WO2021194294A1 - Avatar model platform for evaluating autoimmune diseases - Google Patents

Avatar model platform for evaluating autoimmune diseases Download PDF

Info

Publication number
WO2021194294A1
WO2021194294A1 PCT/KR2021/003756 KR2021003756W WO2021194294A1 WO 2021194294 A1 WO2021194294 A1 WO 2021194294A1 KR 2021003756 W KR2021003756 W KR 2021003756W WO 2021194294 A1 WO2021194294 A1 WO 2021194294A1
Authority
WO
WIPO (PCT)
Prior art keywords
animal model
model
cells
animal
organoid
Prior art date
Application number
PCT/KR2021/003756
Other languages
French (fr)
Korean (ko)
Inventor
박성환
조미라
곽승기
이주하
이재선
문정현
황선희
박진실
박민정
백진아
최정원
나현식
이선영
조근형
Original Assignee
가톨릭대학교 산학협력단
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from KR1020200037761A external-priority patent/KR20210120705A/en
Priority claimed from KR1020200037764A external-priority patent/KR20210120706A/en
Priority claimed from KR1020200037763A external-priority patent/KR20210121366A/en
Priority claimed from KR1020200037762A external-priority patent/KR20210121365A/en
Application filed by 가톨릭대학교 산학협력단 filed Critical 가톨릭대학교 산학협력단
Publication of WO2021194294A1 publication Critical patent/WO2021194294A1/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K67/00Rearing or breeding animals, not otherwise provided for; New breeds of animals
    • A01K67/027New breeds of vertebrates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing

Definitions

  • the present invention relates to an autoimmune disease evaluation avatar model platform.
  • Autoimmune disease is when the body's immune system malfunctions and its own cells attack their own cells.
  • the human immune system basically recognizes foreign antigens against invading microorganisms and cancer cells, and has strong power to attack and remove them, but it does not attack its own cells because of its self-tolerance. This phenomenon is called self-tolerance of the human body.
  • autoreactive T cells that respond to self-cells (or autoantigens) are activated and autoantibodies are generated, constantly destroying self-cells, causing inflammation and immune responses. .
  • T cells that specifically respond to antigens in the immune system include T cells and B cells.
  • T cells meet a specific antigen presented by an antigen presenting cell, they respond according to the antigen.
  • the antigen presented by the antigen presenting cell is recognized as 'non-self', an immune response to remove it , and if recognized as 'self', the immune response is ignored and tolerated.
  • T cells are activated against an antigen, most B cells are activated one after another, and the B cells turn into plasma cells to produce antibodies that specifically react to the recognized antigen. Therefore, when autoimmunity occurs due to the breakdown of tolerance in the body, T cells are activated by abnormal recognition of autoantigens, and B cells are also activated to produce autoantibodies that respond to autoantigens. an immune response occurs.
  • tailored medicine is also called order-made medicine or personalized medicine. means how to treat it.
  • biomarkers for genomic diagnosis various genomic test methods, and medical informatics that can analyze/integrate genomic information derived using the biomarkers or obtained by the test method
  • Analysis methods and targeted treatment technologies to which the results analyzed by the above analysis methods can be applied should be developed in advance and complementary to each other. That is, it is necessary not only to develop a technology for selecting a treatment suitable for a patient, but also to develop a technology for verifying the selected treatment.
  • an organoid is an organ-specific cell aggregate made by three-dimensionally culturing, aggregating, or recombination of stem cells, capable of self-renewal, and is also called a 'mini organ' or 'similar organ'.
  • Organoids are very useful for basic research as they can implement in vitro studies that are difficult to implement in animal models such as molecular signal regulation in a form similar to that of real organs, as well as human development, disease model establishment, drug efficacy evaluation screening, It is a technology that can be very usefully used in various fields such as drug toxicity evaluation platform development and cell therapy drug development.
  • PBMCs peripheral blood mononuclear cells
  • An object of the present invention is to develop a method for producing a humanized lupus disease animal model, comprising injecting PBMC isolated from a lupus disease patient into immunodeficient mice.
  • An object of the present invention is to develop a method for producing an animal model of rheumatoid arthritis accompanied by lung fibrosis, comprising the step of inducing lung fibrosis by administering beta-glucan or a fungicide to a mouse with rheumatoid arthritis.
  • An object of the present invention is to develop an animal model of osteoarthritis accompanied by gout, in which gout is generated by administering potassium oxonate (PO) or hypoxanthine (HX) to an animal with osteoarthritis.
  • PO potassium oxonate
  • HX hypoxanthine
  • An object of the present invention is to develop a method for producing an animal model of osteoarthritis accompanied by gout, including the step of inducing gout by administering potassium oxonate or hypoxanthine to an animal with osteoarthritis.
  • An object of the present invention is a mouse To develop an organoid model of Sjogren's syndrome in which Interleukin-17 is administered to an organoid obtained by culturing the derived salivary gland stem cells.
  • An object of the present invention is a mouse culturing and obtaining derived salivary gland stem cells
  • an organoid model production method for Sjogren's syndrome which includes the step of culturing the cells to generate an organoid and then administering Interleukin-17.
  • the present invention provides a humanized lupus (systemic lupus erythematosus) disease animal model in which immunodeficient mice are administered with PBMCs (peripheral blood mononuclear cells) derived from lupus disease patients.
  • PBMCs peripheral blood mononuclear cells
  • the present invention provides a method for manufacturing a humanized lupus disease animal model, comprising injecting PBMC isolated from a lupus disease patient into an immunodeficient mouse.
  • the present invention provides an animal model of rheumatoid arthritis accompanied by pulmonary fibrosis, in which pulmonary fibrosis is generated by administering beta-glucan or a fungicide to an animal with rheumatoid arthritis.
  • the present invention provides a method for preparing an animal model of rheumatoid arthritis accompanied by lung fibrosis, comprising the step of inducing lung fibrosis by administering beta-glucan or a fungicide to a mouse with rheumatoid arthritis.
  • the present invention provides an animal model of osteoarthritis accompanied by gout, in which gout is generated by administering potassium oxonate (PO) or hypoxanthine (HX) to an animal with osteoarthritis.
  • gout is generated by administering potassium oxonate (PO) or hypoxanthine (HX) to an animal with osteoarthritis.
  • the present invention provides a method for producing an animal model of osteoarthritis accompanied by gout, comprising the step of inducing gout by administering potassium oxonate or hypoxanthine to an animal with osteoarthritis.
  • the present invention is a mouse Interleukin-17 (Interleukin-17) is administered to an organoid obtained by culturing the derived salivary gland stem cells, Sjogren's syndrome provides an organoid model.
  • the present invention provides a Sjogren's syndrome organoid model, including a Sjogren patient-derived salivary gland epithelial cell (Patient-derived salivary gland epithelial cell).
  • the present invention is a mouse culturing and obtaining derived salivary gland stem cells; And it provides a method for producing an organoid model of Sjogren's syndrome, comprising the step of culturing the cells to generate an organoid and then administering Interleukin-17.
  • the present invention provides a method for manufacturing a Sjogren's syndrome organoid model, comprising the step of culturing Sjogren patient-derived salivary gland epithelial cells.
  • the autoimmune disease model according to the present invention reflects the immune status of the autoimmune disease patient well, it can be used not only as a preclinical model for monitoring the patient's immune status, but also can be used for screening of therapeutic substances for autoimmune diseases. can Therefore, there is no treatment drug, which was a problem in the past, and monitoring drugs at the current animal test level can overcome the limitations of not being successful in clinical practice. It can be used in the field or industry related to autoimmune disease.
  • FIG. 1 is a diagram schematically illustrating the lupus patient imitation avatar model construction protocol of the present invention.
  • FIG. 2 is a diagram showing the results of reacting with human CD4, CD8 and CD19 antibodies to confirm human cell engraftment in a lupus patient mimic avatar model.
  • FIG. 3 is a diagram illustrating the degree of infiltration of human immune cell subtypes (CD3, CD4, CD8, CD19) in tissues in order to confirm human cell engraftment in a lupus patient mimic avatar model.
  • 4A, 4B and 4C are diagrams showing the results of measuring the dsDNA level measurement method, the urine human albumin concentration method, and the creatine concentration in order to confirm the autoantibody and proteinuria changes in the simulated avatar model of a lupus patient.
  • FIG. 5 is a view confirming the infiltration of inflammatory cells in order to confirm the histological change of the kidney in the lupus patient simulated avatar model.
  • FIG. 6 is a diagram comparing the arthritis scores for the curdlan-injected rheumatoid arthritis SKG animal model with lung fibrosis and the control group.
  • FIG. 7 is a diagram comparing the weight change of the curdlan-injected rheumatoid arthritis SKG animal model with lung fibrosis and the control group.
  • 9A, 9B, 9C, 9D, 9E and 9F show the curdlan-injected rheumatoid arthritis SKG animal model and control group of GDF (Growth heart, lung, liver, muscle, joint and spine SUVs ( It is a diagram showing the results of evaluating the degree of inflammation increase according to the uptake level of GDF (Growth differentiation factor) according to the Standardized Uptake Value.
  • FIG. 10 is a diagram confirming lung damage due to pulmonary fibrosis in the rheumatoid arthritis SKG animal model and control group injected with curdlan.
  • 11A, 11B, and 11C are rheumatoid arthritis mice with PHMG lung fibrosis and control, confirming the arthritis index (A), alveolar bronchiolization (B), lung tissue and weight change (C) is a confirmation of
  • 12A and 12B are diagrams confirming the degree of increase in macrophages, neutrophils and lymphocytes, which are immune macrophages, in rheumatoid arthritis mice with PHMG lung fibrosis and in the control group.
  • FIG. 13 is a diagram confirming immune cell regulation according to the levels of Th1, Th2, Treg, and Th17 cells in rheumatoid arthritis mice with PHMG lung fibrosis and a control group.
  • FIG. 14 is a graph showing the pain level (Procimal metatasal) for the animal model of osteoarthritis accompanied by gout (MIA+PO+HX) of the present invention, the time (sec) and the applied force ( g) is a diagram showing the measurement result.
  • 16A, 16B, 16C, and 16D are views illustrating the deepening of osteoporosis according to the comparison of cartilage destruction and cartilage volume in the animal model of gout accompanying osteoarthritis (MIA+PO+HX) of the present invention, Trabecular bone (TB). ) (%), bone mineral density (BMD) (mg/cm3), and structure separation (St.sp) (U) were measured and confirmed.
  • 17 is a diagram showing the results of observing the morphology of salivary gland epithelial cells derived from a patient with Sjogren's syndrome (passage-0).
  • 18 is a diagram showing the results of observing the morphology of organoids using salivary gland epithelial cells (passage-1) derived from a patient with Sjogren's syndrome.
  • FIG. 19 shows the results of treatment with isoproterenol and calcium chloride when culturing organoids using salivary gland epithelial cells derived from a Sjogren's syndrome patient
  • FIG. 20 is a diagram confirming the increase in amylase of the organoid. am.
  • 21 is a result of analysis by confocal microscopy of aquaporin-5, cytokeratin-19, and amylase A expression of organoids using salivary gland epithelial cells derived from a Sjogren's syndrome patient. is a diagram showing
  • 22 is a view showing the result of confirming the degree of damage after treatment with IL-17 in order to prepare an organoid of mouse-derived salivary gland stem cells.
  • FIG. 23 is a diagram showing the results of comparing the formation rate of salivary gland stem cells of the mouse-derived salivary gland stem cells organoids and the production level of alpha amylase, which is the main component of saliva, with the control group.
  • Figure 24a-d is a diagram confirming the expression level of aquaporin-5 (Aquaporin-5), nanog (Nanog), amylase 1 (Amylase 1), keratin 18 (Keratin 18) for organoids of mouse-derived salivary gland stem cells am.
  • the present invention provides a humanized animal model of systemic lupus erythematosus in which immunodeficient mice are administered with PBMCs (peripheral blood mononuclear cells) derived from lupus disease patients.
  • PBMCs peripheral blood mononuclear cells
  • the term "patient” means any single individual in need of treatment, including humans, apes, monkeys, cattle, dogs, guinea pigs, rabbits, mice, rats, chickens, insects, and the like. Also included in the subject are any subjects who participated in a clinical study trial without any clinical manifestations of any disease or subjects who participated in epidemiological studies or subjects used as controls.
  • immunodeficient mouse means a mouse characterized by one or more of the following lists: defects in functional immune cells such as T cells and B cells; DNA repair defects; defects in the rearrangement of genes encoding antigen-specific receptors in lymphocytes; and defects in immune function molecules such as IgM, IgG1, IgG2a, IgG2b, IgG3 and IgA.
  • immunodeficient mice can be characterized by a deficiency in one or more genes involved in immune function, such as Rag1 and Rag2 (Oettinger et al, Science, 248:1517-1523, 1990; and Schatz et al, Cell, 59:1035-1048, 1989), immunodeficient mice may have these or other defects that result in abnormal immune function in the mouse.
  • Particularly useful immunodeficient mouse strains include NOD, Cg-PrkdcscidIl2rgtml Wjl/SzJ, commonly referred to as NOD scid gamma (NSG) mice, as detailed in Shultz et al,, J Immunol, 174: 6477-6489, 2005. and NOD.Cg-Rag1tmlMoml12rgtml Wjl/SzJ, which generally refers to NRG mice (Shultz et al, Clin Exp Immunol, 154(2):270-284, 2008). In an embodiment of the present invention, NSG mice were used.
  • SCID severe combined immune deficiency
  • SCID mutations refer to mutations that result in the deletion of functional B cells and T cells.
  • SCID mice CB-17-Prkdcscid
  • BCR B cell receptor
  • TCR T cell receptor
  • the NSG (NOD scid gamma) mouse is also referred to as a NOD/SCID ⁇ null mouse or a NOD/SCID IL-2R ⁇ KO mouse.
  • NSG mice were developed in the laboratory of Dr Leonard Shultz at The Jackson Laboratory.
  • the NSG mice can be purchased commercially from Jackson Laboratories, or prepared according to a known method (Shultz LD, et al, J Immunol 2005; 169: 204-209).
  • NSG mice can be obtained by backcross mating C57BL/6J- ⁇ null mice and NOD/SCID mice 9 times. It is known that NSG mice lack functional T cells and functional B cells, have reduced macrophage function, abolished NK cells or NK activity, and reduced dendritic cell function.
  • NSG mice are known to have a higher engraftment level of xenografts compared to NOD/SCID mice or ⁇ 2-microglobulin-deficient NOD/LtSz-SCID (NOD/SCID/ ⁇ 2m null).
  • NSG mice Cg-Prkdcscid NSG mice have multiple immunodeficiency based on NOD/ShiLtJ, severe combined immunodeficiency, and interleukin-2 receptor gamma chain. As a result, NSG mice lack cytokine signaling.NSG mice lack IL2R- ⁇ (gamma c) expression, no detectable serum immunoglobulin, no hemolytic complement. , characterized by the absence of mature T lymphocytes and the absence of mature NK cells.
  • patient-specific lupus can be studied in the humanized mouse model.
  • the effectiveness, safety (e.g., any relevant adverse effects on the immune system) and efficacy of these test compounds or drugs can be studied. have.
  • These methods are particularly effective for studies involving the interaction between lupus or immunomodulators, or diseased tissues (eg, cancer, autoimmune diseases) and the immune system.
  • This humanized mouse model can also be used to study any patient-derived xenograft in the presence of an engrafted human hematopoietic system. This includes tumor histology studies, omic studies or profiling (proteomic, genomic, metablomic, etc.).
  • the patient-derived xenograft under study is Information on this can be obtained from the Mouse Tumor Biology Database (MTB), for areas such as selecting experimental models, examining patterns of mutations in specific cancers, and identifying genes in which mutations commonly occur across the cancer spectrum. designed to help researchers in MTB.
  • MTB Mouse Tumor Biology Database
  • the present invention provides a method for preparing a humanized lupus disease animal model, comprising injecting PBMC isolated from a lupus disease patient into an immunodeficient mouse.
  • the present invention provides a method for screening a lupus therapeutic agent, comprising the step of treating a candidate substance in a humanized lupus animal model.
  • the present invention provides an animal model of rheumatoid arthritis accompanied by pulmonary fibrosis, in which pulmonary fibrosis occurs by administering beta-glucan or a fungicide to an animal with rheumatoid arthritis.
  • rheumatoid arthritis-inducing animal may use SKG mice used as an animal model for rheumatoid arthritis because there is a genetic defect in the T-cell immune system and thus T-cell-induced chronic autoimmune disease is induced.
  • CIA collagen-in-duced arthritis
  • SCID humanized severe combined immunodeficiency
  • the types of laboratory mice that can be used for the preparation of the animal model according to the present invention include, but are not limited to, ICR or DDY belonging to a closed colony according to phylogenetic classification; BALB/cA, C57BL/6N, C3H/HeN, DBA/2N or CBA/N belonging to Inbred; BDF1 (C57BL/6 x DBA/2), CDF1 (CBA/N x DBA/2) or B6C3F1 (C57BL6 x C3H/HeN) belonging to the hybrid; Any one used as an experimental mouse, such as BALB/c-nu or C,B-17SCID belonging to the mutant family, can be used.
  • the rheumatoid arthritis animal model has a limitation in that only some lung fibrosis occurs, and the incidence rate cannot be arbitrarily controlled. got to build
  • the beta glucan is curdlan, and the beta glucan may be administered at a concentration of 1 to 500 mg/kg, but is not limited thereto.
  • the disinfectant is polyhexamethylene guanidine (PHMG), hydrochloric acid polyhexamethylene biguanide (PHMB), methyl isothiazolinone (MIT), ethoxyethyl guanidine chloride (Chloride Ethoxyethyl Guanidine, PGH) , naphthalene, phthalate, didecyl methyl ammonium chloride (DDAC), benzisocyazolinone (BIT), chloroxylenol, titanium dioxide, zinc oxide and At least one selected from the group consisting of acetone, preferably polyhexamethylene guanidine (PHMG), which is a component of guanine series, polyhexamethylene biguanide hydrochloride (PHMB), ethoxyethyl chloride Adinine (Chloride Ethoxyethyl Guanidine, PGH), more preferably polyhexamethylene guanidine (Polyhexamethylene guanidine, PHMG), but is not
  • the term "polyhexamethylene guanidine (PHMG)” has been used as a major component of a humidifier disinfectant, and when exposed to the lungs at high concentrations, cold or pneumonia symptoms occur and develop into interstitial pneumonia, causing the lungs to harden. It may also cause shortness of breath. Lung damage cannot be repaired, and if a lung is not transplanted due to the fixed deterioration of lung function, it leads to death.
  • the administration may be selected from the group consisting of oral, intravenous, intramuscular, subcutaneous and intraperitoneal injections, and preferably, when administering the curdlan, it may be administered by oral administration and intraperitoneal injection.
  • Curdlan may be administered in a weight ratio of 1:2, but is not limited thereto.
  • the administration may be administered 1-5 times for 1-5 weeks, preferably 1-3 times for 1-3 weeks, but is not limited thereto.
  • the animal is one or more selected from the group consisting of mice, rats and hamsters, rabbits, horses, cattle, dogs, cats, monkeys and guinea pigs, but preferably mice, but not limited thereto.
  • the lung fibrosis may cause one or more lung diseases selected from the group consisting of pulmonary fibrosis accompanying rheumatoid arthritis, common interstitial pneumonia, non-specific interstitial pneumonia, inflammatory airway disease, and organized pneumonia.
  • the arthritis index was higher than that of the control group, and it was confirmed that the mouse weight decreased.
  • inflammation was increased in the heart, lung, liver, muscle, joint and spine of mice, and damage to the lungs due to pulmonary fibrosis appeared.
  • alveolar bronchiolization occurred significantly compared to the control group, macrophages, neutrophils, and lymphocytes were increased to increase immune response, and inflammatory cells, Th17 It was confirmed that the cells were increased and the Treg cells controlling the immune response were decreased.
  • the present invention also provides a method for preparing an animal model of rheumatoid arthritis accompanied by lung fibrosis, comprising the step of inducing pulmonary fibrosis by administering beta-glucan or a fungicide to a mouse with rheumatoid arthritis.
  • the present invention also provides a method for screening a rheumatoid arthritis therapeutic agent with lung fibrosis, comprising the step of treating the candidate substance to the rheumatoid arthritis animal accompanied by pulmonary fibrosis.
  • the present invention provides an animal model of osteoarthritis accompanied by gout, in which gout is developed by administering potassium oxonate (PO) or hypoxanthine (HX) to an animal with osteoarthritis.
  • PO potassium oxonate
  • HX hypoxanthine
  • the term "potassium oxonate (PO)" is a uric acid lyase inhibitor, which can induce hyperuricemia.
  • hypoxanthine is a naturally occurring purine derivative. Hypoxanthin is often found as a component of nucleic acids and is present in the form of inosine, a nucleoside, in the anticodon of tRNA. Hypoxanthin is an additive necessary for the culture of certain cells, bacteria, and parasites as a substrate and nitrogen source.
  • osteoarthritis-inducing animal refers to an animal that has developed osteoarthritis by an anterior cruciate ligament resection method, a meniscus resection method, a transgenic/knockout method, a chemical injection method, a naturally occurring mouse, or the like.
  • sheep, dogs, rabbits, and rat animals using the anterior cruciate ligament resection method may be included, and may include meniscus-resected sheep, dogs, rabbits, guinea pigs, rats, mice, and the like.
  • it may include ADAM-15 KO, MMP-14 KO, MMP-13 transgenic mice constructed by the transgenic/knockout method, and include mice, rabbits, and rats by collagenase constructed by the chemical injection method. can do.
  • MIA Monitoringosodium iodoacetate
  • injection may include mice, rats, rabbits, and guinea pigs.
  • gout promotes bone destruction in osteoarthritis patients
  • the animal model of osteoarthritis accompanied by gout of the present invention overcomes the limitation that some gout occurs in the conventional animal model of osteoarthritis, but the incidence rate cannot be arbitrarily controlled. , established an animal model of osteoarthritis with clear gout.
  • the administration may be selected from the group consisting of oral, intravenous, intramuscular, subcutaneous and intraperitoneal injection, oral administration may be administered with potassium oxonate, and intraperitoneal administration may be administered with hypoxanthine, but is not limited thereto.
  • the potassium oxonate and hypoxanthine may be administered in a weight ratio of 1:1, may be administered 1-10 times for 1-5 weeks, preferably 1-7 times for 1-3 weeks, but , but not limited thereto.
  • the present invention also provides a method for producing an animal model of osteoarthritis accompanied by gout, comprising the step of inducing gout by administering potassium oxonate or hypoxanthine to an animal with osteoarthritis.
  • the present invention provides a method for screening a treatment material for osteoarthritis accompanied by gout, comprising the step of treating the candidate material to the animal model of osteoarthritis accompanied by gout.
  • the present invention provides an organoid model of Sjogren's syndrome in which Interleukin-17 is administered to an organoid obtained by culturing mouse-derived salivary gland stem cells.
  • organoid means that cells derived from tissues or embryonic stem cells can be cultured in a 3D form to form an artificial organ.
  • Organoid is a suffix that has the same meaning as 'organ' of an organ, and has the word 'organ-like'.
  • Organoids have a more well-arranged cell and cell function through a three-dimensional culture method, and have a functional organ-like form and function. Organoids are attracting attention along with research on optimization of growth and differentiation factors that can differentiate into various tissues, with stem cell research and 3D cell culture being developed.
  • saliva is a mixed solution secreted from the parotid, submandibular, sublingual, and mucous glands present in the oral mucosa.
  • Saliva is a key component of the human body, produced in the salivary glands and discharged into the oral cavity.
  • Saliva is an essential component of the human body and contains bioactive proteins, digestive enzymes, mucus, immunoglobulins, and various salts. Saliva plays a very important role in maintaining the homeostasis of the human body as well as oral health.
  • saliva the main components of saliva, mucin, immunoglobulin, etc., play a primary defense role against external infection, and protect the oral mucosa and teeth through lubrication and moisture maintenance of the oral cavity and teeth, and neutralizing pH.
  • saliva contains digestive enzymes such as amylase, such as ptyalin, which promotes digestion by decomposing starch to maltose units.
  • amylase such as ptyalin
  • ptyalin ptyalin
  • body's water metabolism and body temperature control can be achieved by the secretion of saliva, and toxic substances (I, Hg, Pb, etc.) are excreted.
  • saliva gland refers to an organ that produces and secretes saliva, such as parotid gland, submandibular gland, and sublingual gland. It is a minor salivary gland distributed in various parts of the oral mucosa, such as major salivary glands and mucous glands existing in the mucous membrane of oral cavity, such as mucous glands. classified.
  • the derived mouse is not limited, and general laboratory mice, immunodeficient mice, and the like may be used.
  • the present invention provides an organoid model of Sjogren's syndrome, including Patient-derived salivary gland epithelial cells.
  • the present invention is a mouse culturing and obtaining derived salivary gland stem cells; And it provides a method for producing an organoid model of Sjogren's syndrome, comprising the step of culturing the cells to generate an organoid and then administering Interleukin-17.
  • the present invention also provides a method for producing a Sjogren's syndrome organoid model, comprising culturing the salivary gland epithelial cells derived from a Sjogren patient.
  • the present invention provides a method for screening a Sjogren's syndrome therapeutic material, including; treating a candidate material in the Sjogren's syndrome organoid model.
  • a humanized mouse model of lupus equipped with a human immune system
  • PBMCs peripheral blood mononuclear cells
  • mice were sacrificed 4 weeks after cell transplantation to confirm the infiltration of human cells and histological changes in the tissue ( FIG. 1 ).
  • Example 1-1 In order to confirm that the lupus patient mimic avatar model was properly constructed in Example 1-1, engraftment of human cells was confirmed by analyzing T cells and B cells in the blood. Specifically, blood was obtained from the lupus humanized mice of Example 1-1 (normal PBMC-injected group and lupus patient PBMC-injected group), and cells that reacted with human CD4, CD8 and CD19 antibodies were analyzed through flow cytometry. .
  • CD4+T, CD8+T, and Cd19+T cells were detected, confirming that human cells were well engrafted ( FIG. 2 ).
  • Example 1-1 In order to confirm the engraftment of human cells in the tissue in the humanized lupus mouse model established in Example 1-1, the humanized lupus mice were sacrificed 4 weeks after Example 1 and the kidneys were removed, and human immune cells in the tissue Subtypes (CD3, CD4, CD8, CD19) were stained with IHC to infiltrate. The excised tissue was fixed with formalin and then embedded in paraffin to create a 5 ⁇ m thick section. In order to observe the immune cells in the tissue, immunohistochemical analysis was performed by reacting with human CD3, CD4, CD8, and CD19 antibodies on the section slides.
  • tissue Subtypes CD3, CD4, CD8, CD19
  • dsDNA human anti-double stranded DNA
  • IgG human anti-double stranded DNA
  • the kidney tissue of the humanized lupus mouse of Example 1-1 was stained with H&E (haematoxylin and eosin). Specifically, the excised tissue was fixed with formalin and then embedded in paraffin to generate a 5 ⁇ m thick section, and the section slide was deparaffinized with xylene for H&E staining. Thereafter, the cell nucleus was stained with hematoxylin, the cytoplasm with eosin, and periodic acid-Schiff (PAS) was stained to compare and analyze the pathology of the kidney tissue.
  • H&E haematoxylin and eosin
  • SKG mice were used. Specifically, it is known that the SKG mouse has a genetic defect in the T-cell immune system and causes T-cell-induced chronic autoimmune disease.
  • the following control and experimental groups were constructed in SKG male mice (within 8 weeks of age). The control group was treated with PBS, and the experimental group was injected with curdlan at 6 mg/kg intraperitoneal injection and 3 mg/kg oral injection once a week before the start of the experiment. built.
  • Score 2 Mild swelling and redness from the ankle joint to the metatarsal.
  • Score 3 Moderate swelling and redness from the ankle joint to the tarsal bone.
  • the best arthritic index per mouse is 4 points, so the best disease index per mouse is 16.
  • mice weight changes were compared to the SKG animal model of rheumatoid arthritis with lung fibrosis constructed in Example 2-1 (24 weeks old, 17 weeks after curdlan injection) and control SKG mice (24 weeks old).
  • Collagen-induced rheumatoid arthritis animal model (CIA) was injected with polyhexamethylene guandinine (PHMG). Specifically, in order to increase the sensitivity, after immunization at week 0, 1% of 100ul polyhexamethylene guanine (PHMG) was intratracheal instillation at week 6, and administered only once at week 6 Then, the mice were sacrificed and the analysis was performed.
  • PHMG polyhexamethylene guandinine
  • Example 2-4 Effect of an animal model of rheumatoid arthritis with lung fibrosis through polyhexamethylene guandinine (PHMG) injection
  • Arthritis index for the PHMG-injected rheumatoid arthritis animal model with lung fibrosis and control mice constructed in Example 2-3 was confirmed. Specifically, the arthritis index was measured by the method described in 2-2-1 above, and the arthritis index from week 0 to week 7 when rheumatoid arthritis occurred was confirmed.
  • Example 2-3 The effect of PHMG injection constructed in Example 2-3 on the lung tissue in the animal model of rheumatoid arthritis accompanied by lung fibrosis and the control mouse was confirmed.
  • alveolar bronchiolization through alveolar bronchiolization, it is a dysplastic lesion characterized by cells similar to the bronchial epithelium covering the normal or thickened alveolar wall, in pathological conditions exposed to inflammation, chemical stimuli, etc. It is characterized by occurrence.
  • Th1 cells type 1 helper cells
  • Th2 cells type 2 helper cells
  • Treg cells have the property of controlling the inflammatory response by inhibiting the function of abnormally activated cotton cells, and unlike Treg cells, Th17 cells are involved in the forefront of the inflammatory response seen in immune diseases and maximize the signal of the inflammatory response to disease. accelerate the progress of
  • Th17 cells were IL-17+in CD4+
  • Treg cells were CD25+Foxp+inCD4+
  • Th1 cells were IFN- ⁇ +in CD4+ cells were identified
  • Th2 cells were identified as IL-4+in CD4+ cells.
  • the arthritis index was higher than that of the control group, and it was confirmed that the mouse weight decreased.
  • inflammation was increased in the heart, lung, liver, muscle, joint and spine of mice, and damage to the lungs due to pulmonary fibrosis appeared.
  • alveolar bronchiolization occurred significantly compared to the control group, macrophages, neutrophils, and lymphocytes were increased to increase immune response, and inflammatory cells, Th17 It was confirmed that the cells were increased and the Treg cells controlling the immune response were decreased.
  • MIA Monosodium Iodoacetate
  • saline for injection at a concentration of 60 mg/ml, and prepared on the day of the start of the experiment (day 0).
  • 50ul MIA 3mg/body
  • MIA was injected into the right knee joint with a 26.5 gauge needle through the infrapatellar ligament. was injected to induce osteoarthritis (MIA).
  • Example 3-1 In order to measure the pain level of the animal model of osteoarthritis accompanied by gout constructed in Example 3-1, a Dynamic planter esthesiometer (UgoBasile, Comerio, Itaily) of the animal model of the control group and the present invention was used. Place the stimulator on the underside of the rat, adjust a 0.5mm thick plastic stimulation needle (Stimulating microfilament) to be positioned on the hind leg, and operate the machine. The time (sec) and applied force (g) until the rat could not withstand the stimulation and released the foot were measured by increasing it. Each measurement was performed three times and averaged.
  • UgoBasile, Comerio, Itaily Place the stimulator on the underside of the rat, adjust a 0.5mm thick plastic stimulation needle (Stimulating microfilament) to be positioned on the hind leg, and operate the machine. The time (sec) and applied force (g) until the rat could not withstand the stimulation and released the foot were measured by increasing it
  • the time (sec) and force (g) until taking off the foot without enduring the stimulus showed a significant difference in pain level (Procimal metatasal) in the animal model of osteoarthritis accompanied by gout. It was confirmed that the animal model of osteoarthritis accompanied by gout of the present invention was constructed to accompany gout while maintaining the pain characteristic of the animal model of osteoarthritis (MIA) (FIG. 14).
  • Example 3-1 In order to measure the pain level for the osteoarthritis animal model with gout constructed in Example 3-1, using an Incapacitance Meter (IITC, Victory Boulevard Woodland Hills, CA, USA), the weight on the right hind limb (Weight on right hind rimb) ) (%) was measured.
  • IITC Incapacitance Meter
  • the femur (Femur) and tibia (Tibia) of the control group (WT), osteoarthritis animal model (MIA), and gout-accompanied osteoarthritis animal model (MIA+PO+HX) were collected and stained with India ink and then microcomputerized.
  • the degree of destruction of cartilage and cartilage volume were analyzed by tomography (micro-CT image).
  • Trabecular bone (TB) (%) decreases as osteoporosis intensifies
  • bone mineral density (BMD) mg/cm 3 ) is an indicator that decreases as osteoporosis intensifies.
  • the structure separation (St.sp) (U) is the gap between the cancellous bones, and when osteoporosis occurs, the decrease in the cancellous bone (TB) occurs.
  • the degree of osteoporosis was measured by increasing structural separation (St.sp).
  • the animal model of osteoarthritis accompanied by gout (MIA+PO+HX) of the present invention confirmed that osteoporosis was aggravated as it was accompanied by gout, compared with the animal model of osteoarthritis (MIA).
  • Sjogren's patient-derived salivary gland epithelial cells were diluted 20-fold in PureCol (Advanced biomatrix #5005) distilled water, put in a Petri dish, and left at 37°C for 1 hour or more, then removed and washed with PBS or medium before use.
  • 1U/mL dispase II solution (Stemcell#07923) and 2mg/mL collagenase IV (Gibco#17104-019) were prepared. Thereafter, 3 mL of the washed thing was put in a 60 mm Petri dish, and the tissue was placed on it and cut as finely as possible. Incubated at 37°C, taken out every 15 minutes and resuspended.
  • tissue When the tissue became almost limp, it was transferred to a tube, centrifuged at 1500 rpm, 5 minutes, and 4°C, resuspended in medium, and incubated at 37°C.
  • the salivary gland epithelial cells derived from the Sjogren patient were resuspended in 1 mL of 90% fetal bovine serum and 10% dimethyl sulfoxide solution and freeze-dried.
  • subculture was performed to maintain Sjogren's patient-derived salivary gland epithelial cells, and the cells were injected at a concentration of 1 x 10 5 cells/24 well plate and 5 x 10 5 cells/6 well plate.
  • PureCol Advanced biomatrix #5005
  • PureCol was diluted 20 times in distilled water, put in a Petri dish, left at 37°C for 1 hour or more, then removed and washed with PBS or medium before use. After removing the media and washing with PBS, 2mL of 0.25% trypsin-EDTA was placed in a Petri dish and incubated at 37°C for 10 minutes.
  • the resuspended cells were transferred to a 50 mL tube and centrifuged at 1000 rpm at 4°C for 5 minutes. The supernatant was discarded, resuspended in culture medium, and cultured in a Petri dish.
  • Organoid culture medium includes Keratinocyte-Serum Free Media with EGF, bovine pituitary extract (Gibco#37010-022), 0.09mM Calcium chloride (Amresco) #E506-1) was used.
  • the organoid culture method is to dilute Matrigel matrix (BD#354234) with refrigerated K-SFM medium at a ratio of 2:1 and place it on a 48 well-plate placed on ice, which is then placed in a 37 degree cell incubator. Transfer and coating for 1 hour.
  • the salivary gland epithelial cells derived from Sjogren's patient cultured in 4-1-1 were resuspended at 37°C, and 5 x 10 4 cells/500 ⁇ L/well were dispensed to the coated ones. Organoids were observed while changing the medium every 3 days.
  • organoids using salivary gland epithelial cells derived from patients with Sjogren's syndrome were constructed in the form of spheroids after 1 to 2 days of culture, and duct organoids and acinar organoids after 7 to 8 days. (Fig. 18).
  • Example 4-2 Analysis of amylase secretion ability of organoids derived from salivary gland epithelial cells derived from Sjogren patients
  • the patient-derived salivary gland organoids were cultured in a medium supplemented with 0.09 mM calcium chloride to obtain spiroids, and then with 10 mM isoproterenol and 2 mM calcium chloride. Stimulated for 18 hours. Thereafter, the supernatant was removed after microscopy, and amylase activity was measured using the cells (amylase activity assay kit, abcam#102523).
  • Example 4-3 Confocal microscopic analysis of aquaporin-5, cytokeratin-19, and amylase A expression of organoids derived from salivary gland epithelial cells derived from Sjogren's patient
  • rabbit anti-human aquaporin-5 antibody (Abcam#ab92320, diluted 1:100) and mouse anti-human amylase A antibody (Abcam#ab201450, diluted 1:100) were used.
  • Secondary antibodies were Alexa488 donated goat anti-rabbit IgG (H+L) (Invitrogen# A32731, 1:500 dilution), Alexa594 donated goat anti-rabbit-mouse IgG (H+L) (Invitrogen#A32742, 1: 500 dilution) was used.
  • DAPI 4,6-Diamidino-2-Phenylindole, Dilactate, Invitrogen#D3571
  • DAPI DAPI-4',6-Diamidino-2-Phenylindole, Dilactate, Invitrogen#D3571
  • images were taken with a confocal microscope using Zeiss LSM700 equipment, Aquaporin-5 (Aquaporin-5), Cytokeratin Cell nuclei of -19 (Cytokeratin-19) and amylase A were observed.
  • the organoid was treated with IL-17, and the salivary gland stem cells were damaged by treatment with IL-17 at a concentration of 10, 20, or 50 ng/ml. After that, the formation rate of salivary gland stem cells and the production level of alpha amylase, which is the main component of saliva, were confirmed.
  • Example 4-4 In order to confirm that the organoid of the mouse-derived salivary gland stem cells prepared in Example 4-4 was properly constructed as a model for Sjogren's syndrome, aquaporin-5, keratin 18 (genetic markers of salivary gland stem cells) ( Keratin 18), the expression of nanog (Nanog) was confirmed. In addition, the expression of amylase 1 involved in salivary secretion was confirmed. It confirmed the expression of each mRNA through quantitative PCR.

Abstract

The present invention relates to an avatar model platform for evaluating autoimmune diseases. An autoimmune disease model according to the present invention reflects the immune status of patients with autoimmune diseases well, and thus not only can be used as a preclinical model for monitoring the immune status of patients, but can also be used to screen for therapeutic substances for autoimmune diseases. Thus, the model can overcome limitations such as the absence of therapeutic drugs and the lack of clinical success of monitoring drugs with the current level of animal testing, which have been problems in the past, and thus can be used not only to evaluate drugs for autoimmune diseases but also to understand pathological mechanisms and develop tailored disease diagnosis and control systems. Therefore, the model can be effectively used in autoimmune disease-related fields or industries.

Description

자가면역질환 평가 아바타 모델 플랫폼Autoimmune disease evaluation avatar model platform
본 발명은 자기면역질환 평가 아바타 모델 플랫폼에 관한 것이다.The present invention relates to an autoimmune disease evaluation avatar model platform.
자가면역질환은 인체의 면역계가 이상을 일으켜, 자기세포가 자기세포를 공격하는 것이다. 인간의 면역계는 기본적으로 인체에 침입한 미생물 및 암세포 발생 등에 대하여 외부 항원으로 인식하고, 이를 공격하여 제거하는 강력한 힘을 지녔지만 자기 관용성이 있어서 자기세포에 대해서는 공격하지 않는다. 이를 인체의 자기 관용(self-tolerance) 현상이라 한다. 그러나 면역계의 자기 관용이 파괴될 경우, 인체는 자기세포(또는 자가항원)에 반응하는 자가반응 T 세포가 활성화되고 자가항체(autoantibody)가 생성되면서 끊임없이 자기세포를 파괴하며 염증 및 면역반응을 일으키게 된다.Autoimmune disease is when the body's immune system malfunctions and its own cells attack their own cells. The human immune system basically recognizes foreign antigens against invading microorganisms and cancer cells, and has strong power to attack and remove them, but it does not attack its own cells because of its self-tolerance. This phenomenon is called self-tolerance of the human body. However, when the self-tolerance of the immune system is disrupted, autoreactive T cells that respond to self-cells (or autoantigens) are activated and autoantibodies are generated, constantly destroying self-cells, causing inflammation and immune responses. .
면역계에서 항원에 특이적으로 반응하는 세포에는 T 세포와 B 세포가 있다. T 세포는 항원제시세포(antigenpresenting cell)에 의해 제시되는 특정 항원을 만났을 때 그 항원에 따라 반응을 나타내게 되는데, 항원제시세포에서 제시되는 항원이 'non-self'로 인식이 되면 이를 제거하려는 면역반응을 나타내고, 'self'로 인식되면 면역반응이 무시되는 관용성을 보인다. T 세포가 항원에 대하여 활성화되면, 대부분 B 세포가 잇달아 활성화되며 B 세포는 형질세포(plasma cell)로 변하여 인식한 특정항원에 대하여 특이적으로 반응하는 항체를 생성하게된다. 따라서 인체에 관용성이 깨지면서 자가면역이 발생될 경우 T 세포가 자가항원을 비정상적으로 인식하여 활성화되고, B 세포도 활성화되면서 자가항원에 반응하는 자가항체를 생성하게 되며, 우리 몸에서는 자기세포를 공격하는 면역반응이 일어나게 된다.Cells that specifically respond to antigens in the immune system include T cells and B cells. When T cells meet a specific antigen presented by an antigen presenting cell, they respond according to the antigen. When the antigen presented by the antigen presenting cell is recognized as 'non-self', an immune response to remove it , and if recognized as 'self', the immune response is ignored and tolerated. When T cells are activated against an antigen, most B cells are activated one after another, and the B cells turn into plasma cells to produce antibodies that specifically react to the recognized antigen. Therefore, when autoimmunity occurs due to the breakdown of tolerance in the body, T cells are activated by abnormal recognition of autoantigens, and B cells are also activated to produce autoantibodies that respond to autoantigens. an immune response occurs.
한편, 통상적으로, 맞춤의학(tailored medicine)이란 맞춤의료(order-made medicine) 또는 환자 맞춤형 의학(personalized medicine)이라고도 하며, 환자 개인의 체질이나 환경을 개별적으로 조사하여, 여기에 적합한 치료법을 결정하여 치료하는 방법을 의미한다. 유전정보를 기반으로 한 맞춤의학을 구현하기 위하여는, 유전체 진단용 바이오마커, 다양한 유전체 검사 방법, 상기 바이오마커를 이용하여 도출되거나 또는 상기 검사방법으로 얻어진 유전체 정보를 분석/통합할 수 있는 의료정보학적 분석법, 상기 분석법에 의해 분석된 결과를 적용할 수 있는 표적화 치료기술 등을 상호보완적으로 선행개발하여야 한다. 즉, 환자에게 적합한 치료법을 선별할 수 있는 기술 개발이 필요할 뿐만 아니라, 상기 선별된 치료법을 검증할 수 있는 기술이 개발되어야만 한다.On the other hand, in general, tailored medicine is also called order-made medicine or personalized medicine. means how to treat it. In order to implement customized medicine based on genetic information, biomarkers for genomic diagnosis, various genomic test methods, and medical informatics that can analyze/integrate genomic information derived using the biomarkers or obtained by the test method Analysis methods and targeted treatment technologies to which the results analyzed by the above analysis methods can be applied should be developed in advance and complementary to each other. That is, it is necessary not only to develop a technology for selecting a treatment suitable for a patient, but also to develop a technology for verifying the selected treatment.
맞춤의학을 실현하기 위한 방법으로 환자에게 적합한 약물을 스크리닝하거나 치료방법을 선별하는 방법을 개발할 필요가 있는데, 특히 상기와 같은 쇼그렌 증후군 등의 면역질환에 있어서는 개인마다 각 약물에 대해 나타내는 면역반응이 상이하여, 개인에 생체 특성을 재현하는 in vivo 환경을 조성할 수 있는 전임상 연구를 위한 동물모델의 개발이 필요하다.As a method for realizing personalized medicine, it is necessary to develop a method for screening a drug suitable for a patient or a method for selecting a treatment method. Therefore, it is necessary to develop an animal model for preclinical research that can create an in vivo environment that reproduces biological characteristics in individuals.
한편, 오가노이드란 줄기세포를 3차원적으로 배양하거나 응집 또는 재조합하여 만든 장기 특이적 세포 집합체로 자기 재생(self-renewal)이 가능하며 '미니 장기' 또는 '유사 장기'라고도 불린다. 오가노이드는 실제 장기와 유사한 형태로 분자 신호 조절 등과 같은 동물 모델에서는 구현하기 힘든 연구를 in vitro 상에서 구현할 수 있어 기초 연구에 매우 유용함은 물론, 인간의 발생과정, 질환 모델 확립, 의약품 유효성 평가 스크리닝, 의약품 독성 평가 플랫폼 개발, 세포치료제 개발 등 다양한 분야에 매우 유용하게 사용될 수 있는 기술이다.On the other hand, an organoid is an organ-specific cell aggregate made by three-dimensionally culturing, aggregating, or recombination of stem cells, capable of self-renewal, and is also called a 'mini organ' or 'similar organ'. Organoids are very useful for basic research as they can implement in vitro studies that are difficult to implement in animal models such as molecular signal regulation in a form similar to that of real organs, as well as human development, disease model establishment, drug efficacy evaluation screening, It is a technology that can be very usefully used in various fields such as drug toxicity evaluation platform development and cell therapy drug development.
본 발명의 목적은 면역결핍 마우스에 루푸스 질환 환자 유래 PBMC(Peripheral blood mononuclear cell)이 투여된, 인간화된 루푸스(systemic lupus erythematosus) 질환 동물 모델을 개발하는 것이다.It is an object of the present invention to develop a humanized animal model of systemic lupus erythematosus in which immunodeficient mice are administered with PBMCs (peripheral blood mononuclear cells) derived from lupus disease patients.
본 발명의 목적은 면역결핍 마우스에 루푸스 질환 환자로부터 분리한 PBMC를 주입하는 단계를 포함하는, 인간화된 루푸스 질환 동물 모델을 제작하는 방법을 개발하는 것이다.An object of the present invention is to develop a method for producing a humanized lupus disease animal model, comprising injecting PBMC isolated from a lupus disease patient into immunodeficient mice.
본 발명의 목적은 류마티스 관절염 발생 동물에 베타 글루칸 또는 살균제가 투여되어 폐 섬유화가 발생된, 폐 섬유화가 동반된 류마티스 관절염 동물 모델을 개발하는 것이다.It is an object of the present invention to develop an animal model of rheumatoid arthritis accompanied by pulmonary fibrosis, in which pulmonary fibrosis occurred by administering beta-glucan or a fungicide to an animal with rheumatoid arthritis.
본 발명의 목적은 류마티스 관절염 발생 마우스에 베타 글루칸 또는 살균제를 투여하여 폐 섬유화를 유도하는 단계;를 포함하는, 폐 섬유화가 동반된 류마티스 관절염 동물 모델의 제조 방법을 개발하는 것이다.An object of the present invention is to develop a method for producing an animal model of rheumatoid arthritis accompanied by lung fibrosis, comprising the step of inducing lung fibrosis by administering beta-glucan or a fungicide to a mouse with rheumatoid arthritis.
본 발명의 목적은 골관절염 발생 동물에 칼륨 옥소네이트(potassium oxonate, PO) 또는 히포잔틴(Hypoxanthine, HX)가 투여되어 통풍이 발생된, 통풍이 동반된 골관절염 동물 모델을 개발하는 것이다.An object of the present invention is to develop an animal model of osteoarthritis accompanied by gout, in which gout is generated by administering potassium oxonate (PO) or hypoxanthine (HX) to an animal with osteoarthritis.
본 발명의 목적은 골관절염 발생 동물에 칼륨 옥소네이트 또는 히포잔틴을 투여하여 통풍을 유도하는 단계;를 포함하는, 통풍이 동반된 골관절염 동물 모델의 제조 방법을 개발하는 것이다.An object of the present invention is to develop a method for producing an animal model of osteoarthritis accompanied by gout, including the step of inducing gout by administering potassium oxonate or hypoxanthine to an animal with osteoarthritis.
본 발명의 목적은 마우스 유래 침샘 줄기세포를 배양하여 수득한 오가노이드에 인터루킨-17(Interleukin-17)이 투여된, 쇼그렌 증후군(Sjogren's syndrome) 오가노이드(organoid) 모델을 개발하는 것이다.An object of the present invention is a mouse To develop an organoid model of Sjogren's syndrome in which Interleukin-17 is administered to an organoid obtained by culturing the derived salivary gland stem cells.
본 발명의 목적은 쇼그렌 환자 유래 타액샘 상피세포(Patient-derived salivary gland epithelial cell)를 포함하는, 쇼그렌증후군(Sjogren's syndrome) 오가노이드(organoid) 모델을 개발하는 것이다.It is an object of the present invention to develop a Sjogren's syndrome organoid model, including Patient-derived salivary gland epithelial cells.
본 발명의 목적은 마우스 유래 침샘 줄기세포를 배양하여 수득하는 단계; 및An object of the present invention is a mouse culturing and obtaining derived salivary gland stem cells; and
상기 세포를 배양하여 오가노이드를 생성 후 인터루킨-17(Interleukin-17)이 투여하는 단계를 포함하는, 쇼그렌증후군(Sjogren's syndrome) 오가노이드(organoid) 모델 제작 방법을 개발하는 것이다.To develop an organoid model production method for Sjogren's syndrome, which includes the step of culturing the cells to generate an organoid and then administering Interleukin-17.
본 발명의 목적은 쇼그렌 환자 유래 타액샘 상피세포를 배양하는 단계를 포함하는, 쇼그렌증후군(Sjogren's syndrome) 오가노이드(organoid) 모델 제작 방법을 개발하는 것이다.It is an object of the present invention to develop a method for manufacturing a Sjogren's syndrome organoid model, which includes culturing Sjogren's patient-derived salivary gland epithelial cells.
상기 과제를 해결하기 위하여, 본 발명은 면역결핍 마우스에 루푸스 질환 환자 유래 PBMC(Peripheral blood mononuclear cell)이 투여된, 인간화된 루푸스(systemic lupus erythematosus) 질환 동물 모델을 제공한다.In order to solve the above problems, the present invention provides a humanized lupus (systemic lupus erythematosus) disease animal model in which immunodeficient mice are administered with PBMCs (peripheral blood mononuclear cells) derived from lupus disease patients.
상기 과제를 해결하기 위하여, 본 발명은 면역결핍 마우스에 루푸스 질환 환자로부터 분리한 PBMC를 주입하는 단계를 포함하는, 인간화된 루푸스 질환 동물 모델을 제작하는 방법을 제공한다.In order to solve the above problems, the present invention provides a method for manufacturing a humanized lupus disease animal model, comprising injecting PBMC isolated from a lupus disease patient into an immunodeficient mouse.
상기 과제를 해결하기 위하여, 본 발명은 류마티스 관절염 발생 동물에 베타 글루칸 또는 살균제가 투여되어 폐 섬유화가 발생된, 폐 섬유화가 동반된 류마티스 관절염 동물 모델을 제공 한다.In order to solve the above problems, the present invention provides an animal model of rheumatoid arthritis accompanied by pulmonary fibrosis, in which pulmonary fibrosis is generated by administering beta-glucan or a fungicide to an animal with rheumatoid arthritis.
상기 과제를 해결하기 위하여, 본 발명은 류마티스 관절염 발생 마우스에 베타 글루칸 또는 살균제를 투여하여 폐 섬유화를 유도하는 단계;를 포함하는, 폐 섬유화가 동반된 류마티스 관절염 동물 모델의 제조 방법을 제공한다.In order to solve the above problems, the present invention provides a method for preparing an animal model of rheumatoid arthritis accompanied by lung fibrosis, comprising the step of inducing lung fibrosis by administering beta-glucan or a fungicide to a mouse with rheumatoid arthritis.
상기 과제를 해결하기 위하여, 본 발명은 골관절염 발생 동물에 칼륨 옥소네이트(potassium oxonate, PO) 또는 히포잔틴(Hypoxanthine, HX)가 투여되어 통풍이 발생된, 통풍이 동반된 골관절염 동물 모델을 제공한다.In order to solve the above problems, the present invention provides an animal model of osteoarthritis accompanied by gout, in which gout is generated by administering potassium oxonate (PO) or hypoxanthine (HX) to an animal with osteoarthritis.
상기 과제를 해결하기 위하여, 본 발명은 골관절염 발생 동물에 칼륨 옥소네이트 또는 히포잔틴을 투여하여 통풍을 유도하는 단계;를 포함하는, 통풍이 동반된 골관절염 동물 모델의 제조 방법을 제공한다.In order to solve the above problems, the present invention provides a method for producing an animal model of osteoarthritis accompanied by gout, comprising the step of inducing gout by administering potassium oxonate or hypoxanthine to an animal with osteoarthritis.
상기 과제를 해결하기 위하여, 본 발명은 마우스 유래 침샘 줄기세포를 배양하여 수득한 오가노이드에 인터루킨-17(Interleukin-17)이 투여된, 쇼그렌 증후군(Sjogren's syndrome) 오가노이드(organoid) 모델을 제공한다.In order to solve the above problems, the present invention is a mouse Interleukin-17 (Interleukin-17) is administered to an organoid obtained by culturing the derived salivary gland stem cells, Sjogren's syndrome provides an organoid model.
상기 과제를 해결하기 위하여, 본 발명은 쇼그렌 환자 유래 타액샘 상피세포(Patient-derived salivary gland epithelial cell)를 포함하는, 쇼그렌증후군(Sjogren's syndrome) 오가노이드(organoid) 모델을 제공한다.In order to solve the above problems, the present invention provides a Sjogren's syndrome organoid model, including a Sjogren patient-derived salivary gland epithelial cell (Patient-derived salivary gland epithelial cell).
상기 과제를 해결하기 위하여, 본 발명은 마우스 유래 침샘 줄기세포를 배양하여 수득하는 단계; 및 상기 세포를 배양하여 오가노이드를 생성 후 인터루킨-17(Interleukin-17)이 투여하는 단계를 포함하는, 쇼그렌증후군(Sjogren's syndrome) 오가노이드(organoid) 모델 제작 방법을 제공한다.In order to solve the above problems, the present invention is a mouse culturing and obtaining derived salivary gland stem cells; And it provides a method for producing an organoid model of Sjogren's syndrome, comprising the step of culturing the cells to generate an organoid and then administering Interleukin-17.
상기 과제를 해결하기 위하여, 본 발명은 쇼그렌 환자 유래 타액샘 상피세포를 배양하는 단계를 포함하는, 쇼그렌증후군(Sjogren's syndrome) 오가노이드(organoid) 모델 제작 방법을 제공한다.In order to solve the above problems, the present invention provides a method for manufacturing a Sjogren's syndrome organoid model, comprising the step of culturing Sjogren patient-derived salivary gland epithelial cells.
본 발명에 따른 자가면역질환 모델은, 자가면역질환 환자의 면역 상태를 잘 반영하고 있으므로, 이를 환자의 면역 상태를 모니터링 하기 위한 전임상 모델로 사용할 수 있을 뿐만 아니라, 자가면역질환 치료 물질의 스크리닝에 이용할 수 있다. 이에 종래 문제점이었던 치료 약제가 부재하며 현재 동물실험 수준에서 모니터링 약물은 임상에서 성공이 안되는 한계점을 극복할 수 있는바, 자가면역질환의 약물 평가 뿐만 아니라 병리 기전을 이해하고 맞춤형 질병 진단 및 조절 시스템 개발에 활용될 수 있어, 자가질환 면역 관련 분야 또는 산업에 유용하게 이용될 수 있다.Since the autoimmune disease model according to the present invention reflects the immune status of the autoimmune disease patient well, it can be used not only as a preclinical model for monitoring the patient's immune status, but also can be used for screening of therapeutic substances for autoimmune diseases. can Therefore, there is no treatment drug, which was a problem in the past, and monitoring drugs at the current animal test level can overcome the limitations of not being successful in clinical practice. It can be used in the field or industry related to autoimmune disease.
도 1은 본 발명의 루푸스 환자 모사 아바타 모델 구축 프로토콜을 모식화한 도이다. 1 is a diagram schematically illustrating the lupus patient imitation avatar model construction protocol of the present invention.
도 2는 루푸스 환자 모사 아바타 모델에서 인간 세포 생착을 확인하기 위하여, 인간 CD4, CD8 및 CD19 항체와 반응시킨 결과를 나타낸 도이다.2 is a diagram showing the results of reacting with human CD4, CD8 and CD19 antibodies to confirm human cell engraftment in a lupus patient mimic avatar model.
도 3은 루푸스 환자 모사 아바타 모델에서 인간 세포 생착을 확인하기 위하여, 조직 내 인간의 면역세포아형(CD3, CD4, CD8, CD19)의 침윤 정도를 확인한 도이다.3 is a diagram illustrating the degree of infiltration of human immune cell subtypes (CD3, CD4, CD8, CD19) in tissues in order to confirm human cell engraftment in a lupus patient mimic avatar model.
도 4a, 도4b 및 도4c는 루푸스 환자 모사 아바타 모델에서 자가항체 및 단백뇨 변화를 확인하기 위하여, dsDNA level 측정방법, 오줌 내 인간 알부민의 농도 측정 방법, 크레아틴 농도를 측정한 결과를 나타낸 도이다. 4A, 4B and 4C are diagrams showing the results of measuring the dsDNA level measurement method, the urine human albumin concentration method, and the creatine concentration in order to confirm the autoantibody and proteinuria changes in the simulated avatar model of a lupus patient.
도 5는 루푸스 환자 모사 아바타 모델에서 신장의 조직학적 변화를 확인하기 위하여, 염증세포의 침윤을 확인한 도이다.5 is a view confirming the infiltration of inflammatory cells in order to confirm the histological change of the kidney in the lupus patient simulated avatar model.
도 6은 커들란 주입된 폐 섬유화 동반 류마티스 관절염 SKG 동물 모델 및 대조군에 대한 관절염 지수(arthritis score)를 비교한 도이다.6 is a diagram comparing the arthritis scores for the curdlan-injected rheumatoid arthritis SKG animal model with lung fibrosis and the control group.
도 7는 커들란 주입된 폐 섬유화 동반 류마티스 관절염 SKG 동물 모델 및 대조군에 대한 체중 변화를 비교한 도이다.7 is a diagram comparing the weight change of the curdlan-injected rheumatoid arthritis SKG animal model with lung fibrosis and the control group.
도 8은 커들란 주입된 폐 섬유화 동반 류마티스 관절염 SKG 동물 모델 및 대조군에 대한 각 장기와 조직의 생화학적 변화를 Pet-CT 촬영하여 확인한 도이다.8 is a view confirming the biochemical changes of each organ and tissue in the curdlan-injected rheumatoid arthritis SKG animal model and control group injected with pulmonary fibrosis by Pet-CT.
도 9a, 도 9b, 도 9c, 도 9d, 도 9e 및 도 9f는 커들란 주입된 폐 섬유화 동반 류마티스 관절염 SKG 동물 모델 및 대조군의 GDF(Growth 심장, 폐, 간, 근육, 관절 및 척추의 SUV(Standardized Uptake Value, 표준섭취계수) 값에 따른 GDF(Growth differentiation factor) 흡수(uptake) 정도에 따른 염증 증가 정도를 평가한 결과를 나타낸 도이다. 9A, 9B, 9C, 9D, 9E and 9F show the curdlan-injected rheumatoid arthritis SKG animal model and control group of GDF (Growth heart, lung, liver, muscle, joint and spine SUVs ( It is a diagram showing the results of evaluating the degree of inflammation increase according to the uptake level of GDF (Growth differentiation factor) according to the Standardized Uptake Value.
도 10는 커들란 주입된 폐 섬유화 동반 류마티스 관절염 SKG 동물 모델 및 대조군의 폐 섬유화에 의한 폐의 손상을 확인한 도이다.10 is a diagram confirming lung damage due to pulmonary fibrosis in the rheumatoid arthritis SKG animal model and control group injected with curdlan.
도 11a, 도 11b 및 도 11c는 PHMG 폐 섬유화 동반 류마티스 관절염 마우스와 대조군에서, 관절염 지수를 확인하고(A), 폐포의 세 기관지화(alveolar bronchiolization)(B), 폐 조직 및 무게 변화(C)를 확인한 도이다.11A, 11B, and 11C are rheumatoid arthritis mice with PHMG lung fibrosis and control, confirming the arthritis index (A), alveolar bronchiolization (B), lung tissue and weight change (C) is a confirmation of
도 12a, 도 12b는 PHMG 폐 섬유화 동반 류마티스 관절염 마우스와 대조군에서, 면역대식세포인 대식세포, 호중구 및 림프구의 증가 정도를 확인한 도이다.12A and 12B are diagrams confirming the degree of increase in macrophages, neutrophils and lymphocytes, which are immune macrophages, in rheumatoid arthritis mice with PHMG lung fibrosis and in the control group.
도 13은 PHMG 폐 섬유화 동반 류마티스 관절염 마우스와 대조군에서, Th1, Th2, Treg, Th17 세포의 수치에 따른 면역 세포 조절을 확인한 도이다.13 is a diagram confirming immune cell regulation according to the levels of Th1, Th2, Treg, and Th17 cells in rheumatoid arthritis mice with PHMG lung fibrosis and a control group.
도 14는 본 발명의 통풍 동반 골관절염 동물 모델(MIA+PO+HX)에 대한 통증 정도(Procimal metatasal)를 시간이 지남에 따라 자극을 견디지 못하고 발을 뗄 때까지의 시간(sec)과 가해진 힘(g)을 측정한 결과를 나타낸 도이다. 14 is a graph showing the pain level (Procimal metatasal) for the animal model of osteoarthritis accompanied by gout (MIA+PO+HX) of the present invention, the time (sec) and the applied force ( g) is a diagram showing the measurement result.
도 15는 본 발명의 통풍 동반 골관절염 동물 모델(MIA+PO+HX)에 대한 체중 부하(Weight bearing)를 시간이 지남에 따라 오른쪽 뒷다리 무게(Weight on right hind rimb)(%)을 측정하여 확인한 결과를 나타낸 도이다. 15 is a result of measuring and confirming the weight bearing of the gout-accompanied osteoarthritis animal model (MIA+PO+HX) over time (Weight on right hind rimb) (%) of the present invention. is a diagram showing
도 16a, 도 16b, 도 16c 및 도 16d은 본 발명의 통풍 동반 골관절염 동물 모델(MIA+PO+HX)에 대한 연골 파괴 및 연골 볼륨 비교에 따른 골다공증 심화를 확인하기 위하여, 해면골(Trabecular bone, TB)(%), 골밀도(Bone Mineral Density, BMD)(mg/cm3) 및 구조 분리(Structure separation, St.sp)(U)를 측정하여 확인한 결과를 나타낸 도이다.16A, 16B, 16C, and 16D are views illustrating the deepening of osteoporosis according to the comparison of cartilage destruction and cartilage volume in the animal model of gout accompanying osteoarthritis (MIA+PO+HX) of the present invention, Trabecular bone (TB). ) (%), bone mineral density (BMD) (mg/cm3), and structure separation (St.sp) (U) were measured and confirmed.
도 17은 쇼그렌증후군 환자 유래 타액샘상피세포의 형태를 관찰한 결과를 나타낸 도이다(passage-0).17 is a diagram showing the results of observing the morphology of salivary gland epithelial cells derived from a patient with Sjogren's syndrome (passage-0).
도 18는 쇼그렌증후군 환자 유래 타액샘상피세포(passage-1)를 이용한 오가노이드의 형태를 관찰한 결과를 나타낸 도이다. 18 is a diagram showing the results of observing the morphology of organoids using salivary gland epithelial cells (passage-1) derived from a patient with Sjogren's syndrome.
도 19는 쇼그렌증후군 환자 유래 타액샘상피세포를 이용한 오가노이드의 배양 시, 이소프로테레놀(Isopreterenol)과 염화 칼슘(Calcium chloride)를 처리한 결과를, 도 20는 오가노이드의 아밀라아제 증가를 확인한 도이다.19 shows the results of treatment with isoproterenol and calcium chloride when culturing organoids using salivary gland epithelial cells derived from a Sjogren's syndrome patient, and FIG. 20 is a diagram confirming the increase in amylase of the organoid. am.
도 21는 쇼그렌증후군 환자 유래 타액샘상피세포를 이용한 오가노이드의 아쿠아포린-5(Aquaporin-5), 사이토케라틴-19(Cytokeratin-19), 아밀라아제 A(Amylase A) 발현 공초점현미경으로 분석한 결과를 나타낸 도이다.21 is a result of analysis by confocal microscopy of aquaporin-5, cytokeratin-19, and amylase A expression of organoids using salivary gland epithelial cells derived from a Sjogren's syndrome patient. is a diagram showing
도 22는 마우스 유래 침샘 줄기 세포의 오가노이드를 제작하기 위하여, IL-17를 처리한 후 손상 정도를 확인한 결과를 나타낸 도이다.22 is a view showing the result of confirming the degree of damage after treatment with IL-17 in order to prepare an organoid of mouse-derived salivary gland stem cells.
도 23은 마우스 유래 침샘 줄기 세포의 오가노이드의 침샘 줄기세포의 형성률과 침의 주성분인 알파 아밀라아제(alpha amylase)의 생성 정도를 대조군과 비교한 결과를 나타낸 도이다.23 is a diagram showing the results of comparing the formation rate of salivary gland stem cells of the mouse-derived salivary gland stem cells organoids and the production level of alpha amylase, which is the main component of saliva, with the control group.
도 24a-d은 마우스 유래 침샘 줄기 세포의 오가노이드에 대한 아쿠아포린-5(Aquaporin-5), 나노그(Nanog), 아밀라아제 1(Amylase 1), 케라틴 18(Keratin 18)의 발현 정도를 확인한 도이다. Figure 24a-d is a diagram confirming the expression level of aquaporin-5 (Aquaporin-5), nanog (Nanog), amylase 1 (Amylase 1), keratin 18 (Keratin 18) for organoids of mouse-derived salivary gland stem cells am.
본 발명은 면역결핍 마우스에 루푸스 질환 환자 유래 PBMC(Peripheral blood mononuclear cell)이 투여된, 인간화된 루푸스(systemic lupus erythematosus) 질환 동물 모델을 제공한다.The present invention provides a humanized animal model of systemic lupus erythematosus in which immunodeficient mice are administered with PBMCs (peripheral blood mononuclear cells) derived from lupus disease patients.
본 발명에서 용어, "환자"는 인간, 유인원, 원숭이, 소, 개, 기니아 피그, 토끼, 마우스, 래트, 닭, 곤충 등을 포함하여 치료가 요구되는 임의의 단일 개체를 의미한다. 또한, 임의의 질병 임상 소견을 보이지 않는 임상 연구 시험에 참여한 임의의 대상 또는 역학 연구에 참여한 대상 또는 대조군으로 사용된 대상이 대상에 포함된다. As used herein, the term "patient" means any single individual in need of treatment, including humans, apes, monkeys, cattle, dogs, guinea pigs, rabbits, mice, rats, chickens, insects, and the like. Also included in the subject are any subjects who participated in a clinical study trial without any clinical manifestations of any disease or subjects who participated in epidemiological studies or subjects used as controls.
본 발명에서 용어,"면역결핍 마우스"는 하나 또는 그 이상의 하기 목록에 의해 특징되는 마우스를 의미한다: T 세포 및 B 세포와 같은 기능적 면역 세포의 결함; DNA 복구 결함; 림프구에서 항원 특이적 수용체를 코딩하는 유전자의 재배치 상의 결함; 및 IgM, IgG1, IgG2a, IgG2b, IgG3 및 IgA와 같은 면역 기능 분자들의 결함. 일 구현예에서, 면역결핍 마우스는 면역 기능에 관여하는 Rag1 및 Rag2와 같은 유전자의 하나 이상의 결핍에 의해 특징될 수 있으며 (Oettinger et al, Science, 248:1517-1523, 1990; 및 Schatz et al, Cell, 59:1035-1048, 1989), 면역결핍 마우스는 마우스에서 비정상적인 면역 기능을 초래하는 이들 또는 다른 결함을 가질 수 있다.As used herein, the term "immunodeficient mouse" means a mouse characterized by one or more of the following lists: defects in functional immune cells such as T cells and B cells; DNA repair defects; defects in the rearrangement of genes encoding antigen-specific receptors in lymphocytes; and defects in immune function molecules such as IgM, IgG1, IgG2a, IgG2b, IgG3 and IgA. In one embodiment, immunodeficient mice can be characterized by a deficiency in one or more genes involved in immune function, such as Rag1 and Rag2 (Oettinger et al, Science, 248:1517-1523, 1990; and Schatz et al, Cell, 59:1035-1048, 1989), immunodeficient mice may have these or other defects that result in abnormal immune function in the mouse.
특히 유용한 면역결핍 마우스 종자(strains)는 Shultz et al,, J Immunol, 174: 6477-6489, 2005에 상세히 기재된 바와 같이 일반적으로 NOD scid 감마 (NSG) 마우스를 의미하는 NOD, Cg-PrkdcscidIl2rgtml Wjl/SzJ 및 일반적으로 NRG 마우스를 의미하는 NOD.Cg-Rag1tmlMomIl2rgtml Wjl/SzJ (Shultz et al, Clin Exp Immunol, 154(2):270-284, 2008)가 있다. 본 발명의 일 실시예에서는 NSG 마우스를 사용하였다.Particularly useful immunodeficient mouse strains include NOD, Cg-PrkdcscidIl2rgtml Wjl/SzJ, commonly referred to as NOD scid gamma (NSG) mice, as detailed in Shultz et al,, J Immunol, 174: 6477-6489, 2005. and NOD.Cg-Rag1tmlMoml12rgtml Wjl/SzJ, which generally refers to NRG mice (Shultz et al, Clin Exp Immunol, 154(2):270-284, 2008). In an embodiment of the present invention, NSG mice were used.
본 발명에서 용어, "중증 복합성 면역결핍증(severe combined immune deficiency; SCID)"은 T 세포의 부재 및 B 세포 기능의 부재를 특징으로 하는 증상을 의미한다. SCID 돌연변이는 기능적 B 세포 및 T 세포의 결손을 야기시키는 돌연변이를 말한다. 예를 들면, SCID 마우스 (CB-17-Prkdcscid)는 B 세포 수용체 (B cell receptor: BCR) 및 T 세포 수용체(T cell receptor: TCR)의 재배열에 결함이 있어, 기능적 B 세포 및 T 세포의 결손이 야기된다. 상기 NSG (NOD scid gamma) 마우스는 NOD/SCID γnull 마우스 또는 NOD/SCID IL-2RγKO 마우스라고도 한다. NSG 마우스는 잭슨 라보라토리 (The Jackson Laboratory)의 레오나르드 슐츠 박사 (Dr Leonard Shultz)의 실험실에서 개발되었다. 상기 NSG 마우스는 잭슨 라보라토리로부터 상업적으로 구입하거나, 공지된 방법에 따라 제조할 수 있다 (Shultz LD, et al, J Immunol 2005; 169: 204-209). 예를 들면, NSG 마우스는 C57BL/6J-γ null마우스와 NOD/SCID 마우스를 9회 역교배 (backcross mating)하여 얻어질 수 있다. NSG 마우스는 기능적 T 세포 및 기능적 B 세포가 결여되어 있고, 대식구 기능이 감소되어 있고, NK 세포 또는 NK 활성이 제거되어 있고, 수지상 세포 기능이 감소되어 있는 것으로 알려져 있다. NSG 마우스는 NOD/SCID 마우스 또는 γ2-마이크로글로불린-결여 NOD/LtSz-SCID (NOD/SCID/γ2m null)에 비하여 이종조직 (xenograft)의 생착율(engraftment level)이 높은 것으로 알려져 있다.As used herein, the term "severe combined immune deficiency (SCID)" refers to a symptom characterized by the absence of T cells and the absence of B cell function. SCID mutations refer to mutations that result in the deletion of functional B cells and T cells. For example, SCID mice (CB-17-Prkdcscid) are defective in rearrangement of B cell receptor (BCR) and T cell receptor (TCR), resulting in functional B and T cell deficits. this is caused The NSG (NOD scid gamma) mouse is also referred to as a NOD/SCID γnull mouse or a NOD/SCID IL-2RγKO mouse. NSG mice were developed in the laboratory of Dr Leonard Shultz at The Jackson Laboratory. The NSG mice can be purchased commercially from Jackson Laboratories, or prepared according to a known method (Shultz LD, et al, J Immunol 2005; 169: 204-209). For example, NSG mice can be obtained by backcross mating C57BL/6J-γ null mice and NOD/SCID mice 9 times. It is known that NSG mice lack functional T cells and functional B cells, have reduced macrophage function, abolished NK cells or NK activity, and reduced dendritic cell function. NSG mice are known to have a higher engraftment level of xenografts compared to NOD/SCID mice or γ2-microglobulin-deficient NOD/LtSz-SCID (NOD/SCID/γ2m null).
본 발명에서 상기 용어들 "NOD scid 감마"와 "NSG는 상호 교환되어 사용될 수 있다. Cg-Prkdcscid NSG 마우스는 NOD/ShiLtJ를 바탕으로 한 다중 면역 결핍, 중증 복합성 면역결핍증 및 인터루킨-2 수용체 감마 사슬의 완전한 넉아웃(knockout)을 가지고 있다. 그 결과, NSG 마우스는 사이토카인 신호가 결핍되어 있다. NSG 마우스는 IL2R-γ(감마 c) 발현의 결핍, 검출 가능한 혈청 면역 글로불린의 부존재, 용혈 보체 부존재, 성숙한 T 림프구의 부존재 및 성숙한 NK 세포의 부존재로 특징된다.In the present invention, the terms "NOD scid gamma" and "NSG" may be used interchangeably. Cg-Prkdcscid NSG mice have multiple immunodeficiency based on NOD/ShiLtJ, severe combined immunodeficiency, and interleukin-2 receptor gamma chain. As a result, NSG mice lack cytokine signaling.NSG mice lack IL2R-γ (gamma c) expression, no detectable serum immunoglobulin, no hemolytic complement. , characterized by the absence of mature T lymphocytes and the absence of mature NK cells.
상기 인간화된 마우스 모델을 전임상 연구에 사용함으로써, 환자 특이 루푸스를 상기 인간화된 마우스 모델에서 연구할 수 있다. 임의의 시험 화합물 또는 약물을 상기 인간화된 마우스 모델에 하나 또는 그 이상의 투약 요법에 따라 투여함으로써, 이들 시험 화합물 또는 약물의 효과, 안전성 (예, 면역계에 대한 여하한 관련 부작용) 및 효능을 연구할 수 있다. 이러한 방법은 루프스나 면역 조절제, 또는 질병 조직 (예, 암, 자가 면역 질환)과 면역계 사이의 상호 작용을 포함하는 연구에 특히 효과적이다.By using the humanized mouse model for preclinical studies, patient-specific lupus can be studied in the humanized mouse model. By administering any test compound or drug to the humanized mouse model according to one or more dosing regimens, the effectiveness, safety (e.g., any relevant adverse effects on the immune system) and efficacy of these test compounds or drugs can be studied. have. These methods are particularly effective for studies involving the interaction between lupus or immunomodulators, or diseased tissues (eg, cancer, autoimmune diseases) and the immune system.
상기 인간화된 마우스 모델은 생착된(engrafted) 인간 조혈계의 존재 하에 임의의 환자-유래 이종 이식을 연구하는 데에도 사용될 수 있다. 여기에는 종양 조직학 연구, 오믹 연구 또는 프로파일링 (프로테오믹(proteomic), 게노믹(genomic, 메타블로믹(metablomic 등)이 포함된다. 특정 구현예들에 있어서, 연구 중인 환자-유래 이종 이식에 관한 정보는 마우스 종양 생물학 데이터베이스 (MTB)에서 얻을 수 있으며, 이는 실험 모델을 선택하고, 특정 암에서 돌연변이의 패턴을 검토하고, 암 스펙트럼 전반에 걸쳐 돌연변이가 통상적으로 발생하는 유전자들을 동정하는 것과 같은 분야의 연구자를 돕기 위해 고안되었다.This humanized mouse model can also be used to study any patient-derived xenograft in the presence of an engrafted human hematopoietic system. This includes tumor histology studies, omic studies or profiling (proteomic, genomic, metablomic, etc.). In certain embodiments, the patient-derived xenograft under study is Information on this can be obtained from the Mouse Tumor Biology Database (MTB), for areas such as selecting experimental models, examining patterns of mutations in specific cancers, and identifying genes in which mutations commonly occur across the cancer spectrum. designed to help researchers in
본 발명은 면역결핍 마우스에 루푸스 질환 환자로부터 분리한 PBMC를 주입하는 단계를 포함하는, 인간화된 루푸스 질환 동물 모델을 제작하는 방법을 제공한다.The present invention provides a method for preparing a humanized lupus disease animal model, comprising injecting PBMC isolated from a lupus disease patient into an immunodeficient mouse.
본 발명은 인간화된 루푸스 동물 모델에 후보 물질을 처리하는 단계;를 포함하는, 루푸스 치료 물질을 스크리닝하는 방법을 제공한다.The present invention provides a method for screening a lupus therapeutic agent, comprising the step of treating a candidate substance in a humanized lupus animal model.
본 발명은 류마티스 관절염 발생 동물에 베타 글루칸 또는 살균제가 투여되어 폐 섬유화가 발생된, 폐 섬유화가 동반된 류마티스 관절염 동물 모델을 제공한다.The present invention provides an animal model of rheumatoid arthritis accompanied by pulmonary fibrosis, in which pulmonary fibrosis occurs by administering beta-glucan or a fungicide to an animal with rheumatoid arthritis.
본 발명에서 용어 "류마티스 관절염 발생 동물"은 T세포 면역 체계의 유전적 결함이 있어 T세포 유발 만성 자가면역질환이 유발되어 류마티스 관절염의 동물 모델로 이용되는 SKG 마우스를 이용할 수 있다. 또한, 콜라겐 유도성 관절염 모델(Collagen-in-duced arthritis, CIA), 유전자 변형 자연발생 관절염 모델, 항체 유도성 관절염 모델 및 인간화 모델인 severe combined immunodeficiency(SCID)-HuRAg 마우스 등을 이용할 수 있다. 또한, 본 발명에 따른 동물 모델의 제조를 위해 사용될 수 있는 실험용 쥐의 종류로는, 이에 제한되는 것은 아니나, 계통별 분류상 폐쇄군(Closed Colony)에 속하는 ICR 또는 DDY; 근교계(Inbred)에 속하는 BALB/cA, C57BL/6N, C3H/HeN, DBA/2N 또는 CBA/N; 교잡군(hybrid)에 속하는 BDF1(C57BL/6 x DBA/2), CDF1(CBA/N x DBA/2) 또는 B6C3F1(C57BL6 x C3H/HeN); 돌연변이계(mutant)에 속하는 BALB/c-nu 또는 C,B-17SCID 등 실험용 쥐로 사용되는 것이면 모두 이용할 수 있다.As used herein, the term "rheumatoid arthritis-inducing animal" may use SKG mice used as an animal model for rheumatoid arthritis because there is a genetic defect in the T-cell immune system and thus T-cell-induced chronic autoimmune disease is induced. In addition, a collagen-in-duced arthritis (CIA) model, a genetically modified naturally occurring arthritis model, an antibody-induced arthritis model, and a humanized severe combined immunodeficiency (SCID)-HuRAg mouse can be used. In addition, the types of laboratory mice that can be used for the preparation of the animal model according to the present invention include, but are not limited to, ICR or DDY belonging to a closed colony according to phylogenetic classification; BALB/cA, C57BL/6N, C3H/HeN, DBA/2N or CBA/N belonging to Inbred; BDF1 (C57BL/6 x DBA/2), CDF1 (CBA/N x DBA/2) or B6C3F1 (C57BL6 x C3H/HeN) belonging to the hybrid; Any one used as an experimental mouse, such as BALB/c-nu or C,B-17SCID belonging to the mutant family, can be used.
본 발명의 목적 상, 상기 류마티스 관절염 동물 모델은 폐 섬유화가 일부 발생할 뿐, 발생율을 임의 조절할 수 없는 한계점이 존재하는 바, 이를 개선하기 위하여, 본 발명의 명확한 폐 섬유화가 동반된 류마티스 관절염 동물 모델을 구축하게 되었다.For the purpose of the present invention, the rheumatoid arthritis animal model has a limitation in that only some lung fibrosis occurs, and the incidence rate cannot be arbitrarily controlled. got to build
상기 베타 클루칸은 커들란(Curdlan)이고, 베타 클루칸은 1~500mg/kg의 농도로 투여될 수 있으나, 이에 제한되지 않는다.The beta glucan is curdlan, and the beta glucan may be administered at a concentration of 1 to 500 mg/kg, but is not limited thereto.
상기 살균제는 폴리헥사메틸렌 구아디닌(Polyhexamethylene guanidine, PHMG), 염산폴리헥사메틸렌비구아니드(PHMB), 메칠이소치아졸리논(MIT), 염화에톡시에틸구아디닌(Chloride Ethoxyethyl Guanidine, PGH), 나프탈렌(naphthalene), 프탈레이트(phthalate), 디데실메틸염화암모늄(Didecyl methyl ammonium chloride, DDAC), 벤즈아이소사이아졸리논(BIT), 클로록실레놀(chloroxylenol), 티타늄 다이옥사이드, 징크 옥사이트 및 아세톤으로 이루어진 군에서 선택된 1종 이상이고, 바람직하게는 구아디닌 계열의 성분인 폴리헥사메틸렌 구아디닌(Polyhexamethylene guanidine, PHMG), 염산폴리헥사메틸렌비구아니드(PHMB), 염화에톡시에틸구아디닌(Chloride Ethoxyethyl Guanidine, PGH)이며, 더욱 바람직하게는 폴리헥사메틸렌 구아디닌(Polyhexamethylene guanidine, PHMG)이나, 이에 제한되지 않는다. 상기 살균제는 1~500mg/kg의 농도로 투여될 수 있으나, 이에 제한되지 않는다.The disinfectant is polyhexamethylene guanidine (PHMG), hydrochloric acid polyhexamethylene biguanide (PHMB), methyl isothiazolinone (MIT), ethoxyethyl guanidine chloride (Chloride Ethoxyethyl Guanidine, PGH) , naphthalene, phthalate, didecyl methyl ammonium chloride (DDAC), benzisocyazolinone (BIT), chloroxylenol, titanium dioxide, zinc oxide and At least one selected from the group consisting of acetone, preferably polyhexamethylene guanidine (PHMG), which is a component of guanine series, polyhexamethylene biguanide hydrochloride (PHMB), ethoxyethyl chloride Adinine (Chloride Ethoxyethyl Guanidine, PGH), more preferably polyhexamethylene guanidine (Polyhexamethylene guanidine, PHMG), but is not limited thereto. The disinfectant may be administered at a concentration of 1-500 mg/kg, but is not limited thereto.
본 발명에서 용어 "폴리헥사메틸렌 구아디닌(Polyhexamethylene guanidine, PHMG)"는 가습기 살균제의 주요 성분으로서 사용되어 왔고, 고농도로 폐에 노출되면 감기나 폐렴 증상이 발생하고 간질성 폐렴으로 진전돼 폐가 딱딱해져 호흡곤란이 발생하기도 한다. 폐 손상은 회복되지 못하고 고착성 폐 기능 저하로 폐를 이식하지 않으면 사망에 이르게 된다. In the present invention, the term "polyhexamethylene guanidine (PHMG)" has been used as a major component of a humidifier disinfectant, and when exposed to the lungs at high concentrations, cold or pneumonia symptoms occur and develop into interstitial pneumonia, causing the lungs to harden. It may also cause shortness of breath. Lung damage cannot be repaired, and if a lung is not transplanted due to the fixed deterioration of lung function, it leads to death.
상기 투여는 경구, 정맥, 근육, 피하 및 복강내 주사로 이루어진 군에서 선택될 수 있고, 바람직하게는 상기 커들란의 투여 시, 경구 투여 및 복강 내 주사로 투여할 수 있으며, 상기 각 투여 방법에 의한 커들란은 1:2의 중량 비율로 투여될 수 있으나, 이에 제한되지 않는다.The administration may be selected from the group consisting of oral, intravenous, intramuscular, subcutaneous and intraperitoneal injections, and preferably, when administering the curdlan, it may be administered by oral administration and intraperitoneal injection. Curdlan may be administered in a weight ratio of 1:2, but is not limited thereto.
상기 투여는 1-5주 동안 1-5회 투여될 수 있고, 바람직하게는 1-3주 동안 1-3회 투여될 수 있으나, 이에 제한되지 않는다.The administration may be administered 1-5 times for 1-5 weeks, preferably 1-3 times for 1-3 weeks, but is not limited thereto.
상기 동물은 마우스, 래트 및 햄스터, 토끼, 말, 소, 개, 고양이, 원숭이 및 기니 피그로 이루어진 군에서 선택된 1종 이상이나, 바람직하게는 마우스이나 이에 제한되지 않는다. The animal is one or more selected from the group consisting of mice, rats and hamsters, rabbits, horses, cattle, dogs, cats, monkeys and guinea pigs, but preferably mice, but not limited thereto.
상기 폐 섬유화는 류마티스 관절염에서 동반되는 폐 섬유증, 통상성 간질성 폐렴, 비특이성 간질성 폐렴, 염증성 기도 질환 및 기질화 폐렴으로 이루어진 군에서 선택된 1종 이상의 폐 질환을 일으킬 수 있다.The lung fibrosis may cause one or more lung diseases selected from the group consisting of pulmonary fibrosis accompanying rheumatoid arthritis, common interstitial pneumonia, non-specific interstitial pneumonia, inflammatory airway disease, and organized pneumonia.
상기 구축된 폐 섬유화를 동반한 류마티스 관절염 동물 모델은 관절염 지수가 대조군 보다 높고, 마우스의 체중이 감소되는 변화를 확인하였다. 또한, 마우스의 심장, 폐, 간, 근육, 관절 및 척추에서 염증이 증가되고, 폐 섬유화에 의한 폐의 손상이 나타남을 확인하였다. 또한, 폐포의 세 기관지화(alveolar bronchiolization)가 대조군과 비교하여 유의적으로 발생하고, 대식세포(macrophage), 호중구(neutrophil), 림프구(lymphocyte)가 증가되어 면역 반응이 증대되고, 염증성 세포인 Th17 세포가 증가되고, 면역 반응을 제어하는 Treg 세포는 감소되는 특징이 나타남을 확인하였다.In the constructed animal model of rheumatoid arthritis with lung fibrosis, the arthritis index was higher than that of the control group, and it was confirmed that the mouse weight decreased. In addition, it was confirmed that inflammation was increased in the heart, lung, liver, muscle, joint and spine of mice, and damage to the lungs due to pulmonary fibrosis appeared. In addition, alveolar bronchiolization occurred significantly compared to the control group, macrophages, neutrophils, and lymphocytes were increased to increase immune response, and inflammatory cells, Th17 It was confirmed that the cells were increased and the Treg cells controlling the immune response were decreased.
또한 본 발명은 류마티스 관절염 발생 마우스에 베타 글루칸 또는 살균제를 투여하여 폐 섬유화를 유도하는 단계;를 포함하는, 폐 섬유화가 동반된 류마티스 관절염 동물 모델의 제조 방법을 제공한다.The present invention also provides a method for preparing an animal model of rheumatoid arthritis accompanied by lung fibrosis, comprising the step of inducing pulmonary fibrosis by administering beta-glucan or a fungicide to a mouse with rheumatoid arthritis.
또한 본 발명은 상기 폐 섬유화가 동반된 류마티스 관절염 동물에 후보 물질을 처리하는 단계;를 포함하는, 폐 섬유화가 동반된 류마티스 관절염 치료 물질을 스크리닝하는 방법을 제공한다.The present invention also provides a method for screening a rheumatoid arthritis therapeutic agent with lung fibrosis, comprising the step of treating the candidate substance to the rheumatoid arthritis animal accompanied by pulmonary fibrosis.
본 발명은 골관절염 발생 동물에 칼륨 옥소네이트(potassium oxonate, PO) 또는 히포잔틴(Hypoxanthine, HX)가 투여되어 통풍이 발생된, 통풍이 동반된 골관절염 동물 모델을 제공한다.The present invention provides an animal model of osteoarthritis accompanied by gout, in which gout is developed by administering potassium oxonate (PO) or hypoxanthine (HX) to an animal with osteoarthritis.
본 발명에서 용어 " 칼륨 옥소네이트(potassium oxonate, PO)"은 요산 분해 효소 저해제로서, 고요산혈증(hyperuricemia)을 유도할 수 있다.In the present invention, the term "potassium oxonate (PO)" is a uric acid lyase inhibitor, which can induce hyperuricemia.
본 발명에서 용어 "히포잔틴(Hypoxanthine, HX)"은 자연적으로 생성되는 퓨린 유도체이다. 하이포잔틴은 핵산의 구성 성분으로 종종 발견되며, tRNA의 안티코돈에 뉴클레오사이드인 이노신의 형태로 존재한다. 하이포잔틴은 기질 및 질소 공급원으로 특정 세포, 세균, 기생충의 배양에 필요한 첨가물이다.As used herein, the term "hypoxanthine (HX)" is a naturally occurring purine derivative. Hypoxanthin is often found as a component of nucleic acids and is present in the form of inosine, a nucleoside, in the anticodon of tRNA. Hypoxanthin is an additive necessary for the culture of certain cells, bacteria, and parasites as a substrate and nitrogen source.
본 발명에서 용어 "골관절염 발생 동물"은 전방십자인대 절제 방법, 반월판 절제 방법, 트랜스제닉/녹아웃 방법, 화학적 주사 방법, 자연발생 마우스 등으로 골관절염이 발생된 동물을 의미한다. As used herein, the term "osteoarthritis-inducing animal" refers to an animal that has developed osteoarthritis by an anterior cruciate ligament resection method, a meniscus resection method, a transgenic/knockout method, a chemical injection method, a naturally occurring mouse, or the like.
본 발명의 목적 상, 상기 전방십자인대 절제 방법을 이용한 양, 개, 토끼, 래트 동물이 포함될 수 있으며, 반월판 절제한 양, 개, 토끼, 기니피그, 래트, 마우스 등을 포함할 수 있다. 또한, 트랜스제닉/녹아웃 방법으로 구축된 ADAM-15 KO, MMP-14 KO, MMP-13 트랜스제닉 마우스를 포함할 수 있으며, 화학적 주사 방법으로 구축된 콜라게나제에 의한 마웃, 토끼, 래트를 포함할 수 있다. MIA(Monosodium iodoacetate) 주사한 마우스, 래트, 토끼, 기니피그를 포함할 수 있다. 또한, 자연발생한 DBA/1(마우스), STR/ORT(마우스), C57BL6(마우스), 하틀리 기니피그, 레수스 마카크(rhesus macaque; 원숭이), 시노몰거스 마카크(Cynomolgus macaque; 원숭이)를 포함할 수 있고, 통풍 동반 골관절염 동물 모델을 구축하기 위해서라면, 이에 제한되지 않는다. For the purposes of the present invention, sheep, dogs, rabbits, and rat animals using the anterior cruciate ligament resection method may be included, and may include meniscus-resected sheep, dogs, rabbits, guinea pigs, rats, mice, and the like. In addition, it may include ADAM-15 KO, MMP-14 KO, MMP-13 transgenic mice constructed by the transgenic/knockout method, and include mice, rabbits, and rats by collagenase constructed by the chemical injection method. can do. MIA (Monosodium iodoacetate) injection may include mice, rats, rabbits, and guinea pigs. Also includes naturally occurring DBA/1 (mouse), STR/ORT (mouse), C57BL6 (mouse), Hartley guinea pig, rhesus macaque (monkey), Cynomolgus macaque (monkey) and, if it is to construct an animal model of osteoarthritis accompanied by gout, it is not limited thereto.
본 발명의 목적 상, 통풍은 골관절염 환자에서 골파괴를 촉진시키는 바, 본 발명의 통풍이 동반된 골관절염 동물 모델은 종래 골관절염 동물모델에서도 일부 통풍이 발생하지만 발생율을 임의로 조절할 수 없었던 한계를 극복한 것으로, 명확한 통풍을 동반한 골관절염 동물 모델을 구축하였다.For the purpose of the present invention, gout promotes bone destruction in osteoarthritis patients, and the animal model of osteoarthritis accompanied by gout of the present invention overcomes the limitation that some gout occurs in the conventional animal model of osteoarthritis, but the incidence rate cannot be arbitrarily controlled. , established an animal model of osteoarthritis with clear gout.
상기 투여는 경구, 정맥, 근육, 피하 및 복강내 주사로 이루어진 군에서 선택될 수 있고, 경구 투여는 칼륨 옥소네이트로 투여하고, 복강 내 투여는 히포잔틴을 투여할 수 있으나, 이에 제한되지 않는다. The administration may be selected from the group consisting of oral, intravenous, intramuscular, subcutaneous and intraperitoneal injection, oral administration may be administered with potassium oxonate, and intraperitoneal administration may be administered with hypoxanthine, but is not limited thereto.
상기 칼륨 옥소네이트 및 히포잔틴는 1:1 의 중량 비율로 투여될 수 있고, 1-5주 동안 1-10회 투여될 수 있고, 바람직하게는 1-3주 동안 1-7회 투여될 수 있으나, 이에 제한되지 않는다.The potassium oxonate and hypoxanthine may be administered in a weight ratio of 1:1, may be administered 1-10 times for 1-5 weeks, preferably 1-7 times for 1-3 weeks, but , but not limited thereto.
또한 본 발명은 골관절염 발생 동물에 칼륨 옥소네이트 또는 히포잔틴을 투여하여 통풍을 유도하는 단계를 포함하는, 통풍이 동반된 골관절염 동물 모델의 제조 방법을 제공한다.The present invention also provides a method for producing an animal model of osteoarthritis accompanied by gout, comprising the step of inducing gout by administering potassium oxonate or hypoxanthine to an animal with osteoarthritis.
또한 본 발명은 상기 통풍이 동반된 골관절염 동물 모델에 후보 물질을 처리하는 단계;를 포함하는, 통풍이 동반된 골관절염 치료 물질을 스크리닝하는 방법을 제공한다.In addition, the present invention provides a method for screening a treatment material for osteoarthritis accompanied by gout, comprising the step of treating the candidate material to the animal model of osteoarthritis accompanied by gout.
본 발명은 마우스 유래 침샘 줄기세포를 배양하여 수득한 오가노이드에 인터루킨-17(Interleukin-17)이 투여된, 쇼그렌증후군(Sjogren's syndrome) 오가노이드(organoid) 모델을 제공한다.The present invention provides an organoid model of Sjogren's syndrome in which Interleukin-17 is administered to an organoid obtained by culturing mouse-derived salivary gland stem cells.
본 발명에서 용어, "오가노이드(organoid)"란 조직에서나 혹은 배아줄기세포에서 유래된 세포를 이용하여 이를 3D 형태로 배양을 하여 마치 인공장기와 같은 형태로 만들 수 있는 것을 의미한다. 오가노이드는 장기의 'organ'과 같은 의미를 가진 접미어로 '장기와 유사한 것'이라는 말을 지니고 있다. 오가노이드는 3차원 배양방법을 통하여 세포와 세포의 기능이 좀 더 잘 배열되고, 기능성을 가지는 기관 같은 형태와 기능을 지닌다. 오가노이드는 줄기세포연구와 3D 세포배양 등이 개발되고, 다양한 조직으로 분화시킬 수 있는 생장 및 분화 인자의 최적화 연구와 함께 주목을 받고 있다.As used herein, the term "organoid" means that cells derived from tissues or embryonic stem cells can be cultured in a 3D form to form an artificial organ. Organoid is a suffix that has the same meaning as 'organ' of an organ, and has the word 'organ-like'. Organoids have a more well-arranged cell and cell function through a three-dimensional culture method, and have a functional organ-like form and function. Organoids are attracting attention along with research on optimization of growth and differentiation factors that can differentiate into various tissues, with stem cell research and 3D cell culture being developed.
본 발명에서 용어, "타액(saliva)"은 이하선, 악하선, 설하선 및 구강점막에 존재하는 점액선 등으로부터 분비되는 혼합액이다. 타액은 인체의 핵심 성분으로 타액선에서 생성되어 구강 내로 배출된다. 타액은 인체의 필수 성분으로서, 생체 활성단백질, 소화효소, 점액, 면역글로불린, 각종 염류 등이 포함되어 있다. 타액은 구강 내 건강뿐만 아니라 인체의 항상성 유지에 매우 중요한 역할을 수행한다. 예를 들어 타액의 주요 성분인 뮤신(mucin), 면역글로불린 등은 외부의 감염으로부터 1차적인 방어 역할을 수행하며, 구강 및 치아의 윤활 및 수분 유지, pH 중화 기능을 통하여 구강점막 및 치아를 보호한다. 뿐만 아니라 타액에는 프티알린(ptyalin) 등의 아밀라아제(amylase)와 같은 소화 효소들이 있어 전분을 말토오스 단위까지 분해하는 등 소화를 촉진시키는 기능을 담당한다. 또한 타액의 분비에 의하여 인체의 수분대사 및 체온 조절이 이루어질 수 있으며, 유독물(I, Hg, Pb 등)을 배설하기도 한다.As used herein, the term "saliva" is a mixed solution secreted from the parotid, submandibular, sublingual, and mucous glands present in the oral mucosa. Saliva is a key component of the human body, produced in the salivary glands and discharged into the oral cavity. Saliva is an essential component of the human body and contains bioactive proteins, digestive enzymes, mucus, immunoglobulins, and various salts. Saliva plays a very important role in maintaining the homeostasis of the human body as well as oral health. For example, the main components of saliva, mucin, immunoglobulin, etc., play a primary defense role against external infection, and protect the oral mucosa and teeth through lubrication and moisture maintenance of the oral cavity and teeth, and neutralizing pH. do. In addition, saliva contains digestive enzymes such as amylase, such as ptyalin, which promotes digestion by decomposing starch to maltose units. In addition, the body's water metabolism and body temperature control can be achieved by the secretion of saliva, and toxic substances (I, Hg, Pb, etc.) are excreted.
본 발명에서 용어, "타액선(침샘, salivary gland)"은 타액을 생성, 분비하는 기관으로 이하선(귀밑샘, parotid gland), 악하선(턱밑샘, submaxillary gland), 설하선(혀밑샘, sublingual gland)과 같은 주타액선(major salivary gland)과, 점액선(mucous gland)과 같이 구강점막(mucous membrane of oral cavity)에 존재하는 점액선(mucous gland)과 같이 구강점막의 여러 부위에 분포하는 소타액선(minor salivary gland)으로 분류된다.As used herein, the term "salivary gland" refers to an organ that produces and secretes saliva, such as parotid gland, submandibular gland, and sublingual gland. It is a minor salivary gland distributed in various parts of the oral mucosa, such as major salivary glands and mucous glands existing in the mucous membrane of oral cavity, such as mucous glands. classified.
본 발명에 있어서, 유래되는 마우스는 제한되지 않고, 일반 실험용 마우스, 면역결핍 마우스 등이 이용될 수 있다. In the present invention, the derived mouse is not limited, and general laboratory mice, immunodeficient mice, and the like may be used.
또한 본 발명은 쇼그렌 환자 유래 타액샘 상피세포(Patient-derived salivary gland epithelial cell)를 포함하는, 쇼그렌증후군(Sjogren's syndrome) 오가노이드(organoid) 모델을 제공한다.In addition, the present invention provides an organoid model of Sjogren's syndrome, including Patient-derived salivary gland epithelial cells.
쇼그렌 환자 유래 타액샘 상피세포를 단일층으로 배양할 시 3D구조에서 기능하는 acinar cell의 정확한 기능-예를 들면 분비단백질 생성능, water channel- 그리고 acinar cell polarization 등을 확인하기 어려운 현실이다. 정상인과 쇼그렌 환자의 침샘세포로 오가노이드 제작 시 뚜렷한 특징이 나타나지는 않고, 침샘줄기세포의 숫자감소, 침 분비와 관련있는 유전자 (Amy1, Aquaporin5 등)의 감소가 일어난다. When culturing Sjogren's patient-derived salivary gland epithelial cells as a single layer, it is difficult to ascertain the exact functions of acinar cells functioning in a 3D structure - for example, secretory protein production ability, water channel - and acinar cell polarization. When making organoids with salivary gland cells from normal individuals and Sjogren patients, no distinct characteristics appear, and the number of salivary gland stem cells decreases and salivation-related genes (Amy1, Aquaporin5, etc.) decrease.
또한 본 발명은 마우스 유래 침샘 줄기세포를 배양하여 수득하는 단계; 및 상기 세포를 배양하여 오가노이드를 생성 후 인터루킨-17(Interleukin-17)이 투여하는 단계를 포함하는, 쇼그렌 증후군(Sjogren's syndrome) 오가노이드(organoid) 모델 제작 방법을 제공한다.In addition, the present invention is a mouse culturing and obtaining derived salivary gland stem cells; And it provides a method for producing an organoid model of Sjogren's syndrome, comprising the step of culturing the cells to generate an organoid and then administering Interleukin-17.
또한 본 발명은 쇼그렌 환자 유래 타액샘 상피세포를 배양하는 단계를 포함하는, 쇼그렌 증후군(Sjogren's syndrome) 오가노이드(organoid) 모델 제작 방법을 제공한다.The present invention also provides a method for producing a Sjogren's syndrome organoid model, comprising culturing the salivary gland epithelial cells derived from a Sjogren patient.
또한 본 발명은 상기 쇼그렌증후군(Sjogren's syndrome) 오가노이드(organoid) 모델에 후보물질을 처리하는 단계;를 포함하는, 쇼그렌증후군 치료물질을 스크리닝 하는 방법을 제공한다.In addition, the present invention provides a method for screening a Sjogren's syndrome therapeutic material, including; treating a candidate material in the Sjogren's syndrome organoid model.
이하, 첨부된 도면을 참조하여 본 발명의 구현예로 본 발명을 상세히 설명하기로 한다. 다만, 하기 구현예는 본 발명에 대한 예시로 제시되는 것으로, 당업자에게 주지 저명한 기술 또는 구성에 대한 구체적인 설명이 본 발명의 요지를 불필요하게 흐릴 수 있다고 판단되는 경우에는 그 상세한 설명을 생략할 수 있고, 이에 의해 본 발명이 제한되지는 않는다. 본 발명은 후술하는 특허 청구범위의 기재 및 그로부터 해석되는 균등 범주 내에서 다양한 변형 및 응용이 가능하다.Hereinafter, the present invention will be described in detail as an embodiment of the present invention with reference to the accompanying drawings. However, the following embodiments are presented as examples of the present invention, and when it is determined that detailed descriptions of well-known techniques or configurations known to those skilled in the art may unnecessarily obscure the gist of the present invention, the detailed description may be omitted, and , the present invention is not limited thereby. Various modifications and applications of the present invention are possible within the scope of equivalents interpreted therefrom and the description of the claims to be described later.
하기의 실시예를 통하여 본 발명을 보다 상세하게 설명한다. 그러나 하기 실시예는 본 발명의 내용을 구체화하기 위한 것일 뿐 이에 의해 본 발명이 한정되는 것은 아니다. The present invention will be described in more detail through the following examples. However, the following examples are only for specifying the contents of the present invention, and the present invention is not limited thereto.
<실시예 1><Example 1> 루푸스 환자 모사 아바타 모델 평가 플랫폼Lupus patient imitation avatar model evaluation platform
<실시예 1-1> 루푸스 환자 모사 아바타 모델 평가 플랫폼 구축<Example 1-1> Establishment of an evaluation platform for lupus patient imitation avatar model
인간 면역 시스템을 탑재한 루푸스 인간화 마우스 모델 (아바타 모델)을 수A humanized mouse model of lupus (avatar model) equipped with a human immune system
립하기 위해, 8-10주령 NSG 마우스에 정상인 또는 루푸스 환자 유래 PBMC(Peripheral blood mononuclear cell)을 5x106/mice로 복강내 주사한 뒤, 3주In order to establish liposomes, PBMCs (peripheral blood mononuclear cells) derived from normal or lupus patients were intraperitoneally injected into 8-10 week old NSG mice at 5x106/mice, and then 3 weeks
후 혈액 내 정상인 또는 루푸스 환자 세포의 생착 확인을 혈청 및 오줌으로 확인하After confirming the engraftment of normal or lupus patient cells in the blood, serum and urine
였다. 그 후, 세포이식 4주 후 마우스를 희생하여 조직 내 인간 세포의 침윤 및 조직학적 변화를 확인하였다(도 1).it was Thereafter, the mice were sacrificed 4 weeks after cell transplantation to confirm the infiltration of human cells and histological changes in the tissue ( FIG. 1 ).
<실시예 1-2> 루푸스 환자 모사 아바타 모델의 혈액 내 인간 세포 생착 확인<Example 1-2> Confirmation of engraftment of human cells in blood of lupus patient imitation avatar model
상기 실시예 1-1에서 루푸스 환자 모사 아바타 모델이 제대로 구축되었는지 확인하기 위해, 인간 세포의 생착을 혈액 내 T 세포 및 B 세포를 분석함으로써 확인하였다. 구체적으로, 실시예 1-1의 루푸스 인간화 마우스 (정상인 PBMC 주입군 및 루푸스 환자 PBMC 주입군)로부터 혈액을 수득한 뒤, 인간 CD4, CD8 및 CD19 항체와 반응하여 양성인 세포를 유세포 분석을 통해 분석하였다.In order to confirm that the lupus patient mimic avatar model was properly constructed in Example 1-1, engraftment of human cells was confirmed by analyzing T cells and B cells in the blood. Specifically, blood was obtained from the lupus humanized mice of Example 1-1 (normal PBMC-injected group and lupus patient PBMC-injected group), and cells that reacted with human CD4, CD8 and CD19 antibodies were analyzed through flow cytometry. .
그 결과, 정상인과 루푸스 환자의 PBMC 주입에 의해 마우스 혈액 내 인간As a result, human blood in mouse blood by PBMC injection of normal subjects and lupus patients
CD4+T, CD8+T, Cd19+T 세포가 검출되는 것으로 보아 인간 세포가 잘 생착됨을 확인하였다(도 2).CD4+T, CD8+T, and Cd19+T cells were detected, confirming that human cells were well engrafted ( FIG. 2 ).
<실시예 1-3> 루푸스 환자 모사 아바타 모델에서 인간의 면역세포아형 침윤 확인<Example 1-3> Confirmation of infiltration of human immune cell subtypes in a simulated avatar model of a lupus patient
상기 실시예 1-1에서 수립한 루푸스 인간화 마우스 모델에서의 조직 내 인간 세포의 생착을 확인하기 위하여, 실시예 1의 4주후 루푸스 인간화 마우스를 희생시켜 신장을 적출한 뒤, 조직 내 인간의 면역세포아형(CD3, CD4, CD8, CD19)이 침윤 되었는지 IHC 염색하였다. 적출한 조직은 포르말린으로 고정한 후, 파라핀에 임베 딩하여 5㎛ 두께의 절편을 생성하였다. 조직 내 면역세포를 관찰하기 위해 상기 절편 슬라이드에서 인간 CD3, CD4, CD8, CD19 항체와 반응시켜 면역조직화학분석을 수행하였다.In order to confirm the engraftment of human cells in the tissue in the humanized lupus mouse model established in Example 1-1, the humanized lupus mice were sacrificed 4 weeks after Example 1 and the kidneys were removed, and human immune cells in the tissue Subtypes (CD3, CD4, CD8, CD19) were stained with IHC to infiltrate. The excised tissue was fixed with formalin and then embedded in paraffin to create a 5 μm thick section. In order to observe the immune cells in the tissue, immunohistochemical analysis was performed by reacting with human CD3, CD4, CD8, and CD19 antibodies on the section slides.
그 결과, 실시예 1-1의 정상인과 루푸스 환자의 PBMC를 주입한 마우스 신장 조직 내 인간 CD3+T, CD4+T, CD8+T, Cd19+T 세포가 검출되어 인간 세포가 잘 생착된 것을 알 수 있었다(도 3).As a result, human CD3+T, CD4+T, CD8+T, and Cd19+T cells were detected in the mouse kidney tissue injected with the PBMCs of normal persons of Example 1-1 and lupus patients, indicating that human cells were well engrafted. could be (Fig. 3).
<실시예 1-4> 루푸스 환자 모사 아바타 모델에서 자가항체 및 단백뇨 변화 관찰<Example 1-4> Observation of changes in autoantibodies and proteinuria in a simulated avatar model of a lupus patient
상기 일련의 실시예에 따라, 실시예 1-1에서 수립한 루푸스 인간화 마우스 모델에서 인간 세포의 생착 확인을 통해 인간화된 마우스의 시스템이 잘 구축되었음을 확인하였다. 따라서, 정상인과 루푸스 환자의 세포에 따른 루푸스 phenotype에차이가 있는지 확인하기 위하여, 자가 항체 및 단백뇨 변화를 관찰하였다.According to the above series of examples, it was confirmed that the humanized mouse system was well established by confirming the engraftment of human cells in the lupus humanized mouse model established in Example 1-1. Therefore, in order to confirm whether there is a difference in the lupus phenotype according to the cells of normal people and lupus patients, changes in autoantibody and proteinuria were observed.
dsDNA는 마우스의 혈청을 1/20로 희석한 뒤 human anti-double stranded DNA(dsDNA) antibody(IgG) ELISA Kit를 이용하여 혈청 내 dsDNA양을 측정하였다. 또한, 마우스로부터 분리한 urine으로부터 인간 알부민과 크레아틴의 농도 측정은For dsDNA, the amount of dsDNA in the serum was measured using a human anti-double stranded DNA (dsDNA) antibody (IgG) ELISA Kit after diluting mouse serum to 1/20. In addition, the concentration of human albumin and creatine from urine isolated from mice was measured.
mouse albumin ELISA Quantitation kit와 creatinine kit을 사용하여 측정하였다.It was measured using mouse albumin ELISA Quantitation kit and creatinine kit.
구체적으로, 루푸스 인간화 마우스 모델과 정상인 모델의 dsDNA level의 발현 수준을 확인한 결과, 환자의 PBMC주입에 의해 anti-dsDNA level이 루푸스 인간화 마우스 모델에서 증가됨을 확인하였다. 또한, 오줌 내 인간 알부민과 크레아틴(creatine)의 농도를 확인한 결과, 루푸스 인간화 마우스 모델에서 그 농도가 증가됨을 확인하였다.Specifically, as a result of confirming the expression level of the dsDNA level in the humanized lupus mouse model and the normal human model, it was confirmed that the anti-dsDNA level was increased in the humanized lupus mouse model by PBMC injection of the patient. In addition, as a result of confirming the concentrations of human albumin and creatine in the urine, it was confirmed that the concentrations were increased in the humanized lupus mouse model.
또한, 인간 크레아틴(creatine) 수준을 비교하고, 알부민/크레아틴의 수준을 비교한 결과, 모두 루푸스 인간화 마우스 모델에서 증가됨을 확인하였다(도 4)In addition, as a result of comparing human creatine (creatine) levels and comparing the levels of albumin / creatine, it was confirmed that all of them were increased in the lupus humanized mouse model (Fig. 4).
<실시예 1-5> 루푸스 환자 모사 아바타 모델에서 신장의 조직학적 변화 확인<Example 1-5> Confirmation of histological changes in the kidney in the lupus patient simulated avatar model
상기 실시예 1-1에서 수립한 루푸스 인간화 마우스 모델의 신장 조직을 조직학적으로 분석하기 위하여, 실시예 1-1의 루푸스 인간화 마우스의 신장 조직을 H&E(haematoxylin and eosin) 염색하였다. 구체적으로, 적출한 조직은 포르말린으로 고정한 후, 파라핀에 임베딩하여 5㎛ 두께의 절편을 생성하였으며, H&E 염색을위하여 절편 슬라이드를 자일렌(xylene)으로 탈파라핀화하였다. 이 후, 헤마톡실린(Hematoxylin)으로 세포핵을, 에오신(eosin)으로 세포질을 염색하고, periodic acid-Schiff (PAS)를 염색하여 신장 조직의 병리를 비교 분석하였다.In order to histologically analyze the kidney tissue of the humanized lupus mouse model established in Example 1-1, the kidney tissue of the humanized lupus mouse of Example 1-1 was stained with H&E (haematoxylin and eosin). Specifically, the excised tissue was fixed with formalin and then embedded in paraffin to generate a 5 μm thick section, and the section slide was deparaffinized with xylene for H&E staining. Thereafter, the cell nucleus was stained with hematoxylin, the cytoplasm with eosin, and periodic acid-Schiff (PAS) was stained to compare and analyze the pathology of the kidney tissue.
그 결과, PBMC 주입한 루푸스 마우스 모델에서 정상인 대비 염증세포의 침윤As a result, infiltration of inflammatory cells compared to normal in the lupus mouse model injected with PBMC.
이 증가한 것을 확인할 수 있었다(도 5).It was confirmed that this increased (FIG. 5).
<실시예 2> 류마티스 관절염 환자 폐섬유화 동반 질환 모델 평가 플랫폼<Example 2> Model evaluation platform for pulmonary fibrosis comorbidity in rheumatoid arthritis patients
<실시예 2-1> 커들란(Curdlan) 주입을 통한 폐 섬유화를 동반한 류마티스 관절염 모델 제작<Example 2-1> Preparation of rheumatoid arthritis model with lung fibrosis through Curdlan injection
2-1-1 커들란(Curdlan) 주입을 통한 폐 섬유화 동반 류마티스 관절염 동물모델 제작(IL-1Ra KO 모델)2-1-1 Production of an animal model for rheumatoid arthritis with lung fibrosis through Curdlan injection (IL-1Ra KO model)
폐 섬유화가 동반된 류마티스 관절염 동물 모델을 제작하기 위하여, SKG 마우스를 이용하였다. 구체적으로, 상기 SKG 마우스는 T세포 면역 체계의 유전적 결함이 있어 T세포 유발 만성 자가면역질환이 유발하는 것으로 알려져 있다. SKG 수컷 마우스(8주령 이내)에 다음과 같은 대조군 및 실험군을 구성하였다. 대조군은 PBS를 처리하였고, 실험군은 커들란을 6mg/kg로 복강내 주입(Intraperitoneal Injection) 및 3mg/kg 구강 주입을 실험 시작 1주일 전에 한번 주입하는 것으로, 폐 섬유화가 동반된 류마티스 관절염 동물 모델을 구축하였다.To construct an animal model of rheumatoid arthritis with lung fibrosis, SKG mice were used. Specifically, it is known that the SKG mouse has a genetic defect in the T-cell immune system and causes T-cell-induced chronic autoimmune disease. The following control and experimental groups were constructed in SKG male mice (within 8 weeks of age). The control group was treated with PBS, and the experimental group was injected with curdlan at 6 mg/kg intraperitoneal injection and 3 mg/kg oral injection once a week before the start of the experiment. built.
<실시예 2-2> 커들란(Curdlan) 주입을 통한 폐 섬유화 동반 류마티스 관절염 동물 모델의 효과<Example 2-2> Effect of rheumatoid arthritis animal model with lung fibrosis through Curdlan injection
2-2-1 커들란 주입된 폐 섬유화 동반 류마티스 관절염 마우스의 관절염 지수 확인2-2-1 Confirmation of Arthritis Index of Curdlan-Injected Rheumatoid Arthritis Mice with Lung Fibrosis
상기 실시예 2-1에서 구축한 폐 섬유화 동반 류마티스 관절염 SKG 동물 모델 및 대조군에 대한 관절염 지수(arthritis score)를 확인하였다. 관절염 평가에 따른 점수와 기준은 다음과 같다.Arthritis scores were confirmed for the SKG animal model of rheumatoid arthritis with lung fibrosis constructed in Example 2-1 and the control group. The scores and criteria for the evaluation of arthritis are as follows.
-평가 기준--Evaluation standard-
0점: 부종이나 종창이 없다.0 points: No edema or swelling.
1점: 발 또는 발목관절에 국한된 경한 부종과 발적Score 1: Mild swelling and redness confined to the foot or ankle joint
2점: 발목관절에서 족근골(metatarsal)에 걸친 경한 부종과 발적Score 2: Mild swelling and redness from the ankle joint to the metatarsal.
3점: 발목관절에서 족근골에 걸친 중등도의 부종과 발적Score 3: Moderate swelling and redness from the ankle joint to the tarsal bone.
4점: 발목에서 다리 전체에 걸쳐 부종과 발적이 있는 경우4 points: swelling and redness from the ankle to the entire leg
마리당 최고의 관절염 지수는 4점이므로 마우스 1 마리당 최고의 질병 지수는 16 이다.The best arthritic index per mouse is 4 points, so the best disease index per mouse is 16.
그 결과, 폐 섬유화 동반 류마티스 관절염 SKG 동물 모델에서는 375의 값이As a result, in the SKG animal model of rheumatoid arthritis with lung fibrosis, a value of 375 was
나타났고, 대조군에서는 20의 값이 나타남에 따라, 본 발명에서 구축된 동물 모델As the value of 20 appeared in the control group, the animal model constructed in the present invention
에서 관절염 지수가 높음을 확인하여, 류머티스 관절염이 제대로 발현된 동물 모델을 구축함을 확인하였다(도 6).By confirming that the arthritis index was high in the , it was confirmed that an animal model in which rheumatoid arthritis was properly expressed was constructed ( FIG. 6 ).
2-2-2 커들란 주입된 폐 섬유화 동반 류마티스 관절염 마우스의 체중 변화 확인Confirmation of body weight change in rheumatoid arthritis mice with 2-2-2 Curdlan-injected pulmonary fibrosis
상기 실시예 2-1에서 구축한 폐 섬유화 동반 류마티스 관절염 SKG 동물 모델(24주령, 커들란 주입 후 17주)과 대조군 SKG 마우스(24주령)에 대한 마우스 체중 변화를 비교하였다.The mouse weight changes were compared to the SKG animal model of rheumatoid arthritis with lung fibrosis constructed in Example 2-1 (24 weeks old, 17 weeks after curdlan injection) and control SKG mice (24 weeks old).
그 결과, 대조군 SKG 마우스는 277g이고, 폐 섬유화 동반 류마티스 관절염SKG 동물 모델은 158g인 것으로 확인되는 바, 폐 섬유화 동반 류마티스 관절염 SKG 동물 모델의 체중이 감소함을 확인하였다(도 7).As a result, it was confirmed that the control SKG mouse weighed 277 g and the rheumatoid arthritis SKG animal model with lung fibrosis weighed 158 g, confirming that the weight of the rheumatoid arthritis SKG animal model with lung fibrosis decreased ( FIG. 7 ).
2-2-3 커들란 주입된 폐 섬유화 동반 류마티스 관절염 마우스의 염증반응 확인Confirmation of Inflammatory Response in Rheumatoid Arthritis Mice with 2-2-3 Curdlan Injection with Lung Fibrosis
상기 실시예 2-1에서 구축한 폐 섬유화 동반 류마티스 관절염 SKG 동물 모델(24주령, 커들란 주입 후 17주)과 대조군 SKG 마우스(24주령)에 대한 마우스의 장기들에서 염증반응을 확인하였다.Inflammatory responses were confirmed in the organs of the mouse compared to the SKG animal model of rheumatoid arthritis with lung fibrosis constructed in Example 2-1 (24 weeks old, 17 weeks after curdlan injection) and the control SKG mouse (24 weeks old).
구체적으로, 상기 실험군 및 대조군 마우스를 Pet-CT 촬영하여, 생체 내부의각 장기와 조직의 생화학적 및 기능적 변화를 영상으로 관찰하였으며, 장기별 특정관심 영역에 따른 ROI(region of interest) 값을 구하였다. 각 마우스의 심장, 폐,간, 근육, 관절(articulation) 및 척추(vertebra)의 염증에 대하여 SUV(Standardized Uptake Value, 표준섭취계수) 값을 얻었으며, GDF(Growth differentiation factor) 흡수(uptake) 정도에 따른 염증 증가 정도를 평가하였다.Specifically, by Pet-CT imaging of the experimental group and control mice, biochemical and functional changes of each organ and tissue inside the living body were observed as images, and ROI (region of interest) values were obtained according to specific regions of interest for each organ. did. Standardized Uptake Value (SUV) values were obtained for inflammation of the heart, lung, liver, muscle, joint (articulation) and spine (vertebra) of each mouse, and the degree of growth differentiation factor (GDF) uptake The degree of increase in inflammation was evaluated.
그 결과, 대조군 SKG 마우스과 비교하여 심장, 폐, 간, 근육, 관절 및 척추에서 폐 섬유화 동반 류마티스 관절염 SKG 동물 모델의 염증이 증가됨을 확인하였다. 따라서, 본 발명의 동물 모델이 염증 반응이 나타나 폐 섬유화 동반 류마티스관절염이 제대로 나타남을 확인하였다(도 8 및 도 9a-f)As a result, it was confirmed that inflammation in the SKG animal model of rheumatoid arthritis with lung fibrosis was increased in the heart, lung, liver, muscle, joint and spine compared to the control SKG mouse. Therefore, it was confirmed that the animal model of the present invention showed an inflammatory response and properly exhibited rheumatoid arthritis accompanied by pulmonary fibrosis ( FIGS. 8 and 9a-f ).
2-2-4 커들란 주입된 폐 섬유화 동반 류마티스 관절염 마우스의 폐 섬유화에의한 손상 확인Confirmation of damage caused by lung fibrosis in rheumatoid arthritis mice with 2-2-4 curdlan-injected lung fibrosis
상기 실시예 2-1에서 구축한 폐 섬유화 동반 류마티스 관절염 SKG 동물 모델(24주령, 커들란 주입 후 17주)과 대조군 SKG 마우스(24주령)에 대한 마우스의 폐 섬유화에 의한 손상을 확인하였다.Damage caused by lung fibrosis was confirmed in the SKG animal model of rheumatoid arthritis with lung fibrosis constructed in Example 2-1 (24 weeks old, 17 weeks after curdlan injection) and control SKG mice (24 weeks old).
그 결과, 대조군 SKG 마우스과 비교하여 폐 섬유화 동반 류마티스 관절염 SKG 동물 모델에서, 폐 섬유화에 의한 폐의 손상이 확인됨에 따라, 본 발명의 동물모델은 폐 섬유화를 명확하게 발현시킴으로서, 폐 섬유화를 동반한 류마티스 관절염 동물 모델을 구축함을 시사한다(도 10)As a result, as compared to control SKG mice, lung damage due to lung fibrosis was confirmed in the SKG animal model of rheumatoid arthritis with pulmonary fibrosis, and the animal model of the present invention clearly expressed lung fibrosis, resulting in rheumatoid arthritis with lung fibrosis. It is suggested to construct an arthritic animal model ( FIG. 10 ).
<실시예 2-3> 폴리헥사메틸렌 구아디닌(Polyhexamethylene guanidine, PHMG) 주입을 통한 폐 섬유화 동반 류마티스 관절염 동물 모델 제작<Example 2-3> Production of an animal model of rheumatoid arthritis accompanied by lung fibrosis through polyhexamethylene guanidine (PHMG) injection
콜라겐에 의한 류마티스 관절염 동물 모델(CIA)에 폴리헥사메틸렌 구아디닌(PHMG)를 주입하였다. 구체적으로, 감응성을 높이기 위하여 0주차에 면역 처리(Immunization)한 후, 6주차에 1%의 100ul 폴리헥사메틸렌 구아디닌(PHMG)를 기관 내 점적 주입(intratracheal instillation)하였으며, 6주차에 한번만 투여하였고, 이후 마우스를 희생시키고 분석을 수행하였다.Collagen-induced rheumatoid arthritis animal model (CIA) was injected with polyhexamethylene guandinine (PHMG). Specifically, in order to increase the sensitivity, after immunization at week 0, 1% of 100ul polyhexamethylene guanine (PHMG) was intratracheal instillation at week 6, and administered only once at week 6 Then, the mice were sacrificed and the analysis was performed.
<실시예 2-4> 폴리헥사메틸렌 구아디닌(PHMG) 주입을 통한 폐 섬유화 동반 류마티스 관절염 동물 모델의 효과<Example 2-4> Effect of an animal model of rheumatoid arthritis with lung fibrosis through polyhexamethylene guandinine (PHMG) injection
2-4-1 PHMG 폐 섬유화 동반 류마티스 관절염 마우스의 관절염 지수 확인2-4-1 Confirmation of Arthritis Index of Rheumatoid Arthritis Mice with PHMG Lung Fibrosis
상기 실시예 2-3에서 구축한 PHMG 주입 폐 섬유화 동반 류마티스 관절염 동물모델과 대조군 마우스에 대한 관절염 지수를 확인하였다. 구체적으로, 관절염 지수는 상기 2-2-1에 기재된 방법으로 측정하였으며, 류머티스 관절염이 발생한 0주-7주차까지의 관절염 지수를 확인하였다.Arthritis index for the PHMG-injected rheumatoid arthritis animal model with lung fibrosis and control mice constructed in Example 2-3 was confirmed. Specifically, the arthritis index was measured by the method described in 2-2-1 above, and the arthritis index from week 0 to week 7 when rheumatoid arthritis occurred was confirmed.
그 결과, 본 발명에서 구축한 PHMG 주입 폐 섬유화 동반 류마티스 관절염 동물 모델은, 관절염의 붓기는 감소되지만, 류마티스 관절염 동물 모델과 비교하여 관절염 지수가 유지되면서 염증반응이 유발됨에 따라 폐 섬유화가 유발됨을 확인하였다(도 11a)As a result, it was confirmed that the PHMG-injected rheumatoid arthritis animal model with lung fibrosis constructed in the present invention reduces the swelling of arthritis, but as the inflammatory response is induced while the arthritis index is maintained compared to the rheumatoid arthritis animal model, lung fibrosis is induced. (Fig. 11a)
2-4-2 PHMG 폐 섬유화 동반 류마티스 관절염 마우스의 폐 조직 및 무게 변화 확인2-4-2 Confirmation of lung tissue and weight change in rheumatoid arthritis mice with PHMG lung fibrosis
상기 실시예 2-3에서 구축한 PHMG 주입 폐 섬유화 동반 류마티스 관절염 동물모델과 대조군 마우스에 대한 폐 조직에 미치는 영향을 확인하였다.The effect of PHMG injection constructed in Example 2-3 on the lung tissue in the animal model of rheumatoid arthritis accompanied by lung fibrosis and the control mouse was confirmed.
구체적으로, 폐포의 세 기관지화(alveolar bronchiolization)를 통하여, 정상 또는 두꺼워진 폐포 벽을 덮는 기관지 상피와 유사한 세포를 특징으로 하는 이형성 병변으로서, 염증, 화학성 자극 등에 노출된 조건엣의 병리학적 조건에서 발생하는 것이 특징이다.Specifically, through alveolar bronchiolization, it is a dysplastic lesion characterized by cells similar to the bronchial epithelium covering the normal or thickened alveolar wall, in pathological conditions exposed to inflammation, chemical stimuli, etc. It is characterized by occurrence.
본 발명에서 구축한 PHMG 주입 폐 섬유화 동반 류마티스 관절염 동물 모델의폐포의 세 기관지화가 매우 유의적으로 대조군과 비교하여 발생함을 확인하였다(도11b) 또한, 폐의 무게 및 몸무게 대비 폐의 질량을 측정한 결과, 대조군과 비교하여폐의 무게가 증가하고, 몸무게 대비 폐의 질량 역시 증가됨을 확인하였다. 따라서,본 발명에서 구축한 PHMG 주입 폐 섬유화 동반 류마티스 관절염 동물 모델은 폐 섬유화가 진행되어 폐 조직에 영향을 미쳤음을 확인하였다(도 11c).It was confirmed that tribronchialization of the alveoli of the PHMG-injected rheumatoid arthritis animal model with lung fibrosis constructed in the present invention occurred significantly compared to the control group (Fig. 11b). As a result, it was confirmed that the weight of the lungs increased compared to the control group, and the mass of the lungs compared to the body weight also increased. Therefore, the PHMG-injected rheumatoid arthritis animal model constructed in the present invention confirmed that lung fibrosis progressed and affected the lung tissue (FIG. 11c).
2-4-3 PHMG 폐 섬유화 동반 류마티스 관절염 마우스의 폐 조직에서 면역 세포 확인2-4-3 Immune Cell Identification in Lung Tissues from Rheumatoid Arthritis Mice with PHMG Lung Fibrosis
상기 실시예 2-3에서 구축한 PHMG 주입 폐 섬유화 동반 류마티스 관절염 동물모델과 대조군 마우스의 폐 조직에 대하여, 면역세포인 대식세포(macrophage), 호중구(neutrophil), 림프구(lymphocyte)의 증가 정도를 확인하였다.In the lung tissue of the PHMG-injected rheumatoid arthritis animal model with lung fibrosis and control mice constructed in Example 2-3, the degree of increase in immune cells, macrophages, neutrophils, and lymphocytes, was confirmed. did.
그 결과, 본 발명에서 구축한 PHMG 주입 폐 섬유화 동반 류마티스 관절염 동물 모델의 폐 조직에서는 대식세포, 호중구 및 림프구가 모두 유의적으로 증가함을확인하였다(도 12a) 또한, 폐 조직에서의 면역세포의 침강율(infiltration score)을 확인한 결과, 본 발명에서 구축한 동물 모델에서 유의적으로 증가함에 따라, 면역 반응이 유도됨을 확인하였다(도 12b).As a result, it was confirmed that macrophages, neutrophils and lymphocytes all significantly increased in the lung tissue of the PHMG-injected rheumatoid arthritis animal model with lung fibrosis constructed in the present invention (Fig. 12a). As a result of confirming the infiltration score, it was confirmed that the immune response was induced as it significantly increased in the animal model constructed in the present invention ( FIG. 12b ).
2-4-4 PHMG 폐 섬유화 동반 류마티스 관절염 마우스의 면역 세포 조절 확인2-4-4 PHMG Confirmation of Immune Cell Regulation in Rheumatoid Arthritis Mice with Lung Fibrosis
분화를 완료한 T 세포는 그 기능에 따라 크게 1형 보조 세포(Th1)와 2형 보조 세포(Th2)로 구분되며, Th1 세포의 주된 기능은 세포 매개성 면역에 관여하고,Th2 세포는 체액성 면역에 관여한다. Treg 세포는 비정상적으로 활성화된 면세포의 기능을 억제하여 염증 반응을 제어하는 특성이 있고, Th17 세포는 Treg 세포와는 달리 면역질환에서 보이는 염증반응의 최전방에서 관여하여 염증 반응의 신호를 최대화시켜 질병의 진행을 가속화시킨다.T cells that have completed differentiation are largely divided into type 1 helper cells (Th1) and type 2 helper cells (Th2) according to their functions. The main function of Th1 cells is cell-mediated immunity, and Th2 cells are humoral. involved in immunity. Treg cells have the property of controlling the inflammatory response by inhibiting the function of abnormally activated cotton cells, and unlike Treg cells, Th17 cells are involved in the forefront of the inflammatory response seen in immune diseases and maximize the signal of the inflammatory response to disease. accelerate the progress of
상기 실시예 2-1-2에서 구축한 PHMG 주입 폐 섬유화 동반 류마티스 관절염 동물 모델과 대조군 마우스에 대하여, Th17 세포는 IL-17+in CD4+를, Treg 세포는CD25+Foxp+inCD4+를, Th1 세포는 IFN- γ +in CD4+를, Th2 세포는 IL-4+in CD4+ 세포를 확인하였다.For the PHMG-injected rheumatoid arthritis animal model with lung fibrosis and control mice constructed in Example 2-1-2, Th17 cells were IL-17+in CD4+, Treg cells were CD25+Foxp+inCD4+, and Th1 cells were IFN-γ +in CD4+ cells were identified, and Th2 cells were identified as IL-4+in CD4+ cells.
그 결과, PHMG 주입 폐 섬유화 동반 류마티스 관절염 동물 모델염증형 세포인 Th17가 증가되고, 면역 반응을 제어하는 Treg 세포의 수치는 감소됨을 확인함을확인하였다(도 13).As a result, it was confirmed that Th17, an inflammatory cell, was increased, and the level of Treg cells controlling the immune response decreased (FIG. 13).
상기 구축된 폐 섬유화를 동반한 류마티스 관절염 동물 모델은 관절염 지수가 대조군 보다 높고, 마우스의 체중이 감소되는 변화를 확인하였다. 또한, 마우스의 심장, 폐, 간, 근육, 관절 및 척추에서 염증이 증가되고, 폐 섬유화에 의한 폐의 손상이 나타남을 확인하였다. 또한, 폐포의 세 기관지화(alveolar bronchiolization)가 대조군과 비교하여 유의적으로 발생하고, 대식세포(macrophage), 호중구(neutrophil), 림프구(lymphocyte)가 증가되어 면역 반응이 증대되고, 염증성 세포인 Th17 세포가 증가되고, 면역 반응을 제어하는 Treg 세포는 감소되는 특징이 나타남을 확인하였다.In the constructed animal model of rheumatoid arthritis with lung fibrosis, the arthritis index was higher than that of the control group, and it was confirmed that the mouse weight decreased. In addition, it was confirmed that inflammation was increased in the heart, lung, liver, muscle, joint and spine of mice, and damage to the lungs due to pulmonary fibrosis appeared. In addition, alveolar bronchiolization occurred significantly compared to the control group, macrophages, neutrophils, and lymphocytes were increased to increase immune response, and inflammatory cells, Th17 It was confirmed that the cells were increased and the Treg cells controlling the immune response were decreased.
<실시예 3> 통풍 동반 관절염 모델 평가 플랫폼<Example 3> Gout-accompanied arthritis model evaluation platform
<실시예 3-1> 골관절염 환자 모사 통풍 동반 골관절염 모델 제작<Example 3-1> Preparation of osteoarthritis model with gout simulated in osteoarthritis patients
Monosodium Iodoacetate (MIA, I2512, Sigma, Poole, UK)를 주사용 saline에 60mg/ml 농도로 용해하여 실험 개시 당일 (day 0) 조제하였다. 실험개시일에 랫(Wistar rat, 6주령, 180g~220g)에 Isoflurane을 이용하여 호흡 마취를 한 후, MIA를 26.5 gauge needle 을 사용하여 우측 슬관절강내로 infrapatellar ligament를 경유하여 50ul (MIA 3mg/body)를 주사하여 골관절염(MIA)을 유도하였다.Monosodium Iodoacetate (MIA, I2512, Sigma, Poole, UK) was dissolved in saline for injection at a concentration of 60 mg/ml, and prepared on the day of the start of the experiment (day 0). After respiratory anesthesia using isoflurane to rats (Wistar rats, 6 weeks old, 180g~220g) on the day of the start of the experiment, 50ul (MIA 3mg/body) MIA was injected into the right knee joint with a 26.5 gauge needle through the infrapatellar ligament. was injected to induce osteoarthritis (MIA).
상기 유도된 골관절염 마우스에 300 mg/kg 칼륨 옥소네이트(potassium oxonate, PO)를 복강 내 주입하였고, 300 mg/kg 히포잔틴(Hypoxanthine, HX)을 구강 주입하였으며, 이를 주 7회 실험 종료 시점까지 주입하여, 통풍 동반 골관절염 동물 모델을 구축하였다.300 mg/kg potassium oxonate (PO) was intraperitoneally injected into the induced osteoarthritis mice, and 300 mg/kg hypoxanthine (HX) was orally injected, which was injected 7 times a week until the end of the experiment Thus, an animal model of osteoarthritis accompanied by gout was constructed.
<실시예 3-2> 골관절염 환자 모사 통풍 동반 관절염 모델의 통증 정도(Procimal metatasal) 확인<Example 3-2> Confirmation of pain level (Procimal metatasal) of osteoarthritis patient-simulated gout-accompanied arthritis model
상기 실시예 3-1에서 구축한 통풍 동반 골관절염 동물 모델에 대한 통증 정도를 측정하기 위하여, 대조군 및 본 발명의 동물 모델의 Dynamic planter esthesiometer(UgoBasile, Comerio, Itaily)를 사용하였다. Stimulator를 랫 아래쪽에 놓고, 0.5mm 두께의 플라스틱 자극 침(Stimulating microfilament)을 뒷다리 쪽에 위치되도록 조정하고 기계를 작동시키면 자극침이 일정한 속도와 힘으로 뒤쪽 발바닥(Procimal metatasal)을 누르면서, 점차로 누르는 힘을 증가시켜 랫이 자극을 견디지 못하고 발을 뗄 때까지의 시간(sec)과 가해진 힘(g)을 측정하였다. 각 3회 측정하여 평균을 내었다.In order to measure the pain level of the animal model of osteoarthritis accompanied by gout constructed in Example 3-1, a Dynamic planter esthesiometer (UgoBasile, Comerio, Itaily) of the animal model of the control group and the present invention was used. Place the stimulator on the underside of the rat, adjust a 0.5mm thick plastic stimulation needle (Stimulating microfilament) to be positioned on the hind leg, and operate the machine. The time (sec) and applied force (g) until the rat could not withstand the stimulation and released the foot were measured by increasing it. Each measurement was performed three times and averaged.
그 결과, 자극을 견디지 못하고 발을 뗄 때까지의 시간(sec)과 가해진 힘(g)은 정상군과 비교하여 통풍 동반 골관절염 동물 모델은 통증 정도(Procimal metatasal)는 유의적인 차이가 확인되어, 본 발명의 통풍 동반 골관절염 동물 모델은 골관절염 동물 모델(MIA)의 특성인 통증을 그대로 유지하면서 통풍을 동반하도록 동물 모델이 구축됨을 확인하였다(도 14).As a result, compared to the normal group, the time (sec) and force (g) until taking off the foot without enduring the stimulus showed a significant difference in pain level (Procimal metatasal) in the animal model of osteoarthritis accompanied by gout. It was confirmed that the animal model of osteoarthritis accompanied by gout of the present invention was constructed to accompany gout while maintaining the pain characteristic of the animal model of osteoarthritis (MIA) (FIG. 14).
<실시예 3-3> 골관절염 환자 모사 통풍 동반 관절염 모델의 체중 부하(Weight bearing) 정도 확인<Example 3-3> Confirmation of weight bearing degree of osteoarthritis patient simulated gout-accompanied arthritis model
상기 실시예 3-1에서 구축한 통풍 동반 골관절염 동물 모델에 대한 통증 정도를 측정하기 위하여,Incapacitance Meter(IITC, Victory Blvd Woodland Hills, CA, USA)를 사용하여, 오른쪽 뒷다리 무게(Weight on right hind rimb)(%)를 측정하였다. In order to measure the pain level for the osteoarthritis animal model with gout constructed in Example 3-1, using an Incapacitance Meter (IITC, Victory Blvd Woodland Hills, CA, USA), the weight on the right hind limb (Weight on right hind rimb) ) (%) was measured.
그 결과, 통풍 동반 골관절염 동물 모델은 오른쪽 뒷다리 무게(Weight on right hind rimb)(%)의 유의적인 차이가 존재하는 바, 체중 부하의 차이가 있는 것으로 판단되며, 본 발명의 통풍 동반 골관절염 동물 모델은 골관절염 동물 모델(MIA)의 특성인 체중부하의 차이를 그대로 유지하면서 통풍을 동반하도록 동물 모델이 구축됨을 확인하였다(도 15).As a result, it is determined that there is a significant difference in weight on right hind rimb (%) in the animal model of osteoarthritis accompanied by gout, and that there is a difference in weight bearing, and the animal model of osteoarthritis accompanied by gout of the present invention is It was confirmed that the animal model was constructed to accompany gout while maintaining the difference in weight bearing, which is a characteristic of the osteoarthritis animal model (MIA) (FIG. 15).
<실시예 3-4> 골관절염 환자 모사 통풍 동반 관절염 모델의 연골 파괴 및 연골 볼륨 비교에 따른 골다공증 심화 확인<Example 3-4> Confirmation of deepening osteoporosis according to comparison of cartilage destruction and cartilage volume in an osteoarthritis patient simulated gout-accompanied arthritis model
상기 실시예 3-1에서 구축한 통풍 동반 골관절염 동물 모델에 대한 연골 파괴 및 연골 볼륨을 확인하는 것으로, 골다공증 심화 정도를 확인하였다.By confirming the cartilage destruction and cartilage volume in the animal model of osteoarthritis accompanied by gout constructed in Example 3-1, the degree of deepening of osteoporosis was confirmed.
구체적으로, 대조군(WT), 골관절염 동물 모델(MIA), 통풍 동반 골관절염 동물 모델(MIA+PO+HX)의 대퇴골(Femur) 및 경골(Tibia)을 채취하여 인디아 잉크(india ink) 염색 후 미세 컴퓨터단층촬영(micro-CT image)으로 연골의 파괴 정도 및 연골 볼륨을 분석하였다. 해면골(Trabecular bone, TB)(%)은 골다공증이 심화될수록 해면골(TB)가 감소되고, 골밀도(Bone Mineral Density, BMD)(mg/cm3)은 골다공증이 심화될수록 감소되는 지표이다. 또한, 구조 분리(Structure separation, St.sp)(U)는 해면골 사이의 간격으로 골다공증이 발생 시 해면골(TB)의 감소가 일어나게 되면서 구조 분리(St.sp) 가 증가되는 것으로 골다공증 정도를 측정하였다. Specifically, the femur (Femur) and tibia (Tibia) of the control group (WT), osteoarthritis animal model (MIA), and gout-accompanied osteoarthritis animal model (MIA+PO+HX) were collected and stained with India ink and then microcomputerized. The degree of destruction of cartilage and cartilage volume were analyzed by tomography (micro-CT image). Trabecular bone (TB) (%) decreases as osteoporosis intensifies, and bone mineral density (BMD) (mg/cm 3 ) is an indicator that decreases as osteoporosis intensifies. In addition, the structure separation (St.sp) (U) is the gap between the cancellous bones, and when osteoporosis occurs, the decrease in the cancellous bone (TB) occurs. The degree of osteoporosis was measured by increasing structural separation (St.sp).
그 결과, 대조군(WT), 골관절염 동물 모델(MIA)과 비교하여 통풍 동반 골관절염 동물 모델(MIA+PO+HX)에서는 해면골(Trabecular bone, TB)(%) 및 골밀도(Bone Mineral Density, BMD)(mg/cm3)가 감소되었으며, 해면골 사이의 간격이 증가되어 구조 분리(St.sp)가 증가됨을 확인하였다. 따라서, 본 발명의 통풍 동반 골관절염 동물 모델(MIA+PO+HX)은 골관절염 동물 모델(MIA)과 비교하여 통풍이 동반됨에 따라 골다공증이 심화되었음을 확인하였다.As a result, compared with the control group (WT) and the animal model of osteoarthritis (MIA), the trabecular bone (TB) (%) and bone mineral density (BMD) ( mg/cm 3 ) was decreased, and the spacing between cancellous bones was increased, confirming that the structural separation (St.sp) was increased. Therefore, the animal model of osteoarthritis accompanied by gout (MIA+PO+HX) of the present invention confirmed that osteoporosis was aggravated as it was accompanied by gout, compared with the animal model of osteoarthritis (MIA).
<실시에 4> 쇼그렌 질환 침샘 오가노이드 모델 평가 플랫폼<Example 4> Sjogren's disease salivary gland organoid model evaluation platform
<실시예 4-1> 쇼그렌 질환 침샘 오가노이드 모델 플랫폼 구축<Example 4-1> Sjogren's disease salivary gland organoid model platform construction
4-1-1. 쇼그렌 환자 유래 타액샘 상피세포 준비 및 배양4-1-1. Preparation and culture of Sjogren patient-derived salivary gland epithelial cells
쇼그렌 환자 유래 타액샘 상피세포(Patient-derived salivary gland epithelial cell)를 준비하였다. 상기 세포의 배양액을 수득하기 위하여, 배지는 2.5% 태아 소 혈청(Fetal bovine serum)(영인프런티어#US-FBS-500), DMEM/F12(1:3 mixture, Gibco#11330-032), 1% 페니실린-스트렙토마이신(penicillin-streptomycin)(Gibco#15140-122)로 준비하고, 여기에 추가적으로 0.4 ㎍/mL 하이드로코르티손(hydrocortisone)(Sigma#H0888), 10 ng/mL 마우스 EGF (human/mouse 공용, Biolegend#585608), 0.5 ㎍/mL 인슐린(Gibco#12585-014)을 포함시켰다. 상기 배지를 이용하여 쇼그렌 환자 유래 타액샘 상피세포를 PureCol(Advanced biomatrix#5005) 증류수에 20배 희석하여 페트리 디쉬에 넣고 37도 1시간 이상 방치 후 걷어내고 PBS 또는 배지로 세척하여 사용하였다. 1U/mL dispase II solution(Stemcell#07923) 및 2mg/mL 콜라게나아제 IV (collagenase IV)(Gibco#17104-019) 준비하였다. 그 후, 60 mm 페트리 디쉬에 상기 세척한 것을 3 mL 넣고 조직을 올려놓고 최대한 잘게 잘랐다. 37도에서 배양하고, 15분마다 꺼내서 재현탁(resuspension)하였다. 조직이 거의 흐물흐물해지면 튜브에 옮기고 원심분리, 1500 rpm, 5분, 4도의 조건으로 수행 후, 배지에 재현탁하여 37도에서 배양하였다. 상기 쇼그렌 환자 유래 타액샘 상피세포는 90% 태아소혈청과 10% 디메틸술폭시드(dimethyl sulfoxide) 용액 1mL로 재부유하여 동결건조하였다.Patient-derived salivary gland epithelial cells from Sjogren's patients were prepared. To obtain a culture solution of the cells, the medium was 2.5% Fetal bovine serum (Young In Frontier #US-FBS-500), DMEM/F12 (1:3 mixture, Gibco#11330-032), 1% Prepared with penicillin-streptomycin (Gibco#15140-122), additionally 0.4 μg/mL hydrocortisone (Sigma#H0888), 10 ng/mL mouse EGF (human/mouse common, Biolegend#585608), 0.5 μg/mL insulin (Gibco#12585-014). Using the medium, Sjogren's patient-derived salivary gland epithelial cells were diluted 20-fold in PureCol (Advanced biomatrix #5005) distilled water, put in a Petri dish, and left at 37°C for 1 hour or more, then removed and washed with PBS or medium before use. 1U/mL dispase II solution (Stemcell#07923) and 2mg/mL collagenase IV (Gibco#17104-019) were prepared. Thereafter, 3 mL of the washed thing was put in a 60 mm Petri dish, and the tissue was placed on it and cut as finely as possible. Incubated at 37°C, taken out every 15 minutes and resuspended. When the tissue became almost limp, it was transferred to a tube, centrifuged at 1500 rpm, 5 minutes, and 4°C, resuspended in medium, and incubated at 37°C. The salivary gland epithelial cells derived from the Sjogren patient were resuspended in 1 mL of 90% fetal bovine serum and 10% dimethyl sulfoxide solution and freeze-dried.
그 결과, 쇼그렌증후군 환자 유래 타액샘상피세포가 상피 유사 형태(epithelial-like morphology)를 이루며 단일층으로 자라는 모습을 확인할 수 있다(도 17).As a result, it can be confirmed that the salivary gland epithelial cells derived from Sjogren's syndrome patients form an epithelial-like morphology and grow as a single layer (FIG. 17).
또한, 쇼그렌 환자 유래 타액샘 상피세포를 유지하기 위하여 계대 배양하였으며, 세포의 주입은 1 x 105 cells/24 웰 플레이트, 5 x 105 cells/6 웰 플레이트 농도로 이용하였다. PureCol(Advanced biomatrix#5005) 증류수에 20배 희석하여 페트리 디쉬에 넣고 37도 1시간 이상 방치 후 걷어내고 PBS 또는 배지로 세척 후 사용하였다. Media를 걷어 버리고 PBS로 세척 후 0.25% 트립신-EDTA 2mL를 패트리 디쉬에 넣고 37도, 10분 조건으로 배양하였다. 세포 재현탁한 것은 50 mL tube로 옮기고 1000 rpm, 5분간 4도에서 원심분리 하였다. 상층액을 버리고 배양 배지로 재현탁하여 패트리 디쉬에 넣어 배양하였다.In addition, subculture was performed to maintain Sjogren's patient-derived salivary gland epithelial cells, and the cells were injected at a concentration of 1 x 10 5 cells/24 well plate and 5 x 10 5 cells/6 well plate. PureCol (Advanced biomatrix #5005) was diluted 20 times in distilled water, put in a Petri dish, left at 37°C for 1 hour or more, then removed and washed with PBS or medium before use. After removing the media and washing with PBS, 2mL of 0.25% trypsin-EDTA was placed in a Petri dish and incubated at 37°C for 10 minutes. The resuspended cells were transferred to a 50 mL tube and centrifuged at 1000 rpm at 4°C for 5 minutes. The supernatant was discarded, resuspended in culture medium, and cultured in a Petri dish.
4-1-2. 쇼그렌 환자 유래 타액샘 상피세포 유래 오가노이드 제작4-1-2. Production of organoids derived from salivary gland epithelial cells derived from Sjogren patients
오가노이드 배양 배지는 EGF를 포함한 각질 세포-혈청 무 함유 배지(Keratinocyte-Serum Free Media with EGF), 소 뇌하수체 추출물(bovine pituitary extract)(Gibco#37010-022), 0.09mM 염화칼슘(Calcium chloride)(Amresco#E506-1)를 이용하였다. 오가노이드 배양 방법은, 마트리겔(Matrigel matrix)(BD#354234)을 냉장 상태의 K-SFM 배지로 2:1의 비율로 희석하여 아이스에 올린 48 well-plate에 깔고, 이를 37도 세포배양기로 옮겨 1시간 동안 코팅하였다. 상기 4-1-1에서 배양한 쇼그렌 환자 유래 타액샘 상피세포를 37도 에 재현탁하여, 상기 코팅된 것에 5 x 104 cells/500 μL/well로 분주하였다. 3일마다 배지를 교환하면서 오가노이드를 관찰하였다.Organoid culture medium includes Keratinocyte-Serum Free Media with EGF, bovine pituitary extract (Gibco#37010-022), 0.09mM Calcium chloride (Amresco) #E506-1) was used. The organoid culture method is to dilute Matrigel matrix (BD#354234) with refrigerated K-SFM medium at a ratio of 2:1 and place it on a 48 well-plate placed on ice, which is then placed in a 37 degree cell incubator. Transfer and coating for 1 hour. The salivary gland epithelial cells derived from Sjogren's patient cultured in 4-1-1 were resuspended at 37°C, and 5 x 10 4 cells/500 μL/well were dispensed to the coated ones. Organoids were observed while changing the medium every 3 days.
그 결과, 쇼그렌증후군 환자 유래 타액샘상피세포를 이용한 오가노이드는 배양 1 ~ 2일후 스페로이드(spheroid) 형태로 구축되며, 7 ~ 8일 후 duct 형태 오가노이드 및 acinar 형태 오가노이드로 구축됨을 확인하였다(도 18).As a result, it was confirmed that organoids using salivary gland epithelial cells derived from patients with Sjogren's syndrome were constructed in the form of spheroids after 1 to 2 days of culture, and duct organoids and acinar organoids after 7 to 8 days. (Fig. 18).
<실시예 4-2> 쇼그렌 환자 유래 타액샘 상피세포 유래 오가노이드의 아밀라아제 분비능 분석<Example 4-2> Analysis of amylase secretion ability of organoids derived from salivary gland epithelial cells derived from Sjogren patients
본 발명의 상기 실시예 4-1-2에서 제조한 쇼그렌 환자 유래 타액샘 상피세포 유래 오가노이드의 아밀라아제 분비능을 분석하기 위하여, 자극을 달리하여 확인하였다.In order to analyze the amylase secretion ability of the organoids derived from salivary gland epithelial cells derived from Sjogren's patients prepared in Example 4-1-2 of the present invention, different stimuli were used.
구체적으로, 환자 유래 타액샘 오가노이드를 0.09 mM 염화 칼슘(Calcium chloride)를 첨가한 배지로 배양하여 스피로이드를 얻은 다음, 10 mM 이소프로테레놀(Isopreterenol)과 2 mM 염화 칼슘(Calcium chloride)으로 18시간 동안 자극하였다. 그 후, 현미경 촬영 후 상등액을 걷어내고, 세포를 이용하여 아밀라아제 활성을 측정하였다(amylase activity assay kit, abcam#102523).Specifically, the patient-derived salivary gland organoids were cultured in a medium supplemented with 0.09 mM calcium chloride to obtain spiroids, and then with 10 mM isoproterenol and 2 mM calcium chloride. Stimulated for 18 hours. Thereafter, the supernatant was removed after microscopy, and amylase activity was measured using the cells (amylase activity assay kit, abcam#102523).
그 결과, 배양된 오가노이드에 이소프로테레놀 및 염화 칼슘으로 자극하면, 오가노이드 주변으로 단백질이 생성된 모습이 관찰됨을 확인하였다(도 19). 또한, 오가노이드 배양상등액에서 아밀라아제가 매우 현저하게 증가함을 확인하였다(도 20).As a result, it was confirmed that when the cultured organoids were stimulated with isoproterenol and calcium chloride, proteins were observed around the organoids (FIG. 19). In addition, it was confirmed that amylase was very significantly increased in the organoid culture supernatant (FIG. 20).
<실시예 4-3> 쇼그렌 환자 유래 타액샘 상피세포 유래 오가노이드의 아쿠아포린-5(Aquaporin-5), 사이토케라틴-19(Cytokeratin-19), 아밀라아제 A(Amylase A) 발현 공초점현미경 분석<Example 4-3> Confocal microscopic analysis of aquaporin-5, cytokeratin-19, and amylase A expression of organoids derived from salivary gland epithelial cells derived from Sjogren's patient
1차항체로는 토끼 항 인간 아쿠아포린-5 항체(Abcam#ab92320, 1:100희석), 마우스 항 인간 아밀라아제 A 항체(Abcam#ab201450, 1:100희석)를 이용하였다. 2차항체는 Alexa488 공여된 염소 항-토끼 IgG(H+L)(Invitrogen# A32731, 1:500 희석), Alexa594 공여된 염소 항-토끼-마우스 IgG (H+L)(Invitrogen#A32742, 1:500 희석)를 이용하였다. 그 후, DAPI(4',6-Diamidino-2-Phenylindole, Dilactate, Invitrogen#D3571)를 염색한 후, Zeiss LSM700 장비로 공초점 현미경으로 촬영하여, 아쿠아포린-5(Aquaporin-5), 사이토케라틴-19(Cytokeratin-19), 아밀라아제 A(Amylase A)의 세포핵을 관찰하였다.As primary antibodies, rabbit anti-human aquaporin-5 antibody (Abcam#ab92320, diluted 1:100) and mouse anti-human amylase A antibody (Abcam#ab201450, diluted 1:100) were used. Secondary antibodies were Alexa488 donated goat anti-rabbit IgG (H+L) (Invitrogen# A32731, 1:500 dilution), Alexa594 donated goat anti-rabbit-mouse IgG (H+L) (Invitrogen#A32742, 1: 500 dilution) was used. After that, DAPI (4',6-Diamidino-2-Phenylindole, Dilactate, Invitrogen#D3571) was stained, and images were taken with a confocal microscope using Zeiss LSM700 equipment, Aquaporin-5 (Aquaporin-5), Cytokeratin Cell nuclei of -19 (Cytokeratin-19) and amylase A were observed.
그 결과, 오가노이드에서 세포특이적 단백질인 아쿠아포린-5가 발현됨을 확인하였으며, 오가노이드 배양 배지에 첨가된 염화 칼슘에 의해 오가노이드 사이에 분비된 아밀라아제 A가 존재하는 현상을 확인하였다(도 21).As a result, it was confirmed that aquaporin-5, a cell-specific protein, was expressed in the organoids, and it was confirmed that the presence of amylase A secreted between the organoids by calcium chloride added to the organoid culture medium (FIG. 21) ).
<실시예 4-4> 마우스 유래 침샘 줄기 세포의 오가노이드 제작<Example 4-4> Organoid production of mouse-derived salivary gland stem cells
마우스에서 턱밑샘(submandibular)에서 세포를 분리하고, 분리(chopping)작업은 칼슘, 마그네슘, HBSS(Hank'sbalanced saltsolution), 히알루론산 분해 효소(hyaluronidase), 제2형 콜라겐으로 수행하였다. 그 후, 첫번째 strain은 100um로, 두번째 strain은 40um로 단일세포형태로 분리하기 위해 strainer 를 이용하여 sorting 하였다. 그 후, 상기 실시예 4-1과 동일한 조건으로 배양하여 침샘 줄기 세포를 수득하였다. 이를 Y-27632와 함께 오랜 시간 배양하여 오가노이드를 수득하였다.Cells were isolated from the submandibular gland in mice, and the chopping operation was performed with calcium, magnesium, HBSS (Hank's balanced salt solution), hyaluronic acid degrading enzyme (hyaluronidase), and type II collagen. After that, the first strain was 100 μm and the second strain was 40 μm, and sorting was performed using a strainer to isolate single cells. Thereafter, salivary gland stem cells were obtained by culturing under the same conditions as in Example 4-1. This was cultured with Y-27632 for a long time to obtain organoids.
상기 오가노이드에 IL-17을 처리하여 침샘줄기세포에 10, 20, 50ng/ml 농도의 IL-17를 처리하여 손상을 주었다. 그 후, 침샘 줄기세포의 형성률과 침의 주성분인 알파 아밀라아제(alpha amylase)의 생성 정도를 확인하였다.The organoid was treated with IL-17, and the salivary gland stem cells were damaged by treatment with IL-17 at a concentration of 10, 20, or 50 ng/ml. After that, the formation rate of salivary gland stem cells and the production level of alpha amylase, which is the main component of saliva, were confirmed.
그 결과, 또한, 오가노이드 배양 배지에 ROCK inhibitor인 Y-27632을 섞어 배양하면 스페로이드의 크기가 커지는 형태로 구축됨을 확인하였다(도 22). 또한, IL-17 처리에 의해 침샘줄기세포에 손상이 가해짐을 확인하였다(도 22). 또한, 상기 IL-17 처리된 침샘줄기세포의 형성율이 대조군과 비교하여 2배 이상 유의적으로 감소함을 확인하였고, 알파 아밀라아제(alpha amylase) 또한 유의적으로 감소됨을 확인하였다(도 23). 따라서, 본 발명의 마우스 침샘 줄기세포 오가노이드는 쇼그렌증후군의 모델로서 우수함을 확인하였다. As a result, it was also confirmed that when the organoid culture medium was mixed with Y-27632, a ROCK inhibitor, and cultured, the size of the spheroids was increased (FIG. 22). In addition, it was confirmed that the salivary gland stem cells were damaged by the IL-17 treatment (FIG. 22). In addition, it was confirmed that the formation rate of the IL-17-treated salivary gland stem cells was significantly reduced more than two-fold compared to the control group, and it was confirmed that alpha amylase was also significantly reduced ( FIG. 23 ). Therefore, it was confirmed that the mouse salivary gland stem cell organoid of the present invention is excellent as a model for Sjogren's syndrome.
<실시예 4-5> 마우스 유래 침샘 줄기 세포의 오가노이드의 아쿠아포린-5(Aquaporin-5), 나노그(Nanog), 아밀라아제 1(Amylase 1), 케라틴 18(Keratin 18) 발현 확인<Example 4-5> Confirmation of expression of aquaporin-5, nanog, amylase 1, and keratin 18 in organoids of mouse-derived salivary gland stem cells
상기 실시예 4-4에서 제작된 마우스 유래 침샘 줄기 세포의 오가노이드가 쇼그렌증후군의 모델로서 제대로 구축되었는지 확인하기 위하여, 침샘줄기세포의 유전자 마커인 아쿠아포린-5(Aquaporin-5), 케라틴 18(Keratin 18), 나노그(Nanog)의 발현을 확인하였다. 또한, 침 분비 기능에 관여하는 아밀라아제 1(Amylase 1)발현을 확인하였다. 이는 quantitative PCR 을 통하여 각 mRNA의 발현을 확인하였다.In order to confirm that the organoid of the mouse-derived salivary gland stem cells prepared in Example 4-4 was properly constructed as a model for Sjogren's syndrome, aquaporin-5, keratin 18 (genetic markers of salivary gland stem cells) ( Keratin 18), the expression of nanog (Nanog) was confirmed. In addition, the expression of amylase 1 involved in salivary secretion was confirmed. It confirmed the expression of each mRNA through quantitative PCR.
그 결과, 마우스 유래 침샘 줄기 세포의 오가노이드는 아쿠아포린-5(Aquaporin-5), 케라틴 18(Keratin 18), 나노그(Nanog)의 발현이 유의적으로 감소됨을 확인하였다. 또한, 아밀라아제 1(Amylase 1)발현이 감소됨을 확인함에 따라, 본 발명의 마우스 침샘 줄기세포 오가노이드는 쇼그렌증후군의 모델로서 우수함을 확인하였다(도 24a-d).As a result, it was confirmed that the expression of aquaporin-5, keratin 18, and nanog was significantly reduced in mouse-derived salivary gland stem cell organoids. In addition, as it was confirmed that the expression of amylase 1 was reduced, it was confirmed that the mouse salivary gland stem cell organoid of the present invention was excellent as a model for Sjogren's syndrome (FIGS. 24a-d).

Claims (29)

  1. 면역결핍 마우스에 루푸스 질환 환자 유래 PBMC(Peripheral blood mononuclear cell)이 투여된, 인간화된 루푸스(systemic lupus erythematosus) 질환 동물 모델.A humanized animal model of systemic lupus erythematosus, in which immunodeficient mice were administered with PBMCs (peripheral blood mononuclear cells) derived from lupus disease patients.
  2. 제1항에 있어서, 상기 동물 모델은 혈청 내 dsDNA가 증가되고, 오줌 내 알부민 및 크레아틴의 농도가 증가된 것인, 동물 모델.The animal model according to claim 1, wherein the animal model has increased dsDNA in serum and increased concentrations of albumin and creatine in urine.
  3. 면역결핍 마우스에 루푸스 질환 환자로부터 분리한 PBMC를 주입하는 단계를 포함하는, 인간화된 루푸스 질환 동물 모델을 제작하는 방법.A method for producing a humanized lupus disease animal model, comprising injecting PBMC isolated from a lupus disease patient into an immunodeficient mouse.
  4. 제1항의 인간화된 루푸스 동물 모델에 후보 물질을 처리하는 단계;를 포함하는, 루푸스 치료 물질을 스크리닝하는 방법.A method of screening a lupus therapeutic agent, comprising; treating a candidate substance to the humanized lupus animal model of claim 1 .
  5. 류마티스 관절염 발생 동물에 베타 글루칸 또는 살균제가 투여되어 폐 섬유화가 발생된, 폐 섬유화가 동반된 류마티스 관절염 동물 모델.An animal model of rheumatoid arthritis with lung fibrosis, in which pulmonary fibrosis was developed by administration of beta-glucan or a fungicide to an animal with rheumatoid arthritis.
  6. 제5항에 있어서, 상기 베타 클루칸은 커들란(Curdlan)인 것인, 동물 모델.The animal model of claim 5, wherein the beta glucan is Curdlan.
  7. 제5항에 있어서, 상기 살균제는 폴리헥사메틸렌 구아디닌(Polyhexamethylene guanidine, PHMG), 염산폴리헥사메틸렌비구아니드(PHMB), 메칠이소치아졸리논(MIT), 염화에톡시에틸구아디닌(Chloride Ethoxyethyl Guanidine, PGH), 나프탈렌(naphthalene), 프탈레이트(phthalate), 디데실메틸염화암모늄(Didecyl methyl ammonium chloride, DDAC), 벤즈아이소사이아졸리논(BIT), 클로록실레놀(chloroxylenol), 티타늄 다이옥사이드, 징크 옥사이트 및 아세톤으로 이루어진 군에서 선택된 1종 이상인 것인, 동물 모델.According to claim 5, wherein the disinfectant is polyhexamethylene guanidine (Polyhexamethylene guanidine, PHMG), hydrochloric acid polyhexamethylene biguanide (PHMB), methyl isothiazolinone (MIT), ethoxyethyl guanidine chloride ( Chloride Ethoxyethyl Guanidine, PGH), naphthalene, phthalate, Didecyl methyl ammonium chloride (DDAC), benzisocyazolinone (BIT), chloroxylenol, titanium At least one selected from the group consisting of dioxide, zinc oxide and acetone, an animal model.
  8. 제5항에 있어서, 상기 투여는 경구, 정맥, 근육, 피하 및 복강내 주사로 이루어진 군에서 선택된 것인, 동물 모델.The animal model of claim 5, wherein the administration is selected from the group consisting of oral, intravenous, intramuscular, subcutaneous and intraperitoneal injection.
  9. 제8항에 있어서, 상기 경구 투여 및 복강 내 주사는 베타 글루칸 투여 시1:2의 중량 비율로 투여되는 것인, 동물 모델.The animal model according to claim 8, wherein the oral administration and intraperitoneal injection are administered in a weight ratio of 1:2 when beta-glucan is administered.
  10. 제5항에 있어서, 상기 동물 모델은 대식세포(macrophage), 호중구(neutrophil), 림프구(lymphocyte)가 증가되어 면역 반응이 증대된 것인, 동물 모델.The animal model of claim 5, wherein the animal model has an increased immune response by increasing macrophages, neutrophils, and lymphocytes.
  11. 제5항에 있어서, 상기 동물 모델은 염증성 세포인 Th17 세포가 증가되고, 면역 반응을 제어하는 Treg 세포는 감소된 것인, 동물 모델.The animal model according to claim 5, wherein in the animal model, Th17 cells, which are inflammatory cells, are increased, and Treg cells that control the immune response are decreased.
  12. 제5항에 있어서, 상기 폐 섬유화는 류마티스 관절염에서 동반되는 폐 섬유증, 통상성 간질성 폐렴, 비특이성 간질성 폐렴, 염증성 기도 질환 및 기질화 폐렴으로 이루어진 군에서 선택된 1종 이상의 폐 질환인 것인, 동물 모델. The method according to claim 5, wherein the lung fibrosis is at least one lung disease selected from the group consisting of pulmonary fibrosis accompanying rheumatoid arthritis, common interstitial pneumonia, non-specific interstitial pneumonia, inflammatory airway disease, and organized pneumonia. , animal models.
  13. 류마티스 관절염 발생 마우스에 베타 글루칸 또는 살균제를 투여하여 폐 섬유화를 유도하는 단계;를 포함하는, 폐 섬유화가 동반된 류마티스 관절염 동물 모델의 제조 방법.Inducing lung fibrosis by administering beta-glucan or a fungicide to a mouse with rheumatoid arthritis;
  14. 제5항의 폐 섬유화가 동반된 류마티스 관절염 동물에 후보 물질을 처리하는 단계;를 포함하는, 폐 섬유화가 동반된 류마티스 관절염 치료 물질을 스크리닝하는 방법. The method of screening a rheumatoid arthritis treatment material with lung fibrosis, comprising the; treating the candidate substance to the rheumatoid arthritis animal accompanied by pulmonary fibrosis of claim 5.
  15. 골관절염 발생 동물에 칼륨 옥소네이트(potassium oxonate, PO) 또는 히포잔틴(Hypoxanthine, HX)가 투여되어 통풍이 발생된, 통풍이 동반된 골관절염 동물 모델.An animal model of osteoarthritis with gout in which gout was developed by administration of potassium oxonate (PO) or hypoxanthine (HX) to an animal with osteoarthritis.
  16. 제15항에 있어서, 상기 투여는 경구, 정맥, 근육, 피하 및 복강내 주사로 이루어진 군에서 선택된 것인, 동물 모델.The animal model of claim 15 , wherein the administration is selected from the group consisting of oral, intravenous, intramuscular, subcutaneous and intraperitoneal injection.
  17. 제16항에 있어서, 상기 경구 투여는 칼륨 옥소네이트로 투여하고, 복강 내 투여는 히포잔틴을 투여하는 것인, 동물 모델.The animal model according to claim 16, wherein the oral administration is administered with potassium oxonate, and the intraperitoneal administration is hypoxanthine administration.
  18. 제17항에 있어서, 상기 칼륨 옥소네이트 및 히포잔틴는 1:1의 중량 비율로 투여되는 것인, 동물 모델.The animal model according to claim 17, wherein the potassium oxonate and hypoxanthine are administered in a weight ratio of 1:1.
  19. 제15항에 있어서, 상기 동물 모델은 해면골(Trabecular bone, TB) 및 골밀도(Bone Mineral Density, BMD)가 감소되고, 해면골 사이의 간격이 증가되어 구조 분리(St.sp)가 증가되어 골다공증이 대조군과 비교하여 심화된 것인, 동물 모델. 16. The method of claim 15, wherein in the animal model, Trabecular bone (TB) and Bone Mineral Density (BMD) are decreased, and the gap between the cancellous bones is increased to increase structural separation (St.sp) so that osteoporosis is a control group Compared to and intensified, the animal model.
  20. 골관절염 발생 동물에 칼륨 옥소네이트 또는 히포잔틴을 투여하여 통풍을 유도하는 단계;를 포함하는, 통풍이 동반된 골관절염 동물 모델의 제조 방법.Inducing gout by administering potassium oxonate or hypoxanthine to an animal with osteoarthritis; A method for producing an animal model of osteoarthritis accompanied by gout.
  21. 제15항의 통풍이 동반된 골관절염 동물 모델에 후보 물질을 처리하는 단계;를 포함하는, 통풍이 동반된 골관절염 치료 물질을 스크리닝하는 방법. A method of screening for a treatment material for osteoarthritis accompanied by gout, comprising the; treatment of a candidate material to the animal model of osteoarthritis accompanied by gout of claim 15.
  22. 마우스 유래 침샘 줄기세포를 배양하여 수득한 오가노이드에 인터루킨-17(Interleukin-17)이 투여된, 쇼그렌 증후군(Sjogren's syndrome) 오가노이드(organoid) 모델.mouse Interleukin-17 (Interleukin-17) is administered to the organoid obtained by culturing the derived salivary gland stem cells, Sjogren's syndrome organoid model.
  23. 제22항에 있어서, 상기 인터루킨-17(Interleukin-17)는 0-100 ng/ml의 농도로 투여하는 것인, 동물 모델.The animal model of claim 22, wherein the Interleukin-17 is administered at a concentration of 0-100 ng/ml.
  24. 제22항에 있어서, 상기 오가노이드 모델은 아쿠아포린-5(Aquaporin-5), 나노그(Nanog), 아밀라아제 1(Amylase 1) 및 케라틴 18(Keratin 18)의 발현이 저해된 것인, 오가노이드 모델.23. The method of claim 22, wherein the organoid model aquaporin-5 (Aquaporin-5), nanog (Nanog), amylase 1 (Amylase 1) and keratin 18 (Keratin 18) expression of the inhibited, the organoid Model.
  25. 제22항에 있어서, 상기 투여는 IL-17을 1-20 일동안 처리하는 것인, 오가노이드 모델.23. The organoid model of claim 22, wherein said administering treats IL-17 for 1-20 days.
  26. 쇼그렌 환자 유래 타액샘 상피세포(Patient-derived salivary gland epithelial cell)를 포함하는, 쇼그렌증후군(Sjogren's syndrome) 오가노이드(organoid) 모델.Sjogren's syndrome organoid model, including Patient-derived salivary gland epithelial cells.
  27. 마우스 유래 침샘 줄기세포를 배양하여 수득하는 단계; 및mouse culturing and obtaining derived salivary gland stem cells; and
    상기 세포를 배양하여 오가노이드를 생성 후 인터루킨-17(Interleukin-17)이 투여하는 단계를 포함하는, 쇼그렌증후군(Sjogren's syndrome) 오가노이드(organoid) 모델 제작 방법.After culturing the cells to generate an organoid, interleukin-17 (Interleukin-17) is administered, Sjogren's syndrome organoid model production method.
  28. 쇼그렌 환자 유래 타액샘 상피세포를 배양하는 단계를 포함하는, 쇼그렌증후군(Sjogren's syndrome) 오가노이드(organoid) 모델 제작 방법.Sjogren's syndrome (Sjogren's syndrome) organoid model production method comprising the step of culturing the salivary gland epithelial cells derived from the patient.
  29. 제22항 또는 제26항의 쇼그렌증후군(Sjogren's syndrome) 오가노이드(organoid) 모델에 후보물질을 처리하는 단계;를 포함하는, 쇼그렌증후군 치료물질을 스크리닝 하는 방법. The method of claim 22 or claim 26, wherein the Sjogren's syndrome (Sjogren's syndrome) comprising the step of processing a candidate material in the organoid model;
PCT/KR2021/003756 2020-03-27 2021-03-26 Avatar model platform for evaluating autoimmune diseases WO2021194294A1 (en)

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
KR1020200037761A KR20210120705A (en) 2020-03-27 2020-03-27 Lupus patient simulation avatar model evaluation platform
KR1020200037764A KR20210120706A (en) 2020-03-27 2020-03-27 Arthritis with gout model evaluation platform
KR10-2020-0037762 2020-03-27
KR10-2020-0037761 2020-03-27
KR1020200037763A KR20210121366A (en) 2020-03-27 2020-03-27 Rheumatoid arthritis model with lung fibrosis evaluation platfor
KR10-2020-0037763 2020-03-27
KR1020200037762A KR20210121365A (en) 2020-03-27 2020-03-27 Sjogren's disease salivary gland organoid model evaluation platform
KR10-2020-0037764 2020-03-27

Publications (1)

Publication Number Publication Date
WO2021194294A1 true WO2021194294A1 (en) 2021-09-30

Family

ID=77892091

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2021/003756 WO2021194294A1 (en) 2020-03-27 2021-03-26 Avatar model platform for evaluating autoimmune diseases

Country Status (1)

Country Link
WO (1) WO2021194294A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115281152A (en) * 2022-08-12 2022-11-04 浙江中医药大学 Method for constructing mouse lupus encephalopathy model

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR101677945B1 (en) * 2008-03-13 2016-11-29 웰스태트 테러퓨틱스 코포레이션 Compounds and method for reducing uric acid

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR101677945B1 (en) * 2008-03-13 2016-11-29 웰스태트 테러퓨틱스 코포레이션 Compounds and method for reducing uric acid

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
DANIELI ANDRADE, PATRICIA B. REDECHA, MILENA VUKELIC, XIAOPING QING, GIORGIO PERINO, JANE E. SALMON, GLORIA C. KOO: "Engraftment of peripheral blood mononuclear cells from systemic lupus erythematosus and antiphospholipid syndrome patient donors into BALB-RAG-2−/−IL-2Rγ−/− mice: A promising model for studying human disease", ARTHRITIS & RHEUMATISM, vol. 63, no. 9, 1 September 2011 (2011-09-01), US, pages 2764 - 2773, XP055582280, ISSN: 0004-3591, DOI: 10.1002/art.30424 *
HEMRAJ VASHIST, GUPTA AVNEET, UPMANYU NEERAJ: "Anti-Arthritics Activity of Cissampelos pareira Leaves and Stephania glabra Rhizome Ethanolic Extract on Adjuvant and Potassium Oxonate Treated Rat", THE OPEN RHEUMATOLOGY JOURNAL, vol. 13, no. 1, 28 March 2019 (2019-03-28), pages 45 - 52, XP055853417, ISSN: 1874-3129, DOI: 10.2174/1874312901913010045 *
KWON OH CHAN, LEE EUN-JU, CHANG EUN-JU, YOUN JEEHEE, GHANG BYEONGZU, HONG SEOKCHAN, LEE CHANG-KEUN, YOO BIN, KIM YONG-GIL: "IL-17A+GM-CSF+ neutrophils are the major infiltrating cells in interstitial lung disease in an autoimmune arthritis model", FRONTIERS IN IMMUNOLOGY, vol. 9, 1544, 2 July 2018 (2018-07-02), pages 1 - 11, XP055853405, DOI: 10.3389/fimmu.2018.01544 *
NGUYEN CUONG Q, YIN HONGEN, LEE BYUNG HA, CHIORINI JOHN A, PECK AMMON B: "IL17: potential therapeutic target in Sjögren's syndrome using adenovirus-mediated gene transfer", LABORATORY INVESTIGATION, vol. 91, no. 1, 1 January 2011 (2011-01-01), pages 54 - 62, XP055853414, ISSN: 0023-6837, DOI: 10.1038/labinvest.2010.164 *
PRINGLE SARAH, WANG XIAOYAN, VERSTAPPEN GWENNY M. P. J., TERPSTRA JANNEKE H., ZHANG CLARENCE K., HE AIQING, PATEL VISHAL, JONES RH: "Salivary Gland Stem Cells Age Prematurely in Primary Sjögren's Syndrome", ARTHRITIS & RHEUMATOLOGY, vol. 71, no. 1, 1 January 2019 (2019-01-01), US, pages 133 - 142, XP055853407, ISSN: 2326-5191, DOI: 10.1002/art.40659 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115281152A (en) * 2022-08-12 2022-11-04 浙江中医药大学 Method for constructing mouse lupus encephalopathy model
CN115281152B (en) * 2022-08-12 2024-03-12 浙江中医药大学 Method for constructing mouse lupus encephalopathy model

Similar Documents

Publication Publication Date Title
Salinas et al. Fish mucosal immunity: intestine
Sambasivan et al. Distinct regulatory cascades govern extraocular and pharyngeal arch muscle progenitor cell fates
Askary et al. Ancient origin of lubricated joints in bony vertebrates
McDONALD et al. Immune responses to Cryptosporidium muris and Cryptosporidium parvum in adult immunocompetent or immunocompromised (nude and SCID) mice
Ostanin et al. T cell transfer model of chronic colitis: concepts, considerations, and tricks of the trade
Giacomin et al. Epithelial-intrinsic IKKα expression regulates group 3 innate lymphoid cell responses and antibacterial immunity
Ram et al. Fates and targets of male accessory gland proteins in mated female Drosophila melanogaster
King et al. Host–parasite interactions promote disease tolerance to intestinal helminth infection
McCauley et al. Conservation of Pax gene expression in ectodermal placodes of the lamprey
Abraham et al. The zebrafish as a model system for forebrain GnRH neuronal development
WO2021194294A1 (en) Avatar model platform for evaluating autoimmune diseases
Pukhlyakova et al. A cadherin switch marks germ layer formation in the diploblastic sea anemone Nematostella vectensis
WO2015026215A9 (en) Pharmaceutical composition for preventing or treating immune diseases or inflammatory diseases, containing biguanide derivative compound as active ingredient
Yahaya Understanding cellular mechanisms underlying airway epithelial repair: selecting the most appropriate animal models
Marega et al. Cross-talk between inflammation and fibroblast growth factor 10 during organogenesis and pathogenesis: lessons learnt from the lung and other organs
Waldmann et al. The role of Gdf5 in the development of the zebrafish fin endoskeleton
Murata et al. Targeted deletion of liver-expressed Choriogenin L results in the production of soft eggs and infertility in medaka, Oryzias latipes
WO2014109617A1 (en) Composition for preventing or treating autoimmune diseases, containing htra2 as active ingredient
Moreno et al. Local and systemic immune responses to Echinococcus granulosus in experimentally infected dogs
WO2018117647A1 (en) Mutated tau protein fragment and use thereof
KR20210121365A (en) Sjogren&#39;s disease salivary gland organoid model evaluation platform
WO2016088799A1 (en) Non-human mammal
WO2022158882A1 (en) Intestinal organoid immune-control integrated platform for theragnosis of microbiome and immune cell
WO2023224431A1 (en) Pharmaceutical composition for preventing and treating geriatric diseases comprising neural stem cells derived from three-dimensional hypothalamus organoid and use thereof
WO2014189201A1 (en) Composition for preventing or treating autoimmune disease, comprising, as active ingredient, pink1 protein or polynucleotide encoding same

Legal Events

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

Ref document number: 21774343

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 21774343

Country of ref document: EP

Kind code of ref document: A1