WO2014171721A1 - Ik factor and pharmaceutical use of nucleic acid encoding ik factor - Google Patents

Ik factor and pharmaceutical use of nucleic acid encoding ik factor Download PDF

Info

Publication number
WO2014171721A1
WO2014171721A1 PCT/KR2014/003282 KR2014003282W WO2014171721A1 WO 2014171721 A1 WO2014171721 A1 WO 2014171721A1 KR 2014003282 W KR2014003282 W KR 2014003282W WO 2014171721 A1 WO2014171721 A1 WO 2014171721A1
Authority
WO
WIPO (PCT)
Prior art keywords
factor
seq
fragment
nucleic acid
pharmaceutical composition
Prior art date
Application number
PCT/KR2014/003282
Other languages
French (fr)
Korean (ko)
Inventor
남재환
박혜림
이상명
이동희
Original Assignee
가톨릭대학교 산학협력단
(주)셀인바이오
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 KR1020140038809A external-priority patent/KR101625112B1/en
Application filed by 가톨릭대학교 산학협력단, (주)셀인바이오 filed Critical 가톨릭대학교 산학협력단
Priority to US14/784,614 priority Critical patent/US9737588B2/en
Publication of WO2014171721A1 publication Critical patent/WO2014171721A1/en
Priority to US15/481,787 priority patent/US9943567B2/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/713Double-stranded nucleic acids or oligonucleotides
    • 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]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/005Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'active' part of the composition delivered, i.e. the nucleic acid delivered
    • 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
    • C12N2750/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
    • C12N2750/00011Details
    • C12N2750/14011Parvoviridae
    • C12N2750/14111Dependovirus, e.g. adenoassociated viruses
    • C12N2750/14141Use of virus, viral particle or viral elements as a vector
    • C12N2750/14143Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector

Definitions

  • the present invention relates to pharmaceutical uses of peptides derived from living organisms and nucleic acids encoding them, and more particularly to pharmaceutical uses of IK factors or fragments thereof and / or nucleic acids encoding them.
  • Arthritis is a disease involving joint stiffness and persistent pain around the joints due to inflammation of one or more joints. Arthritis can be largely divided into acute arthritis and chronic arthritis. Chronic arthritis, such as gouty arthritis, polyarthritis, rheumatoid arthritis, and osteoarthritis due to aging of bones or joints, causes persistent pain in the joint area.
  • rheumatoid arthritis is an unexplained chronic inflammatory disease characterized by multiple arthritis. To date, the exact cause of rheumatoid arthritis is unknown. However, genetic predisposition, bacterial and viral infections are generally thought to be the cause of rheumatoid arthritis, and hormones are also believed to be involved in rheumatoid arthritis. Rheumatoid arthritis not only affects the joints, but also adversely affects other organs of the body. Thus, patients with rheumatoid arthritis are not only unable to use or walk their hands or feet, but also frequently feel tired and uncomfortable.
  • rheumatoid arthritis causes symptoms such as weight loss and sleep deprivation, and if rheumatoid arthritis persists, physical activity, such as muscle weakness, may be restricted, which may greatly affect personal and social activities.
  • physical activity is limited by rheumatoid arthritis, the likelihood of obesity increases, can lead to heart disease due to cholesterol levels, and can lead to depression.
  • Rheumatoid arthritis occurs in 0.2 men per 1,000 men and 0.4 women per 1,000 women per year, and the prevalence of rheumatoid arthritis is 0.4-1.4% worldwide. Moreover, the market for rheumatoid arthritis drugs is expected to grow rapidly as the elderly population grows.
  • drugs for treating rheumatoid arthritis include nonsteroidal anti-inflammatory drugs (NSAIDs); steroid; Non-biologic disease-modifying antirheumatic drug, DMARD such as antimalarial agent, hydroxychloroquinone (HCQ), sulfasalazine, methotrexate (MTX), leflunomide ); Biological antirheumatic drugs such as tumor necrosis factor (TNF) inhibitors or IL-6 inhibitors are used.
  • NSAIDs nonsteroidal anti-inflammatory drugs
  • DMARD such as antimalarial agent, hydroxychloroquinone (HCQ), sulfasalazine, methotrexate (MTX), leflunomide
  • Biological antirheumatic drugs such as tumor necrosis factor (TNF) inhibitors or IL-6 inhibitors are used.
  • tumor necrosis factor (TNF) inhibitors such as etanercept, adalimumab, infliximab, golomumab, certolizumab; IL-6 inhibitors such as Tocilizumab; Rituximab, a monoclonal antibody that is a B-cell scavenger; Abatacept, which blocks an immune response between antigen presenting cells and T-cells; As small molecule inhibitors, tofacitinib, which selectively blocks JAK (Janue activated kinase), has been developed.
  • TNF tumor necrosis factor
  • nonsteroidal anti-inflammatory drugs cause gastrointestinal side effects
  • nonsteroidal anti-inflammatory drugs including selective COX-2 inhibitors
  • cardiovascular side effects can cause serious cardiovascular side effects.
  • nonsteroidal anti-inflammatory drugs are known to have side effects such as renal failure, interstitial nephritis, and acute renal failure. Steroids can cause hypothalamic-pituitary-adrenal atrophy, resulting in adrenal insufficiency, and side effects such as glaucoma, cataracts, osteonecrosis, osteoporosis, hypertension and hypokalemia.
  • nonsteroidal anti-inflammatory drugs and steroids are not only to relieve inflammation, it does not inhibit the progress of the actual arthritis, and there is a disadvantage that the prevention effect of the joint damage is insufficient.
  • the anti-rheumatic drugs currently developed may inhibit the progression of early rheumatoid arthritis, but do not cure rheumatoid arthritis and some side effects have been reported.
  • abiotic antirheumatic drugs have side effects such as vision disorders, retinal lesions, skin rashes, abnormal liver function levels, and nausea (nausea).
  • tumor necrosis factor inhibitors among biological antirheumatic drugs are problematic for blood abnormalities, heart failure, liver dysfunction, and opportunistic infections, and IL-6 inhibitors may cause side effects such as liver dysfunction and white blood cell reduction.
  • Other biological antirheumatic drugs have been reported with side effects such as infection problems, tumor potential, gastrointestinal perforation and hyperlipidemia.
  • the present invention has been proposed to solve the above-mentioned problems of the prior art, and an object of the present invention is a pharmaceutical for treating and / or preventing arthritis in which safety is ensured due to no side effects and toxicity to a living body despite prolonged administration. It is to provide a composition.
  • the present invention provides a pharmaceutical composition for treating or preventing arthritis, containing a pharmaceutically effective amount of an IK factor or fragment of an IK factor.
  • the fragment of IK factor comprises a peptide having the amino acid sequence of SEQ ID NO: 4 or a partial peptide of SEQ ID NO: 4.
  • the fragment of the IK factor consists of at least one amino acid residue selected from 382 th serine, 489 tyrosine, and 492 tyrosine of the amino acid sequence of SEQ ID NO: 2 and adjacent amino acids of these selected amino acid residues. It may consist of peptides having 10 or more amino acids.
  • the fragment of the IK factor is phosphorylated with at least one amino acid consisting of 382 th serine, 489 th tyrosine, and 492 th tyrosine of the amino acid sequence of SEQ ID NO: 2.
  • the fragment of IK factor is a fragment of a peptide having an amino acid sequence of SEQ ID NO: 2, wherein at least one selected from among 382 th serine and 489 th tyrosine of the amino acid sequence of SEQ ID NO: 2 It may consist of a peptide having at least 10 amino acids consisting of one amino acid residue and contiguous amino acids of these selected amino acid residues.
  • the fragment of the IK factor may be composed of a peptide having 10 or more amino acids in which two or more adjacent amino acids are linked to the N-terminus and C-terminus of the selected amino acid, respectively.
  • the fragment of the IK factor may be composed of a peptide having at least one amino acid sequence selected from the group consisting of SEQ ID NO: 4, SEQ ID NO: 47, SEQ ID NO: 49.
  • compositions containing the IK factor or fragments of IK factor can be used to treat rheumatoid arthritis.
  • the IK factor or fragment of IK factor may be contained in the pharmaceutical composition at a concentration of 1.0 ng / mL to 10 ⁇ g / mL.
  • the present invention also provides a pharmaceutical composition for treating or preventing arthritis, comprising a gene carrier comprising a nucleic acid molecule encoding an IK factor or fragment of an IK factor.
  • the nucleic acid encoding a fragment of the IK factor includes a nucleic acid having a nucleotide sequence of SEQ ID NO: 3 or a partial nucleic acid of SEQ ID NO: 3.
  • the nucleic acid encoding a fragment of the IK factor is at least one selected from nucleotide residues 1144-1146, 1465-1467 nucleotide residues, and 1474-1476 nucleotide residues of the nucleotide sequence of SEQ ID NO: 1. It may consist of a nucleic acid having at least 30 nucleotides consisting of one nucleotide residue and adjacent nucleotides of these selected nucleotide residues.
  • the nucleic acid encoding a fragment of the IK factor comprises at least one nucleotide residue selected from the set of 1144-1146 nucleotides and the set of 1465-1467 nucleotides of the nucleotide sequence of SEQ ID NO: 1. And nucleic acids having 30 or more nucleotides consisting of contiguous nucleotides of these selected nucleotide residues.
  • the nucleic acid encoding a fragment of the IK factor consists of a nucleic acid having at least 30 nucleotides each having at least 6 contiguous nucleotides linked to the 5 'and 3' ends of the selected nucleotide residue. Can be.
  • the nucleic acid encoding the fragment of the IK factor encodes a peptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 4, SEQ ID NO: 47 and SEQ ID NO: 49. And a nucleic acid having a nucleotide sequence.
  • the nucleic acid encoding the fragment of the IK factor may be composed of a nucleic acid having at least one nucleotide sequence selected from the group consisting of SEQ ID NO: 3, SEQ ID NO: 46 and SEQ ID NO: 48. have.
  • Nucleic acids encoding the IK factor or fragments of the IK factor can be used to treat rheumatoid arthritis.
  • the nucleic acid encoding the IK factor or fragment of IK factor may be contained in the pharmaceutical composition at a concentration of 1.0 ng / mL to 10 ⁇ g / mL.
  • the gene carrier may have the form of a naked nucleic acid molecule, a plasmid, a viral vector and a liposome or niosome containing the plasmid or the viral vector.
  • the viral vector may be selected from the group consisting of adenovirus, adeno-associated virus, retrovirus, lentivirus, baculovirus, herpes simplex virus, vaccinia virus.
  • the present invention also relates to an IK factor or fragment thereof for treating arthritis, a nucleic acid encoding an IK factor or a fragment thereof, and / or a vector or gene carrier having these nucleic acids.
  • the present invention also provides a step of administering an IK factor or fragment of IK factor, a nucleic acid encoding IK factor or a fragment thereof, and / or a vector or gene delivery carrier having these nucleic acids in a therapeutically effective amount or in a pharmaceutically effective amount. It includes a method of treating arthritis, which includes.
  • the IK factor or fragments thereof and / or nucleic acids encoding these peptides are involved in the upstream level of reaction mechanisms associated with arthritis, for example rheumatoid arthritis, thereby promoting the development of rheumatoid arthritis.
  • the IK factor or fragment thereof and / or nucleic acid encoding them used as an active ingredient according to the present invention can be secured because it is an immune balance modulator in the human body that overall regulates the activity of immune cells affecting arthritis. have.
  • the IK factor or fragment thereof is an endogenous substance produced in the human body, it has low immunogenicity and does not significantly affect the entire population of immune cells, and therefore, long-term administration is expected to have no side effects.
  • HA in Figure 1 represents a hemagglutinin tag (haemagglutinin) tag.
  • FIG. 2 schematically shows the position of nucleotides constituting a nucleic acid fragment (tIK nucleic acid fragment) in which some nucleotides are cleaved relative to a nucleotide encoding a full-length IK factor according to an exemplary embodiment of the present invention.
  • tIK nucleic acid fragment a nucleic acid fragment
  • FIG. 2 shows the positions of the amino acids and substituted amino acids of the active sites predicted to be involved in the physiological function of the IK factor relative to the construction of the variant IK factor relative to the exemplary amino acid sequence of the full length IK factor.
  • HA in Figure 2 represents a hemagglutinin (haemagglutinin) tag (tag).
  • Figure 3 shows MHC by expression of a truncated nucleic acid fragment of IK factor (tIK nucleic acid fragment) and mutant nucleic acid fragments (S382A, Y489F, Y492F, Y489492F, S382AY489492F) in which some nucleotides of the tIK nucleic acid fragment are substituted with other nucleotides.
  • This is a graph measuring the degree of inhibition of the expression of CNOT1, CDCA3 and MAPK1, which are higher regulators that inhibit the expression of CIITA, a class II transactivator.
  • * means P ⁇ 0.05 as compared to negative control
  • # means P ⁇ 0.05 when mutant nucleic acid fragments are expressed as compared with wild type tIK.
  • Figure 4a is a mouse produced by crossing the IL-1 Receptor antagonist knock-out (IL1RaKO) mouse with arthritis naturally induced, IL1RaKO mouse and transgenic mice expressing some cleaved IK factor (tIK) according to the present invention (tIK-IL1RaKO) is a graph measuring arthritis index over time.
  • IL1RaKO IL-1 Receptor antagonist knock-out
  • tIK-IL1RaKO transgenic mice expressing some cleaved IK factor according to the present invention
  • Figure 4b is a graph measuring the incidence of arthritis over time in IL1RaKO mice and tIK-IL1RaKO mice.
  • FIG. 5 is a photograph of the joint region of IL1RaKO mice naturally induced arthritis, and tIK-IL1RaKO mouse produced according to the present invention.
  • Figure 6a shows the results of measurement of arthritis-derived joint tissue from IL1RaKO mice and tIK-IL1RaKO mice produced according to the present invention using H & E staining.
  • the left side is a micrograph of the joint tissue, and the right side is a graph analyzing the degree of inflammation in the joint tissue.
  • * means P ⁇ 0.05 as compared to negative control.
  • FIG. 6B shows the results of measurement of joint tissue derived from IL1RaKO mice naturally induced arthritis and tIK-IL1RaKO mice prepared according to the present invention using safranin O staining.
  • the left side is a micrograph of the joint tissue and the right side is a graph analyzing the degree of cartilage erosion in the joint tissue.
  • * means P ⁇ 0.05 as compared to negative control.
  • Figure 7 is a photograph of the degree of bone damage in the joint area of IL1RaKO mice naturally induced arthritis, and tIK-IL1RaKO mouse produced according to the present invention.
  • Figure 8a is a graph measuring the mRNA expression level of inflammatory cytokines in the joint region of IL1RaKO mice naturally induced arthritis and tIK-IL1RaKO mouse prepared according to the present invention.
  • * denotes P ⁇ 0.05 and ** denotes P ⁇ 0.01 compared to negative control.
  • Figure 8b is a photograph of the degree of expression of inflammatory cytokines after staining joint tissue of IL1RaKO mice naturally induced arthritis, and tIK-IL1RaKO mice prepared according to the present invention.
  • FIG. 9 is a graph measuring the mRNA expression level of inflammatory cytokines in serum of IL1RaKO mice naturally induced arthritis and tIK-IL1RaKO mice prepared according to the present invention. ** in FIG. 9 means P ⁇ 0.01 compared to negative control.
  • Figure 10a is to examine the degree of differentiation of etiological T cells in spleen cells of IL-1 Receptor antagonist knock-out (IL1RaKO) mice and arthritis induced naturally, according to the invention (tIK-IL1RaKO), Plots and graphs measuring the amount of Th1 cells, which are pathogenic T cells.
  • Figure 10b is to examine the degree of differentiation of etiological T cells in spleen cells of arthritis induced IL-1 Receptor antagonist knock-out (IL1RaKO) mouse, and mouse (tIK-IL1RaKO) produced according to the present invention, Plots and graphs measuring the amount of Th17 cells, the etiological T cells.
  • IL1RaKO arthritis induced IL-1 Receptor antagonist knock-out
  • tIK-IL1RaKO mouse
  • Figure 11 is a graph measuring the amount of macrophage activator factor in splenocytes of IL-1 Receptor antagonist knock-out (IL1RaKO) mice in which arthritis is naturally induced and mice prepared according to the present invention (tIK-IL1RaKO). .
  • * means P ⁇ 0.05 as compared to negative control.
  • Figure 12a is a graph measuring the expression level of inflammatory cytokines in cells after transfection of macrophages with peptides containing amino acids involved in the activity of IK factor in accordance with the present invention.
  • * denotes P ⁇ 0.05 and ** denotes P ⁇ 0.01 compared to negative control.
  • Figure 12b is a graph measuring the expression level of inflammatory cytokines in cell culture after transfection of macrophages with peptides containing amino acids involved in the activity of IK factor according to the present invention.
  • Figure 13a is a plot and a graph analyzing the degree of differentiation of Th0 cells in spleen cells of wild-type mice injected with peptides containing amino acids involved in the activity of IK factor according to the present invention.
  • Figure 13b is a plot and graph analyzing the degree of differentiation of Th1 cells in spleen cells of wild-type mice injected with a peptide containing amino acids involved in the activity of tIK according to the present invention.
  • Figure 13c is a plot and graph analyzing the degree of differentiation of Th17 cells in spleen cells of wild-type mice injected with a peptide containing amino acids involved in the activity of tIK according to the present invention.
  • Figure 14a is a graph measuring the arthritis index over time after injecting the IK factor-derived peptide into IL1RaKO mice, a naturally occurring arthritis model according to the present invention
  • Figure 14b is arthritis over time in the mouse It is a graph measuring the incidence rate.
  • Figure 15a is a plot and graph of the differentiation of Th17 cells in spleen cells of the mouse after injecting a peptide derived from IK factor in accordance with the present invention into a mouse that is a naturally occurring arthritis model.
  • * means P ⁇ 0.05 as compared to negative control.
  • Figure 15b is a photograph of the degree of expression of inflammatory cytokines after staining the joint tissue of a mouse, a naturally occurring arthritis model injected with IK factor-derived peptides according to the present invention.
  • Figure 16 is a SDS-PAGE measurement photograph confirming that the IK factor was expressed after injecting a truncated IK gene fragment (tIK) having the Fc tag sequence added to the 3 'end into insect cells using a Baculovirus expression system. .
  • tIK truncated IK gene fragment
  • FIG. 17 shows the IK factor in recombinant adeno-associated virus (tIK-AAV) in which a truncated IK gene fragment (a gene fragment encoding tIK) having an HA tag sequence added to its 5 'end was inserted into an adeno-associated virus.
  • Western-blotting results show expression.
  • Figure 18a is a graph measuring the arthritis index over time after injecting the adeno-associated virus vector inserted with the truncated IK gene fragment (gene fragment encoding tIK) into IL1RaKO mice, a naturally occurring arthritis model
  • 18b is a graph measuring the incidence of arthritis over time in the mouse.
  • FIG. 19 is a Western-blotting result showing that the plasmid inserted with the truncated IK gene fragment (tIK) having the HA tag sequence added to the 5 'end was injected into CHO cells, and that the IK factor was expressed in CHO cells. .
  • tIK truncated IK gene fragment
  • the present inventors have studied new functions of IK factor, a type of cytokine expressed in vivo, so that gene carriers having IK factors or fragments thereof and / or nucleic acids encoding them are for example rheumatoid arthritis.
  • the present invention was completed by clarifying that the progression of arthritis as described above. EMBODIMENT OF THE INVENTION
  • this invention is demonstrated in detail, referring an accompanying drawing.
  • amino acid is used herein in its broadest sense and is intended to include naturally-occurring L-amino acids or residues. 1- and 3-letter abbreviations commonly used for naturally-occurring amino acids are used herein (Lehninger, Biochemistry, 2d ed., Pp. 71-92, (Worth Publishers: New York, 1975)). Amino acids are chemicals having properties known in the art to be characteristic of not only D-amino acids but also chemically-modified amino acids such as amino acid analogs, naturally-occurring amino acids such as norleucine, and amino acids that are not commonly incorporated into proteins.
  • synthetically-synthesized compounds include, for example, analogs or mimetics of phenylalanine or proline that allow conformational limitations of the same peptide compound as natural phenylalanine (Phe) or proline (Pro).
  • Analogs and mimetics are referred to herein as "functional equivalents" of amino acids
  • Other examples of amino acids are described in Roberts and Vellaccio, The Peptide. s: Analysis, Synthesis, Biology, Eds. Gross and Meiehofer, Vol. 5, p. 341 (Academic Press, Inc .: N.Y. 1983).
  • amino acid analogs amino acids not encoded by the genetic code are referred to herein as "amino acid analogs" and are described, for example, in WO 90/01940.
  • amino acid analogs include 2-amino adipic acid (Aad) for Glu and Asp; 2-aminopimelic acid (Apm) for Glu and Asp; 2-aminobutyric acid (Abu) for Met, Leu and other aliphatic amino acids; 2-aminoheptanoic acid (Ahe) for Met, Leu and other aliphatic amino acids; 2-aminobutyric acid (Aib) for Gly; Cyclohexylalanine (Cha) for Val, Leu and Ile; Homoarginine (Har) for Arg and Lys; 2,3-diaminopropionic acid (Dap) for Lys, Arg and His; N-ethylglycine (EtGly) for Gly, Pro and Ala; N-ethylglycine (EtGly) for Gly, Pro and Ala; N-ethylasparagine (EtAsn) for Asn and Gln; Hydr
  • peptide includes all proteins, protein fragments and peptides that have been isolated from naturally occurring, synthesized by a recombinant technique or chemically synthesized.
  • the peptide of the present invention consists of at least 5, preferably 10 amino acids.
  • variants of a compound are provided, such as peptide variants having one or more amino acid substitutions.
  • peptide variants refers to the original amino acid while one or more amino acids are substituted, deleted, added and / or inserted into the amino acid sequence of the peptide. It is said to exert almost the same biological function as the peptide consisting of.
  • Peptide variants should have at least 70% identity, preferably at least 90% and more preferably at least 95% identity with the original peptide.
  • substituents may include amino acid substituents known as "conservatives”.
  • Variants may also include nonconservative changes.
  • the sequence of the variant polypeptide differs from the original sequence by replacing, deleting, adding or inserting five or fewer amino acids.
  • Variants may also be altered by deletion or addition of amino acids with minimal impact on the immunogenicity, secondary structure and hydropathic nature of the peptide.
  • “conservative” substitutions herein means that there is no significant change in properties such as secondary structure and hydropathic nature of the polypeptide even when one amino acid is substituted with another amino acid.
  • Amino acid variations with respect to such conservative substitutions may be attributed to the relative similarity of amino acid side chain substituents such as polarity, charge, solubility, hydrophobicity, hydrophilicity and / or amphipathic nature. Can be obtained based on similarity.
  • amino acids are based on common side chain properties: 1) hydrophobic (leucine, methionine, alanine, valine, leucine, isoleucine) 2) neutral hydrophilic (cysteine, serine, threonine, asparagine, glutamine), 3) acidic (aspartic acid, Glutamic acid), 4) basic (histidine, lysine, arginine), 5) residues that affect chain orientation (glycine, proline), and 6) aromatics (tryptophan, tyrosine, phenylalanine).
  • Conservative substitutions will entail exchanging a member of one of each of these classes for another member of the same class.
  • arginine, lysine and histidine are all positively charged residues; Alanine, glycine and serine have similar sizes; It can be seen that phenylalanine, tryptophan and tyrosine have a similar shape.
  • arginine, lysine and histidine; Alanine, glycine and serine; Phenylalanine, tryptophan, and tyrosine are biologically equivalent functions.
  • hydropathic idex of amino acids can be considered.
  • Each amino acid is assigned a hydrophobicity index depending on its hydrophobicity and charge: isoleucine (+4.5); Valine (+4.2); Leucine (+3.8); Phenylalanine (+2.8); Cysteine / cysteine (+2.5); Methionine (+1.9); Alanine (+1.8); Glycine (-0.4); Threonine (-0.7); Serine (-0.8); Tryptophan (-0.9); Tyrosine (-1.3); Proline (-1.6); Histidine (-3.2); Glutamic acid (-3.5); Glutamine (-3.5); Aspartic acid (-3.5); Asparagine (-3.5); Lysine (-3.9); And arginine (-4.5).
  • the hydrophobic amino acid index is very important in conferring the interactive biological function of proteins. It is known that substitution with amino acids having similar hydrophobicity indexes can retain similar biological activity. When introducing mutations with reference to the hydrophobicity index, substitutions are made between amino acids which exhibit a hydrophobicity index difference of preferably within ⁇ 2, more preferably within ⁇ 1, even more preferably within ⁇ 0.5.
  • polynucleotide or “nucleic acid” is used interchangeably and refers to a polymer of nucleotides of any length and encompasses DNA (such as cDNA) and RNA molecules inclusively.
  • the “nucleotide”, a structural unit of a nucleic acid molecule, can be incorporated into a polymer by deoxyribonucleotides, ribonucleotides, modified nucleotides or nucleotides, and / or analogs thereof, or by DNA or RNA polymerase, or by synthetic reactions. May be any substrate present.
  • Polynucleotides may include modified nucleotides, sugars or analogs in which the nucleotides are modified, such as methylated nucleotides and analogs thereof (Scheit, Nucleotide Analogs, John Wiley, New York (1980); Uhlman and Peyman, Chemical Reviews , 90: 543-584 (1990).
  • nucleotides do not result in mutations in proteins.
  • Such nucleic acids include nucleic acid molecules comprising a codon that is functionally equivalent or a codon that encodes the same amino acid, or a codon that encodes a biologically equivalent amino acid.
  • variations in nucleotides may result in changes in the protein itself. Even in the case of mutations that bring about changes in amino acids of proteins, those exhibiting almost the same activity as the proteins of the present invention can be obtained.
  • the peptides and nucleic acid molecules of the present invention are amino acid sequences or nucleotide sequences listed in the sequence listing. It should be appreciated by those skilled in the art that the present invention is not limited thereto.
  • the biological functional equivalents that may be included in the peptide of the present invention may be a peptide having a variation in the amino acid sequence exhibiting biological activity equivalent to that of the peptide of the present invention.
  • the peptides (including fusion proteins) and polynucleotides mentioned herein are isolated.
  • An “isolated” peptide or polynucleotide is one that is removed from its original environment. For example, proteins that exist in nature are separated by removing some or all of the substances that exist together in that state. Such polypeptides should be at least 90% pure, preferably at least 95%, more preferably at least 99% pure.
  • Polynucleotides are isolated by cloning in a vector.
  • a vector refers to a construct that is capable of delivery to a host cell and preferably enables expression of one or more desired genes or sequences.
  • a vector may include viral vectors, DNA or RNA expression vectors, plasmids, cosmids or phage vectors, DNA linked to cationic condensing agents, or RNA expression vectors, DNA or RNA expression vectors packaged in liposomes, specific eukaryotic cells such as producer cells, and the like.
  • expression control sequence refers to a nucleic acid sequence that controls the transcription of a nucleic acid.
  • Expression control sequences include promoters or enhancers, such as constitutive promoters or inducible promoters.
  • the expression control sequence is linked to the nucleic acid sequence to be transcribed.
  • operatively linked refers to a functional binding between a nucleic acid expression control sequence (eg, an array of promoters, signal sequences, or transcriptional regulator binding sites) and other nucleic acid sequences, The regulatory sequence thereby controls the transcription and / or translation of the other nucleic acid sequence.
  • a nucleic acid expression control sequence eg, an array of promoters, signal sequences, or transcriptional regulator binding sites
  • the term "pharmaceutically effective amount” or “therapeutically effective amount” means an amount sufficient to achieve the efficacy or activity of the active ingredient peptide or fragments thereof and / or nucleic acids encoding them.
  • a pharmaceutical composition containing a gene carrier comprising a peptide according to the present invention or a nucleic acid encoding these peptides can be used to treat and / or treat chronic arthritis, including gouty arthritis, polyarthritis, deformable arthritis, including rheumatoid arthritis. Can be applied to prevent.
  • IK factor is a cytokine that inhibits the expression of major histocompatibility complex II (MHC II) on the surface of antigen presenting cells (APCs) capable of presenting antigens to CD4 T cells.
  • MHC II major histocompatibility complex II
  • APCs antigen presenting cells
  • Cain. IK factor is known to activate cAMP to inhibit the expression of MHC class II expressed by interferon-gamma (IFN- ⁇ , interferon- ⁇ ) induced by viral infection.
  • IFN- ⁇ interferon-gamma
  • the full-nucleic acid encoding the IK factor has a Nuclear Localization Signal Sequence (NLS) and a trimeric coiled-coil motif in the N-terminal (5 'end) region. have.
  • the C-terminal (3 'end) region of the full-length nucleic acid encoding the IK factor repeats the Arg (R) -Asp (D) and Arg (R) -Glu (E) sequences found mainly in nuclear proteins. It has a RED domain and three NLSs.
  • Exemplary full-nucleic acid encoding an IK factor is a polynucleotide consisting of 1,674 nucleotides represented by SEQ ID NO: 1.
  • the IK factor may be expressed except for the last transcription termination codon of SEQ ID NO: 1, and may consist of 557 amino acids of SEQ ID NO: 2.
  • the IK factor that can be used in the present invention and the IK nucleic acid encoding the same are not limited to those listed in the sequence listing.
  • Joints are organically connected between the bone and bone of the synovial membrane, cartilage, bone, synovial fluid, and is a complex tissue that affects each other through cytokines. Symptoms such as chronic inflammation, tissue necrosis, cell infiltration, neovascularization, and joint destruction in joints are known as the major etiology of rheumatoid arthritis. Synovial tissue called pannus, known as an essential pathology for the development of rheumatoid arthritis, invades tissue around the joint and causes bone resorption. In Pannus, antigen-presenting cells and T cells are observed at a very high density. The interaction of these cells is known to initiate and widen the immune response induced by T cells.
  • antigen-presenting cells expressing MHC class II are observed in Pannus patients with rheumatoid arthritis, and HLA-DR-positive cells and T cells are located at the same site. Therefore, to treat rheumatoid arthritis, it may be effective to suppress the interaction between antigen-presenting cells expressing MHC class II and T cells.
  • IL-17 + T cells and functionally activated IL-17 in the synovial membrane, a lesion of rheumatoid arthritis.
  • IL-17 is highly detected in the serum of rheumatoid arthritis patients, among which IL-17A induces the expression of IL-1 ⁇ and IL-6 in synovial cells of rheumatoid arthritis patients.
  • IL-17 inhibits the production of matrix in chondrocytes and osteoblasts, causing joint damage and deficient tissue regeneration.
  • IL-17 also activates the expression and function of matrix metalloproteinases (MMPs) and causes irreversible cartilage damage in mouse models with tumor necrosis factor (TNF).
  • MMPs matrix metalloproteinases
  • Th17 cells and synovial cells in the inflammatory response promotes the secretion of MMPs and expression of IL-1 ⁇ and IL-6 in synovial cells.
  • IL-17 amplifies RANK signals in osteoclasts by increasing the expression of receptor activator of NF- ⁇ B ligand (RANKL) on osteoblasts.
  • RANKL NF- ⁇ B ligand
  • Th17 cells expressing RANKL are known to play an important role in the osteoclast differentiation process.
  • the nucleic acid encoding the IK factor or fragment thereof can inhibit and prevent the progression of arthritis.
  • these nucleic acids inhibit the secretion of cytokines associated with rheumatoid arthritis and block the differentiation of immune cells.
  • a nucleic acid fragment encoding a fragment of an IK factor eg, an N-terminally truncated fragment (which may be abbreviated herein as "tIK factor” or "tIK”) tIK nucleic acid fragments) can be expressed in an appropriate expression system.
  • These nucleic acid fragments are expressed to inhibit the expression of class II MHC (MHC II) involved in the inflammatory immune response, one of the main symptoms of arthritis.
  • MHC II class II MHC
  • a tITA nucleic acid fragment in which some nucleotides are cleaved and / or a mutant tIK nucleic acid fragment in which some nucleotides are substituted is involved in the expression of MHC II (class II, major histocompatibility complex).
  • MHC II class II, major histocompatibility complex
  • CNOT1 CCR4-NOT transcription complex subunit 1
  • MAPK1 miogen-activated protein kinase 1
  • mutant tIK substituted with some nucleotides was found to be able to halve this effect, confirming that a specific site in the IK factor is a physiologically active site involved in the expression of a higher regulatory factor of MHC II.
  • tIK-IL1RaKO metastatic mice induced to express fragments of the IK factor, e.g., tIK, a truncated fragment of the N-terminus
  • tIK-IL1RaKO significantly reduced the incidence of arthritis
  • FIGS. 4A, 4B and 5 the degree of inflammation or cartilage damage in the articular tissue of tIK-IL1RaKO mice was significantly reduced in the articular tissues of IK1RaKO mice (see FIGS. 6A and 6B), and the degree of bone damage was also greatly reduced (see FIG. 7).
  • tIK-IL1RaKO mice have significantly reduced secretion of cytokines associated with arthritis in joint tissues and serum compared to IL1RaKO mice.
  • the metastatic mouse (tIK-IL1RaKO) induced to express the cleaved IK factor (tIK factor) is compared to the arthritis-induced mice (IL-1 antagonist-receptor knockout mouse, IL1RaKO mouse).
  • IL-1 antagonist-receptor knockout mouse IL-1 antagonist-receptor knockout mouse
  • the nucleic acid encoding a fragment of the IK factor is expressed through an appropriate expression system and is a cytokine involved in the inflammatory immune response associated with arthritis Interleukin-1 beta, IL-1 ⁇ . ), Expression of monocyte chemotactic protein-1 (MCP-1), also known as interleukin-6 (IL-6), interleukin-17 (IL-7), and CCL2 (Chemokine (CC motif) ligand 2). Suppress These results indicate that the injection of nucleic acid encoding an IK factor or fragment thereof into the human body through an appropriate vector or gene transporter can inhibit the progression of arthritis, and in particular can affect the systemic immune response.
  • MCP-1 monocyte chemotactic protein-1
  • IL-6 interleukin-6
  • IL-7 interleukin-17
  • CCL2 Chemokine (CC motif) ligand 2
  • the metastatic mouse (tIK-IL1RaKO) induced to express the IK factor suppresses the differentiation of T cells, which are arthritis diseases, as compared to naturally induced arthritis-induced mice (IL1RaKO) mice (see FIGS. 10A and 10B). It also affects the activator of macrophages (see FIG. 11).
  • relatively small size IK factor fragments comprising an active site, such as peptides before and after the active site and 10 amino acids adjacent thereto, can also inhibit the progression of arthritis.
  • These peptides inhibit the expression of tumor necrosis factor-alpha (TNF- ⁇ ), IL-6, IL-17, inflammatory cytokines in macrophages (see FIGS. 12A, 12B and 15B) and are involved in arthritis It also inhibits the differentiation of Th1 cells and Th17 cells, which are pathogenic cells (see FIGS. 13A-13C).
  • TNF- ⁇ tumor necrosis factor-alpha
  • IL-6 IL-6
  • IL-17 inflammatory cytokines in macrophages
  • Th1 cells and Th17 cells which are pathogenic cells
  • FIGS. 13A-13C are pathogenic cells.
  • These peptides also inhibit arthritis in IL1RaKO mice, naturally occurring arthritis induced mice (see FIGS. 14A and 14B), as well as inhibit the differentiation of Th17 cells in arthritis induced mice (see
  • the nucleic acid encoding the IK factor or fragment thereof can be injected using an appropriate expression system or gene delivery vehicle, which can prevent the progression of arthritis or treat the induced arthritis (FIGS. 19A and 19B). Reference).
  • IL-17 which is secreted according to the differentiation of Th17 cells, plays an important role in rheumatoid arthritis.
  • the IK factor or fragment thereof and / or nucleic acid encoding them inhibits the differentiation of Th17 cells, which has been noted as the root cause of rheumatoid arthritis, which makes treatment much more efficient than conventional arthritis therapeutics. .
  • the present invention therefore relates to an IK factor or fragment thereof for treating arthritis.
  • an active ingredient for treating arthritis preferably, some amino acids of the full-length IK factor are fragments of the cleaved IK factor, more preferably fragments of the N-terminal amino acids of the full-length IK factor are cleaved.
  • the full-length IK factor may be a peptide having an amino acid sequence of SEQ ID NO: 2.
  • the sequence of SEQ ID NO: 4 having 242 amino acids of the C-terminal It may be a peptide such as truncated IK (tIK) having an amino acid sequence or a partial peptide of SEQ ID NO: 4.
  • fragments of the IK factor peptide fragments whose N-terminus are cut out among the amino acids constituting the full-length IK factor are not only easy to manufacture because they are relatively small, but also in vivo and / or in vivo. It may be advantageous to exert biological functions in comparison to full-length IK factor ex vivo.
  • amino acid serine (S382), 489 th (SEQ ID NO: 4 peptide) of the 382th (67th in the peptide of SEQ ID NO: 4), the peptide of SEQ ID NO: 174th) amino acid tyrosine (Y489) and / or 492th (177th in peptide of SEQ ID NO: 4) amino acid tyrosine (Y492) is involved in the biological activity of the IK factor and / or fragments thereof.
  • the 382th amino acid serine (S382) and / or the 489th amino acid tyrosine (Y489) in the peptide of SEQ ID NO: 2 is predicted to be the active site of the IK factor and / or fragment of the IK factor.
  • fragments of relatively short length IK factor containing amino acids of these active sites can be used.
  • fragments of the IK factor that can be used in accordance with the present invention include the serine (S382), which is the 382th amino acid, the tyrosine (Y489), which is the 489th amino acid, and the tyrosine (Y489), which is the 492th amino acid, in the peptide of SEQ ID NO: 2.
  • the fragment of the IK factor is at least one selected from the 82nd amino acid serine (S382), the 489th amino acid tyrosine (Y489) and the 492th amino acid tyrosine (Y489) in the peptide of SEQ ID NO: 2 And a peptide consisting of 10 or more and 100 or less, preferably 10 or more and 50 or less, more preferably 10 or more and 30 or less amino acids, each consisting of the amino acid residues of the selected amino acid residues and adjacent amino acids of these selected amino acid residues. do. At least one amino acid of these selected amino acid residues may be a modified amino acid such as phosphorylation.
  • Exemplary fragments of the IK factor that may be used in accordance with the present invention are peptides or partial peptides thereof having the amino acid sequence of SEQ ID NO: 2.
  • the fragment of the IK factor consists of at least one amino acid residue selected from serine, which is the 382th amino acid, and tyrosine, the 489th amino acid, in the peptide of SEQ ID NO: 2 and adjacent amino acids of these selected amino acid residues. It is a peptide which has 10 or more, for example 10 or more and 100 or less, preferably 10 or more and 50 or less, more preferably 10 or more and 30 or less amino acids.
  • peptides that can be used as fragments of the IK factor include the serine (S382), the 382th amino acid, the tyrosine (Y489), the 489th amino acid, and the tyrosine (Y492), the 492th amino acid, in the peptide of SEQ ID NO: 2.
  • An active amino acid such as may not be located at the N-terminus or C-terminus of the peptide.
  • Peptides may be used in which two or more adjacent amino acids are linked to each of the N-terminus and C-terminus of these selected active amino acids so that these selected amino acids that are active amino acids can be arranged in the middle of the peptide.
  • 2 to 8 preferably 3 to 7 or more adjacent amino acids may be linked to the N-terminus and C-terminus of the active amino acid, respectively.
  • 2 to 100 such as 3 to 70, preferably 3 to 30, more preferably 3 to 20 contiguous amino acids may be linked to the N-terminus and C-terminus of the active amino acid, respectively.
  • the fragment of the IK factor is a peptide of SEQ ID NO: 47 consisting of 377 amino acid glutamic acid-391 amino acid aspartic acid in the peptide of SEQ ID NO: 2 and / or the 484th Also included is a peptide of SEQ ID NO: 49 consisting of the amino acid aspartic acid—the 496th amino acid lysine.
  • the full length IK factor of SEQ ID NO: 2 consists of 557 amino acids
  • the fragment of the IK factor of SEQ ID NO: 4, wherein the N-terminus of the IK factor is cleaved is 242 amino acids. It consists of.
  • the fragment of IK factor according to the present invention may be composed of up to about 300 to 500 amino acids of the amino acid of SEQ ID NO: 2.
  • fragments of IK factor that can be used in accordance with the present invention are not limited thereto.
  • the remaining amino acids may be substituted with other amino acids, for example by conservative substitutions, If necessary some amino acids may be deleted, added and / or inserted.
  • Peptides that are IK factors or fragments thereof can be isolated by preparation through recombinant means or chemical synthesis.
  • peptides expressed by the nucleic acid sequences mentioned herein can be readily prepared by known methods, using any of a number of known expression vectors. Expression can be realized in a suitable host cell transformed with an expression vector comprising a DNA sequence encoding the peptide.
  • suitable host cells include prokaryotes, yeasts and eukaryotes. E. coli, yeast or mammalian cell lines (such as Cos or CHO) are preferably used as host cells.
  • the supernatant containing recombinant protein secreted into the culture is concentrated using a commercially available filter.
  • the concentrate obtained above is purified using an appropriate purification matrix such as an affinity matrix or an ion exchange resin.
  • pure protein can be obtained by performing one or several reverse phase HPLC.
  • Fragments or variants consisting of up to 100, typically up to 50 amino acids can be prepared synthetically.
  • polypeptides can be synthesized by commercially available solid-phase techniques, the Merrifield solid-phase synthesis method, which sequentially adds amino acids to growing amino acid chains ( Merrifield, 1963, J. Am. Chem. Soc. 85: 2146-2149.
  • Equipment for automatic synthesis of polypeptides can be purchased from the supplier and can be manipulated according to the supplier's manual.
  • peptides described herein expressed in a microorganism can be secreted into and recovered from the periplasm of the host cell.
  • protein recovery generally involves grinding the microorganism by means such as osmotic shock, sonication or lysis. Once the cells are destroyed, cell debris or whole cells can be removed by centrifugation or filtration. Proteins can be further purified, for example, by affinity resin chromatography. Alternatively, the protein can be transferred to and separated from the culture medium. The cells can be removed from the culture and the culture supernatant filtered and concentrated to further purify the protein produced.
  • Expressed polypeptides are commonly known methods, such as fractional distillation on immunoaffinity or ion-exchange columns; Ethanol precipitation; Reverse phase HPLC; Chromatography on silica or cation exchange resins such as DEAE; Chromatofocusing; SDS-PAGE; Ammonium sulfate precipitation; Gel filtration using, for example, Sephadex G-75; Hydrophobic affinity resins, ligand affinity using suitable antigens immobilized on a matrix, and Western blot assays can be further isolated and identified.
  • the peptides produced can be purified to obtain a substantially homogeneous formulation for further assays and uses.
  • Standard protein purification methods known in the art can be used.
  • the following procedures are examples of suitable purification procedures: fractional distillation on immunoaffinity or ion-exchange columns, ethanol precipitation, reverse phase HPLC, silica or cation-exchange resins, such as chromatography on DEAE, chromatographic focusing, SDS-PAGE, ammonium sulfate Precipitation, and gel filtration using, for example, Sephadex G-75.
  • the invention also relates to a nucleic acid or polynucleotide encoding an IK factor or fragment thereof for treating arthritis. Similar to peptides, preferably the IK nucleic acid fragments from which some nucleotides of the nucleic acid encoding the full length IK are cleaved are preferred, and particularly preferably the IK nucleic acid fragments from which the nucleotides of the 5 'end are cleaved.
  • the nucleic acid encoding the full-length IK factor may be a nucleic acid having a nucleotide sequence of SEQ ID NO: 1.
  • a nucleic acid encoding a fragment of an IK factor is a truncated IK factor having a nucleotide sequence of SEQ ID NO: 3 wherein 945 nucleotides constituting the 5 'end of the nucleic acid of SEQ ID NO: 1 are cleaved. It may be a nucleic acid (tIK nucleic acid fragment) encoding a portion or a nucleic acid consisting of some nucleotides of SEQ ID NO: 3.
  • IK fragment nucleic acids having the nucleotide sequence of SEQ ID NO: 3
  • IK fragment nucleic acids consisting of nucleotides of relatively short length
  • the IK fragment nucleic acid according to the present invention is a nucleotide residue of 1144-1146 (SEQ ID NO: 199-201) of SEQ ID NO: 1, 1465-1467 (SEQ ID NO: 1) 520-522) nucleotide residues of No. 3, 1474-1476 (529-531 of SEQ ID NO: 3) of SEQ ID NO: 1 and at least 30 adjacent to these selected nucleotide residues, eg For example, it may consist of more than 300 or more than 600 nucleotides.
  • the nucleic acid encoding a fragment of the IK factor comprises a 1144-1146th nucleotide residue of SEQ ID NO: 1, a 1465-1467th nucleotide residue of SEQ ID NO: 1, Nucleotide residues of 1474-1476 and 30 or more and 300 or less, preferably 30 or more and 150 or less, more preferably 30 or more and 90 or less nucleotides each consisting of adjacent nucleotides of these selected nucleotide residues Nucleic acids to be included.
  • the IK fragment nucleic acid is a nucleic acid having a nucleotide sequence of SEQ ID NO: 1 or a partial fragment thereof.
  • the nucleic acid encoding the partial fragment of the IK factor is selected from at least one nucleotide residue selected from nucleotide residues 1144-1146 and 1465-1467 nucleotide residues of the nucleotide sequence of SEQ ID NO: 1; It is a nucleic acid having 30 or more, for example, 30 or more and 300 or less, preferably 30 or more and 150 or less, more preferably 30 or more and 90 or less nucleotides composed of contiguous nucleotides of a nucleotide set.
  • Six or more contiguous nucleotides may each be linked to the 5 'and 3' ends of these selected nucleotide sets so that the selected nucleotide residues can be arranged in the middle of the polynucleotide.
  • 6 to 24, preferably 9 to 21 or more contiguous nucleotides may be linked to the 5 'end and 3' end of the selected nucleotide residue.
  • 6-300, such as 9-210, preferably 9-90, more preferably 9-60 contiguous nucleotides can be linked to the 5 'end and 3' end of the selected nucleotide residue, respectively.
  • the nucleic acid encoding a fragment of the IK factor may be a nucleic acid having the nucleotide sequence of SEQ ID NO: 46 and / or a nucleic acid having the nucleotide sequence of SEQ ID NO: 48.
  • the nucleic acid encoding a fragment of the IK factor may be composed of up to about 900 to 1500 nucleotides among the nucleotides of SEQ ID NO: 1, but the present invention is not limited thereto.
  • amino acids of SEQ ID NO: 2 1144-1146 of SEQ ID NO: 1 coding for serine (S382), 489 amino acid tyrosine (Y489), and / or tyrosine (Y492) 492 amino acid, respectively.
  • the set of nucleotides, the set of 1465-1467 nucleotides and / or the set of 1474-1476 nucleotides may have different nucleotide sequences encoding serine and tyrosine, respectively.
  • nucleic acids encoding an IK factor or fragment thereof can be included in a suitable vector.
  • the vector also includes an expression control sequence linked to the polynucleotide of the present invention.
  • the vector may also comprise one or more polynucleotides encoding other molecules of interest. Polynucleotides of the present invention may also bind to other polynucleotides to encode fusion proteins.
  • the polynucleotides of the invention are formulated to enter into and express in mammalian cells. Such compositions are particularly useful for use for therapeutic purposes.
  • polynucleotides in host cells There are many ways to express polynucleotides in host cells, and any suitable method can be used.
  • one polynucleotide is an adenovirus, an adeno-associated virus, a retrovirus, a vaccinia, a lentivirus baculovirus or another. It can be inserted into viral vectors such as pox virus (eq, avian pox virus). Techniques for inserting DNA into such vectors are well known.
  • Retroviral vectors include targeting sites such as genes for selectable markers that facilitate the identification or selection of transduced cells and / or genes that encode ligands that act as receptors for specific target cells. can additionally insert a targeting moiety. Targeting can also be accomplished by known methods using antibodies.
  • Vectors available and known in the art can be used for the purposes of the present invention. Selection of the appropriate vector will depend primarily on the size of the nucleic acid inserted into the vector and the particular host cell transformed with the vector. Each vector contains various components depending on its function (amplification or expression of heterologous polynucleotides, or both) and compatibility with the particular host cell in which the vector is present. Vector components generally include, but are not limited to, origin of replication (especially when the vector is inserted into prokaryotic cells), selection marker genes, promoters, ribosomal binding sites (RBS), signal sequences, heterologous nucleic acid inserts, and transcription termination sequences. .
  • recombinant vectors of the present invention may be used to express expression control sequences that may affect the expression of proteins, such as initiation codons, termination codons, polyadenylation signals, enhancers, signal sequences for membrane targeting or secretion, and the like. It may include.
  • Polyadenylation signals increase the stability of transcripts or facilitate cellular transport.
  • Enhancer sequences are nucleic acid sequences that are located at various sites in the promoter and increase transcriptional activity as compared to the transcriptional activity by the promoter in the absence of the enhancer sequence.
  • the host when the host is Escherichia (Escherichia) in gyunin include PhoA signal sequence, OmpA signal sequence, etc., the host is Bacillus (Bacillus) when in gyunin include ⁇ - amylase signal sequence, subtilisin signal sequence, etc. Shin, If the host is a yeast, MF- ⁇ signal sequence, SUC2 signal sequence, etc., if the host is an animal cell, insulin signal sequence, a-interferon signal sequence, antibody molecular signal sequence, etc. may be used, but the present invention is limited thereto. It doesn't work.
  • vectors refers to a circular double stranded DNA loop into which additional DNA segments can be ligated therein.
  • a phage vector refers to a viral vector in which additional DNA segments can be ligated into the viral genome.
  • Certain vectors are capable of autonomous replication in the host cell into which they are introduced (eg, bacterial vectors and episomal mammalian vectors with bacterial origins of replication). Other vectors (eg, non-episomal mammalian vectors) can be integrated into the genome of the host cell upon introduction into the host cell and thereby replicate with the host genome.
  • certain vectors may direct expression of the gene to which the vector is operably linked. Such vectors are referred to herein as "recombinant expression vectors" (or simply, “recombinant vectors”). In general, expression vectors of utility in recombinant DNA techniques are often in the form of plasmids.
  • vectors that can be used in the present invention include plasmids often used in the art (eg pSC101, ColE1, pBR322, pUC8 / 9, pHC79, pUC19, pET, etc.), phage (eg ⁇ gt4 ⁇ B, ⁇ -Charon , ⁇ z1, ⁇ GEM.TM.-11 and M13, etc.) or viruses (eg SV40, etc.).
  • the nucleic acid is inserted into a host cell using a virus expression system (vaccinia or other pox virus, retrovirus, lentivirus, baculovirus, adenovirus or adeno-associated virus).
  • virus expression system vaccinia or other pox virus, retrovirus, lentivirus, baculovirus, adenovirus or adeno-associated virus.
  • viral vectors include retroviral vectors derived from HIV, SIV, murine retroviruses, gibbon ape leukemia virus, adeno-associate viruses, and adenoviruses, and the like. But not limited thereto (Miller et al., 1990, Mol. Cell Biol. 10: 4239; J. Kolberg 1992, NIH Res. 4:43; Cornetta et al., 1991, Hum. Gene Ther. 2: 215).
  • Vectors are widely used (Buchscher et al., 1992, J. Virol, 66 (5): 2731-2739; Johann et al., 1992, J. Virol. 66 (5): 1635-1640; Sommerfelt et al. , 1990, Virol. 176: 58-59; Wilson et al., 1989, J. Virol. 63: 2374-2378; Miller et al., 1991, J. Virol.
  • Constitutive or inducible promoters can be used in the present invention depending on the needs of a particular situation that can be identified by one skilled in the art. Many promoters that are recognized by a variety of possible host cells are well known.
  • the selected promoter may be operably linked to the cistron DNA encoding the polypeptides described herein by removing the promoter from the source DNA via restriction enzyme digestion and inserting the isolated promoter sequence into the selection vector.
  • Both native promoter sequences and multiple heterologous promoters can be used to direct amplification and / or expression of a target gene. However, heterologous promoters are generally preferred because they allow greater transcription and higher yield of expressed target genes as compared to native target polypeptide promoters.
  • a promoter derived from the mammalian cell genome for example, metallothionine promoter
  • a promoter derived from a mammalian virus for example, adeno Late viral promoter, vaccinia virus 7.5K promoter, SV40 promoter, cytomegalovirus promoter and tk promoter of HSV
  • a promoter derived from the mammalian cell genome for example, metallothionine promoter
  • a promoter derived from a mammalian virus for example, adeno Late viral promoter, vaccinia virus 7.5K promoter, SV40 promoter, cytomegalovirus promoter and tk promoter of HSV
  • the recombinant vector of the present invention when it is a replicable expression vector, it may include a replication origin, which is a specific nucleic acid sequence from which replication is initiated.
  • the recombinant vector may include a selection marker.
  • the selection marker is for selecting cells transformed with the vector, and markers conferring a selectable phenotype such as drug resistance, nutritional requirements, resistance to cytotoxic agents or expression of surface proteins can be used.
  • Vectors of the invention, as markers include antibiotic resistance genes commonly used in the art and include, for example, ampicillin, gentamicin, carbenicillin, chloramphenicol, streptomycin, kanamycin, geneticin, neomycin and There is a resistance gene for tetracycline.
  • selection markers include ura4, leu1, his3, and the like, which are nutritional markers, but the types of selection markers that can be used in the present invention are not limited by the above examples.
  • RNA polymerase chain reaction PCR
  • LCR ligand chain reaction
  • Q ⁇ -replicase amplification Q ⁇ -replicase amplification
  • RNA polymerases Sambrook et al., 1989, Molecular Cloning-A Laboratory Manual. (2nd Ed) 1-3; US Patent No. 4,683,202; PCR protocols A Guide to Methods and Applications, Innis et al., Eds.Academic Press Inc. San Diego, CA 1990.
  • Improved methods of cloning in vitro amplified nucleic acids are described in US Patent No. 5,426,039.
  • Vectors of the present invention may also be fused with other sequences to facilitate purification of the peptides expressed therefrom.
  • Sequences to be fused include, for example, glutathione S-transferase (Pharmacia, USA), maltose binding protein (NEB, USA), FLAG (IBI, USA) and 6x His (hexahistidine; Quiagen, USA), and most preferably Is 6 ⁇ His. Because of the additional sequence for this purification, the protein expressed in the host is purified quickly and easily through affinity chromatography. If desired, sequences encoding Fc fragments may be fused to facilitate extracellular secretion of these peptides.
  • peptides expressed by a vector containing a nucleotide sequence encoding an IK factor or partial fragment thereof are purified by affinity chromatography.
  • glutathione-S-transferase which is a substrate of this enzyme, can be used, and when 6x His is used, a desired peptide can be obtained using a Ni-NTA His-binding resin column (Novagen, USA). Can be obtained quickly and easily.
  • Host cells capable of continuously cloning and expressing the above-described vectors while being stable are known in the art and can use any host cell.
  • yeast Saccharomyce cerevisiae
  • insect cells such as SF9 cells
  • human cells such as CHO cell lines (Chinese hamster ovary), W138, BHK, COS-7, 293, HepG2, 3T3, RIN and MDCK cell lines) and the like.
  • the vectors of the invention can be used to genetically modify cells in vitro, ex vivo or in vitro .
  • Methods for genetically modifying cells include infection or transduction of cells with viral vectors, calcium phosphate precipitation, bacterial protoplasts containing DNA of receptor cells. Fusion methods, treatment of liposomes or microspheres containing DNA in receptor cells, DEAE dextran, receptor-mediated endocytosis, electroporation, microinjection Various methods are known, including micro-injection and gene bombardment.
  • the present invention provides a therapeutically effective amount of the aforementioned IK factor or fragment thereof; And it relates to a pharmaceutical composition for the treatment of arthritis comprising a pharmaceutically acceptable carrier as needed.
  • the IK factor or fragment thereof peptide can be administered directly to the subject.
  • the present invention provides a gene carrier having a nucleic acid molecule encoding an IK factor or fragment thereof; And it relates to a pharmaceutical composition for the treatment of arthritis comprising a pharmaceutically acceptable carrier as needed.
  • the use of gene carriers is intended for so-called gene therapy.
  • the present invention provides a method of administering to a subject a pharmaceutical composition or a pharmaceutical composition containing as an active ingredient a gene carrier comprising a nucleic acid encoding an IK factor or a fragment thereof and a nucleic acid encoding the IK factor or a fragment thereof. It relates to a method of treating arthritis having a step.
  • the active ingredient may be contained in the pharmaceutical composition at a concentration of 1.0 ng / mL to 10 ⁇ g / mL.
  • a pharmaceutical composition or medicament containing a compound of the invention and a therapeutically inert carrier, diluent or excipient, as well as a method of using the compound of the invention to make such a composition and medicament.
  • the compound may be formulated by mixing in a desired degree of purity at ambient temperature, appropriate pH, together with a physiologically acceptable carrier, that is, a carrier that is nontoxic to the recipient at the dosages and concentrations employed in the herbal dosage form.
  • the pH of the formulation mainly depends on the specific use and concentration of the compound, but is preferably in the range of about 3 to about 8.
  • the compound is formulated in acetate buffer at pH 5.
  • the compound is sterile.
  • the compound may be stored, for example, as a solid or amorphous composition, lyophilized formulation or aqueous solution.
  • compositions are formulated, dosed, and administered in a fashion consistent with good medical practice.
  • Factors to consider in this regard include the particular disorder to be treated, the particular patient to be treated, the clinical condition of the individual patient, the cause of the disorder, the site of delivery of the agent, the method of administration, the dosing schedule, and other factors known to the practitioner.
  • sustained-release preparations may be prepared. Suitable examples of sustained-release preparations include semipermeable matrices of solid hydrophobic polymers containing the compound, which matrices are in the form of shaped articles, eg films, or microcapsules. Examples of sustained-release matrices include polyesters, hydrogels (eg, poly (2-hydroxyethyl-methacrylate), or poly (vinyl alcohol)), polylactide, L-glutamic acid, and gamma-ethyl- Copolymers of L-glutamate, non-degradable ethylene-vinyl acetate, degradable lactic acid-glycolic acid copolymers and poly-D-(-)-3-hydroxybutyric acid.
  • sustained-release preparations include polyesters, hydrogels (eg, poly (2-hydroxyethyl-methacrylate), or poly (vinyl alcohol)), polylactide, L-glutamic acid, and gamma-ethyl- Copo
  • a pharmaceutically effective amount of a compound of the invention administered parenterally per dose will range from about 0.01-100 mg / kg, alternatively about 0.1 to 20 mg / kg, based on the patient's body weight per day, Typical initial ranges for compounds are 0.3 to 15 mg / kg / day.
  • oral unit dosage forms such as tablets and capsules preferably contain about 5-100 mg of the compound of the present invention.
  • the compounds used as active ingredients according to the invention can be used in any suitable means, for example oral, topical (including buccal and sublingual), rectal, vaginal, transdermal, parenteral, subcutaneous, intraperitoneal, intrapulmonary, intradermal, intradural and Epidural and intranasal, and if desired, by topical treatment, by means for intralesional administration.
  • Parenteral infusions include intramuscular, intravenous, intraarterial, intraperitoneal or subcutaneous administration.
  • compositions can be administered in any convenient dosage form, for example tablets, powders, capsules, solutions, dispersions, suspensions, syrups, sprays, suppositories, gels, emulsions, patches and the like.
  • Such compositions may contain components conventional to pharmaceutical formulations, such as diluents, carriers, pH adjusters, sweeteners, bulking agents and additional active agents.
  • Typical formulations are prepared by mixing the compound of the invention with a carrier or excipient.
  • Suitable carriers and excipients are well known to those skilled in the art and are described, for example, in Ansel, Howard C., et al., Ansel's Pharmaceutical Dosage Forms and Drug Delivery Systems. Philadelphia: Lippincott, Williams & Wilkins, 2004; Gennaro, Alfonso R., et al. Remington: The Science and Practice of Pharmacy. Philadelphia: Lippincott, Williams & Wilkins, 2000; And Rowe, Raymond C. Handbook of Pharmaceutical Excipients. Chicago, Pharmaceutical Press, 2005.
  • the pharmaceutically acceptable carrier included in the pharmaceutical composition of the present invention is conventionally used in the preparation, lactose, dextrose, sucrose, sorbitol, mannitol, starch, acacia rubber, calcium phosphate, know Nate, gelatin, calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, water, syrup, methyl cellulose, methylhydroxybenzoate, propylhydroxybenzoate, talc, magnesium stearate and mineral oils
  • the present invention is not limited thereto.
  • the formulations also contain one or more buffers, stabilizers, surfactants, wetting agents, lubricants, emulsifiers, suspending agents, preservatives, antioxidants, opacifiers, glidants, processing aids, colorants, sweeteners, flavorings, flavoring agents, diluents, and drugs (Ie, compounds of the present invention or pharmaceutical compositions thereof) may include other known additives to provide a nice appearance or to aid in the manufacture of a pharmaceutical product (ie, a medicament).
  • compositions of the present invention may be prepared in unit dose form by formulating with a pharmaceutically acceptable carrier and / or excipient according to methods which can be easily carried out by those skilled in the art. Or may be prepared by incorporating into a multi-dose container.
  • the formulation may be in the form of a solution, suspension or emulsion in an oil or an aqueous medium, or may be in the form of extracts, powders, granules, tablets or capsules, and may further include a dispersant or stabilizer.
  • compositions are dried, granulated, mixed with magnesium stearate and compressed into tablet form using conventional equipment.
  • aerosol formulations can be prepared, for example, by dissolving 5-400 mg of the compound of the invention in a suitable buffer solution such as phosphate buffer and, if desired, by adding a salt such as isotonic agent, for example sodium chloride. have. Impurities and contaminants can be removed by filtering the solution using, for example, a 0.2 micron filter.
  • one embodiment includes a pharmaceutical composition comprising a compound or stereoisomer or pharmaceutically acceptable salt thereof.
  • Further embodiments include pharmaceutical compositions comprising a compound or stereoisomer or pharmaceutically acceptable salt thereof together with a pharmaceutically acceptable carrier or excipient.
  • the gene therapy agent of the present invention described above includes a gene carrier comprising a nucleic acid molecule of interest as an active ingredient.
  • the gene carrier is designed to transport and express the nucleic acid molecule of interest, and details of the nucleic acid molecule of interest as a carrier object are omitted in order to avoid overlapping with the above description.
  • the nucleic acid molecule of interest is preferably present in a suitable expression construct.
  • the nucleic acid molecule of interest is preferably operably linked to a promoter.
  • operably linked means functional binding between a nucleic acid expression control sequence (eg, an array of promoters, signal sequences, or transcriptional regulator binding sites) and other nucleic acid sequences, thereby The regulatory sequence will control the transcription and / or translation of said other nucleic acid sequence.
  • a promoter bound to a nucleic acid molecule of interest is preferably a promoter derived from a mammalian virus that can operate in animal cells, more preferably mammalian cells to regulate transcription of the nucleic acid molecule of interest.
  • the expression construct used in the present invention comprises a polyadenylation sequence (e.g., a calcined hormone hormone terminator and an SV40
  • Gene carriers can be produced in a variety of forms, for example, 1) naked (recombinant) DNA molecules, 2) plasmids, 3) viral vectors, and 4) the naked (recombinant) DNA molecules or plasmids. It can be prepared in the form of nesting liposomes or niosomes.
  • DNA is naked (Ulmer et al., 1993, Science 259: 1745-1749; Cohen, 1993, Science 259: 1691-1692), DNA is coated into biodegradable beads and further into the cell. Can be delivered efficiently.
  • the nucleic acid molecule of interest can be applied to all gene delivery systems used in conventional gene therapy, preferably plasmids, adenoviruses (Lockett LJ, et al., Clin. Cancer Res. 3: 2075-2080, 1997), Adeno-associated viruses (AAV, Lashford LS., Et al., Gene Therapy Technologies, Applications and Regulations Ed.A. Meager, 1999), retroviruses (Gunzburg WH, et al., Retroviral vectors. Therapy Technologies, Applications and Regulations Ed.A. Meager, 1999), lentiviruses (Wang G. et al., J. Clin. Invest.
  • the method of introducing the above-described gene carrier into a cell may be carried out through various methods known in the art.
  • the gene carrier when the gene carrier is produced based on a viral vector, it is carried out according to a virus infection method known in the art. Infection of host cells with viral vectors is described in the references cited above.
  • a Basophilic serine / threonine kinase
  • b Protein Kinase A
  • c Src homology 2 group
  • d Insulin Receptor Kinase
  • e Tyrosine kinase group
  • f Src Kinase
  • tIK nucleic acid fragment having a mutation in which a tIK nucleic acid fragment and an amino acid of an active site were replaced with another amino acid was designed.
  • tIK nucleic acid fragment a gene fragment of SEQ ID NO: 3 was used, and a point mutant nucleic acid fragment in which amino acids (S382, Y489, Y492) predicted to play an important role in the function of tIK as kinase motifs were replaced with structurally similar amino acids. was produced.
  • a mutant gene in which the nucleotide encoding tyrosine, which is the 489th amino acid among the amino acids of SEQ ID NO: 2 (TAC, which is the 1465-1467 nucleotide among the nucleotides of SEQ ID NO: 1), was substituted with the nucleotide (TTC) encoding phenylalanine A fragment (Y489F) and a nucleotide encoding tyrosine, which is the 492th amino acid among the amino acids of SEQ ID NO: 2, as a nucleotide (TTC) encoding phenylalanine A substituted double mutant gene fragment (Y492F) was produced.
  • TTC nucleotide encoding tyrosine
  • a mutant gene fragment in which the nucleotides encoding the 489th and the 492th tyrosine among the amino acids of SEQ ID NO: 2 were replaced with the nucleotides encoding the phenylalanine (Y489492F), and the 389th serine and the 489th amino acids of the amino acid of SEQ ID NO: 2; And a triple mutant gene fragment (S382AY489492F) in which the 492th tyrosine was substituted with nucleotides encoding alanine and phenylalanine, respectively.
  • HA-tag sequence (hemagglutinin sequence; SEQ ID NO: 54) was added to the 5 'end of each gene fragment for detection, isolation and purification of the expressed protein.
  • the tIK nucleic acid fragments used in this example and the nucleotide sequences of the mutant nucleic acid fragments are shown in Table 2 below.
  • the tIK fragment and the mutant nucleic acid fragment were prepared by inserting the amplified insert into the pcDNA 3.1 vector using fusion PCR.
  • a PCR primer was prepared using a reverse primer set having a forward primer complementary to the 5 'portion of the tIK nucleic acid from the HA tag, and a nucleotide sequence capable of encoding alanine to replace the 382 th serine with alanine. It was.
  • the first fragment from HA tag to alanine (Ala) 382 was amplified using the tIK gene as a template.
  • a set of forward primers containing nucleotides encoding alanine and complementary reverse primer sets at the 3 ′ end of the tIK gene were used to replace the 382 th serine with alanine.
  • the first section was amplified. 1 ⁇ l each of the two fragments obtained by PCR was added to 500 ⁇ l microtube, and 1 ⁇ l of complementary forward primer complementary to the front of the tIK gene and 1 ⁇ l of complementary reverse primer to the terminal of the tIK gene. And fusion PCR was performed by adding dNTP, polymerase, and distilled water.
  • a including EcoRI recognition site
  • b including XhoI recognition site
  • c HindIII recognition site included
  • tIK and mutant nucleic acids were confirmed by the mRNA expression level of the upper regulators that regulate the expression of CIITA, a class II transactivator that promotes MHC class II expression.
  • CNOT1 CCR4-NOT transcription complex subunit 1
  • CDCA3 Cell division cycle-associated protein 3
  • MAPK1 Mitogen-activated protein kinase 1
  • tIK nucleic acid fragments were thought to ultimately influence the inhibition of expression of MHC class II by increasing the factors that inhibit CIITA. Based on these results, it was confirmed whether the same phenomenon occurs in the tIK mutant nucleic acid fragment in which the kinase motif is replaced with another amino acid.
  • cDNA was prepared by transfection of Raji B cells with electroplasmic plasmids in which the tIK mutants S382A, Y489F, Y492F, Y489492F, and S382AY489492F were inserted. Plasmids in which tIK nucleic acid fragments were inserted were also used, and cDNAs obtained by transfection of the pcDNA 3.1 plasmids into which these nucleic acid fragments were not inserted were used as control groups.
  • FIG. 3 The results of measuring the expression of factors regulating the expression of CIITA according to the present embodiment are shown in FIG. 3. Based on the group transfected with pcDNA 3.1, a negative control vector, it was found that CDCA3, CNOT1, and MAPK1, which are factors that inhibit CIITA expression, were increased in the tIK transfected group. In addition, the mutation of Y492F, Y489492F, and S382AY489492F was replaced by another amino acid at the corresponding sequence position, and the expression of the factor that inhibits the expression of CIITA was reduced as compared with the positive control tIK.
  • the S382 and Y489F mutations significantly reduced the expression of the CIITA expression inhibitory gene, which was increased due to tIK expression. Therefore, it was confirmed that the 382th and 492th amino acids in the full-length IK gene can act as an active site in particular responsible for the physiological function of IK.
  • IL-1 receptor antagonist knock out mice (IL1RaKO, from Balb / c), an animal model that is immunologically sensitive and spontaneously develops arthritis to increase the sensitivity to arthritis inducing substances, and the nucleotides of SEQ ID NO: 3
  • tIK-IL1RaKO autoimmune arthritis-induced animal mouse
  • tIK, Balb / c cross-breeding tIK-Transgenic mice
  • tIK nucleic acid fragment Developed a model.
  • tIK-pcDNA 3.1 clone containing the nucleic acid of SEQ ID NO: 3 was micro-injected into fertilized eggs obtained from Balb / c female. The baby was implanted 3 weeks after implantation into the uterus of Balb / c female mouse of fertility.
  • tail lysis buffer pH8.0 Tris buffer 50mM, EDTA 50mM, 0.5% SDS in distilled water, 20mg / ml proteinase K
  • tail lysis buffer pH8.0 Tris buffer 50mM, EDTA 50mM, 0.5% SDS in distilled water, 20mg / ml proteinase K
  • Amplified genomic DNA derived from a transgenic mouse into which tIK nucleic acid fragment was inserted was amplified by PCR using a primer set for tIK genotyping (forward primer is SEQ ID NO: 33; the reverse primer is SEQ ID NO: 34).
  • forward primer is SEQ ID NO: 33
  • reverse primer is SEQ ID NO: 34
  • amplification by PCR method using a primer set for IL1RaKO genotyping forward primer is SEQ ID NO: 35, the reverse primer is SEQ ID NO: 36 and 37
  • electrophoresis of two amplification products in 1% Agarose The degree was compared. Based on the results of electrophoresis, both amplification products were identified as tIK-IL1RaKO double positive transgenic mice obtained through cross-breeding.
  • tIK-IL1RaKO mice and IL1RaKO mice which are new autoimmune arthritis animal models obtained through cross-breeding of Example 2.
  • 10 numbers of tIK-IL1RaKO mice and 10 numbers of IL1RaKO mice were bred in cages under the same conditions.
  • the degree of arthritis was confirmed by checking the degree of swelling of the mouse ankle area every week.
  • the detailed score counting method is as follows. Arthritis evaluation was recorded based on the mean arthritic index by Rosolinec, etc., and the monitoring was performed three times a week in the range that the maximum score of one ankle was 4 points and the four feet of the mice did not exceed 16 points. Continued.
  • FIG. 4a is a graph measuring the arthritis index
  • Figure 4b is a graph measuring the incidence of arthritis
  • Figure 5 is a photograph taken a joint picture observed in the mouse of each group. As shown, the tIK-IL1RaKO mouse group showed a significantly lower arthritis index than the IL1RaKO mouse group, and the incidence rate also showed a 60% lower incidence of arthritis. In addition, it was confirmed that the degree of inflammation of the mouse ankle region was significantly alleviated in the tIK-IL1RaKO group compared to the IL1RaKO group.
  • the joint sites were obtained by monitoring the arthritis spontaneously occurring group at the 16th week of each group (tIK-IL1RaKO group; IL1RaKO group). Joint sites were fixed in 10% neutral formalin and demineralized by soaking in demineralization solution for 7 hours. The demineralized joint area was washed with water for 16 hours, and a paraffin block was prepared by dehydration, clarity, and infiltration. Paraffin blocks were cut using a slicer, 7 ⁇ m slices were prepared, attached to slides, and stained with hematoxylin and eosin (H & E). The process of H & E staining is as follows.
  • cartilage erosion was confirmed by safranin O staining. Safranin O staining was followed by deparaffinization of tissue sections, followed by water treatment, hematoxylin staining, safranin O staining, fast green staining, dehydration, clearing and encapsulation. The results are shown in Figure 6b.
  • the degree of erosion of cartilage can be confirmed by changing the color of the stained area.In the articular tissue derived from IL1RaKO, the cartilage is destroyed in a large part, but the tissue of the joint derived from tIK-IL1RaKO is well maintained. .
  • joints were obtained by autopsy of mice in each group at the 16th week of monitoring of arthritis naturally occurring groups. After the joints were ground in LN 2 , 400 ⁇ l of Trizol reagent was used for RNA isolation. Chloroform was added and strongly vortexed and then incubated at room temperature for 5 minutes. After centrifugation (12500 rpm, 15 min, 4 ° C.), the supernatant was transferred to a new micro tube. After isopropanol and glycoblue in the supernatant was incubated at room temperature for 5 minutes. The supernatant was removed after centrifugation (12500 rpm, 10 min, 4 ° C.).
  • RNA is QuantiTect CDNA was prepared using Reverse Transcription Kit (Qiagen). Quantitative real-time PCR was performed using the prepared cDNA (cDNA 1 ⁇ l, primer set 1 ⁇ l, 2XSYBR green mix 12.5 ⁇ l, Distilled water 9.5 ⁇ l, 40 cycles).
  • the measured result value (Ct value) for each group was calculated as a relative value based on the results of the wild type mice group using the 2 (- ⁇ Ct) method.
  • IL-1 ⁇ , IL-6, IL-17A inflammatory cytokines
  • Primer sequences used to amplify inflammatory cytokines (IL-1 ⁇ , IL-6, IL-17A) using cDNA as a template are shown in Table 5, and the measurement results are shown in FIG. 8A.
  • Inflammatory cytokines (IL-1 ⁇ , IL-6), which are involved in inflamed joints, showed higher expression levels in IL1RaKO mice than in tIK-IL1RaKO mice.
  • IL-17 a major factor that inhibits tissue production in chondrocytes and osteoblasts, induces joint damage, and depletes tissue regeneration, was significantly lowered in joint tissues derived from tIK-IL1RaKO mice.
  • the joint sites were obtained by autopsy of mice in each group (tIK-IL-1RaKO group; IL-1RaKO group) at week 16 of monitoring the arthritis naturally occurring group.
  • Joint area was fixed by 10% neutral formalin and then demineralized by immersion in demineralization solution for 7 hours.
  • the demineralized joint area was washed with water for 16 hours, and a paraffin block was prepared by dehydration, clarity, and infiltration. Paraffin blocks were cut using a slicer to make 7 ⁇ m sections, attached to slides, and stained with inflammatory cytokines (IL-17, IL-1 ⁇ , TNF ⁇ ). The paraffin fragment was deparaffinized and then functioned.
  • mice of each group were autopsied at week 16 of monitoring the arthritis spontaneous generation, and the blood was obtained by heart blood collection.
  • the obtained blood was centrifuged (4000 rpm, 15 min, 4 ° C) to separate only serum, and the expression of inflammatory cytokines in serum was confirmed by ELISA.
  • MCP-1 ELISA kit R & D system
  • the standard provided by the kit is placed on a plate pre-coated with anti-MCP-1 primary antibody, and the serum stock solution is added for 2 hours at room temperature. incubation. After incubation, all the samples in the wells were removed and washed four times with 0.05% PBST (Phosphate Buffered Saline Tween-20).
  • PBST Phosphate Buffered Saline Tween-20
  • a conjugate solution capable of binding to MCP-1 bound to the anti-MCP-1 antibody in the well was added and incubated at room temperature for 2 hours. After incorporation, the substrate solution was added and monitored for color development by reacting with the conjugate.
  • the concentration of MCP-1 indicating the absorbance of the sample was determined based on the absorbance of the standard concentration, and cytokines other than MCP-1 were measured using the same method as above.
  • a forward primer of SEQ ID NO: 44 and a reverse primer of SEQ ID NO: 45 were used, and the remaining cytokines were amplified using the same primers as in Example 6.
  • Example 6 the chemokine MCP-1, IL-known to be involved in the differentiation of pathogenic T cells in the serum of tIK-IL1RaKO mice than IL1RaKO mice 6, and the expression of IL-17, a major factor in the development of arthritis, was found to show a significantly low pattern. It is expected that tIK may also affect systemic immune responses and contribute to lower arthritis levels.
  • T cells one of the immune cells, have been found to be important cells that cause disturbance of the immune system.
  • Activated T cells differentiate into effector T cells, Th1, Th2, Th17 and regulatory T cells, respectively.
  • Th1 and Th17 cells secrete IFN- ⁇ and IL-17, respectively, and are known to form the central axis of inflammatory bowel diseases such as diabetes, rheumatoid arthritis and Crohn's disease.
  • mice in each group were autopsied to obtain spleens.
  • a 100 ⁇ m strainer was placed in a 100 mm petri dish containing Serum free RPMI media, and the spleen was placed on the strainer.
  • the spleen was then torn using a syringe needle to allow immune cells in the spleen to exit the dish containing the media through the stainer. .
  • the cells spread on the media were collected in a tube and centrifuged at 1500 rpm, 4 ° C for 5 minutes to obtain only pellets.
  • Red blood cell lysis buffer was added to the pellet and incubated at 4 ° C. for 5 minutes. Again, centrifuged at 1500rpm, 4 °C, 5 minutes to remove the red blood cells to finally obtain the splenocytes.
  • splenocytes were suspended in RPMI media containing FBS, only 1 ⁇ 10 6 cells were taken and stained for flow cytometry analysis of the etiology of Th17 cells.
  • PMA 100 ng / ml
  • ionomycin 200 ng / ml
  • golgi stop reagent for staining intracellular IL-17 was added.
  • Cells stimulated with PMA and ionomycin were washed with cold PBS and reacted for 30 minutes at 4 ° C. with an antibody against CD4 (PE-Cy5 conjugated), a surface molecule of etiological Th17 cells.
  • the antibody against IL-17 was added and reacted at room temperature for 30 minutes. . After completion of the reaction, the cells were washed with a fixative solution and finally stained with 1% paraformaldehyde solution, and used for flow cytometry analysis.
  • Th1 cells secreting IFN- ⁇ were 6.09% in IL1RaKO mice, but 3.24% in tIK-IL1RaKO mice, which was lower than that of IL1RaKO (FIG. 10A).
  • Th17 cells secreting IL-17 were also shown to be 1.78% in IL1RaKO mice but 1.2% in tIK-IL1RaKO mice, which showed a 1.5-fold reduction in the expression of etiological Th17 cells compared to IL1RaKO (FIG. 10B).
  • Macrophages activated in rheumatoid arthritis secrete inflammatory cytokines and interact with various immune cells to initiate and maintain joint inflammation. Macrophages not only induce T cell activity by presenting antigens to T cells, but also induce osteoclast activation along with T cells by releasing various cytokines. Macrophages play an important role in maintaining the inflammatory response of arthritis, so in this example it was confirmed whether macrophages are involved in the arthritis inhibition phenomenon by IK.
  • Spleen cells of IL1RaKO group mice and tIK-IL1RaKO group were obtained at week 16 of spontaneous arthritis monitoring, and splenocytes were obtained by the same procedure as in Example 8.
  • the obtained splenocytes had an antibody against macrophage factor CD11b (APC conjugated), a pathogenous macrophage factor F4 / 80 antibody (PE-cy7 conjugated), and a macrophage active factor CD86 antibody (FITC conjugated).
  • Spleen cells were stained. Stained cells were fixed in 1% paraformaldehyde solution and used for flow cytometry analysis.
  • the active peptide which is a partial fragment of the short-length IK factor, which is easier to express and purify, was prepared and confirmed to function when applied to the arthritis model.
  • a peptide comprising two amino acids (S382, Y489) expected to play a major role in the function of the IK factor was synthesized.
  • a peptide consisting of 14 amino acids including S382 (SEQ ID NO: 47, S peptide), and a peptide consisting of 13 amino acids including Y489 (SEQ ID NO: 49, Y peptide) were synthesized.
  • S382 and Y489 were synthesized to be substituted with phosphoric acid groups, respectively.
  • Each active peptide was synthesized by Fmoc solid phase synthesis. The remaining reagents were removed by repeating the coupling-wash-deprotection-wash procedure, and a multi-channel automated synthesizer, a COMDEL instrument, was used. Through this synthesis, peptides were synthesized from the C-terminus to the N-terminus of each active peptide. After all reactions were completed, the synthesized peptide was separated from the resin using reagent K. The separated peptide was precipitated by addition of cold diethyl ether, and once again washed with diethyl ether and dried under vacuum.
  • the molecular weight of the synthesized peptide was measured using a mass spectrometer, and purification was performed when the molecular weight was the same as the expected molecular weight. Purification was performed under reverse-HPLC, C18 column, and 220nm wavelength purification conditions.
  • S and Y peptide were confirmed the degree of macrophage activation under the conditions of stimulation of lipopolysaccharide (lipspolysaccharide, LPS).
  • lipopolysaccharide lipopolysaccharide, LPS
  • S and Y peptides were simultaneously transfected at a concentration of 500 ng / mL and treated with LPS (O111: B4) to confirm the expression of inflammatory cytokines.
  • mRNA expression of inflammatory cytokines TNF- ⁇ , IL-6 was low in the group treated with the active peptide (FIG. 12A).
  • Example 11 Confirmation of inhibition of T helper cell differentiation of etiology by active peptide
  • splenocytes were obtained by the method shown in Example 6 using spleens of wild-type mice. Only naive CD4 T cells were selectively isolated from splenocytes, and pretreated with S peptide and Y peptide (100 ng / mL or 500 ng / mL) for 1 hour. A culture medium containing various cytokines and neutralizing antibodies capable of differentiating into etiological T helper cells was added, and when the condition medium was added, the same concentration of S peptide and Y peptide was again obtained (100 ng / mL or 500 ng / mL). Was added.
  • culture medium containing anti-CD3, anti-CD28, IL-2 was added to the cells.
  • culture medium containing anti-CD3, anti-CD28, IL-2, IL-12 and anti-IL-4 was added.
  • culture medium containing anti-CD3, anti-CD28, IL-6, TGF- ⁇ , anti-IL-4, and anti-IFN ⁇ was added to the cells.
  • the culture medium containing various cytokines and neutralizing antibodies was added to differentiate into T helper cells under the same conditions. After 2 days, the pathogenic T helper cells were stained for flow cytometry analysis.
  • phorbol 12-myristate 13-acetate (PMA, 50 ng / mL) and ionomycin (200 ng / mL) were added to the cultured cells. While incubating for 4 hours in a 37 ° C. incubator, golgi stop (monensin) was added to stain intracellular IL-17. After incubation the cells were washed with cold PBS. In order to stain intracellular cytokine (IL-17, IFN ⁇ ), the cell permeability was increased to allow the antibody to penetrate into the cell, and the antibody against IL-17 and IFN ⁇ was added and reacted on ice for 30 minutes. After the reaction, the cells were washed with a fixative solution, and finally, cells stained with 1% paraformaldehyde solution were used for flow cytometry analysis.
  • PMA phorbol 12-myristate 13-acetate
  • ionomycin 200 ng / mL
  • the treatment of the active peptide showed a tendency to decrease the differentiation of CD4 + IFN ⁇ + T cells under Th0 cell differentiation conditions and Th1 cell differentiation conditions.
  • the differentiation of CD4 + IL-17 + T cells was inhibited by the treatment of the active peptide.
  • mice were necropsied at week 8 of the active peptide injection to obtain spleens of mice in each group.
  • the spleens were ground to obtain splenocytes, and PMA (50 ng / mL) and ionomycin (200 ng / mL) were added.
  • PMA 50 ng / mL
  • ionomycin 200 ng / mL
  • the cells were washed with cold PBS, and an antibody (APC conjugated) to CD4, a surface molecule of etiological Th17 cells, was added and reacted at 4 ° C. for 30 minutes.
  • an antibody (APC conjugated) to CD4 a surface molecule of etiological Th17 cells
  • the reaction is complete, the cells are washed, the cell permeability is increased to allow the antibody to penetrate into the cells to stain the IL-17, and then the antibody to the IL-17 (PE conjugated) is added and the reaction is performed at room temperature for 30 minutes. Reacted.
  • the cells were washed with a fixative solution and finally stained with 1% paraformaldehyde solution, and used for flow cytometry analysis.
  • Th17 cells As a result of analysis of etiological Th17 cells in splenocytes of arthritis-induced mice in the peptide injection group and the PBS injection group, it can be seen that fewer Th17 cells appear in the spleen of IL1RaKO injected with the active peptide (FIG. 15A). This means that the active peptide of IK factor has the effect of inhibiting the differentiation and proliferation of Th17 cells, which play a major role in the arthritis etiology.
  • mice were autopsied at week 8 of the active peptide injection to obtain joint sites. Joint area was fixed by 10% neutral formalin and demineralized by soaking in demineralized solution for 7 hours. The demineralized joint area was washed with water for 16 hours, and a paraffin block was prepared by dehydration, clarity, and infiltration. Paraffin blocks were cut using a slicer to make 7 ⁇ m sections, attached to slides, and stained with inflammatory cytokines (IL-17, IL-1 ⁇ , TNF ⁇ ). The paraffin fragment was deparaffinized and then functioned.
  • inflammatory cytokines IL-17, IL-1 ⁇ , TNF ⁇
  • IK fragment (tIK) protein For mass expression of IK fragment (tIK) protein, the tIK plasmid tagged with Fc was transfected into baculovirus and then expressed in insect cells (SF9 cells). A nucleic acid (SEQ ID NO: 50) encoding a thrombin recognition site was linked to the 3 ′ end of the tIK sequence and an Fc tag sequence (SEQ ID NO: 51). After the virus seed was produced, baculovirus expressing tIK-Fc was gradually infected to a larger number of insect cells over the first (100 mm dish scale), the second (T75 flask scale), and the third (1 L conical flask).
  • TIK-Fc protein present in the cell culture was purified using immunoprecipitation method.
  • rProtein A resin was added to the Sepharose column and the culture solution was injected, the Fc portion of the tIK-Fc protein contained in the culture medium was combined with the resin, thereby remaining in the column.
  • Expression of tIK-Fc protein was primarily confirmed by Commassie blue staining (result not shown).
  • Example 15 Establishment of a Recombinant Adeno-associated Virus (rAAV) Vector System for tIK Gene Delivery
  • rAAV Recombinant Adeno-associated Virus
  • Adeno-associated viruses unlike retroviruses, do not cause disease and are selectively integrated into chromosome 19 of human cells, and thus are used as gene therapy vectors for chronic diseases.
  • a rAAV2 capable of delivering a tIK nucleic acid fragment into a cell is subjected to a gene cloning step in which the tIK nucleic acid fragment represented by SEQ ID NO: 3 is surrounded by an inverted terminal repeat (ITR) form of AAV.
  • ITR inverted terminal repeat
  • a HA tag sequence (SEQ ID NO: 54) is linked to the 5 'end of the tIK nucleic acid fragment, and a primer complementary to both ends of the tIK nucleic acid fragment (forward primer is SEQ ID NO: 55; the reverse primer is SEQ ID NO: 56 PCR was performed using).
  • forward primer is SEQ ID NO: 55; the reverse primer is SEQ ID NO: 56 PCR was performed using.
  • the ends of the amplified fragments thus obtained were cut with restriction enzymes and inserted into expression vectors to prepare tIK-HA-sp72-XP7-scAAV2 recombinant expression vectors. After transforming the prepared recombinant expression vector into Escherichia coli, the plasmid into which the tIK amplification fragment was inserted could be selected.
  • the tIK gene linked to adeno-associated virus was injected into 293T cells by transfection with polyethylenimine (PEI). Viruses that were found to express tIK using PEI were again infected with 293T cells and confirmed by Western blotting (FIG. 17).
  • RAAV2-tIK prepared in Example 15 was applied to an in vivo disease model to confirm the therapeutic effect of arthritis according to the expression of tIK nucleic acid.
  • AAV2-tIK and AAV2-GFP viruses were injected intravenously through the tail of IL1RaKO, a naturally occurring arthritis at a concentration of 1 ⁇ 10 11 vg / 100 ul, respectively.
  • RAAV2-tIK and rAAV-GFP were injected at two week intervals, and once a week mouse joint condition and incidence were measured.
  • IL1RaKO injected with rAAV2-tIK showed lower arthritis incidence and arthritis index than IL1RaKO injected with rAAV2-GFP (FIG. 18A), and the incidence of arthritis was low (FIG. 18B).
  • IK nucleic acid was delivered using a virus vector system, which confirmed the possibility of lowering the incidence of arthritis and arthritis index.
  • CHO cells have high expression efficiency during gene injection and are widely used as animal cell lines for producing recombinant protein pharmaceutical products.
  • tIK plasmids tIK-PcDNA3.1 having a HA tag sequence (SEQ ID NO: 54) linked to the 5 'end were injected into CHO cells.
  • SEQ ID NO: 54 a forward primer of SEQ ID NO: 55 and a reverse primer of SEQ ID NO: 57 were used.
  • Expression of tIK in CHO cells was confirmed by western blotting (FIG. 19).

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Organic Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Toxicology (AREA)
  • Rheumatology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Epidemiology (AREA)
  • Pain & Pain Management (AREA)
  • Zoology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The present invention relates to a pharmaceutical composition for treating and/or preventing arthritis, containing, as an active ingredient, a gene carrier in which an IK factor or a fragment thereof, or a nucleic acid encoding the same is inserted. The IK factor or the fragment thereof, and the nucleic acid encoding the same, which are the active ingredients of the pharmaceutical composition according to the present invention, are derived from an organism, have no side effects if administered for a long time, thereby ensuring safety, and are expected to effectively treat arthritis by being involved in the high level mechanism for arthritis treatment.

Description

아이케이 인자 및 아이케이 인자를 코딩하는 핵산의 약학적 용도Pharmaceutical Use of IK Factor and Nucleic Acids Encoding IK Factor
본 발명은 생체 유래의 펩타이드 및 이를 코딩하는 핵산의 약학적 용도에 관한 것으로, 더욱 상세하게는 아이케이 인자 또는 그 단편 및/또는 이들을 코딩하는 핵산의 약학적 용도에 관한 것이다. FIELD OF THE INVENTION The present invention relates to pharmaceutical uses of peptides derived from living organisms and nucleic acids encoding them, and more particularly to pharmaceutical uses of IK factors or fragments thereof and / or nucleic acids encoding them.
관절염(arthritis)은 하나 이상의 관절의 염증으로 인하여 관절의 경직 및 관절 주변의 지속적인 통증을 수반하는 질병이다. 관절염은 크게 급성 관절염과 만성 관절염으로 구분될 수 있다. 통풍성 관절염, 다발성 관절염, 류마티스성 관절염, 뼈나 관절의 노화로 인한 변형성 관절염과 같은 만성 관절염은 관절 부위에 지속적인 통증을 유발한다. Arthritis is a disease involving joint stiffness and persistent pain around the joints due to inflammation of one or more joints. Arthritis can be largely divided into acute arthritis and chronic arthritis. Chronic arthritis, such as gouty arthritis, polyarthritis, rheumatoid arthritis, and osteoarthritis due to aging of bones or joints, causes persistent pain in the joint area.
만성 관절염 중에서도 류마티스성 관절염(Rheumatoid arthritis)은 다발성 관절염을 특징으로 하는 원인 불명의 만성 염증성 질환이다. 현재까지 류마티스성 관절염에 대한 정확한 원인은 밝혀지지 않았다. 하지만, 일반적으로는 유전적 소인, 세균이나 바이러스 감염 등이 류마티스성 관절염의 원인으로 생각되고 있으며, 호르몬도 류마티스성 관절염에 관여하는 것으로 추측되고 있다. 류마티스성 관절염은 단순히 관절에만 영향을 미치는 것이 아니라 신체의 다른 기관에도 악영향을 미친다. 따라서 류마티스성 관절염이 유발된 환자는 손이나 발을 사용할 수 없거나 걷지 못할 뿐만 아니라, 피곤함과 불쾌감을 빈번하게 느끼게 된다. 더욱이 류마티스 관절염으로 인하여 체중 감소, 수면 부족 등의 증상이 초래되고, 류마티스성 관절염이 지속되면 근육의 약화 등 신체적 활동을 제한하여 개인적, 사회적 활동에 큰 영향을 미칠 수 있다. 류마티스성 관절염에 의하여 신체적 활동이 제한되는 경우, 비만 가능성이 증가하고, 콜레스테롤 수치로 인한 심장병을 초래할 수 있으며, 우울증을 야기할 수 있다. Among chronic arthritis, rheumatoid arthritis is an unexplained chronic inflammatory disease characterized by multiple arthritis. To date, the exact cause of rheumatoid arthritis is unknown. However, genetic predisposition, bacterial and viral infections are generally thought to be the cause of rheumatoid arthritis, and hormones are also believed to be involved in rheumatoid arthritis. Rheumatoid arthritis not only affects the joints, but also adversely affects other organs of the body. Thus, patients with rheumatoid arthritis are not only unable to use or walk their hands or feet, but also frequently feel tired and uncomfortable. Furthermore, rheumatoid arthritis causes symptoms such as weight loss and sleep deprivation, and if rheumatoid arthritis persists, physical activity, such as muscle weakness, may be restricted, which may greatly affect personal and social activities. When physical activity is limited by rheumatoid arthritis, the likelihood of obesity increases, can lead to heart disease due to cholesterol levels, and can lead to depression.
류마티스서 관절염은 1년에 남자는 1,000명당 0.2명, 여자는 1,000명당 0.4명꼴로 발생하며, 전 세계적으로 류마티스성 관절염의 유병율은 0.4-1.4%로 비교적 고른 분포를 보이고 있다. 더욱이 노인 인구가 점차로 증가하면서 류마티스성 관절염 치료제 시장은 급격하게 성장할 것으로 예측되고 있다. Rheumatoid arthritis occurs in 0.2 men per 1,000 men and 0.4 women per 1,000 women per year, and the prevalence of rheumatoid arthritis is 0.4-1.4% worldwide. Moreover, the market for rheumatoid arthritis drugs is expected to grow rapidly as the elderly population grows.
현재까지 류마티스성 관절염을 치료하기 위한 약품으로는 비스테로이드 항염제(nonsteroidal anti-inflammatory drug, NSAID); 스테로이드; 항말라리아제, 하이드록시클로로퀴논(hydroxychloroquinone, HCQ), 설파살라진(sulfasalazine), 메토트렉세이트(methotrexate, MTX), 레플루노마이드(leflunomide)와 같은 비생물학적 항류마티스 약제(non-biologic disease-modifying antirheumatic drug, DMARD); 종양괴사인자(tumor necrosis factor, TNF) 억제제나 IL-6 억제제와 같은 생물학적 항류마티스 약제가 사용되고 있다. To date, drugs for treating rheumatoid arthritis include nonsteroidal anti-inflammatory drugs (NSAIDs); steroid; Non-biologic disease-modifying antirheumatic drug, DMARD such as antimalarial agent, hydroxychloroquinone (HCQ), sulfasalazine, methotrexate (MTX), leflunomide ); Biological antirheumatic drugs such as tumor necrosis factor (TNF) inhibitors or IL-6 inhibitors are used.
생물학적 항류마티스 약제로서, 에타너셉트(etanercept), 아달리무맙(adalimumab), 인플릭시맙(infliximab), 골로무맵(golomumab), 세르토리주맵(certolizumab)과 같은 종양괴사인자(TNF) 억제제; 토실리주맵(Tocilizumab)과 같은 IL-6 억제제; B-세포 제거제인 단일클론항체인 리툭시맵(Rituximab); 항원 제시세포와 T-세포 사이의 면역 반응을 차단하는 아바타셉트(Abatacept); 소분자 억제제로서 JAK(Janue activated kinase)를 선택적으로 차단하는 토파시티닙(tofacitinib) 등이 개발되었다. As biological antirheumatic agents, tumor necrosis factor (TNF) inhibitors such as etanercept, adalimumab, infliximab, golomumab, certolizumab; IL-6 inhibitors such as Tocilizumab; Rituximab, a monoclonal antibody that is a B-cell scavenger; Abatacept, which blocks an immune response between antigen presenting cells and T-cells; As small molecule inhibitors, tofacitinib, which selectively blocks JAK (Janue activated kinase), has been developed.
하지만, 이들 류마티스성 관절염 치료제들은 대부분 심각한 부작용을 유발한다. 예를 들어, 비스테로이드 항염제는 위장관 부작용을 야기하고 있으며, 선택적 COX-2 억제제를 비롯한 비스테로이드 항염제는 심각한 심혈관계 부작용을 유발할 수 있다. 그 외에도 비스테로이드 항염제는 신장 기능 저하, 간질성 신염, 급성신부전 등의 부작용이 있는 것으로 알려져 있다. 스테로이드는 장기간 복용할 경우 시상하부-뇌하수체-부신 위축이 야기되어 부신 기능 부전이 발생할 수 있으며, 녹내장, 백내장, 골괴사, 골다공증, 고혈압, 저칼륨혈증 등의 부작용이 발생할 수 있다. 특히 비스테로이드 항염제나 스테로이드는 단지 염증을 완화시켜주는 것에 불과하기 때문에 실제 관절염의 진행을 억제하지 못하고, 관절 손상의 예방 효과가 미비하다는 단점이 있다. However, most of these rheumatoid arthritis drugs cause serious side effects. For example, nonsteroidal anti-inflammatory drugs cause gastrointestinal side effects, and nonsteroidal anti-inflammatory drugs, including selective COX-2 inhibitors, can cause serious cardiovascular side effects. In addition, nonsteroidal anti-inflammatory drugs are known to have side effects such as renal failure, interstitial nephritis, and acute renal failure. Steroids can cause hypothalamic-pituitary-adrenal atrophy, resulting in adrenal insufficiency, and side effects such as glaucoma, cataracts, osteonecrosis, osteoporosis, hypertension and hypokalemia. In particular, nonsteroidal anti-inflammatory drugs and steroids are not only to relieve inflammation, it does not inhibit the progress of the actual arthritis, and there is a disadvantage that the prevention effect of the joint damage is insufficient.
한편 현재 개발된 항류마티스 약제는 조기 류마티스성 관절염의 진행 속도를 억제할 수는 있으나, 류마티스성 관절염을 완치시키지는 못하며 일부 부작용도 보고되고 있다. 예를 들어, 비생물학적 항류마티스 약제는 시력 장애, 망막 병변, 피부 발진, 간기능 수치 이상, 오심(메스꺼움) 등의 부작용이 있다. 또한, 생물학적 항류마티스 약제 중에서 종양괴사인자 억제제들은 혈액 이상, 심부전, 간 기능 이상 및 기회 감염이 문제가 되고 있으며, IL-6 억제제는 간기능 이상, 백혈구 감소 등의 부작용이 발생할 수 있다. 그 이외의 생물학적 항류마티스 약제는 감염의 문제와 종양 발생 가능성, 위장관 천공 및 고지혈증 등의 부작용이 보고된 바 있다. Meanwhile, the anti-rheumatic drugs currently developed may inhibit the progression of early rheumatoid arthritis, but do not cure rheumatoid arthritis and some side effects have been reported. For example, abiotic antirheumatic drugs have side effects such as vision disorders, retinal lesions, skin rashes, abnormal liver function levels, and nausea (nausea). In addition, tumor necrosis factor inhibitors among biological antirheumatic drugs are problematic for blood abnormalities, heart failure, liver dysfunction, and opportunistic infections, and IL-6 inhibitors may cause side effects such as liver dysfunction and white blood cell reduction. Other biological antirheumatic drugs have been reported with side effects such as infection problems, tumor potential, gastrointestinal perforation and hyperlipidemia.
특히, 대부분의 생물학적 항류마티스 약제들은 피하 투여나 혈관 주사를 통한 투여만 가능하다는 치명적 단점이 있고, 시간이 경과하면서 인체의 면역원성이 약품에 대한 반응 감소를 유발한다. 또한 현재 개발된 생물학적 항류마티스 약제는 전반적인 면역 시스템을 억제하는 결과 기회 감염 및 암 억제 등에 취약점을 노출하고 있다. 아울러, 이들 생물학적 항류마티스 약제는 상당히 고가이기 때문에 환자들에게 상당한 경제적 부담을 초래한다. 특히 기존에 사용되었던 대표적인 생물학적 항류마티스 약제인 항체는 적지 않은 류마티스성 관절염 환자에게 반응하지 않는 문제점이 대두되고 있다. In particular, most biological antirheumatic drugs have a fatal disadvantage that they can only be administered by subcutaneous administration or by vascular injection, and over time, the immunogenicity of the human body causes a decrease in response to the drug. In addition, the currently developed biological antirheumatic drugs expose the vulnerability to opportunistic infection and cancer suppression as a result of suppressing the overall immune system. In addition, these biological antirheumatic drugs are quite expensive, resulting in significant economic burden on patients. In particular, antibodies, which are representative biological anti-rheumatic drugs, which have been used in the past, have a problem of not responding to many patients with rheumatoid arthritis.
본 발명은 전술한 종래 기술의 문제점을 해소하기 위하여 제안된 것으로, 본 발명의 목적은 장기간 투여에도 불구하고 생체에 대한 부작용 및 독성이 없어 안전성이 담보되는 관절염을 치료 및/또는 예방하기 위한 약학적 조성물을 제공하고자 하는 것이다. The present invention has been proposed to solve the above-mentioned problems of the prior art, and an object of the present invention is a pharmaceutical for treating and / or preventing arthritis in which safety is ensured due to no side effects and toxicity to a living body despite prolonged administration. It is to provide a composition.
본 발명의 다른 목적은 효능 회피가 없으며, 저렴하게 대량으로 제조할 수 있어서 환자의 부담을 줄일 수 있는 관절염을 치료 및/또는 예방하기 위한 약학적 조성물을 제공하고자 하는 것이다. It is another object of the present invention to provide a pharmaceutical composition for treating and / or preventing arthritis, which can be prepared in large quantities at low cost without avoiding efficacy, thereby reducing the burden on the patient.
본 발명은 약학적 유효량의 아이케이 인자(IK factor) 또는 아이케이 인자의 단편을 함유하는, 관절염을 치료 또는 예방하는 약학적 조성물을 제공한다. The present invention provides a pharmaceutical composition for treating or preventing arthritis, containing a pharmaceutically effective amount of an IK factor or fragment of an IK factor.
하나의 예시적인 실시형태에서, 상기 아이케이 인자의 단편은 서열식별번호:4의 아미노산 서열을 가지는 펩타이드 또는 서열식별번호:4의 부분 펩타이드를 포함한다. In one exemplary embodiment, the fragment of IK factor comprises a peptide having the amino acid sequence of SEQ ID NO: 4 or a partial peptide of SEQ ID NO: 4.
예를 들어, 상기 아이케이 인자의 단편은 서열식별번호:2의 아미노산 서열의 382번째 세린, 489번째 티로신 및 492번째 티로신 중에서 선택되는 적어도 하나의 아미노산 잔기 및 이들 선택된 아미노산 잔기의 인접 아미노산으로 구성되는 10개 이상의 아미노산을 가지는 펩타이드로 구성될 수 있다. For example, the fragment of the IK factor consists of at least one amino acid residue selected from 382 th serine, 489 tyrosine, and 492 tyrosine of the amino acid sequence of SEQ ID NO: 2 and adjacent amino acids of these selected amino acid residues. It may consist of peptides having 10 or more amino acids.
예시적으로, 상기 아이케이 인자의 단편은 서열식별번호:2의 아미노산 서열의 382번째 세린, 489번째 티로신 및 492번째 티로신으로 구성되는 적어도 하나의 아미노산이 인산화되어 있다. In exemplary embodiments, the fragment of the IK factor is phosphorylated with at least one amino acid consisting of 382 th serine, 489 th tyrosine, and 492 th tyrosine of the amino acid sequence of SEQ ID NO: 2.
하나의 선택적인 실시형태에서, 상기 아이케이 인자의 단편은 서열식별번호:2의 아미노산 서열을 가지는 펩타이드의 단편으로서, 서열식별번호:2의 아미노산 서열의 382번째 세린 및 489번째 티로신 중에서 선택되는 적어도 하나의 아미노산 잔기 및 이들 선택된 아미노산 잔기의 인접 아미노산으로 구성되는 10개 이상의 아미노산을 가지는 펩타이드로 구성될 수 있다. In one optional embodiment, the fragment of IK factor is a fragment of a peptide having an amino acid sequence of SEQ ID NO: 2, wherein at least one selected from among 382 th serine and 489 th tyrosine of the amino acid sequence of SEQ ID NO: 2 It may consist of a peptide having at least 10 amino acids consisting of one amino acid residue and contiguous amino acids of these selected amino acid residues.
구체적인 실시 형태에서, 상기 아이케이 인자의 단편은 상기 선택된 아미노산의 N-말단과 C-말단으로 각각 2개 이상의 인접 아미노산이 연결되어 있는 10개 이상의 아미노산을 가지는 펩타이드로 구성될 수 있다. In a specific embodiment, the fragment of the IK factor may be composed of a peptide having 10 or more amino acids in which two or more adjacent amino acids are linked to the N-terminus and C-terminus of the selected amino acid, respectively.
예시적인 실시예에서, 상기 아이케이 인자의 단편은 서열식별번호:4, 서열식별번호:47, 서열식별번호:49로 구성되는 군에서 적어도 하나 선택되는 아미노산 서열을 가지는 펩타이드로 구성될 수 있다. In an exemplary embodiment, the fragment of the IK factor may be composed of a peptide having at least one amino acid sequence selected from the group consisting of SEQ ID NO: 4, SEQ ID NO: 47, SEQ ID NO: 49.
상기 아이케이 인자 또는 상기 아이케이 인자의 단편을 함유하는 약학적 조성물은 류마티스성 관절염을 치료하는데 사용될 수 있다. Pharmaceutical compositions containing the IK factor or fragments of IK factor can be used to treat rheumatoid arthritis.
예시적인 실시 형태에서, 상기 아이케이 인자 또는 상기 아이케이 인자의 단편은 상기 약학적 조성물 중에 1.0 ng/mL ~ 10 ㎍/mL의 농도로 함유될 수 있다. In an exemplary embodiment, the IK factor or fragment of IK factor may be contained in the pharmaceutical composition at a concentration of 1.0 ng / mL to 10 μg / mL.
또한, 본 발명은 아이케이 인자(IK factor) 또는 아이케이 인자의 단편을 코딩하는 핵산 분자를 포함하는 유전자 전달체를 함유하는, 관절염을 치료 또는 예방하는 약학적 조성물을 제공한다. The present invention also provides a pharmaceutical composition for treating or preventing arthritis, comprising a gene carrier comprising a nucleic acid molecule encoding an IK factor or fragment of an IK factor.
예를 들어, 상기 아이케이 인자의 단편을 코딩하는 핵산은 서열식별번호:3의 뉴클레오티드 서열을 가지는 핵산 또는 서열식별번호:3의 부분 핵산을 포함한다. For example, the nucleic acid encoding a fragment of the IK factor includes a nucleic acid having a nucleotide sequence of SEQ ID NO: 3 or a partial nucleic acid of SEQ ID NO: 3.
예시적인 실시형태에서, 상기 아이케이 인자의 단편을 코딩하는 핵산은 서열식별번호:1의 뉴클레오티드 서열의 1144-1146번의 뉴클레오티드 잔기, 1465-1467번째 뉴클레오티드 잔기 및 1474-1476번째 뉴클레오티드 잔기 중에서 선택되는 적어도 하나의 뉴클레오티드 잔기와 이들 선택된 뉴클레오티드 잔기의 인접 뉴클레오티드로 구성되는 30개 이상의 뉴클레오티드를 가지는 핵산으로 구성될 수 있다.In an exemplary embodiment, the nucleic acid encoding a fragment of the IK factor is at least one selected from nucleotide residues 1144-1146, 1465-1467 nucleotide residues, and 1474-1476 nucleotide residues of the nucleotide sequence of SEQ ID NO: 1. It may consist of a nucleic acid having at least 30 nucleotides consisting of one nucleotide residue and adjacent nucleotides of these selected nucleotide residues.
예시적인 하나의 실시 형태에서, 상기 아이케이 인자의 단편을 코딩하는 핵산은 서열식별번호:1의 뉴클레오티드 서열의 1144-1146번의 뉴클레오티드 세트 및 1465-1467번째 뉴클레오티드 세트 중에서 선택되는 적어도 하나의 뉴클레오티드 잔기와, 이들 선택된 뉴클레오티드 잔기의 인접 뉴클레오티드로 구성되는 30개 이상의 뉴클레오티드를 가지는 핵산으로 구성될 수 있다. In one exemplary embodiment, the nucleic acid encoding a fragment of the IK factor comprises at least one nucleotide residue selected from the set of 1144-1146 nucleotides and the set of 1465-1467 nucleotides of the nucleotide sequence of SEQ ID NO: 1. And nucleic acids having 30 or more nucleotides consisting of contiguous nucleotides of these selected nucleotide residues.
하나의 구체적인 실시형태에서, 상기 아이케이 인자의 단편을 코딩하는 핵산은 상기 선택된 뉴클레오티드 잔기의 5' 말단과 3' 말단으로 각각 6개 이상의 인접 뉴클레오티드가 연결되어 있는 30개 이상의 뉴클레오티드를 가지는 핵산으로 구성될 수 있다. In one specific embodiment, the nucleic acid encoding a fragment of the IK factor consists of a nucleic acid having at least 30 nucleotides each having at least 6 contiguous nucleotides linked to the 5 'and 3' ends of the selected nucleotide residue. Can be.
예시적인 실시예에서, 상기 아이케이 인자의 단편을 코딩하는 핵산은 서열식별번호:4, 서열식별번호:47 및 서열식별번호:49로 구성되는 군에서 적어도 하나 선택되는 아미노산 서열을 가지는 펩타이드를 코딩하는 뉴클레오티드 서열을 갖는 핵산으로 구성된다. In an exemplary embodiment, the nucleic acid encoding the fragment of the IK factor encodes a peptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 4, SEQ ID NO: 47 and SEQ ID NO: 49. And a nucleic acid having a nucleotide sequence.
예를 들어, 상기 아이케이 인자의 단편을 코딩하는 핵산은 서열식별번호:3, 서열식별번호:46 및 서열식별번호:48로 구성되는 군에서 적어도 하나 선택되는 뉴클레오티드 서열을 가지는 핵산으로 구성될 수 있다. For example, the nucleic acid encoding the fragment of the IK factor may be composed of a nucleic acid having at least one nucleotide sequence selected from the group consisting of SEQ ID NO: 3, SEQ ID NO: 46 and SEQ ID NO: 48. have.
상기 아이케이 인자 또는 상기 아이케이 인자의 단편을 코딩하는 핵산은 류마티스성 관절염을 치료하는데 사용될 수 있다. Nucleic acids encoding the IK factor or fragments of the IK factor can be used to treat rheumatoid arthritis.
예시적인 실시형태에서, 상기 아이케이 인자 또는 상기 아이케이 인자의 단편을 코딩하는 핵산은 상기 약학적 조성물 중에 1.0 ng/mL ~ 10 ㎍/mL의 농도로 함유될 수 있다. In an exemplary embodiment, the nucleic acid encoding the IK factor or fragment of IK factor may be contained in the pharmaceutical composition at a concentration of 1.0 ng / mL to 10 μg / mL.
예를 들어, 상기 유전자 전달체는 네이키드 핵산 분자, 플라스미드, 바이러스 벡터 및 상기 플라스미드 또는 상기 바이러스 벡터를 함유하는 리포좀 또는 니오좀의 형태를 가질 수 있다. For example, the gene carrier may have the form of a naked nucleic acid molecule, a plasmid, a viral vector and a liposome or niosome containing the plasmid or the viral vector.
이때, 예시적으로 상기 바이러스 벡터는 아데노바이러스, 아데노-연관 바이러스, 레트로 바이러스, 렌티바이러스, 바큘로바이러스, 헤르페스 심플렉스 바이러스, 백시니아 바이러스로 구성되는 군에서 선택될 수 있다. At this time, for example, the viral vector may be selected from the group consisting of adenovirus, adeno-associated virus, retrovirus, lentivirus, baculovirus, herpes simplex virus, vaccinia virus.
또한 본 발명은 관절염 치료를 위한 아이케이 인자 또는 그 단편, 아이케이 인자 또는 그 단편을 코딩하는 핵산, 및/또는 이들 핵산을 가지는 벡터 또는 유전자 전달체에 관한 것이다. The present invention also relates to an IK factor or fragment thereof for treating arthritis, a nucleic acid encoding an IK factor or a fragment thereof, and / or a vector or gene carrier having these nucleic acids.
또한 본 발명은 아이케이 인자 또는 아이케이 인자의 단편, 아이케이 인자 또는 그 단편을 코딩하는 핵산, 및/또는 이들 핵산을 가지는 벡터 또는 유전자 전달체를 치료학적 유효량, 또는 약학적 유효량으로 투여하는 단계를 포함하는, 관절염을 치료하는 방법에 관한 것이다.The present invention also provides a step of administering an IK factor or fragment of IK factor, a nucleic acid encoding IK factor or a fragment thereof, and / or a vector or gene delivery carrier having these nucleic acids in a therapeutically effective amount or in a pharmaceutically effective amount. It includes a method of treating arthritis, which includes.
본 발명에 따르면, 아이케이 인자 또는 그 단편 및/또는 이들 펩타이드를 코딩하는 핵산은 관절염, 예를 들어 류마티스성 관절염과 관련한 상위 단계(upstream level)의 반응 기전에 관여하여, 류마티스성 관절염의 진행을 억제한다. According to the present invention, the IK factor or fragments thereof and / or nucleic acids encoding these peptides are involved in the upstream level of reaction mechanisms associated with arthritis, for example rheumatoid arthritis, thereby promoting the development of rheumatoid arthritis. Suppress
본 발명에 따라 유효 성분으로 사용되는 아이케이 인자 또는 그 단편 및/또는 이들을 코딩하는 핵산은 관절염에 영향을 주는 면역세포들의 활성을 전반적으로 조절하는 인체 내의 면역 밸런스 조절 물질이기 때문에 안전성이 담보될 수 있다. The IK factor or fragment thereof and / or nucleic acid encoding them used as an active ingredient according to the present invention can be secured because it is an immune balance modulator in the human body that overall regulates the activity of immune cells affecting arthritis. have.
특히, 아이케이 인자 또는 그 단편은 인체 내에서 생성되는 내재성 물질로서 면역원성이 낮을 뿐만 아니라, 면역 세포의 전체 population에는 크게 관여하지 않으므로, 장기간 투여에도 부작용은 없을 것으로 예측된다. In particular, since the IK factor or fragment thereof is an endogenous substance produced in the human body, it has low immunogenicity and does not significantly affect the entire population of immune cells, and therefore, long-term administration is expected to have no side effects.
또한 예를 들어 단일클론항체로 개발되고 있는 기존의 류마티스 관절염 치료제와 비교하여 훨씬 저렴한 비용으로 제조가 가능하다. 아울러, 기존 관절염 치료제가 가지고 있는 효능 회피의 문제점을 해소할 수 있을 것으로 기대된다.It can also be manufactured at a much lower cost than, for example, conventional rheumatoid arthritis therapies being developed as monoclonal antibodies. In addition, it is expected to solve the problem of avoiding the efficacy of the existing arthritis treatments.
도 1은 아이케이(IK) 인자를 코딩하는 전장 핵산과, 이 전장 핵산의 N-말단 뉴클레오티드를 제외하고 C-말단의 뉴클레오티드만으로 구성된 절단 핵산 단편(tIK 핵산 단편)의 위치를 개략적으로 도시한 모식도이다. 도 1에서 "HA"는 헤마글루티닌(haemagglutinin) 태그(tag)를 나타낸 것이다. 1 is a schematic diagram showing the location of a full-length nucleic acid encoding an IK factor and a truncated nucleic acid fragment (tIK nucleic acid fragment) consisting of only the C-terminal nucleotide except for the N-terminal nucleotide of the full-length nucleic acid. to be. "HA" in Figure 1 represents a hemagglutinin tag (haemagglutinin) tag.
도 2의 가장 윗 부분은 본 발명의 예시적인 실시 형태에 따라 전장 아이케이 인자를 코딩하는 뉴클레오티드를 기준으로 일부 뉴클레오티드가 절단된 핵산 단편(tIK 핵산 단편)을 구성하는 뉴클레오티드의 위치를 개략적으로 도시한 모식도이다. 도 2의 나머지 부분은 변이 아이케이 인자의 제작과 관련하여 전장 아이케이 인자의 예시적인 아미노산 서열을 기준으로 아이케이 인자의 생리적 기능에 관여하는 것으로 예측되는 활성 부위의 아미노산의 위치와 치환된 아미노산의 종류를 개략적으로 도시한 모식도이다. 도 2에서 "HA"는 헤마글루티닌(haemagglutinin) 태그(tag)를 나타낸 것이다. The top part of FIG. 2 schematically shows the position of nucleotides constituting a nucleic acid fragment (tIK nucleic acid fragment) in which some nucleotides are cleaved relative to a nucleotide encoding a full-length IK factor according to an exemplary embodiment of the present invention. It is a schematic diagram. The remainder of FIG. 2 shows the positions of the amino acids and substituted amino acids of the active sites predicted to be involved in the physiological function of the IK factor relative to the construction of the variant IK factor relative to the exemplary amino acid sequence of the full length IK factor. It is a schematic diagram which shows the kind schematically. "HA" in Figure 2 represents a hemagglutinin (haemagglutinin) tag (tag).
도 3은 아이케이 인자의 절단된 핵산 단편(tIK 핵산 단편)과, tIK 핵산 단편의 일부 뉴클레오티드를 다른 뉴클레오티드로 치환한 돌연변이 핵산 단편(S382A, Y489F, Y492F, Y489492F, S382AY489492F)의 발현에 의하여, MHC class Ⅱ의 transactivator인 CIITA의 발현을 억제하는 상위 조절 인자인 CNOT1, CDCA3 및 MAPK1의 발현을 억제하는 정도를 측정한 그래프이다. 도 3에서 *는 네거티브 컨트롤에 비하여 P < 0.05를 의미하며, #는 야생형 tIK가 발현된 것에 비하여 돌연변이 핵산 단편이 발현된 경우 P < 0.05를 의미한다. Figure 3 shows MHC by expression of a truncated nucleic acid fragment of IK factor (tIK nucleic acid fragment) and mutant nucleic acid fragments (S382A, Y489F, Y492F, Y489492F, S382AY489492F) in which some nucleotides of the tIK nucleic acid fragment are substituted with other nucleotides. This is a graph measuring the degree of inhibition of the expression of CNOT1, CDCA3 and MAPK1, which are higher regulators that inhibit the expression of CIITA, a class II transactivator. In FIG. 3, * means P <0.05 as compared to negative control, and # means P <0.05 when mutant nucleic acid fragments are expressed as compared with wild type tIK.
도 4a는 관절염이 자연적으로 유도되는 IL-1 Receptor antagonist knock-out (IL1RaKO) 마우스와, IL1RaKO 마우스와 본 발명에 따라 일부 절단된 아이케이 인자(tIK)가 발현된 transgenic 마우스를 교배하여 제작된 마우스(tIK-IL1RaKO)에서 시간 경과에 따른 관절염 지수를 측정한 그래프이다. 도 4a에서 *는 네거티브 컨트롤에 비하여 P < 0.05를 의미하고, **는 네거티브 컨트롤에 비하여 P < 0.01을 의미하며, ***는 P < 0.001을 의미한다. Figure 4a is a mouse produced by crossing the IL-1 Receptor antagonist knock-out (IL1RaKO) mouse with arthritis naturally induced, IL1RaKO mouse and transgenic mice expressing some cleaved IK factor (tIK) according to the present invention (tIK-IL1RaKO) is a graph measuring arthritis index over time. In FIG. 4A, * means P <0.05 compared to negative control, ** means P <0.01 compared to negative control, and *** means P <0.001.
도 4b는 IL1RaKO 마우스와 tIK-IL1RaKO 마우스에서 시간 경과에 따른 관절염 발병율을 측정한 그래프이다. Figure 4b is a graph measuring the incidence of arthritis over time in IL1RaKO mice and tIK-IL1RaKO mice.
도 5는 관절염이 자연적으로 유도되는 IL1RaKO 마우스와, 본 발명에 따라 제작된 tIK-IL1RaKO 마우스의 관절 부위를 촬영한 사진이다. 5 is a photograph of the joint region of IL1RaKO mice naturally induced arthritis, and tIK-IL1RaKO mouse produced according to the present invention.
도 6a는 관절염이 자연적으로 유도되는 IL1RaKO 마우스와, 본 발명에 따라 제작된 tIK-IL1RaKO 마우스 유래의 관절 조직을 H&E 염색법을 이용하여 측정한 결과를 도시하고 있다. 좌측은 관절 조직에 대한 현미경 사진이고, 우측은 관절 조직에서의 염증 정도를 분석한 그래프이다. 도 6a에서 *는 네거티브 컨트롤에 비하여 P < 0.05를 의미한다. Figure 6a shows the results of measurement of arthritis-derived joint tissue from IL1RaKO mice and tIK-IL1RaKO mice produced according to the present invention using H & E staining. The left side is a micrograph of the joint tissue, and the right side is a graph analyzing the degree of inflammation in the joint tissue. In FIG. 6A, * means P <0.05 as compared to negative control.
도 6b는 관절염이 자연적으로 유도되는 IL1RaKO 마우스와, 본 발명에 따라 제작된 tIK-IL1RaKO 마우스 유래의 관절 조직을 safranin O 염색법을 이용하여 측정한 결과를 도시하고 있다. 좌측은 관절 조직에 대한 현미경 사진이고, 우측은 관절 조직에서의 연골 침식 정도를 분석한 그래프이다. 도 6b에서 *는 네거티브 컨트롤에 비하여 P < 0.05를 의미한다.FIG. 6B shows the results of measurement of joint tissue derived from IL1RaKO mice naturally induced arthritis and tIK-IL1RaKO mice prepared according to the present invention using safranin O staining. The left side is a micrograph of the joint tissue and the right side is a graph analyzing the degree of cartilage erosion in the joint tissue. In FIG. 6B, * means P <0.05 as compared to negative control.
도 7은 관절염이 자연적으로 유도되는 IL1RaKO 마우스와, 본 발명에 따라 제작된 tIK-IL1RaKO 마우스에서 관절 부위의 골 손상 정도를 촬영한 사진이다.Figure 7 is a photograph of the degree of bone damage in the joint area of IL1RaKO mice naturally induced arthritis, and tIK-IL1RaKO mouse produced according to the present invention.
도 8a는 관절염이 자연적으로 유도되는 IL1RaKO 마우스와, 본 발명에 따라 제작된 tIK-IL1RaKO 마우스의 관절 부위의 염증성 사이토카인의 mRNA 발현 정도를 측정한 그래프이다. 도 8a에서 *는 네거티브 컨트롤에 비하여 P < 0.05를, **는 P < 0.01을 의미한다. Figure 8a is a graph measuring the mRNA expression level of inflammatory cytokines in the joint region of IL1RaKO mice naturally induced arthritis and tIK-IL1RaKO mouse prepared according to the present invention. In FIG. 8A, * denotes P <0.05 and ** denotes P <0.01 compared to negative control.
도 8b는 관절염이 자연적으로 유도되는 IL1RaKO 마우스와, 본 발명에 따라 제작된 tIK-IL1RaKO 마우스의 관절 조직을 염색한 뒤 염증성 사이토카인의 발현 정도를 촬영한 사진이다. Figure 8b is a photograph of the degree of expression of inflammatory cytokines after staining joint tissue of IL1RaKO mice naturally induced arthritis, and tIK-IL1RaKO mice prepared according to the present invention.
도 9는 관절염이 자연적으로 유도되는 IL1RaKO 마우스와, 본 발명에 따라 제작된 tIK-IL1RaKO 마우스의 혈청 내 염증성 사이토카인의 mRNA 발현 정도를 측정한 그래프이다. 도 9에서 **는 네거티브 컨트롤에 비하여 P < 0.01을 의미한다. 9 is a graph measuring the mRNA expression level of inflammatory cytokines in serum of IL1RaKO mice naturally induced arthritis and tIK-IL1RaKO mice prepared according to the present invention. ** in FIG. 9 means P <0.01 compared to negative control.
도 10a는 관절염이 자연적으로 유도되는 IL-1 Receptor antagonist knock-out(IL1RaKO) 마우스와, 본 발명에 따라 제작된 마우스(tIK-IL1RaKO)의 비장 세포에서 병인 T세포의 분화 정도를 살펴보기 위하여, 병인 T세포인 Th1세포의 양을 측정한 플롯 및 그래프이다. Figure 10a is to examine the degree of differentiation of etiological T cells in spleen cells of IL-1 Receptor antagonist knock-out (IL1RaKO) mice and arthritis induced naturally, according to the invention (tIK-IL1RaKO), Plots and graphs measuring the amount of Th1 cells, which are pathogenic T cells.
도 10b는 관절염이 자연적으로 유도되는 IL-1 Receptor antagonist knock-out(IL1RaKO) 마우스와, 본 발명에 따라 제작된 마우스(tIK-IL1RaKO)의 비장 세포에서 병인 T세포의 분화 정도를 살펴보기 위하여, 병인 T세포인 Th17세포의 양을 측정한 플롯 및 그래프이다 . Figure 10b is to examine the degree of differentiation of etiological T cells in spleen cells of arthritis induced IL-1 Receptor antagonist knock-out (IL1RaKO) mouse, and mouse (tIK-IL1RaKO) produced according to the present invention, Plots and graphs measuring the amount of Th17 cells, the etiological T cells.
도 11은 관절염이 자연적으로 유도되는 IL-1 Receptor antagonist knock-out (IL1RaKO) 마우스와, 본 발명에 따라 제작된 마우스(tIK-IL1RaKO)의 비장세포에서 대식세포 활성인자의 양을 측정한 그래프이다. 도 11에서 *는 네거티브 컨트롤에 비하여 P < 0.05를 의미한다.Figure 11 is a graph measuring the amount of macrophage activator factor in splenocytes of IL-1 Receptor antagonist knock-out (IL1RaKO) mice in which arthritis is naturally induced and mice prepared according to the present invention (tIK-IL1RaKO). . In FIG. 11, * means P <0.05 as compared to negative control.
도 12a는 본 발명에 따라 아이케이 인자의 활성에 관여하는 아미노산을 포함하는 펩타이드를 대식세포에 transfection한 뒤, 세포에서 염증성 사이토카인의 발현 정도를 측정한 그래프이다. 도 12a에서 *는 네거티브 컨트롤에 비하여 P < 0.05를, **는 P < 0.01을 의미한다. Figure 12a is a graph measuring the expression level of inflammatory cytokines in cells after transfection of macrophages with peptides containing amino acids involved in the activity of IK factor in accordance with the present invention. In FIG. 12A, * denotes P <0.05 and ** denotes P <0.01 compared to negative control.
도 12b는 본 발명에 따라 아이케이 인자의 활성에 관여하는 아미노산을 포함하는 펩타이드를 대식세포에 transfection한 뒤, 세포 배양액에서 염증성 사이토카인의 발현 정도를 측정한 그래프이다. 도 12b에서 **는 네거티브 컨트롤에 비하여 P < 0.01를 ***는 P < 0.001을 의미한다. Figure 12b is a graph measuring the expression level of inflammatory cytokines in cell culture after transfection of macrophages with peptides containing amino acids involved in the activity of IK factor according to the present invention. In FIG. 12B, ** denotes P <0.01 and *** denotes P <0.001 as compared to the negative control.
도 13a는 본 발명에 따라 아이케이 인자의 활성에 관여하는 아미노산을 포함하는 펩타이드를 주입한 야생형 마우스의 비장 세포에서 Th0 세포의 분화 정도를 분석한 플롯 및 그래프이다. Figure 13a is a plot and a graph analyzing the degree of differentiation of Th0 cells in spleen cells of wild-type mice injected with peptides containing amino acids involved in the activity of IK factor according to the present invention.
도 13b는 본 발명에 따라 tIK의 활성에 관여하는 아미노산을 포함하는 펩타이드를 주입한 야생형 마우스의 비장 세포에서 Th1 세포의 분화 정도를 분석한 플롯 및 그래프이다. Figure 13b is a plot and graph analyzing the degree of differentiation of Th1 cells in spleen cells of wild-type mice injected with a peptide containing amino acids involved in the activity of tIK according to the present invention.
도 13c는 본 발명에 따라 tIK의 활성에 관여하는 아미노산을 포함하는 펩타이드를 주입한 야생형 마우스의 비장 세포에서 Th17 세포의 분화 정도를 분석한 플롯 및 그래프이다. Figure 13c is a plot and graph analyzing the degree of differentiation of Th17 cells in spleen cells of wild-type mice injected with a peptide containing amino acids involved in the activity of tIK according to the present invention.
도 14a는 본 발명에 따라 아이케이 인자 유래의 펩타이드를 자연 발생 관절염 모델인 IL1RaKO 마우스에 주입한 뒤, 시간 경과에 따른 관절염 지수를 측정한 그래프이고, 도 14b는 해당 마우스에서의 시간 경과에 따른 관절염 발병율을 측정한 그래프이다. Figure 14a is a graph measuring the arthritis index over time after injecting the IK factor-derived peptide into IL1RaKO mice, a naturally occurring arthritis model according to the present invention, Figure 14b is arthritis over time in the mouse It is a graph measuring the incidence rate.
도 15a는 본 발명에 따라 아이케이 인자 유래의 펩타이드를 자연 발생 관절염 모델인 마우스에 주입한 뒤, 해당 마우스의 비장 세포에서 Th17 세포의 분화 정도를 분석한 플롯 및 그래프이다. 도 15a에서 *는 네거티브 컨트롤에 비하여 P < 0.05를 의미한다.Figure 15a is a plot and graph of the differentiation of Th17 cells in spleen cells of the mouse after injecting a peptide derived from IK factor in accordance with the present invention into a mouse that is a naturally occurring arthritis model. In FIG. 15A, * means P <0.05 as compared to negative control.
도 15b는 본 발명에 따라 아이케이 인자 유래의 펩타이드가 주입된 자연 발생 관절염 모델인 마우스의 관절 조직을 염색한 뒤 염증성 사이토카인의 발현 정도를 촬영한 사진이다. Figure 15b is a photograph of the degree of expression of inflammatory cytokines after staining the joint tissue of a mouse, a naturally occurring arthritis model injected with IK factor-derived peptides according to the present invention.
도 16은 3' 말단에 Fc tag 서열이 첨가된 절단된 아이케이 유전자 단편(tIK)을 Baculovirus 발현 시스템을 이용하여 곤충 세포에 주입한 뒤, 아이케이 인자가 발현되었는지를 확인한 SDS-PAGE 측정 사진이다. Figure 16 is a SDS-PAGE measurement photograph confirming that the IK factor was expressed after injecting a truncated IK gene fragment (tIK) having the Fc tag sequence added to the 3 'end into insect cells using a Baculovirus expression system. .
도 17은 5' 말단에 HA tag 서열이 첨가된 절단된 아이케이 유전자 단편(tIK를 코딩하는 유전자 단편)을 아데노-연관바이러스에 삽입한 재조합 아데노-연관바이러스(tIK-AAV)에서 아이케이 인자가 발현된 것을 보여주는 웨스턴-블롯팅 결과이다. 17 shows the IK factor in recombinant adeno-associated virus (tIK-AAV) in which a truncated IK gene fragment (a gene fragment encoding tIK) having an HA tag sequence added to its 5 'end was inserted into an adeno-associated virus. Western-blotting results show expression.
도 18a는 절단된 아이케이 유전자 단편(tIK를 코딩하는 유전자 단편)이 삽입된 아데노-연관바이러스 벡터를 자연 발생 관절염 모델인 IL1RaKO 마우스에 주입한 뒤 시간 경과에 따른 관절염 지수를 측정한 그래프이고, 도 18b는 해당 마우스에서의 시간 경과에 따른 관절염 발병율을 측정한 그래프이다. Figure 18a is a graph measuring the arthritis index over time after injecting the adeno-associated virus vector inserted with the truncated IK gene fragment (gene fragment encoding tIK) into IL1RaKO mice, a naturally occurring arthritis model, 18b is a graph measuring the incidence of arthritis over time in the mouse.
도 19는 5' 말단에 HA tag 서열이 첨가된 절단된 아이케이 유전자 단편(tIK)이 삽입된 플라스미드를 CHO 세포에 주입하고, CHO 세포에서 아이케이 인자가 발현된 것을 보여주는 웨스턴-블롯팅 결과이다. 19 is a Western-blotting result showing that the plasmid inserted with the truncated IK gene fragment (tIK) having the HA tag sequence added to the 5 'end was injected into CHO cells, and that the IK factor was expressed in CHO cells. .
본 발명자들은 생체에서 발현되는 사이토카인의 일종인 아이케이 인자(IK factor)의 새로운 기능을 연구하여, 아이케이 인자 또는 그 단편 및/또는 이들을 코딩하는 핵산을 갖는 유전자 전달체가 예를 들어 류마티스성 관절염과 같은 관절염의 진행을 억제한다는 점을 규명하여 본 발명을 완성하였다. 이하, 첨부하는 도면을 참조하면서 본 발명을 상세하게 설명한다.The present inventors have studied new functions of IK factor, a type of cytokine expressed in vivo, so that gene carriers having IK factors or fragments thereof and / or nucleic acids encoding them are for example rheumatoid arthritis. The present invention was completed by clarifying that the progression of arthritis as described above. EMBODIMENT OF THE INVENTION Hereinafter, this invention is demonstrated in detail, referring an accompanying drawing.
정의Justice
본 명세서에서 용어 "아미노산"은 가장 넓은 의미로 사용되고, 자연-발생 L-아미노산 또는 잔기를 포함하는 것으로 의도된다. 자연-발생 아미노산에 대해 통상적으로 사용되는 1- 및 3-문자 약어가 본 명세서에 사용된다(문헌 [Lehninger, Biochemistry, 2d ed., pp. 71-92, (Worth Publishers: New York, 1975]). 아미노산은 D-아미노산뿐만 아니라 화학적으로-변형된 아미노산, 예컨대 아미노산 유사체, 단백질에 통상적으로 혼입되지 않는 자연-발생 아미노산, 예컨대 노르류신, 및 아미노산의 특징인 것으로 당업계에 공지된 특성을 갖는 화학적으로-합성된 화합물을 포함한다. 예를 들어, 천연 페닐알라닌(Phe) 또는 프롤린(Pro)과 동일한 펩티드 화합물의 입체형태 제한을 허용하는 페닐알라닌 또는 프롤린의 유사체 또는 모방체가 아미노산의 정의 내에 포함된다. 이러한 유사체 및 모방체는 본 명세서에서 아미노산의 "기능적 균등물"로서 지칭된다. 아미노산의 다른 예는 문헌 [Roberts and Vellaccio, The Peptides: Analysis, Synthesis, Biology, Eds. Gross and Meiehofer, Vol. 5, p. 341(Academic Press, Inc.: N.Y. 1983)]에 열거되어 있다.The term "amino acid" is used herein in its broadest sense and is intended to include naturally-occurring L-amino acids or residues. 1- and 3-letter abbreviations commonly used for naturally-occurring amino acids are used herein (Lehninger, Biochemistry, 2d ed., Pp. 71-92, (Worth Publishers: New York, 1975)). Amino acids are chemicals having properties known in the art to be characteristic of not only D-amino acids but also chemically-modified amino acids such as amino acid analogs, naturally-occurring amino acids such as norleucine, and amino acids that are not commonly incorporated into proteins. And synthetically-synthesized compounds include, for example, analogs or mimetics of phenylalanine or proline that allow conformational limitations of the same peptide compound as natural phenylalanine (Phe) or proline (Pro). Analogs and mimetics are referred to herein as "functional equivalents" of amino acids Other examples of amino acids are described in Roberts and Vellaccio, The Peptide. s: Analysis, Synthesis, Biology, Eds. Gross and Meiehofer, Vol. 5, p. 341 (Academic Press, Inc .: N.Y. 1983).
예를 들어, 표준 고체-상 합성 기술에 의해 합성된 합성 펩티드는 유전자에 의해 코딩되는 아미노산으로 제한되지 않으며, 이에 따라 주어진 아미노산에 대해 보다 광범위하게 다양한 치환을 허용한다. 유전자 코드에 의해 코딩되지 않은 아미노산은 본 명세서에서 "아미노산 유사체"로서 지칭되며, 예를 들어 WO 90/01940에 기재되어 있다. 예를 들어, 아미노산 유사체는 Glu 및 Asp에 대한 2-아미노 아디프산 (Aad); Glu 및 Asp에 대한 2-아미노피멜산 (Apm); Met, Leu 및 다른 지방족 아미노산에 대한 2-아미노부티르산 (Abu); Met, Leu 및 다른 지방족 아미노산에 대한 2-아미노헵탄산 (Ahe); Gly에 대한 2-아미노부티르산 (Aib); Val, Leu 및 Ile에 대한 시클로헥실알라닌 (Cha); Arg 및 Lys에 대한 호모아르기닌 (Har); Lys, Arg 및 His에 대한 2,3-디아미노프로피온산 (Dap); Gly, Pro 및 Ala에 대한 N-에틸글리신 (EtGly); Gly, Pro 및 Ala에 대한 N-에틸글리신 (EtGly); Asn 및 Gln에 대한 N-에틸아스파라긴 (EtAsn); Lys에 대한 히드록시리신 (Hyl); Lys에 대한 알로히드록시리신 (AHyl); Pro, Ser 및 Thr에 대한 3-(및 4-)히드록시프롤린 (3Hyp, 4Hyp); Ile, Leu 및 Val에 대한 알로-이소류신(AIle); Arg에 대한 4-아미디노페닐알라닌; Gly, Pro 및 Ala에 대한 N-메틸글리신 (MeGly, 사르코신); Ile에 대한 N-메틸이소류신 (MeIle); Met 및 다른 지방족 아미노산에 대한 노르발린 (Nva); Met 및 다른 지방족 아미노산을 위한 노르류신 (Nle); Lys, Arg 및 His에 대한 오르니틴 (Orn); Thr, Asn및 Gln에 대한 시트룰린 (Cit) 및 메티오닌 술폭시드 (MSO); 및 Phe에 대한 N-메틸페닐알라닌 (MePhe), 트리메틸페닐알라닌, 할로-(F-, Cl-, Br- 또는 I-)페닐알라닌 또는 트리플루오릴페닐알라닌을 포함한다.For example, synthetic peptides synthesized by standard solid-phase synthesis techniques are not limited to amino acids encoded by genes, thus allowing a wider variety of substitutions for a given amino acid. Amino acids not encoded by the genetic code are referred to herein as "amino acid analogs" and are described, for example, in WO 90/01940. For example, amino acid analogs include 2-amino adipic acid (Aad) for Glu and Asp; 2-aminopimelic acid (Apm) for Glu and Asp; 2-aminobutyric acid (Abu) for Met, Leu and other aliphatic amino acids; 2-aminoheptanoic acid (Ahe) for Met, Leu and other aliphatic amino acids; 2-aminobutyric acid (Aib) for Gly; Cyclohexylalanine (Cha) for Val, Leu and Ile; Homoarginine (Har) for Arg and Lys; 2,3-diaminopropionic acid (Dap) for Lys, Arg and His; N-ethylglycine (EtGly) for Gly, Pro and Ala; N-ethylglycine (EtGly) for Gly, Pro and Ala; N-ethylasparagine (EtAsn) for Asn and Gln; Hydroxylysine (Hyl) for Lys; Allohydroxylysine (AHyl) for Lys; 3- (and 4-) hydroxyproline (3Hyp, 4Hyp) for Pro, Ser and Thr; Allo-isoleucine (AIle) for Ile, Leu and Val; 4-amidinophenylalanine for Arg; N-methylglycine (MeGly, Sarcosine) for Gly, Pro and Ala; N-methylisoleucine (MeIle) for Ile; Norvaline (Nva) for Met and other aliphatic amino acids; Norleucine (Nle) for Met and other aliphatic amino acids; Ornithine (Orn) for Lys, Arg and His; Citrulline (Cit) and methionine sulfoxide (MSO) for Thr, Asn and Gln; And N-methylphenylalanine (MePhe), trimethylphenylalanine, halo- (F-, Cl-, Br- or I-) phenylalanine or trifluorylphenylalanine for Phe.
본 명세서에서 용어 "펩타이드"는 자연적으로 존재하는 것으로부터 분리하였거나, 재조합 기술(recombinant technique)에 의하여 또는 화학적으로 합성된 단백질, 단백질 단편 및 펩타이드를 모두 포함한다. 예를 들어 본 발명의 펩타이드는 적어도 5개, 바람직하게는 10개의 아미노산으로 구성되어 있다. As used herein, the term "peptide" includes all proteins, protein fragments and peptides that have been isolated from naturally occurring, synthesized by a recombinant technique or chemically synthesized. For example, the peptide of the present invention consists of at least 5, preferably 10 amino acids.
특정 실시양태에서, 화합물의 변이체, 예컨대 하나 이상의 아미노산 치환을 갖는 펩타이드 변이체가 제공된다. 본 명세서에서 "펩타이드 변이체(peptide variants)"란 하나 또는 그 이상의 아미노산이 펩타이드의 아미노산 서열에 치환(substitutions), 결실(deletions), 첨가(additions) 및/또는 삽입(insertions)되어 있으면서, 원래의 아미노산으로 구성된 펩타이드와 거의 동일한 생물학적 기능을 발휘하는 것을 말한다. 펩타이드 변이체는 원래의 펩타이드와 70% 이상, 바람직하게는 90% 이상, 더욱 바람직하게는 95% 이상의 동일성(identity)을 가지고 있어야 한다. 이러한 치환체로서 "보존성"이라고 알려진 아미노산 치환제가 포함될 수 있다. 변이체는 또한 비보존성(nonconservative) 변화를 포함할 수도 있다. 하나의 예시적인 실시양태에서, 변이체 폴리펩타이드의 서열은 5개 또는 그 이하의 아미노산이 치환, 결실, 부가 또는 삽입됨으로서 원래의 서열과 달라진다. 변이체들은 또한 펩타이드의 면역원성(immunogenicity), 2차 구조(secondary structure) 및 수치료성(hydropathic nature)에 최소한의 영향을 주는 아미노산들의 결실 또는 부가에 의해 변화될 수 있다.In certain embodiments, variants of a compound are provided, such as peptide variants having one or more amino acid substitutions. As used herein, "peptide variants" refers to the original amino acid while one or more amino acids are substituted, deleted, added and / or inserted into the amino acid sequence of the peptide. It is said to exert almost the same biological function as the peptide consisting of. Peptide variants should have at least 70% identity, preferably at least 90% and more preferably at least 95% identity with the original peptide. Such substituents may include amino acid substituents known as "conservatives". Variants may also include nonconservative changes. In one exemplary embodiment, the sequence of the variant polypeptide differs from the original sequence by replacing, deleting, adding or inserting five or fewer amino acids. Variants may also be altered by deletion or addition of amino acids with minimal impact on the immunogenicity, secondary structure and hydropathic nature of the peptide.
달리 언급되지 않는 한, 본 명세서에서"보존성" 치환이란 하나의 아미노산이 다른 아미노산으로 치환되었을 때에도 폴리펩타이드의 2차 구조 및 수치료성(hydropathic nature) 등의 특성에 큰 변화가 없는 것을 의미한다. 이러한 보존성 치환과 관련하여 아미노산 변이는 아미노산 곁사슬 치환체의 상대적 유사성, 예컨대 극성(polarity), 전하(charge), 수용성(solubility), 소수성(hydrophobicity), 친수성(hydrophilicity) 및/또는 양친화성(amphipathic nature) 등의 유사성을 기초로 하여 얻어질 수 있다. Unless stated otherwise, “conservative” substitutions herein means that there is no significant change in properties such as secondary structure and hydropathic nature of the polypeptide even when one amino acid is substituted with another amino acid. Amino acid variations with respect to such conservative substitutions may be attributed to the relative similarity of amino acid side chain substituents such as polarity, charge, solubility, hydrophobicity, hydrophilicity and / or amphipathic nature. Can be obtained based on similarity.
예를 들어 아미노산은 공통적인 곁사슬 특성에 따라 1) 소수성(류신, 메티오닌, 알라닌, 발린, 류신, 이소류신) 2) 중성 친수성(시스테인, 세린, 트레오닌, 아스파라긴, 글루타민), 3) 산성(아스파르트산, 글루탐산), 4) 염기성(히스티딘, 리신, 아르기닌), 5) 쇄 방향에 영향을 미치는 잔기 (글리신, 프롤린), 6) 방향족 (트립토판, 티로신, 페닐알라닌)으로 분류될 수 있다. 보존적 치환은 이들 각각의 클래스 중 하나의 구성원을 동일한 클래스의 다른 구성원으로 교환하는 것을 수반할 것이다. For example, amino acids are based on common side chain properties: 1) hydrophobic (leucine, methionine, alanine, valine, leucine, isoleucine) 2) neutral hydrophilic (cysteine, serine, threonine, asparagine, glutamine), 3) acidic (aspartic acid, Glutamic acid), 4) basic (histidine, lysine, arginine), 5) residues that affect chain orientation (glycine, proline), and 6) aromatics (tryptophan, tyrosine, phenylalanine). Conservative substitutions will entail exchanging a member of one of each of these classes for another member of the same class.
아미노산 곁사슬 치환체의 크기, 모양 및 종류에 대한 분석에 의하여, 아르기닌, 리신과 히스티딘은 모두 양전하를 띤 잔기이고; 알라닌, 글리신과 세린은 유사한 크기를 갖으며; 페닐알라닌, 트립토판과 티로신은 유사한 모양을 갖는다는 것을 알 수 있다. 따라서 이러한 고려 사항에 기초하여, 아르기닌, 라이신과 히스티딘; 알라닌, 글리신과 세린; 그리고 페닐알라닌, 트립토판과 티로신은 생물학적으로 기능 균등물이라 할 수 있다.By analysis of the size, shape and type of amino acid side chain substituents, arginine, lysine and histidine are all positively charged residues; Alanine, glycine and serine have similar sizes; It can be seen that phenylalanine, tryptophan and tyrosine have a similar shape. Thus, based on these considerations, arginine, lysine and histidine; Alanine, glycine and serine; Phenylalanine, tryptophan, and tyrosine are biologically equivalent functions.
변이를 도입하는 데 있어서, 아미노산의 소수성 인덱스 (hydropathic idex)가 고려될 수 있다. 각각의 아미노산은 소수성과 전하에 따라 소수성 인덱스가 부여되어 있다: 이소류신 (+4.5); 발린 (+4.2); 류신 (+3.8); 페닐알라닌(+2.8); 시스테인/시스타인 (+2.5); 메티오닌 (+1.9); 알라닌 (+1.8); 글리신 (-0.4); 트레오닌(-0.7); 세린 (-0.8); 트립토판 (-0.9); 티로신 (-1.3); 프롤린 (-1.6); 히스티딘 (-3.2); 글루탐산 (-3.5); 글루타민 (-3.5); 아스파르트산 (-3.5); 아스파라긴 (-3.5); 라이신 (-3.9); 및 아르기닌 (-4.5).In introducing variants, hydropathic idex of amino acids can be considered. Each amino acid is assigned a hydrophobicity index depending on its hydrophobicity and charge: isoleucine (+4.5); Valine (+4.2); Leucine (+3.8); Phenylalanine (+2.8); Cysteine / cysteine (+2.5); Methionine (+1.9); Alanine (+1.8); Glycine (-0.4); Threonine (-0.7); Serine (-0.8); Tryptophan (-0.9); Tyrosine (-1.3); Proline (-1.6); Histidine (-3.2); Glutamic acid (-3.5); Glutamine (-3.5); Aspartic acid (-3.5); Asparagine (-3.5); Lysine (-3.9); And arginine (-4.5).
단백질의 상호적인 생물학적 기능(interactive biological function)을 부여하는 데 있어서 소수성 아미노산 인덱스는 매우 중요하다. 유사한 소수성 인덱스를 가지는 아미노산으로 치환하여야 유사한 생물학적 활성을 보유할 수 있다는 것은 공지된 사실이다. 소수성 인덱스를 참조하여 변이를 도입시키는 경우, 바람직하게는 ± 2 이내, 보다 바람직하게는 ± 1 이내, 보다 더 바람직하게는 ± 0.5 이내의 소수성 인덱스 차이를 나타내는 아미노산 사이에 치환을 한다. The hydrophobic amino acid index is very important in conferring the interactive biological function of proteins. It is known that substitution with amino acids having similar hydrophobicity indexes can retain similar biological activity. When introducing mutations with reference to the hydrophobicity index, substitutions are made between amino acids which exhibit a hydrophobicity index difference of preferably within ± 2, more preferably within ± 1, even more preferably within ± 0.5.
한편, 유사한 친수성 값(hydrophilicity value)을 가지는 아미노산 사이의 치환이 균등한 생물학적 활성을 갖는 단백질을 초래한다는 것도 잘 알려져 있다. 미국 특허 제4,554,101호에 개시된 바와 같이, 다음의 친수성 값이 각각의 아미노산 잔기에 부여되어 있다: 아르기닌 (+3.0); 리신 (+3.0); 아스파르트산(+3.0 ±1); 글루탐산(+3.0 ±1); 세린 (+0.3); 아스파라긴 (+0.2); 글루타민 (+0.2); 글리신 (0); 트레오닌 (-0.4); 프롤린 (-0.5 ± 1); 알라닌 (-0.5); 히스티딘 (-0.5); 시스테인 (-1.0); 메티오닌 (-1.3); 발린 (-1.5); 리신(-1.8); 이소류신 (-1.8); 티로신 (-2.3); 페닐알라닌 (-2.5); 트립토판 (-3.4). 친수성 값을 참조하여 변이를 도입시키는 경우, 바람직하게는 ± 2 이내, 보다 바람직하게는 ± 1 이내, 보다 더 바람직하게는 ± 0.5 이내의 친수성 값 차이를 나타내는 아미노산 사이에 치환을 한다. On the other hand, it is also well known that substitutions between amino acids having similar hydrophilicity values result in proteins with equivalent biological activity. As disclosed in US Pat. No. 4,554,101, the following hydrophilicity values are assigned to each amino acid residue: arginine (+3.0); Lysine (+3.0); Aspartic acid (+ 3.0 ± 1); Glutamic acid (+ 3.0 ± 1); Serine (+0.3); Asparagine (+0.2); Glutamine (+0.2); Glycine (0); Threonine (-0.4); Proline (-0.5 ± 1); Alanine (-0.5); Histidine (-0.5); Cysteine (-1.0); Methionine (-1.3); Valine (-1.5); Lysine (-1.8); Isoleucine (-1.8); Tyrosine (-2.3); Phenylalanine (-2.5); Tryptophan (-3.4). When introducing mutations with reference to hydrophilicity values, substitutions are made between amino acids which exhibit a hydrophilicity value difference of preferably within ± 2, more preferably within ± 1 and even more preferably within ± 0.5.
분자의 활성을 전체적으로 변경시키지 않는 단백질에서의 아미노산 교환은 당해 분야에 공지되어 있다(H.Neurath, R.L.Hill, The Proteins, Academic Press, New York, 1979). 가장 통상적으로 일어나는 교환은 아미노산 잔기 Ala/Ser, Val/Ile, Asp/Glu, Thr/Ser, Ala/Gly, Ala/Thr, Ser/Asn, Ala/Val, Ser/Gly, Thy/Phe, Ala/Pro, Lys/Arg, Asp/Asn, Leu/Ile, Leu/Val, Ala/Glu, Asp/Gly 간의 교환이다.Amino acid exchange in proteins that do not alter the activity of the molecule as a whole is known in the art (H. Neurode, R. L. Hill, The Proteins, Academic Press, New York, 1979). The most commonly occurring exchanges are amino acid residues Ala / Ser, Val / Ile, Asp / Glu, Thr / Ser, Ala / Gly, Ala / Thr, Ser / Asn, Ala / Val, Ser / Gly, Thy / Phe, Ala / Exchange between Pro, Lys / Arg, Asp / Asn, Leu / Ile, Leu / Val, Ala / Glu, Asp / Gly.
본 명세서에서 "폴리뉴클레오티드" 또는 "핵산"은 교환 가능하게 사용되며, 임의 길이의 뉴클레오티드의 중합체를 지칭하고 DNA(예컨대 cDNA) 및 RNA 분자를 포괄적으로 포함한다. 핵산 분자의 구성단위인 "뉴클레오티드"는 디옥시리보뉴클레오티드, 리보뉴클레오티드, 변형된 뉴클레오티드 또는 뉴클레오티드, 및/또는 그 유사체, 또는 DNA 또는 RNA 폴리머라제(polymerase)에 의해, 또는 합성 반응에 의해 중합체 내로 혼입될 수 있는 임의의 기질일 수 있다. 폴리뉴클레오티드는 변형된 뉴클레오티드, 당 또는 뉴클레오티드가 변형된 유사체(analogue), 예컨대 메틸화 뉴클레오티드 및 그 유사체를 포함할 수 있다(Scheit, Nucleotide Analogs, John Wiley, New York(1980); Uhlman 및 Peyman, Chemical Reviews, 90:543-584(1990)).As used herein, “polynucleotide” or “nucleic acid” is used interchangeably and refers to a polymer of nucleotides of any length and encompasses DNA (such as cDNA) and RNA molecules inclusively. The “nucleotide”, a structural unit of a nucleic acid molecule, can be incorporated into a polymer by deoxyribonucleotides, ribonucleotides, modified nucleotides or nucleotides, and / or analogs thereof, or by DNA or RNA polymerase, or by synthetic reactions. May be any substrate present. Polynucleotides may include modified nucleotides, sugars or analogs in which the nucleotides are modified, such as methylated nucleotides and analogs thereof (Scheit, Nucleotide Analogs, John Wiley, New York (1980); Uhlman and Peyman, Chemical Reviews , 90: 543-584 (1990).
뉴클레오티드에서의 변이는 단백질에서 변이를 가져오지 않는 것도 있다. 이러한 핵산은 기능적으로 균등한 코돈 또는 동일한 아미노산을 코딩하는 코돈, 또는 생물학적으로 균등한 아미노산을 코딩하는 코돈을 포함하는 핵산분자를 포함한다. 또한, 뉴클레오티드에서의 변이가 단백질 자체에 변화를 가져올 수도 있다. 단백질의 아미노산에 변화를 가져오는 변이인 경우에도 본 발명의 단백질과 거의 동일한 활성을 나타내는 것이 얻어질 수 있다.Variations in nucleotides do not result in mutations in proteins. Such nucleic acids include nucleic acid molecules comprising a codon that is functionally equivalent or a codon that encodes the same amino acid, or a codon that encodes a biologically equivalent amino acid. In addition, variations in nucleotides may result in changes in the protein itself. Even in the case of mutations that bring about changes in amino acids of proteins, those exhibiting almost the same activity as the proteins of the present invention can be obtained.
본 발명의 아이케이 인자 및 그 단편 및/또는 이들을 코딩하는 핵산이 가지는 특징, 예를 들어 관절염을 치료하는 효과를 갖는 범위에서, 본 발명의 펩타이드 및 핵산 분자는 서열목록에 기재된 아미노산 서열 또는 뉴클레오티드 서열에 한정되지 않는다는 것은 통상의 기술자라면 쉽게 인식할 것이다. 예를 들어, 본 발명의 펩타이드에 포함될 수 있는 생물학적 기능 균등물은 본 발명의 펩타이드와 균등한 생물학적 활성을 발휘하는 아미노산 서열의 변이를 가지는 펩타이드일 수 있다. In the scope of having the effect of the IK factor of the present invention and fragments thereof and / or nucleic acids encoding them, such as arthritis, the peptides and nucleic acid molecules of the present invention are amino acid sequences or nucleotide sequences listed in the sequence listing. It should be appreciated by those skilled in the art that the present invention is not limited thereto. For example, the biological functional equivalents that may be included in the peptide of the present invention may be a peptide having a variation in the amino acid sequence exhibiting biological activity equivalent to that of the peptide of the present invention.
일반적으로, 본 명세서상에 언급되어 있는 펩타이드들(융합 단백질 포함) 및 폴리뉴클레오티드들은 분리된(isolated) 것이다. "분리된(isolated)" 펩타이드 또는 폴리뉴클레오티드란 원래의 환경으로부터 제거된 것이다. 예를 들어, 자연 상태로 존재하는 단백질은 그 상태에서 함께 존재하는 물질들 전부 혹은 일부를 제거함으로써 분리되는 것이다. 이러한 폴리펩타이드는 적어도 90% 이상의 순도를 지녀야 하며, 바람직하게는 95%, 더욱 바람직하게는 99% 이상의 순도를 가져야 한다. 폴리뉴클레오티드는 벡터 내에서 클로닝 시킴으로써 분리된다. In general, the peptides (including fusion proteins) and polynucleotides mentioned herein are isolated. An “isolated” peptide or polynucleotide is one that is removed from its original environment. For example, proteins that exist in nature are separated by removing some or all of the substances that exist together in that state. Such polypeptides should be at least 90% pure, preferably at least 95%, more preferably at least 99% pure. Polynucleotides are isolated by cloning in a vector.
본 명세서에서 사용된 용어 "벡터"는 숙주세포에 전달이 가능하며 바람직하게는 하나 이상의 목적 유전자 또는 서열의 발현이 가능하도록 만들어진 구조물을 의미한다. 예를 들면 벡터에는 바이러스 벡터(viral vectors), DNA 또는 RNA 발현 벡터(expression vectors), 플라스미드(plasmid), 코스미드(cosmid) 또는 파지벡터(phage vectors), CCA(cationic condensing agents)와 연결된 DNA 또는 RNA 발현 벡터, 리포좀(liposomes)으로 포장된 DNA 또는 RNA 발현 벡터, 프로듀서 세포(producer cells)와 같은 특정 진핵세포(eukaryotic cells) 등이 포함된다. As used herein, the term "vector" refers to a construct that is capable of delivery to a host cell and preferably enables expression of one or more desired genes or sequences. For example, a vector may include viral vectors, DNA or RNA expression vectors, plasmids, cosmids or phage vectors, DNA linked to cationic condensing agents, or RNA expression vectors, DNA or RNA expression vectors packaged in liposomes, specific eukaryotic cells such as producer cells, and the like.
본 명세서에서 "발현 조절 서열(expression control sequence)"은 핵산의 전사(transcription)를 조절하는 핵산 서열을 의미한다. 발현 조절 서열에는 구조 프로모터(constitutive promoter) 또는 유도 프로모터(inducible promoter)와 같은 프로모터 또는 인핸서(enhancer) 등이 있다. 발현 조절 서열은 전사될 핵산 서열에 연결되어 있다.As used herein, "expression control sequence" refers to a nucleic acid sequence that controls the transcription of a nucleic acid. Expression control sequences include promoters or enhancers, such as constitutive promoters or inducible promoters. The expression control sequence is linked to the nucleic acid sequence to be transcribed.
본 명세서에서 용어 "작동가능하게 결합된(operatively linked)"은 핵산 발현 조절 서열(예: 프로모터, 시그널서열, 또는 전사조절인자 결합 위치의 어레이)과 다른 핵산 서열사이의 기능적인 결합을 의미하며, 이에 의해 상기 조절 서열은 상기 다른 핵산 서열의 전사 및/또는 해독을 조절하게 된다. As used herein, the term “operatively linked” refers to a functional binding between a nucleic acid expression control sequence (eg, an array of promoters, signal sequences, or transcriptional regulator binding sites) and other nucleic acid sequences, The regulatory sequence thereby controls the transcription and / or translation of the other nucleic acid sequence.
본 명세서에서 사용된 용어 "약학적 유효량" 또는 "치료학적 유효량"이란, 유효 성분인 펩타이드 또는 그 단편 및/또는 이들을 코딩하는 핵산의 효능 또는 활성을 달성하는데 충분한 양을 의미한다. 예를 들어 본 발명에 따른 펩타이드 또는 이들 펩타이드를 코딩하는 핵산을 포함하는 유전자 전달체를 함유하는 약학적 조성물은 류마티스성 관절염을 비롯하여, 통풍성 관절염, 다발성 관절염, 변형성 관절염과 같은 만성 관절염을 치료 및/또는 예방하는데 적용될 수 있다.As used herein, the term "pharmaceutically effective amount" or "therapeutically effective amount" means an amount sufficient to achieve the efficacy or activity of the active ingredient peptide or fragments thereof and / or nucleic acids encoding them. For example, a pharmaceutical composition containing a gene carrier comprising a peptide according to the present invention or a nucleic acid encoding these peptides can be used to treat and / or treat chronic arthritis, including gouty arthritis, polyarthritis, deformable arthritis, including rheumatoid arthritis. Can be applied to prevent.
펩타이드, 핵산 및 벡터Peptides, Nucleic Acids and Vectors
아이케이 인자(IK factor)는 항원을 CD4 T세포에 제시할 수 있는 항원제시세포(APC; antigen presenting cell) 표면의 주조직적합성 복합체 Ⅱ(MHC Ⅱ; Major histocompatibility complex Ⅱ)의 발현을 억제하는 사이토카인이다. 아이케이 인자는 cAMP를 활성화시켜, 바이러스 감염에 의해 유도된 인터페론-감마(IFN-γ, interferon-γ)에 의해 발현되는 MHC class Ⅱ의 발현을 억제하는 것으로 알려져 있다. IK factor is a cytokine that inhibits the expression of major histocompatibility complex II (MHC II) on the surface of antigen presenting cells (APCs) capable of presenting antigens to CD4 T cells. Cain. IK factor is known to activate cAMP to inhibit the expression of MHC class II expressed by interferon-gamma (IFN-γ, interferon-γ) induced by viral infection.
도 1에 예시적으로 도시된 것과 같이, 아이케이 인자(IK factor)를 코딩하는 전장-핵산은 N-말단(5' 말단) 영역에 Nuclear Localization Signal Sequence(NLS)와 trimeric coiled-coil motif를 지니고 있다. 또한 아이케이 인자를 코딩하는 전장 핵산의 C-말단(3' 말단) 영역에는 주로 핵단백질에서 발견되는 Arg(R)-Asp(D)와 Arg(R)-Glu(E) 서열이 반복되어 있는 RED domain과 3개의 NLS를 가지고 있다. 예시적으로 아이케이 인자를 코딩하는 전장-핵산은 서열식별번호:1로 표시되는 1,674개의 뉴클레오티드로 구성되는 폴리뉴클레오티드이다. 아이케이 인자는 서열식별번호:1의 마지막의 전사 종결 코돈을 제외하고 발현되어, 서열식별번호:2의 557개의 아미노산으로 구성될 수 있다. 다만, 본 발명이 속하는 기술분야에서 통상의 지식을 가진 자라면, 본 발명에서 사용될 수 있는 아이케이 인자 및 이를 코딩하는 아이케이 핵산이 서열목록에 기재된 것으로 제한되지 않는다는 점을 인식할 것이다. As exemplarily shown in FIG. 1, the full-nucleic acid encoding the IK factor has a Nuclear Localization Signal Sequence (NLS) and a trimeric coiled-coil motif in the N-terminal (5 'end) region. have. In addition, the C-terminal (3 'end) region of the full-length nucleic acid encoding the IK factor repeats the Arg (R) -Asp (D) and Arg (R) -Glu (E) sequences found mainly in nuclear proteins. It has a RED domain and three NLSs. Exemplary full-nucleic acid encoding an IK factor is a polynucleotide consisting of 1,674 nucleotides represented by SEQ ID NO: 1. The IK factor may be expressed except for the last transcription termination codon of SEQ ID NO: 1, and may consist of 557 amino acids of SEQ ID NO: 2. However, those skilled in the art will recognize that the IK factor that can be used in the present invention and the IK nucleic acid encoding the same are not limited to those listed in the sequence listing.
관절은 뼈와 뼈 사이에 활막, 연골, 뼈, 활액 등이 유기적으로 연결되어 있으며, 사이토카인을 통하여 서로 영향을 미치는 복합 조직이다. 관절 내에 만성 염증, 조직 괴사, 세포침윤, 신생혈관 생성, 관절 파괴 등의 현상은 류마티스 관절염의 주요한 병인으로 알려져 있다. 류마티스 관절염의 병인에 있어서 필수적인 병리 소견으로 알려져 있는 판누스라고 불리는 활막 조직은 관절 주위의 조직을 침범하여 골흡수가 유발된다. 판누스에는 항원제시세포와 T세포가 매우 높은 밀도로 관찰되는데, 이들 세포의 상호작용으로 인하여 T세포에 의해 유도되는 면역 반응의 시작과 확대가 일어나는 것으로 알려져 있다. 실제 류마티스 관절염 환자의 판누스에는 MHC class Ⅱ를 발현하는 항원제시세포가 많이 관찰되고, 같은 부위에 HLA-DR-양성 세포와 T세포가 위치한다. 따라서 류마티스 관절염을 치료를 위해서는 MHC class Ⅱ를 발현하는 항원제시세포와 T세포 사이의 상호작용을 억제하는 것이 효과적일 수 있다. Joints are organically connected between the bone and bone of the synovial membrane, cartilage, bone, synovial fluid, and is a complex tissue that affects each other through cytokines. Symptoms such as chronic inflammation, tissue necrosis, cell infiltration, neovascularization, and joint destruction in joints are known as the major etiology of rheumatoid arthritis. Synovial tissue called pannus, known as an essential pathology for the development of rheumatoid arthritis, invades tissue around the joint and causes bone resorption. In Pannus, antigen-presenting cells and T cells are observed at a very high density. The interaction of these cells is known to initiate and widen the immune response induced by T cells. In fact, many antigen-presenting cells expressing MHC class II are observed in Pannus patients with rheumatoid arthritis, and HLA-DR-positive cells and T cells are located at the same site. Therefore, to treat rheumatoid arthritis, it may be effective to suppress the interaction between antigen-presenting cells expressing MHC class II and T cells.
특히, 류마티스성 관절염의 병소인 활막 내에는 IL-17+ T세포 및 기능적으로 활성화된 IL-17이 존재한다. IL-17은 류마티스성 관절염 환자의 혈청에서 높게 검출되는데, 그 중에서도 IL-17A는 류마티스 관절염 환자의 활막세포 내 IL-1β와 IL-6의 발현을 유도한다. 관절염과 관련해서 IL-17은 연골세포와 조골세포 내 기질 생산을 억제하여 관절의 손상을 일으키고, 조직 재생이 결핍되게 한다. 또한 IL-17은 matrix metalloproteinases(MMPs)의 발현과 기능을 활성화시키고 종양괴사인자(TNF)와 함께 마우스 모델 내 비가역적인 연골손상을 일으킨다. 염증 반응에서 Th17세포와 활막세포와의 국소적인 상호작용을 통해 활막세포 내 MMP의 분비 및 IL-1β와 IL-6의 발현이 촉진된다. 골 파괴와 관련하여 IL-17은 조골세포 상에 receptor activator of NF-κB ligand (RANKL)의 발현을 증가시켜 파골세포(osteoclasts)내 RANK 신호를 증폭시킨다. 특히 RANKL을 발현하는 Th17세포는 파골세포 분화 과정에 중요한 역할을 하는 것으로 알려져 있다. In particular, there are IL-17 + T cells and functionally activated IL-17 in the synovial membrane, a lesion of rheumatoid arthritis. IL-17 is highly detected in the serum of rheumatoid arthritis patients, among which IL-17A induces the expression of IL-1β and IL-6 in synovial cells of rheumatoid arthritis patients. In connection with arthritis, IL-17 inhibits the production of matrix in chondrocytes and osteoblasts, causing joint damage and deficient tissue regeneration. IL-17 also activates the expression and function of matrix metalloproteinases (MMPs) and causes irreversible cartilage damage in mouse models with tumor necrosis factor (TNF). Local inflammation of Th17 cells and synovial cells in the inflammatory response promotes the secretion of MMPs and expression of IL-1β and IL-6 in synovial cells. In connection with bone destruction, IL-17 amplifies RANK signals in osteoclasts by increasing the expression of receptor activator of NF-κB ligand (RANKL) on osteoblasts. In particular, Th17 cells expressing RANKL are known to play an important role in the osteoclast differentiation process.
본 발명에 따르면, 아이케이 인자 또는 그 단편을 코딩하는 핵산은 관절염의 진행을 억제, 예방할 수 있다. 특히 이들 핵산은 류마티스성 관절염과 관련한 사이토카인의 분비를 억제하고, 면역 세포의 분화를 차단한다. According to the present invention, the nucleic acid encoding the IK factor or fragment thereof can inhibit and prevent the progression of arthritis. In particular, these nucleic acids inhibit the secretion of cytokines associated with rheumatoid arthritis and block the differentiation of immune cells.
또한 본 발명의 예시적인 실시 형태에서, 아이케이 인자의 단편, 예를 들어 N-말단이 절단된 단편(본 명세서에서 "tIK 인자" 또는 "tIK"로 약칭될 수 있다)을 코딩하는 핵산 단편(tIK 핵산 단편)은 적절한 발현 시스템에서 발현될 수 있다. 이들 핵산 단편이 발현되어, 관절염의 주요 증상의 하나인 염증 면역 반응에 관여하는 class Ⅱ MHC(MHC Ⅱ)의 발현을 억제한다. 구체적으로 본 발명의 예시적인 실시예에 따르면, 일부 뉴클레오티드가 절단된 tIK 핵산 단편 및/또는 그 중 일부 뉴클레오티드를 치환한 돌연변이 tIK 핵산 단편이 MHC Ⅱ의 발현에 관여하는 CIITA(class Ⅱ, major histocompatibility complex, transactivator)의 발현을 억제하는 상위 조절 인자인 CDCA3(Cell division cycle-associated protein 3), CNOT1(CCR4-NOT transcription complex subunit 1) 및 MAPK1(Mitogen-activated protein kinase 1)의 발현을 증가시킨다(도 3 참조). 또한 일부 뉴클레오티드가 치환된 돌연변이 tIK는 이러한 효과가 반감될 수 있는 것으로 보아 아이케이 인자 중의 특정 부위가 MHC Ⅱ의 상위 조절 인자의 발현에 관여하는 생리적 활성 부위라는 점을 확인하였다. Also in an exemplary embodiment of the invention, a nucleic acid fragment encoding a fragment of an IK factor, eg, an N-terminally truncated fragment (which may be abbreviated herein as "tIK factor" or "tIK") tIK nucleic acid fragments) can be expressed in an appropriate expression system. These nucleic acid fragments are expressed to inhibit the expression of class II MHC (MHC II) involved in the inflammatory immune response, one of the main symptoms of arthritis. Specifically, according to an exemplary embodiment of the present invention, a tITA nucleic acid fragment in which some nucleotides are cleaved and / or a mutant tIK nucleic acid fragment in which some nucleotides are substituted is involved in the expression of MHC II (class II, major histocompatibility complex). Increasing the expression of cell division cycle-associated protein 3 (CDCA3), CCR4-NOT transcription complex subunit 1 (CNOT1) and miogen-activated protein kinase 1 (MAPK1), which are high regulatory factors that inhibit the expression of transactivator (FIG. 3). In addition, the mutant tIK substituted with some nucleotides was found to be able to halve this effect, confirming that a specific site in the IK factor is a physiologically active site involved in the expression of a higher regulatory factor of MHC II.
본 발명의 예시적인 실시형태에 따르면, 관절염이 자연적으로 유발되는 마우스(IL1RaKO)에 비하여, 아이케이 인자의 단편, 예를 들어 N-말단이 절단된 단편인 tIK를 발현하도록 유도된 전이 마우스(본 명세서에서 "tIK-IL1RaKO")에서 관절염의 유발 정도가 크게 감소하였다(도 4a, 도 4b 및 도 5 참조). 아울러, tIK-IL1RaKO 마우스의 관절 조직에서 염증의 유발 정도나 연골 손상 정도는 IK1RaKO 마우스의 관절 조직에서 훨씬 줄었으며(도 6a 및 도 6b 참조), 골 손상 정도도 크게 감소하였다(도 7 참조). According to exemplary embodiments of the invention, compared to mice naturally induced by arthritis (IL1RaKO), metastatic mice induced to express fragments of the IK factor, e.g., tIK, a truncated fragment of the N-terminus (see In the specification, "tIK-IL1RaKO") significantly reduced the incidence of arthritis (see FIGS. 4A, 4B and 5). In addition, the degree of inflammation or cartilage damage in the articular tissue of tIK-IL1RaKO mice was significantly reduced in the articular tissues of IK1RaKO mice (see FIGS. 6A and 6B), and the degree of bone damage was also greatly reduced (see FIG. 7).
특히, tIK-IL1RaKO 마우스는 IL1RaKO 마우스에 비하여 관절 조직 및 혈청에서 관절염과 관련한 사이토카인의 분비가 크게 감소되어 있다. 구체적으로 절단된 아이케이 인자(tIK 인자)가 발현되도록 유도된 전이 마우스(tIK-IL1RaKO)는 자연적으로 관절염이 유발된 마우스(IL-1 antagonist-receptor knockout mouse, IL1RaKO 마우스)와 비교해서 관절 부위는 물론(도 8a 및 도 8b 참조), 혈청 내에서도 염증을 유발하는 사이토카인의 발현이 크게 감소한다(도 9 참조). In particular, tIK-IL1RaKO mice have significantly reduced secretion of cytokines associated with arthritis in joint tissues and serum compared to IL1RaKO mice. Specifically, the metastatic mouse (tIK-IL1RaKO) induced to express the cleaved IK factor (tIK factor) is compared to the arthritis-induced mice (IL-1 antagonist-receptor knockout mouse, IL1RaKO mouse). Of course (see FIGS. 8A and 8B), the expression of cytokine causing inflammation also decreases significantly in serum (see FIG. 9).
본 발명의 예시적인 실시예에서 아이케이 인자의 단편을 코딩하는 핵산은 적절한 발현 시스템을 통해 발현되어 관절염과 관련된 염증성 면역 반응에 관여하는 사이토카인인 인터류킨-1 베타(Interleukin-1 beta, IL-1β), 인터류킨-6(Interleukin-6, IL-6), 인터류킨-17(IL-7) 및 CCL2(Chemokine (C-C motif) ligand 2)로도 알려져 있는 monocyte chemotactic protein-1(MCP-1)의 발현을 억제한다. 이러한 결과는 아이케이 인자 또는 그 단편을 코딩하는 핵산을 적절한 벡터나 유전자 전달체를 통하여 인체에 주입하면 관절염의 진행을 억제할 수 있으며 특히 systemic 면역 반응에도 영향을 미칠 수 있다는 것을 의미한다. 뿐만 아니라, 아이케이 인자가 발현되도록 유도된 전이 마우스(tIK-IL1RaKO)는 자연적으로 관절염이 유발된 마우스(IL1RaKO) 마우스와 비교해서 관절염 병인 T세포의 분화를 억제하며(도 10a, 10b 참조), 대식세포의 활성인자에도 영향을 미친다(도 11 참조).In an exemplary embodiment of the invention, the nucleic acid encoding a fragment of the IK factor is expressed through an appropriate expression system and is a cytokine involved in the inflammatory immune response associated with arthritis Interleukin-1 beta, IL-1β. ), Expression of monocyte chemotactic protein-1 (MCP-1), also known as interleukin-6 (IL-6), interleukin-17 (IL-7), and CCL2 (Chemokine (CC motif) ligand 2). Suppress These results indicate that the injection of nucleic acid encoding an IK factor or fragment thereof into the human body through an appropriate vector or gene transporter can inhibit the progression of arthritis, and in particular can affect the systemic immune response. In addition, the metastatic mouse (tIK-IL1RaKO) induced to express the IK factor suppresses the differentiation of T cells, which are arthritis diseases, as compared to naturally induced arthritis-induced mice (IL1RaKO) mice (see FIGS. 10A and 10B). It also affects the activator of macrophages (see FIG. 11).
또한, 본 발명의 예시적인 실시형태에서 활성 부위를 포함하는 상대적으로 작은 크기의 아이케이 인자 단편, 예를 들어 활성 부위 및 이에 인접한 아미노산 10개 전후의 펩타이드도 관절염의 진행을 억제할 수 있다. 이들 펩타이드는 대식세포에서의 염증성 사이토카인인 종양괴사인자-알파(TNF-α), IL-6, IL-17의 발현을 억제하며(도 12a, 12b 참조 및 도 15b 참조), 관절염에 관여하는 병인 세포인 Th1세포 및 Th17세포의 분화를 또한 억제한다(도 13a 내지 도 13c 참조). 이들 펩타이드는 또한 자연발생 관절염 유발 마우스인 IL1RaKO 마우스에서 관절염을 억제할 뿐만 아니라(도 14a 및 14b 참조), 관절염 유발 마우스에서의 Th17세포의 분화를 억제한다(도 15a 참조).In addition, in exemplary embodiments of the present invention, relatively small size IK factor fragments comprising an active site, such as peptides before and after the active site and 10 amino acids adjacent thereto, can also inhibit the progression of arthritis. These peptides inhibit the expression of tumor necrosis factor-alpha (TNF-α), IL-6, IL-17, inflammatory cytokines in macrophages (see FIGS. 12A, 12B and 15B) and are involved in arthritis It also inhibits the differentiation of Th1 cells and Th17 cells, which are pathogenic cells (see FIGS. 13A-13C). These peptides also inhibit arthritis in IL1RaKO mice, naturally occurring arthritis induced mice (see FIGS. 14A and 14B), as well as inhibit the differentiation of Th17 cells in arthritis induced mice (see FIG. 15A).
예시적인 실시 형태에서 아이케이 인자 또는 그 단편을 코딩하는 핵산은 적절한 발현 시스템 또는 유전자 전달체를 이용하여 주입될 수 있는데, 관절염의 진행을 방지하거나, 유발된 관절염을 치료할 수 있다(도 19a 및 도 19b 참조). In an exemplary embodiment, the nucleic acid encoding the IK factor or fragment thereof can be injected using an appropriate expression system or gene delivery vehicle, which can prevent the progression of arthritis or treat the induced arthritis (FIGS. 19A and 19B). Reference).
전술한 것과 같이, 류마티스성 관절염과 관련해서 Th17세포의 분화에 따라 분비되는 IL-17이 중요한 역할을 수행한다. 본 발명에 따라 아이케이 인자 또는 그 단편 및/또는 이들을 코딩하는 핵산이 류마티스 관절염의 근본 원인으로 주목받고 있는 Th17세포의 분화를 억제한다는 점에서, 기존의 관절염 치료제와 비교해서 훨씬 효율적인 치료가 가능하다. As mentioned above, IL-17, which is secreted according to the differentiation of Th17 cells, plays an important role in rheumatoid arthritis. In accordance with the present invention, the IK factor or fragment thereof and / or nucleic acid encoding them inhibits the differentiation of Th17 cells, which has been noted as the root cause of rheumatoid arthritis, which makes treatment much more efficient than conventional arthritis therapeutics. .
뿐만 아니라, 단일클론항체로 대표되는 대부분의 생물의약품은 인체에 주입하였을 경우에 면역 반응이 유발될 수 있다는 점과, 제조 과정이 복잡하기 때문에 상당히 고가로 판매된다. 이에 반하여, 본 발명에서 그 효능이 확인된 아이케이 인자는 인체에 내재하는 사이토카인이라는 점에서 면역 반응을 유발할 가능성이 거의 없어 안전하며, 상대적으로 저가로 제조가 가능하다. In addition, most biopharmaceuticals represented by monoclonal antibodies are quite expensive because they can cause an immune response when injected into the human body, and because of the complexity of the manufacturing process. On the contrary, in the present invention, the IK factor, whose efficacy has been confirmed, is safe since it is almost impossible to induce an immune response in that it is a cytokine inherent in the human body, and thus it can be manufactured at relatively low cost.
따라서 본 발명은 관절염을 치료하기 위한 아이케이 인자 또는 그 단편에 관한 것이다. 관절염을 치료하기 위한 유효 성분으로서 바람직하게는 전장 아이케이 인자의 일부 아미노산이 절단된 아이케이 인자의 단편이며, 더욱 바람직하게는 전장 아이케이 인자의 N-말단의 아미노산이 절단된 단편이다. The present invention therefore relates to an IK factor or fragment thereof for treating arthritis. As an active ingredient for treating arthritis, preferably, some amino acids of the full-length IK factor are fragments of the cleaved IK factor, more preferably fragments of the N-terminal amino acids of the full-length IK factor are cleaved.
예시적으로, 전장 아이케이 인자는 서열식별번호:2의 아미노산 서열을 가지는 펩타이드일 수 있다. 아이케이 인자의 단편의 예시적인 형태로서 서열식별번호:2의 전장 아이케이 인자 중에서 N-말단을 구성하는 315개의 아미노산이 절단되어 있으며, C-말단의 242개의 아미노산을 가지는 서열식별번호:4의 아미노산 서열을 가지는 절단된 아이케이 인자(truncated IK, tIK)와 같은 펩타이드 또는 서열식별번호:4의 부분 펩타이드일 수 있다. 아이케이 인자의 단편의 일예로서, 전장 아이케이 인자를 구성하는 아미노산 중에서 N-말단이 절단된 펩타이드 단편은 상대적으로 크기가 작기 때문에 제조가 용이할 뿐만 아니라, 생체 내(in vivo) 및/또는 생체 외(ex vivo)에서 전장 아이케이 인자에 비하여 생물학적 기능을 발휘하는데 유리할 수 있다. In exemplary embodiments, the full-length IK factor may be a peptide having an amino acid sequence of SEQ ID NO: 2. As an exemplary form of a fragment of the IK factor, 315 amino acids constituting the N-terminus of the full-length IK factor of SEQ ID NO: 2 are cleaved, and the sequence of SEQ ID NO: 4 having 242 amino acids of the C-terminal It may be a peptide such as truncated IK (tIK) having an amino acid sequence or a partial peptide of SEQ ID NO: 4. As an example of fragments of the IK factor, peptide fragments whose N-terminus are cut out among the amino acids constituting the full-length IK factor are not only easy to manufacture because they are relatively small, but also in vivo and / or in vivo. It may be advantageous to exert biological functions in comparison to full-length IK factor ex vivo.
본 발명의 예시적인 실시예에 따르면, 서열식별번호:2의 펩타이드에서 382번째(서열식별번호:4의 펩타이드에서 67번째) 아미노산인 세린(S382), 489번째(서열식별번호:4의 펩타이드에서 174번째)아미노산인 티로신(Y489) 및/또는 492번째(서열식별번호:4의 펩타이드에서 177번째) 아미노산인 티로신(Y492)이 아이케이 인자 및/또는 그 단편의 생물학적 활성에 관여한다. 특히, 서열식별번호:2의 펩타이드에서 382번째 아미노산인 세린(S382) 및/또는 489번째 아미노산인 티로신(Y489)은 아이케이 인자 및/또는 아이케이 인자의 단편의 활성 부위인 것으로 예측된다. According to an exemplary embodiment of the present invention, the amino acid serine (S382), 489 th (SEQ ID NO: 4 peptide) of the 382th (67th in the peptide of SEQ ID NO: 4), the peptide of SEQ ID NO: 174th) amino acid tyrosine (Y489) and / or 492th (177th in peptide of SEQ ID NO: 4) amino acid tyrosine (Y492) is involved in the biological activity of the IK factor and / or fragments thereof. In particular, the 382th amino acid serine (S382) and / or the 489th amino acid tyrosine (Y489) in the peptide of SEQ ID NO: 2 is predicted to be the active site of the IK factor and / or fragment of the IK factor.
따라서 서열식별번호:4의 아미노산 서열을 가지는 아이케이 인자의 단편 외에도, 이들 활성 부위의 아미노산을 포함하는 상대적으로 짧은 길이의 아이케이 인자의 단편이 사용될 수 있다. 예시적으로, 본 발명에 따라 사용될 수 있는 아이케이 인자의 단편은 서열식별번호:2의 펩타이드에서 382번째 아미노산인 세린(S382), 489번째 아미노산인 티로신(Y489) 및 492번째 아미노산인 티로신(Y489) 중에서 선택되는 적어도 하나의 아미노산 잔기와, 이들 선택된 아미노산 잔기의 인접 아미노산으로 구성되는 10개 이상, 예를 들어 100개 이상 또는 200개 이상의 아미노산으로 구성되는 펩타이드를 포함한다. 예시적인 실시 형태에서 아이케이 인자의 단편은 서열식별번호:2의 펩타이드에서 82번째 아미노산인 세린(S382), 489번째 아미노산인 티로신(Y489) 및 492번째 아미노산인 티로신(Y489) 중에서 선택되는 적어도 하나의 아미노산 잔기와, 이들 선택된 아미노산 잔기의 인접 아미노산으로 구성되는 10개 이상 100개 이하, 바람직하게는 10개 이상 50개 이하, 더욱 바람직하게는 10개 이상 30개 이하의 아미노산으로 구성되는 펩타이드를 포함한다. 이들 선택된 아미노산 잔기들 중 적어도 하나의 아미노산은 인산화와 같이 변형된 아미노산일 수 있다. Thus, in addition to fragments of IK factor having the amino acid sequence of SEQ ID NO: 4, fragments of relatively short length IK factor containing amino acids of these active sites can be used. Illustratively, fragments of the IK factor that can be used in accordance with the present invention include the serine (S382), which is the 382th amino acid, the tyrosine (Y489), which is the 489th amino acid, and the tyrosine (Y489), which is the 492th amino acid, in the peptide of SEQ ID NO: 2. And at least one amino acid residue selected from C) and at least 10, eg, at least 100 or at least 200 amino acids, consisting of contiguous amino acids of these selected amino acid residues. In an exemplary embodiment the fragment of the IK factor is at least one selected from the 82nd amino acid serine (S382), the 489th amino acid tyrosine (Y489) and the 492th amino acid tyrosine (Y489) in the peptide of SEQ ID NO: 2 And a peptide consisting of 10 or more and 100 or less, preferably 10 or more and 50 or less, more preferably 10 or more and 30 or less amino acids, each consisting of the amino acid residues of the selected amino acid residues and adjacent amino acids of these selected amino acid residues. do. At least one amino acid of these selected amino acid residues may be a modified amino acid such as phosphorylation.
예시적으로 본 발명에 따라 사용될 수 있는 아이케이 인자의 단편은 서열식별번호:2의 아미노산 서열을 가지는 펩타이드 또는 그 부분 펩타이드이다. 예시적인 실시 형태에서, 아이케이 인자의 단편은 서열식별번호:2의 펩타이드에서 382번째 아미노산인 세린 및 489번째 아미노산인 티로신 중에서 선택되는 적어도 하나의 아미노산 잔기 및 이들 선택된 아미노산 잔기의 인접 아미노산으로 구성되는 10개 이상, 예를 들어 10개 이상 100개 이하, 바람직하게는 10개 이상 50개 이하, 더욱 바람직하게는 10개 이상 30개 이하의 아미노산을 가지는 펩타이드이다. Exemplary fragments of the IK factor that may be used in accordance with the present invention are peptides or partial peptides thereof having the amino acid sequence of SEQ ID NO: 2. In an exemplary embodiment, the fragment of the IK factor consists of at least one amino acid residue selected from serine, which is the 382th amino acid, and tyrosine, the 489th amino acid, in the peptide of SEQ ID NO: 2 and adjacent amino acids of these selected amino acid residues. It is a peptide which has 10 or more, for example 10 or more and 100 or less, preferably 10 or more and 50 or less, more preferably 10 or more and 30 or less amino acids.
예시적으로, 아이케이 인자의 단편으로 사용될 수 있는 펩타이드는, 서열식별번호:2의 펩타이드에서 382번째 아미노산인 세린(S382), 489번째 아미노산인 티로신(Y489) 및 492번째 아미노산인 티로신(Y492)과 같은 활성 아미노산이 펩타이드의 N-말단이나 C-말단에 위치하지 않을 수 있다. 활성 아미노산인 이들 선택된 아미노산이 펩타이드의 중간에 배열될 수 있도록, 이들 선택된 활성 아미노산의 N-말단과 C-말단으로 각각 2개 이상의 인접 아미노산이 연결되는 펩타이드가 사용될 수 있다. 예시적으로 활성 아미노산의 N-말단과 C-말단으로 각각 2 ~ 8개, 바람직하게는 3 ~ 7개 이상의 인접 아미노산이 연결될 수 있다. 예를 들어 활성 아미노산의 N-말단과 C-말단으로 각각 2 ~ 100개, 예컨대 3 ~ 70개, 바람직하게는 3 ~ 30개, 더욱 바람직하게는 3 ~ 20개의 인접 아미노산이 연결될 수 있다. For example, peptides that can be used as fragments of the IK factor include the serine (S382), the 382th amino acid, the tyrosine (Y489), the 489th amino acid, and the tyrosine (Y492), the 492th amino acid, in the peptide of SEQ ID NO: 2. An active amino acid such as may not be located at the N-terminus or C-terminus of the peptide. Peptides may be used in which two or more adjacent amino acids are linked to each of the N-terminus and C-terminus of these selected active amino acids so that these selected amino acids that are active amino acids can be arranged in the middle of the peptide. For example, 2 to 8, preferably 3 to 7 or more adjacent amino acids may be linked to the N-terminus and C-terminus of the active amino acid, respectively. For example, 2 to 100, such as 3 to 70, preferably 3 to 30, more preferably 3 to 20 contiguous amino acids may be linked to the N-terminus and C-terminus of the active amino acid, respectively.
하나의 예시적인 실시형태에서, 아이케이 인자의 단편은 서열식별번호:2의 펩타이드에서 377번째 아미노산인 글루탐산 - 391번째 아미노산인 아스파르트산으로 구성되는 서열식별번호:47의 펩타이드 및/또는 484번째의 아미노산인 아스파르트산 - 496번째의 아미노산인 리신으로 구성되는 서열식별번호:49의 펩타이드를 또한 포함한다. In one exemplary embodiment, the fragment of the IK factor is a peptide of SEQ ID NO: 47 consisting of 377 amino acid glutamic acid-391 amino acid aspartic acid in the peptide of SEQ ID NO: 2 and / or the 484th Also included is a peptide of SEQ ID NO: 49 consisting of the amino acid aspartic acid—the 496th amino acid lysine.
예시적인 실시예에 따라 서열식별번호:2의 전장 아이케이 인자가 557개의 아미노산으로 구성되며, 아이케이 인자의 N-말단이 절단되어 있는 서열식별번호:4의 아이케이 인자의 단편이 242개의 아미노산으로 구성된다. 이 점을 고려해 볼 때, 본 발명에 따른 아이케이 인자의 단편은 서열식별번호:2의 아미노산 중에서 최대 약 300~500개 정도의 아미노산으로 구성될 수 있다. 하지만, 본 발명에 따라 사용될 수 있는 아이케이 인자의 단편이 이에 한정되지는 않는다. According to an exemplary embodiment, the full length IK factor of SEQ ID NO: 2 consists of 557 amino acids, and the fragment of the IK factor of SEQ ID NO: 4, wherein the N-terminus of the IK factor is cleaved, is 242 amino acids. It consists of. In consideration of this point, the fragment of IK factor according to the present invention may be composed of up to about 300 to 500 amino acids of the amino acid of SEQ ID NO: 2. However, fragments of IK factor that can be used in accordance with the present invention are not limited thereto.
또한, 예를 들어, 아이케이 인자 및/또는 아이케이 인자의 단편 중에서 S382, Y489 및 Y492와 같은 활성 아미노산을 제외하고, 나머지 아미노산들은 예를 들어 보존적 치환에 의하여 다른 아미노산으로 치환될 수 있으며, 필요한 경우에 일부 아미노산이 결실, 첨가 및/또는 삽입될 수 있다. Also, for example, except for active amino acids such as S382, Y489 and Y492 in the IK factor and / or fragments of IK factor, the remaining amino acids may be substituted with other amino acids, for example by conservative substitutions, If necessary some amino acids may be deleted, added and / or inserted.
아이케이 인자 또는 그 단편인 펩타이드는 재조합 방법(recombinant means) 또는 화학적 합성을 통해 제조함으로써 분리될 수 있다. 예시적으로 본 명세서에 언급된 핵산 서열에 의해 발현되는(expressed) 펩타이드는 알려져 있는 많은 발현 벡터들 중 어느 것을 이용하든지 간에 공지의 방법으로 손쉽게 제조될 수 있다. 발현은 펩타이드를 코딩하는 DNA 서열을 포함하는 발현벡터로 형질전환된 적절한 숙주 세포(host cell) 내에서 실현될 수 있다. 적절한 숙주 세포에는 원핵세포들(prokaryotes), 효모(yeast) 및 진핵세포들(eukaryotes)이 포함된다. 대장균, 효모 또는 포유동물 세포주(Cos 또는 CHO 등의)를 숙주세포로 이용하는 것이 바람직하다. 단백질의 정제를 위해서는 먼저 수용성의 숙주/벡터 시스템에서 얻은, 배양액으로 분비된 재조합 단백질을 포함하는 상등액을 시판되는 필터를 이용하여 농축시킨다. 다음 단계로 상기에서 얻은 농축액을 친화 매트릭스(affinity matrix) 또는 이온교환수지(ion exchange resin) 등의 적절한 정제 매트릭스(purification matrix)를 이용하여 정제한다. 마지막으로 한 단계 또는 여러 단계의 역상(reverse phase) HPLC 를 수행함으로써 순수한 단백질을 얻을 수 있다.Peptides that are IK factors or fragments thereof can be isolated by preparation through recombinant means or chemical synthesis. By way of example, peptides expressed by the nucleic acid sequences mentioned herein can be readily prepared by known methods, using any of a number of known expression vectors. Expression can be realized in a suitable host cell transformed with an expression vector comprising a DNA sequence encoding the peptide. Suitable host cells include prokaryotes, yeasts and eukaryotes. E. coli, yeast or mammalian cell lines (such as Cos or CHO) are preferably used as host cells. For purification of the protein, the supernatant containing recombinant protein secreted into the culture, first obtained in a water-soluble host / vector system, is concentrated using a commercially available filter. In the next step, the concentrate obtained above is purified using an appropriate purification matrix such as an affinity matrix or an ion exchange resin. Finally, pure protein can be obtained by performing one or several reverse phase HPLC.
100개 이하, 일반적으로는 50개 이하의 아미노산으로 구성된 단편이나 변이체들은 합성에 의해 제조될 수 있다. 예를 들어, 이러한 폴리펩타이드들은 상용화되어 있는 고상 기법(solid-phase techniques), 즉 성장하는 아미노산 사슬에 순차적으로 아미노산을 부가시키는 메리필드 고상법(Merrifield solid-phase synthesis method)으로 합성할 수 있다(Merrifield, 1963, J. Am. Chem. Soc. 85:2146-2149). 폴리펩타이드의 자동 합성을 위한 장비는 공급사로부터 구입할 수 있으며, 공급자의 매뉴얼에 따라 조작이 가능하다. Fragments or variants consisting of up to 100, typically up to 50 amino acids can be prepared synthetically. For example, such polypeptides can be synthesized by commercially available solid-phase techniques, the Merrifield solid-phase synthesis method, which sequentially adds amino acids to growing amino acid chains ( Merrifield, 1963, J. Am. Chem. Soc. 85: 2146-2149. Equipment for automatic synthesis of polypeptides can be purchased from the supplier and can be manipulated according to the supplier's manual.
예를 들어 미생물에서 발현되는 본 명세서에 기재된 펩타이드는 숙주 세포의 주변 세포질로 분비되고 그로부터 회수될 수 있다. 전형적으로, 단백질 회수는 일반적으로 삼투압 충격, 초음파처리 또는 용해와 같은 수단에 의해 미생물을 분쇄하는 것을 포함한다. 세포가 파괴되면, 세포 잔해 또는 전세포를 원심분리 또는 여과에 의해 제거할 수 있다. 단백질은 예를 들어 친화성 수지 크로마토그래피에 의해 추가로 정제할 수 있다. 대안적으로, 단백질을 배양 배지로 옮기고, 그로부터 분리할 수 있다. 세포를 배양물로부터 제거하고, 배양 상등액을 여과하고 농축하여 생산된 단백질을 추가로 정제할 수 있다. 발현된 폴리펩티드는 통상적으로 공지된 방법, 예컨대 면역친화성 또는 이온-교환 칼럼 상의 분별 증류; 에탄올 침전; 역상 HPLC; 실리카 또는 양이온 교환수지, 예컨대 DEAE 상의 크로마토그래피; 크로마토포커싱; SDS-PAGE; 황산암모늄 침전; 예를 들어 세파덱스 G-75를 사용하는 겔 여과; 소수성 친화도 수지, 매트릭스 상에 고정화된 적합한 항원을 사용하는 리간드 친화도 및 웨스턴 블롯 검정을 이용하여 추가로 분리 및 확인될 수 있다. For example, peptides described herein expressed in a microorganism can be secreted into and recovered from the periplasm of the host cell. Typically, protein recovery generally involves grinding the microorganism by means such as osmotic shock, sonication or lysis. Once the cells are destroyed, cell debris or whole cells can be removed by centrifugation or filtration. Proteins can be further purified, for example, by affinity resin chromatography. Alternatively, the protein can be transferred to and separated from the culture medium. The cells can be removed from the culture and the culture supernatant filtered and concentrated to further purify the protein produced. Expressed polypeptides are commonly known methods, such as fractional distillation on immunoaffinity or ion-exchange columns; Ethanol precipitation; Reverse phase HPLC; Chromatography on silica or cation exchange resins such as DEAE; Chromatofocusing; SDS-PAGE; Ammonium sulfate precipitation; Gel filtration using, for example, Sephadex G-75; Hydrophobic affinity resins, ligand affinity using suitable antigens immobilized on a matrix, and Western blot assays can be further isolated and identified.
아울러, 생산된 펩타이드는 추가의 검정 및 용도를 위해 실질적으로 균질한 제제를 수득하기 위해 정제될 수 있다. 당업계에 공지된 표준 단백질 정제 방법이 이용될 수 있다. 하기 절차는 적합한 정제 절차의 예이다: 면역친화성 또는 이온-교환 칼럼 상의 분별 증류, 에탄올 침전, 역상 HPLC, 실리카 또는 양이온-교환 수지, 예컨대 DEAE 상의 크로마토그래피, 크로마토포커싱, SDS-PAGE, 황산암모늄 침전, 및 예를 들어 세파덱스 G-75를 사용하는 겔 여과.In addition, the peptides produced can be purified to obtain a substantially homogeneous formulation for further assays and uses. Standard protein purification methods known in the art can be used. The following procedures are examples of suitable purification procedures: fractional distillation on immunoaffinity or ion-exchange columns, ethanol precipitation, reverse phase HPLC, silica or cation-exchange resins, such as chromatography on DEAE, chromatographic focusing, SDS-PAGE, ammonium sulfate Precipitation, and gel filtration using, for example, Sephadex G-75.
또한, 본 발명은 관절염을 치료하기 위한 아이케이 인자 또는 그 단편을 코딩하는 핵산 또는 폴리뉴클레오티드에 관한 것이다. 펩타이드에서와 유사하게, 바람직하게는 전장 아이케이를 코딩하는 핵산의 일부 뉴클레오티드가 절단된 아이케이 핵산 단편이 바람직하고, 특히 바람직하게는 5' 말단의 뉴클레오티드가 절단된 아이케이 핵산 단편이다. The invention also relates to a nucleic acid or polynucleotide encoding an IK factor or fragment thereof for treating arthritis. Similar to peptides, preferably the IK nucleic acid fragments from which some nucleotides of the nucleic acid encoding the full length IK are cleaved are preferred, and particularly preferably the IK nucleic acid fragments from which the nucleotides of the 5 'end are cleaved.
예시적으로 전장 아이케이 인자를 코딩하는 핵산은 서열식별번호:1의 뉴클레오티드 서열을 가지는 핵산일 수 있다. 예시적으로 아이케이 인자의 부분 단편을 코딩하는 핵산은, 서열식별번호:1의 핵산 중에서 5' 말단을 구성하는 945개의 뉴클레오티드가 절단된 서열식별번호:3의 뉴클레오티드 서열을 가지는 절단된 아이케이 인자를 코딩하는 핵산(tIK 핵산 단편) 또는 서열식별번호:3의 일부 뉴클레오티드로 구성되는 핵산일 수 있다.For example, the nucleic acid encoding the full-length IK factor may be a nucleic acid having a nucleotide sequence of SEQ ID NO: 1. For example, a nucleic acid encoding a fragment of an IK factor is a truncated IK factor having a nucleotide sequence of SEQ ID NO: 3 wherein 945 nucleotides constituting the 5 'end of the nucleic acid of SEQ ID NO: 1 are cleaved. It may be a nucleic acid (tIK nucleic acid fragment) encoding a portion or a nucleic acid consisting of some nucleotides of SEQ ID NO: 3.
서열식별번호:2의 펩타이드에서 382번째 아미노산인 세린(S382), 489번째 아미노산인 티로신(Y489) 및 492번째 아미노산인 티로신(Y492)은 각각 서열식별번호:1의 1144-1146(서열식별번호:3의 199-201)번째의 뉴클레오티드 잔기, 서열식별번호:1의 1465-1467(서열식별번호:3의 520-522)번째의 뉴클레오티드 잔기, 서열식별번호:1의 1474-1476(서열식별번호:3의 529-531)번째의 뉴클레오티드 잔기에 의해 발현될 수 있다. In the peptide of SEQ ID NO: 2, the serine (S382), 389 amino acid tyrosine (Y489) and the 492 amino acid tyrosine (Y492), respectively, were 1144-1146 (SEQ ID NO: 1). 199-201) nucleotide residue of 3, 1465-1467 (520-522 of SEQ ID NO: 3) of SEQ ID NO: 1, 1474-1476 of SEQ ID NO: 1 (SEQ ID NO: 529-531) nucleotide residues of 3 can be expressed.
따라서 서열식별번호:3의 뉴클레오티드 서열을 가지는 아이케이 단편 핵산 이외에도, 상대적으로 짧은 길이의 뉴클레오티드로 구성되는 아이케이 단편 핵산이 또한 사용될 수 있다. 예시적으로 본 발명에 따른 아이케이 단편 핵산은, 서열식별번호:1의 1144-1146(서열식별번호:3의 199-201)번째의 뉴클레오티드 잔기, 서열식별번호:1의 1465-1467(서열식별번호:3의 520-522)번째의 뉴클레오티드 잔기, 서열식별번호:1의 1474-1476(서열식별번호:3의 529-531)번째의 뉴클레오티드 잔기와, 이들 선택된 뉴클레오티드 잔기에 인접한 30개 이상, 예를 들어 300개 이상 또는 600개 이상의 뉴클레오티드로 구성될 수 있다. 예시적인 실시 형태에서 아이케이 인자의 단편을 코딩하는 핵산은, 서열식별번호:1의 1144-1146번째의 뉴클레오티드 잔기, 서열식별번호:1의 1465-1467번째의 뉴클레오티드 잔기, 서열식별번호:1의 1474-1476번째의 뉴클레오티드 잔기와, 이들 선택된 뉴클레오티드 잔기의 인접 뉴클레오티드로 구성되는 30개 이상 300개 이하, 바람직하게는 30개 이상 150개 이하, 더욱 바람직하게는 30개 이상 90개 이하의 뉴클레오티드로 구성되는 핵산을 포함한다. Thus, in addition to IK fragment nucleic acids having the nucleotide sequence of SEQ ID NO: 3, IK fragment nucleic acids consisting of nucleotides of relatively short length can also be used. For example, the IK fragment nucleic acid according to the present invention is a nucleotide residue of 1144-1146 (SEQ ID NO: 199-201) of SEQ ID NO: 1, 1465-1467 (SEQ ID NO: 1) 520-522) nucleotide residues of No. 3, 1474-1476 (529-531 of SEQ ID NO: 3) of SEQ ID NO: 1 and at least 30 adjacent to these selected nucleotide residues, eg For example, it may consist of more than 300 or more than 600 nucleotides. In an exemplary embodiment, the nucleic acid encoding a fragment of the IK factor comprises a 1144-1146th nucleotide residue of SEQ ID NO: 1, a 1465-1467th nucleotide residue of SEQ ID NO: 1, Nucleotide residues of 1474-1476 and 30 or more and 300 or less, preferably 30 or more and 150 or less, more preferably 30 or more and 90 or less nucleotides each consisting of adjacent nucleotides of these selected nucleotide residues Nucleic acids to be included.
예시적으로, 아이케이 단편 핵산은 서열식별번호:1의 뉴클레오티드 서열을 가지는 핵산 또는 그 부분 단편이다. 예시적인 실시형태에서 아이케이 인자의 부분 단편을 코딩하는 핵산은 서열식별번호:1의 뉴클레오티드 서열의 1144-1146번의 뉴클레오티드 잔기 및 1465-1467번째 뉴클레오티드 잔기 중에서 선택되는 적어도 하나의 뉴클레오티드 잔기와, 이들 선택된 뉴클레오티드 세트의 인접 뉴클레오티드로 구성되는 30개 이상, 예를 들어 30개 이상 300개 이하, 바람직하게는 30개 이상 150개 이하, 더욱 바람직하게는 30개 이상 90개 이하의 뉴클레오티드를 가지는 핵산이다. Illustratively, the IK fragment nucleic acid is a nucleic acid having a nucleotide sequence of SEQ ID NO: 1 or a partial fragment thereof. In an exemplary embodiment the nucleic acid encoding the partial fragment of the IK factor is selected from at least one nucleotide residue selected from nucleotide residues 1144-1146 and 1465-1467 nucleotide residues of the nucleotide sequence of SEQ ID NO: 1; It is a nucleic acid having 30 or more, for example, 30 or more and 300 or less, preferably 30 or more and 150 or less, more preferably 30 or more and 90 or less nucleotides composed of contiguous nucleotides of a nucleotide set.
선택된 뉴클레오티드 잔기가 폴리뉴클레오티드의 중간에 배열될 수 있도록, 이들 선택된 뉴클레오티드 세트의 5' 말단과 3' 말단으로 각각 6개 이상의 인접 뉴클레오티드가 연결될 수 있다. 예시적으로 선택된 뉴클레오티드 잔기의 5' 말단과 3' 말단으로 각각 6 ~ 24개, 바람직하게는 9 ~ 21개 이상의 인접 뉴클레오티드가 연결될 수 있다. 예를 들어 선택된 뉴클레오티드 잔기의 5' 말단과 3' 말단으로 각각 6-300개, 예컨대 9 ~ 210개, 바람직하게는 9 ~ 90개, 더욱 바람직하게는 9 ~ 60개의 인접 뉴클레오티드가 연결될 수 있다. Six or more contiguous nucleotides may each be linked to the 5 'and 3' ends of these selected nucleotide sets so that the selected nucleotide residues can be arranged in the middle of the polynucleotide. For example, 6 to 24, preferably 9 to 21 or more contiguous nucleotides may be linked to the 5 'end and 3' end of the selected nucleotide residue. For example, 6-300, such as 9-210, preferably 9-90, more preferably 9-60 contiguous nucleotides can be linked to the 5 'end and 3' end of the selected nucleotide residue, respectively.
하나의 예시적인 실시형태에서, 아이케이 인자의 단편을 코딩하는 핵산은 서열식별번호:46의 뉴클레오티드 서열을 가지는 핵산 및/또는 서열식별번호:48의 뉴클레오티드 서열을 가지는 핵산일 수 있다. 예시적으로, 아이케이 인자의 단편을 코딩하는 핵산은 서열식별번호:1의 뉴클레오티드 중에서 최대 약 900 ~ 1500개 정도의 뉴클레오티드로 구성될 수 있지만, 본 발명이 이에 한정되지 않는다. 서열식별번호:2의 아미노산 중에서 382번째 아미노산인 세린(S382), 489번째 아미노산인 티로신(Y489) 및/또는 492번째 아미노산인 티로신(Y492)을 각각 코딩하는 서열식별번호:1의 1144-1146번째의 뉴클레오티드 세트, 1465-1467번째의 뉴클레오티드 세트 및/또는 1474-1476번째의 뉴클레오티드는 각각 세린과 티로신을 코딩하는 다른 뉴클레오티드 서열을 가질 수 있다. In one exemplary embodiment, the nucleic acid encoding a fragment of the IK factor may be a nucleic acid having the nucleotide sequence of SEQ ID NO: 46 and / or a nucleic acid having the nucleotide sequence of SEQ ID NO: 48. For example, the nucleic acid encoding a fragment of the IK factor may be composed of up to about 900 to 1500 nucleotides among the nucleotides of SEQ ID NO: 1, but the present invention is not limited thereto. Of the amino acids of SEQ ID NO: 2, 1144-1146 of SEQ ID NO: 1 coding for serine (S382), 489 amino acid tyrosine (Y489), and / or tyrosine (Y492) 492 amino acid, respectively The set of nucleotides, the set of 1465-1467 nucleotides and / or the set of 1474-1476 nucleotides may have different nucleotide sequences encoding serine and tyrosine, respectively.
본 발명에 따라 아이케이 인자 또는 그 단편을 코딩하는 핵산은 적절한 벡터 안에 포함될 수 있다. 벡터는 또한 본 발명의 폴리뉴클레오티드와 연결된 발현조절서열(expression control sequence)을 포함한다. 본 발명의 예시적인 실시형태에서, 벡터는 목적하는 다른 분자들을 코딩하는 하나 이상의 폴리뉴클레오티드를 포함하기도 한다. 본 발명의 폴리뉴클레오티드는 다른 폴리뉴클레오티드와 결합하여 융합 단백질을 코딩하기도 한다. In accordance with the present invention, nucleic acids encoding an IK factor or fragment thereof can be included in a suitable vector. The vector also includes an expression control sequence linked to the polynucleotide of the present invention. In an exemplary embodiment of the invention, the vector may also comprise one or more polynucleotides encoding other molecules of interest. Polynucleotides of the present invention may also bind to other polynucleotides to encode fusion proteins.
예시적으로, 본 발명의 폴리뉴클레오티드들은 포유동물 세포내로 들어가서 발현되도록 조성되어 있다. 이러한 조성은 치료 목적에 사용하는 데에 특히 유용하다. 폴리뉴클레오티드를 숙주세포 내에서 발현시키는 데에는 많은 방법이 있으며 적절한 어느 방법도 사용 가능하다. 예를 들어 하나의 폴리뉴클레오티드는 아데노바이러스(adenovirus), 아데노-연관 바이러스(adeno-associated virus), 레트로 바이러스(retrovirus), 백시니아(vaccinia), 렌티바이러스(lentivirus) 바큘로바이러스(baculovirus) 또는 다른 폭스 바이러스(e.q., avian pox virus) 등의 바이러스 벡터에 삽입이 가능하다. DNA를 이러한 벡터에 삽입시키는 기술에 관해서는 이미 잘 알려져 있다. 레트로 바이러스 벡터에는 형질도입된 세포들의 확인 또는 선택을 쉽게 해 주는 선택 마커(selectable marker)에 대한 유전자 및/또는 특정한 타겟 세포에 대한 수용체 역할을 하는 리간드(ligand)를 코딩하는 유전자와 같은 타겟팅 부위(targeting moiety)를 부가적으로 삽입시킬 수 있다. 타겟팅은 또한 항체를 이용한 공지의 방법에 의해서도 이루어질 수 있다. Illustratively, the polynucleotides of the invention are formulated to enter into and express in mammalian cells. Such compositions are particularly useful for use for therapeutic purposes. There are many ways to express polynucleotides in host cells, and any suitable method can be used. For example, one polynucleotide is an adenovirus, an adeno-associated virus, a retrovirus, a vaccinia, a lentivirus baculovirus or another. It can be inserted into viral vectors such as pox virus (eq, avian pox virus). Techniques for inserting DNA into such vectors are well known. Retroviral vectors include targeting sites such as genes for selectable markers that facilitate the identification or selection of transduced cells and / or genes that encode ligands that act as receptors for specific target cells. can additionally insert a targeting moiety. Targeting can also be accomplished by known methods using antibodies.
입수 가능하고 당업계에 공지된 다수의 벡터를 본 발명의 목적에 사용할 수 있다. 적절한 벡터의 선택은 주로 벡터에 삽입되는 핵산의 크기 및 벡터로 형질전환되는 특정한 숙주 세포에 따라 달라질 것이다. 각각의 벡터는 그의 기능(이종 폴리뉴클레오티드의 증폭 또는 발현, 또는 둘 다) 및 벡터가 존재하는 특정한 숙주 세포와의 상용성에 따라 다양한 성분을 함유한다. 벡터 성분은 일반적으로 복제 기점(특히 벡터가 원핵세포에 삽입되는 경우), 선택 마커 유전자, 프로모터, 리보솜 결합 부위 (RBS), 신호 서열, 이종 핵산 삽입물 및 전사 종결 서열을 포함하나 이에 제한되지는 않는다.Many vectors available and known in the art can be used for the purposes of the present invention. Selection of the appropriate vector will depend primarily on the size of the nucleic acid inserted into the vector and the particular host cell transformed with the vector. Each vector contains various components depending on its function (amplification or expression of heterologous polynucleotides, or both) and compatibility with the particular host cell in which the vector is present. Vector components generally include, but are not limited to, origin of replication (especially when the vector is inserted into prokaryotic cells), selection marker genes, promoters, ribosomal binding sites (RBS), signal sequences, heterologous nucleic acid inserts, and transcription termination sequences. .
예를 들어, 본 발명의 재조합 벡터는 단백질의 발현에 영향을 미칠 수 있는 발현 조절 서열, 예를 들어, 개시코돈, 종결코돈, 폴리아데닐화 시그널, 인핸서, 막 표적화 또는 분비를 위한 신호서열 등을 포함할 수 있다. 폴리아데닐화 시그널은 전사체의 안정성을 증가시키거나 세포질 수송을 용이하게 한다. 인핸서 서열은 프로모터에서 다양한 부위에 위치하여 인핸서 서열이 없을 때의 프로모터에 의한 전사 활성과 비교하여, 전사 활성을 증가시키는 핵산 염기서열이다. 신호서열로서, 숙주가 에스케리키아(Escherichia) 속 균인 경우에는 PhoA 신호서열, OmpA 신호서열 등이, 숙주가 바실러스(Bacillus)속 균인 경우에는 α-아밀라아제 신호서열, 서브틸리신 신호서열 등이, 숙주가 효모인 경우에는 MF-α 신호서열, SUC2 신호서열 등이, 숙주가 동물세포인 경우에는 인슐린 신호서열, a-인터페론 신호서열, 항체 분자 신호서열 등을 이용할 수 있으나, 본 발명이 이에 제한되지 않는다.For example, recombinant vectors of the present invention may be used to express expression control sequences that may affect the expression of proteins, such as initiation codons, termination codons, polyadenylation signals, enhancers, signal sequences for membrane targeting or secretion, and the like. It may include. Polyadenylation signals increase the stability of transcripts or facilitate cellular transport. Enhancer sequences are nucleic acid sequences that are located at various sites in the promoter and increase transcriptional activity as compared to the transcriptional activity by the promoter in the absence of the enhancer sequence. As a signal sequence, when the host is Escherichia (Escherichia) in gyunin include PhoA signal sequence, OmpA signal sequence, etc., the host is Bacillus (Bacillus) when in gyunin include α- amylase signal sequence, subtilisin signal sequence, etc. Shin, If the host is a yeast, MF-α signal sequence, SUC2 signal sequence, etc., if the host is an animal cell, insulin signal sequence, a-interferon signal sequence, antibody molecular signal sequence, etc. may be used, but the present invention is limited thereto. It doesn't work.
벡터의 한 유형은 추가의 DNA 절편이 그 내부에 라이게이션될 수 있는 원형 이중 가닥 DNA 루프를 지칭하는 "플라스미드"이다. 또 다른 유형의 벡터는 파지 벡터이다. 또 다른 유형의 벡터는 추가의 DNA 절편이 바이러스 게놈 내로 라이게이션될 수 있는 바이러스 벡터이다. 특정 벡터는 그것이 도입된 숙주 세포 내에서 자율 복제가 가능하다(예를 들어, 박테리아 복제 기점을 갖는 박테리아 벡터 및 에피솜 포유동물 벡터). 다른 벡터(예를 들어, 비-에피솜 포유동물 벡터)는 숙주 세포 내로 도입시에 숙주 세포의 게놈 내로 통합될 수 있고, 이에 의해 숙주 게놈과 함께 복제된다. 또한, 특정 벡터는 벡터가 작동가능하게 연결된 유전자의 발현을 지시할 수 있다. 이러한 벡터가 본 명세서에서 "재조합 발현 벡터"(또는 간단히, "재조합 벡터")로서 지칭된다. 일반적으로, 재조합 DNA 기술에 유용한 발현 벡터는 종종 플라스미드의 형태로 존재한다. One type of vector is a "plasmid" which refers to a circular double stranded DNA loop into which additional DNA segments can be ligated therein. Another type of vector is a phage vector. Another type of vector is a viral vector in which additional DNA segments can be ligated into the viral genome. Certain vectors are capable of autonomous replication in the host cell into which they are introduced (eg, bacterial vectors and episomal mammalian vectors with bacterial origins of replication). Other vectors (eg, non-episomal mammalian vectors) can be integrated into the genome of the host cell upon introduction into the host cell and thereby replicate with the host genome. In addition, certain vectors may direct expression of the gene to which the vector is operably linked. Such vectors are referred to herein as "recombinant expression vectors" (or simply, "recombinant vectors"). In general, expression vectors of utility in recombinant DNA techniques are often in the form of plasmids.
본 발명의 벡터 시스템은 당업계에 공지된 다양한 방법을 통해 구축될 수 있으며, 이에 대한 구체적인 방법은 Sambrook et al., Molecular Cloning, A Laboratory Manual, Cold Spring Harbor Laboratory Press(2001)에 개시되어 있으며, 이 문헌은 본 명세서에 참조로서 삽입된다. 예를 들어, 본 발명에 이용될 수 있는 벡터는 당업계에서 종종 사용되는 플라스미드 (예: pSC101, ColE1, pBR322, pUC8/9, pHC79, pUC19, pET 등), 파지 (예: λgt4λB, λ-Charon, λΔz1, λGEM.TM.-11 및 M13 등) 또는 바이러스 (예: SV40 등)를 조작하여 제작될 수 있다. The vector system of the present invention may be constructed through various methods known in the art, and specific methods thereof are disclosed in Sambrook et al., Molecular Cloning, A Laboratory Manual, Cold Spring Harbor Laboratory Press (2001), This document is incorporated herein by reference. For example, vectors that can be used in the present invention include plasmids often used in the art (eg pSC101, ColE1, pBR322, pUC8 / 9, pHC79, pUC19, pET, etc.), phage (eg λgt4λB, λ-Charon , λΔz1, λGEM.TM.-11 and M13, etc.) or viruses (eg SV40, etc.).
본 발명의 구체적인 실시형태에서, 핵산은 바이러스 발현 시스템(백시니아 또는 다른 폭스 바이러스, 레트로 바이러스, 렌티바이러스, 바큘로바이러스, 아데노바이러스 또는 아데노-연관바이러스)을 이용하여 숙주 세포로 삽입된다. 예시적으로, 바이러스 벡터에는 HIV, SIV, 설치류 레트로 바이러스(murine retroviruses), 기본 백혈병 바이러스(gibbon ape leukemia virus), AAVs(adeno-associate viruses) 및 아데노바이러스(adenoviruses) 등에서 유래된 레트로 바이러스 벡터가 포함되나 여기에 한정되는 것은 아니다(Miller et al., 1990, Mol. Cell Biol. 10:4239; J. Kolberg 1992, NIH Res. 4:43; Cornetta et al., 1991, Hum. Gene Ther. 2:215). 설치류 백혈병 바이러스(murine leukemia virus, MuLV), 기본 백혈병 바이러스(gibbon ape leukemia virus, GaLV), 에코트로픽 레트로바이러스(ecotropic retroviruses), SIV(simian immunodeficiency virus), HIV(human immunodeficiency virus) 등에서 유래된 레트로 바이러스 벡터들이 널리 사용되고 있다(Buchscher et al., 1992, J. Virol, 66(5):2731-2739; Johann et al., 1992, J. Virol. 66(5):1635-1640; Sommerfelt et al., 1990, Virol. 176:58-59; Wilson et al., 1989, J. Virol. 63:2374-2378; Miller et al., 1991, J. Virol. 65:2220-2224; Rosenberg and Fauci 1993 in Fundermental Immunology, Third Edition, W. E. Paul(ed.) Raven Press, Ltd., New York and the references therein; Miller et al., 1990, Mol. Cell. Biol. 10:4239; R. Kolberg 1992, J. NIH Res. 4:43;Cornetta et al., 1991, Hum. Gene Ther.2:215).In a specific embodiment of the invention, the nucleic acid is inserted into a host cell using a virus expression system (vaccinia or other pox virus, retrovirus, lentivirus, baculovirus, adenovirus or adeno-associated virus). Exemplary viral vectors include retroviral vectors derived from HIV, SIV, murine retroviruses, gibbon ape leukemia virus, adeno-associate viruses, and adenoviruses, and the like. But not limited thereto (Miller et al., 1990, Mol. Cell Biol. 10: 4239; J. Kolberg 1992, NIH Res. 4:43; Cornetta et al., 1991, Hum. Gene Ther. 2: 215). Retroviruses derived from rodent leukemia virus (MulVe), basic leukemia virus (GIBBON ape leukemia virus, GaLV), ecotropic retroviruses, simian immunodeficiency virus (SIV), human immunodeficiency virus (HIV), etc. Vectors are widely used (Buchscher et al., 1992, J. Virol, 66 (5): 2731-2739; Johann et al., 1992, J. Virol. 66 (5): 1635-1640; Sommerfelt et al. , 1990, Virol. 176: 58-59; Wilson et al., 1989, J. Virol. 63: 2374-2378; Miller et al., 1991, J. Virol. 65: 2220-2224; Rosenberg and Fauci 1993 in Fundermental Immunology, Third Edition, WE Paul (ed.) Raven Press, Ltd., New York and the references therein; Miller et al., 1990, Mol. Cell.Biol. 10: 4239; R. Kolberg 1992, J. NIH Res. 4:43; Cornetta et al., 1991, Hum. Gene Ther. 2: 215).
구성적 또는 유도성 프로모터는 당업자에 의해 확인될 수 있는 특정한 상황의 필요에 따라 본 발명에 사용될 수 있다. 다양한 가능한 숙주 세포에 의해 인식되는 다수의 프로모터가 널리 공지되어 있다. 선택된 프로모터는 제한 효소 소화를 통해 공급원 DNA로부터 프로모터를 제거하고 단리된 프로모터 서열을 선택 벡터 내로 삽입함으로써 본원에 기재된 폴리펩티드를 코딩하는 시스트론 DNA에 작동가능하게 연결될 수 있다. 천연 프로모터 서열 및 다수의 이종 프로모터를 둘 다 사용하여 표적 유전자의 증폭 및/또는 발현을 지시할 수 있다. 그러나, 이종 프로모터는 일반적으로 천연 표적 폴리펩티드 프로모터와 비교하여 발현된 표적 유전자의 보다 큰 전사 및 보다 높은 수율을 허용하게 하기 때문에 바람직하다.Constitutive or inducible promoters can be used in the present invention depending on the needs of a particular situation that can be identified by one skilled in the art. Many promoters that are recognized by a variety of possible host cells are well known. The selected promoter may be operably linked to the cistron DNA encoding the polypeptides described herein by removing the promoter from the source DNA via restriction enzyme digestion and inserting the isolated promoter sequence into the selection vector. Both native promoter sequences and multiple heterologous promoters can be used to direct amplification and / or expression of a target gene. However, heterologous promoters are generally preferred because they allow greater transcription and higher yield of expressed target genes as compared to native target polypeptide promoters.
한편, 본 발명의 벡터가 발현 벡터이고, 진핵 세포를 숙주로 하는 경우에는, 포유동물 세포의 게놈으로부터 유래된 프로모터(예: 메탈로티오닌 프로모터) 또는 포유동물 바이러스로부터 유래된 프로모터 (예: 아데노바이러스 후기 프로모터, 백시니아 바이러스 7.5K 프로모터, SV40 프로모터, 사이토메갈로바이러스 프로모터 및 HSV의 tk 프로모터)가 이용될 수 있으며, 전사 종결 서열로서 폴리아데닐화 서열을 일반적으로 갖는다. On the other hand, when the vector of the present invention is an expression vector and the eukaryotic cell is a host, a promoter derived from the mammalian cell genome (for example, metallothionine promoter) or a promoter derived from a mammalian virus (for example, adeno) Late viral promoter, vaccinia virus 7.5K promoter, SV40 promoter, cytomegalovirus promoter and tk promoter of HSV) can be used and generally have a polyadenylation sequence as a transcription termination sequence.
또한, 본 발명의 재조합 벡터가 복제 가능한 발현 벡터인 경우, 복제가 개시되는 특정 핵산 서열인 복제원점(replication origin)을 포함할 수 있다. 또한, 재조합 벡터는 선택마커(selection marker)를 포함할 수 있다. 선택마커는 벡터로 형질전환된 세포를 선별하기 위한 것으로, 약물 내성, 영양 요구성, 세포 독성제에 대한 내성 또는 표면 단백질의 발현과 같은 선택가능 표현형을 부여하는 마커들이 사용될 수 있다. 본 발명의 벡터는 선택표지로서, 해당 기술분야에서 통상적으로 이용되는 항생제 내성 유전자를 포함하며, 예를 들어 암피실린, 겐타마이신, 카베니실린, 클로람페니콜, 스트렙토마이신, 카나마이신, 게네티신, 네오마이신 및 테트라사이클린에 대한 내성 유전자가 있다. 선택제(selective agent)로 처리된 환경에서 선별 마커를 발현하는 세포만 생존하므로 형질전환된 세포를 선별 가능하다. 선택마커의 대표적인 예로써, 영양 요구 마커(auxotrophic marker)인 ura4, leu1, his3 등을 들 수 있으나 상기 예에 의해 본 발명에서 사용될 수 있는 선택마커의 종류가 제한되는 것은 아니다.In addition, when the recombinant vector of the present invention is a replicable expression vector, it may include a replication origin, which is a specific nucleic acid sequence from which replication is initiated. In addition, the recombinant vector may include a selection marker. The selection marker is for selecting cells transformed with the vector, and markers conferring a selectable phenotype such as drug resistance, nutritional requirements, resistance to cytotoxic agents or expression of surface proteins can be used. Vectors of the invention, as markers, include antibiotic resistance genes commonly used in the art and include, for example, ampicillin, gentamicin, carbenicillin, chloramphenicol, streptomycin, kanamycin, geneticin, neomycin and There is a resistance gene for tetracycline. Since only cells expressing a selection marker survive in an environment treated with a selective agent, transformed cells can be selected. Representative examples of selection markers include ura4, leu1, his3, and the like, which are nutritional markers, but the types of selection markers that can be used in the present invention are not limited by the above examples.
발현 벡터에 서브클론된 서열을 증폭시키는 여러 가지 시험관내 증폭 기법들(in vitroamplification techniques)이 알려져 있다. 이러한 기법에는 PCR(polymerase chain reaction), LCR(ligase chain reaction), Qβ-복제효소 증폭(replicase amplification) 및 다른 RNA 중합효소를 이용한 기법들이 있다(Sambrook et al., 1989, Molecular Cloning-A Laboratory Manual(2nd Ed) 1-3; U.S. Patent No. 4,683,202; PCR protocols A Guide to Methods and Applications, Innis et al., eds. Academic Press Inc. San Diego, CA 1990. Improved methods of cloning in vitro amplified nucleic acids are described in U.S. Patent No. 5,426,039). Various in vitro amplification techniques are known for amplifying a subcloned sequence in an expression vector. Such techniques include polymerase chain reaction (PCR), ligand chain reaction (LCR), Qβ-replicase amplification, and other RNA polymerases (Sambrook et al., 1989, Molecular Cloning-A Laboratory Manual). (2nd Ed) 1-3; US Patent No. 4,683,202; PCR protocols A Guide to Methods and Applications, Innis et al., Eds.Academic Press Inc. San Diego, CA 1990. Improved methods of cloning in vitro amplified nucleic acids are described in US Patent No. 5,426,039.
본 발명의 벡터는 그로부터 발현되는 펩타이드의 정제를 용이하게 하기 위하여, 다른 서열과 융합될 수도 있다. 융합되는 서열은 예컨대, 글루타티온 S-트랜스퍼라제(Pharmacia, USA), 말토스 결합 단백질 (NEB, USA), FLAG (IBI, USA) 및 6x His (hexahistidine; Quiagen, USA) 등이 있고, 가장 바람직하게는 6x His이다. 상기 정제를 위한 추가적인 서열 때문에, 숙주에서 발현된 단백질은 친화성 크로마토그래피를 통하여 신속하고, 용이하게 정제된다. 필요한 경우에 이들 펩타이드의 세포외 분비를 촉진할 수 있도록 Fc 절편을 코딩하는 서열이 융합될 수도 있다. Vectors of the present invention may also be fused with other sequences to facilitate purification of the peptides expressed therefrom. Sequences to be fused include, for example, glutathione S-transferase (Pharmacia, USA), maltose binding protein (NEB, USA), FLAG (IBI, USA) and 6x His (hexahistidine; Quiagen, USA), and most preferably Is 6 × His. Because of the additional sequence for this purification, the protein expressed in the host is purified quickly and easily through affinity chromatography. If desired, sequences encoding Fc fragments may be fused to facilitate extracellular secretion of these peptides.
본 발명의 예시적인 실시형태에 따르면, 아이케이 인자 또는 그 부분 단편을 코딩하는 뉴클레오티드 서열이 포함되어 있는 벡터에 의해 발현된 펩타이드는 친화성 크로마토그래피에 의해 정제된다. 예컨대, 글루타티온-S-트랜스퍼라제가 융합된 경우에는 이 효소의 기질인 글루타티온을 이용할 수 있고, 6x His이 이용된 경우에는 Ni-NTA His-결합 레진 컬럼 (Novagen, USA)을 이용하여 소망하는 펩타이드를 신속하고 용이하게 얻을 수 있다. According to an exemplary embodiment of the present invention, peptides expressed by a vector containing a nucleotide sequence encoding an IK factor or partial fragment thereof are purified by affinity chromatography. For example, when glutathione-S-transferase is fused, glutathione, which is a substrate of this enzyme, can be used, and when 6x His is used, a desired peptide can be obtained using a Ni-NTA His-binding resin column (Novagen, USA). Can be obtained quickly and easily.
전술한 벡터를 안정되면서 연속적으로 클로닝 및 발현시킬 수 있는 숙주 세포는 당업계에 공지되어 어떠한 숙주 세포도 이용할 수 있다. 예컨대, E. coli JM109, E. coli BL21(DE3), E. coli RR1, E. coli LE392, E. coli B, E. coli X 1776, E. coli W3110, 바실러스 서브틸리스, 바실러스 츄린겐시스와 같은 바실러스 속 균주, 그리고 살모넬라 티피무리움, 세라티아 마르세슨스 및 다양한 슈도모나스 종과 같은 장내균과 균주 등이 있다.Host cells capable of continuously cloning and expressing the above-described vectors while being stable are known in the art and can use any host cell. For example, E. coli JM109, E. coli BL21 (DE3), E. coli RR1, E. coli LE392, E. coli B, E. coli X 1776, E. coli W3110, Bacillus subtilis, Bacillus thuringiensis Strains of the genus Bacillus, and enterobacteria and strains such as Salmonella typhimurium, Serratia marsons and various Pseudomonas species.
또한, 본 발명의 벡터를 진핵 세포에 형질전환시키는 경우에는 숙주 세포로서, 이스트 (Saccharomyce cerevisiae), 곤충 세포(예컨대 SF9 세포) 및 사람 세포 (예컨대, CHO 세포주 (Chinese hamster ovary), W138, BHK, COS-7, 293, HepG2, 3T3, RIN 및 MDCK 세포주) 등이 이용될 수 있다. In addition, when transforming a vector of the present invention into eukaryotic cells, as host cells, yeast ( Saccharomyce cerevisiae ), insect cells (such as SF9 cells) and human cells (such as CHO cell lines (Chinese hamster ovary), W138, BHK, COS-7, 293, HepG2, 3T3, RIN and MDCK cell lines) and the like.
본 발명의 벡터는 세포를 세포내외(in vivo, ex vivo) 또는 시험관 내(in vitro)에서 유전적으로 변형시키는데 이용될 수 있다. 세포를 유전적으로 변형시키는 방법으로는 세포를 바이러스 벡터로 감염 또는 형질도입 시키는 방법, 인산칼슘 침전법(calcium phosphate precipitation), 수용체 세포(recipient cells)를 DNA를 포함하는 세균 프로토플라스트(bacterial protoplasts)와 융합시키는 방법, 수용체 세포에 DNA를 포함하고 있는 리포좀(liposome) 또는 미세소구(microspheres)를 처리하는 방법, 엔도사이토시스(DEAE dextran, receptor-mediated endocytosis), 일렉트로포레이션(electroporation), 마이크로인젝션(micro-injection), 유전자 밤바드먼트 등을 포함한 여러 가지 방법이 알려져 있다. The vectors of the invention can be used to genetically modify cells in vitro, ex vivo or in vitro . Methods for genetically modifying cells include infection or transduction of cells with viral vectors, calcium phosphate precipitation, bacterial protoplasts containing DNA of receptor cells. Fusion methods, treatment of liposomes or microspheres containing DNA in receptor cells, DEAE dextran, receptor-mediated endocytosis, electroporation, microinjection Various methods are known, including micro-injection and gene bombardment.
약학적 조성물 및 투여 Pharmaceutical Compositions and Administration
본 발명의 다른 양태에 따르면, 본 발명은 전술한 아이케이 인자 또는 그 단편의 치료학적 유효량; 및 필요에 따라 약학적으로 허용되는 담체를 포함하는 관절염 치료를 위한 약학적 조성물에 관한 것이다. 이 경우 아이케이 인자 또는 그 단편인 펩타이드는 피험자에게 직접 투여될 수 있다. According to another aspect of the present invention, the present invention provides a therapeutically effective amount of the aforementioned IK factor or fragment thereof; And it relates to a pharmaceutical composition for the treatment of arthritis comprising a pharmaceutically acceptable carrier as needed. In this case, the IK factor or fragment thereof peptide can be administered directly to the subject.
본 발명의 다른 양태에 따르면, 본 발명은 아이케이 인자 또는 그 단편을 코딩하는 핵산 분자를 가지는 유전자 전달체; 및 필요에 따라 약학적으로 허용되는 담체를 포함하는 관절염 치료를 위한 약학적 조성물에 관한 것이다. 유전자 전달체를 사용하는 것은 이른바 유전자 치료를 목적으로 하는 것이다. According to another aspect of the present invention, the present invention provides a gene carrier having a nucleic acid molecule encoding an IK factor or fragment thereof; And it relates to a pharmaceutical composition for the treatment of arthritis comprising a pharmaceutically acceptable carrier as needed. The use of gene carriers is intended for so-called gene therapy.
따라서 본 발명은 전술한 아이케이 인자 또는 그 단편인 펩타이드 및/또는 아이케이 인자 또는 그 단편을 코딩하는 핵산을 포함하는 유전자 전달체를 유효 성분으로 함유하는 약학적 조성물 또는 이러한 유효 성분을 피험자에게 투여하는 단계를 가지는 관절염 치료 방법에 관한 것이다. 이때 유효 성분은 약학적 조성물 중에 1.0 ng/mL ~ 10 ㎍/mL의 농도로 함유될 수 있다. Accordingly, the present invention provides a method of administering to a subject a pharmaceutical composition or a pharmaceutical composition containing as an active ingredient a gene carrier comprising a nucleic acid encoding an IK factor or a fragment thereof and a nucleic acid encoding the IK factor or a fragment thereof. It relates to a method of treating arthritis having a step. In this case, the active ingredient may be contained in the pharmaceutical composition at a concentration of 1.0 ng / mL to 10 μg / mL.
하나의 실시형태에서, 본 발명의 화합물 및 치료상 불활성 담체, 희석제 또는 부형제를 함유하는 제약 조성물 또는 의약, 뿐만 아니라 이러한 조성물 및 의약을 제조하기 위해 본 발명의 화합물을 사용하는 방법을 제공한다. 한 예에서, 화합물은 생리학상 허용되는 담체, 즉 생약 투여 형태로 사용되는 투여량 및 농도에서 수용자에게 비독성인 담체와 함께, 주위 온도, 적절한 pH에서, 원하는 정도의 순도로 혼합됨으로써 제제화될 수 있다. 제제의 pH는 주로 화합물의 특정한 용도 및 농도에 따라 달라지지만, 바람직하게는 약 3 내지 약 8의 범위이다. 한 예에서, 화합물은 pH 5에서 아세테이트 완충제 중에서 제제화된다. 또 다른 실시양태에서, 화합물은 멸균된다. 화합물은 예를 들어 고체 또는 무정형 조성물, 동결건조 제제 또는 수용액으로서 저장될 수 있다.In one embodiment, there is provided a pharmaceutical composition or medicament containing a compound of the invention and a therapeutically inert carrier, diluent or excipient, as well as a method of using the compound of the invention to make such a composition and medicament. In one example, the compound may be formulated by mixing in a desired degree of purity at ambient temperature, appropriate pH, together with a physiologically acceptable carrier, that is, a carrier that is nontoxic to the recipient at the dosages and concentrations employed in the herbal dosage form. . The pH of the formulation mainly depends on the specific use and concentration of the compound, but is preferably in the range of about 3 to about 8. In one example, the compound is formulated in acetate buffer at pH 5. In another embodiment, the compound is sterile. The compound may be stored, for example, as a solid or amorphous composition, lyophilized formulation or aqueous solution.
조성물은 우수한 의료 행위와 일치하는 방식으로 제제화, 투약 및 투여된다. 이와 관련하여 고려할 인자는 치료할 특정한 장애, 치료할 특정한 환자, 개별 환자의 임상적 상태, 장애의 원인, 작용제의 전달 부위, 투여 방법, 투여 스케줄, 및 의료진에게 공지된 다른 인자를 포함한다.The compositions are formulated, dosed, and administered in a fashion consistent with good medical practice. Factors to consider in this regard include the particular disorder to be treated, the particular patient to be treated, the clinical condition of the individual patient, the cause of the disorder, the site of delivery of the agent, the method of administration, the dosing schedule, and other factors known to the practitioner.
필요한 경우, 지속-방출 제제를 제조할 수 있다. 지속-방출 제제의 적합한 예는 화합물을 함유하는 고체 소수성 중합체의 반투과성 매트릭스를 포함하고, 이 매트릭스는 성형품, 예를 들어 필름 또는 마이크로캡슐의 형태이다. 지속-방출 매트릭스의 예는 폴리에스테르, 히드로겔 (예를 들어, 폴리(2-히드록시에틸-메타크릴레이트), 또는 폴리(비닐알콜)), 폴리락티드, L-글루탐산 및 감마-에틸-L-글루타메이트의 공중합체, 비-분해성 에틸렌-비닐 아세테이트, 분해성 락트산-글리콜산 공중합체 및 폴리-D-(-)-3-히드록시부티르산을 포함한다. If necessary, sustained-release preparations may be prepared. Suitable examples of sustained-release preparations include semipermeable matrices of solid hydrophobic polymers containing the compound, which matrices are in the form of shaped articles, eg films, or microcapsules. Examples of sustained-release matrices include polyesters, hydrogels (eg, poly (2-hydroxyethyl-methacrylate), or poly (vinyl alcohol)), polylactide, L-glutamic acid, and gamma-ethyl- Copolymers of L-glutamate, non-degradable ethylene-vinyl acetate, degradable lactic acid-glycolic acid copolymers and poly-D-(-)-3-hydroxybutyric acid.
한 예에서, 용량당 비경구 투여되는 본 발명의 화합물의 제약 유효량은 하루에 환자 체중을 기준으로 약 0.01-100 mg/kg, 대안적으로 약 0.1 내지 20 mg/kg의 범위일 것이며, 사용되는 화합물의 전형적인 초기 범위는 0.3 내지 15 mg/kg/일이다. 또 다른 실시양태에서, 경구 단위 투여 형태, 예컨대 정제 및 캡슐은 바람직하게는 본 발명의 화합물의 약 5-100 mg을 함유한다. In one example, a pharmaceutically effective amount of a compound of the invention administered parenterally per dose will range from about 0.01-100 mg / kg, alternatively about 0.1 to 20 mg / kg, based on the patient's body weight per day, Typical initial ranges for compounds are 0.3 to 15 mg / kg / day. In another embodiment, oral unit dosage forms such as tablets and capsules preferably contain about 5-100 mg of the compound of the present invention.
본 발명에 따라 유효 성분으로 사용되는 화합물은 임의의 적합한 수단, 예를 들어 경구, 국소 (협측 및 설하 포함), 직장, 질, 경피, 비경구, 피하, 복강내, 폐내, 피내, 경막내 및 경막외 및 비강내, 및 원하는 경우에 국부 치료, 병변내 투여를 위한 수단에 의해 투여될 수 있다. 비경구 주입은 근육내, 정맥내, 동맥내, 복강내 또는 피하 투여를 포함한다. The compounds used as active ingredients according to the invention can be used in any suitable means, for example oral, topical (including buccal and sublingual), rectal, vaginal, transdermal, parenteral, subcutaneous, intraperitoneal, intrapulmonary, intradermal, intradural and Epidural and intranasal, and if desired, by topical treatment, by means for intralesional administration. Parenteral infusions include intramuscular, intravenous, intraarterial, intraperitoneal or subcutaneous administration.
본 발명의 유효 성분인 화합물은 임의의 편리한 투여 형태, 예를 들어 정제, 분말, 캡슐, 용액, 분산액, 현탁액, 시럽, 분무제, 좌제, 겔, 에멀젼, 패치 등으로 투여될 수 있다. 이러한 조성물은 제약 제제에 통상적인 성분, 예를 들어 희석제, 담체, pH 조정제, 감미제, 벌킹제 및 추가의 활성제를 함유할 수 있다.Compounds which are active ingredients of the present invention can be administered in any convenient dosage form, for example tablets, powders, capsules, solutions, dispersions, suspensions, syrups, sprays, suppositories, gels, emulsions, patches and the like. Such compositions may contain components conventional to pharmaceutical formulations, such as diluents, carriers, pH adjusters, sweeteners, bulking agents and additional active agents.
전형적인 제제는 본 발명의 화합물과 담체 또는 부형제를 혼합하여 제조된다. 적합한 담체 및 부형제는 당업자에게 널리 공지되어 있고, 예를 들어 문헌 [Ansel, Howard C., et al., Ansel's Pharmaceutical Dosage Forms and Drug Delivery Systems. Philadelphia: Lippincott, Williams & Wilkins, 2004; Gennaro, Alfonso R., et al. Remington: The Science and Practice of Pharmacy. Philadelphia: Lippincott, Williams & Wilkins, 2000; 및 Rowe, Raymond C. Handbook of Pharmaceutical Excipients. Chicago, Pharmaceutical Press, 2005]에 상세히 기재되어 있다. Typical formulations are prepared by mixing the compound of the invention with a carrier or excipient. Suitable carriers and excipients are well known to those skilled in the art and are described, for example, in Ansel, Howard C., et al., Ansel's Pharmaceutical Dosage Forms and Drug Delivery Systems. Philadelphia: Lippincott, Williams & Wilkins, 2004; Gennaro, Alfonso R., et al. Remington: The Science and Practice of Pharmacy. Philadelphia: Lippincott, Williams & Wilkins, 2000; And Rowe, Raymond C. Handbook of Pharmaceutical Excipients. Chicago, Pharmaceutical Press, 2005.
예를 들어, 본 발명의 약학적 조성물에 포함되는 약제학적으로 허용되는 담체는 제제 시에 통상적으로 이용되는 것으로서, 락토스, 덱스트로스, 수크로스, 솔비톨, 만니톨, 전분, 아카시아 고무, 인산 칼슘, 알기네이트, 젤라틴, 규산칼슘, 미세결정성 셀룰로스, 폴리비닐피롤리돈, 셀룰로스, 물, 시럽, 메틸 셀룰로스, 메틸히드록시벤조에이트, 프로필히드록시벤조에이트, 활석, 스테아르산 마그네슘 및 미네랄 오일 등을 포함하나, 이에 한정되는 것은 아니다.For example, the pharmaceutically acceptable carrier included in the pharmaceutical composition of the present invention is conventionally used in the preparation, lactose, dextrose, sucrose, sorbitol, mannitol, starch, acacia rubber, calcium phosphate, know Nate, gelatin, calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, water, syrup, methyl cellulose, methylhydroxybenzoate, propylhydroxybenzoate, talc, magnesium stearate and mineral oils However, the present invention is not limited thereto.
상기 제제는 또한 하나 이상의 완충제, 안정화제, 계면활성제, 습윤제, 윤활제, 유화제, 현탁화제, 보존제, 항산화제, 불투명화제, 활택제, 가공 보조제, 착색제, 감미제, 향료, 향미제, 희석제, 및 약물 (즉, 본 발명의 화합물 또는 그의 제약 조성물)의 멋진 외양을 제공하거나 제약 제품 (즉, 의약)의 제조에 도움이 되는 다른 공지된 첨가제를 포함할 수 있다. The formulations also contain one or more buffers, stabilizers, surfactants, wetting agents, lubricants, emulsifiers, suspending agents, preservatives, antioxidants, opacifiers, glidants, processing aids, colorants, sweeteners, flavorings, flavoring agents, diluents, and drugs (Ie, compounds of the present invention or pharmaceutical compositions thereof) may include other known additives to provide a nice appearance or to aid in the manufacture of a pharmaceutical product (ie, a medicament).
본 발명의 약제학적 조성물은 당해 발명이 속하는 기술분야에서 통상의 지식을 가진 자가 용이하게 실시할 수 있는 방법에 따라, 약제학적으로 허용되는 담체 및/또는 부형제를 이용하여 제제화함으로써 단위 용량 형태로 제조되거나 또는 다용량 용기내에 내입시켜 제조될 수 있다. 이때 제형은 오일 또는 수성 매질중의 용액, 현탁액 또는 유화액 형태이거나 엑스제, 분말제, 과립제, 정제 또는 캅셀제 형태일 수도 있으며, 분산제 또는 안정화제를 추가적으로 포함할 수 있다.The pharmaceutical compositions of the present invention may be prepared in unit dose form by formulating with a pharmaceutically acceptable carrier and / or excipient according to methods which can be easily carried out by those skilled in the art. Or may be prepared by incorporating into a multi-dose container. In this case, the formulation may be in the form of a solution, suspension or emulsion in an oil or an aqueous medium, or may be in the form of extracts, powders, granules, tablets or capsules, and may further include a dispersant or stabilizer.
생성된 조성물은 건조시키고, 과립화되고, 스테아르산마그네슘과 혼합되고, 통상의 장비를 이용하여 정제 형태로 압축된다. 에어로졸 제제의 예는, 예를 들어 5-400 mg의 본 발명 화합물을 적합한 완충 용액, 예를 들어 포스페이트 완충제에 용해시키고, 원하는 경우에 등장화제, 예를 들어 염화나트륨과 같은 염을 첨가함으로써 제조될 수 있다. 용액을 예를 들어 0.2 마이크로미터 필터를 사용하여 여과함으로써 불순물 및 오염물을 제거할 수 있다.The resulting composition is dried, granulated, mixed with magnesium stearate and compressed into tablet form using conventional equipment. Examples of aerosol formulations can be prepared, for example, by dissolving 5-400 mg of the compound of the invention in a suitable buffer solution such as phosphate buffer and, if desired, by adding a salt such as isotonic agent, for example sodium chloride. have. Impurities and contaminants can be removed by filtering the solution using, for example, a 0.2 micron filter.
따라서 한 실시양태는 화합물 또는 그의 입체이성질체 또는 약학적으로 허용되는 염을 포함하는 약학적 조성물을 포함한다. 추가 실시양태는 화합물 또는 그의 입체이성질체 또는 약학적으로 허용되는 염을 약학적으로 허용되는 담체 또는 부형제와 함께 포함하는 약학적 조성물을 포함한다.Thus one embodiment includes a pharmaceutical composition comprising a compound or stereoisomer or pharmaceutically acceptable salt thereof. Further embodiments include pharmaceutical compositions comprising a compound or stereoisomer or pharmaceutically acceptable salt thereof together with a pharmaceutically acceptable carrier or excipient.
상술한 본 발명의 유전자 치료제는 목적의 핵산분자를 포함하는 유전자 전달체를 유효성분으로 포함한다. 유전자 전달체는 목적의 핵산분자를 운반 및 발현시키기 위하여 제작된 것으로서, 운반 객체인 목적의 핵산분자에 대한 상세한 내용은 위에 기재된 내용과의 중복기재를 피하기 위하여 그 기재를 생략한다.The gene therapy agent of the present invention described above includes a gene carrier comprising a nucleic acid molecule of interest as an active ingredient. The gene carrier is designed to transport and express the nucleic acid molecule of interest, and details of the nucleic acid molecule of interest as a carrier object are omitted in order to avoid overlapping with the above description.
유전자 전달체를 제조하기 위해, 목적의 핵산분자는 적합한 발현 컨스트럭트 (expression construct) 내에 존재하는 것이 바람직하다. 상기 발현 컨스트럭트에서, 목적의 핵산분자는 프로모터에 작동적으로 연결되는 것이 바람직하다. 본 명세서에서, 용어 "작동적으로 결합된"은 핵산 발현 조절 서열 (예: 프로모터, 시그널서열, 또는 전사조절인자 결합 위치의 어레이)과 다른 핵산 서열사이의 기능적인 결합을 의미하며, 이에 의해 상기 조절 서열은 상기 다른 핵산 서열의 전사 및/또는 해독을 조절하게 된다. 본 발명에 있어서, 목적의 핵산 분자에 결합된 프로모터는, 바람직하게는 동물세포, 보다 바람직하게는 포유동물 세포에서 작동하여 목적의 핵산분자의 전사를 조절할 수 있는 것으로서, 포유동물 바이러스로부터 유래된 프로모터 및 포유동물 세포의 게놈으로부터 유래된 프로모터를 포함하며, 예컨대, CMV (cytomegalo virus) 프로모터, 아데노바이러스 후기 프로모터, 백시니아 바이러스 7.5K 프로모터, SV40 프로모터, HSV의 tk 프로모터, RSV 프로모터, EF1 알파프로모터, 메탈로티오닌 프로모터, 베타-액틴 프로모터, 인간 IL-2 유전자의 프로모터, 인간 IFN 유전자의 프로모터, 인간 IL-4 유전자의 프로모터, 인간 림포톡신 유전자의 프로모터, 인간 GM-CSF 유전자의 프로모터, 암세포 특이적 프로모터 (예컨대, TERT 프로모터, PSA 프로모터, PSMA 프로모터, CEA 프로모터, E2F 프로모터 및 AFP 프로모터) 및 조직 특이적 프로모터 (예컨대, 알부민 프로모터)를 포함하나, 이에 한정되는 것은 아니다. 바람직하게는, 본 발명에 이용되는 발현 컨스트럭트는 폴리아데닐화 서열을 포함한다(예: 소성장 호르몬 터미네이터 및 SV40 유래 폴리아데닐화 서열). To prepare a gene carrier, the nucleic acid molecule of interest is preferably present in a suitable expression construct. In the expression construct, the nucleic acid molecule of interest is preferably operably linked to a promoter. As used herein, the term “operably linked” means functional binding between a nucleic acid expression control sequence (eg, an array of promoters, signal sequences, or transcriptional regulator binding sites) and other nucleic acid sequences, thereby The regulatory sequence will control the transcription and / or translation of said other nucleic acid sequence. In the present invention, a promoter bound to a nucleic acid molecule of interest is preferably a promoter derived from a mammalian virus that can operate in animal cells, more preferably mammalian cells to regulate transcription of the nucleic acid molecule of interest. And promoters derived from the genome of mammalian cells, such as, for example, the cytomegalo virus (CMV) promoter, the adenovirus late promoter, the vaccinia virus 7.5K promoter, the SV40 promoter, the tk promoter of HSV, the RSV promoter, the EF1 alpha promoter, Metallothionine promoter, beta-actin promoter, promoter of human IL-2 gene, promoter of human IFN gene, promoter of human IL-4 gene, promoter of human lymphotoxin gene, promoter of human GM-CSF gene, cancer cell specific Red promoters (eg, TERT promoter, PSA promoter, PSMA promoter, CEA promoter, E2F Promoters and AFP promoters) and tissue specific promoters (eg, albumin promoters). Preferably, the expression construct used in the present invention comprises a polyadenylation sequence (e.g., a calcined hormone hormone terminator and an SV40 derived polyadenylation sequence).
유전자 전달체는 다양한 형태로 제작할 수 있는 데, 예시적으로 1) 네이키드(naked) (재조합) DNA 분자, 2) 플라스미드, 3) 바이러스 벡터, 및 4) 상기 네이키드 (재조합) DNA 분자 또는 플라스미드를 내포하는 리포좀 또는 니오좀의 형태로 제작할 수 있다. DNA가 네이키드 상태인 경우((Ulmer et al., 1993, Science 259:1745-1749; Cohen, 1993, Science 259:1691-1692), DNA는 생분해성 구슬(biodegradable beads)에 코팅됨으로써 세포내로 더욱 효율적으로 전달될 수 있다.Gene carriers can be produced in a variety of forms, for example, 1) naked (recombinant) DNA molecules, 2) plasmids, 3) viral vectors, and 4) the naked (recombinant) DNA molecules or plasmids. It can be prepared in the form of nesting liposomes or niosomes. When DNA is naked (Ulmer et al., 1993, Science 259: 1745-1749; Cohen, 1993, Science 259: 1691-1692), DNA is coated into biodegradable beads and further into the cell. Can be delivered efficiently.
목적의 핵산 분자는 통상적인 유전자 치료에 이용되는 모든 유전자 전달 시스템에 적용될 수 있으며, 바람직하게는 플라스미드, 아데노바이러스(Lockett LJ, et al., Clin. Cancer Res. 3:2075-2080, 1997), 아데노-연관 바이러스 (Adeno-associated viruses: AAV, Lashford LS., et al., Gene Therapy Technologies, Applications and Regulations Ed. A. Meager, 1999), 레트로 바이러스 (Gunzburg WH, et al., Retroviral vectors. Gene Therapy Technologies, Applications and Regulations Ed. A. Meager, 1999), 렌티바이러스 (Wang G. et al.,J. Clin. Invest. 104(11):R55-62(1999)), 헤르페스 심플렉스 바이러스 (Chamber R., et al., Proc. Natl.Acad. Sci USA 92:1411-1415, 1995), 백시니아 바이러스 (Puhlmann M. et al., Human Gene Therapy 10:649-657, 1999), 바큘로바이러스 (King LA. and Possee RD., The Baculovirus Expression System, Springer, 1992), 리포좀 (Methods in Molecular Biology, Vol 199, S.C. Basu and M. Basu (Eds.), Human Press 2002) 또는 니오좀에 적용될 수 있다.The nucleic acid molecule of interest can be applied to all gene delivery systems used in conventional gene therapy, preferably plasmids, adenoviruses (Lockett LJ, et al., Clin. Cancer Res. 3: 2075-2080, 1997), Adeno-associated viruses (AAV, Lashford LS., Et al., Gene Therapy Technologies, Applications and Regulations Ed.A. Meager, 1999), retroviruses (Gunzburg WH, et al., Retroviral vectors. Therapy Technologies, Applications and Regulations Ed.A. Meager, 1999), lentiviruses (Wang G. et al., J. Clin. Invest. 104 (11): R55-62 (1999)), herpes simplex virus (Chamber R., et al., Proc. Natl. Acad. Sci USA 92: 1411-1415, 1995), vaccinia virus (Puhlmann M. et al., Human Gene Therapy 10: 649-657, 1999), baculovirus (King LA. And Possee RD., The Baculovirus Expression System, Springer, 1992), Methods in Molecular Biology, Vol 199, SC Basu and M. Basu (Eds.) , Human Press 2002) or niosomes.
상술한 유전자 전달체를 세포내로 도입하는 방법은 당업계에 공지된 다양한 방법을 통해 실시될 수 있다. 본 발명에서, 유전자 전달체가 바이러스 벡터에 기초하여 제작된 경우에는, 당업계에 공지된 바이러스 감염 방법에 따라 실시된다. 바이러스 벡터를 이용한 숙주 세포의 감염은 상술한 인용문헌에 기재되어 있다. The method of introducing the above-described gene carrier into a cell may be carried out through various methods known in the art. In the present invention, when the gene carrier is produced based on a viral vector, it is carried out according to a virus infection method known in the art. Infection of host cells with viral vectors is described in the references cited above.
본 발명에서 유전자 전달 시스템이 네이키드(naked) 재조합 DNA 분자 또는 플라스미드인 경우에는, 미세 주입법(Capecchi, M.R., Cell, 22:479, 1980); 및 Harland와 Weintraub, J. Cell Biol. 101:1094-1099, 1985)), 칼슘 포스페이트 침전법 (Graham, F.L. et al.,Virology, 52:456, 1973); 및 Chen과 Okayama, Mol. Cell. Biol. 7:2745-2752, 1987), 전기 천공법 (Neumann, E. et al., EMBO J.,1:841, 1982; 및 Tur-Kaspa et al., Mol. Cell Biol., 6:716-718(1986)), 리포좀-매개 형질감염법 (Wong, T.K. et al., Gene, 10:87, 1980); Nicolau 및 Sene, Biochim. Biophys. Acta, 721:185-190, 1982; 및 Nicolau et al., Methods Enzymol., 149:157-176, 1987)), DEAE-덱스트란 처리법 (Gopal, Mol. Cell Biol., 5:1188-1190, 1985)), 및 유전자 밤바드먼트(Yang et al., Proc. Natl. Acad. Sci., 87:9568-9572, 1990)) 방법에 의해 유전자를 세포내로 이입시킬 수 있다.In the present invention, when the gene delivery system is naked recombinant DNA molecules or plasmids, micro-injection (Capecchi, M.R., Cell, 22: 479, 1980); And Harland and Weintraub, J. Cell Biol. 101: 1094-1099, 1985)), calcium phosphate precipitation (Graham, F.L. et al., Virology, 52: 456, 1973); And Chen and Okayama, Mol. Cell. Biol. 7: 2745-2752, 1987), electroporation (Neumann, E. et al., EMBO J., 1: 841, 1982; and Tur-Kaspa et al., Mol. Cell Biol., 6: 716-718 (1986)), liposome-mediated transfection (Wong, TK et al., Gene, 10:87, 1980); Nicolau and Sene, Biochim. Biophys. Acta, 721: 185-190, 1982; And Nicolau et al., Methods Enzymol., 149: 157-176, 1987)), DEAE-dextran treatment (Gopal, Mol. Cell Biol., 5: 1188-1190, 1985)), and gene bombardment ( Yang et al., Proc. Natl. Acad. Sci., 87: 9568-9572, 1990)) can be used to introduce the gene into the cell.
이하, 예시적인 실시예를 통하여 본 발명을 더욱 상세하게 설명하지만, 본 발명이 하기 실시예에 기재된 발명으로 한정되지 않는다. Hereinafter, the present invention will be described in more detail with reference to examples, but the present invention is not limited to the invention described in the following examples.
실시예 1 : 아이케이 인자 단편의 CIITA 상위 조절 인자 발현 측정Example 1 Measurement of CIITA Higher Regulatory Factor Expression of IK Factor Fragments
1) IK 인자의 활성 부위 선별1) Selection of active site of IK factor
컴퓨터 프로그램을 사용하여 아이케이 인자의 활성 부위를 선별(screening)하였다. 웹사이트 http://scansite.mit.edu/motifscan_seq.phtml에서 제공하는 in silico program 'scanlite'에서 서열식별번호:3의 아이케이 인자의 절단된 단편(tIK) 아미노산 서열을 입력, 설정하여 선별하였다. 모든 모티프(motif) 분석 데이터는 SWISS-PROT 데이터베이스를 바탕으로 하였다. 하기 표 1에서 아이케이 인자의 활성 부위를 선별한 결과를 표시하고 있다. 표 1에서 score는 tIK의 해당 부위가 인산화효소(kinase)와 실제 상호작용할 가능성을 수치화한 것이고, percnetile은 SWISS-PROT 데이터베이스의 인산화효소 모티프들 중에서 tIK와 연관성을 가는 인산화효소 모티프가 상위 몇%에 해당하는지를 나타내는 수치이다. S382 부위와 Y482 부위는 high/medium/low stringency의 모든 선별 조건에서 제시된 모티프 후보이며, Y489 부위는 medium/low stringency 조건에서 제시된 모티프 후보이다. Computer programs were used to screen the active sites of IK factor. In silico program 'scanlite' provided on the website http://scansite.mit.edu/motifscan_seq.phtml, the fragment fragment (tIK) amino acid sequence of the IK factor of SEQ ID NO: 3 was input and set and selected. . All motif analysis data was based on the SWISS-PROT database. Table 1 shows the results of screening the active site of IK factor. In Table 1, the score quantifies the probability that the site of tIK actually interacts with the kinase, and the percnetile is the highest percentage of kinase motifs associated with tIK among the kinase motifs in the SWISS-PROT database. A number that indicates whether or not The S382 site and the Y482 site are motif candidates presented under all selection conditions of high / medium / low stringency, and the Y489 site is motif candidates presented under medium / low stringency conditions.
표 1 아이케이 인자의 활성 부위 선별
Score percentile Motif Motif group site 서열
0.482 0.416% Aurora A(AuroA) Baso_ST_kina S382 EKKRH S YFEKPKV
0.392 0.273% PKC epsion(PKC-epsion) Baso_ST_kina S382 EKKRH S YFEKPKV
0.323 0.177% (PKA_kin)b Baso_ST_kina S382 EKKRH S YFEKPKV
0.408 0.163% Grb2 SH2(Grb2_SH2) SH2c Y492 TQEEYSE Y MNNKEAL
0.574 0.378% InsR_Kind Y_Kine Y489 DFDTQEE Y SEYMNNK
0.484 0.638% Src_Kinf Y_Kine Y489 DFDTQEE Y SEYMNNK
Table 1 Active site screening of IK factor
Score percentile Motif Motif group site order
0.482 0.416% Aurora A (AuroA) Baso_ST_kin a S382 EKKRH S YFEKPKV
0.392 0.273% PKC epsion (PKC-epsion) Baso_ST_kin a S382 EKKRH S YFEKPKV
0.323 0.177% (PKA_kin) b Baso_ST_kin a S382 EKKRH S YFEKPKV
0.408 0.163% Grb2 SH2 (Grb2_SH2) SH2 c Y492 TQEEYSE Y MNNKEAL
0.574 0.378% InsR_Kin d Y_Kin e Y489 DFDTQEE Y SEYMNNK
0.484 0.638% Src_Kin f Y_Kin e Y489 DFDTQEE Y SEYMNNK
a: Basophilic serine/threonine kinase; b: Protein Kinase A; c: Src homology 2 group; d: Insulin Receptor Kinase; e: Tyrosine kinase group; f: Src Kinase a : Basophilic serine / threonine kinase; b : Protein Kinase A; c : Src homology 2 group; d : Insulin Receptor Kinase; e : Tyrosine kinase group; f : Src Kinase
2) 일부 뉴클레오티드가 절단된 핵산 단편의 설계2) Design of Nucleic Acid Fragments with Some Nucleotide Truncated
도 2에 개략적으로 도시한 것과 같이, 1)에서의 선별 결과를 토대로 tIK 핵산 단편 및 활성 부위의 아미노산을 다른 아미노산으로 치환한 돌연변이를 갖는 변이 tIK 핵산 단편을 설계하였다. tIK 핵산 단편으로서 서열식별번호:3의 유전자 단편을 사용하였으며, kinase 모티프로서 tIK의 기능에 중요한 역할을 할 것으로 예측되는 아미노산(S382, Y489, Y492)을 구조적으로 유사한 아미노산으로 대체한 포인트 돌연변이 핵산 단편을 제작하였다. As schematically shown in FIG. 2, based on the selection result in 1), a mutant tIK nucleic acid fragment having a mutation in which a tIK nucleic acid fragment and an amino acid of an active site were replaced with another amino acid was designed. As the tIK nucleic acid fragment, a gene fragment of SEQ ID NO: 3 was used, and a point mutant nucleic acid fragment in which amino acids (S382, Y489, Y492) predicted to play an important role in the function of tIK as kinase motifs were replaced with structurally similar amino acids. Was produced.
서열식별번호:2의 아미노산 중에서 382번째 아미노산인 세린을 코딩하는 뉴클레오티드(서열식별번호:1의 뉴클레오티드 중에서 1144-1146번째 뉴클레오티드인 AGC)를 알라닌을 코딩하는 뉴클레오티드(GCC)로 치환한 돌연변이 유전자 단편(S382A)을 제작하였다. 또한, 서열식별번호:2의 아미노산 중에서 489번째 아미노산인 티로신을 코딩하는 뉴클레오티드(서열식별번호:1의 뉴클레오티드 중에서 1465-1467번째 뉴클레오티드인 TAC)를 페닐알라닌을 코딩하는 뉴클레오티드(TTC)로 치환한 돌연변이 유전자 단편(Y489F)과, 서열식별번호:2의 아미노산 중에서 492번째 아미노산인 티로신을 코딩하는 뉴클레오티드(서열식별번호:1의 뉴클레오티드 중에서1474-1475번째 뉴클레오티드인 TAC)를 페닐알라닌을 코딩하는 뉴클레오티드(TTC)로 치환한 이중 돌연변이 유전자 단편(Y492F)을 제작하였다. A mutant gene fragment in which the nucleotide encoding serine (the amino acid of SEQ ID NO: 2), which is the 3rd amino acid (AGC, which is the 1144-1146 nucleotide among the nucleotides of SEQ ID NO: 1), is replaced with the nucleotide (GCC) encoding alanine ( S382A) was produced. A mutant gene in which the nucleotide encoding tyrosine, which is the 489th amino acid among the amino acids of SEQ ID NO: 2 (TAC, which is the 1465-1467 nucleotide among the nucleotides of SEQ ID NO: 1), was substituted with the nucleotide (TTC) encoding phenylalanine A fragment (Y489F) and a nucleotide encoding tyrosine, which is the 492th amino acid among the amino acids of SEQ ID NO: 2, as a nucleotide (TTC) encoding phenylalanine A substituted double mutant gene fragment (Y492F) was produced.
동시에 서열식별번호:2의 아미노산 중에서 489번째 및 492번째 티로신을 코딩하는 뉴클레오티드를 페닐알라닌을 코딩하는 뉴클레오티드로 치환한 돌연변이 유전자 단편(Y489492F)과, 서열식별번호:2의 아미노산 중에서 382번째 세린, 489번째 및 492번째 티로신을 각각 알라닌과 페닐알라닌을 코딩하는 뉴클레오티드로 치환한 삼중 돌연변이 유전자 단편(S382AY489492F)을 제작하였다. 발현되는 단백질의 탐지, 분리 및 정제 등을 위하여 각각의 유전자 단편의 5' 말단에 HA-tag sequence(hemagglutinin sequence; 서열식별번호:54)를 첨가하였다. 본 실시예에서 사용된 tIK 핵산 단편과, 돌연변이 핵산 단편의 뉴클레오티드 서열은 하기 표 2에 표시되어 있다. A mutant gene fragment (Y489492F) in which the nucleotides encoding the 489th and the 492th tyrosine among the amino acids of SEQ ID NO: 2 were replaced with the nucleotides encoding the phenylalanine (Y489492F), and the 389th serine and the 489th amino acids of the amino acid of SEQ ID NO: 2; And a triple mutant gene fragment (S382AY489492F) in which the 492th tyrosine was substituted with nucleotides encoding alanine and phenylalanine, respectively. HA-tag sequence (hemagglutinin sequence; SEQ ID NO: 54) was added to the 5 'end of each gene fragment for detection, isolation and purification of the expressed protein. The tIK nucleic acid fragments used in this example and the nucleotide sequences of the mutant nucleic acid fragments are shown in Table 2 below.
표 2 아이케이 핵산 단편 및 돌연변이 핵산 단편
단편 뉴클레오티드서열 아미노산서열 비고
tIK 서열식별번호:3 서열식별번호:4
S382A 서열식별번호:5 서열식별번호:6 Ser→Ala
Y489F 서열식별번호:7 서열식별번호:8 Tyr→Phe
Y492F 서열식별번호:9 서열식별번호:10 Tyr→Phe
Y489492F 서열식별번호:11 서열식별번호:12 Tyr→Phe
S382AY489492F 서열식별번호:13 서열식별번호:14 Ser→Ala; Tyr→Phe
TABLE 2 IK nucleic acid fragments and mutant nucleic acid fragments
snippet Nucleotide Sequence Amino acid sequence Remarks
tIK SEQ ID NO: 3 SEQ ID NO: 4
S382A SEQ ID NO: 5 SEQ ID NO: 6 Ser → Ala
Y489F SEQ ID NO: 7 SEQ ID NO: 8 Tyr → Phe
Y492F SEQ ID NO: 9 SEQ ID NO: 10 Tyr → Phe
Y489492F SEQ ID NO: 11 SEQ ID NO: 12 Tyr → Phe
S382AY489492F SEQ ID NO: 13 SEQ ID NO: 14 Ser → Ala; Tyr → Phe
tIK 단편 및 돌연변이 핵산 단편은 fusion PCR 법을 이용하여 증폭시킨 insert를 pcDNA 3.1 vector에 삽입하여 제작하였다. S382A 돌연변이의 경우, HA tag부터 tIK 핵산의 5' 부분과 상보적인 정방향 프라이머와, 382번째 serine을 alanine으로 치환 할 수 있도록 alanine을 coding할 수 있는 nucleotide 서열을 지니는 역방향 프라이머 세트를 제작하여 PCR에 이용하였다. 이 프라이머 세트를 이용하여 tIK 유전자를 template로 사용하여 HA tag부터 382번 alanine(Ala)에 이르는 첫 번째 절편을 증폭하였다. 이어서 382번째 serine을 alanine으로 치환할 수 있도록 alanine을 coding할 수 있는 nucleotide를 포함하는 정방향 프라이머와 tIK 유전자의 3'말단 부분에 상보적인 역방향 프라이머 세트를 제작하여 382번 alanine부터 tIK 핵산 말단에 이르는 두 번째 절편을 증폭하였다. PCR을 통해 얻은 두 가지 절편을 500 ㎕ micro tube에 각각 1㎕씩 넣은 후 HA tag부터 tIK 유전자의 앞부분에 상보적인 정방향 프라이머와 tIK 유전자의 말단 부분에 상보적인 역방향 프라이머를 1 ㎕씩 첨가 후 polymerase buffer와 dNTP, polymerase, distilled water를 첨가하여 fusion PCR을 진행하였다. PCR을 통해 얻은 증폭절편의 말단을 제한효소로 자른 뒤 pcDNA 3.1에 삽입하고 transformation함으로써 아미노산이 치환된 insert가 삽입된 플라스미드를 selection할 수 있었으며, 아미노산의 치환여부는 sequencing을 통해 최종적으로 확인하였다. 다른 돌연변이도 위와 같은 방법으로 제작하였다. 하기 표 3은 본 실시예에 따라 tIK 핵산 단편 및 돌연변이 tIK 핵산 단편의 증폭을 위해 사용된 프라이머 세트를 표시하고 있다. The tIK fragment and the mutant nucleic acid fragment were prepared by inserting the amplified insert into the pcDNA 3.1 vector using fusion PCR. In the case of the S382A mutant, a PCR primer was prepared using a reverse primer set having a forward primer complementary to the 5 'portion of the tIK nucleic acid from the HA tag, and a nucleotide sequence capable of encoding alanine to replace the 382 th serine with alanine. It was. Using this primer set, the first fragment from HA tag to alanine (Ala) 382 was amplified using the tIK gene as a template. Subsequently, a set of forward primers containing nucleotides encoding alanine and complementary reverse primer sets at the 3 ′ end of the tIK gene were used to replace the 382 th serine with alanine. The first section was amplified. 1μl each of the two fragments obtained by PCR was added to 500μl microtube, and 1μl of complementary forward primer complementary to the front of the tIK gene and 1μl of complementary reverse primer to the terminal of the tIK gene. And fusion PCR was performed by adding dNTP, polymerase, and distilled water. By cutting the ends of the amplification fragments obtained by PCR with restriction enzymes and inserting them into pcDNA 3.1 and transforming, the plasmid containing the amino acid-substituted insert was selected, and the amino acid substitution was finally confirmed by sequencing. Other mutations were made in the same way as above. Table 3 below shows the primer sets used for amplification of tIK nucleic acid fragments and mutant tIK nucleic acid fragments according to this example.
표 3 tIK 핵산 단편 및 돌연변이 핵산 단편 증폭에 사용된 프라이머 세트
plasmid(HA tagged) 제 1 절편 프라이머 제 2 절편 프라이머
tIK - 정방향 : 서열식별번호:15a- 역방향 : 서열식별번호:16b
S382A - 정방향 : 서열식별번호:17*- 역방향 : 서열식별번호:19 - 정방향 : 서열식별번호:20- 역방향 : 서열식별번호:16
Y489F - 정방향 : 서열식별번호:18c- 역방향 : 서열식별번호:21 - 정방향 : 서열식별번호:22- 역방향 : 서열식별번호:16
Y492F - 정방향 : 서열식별번호:17- 역방향 : 서열식별번호:23 - 정방향 : 서열식별번호:24- 역방향 : 서열식별번호:16
Y489492F - 정방향 : 서열식별번호:18c- 역방향 : 서열식별번호:25 - 정방향 : 서열식별번호:26- 역방향 : 서열식별번호:16
S382Y489492F S67A 절편과 Y174_177F 절편을 template로 함.정방향 프라이머로 서열식별번호:18c을, 역방향 프라이머로 서열식별번호:16을 사용하여 세 곳이 mutation된 insert를 얻었음.
TABLE 3 Primer sets used for amplification of tIK nucleic acid fragments and mutant nucleic acid fragments
plasmid (HA tagged) Section 1 Primer 2nd section primer
tIK -Forward: SEQ ID NO: 15 a -Reverse: SEQ ID NO: 16 b
S382A -Forward: SEQ ID NO: 17 * -Reverse: SEQ ID NO: 19 -Forward: SEQ ID NO: 20- Reverse: SEQ ID NO: 16
Y489F -Forward: SEQ ID NO: 18 c -Reverse: SEQ ID NO: 21 -Forward: SEQ ID NO: 22- Reverse: SEQ ID NO: 16
Y492F -Forward: SEQ ID NO: 17- Reverse: SEQ ID NO: 23 -Forward: SEQ ID NO: 24- Reverse: SEQ ID NO: 16
Y489492F -Forward: SEQ ID NO: 18 c -Reverse: SEQ ID NO: 25 -Forward: SEQ ID NO: 26- Reverse: SEQ ID NO: 16
S382Y489492F Using the S67A fragment and the Y174_177F fragment as a template, three mutated inserts were obtained using SEQ ID NO: 18 c as the forward primer and SEQ ID NO: 16 as the reverse primer.
a: EcoRⅠ 인식 부위 포함; b: XhoⅠ 인식부위 포함; c: HindⅢ 인식부위 포함 a : including EcoRI recognition site; b : including XhoI recognition site; c : HindIII recognition site included
MHC class Ⅱ의 발현을 촉진하는 class Ⅱ transactivator인 CIITA의 발현을 조절하는 상위 조절인자들의 mRNA 발현 정도를 통해 tIK 및 돌연변이 핵산의 기능을 확인하였다. CIITA의 발현을 조절하는 상위 조절인자로서 CNOT1(CCR4-NOT transcription complex subunit 1), CDCA3(Cell division cycle-associated protein 3), 및 MAPK1(Mitogen-activated protein kinase 1)을 선정하였다. 내재성 tIK를 가지고 있지 않은 human Raji B세포에 tIK가 삽입된 플라스미드를 transfection하는 경우, control vector를 transfection한 그룹에 비해 CIITA의 발현을 억제하는 인자들(CNOT1, CDCA3, MAPK1)의 발현을 증가시키는 것을 확인하였다. tIK 핵산 단편은 CIITA를 억제하는 인자들을 증가시킴으로써 최종적으로 MHC class II의 발현 억제에 영향을 미치는 것이라고 생각되었다. 이와 같은 결과를 바탕으로 kinase motif를 다른 아미노산으로 치환한 tIK 돌연변이 핵산 단편에서도 이와 같은 현상이 나타나는지를 확인하였다. The function of tIK and mutant nucleic acids was confirmed by the mRNA expression level of the upper regulators that regulate the expression of CIITA, a class II transactivator that promotes MHC class II expression. CNOT1 (CCR4-NOT transcription complex subunit 1), CDCA3 (Cell division cycle-associated protein 3), and Mitogen-activated protein kinase 1 (MAPK1) were selected as top regulators that regulate the expression of CIITA. Transfection of tIK-inserted plasmids into human Raji B cells without endogenous tIK increased expression of factors that inhibit the expression of CIITA (CNOT1, CDCA3, MAPK1) compared to the control vector-transfected group. It was confirmed. tIK nucleic acid fragments were thought to ultimately influence the inhibition of expression of MHC class II by increasing the factors that inhibit CIITA. Based on these results, it was confirmed whether the same phenomenon occurs in the tIK mutant nucleic acid fragment in which the kinase motif is replaced with another amino acid.
구체적으로 tIK 돌연변이체인 S382A, Y489F, Y492F, Y489492F, S382AY489492F 단편이 각각 삽입된 플라스미드를 Raji B 세포에 electrophoration을 통해 transfection한 후 세포에서 RNA를 분리하여 cDNA를 제조하였다. tIK 핵산 단편이 삽입된 플라스미드를 또한 사용하였으며, 이들 핵산 단편이 삽입되지 않은 pcDNA 3.1 플라스미드를 각각 transfection하여 얻은 cDNA를 control 그룹으로 사용하였다. 제조한 cDNA를 이용해 quantitative real-time PCR을 수행하였으며, 측정된 결과 값 (Ct 값)은 2(-ΔΔCt) method를 이용하여 pcDNA 3.1 플라스미드를 transfection한 그룹의 결과를 기준으로 하는 상대적인 값으로 계산하였다. CIITA의 발현을 조절하는 인자들(CDCA3, CNOT1, MAPK1)의 cDNA를 template로 사용하였으며, 이들 인자의 발현을 확인하기 위하여 사용된 프라이머 서열은 표 4에 표시되어 있다. Specifically, cDNA was prepared by transfection of Raji B cells with electroplasmic plasmids in which the tIK mutants S382A, Y489F, Y492F, Y489492F, and S382AY489492F were inserted. Plasmids in which tIK nucleic acid fragments were inserted were also used, and cDNAs obtained by transfection of the pcDNA 3.1 plasmids into which these nucleic acid fragments were not inserted were used as control groups. Quantitative real-time PCR was performed using the prepared cDNA, and the measured result value (Ct value) was calculated as a relative value based on the result of the group transfected with pcDNA 3.1 plasmid using the 2 (-ΔΔCt) method. . CDNA of the factors that regulate the expression of CIITA (CDCA3, CNOT1, MAPK1) was used as a template, and the primer sequences used to confirm the expression of these factors are shown in Table 4.
표 4
CIITA 조절 인자 정방향 프라이머 역방향 프라이머
CDCA3 서열식별번호:27 서열식별번호:28
CNOT1 서열식별번호:29 서열식별번호:30
MAPK1 서열식별번호:31 서열식별번호:32
Table 4
CIITA regulator Forward primer Reverse primer
CDCA3 SEQ ID NO: 27 SEQ ID NO: 28
CNOT1 SEQ ID NO: 29 SEQ ID NO: 30
MAPK1 SEQ ID NO: 31 SEQ ID NO: 32
본 실시예에 따라 CIITA의 발현을 조절하는 인자들의 발현을 측정한 결과는 도 3에 도시되어 있다. Negative Control vector인 pcDNA 3.1을 transfection한 그룹을 기준으로 하였을 때, tIK가 transfection된 그룹에서는 CIITA의 발현을 억제하는 인자인 CDCA3, CNOT1, MAPK1이 모두 증가되어 있는 것을 확인할 수 있다. 또한 Y492F, Y489492F, S382AY489492F는 돌연변이는 해당 서열 위치의 아미노산이 다른 아미노산으로 치환되어, positive control인 tIK에 비하여 CIITA의 발현을 억제하는 인자의 발현이 감소하였다. 특히, S382와 Y489F 돌연변이는 tIK 발현으로 인해 증가했던 CIITA 발현 억제 유전자의 발현을 크게 감소시켰다. 따라서 전장 아이케이 유전자에서 382번째 및 492번째 아미노산이 특히 아이케이의 생리적 기능을 담당하는 활성 부위로 작용할 수 있다는 점을 확인하였다. The results of measuring the expression of factors regulating the expression of CIITA according to the present embodiment are shown in FIG. 3. Based on the group transfected with pcDNA 3.1, a negative control vector, it was found that CDCA3, CNOT1, and MAPK1, which are factors that inhibit CIITA expression, were increased in the tIK transfected group. In addition, the mutation of Y492F, Y489492F, and S382AY489492F was replaced by another amino acid at the corresponding sequence position, and the expression of the factor that inhibits the expression of CIITA was reduced as compared with the positive control tIK. In particular, the S382 and Y489F mutations significantly reduced the expression of the CIITA expression inhibitory gene, which was increased due to tIK expression. Therefore, it was confirmed that the 382th and 492th amino acids in the full-length IK gene can act as an active site in particular responsible for the physiological function of IK.
실시예 2 : IL-1 Receptor antagonist knock-out mice 및 IK Transgenic mice를 활용한 동물 모델 개발Example 2 Animal Model Development Using IL-1 Receptor Antagonist Knock-out Mice and IK Transgenic Mice
관절염 유도 물질에 대한 감수성을 높이기 위해 면역학적으로 감수성이 높으며, 자발적으로 관절염이 생성되는 동물모델인 IL-1 receptor antagonist knock out mice (IL1RaKO, Balb/c 유래)와, 서열식별번호:3의 뉴클레오티드 서열을 가지는 절단된 아이케이 인자를 코딩하는 핵산(tIK 핵산 단편)을 발현시킬 수 있는 tIK-Transgenic mice (tIK, Balb/c 유래)를 cross-breeding시켜서 새로운 자가 면역 관절염 유발 동물 마우스(tIK-IL1RaKO) 모델을 개발하였다. Truncated IK(tIK)를 발현하는 transgenic mice를 제작하기 위하여 서열식별번호:3의 핵산이 삽입되어 있는 tIK-pcDNA 3.1 클론을 Balb/c female로부터 얻은 수정란에 micro-injection하였다. 이를 가임신기의 Balb/c female mouse의 자궁에 이식하여 착상시킨 후 3주 후 새끼를 얻었다. Cross-breeding을 통해 얻은 mice의 유전자를 얻기 위해 생후 3주 후에 꼬리를 0.3 mm로 자른 후 tail lysis buffer (pH8.0 Tris buffer 50mM, EDTA 50mM, 0.5% SDS in distilled water, 20mg/ml proteinase K)를 넣어 55℃ 항온수조에서 18시간 incubatoin하여 tail lysate를 얻었다. IL-1 receptor antagonist knock out mice (IL1RaKO, from Balb / c), an animal model that is immunologically sensitive and spontaneously develops arthritis to increase the sensitivity to arthritis inducing substances, and the nucleotides of SEQ ID NO: 3 A new autoimmune arthritis-induced animal mouse (tIK-IL1RaKO) by cross-breeding tIK-Transgenic mice (tIK, Balb / c) capable of expressing a nucleic acid encoding a truncated IK factor with sequence (tIK nucleic acid fragment) Developed a model. To prepare truncated IK (tIK) transgenic mice, tIK-pcDNA 3.1 clone containing the nucleic acid of SEQ ID NO: 3 was micro-injected into fertilized eggs obtained from Balb / c female. The baby was implanted 3 weeks after implantation into the uterus of Balb / c female mouse of fertility. To obtain the gene of mice obtained through cross-breeding, after 3 weeks of age, the tail was cut to 0.3 mm and tail lysis buffer (pH8.0 Tris buffer 50mM, EDTA 50mM, 0.5% SDS in distilled water, 20mg / ml proteinase K) Incubated for 18 hours in a 55 ℃ constant temperature water bath to obtain a tail lysate.
이를 vacutainer에 옮기고 phenol/chloroform/isoamylalcohol(25:24:1)를 넣고 50회 inverting 후 원심분리(2000 rpm, 5 min, 실온)하여 genomic DNA를 제외한 나머지 debris를 제거하였다. 여기에 chloroform을 넣어 다시 한 번 inverting하고 원심분리(2000 rpm, 5 min, 실온)를 함으로써 genomic DNA층만을 분리하였다. 분리 후 얻은 투명한 상층액을 새로운 micro tube에 옮기고, 여기에 100% 에탄올을 첨가한 후 inverting 하여 하얀 실타래 모양의 genomic DNA를 얻었다. 이를 파이펫 팁으로 건져 올린 후 75% 에탄올을 이용하여 세척하고 10분 동안 air dry한 후 distilled water에 녹였다. tIK 핵산 단편이 삽입된 transgenic mouse에서 유래한 녹인 genomic DNA를 template로, tIK genotyping용 프라이머 세트(정방향 프라이머는 서열식별번호:33; 역방향 프라이머는 서열식별번호:34)를 이용하여 PCR 법으로 증폭하였다. 한편, IL1RaKO genotyping용 프라이머 세트(정방향 프라이머는 서열식별번호:35, 역방향 프라이머는 서열식별번호:36 및 37)를 이용하여 PCR 법으로 증폭한 후 두 가지 증폭 산물을 1% Agarose에 전기영동하여 증폭 정도를 비교하였다. 전기영동 결과를 토대로 두 가지 증폭 산물이 모두 확인된 경우를 cross-breeding을 통해 얻은 tIK-IL1RaKO double positive transgenic mice로 구분하였다. This was transferred to a vacutainer, phenol / chloroform / isoamylalcohol (25: 24: 1) was added and inverted 50 times, followed by centrifugation (2000 rpm, 5 min, room temperature) to remove the remaining debris except genomic DNA. The chloroform was added to the inverting and centrifugation (2000 rpm, 5 min, room temperature) to separate only the genomic DNA layer. The transparent supernatant obtained after the separation was transferred to a new micro tube, and 100% ethanol was added thereto, followed by inverting to obtain a white thread-like genomic DNA. The pipette tip was lifted up, washed with 75% ethanol, air dried for 10 minutes, and dissolved in distilled water. Amplified genomic DNA derived from a transgenic mouse into which tIK nucleic acid fragment was inserted was amplified by PCR using a primer set for tIK genotyping (forward primer is SEQ ID NO: 33; the reverse primer is SEQ ID NO: 34). . On the other hand, amplification by PCR method using a primer set for IL1RaKO genotyping (forward primer is SEQ ID NO: 35, the reverse primer is SEQ ID NO: 36 and 37) and then amplified by electrophoresis of two amplification products in 1% Agarose The degree was compared. Based on the results of electrophoresis, both amplification products were identified as tIK-IL1RaKO double positive transgenic mice obtained through cross-breeding.
실시예 3 : 자가면역 관절염 동물 모델을 이용하여 IK의 효능 분석Example 3 Efficacy Analysis of IK Using an Autoimmune Arthritis Animal Model
실시예 2의 Cross-breeding을 통해 얻은 새로운 자가면역 관절염 동물모델인 tIK-IL1RaKO mice와 IL1RaKO mice를 대상으로 관절염의 유발 정도를 관찰하였다. 자연적인 상태에서 관절염이 유발 되는 정도를 비교하기 위하여, 4주령의 tIK-IL1RaKO mice 10수와 IL1RaKO mice 10수를 케이지에서 동일한 조건에서 사육하였다. 사육하는 동안 매 일주일마다 관절염의 정도를 마우스 발목 부위의 부종 정도로 확인하여 점수화하였다. 상세한 점수 개수 방법은 다음과 같다. 관절염 평가는 Rosoliniec 등에 의한 평균 관절염 지수(mean arthritic index)에 기준하여 기록하였으며, 한쪽 발목의 최대 점수를 4점으로 하고 mice의 네 개 발이 16점을 초과하지 않는 범위에서 1주일에 3회씩 모니터링을 지속적으로 진행하였다. 또한 모니터링 시기별로 관절염이 발생하는 정도를 %로 나타내어 발병률로 나타냈다. 도 4a는 관절염 지수를 측정한 그래프이고, 도 4b는 관절염의 발병률을 측정한 그래프이다. 또한 도 5는 각 군의 마우스에서 관찰된 관절 사진을 촬영한 사진이다. 도시된 것과 같이 tIK-IL1RaKO 마우스 그룹은 IL1RaKO 마우스 그룹에 비하여 유의미하게 낮은 관절염 지수를 나타냈으며, 발병율 역시 60% 이상 낮은 관절염 유발 정도를 보여주었다. 또한 마우스 발목 부위의 염증의 정도도 tIK-IL1RaKO 그룹이 IL1RaKO 그룹에 비해 상당히 완화되어 있는 것을 확인하였다.The incidence of arthritis was observed in tIK-IL1RaKO mice and IL1RaKO mice, which are new autoimmune arthritis animal models obtained through cross-breeding of Example 2. To compare the incidence of arthritis in the natural state, 10 numbers of tIK-IL1RaKO mice and 10 numbers of IL1RaKO mice were bred in cages under the same conditions. During breeding, the degree of arthritis was confirmed by checking the degree of swelling of the mouse ankle area every week. The detailed score counting method is as follows. Arthritis evaluation was recorded based on the mean arthritic index by Rosoliniec, etc., and the monitoring was performed three times a week in the range that the maximum score of one ankle was 4 points and the four feet of the mice did not exceed 16 points. Continued. In addition, the incidence rate of arthritis was expressed in% by monitoring period. Figure 4a is a graph measuring the arthritis index, Figure 4b is a graph measuring the incidence of arthritis. In addition, Figure 5 is a photograph taken a joint picture observed in the mouse of each group. As shown, the tIK-IL1RaKO mouse group showed a significantly lower arthritis index than the IL1RaKO mouse group, and the incidence rate also showed a 60% lower incidence of arthritis. In addition, it was confirmed that the degree of inflammation of the mouse ankle region was significantly alleviated in the tIK-IL1RaKO group compared to the IL1RaKO group.
실시예 4 : 마우스의 관절 부위 병인 상태 확인Example 4 Checking the Pathogenesis of Mouse Joints
관절염이 유도된 마우스 관절의 병인상태를 확인하기 위하여 관절염 자연 발생 그룹의 모니터링 16주차에 각 그룹(tIK-IL1RaKO 그룹; IL1RaKO 그룹)의 마우스를 부검하여 관절 부위를 얻었다. 관절 부위를 10% 중성 포르말린에 고정하고, 탈회 용액에 7시간 동안 침지하여 탈회하였다. 탈회된 관절 부위를 16시간에 걸쳐 수세하고, 탈수, 청명, 침투 과정을 거쳐 파라핀 블록(paraffin block)을 제조하였다. 절편 기기를 사용하여 파라핀 블록을 절단하여 7 ㎛ 절편을 만들고 슬라이드에 붙인 후 헤마톡실린과 에오진(hematoxylin & eosin, H&E) 염색을 하였다. H&E 염색의 과정은 다음과 같다. 파라핀 절편에서 파라핀을 제거(deparaffinization)한 후 함수 과정을 진행하고, 헤마톡실린으로 핵을 염색하였다. 이를 세척한 뒤, 에오진을 이용하여 세포질을 염색하였다. 이를 다시 탈수, 투명, 봉입 과정을 거쳐 최종적으로 발목 부분의 tarsal bone의 구조적 상태 및 다양한 면역세포들의 침윤을 광학현미경으로 확인하였다. 결과는 도 6a에 도시되어 있다. tIK-IL1RaKO 마우스 유래의 관절 조직은, IL1RaKO 마우스 유래의 관절 조직보다 tarsal bone의 구조적 변화가 작고 면역세포의 침윤이 덜 일어났다. 이를 통해 윤활관절부위의 염증(synovial inflammation)이 tIK-IL1RaKO 그룹에서 낮게 발생하였음을 확인할 수 있었다. In order to confirm the pathogenesis of arthritis-induced mouse joints, the joint sites were obtained by monitoring the arthritis spontaneously occurring group at the 16th week of each group (tIK-IL1RaKO group; IL1RaKO group). Joint sites were fixed in 10% neutral formalin and demineralized by soaking in demineralization solution for 7 hours. The demineralized joint area was washed with water for 16 hours, and a paraffin block was prepared by dehydration, clarity, and infiltration. Paraffin blocks were cut using a slicer, 7 μm slices were prepared, attached to slides, and stained with hematoxylin and eosin (H & E). The process of H & E staining is as follows. Paraffin was removed from the paraffin sections (deparaffinization), followed by hydration, and nuclei stained with hematoxylin. After washing, the cytoplasm was stained with eogene. After dehydration, transparency, and encapsulation, the optical status of the tarsal bone in the ankle and the infiltration of various immune cells were confirmed. The results are shown in Figure 6a. Articular tissues derived from tIK-IL1RaKO mice had smaller structural changes in tarsal bone and less infiltration of immune cells than those from IL1RaKO mice. It was confirmed that synovial inflammation was low in the tIK-IL1RaKO group.
또한 safranin O 염색법을 통해 연골부위의 침식 정도(cartilage erosion)를 확인하였다. Safranin O 염색은 조직 절편의 deparaffinization 과정에 이어 함수 처리, 헤마톡실린 염색, safranin O 염색, fast green 염색, 탈수, 투명, 봉입의 과정을 거쳐 진행하였다. 결과는 도 6b에 도시되어 있다. 연골의 침식 정도는 염색된 부위의 색채 변화를 통해서 확인할 수 있는데, IL1RaKO 유래의 관절 조직은 연골이 상당부분 파괴되어 있으나 tIK-IL1RaKO 유래의 관절의 조직에서는 연골부분이 양호하게 유지되어 있음을 확인하였다. In addition, cartilage erosion was confirmed by safranin O staining. Safranin O staining was followed by deparaffinization of tissue sections, followed by water treatment, hematoxylin staining, safranin O staining, fast green staining, dehydration, clearing and encapsulation. The results are shown in Figure 6b. The degree of erosion of cartilage can be confirmed by changing the color of the stained area.In the articular tissue derived from IL1RaKO, the cartilage is destroyed in a large part, but the tissue of the joint derived from tIK-IL1RaKO is well maintained. .
실시예 5: 관절염에 의한 골 손상 정도 분석Example 5 Analysis of Bone Damage by Arthritis
관절염이 유도된 마우스에서 염증으로 인하여 손상된 뼈 부분을 IL1RaKO 그룹과 tIK-IL1RaKO 그룹에서 비교분석하기 위하여 관절염 자연발생 그룹의 모니터링 16주차에 각 그룹별 마우스를 부검하여 관절 부위를 얻었다. 조직을 10% 중성 포르말린에 18시간 고정 후 skyscan1172 장비를 이용하여 micro-CT를 촬영하였다. 50kV, 200uA의 X-ray source를 이용하고, 0,5mm의 필터를 사용하여 15μm의 픽셀 해상도로 조직의 단면영상을 촬영하였다. IL1RaKO 마우스 유래의 관절은 염증으로 인해 뼈가 많이 손상되고 구조적 변형이 관찰되었으나, tIK-IL1RaKO 마우스 유래의 관절의 경우 뼈의 손상이 거의 없는 본래의 구조를 유지하고 있었다(도 7). 이를 통해 아이케이의 발현이 관절염에 의한 뼈의 파괴를 억제하는 데에도 관여한다는 것을 확인하였다.In order to compare the bone damage due to inflammation in arthritis-induced mice in the IL1RaKO group and the tIK-IL1RaKO group, the joint sites were obtained at the 16th week of monitoring of the arthritis spontaneous group. Tissues were fixed in 10% neutral formalin for 18 hours and micro-CT was photographed using skyscan1172. X-ray sources of 50 kV and 200 uA were used, and a cross-sectional image of the tissue was taken at a pixel resolution of 15 μm using a filter of 0,5 mm. Joints derived from IL1RaKO mice were found to have many bone damages and structural deformations due to inflammation, but the joints derived from tIK-IL1RaKO mice maintained their original structure with little bone damage (FIG. 7). It was confirmed that IK expression is also involved in suppressing bone destruction caused by arthritis.
실시예 6 : 관절부위 염증 사이토카인의 발현 확인Example 6 Expression of Inflammatory Cytokines at the Joint Site
(1) mRNA 발현 확인(1) mRNA expression confirmation
관절염이 유도된 마우스 관절에서의 염증 사이토카인 발현정도를 확인하기 위하여 관절염 자연발생 그룹의 모니터링 16주차에 각 그룹의 마우스를 부검하여 관절 부위를 얻었다. 관절 부위를 LN2에 마쇄한 후 Trizol reagent 400 ㎕에 넣어 RNA isolation을 진행하였다. Chloroform을 넣고 강하게 vortex한 후 5분 동안 상온에서 incubation하였다. 원심분리(12500 rpm, 15 min, 4℃) 후 상층액을 새로운 micro tube로 옮겼다. 이후 상층액에 isopropanol과 glycoblue를 넣고 5분 동안 상온에서 incubation하였다. 원심분리(12500 rpm, 10 min, 4℃) 후 상층액을 제거하였다. 상층액 제거 후 남은 pellet을 70% 에탄올로 세척하고, 원심분리(9500 rpm, 5 min, 4℃)한 뒤, pellet을 air dry 하고 DEPC-water로 녹여주었다. 분리된 RNA는 QuantiTect Reverse Transcription Kit (Qiagen)을 이용하여 cDNA로 제조하였다. 제조한 cDNA를 이용해 quantitative real-time PCR을 수행하였다(cDNA 1 ㎕, primer set 각 1 ㎕, 2XSYBR green mix 12.5 ㎕, Distilled water 9.5 ㎕, 40cycle). In order to confirm the expression level of inflammatory cytokines in arthritis-induced mouse joints, joints were obtained by autopsy of mice in each group at the 16th week of monitoring of arthritis naturally occurring groups. After the joints were ground in LN 2 , 400 μl of Trizol reagent was used for RNA isolation. Chloroform was added and strongly vortexed and then incubated at room temperature for 5 minutes. After centrifugation (12500 rpm, 15 min, 4 ° C.), the supernatant was transferred to a new micro tube. After isopropanol and glycoblue in the supernatant was incubated at room temperature for 5 minutes. The supernatant was removed after centrifugation (12500 rpm, 10 min, 4 ° C.). After removing the supernatant, the remaining pellet was washed with 70% ethanol, centrifuged (9500 rpm, 5 min, 4 ℃), the pellet was air dried and dissolved in DEPC-water. Isolated RNA is QuantiTect CDNA was prepared using Reverse Transcription Kit (Qiagen). Quantitative real-time PCR was performed using the prepared cDNA (cDNA 1 μl, primer set 1 μl, 2XSYBR green mix 12.5 μl, Distilled water 9.5 μl, 40 cycles).
그룹별 측정된 결과 값(Ct 값)은 2(-ΔΔCt) method를 이용하여 wild type mice 그룹의 결과를 기준으로 하는 상대적인 값으로 계산하였다. cDNA를 template로 하여 관절염 발병 시 발현이 증가되는 것으로 알려져 있는 염증성 사이토카인 (IL-1β, IL-6, IL-17A)의 발현을 측정하였다. cDNA를 template로 하여 염증성 사이토카인(IL-1β, IL-6, IL-17A)을 증폭하기 위해 사용한 프라이머 서열이 표 5에 표시되어 있으며, 측정 결과는 도 8a에 도시되어 있다. The measured result value (Ct value) for each group was calculated as a relative value based on the results of the wild type mice group using the 2 (-ΔΔCt) method. Using cDNA as a template, the expression of inflammatory cytokines (IL-1β, IL-6, IL-17A), which are known to increase expression in the onset of arthritis, was measured. Primer sequences used to amplify inflammatory cytokines (IL-1β, IL-6, IL-17A) using cDNA as a template are shown in Table 5, and the measurement results are shown in FIG. 8A.
표 5
염증성 사이토카인 정방향 프라이머 역방향 프라이머
IL-1β 서열식별번호:38 서열식별번호:39
IL-6 서열식별번호:40 서열식별번호:41
IL-17A 서열식별번호:42 서열식별번호:43
Table 5
Inflammatory cytokines Forward primer Reverse primer
IL-1β SEQ ID NO: 38 SEQ ID NO: 39
IL-6 SEQ ID NO: 40 SEQ ID NO: 41
IL-17A SEQ ID NO: 42 SEQ ID NO: 43
관절부위 염증유발에 관여하는 염증성 사이토카인(IL-1β, IL-6)은 tIK-IL1RaKO mice의 관절에 비해 IL1RaKO mice의 관절에서 보다 높은 발현 양상을 나타냈다. 또한 연골세포와 조골세포 내 기질 생산을 억제시켜 관절 손상을 유발하여, 조직재생을 결핍시키는 주요 인자인 IL-17은 tIK-IL1RaKO 마우스 유래의 관절 조직에 현저히 낮게 발현되었다. 이러한 결과를 통해 아이케이 유래의 핵산 단편이 관절염이 유도될 때 발현하는 염증성 사이토카인의 발현을 조절할 수 있으며, 이에 따라 관절염 수치를 낮출 수 있다는 점을 확인하였다.Inflammatory cytokines (IL-1β, IL-6), which are involved in inflamed joints, showed higher expression levels in IL1RaKO mice than in tIK-IL1RaKO mice. In addition, IL-17, a major factor that inhibits tissue production in chondrocytes and osteoblasts, induces joint damage, and depletes tissue regeneration, was significantly lowered in joint tissues derived from tIK-IL1RaKO mice. These results confirm that IK-derived nucleic acid fragments can regulate the expression of inflammatory cytokines expressed when arthritis is induced, thereby lowering arthritis levels.
(2) 조직 염색을 이용한 사이토카인 발현 확인(2) Confirmation of cytokine expression using tissue staining
관절염이 유도된 마우스 관절의 병인상태를 확인하기 위하여 관절염 자연 발생 그룹의 모니터링 16주차에 각 그룹(tIK-IL-1RaKO 그룹; IL-1RaKO 그룹)의 마우스를 부검하여 관절 부위를 얻었다. 관절 부위를 10% 중성 포르말린에 고정 후 탈회 용액에 7시간 동안 침지하여 탈회하였다. 탈회된 관절 부위를 16시간에 걸쳐 수세하고, 탈수, 청명, 침투 과정을 거쳐 파라핀 블록(paraffin block)을 제조하였다. 절편 기기를 사용하여 파라핀 블록을 절단하여 7 ㎛ 절편을 만들고 슬라이드에 붙인 후 염증성 사이토카인(IL-17, IL-1β, TNFα) 염색을 진행하였다. 파라핀 절편을 deparaffinization한 후 함수 과정을 진행하였다. IL-17, IL-1β, TNFα에 대한 1차 항체를 16시간 붙인 뒤, biotin이 label되어 있는 2차 항체를 붙였다. ABC reagent로 incubation 후 peroxidase substrate solution으로 발색시킨 후 이를 수세하여 관절 synovial 부위를 중심으로 염증성 사이토카인의 발현을 확인 후 현미경으로 관찰하였다. 결과는 도 8b에 도시되어 있다. tIK-IL1RaKO 마우스 유래의 관절 조직은, IL1RaKO 마우스 유래의 관절 조직보다 synovial 부위 내 면역세포의 침윤이 덜 일어났으며, 특히 IL-17의 발현이 낮게 나타났다. 이를 통해 윤활관절부위의 염증(synovial inflammation)이 tIK-IL1RaKO 그룹에서 낮게 발생했음을 확인할 수 있었다.In order to confirm the pathogenesis of arthritis-induced mouse joints, the joint sites were obtained by autopsy of mice in each group (tIK-IL-1RaKO group; IL-1RaKO group) at week 16 of monitoring the arthritis naturally occurring group. Joint area was fixed by 10% neutral formalin and then demineralized by immersion in demineralization solution for 7 hours. The demineralized joint area was washed with water for 16 hours, and a paraffin block was prepared by dehydration, clarity, and infiltration. Paraffin blocks were cut using a slicer to make 7 μm sections, attached to slides, and stained with inflammatory cytokines (IL-17, IL-1β, TNFα). The paraffin fragment was deparaffinized and then functioned. After attaching the primary antibody against IL-17, IL-1β, and TNFα for 16 hours, the secondary antibody labeled with biotin was attached. After incubation with ABC reagent, the cells were colored with peroxidase substrate solution and washed with water to confirm the expression of inflammatory cytokines around synovial joints. The results are shown in Figure 8b. Joint tissues derived from tIK-IL1RaKO mice showed less infiltration of immune cells in the synovial site than joint tissues derived from IL1RaKO mice, and showed particularly low expression of IL-17. It was confirmed that synovial inflammation was low in the tIK-IL1RaKO group.
실시예 7 : 혈청 내 염증 사이토카인의 발현 확인Example 7 Expression of Inflammatory Cytokines in Serum
관절염이 유도된 마우스의 혈액 내 염증 사이토카인의 발현을 확인하기 위하여 관절염 자연발생 그룹의 모니터링 16주차에 각 그룹의 마우스를 부검하여 심장채혈을 통해 혈액을 얻었다. 얻은 혈액을 원심분리 (4000 rpm, 15 min, 4℃)하여 혈청만을 분리해냈고, 이를 이용하여 ELISA 방법으로 혈청 내 염증 사이토카인의 발현을 확인하였다. In order to confirm the expression of inflammatory cytokines in the blood of arthritis-induced mice, the mice of each group were autopsied at week 16 of monitoring the arthritis spontaneous generation, and the blood was obtained by heart blood collection. The obtained blood was centrifuged (4000 rpm, 15 min, 4 ° C) to separate only serum, and the expression of inflammatory cytokines in serum was confirmed by ELISA.
MCP-1 ELISA kit(R&D system)을 예로 들어 설명하면, anti-MCP-1 primary antibody가 pre-coating 되어 있는 plate에 kit에서 제공하는 standard를 serial dilution하여 넣고, serum 원액을 넣은 후 상온에서 2시간 incubation하였다. Incubation이 끝나면 well에 넣어주었던 모든 sample을 제거하고 0.05% PBST(Phosphate Buffered Saline Tween-20)로 4번 washing하였다. Well 내 anti-MCP-1 antibody에 binding한 MCP-1에 binding할 수 있는 conjugate 용액을 넣고 상온에서 2시간 incubation하였다. Incubation후에 substrate solution을 넣고 conjugate와 반응하여 발색되는 것을 모니터링 하였다. Standard의 발색 범위 내에 측정하고자 하는 serum sample의 발색정도가 들어오면 stop solution (2N H2SO4)을 넣고 ELISA reader기를 이용하여 450 nm 파장대에서 흡광도를 측정하였다. 이를 바탕으로 standard 농도에 대한 흡광도를 기준으로 하여 샘플의 흡광도가 나타내는 MCP-1의 농도를 구할 수 있었으며, MCP-1 이외의 다른 사이토카인도 위와 같은 방법을 이용하여 농도를 측정하였다. MCP-1 사이토카인을 증폭시키기 위하여 서열식별번호:44의 정방향 프라이머와, 서열식별번호:45의 역방향 프라이머를 사용하였으며, 나머지 사이토카인은 실시예 6과 동일한 프라이머를 사용하여 증폭하였다. Using the MCP-1 ELISA kit (R & D system) as an example, the standard provided by the kit is placed on a plate pre-coated with anti-MCP-1 primary antibody, and the serum stock solution is added for 2 hours at room temperature. incubation. After incubation, all the samples in the wells were removed and washed four times with 0.05% PBST (Phosphate Buffered Saline Tween-20). A conjugate solution capable of binding to MCP-1 bound to the anti-MCP-1 antibody in the well was added and incubated at room temperature for 2 hours. After incorporation, the substrate solution was added and monitored for color development by reacting with the conjugate. When the color development degree of the serum sample to be measured within the color development range of the standard was added to the stop solution (2N H 2 SO 4 ) and the absorbance was measured at 450 nm wavelength using an ELISA reader. Based on this, the concentration of MCP-1 indicating the absorbance of the sample was determined based on the absorbance of the standard concentration, and cytokines other than MCP-1 were measured using the same method as above. In order to amplify the MCP-1 cytokine, a forward primer of SEQ ID NO: 44 and a reverse primer of SEQ ID NO: 45 were used, and the remaining cytokines were amplified using the same primers as in Example 6.
분석 결과는 도 9에 도시되어 있다. 실시예 6에서 관절 부위 염증성 사이토카인의 mRNA 발현에서 나타난 결과와 유사하게, IL1RaKO mice보다 tIK-IL1RaKO mice의 혈청에서 chemokine의 일종인 MCP-1, 병인 T 세포의 분화에 관여하는 것으로 알려져 있는 IL-6, 및 관절염 발병의 주요 인자인 IL-17의 발현이 유의미하게 낮은 양상을 나타내는 것을 확인하였다. 이를 통해 tIK가 systemic한 면역반응에도 영향을 미쳐 관절염 수치를 낮추는데 기여할 수 있을 것으로 예상되었다. The analysis results are shown in FIG. Similar to the results of mRNA expression of inflammatory cytokines in the joint region, Example 6, the chemokine MCP-1, IL-known to be involved in the differentiation of pathogenic T cells in the serum of tIK-IL1RaKO mice than IL1RaKO mice 6, and the expression of IL-17, a major factor in the development of arthritis, was found to show a significantly low pattern. It is expected that tIK may also affect systemic immune responses and contribute to lower arthritis levels.
실시예 8 : 병인 T-helper 세포의 발현 측정Example 8 Measurement of Expression of Pathogenic T-helper Cells
면역 세포 중 하나인 T 세포는 면역 체계의 교란을 유발하는 중요한 세포임이 밝혀져 있다. 활성화된 T 세포는, 각각 작동 T 세포 (effector T cell) 인 Th1, Th2, Th17, regulatory T 세포로 분화한다. 어느 쪽으로의 분화 경로가 촉진되는지의 여부에 따라 질병의 발생 및 경과가 결정된다. Th1세포와 Th17 세포는 각각 IFN-γ와 IL-17을 분비하며, 당뇨, 류마티스성 관절염, 크론병과 같은 염증성 장 질환의 병인의 중심축을 이루는 것으로 알려져 있다. T cells, one of the immune cells, have been found to be important cells that cause disturbance of the immune system. Activated T cells differentiate into effector T cells, Th1, Th2, Th17 and regulatory T cells, respectively. The incidence and course of the disease depends on which way the differentiation pathway is promoted. Th1 and Th17 cells secrete IFN-γ and IL-17, respectively, and are known to form the central axis of inflammatory bowel diseases such as diabetes, rheumatoid arthritis and Crohn's disease.
본 실시예에서는 아이케이 유래의 핵산 단편에 의하여 관절염이 억제될 때, 병인 T hepler 세포의 발현 양상이 관여하는지를 확인하였다. 관절염 자연발생 그룹의 모니터링 16주차에 각 그룹의 마우스를 부검하여 비장을 얻었다. Serum free RPMI media가 담긴 100 mm petri dish에 100 ㎛ strainer를 넣고, strainer 위에 비장을 올린 후 주사기 바늘을 이용하여 비장을 찢어주면서 비장의 면역세포들이 stainer를 통해 media가 있는 dish로 빠져나갈 수 있도록 하였다. 비장을 모두 갈아낸 후, media 상에 퍼져있는 세포를 튜브에 모아 1500rpm, 4℃, 5분 동안 원심분리하여 pellet만을 얻어냈다. Pellet에 Red blood cell lysis buffer를 넣고 4℃에서 5분 동안 incubation하였다. 다시 1500rpm, 4℃, 5분 동안 원심분리하여 적혈구를 제거하여 최종적으로 비장세포를 얻어냈다. 비장세포를 FBS가 포함되어 있는 RPMI media로 부유시킨 후 1X106 개의 세포만을 취하여 병인 Th17 세포의 양을 flow cytometry로 분석하기 위한 염색 과정을 진행하였다. 그룹별로 얻은 비장세포에 PMA(100ng/ml)와 ionomycin (200ng/ml)을 넣고 37℃ 배양기에서 4시간 incubation하고, 세포 내 IL-17을 염색하기 위한 golgi stop 시약을 첨가해주었다. PMA와 ionomycin을 통해 자극된 세포를 차가운 PBS로 washing하고 병인 Th17 세포의 표면분자인 CD4에 대한 항체 (PE-Cy5 conjugated)를 넣어 4℃에서 30분간 반응시켰다. 그 후 차가운 PBS로 washing하고, 세포 내 IL-17을 염색하기 위해 세포 내로 항체가 투과할 수 있도록 세포의 permeability를 증가시킨 후 IL-17에 대한 항체 (PE conjugated)를 넣어 실온에서 30분간 반응시켰다. 반응이 모두 끝난 후에 fixative 용액을 이용하여 washing하고 최종적으로 1% paraformaldehyde 용액으로 염색된 세포를 고정시켜 flow cytometry 분석에 이용하였다. In this example, when arthritis is inhibited by the nucleic acid fragment derived from IK, it was confirmed whether the expression pattern of the pathogenic T hepler cells is involved. Monitoring of Spontaneous Arthritis Groups At week 16, mice in each group were autopsied to obtain spleens. A 100 μm strainer was placed in a 100 mm petri dish containing Serum free RPMI media, and the spleen was placed on the strainer. The spleen was then torn using a syringe needle to allow immune cells in the spleen to exit the dish containing the media through the stainer. . After removing all the spleens, the cells spread on the media were collected in a tube and centrifuged at 1500 rpm, 4 ° C for 5 minutes to obtain only pellets. Red blood cell lysis buffer was added to the pellet and incubated at 4 ° C. for 5 minutes. Again, centrifuged at 1500rpm, 4 ℃, 5 minutes to remove the red blood cells to finally obtain the splenocytes. After splenocytes were suspended in RPMI media containing FBS, only 1 × 10 6 cells were taken and stained for flow cytometry analysis of the etiology of Th17 cells. PMA (100 ng / ml) and ionomycin (200 ng / ml) were added to the splenocytes obtained by each group, and incubated for 4 hours in a 37 ° C incubator, and golgi stop reagent for staining intracellular IL-17 was added. Cells stimulated with PMA and ionomycin were washed with cold PBS and reacted for 30 minutes at 4 ° C. with an antibody against CD4 (PE-Cy5 conjugated), a surface molecule of etiological Th17 cells. After washing with cold PBS, to increase the permeability of the cells so that the antibody can penetrate the cells to stain the intracellular IL-17, the antibody against IL-17 (PE conjugated) was added and reacted at room temperature for 30 minutes. . After completion of the reaction, the cells were washed with a fixative solution and finally stained with 1% paraformaldehyde solution, and used for flow cytometry analysis.
IFN-γ를 분비하는 Th1세포는 IL1RaKO 마우스에서 6.09%로 나타났으나 tIK-IL1RaKO 마우스의 경우 3.24%로 IL1RaKO에 비해 낮은 양상을 나타냈다(도 10a). IL-17을 분비하는 Th17세포의 경우도 마찬가지로 IL1RaKO 마우스에서 1.78%로 나타났으나 tIK-IL1RaKO 마우스의 경우 1.2%로 IL1RaKO에 비해 병인 Th17세포 발현이 1.5배 감소한 양상이 나타났다(도 10b).Th1 cells secreting IFN-γ were 6.09% in IL1RaKO mice, but 3.24% in tIK-IL1RaKO mice, which was lower than that of IL1RaKO (FIG. 10A). Th17 cells secreting IL-17 were also shown to be 1.78% in IL1RaKO mice but 1.2% in tIK-IL1RaKO mice, which showed a 1.5-fold reduction in the expression of etiological Th17 cells compared to IL1RaKO (FIG. 10B).
이러한 분석 결과는, 실시예 6과 실시예 7에서 마우스의 관절 부위와 혈청에서 확인하였던 Th17 세포의 분화에 관여하는 것으로 알려져 있는 염증성 사이토카인의 발현 분석 결과와 일치한다. 따라서 관절염이 유발되었을 때, 아이케이 유래의 핵산 단편이 염증 반응에 크게 기여하는 병인 Th17세포 및 Th1세포의 발현을 억제함으로써 관절염의 염증반응을 억제할 수 있는 가능성을 확인할 수 있었다.The results of this analysis are consistent with the results of expression analysis of inflammatory cytokines known to be involved in the differentiation of Th17 cells identified in the joints and serum of mice in Examples 6 and 7. Therefore, when arthritis was induced, it was confirmed that IK-derived nucleic acid fragments could suppress the inflammatory response of arthritis by inhibiting the expression of Th17 cells and Th1 cells, which are diseases that contribute greatly to the inflammatory response.
실시예 9 : 대식세포 활성 인자 발현 분석Example 9 Macrophage Activator Factor Expression Analysis
류마티스 관절염에서 활성화된 대식세포는 염증성 사이토카인을 분비하고 다양한 면역세포들과의 상호작용을 통해 관절부위 염증 반응을 시작하고 유지한다. 대식세포는 T세포에 항원을 제시하여 T세포의 활성을 유도할 뿐 아니라, 다양한 사이토카인을 분비하여 T 세포와 함께 파골세포의 활성화도 유도한다. 대식세포는 관절염의 염증 반응 유지에 중요한 역할을 하므로, 본 실시예에서는 아이케이에 의한 관절염 억제 현상에 대식세포가 관여하는지의 여부를 확인하였다. Macrophages activated in rheumatoid arthritis secrete inflammatory cytokines and interact with various immune cells to initiate and maintain joint inflammation. Macrophages not only induce T cell activity by presenting antigens to T cells, but also induce osteoclast activation along with T cells by releasing various cytokines. Macrophages play an important role in maintaining the inflammatory response of arthritis, so in this example it was confirmed whether macrophages are involved in the arthritis inhibition phenomenon by IK.
자연발생 관절염 모니터링 16주차에 IL1RaKO 그룹 마우스와 tIK-IL1RaKO 그룹의 비장을 얻어 실시예 8과 같은 절차를 통하여 비장세포를 얻었다. 얻어진 비장세포에 대식세포 인자인 CD11b에 대한 항체(APC conjugated)와 병인대식세포 인자인 F4/80에 대한 항체 (PE-cy7 conjugated), 대식세포의 활성인자인 CD86에 대한 항체 (FITC conjugated)를 이용하여 비장세포를 염색하였다. 염색된 세포를 1% paraformaldehyde 용액으로 고정시켜 flow cytometry 분석에 이용하였다. 도 11에 도시된 것과 같이, tIK-IL1RaKO 유래의 비장세포에서는 대식세포 활성인자의 발현이 IL1RaKO에 비해 감소되어 있었다. 따라서 아이케이의 발현은 병인 Th17세포뿐만 아니라 대식세포의 활성억제에도 영향을 미치는 것을 확인할 수 있었다. Spleen cells of IL1RaKO group mice and tIK-IL1RaKO group were obtained at week 16 of spontaneous arthritis monitoring, and splenocytes were obtained by the same procedure as in Example 8. The obtained splenocytes had an antibody against macrophage factor CD11b (APC conjugated), a pathogenous macrophage factor F4 / 80 antibody (PE-cy7 conjugated), and a macrophage active factor CD86 antibody (FITC conjugated). Spleen cells were stained. Stained cells were fixed in 1% paraformaldehyde solution and used for flow cytometry analysis. As shown in Figure 11, the expression of macrophage activator in the spik cells derived from tIK-IL1RaKO was reduced compared to IL1RaKO. Therefore, it was confirmed that IK expression affects the inhibition of macrophage activity as well as etiological Th17 cells.
실시예 10 : 활성 펩타이드의 합성 및 대식세포 활성 억제 확인Example 10 Synthesis of Active Peptides and Confirmation of Inhibition of Macrophage Activity
tIK에 비하여 발현 및 정제가 용이한 짧은 길이의 아이케이 인자의 부분 단편인 활성 펩타이드를 제작하여 관절염 모델에 적용하는 경우에 기능을 발휘하는지를 확인하였다. 실시예 1을 통하여 아이케이 인자의 기능에 주요한 역할을 담당할 것으로 예상되는 2개의 아미노산(S382, Y489)을 포함하는 펩타이드를 합성하였다. S382를 포함하는 14개 아미노산으로 구성되는 펩타이드(서열식별번호:47, S 펩타이드), Y489를 포함하는 13개 아미노산으로 구성되는 펩타이드(서열식별번호:49, Y 펩타이드)를 합성하였다. S382와 Y489에는 각각 인산기로 치환될 수 있도록 합성하였다. 각각의 활성 펩타이드는 Fmoc solid phase 합성방법을 통해 합성되었다. Coupling-wash-deprotection-wash 과정을 반복하여 남아있는 시약을 제거하였으며, multi-channel automated synthesizer인 COMDEL 기기를 사용하였다. 이러한 합성을 통해 각각의 활성 펩타이드의 C-말단에서 N-말단을 향해 펩타이드를 합성하였으며, 모든 반응이 종결된 후 합성된 펩타이드를 reagent K를 이용하여 resin에서 분리하였다. 분리된 펩타이드를 차가운 디에틸에테르를 첨가하여 침전시키고, 다시 한 번 디에틸에테르를 이용하셔 세척한 뒤 vacuum 상태에서 건조하였다. mass spectrometer를 사용하여 합성된 펩타이드의 분자량을 측정하였으며, 예상한 분자량과 동일한 경우 정제를 진행하였다. Reverse-HPLC, C18 column, 220nm wavelength 정제조건에서 정제를 진행하였다.  Compared to tIK, the active peptide, which is a partial fragment of the short-length IK factor, which is easier to express and purify, was prepared and confirmed to function when applied to the arthritis model. Through Example 1 a peptide comprising two amino acids (S382, Y489) expected to play a major role in the function of the IK factor was synthesized. A peptide consisting of 14 amino acids including S382 (SEQ ID NO: 47, S peptide), and a peptide consisting of 13 amino acids including Y489 (SEQ ID NO: 49, Y peptide) were synthesized. S382 and Y489 were synthesized to be substituted with phosphoric acid groups, respectively. Each active peptide was synthesized by Fmoc solid phase synthesis. The remaining reagents were removed by repeating the coupling-wash-deprotection-wash procedure, and a multi-channel automated synthesizer, a COMDEL instrument, was used. Through this synthesis, peptides were synthesized from the C-terminus to the N-terminus of each active peptide. After all reactions were completed, the synthesized peptide was separated from the resin using reagent K. The separated peptide was precipitated by addition of cold diethyl ether, and once again washed with diethyl ether and dried under vacuum. The molecular weight of the synthesized peptide was measured using a mass spectrometer, and purification was performed when the molecular weight was the same as the expected molecular weight. Purification was performed under reverse-HPLC, C18 column, and 220nm wavelength purification conditions.
정제된 S 펩타이드와 Y 펩타이드를 리포다당(lipopolysaccharide, LPS)의 자극 조건에서 대식세포의 활성화 정도를 확인하였다. J774a.1 대식세포주에 S 펩타이드와 Y 펩타이드를 각각 500 ng/mL의 농도로 동시에 transfection 후 LPS (O111:B4)를 처리하여 염증성 사이토카인의 발현을 확인하였다. LPS 처리 후 24시간째에 얻은 대식세포에서, 활성 펩타이드를 처리한 그룹에서 염증성 사이토카인(TNF-α, IL-6)의 mRNA 발현이 낮게 나타났다(도 12a). 세포 배양액을 이용한 측정에서도 tIK active peptide가 주어진 조건에서 대조군에 비해 염증성 사이토카인이 낮게 발현되었다(도 12b). 따라서 활성 펩타이드가 관절염과 관련된 염증성 사이토카인의 발현을 억제한다는 점을 확인하였다. Purified S peptide and Y peptide were confirmed the degree of macrophage activation under the conditions of stimulation of lipopolysaccharide (lipspolysaccharide, LPS). In the J774a.1 macrophage line, S and Y peptides were simultaneously transfected at a concentration of 500 ng / mL and treated with LPS (O111: B4) to confirm the expression of inflammatory cytokines. In macrophages obtained 24 hours after LPS treatment, mRNA expression of inflammatory cytokines (TNF-α, IL-6) was low in the group treated with the active peptide (FIG. 12A). In the cell culture solution, inflammatory cytokines were expressed lower than that of the control group in the condition of tIK active peptide (FIG. 12B). Therefore, it was confirmed that the active peptide inhibits the expression of inflammatory cytokines associated with arthritis.
실시예 11 : 활성 펩타이드에 의한 병인 T helper 세포 분화 억제 확인Example 11: Confirmation of inhibition of T helper cell differentiation of etiology by active peptide
아이케이 인자 활성 펩타이드가 병인 T helper 세포의 분화에 미치는 영향을 확인하기 위하여, 야생형 마우스의 비장을 이용하여 실시예 6에서 제시된 방법을 통하여 비장세포를 얻었다. 비장세포에서 naive CD4 T 세포만을 선택적으로 분리하고, S 펩타이드와 Y 펩타이드(100 ng/mL or 500 ng/mL)를 1시간 동안 전-처리하였다. 병인 T helper 세포로 분화할 수 있는 각종 사이토카인 및 중화항체를 포함하는 배양액을 첨가해주었으며, condition 배양액을 첨가할 때 다시 S 펩타이드와 Y 펩타이드를 동일한 농도(100 ng/mL or 500 ng/mL)로 첨가하였다. Th0 세포로의 분화를 위해 anti-CD3, anti-CD28, IL-2를 첨가한 배양액을 세포에 첨가하였다. Th1 세포로의 분화를 위해 anti-CD3, anti-CD28, IL-2, IL-12, anti-IL-4를 첨가한 배양액을 첨가해주었다. 또한, Th17 세포로의 분화를 위해서는 anti-CD3, anti-CD28, IL-6, TGF-β, anti-IL-4, anti-IFNγ를 첨가한 배양액을 세포에 첨가해주었다. 배양 2일 후 같은 조건으로 T helper 세포로 분화할 수 있도록 각종 사이토카인과 중화항체가 포함된 배양액을 첨가해준 후 2일 후에 병인 T helper 세포를 flow cytometry로 분석하기 위하여 염색하였다. In order to confirm the effect of the IK factor active peptide on the differentiation of pathogenic T helper cells, splenocytes were obtained by the method shown in Example 6 using spleens of wild-type mice. Only naive CD4 T cells were selectively isolated from splenocytes, and pretreated with S peptide and Y peptide (100 ng / mL or 500 ng / mL) for 1 hour. A culture medium containing various cytokines and neutralizing antibodies capable of differentiating into etiological T helper cells was added, and when the condition medium was added, the same concentration of S peptide and Y peptide was again obtained (100 ng / mL or 500 ng / mL). Was added. For differentiation into Th0 cells, culture medium containing anti-CD3, anti-CD28, IL-2 was added to the cells. For the differentiation into Th1 cells, the culture medium containing anti-CD3, anti-CD28, IL-2, IL-12 and anti-IL-4 was added. In addition, for the differentiation into Th17 cells, culture medium containing anti-CD3, anti-CD28, IL-6, TGF-β, anti-IL-4, and anti-IFNγ was added to the cells. After 2 days of culture, the culture medium containing various cytokines and neutralizing antibodies was added to differentiate into T helper cells under the same conditions. After 2 days, the pathogenic T helper cells were stained for flow cytometry analysis.
먼저 배양된 세포에 phorbol 12-myristate 13-acetate (PMA, 50 ng/mL)와 ionomycin(200 ng/mL)을 첨가하였다. 37℃ 배양기에서 4시간 incubation하면서, 세포 내 IL-17을 염색하기 위한 golgi stop (monensin)을 첨가하였다. Incubation 후 세포를 차가운 PBS로 세척하였다. 세포 내 cytokine (IL-17, IFNγ)을 염색하기 위해 세포 내로 항체가 투과할 수 있도록 세포의 투과도를 증가시킨 후 IL-17, IFNγ에 대한 항체를 넣고 ice에서 30분간 반응시켰다. 반응 후 fixative 용액을 이용하여 세척하고, 최종적으로 1% paraformaldehyde 용액으로 염색된 세포를 고정시켜 flow cytometry 분석에 이용하였다. First, phorbol 12-myristate 13-acetate (PMA, 50 ng / mL) and ionomycin (200 ng / mL) were added to the cultured cells. While incubating for 4 hours in a 37 ° C. incubator, golgi stop (monensin) was added to stain intracellular IL-17. After incubation the cells were washed with cold PBS. In order to stain intracellular cytokine (IL-17, IFNγ), the cell permeability was increased to allow the antibody to penetrate into the cell, and the antibody against IL-17 and IFNγ was added and reacted on ice for 30 minutes. After the reaction, the cells were washed with a fixative solution, and finally, cells stained with 1% paraformaldehyde solution were used for flow cytometry analysis.
도 13a 및 도 13b에 도시된 것과 같이, 활성 펩타이드를 처리하면, Th0 세포 분화 조건 및 Th1 세포의 분화조건에서 CD4+IFNγ+T 세포의 분화가 감소하는 경향을 나타냈다. 또한 도 13c에 도시된 것과 같이, 활성 펩타이드의 처리에 의하여 CD4+IL-17+T 세포의 분화가 억제되었다. 이러한 결과는 아이케이 인자의 활성 부위 아미노산을 가지는 짧은 활성 펩타이드가 염증 질환에서의 병인 T helper 세포로 작용하는 Th1 세포, Th17 세포로의 분화를 억제할 수 있음을 의미한다. As shown in FIGS. 13A and 13B, the treatment of the active peptide showed a tendency to decrease the differentiation of CD4 + IFNγ + T cells under Th0 cell differentiation conditions and Th1 cell differentiation conditions. In addition, as shown in Figure 13c, the differentiation of CD4 + IL-17 + T cells was inhibited by the treatment of the active peptide. These results indicate that the short active peptide having the active site amino acid of IK factor can inhibit the differentiation into Th1 cells and Th17 cells, which act as T helper cells, which are pathogenesis in inflammatory diseases.
실시예 12 : 활성 펩타이드의 주입을 통한 관절염 억제Example 12 Inhibition of Arthritis by Infusion of Active Peptides
아이케이 인자의 활성 펩타이드인 S 펩타이드와 Y 펩타이드의 관절염 억제 효과를 알아보기 위하여, 이들 펩타이드를 자연 발생 관절염 모델인 IL1RaKO 마우스에 주입하였다. 각각의 그룹에 7마리의 IL1RaKO 마우스를 이용하였으며, 활성 펩타이드로서 S 펩타이드와 Y 펩타이드를 동시에 10 mg/kg (S peptide 5 mg/kg, Y peptide 5 mg/kg)의 농도로 복강 주사하여 주입하였다. 대조군에는 동일한 부피의 인산환충식염수(phosphate buffered saline, PBS)를 복강 주사하였다. 펩타이드는 2~3 일 간격으로 주입하였으며, Peptide 주입 시마다 마우스의 몸무게, 관절염 발병 여부 및 지수를 기록하였다. 펩타이드 주입 후 7주째 마우스의 관절염 지수는 PBS를 주입한 대조군에 비하여 낮은 수치를 나타냈으며(도 14a), 펩타이드 주입 그룹에서 관절염 발병률이 낮았다(도 14b).In order to examine the arthritis inhibitory effects of S peptides and Y peptides, active peptides of IK factor, these peptides were injected into IL1RaKO mice, a model of naturally occurring arthritis. Seven IL1RaKO mice were used in each group, and S peptide and Y peptide were simultaneously intraperitoneally injected at a concentration of 10 mg / kg (S peptide 5 mg / kg, Y peptide 5 mg / kg) as active peptides. . The control group was intraperitoneally injected with the same volume of phosphate buffered saline (PBS). Peptides were injected at intervals of 2 to 3 days, and the weight of mice, the incidence of arthritis, and the index were recorded for each Peptide injection. Arthritis index of mice 7 weeks after the peptide injection was lower than the control group injected with PBS (Fig. 14a), the incidence of arthritis in the peptide injection group (Fig. 14b).
실시예 13 : 활성 펩타이드 주입후 관절염 모델에서 병인 세포 분석Example 13: Pathogenesis Cell Analysis in Arthritis Models After Active Peptide Injection
(1) flow cytometry 분석(1) flow cytometry analysis
아이케이 인자의 활성 펩타이드 주입에 의하여, 관절염 염증 반응의 시작과 유지에 관여하는 주요 인자인 Th17세포의 분포를 측정하였다. 실시예 12에 따라, 활성 펩타이드 주입 8주차에 마우스를 부검하여 그룹별 마우스의 비장을 얻었다. 비장을 갈아 비장세포를 얻은 후 PMA(50 ng/mL)와 ionomycin (200 ng/mL)을 첨가하였다. 37℃ 배양기에서 4시간 incubation하면서, 세포 내 IL-17을 염색하기 위한 golgi stop (monensin)을 첨가하였다. Incubation 후 세포를 차가운 PBS로 세척하고 병인 Th17 세포의 표면분자인 CD4에 대한 항체(APC conjugated)를 넣어 4℃에서 30분간 반응시켰다. 반응이 완료되면, 세포를 세척하고, 세포 내 IL-17을 염색하기 위해 세포 내로 항체가 투과할 수 있도록 세포의 투과도를 증가시킨 후 IL-17에 대한 항체(PE conjugated)를 넣고 실온에서 30분간 반응시켰다. 반응시킨 후 fixative 용액을 이용하여 세척하고 최종적으로 1% paraformaldehyde 용액으로 염색된 세포를 고정시켜 flow cytometry 분석에 이용하였다. By injecting the active peptide of IK factor, the distribution of Th17 cells, a major factor involved in the initiation and maintenance of arthritis inflammatory response, was measured. According to Example 12, mice were necropsied at week 8 of the active peptide injection to obtain spleens of mice in each group. The spleens were ground to obtain splenocytes, and PMA (50 ng / mL) and ionomycin (200 ng / mL) were added. While incubating for 4 hours in a 37 ° C. incubator, golgi stop (monensin) was added to stain intracellular IL-17. After incubation, the cells were washed with cold PBS, and an antibody (APC conjugated) to CD4, a surface molecule of etiological Th17 cells, was added and reacted at 4 ° C. for 30 minutes. When the reaction is complete, the cells are washed, the cell permeability is increased to allow the antibody to penetrate into the cells to stain the IL-17, and then the antibody to the IL-17 (PE conjugated) is added and the reaction is performed at room temperature for 30 minutes. Reacted. After the reaction, the cells were washed with a fixative solution and finally stained with 1% paraformaldehyde solution, and used for flow cytometry analysis.
펩타이드 주입 그룹과 PBS 주입 그룹에서 관절염이 유발된 마우스의 비장세포 내에서 병인 Th17세포의 분석결과를 보면 활성 펩타이드를 주입한 IL1RaKO의 비장에서 Th17 세포가 적게 나타남을 알 수 있다(도 15a). 이는 아이케이 인자의 활성 펩타이드가 관절염 병인에 주요한 역할을 하는 Th17세포의 분화 및 증식을 억제하는 효과를 갖는다는 것을 의미한다. As a result of analysis of etiological Th17 cells in splenocytes of arthritis-induced mice in the peptide injection group and the PBS injection group, it can be seen that fewer Th17 cells appear in the spleen of IL1RaKO injected with the active peptide (FIG. 15A). This means that the active peptide of IK factor has the effect of inhibiting the differentiation and proliferation of Th17 cells, which play a major role in the arthritis etiology.
(2) 조직 염색을 이용한 사이토카인 발현 확인(2) Confirmation of cytokine expression using tissue staining
아이케이 활성 펩타이드 주입 후, 관절 synovial부위에서의 염증성 사이토카인의 발현을 확인하였다. 실시예 12에 따라, 활성 펩타이드 주입 8주차에 마우스를 부검하여 관절 부위를 얻었다. 관절 부위를 10% neutral formalin에 고정하고 탈회 용액에 7시간 동안 침지하여 탈회하였다. 탈회된 관절 부위를 16시간에 걸쳐 수세하고, 탈수, 청명, 침투 과정을 거쳐 파라핀 블록(paraffin block)을 제조하였다. 절편 기기를 사용하여 파라핀 블록을 절단하여 7 ㎛ 절편을 만들고 슬라이드에 붙인 후 염증성 사이토카인(IL-17, IL-1β, TNFα) 염색을 진행하였다. 파라핀 절편을 deparaffinization한 후 함수 과정을 진행하였다. 여기에 IL-17, IL-1β, TNFα에 대한 1차 항체를 16시간 붙인 후 biotin이 label되어 있는 2차 항체를 붙였다. ABC reagent로 incubation 후 peroxidase substrate solution으로 발색시킨 후 이를 수세하여 관절 synovial 부위를 중심으로 염증성 사이토카인의 발현을 확인 후 현미경으로 관찰하였다. 결과는 도 15b에 도시되어 있다. 활성 펩타이드를 주입한 마우스의 경우 관절 부위의 염증성 사이토카인의 발현이 낮고, 염증세포의 침윤이 현저히 감소되어 있는 것을 확인하였다.After injection of IK active peptide, the expression of inflammatory cytokines in the synovial area of the joint was confirmed. According to Example 12, mice were autopsied at week 8 of the active peptide injection to obtain joint sites. Joint area was fixed by 10% neutral formalin and demineralized by soaking in demineralized solution for 7 hours. The demineralized joint area was washed with water for 16 hours, and a paraffin block was prepared by dehydration, clarity, and infiltration. Paraffin blocks were cut using a slicer to make 7 μm sections, attached to slides, and stained with inflammatory cytokines (IL-17, IL-1β, TNFα). The paraffin fragment was deparaffinized and then functioned. After attaching the primary antibody against IL-17, IL-1β, and TNFα for 16 hours, the secondary antibody labeled with biotin was attached. After incubation with ABC reagent, the cells were colored with peroxidase substrate solution and washed with water to confirm the expression of inflammatory cytokines around synovial joints. The results are shown in FIG. 15B. In the case of mice injected with the active peptide, it was confirmed that the expression of inflammatory cytokines in the joint region was low and the infiltration of inflammatory cells was significantly reduced.
실시예 14 : Baculovirus expression system을 이용한 tIK 발현 및 정제Example 14 tIK Expression and Purification Using a Baculovirus Expression System
아이케이 단편(tIK) 단백질의 대량 발현을 위해 Fc가 tagging되어 있는 tIK 플라스미드를 baculovirus에 transfection한 후, 곤충세포(SF9 세포)에서 발현하였다. tIK 서열의 3' 말단으로 thrombin 인식 부위를 코딩하는 핵산(서열식별번호:50)과 Fc 태그 서열(서열식별번호:51)을 연결하였다. Virus seed를 만든 후, 1차(100 mm dish scale), 2차(T75 flask scale), 3차(1L 삼각플라스크)에 걸쳐 점차 많은 수의 곤충세포에 tIK-Fc를 발현하는 baculovirus를 infection하였다. 최종적으로 3차 바이러스 infection 후 3일차에 tIK-Fc가 secretion 되어 있는 1L 부피의 세포 배양액을 얻었다. 세포 배양액 내에 존재하는 tIK-Fc 단백질은 immunoprecipitation방법을 이용하여 정제하였다. Sepharose column에 rProtein A resin을 넣고 배양액을 주입하면, 배양액 내 함유된 tIK-Fc 단백질의 Fc 부분이 resin과 결합함으로써 column에 남게 되는 특성을 이용하였다. tIK-Fc 단백질의 발현(약 55 kDa)은 Commassie blue 염색법을 통해 일차적으로 확인하였다(결과 미도시). 순수한 tIK-Fc 단백질을 얻기 위해 rProtein A와 Fc 영역 사이의 비-공유결합을 끊어서 resin으로부터 tIK와 Fc region 결합체를 분리하고자 하였다. 비-공유결합을 끊기 위해서 Elute buffer (0.1M Glycine buffer, pH2.8)를 이용하여 용출시킨 뒤 Neutralization buffer (1M Tris-HCl, pH8.0)로 Neutralization과정을 거쳐 단백질의 구조 변성을 방지하였다. Column 세척 과정은 washing buffer (0.15M NaCl-20mM Na2HPO4, pH8.0)를 이용하였다. Affinity chromatography를 통해 총 7개의 fraction으로 분리된 용액을 SDS-PAGE를 통해 확인한 결과, 단백질이 용출된 것을 확인 할 수 있었다. 단백질의 크기를 확인한 결과 tIK-Fc protein이 결합된 크기인 55 kDa을 확인하였다. 이를 통해 Fc region과 fusion protein이 결합체를 분리할 수 있음을 확인하였다(도 16).For mass expression of IK fragment (tIK) protein, the tIK plasmid tagged with Fc was transfected into baculovirus and then expressed in insect cells (SF9 cells). A nucleic acid (SEQ ID NO: 50) encoding a thrombin recognition site was linked to the 3 ′ end of the tIK sequence and an Fc tag sequence (SEQ ID NO: 51). After the virus seed was produced, baculovirus expressing tIK-Fc was gradually infected to a larger number of insect cells over the first (100 mm dish scale), the second (T75 flask scale), and the third (1 L conical flask). Finally, 3 days after tertiary virus infection, 1 L volume of cell culture containing secreted tIK-Fc was obtained. TIK-Fc protein present in the cell culture was purified using immunoprecipitation method. When rProtein A resin was added to the Sepharose column and the culture solution was injected, the Fc portion of the tIK-Fc protein contained in the culture medium was combined with the resin, thereby remaining in the column. Expression of tIK-Fc protein (approximately 55 kDa) was primarily confirmed by Commassie blue staining (result not shown). To obtain the pure tIK-Fc protein, we tried to isolate the tIK and Fc region conjugate from the resin by breaking the non-covalent bond between rProtein A and Fc region. In order to break the non-covalent bond, the protein was eluted with Elute buffer (0.1M Glycine buffer, pH2.8) and then neutralized with Neutralization buffer (1M Tris-HCl, pH8.0) to prevent protein denaturation. Column washing was performed using washing buffer (0.15M NaCl-20mM Na 2 HPO 4 , pH8.0). As a result of confirming the solution separated into a total of seven fractions through affinity chromatography through SDS-PAGE, the protein was eluted. As a result of checking the size of the protein, 55 kDa, which is the size of the tIK-Fc protein, was bound. This confirmed that the Fc region and the fusion protein can separate the conjugate (FIG. 16).
실시예 15 : tIK 유전자 전달을 위한 재조합 아데노-연관바이러스(rAAV) 벡터 시스템 확립Example 15: Establishment of a Recombinant Adeno-associated Virus (rAAV) Vector System for tIK Gene Delivery
아데노-연관바이러스(AAV)는 레트로 바이러스와는 달리 질병을 일으키지 않고, 인체세포의 제 19번 염색체에 선택적으로 통합되므로 만성질환의 유전자 치료벡터로 활용되고 있다. 이러한 특성을 이용하여 본 실시예에서는 서열식별번호:3으로 표시되는 tIK 핵산 단편을 AAV의 inverted terminal repeat(ITR)로 둘러싼 형태로 만드는 유전자 클로닝 단계를 거쳐, tIK 핵산 단편을 세포 내로 전달할 수 있는 rAAV2-tIK 벡터 시스템을 확립하였다. GFP 발현이 가능한 rAAV viral vector인 pSP72-XP7-scAAV2-CMV-GFP를 발현 vector로 이용하였다. tIK 핵산 단편의 5' 말단에 HA 태그 서열(서열식별번호:54)을 연결하고, tIK 핵산 단편의 양쪽 말단에 상보적인 프라이머(정방향 프라이머는 서열식별번호:55; 역방향 프라이머는 서열식별번호:56)를 이용하여 PCR을 진행하였다. 이를 통해 얻은 증폭 절편의 말단을 제한효소로 자른 뒤, 발현 vector에 삽입하여 tIK-HA-sp72-XP7-scAAV2 재조합 발현 벡터를 제조하였다. 제조된 재조합 발현 벡터를 대장균에 transformation한 뒤, tIK 증폭 절편이 삽입된 플라스미드를 선택할 수 있었다. 아데노-연관바이러스에 연결된 tIK 유전자를 폴리에틸렌이민(polyethylenimine, PEI)을 이용한 transfection으로 293T 세포에 주입하였다. PEI를 이용하여 tIK를 발현하는 것으로 확인된 바이러스를 다시 293T 세포에 감염시키고 웨스턴 블롯팅을 이용하여 발현을 확인하였다(도 17). Adeno-associated viruses (AAV), unlike retroviruses, do not cause disease and are selectively integrated into chromosome 19 of human cells, and thus are used as gene therapy vectors for chronic diseases. Using this property, in the present embodiment, a rAAV2 capable of delivering a tIK nucleic acid fragment into a cell is subjected to a gene cloning step in which the tIK nucleic acid fragment represented by SEQ ID NO: 3 is surrounded by an inverted terminal repeat (ITR) form of AAV. -tIK vector system was established. PSP72-XP7-scAAV2-CMV-GFP, a rAAV viral vector capable of GFP expression, was used as an expression vector. a HA tag sequence (SEQ ID NO: 54) is linked to the 5 'end of the tIK nucleic acid fragment, and a primer complementary to both ends of the tIK nucleic acid fragment (forward primer is SEQ ID NO: 55; the reverse primer is SEQ ID NO: 56 PCR was performed using). The ends of the amplified fragments thus obtained were cut with restriction enzymes and inserted into expression vectors to prepare tIK-HA-sp72-XP7-scAAV2 recombinant expression vectors. After transforming the prepared recombinant expression vector into Escherichia coli, the plasmid into which the tIK amplification fragment was inserted could be selected. The tIK gene linked to adeno-associated virus was injected into 293T cells by transfection with polyethylenimine (PEI). Viruses that were found to express tIK using PEI were again infected with 293T cells and confirmed by Western blotting (FIG. 17).
실시예 16 : AAV2-tIK 주입을 통한 관절염 억제 확인Example 16: Confirmation of Arthritis Inhibition through AAV2-tIK Injection
실시예 15에서 제작한 rAAV2-tIK를 in vivo 질환 모델에 적용하여, tIK 핵산의 발현에 따른 관절염의 치료 효과를 확인하였다. AAV2-tIK와 AAV2-GFP 바이러스를 각각 1 x 1011 vg/100 ul의 농도로 자연발생 관절염을 나타내는 IL1RaKO의 꼬리를 통해 정맥 주사하였다. 2주 간격으로 rAAV2-tIK와 rAAV-GFP를 주입하였고, 일주일에 1회 마우스 관절상태와 발병률을 측정하였다. rAAV2-tIK를 주입한 IL1RaKO에서 rAAV2-GFP를 주입한 IL1RaKO보다 낮은 관절염 발병률과 관절염 지수를 나타냈으며(도 18a), 관절염의 발병율이 낮았다(도 18b). 이를 통해 virus vector 시스템을 이용하여 아이케이 핵산을 전달하여, 관절염의 발병률 및 관절염 지수를 낮출 수 있다는 가능성을 확인하였다. RAAV2-tIK prepared in Example 15 was applied to an in vivo disease model to confirm the therapeutic effect of arthritis according to the expression of tIK nucleic acid. AAV2-tIK and AAV2-GFP viruses were injected intravenously through the tail of IL1RaKO, a naturally occurring arthritis at a concentration of 1 × 10 11 vg / 100 ul, respectively. RAAV2-tIK and rAAV-GFP were injected at two week intervals, and once a week mouse joint condition and incidence were measured. IL1RaKO injected with rAAV2-tIK showed lower arthritis incidence and arthritis index than IL1RaKO injected with rAAV2-GFP (FIG. 18A), and the incidence of arthritis was low (FIG. 18B). Through this, IK nucleic acid was delivered using a virus vector system, which confirmed the possibility of lowering the incidence of arthritis and arthritis index.
실시예 17 : CHO cell line에서 tIK 발현 시스템 구축Example 17 tIK expression system construction in CHO cell line
Chinese hamster ovary (CHO) 세포는 유전자 주입 시 그 발현 효율이 높으며, 유전자 재조합 단백질 의약품 생산용 동물 세포주로 많이 이용되고 있다. CHO cell에서의 tIK 발현 효율을 확인하기 위하여, 5' 말단에 HA 태그 서열(서열식별번호:54)이 연결되어 있는 tIK 플라스미드(tIK-PcDNA3.1)를 CHO 세포에 주입하였다. tIK의 증폭을 위해서 서열식별번호:55의 정방향 프라이머와 서열식별번호:57의 역방향 프라이머를 사용하였다. CHO 세포에서 tIK의 발현을 웨스턴 블롯팅으로 확인하였다(도 19). Chinese hamster ovary (CHO) cells have high expression efficiency during gene injection and are widely used as animal cell lines for producing recombinant protein pharmaceutical products. To confirm the efficiency of tIK expression in CHO cells, tIK plasmids (tIK-PcDNA3.1) having a HA tag sequence (SEQ ID NO: 54) linked to the 5 'end were injected into CHO cells. For amplification of tIK, a forward primer of SEQ ID NO: 55 and a reverse primer of SEQ ID NO: 57 were used. Expression of tIK in CHO cells was confirmed by western blotting (FIG. 19).

Claims (20)

  1. 약학적 유효량의 아이케이 인자(IK factor) 또는 아이케이 인자의 단편을 함유하는 관절염을 치료 또는 예방하는 약학적 조성물.A pharmaceutical composition for treating or preventing arthritis containing a pharmaceutically effective amount of an IK factor or fragment of an IK factor.
  2. 제 1항에 있어서, 상기 아이케이 인자의 단편은 서열식별번호:4의 아미노산 서열을 가지는 펩타이드 또는 서열식별번호:4의 부분 펩타이드를 포함하는 약학적 조성물. The pharmaceutical composition of claim 1, wherein the fragment of IK factor comprises a peptide having an amino acid sequence of SEQ ID NO: 4 or a partial peptide of SEQ ID NO: 4.
  3. 제 1항에 있어서, 상기 아이케이 인자의 단편은 서열식별번호:2의 아미노산 서열의 382번째 세린, 489번째 티로신 및 492번째 티로신 중에서 선택되는 적어도 하나의 아미노산 잔기 및 이들 선택된 아미노산 잔기의 인접 아미노산으로 구성되는 10개 이상의 아미노산을 가지는 펩타이드로 구성되는 약학적 조성물.The fragment of claim 1, wherein the fragment of the IK factor comprises at least one amino acid residue selected from 382 th serine, 489 tyrosine, and 492 tyrosine of the amino acid sequence of SEQ ID NO: 2, and a contiguous amino acid of these selected amino acid residues. Pharmaceutical composition consisting of a peptide having at least 10 amino acids consisting of.
  4. 제 3항에 있어서, 상기 아이케이 인자의 단편은 서열식별번호:2의 아미노산 서열의 382번째 세린, 489번째 티로신 및 492번째 티로신으로 구성되는 적어도 하나의 아미노산은 인산화되어 있는 것을 특징으로 하는 약학적 조성물.4. The pharmaceutical composition according to claim 3, wherein the fragment of IK factor is phosphorylated at least one amino acid consisting of 382 th serine, 489 th tyrosine and 492 th tyrosine of the amino acid sequence of SEQ ID NO: 2. Composition.
  5. 제 3항에 있어서, 상기 아이케이 인자의 단편은 서열식별번호:2의 아미노산 서열을 가지는 펩타이드의 단편으로서, 서열식별번호:2의 아미노산 서열의 382번째 세린 및 489번째 티로신 중에서 선택되는 적어도 하나의 아미노산 잔기 및 이들 선택된 아미노산 잔기의 인접 아미노산으로 구성되는 10개 이상의 아미노산을 가지는 펩타이드로 구성되는 약학적 조성물. The fragment of claim 3, wherein the fragment of the IK factor is a fragment of a peptide having an amino acid sequence of SEQ ID NO: 2, wherein at least one selected from 382 th serine and 489 th tyrosine of the amino acid sequence of SEQ ID NO: 2. A pharmaceutical composition consisting of a peptide having at least 10 amino acids consisting of amino acid residues and contiguous amino acids of these selected amino acid residues.
  6. 제 5항에 있어서, 상기 아이케이 인자의 단편은 상기 선택된 아미노산의 N-말단과 C-말단으로 각각 2개 이상의 인접 아미노산이 연결되어 있는 10개 이상의 아미노산을 가지는 펩타이드로 구성되는 약학적 조성물. The pharmaceutical composition according to claim 5, wherein the fragment of the IK factor is composed of a peptide having 10 or more amino acids each having two or more adjacent amino acids linked to the N-terminus and C-terminus of the selected amino acid.
  7. 제 1항에 있어서, 상기 아이케이 인자의 단편은 서열식별번호:4, 서열식별번호:47 및 서열식별번호:49로 구성되는 군에서 적어도 하나 선택되는 아미노산 서열을 가지는 펩타이드로 구성되는 약학적 조성물. The pharmaceutical composition of claim 1, wherein the fragment of IK factor is composed of a peptide having an amino acid sequence selected from at least one group consisting of SEQ ID NO: 4, SEQ ID NO: 47, and SEQ ID NO: 49. .
  8. 제 1항에 있어서, 상기 약학적 조성물은 류마티스성 관절염을 치료하는데 사용되는 약학적 조성물. The pharmaceutical composition of claim 1, wherein the pharmaceutical composition is used to treat rheumatoid arthritis.
  9. 제 1항에 있어서, 상기 아이케이 인자 또는 상기 아이케이 인자의 단편은 약학적 조성물 중에 1.0 ng/mL ~ 10 ㎍/mL의 농도로 함유되어 있는 약학적 조성물. The pharmaceutical composition of claim 1, wherein the IK factor or fragment of IK factor is contained in a pharmaceutical composition at a concentration of 1.0 ng / mL to 10 μg / mL.
  10. 아이케이 인자(IK factor) 또는 아이케이 인자의 단편을 코딩하는 핵산 분자를 포함하는 유전자 전달체를 함유하는, 관절염을 치료 또는 예방하는 약학적 조성물. A pharmaceutical composition for treating or preventing arthritis, comprising a gene carrier comprising a nucleic acid molecule encoding an IK factor or fragment of an IK factor.
  11. 제 10항에 있어서, 상기 아이케이 인자의 단편을 코딩하는 핵산은 서열식별번호:3의 뉴클레오티드 서열을 가지는 핵산 또는 서열식별번호:3의 부분 핵산을 포함하는 약학적 조성물. The pharmaceutical composition of claim 10, wherein the nucleic acid encoding the fragment of IK factor comprises a nucleic acid having a nucleotide sequence of SEQ ID NO: 3 or a partial nucleic acid of SEQ ID NO: 3.
  12. 제 10항에 있어서, 상기 아이케이 인자의 단편을 코딩하는 핵산은 서열식별번호:1의 뉴클레오티드 서열의 1144-1146번의 뉴클레오티드 잔기, 1465-1467번째 뉴클레오티드 잔기 및 1474-1476번째 뉴클레오티드 잔기 중에서 선택되는 적어도 하나의 뉴클레오티드 잔기와, 이들 선택된 뉴클레오티드 잔기의 인접 뉴클레오티드로 구성되는 30개 이상의 뉴클레오티드를 가지는 핵산으로 구성되는 약학적 조성물. The nucleic acid encoding a fragment of the IK factor of claim 10, wherein the nucleic acid encoding a fragment of the IK factor is at least one selected from nucleotide residues 1144-1146, 1465-1467 nucleotide residues, and 1474-1476 nucleotide residues of the nucleotide sequence of SEQ ID NO: 1. A pharmaceutical composition consisting of a nucleic acid having one nucleotide residue and at least 30 nucleotides consisting of adjacent nucleotides of these selected nucleotide residues.
  13. 제 12항에 있어서, 상기 아이케이 인자의 단편을 코딩하는 핵산은 서열식별번호:1의 뉴클레오티드 서열의 1144-1146번의 뉴클레오티드 잔기 및 1465-1467번째 뉴클레오티드 잔기 중에서 선택되는 적어도 하나의 뉴클레오티드 잔기와, 이들 선택된 뉴클레오티드 잔기의 인접 뉴클레오티드로 구성되는 30개 이상의 뉴클레오티드를 가지는 핵산으로 구성되는 약학적 조성물.13. The nucleic acid encoding a fragment of the IK factor according to claim 12, wherein the nucleic acid encoding the fragment of IK factor comprises at least one nucleotide residue selected from nucleotide residues 1144-1146 and 1465-1467 nucleotide residues of the nucleotide sequence of SEQ ID NO: 1; A pharmaceutical composition consisting of a nucleic acid having at least 30 nucleotides consisting of contiguous nucleotides of a selected nucleotide residue.
  14. 제 13항에 있어서, 상기 아이케이 인자의 단편을 코딩하는 핵산은 상기 선택된 뉴클레오티드 잔기의 5' 말단과 3' 말단으로 각각 6개 이상의 인접 뉴클레오티드가 연결되어 있는 30개 이상의 뉴클레오티드를 가지는 핵산으로 구성되는 약학적 조성물. The nucleic acid encoding a fragment of the IK factor is composed of nucleic acids having 30 or more nucleotides each having at least 6 adjacent nucleotides linked to the 5 'and 3' ends of the selected nucleotide residue. Pharmaceutical compositions.
  15. 제 10항에 있어서, 상기 아이케이 인자의 단편을 코딩하는 핵산은 서열식별번호:4, 서열식별번호:47 및 서열식별번호:49로 구성되는 군에서 적어도 하나 선택되는 아미노산 서열을 가지는 펩타이드를 코딩하는 뉴클레오티드 서열을 갖는 핵산으로 구성되는 약학적 조성물. The method of claim 10, wherein the nucleic acid encoding a fragment of the IK factor encodes a peptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 4, SEQ ID NO: 47 and SEQ ID NO: 49 Pharmaceutical composition consisting of a nucleic acid having a nucleotide sequence.
  16. 제 10항에 있어서, 상기 아이케이 인자의 단편을 코딩하는 핵산은 서열식별번호:3, 서열식별번호:46 및 서열식별번호:48로 구성되는 군에서 적어도 하나 선택되는 뉴클레오티드 서열을 가지는 핵산으로 구성되는 약학적 조성물. The nucleic acid encoding a fragment of the IK factor is composed of a nucleic acid having at least one nucleotide sequence selected from the group consisting of SEQ ID NO: 3, SEQ ID NO: 46 and SEQ ID NO: 48. Pharmaceutical compositions.
  17. 제 10항에 있어서, 상기 약학적 조성물은 류마티스성 관절염을 치료하는데 사용되는 약학적 조성물. The pharmaceutical composition of claim 10, wherein the pharmaceutical composition is used to treat rheumatoid arthritis.
  18. 제 10항에 있어서, 상기 아이케이 인자 또는 상기 아이케이 인자의 단편을 코딩하는 핵산은 상기 약학적 조성물 중에 1.0 ng/mL ~ 10 ㎍/mL의 농도로 함유되어 있는 약학적 조성물. The pharmaceutical composition of claim 10, wherein the nucleic acid encoding the IK factor or a fragment of the IK factor is contained in the pharmaceutical composition at a concentration of 1.0 ng / mL to 10 μg / mL.
  19. 제 10항에 있어서, 상기 유전자 전달체는 네이키드 핵산 분자, 플라스미드, 바이러스 벡터 및 상기 플라스미드 또는 상기 바이러스 벡터를 함유하는 리포좀 또는 니오좀의 형태를 가지는 약학적 조성물.The pharmaceutical composition of claim 10, wherein the gene carrier is in the form of a naked nucleic acid molecule, a plasmid, a viral vector and a liposome or niosome containing the plasmid or the viral vector.
  20. 제 19항에 있어서, 상기 바이러스 벡터는 아데노바이러스, 아데노-관련 바이러스, 레트로 바이러스, 렌티바이러스, 바큘로바이러스, 헤르페스 심플렉스 바이러스, 백시니아 바이러스로 구성되는 군에서 선택되는 약학적 조성물. The pharmaceutical composition of claim 19, wherein the viral vector is selected from the group consisting of adenovirus, adeno-associated virus, retrovirus, lentivirus, baculovirus, herpes simplex virus, vaccinia virus.
PCT/KR2014/003282 2013-04-16 2014-04-16 Ik factor and pharmaceutical use of nucleic acid encoding ik factor WO2014171721A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US14/784,614 US9737588B2 (en) 2013-04-16 2014-04-16 Method for treating arthritis using IK factor or nucleic acid encoding IK factor
US15/481,787 US9943567B2 (en) 2013-04-16 2017-04-07 Method for treating arthritis using IK factor or nucleic acid encoding IK factor

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
KR10-2013-0041771 2013-04-16
KR20130041771 2013-04-16
KR10-2014-0038809 2014-04-01
KR1020140038809A KR101625112B1 (en) 2013-04-16 2014-04-01 Pharmaceutical use of ik factor and nucleic acids encoding ik factor

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US14/784,614 A-371-Of-International US9737588B2 (en) 2013-04-16 2014-04-16 Method for treating arthritis using IK factor or nucleic acid encoding IK factor
US15/481,787 Division US9943567B2 (en) 2013-04-16 2017-04-07 Method for treating arthritis using IK factor or nucleic acid encoding IK factor

Publications (1)

Publication Number Publication Date
WO2014171721A1 true WO2014171721A1 (en) 2014-10-23

Family

ID=51731591

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2014/003282 WO2014171721A1 (en) 2013-04-16 2014-04-16 Ik factor and pharmaceutical use of nucleic acid encoding ik factor

Country Status (1)

Country Link
WO (1) WO2014171721A1 (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20020034185A (en) * 1999-09-09 2002-05-08 둘락 노먼 씨. Mammalian cytokines, related reagents and methods
KR20040031031A (en) * 2001-08-29 2004-04-09 제넨테크, 인크. Compositions and Methods for the Treatment of Immune Related Diseases

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20020034185A (en) * 1999-09-09 2002-05-08 둘락 노먼 씨. Mammalian cytokines, related reagents and methods
KR20040031031A (en) * 2001-08-29 2004-04-09 제넨테크, 인크. Compositions and Methods for the Treatment of Immune Related Diseases

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
DATABASE GENBANK 15 August 2012 (2012-08-15), "protein Red [Mus musculus", accession no. P_036009. *
MASATAKE MURAOKA ET AL.: "IK Cytokine Ameliorates the Progression of Lupus Nephritis in MRL/1pr Mice", ARTHRITIS & RHEUMATISM., vol. 54, no. 11, 2006, pages 3591 - 3600 *
TAKAHIRO OKABE ET AL.: "Detection of gene expression in synovium of patients with osteoarthritis using a random sequencing method", ACTA ORTHOPAEDICA, vol. 78, no. 5, 2007, pages 687 - 692 *

Similar Documents

Publication Publication Date Title
CA2285040C (en) Death domain containing receptor 5
JP4411330B2 (en) Tumor necrosis factor-related ligand
US20020150993A1 (en) Novel tumor necrosis factor receptor homolog and nucleic acids encoding the same
AU732793B2 (en) Modulators of TNF receptor associated factor (TRAF), their preparation and use
US20030100745A1 (en) Antibody for 4-1BB
WO2013169077A1 (en) Composition for preventing or treating cachexia
JP2010187691A (en) MAMMALIAN TRANSFORMING GROWTH FACTOR beta-9
WO2000017350A1 (en) New human growth differentiation factor encoding sequence and polypeptide encoded by such dna sequence and producing method thereof
KR100781666B1 (en) Human Interferon-beta Mutein
JP2011155971A (en) Il-1 related polypeptide
CA2270913C (en) Human tumor necrosis factor receptor-like 2
US20070048827A1 (en) Mammalian transforming growth factor beta-9
US9943567B2 (en) Method for treating arthritis using IK factor or nucleic acid encoding IK factor
WO2015072750A1 (en) Peptide having anti-inflammatory activity and composition comprising same
WO2014171721A1 (en) Ik factor and pharmaceutical use of nucleic acid encoding ik factor
NO327854B1 (en) DNA sequence, replicable expression vector, transformed eukaryotic or prokaryotic host cell; RAP-2 protein, isoform, fragment, functional analogue or derivatives thereof, and method of preparation and use thereof; pharmaceutical composition, use of a recombinant animal virus vector, and a ribozyme coding vector.
US20230203204A1 (en) Fusion protein, preparation method therefor and use thereof
TW202216747A (en) Fusion protein comprising glucagon-like peptide-1 and interleukin-1 receptor antagonist, and use thereof
JPH10504714A (en) Human potassium channel 1 and 2 proteins
US20030211991A1 (en) Human sez6 nucleic acids and polypeptides
WO2001083552A2 (en) Human sez6 nucleic acids and polypeptides
KR102365266B1 (en) Polypeptide derived from oenanthe javanica and phamaceutical composition comprising polypeptide thereof
US20070256145A1 (en) Mammalian transforming growth factor beta-9
WO1998025957A2 (en) HUMAN Prt1-LIKE SUBUNIT PROTEIN (hPrt1) AND HUMAN eIF4G-LIKE PROTEIN (p97) GENES
WO2002002771A2 (en) Human sparc-homologous (hsparc-h1) gene and methods and uses thereof

Legal Events

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

Ref document number: 14785060

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 14784614

Country of ref document: US

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 14785060

Country of ref document: EP

Kind code of ref document: A1