, CA, USA) was used, and bovine serum albumin (BSA) was used as a standard material. The protein extracts were separated on 10% polyacrylamide gel, and then transferred to a polyvinylidene fluoride membrane (Gelman Laboratory, MI, USA). Then, the polyvinylidene fluoride membrane was blocked with 5% skimmed milk, and then incubated with each of an anti-VEGF polyclonal antibody (1: 500, Santa Cruz, USA) and an anti-beta-actin antibody. The membrane was washed with water, incubated with peroxidase-conjugated secondary antibody IgG (SantaCruz, USA), washed again with water, and then color-developed with an ECL detection kit.
  • As a result, as can be seen in FIG. 7, the expression level of VEGF in the wild-type mice was increased due to UVB irradiation, but the expression of VEGF in the EC-SOD-overexpressed mice irradiated with UVB was significantly reduced. These results confirm again that the EC-SOD protein has the effect of inhibiting angiogenesis.
  • <2-3> Immunohistochemical Examination
  • Whether EC-SOD inhibits the expression of VEGF was examined again by an immunohistochemical method.
  • The back skin tissue of each of the EC-SOD-overexpressing mice, sacrificed in Example 2-1, and the wild-type mice, was fixed with 4% paraformaldehyde in 0.5M phosphate buffered saline (pH 7.4). The fixed tissue was washed with running water, dehydrated with ethanol, embedded in paraffin, sectioned to a thickness of 5 mm and attached to glass slides. The sections were deparaffinized with xylene solution, hydrated with ethanol, and then stained with hematoxylin and eosin. The deparaffinized tissue segments were blocked by treatment with 3% hydrogen peroxide solution. To reduce nonspecific binding and background staining, the sections were treated with 10% goat serum. Each of the tissue slides was treated with an anti-VEGF polyclonal antibody (1: 500, Santa Cruz, USA) and visualized by the avidin-biotin-immunoperoxidase complex method using a streptavidin-biotin kit (LSAB, Dako, Calif., USA). The stained tissue was washed with water, and then color-developed with diaminobenzidine (DAB), and the tissue sections were counterstained with 0.2% Mayer's hematoxylin (Sigma, Mo., USA). The staining results are shown in FIG. 8.
  • As shown in FIG. 8, in the wild-type mice, VEGF was highly expressed by UVB irradiation, but in the EC-SOD-overexpressed mice, the expression of VEGF was significantly inhibited compared to that in the wild-type mice. These results were consistent with the results of Example 1-2. These results suggest that natural EC-SOD has the effect of inhibiting the expression of angiogenesis-inducing VEGF expression in vivo to inhibit angiogenesis.
  • <2-4> Examination of Expression Level of MMP-9 in EC-SOD-Overexpressing Mice
  • The MMP protein degrading an extracellular matrix protein is an important angiogenesis regulatory factor together with VEGF. Particularly, MMP9 is known to be a factor regulating the tumor angiogenesis process through the VEGF-VEGFR system (Bergers et al., Nat. Cell Biol., 2: 737-744, 2000).
  • Thus, the present inventors examined the expression level of MMP-9 in EC-SOD-overexpressing mice by UVB irradiation.
  • The back skin tissue of each of the EC-SOD-overexpressing mice, sacrificed in Example 2-1, and the wild-type mice, was mixed with 1 mg/ml gelatin (100 mg/ml) and subjected to SDS-PAGE. Then, the gel was treated with 2.5% Triton X-100 for 20 minutes and washed with water. The gel was incubated with a Zymography reaction solution (1M Tris (pH7.5), 1M CaCl2, 5M NaCl, 0.2 mM ZnCl2, 25% TX-100, 0.2% NaN3) at 37 Cfor 16 hours, stained for 1 hour, decolorized for 1 hour, and then dried in an over for 1 hour. The dried gel was analyzed, and the analysis results are shown in FIG. 9.
  • As can be seen in FIG. 9, there was no significant difference in the expression level of MMP-9 between the wild-type mice and the EC-SOD-overexpressed mice before irradiation with UV light. However, due to UVB irradiation, the expression of MMP-9 in the wild-type was increased, but the expression of MMP-9 in the EC-SOD-overexpressed mice was significantly inhibited compared to that in the wild-type mice.
  • From the above results, it can be seen that the natural EC-SOD protein inhibits the expression of VEGF in vivo, and in addition, inhibits the expression of MMP-9, which is involved in angiogenesis-related signaling through the VEGF-VEGFR system, thus effectively inhibiting angiogenesis.
  • Example 3 Angiogenesis Inhibitory Effect of Recombinant EC-SOD Protein
  • Whether the recombinant EC-SOD protein, separated and purified in Example 1, has the effect of inhibiting angiogenesis was examined. Keratinocyte HaCaT cells were plated on 18-mm cover slips at a concentration of 1×104 cells/cover slip. The cells were cultured in DME medium (Dulbecco's minimum essential medium, GIBCO), containing 10% fetal bovine serum (FBS, GIBCO) and 100 units penicillin and 100 g streptomycin/ml of medium, in 5% CO2 at 37° C. When the adhered cells reached a confluence of 30-40%, the fetal bovine serum was removed, and then cultured for 24 hours. When the cells reached a confluence of 50-60%, the cells were pretreated with 10 μg of EC-SOD. After the cells were treated with the purified recombinant EC-SOD protein for 30 minutes, the cells were treated with UV light at a dose of 100 J, and after 24 hours, the skin keratinocyte cells were fixed with paraformaldehyde and immunostained. The fixed cells were blocked with 10% normal goat serum and washed three times with phosphate buffered saline (PBS). For immunostaining for VEGF (vascular endothelial growth factor), the cells were treated with a rabbit anti-VEGF antibody (Santacruz, USA), and then incubated at 4° C. for at least 16 hours. Then, the cells were fluorescence-stained with a FITC-conjugated anti-mouse-IgG secondary antibody (Zymed, USA), and then observed with a fluorescent microscope (Carl Zeiss, Germany).
  • As a result, as shown in FIG. 10, in the UVB group where the cells were irradiated with UVB, the expression of VEGF in the cells was significantly increased compared to that in the control group not irradiated with UVB. Also, in the EC-SOD/UVB group where the group irradiated with UVB was treated with the recombinant protein, the expression of VEGF was significantly inhibited, suggesting that angiogenesis was inhibited by the recombinant EC-SOD protein.
  • Example 4 Effect of Recombinant EC-SOD Protein on Proliferation and Differentiation of T Cells <4-1> Examination of Effect of Recombinant EC-SOD on Proliferation of T Cells
  • Whether EC-SOD has an effect on immune T cells, the following test was examined in the following manner using specific antigen (ovalbumin)-reactive TCR (T cell receptor)-transfected mice (DO.11.10 provided by professor Young-Chul Sung, Pohang University of gcience and Technology).
  • In order to examine whether EC-SOD has an effect on the proliferation of T cells, cells isolated from the spleen of the transfected mice were cultured with the antigen ova peptide for 72 hours, and then cultured with 3H for 16 hours. Then, the radiation dose of the cultured cells was measured to examine the proliferation.
  • As a result, as can be seen in FIG. 11, there is little or no difference in radiation doses between the group treated without EC-SOD and the groups treated with EC-SOD at concentrations of 1 μg/ml and 10 μg/ml, respectively, suggesting that EC-SOD had little or no effect on the proliferation of T cells.
  • <4-2> Examination of Effect of Recombinant EC-SOD on Differentiation of T Cells
  • Although recombinant EC-SOD has no effect on the proliferation of T cells, it can have an effect on the differentiation of T cells. For this reason, T cell cytokines were measured to examine the differentiation of T cells.
  • Spleen cells were cultured with the antigen ova peptide for 48 hours, and then IL-4 (interleukin-4) produced in the culture medium was measured. The measurement was carried out using an ELISA-kit (Becton, Dickinson and Company, USA) according to the manufacturer's instruction.
  • As a result, as can be seen in FIG. 12, EC-SOD reduced the formation of IL-4. This suggests that EC-SOD has an effect on the differentiation of T cells. In FIG. 12, neg.: a test group not treated with an antigen; −: a test group treated only with an antigen (Ova peptide) p: a test group treated with an antigen (Ova peptide) together with EC-SOD in PBS NB: a test group treated with an antigen (Ova peptide) together with EC-SOD in sodium borate. All the test groups were treated in culture media for 48 hours.
  • Meanwhile, in order to specifically examine the effect of the inventive recombinant EC-SOD on the differentiation of T cells, the recombinant EC-SOD protein was administered into the T-cells of mice, and the amounts of production of cytokines, such as IL-4 and IFN-γ, were compared.
  • Specifically, from the spleen of DO 11.10 mice provided by Professor Young-Chul Sung, Pohang University of Science and Technology) overexpressing a TCR (T cell receptor) gene recognizing a specific antigen Ova peptide, CD4 T cells were isolated using MACS (Miltenyi Biotech). As antigen-presenting cells, the spleen cells of normal Balb/c mice were subjected to γ-irradiation, and then cultured with the Ova peptide (1 μg/ml, Ova323-339, Peptron) and the CD4 T cells isolated using MACS, in RPMI1640 medium supplemented with 10% FBS. During the culture process, the recombinant EC-SOD protein, separated and purified in Example 1, was added to the culture medium. After 72 hours, the culture medium was collected, and the amounts of IL-4 and IFN-γ in the culture medium were measured using ELISA (antibody: Becton Dickinson).
  • As a result, as can be seen in FIG. 13, the recombinant EC-SOD protein, separated and purified in Example 1, induced the formation of Th1 cytokine IFN-γ in T cells and inhibited the formation of Th2 cytokine IL-4. This suggests that the recombinant EC-SOD protein had an effect on immune cells, particularly on the differentiation of T cells.
  • Accordingly, it can be seen that the recombinant EC-SOD protein of the present invention blocks the over-differentiation of Th2 cells inducing allergic diseases and induces differentiation into Th1 cells, and thus it can be advantageously used to treat or prevent allergic diseases.
  • Example 5 Effect of Natural EC-SOD Protein on Differentiation of T Cells
  • An Ova protein and an immune adjuvant CFA (Complete Freund's Adjuvant, Sigma) inducing Th1 immunity were mixed with alum (aluminum hydroxide, Pierce) inducing Th2 immunity, and 25 μg of the mixture was immunized into the foot sole and tail of the human EC-SOD-overexpressed mice, prepared in Test Example 1. After 11 days, from lymphocytes having inflammation occurred therein, cells were isolated and observed to examine whether EC-SOD had effects on cell proliferation and cytokine production.
  • In the observation of cell proliferation, 5×105 lymphocytes were cultured in a 96-well plate, and after 3 days of the culture, isotope 3H was added to the culture medium. After 16 hours, the proliferation of the cells was observed by measuring the content of the isotope in the cells using a β-counter. Also, in order to examine whether the reaction is an antigen-specific reaction, cell proliferation was observed using varying amounts of the Ova protein. To observe the expression of cytokines, 5×106 lymphocytes were cultured in a 46-well plate, and after 72 hours, the culture medium was collected, and the amounts of IL-4 and IFN-γ in the culture medium were measured using ELISA. The measurement results are shown in FIGS. 14 and 15.
  • As shown in FIGS. 14 and 15, it was observed that, when CFA inducing Th1 differentiation was used as an immune adjuvant, the proliferation of lymphocytes was slightly increased compared to the expression of lymphocytes in the normal mice, and the expression of IFN-γ was increased. It was also observed that, when alum inducing Th2 differentiation was used as an immune adjuvant, the proliferation of lymphocytes was reduced compared to the proliferation of lymphocyte in the normal mice, and the expression of IL-4 was reduced. Such results suggest that EC-SOD has an effect on the differentiation of T cells, that is, it stimulates differentiation into Th1 and inhibits differentiation into Th2.
  • Accordingly, it can be seen that the natural EC-SOD protein blocks the over-differentiation of Th2 cells inducing allergic diseases and induces differentiation into Th1 cells, and thus it can be advantageously used to treat or prevent allergic diseases.
  • Example 6 Examination of Effect of Recombinant EC-SOD Protein on Inhibition of Transcriptional Factor Activity
  • In order to examine the effect of EC-SOD on NF-κ B activation caused by UV light, HaCaT cells were plated on a 14-mm culture slide, and then cultured in 10% fetal bovine serum-containing DMEM (Dulbecco's modified Eagle's medium) for 24 hours. After 24 hours of the culture, the cells were starved in 1% fetal bovine serum-containing DMEM medium for 24 hours. After the starvation, the cells were pretreated with 10 μg of EC-SOD for 30 minutes, and then washed three times with phosphate buffered saline (PBS). To induce the activation of NF-κ B, the cells were then irradiated with UV light at a dose of 100 J/m2. Immediately after the UV light irradiation, the cells were washed again with phosphate buffered saline, treated with 10 μg of EC-SOD, and then further cultured in 1% fetal bovine serum-containing DMEM medium for 4 hours. Then, the cells were fixed with methanol and immunostained. The fixed cells were blocked with 10% normal goat serum and washed three times with phosphate buffered saline (PBS). For immunostaining for EC-SOD and NF-κ B, the cells were treated with each of a mouse anti-EC-SOD antibody (Lab Frontier, Korea) and a rabbit anti-NF-kB antibody (Santa Cruz, USA) and incubated at 4° C. for at least 16 hours. Then, the cells were fluorescence-stained with TRITC (tetramethyl rhodamine isothiocyanate)-conjugated anti-rabbit IgG secondary antibody, Serotec, GB) and a FITC-fused anti-mouse IgG secondary antibody (Zymed, USA), and then counterstained with Hoechst (Sigma, USA).
  • As a result, as shown in FIG. 16 (200× magnification), it could be observed that, in the control group where the HaCaT cells transfected with EC-SOD were not irradiated with UV light, NF-κ B was not immunostained in the cytoplasm and the nucleus, suggesting that NF-κ B was inactivated (FIGS. 16A, D, G and J). It was also observed that, in the UVB group (where the HaCaT cells were irradiated with UVB at a dose of 100 J/m2), NF-κ B was rapidly activated at the nuclear site and the perinuclear site (FIGS. 16B, E, H and K). In comparison with this, it was observed that, in the EC-SOD/UVB group (where HaCaT cells were treated with the inventive recombinant EC-SOD protein and irradiated with 100 J/m2 of UVB), the activation of NF-κ Bat the nuclear site was significantly inhibited compared to that in the UVB group (FIGS. 16C, F, I and L). Such results revealed that the recombinant EC-SOD protein of the present invention inhibits the activation of the transcriptional factor NF-κ B.
  • Accordingly, it can be seen that the recombinant EC-SOD protein of the present invention inhibits the abnormal activation of NF-κ B, and thus it can be used to prevent or treat autoimmune diseases, such as allergies.
  • Example 7 Examination of Effect of EC-SOD Protein on Reduction of Human Mast Cell Degranulation
  • In order to examine whether EC-SOD has an effect on the degranulation of human mast cells, mast cells were stained with toluidine blue, and then the number of degranulated cells among 100 stained cells was counted.
  • Human mast cells (semi-adhesion, Human Mast Cell-1 (HMC-1) provided by professor Hyung-Min Kim, Kyunghee University) were plated on 18×18 mm cover slips at a concentration of 1×104 cells/cover slip. The cells were cultured in IMDM medium (Isocove's modified Dulbecco's medium, GIBCO), containing 10% fetal bovine serum (FBS, GIBCO) and 100 units penicillin and 100 g streptomycin/ml of medium, in 5% CO2 at 37° C. Because the cells were semi-adhesion cells, the ratio of adhered cells to non-adhered cells was about 4:6. When the adhered cells reached a confluence of about 30-40%, the cells were pretreated with 5 μg of EC-SOD for 1 hour, and then treated with 1 μM of calcium channel A23187 and 50 nM of phorbol 12-myristate 13-acetate (PMA) to increase the concentration of calcium in the cells.
  • After 12 hours, the human mast cells were washed with phosphate buffered saline (PBS), and then fixed with 4% paraformaldehyde for 10 minutes. After the cells were washed with phosphate buffered saline, they were stained with a solution of 0.1% toluidine blue in 1% acidic saline (pH 2.3) for 2-3 minutes. After the cells were washed again with water, the cells were dehydrated with 95% and 100% alcohol and fixed with xylene, and the number of degranulated cells among 100 stained cells was counted.
  • As a result, as shown in FIG. 17A, the number of degranulated cells in the human mast cell group (control) not treated with anything was 10, and the number of degranulated cells in the group (EC-SOD) treated with EC-SOD was 16. In comparison with this, in the group (A+P) where the concentration of calcium in the cells was increased to induce the activation and degranulation of the mast cells, the number of degranulated cells was increased five-fold to 55, but in the group (A+P+EC-SOD) where the cells were pretreated with EC-SOD and treated to increase the concentration of calcium in the cells, the number of degranulated cells was reduced again to 25. This indicates that the reduction of mast cell degranulation occurred due to the inhibition of reactive oxygen species caused by the action of EC-SOD. A micrograph showing the degranulation of the mast is shown in FIG. 17B.
  • Accordingly, in view of the fact that mediators, such as histamine, stored in mast cells, are released due to the degranulation of the mast cells to cause inflammatory reactions, such as allergies, it can be seen that the recombinant EC-SOD protein of the present invention can be advantageously used to prevent or treat allergies.
  • INDUSTRIAL APPLICABILITY
  • An EC-SOD protein or a vector having a polynucleotide encoding the EC-SOD protein has the effect on inhibiting angiogenesis by inhibiting the expression of VEGF and MMP-9 which induce angiogenesis. Therefore, the EC-SOD protein or the vector may be useful for preventing or treating angiogenesis-mediated diseases. And the EC-SOD protein or the vector having a polynucleotide encoding said EC-SOD protein also has the effect on inhibiting over-differentiation of Th2 cells which cause allergic diseases, inhibiting transcription factor (NF-κ B), and reducing degranulation of human mast cells. Accordingly, the EC-SOD protein or the vector may be useful for preventing or treating allergy diseases. In addition, the inventive method for preparing an EC-SOD protein may be used in preparing an EC-SOD protein having the activity on a large scale. Therefore, the inventive method may be useful industrially.

Claims (26)

1. A composition for preventing or treating angiogenesis-mediated diseases comprising an EC-SOD protein as an active ingredient.
2. The composition for preventing or treating angiogenesis-mediated diseases of claim 1, wherein the EC-SOD protein has an amino acid sequence selected from the group consisting of SEQ ID NOs: 1, 3, and 5.
3. The composition for preventing or treating angiogenesis-mediated diseases of claim 1, wherein the diseases is selected from the group consisting of cancer, diabetes, rheumatoid arthritis, arteriosclerosis, hemangioma, angiofibroma, diabetic retinopathy, retinopathy of prematurity, neovascular glaucoma, corneal diseases caused by angiogenesis, degenerative spots, macular degeneration, pterygium, retinal degeneration, retrolental fibroplasia, Granular conjunctivitis, telangiectasia, pyogenic granuloma and acne.
4. A composition for preventing or treating angiogenesis-mediated diseases comprising a vector having a polynucleotide encoding the EC-SOD protein of claim 1 as an active ingredient.
5. The composition for preventing or treating angiogenesis-mediated diseases of claim 4, wherein the polynucleotide has a nucleotide sequence selected from the group consisting of SEQ ID NOs: 2, 4, and 6.
6. The composition for preventing or treating angiogenesis-mediated diseases of claim 4, wherein the diseases is selected from the group consisting of cancer, diabetes, rheumatoid arthritis, arteriosclerosis, hemangioma, angiofibroma, diabetic retinopathy, retinopathy of prematurity, neovascular glaucoma, corneal diseases caused by angiogenesis, degenerative spots, macular degeneration, pterygium, retinal degeneration, retrolental fibroplasia, Granular conjunctivitis, telangiectasia, pyogenic granuloma and acne.
7. A composition for preventing or treating allergic diseases comprising an EC-SOD protein as an active ingredient.
8. The composition for preventing or treating allergic diseases of claim 7, wherein the EC-SOD protein has an amino acid sequence selected from the group consisting of SEQ ID NOs: 1, 3, and 5.
9. The composition for preventing or treating allergic diseases of claim 7, wherein the allergic diseases is selected from the group consisting of allergic asthma, allergic rhinitis, allergic otitis media, anaphylatic shock, allergic skin disease, atopic dermatitis, psoriasis, contact allergic dermatitis and urticaria.
10. A composition for preventing or treating allergic diseases comprising a vector having a nucleotide encoding the EC-SOD protein of claim 7 as an active ingredient.
11. The composition for preventing or treating allergic diseases of claim 10, wherein the polynucleotide has a nucleotide sequence selected from the group consisting of SEQ ID NOs: 2, 4, and 6.
12. The composition for preventing or treating allergic diseases of claim 10, wherein the allergic diseases is selected from the group consisting of allergic asthma, allergic rhinitis, allergic otitis media, anaphylatic shock, allergic skin disease, atopic dermatitis, psoriasis, contact allergic dermatitis and urticaria.
13. (canceled)
14. (canceled)
15. (canceled)
16. (canceled)
17. (canceled)
18. (canceled)
19. A method for preventing or treating angiogenesis-mediated disease, comprising administering to a subject in need thereof an effective amount of an EC-SOD protein.
20. Use of an EC-SOD protein for preparation a therapeutic agent for angiogenesis-mediated diseases.
21. A method for preventing or treating angiogenesis-mediated disease, comprising administering to a subject in need thereof an effective amount of a vector having a polynucleotide encoding an EC-SOD protein.
22. Use of a vector having a polynucleotide encoding an EC-SOD protein for preparation a therapeutic, agent for angiogenesis-mediated diseases.
23. A method for preventing or treating allergic diseases comprising administering to a subject in need thereof an effective amount of an EC-SOD protein.
24. Use of an EC-SOD protein for preparation a therapeutic agent for allergic diseases.
25. A method for preventing or treating allergic diseases comprising administering to a subject in need thereof an effective amount of a vector having a polynucleotide encoding an EC-SOD protein
26. Use of a vector having a polynucleotide encoding an EC-SOD protein for preparation a therapeutic agent for allergic diseases.
US12/532,539 2007-03-22 2008-03-24 Novel use of ec-sod and method for preparing thereof Abandoned US20120093796A1 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
KR10-2007-0028327 2007-03-22
KR1020070028327A KR101019470B1 (en) 2007-03-22 2007-03-22 Composition for preventing or treating angiogenesis-induced diseases containing EC SOD protein
KR10-2007-0056973 2007-06-11
KR1020070056973A KR20080108876A (en) 2007-06-11 2007-06-11 Recombinant ec-sod protein, prepration method and use thereof
PCT/KR2008/001636 WO2008115039A1 (en) 2007-03-22 2008-03-24 Novel use of ec-sod and method for preparing thereof

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2008/001636 A-371-Of-International WO2008115039A1 (en) 2007-03-22 2008-03-24 Novel use of ec-sod and method for preparing thereof

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US14/325,107 Continuation-In-Part US9089564B2 (en) 2007-03-22 2014-07-07 Use of EC-SOD for treating angiogenesis-mediated eye diseases
US14/325,077 Continuation US8980841B2 (en) 2007-03-22 2014-07-07 Use of EC-SOD and method for preparing thereof

Publications (1)

Publication Number Publication Date
US20120093796A1 true US20120093796A1 (en) 2012-04-19

Family

ID=39766094

Family Applications (2)

Application Number Title Priority Date Filing Date
US12/532,539 Abandoned US20120093796A1 (en) 2007-03-22 2008-03-24 Novel use of ec-sod and method for preparing thereof
US14/325,077 Active US8980841B2 (en) 2007-03-22 2014-07-07 Use of EC-SOD and method for preparing thereof

Family Applications After (1)

Application Number Title Priority Date Filing Date
US14/325,077 Active US8980841B2 (en) 2007-03-22 2014-07-07 Use of EC-SOD and method for preparing thereof

Country Status (4)

Country Link
US (2) US20120093796A1 (en)
JP (1) JP5581485B2 (en)
CN (1) CN101678074B (en)
WO (1) WO2008115039A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108430480A (en) * 2015-09-15 2018-08-21 康干细胞生物科技有限公司 A composition for preventing or treating inflammatory diseases comprising stem cells overexpressing SOD3 as an active ingredient

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102580064A (en) * 2011-01-06 2012-07-18 无锡天德生物技术有限公司 Medicinal application of superoxide dismutase (SOD) to treatment of rhinitis
ES2545929B2 (en) * 2014-12-23 2016-07-15 Universidad De Málaga Use of antibodies against the SOD-3 enzyme for the inhibition of the angiogenesis process and applications of said antibodies and said SOD-3 enzyme
CN106955286A (en) * 2017-04-14 2017-07-18 山东省眼科研究所 Application of the manganese benzoic acid porphyrin in the medicine for promoting diabetic corneal injury repair is prepared
CN109486771B (en) * 2018-12-29 2021-02-12 深圳市旷逸生物科技有限公司 CHO cell strain construction method for high expression SOD3 and SOD3 protein purification method

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6011067A (en) * 1999-06-11 2000-01-04 Thione International, Inc. Antioxidant composition for the treatment of psoriasis and related diseases
US7740839B2 (en) * 2003-10-31 2010-06-22 Tae-Yoon Kim EC SOD and cell transducing EC SOD and use thereof
US8318803B2 (en) * 2003-10-31 2012-11-27 Tae-Yoon Kim EC SOD and use thereof

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IL87737A (en) 1987-09-11 1993-08-18 Genentech Inc Method for culturing polypeptide factor dependent vertebrate recombinant cells
DE69024369T2 (en) 1989-11-22 1996-06-13 Genentech Inc LATENCY ASSOCIATED PEPTIDES AND THEIR USE
AU1795400A (en) * 1998-12-24 2000-07-31 Florence Medical Ltd. Expression vectors comprising multiple shear stress responsive elements (ssre) and a gene of interest and methods of use thereof
JP2001136985A (en) * 1999-09-01 2001-05-22 Dnavec Research Inc Use of ec-sod for treatment of systemic autoimmune disease
BR0309676A (en) * 2002-04-30 2005-02-22 Fit Biotech Oyl Plc Medical device

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6011067A (en) * 1999-06-11 2000-01-04 Thione International, Inc. Antioxidant composition for the treatment of psoriasis and related diseases
US7740839B2 (en) * 2003-10-31 2010-06-22 Tae-Yoon Kim EC SOD and cell transducing EC SOD and use thereof
US8318803B2 (en) * 2003-10-31 2012-11-27 Tae-Yoon Kim EC SOD and use thereof

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108430480A (en) * 2015-09-15 2018-08-21 康干细胞生物科技有限公司 A composition for preventing or treating inflammatory diseases comprising stem cells overexpressing SOD3 as an active ingredient
US20180344774A1 (en) * 2015-09-15 2018-12-06 Kangstem Biotech Co., Ltd. Composition for preventing or treating inflammatory diseases, containing, as active ingredient, stem cells overexpressing sod3
US11730766B2 (en) * 2015-09-15 2023-08-22 Kangstem Biotech Co., Ltd. Composition for preventing or treating inflammatory diseases, containing, as active ingredient, stem cells overexpressing SOD3

Also Published As

Publication number Publication date
CN101678074A (en) 2010-03-24
JP5581485B2 (en) 2014-09-03
US8980841B2 (en) 2015-03-17
US20140335067A1 (en) 2014-11-13
WO2008115039A1 (en) 2008-09-25
JP2010522159A (en) 2010-07-01
CN101678074B (en) 2014-04-23

Similar Documents

Publication Publication Date Title
US8980841B2 (en) Use of EC-SOD and method for preparing thereof
JP6408603B2 (en) Human FGFR2B extracellular domain and nucleic acid encoding it
US20200010533A1 (en) Modified serpins for the treatment of bradykinin-mediated disease
KR20230117552A (en) Composition for preventing or treating immune diseases comprising MLS-STAT3
US8318803B2 (en) EC SOD and use thereof
US9089564B2 (en) Use of EC-SOD for treating angiogenesis-mediated eye diseases
US9566314B2 (en) Extracellular vesicles comprising recombinant lysosomal transmembrane protein
EP1692187B1 (en) EC SOD and cell transducing EC SOD and use thereof
WO2015138309A1 (en) Compositions and methods for regulating pancreatic beta cell function using adipsin
CN112402592B (en) Application of SPINK7 protein in preparation of medicine for promoting healing of excessive inflammatory wound surface
CN113209303B (en) WWP1 degradation oncoprotein MUC1 inhibition tumor through lysosome approach and application thereof
KR101019470B1 (en) Composition for preventing or treating angiogenesis-induced diseases containing EC SOD protein
JP2001526063A (en) SAG: apoptosis susceptibility gene
Jiménez et al. Characterization of a Thioredoxin-1 gene from Taenia solium and its encoding product
CN114502529A (en) Antioxidant serine protease inhibitors
US20110092427A1 (en) Polypeptide and pharmaceutical composition containing the polypeptide
JP2009511036A (en) GSE 24.2 nucleotide and peptide sequences of dyskerin inducing telomerase activity, methods of obtaining the same, pharmaceutical compositions and methods of use thereof
KR20180126881A (en) Composition for preventing or treating dry eye syndrome comprising SOD3
US20110300115A1 (en) Use of inductor agents gse24.2 for producing pharmaceutical compositions for treating illnesses relating to cellular senescence
KR20080108876A (en) Recombinant ec-sod protein, prepration method and use thereof
CN116693699A (en) Polypeptide TAT-TRPV1-C and application thereof
US20090062189A1 (en) Tumor suppressor protein and nucleotide encoding same
Chavushyan et al. Cryoglobulins of schizophrenia-affected subjects induce production of monocyte chemoattractant protein-1 by human peripheral mononuclear cells
WO2013028527A1 (en) Compositions and methods for treating cancer
KR19990088583A (en) Process of purifying angiogenesis inhibitors

Legal Events

Date Code Title Description
AS Assignment

Owner name: INDUSTRY ACADEMIC COOPERATION FOUNDATION, THE CATH

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:SHIN, DONG HEON;KIM, TAE YOON;REEL/FRAME:023500/0971

Effective date: 20091023

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION