WO2024112143A1 - Use of stat1-tmd in immune-related diseases - Google Patents

Use of stat1-tmd in immune-related diseases Download PDF

Info

Publication number
WO2024112143A1
WO2024112143A1 PCT/KR2023/019074 KR2023019074W WO2024112143A1 WO 2024112143 A1 WO2024112143 A1 WO 2024112143A1 KR 2023019074 W KR2023019074 W KR 2023019074W WO 2024112143 A1 WO2024112143 A1 WO 2024112143A1
Authority
WO
WIPO (PCT)
Prior art keywords
disease
cells
cancer
tmd
ndstat1
Prior art date
Application number
PCT/KR2023/019074
Other languages
French (fr)
Korean (ko)
Inventor
김정호
김범석
Original Assignee
주식회사 굳티셀
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 주식회사 굳티셀 filed Critical 주식회사 굳티셀
Publication of WO2024112143A1 publication Critical patent/WO2024112143A1/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders

Definitions

  • the present invention relates to a novel fusion protein containing STAT1 (Signal transducer and activator of transcription 1, STAT1) and a protein transduction domain (PTD) and uses thereof.
  • STAT1 Signal transducer and activator of transcription 1, STAT1
  • PTD protein transduction domain
  • T helper (Th) cells play an essential role in various immune responses and inflammation. Homeostatic imbalance in the functional immune network caused by excessively activated CD4 + effector T cells is associated with the initiation and maintenance of inflammatory responses that lead to serious autoimmune diseases. More specifically, T helper 1 (Th1) cells, which secrete interferon- ⁇ (IFN- ⁇ ), and T helper 17 (Th17) cells, which secrete interleukin-17 (IL-17), are involved in long-term diseases, including psoriasis and inflammatory bowel disease. It is known to be related to the pathogenesis of specific inflammatory diseases. In addition, T helper 1 (Th1) cells and T helper 17 (Th17) cells are known to be related to the pathogenesis of various metabolic diseases, including degenerative brain or vascular diseases, obesity, and arteriosclerosis.
  • T helper 1 cells and T helper 17 cells (Th17 cells) play a central role in the pathogenesis of various autoimmune diseases, including psoriasis and inflammatory bowel disease (IBD).
  • STAT1 Signal transducer and activator of transcription 1
  • Th1 and Th17 cell lineage commitment at early stages and maintains immunological functions in vitro and in vivo. Previous strategies to block STAT1 inhibit its ability to treat autoimmune diseases but also cause hyperactivation of Th17 cells.
  • the protein transduction domain is a short peptide with strong hydrophobicity and is known to effectively deliver bioactive molecules such as proteins, DNA, and RNA fused together into cells. Since the protein transport domain can transport bioactive molecules not only to the cytoplasm but also to the nucleus, it has characteristics suitable for delivering various proteins into the nucleus.
  • the purpose of the present invention is to provide a fusion protein containing the transcription modulation domain (TMD) and protein transduction domain (PTD) of STAT1.
  • TMD transcription modulation domain
  • PTD protein transduction domain
  • Another object of the present invention is to obtain a fusion protein containing a plurality of protein transduction domains and a transcription modulation domain by expressing a nucleic acid molecule; and purifying the fusion protein obtained in the above step.
  • Another object of the present invention is to provide a pharmacological composition containing the fusion protein for the treatment, improvement or prevention of immune-related diseases.
  • One embodiment of the present invention provides a fusion protein comprising a transcriptional regulatory domain and a protein transduction domain (PTD) of STAT1 (Signal transducer and activator of transcription 1).
  • PTD protein transduction domain
  • T helper 1 cells (Th1 cells) and T helper 17 cells (Th17 cells) play a pivotal role in the pathogenesis of various autoimmune diseases, including psoriasis and inflammatory bowel disease (IBD).
  • T helper 1 (Th1) cells and T helper 17 (Th17) cells are known to be related to the pathogenesis of various metabolic diseases, including degenerative brain or vascular diseases, various inflammatory diseases, obesity, and arteriosclerosis.
  • STAT1 Signal transducer and activator of transcription 1 regulates Th1 and Th17 cell lineage commitment at early stages and maintains immune function in vitro and in vivo.
  • ndSTAT1-TMD nuclear delivery form of the STAT1 transcriptional regulatory domain that can be delivered to the nucleus of the target to regulate the function of pathogenic Th1 and Th17 cells without genetic modification under normal physiological conditions.
  • the fusion protein provided by the present invention protects cells in a dose- and time-dependent manner without affecting cell viability and T cell activation signaling events.
  • the ndSTAT1-TMD provided by the present invention significantly blocked the differentiation of na ⁇ ve CD4 + T cells into Th1 or Th17 cells through competitive inhibition of endogenous STAT1-mediated transcription, and did not affect Th2 and Treg cell differentiation.
  • ndSTAT1-TMD can be a new therapeutic agent for Th1/17 cell-mediated autoimmune diseases by jointly regulating the functions of pathogenic Th1 and Th17 cells.
  • Th1 cells are known to protect our bodies from invading intracellular pathogens and viruses by secreting IFN- ⁇ to activate both macrophages and dendritic cells (DC). Secreted IFN- ⁇ also acts as a Th1 differentiation inducer by activating the STAT1 signaling pathway in the early stages. Th17 cells secrete IL-17 and IL-23, which are mainly involved in eliminating extracellular bacteria and fungi. Moreover, the presence of IFN- ⁇ during Th17 cell lineage development undermines the full commitment of Th17 cell differentiation. In addition to all these unique properties of Th1 and Th17 cells, hyperactivated Th1 and Th17 cells play an important role in autoimmune disorders by disrupting the immunological balance between helper and regulatory T cells and with subsequent cytokine dysregulation. Do it.
  • STAT1 a member of the STAT family, regulates the axes of Th1/Th2 and Th1/17 cell differentiation by controlling the expression pattern of T helper subset-specific genes during the early stages of CD4 + T cell differentiation.
  • STAT1 has an N-terminal domain (NTD), coiled-coil domain (CCD), DNA binding domain (DBD), linker domain (LD), and SCR2 homology. It consists of six domains, including a SCR2 homology domain (SH2) and a transactivation domain (TAD).
  • STAT1 homodimers translocate to the nucleus and induce and maintain the expression of T-bet, a transcription factor involved in Th1 responses.
  • STAT1 has also been shown to inhibit Th17 cell differentiation by directly binding to the Rorc or Il17a loci, the corresponding chromosomal loci of ROR- ⁇ t and IL-17A expression, respectively, which are critically involved in the regulation of Th17 responses.
  • STAT1-knockout (KO) studies reported that loss of endogenous STAT1 impairs Th17 cell lineage commitment.
  • Functional inhibition of STAT1 using genetic methods has been consistently proposed as a treatment strategy for Th1 or Th17 cell-mediated systemic autoimmune diseases and has shown substantial effects in limiting Th1 cells. However, no significant functional regulation of Th17 cells was detected, and Th17 cells lacking or with low levels of STAT1 displayed a hyperactivated phenotype.
  • transcription modulation domain refers to a domain that constitutes a transcription factor and consists of only a DNA binding site without a transactivation domain.
  • the fusion protein of the present invention does not have a transactivation domain but has a DNA binding site, so it can bind to the desired promoter (e.g. Foxp3 and CD25) but cannot promote transcription. . Therefore, since the fusion protein of the present invention is a dominant negative mutant for the Foxp3 and CD25 genes, it can act as a competitive inhibitor for wild-type STAT1 in cells and inhibit the transcription and activity of STAT1.
  • the “protein transport domain (PTD)” is a short, highly hydrophobic peptide consisting of 7 to 50 amino acids, and refers to a domain capable of delivering not only proteins with a molecular weight of 120 kDa or more, but also DNA or RNA into cells. .
  • the protein transport domains include Hph-1, Mph-1, Sim-2, Tat, VP22, Antp (antennapedia), Pep-1 (peptide-1), PTD-5 (protein transduction domain-5), and 11R. , 7R, and CTP (cytoplamic transduction peptide), or a macromolecule transduction domain may be used, but is not limited thereto, and is commonly used or commercially available in the art. Any protein transport domain can be used without limitation.
  • CTP represented by SEQ ID NO: 1 Hph-1 represented by SEQ ID NO: 2, or Tat represented by SEQ ID NO: 3, Sim-2 represented by SEQ ID NO: 4, Mph-1 represented by SEQ ID NO: 5, VP22 represented by SEQ ID NO: 6, Antp represented by SEQ ID NO: 7, Pep-1 represented by SEQ ID NO: 8, PTD-5 represented by SEQ ID NO: 9, 7R represented by SEQ ID NO: 10, and SEQ ID NO: 11. It may be any one of 11R and CTP represented by SEQ ID NO: 12.
  • the protein transport domains include Hph-1, Mph-1, Sim-2, Tat, VP22, Antp (antennapedia), Pep-1 (peptide-1), PTD-5 (protein transduction domain-5), and 11R. , 7R and CTP (cytoplamic transduction peptide), Hph-1, Mph-1, Sim-2, Tat, VP22, Antp (antennapedia), Pep-1 (peptide-1), PTD
  • One or more domains selected from the group consisting of -5 (protein transduction domain-5), 11R, 7R, and CTP (cytoplamic transduction peptide) are repeated, for example, 2, 3, 4 or more times. can be included.
  • the number of protein transport domains is not particularly limited, but may be 2 or more and 10 or less, 2 or more and 5 or less, and 2 or more and 3 or less.
  • nucleic acid molecule encoding the fusion protein provided by the present invention is provided.
  • the nucleic acid molecule of the present invention includes all nucleic acid molecules in which the amino acid sequence of the fusion protein provided by the present invention has been translated into a polynucleotide sequence as known to those skilled in the art. Therefore, various polynucleotide sequences can be prepared by ORF (open reading frame), and all of these are also included in the nucleic acid molecule of the present invention.
  • ORF open reading frame
  • an expression vector is provided into which the isolated nucleic acid molecule provided by the present invention is inserted.
  • the “vector” is a nucleic acid molecule capable of transporting another nucleic acid to which a nucleic acid molecule is linked.
  • a vector which refers to a circular double-stranded DNA into which additional DNA segments can be ligated.
  • a phage vector Another type of vector is a viral vector, in which additional DNA segments can be ligated into the viral genome.
  • Some vectors are capable of autonomous replication in the host cell into which they are introduced (e.g., bacterial vectors are episomal mammalian vectors that have a bacterial origin of replication).
  • vectors e.g., non-episomal mammalian vectors
  • Other vectors can enter a host cell and become integrated into the host cell's genome, thereby replicating along with the host genome.
  • some vectors can direct the expression of genes to which they are operationally linked.
  • Such vectors are referred to herein as “recombinant expression vectors” or simply “expression vectors”.
  • expression vectors useful in recombinant DNA techniques often exist in the form of plasmids.
  • plasmid and “vector” may be used interchangeably since plasmid is the most commonly used form of vector.
  • the expression vector in the present invention include widely used commercially pCDNA vectors, F, R1, RP1, Col, pBR322, ToL, and Ti vectors; cosmid; Phages such as lambda, lambdoid, M13, Mu, p1 P22, Q ⁇ , T-even, T2, T3, T7; It may be selected from the group consisting of plant viruses, but is not limited thereto, and all expression vectors known to those skilled in the art as expression vectors can be used in the present invention, and when selecting an expression vector, it depends on the properties of the target host cell.
  • Introduction of vectors into host cells may be performed by calcium phosphate transfection, viral infection, DEAE-dextran control transfection, lipofectamine transfection, or electroporation, but is not limited thereto, and those skilled in the art will use the method.
  • An introduction method suitable for the expression vector and host cell can be selected and used.
  • the vector contains one or more selection markers, but is not limited to this, and selection can be made depending on whether or not the product is produced using a vector that does not contain a selection marker. The selection of the selection marker is selected based on the target host cell, and since this method is already known to those skilled in the art, the present invention is not limited thereto.
  • a tag sequence can be inserted and fused into an expression vector.
  • the tag includes, but is not limited to, a hexa-histidine tag, a hemagglutinin tag, a myc tag, or a flag tag, and any tag that facilitates purification known to those skilled in the art can be used in the present invention.
  • a host cell line transformed with the expression vector provided by the present invention is provided.
  • the “host cell” includes an individual cell or cell culture that can be or has been a recipient of vector(s) for incorporation of a polypeptide insert.
  • a host cell includes the progeny of a single host cell, which progeny may not necessarily be completely identical (morphologically or in genomic DNA complement) to the original parent cell due to natural, accidental or intentional mutations.
  • Host cells include cells transfected in vivo with the polypeptide(s) herein.
  • the host cells may include cells of mammalian, plant, insect, fungal, or cellular origin, for example, bacterial cells such as Escherichia coli, Streptomyces, and Salmonella Typhimurium; Fungal cells such as yeast cells and Pichia pastoris; Insect cells such as Drozophila and Spodoptera Sf9 cells; CHO (Chinese hamster ovary cells), SP2/0 (mouse myeloma), human lymphoblastoid, COS, NSO (mouse myeloma), 293T, Bow melanoma cells, HT-1080, BHK Animal cells of (Baby Hamster Kidney cells), HEK (Human Embryonic Kidney cells) or PERC.6 (Human Retinal Cells); Or it may be a plant cell, but is not limited thereto, and any cell that can be used as a host cell line known to those skilled in the art can be used.
  • bacterial cells such as Escherichia
  • lipid nanoparticles containing the nucleic acid molecules are provided.
  • nanoparticles are particles containing one or more lipids and one or more therapeutic agents. Nanoparticles are typically on the order of micrometers or smaller and may include a lipid layer.
  • the lipid nanoparticle (LNP) refers to an endoplasmic reticulum with an adjacent lipid bilayer, such as a spherical endoplasmic reticulum. Lipid nanoparticles can be used in methods to deliver pharmaceutical therapies to targeted locations.
  • Non-limiting examples of LNPs include liposomes, amphiphiles, solid lipid nanoparticles (SLNs), nanostructured lipid carriers (NLCs), and monolayer membrane structures (e.g., archaeosomes and micelles).
  • auxiliary lipids selected from positively charged lipids, neutral lipids, and negatively charged lipids can be additionally used.
  • positively charged lipids and neutral lipids may be used
  • neutral liposomes neutral lipids
  • anionic liposomes negatively charged lipids and neutral lipids may be additionally used.
  • Positively charged lipids, neutral lipids, and negatively charged lipids that can be used for the preparation of lipid nanoparticles are known in the art.
  • the lipid component of the nanoparticle may include one or more phospholipids, such as one or more (poly)unsaturated lipids.
  • Phospholipids can be assembled into one or more lipid layers.
  • phospholipids may include a phospholipid moiety and one or more fatty acid moieties.
  • Phospholipids useful in the compositions and methods include 1,2-distearoyl-sn-glycero-3-phosphocholine (DSPC), 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine ( DOPE), 1,2-dilinoleoyl-sn-glycero-3-phosphocholine (DLPC), 1,2-dimyristoyl-sn-glycero-phosphocholine (DMPC), 1,2 -Dioleoyl-sn-glycero-3-phosphocholine (DOPC), 1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC), 1,2-diundecanoyl- sn-glycero-phosphocholine (DUPC), 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC), 1,2-di-O-octadecenyl-sn- Glycero-3-phosphocholine
  • the lipid component of the nanoparticle may include one or more structural lipids.
  • Structural lipids may be selected from, but are limited to, cholesterol, fecosterol, sitosterol, ergosterol, campesterol, stigmasterol, brassicasterol, tomatidine, tomatine, ursolic acid, alpha-tocopherol, and mixtures thereof. It doesn't work.
  • the lipid component of the nanoparticle may also include one or more PEG or PEG-modified lipids. These lipids may alternatively be referred to as PEGylated lipids.
  • PEG lipids are lipids modified with polyethylene glycol.
  • PEG lipids include PEG-modified phosphatidylethanolamine, PEG-modified phosphatidic acid, PEG-modified ceramide, PEG-modified dialkylamine, PEG-modified diacylglycerol, PEG-modified dialkylglycerol and these. It may be selected from the non-limiting group consisting of mixtures of.
  • PEG lipids include PEG-c-DOMG, PEG-DMG (1,2-dimyristoyl-OT-glycerol methoxypolyethylene glycol, available from Avanti Polar Lipid, Elabaster, AL), PEG- It may be DLPE, PEG-DMPE, PEG-DPPC or PEG-DSPE lipid.
  • a pharmaceutical composition for the prevention or treatment of immune-related diseases containing any one of the above fusion proteins, nucleic acid molecules, lipid nanoparticles (LNPs), expression vectors, and host cells as an active ingredient.
  • LNPs lipid nanoparticles
  • the “immune-related disease” of the present invention may be at least one selected from the group consisting of autoimmune disease, graft-versus-host disease, organ transplant rejection, asthma, atopy, and acute or chronic inflammatory disease, but is not limited thereto. no.
  • the autoimmune diseases of the present invention include psoriasis, acanthosis, parakeratosis, neo-angiogenesis, rheumatoid arthritis, systemic scleroderma, systemic lupus erythematosus, atopic dermatitis, psoriasis, and alopecia areata. , asthma, Crohn's disease, Behce's disease, Sjögren's syndrome, Guillain-Barre syndrome, chronic thyroiditis, multiple sclerosis, polymyositis, ankylosing spondylitis, fibromyositis, and polyarteritis nodosa, but limited thereto. It doesn't work.
  • the present invention relates to the acute or chronic inflammatory diseases such as colitis, sepsis, septic shock, inflammatory bowel disease (IBD), psoriasis, peritonitis, nephritis, acute bronchitis, chronic bronchitis, osteoarthritis, and intestinal spondylitis.
  • chronic obstructive pulmonary disease COPD
  • rheumatoid arthritis acute lung injury
  • broncho-pulmonary dysplasia it is not limited to this.
  • psoriasis is a recurrent inflammatory skin disease characterized by thick, non-pruritic plaques covered with silvery scales of the skin.
  • IBD Inflammatory bowel disease
  • CD Crohn's disease
  • UC ulcerative disease
  • Typical symptoms include diarrhea, rectal bleeding, or weight loss.
  • the exact mechanism of how psoriasis and inflammatory bowel disease (IBD) develop is still unclear, but overly activated Th1 and Th17 cells are believed to be the main cause of irregular immune responses in the skin and intestinal tract.
  • IFN- ⁇ and IL-17A secreted by Th1 and Th17 cells, respectively, are important mediators that induce and accelerate the pathological progression of psoriasis and IBD.
  • therapeutic strategies targeting IFN- ⁇ or IL-17A for psoriasis and IBD still face several limitations that need to be overcome.
  • ndSTAT1-TMD nuclear delivery of the transcription modulation domain of STAT1
  • TMD transcription modulation domain
  • PTD human protein transduction domain
  • ndSTAT1-TMD nuclear delivery of the transcription modulation domain of STAT1
  • TcR-mediated signaling events The ndSTAT1-TMD provided by the present invention specifically inhibited the transcriptional activity of endogenous STAT1 in Th1 and Th17 cells and showed therapeutic potential similar to that of anti-IL-17A antibody when administered to psoriasis and IBD mouse models. This suggests that ndSTAT1-TMD provided by the present invention can be a new and effective treatment reagent for Th1/17 cell-mediated autoimmune diseases.
  • 'metabolic disease means a condition or disease closely related to or caused by obesity, and specifically selected from the group consisting of fatty liver, type 2 diabetes, hyperlipidemia, cardiovascular disease, and arteriosclerosis. There may be more than one.
  • fatty liver refers to a condition or disease in which fat accumulates in excessive amounts in liver cells due to a disorder of liver fat metabolism.
  • hyperlipidemia refers to a condition or disease in which the concentration of fat components in the blood, especially cholesterol and triglyceride, is higher than normal, and includes all conditions requiring lowering the lipid concentration in the blood. It is used in a broad sense.
  • arteriosclerosis refers to a condition or disease in which blood circulation to organs and tissues in the body is reduced due to thickening of the artery wall and reduction in elasticity, and the deposition of fat, cholesterol, and other substances on the inner wall of the artery, forming plaque ( It has a meaning that includes “atherosclerosis,” which refers to a condition or disease in which blood circulation is reduced by forming plaques and narrowing the lumen.
  • Atherosclerosis can occur anywhere in the body. If it occurs in blood vessels within the heart, it can cause coronary artery disease such as angina pectoris or myocardial infarction. If it occurs in the brain, it can cause cerebral infarction. If it occurs in the kidneys, it can cause renal failure. It can be triggered.
  • the “neurodegenerative disease” or “degenerative brain or vascular disease” may refer to a disease caused by decreased or lost function of nerve cells
  • the “neuroinflammatory disease” may refer to a disease caused by excessive inflammatory response in the nervous system. It may mean a disease that occurs.
  • Specific examples of the neurodegenerative disease or neuroinflammatory disease in the present invention include stroke, dementia, Alzheimer's disease, Parkinson's disease, Huntington's disease, and Niemann-Pick disease. , multiple sclerosis, prion disease, Creutzfeldt-Jakob disease, frontotemporal dementia, Lewy dementia, amyotrophic lateral sclerosis, paraneoplastic syndrome, corticobasal degeneration.
  • multiple system atrophy disease progressive supranuclear palsy, nervous system autoimmune disease, spinocerebellar ataxia, inflammatory and neuropathic pain, cerebrovascular disease, spinal cord injury, and tauopathy. It may be selected from the group, but is not limited thereto.
  • cancer refers to or refers to a physiological condition typically characterized by uncontrolled cell growth in mammals.
  • the cancer subject to prevention, improvement, or treatment in the present invention may be a solid tumor consisting of a lump generated by abnormal cell growth in a solid organ, for example, depending on the part of the solid organ. It may be stomach cancer, liver cancer, glioblastoma, ovarian cancer, colon cancer, head and neck cancer, bladder cancer, renal cell cancer, breast cancer, metastatic cancer, prostate cancer, pancreatic cancer, melanoma, or lung cancer. For example, it may be melanoma. It is not limited.
  • the “object of interest” refers to an individual who has developed, or is likely to develop, an immune-related disease.
  • prevention may include without limitation any act of blocking, suppressing or delaying the symptoms of a disease using the pharmaceutical composition of the present invention.
  • treatment may include, without limitation, any action that improves the symptoms of a disease or provides benefits using the pharmaceutical composition of the present invention.
  • the method for preventing or treating the disease may be a combination therapy that further includes administering a compound or substance having therapeutic activity for one or more diseases.
  • the “combined use” should be understood to indicate simultaneous, separate or sequential administration. If the administration is sequential or separate, the interval between the administrations of the second components should be such that the beneficial effects of the combination are not lost.
  • the pharmaceutical composition may be in the form of a capsule, tablet, granule, injection, ointment, powder, or beverage, and the pharmaceutical composition may be intended for human subjects.
  • the pharmaceutical composition is not limited to these, but can be formulated and used in the form of oral dosage forms such as powders, granules, capsules, tablets, and aqueous suspensions, external preparations, suppositories, and sterile injection solutions according to conventional methods. You can.
  • the pharmaceutical composition of the present invention may include a pharmaceutically acceptable carrier.
  • Pharmaceutically acceptable carriers include binders, lubricants, disintegrants, excipients, solubilizers, dispersants, stabilizers, suspending agents, colorants, flavorings, etc. for oral administration, and buffers, preservatives, and analgesics for injections. Topics, solubilizers, isotonic agents, stabilizers, etc.
  • the dosage form of the pharmaceutical composition of the present invention can be prepared in various ways by mixing it with a pharmaceutically acceptable carrier as described above.
  • a pharmaceutically acceptable carrier as described above.
  • it can be manufactured in the form of tablets, troches, capsules, elixirs, suspensions, syrups, wafers, etc., and in the case of injections, it can be manufactured in the form of unit dose ampoules or multiple doses. there is.
  • it can be formulated as a solution, suspension, tablet, capsule, sustained-release preparation, etc.
  • examples of carriers, excipients and diluents suitable for formulation include lactose, dextrose, sucrose, sorbitol, mannitol, xylitol, erythritol, maltitol, starch, gum acacia, alginate, gelatin, calcium phosphate, calcium silicate, cellulose. , methyl cellulose, microcrystalline cellulose, polyvinylpyrrolidone, water, methylhydroxybenzoate, propylhydroxybenzoate, talc, magnesium stearate, or mineral oil can be used.
  • fillers, anti-coagulants, lubricants, wetting agents, fragrances, emulsifiers, preservatives, etc. may be additionally included.
  • the route of administration of the pharmaceutical composition according to the present invention is not limited to these, but may be administered directly to the eye, orally, intravenously, intramuscularly, intraarterially, intramedullary, intrathecally, intracardiacally, transdermally, subcutaneously, intraperitoneally, or nasally. Includes internal, intestinal, topical, sublingual or rectal. Oral or parenteral administration is preferred. It is more preferable to administer directly to the eye.
  • parenteral includes direct administration to the eye, subcutaneous, intradermal, intravenous, intramuscular, intraarticular, intrasynovial, intrasternal, intrathecal, intralesional and intracranial injection or injection techniques.
  • the pharmaceutical composition of the present invention can also be administered in the form of a suppository for rectal administration. It is preferable to administer directly to the eye.
  • the "usage and dosage" of the composition of the present invention includes the activity of the specific compound used, age, weight, general health, gender, diet, administration time, administration route, excretion rate, drug combination, and the severity of the specific disease to be prevented or treated. It may vary depending on various factors, and the dosage of the pharmaceutical composition may vary depending on the patient's condition, body weight, degree of disease, drug form, administration route and period, but may be appropriately selected by a person skilled in the art, and is 0.0001 per day. It can be administered at 0.001 to 50 mg/kg or 0.001 to 50 mg/kg. Administration may be administered once a day, or may be administered several times. The above dosage does not limit the scope of the present invention in any way.
  • the pharmaceutical composition according to the present invention may be formulated as pills, dragees, capsules, solutions, gels, syrups, slurries, and suspensions.
  • the appropriate total daily amount can be determined by the treating physician within the scope of sound medical judgment, and can be administered once or in several divided doses.
  • the specific therapeutically effective amount for a specific patient is determined by the type and degree of response to be achieved, the composition containing the active ingredient, the patient's age, and whether other agents are used as the case may be.
  • Various factors well known in the medical field including body weight, general health condition, gender and diet, administration time, administration route and secretion rate of the composition containing the active ingredient, treatment period, and drugs used together or simultaneously with the specific composition. It is desirable to apply it differently depending on similar factors.
  • One embodiment of the present invention provides a fusion protein comprising a transcriptional regulatory domain and a protein transduction domain (PTD) of STAT1 (Signal transducer and activator of transcription 1).
  • PTD protein transduction domain
  • STAT1 provides a fusion protein represented by the amino acid sequence of SEQ ID NO: 13.
  • a fusion protein in which the transcriptional regulatory domain of STAT1 is represented by the amino acid sequence of SEQ ID NO: 14.
  • the protein transport domain is Hph-1, Mph-1, Sim-2, Tat, VP22, Antp (antennapedia), Pep-1 (peptide-1), PTD-5 (protein transduction domain) -5), 11R, 7R, and CTP (cytoplamic transduction peptide) are selected from the group consisting of fusion proteins.
  • Another embodiment of the present invention provides a fusion protein, wherein the protein transport domain is represented by any one of the amino acid sequences of SEQ ID NOs: 1 to 12.
  • Another embodiment of the present invention provides a fusion protein that competitively inhibits STAT1.
  • Another embodiment of the present invention provides a fusion protein that inhibits differentiation or function of Th1 or Th17.
  • One embodiment of the present invention provides a nucleic acid molecule encoding the fusion protein.
  • One embodiment of the present invention provides lipid nanoparticles (LNPs) containing the nucleic acid molecules.
  • LNPs lipid nanoparticles
  • One embodiment of the present invention provides an expression vector into which the nucleic acid molecule is inserted.
  • One embodiment of the present invention provides a host cell transfected with the expression vector.
  • a pharmaceutical composition for preventing or treating immune-related diseases comprising any one of the fusion protein, the nucleic acid molecule, the lipid nanoparticle (LNP), the expression vector, and the host cell as an active ingredient. to provide.
  • Another embodiment of the present invention provides a pharmaceutical composition for preventing or treating immune-related diseases, wherein the immune-related diseases occur due to activation or dysfunction of Th1 cells or Th17 cells.
  • the immune-related disease is at least one selected from the group consisting of autoimmune disease, graft-versus-host disease, organ transplant rejection, asthma, atopy, and acute or chronic inflammatory disease,
  • a pharmaceutical composition for preventing or treating immune-related diseases is provided.
  • the autoimmune disease includes psoriasis, acanthosis, parakeratosis, neo-angiogenesis, rheumatoid arthritis, systemic scleroderma, systemic lupus erythematosus, and atopic dermatitis.
  • alopecia areata, asthma, Crohn's disease, Behce's disease, Sjögren's syndrome, Guillain-Barre syndrome, chronic thyroiditis, multiple sclerosis, polymyositis, ankylosing spondylitis, fibromyositis, and polyarteritis nodosa.
  • the acute or chronic inflammatory disease includes colitis, sepsis, septic shock, inflammatory bowel disease (IBD), psoriasis, peritonitis, nephritis, acute bronchitis, chronic bronchitis, In the group consisting of osteoarthritis, enteropathy spondylitis, chronic obstructive pulmonary disease (COPD), rheumatoid arthritis, acute lung injury, and broncho-pulmonary dysplasia.
  • IBD inflammatory bowel disease
  • COPD chronic obstructive pulmonary disease
  • rheumatoid arthritis acute lung injury
  • broncho-pulmonary dysplasia broncho-pulmonary dysplasia.
  • a pharmaceutical composition for preventing or treating immune-related diseases which is characterized in that it contains one or more selected substances.
  • the fusion protein (ndSTAT1-TMD) provided in the present invention can be efficiently delivered to the nucleus of cells without cytotoxicity or affecting TcR-mediated signaling events. Additionally, the fusion protein provided in the present invention specifically inhibited the transcriptional activity of endogenous STAT1 in Th1 and Th17 cells and showed therapeutic potential similar to that of anti-IL-17A antibody when administered to psoriasis and IBD mouse models. Therefore, the fusion protein provided in the present invention can become a new treatment for Th1/17 cell-mediated autoimmune diseases by controlling the functions of pathogenic Th1 and Th17 cells.
  • Figure 1 shows the production of ndSTAT1-TMD capable of nuclear delivery and verification of its intranuclear delivery kinetics without cytotoxicity.
  • Figure 1A shows the protein structures of Hph-1-PTD with mutated valine 426 and threonine 427, ndSTAT-TMD, and STAT1-TMD without ndSTAT1-TMD (V426D, T427D), PTD and protein transduction domain, respectively.
  • protein transduction domain nd, nucleus-transducible
  • TMD transcriptional regulatory domain
  • DBD DNA binding domain
  • ND N-terminal domain
  • LD linker domain
  • TAD stands for transaction activation domain.
  • Figure 1B shows purified STAT1-TMD, ndSTAT1-TMD, and ndSTAT1-TMD (V426D, T427D) confirmed by Western blot (left panel) or SDS-PAGE (right panel).
  • Figures 1C and 1D show dose-dependent and time-dependent intranuclear delivery of ndSTAT1-TMD (0.5-2 ⁇ M) or STAT1-TMD (2 ⁇ M) into mouse splenocytes.
  • Figure 1E shows the intracellular stability of ndSTAT1-TMD after transduction into mouse splenocytes.
  • Figure 1F Subcellular localization of ndSTAT1-TMD in cultured mouse splenocytes. Representative immunofluorescence 63X (left panel) and 189X (right panel) images using confocal microscopy are shown, and the scale bar is 10 ⁇ m.
  • Figure 1G shows cell cytotoxicity evaluation of ndSTAT1-TMD or ndSTAT1-TMD (V426D, T427D) in mouse splenocytes analyzed by CCK-8 assay.
  • Figure 2 shows the functional impact of ndSTAT1-TMD on the differentiation ability of CD4 + naive T cells into various T cell subsets.
  • Figure 2A shows flow cytometric analysis of intracellular T-bet and IFN- ⁇ in Th1 cells treated with various concentrations (0.5-2 ⁇ M) of ndSTAT1-TMD or ndSTAT1-TMD (V426D, T427D) under Th1-skewing conditions. It represents.
  • Figure 2B shows the level of IFN- ⁇ secreted in the Th1 cell supernatant of Figure 2A measured by ELISA.
  • Figure 2C shows flow cytometry analysis of the levels of T-bet + IRF1 + cells in the Th1 cells of Figure 2A.
  • Figure 2D shows ROR- ⁇ t and intracellular IL- levels in Th17 cells treated with various concentrations (0.5-2 ⁇ M) of ndSTAT1-TMD or ndSTAT1-TMD (V426D, T427D) under Th17-skewing conditions. Flow cytometry analysis of 17A is shown.
  • Figures 2E and 2F show the levels of IL-17A or IFN- ⁇ secreted from the Th17 cell supernatant of Figure 2D measured by ELISA. All experiments shown here were repeated three times on independent biological samples with similar results. Data are expressed as mean ⁇ SEM (n ⁇ 3) and statistical analysis was examined using Student's t-test.
  • Figure 3 shows that ndSTAT1-TMD suppresses the expression of STAT1 target genes at the transcription level.
  • Figures 3B and 3C are a representation of the genes upregulated in STAT3 knockout CD4 + T cells compared to wild-type CD4 + T cells in ntSTAT1-treated Th1 cells or in ntSTAT1-TMD treated Th17 cells. This shows GSEA-based enrichment plots for .
  • FIGS 3D and 3E show heatmaps of Th1 or Th17 cell signature genes and naive T cell specific genes in Th1 or Th17 cells treated or not with ndSTAT1-TMD. Each row represents the results from one experimental well and the Z-score was calculated from log2(TPM+1).
  • Figure 4 shows that ndSTAT1-TMD treatment attenuates the disease severity of IMQ-induced psoriasis.
  • Figure 4A shows PBS (IMQ group), anti-IL-17A antibody (60 ⁇ g/mouse), and STAT1-TMD (20 ⁇ g/mouse or 60 ⁇ g/mouse), or ndSTAT1-TMD (V426D, T427D).
  • This shows a treatment method for 5% IMQ-induced psoriasis mice using (60 ⁇ g/mouse).
  • Figure 4B shows a plot of the PASI scores on the back skin of each group in Figure 4A against disease progression.
  • Figure 4C shows representative images of skin damage on the back of the mouse in Figure 4A on day 5 (top panel) and day 7 (bottom panel).
  • Figure 4D shows the change in back skin thickness within each group from Figure 4A on the 5th day, calculated based on the thickness on the 1st day.
  • Figure 4E shows the back skin thickness of mice in each treatment group.
  • Figure 4F shows the change in body weight of each group.
  • Figure 4G shows the levels of proinflammatory cytokines (TNF- ⁇ , IL-1 ⁇ , or IL-6) in the serum of mice from each group measured by ELISA.
  • TNF- ⁇ proinflammatory cytokines
  • IL-1 ⁇ IL-1 ⁇
  • IL-6 proinflammatory cytokines
  • Figure 5 shows that ndSTAT1-TMD treatment restores the destroyed functional state of CD4 + T cells in a psoriasis animal model.
  • Figures 5A and 5B show CD4 + T-bet + T cells (left panel of (A) and (B)) or CD4 + in the spleen (A) and draining lymph node (dLN) (B) of each treatment group.
  • the level of ROR- ⁇ t + T cells (right panel (A) of (A) and (B)) is shown as examined on day 7 by flow cytometry.
  • Figures 5C and 5D show CD4 + IFN- ⁇ + (left panels of (C) and (D)) or CD4 + IL-17A + ( (C) and (D) right panel) levels were analyzed on day 7.
  • Figure 6 shows that ndSTAT1-TMD treatment of a psoriasis animal model prevents leukocytes from infiltrating the back skin.
  • Figure 6A shows representative H&E staining (top panel) or Masson's trichrome staining (bottom panel) results on the back skin of mice from each group at day 7.
  • the scale bar represents 200 ⁇ m for 100X and 100 ⁇ m for 200X.
  • Figure 6B shows changes in the thickness of the skin epidermis on the back of mice in each treatment group measured using the histological image of Figure 6A.
  • Figure 6C shows the level of CD45 + leukocytes infiltrated in the dorsal epidermis (grey) or dermis (blue) of each group on day 7.
  • Figure 6D shows the levels of infiltrating macrophages expressing CD45, CD11b, and F4/80 in the dorsal epidermis (grey) or dermis (blue) region of each group at day 7.
  • Figure 6E shows the level of nitric oxide (NO) in the serum of mice in each treatment group on day 7.
  • FIGS 6F and 6G show infiltration of T-bet + (Figure 6F) or ROR- ⁇ t + ( Figure 6G) T cells in total CD45 + leukocytes in the dorsal epidermis (gray) or dermis (blue) region of each treatment group on day 7. It shows the degree to which it has been done.
  • Figures 6H and 6I show infiltration of IFN- ⁇ + (Figure 6H) or IL-17A + ( Figure 6I) T cells in total CD45 + leukocytes in the dorsal epidermis (gray) or dermis (blue) region of each treatment group on day 7. It shows the degree to which it has been done.
  • Figure 7 shows that ndSTAT1-TMD alleviates the disease progression of DSS-induced colitis.
  • Figure 7A shows 100 ⁇ g of PBS (DSS group), anti-IL-17A antibody (100 ⁇ g/mouse), ndSTAT1-TMD (20 ⁇ g/mouse or 100 ⁇ g/mouse), or ndSTAT1-TMD (V426D, T427D) (100 ⁇ g). /mouse) shows the treatment plan for mice with 2.5% DSS-induced colitis.
  • Figure 7B shows the change in body weight in each treatment group.
  • Figure 7C shows the disease activity index (DAI) score for each treatment group.
  • the DAI score was calculated by measuring the clinical scores of stool, stool occult blood, and weight change in each group.
  • the left panel of Figure 7D shows a comparative analysis of the colon length of mice in each treatment group on day 12, and the right panel shows a representative image of the 7A colon.
  • Figure 7E shows representative vertical (above panel) and horizontal (below panel) colon sections from Figure 7D stained with H&E or PAS.
  • the scale bar represents 500 ⁇ m for 40X and 100 ⁇ m for 200X.
  • Figure 7F shows the thickness of the colonic muscle layer of each treatment group measured from the image in Figure 7E.
  • the plasmid of mouse STAT1 (BC004808) was purchased from Origene Technologies, Inc., USA.
  • the sequence coding for amino acids 317 to 488 of the wild-type STAT1 DNA binding domain (DBD) was amplified by PCR and then inserted into the pET28a(+) expression vector containing Hph-1-PTD, 6x His tag, and FLAG tag. introduced.
  • Point mutation forms of DBD (V426D, T427D) were generated using pfu DNA polymerase (Agilent). After cloning, all generated cloned DNA structures were sequenced to confirm the fidelity of the open reading frame (ORF).
  • Inclusion bodies were pelleted by centrifugation of the cell lysate and further sonicated in native lysis buffer containing 3M urea (10mM imidazole, 50mM NaH 2 PO 4 , 300mM NaCl, 3M Urea, pH 8.0). did. Cell lysates were pelleted, and the supernatant was mixed with Ni-NTA resin (Qiagen). The recombinant protein was washed with a buffer containing 30mM imidazole, 50mM NaH 2 PO 4 , 300mM NaCl, pH 8.0, and eluted with a buffer containing 500mM imidazole, 50mM NaH 2 PO 4 , 300mM NaCl, pH 8.0.
  • the eluted recombinant protein was further purified to remove endotoxin through ion exchange chromatography and desalted using a PD-10 Sephadex G-25 column (GE Healthcare).
  • the purified recombinant protein contained a safe level of endotoxin of approximately 6 EU/ml. In subsequent animal experiments, none of the animals showed signs of an immune response to endotoxin, such as anaphylactic shock.
  • the recombinant protein purified in step 2 above was separated through sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) and transferred to a polyvinylidene fluoride (PVDF, Bio-Rad, Hercules, CA, USA) membrane. . Membranes were blocked with 5% bovine serum albumin in 0.1% Tween-20 in Tris-buffered saline. Recombinant proteins were incubated with anti-6x His-tag antibody (27E8 Clone, Cell Signaling, Danvers, MA, USA) and anti-mouse IgG-HRP antibody (Abcam, Cambridge, MA, USA). Then, ECL reagent (Bio-Rad) was used to visualize the protein in ChemiDoc (Bio-Rad).
  • HEK293T cells were incubated with 10% heat-inactivated fetal bovine serum (FBS, Hyclone), 2mM L-glutamate (Lonza), 100 ⁇ g/ml penicillin-streptomycin (Lonza), 1mM sodium pyruvate (Lonza), and NEAA (Gibco, Waltham). , MA, USA) was cultured in DMEM medium (Lonza, Basel, Switzerland) supplemented with .
  • 2x105 mouse splenocytes were cultured with recombinant proteins in 96-well cell culture plates (Eppendorf) in a dose-dependent manner (0.5-2 ⁇ M, 1 h) or time-dependent manner (2 ⁇ M, 0-72 h) for flow cytometric analysis. was used in 2x106 mouse splenocytes were seeded on coverslips placed on the bottom of a 12-well cell culture plate (SPL, Pocheon, South Korea) and incubated with 1 ⁇ M of ndSTAT1-TMD or STAT1-TMD for 1 h before analysis, followed by confocal analysis. Observation was made using a microscope (LSM 980, Carl Zeiss, Jena, Germany).
  • 2x105 mouse na ⁇ ve CD4 + T cells were cultured in 96-well cell culture plates (Eppendorf) under appropriate T cell subset-polarizing conditions with the addition of recombinant proteins in a dose-dependent manner (0.5-2 ⁇ M).
  • 2x10 6 mouse splenocytes were seeded on a round microscope cover glass with a diameter of 18 mm placed on the bottom of a 12-well cell culture plate (SPL).
  • Cells were cultured in 12-well cell culture plates (SPL) in medium containing PBS, 1 ⁇ M STAT1-TMD, or 1 ⁇ M ndSTAT1-TMD for 1 hour.
  • SPL 12-well cell culture plates
  • cells were washed with PBS, fixed with 10% formalin solution (Sigma-Aldrich), and permeabilized with 0.5% Triton X-100 solution (Sigma-Aldrich). Samples were blocked with 1% BSA solution.
  • the delivered protein was captured with anti-FLAG Alexa 488 antibody (Invitrogen). After washing with PBS, nuclei were counterstained with 4',6-diamino-2-phenylindole (DAPI), and cells were visualized using a confocal microscope (LSM 980, Carl Zeiss).
  • ndSTAT1-TMD and ndSTAT1-TMD were tested using Cell Counting Kit-8 (CCK-8, Dojindo Laboratories, Kumamoto, Japan).
  • 2x10 5 mouse splenocytes were seeded in a 96-well culture plate (SPL) and incubated with various doses of ndSTAT1-TMD or ndSTAT1-TMD (V426D, T427D) for 1 hour.
  • CCK-8 reagent was added to the cultured cells and cultured for an additional 4 hours. Cell viability was measured at absorbance at a wavelength of 450 nm using a microplate reader (Bio-Rad).
  • CD4 + CD62L + naive T cells were purified from RBC-cleared spleen cells extracted from the spleens of 8-week-old female C57BL/6N (B6) mice using a CD4 + CD62L + T cell isolation kit (Miltenyi Biotec, Bergisch Gladbach, Germany). Isolated CD4 + naive T cells were grown in 96-well cell culture plates (Eppendorf) coated with 1 ⁇ g/ml anti-CD3 ⁇ (BD Bioscience, San Diego, CA, USA) and anti-CD28 (BD Bioscience) for 72 h. activated.
  • Th1 25 ng/ml of recombinant mouse IL-12 and 2 ⁇ g/ml of anti- IL-4 antibody
  • Th2 200 ng/ml recombinant mouse IL-4 and 2 ⁇ g/ml anti-IFN- ⁇ antibody
  • Th17 1 ng/ml recombinant mouse TGF- ⁇ 1, 25 ng/ml recombinant mouse IL) -6, 2 ⁇ g/ml anti-IFN- ⁇ antibody and 2 ⁇ g/ml anti-IL-4 antibody
  • iTreg 5 ng/ml recombinant mouse TGF- ⁇ and 20 ng/ml recombinant mouse IL-2) .
  • Cytokines and antibodies were purchased from PeproTech (Rocky Hill, NJ, USA), BioLegend (San Diego, CA, USA), and R&D Systems (Minneapolis, MN, USA). All cells were cultured at 37°C in humid conditions with 5% CO 2 .
  • Mouse serum samples were obtained by centrifugation of blood samples obtained from IMQ-induced psoriatic mice on day 7 of disease induction or from dextran sulfate sodium (DSS)-induced colitis mice on day 12 of disease induction. TNF- ⁇ , IL-1 ⁇ , and IL-6 levels in blood were measured by ELISA according to the manufacturer's protocol (Invitrogen).
  • the concentration of cytokines was measured at absorbance at a wavelength of 450 nm using a microplate reader (Bio-Rad).
  • ndSTAT1-TMD and ndSTAT1-TMD were tested for their binding ability to target gene promoters.
  • 5x10 HEK293T cells were seeded in 6-well culture plates (SPL) and incubated with 1 ⁇ g of pCMV6 vector containing mouse wild-type STAT1 and 1 ⁇ g of IL using Lipofectamine Reagents (Invitrogen) diluted in Opti-MEM (Gibco). -17A promoter-luciferase vector was co-transfected.
  • ndSTAT1-TMD or ndSTAT1-TMD V426D, T427D
  • V426D, T427D a dose-dependent manner (0.5-2 ⁇ M).
  • Cells were then cultured overnight, washed with PBS, and lysed using Cell Culture Lysis 5X Reagent (Promega, Madison, WI, USA).
  • Cell lysates were mixed with Luciferase Assay Substrate (Promega), and luciferase activity was measured with a Luminometer (Promega).
  • Mouse CD4 + T cells differentiated under Th1- or Th17-polarizing conditions with or without the addition of ndSTAT1-TMD were harvested after 72 h.
  • RNA extraction and sequencing were performed by Macrogen (Seoul, South Korea).
  • the Trimmomatic 0.38 program was used to remove low-quality data, adapters, and DNA contamination from the first reads.
  • the truncated data were then mapped to the genomic reference (10 mm) using the HISAT2 program.
  • Transcriptome assembly was performed using the reference-based aligned reads information, and TPM (transcript per million) counts were also obtained from the assembled transcriptome using StringTie.
  • GSEA Gene set enrichment analysis
  • mice 8- to 9-week-old female and male C57BL/6N (B6) mice (8 to 9 weeks) and 9-week-old female C57BL/6J (B6) mice were purchased from Orient Bio (Korea). All mice were bred and maintained in the semi-specific-pathogen-free (SPF) facility at the Yonsei Animal Research Center (YLARC). All animal studies were approved by the Institutional Animal Care and Use Committee (IACUC) of the Yonsei University Laboratory Animal Research Center (YLARC) and were performed in accordance with the guidelines of YLARC-IACUC for ethical use. (IACUC-A-202109-1327-01, IACUC-A-202201-1404-01).
  • SPPF semi-specific-pathogen-free
  • YLARC Yonsei Animal Research Center
  • Psoriasis Induction and scoring of psoriasis area and severity index (PASI)
  • ndSTAT1-TMD (20 or 60 ⁇ g/mouse), ndSTAT1-TMD (V426D, T427D) (60 ⁇ g/mouse), or anti-IL-17A antibody (BioXCell, Lebanon, NH, USA) (60 ⁇ g/mouse).
  • Intraperitoneal injections were administered every other day from days 1 to 5, and body weight and psoriasis area and severity index (PASI) scores were measured daily from days 1 to 7.
  • the exposed back skin of the mice was photographed on days 5 and 7, and both ears of the mice were photographed on day 7.
  • the thickness of the back or right ear skin was measured with a Vernier caliper for 7 consecutive days.
  • the Psoriasis Area and Severity Index (PASI) scores of all groups were assessed daily and measured in a blinded manner according to the following indices:
  • Erythema 0, flesh-colored pink; 1, slight redness over the entire surface; 2, medium red with dark red patches throughout the surface; 3, If there are dark red patches all over the surface.
  • Curing thickness: 0, 1-5%; 1, 5-50%; 2, 50-100%; 3, >100%.
  • Detachment 0, no peeling of skin; 1, minor dry spots without peeling; 2, dry patches over most of the skin with peeling; 3. Dry patches over most of the skin, with moderate to severe peeling over a large surface area.
  • mice After one week of acclimatization, 8-week-old male C57BL/6N mice were provided with sterile water or water containing 2.5% DSS from days 1 to 6. Water containing dextran sulfate sodium (DSS) was changed every other day. After 6 days, all mice were provided with sterile water. Mice were administered ndSTAT1-TMD (20 or 100 ⁇ g/mouse), ndSTAT1-TMD (V426D, T427D) (100 ⁇ g/mouse), or anti-IL-17A antibody (BioXCell, Lebanon, NH, USA) (100 ⁇ g/mouse). was injected intraperitoneally.
  • DAI Body weight and disease activity index
  • Weight change 0, 0-1%; 1, 1-5%; 2, 5-10%; 3, 10-20%; 4, >20%.
  • DSS dextran sulfate sodium
  • Inflammatory cell infiltration 0, no infiltration; 1, light penetration ⁇ 25%; 2, moderate penetration ⁇ 50%; 3, significant penetration >50%.
  • Crypt damage 0, none; 1, Some crypt damage; 2, larger space between crypts; 3, Large area without crypts.
  • Goblet cell depletion 0, none; 1, mild depletion ⁇ 25%; 2, moderate depletion ⁇ 50%; 3, significant depletion >50%.
  • the thickness of the muscle layer of the large intestine was measured using the Image J program. All tissue staining was performed at Korea CFC (Seoul, South Korea).
  • ndSTAT1-TMD transduction efficiency of ndSTAT1-TMD in mouse splenocytes was analyzed by intracellular staining using anti-FLAG-PE (BioLegend) after protein treatment for the indicated times.
  • activated CD4 + T cells were stained with anti-CD69-FITC (BD Bioscience) or anti-CD25-PE (BD Bioscience). After 72 hours of incubation with recombinant proteins, differentiated CD4 + effector T cell subsets were resuspended with Cell-Stimulation Cocktail (500X) (eBioscience, San Diego, CA, USA) for 4 hours . provoked. Each subset was then fixed and permeabilized with Fixation/Permeabilization Buffer (eBioscience) and intracellularly stained for appropriate lineage-defining transcription factors and cytokines. All antibodies were purchased from eBioscience.
  • Immune cells isolated from the spleen or draining lymph nodes (dLN) of IMQ-induced psoriasis mice and the spleen or mesenteric lymph nodes (mLN) of DSS-induced colitis mice were restimulated with Cell Stimulation Cocktail (500X) (eBioscience) for 4 hours. After restimulation, cells were first stained with anti-CD4-AF700 (eBioscience) before fixation and permeabilization. Intracellular cytokines and transcription factors were added with anti-IFN- ⁇ -APC, anti-IL-17A-APC, anti-T-bet-PE, anti-ROR- ⁇ t-PE, or anti-Foxp3-PE (eBioscience). was dyed with
  • Skin-infiltrating leukocytes were isolated from the back and ear skin of mice with IMQ-induced psoriasis. Their properties were analyzed by staining for appropriate surface markers or staining for intracellular transcription factors or cytokines. For intracellular staining, cells were restimulated for 4 h using Cell Stimulation Cocktail (500X) (eBioscience), fixed, and permeabilized. Cells were incubated with anti-CD45-AF700, anti-CD11b-APC, anti-IFN- ⁇ -PE, anti-IL-17A-PE, anti-T-bet-PE, anti-ROR- ⁇ t-PE (eBioscience), and anti- Stained with -F4/80-PE (BD Bioscience).
  • Cell Stimulation Cocktail 500X
  • eBioscience Cell Stimulation Cocktail
  • Nitro oxide (NO) production levels were measured using the Griess Reagent System according to the manufacturer's protocol (Peprotech). Nitric oxide levels in blood supernatant samples were measured by analyzing absorbance at 540 nm using a microplate reader (Bio-Rad).
  • Skin-infiltrating leukocytes were isolated from the back skin or left ear skin of IMQ-induced psoriasis mice. Seven days after IMQ application, a 1 cm 2 area was cut from the mouse back skin and left ear, the remaining IMQ cream was removed by washing with HBSS (Gibco), and the epidermis and dermis were separated using curved forceps. The separated epidermis and dermis were digested at 37°C using Dispase II solution (5 mg/ml, Sigma Aldrich).
  • the dermis was dissected into small pieces ( ⁇ 0.5%) using curved scissors in dermal dissociation buffer (DMEM (Hyclone, Roche) containing 1 mg/ml collagenase P and 100 ⁇ g/ml DNaseI (Hyclone, Roche)). mm) and incubated at 37°C for 1 hour.
  • the fully digested dermal suspension was filtered through a 40- ⁇ m cell strainer (Falcon), rinsed with DMEM medium containing 10% FBS (Hyclone), and separated dermal-infiltrating leukocytes were collected by centrifugation.
  • the front and back were quickly cut into small pieces ( ⁇ 2 mm) with curved scissors, transferred to 0.05% TE (Trypsin-EDTA, Hyclone) buffer, and incubated at 37°C for 5 minutes. After incubation, trypsin neutralizing solution (5% FBS diluted in HBSS) was added to the completely digested epidermis, and the epidermal suspension was filtered through a 40- ⁇ m cell strainer (Falcon). Isolated epidermal-infiltrating leukocytes were collected by centrifugation.
  • Results are expressed as mean ⁇ SEM (n ⁇ 3).
  • Statistical analysis of group differences used unpaired Student's t-test or ANOVA analysis followed by Dunnett's multiple comparison test. The number of asterisks indicates the results of statistical analysis as follows: ns; Not significant, *p ⁇ 0.05, **p ⁇ 0.01, ***p ⁇ 0.001 and ****p ⁇ 0.0001.
  • GraphPad Prism 9 was used for analysis of all data.
  • CD4 + naive T cells can differentiate into Th1 cells by adopting the STAT1 signaling pathway, which includes phosphorylation (p-STAT1), homodimerization, and subsequent processes of STAT1 translocation.
  • STAT1 signaling pathway which includes phosphorylation (p-STAT1)
  • p-STAT1 phosphorylation
  • homodimerization homodimerization
  • subsequent processes of STAT1 translocation Various genetic and therapeutic approaches have been used to analyze STAT1 function in vitro and in vivo .
  • STAT1 signaling pathway which includes phosphorylation (p-STAT1), homodimerization, and subsequent processes of STAT1 translocation.
  • ndSTAT1-TMD specifically and simultaneously inhibits the differentiation of na ⁇ ve CD4+ T cells into Th1 and Th17 T cell subsets.
  • T cell receptor (TcR) stimulation is an essential factor required for the successful differentiation of na ⁇ ve CD4 + T cells into various T cell subsets.
  • TcR T cell receptor
  • ndSTAT1-TMD or ndSTAT1-TMD(V426D, T427D) treatment of T cells affects T cell receptor-mediated early activation signaling events.
  • T The induced surface expression levels of CD69, CD25, and IL-2 secretion were measured after cell receptor stimulation.
  • Figures 1H and 1I the expression levels of CD25, CD69, and interleukin-2 (IL-2) were not affected by treatment with ndSTAT1-TMD or ndSTAT1-TMD (V426D, V427D).
  • na ⁇ ve CD4+ T cells were transfected with ndSTAT1-TMD or ndSTAT1-TMD ( Different concentrations of V426D, T427D) were induced to differentiate into each T cell subset under the appropriate T cell subset-skewing conditions.
  • interferon regulatory factor 1 known as a primary responder to IFN- ⁇ -STAT1 signaling in Th1 cells
  • IRF1 interferon regulatory factor 1
  • the level of interferon regulatory factor 1 (IRF1) decreased after ndSTAT1-TMD treatment, which is consistent with ndSTAT1 Unlike the Th1 group treated with -TMD (V426D, T427D), it decreased ( Figure 2c).
  • the expression levels of ROR- ⁇ t and intracellular IL-17A in Th17 cells were also significantly decreased by ndSTAT1-TMD in a concentration-dependent manner (Figure 2D).
  • ndSTAT1-TMD treatment reduced the secretion of IL-17A and IFN- ⁇ from Th17 cells ( Figures 2E-2F). Meanwhile, it has been suggested that pathogenic Th17 cells can secrete IL-17A and IFN- ⁇ .
  • ndSTAT1-TMD appeared to have no effect on regulating the expression of GATA3 or interleukin-4 (IL-4) in Th2 cells and Foxp3 expression in Treg cells.
  • ndSTAT1-TMD (V426D, T427D) treated cells showed a perceptible decrease in the expression of genes associated with immunological functions of Th1 or Th17 cells.
  • the level of reduction was substantially lower compared to cells treated with the same concentration of ndSTAT1-TMD.
  • Th1/17 cell differentiation by ndSTAT1-TMD is attributed to the transcriptional suppression of STAT1 target genes related to the function of Th1/17 cells of STAT1 target genes associated with the functions of Th1/17 cells).
  • ndSTAT1-TMD during Th1 and Th17 cell differentiation is due to disruption of endogenous STAT1 binding to target gene promoters at the transcriptional level.
  • STAT1 binds to the IL-17A promoter
  • a plasmid in which induction of the IL-17A promoter induces expression of a luciferase reporter gene was co-transfected into HEK293T cells together with a plasmid containing the gene encoding wild-type STAT1. .
  • the transfected cells were then incubated with various concentrations of ndSTAT1-TMD or ndSTAT1-TMD (V426D, T427D).
  • Activated STAT1 is involved in the generation of Th1 cells secreting IFN- ⁇ and Th17 cells secreting IL-17A in the early stage of lineage commitment. Disruption of such functional commitment of Th1 and/or Th17 cells during differentiation often leads to autoimmune diseases and immune dysfunction.
  • the mRNA expression profile of Th1 and Th17 cells treated with ndSTAT1-TMD was analyzed to evaluate the intrinsic role of ndSTAT1-TMD on the expression of genes related to the function of Th1 and Th17 cells.
  • GSEA Gene set enrichment analysis
  • mRNA sequencing data from ndSTAT1-TMD treated Th1 cells showed that genes involved in Th1 cell differentiation and function, such as Stat1 and Tbx21 , and genes required for the IFN- ⁇ response, such as Irf5 , were relatively downregulated. was regulated ( Figure 3D).
  • mRNA sequencing data generated from Th17 cells treated with ndSTAT1-TMD showed that the expression of genes involved in the function of Th17 cells, such as Rorc , Il17ra , and Ccl20, was significantly reduced, while the activity of Th17 cells, such as Osm and Eomes, was significantly reduced. Expression of genes known to be restrictive showed increased (Figure 3E).
  • ndSTAT1-TMD alleviates psoriasis symptoms induced by IMQ (The ndSTAT1-TMD treatment alleviates the symptoms of psoriasis induced by IMQ)
  • ndSTAT1-TMD had therapeutic potential for psoriasis.
  • Psoriasis was induced by applying IMQ (imiquimod) cream to the back and ear skin of mice for 7 consecutive days, and as a positive control, ndSTAT1-TMD, ndSTAT1-TMD (V426D, T427D) or anti-IL-17A antibody was applied 2 days from the initial disease induction date. It was administered via intraperitoneal injection at daily intervals (Figure 4A).
  • the psoriasis area and severity index (PASI) score measured by analyzing the back skin of psoriasis-induced mice, peaked on the 5th day. It then gradually decreased on day 6 or 7 throughout modeling ( Figure 4B).
  • ndSTAT1-TMD treatment showed significant improvement in pathological changes compared to anti-IL-17A antibody treatment ( Figure 4C).
  • the thickness of the back skin doubled in the psoriasis-induced group.
  • ndSTAT1-TMD treatment showed the most effective treatment function in the ear.
  • major pro-inflammatory cytokines secreted in the serum of each group of mice 7 days after IMQ induction such as TNF- ⁇ , IL-1 ⁇ , and IL-6
  • 60 ⁇ g of ndSTAT1-TMD or anti-IL-17A antibody A significant decrease in serum concentration was observed in the treated group ( Figure 4G).
  • Th1 cells and Th17 cells are important factors in the development of psoriasis and ndSTAT1-TMD inhibited the function and differentiation program of both Th1 and Th17 cells in vitro and in vivo
  • ndSTAT1-TMD treatment on animal models of psoriasis may be effective in vivo. It was hypothesized to alter the functional status of CD4 + T cell subsets. Profiles of CD4 + T cell subsets in the spleen and draining lymph nodes (dLNs) of animal models of psoriasis revealed T-bet + or IFN- ⁇ + CD4 + T cells and ROR- ⁇ t + in psoriasis-induced mice.
  • ndSTAT1-TMD restricts the infiltration of leukocytes into the skin of psoriasis-induced mice.
  • the histopathological features of the IMQ-induced psoriasis mouse model are human plaque-type psoriasis, such as acanthosis, parakeratosis, and neovascularization, along with infiltration of various leukocytes consisting of dendritic cells, neutrophils, macrophages, and T cells. It has been proven to be very similar to the multifactorial characteristics of . Additionally, several studies have reported that depletion of CD4 + T cells significantly reduces IMQ-induced skin inflammation.
  • ndSTAT1-TMD V426D, T427D
  • ndSTAT1-TMD improves the inflammatory state of the dermal region of psoriatic skin by limiting activated Th1 and Th17 cells as well as macrophages.
  • Colitis is another inflammatory disease in which Th1 or/and Th17 cells play a pathogenic role.
  • DSS dextran sulfate sodium
  • ndSTAT1-TMD nti-IL-17A antibody treatment served as a positive control.
  • Figure 7A The severity of IBD was assessed by starting treatment with ndSTAT1-TMD, ndSTAT1-TMD (V426D, T427D), or anti-IL-17A antibody on day 6 after colitis induction via intraperitoneal injection and monitoring disease progression daily for another 6 days. monitored.
  • colon sections were stained with H&E or PAS 6 days after intraperitoneal administration. Signs of inflammatory colon, such as increased infiltrating inflammatory cells, goblet cell depletion, crypt damage, and changes in muscle layer thickness, were improved in the anti-IL-17A antibody or ndSTAT1-TMD treated groups ( Figures 7E-7G). On the other hand, in the ndSTAT1-TMD treatment group (V426D, T427D), the severity of colitis remained at a severe level without any improvement ( Figures 7E-7G).
  • ndSTAT1-TMD could improve colitis symptoms by suppressing the pathogenic functions of inflammatory Th1 and Th17 cells.
  • the levels of T-bet + or IFN- ⁇ + CD4 + T cells and ROR- ⁇ t + or IL-17A + CD4 + T cells in the spleen and mesenteric lymph nodes (mLN) in IBD-induced mice treated with PBS It was significantly higher than that of healthy controls.
  • ndSTAT1-TMD or anti-IL-17A antibody treatment resulted in decreased T-bet + or IFN- ⁇ + CD4 + T cells and ROR- ⁇ t + or IL-17A + CD4 + T cells in both the spleen and mesenteric lymph nodes (mLNs). The number of cells was greatly reduced. Consistent with the therapeutic potential of ndSTAT1-TMD in psoriasis animal models, ndSTAT1-TMD showed dual inhibitory functions against Th1 cells and Th17 cells. The decrease in Th1 and Th17 cells by ndSTAT1-TMD treatment was accompanied by an increase in Foxp3 expressing CD4 + Treg cells in the spleen and mesenteric lymph nodes (mLN). Taking these results together, ndSTAT1-TMD treatment showed a therapeutic effect in DSS-induced colitis comparable to anti-IL-17A antibody treatment.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Organic Chemistry (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Immunology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Neurosurgery (AREA)
  • Rheumatology (AREA)
  • Obesity (AREA)
  • Biomedical Technology (AREA)
  • Neurology (AREA)
  • Diabetes (AREA)
  • Zoology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Hematology (AREA)
  • Epidemiology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Toxicology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)

Abstract

A fusion protein (ndSTAT1-TMD) provided in the present invention can be efficiently delivered to the nucleus of cells without affecting cytotoxicity and a TcR-mediated signaling event. In addition, the fusion protein provided in the present invention specifically inhibited the transcriptional activity of endogenous STAT1 in Th1 and Th17 cells, and showed similar therapeutic potential to an anti-IL-17A antibody when administered to psoriasis and IBD mouse models. Therefore, the fusion protein provided in the present invention regulates the functions of pathogenic Th1 and Th17 cells together and thus can be a new medicine for Th1/17 cell-mediated autoimmune diseases.

Description

STAT1-TMD의 면역 관련 질환 용도Use of STAT1-TMD in immune-related diseases
본 발명은 STAT1 (Signal transducer and activator of transcription 1, STAT1)과 단백질 운반 도메인 (Protein transduction domain, PTD)를 포함하는 신규 융합 단백질 및 이의 용도에 관한 것이다.The present invention relates to a novel fusion protein containing STAT1 (Signal transducer and activator of transcription 1, STAT1) and a protein transduction domain (PTD) and uses thereof.
CD4+ T 헬퍼 (Th) 세포는 다양한 면역 반응과 염증에서 필수적인 역할을 한다. 과도하게 활성화된 CD4+ 효과 T 세포로 인한 기능적 면역 네트워크의 항상성 불균형은 심각한 자가면역 질환을 유발하는 염증 반응의 시작 및 유지와 관련이 있다. 보다 구체적으로, 인터페론-γ (IFN-γ)를 분비하는 T 헬퍼 1 (Th1) 세포와 인터루킨-17 (IL-17)을 분비하는 T 헬퍼 17 (Th17) 세포는 건선 및 염증성 장질환을 포함한 장기 특이성 염증 질환의 발병기전과 관련이 있는 것으로 알려져 있다. 또한, T 헬퍼 1 (Th1) 세포와 T 헬퍼 17 (Th17) 세포는 퇴행성 뇌 또는 혈관 질환 및 비만 및 동맥경화를 포함하는 각종 대사성 질환의 발병기전과도 관련이 있는 것으로 알려져 있다.CD4 + T helper (Th) cells play an essential role in various immune responses and inflammation. Homeostatic imbalance in the functional immune network caused by excessively activated CD4 + effector T cells is associated with the initiation and maintenance of inflammatory responses that lead to serious autoimmune diseases. More specifically, T helper 1 (Th1) cells, which secrete interferon-γ (IFN-γ), and T helper 17 (Th17) cells, which secrete interleukin-17 (IL-17), are involved in long-term diseases, including psoriasis and inflammatory bowel disease. It is known to be related to the pathogenesis of specific inflammatory diseases. In addition, T helper 1 (Th1) cells and T helper 17 (Th17) cells are known to be related to the pathogenesis of various metabolic diseases, including degenerative brain or vascular diseases, obesity, and arteriosclerosis.
또한, T 헬퍼 1 세포 (Th1 세포)와 T 헬퍼 17 세포 (Th17 세포)는 건선 및 염증성 장질환(inflammatory bowel disease; IBD)을 포함한 다양한 자가면역질환의 병리 발생에 중추적인 역할을 한다. 또한, STAT1 (Signal transducer and activator of transcription 1)는 초기 단계에서 Th1 및 Th17 세포 계통 약속 (lineage commitment)을 조절하고 시험관 내 및 생체 내에서 면역학적 기능을 유지합니다. STAT1을 차단하는 이전의 전략은 자가면역질환을 치료하는 기능을 억제하지만 동시에 Th17 세포의 과활성화를 유발한다. Additionally, T helper 1 cells (Th1 cells) and T helper 17 cells (Th17 cells) play a central role in the pathogenesis of various autoimmune diseases, including psoriasis and inflammatory bowel disease (IBD). Additionally, STAT1 (Signal transducer and activator of transcription 1) regulates Th1 and Th17 cell lineage commitment at early stages and maintains immunological functions in vitro and in vivo. Previous strategies to block STAT1 inhibit its ability to treat autoimmune diseases but also cause hyperactivation of Th17 cells.
한편, 단백질 운반 도메인 (Protein transduction domain, PTD)은 소수성이 강한 짧은 펩타이드로 함께 융합된 단백질이나 DNA, RNA 등의 생리활성분자를 세포 내로 효과적으로 전달하는 것으로 알려져 있다. 단백질 운반 도메인은 세포질뿐만 아니라 핵까지도 생리활성분자를 운반할 수 있기 때문에, 다양한 단백질을 핵 안으로 전달하는 데 적합한 특성을 지니고 있다.Meanwhile, the protein transduction domain (PTD) is a short peptide with strong hydrophobicity and is known to effectively deliver bioactive molecules such as proteins, DNA, and RNA fused together into cells. Since the protein transport domain can transport bioactive molecules not only to the cytoplasm but also to the nucleus, it has characteristics suitable for delivering various proteins into the nucleus.
본 발명의 목적은 STAT1의 전사 조절 도메인(Transcription modulation domain, TMD) 및 단백질 운반 도메인 (Protein transduction domain, PTD)을 포함하는 융합 단백질을 제공하는 것이다.The purpose of the present invention is to provide a fusion protein containing the transcription modulation domain (TMD) and protein transduction domain (PTD) of STAT1.
본 발명의 다른 목적은 핵산분자를 발현시켜 복수의 단백질 운반 도메인(Protein transduction domain)과 전사 조절 도메인(Transcription modulation domain)를 포함하는 융합 단백질을 얻는 단계; 및 상기 단계에서 얻어진 융합 단백질을 정제하는 단계를 포함하는 복수의 단백질 운반 도메인(Protein transduction domain)과 전사 조절 도메인(Transcription modulation domain)을 포함하는 융합 단백질의 제조방법을 제공하는 것이다.Another object of the present invention is to obtain a fusion protein containing a plurality of protein transduction domains and a transcription modulation domain by expressing a nucleic acid molecule; and purifying the fusion protein obtained in the above step. To provide a method for producing a fusion protein containing a plurality of protein transduction domains and a transcription modulation domain.
본 발명의 또 다른 목적은 상기 융합 단백질을 포함하는 면역관련질환의 치료, 개선 또는 예방용 약리학적 조성물을 제공하는 것이다.Another object of the present invention is to provide a pharmacological composition containing the fusion protein for the treatment, improvement or prevention of immune-related diseases.
그러나 본 발명이 이루고자 하는 기술적 과제는 이상에서 언급한 과제에 제한되지 않으며, 언급되지 않은 또 다른 과제들은 아래의 기재로부터 당 업계에서 통상의 지식을 가진 자에게 명확하게 이해될 수 있을 것이다.However, the technical problem to be achieved by the present invention is not limited to the problems mentioned above, and other problems not mentioned will be clearly understood by those skilled in the art from the description below.
본 발명에 기재된 다양한 실시예가 도면을 참조로 기재된다. 하기 설명에서, 본 발명의 완전한 이해를 위해서, 다양한 특이적 상세 사항, 예컨대, 특이적 형태, 조성물 및 공정 등이 기재되어 있다. 그러나, 특정의 구체예는 이들 특이적 상세 사항 중 하나 이상 없이, 또는 다른 공지된 방법 및 형태와 함께 실행될 수 있다. 다른 예에서, 공지된 공정 및 제조 기술은 본 발명을 불필요하거나 모호하게 하지 않기 위해서, 특정의 상세사항으로 기재되지 않는다. 한 가지 실시예에 대한 본 명세서 전체를 통한 참조는 실시예와 결부되어 기재된 특별한 특징, 형태, 조성 또는 특성이 본 발명의 하나 이상의 실시예에 포함됨을 의미한다. 따라서, 본 명세서 전체에 걸친 다양한 위치에서 표현한 실시예의 상황은 반드시 본 발명의 동일한 실시예를 나타내지는 않는다. 추가로, 특별한 특징, 형태, 조성, 또는 특성은 하나 이상의 실시예에서 어떠한 적합한 방법으로 조합될 수 있다.Various embodiments described in the present invention are described with reference to the drawings. In the following description, various specific details, such as specific forms, compositions, and processes, are set forth in order to provide a thorough understanding of the invention. However, certain embodiments may be practiced without one or more of these specific details or in conjunction with other known methods and forms. In other instances, well-known processes and manufacturing techniques are not described in specific detail so as not to unnecessarily or obscure the invention. Reference throughout this specification to one embodiment means that a particular feature, feature, composition or characteristic described in connection with the embodiment is included in one or more embodiments of the invention. Accordingly, the contexts of embodiments expressed in various places throughout this specification do not necessarily represent the same embodiment of the invention. Additionally, particular features, shapes, compositions, or properties may be combined in any suitable way in one or more embodiments.
1. STAT1 (Signal transducer and activator of transcription 1)의 전사 조절 도메인 및 단백질 운반 도메인 (Protein transduction domain, PTD)을 포함하는 융합 단백질1. Fusion protein containing the transcriptional regulatory domain and protein transduction domain (PTD) of STAT1 (Signal transducer and activator of transcription 1)
본 발명의 일 구현예에서는 STAT1 (Signal transducer and activator of transcription 1)의 전사 조절 도메인 및 단백질 운반 도메인 (Protein transduction domain, PTD)을 포함하는 융합 단백질을 제공한다.One embodiment of the present invention provides a fusion protein comprising a transcriptional regulatory domain and a protein transduction domain (PTD) of STAT1 (Signal transducer and activator of transcription 1).
T 헬퍼 1 세포 (Th1 세포)와 T 헬퍼 17 세포 (Th17 세포)는 건선 및 염증성 장 질환 (inflammatory bowel disease; IBD)을 비롯한 다양한 자가면역 질환의 발병에서 중추적인 역할을 한다. 또한, T 헬퍼 1 (Th1) 세포와 T 헬퍼 17 (Th17) 세포는 퇴행성 뇌 또는 혈관 질환, 각종 염증성 질환 및 비만과 동맥경화를 포함하는 각종 대사성 질환의 발병기전과도 관련이 있는 것으로 알려져 있다. T helper 1 cells (Th1 cells) and T helper 17 cells (Th17 cells) play a pivotal role in the pathogenesis of various autoimmune diseases, including psoriasis and inflammatory bowel disease (IBD). In addition, T helper 1 (Th1) cells and T helper 17 (Th17) cells are known to be related to the pathogenesis of various metabolic diseases, including degenerative brain or vascular diseases, various inflammatory diseases, obesity, and arteriosclerosis.
STAT1 (Signal transducer and activator of transcription 1)은 초기 단계에서 Th1 및 Th17 세포 계통 약속 (lineage commitment)을 조절하고 시험관 내 및 생체 내에서 면역 기능을 유지한다. 자가면역 질환을 치료하기 위해 STAT1 기능을 차단하는 이전 전략은 Th1 세포 활성을 억제하는 동시에 Th17 세포의 과다 활성화를 유발한다. 본 발명에서는 정상적인 생리학적 조건에서 유전적 변형 없이 병원성 Th1 및 Th17 세포의 기능을 조절하기 위해 표적의 핵으로 전달될 수 있는 STAT1 전사 조절 도메인의 핵 전달 형태 (ndSTAT1-TMD)를 생성하였다. 본 발명에서 제공하는 상기 융합 단백질은 세포 생존력과 T 세포 활성화 신호 전달 이벤트에 영향을 주지 않으면서 용량 및 시간 의존 방식으로 세포를 보호한다. 본 발명에서 제공하는 ndSTAT1-TMD는 내인성 STAT1 매개 전사의 경쟁적 억제를 통해 나이브 CD4+ T 세포의 Th1 또는 Th17 세포로의 분화를 유의하게 차단했으며, 이는 Th2 및 Treg 세포 분화에 영향을 미치지 않았다.Signal transducer and activator of transcription 1 (STAT1) regulates Th1 and Th17 cell lineage commitment at early stages and maintains immune function in vitro and in vivo. Previous strategies to block STAT1 function to treat autoimmune diseases suppress Th1 cell activity while simultaneously causing hyperactivation of Th17 cells. In the present invention, we generated a nuclear delivery form (ndSTAT1-TMD) of the STAT1 transcriptional regulatory domain that can be delivered to the nucleus of the target to regulate the function of pathogenic Th1 and Th17 cells without genetic modification under normal physiological conditions. The fusion protein provided by the present invention protects cells in a dose- and time-dependent manner without affecting cell viability and T cell activation signaling events. The ndSTAT1-TMD provided by the present invention significantly blocked the differentiation of naïve CD4 + T cells into Th1 or Th17 cells through competitive inhibition of endogenous STAT1-mediated transcription, and did not affect Th2 and Treg cell differentiation.
또한, 본 발명에서 제공하는 ndSTAT1-TMD 처리 후 Th1 또는 Th17 세포의 유전자 발현 프로파일을 mRNA 서열분석으로 분석한 결과, Th1 또는 Th17 세포의 분화능 및 면역학적 기능에 관여하는 유전자의 발현이 실질적으로 감소하였다. 본 발명에서 제공하는 ndSTAT1-TMD의 치료 가능성은 건선 및 대장염의 동물 모델에서 테스트되었으며, 이의 발병은 주로 Th1 및/또는 Th17 세포에 의해 발생한다. 건선 및 대장염의 증상 및 진행은 항-IL-17A 항체 치료와 유사하게 본 발명에서 제공하는 ndSTAT1-TMD 치료에 의해 상당히 완화되었다. 결론적으로, 본 발명은 ndSTAT1-TMD가 병원성 Th1 및 Th17 세포의 기능을 함께 조절함으로써 Th1/17 세포 매개 자가면역질환에 대한 새로운 치료제가 될 수 있음을 보여준다.In addition, as a result of analyzing the gene expression profile of Th1 or Th17 cells after treatment with ndSTAT1-TMD provided by the present invention by mRNA sequencing, the expression of genes involved in the differentiation capacity and immunological function of Th1 or Th17 cells was substantially reduced. . The therapeutic potential of ndSTAT1-TMD provided by the present invention has been tested in animal models of psoriasis and colitis, the pathogenesis of which is mainly caused by Th1 and/or Th17 cells. The symptoms and progression of psoriasis and colitis were significantly alleviated by the ndSTAT1-TMD treatment provided by the present invention, similar to anti-IL-17A antibody treatment. In conclusion, the present invention shows that ndSTAT1-TMD can be a new therapeutic agent for Th1/17 cell-mediated autoimmune diseases by jointly regulating the functions of pathogenic Th1 and Th17 cells.
Th1 세포는 IFN-γ를 분비하여 대식세포와 수지상 세포 (dendritic cell; DC)를 모두 활성화함으로써 침입한 세포내 병원균과 바이러스로부터 우리 몸을 보호하는 것으로 알려져 있다. 분비된 IFN-γ는 초기 단계에서 STAT1의 신호 전달 경로를 활성화하여 Th1 분화 유도제로도 작용한다. Th17 세포는 주로 세포외 박테리아와 곰팡이 제거에 관여하는 IL-17과 IL-23을 분비한다. 더욱이, Th17 세포 계통 발달 동안 IFN-γ의 존재는 Th17 세포 분화의 완전한 약속을 약화시킨다. Th1 및 Th17 세포가 독특하게 갖고 있는 이러한 모든 특성 외에도, 과활성화된 Th1 및 Th17 세포는 보조 T 세포와 조절 T 세포 사이의 면역학적 균형을 파괴하고 후속 사이토카인 조절 장애를 동반하여 자가면역 장애에서 중요한 역할을 한다.Th1 cells are known to protect our bodies from invading intracellular pathogens and viruses by secreting IFN-γ to activate both macrophages and dendritic cells (DC). Secreted IFN-γ also acts as a Th1 differentiation inducer by activating the STAT1 signaling pathway in the early stages. Th17 cells secrete IL-17 and IL-23, which are mainly involved in eliminating extracellular bacteria and fungi. Moreover, the presence of IFN-γ during Th17 cell lineage development undermines the full commitment of Th17 cell differentiation. In addition to all these unique properties of Th1 and Th17 cells, hyperactivated Th1 and Th17 cells play an important role in autoimmune disorders by disrupting the immunological balance between helper and regulatory T cells and with subsequent cytokine dysregulation. Do it.
STAT 계열의 구성원인 STAT1은 CD4+ T 세포 분화의 초기 단계 동안 T 헬퍼 하위 집합 특이적 유전자의 발현 패턴을 제어하여 Th1/Th2 및 Th1/17 세포 분화의 축을 조절한다. STAT1은 N 말단 도메인 (N-terminal domain; NTD), 코일드 코일 도메인 (coiled-coil domain; CCD), DNA 결합 도메인 (DNA binding domain; DBD), 링커 도메인 (linker domain; LD), SCR2 상동성 도메인 (SCR2 homology domain; SH2), 트랜스활성화 도메인 (transactivation domain; TAD)을 포함한 6개의 도메인으로 구성된다. IFN-γ 및 IL-27이 나이브 CD4+ T 세포 표면의 각각의 수용체에 결합하여 세포내 신호전달이 STAT1의 활성화 및 동종이량체화를 유발한다. STAT1 동종이량체는 핵으로 전위되어 Th1 반응에 관여하는 전사 인자인 T-bet 발현을 유도하고 유지한다. STAT1은 또한 Th17 반응의 조절에 결정적으로 관여하는 각 ROR-γt 및 IL-17A 발현의 해당 염색체 유전자 위치인 Rorc 또는 Il17a 유전자좌에 직접 결합하여 Th17 세포 분화를 억제하는 것으로 나타났다. STAT1-녹아웃(knockout; KO) 연구에서는 내인성 STAT1의 손실이 Th17 세포 계통 약속을 손상시키는 것으로 보고되었다. 유전적 방법을 이용한 STAT1의 기능적 억제가 Th1 또는 Th17 세포 매개 전신 자가면역 질환의 치료 전략으로 지속적으로 제시되어 왔으며, Th1 세포를 제한하는 실질적인 효과를 보여주었다. 그러나 Th17 세포의 중요한 기능적 조절은 감지되지 않았으며 STAT1이 없거나 낮은 수준의 Th17 세포는 과다 활성화된 표현형을 나타냈다.STAT1, a member of the STAT family, regulates the axes of Th1/Th2 and Th1/17 cell differentiation by controlling the expression pattern of T helper subset-specific genes during the early stages of CD4 + T cell differentiation. STAT1 has an N-terminal domain (NTD), coiled-coil domain (CCD), DNA binding domain (DBD), linker domain (LD), and SCR2 homology. It consists of six domains, including a SCR2 homology domain (SH2) and a transactivation domain (TAD). When IFN-γ and IL-27 bind to their respective receptors on the surface of naïve CD4 + T cells, intracellular signaling causes activation and homodimerization of STAT1. STAT1 homodimers translocate to the nucleus and induce and maintain the expression of T-bet, a transcription factor involved in Th1 responses. STAT1 has also been shown to inhibit Th17 cell differentiation by directly binding to the Rorc or Il17a loci, the corresponding chromosomal loci of ROR-γt and IL-17A expression, respectively, which are critically involved in the regulation of Th17 responses. STAT1-knockout (KO) studies reported that loss of endogenous STAT1 impairs Th17 cell lineage commitment. Functional inhibition of STAT1 using genetic methods has been consistently proposed as a treatment strategy for Th1 or Th17 cell-mediated systemic autoimmune diseases and has shown substantial effects in limiting Th1 cells. However, no significant functional regulation of Th17 cells was detected, and Th17 cells lacking or with low levels of STAT1 displayed a hyperactivated phenotype.
본 명세서에서 사용되는 용어 “전사 조절 도메인” (Transcription modulation domain, TMD)은 전사 인자를 구성하는 도메인으로 트랜스 액티베이션 도메인이 없이 DNA 결합 부위만으로 구성된 도메인을 의미한다. 하기 실시예에서 입증한 바와 같이, 본 발명의 융합 단백질은 트랜스 액티베이션 도메인은 없으나 DNA 결합 부위를 가지고 있기 때문에 목적하는 프로모터(예를 들면 Foxp3와 CD25)에 결합할 수는 있지만 전사를 촉진할 수 없다. 따라서 본 발명의 융합 단백질은 Foxp3와 CD25 유전자에 대한 우성음성 돌연변이체(dominant negative mutant)이기 때문에 세포 내의 야생형 STAT1에 대한 경쟁적 억제제로 작용하여 STAT1의 전사 및 활성을 억제할 수 있다.As used herein, the term “transcription modulation domain” (TMD) refers to a domain that constitutes a transcription factor and consists of only a DNA binding site without a transactivation domain. As demonstrated in the following examples, the fusion protein of the present invention does not have a transactivation domain but has a DNA binding site, so it can bind to the desired promoter (e.g. Foxp3 and CD25) but cannot promote transcription. . Therefore, since the fusion protein of the present invention is a dominant negative mutant for the Foxp3 and CD25 genes, it can act as a competitive inhibitor for wild-type STAT1 in cells and inhibit the transcription and activity of STAT1.
본 발명에서 상기 “단백질 운반 도메인(PTD)"은 7~50개의 아미노산으로 이루어져 있는 소수성이 강한 짧은 펩타이드로, 120 kDa 이상의 분자량을 단백질뿐만 아니라, DNA 또는 RNA를 세포 내로 전달할 수 있는 도메인을 의미한다. In the present invention, the “protein transport domain (PTD)” is a short, highly hydrophobic peptide consisting of 7 to 50 amino acids, and refers to a domain capable of delivering not only proteins with a molecular weight of 120 kDa or more, but also DNA or RNA into cells. .
본 발명에서 상기 단백질 운반 도메인은 Hph-1, Mph-1, Sim-2, Tat, VP22, Antp(antennapedia), Pep-1(peptide-1), PTD-5(protein transduction domain-5), 11R, 7R 및 CTP(cytoplamic transduction peptide)로 구성된 군으로부터 선택될 수 있고, 혹은 거대분자 운반 도메인(macromolecule transduction domain)을 사용할 수 있으나, 이에 제한되는 것은 아니며 당해 기술분야에서 일반적으로 사용되거나 시중에 판매되는 단백질 운반 도메인이라면 제한없이 사용될 수 있다. 바람직하게는 서열번호 1로 표시되는 CTP, 서열번호 2로 표시되는 Hph-1, 또는 서열번호 3으로 표시되는 Tat, 서열번호 4로 표시되는 Sim-2, 서열번호 5로 표시되는 Mph-1, 서열번호 6으로 표시되는 VP22, 서열번호 7로 표시되는 Antp, 서열번호 8로 표시되는 Pep-1, 서열번호 9로 표시되는 PTD-5, 서열번호 10으로 표시되는 7R, 서열번호 11로 표시되는 11R, 서열번호 12로 표시되는 CTP 중 어느 하나일 수 있다.In the present invention, the protein transport domains include Hph-1, Mph-1, Sim-2, Tat, VP22, Antp (antennapedia), Pep-1 (peptide-1), PTD-5 (protein transduction domain-5), and 11R. , 7R, and CTP (cytoplamic transduction peptide), or a macromolecule transduction domain may be used, but is not limited thereto, and is commonly used or commercially available in the art. Any protein transport domain can be used without limitation. Preferably CTP represented by SEQ ID NO: 1, Hph-1 represented by SEQ ID NO: 2, or Tat represented by SEQ ID NO: 3, Sim-2 represented by SEQ ID NO: 4, Mph-1 represented by SEQ ID NO: 5, VP22 represented by SEQ ID NO: 6, Antp represented by SEQ ID NO: 7, Pep-1 represented by SEQ ID NO: 8, PTD-5 represented by SEQ ID NO: 9, 7R represented by SEQ ID NO: 10, and SEQ ID NO: 11. It may be any one of 11R and CTP represented by SEQ ID NO: 12.
본 발명에서 상기 단백질 운반 도메인은 Hph-1, Mph-1, Sim-2, Tat, VP22, Antp(antennapedia), Pep-1(peptide-1), PTD-5(protein transduction domain-5), 11R, 7R 및 CTP(cytoplamic transduction peptide)로 구성된 군으로부터 선택되는 경우에는, 상기 Hph-1, Mph-1, Sim-2, Tat, VP22, Antp(antennapedia), Pep-1(peptide-1), PTD-5(protein transduction domain-5), 11R, 7R 및 CTP(cytoplamic transduction peptide)로 구성된 군으로부터 선택되는 하나의 도메인이 1개 이상, 예를 들면, 2개, 3개, 4개 또는 그 이상 반복되어 포함될 수 있다. 본 발명에서 상기 단백질 운반 도메인의 개수는 특별히 제한되지는 않으나 2개 이상, 10개 이하일 수 있고, 2개 이상, 5개 이하일 수 있으며, 2개 이상 3개 이하일 수 있다.In the present invention, the protein transport domains include Hph-1, Mph-1, Sim-2, Tat, VP22, Antp (antennapedia), Pep-1 (peptide-1), PTD-5 (protein transduction domain-5), and 11R. , 7R and CTP (cytoplamic transduction peptide), Hph-1, Mph-1, Sim-2, Tat, VP22, Antp (antennapedia), Pep-1 (peptide-1), PTD One or more domains selected from the group consisting of -5 (protein transduction domain-5), 11R, 7R, and CTP (cytoplamic transduction peptide) are repeated, for example, 2, 3, 4 or more times. can be included. In the present invention, the number of protein transport domains is not particularly limited, but may be 2 or more and 10 or less, 2 or more and 5 or less, and 2 or more and 3 or less.
2. 상기 융합 단백질을 암호화하는 핵산 분자, 지질 나노입자, 발현 벡터, 숙주 세포2. Nucleic acid molecules, lipid nanoparticles, expression vectors, host cells encoding the fusion protein
본 발명의 다른 구현 예에 따르면, 본 발명에서 제공하는 융합 단백질을 암호화하는 핵산 분자를 제공한다. According to another embodiment of the present invention, a nucleic acid molecule encoding the fusion protein provided by the present invention is provided.
본 발명의 핵산 분자는 본 발명에서 제공하는 융합 단백질의 아미노산 서열을 당업자에게 알려진 바와 같이 폴리뉴클레오티드 서열로 번역된 핵산 분자 모두를 포함한다. 그러므로 ORF(open reading frame)에 의한 다양한 폴리뉴클레오티드 서열이 제조될 수 있으며 이 또한 모두 본 발명의 핵산 분자에 포함된다.The nucleic acid molecule of the present invention includes all nucleic acid molecules in which the amino acid sequence of the fusion protein provided by the present invention has been translated into a polynucleotide sequence as known to those skilled in the art. Therefore, various polynucleotide sequences can be prepared by ORF (open reading frame), and all of these are also included in the nucleic acid molecule of the present invention.
본 발명의 또 다른 구현 예에 따르면, 본 발명에서 제공하는 상기 단리된 핵산 분자가 삽입된 발현 벡터를 제공한다.According to another embodiment of the present invention, an expression vector is provided into which the isolated nucleic acid molecule provided by the present invention is inserted.
본 발명에서 상기 "벡터"는 어떤 핵산 분자가 연결된 또 다른 핵산을 수송할 수 있는 상기 핵산 분자이다. 벡터의 한 가지 유형은, 추가적인 DNA 세그멘트가 결찰될 수 있는 원형 이중가닥 DNA를 가리키는 "플라스미드"이다. 또 다른 유형의 벡터는 파지 벡터이다. 또 다른 유형의 벡터는 바이러스성 벡터로, 추가적인 DNA 세그멘트가 바이러스 게놈에 결찰될 수 있다. 어떤 벡터들은 그들이 유입된 숙주세포에서 자율적인 복제를 할 수 있다(예컨대, 박테리아성 벡터는 박테리아성 복제 기원을 갖는 에피솜 포유류 벡터). 기타 벡터(예컨대, 비-에피솜 포유류 벡터)는 숙주세포에 유입되면서 숙주세포의 게놈에 통합될 수 있고, 그럼으로써, 숙주 게놈과 함께 복제된다. 뿐만 아니라, 어떤 벡터는 이들이 작동차원에서 연결된 유전자의 발현을 지시할 수 있다. 이와 같은 벡터는 본원에서 "재조합 발현 벡터" 또는 단순히 "발현 벡터"라 명명된다. 일반적으로 재조합 DNA 기법에서 유용한 발현 벡터는 종종 플라스미드의 형태로 존재한다. 본 명세서에서, "플라스미드"와 "벡터"는, 플라스미드가 벡터 중 가장 통상적으로 사용되는 형태이기 때문에, 상호 교환하여 사용될 수 있다. In the present invention, the “vector” is a nucleic acid molecule capable of transporting another nucleic acid to which a nucleic acid molecule is linked. One type of vector is a "plasmid," which refers to a circular double-stranded DNA into which additional DNA segments can be ligated. 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. Some vectors are capable of autonomous replication in the host cell into which they are introduced (e.g., bacterial vectors are episomal mammalian vectors that have a bacterial origin of replication). Other vectors (e.g., non-episomal mammalian vectors) can enter a host cell and become integrated into the host cell's genome, thereby replicating along with the host genome. In addition, some vectors can direct the expression of genes to which they are operationally linked. Such vectors are referred to herein as “recombinant expression vectors” or simply “expression vectors”. In general, expression vectors useful in recombinant DNA techniques often exist in the form of plasmids. In this specification, “plasmid” and “vector” may be used interchangeably since plasmid is the most commonly used form of vector.
본 발명에서 상기 발현 벡터의 구체적인 예시로는 상업적으로 널리 사용되는 pCDNA 벡터, F, R1, RP1, Col, pBR322, ToL, Ti 벡터; 코스미드; 람다, 람도이드(lambdoid), M13, Mu, p1 P22, Qμμ, T-even, T2, T3, T7 등의 파아지; 식물 바이러스로 이루어진 군으로부터 선택될 수 있으나, 이에 제한되는 것은 아니며, 당업자에게 발현 벡터로 알려진 모든 발현 벡터는 본 발명에 사용 가능하고, 발현 벡터를 선택할 때에는 목적으로 하는 숙주 세포의 성질에 따른다. 숙주세포로의 벡터 도입 시 인산칼슘 트랜스펙션, 바이러스 감염, DEAE-덱스트란 조절 트랜스펙션, 리포펙타민 트랜스펙션 또는 전기천공법에 의해 수행될 수 있으나 이에 한정되는 것은 아니며 당업자는 사용하는 발현 벡터 및 숙주 세포에 알맞은 도입 방법을 선택하여 이용할 수 있다. 바람직하게 벡터는 하나 이상의 선별 마커를 함유하나 이에 한정되지 않으며, 선별 마커를 포함하지 않은 벡터를 이용하여 생산물 생산 여부에 따라 선별이 가능하다. 선별 마커의 선택은 목적하는 숙주 세포에 의해 선별되며, 이는 이미 당업자에게 알려진 방법을 이용하므로 본 발명은 이에 제한을 두지 않는다. Specific examples of the expression vector in the present invention include widely used commercially pCDNA vectors, F, R1, RP1, Col, pBR322, ToL, and Ti vectors; cosmid; Phages such as lambda, lambdoid, M13, Mu, p1 P22, Qμμ, T-even, T2, T3, T7; It may be selected from the group consisting of plant viruses, but is not limited thereto, and all expression vectors known to those skilled in the art as expression vectors can be used in the present invention, and when selecting an expression vector, it depends on the properties of the target host cell. Introduction of vectors into host cells may be performed by calcium phosphate transfection, viral infection, DEAE-dextran control transfection, lipofectamine transfection, or electroporation, but is not limited thereto, and those skilled in the art will use the method. An introduction method suitable for the expression vector and host cell can be selected and used. Preferably, the vector contains one or more selection markers, but is not limited to this, and selection can be made depending on whether or not the product is produced using a vector that does not contain a selection marker. The selection of the selection marker is selected based on the target host cell, and since this method is already known to those skilled in the art, the present invention is not limited thereto.
본 발명의 핵산 분자를 정제를 용이하게 하기 위하여 태그 서열을 발현 벡터 상에 삽입하여 융합시킬 수 있다. 상기 태그로는 헥사-히스티딘 태그, 헤마글루티닌 태그, myc 태그 또는 flag 태그를 포함하나 이에 한정되는 것은 아니며 당업자에게 알려진 정제를 용이하게 하는 태그는 모두 본 발명에서 이용 가능하다. To facilitate purification of the nucleic acid molecule of the present invention, a tag sequence can be inserted and fused into an expression vector. The tag includes, but is not limited to, a hexa-histidine tag, a hemagglutinin tag, a myc tag, or a flag tag, and any tag that facilitates purification known to those skilled in the art can be used in the present invention.
본 발명의 또 다른 구현 예에 따르면, 본 발명에서 제공하는 발현 벡터가 형질 전환된 숙주 세포주를 제공한다.According to another embodiment of the present invention, a host cell line transformed with the expression vector provided by the present invention is provided.
본 발명에서 상기 "숙주 세포"에는 폴리펩티드 삽입물의 편입을 위한 벡터(들)의 수령자(recipient)일 수 있거나 또는 수령자였던 개별적인 세포 또는 세포배양물이 포함된다. 숙주 세포에는 단일 숙주 세포의 자손이 포함되고, 상기 자손은 자연적인, 우발적인 또는 고의의 돌연변이 때문에 반드시 원래 모세포와 완전히 동일(형태학상 또는 게놈 DNA 보완체에서)하지 않을 수 있다. 숙주 세포에는 본원의 폴리펩티드(들)로 체내에서 형질주입된 세포가 포함된다.In the present invention, the “host cell” includes an individual cell or cell culture that can be or has been a recipient of vector(s) for incorporation of a polypeptide insert. A host cell includes the progeny of a single host cell, which progeny may not necessarily be completely identical (morphologically or in genomic DNA complement) to the original parent cell due to natural, accidental or intentional mutations. Host cells include cells transfected in vivo with the polypeptide(s) herein.
본 발명에 있어서, 상기 숙주 세포로는 포유동물, 식물, 곤충, 균류 또는 세포성 기원의 세포를 포함할 수 있고, 예를 들면 대장균, 스트렙토미세스, 살모넬라 티피뮤리움 등의 박테리아 세포; 효모 세포, 피치아 파스 토리스 등의 균류세포; 드로조필라, 스포도프테라 Sf9 세포 등의 곤충 세포; CHO(중국 햄스터 난소 세포, Chinese hamster ovary cells), SP2/0(생쥐 골수종), 인간 림프아구(Human lymphoblastoid), COS, NSO(생쥐 골 수종), 293T, 보우 멜라노마 세포, HT-1080, BHK(베이비 햄스터 신장세포, Baby Hamster Kidney cells), HEK(인간 배아신장 세포, Human Embryonic Kidney cells) 또는 PERC.6(인간 망막 세포)의 동물 세포; 또는 식물 세포일 수 있으나, 이에 제한되는 것은 아니며, 당업자에게 알려진 숙주 세포주로 사용 가능한 세포는 모두 이용 가능하다.In the present invention, the host cells may include cells of mammalian, plant, insect, fungal, or cellular origin, for example, bacterial cells such as Escherichia coli, Streptomyces, and Salmonella Typhimurium; Fungal cells such as yeast cells and Pichia pastoris; Insect cells such as Drozophila and Spodoptera Sf9 cells; CHO (Chinese hamster ovary cells), SP2/0 (mouse myeloma), human lymphoblastoid, COS, NSO (mouse myeloma), 293T, Bow melanoma cells, HT-1080, BHK Animal cells of (Baby Hamster Kidney cells), HEK (Human Embryonic Kidney cells) or PERC.6 (Human Retinal Cells); Or it may be a plant cell, but is not limited thereto, and any cell that can be used as a host cell line known to those skilled in the art can be used.
본 발명의 또 다른 구현예에 따르면 상기 핵산 분자를 포함하는 지질 나노입자 (lipid nanoparticle, LNP)를 제공한다. 본 발명에 있어서, "나노입자"는 하나 이상의 지질 및 하나 이상의 치료제를 포함하는 입자이다. 나노입자는 전형적으로, 대략 마이크로미터 또는 그보다 작은 크기이고 지질층을 포함할 수 있다. 상기 지질 나노입자(LNP)란, 인접한 지질 이중층을 갖는 소포체, 예컨대, 구형 소포체를 지칭한다. 지질 나노입자는 제약 요법이 표적 위치에 전달되도록 하는 방법에 사용될 수 있다. LNP의 비-제한적 예로는 리포솜, 볼양친매체, 고체 지질 나노입자 (SLN), 나노구조화된 지질 운반체 (NLC) 및 단층막 구조체 (예컨대, 아키오좀(archaeosome) 및 미셀)를 포함한다.According to another embodiment of the present invention, lipid nanoparticles (LNPs) containing the nucleic acid molecules are provided. In the present invention, “nanoparticles” are particles containing one or more lipids and one or more therapeutic agents. Nanoparticles are typically on the order of micrometers or smaller and may include a lipid layer. The lipid nanoparticle (LNP) refers to an endoplasmic reticulum with an adjacent lipid bilayer, such as a spherical endoplasmic reticulum. Lipid nanoparticles can be used in methods to deliver pharmaceutical therapies to targeted locations. Non-limiting examples of LNPs include liposomes, amphiphiles, solid lipid nanoparticles (SLNs), nanostructured lipid carriers (NLCs), and monolayer membrane structures (e.g., archaeosomes and micelles).
지질을 이용하여 지질 나노입자를 제조하는 방법은 당업계에 잘 알려져 있다. 다양한 제형을 갖는 지질 나노입자의 제조를 위해서는 리포펩타이드 이외에도 양하전 지질, 중성 지질 및 음하전 지질로부터 선택되는 보조 지질을 추가로 사용할 수 있다. 예를 들어, 양이온성 리포좀의 제조를 위해서는 양하전 지질 및 중성 지질을, 중성 리포좀의 제조를 위해서는 중성 지질을, 음이온성 리포좀의 제조를 위해서는 음하전 지질 및 중성 지질을 추가로 사용하여 제조할 수 있다. 지질 나노입자의 제조를 위해 사용할 수 있는 양하전 지질, 중성 지질 및 음하전 지질은 당업계에 공지되어 있다.Methods for producing lipid nanoparticles using lipids are well known in the art. For the production of lipid nanoparticles with various formulations, in addition to lipopeptides, auxiliary lipids selected from positively charged lipids, neutral lipids, and negatively charged lipids can be additionally used. For example, for the production of cationic liposomes, positively charged lipids and neutral lipids may be used, for the production of neutral liposomes, neutral lipids may be used, and for the production of anionic liposomes, negatively charged lipids and neutral lipids may be additionally used. Positively charged lipids, neutral lipids, and negatively charged lipids that can be used for the preparation of lipid nanoparticles are known in the art.
나노입자의 지질 구성성분은 하나 이상의 인지질, 예컨대 하나 이상의 (폴리)불포화된 지질을 포함할 수 있다. 인지질은 하나 이상의 지질층으로 조립될 수 있다. 일반적으로, 인지질은 인지질 모이어티 및 하나 이상의 지방산 모이어티를 포함할 수 있다. 조성물 및 방법에 유용한 인지질은 1,2-디스테아로일-sn-글리세로-3-포스포콜린(DSPC), 1,2-디올레오일-sn-글리세로-3-포스포에탄올아민(DOPE), 1,2-디리놀레오일-sn-글리세로-3-포스포콜린(DLPC), 1,2-디미리스토일-sn-글리세로-포스포콜린(DMPC), 1,2-디올레오일-sn-글리세로-3-포스포콜린(DOPC), 1,2-디팔미토일-sn-글리세로-3-포스포콜린(DPPC), 1,2-디운데카노일-sn-글리세로-포스포콜린(DUPC), 1-팔미토일-2-올레오일-sn-글리세로-3-포스포콜린(POPC), 1,2-디-O-옥타데세닐-sn-글리세로-3-포스포콜린(18:0 디에테르 PC), 1-올레오일-2-콜레스테릴헤미숙시노일-sn-글리세로-3-포스포콜린(OChemsPC), 1-헥사데실-sn-글리세로-3-포스포콜린(C16 Lyso PC), 1,2-디리놀레노일-sn-글리세로-3-포스포콜린,1,2-디아라키도노일-sn-글리세로-3-포스포콜린, 1,2-디도코사헥사에노일sn-글리세로-3-포스포콜린, 1,2-디피타노일-sn-글리세로-3-포스포에탄올아민(ME 16.0 PE), 1,2-디스테아로일sn-글리세로-3-포스포에탄올아민, 1,2-디리놀레오일-sn-글리세로-3-포스포에탄올아민, 1,2-디리놀레노일-sn-글리세로-3-포스포에탄올아민, 1,2-디아라키도노일-sn-글리세로-3-포스포에탄올아민, 1,2-디도코사헥사에노일-sn글리세로-3-포스포에탄올아민, 1,2-디올레오일-sn-글리세로-3-포스포-rac-(1-글리세롤) 소듐 염(DOPG) 및 스핑 고미엘린으로 이루어진 비-제한적인 군으로부터 선택될 수 있다.The lipid component of the nanoparticle may include one or more phospholipids, such as one or more (poly)unsaturated lipids. Phospholipids can be assembled into one or more lipid layers. Generally, phospholipids may include a phospholipid moiety and one or more fatty acid moieties. Phospholipids useful in the compositions and methods include 1,2-distearoyl-sn-glycero-3-phosphocholine (DSPC), 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine ( DOPE), 1,2-dilinoleoyl-sn-glycero-3-phosphocholine (DLPC), 1,2-dimyristoyl-sn-glycero-phosphocholine (DMPC), 1,2 -Dioleoyl-sn-glycero-3-phosphocholine (DOPC), 1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC), 1,2-diundecanoyl- sn-glycero-phosphocholine (DUPC), 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC), 1,2-di-O-octadecenyl-sn- Glycero-3-phosphocholine (18:0 diether PC), 1-oleoyl-2-cholesterylhemisuccinoyl-sn-glycero-3-phosphocholine (OChemsPC), 1-hexadecyl- sn-glycero-3-phosphocholine (C16 Lyso PC), 1,2-dilinolenoyl-sn-glycero-3-phosphocholine, 1,2-diarachidonoyl-sn-glycero- 3-phosphocholine, 1,2-didocosahexaenoylsn-glycero-3-phosphocholine, 1,2-dipitanoyl-sn-glycero-3-phosphoethanolamine (ME 16.0 PE) , 1,2-distearoyl sn-glycero-3-phosphoethanolamine, 1,2-dilinoleoyl-sn-glycero-3-phosphoethanolamine, 1,2-dilinolenoyl -sn-glycero-3-phosphoethanolamine, 1,2-diarachidonoyl-sn-glycero-3-phosphoethanolamine, 1,2-didocosahexaenoyl-snglycero-3- Can be selected from the non-limiting group consisting of phosphoethanolamine, 1,2-dioleoyl-sn-glycero-3-phospho-rac-(1-glycerol) sodium salt (DOPG) and sphingomyelin. there is.
또한 나노입자의 지질 구성성분은 하나 이상의 구조적 지질을 포함할 수 있다. 구조적 지질은 콜레스테롤, 페코스테롤, 시토스테롤, 에르고스테롤, 캄페스테롤, 스티그마스테롤, 브라씨카스테롤, 토마티딘, 토마틴, 우르솔산, 알 파-토코페롤 및 이들의 혼합물로부터 선택될 수 있으나 이들로 한정되는 것은 아니다.Additionally, the lipid component of the nanoparticle may include one or more structural lipids. Structural lipids may be selected from, but are limited to, cholesterol, fecosterol, sitosterol, ergosterol, campesterol, stigmasterol, brassicasterol, tomatidine, tomatine, ursolic acid, alpha-tocopherol, and mixtures thereof. It doesn't work.
또한 나노입자의 지질 구성성분은 하나 이상의 PEG 또는 PEG-변형된 지질을 포함할 수 있다. 이러한 지질은 대안적으로, PEG화된 지질로 지칭될 수 있다. PEG 지질은 폴리에틸렌 글리콜로 변형된 지질이다. PEG 지질은 PEG-변형된 포스파티딜에탄올아민, PEG-변형된 포스파티드산, PEG-변형된 세라미드, PEG-변형된 디알킬아민, PEG-변형된 디 아실글리세롤, PEG-변형된 디알킬글리세롤 및 이들의 혼합물로 이루어진 비-제한적인 군으로부터 선택될 수 있 다. 예를 들어, PEG 지질은 PEG-c-DOMG, PEG-DMG(미국 앨라배마주 엘라베스터 소재의 Avanti Polar Lipid로부터 입수 가능한 1,2-디미리스토일-OT-글리세롤 메톡시폴리에틸렌 글리콜), PEG-DLPE, PEG-DMPE, PEG-DPPC 또는 PEG-DSPE 지질일 수 있다.The lipid component of the nanoparticle may also include one or more PEG or PEG-modified lipids. These lipids may alternatively be referred to as PEGylated lipids. PEG lipids are lipids modified with polyethylene glycol. PEG lipids include PEG-modified phosphatidylethanolamine, PEG-modified phosphatidic acid, PEG-modified ceramide, PEG-modified dialkylamine, PEG-modified diacylglycerol, PEG-modified dialkylglycerol and these. It may be selected from the non-limiting group consisting of mixtures of. For example, PEG lipids include PEG-c-DOMG, PEG-DMG (1,2-dimyristoyl-OT-glycerol methoxypolyethylene glycol, available from Avanti Polar Lipid, Elabaster, AL), PEG- It may be DLPE, PEG-DMPE, PEG-DPPC or PEG-DSPE lipid.
3. 상기 융합 단백질, 핵산분자, 지질나노입자(LNP), 발현 벡터, 숙주 세포 중 어느 하나를 유효 성분으로 포함하는 면역 관련 질환의 예방 또는 치료용 약학 조성물.3. A pharmaceutical composition for the prevention or treatment of immune-related diseases containing any one of the above fusion proteins, nucleic acid molecules, lipid nanoparticles (LNPs), expression vectors, and host cells as an active ingredient.
본 발명의 “면역관련질환”은 자가면역질환, 이식편대숙주 질환, 장기이식거부반응, 천식, 아토피, 및 급성 또는 만성의 염증 질환으로 이루어진 군으로부터 선택되는 적어도 하나일 수 있으나, 이에 제한되는 것은 아니다. The “immune-related disease” of the present invention may be at least one selected from the group consisting of autoimmune disease, graft-versus-host disease, organ transplant rejection, asthma, atopy, and acute or chronic inflammatory disease, but is not limited thereto. no.
본 발명의 상기 자가면역질환은 건선, 극세포증 (acanthosis), 이상각화증 (parakeratosis), 신생혈관신생증 (neo-angiogenesis), 류마티스성 관절염, 전신성 경피증, 전신 홍반성 낭창, 아토피 피부염, 건선, 원형탈모증, 천식, 크론씨병, 베체시병, 쇼그렌 증후군, 길리아-바레 증후군, 만성 갑상선염, 다발성 경화증, 다발성 근염, 강직성 척추염, 섬유조직염 및 결절성 다발성 동맥염으로 구성된 군으로부터 선택되는 적어도 하나일 수 있으나, 이에 제한되는 것은 아니다.The autoimmune diseases of the present invention include psoriasis, acanthosis, parakeratosis, neo-angiogenesis, rheumatoid arthritis, systemic scleroderma, systemic lupus erythematosus, atopic dermatitis, psoriasis, and alopecia areata. , asthma, Crohn's disease, Behce's disease, Sjögren's syndrome, Guillain-Barre syndrome, chronic thyroiditis, multiple sclerosis, polymyositis, ankylosing spondylitis, fibromyositis, and polyarteritis nodosa, but limited thereto. It doesn't work.
본 발명은 상기 급성 또는 만성의 염증 질환은 대장염, 패혈증(sepsis), 패혈성 쇼크, 염증성 장질환(Inflammatory bowel disease, IBD), 건선, 복막염, 신장염, 급성 기관지염, 만성 기관지염, 골관절염, 장질환 척추염, 만성 폐쇄성 폐질환 (chronic obstructive pulmonary disease, COPD), 류마티스성 관절염(rheumatoid arthritis), 급성 폐손상 (acute lung injury) 및 기관지 폐 형성장애(broncho-pulmonary dysplasia)로 이루어진 군에서 선택된 어느 하나 이상일 수 있으나, 이에 제한되는 것은 아니다.The present invention relates to the acute or chronic inflammatory diseases such as colitis, sepsis, septic shock, inflammatory bowel disease (IBD), psoriasis, peritonitis, nephritis, acute bronchitis, chronic bronchitis, osteoarthritis, and intestinal spondylitis. , chronic obstructive pulmonary disease (COPD), rheumatoid arthritis, acute lung injury, and broncho-pulmonary dysplasia. However, it is not limited to this.
본 발명에서 “건선”은 피부의 은빛 인설로 덮인 두껍고 가려움증이 없는 판 (non-pruritic plaque)을 특징으로 하는 재발성 염증성 피부 질환이다. 크론병 (Chron's disease; CD) 및 궤양성 질환 (ulcerative disease; UC)을 포함한 염증성 장 질환(Inflammatory bowel disease; IBD)은 장내 미생물에 대한 장내 면역 반응의 조절 장애와 뒤이어 위장관 염증이 특징이며, 지속적인 설사, 직장 출혈, 또는 체중 감소 등의 전형적인 증상이 나타난다. 건선과 염증성 장 질환(Inflammatory bowel disease; IBD)가 어떻게 발생하는지에 대한 정확한 메커니즘은 아직 불분명하지만 과도하게 활성화된 Th1 및 Th17 세포는 피부나 장관에서 불규칙한 면역반응을 일으키는 주요 원인으로 추정된다. 또한, 최근 연구에서는 각각 Th1 및 Th17 세포에서 분비되는 IFN-γ 및 IL-17A가 건선 및 IBD의 병리학적 진행을 유도하고 가속화하는 중요한 매개체인 것으로 나타났다. 그러나 건선 및 IBD에 대해 IFN-γ 또는 IL-17A를 표적으로 하는 치료 전략은 여전히 극복해야 할 몇 가지 한계에 직면해 있다.In the present invention, “psoriasis” is a recurrent inflammatory skin disease characterized by thick, non-pruritic plaques covered with silvery scales of the skin. Inflammatory bowel disease (IBD), including Crohn's disease (CD) and ulcerative disease (UC), is characterized by dysregulation of the intestinal immune response to intestinal microorganisms, followed by gastrointestinal inflammation, with persistent inflammation. Typical symptoms include diarrhea, rectal bleeding, or weight loss. The exact mechanism of how psoriasis and inflammatory bowel disease (IBD) develop is still unclear, but overly activated Th1 and Th17 cells are believed to be the main cause of irregular immune responses in the skin and intestinal tract. Additionally, recent studies have shown that IFN-γ and IL-17A, secreted by Th1 and Th17 cells, respectively, are important mediators that induce and accelerate the pathological progression of psoriasis and IBD. However, therapeutic strategies targeting IFN-γ or IL-17A for psoriasis and IBD still face several limitations that need to be overcome.
본 발명에서는 STAT1의 전사 조절 도메인 (Transcription modulation domain, TMD)와 인간 기원 단백질 전달 도메인 (Protein transduction domain, PTD) Hph-1 사이의 융합 단백질인 STAT1 (ndSTAT1-TMD)의 전사 조절 도메인의 핵 전달 가능한 형태를 생성했다. 본 발명에서 제공하는 ndSTAT1-TMD는 세포 독성과 TcR 매개 신호 이벤트에 영향을 미치지 않고 효율적으로 세포의 핵으로 전달될 수 있다. 본 발명에서 제공하는 ndSTAT1-TMD는 Th1 및 Th17 세포에서 내인성 STAT1의 전사 활성을 특이적으로 억제했으며 건선 및 IBD 마우스 모델에 투여할 때 항 IL-17A 항체와 비슷한 치료 잠재력을 보였다. 본 발명에서 제공하는 ndSTAT1-TMD가 Th1/17 세포 매개 자가면역 질환에 대한 새롭고 효과적인 치료 시약이 될 수 있음을 시사한다.In the present invention, nuclear delivery of the transcription modulation domain of STAT1 (ndSTAT1-TMD), which is a fusion protein between the transcription modulation domain (TMD) of STAT1 and the human protein transduction domain (PTD) Hph-1, is possible. created a shape. The ndSTAT1-TMD provided by the present invention can be efficiently delivered to the nucleus of cells without cytotoxicity and without affecting TcR-mediated signaling events. The ndSTAT1-TMD provided by the present invention specifically inhibited the transcriptional activity of endogenous STAT1 in Th1 and Th17 cells and showed therapeutic potential similar to that of anti-IL-17A antibody when administered to psoriasis and IBD mouse models. This suggests that ndSTAT1-TMD provided by the present invention can be a new and effective treatment reagent for Th1/17 cell-mediated autoimmune diseases.
본 발명에서 '대사성 질환'이란, 비만과 밀접하게 연관성이 있거나, 비만에 기인하는 상태 또는 질환을 의미하며, 구체적으로 지방간, 제2형 당뇨, 고지혈증, 심혈관 질환 및 동맥경화증으로 이루어진 군에서 선택되는 하나 이상일 수 있다.In the present invention, 'metabolic disease' means a condition or disease closely related to or caused by obesity, and specifically selected from the group consisting of fatty liver, type 2 diabetes, hyperlipidemia, cardiovascular disease, and arteriosclerosis. There may be more than one.
본 발명에서 지방간(fatty liver)은 간의 지방대사 장애로 지방이 간세포에 과도한 양으로 축적되는 상태 또는 질환을 의미한다.In the present invention, fatty liver refers to a condition or disease in which fat accumulates in excessive amounts in liver cells due to a disorder of liver fat metabolism.
본 발명에서 고지혈증은 혈액 중에 지방 성분, 특히 콜레스테롤(cholesterol)과 중성 지방(triglyceride)의 농도가 정상 수치에 비해 높은 상태 또는 질환을 의미하며, 혈액 중의 지질 농도를 저하시키는 것이 요구되는 모든 상태를 포함하는 광범위한 의미로 사용된다.In the present invention, hyperlipidemia refers to a condition or disease in which the concentration of fat components in the blood, especially cholesterol and triglyceride, is higher than normal, and includes all conditions requiring lowering the lipid concentration in the blood. It is used in a broad sense.
본 명세서에서 동맥경화증(arteriosclerosis)은 동맥벽이 두꺼워지고 탄력성이 감소하여 체내의 장기 및 조직으로의 혈액 순환이 저하되는 상태 또는 질환을 의미하며, 지방, 콜레스테롤 등 기타 물질이 동맥내벽에 침착되어 플라크(plaque)를 형성하여 내강이 좁아짐으로써 혈액 순환이 저하되는 상태 또는 질환을 의미하는 "아테롬성 동맥경화증(atherosclerosis)"을 포함하는 의미를 가진다. 동맥경화증은 체내 어느 부위에서도 발생할 수 있으며, 심장내 혈관에서 발생하는 경우에는 협심증, 심근경색 등 관상동맥질환이 유발되며, 뇌에서 발생하는 뇌경색이 유발될 수 있으며, 신장에서 일어나는 경우에는 신부전증 등이 유발될 수 있다.In this specification, arteriosclerosis refers to a condition or disease in which blood circulation to organs and tissues in the body is reduced due to thickening of the artery wall and reduction in elasticity, and the deposition of fat, cholesterol, and other substances on the inner wall of the artery, forming plaque ( It has a meaning that includes “atherosclerosis,” which refers to a condition or disease in which blood circulation is reduced by forming plaques and narrowing the lumen. Atherosclerosis can occur anywhere in the body. If it occurs in blood vessels within the heart, it can cause coronary artery disease such as angina pectoris or myocardial infarction. If it occurs in the brain, it can cause cerebral infarction. If it occurs in the kidneys, it can cause renal failure. It can be triggered.
본 발명에서 상기 "신경 퇴행성 질환" 또는 “퇴행성 뇌 또는 혈관 질환”은 신경 세포들의 기능 감소 또는 소실에 의해 발생되는 질환을 의미할 수 있고, "신경 염증성 질환"으로는 신경계의 과도한 염증 반응에 의하여 발생되는 질환을 의미할 수 있다. 본 발명에서 상기 신경 퇴행성 질환 또는 신경 염증성 질환의 구체적인 예시로는 뇌졸중, 치매, 알츠하이머병(Alzheimer's disease), 파킨슨병(Parkinson's disease), 헌팅턴병(Huntington's disease), 니만-피크병(Niemann-Pick disease), 다발성 경화증, 프리온병(prion disease), 크로이펠츠-야콥병(Creutzfeldt-Jakob disease), 전두측두치매, 루이치매, 근위축성 측삭경화증(AlzAmyotrophic lateral sclerosis), 부신생물증후군(Paraneoplastic syndrome), 피질기저퇴행증, 다계통위축병, 진행성핵상마비, 신경계 자가면역질환, 척수 소뇌 실조(spinocerebellar ataxia), 염증성 및 신경병증성 통증, 뇌혈관 질환, 척수 손상(spinal cord injury) 및 타우병증(tauopathy)으로 이루어진 군에서 선택될 수 있으나, 이에 제한되는 것은 아니다.In the present invention, the “neurodegenerative disease” or “degenerative brain or vascular disease” may refer to a disease caused by decreased or lost function of nerve cells, and the “neuroinflammatory disease” may refer to a disease caused by excessive inflammatory response in the nervous system. It may mean a disease that occurs. Specific examples of the neurodegenerative disease or neuroinflammatory disease in the present invention include stroke, dementia, Alzheimer's disease, Parkinson's disease, Huntington's disease, and Niemann-Pick disease. , multiple sclerosis, prion disease, Creutzfeldt-Jakob disease, frontotemporal dementia, Lewy dementia, amyotrophic lateral sclerosis, paraneoplastic syndrome, corticobasal degeneration. , multiple system atrophy disease, progressive supranuclear palsy, nervous system autoimmune disease, spinocerebellar ataxia, inflammatory and neuropathic pain, cerebrovascular disease, spinal cord injury, and tauopathy. It may be selected from the group, but is not limited thereto.
본 발명에서 상기 "암"은 포유류에서 전형적으로 조절되지 않는 세포 성장으로 특징 지어진 생리적 상태를 나타내거나 가리킨다. 본 발명에서 예방, 개선 또는 치료의 대상이 되는 암은 고형 장기(solid organ)에서 비정상적으로 세포가 성장하여 발생한 덩어리로 이루어진 고형암(solid tumor)일 수 있고, 예를 들면, 고형 장기의 부위에 따라 위암, 간암, 교세포종, 난소암, 대장암, 두경부암, 방광암, 신장세포암, 유방암, 전이암, 전립선암, 췌장암, 흑색종 또는 폐암 등일 수 있으며, 예를 들면, 흑색종일 수 있으나, 이에 제한되는 것은 아니다.As used herein, “cancer” refers to or refers to a physiological condition typically characterized by uncontrolled cell growth in mammals. The cancer subject to prevention, improvement, or treatment in the present invention may be a solid tumor consisting of a lump generated by abnormal cell growth in a solid organ, for example, depending on the part of the solid organ. It may be stomach cancer, liver cancer, glioblastoma, ovarian cancer, colon cancer, head and neck cancer, bladder cancer, renal cell cancer, breast cancer, metastatic cancer, prostate cancer, pancreatic cancer, melanoma, or lung cancer. For example, it may be melanoma. It is not limited.
본 발명에서 상기 "목적하는 개체"란, 면역 관련 질환이 발병하였거나, 발병 가능성이 높은 개체를 의미한다. In the present invention, the “object of interest” refers to an individual who has developed, or is likely to develop, an immune-related disease.
한편, 본 발명에서, "예방"은 본 발명의 약학 조성물을 이용하여 질환의 증상을 차단하거나, 그 증상을 억제 또는 지연시키는 모든 행위라면 제한없이 포함할 수 있다. Meanwhile, in the present invention, “prevention” may include without limitation any act of blocking, suppressing or delaying the symptoms of a disease using the pharmaceutical composition of the present invention.
또한, 본 발명에서, "치료"는 본 발명의 약학 조성물을 이용하여 질환의 증상이 호전되거나 이롭게 되는 모든 행위라면 제한없이 포함할 수 있다.Additionally, in the present invention, “treatment” may include, without limitation, any action that improves the symptoms of a disease or provides benefits using the pharmaceutical composition of the present invention.
한편, 이에 제한되지 않으나, 상기 질환의 예방 또는 치료 방법은 하나 이상의 질환에 대한 치료적 활성을 가지는 화합물 또는 물질을 투여하는 것을 더 포함하는 병용 요법일 수 있다. Meanwhile, although not limited thereto, the method for preventing or treating the disease may be a combination therapy that further includes administering a compound or substance having therapeutic activity for one or more diseases.
본 발명에서 상기 "병용"은 동시, 개별 또는 순차 투여를 나타내는 것으로 이해되어야 한다. 상기 투여가 순차 또는 개별적인 경우, 2차 성분 투여의 간격은 상기 병용의 이로운 효과를 잃지 않도록 하는 것이어야 한다. In the present invention, the “combined use” should be understood to indicate simultaneous, separate or sequential administration. If the administration is sequential or separate, the interval between the administrations of the second components should be such that the beneficial effects of the combination are not lost.
본 발명에서 상기 약학 조성물은 캡슐, 정제, 과립, 주사제, 연고제, 분말 또는 음료 형태임을 특징으로 할 수 있으며, 상기 약학 조성물은 인간을 대상으로 하는 것을 특징으로 할 수 있다. In the present invention, the pharmaceutical composition may be in the form of a capsule, tablet, granule, injection, ointment, powder, or beverage, and the pharmaceutical composition may be intended for human subjects.
본 발명에서 상기 약학 조성물은 이들로 한정되는 것은 아니지만, 각각 통상의 방법에 따라 산제, 과립제, 캡슐, 정제, 수성 현탁액 등의 경구형 제형, 외용제, 좌제 및 멸균 주사 용액의 형태로 제형화하여 사용될 수 있다. 본 발명의 약학 조성물은 약학적으로 허용 가능한 담체를 포함할 수 있다. 약학적으로 허용되는 담체는 경구 투여 시에는 결합제, 활탁제, 붕해제, 부형제, 가용화제, 분산제, 안정화제, 현탁화제, 색소, 향료 등을 사용할 수 있으며, 주사제의 경우에는 완충제, 보존제, 무통화제, 가용화제, 등장제, 안정화제 등을 혼합하여 사용할 수 있으며, 국소투여용의 경우에는 기제, 부형제, 윤활제, 보존제 등을 사용할 수 있다. 본 발명의 약학 조성물의 제형은 상술한 바와 같은 약제학적으로 허용되는 담체와 혼합하여 다양하게 제조될 수 있다. 예를 들어, 경구 투여시에는 정제, 트로키, 캡슐, 엘릭서(elixir), 서스펜션, 시럽, 웨이퍼 등의 형태로 제조할 수 있으며, 주사제의 경우에는 단위 투약 앰플 또는 다수회 투약 형태로 제조할 수 있다. 기타, 용액, 현탁액, 정제, 캡슐, 서방형 제제 등으로 제형화할 수 있다.In the present invention, the pharmaceutical composition is not limited to these, but can be formulated and used in the form of oral dosage forms such as powders, granules, capsules, tablets, and aqueous suspensions, external preparations, suppositories, and sterile injection solutions according to conventional methods. You can. The pharmaceutical composition of the present invention may include a pharmaceutically acceptable carrier. Pharmaceutically acceptable carriers include binders, lubricants, disintegrants, excipients, solubilizers, dispersants, stabilizers, suspending agents, colorants, flavorings, etc. for oral administration, and buffers, preservatives, and analgesics for injections. Topics, solubilizers, isotonic agents, stabilizers, etc. can be mixed and used, and for topical administration, bases, excipients, lubricants, preservatives, etc. can be used. The dosage form of the pharmaceutical composition of the present invention can be prepared in various ways by mixing it with a pharmaceutically acceptable carrier as described above. For example, for oral administration, it can be manufactured in the form of tablets, troches, capsules, elixirs, suspensions, syrups, wafers, etc., and in the case of injections, it can be manufactured in the form of unit dose ampoules or multiple doses. there is. In addition, it can be formulated as a solution, suspension, tablet, capsule, sustained-release preparation, etc.
한편, 제제화에 적합한 담체, 부형제 및 희석제의 예로는, 락토즈, 덱스트로즈, 수크로즈, 솔비톨, 만니톨, 자일리톨, 에리스리톨, 말티톨, 전분, 아카시아 고무, 알지네이트, 젤라틴, 칼슘 포스페이트, 칼슘 실리케이트, 셀룰로즈, 메틸 셀룰로즈, 미정질 셀룰로즈, 폴리비닐피롤리돈, 물, 메틸하이드록시벤조에이트, 프로필하이드록시벤조에이트, 탈크, 마그네슘 스테아레이트 또는 광물유 등이 사용될 수 있다. 또한, 충진제, 항 응집제, 윤활제, 습윤제, 향료, 유화제, 방부제 등을 추가로 포함할 수 있다.Meanwhile, examples of carriers, excipients and diluents suitable for formulation include lactose, dextrose, sucrose, sorbitol, mannitol, xylitol, erythritol, maltitol, starch, gum acacia, alginate, gelatin, calcium phosphate, calcium silicate, cellulose. , methyl cellulose, microcrystalline cellulose, polyvinylpyrrolidone, water, methylhydroxybenzoate, propylhydroxybenzoate, talc, magnesium stearate, or mineral oil can be used. In addition, fillers, anti-coagulants, lubricants, wetting agents, fragrances, emulsifiers, preservatives, etc. may be additionally included.
본 발명에 상기 약학 조성물의 투여 경로는 이들로 한정되는 것은 아니지만 안구에 직접 투여하거나, 구강, 정맥내, 근육내, 동맥내, 골수내, 경막내, 심장내, 경피, 피하, 복강내, 비강내, 장관, 국소, 설하 또는 직장이 포함된다. 경구 또는 비경구 투하가 바람직하다. 안구에 직접 투여하는 것이 더 바람직하다.The route of administration of the pharmaceutical composition according to the present invention is not limited to these, but may be administered directly to the eye, orally, intravenously, intramuscularly, intraarterially, intramedullary, intrathecally, intracardiacally, transdermally, subcutaneously, intraperitoneally, or nasally. Includes internal, intestinal, topical, sublingual or rectal. Oral or parenteral administration is preferred. It is more preferable to administer directly to the eye.
본 발명에서 상기 "비경구"란, 안구에 직접 투여하거나, 피하, 피내, 정맥내, 근육내, 관절내, 활액낭내, 흉골내, 경막내, 병소내 및 두개골내 주사 또는 주입기술을 포함한다. 본 발명의 약학 조성물은 또한 직장 투여를 위한 좌제의 형태로 투여될 수 있다. 안구에 직접 투여하는 것이 바람직하다.In the present invention, the term “parenteral” includes direct administration to the eye, subcutaneous, intradermal, intravenous, intramuscular, intraarticular, intrasynovial, intrasternal, intrathecal, intralesional and intracranial injection or injection techniques. . The pharmaceutical composition of the present invention can also be administered in the form of a suppository for rectal administration. It is preferable to administer directly to the eye.
본 발명의 조성물의 "용법 및 용량"은 사용된 특정 화합물의 활성, 연령, 체중, 일반적인 건강, 성별, 정식, 투여시간, 투여경로, 배출율, 약물 배합 및 예방 또는 치료될 특정 질환의 중증을 포함한 여러 요인에 따라 다양하게 변할 수 있고, 상기 약학 조성물의 투여량은 환자의 상태, 체중, 질병의 정도, 약무형태, 투여경로 및 기간에 따라 다르지만 당업자에 의해 적절하게 선택될 수 있고, 1일 0.0001 내지 50mg/kg 또는 0.001 내지 50mg/kg으로 투여할 수 있다. 투여는 하루에 한번 투여할 수도 있고, 수회 나누어 투여할 수도 있다. 상기 투여량은 어떠한 면으로든 본 발명의 범위를 한정하는 것은 아니다. 본 발명에 따른 의약 조성물은 환제, 당의정, 캡슐, 액제, 겔, 시럽, 슬러리, 현탁제로 제형될 수 있다.The "usage and dosage" of the composition of the present invention includes the activity of the specific compound used, age, weight, general health, gender, diet, administration time, administration route, excretion rate, drug combination, and the severity of the specific disease to be prevented or treated. It may vary depending on various factors, and the dosage of the pharmaceutical composition may vary depending on the patient's condition, body weight, degree of disease, drug form, administration route and period, but may be appropriately selected by a person skilled in the art, and is 0.0001 per day. It can be administered at 0.001 to 50 mg/kg or 0.001 to 50 mg/kg. Administration may be administered once a day, or may be administered several times. The above dosage does not limit the scope of the present invention in any way. The pharmaceutical composition according to the present invention may be formulated as pills, dragees, capsules, solutions, gels, syrups, slurries, and suspensions.
적합한 총 1일 사용량은 올바른 의학적 판단범위 내에서 처치의에 의해 결정될 수 있으며, 1회 또는 수회로 나누어 투여할 수 있다. 그러나 본 발명의 목적상, 특정 환자에 대한 구체적인 치료적 유효량은 달성하고자 하는 반응의 종류와 정도, 경우에 따라 다른 제제가 사용되는지의 여부를 비롯한 구체적 상기 유효성분을 포함하는 조성물, 환자의 연령, 체중, 일반 건강 상태, 성별 및 식이, 투여 시간, 투여 경로 및 상기 유효성분을 포함하는 조성물의 분비율, 치료기간, 구체적 조성물과 함께 사용되거나 동시 사용되는 약물을 비롯한 다양한 인자와 의약 분야에 잘 알려진 유사 인자에 따라 다르게 적용하는 것이 바람직하다.The appropriate total daily amount can be determined by the treating physician within the scope of sound medical judgment, and can be administered once or in several divided doses. However, for the purposes of the present invention, the specific therapeutically effective amount for a specific patient is determined by the type and degree of response to be achieved, the composition containing the active ingredient, the patient's age, and whether other agents are used as the case may be. Various factors well known in the medical field, including body weight, general health condition, gender and diet, administration time, administration route and secretion rate of the composition containing the active ingredient, treatment period, and drugs used together or simultaneously with the specific composition. It is desirable to apply it differently depending on similar factors.
본 발명의 일 구현예에서는 STAT1 (Signal transducer and activator of transcription 1)의 전사 조절 도메인 및 단백질 운반 도메인 (Protein transduction domain, PTD)을 포함하는 융합 단백질을 제공한다.One embodiment of the present invention provides a fusion protein comprising a transcriptional regulatory domain and a protein transduction domain (PTD) of STAT1 (Signal transducer and activator of transcription 1).
본 발명의 다른 구현예에서는 상기 STAT1은 서열번호 13의 아미노산 서열로 표시되는, 융합 단백질을 제공한다.In another embodiment of the present invention, STAT1 provides a fusion protein represented by the amino acid sequence of SEQ ID NO: 13.
본 발명의 또 다른 구현예에서는 상기 STAT1의 전사 조절 도메인은 서열번호 14의 아미노산 서열로 표시되는, 융합 단백질을 제공한다.In another embodiment of the present invention, a fusion protein is provided in which the transcriptional regulatory domain of STAT1 is represented by the amino acid sequence of SEQ ID NO: 14.
본 발명의 또 다른 구현예에서는 상기 단백질 운반 도메인은 Hph-1, Mph-1, Sim-2, Tat, VP22, Antp(antennapedia), Pep-1(peptide-1), PTD-5(protein transduction domain-5), 11R, 7R 및 CTP(cytoplamic transduction peptide)로 구성된 군으로부터 선택되는 것인, 융합 단백질을 제공한다.In another embodiment of the present invention, the protein transport domain is Hph-1, Mph-1, Sim-2, Tat, VP22, Antp (antennapedia), Pep-1 (peptide-1), PTD-5 (protein transduction domain) -5), 11R, 7R, and CTP (cytoplamic transduction peptide) are selected from the group consisting of fusion proteins.
본 발명의 또 다른 구현예에서는 상기 단백질 운반 도메인은 서열번호 1 내지 12 중 어느 하나의 아미노산 서열로 표시되는, 융합 단백질을 제공한다.Another embodiment of the present invention provides a fusion protein, wherein the protein transport domain is represented by any one of the amino acid sequences of SEQ ID NOs: 1 to 12.
본 발명의 또 다른 구현예에서는 상기 융합 단백질은 STAT1을 경쟁적으로 억제하는 것인, 융합 단백질을 제공한다.Another embodiment of the present invention provides a fusion protein that competitively inhibits STAT1.
본 발명의 또 다른 구현예에서는 상기 융합 단백질은 Th1 또는 Th17의 분화 또는 기능을 억제하는 것인, 융합 단백질을 제공한다.Another embodiment of the present invention provides a fusion protein that inhibits differentiation or function of Th1 or Th17.
본 발명의 일 구현예에서는 상기 융합 단백질을 암호화하는 핵산 분자를 제공한다.One embodiment of the present invention provides a nucleic acid molecule encoding the fusion protein.
본 발명의 일 구현예에서는 상기 핵산 분자를 포함하는 지질 나노입자 (LNP)를 제공한다.One embodiment of the present invention provides lipid nanoparticles (LNPs) containing the nucleic acid molecules.
본 발명의 일 구현예에서는 상기 핵산 분자가 삽입된 발현 벡터를 제공한다.One embodiment of the present invention provides an expression vector into which the nucleic acid molecule is inserted.
본 발명의 일 구현예에서는 상기 발현 벡터가 형질 감염된 숙주 세포를 제공한다.One embodiment of the present invention provides a host cell transfected with the expression vector.
본 발명의 일 구현예에서는 상기 융합 단백질, 상기 핵산분자, 상기 지질 나노입자 (LNP), 상기 발현 벡터, 상기 숙주 세포 중 어느 하나를 유효 성분으로 포함하는 면역 관련 질환의 예방 또는 치료용 약학 조성물을 제공한다.In one embodiment of the present invention, a pharmaceutical composition for preventing or treating immune-related diseases comprising any one of the fusion protein, the nucleic acid molecule, the lipid nanoparticle (LNP), the expression vector, and the host cell as an active ingredient. to provide.
본 발명의 다른 구현예에서는 상기 면역 관련 질환은 Th1 세포 또는 Th17 세포의 활성화 또는 기능 이상에 따라 발생하는 것인, 면역 관련 질환의 예방 또는 치료용 약학 조성물을 제공한다.Another embodiment of the present invention provides a pharmaceutical composition for preventing or treating immune-related diseases, wherein the immune-related diseases occur due to activation or dysfunction of Th1 cells or Th17 cells.
본 발명의 또 다른 구현예에서는 상기 면역 관련 질환은 자가면역질환, 이식편대숙주 질환, 장기이식거부반응, 천식, 아토피, 및 급성 또는 만성의 염증 질환으로 이루어진 군으로부터 선택되는 적어도 하나인 것인, 면역 관련 질환의 예방 또는 치료용 약학 조성물을 제공한다.In another embodiment of the present invention, the immune-related disease is at least one selected from the group consisting of autoimmune disease, graft-versus-host disease, organ transplant rejection, asthma, atopy, and acute or chronic inflammatory disease, A pharmaceutical composition for preventing or treating immune-related diseases is provided.
본 발명의 또 다른 구현예에서는 상기 자가면역질환은 건선, 극세포증 (acanthosis), 이상각화증 (parakeratosis), 신생혈관신생증 (neo-angiogenesis), 류마티스성 관절염, 전신성 경피증, 전신 홍반성 낭창, 아토피 피부염, 원형탈모증, 천식, 크론씨병, 베체시병, 쇼그렌 증후군, 길리아-바레 증후군, 만성 갑상선염, 다발성 경화증, 다발성 근염, 강직성 척추염, 섬유조직염 및 결절성 다발성 동맥염으로 구성된 군으로부터 선택된 어느 하나 이상인 것을 특징으로 하는, 면역 관련 질환의 예방 또는 치료용 약학 조성물을 제공한다.In another embodiment of the present invention, the autoimmune disease includes psoriasis, acanthosis, parakeratosis, neo-angiogenesis, rheumatoid arthritis, systemic scleroderma, systemic lupus erythematosus, and atopic dermatitis. , alopecia areata, asthma, Crohn's disease, Behce's disease, Sjögren's syndrome, Guillain-Barre syndrome, chronic thyroiditis, multiple sclerosis, polymyositis, ankylosing spondylitis, fibromyositis, and polyarteritis nodosa. Provides a pharmaceutical composition for preventing or treating immune-related diseases.
본 발명의 또 다른 구현예에서는 상기 급성 또는 만성의 염증 질환은 대장염, 패혈증(sepsis), 패혈성 쇼크, 염증성 장질환(Inflammatory bowel disease, IBD), 건선, 복막염, 신장염, 급성 기관지염, 만성 기관지염, 골관절염, 장질환 척추염, 만성 폐쇄성 폐질환 (chronic obstructive pulmonary disease, COPD), 류마티스성 관절염(rheumatoid arthritis), 급성 폐손상 (acute lung injury) 및 기관지 폐 형성장애(broncho-pulmonary dysplasia)로 이루어진 군에서 선택된 어느 하나 이상인 것을 특징으로 하는, 면역 관련 질환의 예방 또는 치료용 약학 조성물을 제공한다.In another embodiment of the present invention, the acute or chronic inflammatory disease includes colitis, sepsis, septic shock, inflammatory bowel disease (IBD), psoriasis, peritonitis, nephritis, acute bronchitis, chronic bronchitis, In the group consisting of osteoarthritis, enteropathy spondylitis, chronic obstructive pulmonary disease (COPD), rheumatoid arthritis, acute lung injury, and broncho-pulmonary dysplasia. Provided is a pharmaceutical composition for preventing or treating immune-related diseases, which is characterized in that it contains one or more selected substances.
본 발명에서 제공하는 융합 단백질 (ndSTAT1-TMD)는 세포 독성과 TcR 매개 신호 이벤트에 영향을 미치지 않고 효율적으로 세포의 핵으로 전달될 수 있다. 또한 본 발명에서 제공하는 융합 단백질은 Th1 및 Th17 세포에서 내인성 STAT1의 전사 활성을 특이적으로 억제했으며 건선 및 IBD 마우스 모델에 투여할 때 항 IL-17A 항체와 비슷한 치료 잠재력을 보였다. 그러므로 본 발명에서 제공하는 융합 단백질이 병원성 Th1 및 Th17 세포의 기능을 함께 조절함으로써 Th1/17 세포 매개 자가면역질환에 대한 새로운 치료제가 될 수 있다.The fusion protein (ndSTAT1-TMD) provided in the present invention can be efficiently delivered to the nucleus of cells without cytotoxicity or affecting TcR-mediated signaling events. Additionally, the fusion protein provided in the present invention specifically inhibited the transcriptional activity of endogenous STAT1 in Th1 and Th17 cells and showed therapeutic potential similar to that of anti-IL-17A antibody when administered to psoriasis and IBD mouse models. Therefore, the fusion protein provided in the present invention can become a new treatment for Th1/17 cell-mediated autoimmune diseases by controlling the functions of pathogenic Th1 and Th17 cells.
도 1은 핵 전달 가능한 ndSTAT1-TMD의 생성 및 세포 독성 없이 핵 내 전달 동역학 검증을 나타낸 것이다.Figure 1 shows the production of ndSTAT1-TMD capable of nuclear delivery and verification of its intranuclear delivery kinetics without cytotoxicity.
구체적으로 도 1A는 돌연변이된 발린 426 및 트레오닌 427을 갖는 Hph-1-PTD, ndSTAT-TMD 및 ndSTAT1-TMD (V426D, T427D)가 없는 STAT1-TMD의 단백질 구조를 나타낸 것으로, 각각 PTD, 단백질 전달 도메인 (protein transduction domain); nd, 핵 변환 가능 (nucleus-transducible); TMD, 전사 조절 도메인; DBD, DNA 결합 도메인; ND, N 말단 도메인; LD, 링커 도메인; SH2, SCR2 상동성 도메인; TAD, 트랜잭션 활성화 도메인 (transactivation domain)을 나타낸다. Specifically, Figure 1A shows the protein structures of Hph-1-PTD with mutated valine 426 and threonine 427, ndSTAT-TMD, and STAT1-TMD without ndSTAT1-TMD (V426D, T427D), PTD and protein transduction domain, respectively. (protein transduction domain); nd, nucleus-transducible; TMD, transcriptional regulatory domain; DBD, DNA binding domain; ND, N-terminal domain; LD, linker domain; SH2, SCR2 homology domain; TAD, stands for transaction activation domain.
도 1B는 정제된 STAT1-TMD, ndSTAT1-TMD 및 ndSTAT1-TMD (V426D, T427D)가 웨스턴 블롯(왼쪽 패널) 또는 SDS-PAGE(오른쪽 패널)에 의해 확인되었음을 나타낸 것이다.Figure 1B shows purified STAT1-TMD, ndSTAT1-TMD, and ndSTAT1-TMD (V426D, T427D) confirmed by Western blot (left panel) or SDS-PAGE (right panel).
도 1C 및 도 1D는 마우스 비장세포로의 ndSTAT1-TMD (0.5-2μM) 또는 STAT1-TMD (2μM)의 용량 의존적 및 시간 의존적 핵내 전달을 나타낸다.Figures 1C and 1D show dose-dependent and time-dependent intranuclear delivery of ndSTAT1-TMD (0.5-2 μM) or STAT1-TMD (2 μM) into mouse splenocytes.
도 1E는 마우스 비장세포로의 형질도입 후 ndSTAT1-TMD의 세포내 안정성을 나타낸다. Figure 1E shows the intracellular stability of ndSTAT1-TMD after transduction into mouse splenocytes.
도 1F는 배양된 마우스 비장세포에서 ndSTAT1-TMD의 세포내 국소화. 공초점 현미경을 사용한 대표적인 면역형광 63X (왼쪽 패널) 및 189X (오른쪽 패널) 이미지를 나타낸 것으로 스케일 바는 10㎛ 이다.Figure 1F: Subcellular localization of ndSTAT1-TMD in cultured mouse splenocytes. Representative immunofluorescence 63X (left panel) and 189X (right panel) images using confocal microscopy are shown, and the scale bar is 10 μm.
도 1G는 CCK-8 분석에 의해 분석된 마우스 비장세포에서 ndSTAT1-TMD 또는 ndSTAT1-TMD (V426D, T427D)의 세포 세포독성 평가를 나타낸다.Figure 1G shows cell cytotoxicity evaluation of ndSTAT1-TMD or ndSTAT1-TMD (V426D, T427D) in mouse splenocytes analyzed by CCK-8 assay.
도 1H 및 도 1I는 ndSTAT1-TMD 또는 ndSTAT1-TMD (V426D, T427D)를 유세포 분석 및 ELISA로 분석한 것으로 그래프는 평균 ± SEM(n=3)으로 표시되었으며 통계 분석은 Student's t-test를 사용하여 조사되었다.Figure 1H and Figure 1I show ndSTAT1-TMD or ndSTAT1-TMD (V426D, T427D) analyzed by flow cytometry and ELISA. Graphs are presented as mean ± SEM (n = 3), and statistical analysis was performed using Student's t-test. investigated.
도 2는 CD4+ 나이브 T 세포의 다양한 T 세포 하위 집합으로의 분화 능력에 대한 ndSTAT1-TMD의 기능적 영향을 나타낸 것이다.Figure 2 shows the functional impact of ndSTAT1-TMD on the differentiation ability of CD4 + naive T cells into various T cell subsets.
구체적으로, 도 2A는 Th1-스큐잉 조건 하에서 다양한 농도 (0.5-2μM)의 ndSTAT1-TMD 또는 ndSTAT1-TMD (V426D, T427D)로 처리된 Th1 세포에서 세포내 T-bet 및 IFN-γ의 유세포 분석을 나타낸 것이다.Specifically, Figure 2A shows flow cytometric analysis of intracellular T-bet and IFN-γ in Th1 cells treated with various concentrations (0.5-2 μM) of ndSTAT1-TMD or ndSTAT1-TMD (V426D, T427D) under Th1-skewing conditions. It represents.
도 2B는 도 2A의 Th1 세포 상청액에서 분비된 IFN-γ의 수준을 ELISA로 측정한 것이다.Figure 2B shows the level of IFN-γ secreted in the Th1 cell supernatant of Figure 2A measured by ELISA.
도 2C는 도 2A의 Th1 세포에서 T-bet+ IRF1+ 세포의 수준을 유세포 분석으로 분석한 것이다.Figure 2C shows flow cytometry analysis of the levels of T-bet + IRF1 + cells in the Th1 cells of Figure 2A.
도 2D는 Th17-스큐잉 조건 하에서 (under Th17-skewing condition) 다양한 농도(0.5-2μM)의 ndSTAT1-TMD 또는 ndSTAT1-TMD (V426D, T427D)로 처리된 Th17 세포에서 ROR-γt 및 세포내 IL-17A의 유세포 분석을 나타낸 것이다.Figure 2D shows ROR-γt and intracellular IL- levels in Th17 cells treated with various concentrations (0.5-2 μM) of ndSTAT1-TMD or ndSTAT1-TMD (V426D, T427D) under Th17-skewing conditions. Flow cytometry analysis of 17A is shown.
도 2E 및 도 2F는 도 2D의 Th17 세포 상청액에서 분비된 IL-17A 또는 IFN-γ의 수준을 ELISA로 측정한 것이다. 여기에 표시된 모든 실험은 유사한 결과를 가진 독립적인 생물학적 샘플에 대해 3회 반복되었다. 데이터는 평균 ± SEM(n≥3)으로 표시되었으며 통계 분석은 Student's t-test를 사용하여 조사되었다.Figures 2E and 2F show the levels of IL-17A or IFN-γ secreted from the Th17 cell supernatant of Figure 2D measured by ELISA. All experiments shown here were repeated three times on independent biological samples with similar results. Data are expressed as mean ± SEM (n≥3) and statistical analysis was examined using Student's t-test.
도 3은 ndSTAT1-TMD가 전사 수준에서 STAT1 표적 유전자의 발현을 억제하는 것을 나타낸 것이다.Figure 3 shows that ndSTAT1-TMD suppresses the expression of STAT1 target genes at the transcription level.
구체적으로, 도 3A는 HEK293T 세포를 IL-17A 프로모터에 의해 발현이 유도되는 루시퍼라제 리포터 유전자 및 야생형 STAT1 유전자를 함유하는 플라스미드로 공동 형질감염시킨 것을 나타낸 것이다. 즉, ndSTAT1-TMD 또는 ndSTAT1-TMD (V426D, T427D)를 처리한 후 24시간 후에 세포의 루시퍼라제 활성을 루미노미터 (luminometer)로 측정한 것을 나타낸 것이다. 데이터는 평균 ± SEM(n=5)으로 표시되었으며, 루시퍼라제 활성이 양성 대조군 (pCMV-STAT1 단독)보다 낮을 때 통계적 유의성이 나타났다. 통계 분석은 Student's t-test를 사용하여 조사되었습니다.Specifically, Figure 3A shows that HEK293T cells were co-transfected with a plasmid containing a luciferase reporter gene whose expression is driven by the IL-17A promoter and a wild-type STAT1 gene. That is, the luciferase activity of cells was measured using a luminometer 24 hours after treatment with ndSTAT1-TMD or ndSTAT1-TMD (V426D, T427D). Data are expressed as mean ± SEM (n = 5), and statistical significance was achieved when luciferase activity was lower than the positive control (pCMV-STAT1 alone). Statistical analysis was examined using Student's t-test.
도 3B 및 도 3C는 ntSTAT1 처리된 Th1 세포에서 설정된 IFN-γ 반응 유전자의 특징 또는 ntSTAT1-TMD 처리된 Th17 세포에서 야생형 CD4+ T 세포와 비교하여 STAT3 녹아웃 CD4+ T 세포에서 상향 조절된 유전자의 특징에 대한 GSEA 기반 농축 도표 (GSEA-based enrichment plots)를 나타낸 것이다.Figures 3B and 3C are a representation of the genes upregulated in STAT3 knockout CD4 + T cells compared to wild-type CD4 + T cells in ntSTAT1-treated Th1 cells or in ntSTAT1-TMD treated Th17 cells. This shows GSEA-based enrichment plots for .
도 3D 및 도 3E는 ndSTAT1-TMD를 처리하거나 처리하지 않은 Th1 또는 Th17 세포에서 Th1 또는 Th17 세포 시그니처 유전자 및 나이브 T 세포 특이적 유전자의 히트맵을 나타낸 것이다. 각 열은 하나의 실험 웰로부터의 결과를 의미하고 Z-score는 log2(TPM+1)로부터 계산하였다.Figures 3D and 3E show heatmaps of Th1 or Th17 cell signature genes and naive T cell specific genes in Th1 or Th17 cells treated or not with ndSTAT1-TMD. Each row represents the results from one experimental well and the Z-score was calculated from log2(TPM+1).
도 4는 ndSTAT1-TMD 치료가 IMQ가 유발하는 건선의 질병 중증도를 약화시키는 것을 나타낸 것이다.Figure 4 shows that ndSTAT1-TMD treatment attenuates the disease severity of IMQ-induced psoriasis.
구체적으로, 도 4A는 PBS (IMQ 그룹), 항-IL-17A 항체 (60 ㎍/마우스), 및 STAT1-TMD (20 ㎍/마우스 또는 60 ㎍/마우스), 또는 ndSTAT1-TMD (V426D, T427D) (60 ㎍/마우스)를 이용한, 5% IMQ-유도 건선 마우스의 치료 방법을 나타낸 것이다.Specifically, Figure 4A shows PBS (IMQ group), anti-IL-17A antibody (60 μg/mouse), and STAT1-TMD (20 μg/mouse or 60 μg/mouse), or ndSTAT1-TMD (V426D, T427D). This shows a treatment method for 5% IMQ-induced psoriasis mice using (60 μg/mouse).
도 4B는 질병 진행에 대해 도 4A 각 그룹의 등 피부에 있는 PASI 점수 플롯을 나타낸 것이다.Figure 4B shows a plot of the PASI scores on the back skin of each group in Figure 4A against disease progression.
도 4C는 5일차 (패널 위)와 7일차 (패널 아래)에 도 4A에서 마우스 등 피부 손상의 대표적인 이미지를 나타낸 것이다.Figure 4C shows representative images of skin damage on the back of the mouse in Figure 4A on day 5 (top panel) and day 7 (bottom panel).
도 4D는 1일차 두께를 기준으로 계산한 5일차 도 4A부터 각 그룹 내 등 피부 두께의 변화를 나타낸 것이다. 그래프는 평균 ± SEM (n=8)으로 표현하였으며, 통계 분석은 Student's t-test를 사용하였다.Figure 4D shows the change in back skin thickness within each group from Figure 4A on the 5th day, calculated based on the thickness on the 1st day. The graph was expressed as mean ± SEM (n = 8), and statistical analysis was performed using Student's t-test.
도 4E는 각 치료 그룹의 생쥐의 등 피부 두께를 나타낸 것이다.Figure 4E shows the back skin thickness of mice in each treatment group.
도 4F는 각 그룹의 체중 변화를 나타낸 것이다.Figure 4F shows the change in body weight of each group.
도 4G는 각 그룹의 마우스 혈청에서 전염증성 사이토카인 (TNF-α, IL-1β 또는 IL-6)의 수준을 ELISA로 측정한 것이다. 건선 모델의 실험은 독립적으로 두 번 수행되었다. 그래프는 평균 ± SEM(n=5)으로 표시되었으며 통계 분석은 Student's t-test를 사용하여 조사되었다. Figure 4G shows the levels of proinflammatory cytokines (TNF-α, IL-1β, or IL-6) in the serum of mice from each group measured by ELISA. Experiments in the psoriasis model were performed twice independently. Graphs are presented as mean ± SEM (n = 5) and statistical analysis was examined using Student's t-test.
도 4B, E 및 F의 그래프는 평균 ± SEM(n=8)으로 표시되었으며 통계 분석도 ANOVA 분석과 Dunnett의 다중 비교 테스트를 사용하여 조사되었다. 그래프에서 * IMQ 및 ndSTAT1-TMD 60㎍, # IMQ 및 항-IL-17A 항체 60㎍, + IMQ 및 ndSTAT1-TMD 20㎍, x IMQ 및 ndSTAT1-TMD(V426D, T427D) 60㎍를 각각 나타낸다.Graphs in Figure 4B, E, and F are presented as mean ± SEM (n = 8) and statistical analysis was also examined using ANOVA analysis and Dunnett's multiple comparison test. In the graph, * IMQ and ndSTAT1-TMD 60 μg, # IMQ and anti-IL-17A antibody 60 μg, + IMQ and ndSTAT1-TMD 20 μg, x IMQ and ndSTAT1-TMD (V426D, T427D) 60 μg, respectively.
도 5는 ndSTAT1-TMD 처리는 건선 동물 모델에서 CD4+ T 세포의 파괴된 기능 상태를 복원하는 것을 나타낸 것이다.Figure 5 shows that ndSTAT1-TMD treatment restores the destroyed functional state of CD4 + T cells in a psoriasis animal model.
구체적으로 도 5A 및 도 5B는 각 처리군의 비장(A) 및 배수림프절 (dLN)(B)에서, CD4+ T-bet+ T 세포 ((A) 및 (B)의 좌측 패널) 또는 CD4+ ROR-γt+ T 세포 ((A) 및 (B)의 우측 패널(A))의 레벨을 유세포 분석으로 7일째에 검사한 것을 나타낸 것이다.Specifically, Figures 5A and 5B show CD4 + T-bet + T cells (left panel of (A) and (B)) or CD4 + in the spleen (A) and draining lymph node (dLN) (B) of each treatment group. The level of ROR-γt + T cells (right panel (A) of (A) and (B)) is shown as examined on day 7 by flow cytometry.
도 5C 및 도 5D는 각 처리군의 비장(A) 및 배수림프절 (dLN)(B)에서 CD4+ IFN-γ+((C) 및 (D)의 왼쪽 패널) 또는 CD4+ IL-17A+((C) 및 (D)의 오른쪽 패널)의 수준을 7일차에 분석한 것이다.Figures 5C and 5D show CD4 + IFN-γ + (left panels of (C) and (D)) or CD4 + IL-17A + ( (C) and (D) right panel) levels were analyzed on day 7.
도 5E는 각 치료 그룹의 비장(왼쪽 패널) 또는 배수 림프절(dLN)(오른쪽 패널)의 CD4+ Foxp3+ 조절 T 세포 수준을 나타낸 것이다. 그래프는 평균 ± SEM(n=5)으로 표시되었으며 통계 분석은 Student's t-test를 사용하여 조사되었다.Figure 5E shows CD4 + Foxp3 + regulatory T cell levels in the spleen (left panel) or draining lymph nodes (dLN) (right panel) for each treatment group. Graphs are presented as mean ± SEM (n = 5) and statistical analysis was examined using Student's t-test.
도 6은 건선 동물 모델에 대한 ndSTAT1-TMD 처리는 백혈구가 등 피부로 침투하는 것을 방지하는 것을 나타낸 것이다.Figure 6 shows that ndSTAT1-TMD treatment of a psoriasis animal model prevents leukocytes from infiltrating the back skin.
구체적으로, 도 6A는 7일째 각 그룹의 마우스 등 피부에 대한 대표적인 H&E 염색 (패널 위) 또는 Masson의 삼색 염색 (패널 아래) 결과를 나타낸 것이다. 스케일 바는 100X의 경우 200㎛, 200X의 경우 100㎛를 나타낸다.Specifically, Figure 6A shows representative H&E staining (top panel) or Masson's trichrome staining (bottom panel) results on the back skin of mice from each group at day 7. The scale bar represents 200 μm for 100X and 100 μm for 200X.
도 6B는 각 처리군의 생쥐 등 피부 표피 두께의 변화를 도 6A의 조직학적 이미지로 측정한 것이다. 그래프는 평균 ± SEM(n=8)으로 표시되었으며, 통계적 분석은 Student's t-test를 이용하여 조사하였다.Figure 6B shows changes in the thickness of the skin epidermis on the back of mice in each treatment group measured using the histological image of Figure 6A. The graph was expressed as mean ± SEM (n = 8), and statistical analysis was performed using Student's t-test.
도 6C는 7일차 각 그룹의 등쪽 표피 (회색) 또는 진피 (파란색) 부위에 침윤된 CD45+ 백혈구의 수준을 나타낸 것이다.Figure 6C shows the level of CD45 + leukocytes infiltrated in the dorsal epidermis (grey) or dermis (blue) of each group on day 7.
도 6D는 7일째 각 그룹의 등쪽 표피 (회색) 또는 진피 (파란색) 영역에서 CD45, CD11b 및 F4/80을 발현하는 침윤 대식세포의 수준을 나타낸 것이다.Figure 6D shows the levels of infiltrating macrophages expressing CD45, CD11b, and F4/80 in the dorsal epidermis (grey) or dermis (blue) region of each group at day 7.
도 6E는 7일차 각 처리군의 생쥐 혈청 내 산화질소 (nitric oxide; NO) 수준을 나타낸 것이다.Figure 6E shows the level of nitric oxide (NO) in the serum of mice in each treatment group on day 7.
도 6F 및 도 6G는 7일차 각 처리군의 등쪽 표피 (회색) 또는 진피 (파란색) 부위의 전체 CD45+ 백혈구에서 T-bet+(도 6F) 또는 ROR-γt+(도 6G) T 세포의 침윤된 정도를 나타낸 것이다.Figures 6F and 6G show infiltration of T-bet + (Figure 6F) or ROR-γt + (Figure 6G) T cells in total CD45 + leukocytes in the dorsal epidermis (gray) or dermis (blue) region of each treatment group on day 7. It shows the degree to which it has been done.
도 6H 및 도 6I는 7일차 각 처리군의 등쪽 표피 (회색) 또는 진피 (파란색) 부위의 전체 CD45+ 백혈구에서 IFN-γ+(도 6H) 또는 IL-17A+(도 6I) T 세포의 침윤된 정도를 나타낸 것이다.Figures 6H and 6I show infiltration of IFN-γ + (Figure 6H) or IL-17A + (Figure 6I) T cells in total CD45 + leukocytes in the dorsal epidermis (gray) or dermis (blue) region of each treatment group on day 7. It shows the degree to which it has been done.
도 6C 내지 도 6I의 그래프는 평균 ± SEM(n=6)으로 표시되고, 통계적 분석은 Student's t-test를 이용하여 조사하였으며, IMQ군에 비해 세포군의 비율이 낮을 때 통계적 유의성을 나타내었다.The graphs in Figures 6C to 6I are expressed as mean ± SEM (n = 6), and statistical analysis was performed using Student's t-test, with statistical significance shown when the ratio of the cell group was low compared to the IMQ group.
도 7은 ndSTAT1-TMD는 DSS 유발 대장염의 질병 진행을 완화함을 나타낸 것이다.Figure 7 shows that ndSTAT1-TMD alleviates the disease progression of DSS-induced colitis.
구체적으로 도 7A는 PBS (DSS 그룹), 항-IL-17A 항체 (100㎍/마우스), ndSTAT1-TMD (20㎍/마우스 또는 100㎍/마우스) 또는 ndSTAT1-TMD (V426D, T427D) (100㎍/마우스)를 사용한 2.5% DSS 유발 대장염 마우스의 치료 계획을 나타낸 것이다.Specifically, Figure 7A shows 100 μg of PBS (DSS group), anti-IL-17A antibody (100 μg/mouse), ndSTAT1-TMD (20 μg/mouse or 100 μg/mouse), or ndSTAT1-TMD (V426D, T427D) (100 μg). /mouse) shows the treatment plan for mice with 2.5% DSS-induced colitis.
도 7B는 각 치료군의 체중 변화를 나타낸 것이다.Figure 7B shows the change in body weight in each treatment group.
도 7C는 각 치료 그룹의 질병 활동 지수 (Disease activity index; DAI) 점수를 나타낸 것이다. DAI 점수는 각 군의 대변, 대변잠혈, 체중변화의 임상점수를 측정하여 계산하였다.Figure 7C shows the disease activity index (DAI) score for each treatment group. The DAI score was calculated by measuring the clinical scores of stool, stool occult blood, and weight change in each group.
도 7D의 왼쪽 패널은 12일차에 각 처리 그룹의 마우스 결장 길이에 대한 비교 분석한 것이고, 오른쪽 패널은 더 7A 결장의 대표 이미지를 나타낸 것이다.The left panel of Figure 7D shows a comparative analysis of the colon length of mice in each treatment group on day 12, and the right panel shows a representative image of the 7A colon.
도 7E는 도 7D의 대표적인 수직 (패널 위) 및 수평 (패널 아래) 결장 섹션을 H&E 또는 PAS로 염색했다. 스케일 바는 40X의 경우 500㎛, 200X의 경우 100㎛를 나타낸다.Figure 7E shows representative vertical (above panel) and horizontal (below panel) colon sections from Figure 7D stained with H&E or PAS. The scale bar represents 500 μm for 40X and 100 μm for 200X.
도 7F는 각 처리군의 결장 근육층의 두께를 도 7E 이미지로부터 측정한 것이다.Figure 7F shows the thickness of the colonic muscle layer of each treatment group measured from the image in Figure 7E.
도 7G는 조직병리학적 점수를 나타낸 것으로, 조직병리학적 점수는 염증 세포 침윤, 선와 손상 및 잔 세포 고갈에 의해 계산된다. 그래프는 평균 ± SEM(n=6)으로 표시된다.Figure 7G shows the histopathological score, which is calculated by inflammatory cell infiltration, crypt damage, and goblet cell depletion. Graphs are presented as mean ± SEM (n = 6).
도 7H는 각 치료군의 혈청 내 전염증성 사이토카인 (TNF-α, IL-1β 또는 IL-6)의 수준을 ELISA로 측정한 것이다. 그래프는 평균 ± SEM(n=5)으로 표시된다. IBD 모델의 실험은 독립적으로 두 번 수행되었다.Figure 7H shows the levels of pro-inflammatory cytokines (TNF-α, IL-1β or IL-6) in the serum of each treatment group measured by ELISA. Graphs are presented as mean ± SEM (n = 5). Experiments in the IBD model were performed twice independently.
도 7B 내지 도 7D 및 도 7F의 그래프는 평균 ± SEM(n=7)으로 표시된다. 통계학적 분석은 Student's t-test 또는 ANOVA 분석과 Dunnett의 다중비교시험을 이용하여 조사하였으며, 도 7G의 경우 DSS군에 비해 조직병리학적 점수가 낮을 때 통계적 유의성을 보였다.The graphs in Figures 7B-7D and 7F are presented as mean ± SEM (n=7). Statistical analysis was conducted using Student's t-test or ANOVA analysis and Dunnett's multiple comparison test, and in Figure 7G, statistical significance was shown when the histopathological score was lower than that of the DSS group.
이하, 실시예를 통하여 본 발명을 더욱 상세히 설명하고자 한다. 이들 실시예는 오로지 본 발명을 보다 구체적으로 설명하기 위한 것으로서, 본 발명의 요지에 따라 본 발명의 범위가 이들 실시예에 의해 제한되지 않는다는 것은 당업계에서 통상의 지식을 가진 자에게 있어서 자명할 것이다.Hereinafter, the present invention will be described in more detail through examples. These examples are only for illustrating the present invention in more detail, and it will be apparent to those skilled in the art that the scope of the present invention is not limited by these examples according to the gist of the present invention. .
[준비예][Preparation example]
1. 재조합 단백질 생성을 위한 DNA 클로닝1. DNA cloning for recombinant protein production
마우스 STAT1의 플라스미드 (BC004808)는 미국 Origene Technologies, Inc.에서 구입했다. 야생형 STAT1 DNA 결합 도메인 (DNA binding domain; DBD)의 317~488번 아미노산을 코딩하는 서열을 PCR로 증폭한 후 Hph-1-PTD, 6x His 태그 및 FLAG 태그가 포함된 pET28a(+) 발현 벡터에 도입하였다. pfu DNA 중합효소 (Agilent)를 통해 점 돌연변이 형태의 DBD(V426D, T427D)가 생성되었다. 클로닝 후 오픈 리딩 프레임 (open reading frame, ORF)의 충실도를 확인하기 위해 생성된 모든 클로닝된 DNA 구조를 서열 분석하였다.The plasmid of mouse STAT1 (BC004808) was purchased from Origene Technologies, Inc., USA. The sequence coding for amino acids 317 to 488 of the wild-type STAT1 DNA binding domain (DBD) was amplified by PCR and then inserted into the pET28a(+) expression vector containing Hph-1-PTD, 6x His tag, and FLAG tag. introduced. Point mutation forms of DBD (V426D, T427D) were generated using pfu DNA polymerase (Agilent). After cloning, all generated cloned DNA structures were sequenced to confirm the fidelity of the open reading frame (ORF).
2. STAT1-TMD, ndSTAT1-TMD (V426D, T427D) 및 ndSTAT1-TMD의 발현 및 정제2. Expression and purification of STAT1-TMD, ndSTAT1-TMD (V426D, T427D) and ndSTAT1-TMD
모든 재조합 단백질은 Escherichia coli의 BL21 Codon Plus (DE3) RIPL 균주 (Invitrogen, Waltham, MA, USA)에서 발현되었으며 친화성 크로마토그래피 (affinity chromatography)를 통해 정제하였다. STAT1-TMD, ndSTAT1-TMD 또는 ndSTAT1-TMD (V426D, T427D)를 발현하는 세포를 천연 용해 완충액 (10mM 이미다졸, 50mM NaH2PO4, 300mM NaCl, pH 8.0)에서 수집하고 초음파를 처리하였다. 봉입체 (inclusion bodies)를 세포 용해물의 원심분리를 통해 펠렛화하고 3M 우레아 (10mM 이미다졸, 50mM NaH2PO4, 300mM NaCl, 3M Urea, pH 8.0)를 함유하는 천연 용해 완충액에서 추가로 초음파 처리했다. 세포 용해물을 펠렛화하고, 상층액을 Ni-NTA 수지 (Qiagen)와 혼합했다. 재조합 단백질을 30mM 이미다졸, 50mM NaH2PO4, 300mM NaCl, pH 8.0을 함유하는 완충액으로 세척하고, 500mM 이미다졸, 50mM NaH2PO4, 300mM NaCl, pH 8.0을 함유하는 완충액으로 용출시켰다. 용출된 재조합 단백질을 추가로 정제하여 이온 교환 크로마토그래피를 통해 내독소를 제거하고 PD-10 Sephadex G-25 컬럼 (GE Healthcare)을 사용하여 탈염했다. 정제된 재조합 단백질은 약 6 EU/ml의 안전한 수준의 내독소를 함유하고 있었다. 이후의 동물 실험에서는 아나필락시성 쇼크와 같은 내독소에 대한 면역 반응의 징후를 보이는 동물이 없었다.All recombinant proteins were expressed in the BL21 Codon Plus (DE3) RIPL strain of Escherichia coli (Invitrogen, Waltham, MA, USA) and purified through affinity chromatography. Cells expressing STAT1-TMD, ndSTAT1-TMD, or ndSTAT1-TMD (V426D, T427D) were collected in native lysis buffer (10mM imidazole, 50mM NaH 2 PO 4 , 300mM NaCl, pH 8.0) and sonicated. Inclusion bodies were pelleted by centrifugation of the cell lysate and further sonicated in native lysis buffer containing 3M urea (10mM imidazole, 50mM NaH 2 PO 4 , 300mM NaCl, 3M Urea, pH 8.0). did. Cell lysates were pelleted, and the supernatant was mixed with Ni-NTA resin (Qiagen). The recombinant protein was washed with a buffer containing 30mM imidazole, 50mM NaH 2 PO 4 , 300mM NaCl, pH 8.0, and eluted with a buffer containing 500mM imidazole, 50mM NaH 2 PO 4 , 300mM NaCl, pH 8.0. The eluted recombinant protein was further purified to remove endotoxin through ion exchange chromatography and desalted using a PD-10 Sephadex G-25 column (GE Healthcare). The purified recombinant protein contained a safe level of endotoxin of approximately 6 EU/ml. In subsequent animal experiments, none of the animals showed signs of an immune response to endotoxin, such as anaphylactic shock.
3. 웨스턴 블랏3. Western blot
상기 2.에서 정제한 재조합 단백질을 나트륨 도데실 설페이트-폴리아크릴아미드 겔 전기영동 (SDS-PAGE)을 통해 분리하고 폴리비닐리덴 플루오라이드 (PVDF, Bio-Rad, Hercules, CA, USA) 막으로 옮겼다. 막을 Tris 완충 식염수 중 0.1% Tween-20 중 5% 소 혈청 알부민으로 차단했습니다. 재조합 단백질을 항-6x His-tag 항체 (27E8 Clone, Cell Signaling, Danvers, MA, USA) 및 항-마우스 IgG-HRP 항체 (Abcam, Cambridge, MA, USA)와 함께 배양했다. 그리고 ECL 시약(Bio-Rad)을 사용하여 ChemiDoc (Bio-Rad)의 단백질을 시각화했다.The recombinant protein purified in step 2 above was separated through sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) and transferred to a polyvinylidene fluoride (PVDF, Bio-Rad, Hercules, CA, USA) membrane. . Membranes were blocked with 5% bovine serum albumin in 0.1% Tween-20 in Tris-buffered saline. Recombinant proteins were incubated with anti-6x His-tag antibody (27E8 Clone, Cell Signaling, Danvers, MA, USA) and anti-mouse IgG-HRP antibody (Abcam, Cambridge, MA, USA). Then, ECL reagent (Bio-Rad) was used to visualize the protein in ChemiDoc (Bio-Rad).
4. 세포배양4. Cell culture
HEK293T 세포를 10% 열 불활성화 소 태아 혈청 (FBS, Hyclone), 2mM L-글루타메이트 (Lonza), 100㎍/ml 페니실린-스트렙토마이신 (Lonza), 1 mM 피루브산 나트륨 (Lonza) 및 NEAA (Gibco, Waltham, MA, USA)이 보충된 DMEM 배지 (Lonza, Basel, Switzerland)에서 배양했다.HEK293T cells were incubated with 10% heat-inactivated fetal bovine serum (FBS, Hyclone), 2mM L-glutamate (Lonza), 100 μg/ml penicillin-streptomycin (Lonza), 1mM sodium pyruvate (Lonza), and NEAA (Gibco, Waltham). , MA, USA) was cultured in DMEM medium (Lonza, Basel, Switzerland) supplemented with .
모든 일차 세포는 7.5% 열 활성화 소 태아 혈청 (FBS, Hyclone), 2mM L-글루타메이트 (Lonza), 100㎍/ml 페니실린-스트렙토마이신 (Lonza) 및 50 μM β- Mercaptoethanol (Sigma Aldrich, St. Louis, MO, USA)이 보충된 RPMI 1640 (Lonza) 배지에서 유지되었다. 모든 세포는 5% CO2의 습한 분위기에서 37℃에서 배양되었다. All primary cells were incubated with 7.5% heat-activated fetal bovine serum (FBS, Hyclone), 2mM L-glutamate (Lonza), 100 μg/ml penicillin-streptomycin (Lonza), and 50 μM β-Mercaptoethanol (Sigma Aldrich, St. Louis; MO, USA) was maintained in RPMI 1640 (Lonza) medium supplemented with All cells were cultured at 37°C in a humidified atmosphere of 5% CO 2 .
5. 단백질 처리 (Protein Treatment)5. Protein Treatment
2x105 마우스 비장세포를 96-웰 세포 배양 플레이트 (Eppendorf)에서 재조합 단백질과 함께 용량 의존적 방식 (0.5-2 μM, 1시간) 또는 시간 의존적 방식 (2 μM, 0-72시간)으로 배양되어 유세포 분석에 사용되었다. 2x106 마우스 비장세포를 12-웰 세포 배양 플레이트 (SPL, Pocheon, South Korea) 바닥에 놓인 커버슬립에 파종하고 분석 전 1 μM의 ndSTAT1-TMD 또는 STAT1-TMD와 함께 1시간 동안 배양한 후 공초점 현미경 (LSM 980, Carl Zeiss, Jena, Germany)을 이용하여 관찰하였다. 2x105 마우스 나이브 CD4+ T 세포를 용량 의존적 방식 (0.5-2 μM)으로 재조합 단백질을 첨가하여 적절한 T 세포 서브세트-분극화 조건 하에 96웰 세포 배양 플레이트 (Eppendorf)에서 배양했다. 2x105 mouse splenocytes were cultured with recombinant proteins in 96-well cell culture plates (Eppendorf) in a dose-dependent manner (0.5-2 μM, 1 h) or time-dependent manner (2 μM, 0-72 h) for flow cytometric analysis. was used in 2x106 mouse splenocytes were seeded on coverslips placed on the bottom of a 12-well cell culture plate (SPL, Pocheon, South Korea) and incubated with 1 μM of ndSTAT1-TMD or STAT1-TMD for 1 h before analysis, followed by confocal analysis. Observation was made using a microscope (LSM 980, Carl Zeiss, Jena, Germany). 2x105 mouse naïve CD4 + T cells were cultured in 96-well cell culture plates (Eppendorf) under appropriate T cell subset-polarizing conditions with the addition of recombinant proteins in a dose-dependent manner (0.5-2 μM).
6. 면역세포화학 (Immunocytochemistry)6. Immunocytochemistry
2x106 개의 마우스 비장세포를 12웰 세포 배양 플레이트 (SPL) 바닥에 놓인 직경 18mm의 둥근 현미경 커버 글래스에 접종했다. 세포를 12-웰 세포 배양 플레이트 (SPL)에서 PBS, 1 μM STAT1-TMD, 또는 1 μM ndSTAT1-TMD가 포함된 배지에서 1시간 동안 배양하였다. 재조합 단백질 처리 후, 세포를 PBS로 세척하고, 10% 포르말린 용액 (Sigma-Aldrich)으로 고정한 후, 0.5% Triton X-100 용액 (Sigma-Aldrich)으로 투과화시켰다. 샘플은 1% BSA 용액으로 차단했다. 전달된 단백질은 항-FLAG Alexa 488 항체 (Invitrogen)로 포획하였다. PBS로 세척한 후 핵을 4',6-diamino-2-phenylindole (DAPI)로 대조염색하고 공초점 현미경(LSM 980, Carl Zeiss)으로 세포를 시각화하였다.2x10 6 mouse splenocytes were seeded on a round microscope cover glass with a diameter of 18 mm placed on the bottom of a 12-well cell culture plate (SPL). Cells were cultured in 12-well cell culture plates (SPL) in medium containing PBS, 1 μM STAT1-TMD, or 1 μM ndSTAT1-TMD for 1 hour. After recombinant protein treatment, cells were washed with PBS, fixed with 10% formalin solution (Sigma-Aldrich), and permeabilized with 0.5% Triton X-100 solution (Sigma-Aldrich). Samples were blocked with 1% BSA solution. The delivered protein was captured with anti-FLAG Alexa 488 antibody (Invitrogen). After washing with PBS, nuclei were counterstained with 4',6-diamino-2-phenylindole (DAPI), and cells were visualized using a confocal microscope (LSM 980, Carl Zeiss).
7. 세포 독성 분석 (Cytotoxicity assay)7. Cytotoxicity assay
ndSTAT1-TMD 및 ndSTAT1-TMD (V426D, T427D) 처리 후 세포 생존율을 Cell Counting Kit-8 (CCK-8, Dojindo Laboratories, Kumamoto, Japan)을 사용하여 테스트했습다. 2x105 마우스 비장세포를 96웰 배양 플레이트 (SPL)에 파종하고 다양한 용량의 ndSTAT1-TMD 또는 ndSTAT1-TMD (V426D, T427D)와 함께 1시간 동안 배양했다. 단백질 처리 후, 배양된 세포에 CCK-8 시약을 첨가하고 추가로 4시간 동안 배양하였다. 세포 생존율은 마이크로플레이트 리더 (Bio-Rad)를 사용하여 450nm 파장의 흡광도에서 측정하였다.Cell viability after treatment with ndSTAT1-TMD and ndSTAT1-TMD (V426D, T427D) was tested using Cell Counting Kit-8 (CCK-8, Dojindo Laboratories, Kumamoto, Japan). 2x10 5 mouse splenocytes were seeded in a 96-well culture plate (SPL) and incubated with various doses of ndSTAT1-TMD or ndSTAT1-TMD (V426D, T427D) for 1 hour. After protein treatment, CCK-8 reagent was added to the cultured cells and cultured for an additional 4 hours. Cell viability was measured at absorbance at a wavelength of 450 nm using a microplate reader (Bio-Rad).
8. CD4+ T 세포 분화8. CD4+ T cell differentiation
CD4+CD62L+ T 세포 분리 키트 (Miltenyi Biotec, Bergisch Gladbach, Germany)를 사용하여 8주령 암컷 C57BL/6N (B6) 마우스의 비장에서 추출한 RBC 제거 비장 세포에서 CD4+CD62L+ 순수 T 세포를 정제했다. 분리된 CD4+ 나이브 T 세포를 1㎍/ml의 항-CD3ε (BD Bioscience, San Diego, CA, USA) 및 항-CD28 (BD Bioscience)로 코팅된 96웰 세포 배양 플레이트 (Eppendorf)에서 72시간 동안 활성화시켰다. CD4+ 나이브 T 세포를 Th1, Th2, Th17 또는 Treg 세포로 분화하기 위해 다양한 사이토카인 혼합물과 항체를 배양 플레이트에 첨가했다: Th1 (25ng/ml의 재조합 마우스 IL-12 및 2㎍/ml의 항-IL-4 항체), Th2 (200ng/ml의 재조합 마우스 IL-4 및 2㎍/ml의 항-IFN-γ 항체), Th17 (1ng/ml의 재조합 마우스 TGF-β1, 25ng/ml의 재조합 마우스 IL-6, 2㎍/ml의 항-IFN-γ 항체 및 2㎍/ml의 항-IL-4 항체), iTreg (5ng/ml의 재조합 마우스 TGF-β 및 20ng/ml의 재조합 마우스 IL-2). 사이토카인과 항체는 PeproTech (Rocky Hill, NJ, USA), BioLegend (San Diego, CA, USA) 및 R&D Systems (Minneapolis, MN, USA)에서 구입했다. 모든 세포는 5% CO2의 습한 조건에서 37℃로 배양되었다.CD4 + CD62L + naive T cells were purified from RBC-cleared spleen cells extracted from the spleens of 8-week-old female C57BL/6N (B6) mice using a CD4 + CD62L + T cell isolation kit (Miltenyi Biotec, Bergisch Gladbach, Germany). Isolated CD4 + naive T cells were grown in 96-well cell culture plates (Eppendorf) coated with 1 μg/ml anti-CD3ε (BD Bioscience, San Diego, CA, USA) and anti-CD28 (BD Bioscience) for 72 h. activated. To differentiate CD4 + naive T cells into Th1, Th2, Th17 or Treg cells, various cytokine mixtures and antibodies were added to the culture plates: Th1 (25 ng/ml of recombinant mouse IL-12 and 2 μg/ml of anti- IL-4 antibody), Th2 (200 ng/ml recombinant mouse IL-4 and 2 μg/ml anti-IFN-γ antibody), Th17 (1 ng/ml recombinant mouse TGF-β1, 25 ng/ml recombinant mouse IL) -6, 2 μg/ml anti-IFN-γ antibody and 2 μg/ml anti-IL-4 antibody), iTreg (5 ng/ml recombinant mouse TGF-β and 20 ng/ml recombinant mouse IL-2) . Cytokines and antibodies were purchased from PeproTech (Rocky Hill, NJ, USA), BioLegend (San Diego, CA, USA), and R&D Systems (Minneapolis, MN, USA). All cells were cultured at 37°C in humid conditions with 5% CO 2 .
9. ELISA9.ELISA
활성화되거나 분화된 CD4+ 효과 T 세포 (effector T cells)의 상층액을 수집하여 사이토카인 수준을 측정했다. IFN-γ, IL-4, IL-17A 및 IL-2 수준은 제조업체의 프로토콜 (Invitrogen)에 따라 ELISA로 측정하였다.Supernatants of activated or differentiated CD4 + effector T cells were collected and cytokine levels were measured. Levels of IFN-γ, IL-4, IL-17A, and IL-2 were measured by ELISA according to the manufacturer's protocol (Invitrogen).
마우스 혈청 샘플은 질병 유발 7일차에 IMQ로 유발된 건선 마우스 또는 질병 유발 12일차에 덱스트란 황산나트륨 (dextran sulfate sodium; DSS)으로 유발된 대장염 마우스로부터 얻은 혈액 샘플을 원심분리하여 얻었다. 혈액 내 TNF-α, IL-1β 및 IL-6 수준은 제조업체의 프로토콜 (Invitrogen)에 따라 ELISA로 측정되었다.Mouse serum samples were obtained by centrifugation of blood samples obtained from IMQ-induced psoriatic mice on day 7 of disease induction or from dextran sulfate sodium (DSS)-induced colitis mice on day 12 of disease induction. TNF-α, IL-1β, and IL-6 levels in blood were measured by ELISA according to the manufacturer's protocol (Invitrogen).
사이토카인의 농도는 마이크로플레이트 리더 (Bio-Rad)를 사용하여 450 nm 파장의 흡광도에서 측정되었다.The concentration of cytokines was measured at absorbance at a wavelength of 450 nm using a microplate reader (Bio-Rad).
10. 리포터 유전자 분석 (Reporter Gene Analysis)10. Reporter Gene Analysis
ndSTAT1-TMD 및 ndSTAT1-TMD (V426D, T427D)에 대해 표적 유전자 프로모터에 대한 결합 능력을 테스트하였다. 5x105개 HEK293T 세포를 6-웰 배양 플레이트 (SPL)에 접종하고 Opti-MEM (Gibco)에 희석된 Lipofectamine Reagents (Invitrogen)를 사용하여 마우스 야생형 STAT1을 함유하는 1㎍의 pCMV6 벡터와 1㎍의 IL-17A 프로모터-루시퍼라제 벡터로 공동 형질감염시켰다. 5% CO2, 37℃에서 4시간 인큐베이션한 후, 세포를 ndSTAT1-TMD 또는 ndSTAT1-TMD (V426D, T427D)로 용량 의존적 방식 (0.5-2μM)으로 처리했다. 그런 다음 세포를 밤새 배양하고 PBS로 세척한 후 Cell Culture Lysis 5X Reagent (Promega, Madison, WI, USA)를 사용하여 용해했다. 세포 용해물을 Luciferase Assay Substrate (Promega)와 혼합하고, 루시퍼라제 활성을 Luminometer (Promega)로 측정했다.ndSTAT1-TMD and ndSTAT1-TMD (V426D, T427D) were tested for their binding ability to target gene promoters. 5x10 HEK293T cells were seeded in 6-well culture plates (SPL) and incubated with 1 μg of pCMV6 vector containing mouse wild-type STAT1 and 1 μg of IL using Lipofectamine Reagents (Invitrogen) diluted in Opti-MEM (Gibco). -17A promoter-luciferase vector was co-transfected. After incubation for 4 hours at 37°C with 5% CO 2 , cells were treated with ndSTAT1-TMD or ndSTAT1-TMD (V426D, T427D) in a dose-dependent manner (0.5-2 μM). Cells were then cultured overnight, washed with PBS, and lysed using Cell Culture Lysis 5X Reagent (Promega, Madison, WI, USA). Cell lysates were mixed with Luciferase Assay Substrate (Promega), and luciferase activity was measured with a Luminometer (Promega).
11. mRNA 서열 분석 및 유전자 세트 농축 분석 (mRNA-Sequencing and Gene Set Enrichment Analysis)11. mRNA-Sequencing and Gene Set Enrichment Analysis
ndSTAT1-TMD를 첨가하거나 첨가하지 않고 Th1- 또는 Th17-분극 조건 하에서 분화된 마우스 CD4+ T 세포를 72시간 후에 수확하였다. Macrogen (Seoul, South Korea)에서 RNA 추출 및 시퀀싱을 수행했다. Trimmomatic 0.38 프로그램은 첫 번째 판독의 품질이 낮은 데이터, 어댑터 및 DNA 오염을 제거하는 데 사용되었다. 그런 다음 HISAT2 프로그램을 사용하여 잘린 데이터를 게놈 레퍼런스 (10mm)에 매핑했다. 레퍼런스 기반 정렬 읽기 정보 (the reference-based aligned reads information)를 사용하여 전사체 어셈블리를 수행했으며, StringTie를 통해 조립된 전사체로부터 TPM (백만 개당 전사체) 카운트도 획득했다.Mouse CD4 + T cells differentiated under Th1- or Th17-polarizing conditions with or without the addition of ndSTAT1-TMD were harvested after 72 h. RNA extraction and sequencing were performed by Macrogen (Seoul, South Korea). The Trimmomatic 0.38 program was used to remove low-quality data, adapters, and DNA contamination from the first reads. The truncated data were then mapped to the genomic reference (10 mm) using the HISAT2 program. Transcriptome assembly was performed using the reference-based aligned reads information, and TPM (transcript per million) counts were also obtained from the assembled transcriptome using StringTie.
GSEA v4.1.0을 사용하여 ndSTAT1-TMD 처리 샘플 그룹에 대해 유전자 세트 농축 분석 (Gene set enrichment analysis; GSEA)을 수행했습니다. MSigDB는 Hallmark 유전자 세트 및 ImmuneSigDB를 포함하여 유전자 세트 데이터베이스로 사용되었다. 순열 수 (the number of permutations)는 1000으로 설정되었고 순열 유형 (the permutation type)은 '유전자 세트'로 설정되었습니다. 칩 플랫폼은 'Mouse_Gene_Symbol_Remapping_Human_Orthologs_MSigDB.v7.4로 설정되었다. ndSTAT1-TMD 처리 및 비처리 Th1 세포의 4개 샘플로부터의 17358개 유전자 및 ndSTAT1-TMD 처리 및 비처리 Th17 세포의 4개 샘플로부터의 17201개 유전자에 대한 TPM 값을 분석에 사용했다. Gene set enrichment analysis (GSEA) was performed on a group of ndSTAT1-TMD treated samples using GSEA v4.1.0. MSigDB was used as the gene set database, including Hallmark gene set and ImmuneSigDB. The number of permutations was set to 1000 and the permutation type was set to 'gene set'. The chip platform was set to 'Mouse_Gene_Symbol_Remapping_Human_Orthologs_MSigDB.v7.4. TPM values for 17358 genes from four samples of ndSTAT1-TMD treated and untreated Th1 cells and 17201 genes from four samples of ndSTAT1-TMD treated and untreated Th17 cells were used for analysis.
12. 실험 동물12. Laboratory animals
8~9주령 암컷 및 수컷 C57BL/6N (B6) 마우스 (8~9주)와 9주령 암컷 C57BL/6J (B6) 마우스를 Orient Bio (대한민국)에서 구입하였다. 모든 쥐는 연세동물실험센터 (YLARC)의 반특이성 무병원체 (the semi-specific-pathogen-free; SPF) 시설에서 사육 및 관리되었다. 모든 동물 연구는 연세대 실험실 동물 연구 센터 (YLARC)의 동물 실험 관리 및 사용위원회 (IACUC)의 승인을 받았으며 윤리적 사용을 위해 YLARC-IACUC의 지침에 따라 수행되었다. (IACUC-A-202109-1327-01, IACUC-A-202201-1404-01).8- to 9-week-old female and male C57BL/6N (B6) mice (8 to 9 weeks) and 9-week-old female C57BL/6J (B6) mice were purchased from Orient Bio (Korea). All mice were bred and maintained in the semi-specific-pathogen-free (SPF) facility at the Yonsei Animal Research Center (YLARC). All animal studies were approved by the Institutional Animal Care and Use Committee (IACUC) of the Yonsei University Laboratory Animal Research Center (YLARC) and were performed in accordance with the guidelines of YLARC-IACUC for ethical use. (IACUC-A-202109-1327-01, IACUC-A-202201-1404-01).
13. 건선 유도 및 건선 부위 및 중증도 지수 (PASI) 점수 측정 (Psoriasis Induction and scoring of psoriasis area and severity index (PASI))13. Psoriasis Induction and scoring of psoriasis area and severity index (PASI)
일주일의 순응 후, 8주령 암컷 C57BL/6J 마우스를 면도하고 1일차에 2*4cm 면적의 등 부위에 제모 크림을 바르고 털을 완전히 제거했다. 정상 그룹은 바셀린을 각각 노출된 등 피부 (40㎍) 및 양쪽 귀 피부 (20㎍/귀)에 7일 동안 연속해서 도포했다. 질병 유발군은 1일차부터 5% IMQ 크림(알다라크림, 동아에스앤티, 한국 서울)을 노출된 등 피부 (40㎍)와 양쪽 귀 피부(20㎍/귀)에 도포하였다. ndSTAT1-TMD (20 또는 60㎍/마우스), ndSTAT1-TMD (V426D, T427D)(60㎍/마우스) 또는 항-IL-17A 항체 (BioXCell, Lebanon, NH, USA)(60㎍) /mouse)를 1일부터 5일까지 격일로 복강내 주사하였고, 1일부터 7일까지 매일 체중, 건선 면적 및 중증도 지수(the psoriasis area and severity index; PASI) 점수를 측정하였다. 노출된 쥐의 등 피부는 5일과 7일에 사진을 찍었고, 7일에 쥐의 양쪽 귀를 촬영했다. 등 또는 오른쪽 귀 피부의 두께를 버니어 캘리퍼스 (Vernier caliper)로 연속 7일 동안 측정했다. 모든 그룹의 건선 부위 및 중증도 지수(PASI) 점수를 매일 평가하고 다음 지수에 따라 맹검 방식으로 측정했다.After one week of acclimatization, 8-week-old female C57BL/6J mice were shaved and hair removal cream was applied to a 2*4cm area on the back on day 1 to completely remove hair. The normal group applied petroleum jelly to each exposed back skin (40 μg) and both ear skin (20 μg/ear) continuously for 7 days. In the disease-inducing group, 5% IMQ cream (Aldara Cream, Donga S&T, Seoul, Korea) was applied to the exposed back skin (40㎍) and both ear skin (20㎍/ear) from day 1. ndSTAT1-TMD (20 or 60 μg/mouse), ndSTAT1-TMD (V426D, T427D) (60 μg/mouse), or anti-IL-17A antibody (BioXCell, Lebanon, NH, USA) (60 μg/mouse). Intraperitoneal injections were administered every other day from days 1 to 5, and body weight and psoriasis area and severity index (PASI) scores were measured daily from days 1 to 7. The exposed back skin of the mice was photographed on days 5 and 7, and both ears of the mice were photographed on day 7. The thickness of the back or right ear skin was measured with a Vernier caliper for 7 consecutive days. The Psoriasis Area and Severity Index (PASI) scores of all groups were assessed daily and measured in a blinded manner according to the following indices:
홍반 (발적): 0, 살색 분홍색; 1, 표면 전체에 약간의 붉어짐; 2, 표면 전체에 진한 빨간색 패치가 있는 중간 빨간색; 3, 표면 전체에 진한 빨간색의 진한 빨간색 패치있는 경우.Erythema (redness): 0, flesh-colored pink; 1, slight redness over the entire surface; 2, medium red with dark red patches throughout the surface; 3, If there are dark red patches all over the surface.
경화(두께): 0, 1-5%; 1, 5-50%; 2, 50-100%; 3, >100%. 박리(스케일링): 0, 피부 벗겨짐 없음; 1, 벗겨짐 없는 사소한 건조 반점; 2, 벗겨짐과 함께 피부의 대부분에 걸쳐 건조한 반점; 3, 피부 대부분에 건조한 반점이 있으며, 넓은 표면적에 걸쳐 중간 정도의 벗겨짐이 있고 심한 벗겨짐이 있는 경우.Curing (thickness): 0, 1-5%; 1, 5-50%; 2, 50-100%; 3, >100%. Detachment (scaling): 0, no peeling of skin; 1, minor dry spots without peeling; 2, dry patches over most of the skin with peeling; 3. Dry patches over most of the skin, with moderate to severe peeling over a large surface area.
실험 프로토콜은 연세대학교 동물보호사용위원회 (IACUC)에 의해 승인되었으며, 건선 마우스 모델에 대한 실험은 동물의 윤리적 사용에 대한 IACUC 지침에 따라 수행되었다 (IACUC-A-202201-1404-01). The experimental protocol was approved by the Yonsei University Institutional Animal Care and Use Committee (IACUC), and experiments on the psoriasis mouse model were performed in accordance with the IACUC Guidelines for the Ethical Use of Animals (IACUC-A-202201-1404-01).
13. 대장염 유발 및 질병 활동 지수(DAI) 점수 매기기 (Colitis Induction and Scoring of disease activity index)13. Colitis Induction and Scoring of disease activity index (DAI)
일주일 간의 순응 후, 8주령 수컷 C57BL/6N 마우스에게 1일부터 6일까지 멸균수 또는 2.5% DSS가 포함된 물을 제공했다. 덱스트란 황산나트륨 (dextran sulfate sodium; DSS)이 포함된 물은 격일로 교체했다. 6일 후, 모든 쥐에게 멸균수를 제공했다. 마우스에 ndSTAT1-TMD (20 또는 100㎍/마우스), ndSTAT1-TMD (V426D, T427D) (100㎍/마우스) 또는 항-IL-17A 항체 (BioXCell, Lebanon, NH, USA)(100㎍/마우스)를 복강 내 주사했다. 6일부터 11일까지 1일 1회, 체중 및 질병 활성도 지수(DAI) 점수를 1일부터 11일까지 매일 측정하였다. 모든 마우스를 분석을 위해 12일에 희생시켰다. 길이를 측정하기 위해 결장(맹장에서 직장까지)을 분리했다. DAI 점수의 맹검 측정을 제공하기 위해 다음 지수가 사용되었다:After one week of acclimatization, 8-week-old male C57BL/6N mice were provided with sterile water or water containing 2.5% DSS from days 1 to 6. Water containing dextran sulfate sodium (DSS) was changed every other day. After 6 days, all mice were provided with sterile water. Mice were administered ndSTAT1-TMD (20 or 100 μg/mouse), ndSTAT1-TMD (V426D, T427D) (100 μg/mouse), or anti-IL-17A antibody (BioXCell, Lebanon, NH, USA) (100 μg/mouse). was injected intraperitoneally. Body weight and disease activity index (DAI) scores were measured once a day from days 6 to 11, and daily from days 1 to 11. All mice were sacrificed on day 12 for analysis. The colon (caecum to rectum) was separated to measure length. The following indices were used to provide a blinded measure of DAI score:
체중 변화: 0, 0-1%; 1, 1-5%; 2, 5-10%; 3, 10-20%; 4, >20%. Weight change: 0, 0-1%; 1, 1-5%; 2, 5-10%; 3, 10-20%; 4, >20%.
대변: 0, 정상; 1, 일부는 부드럽지만 여전히 형성됨; 2, 매우 부드러움; 3, 설사. 대변 잠혈: 0, 정상; 1, 양성혈액종혈; 2, 대변의 혈액 흔적; 3, 직장 출혈.Stool: 0, normal; 1, some are soft but still formed; 2, very soft; 3, Diarrhea. Fecal occult blood: 0, normal; 1, benign hematoma; 2, traces of blood in stool; 3, rectal bleeding.
실험 프로토콜은 연세대학교 동물실험연구위원회(IACUC)에 의해 승인되었으며, 대장염 실험은 동물의 윤리적 사용에 대한 IACUC 지침(IACUC-A-202109-1327-01)을 따랐다. The experimental protocol was approved by the Yonsei University Institutional Animal Care and Use Committee (IACUC), and the colitis experiments followed the IACUC Guidelines for the Ethical Use of Animals (IACUC-A-202109-1327-01).
14. 조직학적 검사 (Histological Examination)14. Histological Examination
IMQ 유발 건선 마우스 모델에서는 각 마우스로부터 등 피부와 오른쪽 귀 1cm2를 채취하여 4% 인산염 완충 파라포름알데히드에 고정하고 파라핀에 포매한 후 헤마톡실린 및 에오신 (H&E)과 Masson's 삼색염색 (Masson's trichrome)으로 염색했다. 조직학적 샘플은 광학 현미경 (Olympus CX40)(Olympus Corporation, Shinjuku, Tokyo, Japan)을 통해 이미지화되었다. Image J 프로그램을 이용하여 등 피부 또는 오른쪽 귀 피부의 표피 두께를 측정하였다.In the IMQ-induced psoriasis mouse model, 1 cm 2 of back skin and right ear was collected from each mouse, fixed in 4% phosphate-buffered paraformaldehyde, embedded in paraffin, and stained with hematoxylin and eosin (H&E) and Masson's trichrome. dyed with Histological samples were imaged through a light microscope (Olympus CX40) (Olympus Corporation, Shinjuku, Tokyo, Japan). The epidermal thickness of the back skin or right ear skin was measured using the Image J program.
덱스트란 황산나트륨 (dextran sulfate sodium; DSS) 유발 대장염 마우스 모델에서 각 마우스로부터 결장의 원위 부분을 얻어 4% 인산염 완충 파라포름알데히드로 고정했습니다. 그런 다음 각 샘플을 파라핀에 포매하고 수평 또는 수직으로 절단한 후 H&E 및 PAS (과요오드산-Schiff)로 염색했습니다. 조직학적 샘플은 광학 현미경(Olympus BX51)(Olympus Corporation)을 통해 이미지화되었다. 다음 지표를 사용하여 염증 세포 침윤 정도, 크립트 손상 (Crypt damage) 및 잔세포 고갈 (Goblet cell depletion) 정도를 맹목적으로 측정했다. In the dextran sulfate sodium (DSS)-induced colitis mouse model, distal sections of the colon were obtained from each mouse and fixed in 4% phosphate-buffered paraformaldehyde. Each sample was then embedded in paraffin, sectioned horizontally or vertically, and stained with H&E and PAS (periodic acid-Schiff). Histological samples were imaged via light microscopy (Olympus BX51) (Olympus Corporation). The extent of inflammatory cell infiltration, crypt damage, and goblet cell depletion were blindly measured using the following indicators:
염증 세포 침윤: 0, 침윤 없음; 1, 가벼운 침투 <25%; 2, 적당한 침투 <50%; 3, 현저한 침투 >50%.Inflammatory cell infiltration: 0, no infiltration; 1, light penetration <25%; 2, moderate penetration <50%; 3, significant penetration >50%.
크립트 손상 (Crypt damage): 0, 없음; 1, 일부 크립트 손상; 2, 크립트 사이의 더 큰 공간; 3, 크립트가 없는 넓은 지역.Crypt damage: 0, none; 1, Some crypt damage; 2, larger space between crypts; 3, Large area without crypts.
잔세포 고갈 (Goblet cell depletion): 0, 없음; 1, 경미한 고갈 <25%; 2, 중간 정도의 고갈 <50%; 3, 현저한 고갈 >50%.Goblet cell depletion: 0, none; 1, mild depletion <25%; 2, moderate depletion <50%; 3, significant depletion >50%.
대장의 근육층 두께는 Image J 프로그램을 이용하여 측정하였다. 모든 조직 염색은 Korea CFC (Seoul, South Korea)에서 수행되었다.The thickness of the muscle layer of the large intestine was measured using the Image J program. All tissue staining was performed at Korea CFC (Seoul, South Korea).
15. 유세포 분석(Flow Cytometry)15. Flow Cytometry
마우스 비장세포에서 ndSTAT1-TMD의 형질도입 효율은 표시된 시간 동안 단백질 처리 후 항-FLAG-PE (BioLegend)를 사용한 세포내 염색을 통해 분석되었다.The transduction efficiency of ndSTAT1-TMD in mouse splenocytes was analyzed by intracellular staining using anti-FLAG-PE (BioLegend) after protein treatment for the indicated times.
분석을 위해 활성화된 CD4+ T 세포를 항-CD69-FITC (BD Bioscience) 또는 항-CD25-PE (BD Bioscience)로 염색했다. 재조합 단백질과 함께 72시간 인큐베이션한 후, 분화된 CD4+ 효과 T 세포 하위세트 (CD4+ effector T cell subsets)를 Cell-Stimulation Cocktail (500X) (eBioscience, San Diego, CA, USA)로 4시간 동안 재자극했다. 그런 다음 각 하위 집합을 Fixation/Permeabilization Buffer (eBioscience)로 고정 및 투과화하고 적절한 계통 정의 전사 인자 (appropriate lineage-defining transcription factors) 및 사이토카인에 대해 세포 내 염색했다. 모든 항체는 eBioscience에서 구입했다.For analysis, activated CD4 + T cells were stained with anti-CD69-FITC (BD Bioscience) or anti-CD25-PE (BD Bioscience). After 72 hours of incubation with recombinant proteins, differentiated CD4 + effector T cell subsets were resuspended with Cell-Stimulation Cocktail (500X) (eBioscience, San Diego, CA, USA) for 4 hours . provoked. Each subset was then fixed and permeabilized with Fixation/Permeabilization Buffer (eBioscience) and intracellularly stained for appropriate lineage-defining transcription factors and cytokines. All antibodies were purchased from eBioscience.
IMQ 유발 건선 쥐의 비장 또는 배수 림프절 (dLN)과 DSS 유발 대장염 쥐의 비장 또는 장간막 림프절 (mLN)에서 분리된 면역 세포를 Cell Stimulation Cocktail (500X) (eBioscience)로 4시간 동안 재자극했다. 재자극 후, 고정 및 투과화 전에 세포를 먼저 항-CD4-AF700 (eBioscience)으로 염색했다. 세포내 사이토카인 및 전사 인자는 항-IFN-γ-APC, 항-IL-17A-APC, 항-T-bet-PE, 항-ROR-γt-PE 또는 항-Foxp3-PE (eBioscience)로 추가로 염색되었다.Immune cells isolated from the spleen or draining lymph nodes (dLN) of IMQ-induced psoriasis mice and the spleen or mesenteric lymph nodes (mLN) of DSS-induced colitis mice were restimulated with Cell Stimulation Cocktail (500X) (eBioscience) for 4 hours. After restimulation, cells were first stained with anti-CD4-AF700 (eBioscience) before fixation and permeabilization. Intracellular cytokines and transcription factors were added with anti-IFN-γ-APC, anti-IL-17A-APC, anti-T-bet-PE, anti-ROR-γt-PE, or anti-Foxp3-PE (eBioscience). was dyed with
IMQ로 유발된 건선 쥐의 등과 귀 피부에서 피부 침윤 백혈구를 분리했다. 적절한 표면 마커를 염색하거나 세포내 전사 인자 또는 사이토카인을 염색하여 이들의 특성을 분석했다. 세포내 염색을 위해 세포를 Cell Stimulation Cocktail (500X) (eBioscience)을 사용하여 4시간 동안 재자극하고 고정하고 투과화시켰다. 세포를 항-CD45-AF700, 항-CD11b-APC, 항-IFN-γ-PE, 항-IL-17A-PE, 항-T-bet-PE, 항-ROR-γt-PE (eBioscience) 및 항-F4/80-PE (BD Bioscience)로 염색했습니다.Skin-infiltrating leukocytes were isolated from the back and ear skin of mice with IMQ-induced psoriasis. Their properties were analyzed by staining for appropriate surface markers or staining for intracellular transcription factors or cytokines. For intracellular staining, cells were restimulated for 4 h using Cell Stimulation Cocktail (500X) (eBioscience), fixed, and permeabilized. Cells were incubated with anti-CD45-AF700, anti-CD11b-APC, anti-IFN-γ-PE, anti-IL-17A-PE, anti-T-bet-PE, anti-ROR-γt-PE (eBioscience), and anti- Stained with -F4/80-PE (BD Bioscience).
세포 자극 칵테일 (500x)을 사용하여 재자극된 모든 세포를 5% CO2의 습한 조건에서 37℃로 배양했다. 유세포 분석은 SA3800 (Sony, Tokyo, Japan)을 이용하여 수행하였고, 데이터 분석은 Flowjo V10 프로그램을 이용하였다.All cells restimulated using the cell stimulation cocktail (500x) were cultured at 37°C in humidified conditions with 5% CO 2 . Flow cytometry was performed using SA3800 (Sony, Tokyo, Japan), and data analysis was performed using the Flowjo V10 program.
16. 산화질소 생산 분석 (Nitro Oxide production assay)16. Nitro Oxide production assay
질병 유발 7일차에 IMQ로 유도된 각 군의 건선 마우스로부터 혈액을 채취하고, 원심분리하여 혈청을 분리하였다. 산화질소 (nitro oxide; NO) 생성 수준은 Griess Reagent System을 사용하여 제조업체의 프로토콜 (Peprotech)에 따라 측정되었다. 혈액 상등액 샘플의 산화질소 수준은 마이크로플레이트 리더 (Bio-Rad)를 사용하여 540 nm에서의 흡광도를 분석하여 측정되었다.On the 7th day of disease induction, blood was collected from IMQ-induced psoriasis mice in each group, and serum was separated by centrifugation. Nitro oxide (NO) production levels were measured using the Griess Reagent System according to the manufacturer's protocol (Peprotech). Nitric oxide levels in blood supernatant samples were measured by analyzing absorbance at 540 nm using a microplate reader (Bio-Rad).
17. 피부 침윤 백혈구 분리 (Isolation of Skin-infiltrating Leukocytes)17. Isolation of Skin-infiltrating Leukocytes
IMQ 유발 건선 마우스의 등 피부 또는 왼쪽 귀 피부에서 피부 침윤 백혈구를 분리했다. IMQ 적용 7일 후, 마우스 등 피부와 왼쪽 귀에서 1cm2 면적을 잘라내고 HBSS (Gibco) 세척으로 남은 IMQ 크림을 제거한 후 곡선 겸자를 사용하여 표피와 진피를 분리했습니다. 분리된 표피와 진피를 Dispase II 용액 (5 mg/ml, Sigma Aldrich)을 사용하여 37℃에서 소화시켰다. 첫 번째 소화 후, 진피 해리 완충액 (콜라게나제 P 1mg/ml 및 DNaseI 100㎍/ml (Hyclone, Roche)을 함유하는 DMEM (Hyclone, Roche))에서 곡선 가위를 사용하여 진피를 작은 조각(<0.5mm)으로 신속하게 절단하고 1시간 동안 37℃서 배양했다. 완전히 소화된 진피 현탁액을 40-㎛ 세포 여과기 (Falcon)를 통해 여과하고 10% FBS (Hyclone)가 포함된 DMEM 배지로 헹구고 원심분리를 통해 분리된 진피 침윤 백혈구를 수집했습니다. 첫 번째 소화 후, 표리를 곡선 가위로 빠르게 작은 조각(< 2mm)으로 자르고 0.05% TE (Trypsin-EDTA, Hyclone) 완충액에 옮기고 37℃에서 5분간 배양했다. 배양 후, 완전히 소화된 표피에 트립신 중화 용액 (HBSS로 희석된 5% FBS)을 첨가하고, 표피 현탁액을 40-㎛ 세포 여과기 (Falcon)를 통해 여과하였다. 분리된 표피-침윤 백혈구를 원심분리에 의해 수집하였다.Skin-infiltrating leukocytes were isolated from the back skin or left ear skin of IMQ-induced psoriasis mice. Seven days after IMQ application, a 1 cm 2 area was cut from the mouse back skin and left ear, the remaining IMQ cream was removed by washing with HBSS (Gibco), and the epidermis and dermis were separated using curved forceps. The separated epidermis and dermis were digested at 37°C using Dispase II solution (5 mg/ml, Sigma Aldrich). After the first digestion, the dermis was dissected into small pieces (<0.5%) using curved scissors in dermal dissociation buffer (DMEM (Hyclone, Roche) containing 1 mg/ml collagenase P and 100 μg/ml DNaseI (Hyclone, Roche)). mm) and incubated at 37°C for 1 hour. The fully digested dermal suspension was filtered through a 40-μm cell strainer (Falcon), rinsed with DMEM medium containing 10% FBS (Hyclone), and separated dermal-infiltrating leukocytes were collected by centrifugation. After the first digestion, the front and back were quickly cut into small pieces (< 2 mm) with curved scissors, transferred to 0.05% TE (Trypsin-EDTA, Hyclone) buffer, and incubated at 37°C for 5 minutes. After incubation, trypsin neutralizing solution (5% FBS diluted in HBSS) was added to the completely digested epidermis, and the epidermal suspension was filtered through a 40-μm cell strainer (Falcon). Isolated epidermal-infiltrating leukocytes were collected by centrifugation.
18. 통계 분석18. Statistical analysis
결과는 평균 ± SEM(n ≥3)으로 표시하였다. 그룹 차이의 통계 분석은 쌍을 이루지 않은 Student's t-test 또는 ANOVA 분석을 사용한 후 Dunnett의 다중 비교 검정을 수행했다. 별표의 수는 하기와 같이 통계분석 결과를 나타낸다: ns; 유의하지 않음, *p<0.05, **p<0.01, ***p<0.001 및 ****p<0.0001. 그래프패드 프리즘 9는 모든 데이터의 분석에 사용되었다.Results are expressed as mean ± SEM (n ≥3). Statistical analysis of group differences used unpaired Student's t-test or ANOVA analysis followed by Dunnett's multiple comparison test. The number of asterisks indicates the results of statistical analysis as follows: ns; Not significant, *p<0.05, **p<0.01, ***p<0.001 and ****p<0.0001. GraphPad Prism 9 was used for analysis of all data.
[실시예][Example]
1. 일차 T 세포의 핵으로의 ndSTAT1-TMD의 세포내 전달 동역학 (Intracellular delivery kinetics of ndSTAT1-TMD into the nucleus of primary T cells)1. Intracellular delivery kinetics of ndSTAT1-TMD into the nucleus of primary T cells
CD4+ 나이브 T 세포는 인산화 (p-STAT1), 동종이량체화 및 STAT1의 전좌의 후속 과정을 포함하는 STAT1 신호 전달 경로를 채택하여 Th1 세포로 분화할 수 있다. in vitroin vivo에서 STAT1 기능을 분석하기 위해 다양한 유전적 및 치료적 접근법이 사용되었다. 그러나 in vitroin vivo에서 T 세포 네트워크의 기능적 맥락에서 STAT1의 조절 역할은 여전히 파편화되어 있다. 정상적인 생리학적 조건에서 in vitroin vivo에서 T 세포 하위 집합의 기능적 네트워크에서의 STAT1의 역할을 조사하기 위해 도 1A에 표시된 대로 Hph-1-PTD와 STAT1-TMD 사이의 융합 단백질인 ndSTAT1-TMD를 생성했다. 음성대조군으로 Hph1-PTD가 없는 STAT1-TMD를 제작하였고, DNA 결합에 중요한 두 가지 필수 아미노산 (V426, T427)이 D426 및 D427로 변경되었다. 이러한 모든 융합 단백질은 Escherichia coli 시스템에서 발현되었으며 Ni-NTA 아가로스를 사용하여 기본 조건에서 정제되었다. 이러한 정제된 단백질은 항-6xHis 및 SDS-PAGE를 사용하여 웨스턴 블롯으로 확인되었다 (도 1B).CD4 + naive T cells can differentiate into Th1 cells by adopting the STAT1 signaling pathway, which includes phosphorylation (p-STAT1), homodimerization, and subsequent processes of STAT1 translocation. Various genetic and therapeutic approaches have been used to analyze STAT1 function in vitro and in vivo . However, the regulatory role of STAT1 in the functional context of T cell networks in vitro and in vivo remains fragmented. To investigate the role of STAT1 in the functional network of T cell subsets in vitro and in vivo under normal physiological conditions, ndSTAT1-TMD, a fusion protein between Hph-1-PTD and STAT1-TMD, was used as shown in Figure 1A. created. As a negative control, STAT1-TMD without Hph1-PTD was produced, and two essential amino acids (V426, T427) important for DNA binding were changed to D426 and D427. All these fusion proteins were expressed in the Escherichia coli system and purified under basic conditions using Ni-NTA agarose. These purified proteins were confirmed by Western blot using anti-6xHis and SDS-PAGE (Figure 1B).
다음으로, 1차 T 세포에서 ndSTAT1-TMD의 핵내 형질도입 동역학을 조사하였다. Hph-1-PTD가 없는 STAT1-TMD와 달리, ndSTAT1-TMD는 2 μM 농도로 처리했을 때 용량 및 시간 의존 방식으로 세포 내로 효과적으로 전달되었다 (도 1C-1D). 전달된 ndSTAT1-TMD는 전달 후 72시간까지 세포 내부에서 안정적으로 유지되었다 (도 1E). 또한, 1차 T 세포 내에서 전달된 ndSTAT1-TMD의 세포내 위치를 공초점 현미경을 사용하여 시각화한 경우, ndSTAT1-TMD는 처리된 세포의 핵에 국한되었지만 STAT1-TMD로 처리된 세포에서는 발견되지 않았다 (도 1F).Next, we investigated the intranuclear transduction kinetics of ndSTAT1-TMD in primary T cells. Unlike STAT1-TMD without Hph-1-PTD, ndSTAT1-TMD was efficiently delivered into cells in a dose- and time-dependent manner when treated at a concentration of 2 μM (Figure 1C-1D). The delivered ndSTAT1-TMD remained stably inside the cells up to 72 hours after delivery (Figure 1E). Additionally, when the subcellular localization of delivered ndSTAT1-TMD within primary T cells was visualized using confocal microscopy, ndSTAT1-TMD was localized to the nucleus of treated cells but not found in cells treated with STAT1-TMD. did (Figure 1F).
이들 융합 단백질이 세포 독성을 갖는지 테스트하기 위해 일차 T 세포에 ndSTAT1-TMD 또는 ndSTAT1-TMD (V426D, T427D)를 다양한 농도로 1시간 동안 처리한 후 CCK-8 분석을 통해 세포독성 효과를 분석하였다. ndSTAT1-TMD나 ndSTAT1-TMD(V426D, T427D) 모두 유의미한 세포독성을 나타내지 않았다 (도 1G). 이러한 데이터는 ndSTAT1-TMD가 세포 생존력에 영향을 주지 않고 일차 T 세포의 핵으로 효과적이고 안정적으로 전달될 수 있음을 보여준다. To test whether these fusion proteins have cytotoxicity, primary T cells were treated with ndSTAT1-TMD or ndSTAT1-TMD (V426D, T427D) at various concentrations for 1 hour, and then the cytotoxic effect was analyzed through CCK-8 analysis. Neither ndSTAT1-TMD nor ndSTAT1-TMD(V426D, T427D) showed significant cytotoxicity (Figure 1G). These data show that ndSTAT1-TMD can be effectively and stably delivered into the nucleus of primary T cells without affecting cell viability.
2. ndSTAT1-TMD는 나이브 CD4+ T 세포가 Th1 및 Th17 T 세포 하위 집합으로 분화되는 것을 특이하게, 그리고 동시에 억제함.2. ndSTAT1-TMD specifically and simultaneously inhibits the differentiation of naïve CD4+ T cells into Th1 and Th17 T cell subsets.
T 세포 수용체 (T cell receptor; TcR) 자극은 나이브 CD4+ T 세포가 다양한 T 세포 하위 집합으로 성공적으로 분화하는 데 필요한 필수 요소이다. T 세포의 ndSTAT1-TMD 또는 ndSTAT1-TMD (V426D, T427D) 처리가 T 세포 수용체 매개 초기 활성화 신호 전달 이벤트에 영향을 미치는지 여부를 조사하기 위해 ndSTAT1-TMD 또는 ndSTAT1-TMD (V426D, T427D) 존재 하에 T 세포 수용체 자극 후 CD69, CD25 및 IL-2 분비의 유도된 표면 발현 수준을 측정했다. 도 1H 및 1I에 나타낸 바와 같이, CD25, CD69 및 인터루킨-2(IL-2)의 발현 수준은 ndSTAT1-TMD 또는 ndSTAT1-TMD (V426D, V427D) 처리에 의해 영향을 받지 않았다.T cell receptor (TcR) stimulation is an essential factor required for the successful differentiation of naïve CD4 + T cells into various T cell subsets. To investigate whether ndSTAT1-TMD or ndSTAT1-TMD(V426D, T427D) treatment of T cells affects T cell receptor-mediated early activation signaling events, T The induced surface expression levels of CD69, CD25, and IL-2 secretion were measured after cell receptor stimulation. As shown in Figures 1H and 1I, the expression levels of CD25, CD69, and interleukin-2 (IL-2) were not affected by treatment with ndSTAT1-TMD or ndSTAT1-TMD (V426D, V427D).
다음으로, Th1, Th2, Th17 및 조절 T 세포 (Treg)를 포함한 여러 CD4+ T 세포 하위 집합의 분화 동안 ndSTAT1-TMD의 기능을 조사하기 위해, 나이브 CD4+ T 세포는 ndSTAT1-TMD 또는 ndSTAT1-TMD (V426D, T427D)의 농도가 다른 경우 적절한 T 세포 하위 집합 스큐잉 조건 (the appropriate T cell subset-skewing conditions)에서 각 T 세포 하위 집합으로 분화하도록 유도되었다. Th1 세포에서 T-bet 발현, 세포 내 및 분비 IFN- γ의 수준은 농도 의존적 방식으로 ndSTAT1-TMD 처리에 의해 상당히 감소했으며, Th1 세포 분화에 대한 ndSTAT1-TMD (V426D, T427D)의 기능적 영향은 ndSTAT1-TMD보다 현저히 적었다 (도 2A-2B).Next, to investigate the function of ndSTAT1-TMD during the differentiation of multiple CD4 + T cell subsets, including Th1, Th2, Th17, and regulatory T cells (Treg), naïve CD4+ T cells were transfected with ndSTAT1-TMD or ndSTAT1-TMD ( Different concentrations of V426D, T427D) were induced to differentiate into each T cell subset under the appropriate T cell subset-skewing conditions. T-bet expression, levels of intracellular and secreted IFN-γ in Th1 cells were significantly reduced by ndSTAT1-TMD treatment in a concentration-dependent manner, and the functional impact of ndSTAT1-TMD (V426D, T427D) on Th1 cell differentiation was significantly reduced by ndSTAT1 It was significantly less than -TMD (Figures 2A-2B).
또한, Th1 세포에서 IFN- γ-STAT1 신호전달에 대한 1차 반응자 (as a primary responder)로 알려진 인터페론 조절인자 1 (interferon regulatory factor 1; IRF1)의 수준은 ndSTAT1-TMD 처리 후 감소하였으며, 이는 ndSTAT1-TMD (V426D, T427D)를 처리한 Th1군과 달리 감소하였다 (도 2c). 그리고 Th17 세포에서 ROR- γt 및 세포 내 IL-17A의 발현 수준도 ndSTAT1-TMD에 의해 농도 의존적으로 유의하게 감소하였다 (도 2D). 또한, ndSTAT1-TMD 처리는 Th17 세포로부터 IL-17A 및 IFN- γ의 분비를 감소시키는 것을 발견하였다 (도 2E-2F). 한편 병원성 Th17 세포는 IL-17A 및 IFN- γ을 분비할 수 있음이 제시되었다.In addition, the level of interferon regulatory factor 1 (IRF1), known as a primary responder to IFN-γ-STAT1 signaling in Th1 cells, decreased after ndSTAT1-TMD treatment, which is consistent with ndSTAT1 Unlike the Th1 group treated with -TMD (V426D, T427D), it decreased (Figure 2c). In addition, the expression levels of ROR-γt and intracellular IL-17A in Th17 cells were also significantly decreased by ndSTAT1-TMD in a concentration-dependent manner (Figure 2D). Additionally, we found that ndSTAT1-TMD treatment reduced the secretion of IL-17A and IFN-γ from Th17 cells (Figures 2E-2F). Meanwhile, it has been suggested that pathogenic Th17 cells can secrete IL-17A and IFN-γ.
한편, ndSTAT1-TMD는 Th2 세포에서 GATA3 또는 인터루킨-4 (IL-4)의 발현과 Treg 세포에서 Foxp3 발현을 조절하는 데 아무런 영향을 미치지 않는 것으로 나타났다. ndSTAT1-TMD(V426D, T427D) 처리된 세포는 Th1 또는 Th17 세포의 면역학적 기능과 관련된 유전자 발현의 인지 가능한 감소를 나타냈다. 그러나 감소 수준은 동일한 농도의 ndSTAT1-TMD로 처리된 세포에 비해 실질적으로 낮았다. 이러한 결과는 STAT1이 표적 유전자의 프로모터 영역에 결합하는 것 외에도 STAT1의 또 다른 기능적 도메인이 Th1 또는 Th17 세포 분화 조절에 부분적으로 관여한다는 것을 시사한다. 이러한 결과를 종합하면, ndSTAT1-TMD가 T 세포 수용체 매개 초기 T 세포 활성화 신호 전달 이벤트에 영향을 주지 않으면서 Th1 및 Th17 세포 모두의 분화 및 기능을 특이적으로 억제한다는 것을 보여준다.Meanwhile, ndSTAT1-TMD appeared to have no effect on regulating the expression of GATA3 or interleukin-4 (IL-4) in Th2 cells and Foxp3 expression in Treg cells. ndSTAT1-TMD (V426D, T427D) treated cells showed a perceptible decrease in the expression of genes associated with immunological functions of Th1 or Th17 cells. However, the level of reduction was substantially lower compared to cells treated with the same concentration of ndSTAT1-TMD. These results suggest that, in addition to STAT1 binding to the promoter region of target genes, another functional domain of STAT1 is partially involved in regulating Th1 or Th17 cell differentiation. Taken together, these results show that ndSTAT1-TMD specifically inhibits the differentiation and function of both Th1 and Th17 cells without affecting T cell receptor-mediated early T cell activation signaling events.
3. ndSTAT1-TMD에 의한 Th1/17 세포 분화의 억제는 Th1/17 세포의 기능과 관련된 STAT1 표적 유전자의 전사 억제에 기인함 (Inhibition of Th1/17 cell differentiation by ndSTAT1-TMD is attributed to the transcriptional suppression of STAT1 target genes associated with the functions of Th1/17 cells).3. Inhibition of Th1/17 cell differentiation by ndSTAT1-TMD is attributed to the transcriptional suppression of STAT1 target genes related to the function of Th1/17 cells of STAT1 target genes associated with the functions of Th1/17 cells).
다음으로, Th1 및 Th17 세포 분화 동안 ndSTAT1-TMD의 억제 기능이 전사 수준에서 표적 유전자 프로모터에 대한 내인성 STAT1 결합의 파괴로 인한 것인지 확인하고자 했다. STAT1이 IL-17A 프로모터에 결합하는 것을 감안하여, IL-17A 프로모터의 유도가 루시퍼라제 리포터 유전자의 발현을 유도하는 플라스미드를 야생형 STAT1을 코딩하는 유전자를 포함하는 플라스미드와 함께 HEK293T 세포에 공동 형질감염시켰다. 이어서, 형질감염된 세포를 다양한 농도의 ndSTAT1-TMD 또는 ndSTAT1-TMD (V426D, T427D)와 함께 배양하였다. ndSTAT1-TMD 처리군에서는 농도 의존적으로 루시퍼라제 활성이 유의하게 감소하였다. 반면, ndSTAT1-TMD 처리군 (V426D, T427D)에서는 억제 기능을 나타내지 않았다 (도 3A). 따라서, 이러한 결과는 ndSTAT1-TMD가 표적 세포 내에서 STAT1 매개 전사에 특이적으로 영향을 미친다는 것을 보여준다.Next, we sought to determine whether the suppressive function of ndSTAT1-TMD during Th1 and Th17 cell differentiation is due to disruption of endogenous STAT1 binding to target gene promoters at the transcriptional level. Given that STAT1 binds to the IL-17A promoter, a plasmid in which induction of the IL-17A promoter induces expression of a luciferase reporter gene was co-transfected into HEK293T cells together with a plasmid containing the gene encoding wild-type STAT1. . The transfected cells were then incubated with various concentrations of ndSTAT1-TMD or ndSTAT1-TMD (V426D, T427D). In the ndSTAT1-TMD treatment group, luciferase activity was significantly decreased in a concentration-dependent manner. On the other hand, the ndSTAT1-TMD treated group (V426D, T427D) did not show an inhibitory function (Figure 3A). Therefore, these results show that ndSTAT1-TMD specifically affects STAT1-mediated transcription within target cells.
활성화된 STAT1은 계통 확정의 초기 단계 (the early stage of lineage commitment)에서 IFN-γ를 분비하는 Th1 세포와 IL-17A를 분비하는 Th17 세포의 생성에 관여한다. 분화 과정에서 이와 같은 Th1 및/또는 Th17 세포의 기능적 확정(such functional commitment)이 중단되면 종종 자가면역 질환 및 면역 기능 장애가 발생하게 된다. ndSTAT1-TMD를 처리한 Th1 및 Th17 세포의 mRNA 발현 프로파일을 분석하여 Th1 및 Th17 세포의 기능과 관련된 유전자의 발현에 미치는 ndSTAT1-TMD의 내재적 역할을 평가했다. 유전자 세트 농축 분석(GSEA)은 ndSTAT1-TMD 처리 Th1 세포가 IFN-γ 반응과 관련된 시그니처 유전자에서 상당한 하향 조절을 보였으며 (그림 3B), ndSTAT1-TMD로 처리된 Th17 세포는 STAT3-녹아웃 CD4+ T 세포의 유전자 농축 패턴을 따른다 (그림 3C). Activated STAT1 is involved in the generation of Th1 cells secreting IFN-γ and Th17 cells secreting IL-17A in the early stage of lineage commitment. Disruption of such functional commitment of Th1 and/or Th17 cells during differentiation often leads to autoimmune diseases and immune dysfunction. The mRNA expression profile of Th1 and Th17 cells treated with ndSTAT1-TMD was analyzed to evaluate the intrinsic role of ndSTAT1-TMD on the expression of genes related to the function of Th1 and Th17 cells. Gene set enrichment analysis (GSEA) showed that ndSTAT1-TMD-treated Th1 cells showed significant downregulation in signature genes associated with the IFN-γ response (Figure 3B), while ndSTAT1-TMD-treated Th17 cells showed significant downregulation in STAT3-knockout CD4+ T cells. follows the gene enrichment pattern of (Figure 3C).
이러한 GSEA와 일치하게, ndSTAT1-TMD 처리된 Th1 세포의 mRNA 서열 분석 데이터는 Stat1Tbx21과 같은 Th1 세포 분화 및 기능에 관여하는 유전자와 Irf5와 같은 IFN-γ 반응에 필요한 유전자가 및 Irf7은 비교적 하향 조절되었다 (그림 3D). 마찬가지로, ndSTAT1-TMD로 처리된 Th17 세포에서 생성된 mRNA 서열 분석 데이터는 Rorc, Il17raCcl20과 같은 Th17 세포의 기능에 관여하는 유전자의 발현은 현저히 감소한 반면, OsmEomes와 같은 Th17 세포의 활성을 제한하는 것으로 알려진 유전자의 발현은 증가했음을 보여주었다 (그림 3E). 또한, Th1 세포에서 관찰된 바와 같이 Th17 세포의 Stat1 발현은 감소되었다 (그림 3E). 또한, 나이브 T 세포와 관련된 유전자는 처리되지 않은 그룹에 비해 ndSTAT1-TMD로 처리된 Th1 및 Th17 집단에서 더 풍부하다는 것을 발견했다 (그림 3D-3E). 이러한 결과를 종합하면, ndSTAT1-TMD에 의한 Th1 및 Th17 세포 분화의 억제는 STAT1 표적 유전자의 촉진체 영역에 대한 경쟁적 결합에 기인하며, 결과적으로 Th1 및 Th17 세포의 기능과 관련된 유전자의 발현이 하향 조절되었다.Consistent with this GSEA, mRNA sequencing data from ndSTAT1-TMD treated Th1 cells showed that genes involved in Th1 cell differentiation and function, such as Stat1 and Tbx21 , and genes required for the IFN-γ response, such as Irf5 , were relatively downregulated. was regulated (Figure 3D). Likewise, mRNA sequencing data generated from Th17 cells treated with ndSTAT1-TMD showed that the expression of genes involved in the function of Th17 cells, such as Rorc , Il17ra , and Ccl20, was significantly reduced, while the activity of Th17 cells, such as Osm and Eomes, was significantly reduced. Expression of genes known to be restrictive showed increased (Figure 3E). Additionally, Stat1 expression in Th17 cells was reduced, as observed in Th1 cells (Figure 3E). Additionally, we found that genes associated with naïve T cells were more abundant in the Th1 and Th17 populations treated with ndSTAT1-TMD compared to the untreated group (Figure 3D-3E). Taken together, these results suggest that the inhibition of Th1 and Th17 cell differentiation by ndSTAT1-TMD is due to competitive binding to the promoter region of STAT1 target genes, resulting in downregulation of the expression of genes related to the function of Th1 and Th17 cells. It has been done.
4. ndSTAT1-TMD는 IMQ에 의해 유발된 건선 증상을 완화함 (The ndSTAT1-TMD treatment alleviates the symptoms of psoriasis induced by IMQ)4. ndSTAT1-TMD alleviates psoriasis symptoms induced by IMQ (The ndSTAT1-TMD treatment alleviates the symptoms of psoriasis induced by IMQ)
건선 (psoriasis)은 과활성화된 Th1 및 Th17 세포에 의해 유발되는 질환으로 알려져 있기 때문에, 다음으로 ndSTAT1-TMD가 건선에 대한 치료 가능성이 있는지를 평가했다. 마우스의 등과 귀 피부에 IMQ (imiquimod) 크림을 7일 연속 도포하여 건선을 유도하였고, 양성 대조군으로 ndSTAT1-TMD, ndSTAT1-TMD (V426D, T427D) 또는 항 IL-17A 항체를 초기 질환 유도일로부터 2일 간격으로 복강 내 주사를 통해 투여하였다(그림 4A). 건선 유발 마우스의 등 피부를 분석하여 측정한 건선 면적 및 심각도 지수 (The psoriasis area and severity index; PASI) 점수는 5일차에 최고조에 달했다. 그런 다음 모델링 전반에 걸쳐 6일 또는 7일차에 점차 감소했다 (그림4B). 60㎍의 ndSTAT1-TMD로 치료한 그룹은 치료 그룹 중 5일차에 가장 낮은 PASI 점수를 나타냈다 (그림 4B). 마찬가지로, 5일차와 7일차의 등 피부 출현 상태와 관련하여, ndSTAT1-TMD 처리는 항IL-17A 항체 처리와 비교하여 병리학적 변화에 있어서 유의미한 개선을 보여주었다 (도 4C). 발병 5일째에는 건선 유발군에서 등 피부 두께가 2배 증가했다. 동시에, ndSTAT1-TMD 또는 항-IL-17A 항체 처리는 5일째 및 모델링 기간 전체에 걸쳐 등 피부 두께를 정상 그룹에 가깝게 실질적으로 감소시켰다 (도 4D, 4E). 생쥐의 체중 감소도 질병 심각도를 측정하는 기준 중 하나로 간주된다. ndSTAT1-TMD 또는 항-IL-17A 항체의 투여는 치료 효과와 일치하여 체중 감소를 부분적으로 예방했다 (그림 4F). 그러나, ndSTAT1-TMD(V426D, T427)의 치료 활성은 부분적이거나 미미했다(도 4B-4F).Since psoriasis is known to be a disease caused by overactivated Th1 and Th17 cells, we next evaluated whether ndSTAT1-TMD had therapeutic potential for psoriasis. Psoriasis was induced by applying IMQ (imiquimod) cream to the back and ear skin of mice for 7 consecutive days, and as a positive control, ndSTAT1-TMD, ndSTAT1-TMD (V426D, T427D) or anti-IL-17A antibody was applied 2 days from the initial disease induction date. It was administered via intraperitoneal injection at daily intervals (Figure 4A). The psoriasis area and severity index (PASI) score, measured by analyzing the back skin of psoriasis-induced mice, peaked on the 5th day. It then gradually decreased on day 6 or 7 throughout modeling (Figure 4B). The group treated with 60 μg of ndSTAT1-TMD had the lowest PASI score on day 5 among the treatment groups (Figure 4B). Likewise, with regard to the dorsal skin appearance status on days 5 and 7, ndSTAT1-TMD treatment showed significant improvement in pathological changes compared to anti-IL-17A antibody treatment (Figure 4C). On the 5th day of onset, the thickness of the back skin doubled in the psoriasis-induced group. At the same time, treatment with ndSTAT1-TMD or anti-IL-17A antibody substantially reduced dorsal skin thickness closer to the normal group at day 5 and throughout the modeling period (Figures 4D, 4E). Weight loss in mice is also considered one of the criteria for measuring disease severity. Administration of ndSTAT1-TMD or anti-IL-17A antibody partially prevented weight loss, consistent with the treatment effect (Figure 4F). However, the therapeutic activity of ndSTAT1-TMD (V426D, T427) was partial or minimal (Figures 4B-4F).
PASI 점수 플롯과 귀 피부의 피부 두께 변화와 같은 병리학적 패턴이 등 피부의 병리학적 패턴과 다름에도 불구하고 ndSTAT1-TMD 치료는 귀에서 가장 효과적인 치료 기능을 보여주었다. IMQ 유도 7일 후 각 그룹의 마우스의 혈청에서 TNF-α, IL-1β, IL-6 등 분비된 주요 전염증성 사이토카인의 농도를 평가하였을 때, 60 ㎍의 ndSTAT1-TMD 또는 항 IL-17A 항체를 처리한 그룹에서 혈청 내 농도의 유의한 감소가 관찰되었다 (그림 4G). 이러한 결과는 ndSTAT1-TMD가 항 IL-17A 항체만큼 효과적으로 건선 진행을 예방한다는 것을 보여준다.Despite the fact that the pathological patterns, such as the PASI score plot and skin thickness changes in the ear skin, were different from those in the back skin, ndSTAT1-TMD treatment showed the most effective treatment function in the ear. When evaluating the concentrations of major pro-inflammatory cytokines secreted in the serum of each group of mice 7 days after IMQ induction, such as TNF-α, IL-1β, and IL-6, 60 μg of ndSTAT1-TMD or anti-IL-17A antibody A significant decrease in serum concentration was observed in the treated group (Figure 4G). These results show that ndSTAT1-TMD prevents psoriasis progression as effectively as anti-IL-17A antibody.
5. 건선 동물 모델의 2차 림프 기관에서 CD4+ T 세포 서브세트의 파괴된 상태는 ndSTAT1-TMD 처리에 의해 복원된다 (The disrupted status of CD4+ T cell subsets in secondary lymphoid organs of psoriasis animal model is restored by ndSTAT1-TMD treatment)5. The disrupted status of CD4+ T cell subsets in secondary lymphoid organs of psoriasis animal model is restored by ndSTAT1-TMD treatment -TMD treatment)
Th1 세포와 Th17 세포는 건선 발병의 중요한 요인이고 ndSTAT1-TMD는 시험관 내 및 생체 내에서 Th1 및 Th17 세포 모두의 기능 및 분화 프로그램을 억제했기 때문에, 건선 동물 모델에 대한 ndSTAT1-TMD 치료가 생체 내에서 CD4+ T 세포 하위 집합의 기능적 상태를 변경한다고 가정하였다. 건선 동물 모델의 비장 및 배액 림프절 (draining lymph node; dLN)에서 CD4+ T 세포 서브세트의 프로파일을 조사한 결과, 건선 유발 마우스에서 T-bet+ 또는 IFN-γ+ CD4+ T 세포 및 ROR-γt+ 또는 IL-17A+ CD4+ T 세포의 수준이 높았다 (그림 5A-5D). ndSTAT1-TMD 처리는 비장 및 배액 림프절 (draining lymph node; dLN)에서 T-bet+ 및 ROR-γt+ CD4+ T 세포 둘 다의 수준을 실질적으로 감소시키는 것으로 관찰되었다. 항-IL-17A 항체 치료는 ROR-γt CD4+ T 세포에 상대적으로 특이적인 치료 기능을 보여주었다 (그림 5A-5B). ndSTAT1-TMD 또는 항-IL-17A 항체 치료 후에 이차 림프 기관에서 IFN-γ 및 IL-17A 분비 수준을 조사한 경우에도 일관된 결과가 얻어졌다 (그림 5C-5D). Th1 및 Th17 세포의 증가와 대조적으로, 건선 유발 마우스에서는 CD4+Foxp3+T 세포의 수준이 감소했다. 건선 유발 마우스에서 ndSTAT1-TMD에 의한 Th1 및 Th17 세포의 기능적 억제는 CD4+Foxp3+T 세포의 수준을 실질적으로 회복시켰다 (그림 5E). 종합적으로, 건선 유발 동물의 ndSTAT1-TMD 치료는 치료 효능을 나타냈고 다양한 T 세포 하위 집합의 기능적 상태를 회복시켰다.Because Th1 cells and Th17 cells are important factors in the development of psoriasis and ndSTAT1-TMD inhibited the function and differentiation program of both Th1 and Th17 cells in vitro and in vivo, ndSTAT1-TMD treatment on animal models of psoriasis may be effective in vivo. It was hypothesized to alter the functional status of CD4 + T cell subsets. Profiles of CD4 + T cell subsets in the spleen and draining lymph nodes (dLNs) of animal models of psoriasis revealed T-bet + or IFN-γ + CD4 + T cells and ROR-γt + in psoriasis-induced mice. Alternatively, there were higher levels of IL-17A + CD4 + T cells (Figures 5A-5D). ndSTAT1-TMD treatment was observed to substantially reduce the levels of both T-bet + and ROR-γt + CD4 + T cells in the spleen and draining lymph nodes (dLN). Anti-IL-17A antibody treatment showed relatively specific therapeutic function on ROR-γt CD4 + T cells (Figures 5A-5B). Consistent results were also obtained when we examined levels of IFN-γ and IL-17A secretion in secondary lymphoid organs after treatment with ndSTAT1-TMD or anti-IL-17A antibody (Figures 5C-5D). In contrast to the increase in Th1 and Th17 cells, the levels of CD4 + Foxp3 + T cells were decreased in psoriasis-induced mice. Functional inhibition of Th1 and Th17 cells by ndSTAT1-TMD in psoriasis-induced mice substantially restored the levels of CD4 + Foxp3 + T cells (Figure 5E). Overall, ndSTAT1-TMD treatment of psoriasis-induced animals showed therapeutic efficacy and restored the functional status of various T cell subsets.
6. ndSTAT1-TMD는 건선 유발 쥐의 피부에 백혈구가 침투하는 것을 제한 (ndSTAT1-TMD restricts the infiltration of leukocytes into the skin of psoriasis-induced mice)6. ndSTAT1-TMD restricts the infiltration of leukocytes into the skin of psoriasis-induced mice.
IMQ 유도 건선 마우스 모델의 조직병리학적 특징은 수지상 세포, 호중구, 대식세포 및 T세포로 구성된 다양한 백혈구의 침윤과 함께 극세포증 (acanthosis), 이상각화증 (parakeratosis), 신생혈관신생증과 같은 인간 플라크형 건선의 다인자적 특징과 매우 유사함이 입증되었다. 또한 여러 연구에서 CD4+ T 세포의 고갈이 IMQ로 인한 피부 염증을 크게 감소시키는 것으로 보고되었다. 건선 유발 쥐의 등과 귀 피부의 H&E 염색 부분은 표피의 두꺼워짐, 길쭉한 표피망 능선, 각화증 및 이상각화증을 보여주었다 (그림 6A). 그림 6A-6B에 표시된 것처럼 등과 귀 피부의 건선 관련 염증 매개변수는 60㎍의 ndSTAT1-TMD 또는 항-IL-17A 항체를 투여한 후 크게 감소했으나 ndSTAT-TMD (V426D, T427D)의 효과는 미미했다. 또한, 60 ㎍의 ndSTAT1-TMD 또는 항-IL-17A 항체를 처리한 그룹에서 Masson의 삼색 염색 부위를 조직학적으로 관찰한 결과, 진피 부위에서 각질세포의 침윤이 크게 감소하고 콜라겐 섬유의 점유가 감소하는 것으로 나타났다 (그림 6A).The histopathological features of the IMQ-induced psoriasis mouse model are human plaque-type psoriasis, such as acanthosis, parakeratosis, and neovascularization, along with infiltration of various leukocytes consisting of dendritic cells, neutrophils, macrophages, and T cells. It has been proven to be very similar to the multifactorial characteristics of . Additionally, several studies have reported that depletion of CD4 + T cells significantly reduces IMQ-induced skin inflammation. H&E-stained sections of the back and ear skin of psoriasis-induced mice showed epidermal thickening, elongated epidermal network ridges, keratosis, and dyskeratosis (Figure 6A). As shown in Figures 6A-6B, psoriasis-related inflammatory parameters of back and ear skin were significantly reduced after administration of 60 μg of ndSTAT1-TMD or anti-IL-17A antibody, but the effect of ndSTAT-TMD (V426D, T427D) was minimal. . In addition, histological observation of Masson's trichrome staining areas in the group treated with 60 μg of ndSTAT1-TMD or anti-IL-17A antibody showed that the infiltration of keratinocytes in the dermal area was significantly reduced and the occupancy of collagen fibers was reduced. It was found that (Figure 6A).
다음으로, 침윤하는 백혈구의 개체군 변화를 설명하기 위해 처리된 마우스의 귀와 등 피부의 진피와 표피에서 백혈구를 분리했다. 60㎍의 ndSTAT1-TMD 또는 항-IL-17A 항체를 처리한 그룹에서 귀와 등 피부의 진피에서 CD45+ 백혈구의 수준이 감소했으며, 이는 표피 영역에서는 관찰되지 않았다 (그림 6C). 건선 피부의 진피 부위에 위치한 대식세포가 IFN-γ 또는/및 IL-17A에 의해 활성화된다는 점을 고려하여, ndSTAT1-TMD가 대식세포의 활성화 및 동원을 제한할 수 있는지 여부를 조사했다. 항-IL-17A 항체 또는 ndSTAT1-TMD의 투여는 등과 귀 피부의 진피 영역에서 CD11b+ F4/80+ 대식세포 집단의 상당한 감소를 가져왔다 (그림 6D). 이 결과와 일치하게, 7일째 혈청 내 산화질소(the nitric oxide; NO) 수준은 ndSTAT1-TMD 또는 항-IL17A 항체로 처리한 그룹에서 실질적으로 감소했는데, 이는 ndSTAT1-TMD가 대식세포의 활성화 및 동원을 억제한다는 것을 나타낸다 (그림 6E).Next, we isolated leukocytes from the dermis and epidermis of the ear and back skin of treated mice to elucidate changes in the population of infiltrating leukocytes. In groups treated with 60 μg of ndSTAT1-TMD or anti-IL-17A antibody, the level of CD45 + leukocytes was reduced in the dermis of the ears and back skin, which was not observed in the epidermal area (Figure 6C). Considering that macrophages located in the dermal region of psoriatic skin are activated by IFN-γ or/and IL-17A, we investigated whether ndSTAT1-TMD could limit the activation and recruitment of macrophages. Administration of anti-IL-17A antibody or ndSTAT1-TMD resulted in a significant reduction of the CD11b + F4/80 + macrophage population in the dermal regions of the back and ear skin (Figure 6D). Consistent with these results, the nitric oxide (NO) level in serum at day 7 was substantially reduced in groups treated with ndSTAT1-TMD or anti-IL17A antibody, suggesting that ndSTAT1-TMD activates and recruits macrophages. (Figure 6E).
피부로의 대식세포 활성화 및 모집의 감소가 병원성 T 세포 하위 집합의 전반적인 감소 때문인지 조사하기 위해 귀 및 등 피부의 진피 및 표피 영역에서 백혈구에서 T 세포 하위 집합 특이적 전사 인자 및 사이토카인의 발현을 분석했다. 60㎍ 또는 20㎍의 ndSTAT1-TMD를 처리한 그룹은 진피 백혈구에서 CD45+ T-bet+ T 세포의 유의한 감소를 보였다 (그림 6F). 반면, 진피 백혈구의 CD45+ ROR-γt+ T 세포 집단의 수준은 ndSTAT1-TMD 또는 항-IL-17A 항체의 투여에 의해 크게 영향을 받지 않았다 (그림 6G). 일관되게, 60㎍의 ndSTAT1-TMD로 처리된 그룹은 진피 백혈구로부터 IFN-γ 분비의 실질적인 감소를 보여주었다 (그림 6H). 진피 백혈구로부터의 IL-17A 분비는 또한 ndSTAT1-TMD 또는 항-IL-17A 항체의 처리에 의해 감소되었다 (도 6I). 한편, ndSTAT1-TMD 및 항-IL-17A 항체 치료는 귀 및 등 피부의 표피 백혈구 내에서 대식세포 또는 T 세포에 의미 있는 억제 효과를 발휘하지 않습니다 (그림 6D 및 6F-6I). 또한, ndSTAT1-TMD (V426D, T427D) 처리군은 대식세포 및 T 세포의 기능적 조절에 효과를 나타내지 않았다 (도 6D-6I). 이러한 결과는 ndSTAT1-TMD가 대식세포뿐만 아니라 활성화된 Th1 및 Th17 세포를 제한함으로써 건선 피부의 진피 부위의 염증 상태를 개선한다는 것을 입증한다.To investigate whether the reduction in macrophage activation and recruitment to the skin is due to an overall reduction in pathogenic T cell subsets, we measured the expression of T cell subset-specific transcription factors and cytokines in leukocytes in the dermal and epidermal regions of ear and back skin. analyzed. Groups treated with 60 μg or 20 μg ndSTAT1-TMD showed a significant decrease in CD45 + T-bet + T cells in dermal leukocytes (Figure 6F). In contrast, the levels of the CD45 + ROR-γt + T cell population in dermal leukocytes were not significantly affected by administration of ndSTAT1-TMD or anti-IL-17A antibodies (Figure 6G). Consistently, the group treated with 60 μg of ndSTAT1-TMD showed a substantial reduction in IFN-γ secretion from dermal leukocytes (Figure 6H). IL-17A secretion from dermal leukocytes was also reduced by treatment with ndSTAT1-TMD or anti-IL-17A antibody (Figure 6I). Meanwhile, ndSTAT1-TMD and anti-IL-17A antibody treatment did not exert significant inhibitory effects on macrophages or T cells within the epidermal leukocytes of the ear and back skin (Figures 6D and 6F-6I). Additionally, the ndSTAT1-TMD (V426D, T427D) treatment group showed no effect on the functional regulation of macrophages and T cells (Figures 6D-6I). These results demonstrate that ndSTAT1-TMD improves the inflammatory state of the dermal region of psoriatic skin by limiting activated Th1 and Th17 cells as well as macrophages.
7. DSS 유발 대장염의 중증도는 ndSTAT1-TMD 치료에 의해 약화 (The severity of DSS-induced colitis is attenuated by ndSTAT1-TMD treatment)7. The severity of DSS-induced colitis is attenuated by ndSTAT1-TMD treatment
대장염은 Th1 또는/및 Th17 세포가 병원성 역할을 하는 또 다른 염증성 질환이다. 대장염 동물 모델에서 ndSTAT1-TMD의 치료 가능성을 평가하기 위해 덱스트란 황산나트륨 (dextran sulfate sodium; DSS) 유도 대장염 마우스 모델을 생성하고 동물을 ndSTAT1-TMD로 처리했으며, nti-IL-17A 항체 치료를 양성 대조군으로 사용했다 (도 7A). IBD의 중증도는 대장염 유도 후 6일째에 ndSTAT1-TMD, ndSTAT1-TMD (V426D, T427D) 또는 항-IL-17A 항체 치료를 복강 내 주사를 통해 6일째에 시작했고 또 다른 6일 동안 질병 진행을 매일 모니터링했다.Colitis is another inflammatory disease in which Th1 or/and Th17 cells play a pathogenic role. To evaluate the therapeutic potential of ndSTAT1-TMD in colitis animal models, we generated a dextran sulfate sodium (DSS)-induced colitis mouse model and animals were treated with ndSTAT1-TMD, and nti-IL-17A antibody treatment served as a positive control. was used (Figure 7A). The severity of IBD was assessed by starting treatment with ndSTAT1-TMD, ndSTAT1-TMD (V426D, T427D), or anti-IL-17A antibody on day 6 after colitis induction via intraperitoneal injection and monitoring disease progression daily for another 6 days. monitored.
ndSTAT1-TMD 또는 항IL-17A 항체를 처리한 그룹에서는 체중 감소율이 크게 개선된 반면, ndSTAT1-TMD (V426D, T427D) 처리 그룹에서는 지속적으로 체중이 감소했다 (도 7B). 대장염의 전체 질병 활성 지수(The overall disease activity index; DAI) 점수는 항-IL-17A 항체 치료 그룹과 유사하게 ndSTAT1-TMD 치료 그룹에서 실질적으로 감소했다 (도 7C). 이러한 데이터와 일치하게, 결장 길이는 ndSTAT1-TMD 또는 항-IL-17A 항체 100㎍마다 처리한 그룹에서 잘 유지되었습니다. 그러나 ndSTAT1-TMD (V426D, T427D) 처리군에서는 질병 진행이나 결장 길이의 개선이 나타나지 않았다 (도 7C-7D). 대장의 조직병리학적 상태를 알아보기 위해 복강내 투여 6일 후 대장 절편을 H&E 또는 PAS로 염색하였다. 침윤성 염증 세포의 증가, 술잔세포 고갈, 선와 손상 및 근육층 두께의 변화와 같은 염증성 대장의 징후는 항IL-17A 항체 또는 ndSTAT1-TMD 처리군에서 개선되었다 (그림 7E-7G). 반면, ndSTAT1-TMD 처리군 (V426D, T427D)에서는 대장염 중증도가 아무런 개선 없이 심각한 수준으로 유지되었다 (도 7E-7G).The weight loss rate was significantly improved in the group treated with ndSTAT1-TMD or anti-IL-17A antibody, whereas the group treated with ndSTAT1-TMD (V426D, T427D) continued to lose weight (Figure 7B). The overall disease activity index (DAI) score of colitis was substantially reduced in the ndSTAT1-TMD treatment group, similar to the anti-IL-17A antibody treatment group (Figure 7C). Consistent with these data, colon length was well maintained in groups treated with every 100 μg of ndSTAT1-TMD or anti-IL-17A antibody. However, no improvement in disease progression or colon length was seen in the ndSTAT1-TMD (V426D, T427D) treatment group (Figures 7C-7D). To determine the histopathological status of the colon, colon sections were stained with H&E or PAS 6 days after intraperitoneal administration. Signs of inflammatory colon, such as increased infiltrating inflammatory cells, goblet cell depletion, crypt damage, and changes in muscle layer thickness, were improved in the anti-IL-17A antibody or ndSTAT1-TMD treated groups (Figures 7E-7G). On the other hand, in the ndSTAT1-TMD treatment group (V426D, T427D), the severity of colitis remained at a severe level without any improvement (Figures 7E-7G).
12일째 각 처리군의 혈청에서 TNF-α, IL-1β, IL-6 등의 분비된 전염증성 사이토카인의 수준을 검사하였습니다. 이러한 전염증성 사이토카인의 분비는 용량 의존적으로 ndSTAT1-TMD를 처리한 군에서 유의하게 감소했다 (도 7H).On day 12, the serum of each treatment group was tested for the levels of secreted pro-inflammatory cytokines such as TNF-α, IL-1β, and IL-6. The secretion of these pro-inflammatory cytokines was significantly reduced in the ndSTAT1-TMD-treated group in a dose-dependent manner (Figure 7H).
건선 동물 모델의 결과에 따라 ndSTAT1-TMD가 염증성 Th1 및 Th17 세포의 병원성 기능을 억제하여 대장염 증상을 개선할 수 있다고 가정했다. PBS로 치료한 IBD 유도 마우스에서 비장 및 장간막 림프절 (mesenteric lymph node; mLN)의 T-bet+ 또는 IFN-γ+CD4+ T 세포와 ROR-γt+ 또는 IL-17A+CD4+ T 세포의 수준은 건강한 대조군보다 상당히 높았다. ndSTAT1-TMD 또는 항 IL-17A 항체 치료는 비장과 장간막 림프절 (mesenteric lymph node; mLN) 모두에서 T-bet+ 또는 IFN-γ+CD4+ T 세포와 ROR-γt+ 또는 IL-17A+CD4+ T 세포의 수를 크게 감소시켰다. 건선 동물 모델에서 ndSTAT1-TMD의 치료 잠재력과 일치하게, ndSTAT1-TMD는 Th1 세포와 Th17 세포에 대해 이중 억제 기능을 보였다. ndSTAT1-TMD 치료에 의한 Th1 및 Th17 세포의 감소는 비장 및 장간막 림프절 (mesenteric lymph node; mLN)에서 CD4+ Treg 세포를 발현하는 Foxp3의 증가를 동반했다. 이러한 결과를 종합하면, ndSTAT1-TMD 치료는 항 IL-17A 항체 치료에 필적하는 DSS 유도 대장염에서도 치료 효과를 보였다.Based on the results of psoriasis animal models, we hypothesized that ndSTAT1-TMD could improve colitis symptoms by suppressing the pathogenic functions of inflammatory Th1 and Th17 cells. The levels of T-bet + or IFN-γ + CD4 + T cells and ROR-γt + or IL-17A + CD4 + T cells in the spleen and mesenteric lymph nodes (mLN) in IBD-induced mice treated with PBS It was significantly higher than that of healthy controls. ndSTAT1-TMD or anti-IL-17A antibody treatment resulted in decreased T-bet + or IFN-γ + CD4 + T cells and ROR-γt + or IL-17A + CD4 + T cells in both the spleen and mesenteric lymph nodes (mLNs). The number of cells was greatly reduced. Consistent with the therapeutic potential of ndSTAT1-TMD in psoriasis animal models, ndSTAT1-TMD showed dual inhibitory functions against Th1 cells and Th17 cells. The decrease in Th1 and Th17 cells by ndSTAT1-TMD treatment was accompanied by an increase in Foxp3 expressing CD4 + Treg cells in the spleen and mesenteric lymph nodes (mLN). Taking these results together, ndSTAT1-TMD treatment showed a therapeutic effect in DSS-induced colitis comparable to anti-IL-17A antibody treatment.
이상으로 본 발명의 특정한 부분을 상세히 기술하였는 바, 당업계의 통상의 지식을 가진 자에게 있어서 이러한 구체적인 기술은 단지 바람직한 구현예일 뿐이며, 이에 본 발명의 범위가 제한되는 것이 아닌 점은 명백하다. 따라서, 본 발명의 실질적인 범위는 첨부된 청구항과 그의 등가물에 의하여 정의된다고 할 것이다.As the specific parts of the present invention have been described in detail above, it is clear to those skilled in the art that these specific techniques are merely preferred embodiments and do not limit the scope of the present invention. Accordingly, the substantial scope of the present invention will be defined by the appended claims and their equivalents.

Claims (31)

  1. STAT1 (Signal transducer and activator of transcription 1)의 전사 조절 도메인 및 단백질 운반 도메인 (Protein transduction domain, PTD)을 포함하는 융합 단백질.Fusion protein containing the transcriptional regulatory domain and protein transduction domain (PTD) of STAT1 (Signal transducer and activator of transcription 1).
  2. 제1항에 있어서,According to paragraph 1,
    상기 STAT1은 서열번호 13의 아미노산 서열로 표시되는, 융합 단백질.The STAT1 is a fusion protein represented by the amino acid sequence of SEQ ID NO: 13.
  3. 제1항에 있어서,According to paragraph 1,
    상기 STAT1의 전사 조절 도메인은 서열번호 14의 아미노산 서열로 표시되는, 융합 단백질.The transcriptional regulatory domain of STAT1 is a fusion protein represented by the amino acid sequence of SEQ ID NO: 14.
  4. 제3항에 있어서, According to paragraph 3,
    상기 단백질 운반 도메인은 Hph-1, Mph-1, Sim-2, Tat, VP22, Antp(antennapedia), Pep-1(peptide-1), PTD-5(protein transduction domain-5), 11R, 7R 및 CTP(cytoplamic transduction peptide)로 구성된 군으로부터 선택되는 것인, 융합 단백질.The protein transport domains include Hph-1, Mph-1, Sim-2, Tat, VP22, Antp (antennapedia), Pep-1 (peptide-1), PTD-5 (protein transduction domain-5), 11R, 7R and A fusion protein selected from the group consisting of CTP (cytoplamic transduction peptide).
  5. 제4항에 있어서, According to paragraph 4,
    상기 단백질 운반 도메인은 서열번호 1 내지 12 중 어느 하나의 아미노산 서열로 표시되는, 융합 단백질.A fusion protein wherein the protein transport domain is represented by any one of the amino acid sequences of SEQ ID NOs: 1 to 12.
  6. 제5항에 있어서,According to clause 5,
    상기 융합 단백질은 STAT1 매개 전사를 경쟁적으로 억제하는 것인, 융합 단백질.The fusion protein competitively inhibits STAT1-mediated transcription.
  7. 제6항에 있어서,According to clause 6,
    상기 융합 단백질은 나이브 CD4+ T 세포의 Th1 또는 Th17의 분화를 억제하는 것인, 융합 단백질.The fusion protein inhibits Th1 or Th17 differentiation of naïve CD4+ T cells.
  8. 제1항 내지 제7항 중 어느 한 항의 융합 단백질을 암호화하는 핵산 분자.A nucleic acid molecule encoding the fusion protein of any one of claims 1 to 7.
  9. 제8항의 핵산 분자를 포함하는 지질 나노입자 (LNP).Lipid nanoparticles (LNPs) comprising the nucleic acid molecule of claim 8.
  10. 제9항의 핵산 분자가 삽입된 발현 벡터.An expression vector into which the nucleic acid molecule of claim 9 is inserted.
  11. 제10항의 발현 벡터가 형질 감염된 숙주 세포.A host cell transfected with the expression vector of claim 10.
  12. 제1항 내지 제7항 중 어느 한 항의 융합 단백질, 제8항의 핵산분자, 제9항의 지질 나노입자(LNP), 제10항의 발현 벡터, 제11항의 숙주 세포 중 어느 하나를 유효 성분으로 포함하는 면역 관련 질환의 예방 또는 치료용 약학 조성물.Containing as an active ingredient any one of the fusion protein of any one of claims 1 to 7, the nucleic acid molecule of claim 8, the lipid nanoparticle (LNP) of claim 9, the expression vector of claim 10, and the host cell of claim 11. Pharmaceutical composition for preventing or treating immune-related diseases.
  13. 제12항에 있어서,According to clause 12,
    상기 면역 관련 질환은 Th1 세포 또는 Th17 세포의 활성화 또는 기능 이상에 따라 발생하는 것인, 면역 관련 질환의 예방 또는 치료용 약학 조성물.A pharmaceutical composition for preventing or treating an immune-related disease, wherein the immune-related disease occurs due to activation or dysfunction of Th1 cells or Th17 cells.
  14. 제12항에 있어서,According to clause 12,
    상기 면역 관련 질환은 자가면역질환, 퇴행성 뇌 또는 혈관 질환, 대사성 질환, 이식편대숙주 질환, 장기이식거부반응, 천식, 아토피, 및 급성 또는 만성의 염증 질환으로 이루어진 군으로부터 선택되는 적어도 하나인 것인, 면역 관련 질환의 예방 또는 치료용 약학 조성물.The immune-related disease is at least one selected from the group consisting of autoimmune disease, degenerative brain or vascular disease, metabolic disease, graft-versus-host disease, organ transplant rejection, asthma, atopy, and acute or chronic inflammatory disease. , Pharmaceutical composition for preventing or treating immune-related diseases.
  15. 제14항에 있어서,According to clause 14,
    상기 자가면역질환은 건선, 극세포증 (acanthosis), 이상각화증 (parakeratosis), 신생혈관신생증 (neo-angiogenesis), 염증성 장질환(Inflammatory bowel disease, IBD), 류마티스성 관절염, 전신성 경피증, 전신 홍반성 낭창, 아토피 피부염, 원형탈모증, 천식, 크론씨병, 베체시병, 쇼그렌 증후군, 길리아-바레 증후군, 만성 갑상선염, 다발성 경화증, 다발성 근염, 강직성 척추염, 섬유조직염 및 결절성 다발성 동맥염으로 구성된 군으로부터 선택된 어느 하나 이상인 것을 특징으로 하는, 면역 관련 질환의 예방 또는 치료용 약학 조성물.The autoimmune diseases include psoriasis, acanthosis, parakeratosis, neo-angiogenesis, inflammatory bowel disease (IBD), rheumatoid arthritis, systemic scleroderma, and systemic lupus erythematosus. , atopic dermatitis, alopecia areata, asthma, Crohn's disease, Behce's disease, Sjögren's syndrome, Guillain-Barré syndrome, chronic thyroiditis, multiple sclerosis, polymyositis, ankylosing spondylitis, fibromyositis, and polyarteritis nodosa. A pharmaceutical composition for preventing or treating immune-related diseases.
  16. 제14항에 있어서,According to clause 14,
    상기 급성 또는 만성의 염증 질환은 대장염, 패혈증(sepsis), 패혈성 쇼크, 염증성 장질환(Inflammatory bowel disease, IBD), 건선, 복막염, 신장염, 급성 기관지염, 만성 기관지염, 골관절염, 장질환 척추염, 만성 폐쇄성 폐질환 (chronic obstructive pulmonary disease, COPD), 류마티스성 관절염(rheumatoid arthritis), 급성 폐손상 (acute lung injury) 및 기관지 폐 형성장애(broncho-pulmonary dysplasia)로 이루어진 군에서 선택된 어느 하나 이상인 것을 특징으로 하는, 면역 관련 질환의 예방 또는 치료용 약학 조성물.The acute or chronic inflammatory diseases include colitis, sepsis, septic shock, inflammatory bowel disease (IBD), psoriasis, peritonitis, nephritis, acute bronchitis, chronic bronchitis, osteoarthritis, enteropathy spondylitis, and chronic obstructive inflammatory disease. Characterized by at least one selected from the group consisting of chronic obstructive pulmonary disease (COPD), rheumatoid arthritis, acute lung injury, and broncho-pulmonary dysplasia , Pharmaceutical composition for preventing or treating immune-related diseases.
  17. 제14항에 있어서,According to clause 14,
    상기 퇴행성 뇌 또는 혈관 질환은 뇌졸중, 치매, 알츠하이머병(Alzheimer's disease), 파킨슨병(Parkinson's disease), 헌팅턴병(Huntington's disease), 니만-피크병(Niemann-Pick disease), 다발성 경화증, 프리온병(prion disease), 크로이펠츠-야콥병(Creutzfeldt-Jakob disease), 전두측두치매, 루이치매, 근위축성 측삭경화증(AlzAmyotrophic lateral sclerosis), 부신생물증후군(Paraneoplastic syndrome), 피질기저퇴행증, 다계통위축병, 진행성핵상마비, 신경계 자가면역질환, 척수 소뇌 실조(spinocerebellar ataxia), 염증성 및 신경병증성 통증, 뇌혈관 질환, 척수 손상(spinal cord injury) 및 타우병증(tauopathy)으로 이루어진 군에서 선택되는, 약학 조성물.The degenerative brain or blood vessel diseases include stroke, dementia, Alzheimer's disease, Parkinson's disease, Huntington's disease, Niemann-Pick disease, multiple sclerosis, and prion disease. ), Creutzfeldt-Jakob disease, frontotemporal dementia, Lewy dementia, Amyotrophic lateral sclerosis, Paraneoplastic syndrome, corticobasal degeneration, multiple system atrophy, progressive supranuclear disease A pharmaceutical composition selected from the group consisting of paralysis, nervous system autoimmune diseases, spinocerebellar ataxia, inflammatory and neuropathic pain, cerebrovascular diseases, spinal cord injury and tauopathy.
  18. 제14항에 있어서,According to clause 14,
    상기 대사성 질환은 인슐린 저항성 질환, 지방간, 당뇨병, 제 2형 당뇨, 고지혈증, 고중성지방혈증, 이상지질혈증, 심혈관 질환, 뇌졸증, 심근경색, 과혈당증, 과인슐린혈증, 동맥경화, 고혈압, 심장병, 비만, 간 질환, 신장 손상 및 고혈압으로 이루어진 군으로부터 선택된 1종 이상인 것인, 약학 조성물.The metabolic diseases include insulin resistance disease, fatty liver, diabetes, type 2 diabetes, hyperlipidemia, hypertriglyceridemia, dyslipidemia, cardiovascular disease, stroke, myocardial infarction, hyperglycemia, hyperinsulinemia, arteriosclerosis, hypertension, heart disease, and obesity. , a pharmaceutical composition that is one or more selected from the group consisting of liver disease, kidney damage, and high blood pressure.
  19. 제1항 내지 제7항 중 어느 한 항의 융합 단백질, 제8항의 핵산분자, 제9항의 지질 나노입자(LNP), 제10항의 발현 벡터, 제11항의 숙주 세포 중 어느 하나를 유효 성분으로 포함하는 암의 예방 또는 치료용 약학 조성물.Containing as an active ingredient any one of the fusion protein of any one of claims 1 to 7, the nucleic acid molecule of claim 8, the lipid nanoparticle (LNP) of claim 9, the expression vector of claim 10, and the host cell of claim 11. Pharmaceutical composition for preventing or treating cancer.
  20. 제19항에 있어서,According to clause 19,
    상기 암은 Th1 세포 또는 Th17 세포의 활성화 또는 기능 이상에 따라 발생하는 것인, 암의 예방 또는 치료용 약학 조성물.A pharmaceutical composition for preventing or treating cancer, wherein the cancer occurs due to activation or dysfunction of Th1 cells or Th17 cells.
  21. 제20항에 있어서,According to clause 20,
    상기 암은 위암, 간암, 교세포종, 난소암, 대장암, 두경부암, 방광암, 신장세포암, 유방암, 전이암, 전립선암, 췌장암, 흑색종 또는 폐암인, 약학 조성물.The pharmaceutical composition, wherein the cancer is stomach cancer, liver cancer, glioblastoma, ovarian cancer, colon cancer, head and neck cancer, bladder cancer, renal cell cancer, breast cancer, metastatic cancer, prostate cancer, pancreatic cancer, melanoma, or lung cancer.
  22. 제1항 내지 제7항 중 어느 한 항의 융합 단백질, 제8항의 핵산분자, 제9항의 지질 나노입자(LNP), 제10항의 발현 벡터, 제11항의 숙주 세포 중 어느 하나를 대상체에 대하여 투여하는, 면역 관련 질환의 치료방법.Administering to a subject any one of the fusion protein of any one of claims 1 to 7, the nucleic acid molecule of claim 8, the lipid nanoparticle (LNP) of claim 9, the expression vector of claim 10, and the host cell of claim 11. , Treatment method for immune-related diseases.
  23. 제22항에 있어서,According to clause 22,
    상기 면역 관련 질환은 Th1 세포 또는 Th17 세포의 활성화 또는 기능 이상에 따라 발생하는 것인, 치료방법.A treatment method in which the immune-related disease occurs due to activation or dysfunction of Th1 cells or Th17 cells.
  24. 제23항에 있어서,According to clause 23,
    상기 면역 관련 질환은 자가면역질환, 퇴행성 뇌 또는 혈관 질환, 대사성 질환, 이식편대숙주 질환, 장기이식거부반응, 천식, 아토피, 및 급성 또는 만성의 염증 질환으로 이루어진 군으로부터 선택되는 적어도 하나인 것인, 면역 관련 질환의 치료방법.The immune-related disease is at least one selected from the group consisting of autoimmune disease, degenerative brain or vascular disease, metabolic disease, graft-versus-host disease, organ transplant rejection, asthma, atopy, and acute or chronic inflammatory disease. , Treatment method for immune-related diseases.
  25. 제24항에 있어서,According to clause 24,
    상기 자가면역질환은 건선, 극세포증 (acanthosis), 이상각화증 (parakeratosis), 신생혈관신생증 (neo-angiogenesis), 염증성 장질환(Inflammatory bowel disease, IBD), 류마티스성 관절염, 전신성 경피증, 전신 홍반성 낭창, 아토피 피부염, 원형탈모증, 천식, 크론씨병, 베체시병, 쇼그렌 증후군, 길리아-바레 증후군, 만성 갑상선염, 다발성 경화증, 다발성 근염, 강직성 척추염, 섬유조직염 및 결절성 다발성 동맥염으로 구성된 군으로부터 선택된 어느 하나 이상인 것을 특징으로 하는, 면역 관련 질환의 치료방법.The autoimmune diseases include psoriasis, acanthosis, parakeratosis, neo-angiogenesis, inflammatory bowel disease (IBD), rheumatoid arthritis, systemic scleroderma, and systemic lupus erythematosus. , atopic dermatitis, alopecia areata, asthma, Crohn's disease, Behce's disease, Sjögren's syndrome, Guillain-Barré syndrome, chronic thyroiditis, multiple sclerosis, polymyositis, ankylosing spondylitis, fibromyositis, and polyarteritis nodosa. A method of treating immune-related diseases, characterized in that.
  26. 제24항에 있어서,According to clause 24,
    상기 급성 또는 만성의 염증 질환은 대장염, 패혈증(sepsis), 패혈성 쇼크, 염증성 장질환(Inflammatory bowel disease, IBD), 건선, 복막염, 신장염, 급성 기관지염, 만성 기관지염, 골관절염, 장질환 척추염, 만성 폐쇄성 폐질환 (chronic obstructive pulmonary disease, COPD), 류마티스성 관절염(rheumatoid arthritis), 급성 폐손상 (acute lung injury) 및 기관지 폐 형성장애(broncho-pulmonary dysplasia)로 이루어진 군에서 선택된 어느 하나 이상인 것을 특징으로 하는, 면역 관련 질환의 치료방법.The acute or chronic inflammatory diseases include colitis, sepsis, septic shock, inflammatory bowel disease (IBD), psoriasis, peritonitis, nephritis, acute bronchitis, chronic bronchitis, osteoarthritis, enteropathy spondylitis, and chronic obstructive inflammatory disease. Characterized by at least one selected from the group consisting of chronic obstructive pulmonary disease (COPD), rheumatoid arthritis, acute lung injury, and broncho-pulmonary dysplasia , Treatment method for immune-related diseases.
  27. 제24항에 있어서,According to clause 24,
    상기 퇴행성 뇌 또는 혈관 질환은 뇌졸중, 치매, 알츠하이머병(Alzheimer's disease), 파킨슨병(Parkinson's disease), 헌팅턴병(Huntington's disease), 니만-피크병(Niemann-Pick disease), 다발성 경화증, 프리온병(prion disease), 크로이펠츠-야콥병(Creutzfeldt-Jakob disease), 전두측두치매, 루이치매, 근위축성 측삭경화증(AlzAmyotrophic lateral sclerosis), 부신생물증후군(Paraneoplastic syndrome), 피질기저퇴행증, 다계통위축병, 진행성핵상마비, 신경계 자가면역질환, 척수 소뇌 실조(spinocerebellar ataxia), 염증성 및 신경병증성 통증, 뇌혈관 질환, 척수 손상(spinal cord injury) 및 타우병증(tauopathy)으로 이루어진 군에서 선택되는, 치료방법.The degenerative brain or blood vessel diseases include stroke, dementia, Alzheimer's disease, Parkinson's disease, Huntington's disease, Niemann-Pick disease, multiple sclerosis, and prion disease. ), Creutzfeldt-Jakob disease, frontotemporal dementia, Lewy dementia, Amyotrophic lateral sclerosis, Paraneoplastic syndrome, corticobasal degeneration, multiple system atrophy, progressive supranuclear disease A method of treatment selected from the group consisting of paralysis, nervous system autoimmune diseases, spinocerebellar ataxia, inflammatory and neuropathic pain, cerebrovascular diseases, spinal cord injury, and tauopathy.
  28. 제14항에 있어서,According to clause 14,
    상기 대사성 질환은 인슐린 저항성 질환, 지방간, 당뇨병, 제 2형 당뇨, 고지혈증, 고중성지방혈증, 이상지질혈증, 심혈관 질환, 뇌졸증, 심근경색, 과혈당증, 과인슐린혈증, 동맥경화, 고혈압, 심장병, 비만, 간 질환, 신장 손상 및 고혈압으로 이루어진 군으로부터 선택된 1종 이상인 것인, 치료방법.The metabolic diseases include insulin resistance disease, fatty liver, diabetes, type 2 diabetes, hyperlipidemia, hypertriglyceridemia, dyslipidemia, cardiovascular disease, stroke, myocardial infarction, hyperglycemia, hyperinsulinemia, arteriosclerosis, hypertension, heart disease, and obesity. , liver disease, kidney damage, and high blood pressure.
  29. 제1항 내지 제7항 중 어느 한 항의 융합 단백질, 제8항의 핵산분자, 제9항의 지질 나노입자(LNP), 제10항의 발현 벡터, 제11항의 숙주 세포 중 어느 하나를 대상체에게 투여하는 암의 치료방법.Cancer in which any one of the fusion protein of any one of claims 1 to 7, the nucleic acid molecule of claim 8, the lipid nanoparticle (LNP) of claim 9, the expression vector of claim 10, or the host cell of claim 11 is administered to a subject. treatment method.
  30. 제29항에 있어서,According to clause 29,
    상기 암은 Th1 세포 또는 Th17 세포의 활성화 또는 기능 이상에 따라 발생하는 것인, 암의 치료방법.A method of treating cancer, wherein the cancer occurs due to activation or dysfunction of Th1 cells or Th17 cells.
  31. 제30항에 있어서,According to clause 30,
    상기 암은 위암, 간암, 교세포종, 난소암, 대장암, 두경부암, 방광암, 신장세포암, 유방암, 전이암, 전립선암, 췌장암, 흑색종 또는 폐암인, 치료방법.The cancer is stomach cancer, liver cancer, glioblastoma, ovarian cancer, colon cancer, head and neck cancer, bladder cancer, renal cell cancer, breast cancer, metastatic cancer, prostate cancer, pancreatic cancer, melanoma, or lung cancer.
PCT/KR2023/019074 2022-11-24 2023-11-24 Use of stat1-tmd in immune-related diseases WO2024112143A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
KR20220159214 2022-11-24
KR10-2022-0159214 2022-11-24

Publications (1)

Publication Number Publication Date
WO2024112143A1 true WO2024112143A1 (en) 2024-05-30

Family

ID=91196351

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2023/019074 WO2024112143A1 (en) 2022-11-24 2023-11-24 Use of stat1-tmd in immune-related diseases

Country Status (2)

Country Link
KR (1) KR20240087555A (en)
WO (1) WO2024112143A1 (en)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5716622A (en) * 1995-01-06 1998-02-10 The Rockefeller University Functionally active regions of signal transducer and activators of transcription
US6160092A (en) * 1998-05-29 2000-12-12 The Rockefeller University Crystal of the core portion of a signal transducer and activator of transcription (STAT)
US20130231274A1 (en) * 2010-08-20 2013-09-05 Sang-Kyou Lee Fusion protein having transcription factor transactivation-regulating domain and protein transduction domain, and transcription factor function inhibitor comprising the same
US20190315820A1 (en) * 2016-01-25 2019-10-17 Good T Cells, Inc. A Composition Containing SMAD Protein for Treatment of Autoimmune Diseases, a Fusion Protein Comprising SMAD Protein, an Expression Vector and a Method for Preparing the Same
CN112807425A (en) * 2021-01-14 2021-05-18 南方医科大学深圳医院 tTIM fusion protein vaccine, preparation method and application

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5716622A (en) * 1995-01-06 1998-02-10 The Rockefeller University Functionally active regions of signal transducer and activators of transcription
US6160092A (en) * 1998-05-29 2000-12-12 The Rockefeller University Crystal of the core portion of a signal transducer and activator of transcription (STAT)
US20130231274A1 (en) * 2010-08-20 2013-09-05 Sang-Kyou Lee Fusion protein having transcription factor transactivation-regulating domain and protein transduction domain, and transcription factor function inhibitor comprising the same
US20190315820A1 (en) * 2016-01-25 2019-10-17 Good T Cells, Inc. A Composition Containing SMAD Protein for Treatment of Autoimmune Diseases, a Fusion Protein Comprising SMAD Protein, an Expression Vector and a Method for Preparing the Same
CN112807425A (en) * 2021-01-14 2021-05-18 南方医科大学深圳医院 tTIM fusion protein vaccine, preparation method and application

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
PARK JIYOON, SON MIN-JI; HO CHUN-CHANG; KIM YUNA; AN JAEKYEUNG; LEE SANG-KYOU: "Transcriptional inhibition of STAT1 functions in the nucleus alleviates Th1 and Th17 cell-mediated inflammatory diseases", FRONTIERS IN IMMUNOLOGY, FRONTIERS MEDIA, LAUSANNE, CH, vol. 13, 15 December 2022 (2022-12-15), Lausanne, CH , pages 1 - 19, XP093174175, ISSN: 1664-3224, DOI: 10.3389/fimmu.2022.1054472 *

Also Published As

Publication number Publication date
KR20240087555A (en) 2024-06-19

Similar Documents

Publication Publication Date Title
WO2015156649A1 (en) Peptide having fibrosis inhibitory activity and composition containing same
WO2021010621A1 (en) Peptide therapeutics for autoimmune diseases and inflammatory diseases
WO2016137162A1 (en) Peptide for preventing hearing loss, and composition comprising same
WO2013169077A1 (en) Composition for preventing or treating cachexia
WO2018092955A1 (en) Antimicrobial peptide analog derived from haliotis discus and antimicrobial pharmaceutical composition containing same
WO2013129739A1 (en) Pharmaceutical composition comprising bee venom-phospholipase a2 (bv-pla2) for treating or preventing diseases related to degradation of abnormal regulatory t cell activity
WO2016190660A1 (en) Novel peptide and composition containing the same
WO2019103541A1 (en) Anti-inflammatory composition comprising graphene nano-structure
JP5911869B2 (en) Fusion protein having transcription regulatory domain and protein transduction domain and inhibitor for transcription factor function comprising the same
WO2018056706A1 (en) Composition comprising thioredoxin-interacting protein-derived peptide or polynucleotide encoding same as effective ingredient for reverse-ageing of aged stem cell and use thereof
WO2017034244A1 (en) Peptide having effect of preventing or treating central nervous system diseases and pharmaceutical composition for preventing and treating central nervous system diseases, containing same as active ingredient
WO2017043920A1 (en) Skin permeable peptide and method for using same
WO2012118323A2 (en) Fusion protein comprising albumin and a retinol-binding protein
WO2019147036A1 (en) Mesenchymal stem cells expressing brain-derived neurotrophic factor and use thereof
WO2024112143A1 (en) Use of stat1-tmd in immune-related diseases
WO2020159191A1 (en) Composition comprising mls-stat3 for prevention or treatment of immune disease
WO2020116810A1 (en) Pharmaceutical composition, comprising inhibitory peptide against fas signaling, for prevention or treatment of obesity, fatty liver, or steatohepatitis
WO2013129852A1 (en) Repebody for novel interleukin-6 and use thereof
WO2016027990A1 (en) Pharmaceutical composition containing dusp5 as active ingredient for preventing or treating bone metabolic diseases
WO2018203613A1 (en) Peptide for toll-like receptor (tlr) inhibition and pharmaceutical composition comprising same
DE60222265T2 (en) CELL TOD INDUCTORS FOR MAST CELLS
WO2018021778A1 (en) Composition for preventing or treating impotence, ischemic diseases or peripheral nerve diseases, containing fusion protein comprising lrg1 glycoprotein
WO2015023165A1 (en) Inflammation-controlling composite and stabilized mesenchymal stem cells having optimized immunity control function by blocking stat3 signal molecule
WO2022010319A1 (en) Fusion protein including glucagon-like peptide-1 and interleukin-1 receptor antagonist and use thereof
WO2020241925A1 (en) Lipid nanoparticle complex capturing aptide fused with cell-penetrating material and use thereof