WO2022131750A1 - Composition for treating or preventing myopathy, obesity or diabetes - Google Patents

Composition for treating or preventing myopathy, obesity or diabetes Download PDF

Info

Publication number
WO2022131750A1
WO2022131750A1 PCT/KR2021/018973 KR2021018973W WO2022131750A1 WO 2022131750 A1 WO2022131750 A1 WO 2022131750A1 KR 2021018973 W KR2021018973 W KR 2021018973W WO 2022131750 A1 WO2022131750 A1 WO 2022131750A1
Authority
WO
WIPO (PCT)
Prior art keywords
peptide
muscle
mutant
present
amino acids
Prior art date
Application number
PCT/KR2021/018973
Other languages
French (fr)
Korean (ko)
Inventor
최봉근
이성권
이상협
Original Assignee
주식회사 뉴온바이오
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 주식회사 뉴온바이오 filed Critical 주식회사 뉴온바이오
Publication of WO2022131750A1 publication Critical patent/WO2022131750A1/en

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/48Hydrolases (3) acting on peptide bonds (3.4)
    • C12N9/50Proteinases, e.g. Endopeptidases (3.4.21-3.4.25)
    • C12N9/64Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from animal tissue
    • C12N9/6421Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from animal tissue from mammals
    • C12N9/6489Metalloendopeptidases (3.4.24)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/46Hydrolases (3)
    • A61K38/48Hydrolases (3) acting on peptide bonds (3.4)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/46Hydrolases (3)
    • A61K38/48Hydrolases (3) acting on peptide bonds (3.4)
    • A61K38/4886Metalloendopeptidases (3.4.24), e.g. collagenase
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/48Hydrolases (3) acting on peptide bonds (3.4)
    • C12N9/50Proteinases, e.g. Endopeptidases (3.4.21-3.4.25)
    • C12N9/64Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from animal tissue

Definitions

  • the present invention relates to an L1 mutant peptide and a composition for alleviating, inhibiting, preventing, or treating muscle disease, obesity or diabetes comprising the same as an active ingredient, and a method for alleviating, inhibiting, preventing, or treating muscle disease, obesity or diabetes using the L1 mutant peptide , and to the alleviation, inhibition, prevention or treatment of muscle disease, obesity or diabetes of the L1 mutant peptide.
  • Muscle atrophy refers to a decrease in muscle volume, which can be clearly seen in patients suffering from cancer or various immunodeficiency diseases, and patients with extremely limited physical activity due to long-term bed life It is more pronounced in people with limited mobility and the elderly.
  • the progress of aging causes a sarcopenia phenomenon in which muscle strength decreases due to a continuous decrease in muscle volume, which leads to a dangerous result in which the elderly may be exposed to various life accidents.
  • An increase in fat mass along with a decrease in muscle mass is called sarcopenic obesity. If the increase in body fat and muscle atrophy act synergistically, it is estimated that the risk of functional disorders and metabolic disorders in the body will further increase.
  • body mass index BMI
  • WHR waist circumference
  • the present inventors screened for peptides that have the potential to show an effect on muscle reduction in order to develop a therapeutic agent for alleviation, inhibition, prevention or treatment of muscle disease related to muscle loss, and significant inhibition of muscle reduction of the double L1 mutant peptide
  • the activity was found, and the active protein was isolated and specified, and it was confirmed that the specified mutant peptide had significantly superior activity than the currently used therapeutic agent for muscle disease related to muscle loss, and completed the present invention.
  • Another object of the present invention is to provide a pharmaceutical composition for alleviating, inhibiting, preventing, or treating muscle disease comprising an L1 mutant peptide as an active ingredient.
  • Another object of the present invention is to provide a method for alleviating, inhibiting, preventing or treating muscle loss by administering an L1 mutant peptide to a subject in need thereof in an amount effective to alleviate, inhibit, prevent or treat muscle disease.
  • Another object of the present invention is to provide a use of the L1 mutant peptide used for alleviation, inhibition, prevention or treatment of muscle disease.
  • Another object of the present invention is to provide a method for alleviating, inhibiting, preventing or treating obesity by administering an L1 mutant peptide to a subject in need thereof in an amount effective to alleviate, suppress, prevent, or treat obesity.
  • Another object of the present invention is to provide a use of the L1 mutant peptide for use in alleviating, suppressing, preventing or treating obesity.
  • Another object of the present invention is to provide a method for alleviating, inhibiting, preventing or treating diabetes by administering an L1 mutant peptide to a subject in need thereof in an amount effective to alleviate, inhibit, prevent, or treat diabetes.
  • Another object of the present invention is to provide a use of the L1 mutant peptide for use in alleviating, suppressing, preventing or treating diabetes.
  • the present invention provides an L1 mutant peptide and a composition for alleviating, inhibiting, preventing, or treating muscle disease, obesity or diabetes comprising the same as an active ingredient, and alleviating, inhibiting, preventing, or treating muscle disease, obesity or diabetes using the L1 mutant peptide it's about how
  • the present invention provides an L1 mutant peptide.
  • the L1 mutant peptide of the present invention is a mutant peptide of wild-type ADAMTS1 (A disintegrin and metalloproteinase with thrombospondin motifs 1).
  • ADAMTS1 is an enzyme belonging to a disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS), a multidomain extracellular protease family, and was first discovered among 19 ADAMTS family enzymes found in humans. ADAMTS1 is known to inhibit angiogenesis by interacting with vascular endothelial growth factor A.
  • the L1 mutant peptide may be one in which a part of the peptide consisting of the amino acid sequence of SEQ ID NO: 1 is deleted.
  • the L1 mutant peptide of the present invention is a peptide comprising a deletion of one or more amino acids selected from the group consisting of amino acids at positions 295 to 300 of the peptide consisting of the amino acid sequence of SEQ ID NO: 1 can
  • the L1 mutant peptide of the present invention is histidine at position 295, proline at position 296, serine at position 297 of the peptide consisting of the amino acid sequence of SEQ ID NO: 1 ), position 298 isoleucine (Isoleucine), position 299 arginine (Arginine) and position 300 asparagine (Asparagine) may be a peptide comprising a deletion of one or more positions selected from the group consisting of, for example, 295 to It may be a peptide containing a deletion at position 300.
  • the L1 mutant peptide of the present invention is a peptide further comprising a deletion of one or more amino acids selected from the group consisting of amino acids at positions 301 to 312 of the peptide consisting of the amino acid sequence of SEQ ID NO: 1 can be
  • the L1 mutant peptide of the present invention is a peptide further comprising a deletion of one or more amino acids selected from the group consisting of amino acids at positions 291 to 294 of the peptide consisting of the amino acid sequence of SEQ ID NO: 1 can be
  • the L1 mutant peptide of the present invention has a deletion from aspartic acid at position 413 to serine at position 967 of the peptide consisting of the amino acid sequence of SEQ ID NO: 1 It may be a peptide comprising additionally.
  • the L1 mutant peptide of the present invention may be a peptide further comprising a deletion of amino acids at positions 1 to 252 of the peptide consisting of the amino acid sequence of SEQ ID NO: 1.
  • the L1 mutant peptide of the present invention may be a peptide consisting of the amino acid sequence of SEQ ID NO: 2.
  • the present invention provides a pharmaceutical composition for alleviating, inhibiting or treating muscle disease, comprising the L1 mutant peptide as an active ingredient.
  • the L1 mutant peptide is the same as described above.
  • glucocorticoids such as cortisol, a natural hormone, or many synthetic cortisol analogues (including, for example, prednisone, hydrocortisone and dexamethasone) act on the body through the glucocorticoid receptor (GR).
  • Glucocorticoids play an important role in regulating differentiation decisions both in vivo and ex vivo, promoting adipogenesis and inhibiting muscle formation. Androgen administration has been shown to increase muscle mass while decreasing fat mass by affecting body composition.
  • Muscle insufficiency or weakness is one of the most fatal health problems in children. These muscle disorders affect more than 1 in 3000 people with various congenital myopathy and muscular dystrophy, such as Duchenne Muscular Dystrophy (DMD). These disorders are usually associated with genetic or spontaneous gene mutations. Children with these disorders suffer from a wide range of complications. Currently, due to the lack of effective treatment, the mortality rate is high, and there is a need to develop a new treatment strategy for these muscle diseases.
  • DMD Duchenne Muscular Dystrophy
  • Muscle tissue of adult vertebrates is regenerated from stem cells known as satellite cells or muscle stem cells (MuSC). Satellite cells are distributed throughout the muscle tissue, are similarly stationary in the absence of injury or disease, and are located in anatomically defined crevices. In addition to satellite cells, cell types that may contribute to muscle regeneration include, but are not limited to, mesenchymal cells, bone marrow-derived cells, muscle stromal cells, and mesenchymal stem cells.
  • Tissue engineering seeks to repair or replace damaged or diseased tissue in the body by implanting a combination of cells, biomaterial scaffolds, biologically active molecules and genes.
  • the basic premise of this approach is that exogenously introduced cells will improve the rate and extent of tissue repair.
  • muscle diseases include sarcopenia, amyotrophy, cancer cachexia, muscle damage, muscular dystrophy, cardiac atrophy, atony, muscular dystrophy, and muscle degeneration. It may be selected from the group consisting of myasthenia gravis and myasthenia gravis, but is not limited thereto, and includes all muscle diseases related to muscle loss.
  • sarcopenia was first named by Rosenborg in 1989, and the etymology of sarcopenia is derived from the Greek word "sarco” meaning muscle and "penia” meaning reduced.
  • WHO World Health Organization assigned a disease classification code to sarcopenia, and recognized that muscle mass less than normal was an official disease. It refers to the loss of skeletal muscle mass mainly distributed in the extremities, and is a result of the gradual decrease in skeletal muscle mass associated with aging.
  • Amyotrophy is defined as a loss of muscle mass. It is common when a patient is resting, such as when admitted to a hospital, or when movement is restricted. That is, muscle atrophy often occurs in cachexia, which is a state of being inactive or suffering from two or more diseases such as cancer, AIDS, congestive heart failure, chronic obstructive pulmonary disease, renal failure, and severe burns at the same time. This increases the risk of falls and falls, and is known to be associated with a decrease in energy expenditure, which in turn increases the risk of obesity and metabolic diseases.
  • muscular atrophy the results of serious diseases such as cancer, a metabolic disease, are also the causes of muscular atrophy, which has been demonstrated to be induced by an increase in inflammatory cytokines and activation of inflammatory signaling pathways.
  • cytokines the causes of muscular atrophy
  • metabolic changes such as an increase in the influx of fatty acids in muscle tissue and a decrease in fatty acid oxidation occur, and mitochondrial biosynthesis, structure and dysfunction occurs.
  • the oxidative stress and the inflow of fatty acids in the muscle also cause inflammation, which can lead to muscular atrophy.
  • the present invention provides a method for alleviating, inhibiting or treating muscle disease, comprising administering to a subject a pharmaceutical composition comprising an L1 mutant peptide as an active ingredient.
  • the present invention relates to a composition for alleviating, suppressing, preventing, or treating obesity comprising an L1 mutant peptide as an active ingredient.
  • the L1 mutant peptide is the same as described above.
  • the term “obesity” refers to a state in which adipose tissue is excessively accumulated in the body to the extent that it causes an abnormality in health.
  • the increase in adipose tissue mass in the development of obesity may be due to an increase in the size and number of adipocytes.
  • the increase in cell number may be the result of recruitment of pre-adipose cells from a multipotent stem cell population or from a subpopulation of cells resident in mature white adipose tissue (WAT).
  • WAT white adipose tissue
  • Bone marrow-derived mesenchymal stem cells can differentiate into various cell types including fat, muscle, cartilage and bone. With aging, bone marrow adipogenesis accelerates in vivo, while the bone-forming ability of MSCs decreases. It has been suggested that MSC precursors may be differentiated into adipose rather than bone with interrelationships, contributing to age-related body composition changes.
  • Fat redistribution in the elderly is associated with an increased risk of metabolic syndrome, including diabetes, hypertension, dyslipidemia, atherosclerosis, and relatively increased intra-abdominal fat. There is also a decline in muscle performance associated with muscle aging and normal aging, often with a gradual onset of sarcopenia. Although skeletal muscle has the ability to self-renew, this process is not activated in the elderly. Age-related changes within skeletal muscle tissue and the host environment are known to affect the proliferation and fusion of myoblasts in response to injury in elderly animals.
  • the present invention provides a method for alleviating, suppressing or treating obesity, comprising administering to a subject a pharmaceutical composition comprising an L1 mutant peptide as an active ingredient.
  • the present invention provides a composition for alleviating, inhibiting, preventing, or treating diabetes comprising an L1 mutant peptide as an active ingredient.
  • the L1 mutant peptide is the same as described above.
  • diabetes in the present invention includes all types of diabetes, for example, type 1 diabetes, type 2 diabetes, and hereditary diabetes.
  • Type 1 diabetes is insulin-dependent diabetes mellitus, mainly caused by destruction of ⁇ -cells.
  • Type 2 diabetes is non-insulin-dependent diabetes mellitus, caused by insufficient insulin secretion after a meal or by insulin resistance.
  • the present invention provides a method for alleviating, inhibiting or treating diabetes, comprising administering to a subject a pharmaceutical composition comprising an L1 mutant peptide as an active ingredient.
  • the term 'comprising as an active ingredient' means including an amount sufficient to achieve efficacy or activity of the L1 mutant peptide of the present invention.
  • the content of the peptide as an active ingredient in the composition according to the present invention can be appropriately adjusted depending on the type and purpose of use, the patient's condition, the type and severity of symptoms, etc. 1 to 90.9% by weight, 0.001 to 99% by weight, 0.1 to 99% by weight, 1 to 99% by weight, 0.001 to 90% by weight, 0.1 to 90% by weight, 1 to 90% by weight, 0.001 to 80% by weight, 0.1 to 80% by weight, 1 to 80% by weight, 0.001 to 70% by weight or 0.1 to 70% by weight, for example, may be 1 to 70% by weight, but is not limited thereto. can do.
  • composition according to the present invention may be administered to mammals, including humans, by various routes.
  • the administration method may be any method commonly used, for example, may be administered by oral, dermal, intravenous, intramuscular, subcutaneous, etc. routes, and preferably intravenously.
  • composition of the present invention can be administered in oral dosage forms such as powders, granules, tablets, capsules, ointments, suspensions, emulsions, syrups, and aerosols, or parenteral dosage forms in the form of transdermal preparations, suppositories, and sterile injection solutions according to conventional methods, respectively. It can be formulated and used as such.
  • composition of the present invention may further contain adjuvants such as pharmaceutically suitable and physiologically acceptable carriers, excipients, and diluents in addition to the mixed extract.
  • adjuvants such as pharmaceutically suitable and physiologically acceptable carriers, excipients, and diluents in addition to the mixed extract.
  • Carriers, excipients and diluents that may be included in the composition of the present invention include dextrin, crystalline cellulose, lactose, dextrose, sucrose, sorbitol, mannitol, xylitol, erythritol, maltitol, starch, gum acacia, alginate, gelatin, calcium phosphate, calcium silicate, cellulose, methyl cellulose, microcrystalline cellulose, polyvinyl pyrrolidone, water, methylhydroxybenzoate, propylhydroxybenzoate, talc, magnesium stearate and mineral oil.
  • Solid preparations for oral administration include tablets, pills, powders, granules, capsules, etc., and these solid preparations include at least one excipient in the extract, for example, starch, calcium carbonate, sucrose ( sucrose) or lactose, gelatin, etc. may be mixed and prepared.
  • lubricants such as magnesium stearate and talc are also used.
  • Formulations for oral use include suspensions, solutions, emulsions, syrups, etc.
  • various excipients such as wetting agents, sweeteners, fragrances, and preservatives may be included.
  • Formulations for parenteral administration include sterile aqueous solutions, non-aqueous solutions, suspensions, emulsions, freeze-dried preparations, suppositories, transdermal preparations, and the like.
  • Non-aqueous solvents and suspending agents include propylene glycol, polyethylene glycol, vegetable oils such as olive oil, and injectable esters such as ethyl oleate.
  • witepsol macrogol, tween 61, cacao butter, laurin, glycerogelatin, and the like can be used.
  • the pharmaceutical composition of the present invention may be administered alone, but in general, a pharmaceutical carrier selected in consideration of the mode of administration and standard pharmaceutical practice. They may be mixed and administered.
  • the pharmaceutical composition of the present invention may be in the form of a tablet containing starch or lactose, or in the form of a capsule containing alone or an excipient, or an elixir or suspension containing a flavoring or coloring chemical agent. It may be administered orally, orally or sublingually in the form.
  • Such liquid formulations may contain suspending agents (eg, methylcellulose, semisynthetic glycerides such as witepsol or a mixture of apricot kernel oil and PEG-6 esters or PEG-8 and caprylic/capric glyceride mixtures, such as mixtures of glycerides).
  • suspending agents eg, methylcellulose, semisynthetic glycerides such as witepsol or a mixture of apricot kernel oil and PEG-6 esters or PEG-8 and caprylic/capric glyceride mixtures, such as mixtures of glycerides).
  • the dosage of the pharmaceutical composition of the present invention may vary depending on the patient's age, weight, sex, dosage form, health status and disease level, and may be administered in divided doses at regular time intervals according to the judgment of a doctor or pharmacist.
  • the daily dose is 0.1 to 1000 mg/kg, 0.1 to 900 mg/kg, 0.1 to 800 mg/kg, 0.1 to 700 mg/kg, 0.1 to 600 mg/kg, e.g. For example, it may be 0.5 to 500 mg/kg.
  • the above dosage is an example of an average case, and the dosage may be higher or lower depending on individual differences.
  • the daily dosage of the pharmaceutical composition of the present invention is less than the above dosage, a significant effect cannot be obtained, and when it exceeds that, it is not only uneconomical but also undesirable side effects may occur because it is outside the range of the normal dosage. Therefore, it is good to set it as the above range.
  • the present invention relates to a composition for alleviating, inhibiting, preventing, or treating muscle disease, obesity or diabetes comprising an L1 mutant peptide as an active ingredient; a method for alleviating, inhibiting, preventing, or treating muscle disease, obesity or diabetes using an L1 mutant peptide; And it relates to the alleviation, inhibition, prevention or treatment of muscle disease, obesity or diabetes of the L1 mutant peptide.
  • Figure 1a is a photograph showing the result of confirming the expression of the mutant peptide through western blot according to an embodiment of the present invention.
  • Figure 1b is a schematic diagram of a mutant peptide according to an embodiment of the present invention.
  • FIG. 2 is a photograph showing the result of confirming the length of the differentiation-induced myotubes in the control group through Zener staining according to an embodiment of the present invention.
  • FIG. 3 is a photograph showing the result of confirming the length of wild-type differentiation-induced myotubes through Zener staining according to an embodiment of the present invention.
  • FIG. 4 is a photograph showing the result of confirming the length of the L1 mutant differentiation-induced myotube through Zener staining according to an embodiment of the present invention.
  • FIG. 5 is a photograph showing the result of confirming the length of the L3 mutant differentiation-induced myotube through Zener staining according to an embodiment of the present invention.
  • FIG. 6 is a photograph showing the result of confirming the length of the L4 mutant differentiation-induced myotube through Zener staining according to an embodiment of the present invention.
  • FIG. 7 is a photograph showing the result of confirming the length of the differentiation-induced myotube in the control group through immunofluorescence staining according to an embodiment of the present invention.
  • FIG. 8 is a photograph showing the result of confirming the length of the wild-type differentiation-induced myotube through immunofluorescence staining according to an embodiment of the present invention.
  • FIG. 9 is a photograph showing the result of confirming the length of the L1 mutant differentiation-induced myotube through immunofluorescence staining according to an embodiment of the present invention.
  • FIG. 10 is a graph showing the result of confirming the length of differentiation-induced myotubes through immunofluorescence staining according to an embodiment of the present invention.
  • FIG. 11 is a graph showing the results of confirming the thickness of myotubes induced to differentiate through immunofluorescence staining according to an embodiment of the present invention.
  • FIG. 12 is a diagram showing the differentiation result of adipose-derived mesenchymal stem cells into adipocytes according to an embodiment of the present invention.
  • FIG. 13 is a graph showing the result of confirming the degree of adipogenesis according to an embodiment of the present invention.
  • FIG. 14 is a graph showing the measurement result of intracellular glucose uptake according to an embodiment of the present invention.
  • 15 is a graph showing changes in body weight in an animal model with reduced muscle mass according to an embodiment of the present invention.
  • 16 is a graph showing changes in the weight of GA muscle and TA muscle tissue in an animal model with reduced muscle mass according to an embodiment of the present invention.
  • 17 is a graph showing changes in weight of fat and muscle in an animal model with reduced muscle mass according to an embodiment of the present invention.
  • FIG. 18 is a graph showing the results of checking the gene expression levels of MyoD, MyoG, Pax7, and MRF4, which are genes related to muscle differentiation according to an embodiment of the present invention.
  • FIG. 19 is a graph showing the results of confirming the expression levels of MuRF1, Atrogin1, Hes1 genes, which are genes associated with inhibition of muscle differentiation according to an embodiment of the present invention.
  • 20 is a graph showing the result of confirming the length of the myotube differentiation-induced through Zener staining according to an embodiment of the present invention.
  • 21 is a graph showing the results of confirming the activity level of muscle cells according to an embodiment of the present invention.
  • Example 1 C2C12 cell line culture and differentiation induction
  • the myoblast cell line C2C12 cell line (ATCC, CRL-1772), which is mainly used for myocyte differentiation studies, was cultured in DMEM supplemented with 10% fetal bovine serum (FBS), penicillin 100 U/ml and streptomycin 100 ⁇ g/ml. It was cultured in a cell incubator at 37° C. supplied with 5% CO 2 as a medium for use.
  • FBS fetal bovine serum
  • the C2C12 cell line was inoculated in a 24-well cell culture plate at 5 X 10 4 cells/well and cultured in a culture medium.
  • DMEM medium containing 2% horse serum which is a medium for differentiation, to induce differentiation.
  • the differentiation medium was replaced every 3 days, and differentiation was induced in a cell culture medium at 37° C. supplied with 5% CO 2 for a total of 8 days.
  • ADAMTS1 and mutant ADAMTS1 recombinant proteins produced in the CHO-K1 cell line were separated and purified by the method of Example 2 below, and treated together with the induction of differentiation of the C2C12 cell line. It was treated once daily at a concentration of 10, 100 ng/ml.
  • an ADAMTS1 overexpression vector was constructed so that 6x His was placed at the C-terminus of the recombinant protein.
  • the pEF6/V5-His A vector (Invitrogen, V96120) was digested using KpnI and XhoI restriction enzymes.
  • KpnI was inserted at the 5' end of the CDS part of the ADAMTS1 sequence (NM_006988.5) and a sequence recognized as XhoI was inserted at the 3' end, and then T4 DNA ligase was applied to the previously cut vector. It was manufactured by bonding.
  • L1, L2, L3, L4 mutant ADAMTS1 uses wild-type ADAMTS1 as a template, L1 is cleaved from 883 to 900, L2 from 937 to 951, L3 from 1021 to 1038, and L4 from 1084 to 1098 nucleotides and all the mutations were made by removing all sequences after nucleotide 1237.
  • the CHO-K1 cell line is a 6-well cell culture plate using RPMI1640 medium supplemented with 10% Fetal bovine serum (FBS), penicillin 100 U/ml, streptomycin 100 ⁇ g/ml and 25 mM Hepes as a culture medium. was inoculated, and cultured in a 37° C. cell incubator supplied with 5% CO 2 . The next day, the vector was introduced into the cells by mixing Lipofectamine 2000 (Invitrogen, 11668019) with the prepared ADAMTS1 or ADAMTS1 mutant expression vector and treating CHO-K1 cells.
  • FBS Fetal bovine serum
  • blasticidin-S Bovine serum-S
  • blasticidin-S TM blasticidin-S
  • blasticidin-S was first treated to a concentration of 10 ⁇ g/ml. Thereafter, blasticidin-S was mixed and replaced in a new culture medium to a concentration of 10 ⁇ g/ml over a total of 3 times at 4-day intervals.
  • each colony was inoculated into a new cell culture plate and cultured. After confirming whether each colony expresses ADAMTS1 or ADAMTS1 mutant protein through western blot, it was used as a stabilizing cell line.
  • the CHO-K1 stabilized cell line was inoculated into a 100 mm dish, and when the cell density reached 100%, it was exchanged with RPMI1640 medium and cultured for 2 days. After 2 days, only a portion of the upper culture medium was taken from the cultured dish, purified with Ni-NTA agarose (Qiagen, 30210), and recovered, and used in the differentiation induction experiment of the C2C12 cell line. Specifically, the cell culture medium was passed through a gravity chromatography column (Qiagen, 34964) equipped with Ni-NTA agarose and washed with 10 mM imidazole buffer. Washing was repeated 3 times, and then the protein was purified by eluting with 250 mM imidazole buffer.
  • a gravity chromatography column Qiagen, 34964
  • FIGS. 1A and FIG. 1b Taking the culture medium of the ADAMTS1 mutant-overexpressing CHO-K1 stabilized cell line prepared by the method of Example 2, and confirming the expression of the recombinant protein produced by western blotting using an antibody that recognizes 6x-His, the results are shown in FIGS. 1A and FIG. 1b.
  • the L1, L2, L3, and L4 mutations showed a pattern in which the part including the prodomain disappeared and the mature domain was mainly expressed.
  • the remaining mutations except for the L1 mutation showed a significant decrease in protein expression (see FIG. 1a ).
  • the C2C12 cell line was treated with ADAMTS1 or ADAMTS1 mutant recombinant protein while inducing differentiation for 8 days. After the differentiation-induced cells were fixed with 4% paraformaldehyde for 10 minutes, cell membrane permeabilization was performed for 10 minutes using 0.25% Triton X-100.
  • myosin heavy chain (MHC) antibody was mixed with 2% BSA buffer at a ratio of 1:500 and treated for 1 hour.
  • Alexa Fluor® 555 fluorescent antibody was mixed with 2% BSA buffer at a ratio of 1:200 and treated for 1 hour, and the nucleus was stained by DAPI staining.
  • the plate after the staining process was photographed using a fluorescence microscope (Nikon Ts2-FL), and the length of the myotube was measured and statistically processed using the analysis program (Nikon, NIS Elements) provided by the manufacturer. After the analysis was completed, the plate was refrigerated after blocking the light. The results are shown in FIGS. 7 to 11 and Tables 1 to 2 .
  • the C2C12 cell line in which differentiation was induced by treatment with ADAMTS1 wild-type or mutant was longer in thickness and length. It was confirmed that differentiation into Otube. In addition, it was confirmed that the length of the myotube became longer and thicker in the cell line treated with ADAMTS1 L1 mutation compared to the wild type. However, L3 or L4 mutants did not show a better effect than wild-type ADAMTS1 on the length or thickness of myotubes.
  • An adipose-derived mesenchymal stem cell line (ATCC, PCS-500-011) was prepared using 2% fetal bovine serum sold by ATCC, 5 ng/ml of recombinant human epithelial cell factor, and 5 ng/ml of recombinant human fibroblast growth factor. It was cultured in the included mesenchymal stem cell line-only medium (ATCC, PCS-500-030). At the time of cell differentiation, the medium mixed with StemPro medium (Gibco, A10410-01) and the additive (Gibco, A10065-01) provided by the manufacturer was changed and replaced with a new StemPro medium every 3 days for a total of 14 days. Differentiation was induced. While inducing differentiation, ADAMTS1 wild-type or L1 mutant recombinant protein was treated once daily at a concentration of 1 ng/ml or 100 ng/ml.
  • the Oil Red O staining method was used. Specifically, cells that have been differentiated were fixed with 10% formalin and stained with Oil Red O staining solution. After the stained cells were observed under a microscope, the deposited dye was extracted using 100% isopropanol. The extracted dye was quantified by measuring absorbance at a wavelength of 520 nm, and the results are shown in FIGS. 12 and 13 and Table 3.
  • WT 1 (ng/ml) WT 100 (ng/ml) L1 1 (ng/ml) L1 100 (ng/ml) 0.13439 One 0.71642 0.75039 0.67123 0.67372 0.04509 0 0.0447 0.03343 0.15397 0.08999
  • the degree of glucose uptake of the differentiation-induced C2C12 cell line increases under insulin treatment.
  • the C2C12 cell line in which myocyte differentiation was induced by treatment with L1 mutant ADAMTS1 was treated with the wild-type cell line to induce differentiation. It was confirmed that the glucose uptake was further increased compared to .
  • Example 8 Observation of weight recovery in an animal model of muscle loss
  • dexamethasone was administered via intraperitoneal injection at a concentration of 50 mg/kg daily for 12 days.
  • L1 mutant ADAMTS1 was administered by intraperitoneal injection at a concentration of 0.02 ⁇ g/ ⁇ l (Low), 0.2 ⁇ g/ ⁇ l (Mid), and 2 ⁇ g/ ⁇ l (High), respectively. It was administered for a total of 12 days, and the degree of body weight recovery was observed in animal models.
  • Example 9 Observation of TA, GA muscle tissue weight recovery in sarcopenia animal model
  • Example 10 Observation of fat and muscle recovery in muscle loss animal model
  • mice To 12-week-old C57BL/6 mice, dexamethasone was administered by intraperitoneal injection at a concentration of 50 mg/kg every day for 12 days, and at the same time, L1 mutant ADAMTS1 was administered at a concentration of 2 ⁇ g/ ⁇ l by 100 ⁇ l by intraperitoneal injection. After completion of administration, mice were anesthetized through inhalation anesthesia, and muscle mass and body fat mass were analyzed using a dual-energy X-ray absorptiometry (DXA) device.
  • DXA dual-energy X-ray absorptiometry
  • Example 11 Confirmation of increased expression of genes associated with muscle differentiation in an animal model of sarcopenia
  • dexamethasone was administered at a concentration of 50 mg/kg daily for 12 days via intraperitoneal injection, and at the same time, L1 mutant ADAMTS1 was administered at 0.02 ⁇ g/ ⁇ l (Low), 0.2 ⁇ g/ ⁇ l (Mid), and 2 ⁇ g, respectively. It was administered by intraperitoneal injection by 100 ⁇ l at a concentration of / ⁇ l (High).
  • the GA muscle tissue was excised at an autopsy. GA muscle tissue was subjected to homogenization using a pestle in a frozen state, and then RNA was extracted (Qiagen, 74104).
  • the extracted RNA was prepared as cDNA using cDNA synthesis reagent (Promega, A5000).
  • the synthesized cDNA was compared to the relative mRNA expression levels of MyoD, MyoG, Pax7, and MRF4 genes related to muscle differentiation using real-time polymerase chain reaction equipment (Applied Biosystems, Quantstudio3).
  • Example 12 Confirmation of reduced expression of genes associated with inhibition of muscle differentiation in an animal model of muscle loss
  • dexamethasone was administered at a concentration of 50 mg/kg daily for 12 days via intraperitoneal injection, and at the same time, L1 mutant ADAMTS1 was administered at 0.02 ⁇ g/ ⁇ l (Low), 0.2 ⁇ g/ ⁇ l (Mid), and 2 ⁇ g, respectively. It was administered by intraperitoneal injection by 100 ⁇ l at a concentration of / ⁇ l (High).
  • the GA muscle tissue was excised at an autopsy. GA muscle tissue was subjected to homogenization using a pestle in a frozen state, and then RNA was extracted (Qiagen, 74104).
  • cDNA synthesis reagent Promega, A5000.
  • MuRF1, Atrogin1, and Hes1 which are genes related to inhibition of muscle differentiation, were compared using real-time polymerase chain reaction equipment (Applied Biosystems, Quantstudio3).
  • Example 13 Confirmation of increased muscle differentiation in cells (Cancer cachexia model) inducing muscle differentiation inhibition by anticancer agents
  • the C2C12 cell line In a 24-well cell culture plate, the C2C12 cell line, a myoblast cell line, was treated with a cisplatin anticancer drug at a concentration of 50 ⁇ M and a L1 ADAMTS1 mutant recombinant protein at a concentration of 0.1, 1, and 10 ng/ml, respectively, while inducing differentiation for 8 days. Differentiation-induced cells were fixed with methanol for 10 minutes. After sufficient washing with PBS three times, a Zener dyeing reagent dissolved in methanol at a concentration of 2.5 mg/ml was mixed with distilled water 1:1 and dyed for 10 minutes. The stained plate was washed with distilled water, dried and observed under a microscope.
  • the medium was replaced with a serum-free medium and further cultured for 24 hours.
  • the L1 ADAMTS1 mutant protein was treated with 10 ⁇ M of BrdU at concentrations of 0.01, 0.1, 1, 10, and 100 ng/ml, respectively, and reacted for 24 hours.
  • the absorbance was measured at 450 nm using a BrdU fluorescence measurement ELISA kit (Cell signaling technology, 6813S). When the amount of newly synthesized DNA in muscle cells increases, the measured absorbance increases, which can be seen as an increase in muscle cell activity.

Landscapes

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

Abstract

The present invention relates to: an L1 mutant peptide; a composition for alleviating, inhibiting, preventing or treating myopathy, obesity or diabetes, containing same as an active gradient; a method for alleviating, inhibiting, preventing or treating myopathy, obesity or diabetes by using the L1 mutant peptide; and a use, of the L1 mutant peptide, for alleviating, inhibiting, preventing or treating myopathy, obesity or diabetes.

Description

근육질환, 비만 또는 당뇨병 치료, 또는 예방용 조성물Composition for treating or preventing muscle disease, obesity or diabetes
본 특허출원은 2020년 12월 14일에 대한민국 특허청에 제출된 대한민국 특허출원 제10-2020-0174844호에 대하여 우선권을 주장하며, 상기 특허출원의 개시사항은 본 명세서에 참조로서 삽입된다.This patent application claims priority to Korean Patent Application No. 10-2020-0174844 filed with the Korean Intellectual Property Office on December 14, 2020, the disclosure of which is incorporated herein by reference.
본 발명은 L1 돌연변이 펩타이드 및 이를 유효성분으로 포함하는 근육질환, 비만 또는 당뇨병 완화, 억제, 예방, 또는 치료용 조성물, L1 돌연변이 펩타이드를 이용한 근육질환, 비만 또는 당뇨병 완화, 억제, 예방, 또는 치료 방법, 및 L1 돌연변이 펩타이드의 근육질환, 비만 또는 당뇨병의 완화, 억제, 예방 또는 치료 용도에 관한 것이다. The present invention relates to an L1 mutant peptide and a composition for alleviating, inhibiting, preventing, or treating muscle disease, obesity or diabetes comprising the same as an active ingredient, and a method for alleviating, inhibiting, preventing, or treating muscle disease, obesity or diabetes using the L1 mutant peptide , and to the alleviation, inhibition, prevention or treatment of muscle disease, obesity or diabetes of the L1 mutant peptide.
인간의 신체활동 감소는 현대 사회의 산업화로 인한 생활습관 변화의 결과이며 이에 따른 신체적, 정신적 건강 측면의 부정적 영향에 대한 심각성이 제기 된지는 상당히 오래 전부터의 일이다. 신체활동 감소에 따른 운동부족은 비만과 대사증후군을 비롯한 각종 생활 습관병에 주요 원인이 되고, 그에 따른 근육량의 감소를 유발한다. 체내 근육량은 생존에 있어 그 역할이 매우 중요하며, 골격근 부피의 감소는 기초대사량의 감소, 근력의 감소 등으로 이어져 일상적인 신체활동을 유지하는데 상당한 어려움을 겪게 한다.The decrease in human physical activity is a result of lifestyle changes caused by industrialization in modern society, and the seriousness of the negative impact on physical and mental health has been raised for quite some time. Lack of exercise due to decreased physical activity is a major cause of obesity and various lifestyle-related diseases, including metabolic syndrome, resulting in a decrease in muscle mass. Muscle mass in the body plays a very important role in survival, and a decrease in skeletal muscle volume leads to a decrease in basal metabolic rate and a decrease in muscle strength, leading to considerable difficulties in maintaining daily physical activity.
근위축(muscle atrophy)은 근부피가 감소하는 현상을 말하며, 이는 암이나 각종 면역 결핍성 질병을 겪는 환자들에 있어서 뚜렷하게 그 증상을 볼 수 있고, 장기간의 침상 생활로 인해 신체활동이 극히 제한된 환자나 움직임이 제한적인 신체장애인 그리고, 노인들에게 있어서는 더욱 두드러지게 나타난다. 노화의 진행은 지속적인 근부피의 감소로 인해 근력이 감소하는 근감소증(sarcopenia) 현상을 야기하고, 이는 노인들에 있어서 각종 생활 사고에 노출될 수 있는 위험한 결과를 초래한다.Muscle atrophy refers to a decrease in muscle volume, which can be clearly seen in patients suffering from cancer or various immunodeficiency diseases, and patients with extremely limited physical activity due to long-term bed life It is more pronounced in people with limited mobility and the elderly. The progress of aging causes a sarcopenia phenomenon in which muscle strength decreases due to a continuous decrease in muscle volume, which leads to a dangerous result in which the elderly may be exposed to various life accidents.
나이가 들면서, 남녀 모두 체성분의 변화가 발생하게 되고, 이와같은 변화는 체중의 변화와는 무관하게 일어난다. 일반적으로 체성분의 변화는 체중증가에 따른 에너지 균형의 변화 때문으로 판단되며, 이때 나타나는 체성분의 변화는 체지방은 증가하지만, 근육량은 감소하게 된다. 또한, 근육량의 감소는 약 30대부터 서서히 진행되며, 노인에게서 근감소(sarcopenia)는 기능장애, 삶의 질, 의료비용 등에 심각한 영향을 미친다.As people age, changes in body composition occur in both men and women, and these changes occur irrespective of changes in body weight. In general, the change in body composition is judged to be due to a change in energy balance according to weight gain. In addition, the decrease in muscle mass gradually progresses from about 30 years of age, and sarcopenia in the elderly has a serious impact on dysfunction, quality of life, medical costs, and the like.
근육량의 감소와 함께 지방량이 증가하는 것을 가리켜 근감소성 비만이라고 한다. 체지방의 증가와 근감소(muscle atrophy)가 상승적으로 작용하게 되면 체내 기능장애 및 대사 장애의 위험성이 더욱 상승할 것으로 추정된다.An increase in fat mass along with a decrease in muscle mass is called sarcopenic obesity. If the increase in body fat and muscle atrophy act synergistically, it is estimated that the risk of functional disorders and metabolic disorders in the body will further increase.
임상에서의 접근 용이성으로 비만의 지표로 사용되는 체질량지수(BMI)와 허리둘레(WHR)는 연령이나 운동의 영향, 체중감소로 인한 신체구성 성분의 변화 등을 잘 반영하지 못한다는 제한점이 있다. 특히, 신체구성 중에서 근육량은 대사증후군과 관련이 있는 것으로 보고되고 있으며, 모든 사망률의 원인을 예측하는 인자로 제시되고 있다. 최근에는 건강과 관련해서 인슐린 저항성, 당대사, 지질농도 및 혈압에 보다 관련 있는 요인으로 체지방률 증가와 근손실에 관심을 가질 필요성이 대두되고 있다.Due to their ease of access in clinical practice, body mass index (BMI) and waist circumference (WHR), which are used as indicators of obesity, have limitations in that they do not reflect well the effects of age, exercise, and changes in body composition due to weight loss. In particular, muscle mass among body composition is reported to be related to metabolic syndrome, and is suggested as a predictor of the cause of all mortality. Recently, as factors more related to insulin resistance, glucose metabolism, lipid concentration and blood pressure in relation to health, there is a need to pay attention to increase in body fat percentage and muscle loss.
본 명세서 전체에 걸쳐 다수의 논문 및 특허문헌이 참조되고 그 인용이 표시되어 있다. 인용된 논문 및 특허문헌의 개시 내용은 그 전체로서 본 명세서에 참조로 삽입되어 본 발명이 속하는 기술 분야의 수준 및 본 발명의 내용이 보다 명확하게 설명된다.Numerous papers and patent documents are referenced throughout this specification and their citations are indicated. The disclosures of the cited papers and patent documents are incorporated herein by reference in their entirety to more clearly describe the level of the technical field to which the present invention pertains and the content of the present invention.
이에, 본 발명자들은 근육감소와 관련된 근육질환의 완화, 억제, 예방 또는 치료제를 개발하기 위해, 근육감소에 효력을 보일 가능성이 있는 펩타이드들을 대상으로 스크리닝하였으며, 이중 L1 돌연변이 펩타이드의 유의성 있는 근육감소 억제 활성을 발견하였고, 또한, 활성이 있는 단백질을 분리하고 특정하였으며, 상기 특정된 돌연변이 펩타이드가 현재 사용되는 근육감소와 관련된 근육질환의 치료제보다 활성이 월등히 우수한 것을 확인하고 본 발명을 완성하였다. Accordingly, the present inventors screened for peptides that have the potential to show an effect on muscle reduction in order to develop a therapeutic agent for alleviation, inhibition, prevention or treatment of muscle disease related to muscle loss, and significant inhibition of muscle reduction of the double L1 mutant peptide The activity was found, and the active protein was isolated and specified, and it was confirmed that the specified mutant peptide had significantly superior activity than the currently used therapeutic agent for muscle disease related to muscle loss, and completed the present invention.
따라서, 본 발명의 목적은 L1 돌연변이 펩타이드를 제공하는 것이다.Accordingly, it is an object of the present invention to provide an L1 mutant peptide.
본 발명의 다른 목적은 L1 돌연변이 펩타이드를 유효성분으로 포함하는 근육질환 완화, 억제, 예방 또는 치료용 약제학적 조성물을 제공하는 것이다.Another object of the present invention is to provide a pharmaceutical composition for alleviating, inhibiting, preventing, or treating muscle disease comprising an L1 mutant peptide as an active ingredient.
본 발명의 또 다른 목적은 L1 돌연변이 펩타이드를 근육질환 완화, 억제, 예방 또는 치료 유효량으로 이를 필요로 하는 대상에 투여하여 근육감소 완화, 억제, 예방 또는 치료하는 방법을 제공하는 것이다.Another object of the present invention is to provide a method for alleviating, inhibiting, preventing or treating muscle loss by administering an L1 mutant peptide to a subject in need thereof in an amount effective to alleviate, inhibit, prevent or treat muscle disease.
본 발명의 또 다른 목적은 근육질환 완화, 억제, 예방 또는 치료에 사용되는 L1 돌연변이 펩타이드의 용도를 제공하는 것이다.Another object of the present invention is to provide a use of the L1 mutant peptide used for alleviation, inhibition, prevention or treatment of muscle disease.
본 발명의 목적은 L1 돌연변이 펩타이드를 유효성분으로 포함하는 비만 완화, 억제, 예방 또는 치료용 약제학적 조성물을 제공하는 것이다.It is an object of the present invention to provide a pharmaceutical composition for alleviating, suppressing, preventing or treating obesity comprising an L1 mutant peptide as an active ingredient.
본 발명의 다른 목적은 L1 돌연변이 펩타이드를 비만 완화, 억제, 예방 또는 치료 유효량으로 이를 필요로 하는 대상에 투여하여 비만 완화, 억제, 예방 또는 치료하는 방법을 제공하는 것이다.Another object of the present invention is to provide a method for alleviating, inhibiting, preventing or treating obesity by administering an L1 mutant peptide to a subject in need thereof in an amount effective to alleviate, suppress, prevent, or treat obesity.
본 발명의 또 다른 목적은 비만 완화, 억제, 예방 또는 치료에 사용되는 L1 돌연변이 펩타이드의 용도를 제공하는 것이다.Another object of the present invention is to provide a use of the L1 mutant peptide for use in alleviating, suppressing, preventing or treating obesity.
본 발명의 목적은 L1 돌연변이 펩타이드를 유효성분으로 포함하는 당뇨병 완화, 억제, 예방 또는 치료용 약제학적 조성물을 제공하는 것이다.It is an object of the present invention to provide a pharmaceutical composition for alleviating, inhibiting, preventing or treating diabetes comprising an L1 mutant peptide as an active ingredient.
본 발명의 다른 목적은 L1 돌연변이 펩타이드를 당뇨병 완화, 억제, 예방 또는 치료 유효량으로 이를 필요로 하는 대상에 투여하여 당뇨병 완화, 억제, 예방 또는 치료하는 방법을 제공하는 것이다.Another object of the present invention is to provide a method for alleviating, inhibiting, preventing or treating diabetes by administering an L1 mutant peptide to a subject in need thereof in an amount effective to alleviate, inhibit, prevent, or treat diabetes.
본 발명의 또 다른 목적은 당뇨병 완화, 억제, 예방 또는 치료에 사용되는 L1 돌연변이 펩타이드의 용도를 제공하는 것이다.Another object of the present invention is to provide a use of the L1 mutant peptide for use in alleviating, suppressing, preventing or treating diabetes.
본 발명의 다른 목적 및 이점은 하기의 발명의 상세한 설명, 청구범위 및 도면에 의해 보다 명확하게 된다.Other objects and advantages of the present invention will become more apparent from the following detailed description of the invention, claims and drawings.
본 발명은 L1 돌연변이 펩타이드 및 이를 유효성분으로 포함하는 근육질환, 비만 또는 당뇨병 완화, 억제, 예방, 또는 치료용 조성물, 및 L1 돌연변이 펩타이드를 이용한 근육질환, 비만 또는 당뇨병 완화, 억제, 예방, 또는 치료 방법에 관한 것이다. The present invention provides an L1 mutant peptide and a composition for alleviating, inhibiting, preventing, or treating muscle disease, obesity or diabetes comprising the same as an active ingredient, and alleviating, inhibiting, preventing, or treating muscle disease, obesity or diabetes using the L1 mutant peptide it's about how
이하 본 발명을 더욱 자세히 설명하고자 한다.Hereinafter, the present invention will be described in more detail.
본 발명의 일 양태에 따르면, 본 발명은 L1 돌연변이 펩타이드를 제공한다.According to one aspect of the present invention, the present invention provides an L1 mutant peptide.
본 발명의 L1 돌연변이 펩타이드는 야생형 ADAMTS1(A disintegrin and metalloproteinase with thrombospondin motifs 1)의 돌연변이 펩타이드이다.The L1 mutant peptide of the present invention is a mutant peptide of wild-type ADAMTS1 (A disintegrin and metalloproteinase with thrombospondin motifs 1).
ADAMTS1는 다중 도메인 세포외 단백질 분해효소(multidomain extracellular protease) 패밀리인 ADAMTS(a disintegrin and metalloproteinase with thrombospondin motifs)에 속하는 효소로서, 인간에게서 발견되는 19 종의 ADAMTS 패밀리의 효소들 중 가장 먼저 발견되었다. ADAMTS1은 혈관 내피 성장 인자 A(vascular endothelial growth factor A)와 상호 작용하여 혈관 신생을 억제하는 것으로 알려져 있다.ADAMTS1 is an enzyme belonging to a disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS), a multidomain extracellular protease family, and was first discovered among 19 ADAMTS family enzymes found in humans. ADAMTS1 is known to inhibit angiogenesis by interacting with vascular endothelial growth factor A.
본 발명에 있어서 L1 돌연변이 펩타이드는 서열번호 1의 아미노산 서열로 이루어진 펩타이드의 일부가 결실된 것일 수 있다.In the present invention, the L1 mutant peptide may be one in which a part of the peptide consisting of the amino acid sequence of SEQ ID NO: 1 is deleted.
본 발명의 구체적인 일 구현예에 있어서, 본 발명의 L1 돌연변이 펩타이드는 서열번호 1의 아미노산 서열로 이루어진 펩타이드의 295 내지 300번 위치의 아미노산으로 이루어진 군에서 선택된 1개 이상의 아미노산의 결실을 포함하는 펩타이드일 수 있다.In a specific embodiment of the present invention, the L1 mutant peptide of the present invention is a peptide comprising a deletion of one or more amino acids selected from the group consisting of amino acids at positions 295 to 300 of the peptide consisting of the amino acid sequence of SEQ ID NO: 1 can
본 발명의 구체적인 일 구현예에 있어서, 본 발명의 L1 돌연변이 펩타이드는 서열번호 1의 아미노산 서열로 이루어진 펩타이드의 295번 위치 히스티틴 (Histidine), 296번 위치 프롤린 (Proline), 297번 위치 세린 (Serine), 298번 위치 아이소루신 (Isoleucine), 299번 위치 아르기닌 (Arginine) 및 300번 위치 아스파라긴 (Asparagine)으로 이루어진 군에서 선택된 하나 이상의 위치의 결실을 포함하는 펩타이드일 수 있으며, 예를 들어, 295 내지 300번 위치의 결실을 포함하는 펩타이드일 수 있다.In a specific embodiment of the present invention, the L1 mutant peptide of the present invention is histidine at position 295, proline at position 296, serine at position 297 of the peptide consisting of the amino acid sequence of SEQ ID NO: 1 ), position 298 isoleucine (Isoleucine), position 299 arginine (Arginine) and position 300 asparagine (Asparagine) may be a peptide comprising a deletion of one or more positions selected from the group consisting of, for example, 295 to It may be a peptide containing a deletion at position 300.
본 발명의 구체적인 일 구현예에 있어서, 본 발명의 L1 돌연변이 펩타이드는 서열번호 1의 아미노산 서열로 이루어진 펩타이드의 301 내지 312번 위치의 아미노산으로 이루어진 군에서 선택된 1개 이상의 아미노산의 결실을 추가적으로 포함하는 펩타이드일 수 있다. In a specific embodiment of the present invention, the L1 mutant peptide of the present invention is a peptide further comprising a deletion of one or more amino acids selected from the group consisting of amino acids at positions 301 to 312 of the peptide consisting of the amino acid sequence of SEQ ID NO: 1 can be
본 발명의 구체적인 일 구현예에 있어서, 본 발명의 L1 돌연변이 펩타이드는 서열번호 1의 아미노산 서열로 이루어진 펩타이드의 291 내지 294번 위치의 아미노산으로 이루어진 군에서 선택된 1개 이상의 아미노산의 결실을 추가적으로 포함하는 펩타이드일 수 있다. In a specific embodiment of the present invention, the L1 mutant peptide of the present invention is a peptide further comprising a deletion of one or more amino acids selected from the group consisting of amino acids at positions 291 to 294 of the peptide consisting of the amino acid sequence of SEQ ID NO: 1 can be
본 발명의 구체적인 일 구현예에 있어서, 본 발명의 L1 돌연변이 펩타이드는 서열번호 1의 아미노산 서열로 이루어진 펩타이드의 413번 위치의 아스파르트산(Aspartic acid)부터 967번 위치의 세린(Serine)까지의 결실을 추가적으로 포함하는 펩타이드일 수 있다.In a specific embodiment of the present invention, the L1 mutant peptide of the present invention has a deletion from aspartic acid at position 413 to serine at position 967 of the peptide consisting of the amino acid sequence of SEQ ID NO: 1 It may be a peptide comprising additionally.
본 발명의 구체적인 일 구현예에 있어서, 본 발명의 L1 돌연변이 펩타이드는 서열번호 1의 아미노산 서열로 이루어진 펩타이드의 1 내지 252번 위치의 아미노산의 결실을 추가적으로 포함하는 펩타이드일 수 있다.In a specific embodiment of the present invention, the L1 mutant peptide of the present invention may be a peptide further comprising a deletion of amino acids at positions 1 to 252 of the peptide consisting of the amino acid sequence of SEQ ID NO: 1.
본 발명의 구체적인 일 구현예에 있어서, 본 발명의 L1 돌연변이 펩타이드는 서열번호 2의 아미노산 서열로 이루어진 펩타이드일 수 있다.In a specific embodiment of the present invention, the L1 mutant peptide of the present invention may be a peptide consisting of the amino acid sequence of SEQ ID NO: 2.
본 발명의 다른 일 양태에 따르면, 본 발명은 L1 돌연변이 펩타이드를 유효성분으로 포함하는 근육질환 완화, 억제 또는 치료용 약제학적 조성물을 제공한다.According to another aspect of the present invention, the present invention provides a pharmaceutical composition for alleviating, inhibiting or treating muscle disease, comprising the L1 mutant peptide as an active ingredient.
본 발명에 있어서 L1 돌연변이 펩타이드는 기 설명한 바와 동일하다.In the present invention, the L1 mutant peptide is the same as described above.
근육과 지방 조직의 체질량 분포는 개인의 건강에 중대한 영향을 미친다. 지방 세포와 근육 세포 분화의 균형에는 많은 요인이 관여하는 것으로 알려져 있다. 예를 들어, 코르티솔, 천연 호르몬 또는 다수의 합성 코르티솔 유사체 (예를 들어, 프레드니손, 하이드로 코르티손 및 덱사메타손 포함)와 같은 글루코 코르티코이드는 글루코 코르티코이드 수용체(glucocorticoid receptor, GR)를 통해 몸에 작용한다. 글루코 코르티코이드는 생체 내 및 생체 외 둘 다에서 분화 결정을 조절하는 데 중요한 역할을 하며, 지방 생성을 촉진하고 근육 형성을 억제한다. 안드로겐 투여는 체성분에 영향을 주어 지방량을 줄이면서 근육량을 증가시키는 것으로 나타났다.The distribution of body mass in muscle and adipose tissue has a significant impact on an individual's health. It is known that many factors are involved in the balance of adipocyte and muscle cell differentiation. For example, glucocorticoids such as cortisol, a natural hormone, or many synthetic cortisol analogues (including, for example, prednisone, hydrocortisone and dexamethasone) act on the body through the glucocorticoid receptor (GR). Glucocorticoids play an important role in regulating differentiation decisions both in vivo and ex vivo, promoting adipogenesis and inhibiting muscle formation. Androgen administration has been shown to increase muscle mass while decreasing fat mass by affecting body composition.
근육 불충분 또는 기능 약화는 가장 치명적인 아동 건강 문제 중 하나이다. 이러한 근육 질환은 Duchenne Muscular Dystrophy (DMD)와 같은 다양한 선천성 근병증 및 근이영양증 환자에서 3000 명 중 1 명 이상에게 영향을 미친다. 이러한 장애는 일반적으로 유전적 또는 자발적 유전자 돌연변이와 관련이 있다. 이러한 장애를 가진 어린이는 광범위한 합병증으로 고통받고 있다. 현재 효과적인 치료법의 부족으로 인하여 높은 사망률을 나타내고 있으며, 이러한 근육 질환에 대한 새로운 치료 전략 개발이 필요한 실정이다.Muscle insufficiency or weakness is one of the most fatal health problems in children. These muscle disorders affect more than 1 in 3000 people with various congenital myopathy and muscular dystrophy, such as Duchenne Muscular Dystrophy (DMD). These disorders are usually associated with genetic or spontaneous gene mutations. Children with these disorders suffer from a wide range of complications. Currently, due to the lack of effective treatment, the mortality rate is high, and there is a need to develop a new treatment strategy for these muscle diseases.
성체 척추 동물의 근육 조직은 위성 세포 또는 근육 줄기 세포(MuSC)로 알려진 줄기 세포로부터 재생된다. 위성 세포는 근육 조직 전체에 분포되어 있으며, 상해 또는 질병이 없을 때 유사하게 정지되어 있으며, 해부학적으로 정의된 틈새에 위치한다. 위성 세포 이외에, 근육 재생에 기여할 수 있는 세포 유형은 간엽 모세포, 골수 유래 세포, 근육 간질 세포, 중간엽 줄기 세포를 포함하지만, 이에 제한되지는 않는다.Muscle tissue of adult vertebrates is regenerated from stem cells known as satellite cells or muscle stem cells (MuSC). Satellite cells are distributed throughout the muscle tissue, are similarly stationary in the absence of injury or disease, and are located in anatomically defined crevices. In addition to satellite cells, cell types that may contribute to muscle regeneration include, but are not limited to, mesenchymal cells, bone marrow-derived cells, muscle stromal cells, and mesenchymal stem cells.
조직 공학은 세포, 생체 물질 스캐폴드, 생물학적 활성 분자 및 유전자의 조합을 이식함으로써 신체의 손상되거나 병에 걸린 조직을 복구하거나 대체하려고한다. 이 접근법의 기본 전제는 외인적으로 도입된 세포가 조직 복구 속도 및 범위를 개선할 것이라는 것이다. Tissue engineering seeks to repair or replace damaged or diseased tissue in the body by implanting a combination of cells, biomaterial scaffolds, biologically active molecules and genes. The basic premise of this approach is that exogenously introduced cells will improve the rate and extent of tissue repair.
성인 MuSC는 손상되거나 결함이 있는 골격근에 이식되어 근육 섬유를 재구성하고 기능을 향상시켜 잠재적으로 MuSC에 대한 치료적 적용을 제공할 수 있다. 그러나, 이 기술을 번역하는 데있어 주요 장애물은 차별화 결정이 어떻게 결정되는지에 대한 이해가 부족하고 치료상의 이점에 대한 이러한 결정을 제어하고 촉진하는 도구가 개발되지 않은 것이다. 이러한 영역을 발전시키면 근육량이나 기능이 손상된 개인을 위한 다양한 새로운 치료법을 개척하거나 근육과 지방 조직 사이의 불균형을 해결할 수 있다.Adult MuSCs could be transplanted into damaged or defective skeletal muscle to reconstruct muscle fibers and improve function, potentially providing therapeutic applications for MuSCs. However, a major obstacle to translating this technique is the lack of understanding of how differentiation decisions are determined and the lack of developed tools to control and facilitate these decisions of therapeutic benefit. Advancement in these areas could lead to a variety of new treatments for individuals with impaired muscle mass or function, or to address imbalances between muscle and adipose tissue.
본 발명에 있어서 근육질환은 근감소증(Sarcopenia), 근위축증(Amyotrophy), 암악액질(Cancer cachexia), 근손상, 근육퇴행위축증, 심위축증, 긴장감퇴증(atony), 근이영양증(muscular dystrophy), 근육 퇴화증 및 근무력증으로 이루어진 군에서 선택된 것일 수 있으나, 이에 한정되는 것은 아니며, 근육감소와 관련된 근육질환을 모두 포함하는 것이다. In the present invention, muscle diseases include sarcopenia, amyotrophy, cancer cachexia, muscle damage, muscular dystrophy, cardiac atrophy, atony, muscular dystrophy, and muscle degeneration. It may be selected from the group consisting of myasthenia gravis and myasthenia gravis, but is not limited thereto, and includes all muscle diseases related to muscle loss.
근감소증(sarcopenia)라는 이름은 1989년에 로젠 보그에 의해 최초로 명명되었으며, 근감소증의 어원은 그리스에서 기원한 근육을 뜻하는 "sarco"와 감소되어 있다는 뜻의 "penia"가 합성된 단어이다. 2017년 초 세계보건기구(WHO)는 근감소증에 질병분류 코드를 부여하여, 정상보다 근육량이 적은 것을 정식 질환으로 인정하였다. 주로 사지에 분포한 골격근의 감소(loss of skeletal musclemass)를 의미하며, 노화와 연관되어 나타나는 점진적인 골격근 감소의 결과이다. The name sarcopenia was first named by Rosenborg in 1989, and the etymology of sarcopenia is derived from the Greek word "sarco" meaning muscle and "penia" meaning reduced. In early 2017, the World Health Organization (WHO) assigned a disease classification code to sarcopenia, and recognized that muscle mass less than normal was an official disease. It refers to the loss of skeletal muscle mass mainly distributed in the extremities, and is a result of the gradual decrease in skeletal muscle mass associated with aging.
근위축증(amyotrophy)은 근육의 중량이 감소하는 것으로 정의된다. 환자가 병원에 입원했을 때와 같이 안정을 취하는 것이나, 움직임이 제한되는 경우 흔히 나타난다. 즉, 활동적이지 않거나, 암, AIDS, 울혈성 심부전, 만성폐쇄성폐질환, 신부전, 극심한 화상 등과 같은 질환을 동시에 2개 이상 앓는 상태인 악액질(cachexia)에서 흔히 근위축증이 발생한다. 이는 넘어짐과 낙상의 위험을 증가시키고, 에너지 소비를 감소시켜 그 결과로 비만과 대사질환의 위험도를 증가시키는 것과도 연관 있다고 알려져 있다. 근위축증의 원인 중 대사질환인 암과 같은 심각한 질환들의 결과가 근위축증의 원인이 되기도 하는데, 이는 염증성 사이토카인의 증가와 염증 신호경로의 활성화로 인해 유도되는 것으로 증명되었다. 비만한 환경에서도 이와 유사하게 산화스트레스를 유도하는 활성산소증(reactive oxygen species)의 생성이 증가하고, 근육조직 내 지방산의 유입 증가 및 지방산 산화의 감소 등 대사적 변화가 발생하며, 미토콘드리아 생합성, 구조 및 기능에 장애가 발생한다. 이러한 근육 내 산화스트레스, 지방산의 유입 또한 염증을 유발하여 근위축증을 유발할 수 있다. Amyotrophy is defined as a loss of muscle mass. It is common when a patient is resting, such as when admitted to a hospital, or when movement is restricted. That is, muscle atrophy often occurs in cachexia, which is a state of being inactive or suffering from two or more diseases such as cancer, AIDS, congestive heart failure, chronic obstructive pulmonary disease, renal failure, and severe burns at the same time. This increases the risk of falls and falls, and is known to be associated with a decrease in energy expenditure, which in turn increases the risk of obesity and metabolic diseases. Among the causes of muscular atrophy, the results of serious diseases such as cancer, a metabolic disease, are also the causes of muscular atrophy, which has been demonstrated to be induced by an increase in inflammatory cytokines and activation of inflammatory signaling pathways. Similarly, in an obese environment, the generation of reactive oxygen species that induce oxidative stress increases, and metabolic changes such as an increase in the influx of fatty acids in muscle tissue and a decrease in fatty acid oxidation occur, and mitochondrial biosynthesis, structure and dysfunction occurs. The oxidative stress and the inflow of fatty acids in the muscle also cause inflammation, which can lead to muscular atrophy.
본 발명의 또 다른 일 양태에 따르면, 본 발명은 L1 돌연변이 펩타이드를 유효성분으로 포함하는 약제학적 조성물을 대상(subject)에 투여하는 단계를 포함하는 근육질환 완화, 억제 또는 치료 방법을 제공한다.According to another aspect of the present invention, the present invention provides a method for alleviating, inhibiting or treating muscle disease, comprising administering to a subject a pharmaceutical composition comprising an L1 mutant peptide as an active ingredient.
본 발명의 또 다른 일 양태에 따르면, 본 발명은 L1 돌연변이 펩타이드를 유효성분으로 포함하는 비만 완화, 억제, 예방, 또는 치료용 조성물에 관한 것이다.According to another aspect of the present invention, the present invention relates to a composition for alleviating, suppressing, preventing, or treating obesity comprising an L1 mutant peptide as an active ingredient.
본 발명에 있어서 L1 돌연변이 펩타이드는 기 설명한 바와 동일하다.In the present invention, the L1 mutant peptide is the same as described above.
본 명세서에서 사용되는 용어 "비만"은 건강에 이상을 초래할 정도로 지방 조직이 체내에 과잉으로 축적된 상태를 의미한다.As used herein, the term “obesity” refers to a state in which adipose tissue is excessively accumulated in the body to the extent that it causes an abnormality in health.
비만의 발달에서 지방 조직 질량의 증가는 지방 세포의 크기와 수의 증가로 인한 것일 수 있다. 세포 수의 증가는 다 분화능 줄기 세포 집단으로부터 또는 성숙한 백색 지방 조직 (white adipose tissue, WAT)에 상주하는 세포의 하위 집단으로부터 전 지방 세포 (pre-adipose cell)를 동원한 결과일 수 있다. 골수유래 중간엽 줄기 세포 (bone marrow-derived mesenchymal stem cell, BM-MSC)는 지방, 근육, 연골 및 뼈를 포함한 다양한 세포 유형으로 분화될 수 있다. 노화와 함께, 골수 지방 생성은 생체 내에서 가속화되는 반면, MSC의 뼈 형성 능력은 감소한다. MSC 전구체는 상호 관계를 갖는 뼈가 아닌 지방으로 분화되어 연령 관련 체성분 변화에 기여할 수 있다고 제안되었다. The increase in adipose tissue mass in the development of obesity may be due to an increase in the size and number of adipocytes. The increase in cell number may be the result of recruitment of pre-adipose cells from a multipotent stem cell population or from a subpopulation of cells resident in mature white adipose tissue (WAT). Bone marrow-derived mesenchymal stem cells (BM-MSCs) can differentiate into various cell types including fat, muscle, cartilage and bone. With aging, bone marrow adipogenesis accelerates in vivo, while the bone-forming ability of MSCs decreases. It has been suggested that MSC precursors may be differentiated into adipose rather than bone with interrelationships, contributing to age-related body composition changes.
노인의 지방 재분배는 당뇨병, 고혈압, 이상 지질 혈증, 죽상 동맥 경화증 및 비교적 증가된 복부 내 지방을 포함한 대사 증후군의 위험 증가와 관련이 있다. 또한, 근육 노화와 정상적인 노화와 관련된 근육 성능의 저하가 있으며, 종종 근육 감소증이 점진적으로 시작된다. 골격근에는 자체 재생 능력이 있지만, 노인에서는 이 과정이 활성화되지 않는다. 골격근 조직 및 숙주 환경 내에서 연령 관련 변화는 노령 동물의 손상에 대한 반응으로 근 모세포의 증식 및 융합에 영향을 미치는 것으로 알려져 있다.Fat redistribution in the elderly is associated with an increased risk of metabolic syndrome, including diabetes, hypertension, dyslipidemia, atherosclerosis, and relatively increased intra-abdominal fat. There is also a decline in muscle performance associated with muscle aging and normal aging, often with a gradual onset of sarcopenia. Although skeletal muscle has the ability to self-renew, this process is not activated in the elderly. Age-related changes within skeletal muscle tissue and the host environment are known to affect the proliferation and fusion of myoblasts in response to injury in elderly animals.
본 발명의 다른 일 양태에 따르면, 본 발명은 L1 돌연변이 펩타이드를 유효성분으로 포함하는 약제학적 조성물을 대상(subject)에 투여하는 단계를 포함하는 비만 완화, 억제 또는 치료 방법을 제공한다.According to another aspect of the present invention, the present invention provides a method for alleviating, suppressing or treating obesity, comprising administering to a subject a pharmaceutical composition comprising an L1 mutant peptide as an active ingredient.
본 발명의 다른 일 양태에 따르면, 본 발명은 L1 돌연변이 펩타이드를 유효성분으로 포함하는 당뇨병 완화, 억제, 예방, 또는 치료용 조성물을 제공한다.According to another aspect of the present invention, the present invention provides a composition for alleviating, inhibiting, preventing, or treating diabetes comprising an L1 mutant peptide as an active ingredient.
본 발명에 있어서 L1 돌연변이 펩타이드는 기 설명한 바와 동일하다.In the present invention, the L1 mutant peptide is the same as described above.
본 명세서에서 사용되는 용어 '당뇨'는 포도당-불내성(intolerance)을 초래하는 인슐린의 상대적 또는 절대적 부족으로 특징되는 만성질환을 의미한다. 본 발명의 용어 당뇨는 모든 종류의 당뇨병을 포함하며, 예를 들어, 제1형 당뇨, 제2형 당뇨 및 유전성 당뇨를 포함한다. 제1형 당뇨는 인슐린 의존성 당뇨병으로서, β-세포의 파괴에 의해 주로 초래된다. 제2형 당뇨는 인슐린 비의존성 당뇨병으로서, 식사 후 불충분한 인슐린 분비에 의해 초래되거나 또는 인슐린 내성에 의해 초래된다. As used herein, the term 'diabetes' refers to a chronic disease characterized by a relative or absolute lack of insulin resulting in glucose-intolerance. The term diabetes in the present invention includes all types of diabetes, for example, type 1 diabetes, type 2 diabetes, and hereditary diabetes. Type 1 diabetes is insulin-dependent diabetes mellitus, mainly caused by destruction of β-cells. Type 2 diabetes is non-insulin-dependent diabetes mellitus, caused by insufficient insulin secretion after a meal or by insulin resistance.
본 발명의 또 다른 일 양태에 따르면, 본 발명은 L1 돌연변이 펩타이드를 유효성분으로 포함하는 약제학적 조성물을 대상(subject)에 투여하는 단계를 포함하는 당뇨병 완화, 억제 또는 치료 방법을 제공한다.According to another aspect of the present invention, the present invention provides a method for alleviating, inhibiting or treating diabetes, comprising administering to a subject a pharmaceutical composition comprising an L1 mutant peptide as an active ingredient.
본 명세서에서 용어 '유효성분으로 포함하는'이란 본 발명의 L1 돌연변이 펩타이드의 효능 또는 활성을 달성하는 데 충분한 양을 포함하는 것을 의미한다. As used herein, the term 'comprising as an active ingredient' means including an amount sufficient to achieve efficacy or activity of the L1 mutant peptide of the present invention.
본 발명에 따른 조성물 내의 유효성분으로서의 펩타이드의 함량은 사용 형태 및 목적, 환자 상태, 증상의 종류 및 경중 등에 의하여 적절하게 조절할 수 있으며, 고형분 중량 기준으로 0.001 내지 99.9 중량%, 0.1 내지 99.9 중량%, 1 내지 90.9 중량%, 0.001 내지 99 중량%, 0.1 내지 99 중량%, 1 내지 99 중량%, 0.001 내지 90 중량%, 0.1 내지 90 중량%, 1 내지 90 중량%, 0.001 내지 80 중량%, 0.1 내지 80 중량%, 1 내지 80 중량%, 0.001 내지 70 중량% 또는 0.1 내지 70 중량%, 예를 들어, 1 내지 70 중량%일 수 있으나, 이에 한정되는 것은 아니며, 당업자가 적절한 범위 내에서 선택하여 실시할 수 있다. The content of the peptide as an active ingredient in the composition according to the present invention can be appropriately adjusted depending on the type and purpose of use, the patient's condition, the type and severity of symptoms, etc. 1 to 90.9% by weight, 0.001 to 99% by weight, 0.1 to 99% by weight, 1 to 99% by weight, 0.001 to 90% by weight, 0.1 to 90% by weight, 1 to 90% by weight, 0.001 to 80% by weight, 0.1 to 80% by weight, 1 to 80% by weight, 0.001 to 70% by weight or 0.1 to 70% by weight, for example, may be 1 to 70% by weight, but is not limited thereto. can do.
본 발명에 따른 조성물은 인간을 포함하는 포유동물에 다양한 경로로 투여될 수 있다. 투여 방식은 통상적으로 사용되는 모든 방식일 수 있으며, 예를 들어, 경구, 피부, 정맥, 근육, 피하 등의 경로로 투여될 수 있으며, 바람직하게는 정맥으로 투여될 수 있다. The composition according to the present invention may be administered to mammals, including humans, by various routes. The administration method may be any method commonly used, for example, may be administered by oral, dermal, intravenous, intramuscular, subcutaneous, etc. routes, and preferably intravenously.
본 발명의 조성물은 각각 통상의 방법에 따라 산제, 과립제, 정제, 캡슐제, 연고제, 현탁액, 에멀젼, 시럽, 에어로졸 등의 경구형 제형, 또는 경피제, 좌제 및 멸균 주사용액의 형태의 비경구 제형 등으로 제형화하여 사용될 수 있다.The composition of the present invention can be administered in oral dosage forms such as powders, granules, tablets, capsules, ointments, suspensions, emulsions, syrups, and aerosols, or parenteral dosage forms in the form of transdermal preparations, suppositories, and sterile injection solutions according to conventional methods, respectively. It can be formulated and used as such.
본 발명의 조성물은 상기 혼합 추출물 이외에 약제학적으로 적합하고 생리학적으로 허용되는 담체, 부형제 및 희석제 등의 보조제를 추가로 함유하는 것일 수 있다. The composition of the present invention may further contain adjuvants such as pharmaceutically suitable and physiologically acceptable carriers, excipients, and diluents in addition to the mixed extract.
본 발명의 조성물에 포함될 수 있는 담체, 부형제 및 희석제로는 덱스트린, 결정셀룰로오스, 락토즈, 덱스트로즈, 수크로스, 솔비톨, 만니톨, 자일리톨, 에리스리톨, 말티톨, 전분, 아카시아 고무, 알지네이트, 젤라틴, 칼슘 포스페이트, 칼슘 실리케이트, 셀룰로즈, 메틸 셀룰로즈, 미정질 셀룰로스, 폴리비닐 피롤리돈, 물, 메틸히드록시벤조에이트, 프로필히드록시벤조에이트, 탈크, 마그네슘 스테아레이트 및 광물유를 들 수 있다. Carriers, excipients and diluents that may be included in the composition of the present invention include dextrin, crystalline cellulose, lactose, dextrose, sucrose, sorbitol, mannitol, xylitol, erythritol, maltitol, starch, gum acacia, alginate, gelatin, calcium phosphate, calcium silicate, cellulose, methyl cellulose, microcrystalline cellulose, polyvinyl pyrrolidone, water, methylhydroxybenzoate, propylhydroxybenzoate, talc, magnesium stearate and mineral oil.
제제화할 경우에는 보통 사용하는 충진제, 증량제, 결합제, 습윤제, 붕해제, 계면활성제 등의 희석제 또는 부형제를 사용할 수 있다. 경구투여를 위한 고형제제에는 정제, 환제, 산제, 과립제, 캡슐제 등이 포함되며, 이러한 고형 제제는 상기 추출물에 적어도 하나 이상의 부형제, 예를 들면, 전분, 칼슘카보네이트(calcium carbonate), 수크로스(sucrose) 또는 락토오스(lactose), 젤라틴 등을 섞어 조제될 수 있다. 또한, 단순한 부형제 이외에 마그네슘 스테아레이트 탈크 같은 윤활제들도 사용된다. In the case of formulation, diluents or excipients such as commonly used fillers, extenders, binders, wetting agents, disintegrants, and surfactants may be used. Solid preparations for oral administration include tablets, pills, powders, granules, capsules, etc., and these solid preparations include at least one excipient in the extract, for example, starch, calcium carbonate, sucrose ( sucrose) or lactose, gelatin, etc. may be mixed and prepared. In addition to simple excipients, lubricants such as magnesium stearate and talc are also used.
경구를 위한 제제로는 현탁제, 내용액제, 유제, 시럽제 등이 해당되는데 흔히 사용되는 단순희석제인 물, 리퀴드 파라핀 이외에 여러 가지 부형제, 예를 들면 습윤제, 감미제, 방향제, 보존제 등이 포함될 수 있다. Formulations for oral use include suspensions, solutions, emulsions, syrups, etc. In addition to water and liquid paraffin, which are commonly used simple diluents, various excipients such as wetting agents, sweeteners, fragrances, and preservatives may be included.
비경구 투여를 위한 제제에는 멸균된 수용액, 비수성용제, 현탁제, 유제, 동결건조제제, 좌제, 경피제 등이 포함된다. 비수성용제, 현탁제로는 프로필렌 글리콜(propylene glycol), 폴리에틸렌 글리콜, 올리브 오일과 같은 식물성 기름, 에틸올레이트와 같은 주사 가능한 에스테르 등이 사용될 수 있다. Formulations for parenteral administration include sterile aqueous solutions, non-aqueous solutions, suspensions, emulsions, freeze-dried preparations, suppositories, transdermal preparations, and the like. Non-aqueous solvents and suspending agents include propylene glycol, polyethylene glycol, vegetable oils such as olive oil, and injectable esters such as ethyl oleate.
좌제의 제제로는 위텝솔(witepsol), 마크로골, 트윈(tween) 61, 카카오지, 라우린지, 글리세로제라틴 등이 사용될 수 있다.As the preparation of the suppository, witepsol, macrogol, tween 61, cacao butter, laurin, glycerogelatin, and the like can be used.
본 발명의 조성물을 인간에게 적용하는 구체예에 있어서, 본 발명의 약제학적 조성물은 단독으로 투여될 수 있으나, 일반적으로 투여방식과 표준 약제학적 관행(standard phamaceutical practice)을 고려하여 선택된 약제학적 담체와 혼합되어 투여될 수 있다. In the embodiment of applying the composition of the present invention to humans, the pharmaceutical composition of the present invention may be administered alone, but in general, a pharmaceutical carrier selected in consideration of the mode of administration and standard pharmaceutical practice. They may be mixed and administered.
예를 들면, 본 발명의 약제학적 조성물은 전분 또는 락토오즈를 함유하는 정제 형태로, 또는 단독 또는 부형제를 함유하는 캡슐 형태로, 또는 맛을 내거나 색을 띄게 하는 화학 약품을 함유하는 엘릭시르 또는 현탁제 형태로 경구, 구강 내 또는 혀 밑 투여될 수 있다. For example, the pharmaceutical composition of the present invention may be in the form of a tablet containing starch or lactose, or in the form of a capsule containing alone or an excipient, or an elixir or suspension containing a flavoring or coloring chemical agent. It may be administered orally, orally or sublingually in the form.
이러한 액체 제제는 현탁제(예를 들어, 메틸셀룰로오즈, 위텝솔(witepsol)과 같은 반합성 글리세라이드 또는 행인유(apricot kernel oil)와 PEG-6 에스테르의 혼합물 또는 PEG-8과 카프릴릭/카프릭 글리세라이드의 혼합물과 같은 글리세라이드 혼합물)와 같은 약제학적으로 허용 가능한 첨가제와 함께 제형화 될 수 있다.Such liquid formulations may contain suspending agents (eg, methylcellulose, semisynthetic glycerides such as witepsol or a mixture of apricot kernel oil and PEG-6 esters or PEG-8 and caprylic/capric glyceride mixtures, such as mixtures of glycerides).
본 발명의 약제학적 조성물의 투여 용량은 환자의 나이, 몸무게, 성별, 투여형태, 건강상태 및 질환 정도에 따라 달라질 수 있으며, 의사 또는 약사의 판단에 따라 일정 시간간격으로 분할 투여할 수도 있다. The dosage of the pharmaceutical composition of the present invention may vary depending on the patient's age, weight, sex, dosage form, health status and disease level, and may be administered in divided doses at regular time intervals according to the judgment of a doctor or pharmacist.
예를 들어, 유효성분 함량을 기준으로 1일 투여량이 0.1 내지 1000 ㎎/kg, 0.1 내지 900 ㎎/kg, 0.1 내지 800 ㎎/kg, 0.1 내지 700 ㎎/kg, 0.1 내지 600 ㎎/kg, 예를 들어, 0.5 내지 500 ㎎/kg일 수 있다. 상기한 투여량은 평균적인 경우를 예시한 것으로서 개인적인 차이에 따라 그 투여량이 높거나 낮을 수 있다.For example, based on the active ingredient content, the daily dose is 0.1 to 1000 mg/kg, 0.1 to 900 mg/kg, 0.1 to 800 mg/kg, 0.1 to 700 mg/kg, 0.1 to 600 mg/kg, e.g. For example, it may be 0.5 to 500 mg/kg. The above dosage is an example of an average case, and the dosage may be higher or lower depending on individual differences.
본 발명의 약제학적 조성물의 1일 투여량이 상기 투여 용량 미만이면 유의성 있는 효과를 얻을 수 없으며, 그 이상을 초과하는 경우 비경제적일 뿐만 아니라 정상 용량의 범위를 벗어나므로 바람직하지 않은 부작용이 발생할 우려가 있으므로, 상기 범위로 하는 것이 좋다.If the daily dosage of the pharmaceutical composition of the present invention is less than the above dosage, a significant effect cannot be obtained, and when it exceeds that, it is not only uneconomical but also undesirable side effects may occur because it is outside the range of the normal dosage. Therefore, it is good to set it as the above range.
본 발명은 L1 돌연변이 펩타이드를 유효성분으로 포함하는 근육질환, 비만 또는 당뇨병 완화, 억제, 예방, 또는 치료용 조성물; L1 돌연변이 펩타이드를 이용한 근육질환, 비만 또는 당뇨병 완화, 억제, 예방, 또는 치료 방법; 및 L1 돌연변이 펩타이드의 근육질환, 비만 또는 당뇨병의 완화, 억제, 예방 또는 치료 용도에 관한 것이다.The present invention relates to a composition for alleviating, inhibiting, preventing, or treating muscle disease, obesity or diabetes comprising an L1 mutant peptide as an active ingredient; a method for alleviating, inhibiting, preventing, or treating muscle disease, obesity or diabetes using an L1 mutant peptide; And it relates to the alleviation, inhibition, prevention or treatment of muscle disease, obesity or diabetes of the L1 mutant peptide.
도 1a는 본 발명의 일 실시예에 따라 웨스턴블랏을 통해 돌연변이 펩타이드의 발현을 확인한 결과를 보여주는 사진이다.Figure 1a is a photograph showing the result of confirming the expression of the mutant peptide through western blot according to an embodiment of the present invention.
도 1b는 본 발명의 일 실시예에 따른 돌연변이 펩타이드의 모식도이다.Figure 1b is a schematic diagram of a mutant peptide according to an embodiment of the present invention.
도 2는 본 발명의 일 실시예에 따라 제너 염색법을 통해 대조군의 분화 유도된 마이오튜브의 길이를 확인한 결과를 보여주는 사진이다.2 is a photograph showing the result of confirming the length of the differentiation-induced myotubes in the control group through Zener staining according to an embodiment of the present invention.
도 3은 본 발명의 일 실시예에 따라 제너 염색법을 통해 야생형의 분화 유도된 마이오튜브의 길이를 확인한 결과를 보여주는 사진이다.3 is a photograph showing the result of confirming the length of wild-type differentiation-induced myotubes through Zener staining according to an embodiment of the present invention.
도 4는 본 발명의 일 실시예에 따라 제너 염색법을 통해 L1 돌연변이의 분화 유도된 마이오튜브의 길이를 확인한 결과를 보여주는 사진이다.4 is a photograph showing the result of confirming the length of the L1 mutant differentiation-induced myotube through Zener staining according to an embodiment of the present invention.
도 5는 본 발명의 일 실시예에 따라 제너 염색법을 통해 L3 돌연변이의 분화 유도된 마이오튜브의 길이를 확인한 결과를 보여주는 사진이다.5 is a photograph showing the result of confirming the length of the L3 mutant differentiation-induced myotube through Zener staining according to an embodiment of the present invention.
도 6은 본 발명의 일 실시예에 따라 제너 염색법을 통해 L4 돌연변이의 분화 유도된 마이오튜브의 길이를 확인한 결과를 보여주는 사진이다.6 is a photograph showing the result of confirming the length of the L4 mutant differentiation-induced myotube through Zener staining according to an embodiment of the present invention.
도 7은 본 발명의 일 실시예에 따라 면역형광염색법을 통해 대조군의 분화 유도된 마이오튜브의 길이를 확인한 결과를 보여주는 사진이다.7 is a photograph showing the result of confirming the length of the differentiation-induced myotube in the control group through immunofluorescence staining according to an embodiment of the present invention.
도 8은 본 발명의 일 실시예에 따라 면역형광염색법을 통해 야생형의 분화 유도된 마이오튜브의 길이를 확인한 결과를 보여주는 사진이다.8 is a photograph showing the result of confirming the length of the wild-type differentiation-induced myotube through immunofluorescence staining according to an embodiment of the present invention.
도 9는 본 발명의 일 실시예에 따라 면역형광염색법을 통해 L1 돌연변이의 분화 유도된 마이오튜브의 길이를 확인한 결과를 보여주는 사진이다.9 is a photograph showing the result of confirming the length of the L1 mutant differentiation-induced myotube through immunofluorescence staining according to an embodiment of the present invention.
도 10은 본 발명의 일 실시예에 따라 면역형광염색법을 통해 분화 유도된 마이오튜브의 길이를 확인한 결과 그래프이다.10 is a graph showing the result of confirming the length of differentiation-induced myotubes through immunofluorescence staining according to an embodiment of the present invention.
도 11은 본 발명의 일 실시예에 따라 면역형광염색법을 통해 분화 유도된 마이오튜브의 두께를 확인한 결과 그래프이다.11 is a graph showing the results of confirming the thickness of myotubes induced to differentiate through immunofluorescence staining according to an embodiment of the present invention.
도 12는 본 발명의 일 실시예에 따라 지방유래 중간엽 줄기세포의 지방세포로의 분화 결과를 보여주는 그림이다.12 is a diagram showing the differentiation result of adipose-derived mesenchymal stem cells into adipocytes according to an embodiment of the present invention.
도 13은 본 발명의 일 실시예에 따라 지방생성 정도를 확인한 결과를 보여주는 그래프이다.13 is a graph showing the result of confirming the degree of adipogenesis according to an embodiment of the present invention.
도 14는 본 발명의 일 실시예에 따라 세포 내 글루코오스의 업테이크 측정 결과를 보여주는 그래프이다.14 is a graph showing the measurement result of intracellular glucose uptake according to an embodiment of the present invention.
도 15는 본 발명의 일 실시예에 따라 근감소 동물 모델에서의 체중 변화를 보여주는 그래프이다.15 is a graph showing changes in body weight in an animal model with reduced muscle mass according to an embodiment of the present invention.
도 16은 본 발명의 일 실시예에 따라 근감소 동물 모델에서의 GA 근육 및 TA 근육 조직의 무게 변화를 보여주는 그래프이다.16 is a graph showing changes in the weight of GA muscle and TA muscle tissue in an animal model with reduced muscle mass according to an embodiment of the present invention.
도 17은 본 발명의 일 실시예에 따라 근감소 동물 모델에서의 지방 및 근육의 무게 변화를 보여주는 그래프이다.17 is a graph showing changes in weight of fat and muscle in an animal model with reduced muscle mass according to an embodiment of the present invention.
도 18은 본 발명의 일 실시예에 따라 근육 분화 연관 유전자인 MyoD, MyoG, Pax7, 및 MRF4의 유전자 발현량을 확인한 결과를 보여주는 그래프이다.18 is a graph showing the results of checking the gene expression levels of MyoD, MyoG, Pax7, and MRF4, which are genes related to muscle differentiation according to an embodiment of the present invention.
도 19는 본 발명의 일 실시예에 따라 근육 분화 억제 연관 유전자인 MuRF1, Atrogin1, Hes1 유전자의 발현량을 확인한 결과를 그래프이다.19 is a graph showing the results of confirming the expression levels of MuRF1, Atrogin1, Hes1 genes, which are genes associated with inhibition of muscle differentiation according to an embodiment of the present invention.
도 20은 본 발명의 일 실시예에 따라 제너 염색을 통해 분화 유도된 마이오튜브의 길이를 확인한 결과 그래프이다.20 is a graph showing the result of confirming the length of the myotube differentiation-induced through Zener staining according to an embodiment of the present invention.
도 21은 본 발명의 일 실시예에 따라 근육 세포의 활성 정도를 확인한 결과 그래프이다.21 is a graph showing the results of confirming the activity level of muscle cells according to an embodiment of the present invention.
이하, 실시예를 통하여 본 발명을 더욱 상세히 설명하고자 한다. 이들 실시예는 오로지 본 발명을 보다 구체적으로 설명하기 위한 것으로, 본 발명의 요지에 따라 본 발명의 범위가 이들 실시예에 의해 제한되지 않는다는 것은 당업계에서 통상의 지식을 가진 자에 있어서 자명할 것이다.Hereinafter, the present invention will be described in more detail through examples. These examples are only for illustrating the present invention in more detail, and it will be apparent to those of ordinary skill in the art that the scope of the present invention is not limited by these examples according to the gist of the present invention. .
실시예Example
실시예 1. C2C12 세포주의 배양 및 분화 유도Example 1. C2C12 cell line culture and differentiation induction
근세포 분화 연구에 주로 사용되고 있는 근원세포주 C2C12 세포주(ATCC, CRL-1772)를 10% 우태아혈청 (FBS; Fetal bovine serum), 페니실린 100 U/ml 및 스트렙토마이신 100 ㎍/ml이 첨가된 DMEM을 배양용 배지로 사용하여 5% CO2가 공급되는 37℃ 세포 배양기에서 배양하였다. The myoblast cell line C2C12 cell line (ATCC, CRL-1772), which is mainly used for myocyte differentiation studies, was cultured in DMEM supplemented with 10% fetal bovine serum (FBS), penicillin 100 U/ml and streptomycin 100 μg/ml. It was cultured in a cell incubator at 37° C. supplied with 5% CO 2 as a medium for use.
C2C12 세포주는 24 웰 세포 배양 플레이트에 5 X 104 개/well 로 접종하여 배양용 배지에서 배양하였다. 세포 배양 플레이트에 세포가 90% 수준의 밀도에 도달하였을 때, 분화용 배지인 2% 말혈청이 포함된 DMEM 배지로 교체하여 분화를 유도하였다. 분화용 배지는 3 일 간격으로 새로 교체해 주었고 총 8일간 5% CO2가 공급되는 37℃ 세포 배양기에서 분화를 유도하였다. The C2C12 cell line was inoculated in a 24-well cell culture plate at 5 X 10 4 cells/well and cultured in a culture medium. When the cells in the cell culture plate reached a density of 90%, they were replaced with DMEM medium containing 2% horse serum, which is a medium for differentiation, to induce differentiation. The differentiation medium was replaced every 3 days, and differentiation was induced in a cell culture medium at 37° C. supplied with 5% CO 2 for a total of 8 days.
CHO-K1 세포주에서 생산한 ADAMTS1과 돌연변이 ADAMTS1 재조합 단백질은 하기 실시예 2의 방법으로 분리, 정제 과정을 거쳐 C2C12 세포주의 분화 유도시작과 함께 처리하였고 분화 유도가 종료되는 시점까지 0.01, 0.1, 1, 10, 100 ng/ml의 농도로 매일 1회씩 처리하였다.ADAMTS1 and mutant ADAMTS1 recombinant proteins produced in the CHO-K1 cell line were separated and purified by the method of Example 2 below, and treated together with the induction of differentiation of the C2C12 cell line. It was treated once daily at a concentration of 10, 100 ng/ml.
실시예 2. 재조합 단백질의 생산Example 2. Production of Recombinant Proteins
세포에서 생산한 재조합 단백질의 분리, 정제를 위해 재조합 단백질의 C-말단에 6x His가 달리도록 ADAMTS1 과발현 벡터를 제작하였다. 구체적으로, pEF6/V5-His A 벡터(Invitrogen, V96120)를 KpnI과 XhoI 제한효소를 이용하여 절단하였다. 여기에 ADAMTS1 서열(NM_006988.5)중 CDS부분의 5'말단에 KpnI으로 인식되는 서열을 삽입하고 3'말단에 XhoI으로 인식되는 서열을 삽입한 뒤, 앞서 절단한 벡터에 T4 DNA 라이게이즈를 이용하여 접착시켜 제작하였다. L1, L2, L3, L4 돌연변이 ADAMTS1은 야생형 ADAMTS1을 주형으로 하고 L1은 883번부터 900번, L2는 937번부터 951번, L3는 1021번부터 1038번, L4는 1084번부터 1098번 뉴클레오타이드까지 절단하였고, 상기 모든 돌연변이는 1237번 뉴클레오타이드 이후의 서열을 모두 제거하여 제작하였다. For the isolation and purification of the recombinant protein produced in the cell, an ADAMTS1 overexpression vector was constructed so that 6x His was placed at the C-terminus of the recombinant protein. Specifically, the pEF6/V5-His A vector (Invitrogen, V96120) was digested using KpnI and XhoI restriction enzymes. Here, a sequence recognized as KpnI was inserted at the 5' end of the CDS part of the ADAMTS1 sequence (NM_006988.5) and a sequence recognized as XhoI was inserted at the 3' end, and then T4 DNA ligase was applied to the previously cut vector. It was manufactured by bonding. L1, L2, L3, L4 mutant ADAMTS1 uses wild-type ADAMTS1 as a template, L1 is cleaved from 883 to 900, L2 from 937 to 951, L3 from 1021 to 1038, and L4 from 1084 to 1098 nucleotides and all the mutations were made by removing all sequences after nucleotide 1237.
CHO-K1 세포주는 10% 우태아혈청 (FBS; Fetal bovine serum), 페니실린 100 U/ml, 스트렙토마이신 100 ㎍/ml 및 25 mM 헤페스가 첨가된 RPMI1640 배지를 배양배지로 하여 6 웰 세포 배양 플레이트에 접종하고, 5% CO2가 공급되는 37℃ 세포 배양기에서 배양하였다. 다음날 Lipofectamine 2000 (Invitrogen, 11668019)과 제작한 ADAMTS1 또는 ADAMTS1 돌연변이 발현 벡터를 섞어 CHO-K1세포에 처리하는 방식으로 벡터를 세포 내 도입시켰다. 24 시간 동안 배양한 뒤 세포를 10 배 희석하여 100 mm 세포배양 디쉬로 옮겨준 다음 블라스티시딘-에스(Blasticidin-STM)를 10 ㎍/ml의 농도가 되도록 1차 처리하였다. 이후 4일 간격으로 총 3회에 걸쳐 새로운 배양배지에 블라스티시딘-에스를 10 ㎍/ml의 농도가 되도록 섞어 교체해주었다. 블라스티시딘-에스에 의해 독립된 콜로니가 형성된 후, 각 콜로니를 새로운 세포 배양용 플레이트에 접종하여 배양하였다. 각 콜로니를 웨스턴블랏을 통해 ADAMTS1 또는 ADAMTS1 돌연변이 단백질을 발현하는지 확인한 뒤 안정화 세포주로 사용하였다. CHO-K1 안정화 세포주를 100 mm 디쉬에 접종한 다음 세포의 밀도가 100% 수준에 도달하면 RPMI1640 배지로 교환하여 2 일간 배양하였다. 2 일 뒤 배양하던 디쉬에서 상부 배양액 부분만을 취하여 Ni-NTA 아가로오스 (Qiagen, 30210)로 정제하여 회수한 뒤, C2C12 세포주의 분화 유도 실험에 사용하였다. 구체적으로는, 세포 배양액을 Ni-NTA 아가로오스를 장착한 중력 크로마토그래피 컬럼(Qiagen, 34964)에 통과시킨 뒤 10 mM 이미다졸 버퍼로 세척하였다. 3 회 세척을 반복한 다음 250 mM 이미다졸 버퍼로 용출하여 단백질을 정제하였다.The CHO-K1 cell line is a 6-well cell culture plate using RPMI1640 medium supplemented with 10% Fetal bovine serum (FBS), penicillin 100 U/ml, streptomycin 100 μg/ml and 25 mM Hepes as a culture medium. was inoculated, and cultured in a 37° C. cell incubator supplied with 5% CO 2 . The next day, the vector was introduced into the cells by mixing Lipofectamine 2000 (Invitrogen, 11668019) with the prepared ADAMTS1 or ADAMTS1 mutant expression vector and treating CHO-K1 cells. After culturing for 24 hours, the cells were diluted 10-fold, transferred to a 100 mm cell culture dish, and then blasticidin-S (Blasticidin-S TM ) was first treated to a concentration of 10 μg/ml. Thereafter, blasticidin-S was mixed and replaced in a new culture medium to a concentration of 10 μg/ml over a total of 3 times at 4-day intervals. After independent colonies were formed by blasticidin-S, each colony was inoculated into a new cell culture plate and cultured. After confirming whether each colony expresses ADAMTS1 or ADAMTS1 mutant protein through western blot, it was used as a stabilizing cell line. The CHO-K1 stabilized cell line was inoculated into a 100 mm dish, and when the cell density reached 100%, it was exchanged with RPMI1640 medium and cultured for 2 days. After 2 days, only a portion of the upper culture medium was taken from the cultured dish, purified with Ni-NTA agarose (Qiagen, 30210), and recovered, and used in the differentiation induction experiment of the C2C12 cell line. Specifically, the cell culture medium was passed through a gravity chromatography column (Qiagen, 34964) equipped with Ni-NTA agarose and washed with 10 mM imidazole buffer. Washing was repeated 3 times, and then the protein was purified by eluting with 250 mM imidazole buffer.
실시예 3. 재조합 단백질 발현 확인Example 3. Confirmation of recombinant protein expression
실시예 2의 방법으로 제작한 ADAMTS1 돌연변이 과발현 CHO-K1 안정화 세포주의 배양액을 취하여 6x-His를 인지하는 항체를 사용한 웨스턴블랏을 통해 생성된 재조합 단백질의 발현을 확인하여, 그 결과를 도 1a 및 도 1b에 나타내었다.Taking the culture medium of the ADAMTS1 mutant-overexpressing CHO-K1 stabilized cell line prepared by the method of Example 2, and confirming the expression of the recombinant protein produced by western blotting using an antibody that recognizes 6x-His, the results are shown in FIGS. 1A and FIG. 1b.
도 1b에서 확인할 수 있듯이, L1, L2, L3, L4 돌연변이는 전구영역(Prodomain)이 포함된 부분이 사라지고 성숙영역(Mature domain)이 주요하게 발현되는 양상을 보였다. 한편 L1 돌연변이를 제외한 나머지 돌연변이는 단백질의 발현이 상당부분 감소하는 것으로 나타났다(도 1a 참조).As can be seen in FIG. 1b , the L1, L2, L3, and L4 mutations showed a pattern in which the part including the prodomain disappeared and the mature domain was mainly expressed. On the other hand, the remaining mutations except for the L1 mutation showed a significant decrease in protein expression (see FIG. 1a ).
실시예 4. 제너 염색법 (Jenner's staining)Example 4. Jenner's staining
실시예 3 과 동일한 방법으로 C2C12 세포주를 분화 유도하였고, 분화 세포주를 메탄올로 10 분간 고정하였다. PBS로 3차례 충분히 세척해 준 뒤 2.5 mg/ml 농도로 메탄올에 용해되어 있는 제너 염색시약을 증류수와 1:1로 섞어 10분간 염색하였다. 염색이 끝난 플레이트는 증류수로 세척을 해 준 뒤 건조시켜 현미경으로 관찰하여, 그 결과를 도 2 내지 도 6에 나타내었다. Differentiation was induced in the C2C12 cell line in the same manner as in Example 3, and the differentiated cell line was fixed with methanol for 10 minutes. After sufficient washing with PBS three times, a Zener dyeing reagent dissolved in methanol at a concentration of 2.5 mg/ml was mixed with distilled water 1:1 and dyed for 10 minutes. The stained plate was washed with distilled water, dried, and observed under a microscope, and the results are shown in FIGS. 2 to 6 .
도 2 내지 도 6에서 확인할 수 있듯이, C2C12세포주를 분화 유도할 때 ADAMTS1을 함께 처리하면 마이오튜브의 길이가 길어지는 것을 확인하였고 특히 L1 돌연변이를 처리한 경우 야생형을 처리하여 분화 유도한 세포주보다 마이오튜브의 길이가 길어지고 두께가 두꺼워지는 것을 확인하였다.As can be seen in FIGS. 2 to 6 , it was confirmed that the length of the myotube was increased when ADAMTS1 was treated with the C2C12 cell line when differentiation was induced. It was confirmed that the length of the O-tube was increased and the thickness was increased.
실시예 5. 면역형광염색법 (Immunofluorescence staining)Example 5. Immunofluorescence staining
24 웰 세포 배양 플레이트에서 C2C12 세포주를 8 일간 분화 유도하면서 ADAMTS1 또는 ADAMTS1 돌연변이 재조합 단백질을 처리하였다. 분화가 유도된 세포는 4% 파라포름알데히드로 10 분간 고정한 후, 0.25% Triton X-100을 이용하여 10분간 세포막 투과화를 진행하였다. In a 24-well cell culture plate, the C2C12 cell line was treated with ADAMTS1 or ADAMTS1 mutant recombinant protein while inducing differentiation for 8 days. After the differentiation-induced cells were fixed with 4% paraformaldehyde for 10 minutes, cell membrane permeabilization was performed for 10 minutes using 0.25% Triton X-100.
그 다음, 2% BSA로 30 분간 블로킹을 해준 뒤 마이오신 중사슬(MHC) 항체를 1:500으로 2% BSA 버퍼에 섞어 1시간 동안 처리하였다. 그 다음, Alexa Fluor® 555 형광항체를 1:200으로 2% BSA 버퍼에 섞어 1 시간 동안 처리하였고 DAPI 염색법을 통해 핵을 염색하였다. Then, after blocking with 2% BSA for 30 minutes, myosin heavy chain (MHC) antibody was mixed with 2% BSA buffer at a ratio of 1:500 and treated for 1 hour. Then, Alexa Fluor® 555 fluorescent antibody was mixed with 2% BSA buffer at a ratio of 1:200 and treated for 1 hour, and the nucleus was stained by DAPI staining.
염색 과정이 끝난 플레이트는 형광현미경(Nikon Ts2-FL)을 이용하여 사진을 촬영하였고 마이오튜브의 길이는 제조사에서 제공한 분석 프로그램 (Nikon, NIS Elements)을 이용하여 측정하고 통계처리 하였다. 분석이 종료된 플레이트는 차광하여 냉장보관 하였다. 그 결과를 도 7 내지 도 11 및 표 1 내지 표 2에 나타내었다.The plate after the staining process was photographed using a fluorescence microscope (Nikon Ts2-FL), and the length of the myotube was measured and statistically processed using the analysis program (Nikon, NIS Elements) provided by the manufacturer. After the analysis was completed, the plate was refrigerated after blocking the light. The results are shown in FIGS. 7 to 11 and Tables 1 to 2 .
구분division ng/mlng/ml 평균 (um)average (um) SdSd
CtrlCtrl -- 707.937707.937 119.37119.37
WTWT 0.010.01 1354.071354.07 168.904168.904
1One 1498.861498.86 48.399748.3997
100100 1558.951558.95 117.736117.736
L1 돌연변이L1 mutation 0.010.01 1618.181618.18 136.365136.365
1One 1676.781676.78 111.361111.361
100100 1750.251750.25 126.443126.443
L3 돌연변이L3 mutation 0.010.01 1311.131311.13 217.574217.574
1One 1253.491253.49 204.303204.303
100100 1198.51198.5 168.134168.134
L4 돌연변이L4 mutation 0.010.01 1128.151128.15 89.467789.4677
1One 1170.021170.02 70.000470.0004
100100 1129.451129.45 69.517469.5174
ng/mlng/ml 평균 (um)average (um) SdSd
CtrlCtrl -- 31.495631.4956 5.745725.74572
WTWT 0.010.01 40.093340.0933 6.152546.15254
1One 44.005644.0056 4.551344.55134
100100 42.4642.46 4.145534.14553
L1 돌연변이L1 mutation 0.010.01 40.233340.2333 5.336975.33697
1One 47.0447.04 6.059186.05918
100100 47.672247.6722 3.602093.60209
L3 돌연변이L3 mutation 0.010.01 33.5433.54 4.983234.98323
1One 37.492237.4922 7.182627.18262
100100 38.278938.2789 6.78596.7859
L4 돌연변이L4 mutation 0.010.01 36.577836.5778 3.069143.06914
1One 31.788931.7889 5.002865.00286
100100 31.801131.8011 5.263345.26334
도 7 내지 도 11 및 표 1 내지 표 2에서 확인할 수 있듯이, 분화배지를 처리하여 분화를 유도한 C2C12세포주 대비, ADAMTS1 야생형이나 돌연변이를 처리하면서 분화를 유도한 C2C12 세포주가 더욱 두께와 길이가 긴 마이오튜브로 분화됨을 확인하였다. 또한 마이오튜브의 길이는 야생형에 비해 ADAMTS1 L1 돌연변이를 처리한 세포주에서 더욱 길어지고 두께 또한 두꺼워지는 것을 확인하였다. 그러나 L3나 L4 돌연변이는 마이오튜브의 길이나 두께에 있어서 야생형 ADAMTS1보다 나은 효과를 나타내지 않았다.As can be seen in FIGS. 7 to 11 and Tables 1 to 2, compared to the C2C12 cell line in which differentiation was induced by treatment with a differentiation medium, the C2C12 cell line in which differentiation was induced by treatment with ADAMTS1 wild-type or mutant was longer in thickness and length. It was confirmed that differentiation into Otube. In addition, it was confirmed that the length of the myotube became longer and thicker in the cell line treated with ADAMTS1 L1 mutation compared to the wild type. However, L3 or L4 mutants did not show a better effect than wild-type ADAMTS1 on the length or thickness of myotubes.
실시예 6. 지방생성 정도 확인Example 6. Confirmation of adipogenesis level
지방유래 중간엽 줄기세포주(ATCC, PCS-500-011)는 ATCC에서 판매하는 2%의 우태아혈청과 5 ng/ml의 재조합 인간 상피세포인자, 5 ng/ml의 재조합 인간 섬유아세포 성장인자가 포함된 중간엽 줄기세포주 전용 배지(ATCC, PCS-500-030)에서 배양하였다. 세포의 분화 시에는 StemPro 배지(Gibco, A10410-01)와 제조사가 제공하는 첨가물(Gibco, A10065-01)을 혼합한 배지로 갈아주었고 3 일 간격으로 새로운 StemPro배지로 갈아주면서 총 14 일간 지방세포로의 분화를 유도하였다. 분화를 유도하면서 ADAMTS1 야생형 또는 L1 돌연변이 재조합 단백질은 1 ng/ml 또는 100 ng/ml의 농도로 매일 1 회씩 처리하였다. An adipose-derived mesenchymal stem cell line (ATCC, PCS-500-011) was prepared using 2% fetal bovine serum sold by ATCC, 5 ng/ml of recombinant human epithelial cell factor, and 5 ng/ml of recombinant human fibroblast growth factor. It was cultured in the included mesenchymal stem cell line-only medium (ATCC, PCS-500-030). At the time of cell differentiation, the medium mixed with StemPro medium (Gibco, A10410-01) and the additive (Gibco, A10065-01) provided by the manufacturer was changed and replaced with a new StemPro medium every 3 days for a total of 14 days. Differentiation was induced. While inducing differentiation, ADAMTS1 wild-type or L1 mutant recombinant protein was treated once daily at a concentration of 1 ng/ml or 100 ng/ml.
분화 유도된 지방유래 중간엽 줄기세포의 지방생성 정도를 확인하기 위하여 오일 레드 오 염색 방법을 이용하였다. 구체적으로, 분화가 완료된 세포를 10% 포르말린으로 고정하고, 오일 레드 오 염색 용액을 처리하여 염색하였다. 염색이 완료된 세포는 현미경으로 관찰한 후 100% 이소프로판올을 이용하여 침착된 염료를 추출하였다. 추출한 염료는 520 nm의 파장에서 흡광도를 측정하여 정량하여, 그 결과를 도 12 및 도 13 및 표 3에 나타내었다.In order to confirm the degree of adipogenesis of differentiation-induced adipose-derived mesenchymal stem cells, the Oil Red O staining method was used. Specifically, cells that have been differentiated were fixed with 10% formalin and stained with Oil Red O staining solution. After the stained cells were observed under a microscope, the deposited dye was extracted using 100% isopropanol. The extracted dye was quantified by measuring absorbance at a wavelength of 520 nm, and the results are shown in FIGS. 12 and 13 and Table 3.
Diff. -Diff. - Diff. +Diff. + WT 1 (ng/ml)WT 1 (ng/ml) WT 100 (ng/ml)WT 100 (ng/ml) L1 1 (ng/ml)L1 1 (ng/ml) L1 100 (ng/ml)L1 100 (ng/ml)
0.134390.13439 1One 0.716420.71642 0.750390.75039 0.671230.67123 0.673720.67372
0.045090.04509 00 0.04470.0447 0.033430.03343 0.153970.15397 0.089990.08999
도 12 및 도 13 및 표 3에서 확인할 수 있듯이, ADAMTS1의 야생형과 L1 돌연변이 모두 지방유래 중간엽 줄기세포의 지방세포로의 분화를 억제함을 확인하였다.As can be seen in FIGS. 12 and 13 and Table 3, it was confirmed that both the wild-type and L1 mutants of ADAMTS1 inhibit the differentiation of adipose-derived mesenchymal stem cells into adipocytes.
실시예 7. 세포 내 글루코오스의 업테이크 측정Example 7. Measurement of uptake of intracellular glucose
형광 글루코오스 유사체인 2-NBDG (2-(N-(7-nitrobenz-2-oxa-1,3-diazol-4-yl)amino- 2-deoxygluocose)를 이용하여 근육 분화 증가 활성에 따라 변화하는 세포 내 글루코오스 업테이크를 측정하였다. C2C12 세포주를 8 일간 분화 유도하면서 ADAMTS1 혹은 L1 돌연변이 재조합 단백질을 각각 100 ng/ml의 농도로 매일 1회씩 처리하였다. 분화가 유도된 세포는 글루코오스가 없는 DMEM 배지로 2 시간동안 처리하였고, 이후 100 nM의 인슐린을 15 분간 처리한 뒤 2-NBDG를 최종 100 ㎍/ml의 농도가 되도록 첨가하였다. 글루코오스 업테이크는 익사이테이션(excitation) 485 nm, 에미션(emission) 535 nm에서 측정하여, 그 결과를 도 14 및 표 4에 나타내었다.Cells that change according to the activity of increasing muscle differentiation using 2-NBDG (2-(N-(7-nitrobenz-2-oxa-1,3-diazol-4-yl)amino-2-deoxygluocose), a fluorescent glucose analogue) Glucose uptake was measured.C2C12 cell line was treated with ADAMTS1 or L1 mutant recombinant protein at a concentration of 100 ng/ml, respectively, once daily while inducing differentiation for 8 days.The differentiation-induced cells were treated with DMEM medium without glucose 2 After treatment with 100 nM insulin for 15 minutes, 2-NBDG was added to a final concentration of 100 μg/ml, glucose uptake, excitation 485 nm, emission 535 It was measured in nm, and the results are shown in FIG. 14 and Table 4.
구분division Ins-Ins- Ins+Ins+
-- WTWT L1L1 -- WTWT L1L1
#1#One 1One 1.305431.30543 1.6046921.604692 1.7035631.703563 1.9124191.912419 2.4610152.461015
#2#2 0.8964750.896475 1.337541.33754 1.9135861.913586 1.6347351.634735 1.9547271.954727 2.1100512.110051
#3#3 0.9820870.982087 1.2729341.272934 1.617641.61764 1.6939081.693908 1.7970681.797068 2.5300662.530066
#4#4 0.8900830.890083 1.2948611.294861 1.5936171.593617 1.6647561.664756 1.8316391.831639 2.465922.46592
MeanMean 0.9421610.942161 1.3026911.302691 1.6823831.682383 1.6742411.674241 1.8739631.873963 2.3917632.391763
SDSD 0.0569760.056976 0.0268870.026887 0.1544470.154447 0.0310770.031077 0.0723550.072355 0.1904250.190425
도 14 및 표 4에서 확인할 수 있듯이, 분화 유도된 C2C12 세포주의 글루코오스 업테이크 정도는 인슐린 처리 하에서 증가하는데 특히 L1 돌연변이 ADAMTS1을 처리하여 근육세포 분화를 유도한 C2C12세포주가 야생형을 처리하여 분화 유도한 세포주에 비해 글루코오스 업테이크가 더욱 증가함을 확인하였다.As can be seen in Figure 14 and Table 4, the degree of glucose uptake of the differentiation-induced C2C12 cell line increases under insulin treatment. In particular, the C2C12 cell line in which myocyte differentiation was induced by treatment with L1 mutant ADAMTS1 was treated with the wild-type cell line to induce differentiation. It was confirmed that the glucose uptake was further increased compared to .
실시예 8: 근감소 동물 모델에서의 체중 회복 관찰Example 8: Observation of weight recovery in an animal model of muscle loss
12주령의 C57BL/6 마우스에 12 일간 매일 덱사메타손을 50 mg/kg의 농도로 복강주사를 통해 투여하였다. 동시에 L1 돌연변이 ADAMTS1을 각 0.02 ㎍/㎕(Low), 0.2㎍/㎕(Mid), 2㎍/㎕(High)의 농도로 100 ㎕씩 복강주사를 통해 투여하였다. 총 12 일간 투여하며 동물 모델에서 체중 회복 정도를 관찰하였다. To 12-week-old C57BL/6 mice, dexamethasone was administered via intraperitoneal injection at a concentration of 50 mg/kg daily for 12 days. At the same time, L1 mutant ADAMTS1 was administered by intraperitoneal injection at a concentration of 0.02 μg/μl (Low), 0.2 μg/μl (Mid), and 2 μg/μl (High), respectively. It was administered for a total of 12 days, and the degree of body weight recovery was observed in animal models.
도 15에서 확인할 수 있듯이, 덱사메타손 투여로 근감소를 유도한 동물 모델의 경우, 정상 대조군에 비해 뚜렷한 체중 감소가 확인되었고, 동물 모델에 L1 돌연변이 ADAMTS1을 처리한 경우, Low, Mid 및 High 용량 군에서 모두 유의하게 체중이 회복되는 것을 확인하였으며, 투여된 L1 돌연변이 ADAMTS1의 용량에 의존적으로 체중이 회복되는 것이 관찰되었다.As can be seen in Figure 15, in the case of the animal model inducing muscle loss by administration of dexamethasone, a clear weight loss was confirmed compared to the normal control group, and when the L1 mutant ADAMTS1 was treated in the animal model, Low, Mid and High dose groups In all, it was confirmed that the body weight was recovered significantly, and it was observed that the body weight was recovered depending on the dose of the administered L1 mutant ADAMTS1.
실시예 9: 근감소 동물 모델에서의 TA, GA 근육 조직 무게 회복 관찰Example 9: Observation of TA, GA muscle tissue weight recovery in sarcopenia animal model
12주령의 C57BL/6 마우스에 12 일간 덱사메타손을 투여한 근감소 동물모델에서 근육조직의 무게 변화를 확인하기 위하여 투여 종료 후 부검을 진행하여 TA와 GA 근육조직을 적출하였다. 적출한 근육 조직은 무게를 측정하여 비교하였다.In order to confirm the change in muscle tissue weight in a sarcopenic animal model in which dexamethasone was administered to 12-week-old C57BL/6 mice for 12 days, an autopsy was performed after administration to extract TA and GA muscle tissue. The extracted muscle tissue was compared by measuring the weight.
도 16에서 확인할 수 있듯이, 덱사메타손 투여로 근감소를 유도한 동물 모델의 경우, TA 근육 및 GA 근육 모두에서 정상 대조군에 비해 뚜렷한 조직 무게 감소가 확인되었고, 근 감소 유도 동물 모델에 L1 돌연변이 ADAMTS1을 처리한 경우, Mid 및 High 용량 군에서 유의하게 TA 근육 및 GA 근육 조직의 무게가 회복되는 것을 확인하였다.As can be seen in FIG. 16 , in the case of the animal model induced by dexamethasone administration, a significant decrease in tissue weight was confirmed in both the TA muscle and the GA muscle compared to the normal control group, and the L1 mutant ADAMTS1 was treated in the muscle reduction induced animal model. In one case, it was confirmed that the weight of TA muscle and GA muscle tissue was significantly recovered in the Mid and High dose groups.
실시예 10: 근감소 동물 모델에서의 지방과 근육의 회복 관찰Example 10: Observation of fat and muscle recovery in muscle loss animal model
12주령의 C57BL/6 마우스에 12일간 매일 덱사메타손을 50 mg/kg의 농도로 복강주사를 통해 투여하였고 동시에 L1 돌연변이 ADAMTS1을 2 ㎍/㎕의 농도로 100 ㎕씩 복강주사를 통해 투여하였다. 투여 종료 후 흡입마취를 통해 마우스를 마취시킨 다음 이중 에너지 X선 흡수 (DXA; Dual-energy X-ray Absorptiometry) 장치를 이용한 근육량 및 체지방량 분석을 시행하였다. To 12-week-old C57BL/6 mice, dexamethasone was administered by intraperitoneal injection at a concentration of 50 mg/kg every day for 12 days, and at the same time, L1 mutant ADAMTS1 was administered at a concentration of 2 μg/μl by 100 μl by intraperitoneal injection. After completion of administration, mice were anesthetized through inhalation anesthesia, and muscle mass and body fat mass were analyzed using a dual-energy X-ray absorptiometry (DXA) device.
도 17에서 확인할 수 있듯이, 덱사메타손 투여로 근감소를 유도한 동물 모델의 경우, 지방과 근육이 모두 크게 감소되었다. 근 감소 유도 동물 모델에 L1 돌연변이 ADAMTS1을 처리한 경우, 지방과 근육이 모두 유의하게 회복되는 것을 확인하였다. 특히 근육의 경우는 정상 수준으로 회복되는 것을 확인하였다. As can be seen in FIG. 17 , in the case of an animal model in which muscle loss was induced by administration of dexamethasone, both fat and muscle were significantly reduced. When the L1 mutant ADAMTS1 was treated in the muscle loss induced animal model, it was confirmed that both fat and muscle were significantly recovered. In particular, it was confirmed that the muscle was restored to a normal level.
실시예 11: 근감소 동물 모델에서의 근육 분화 연관 유전자 발현 증가 확인Example 11: Confirmation of increased expression of genes associated with muscle differentiation in an animal model of sarcopenia
12주령의 C57BL/6 마우스에 12일간 매일 덱사메타손을 50 mg/kg의 농도로 복강주사를 통해 투여하였고 동시에 L1 돌연변이 ADAMTS1을 각 0.02 ㎍/㎕(Low), 0.2 ㎍/㎕(Mid), 2 ㎍/㎕(High)의 농도로 100 ㎕씩 복강주사를 통해 투여하였다. 덱사메타손과 L1 돌연변이 ADAMTS1의 투여가 종료된 후 부검하여 GA 근육조직을 적출하였다. GA 근육조직은 동결상태로 페슬을 이용하여 균질화 과정을 거친 뒤 RNA를 추출하였다 (Qiagen, 74104). 추출한 RNA는 cDNA 합성 시약 (Promega, A5000)을 이용하여 cDNA로 제작하였다. 합성된 cDNA는 실시간 중합효소 연쇄반응 장비(Applied Biosystems, Quantstudio3)를 이용하여 근육의 분화와 연관된 유전자인 MyoD, MyoG, Pax7, MRF4의 상대적인 mRNA 발현양을 비교하였다. To 12-week-old C57BL/6 mice, dexamethasone was administered at a concentration of 50 mg/kg daily for 12 days via intraperitoneal injection, and at the same time, L1 mutant ADAMTS1 was administered at 0.02 μg/μl (Low), 0.2 μg/μl (Mid), and 2 μg, respectively. It was administered by intraperitoneal injection by 100 μl at a concentration of /μl (High). After the administration of dexamethasone and L1 mutant ADAMTS1 was completed, the GA muscle tissue was excised at an autopsy. GA muscle tissue was subjected to homogenization using a pestle in a frozen state, and then RNA was extracted (Qiagen, 74104). The extracted RNA was prepared as cDNA using cDNA synthesis reagent (Promega, A5000). The synthesized cDNA was compared to the relative mRNA expression levels of MyoD, MyoG, Pax7, and MRF4 genes related to muscle differentiation using real-time polymerase chain reaction equipment (Applied Biosystems, Quantstudio3).
도 18에서 확인할 수 있듯이, 덱사메타손 처리에 의한 근 감소 유도 동물 모델에 대해 L1 돌연변이 ADAMTS1을 처리한 경우, MyoD, MyoG, Pax7, 및 MRF4 유전자의 발현량이 모두 감소되었고, Low 용량군 및 High 용량군에서 용량의존적으로 덱사메타손 처리군에 비해 유의하게 유전자 발현이 모두 증가되는 것을 확인하였다. As can be seen in FIG. 18 , when L1 mutant ADAMTS1 was treated with respect to the dexamethasone treatment-induced muscle loss induction animal model, the expression levels of MyoD, MyoG, Pax7, and MRF4 genes were all reduced, and in the Low and High dose groups It was confirmed that all gene expression was significantly increased compared to the dexamethasone treatment group in a dose-dependent manner.
이는 L1 돌연변이 ADAMTS1이 근육 분화 연관 유전자의 발현을 증가시킴으로써, 근육 분화를 증가시킨다는 것을 보여준다.This shows that the L1 mutant ADAMTS1 increases muscle differentiation by increasing the expression of genes associated with muscle differentiation.
실시예 12: 근감소 동물 모델에서의 근육 분화 억제 연관 유전자 발현 감소 확인Example 12: Confirmation of reduced expression of genes associated with inhibition of muscle differentiation in an animal model of muscle loss
12주령의 C57BL/6 마우스에 12일간 매일 덱사메타손을 50 mg/kg의 농도로 복강주사를 통해 투여하였고 동시에 L1 돌연변이 ADAMTS1을 각 0.02 ㎍/㎕(Low), 0.2 ㎍/㎕(Mid), 2㎍/㎕(High)의 농도로 100 ㎕씩 복강주사를 통해 투여하였다. 덱사메타손과 L1 돌연변이 ADAMTS1의 투여가 종료된 후 부검하여 GA 근육조직을 적출하였다. GA 근육조직은 동결상태로 페슬을 이용하여 균질화 과정을 거친 뒤 RNA를 추출하였다 (Qiagen, 74104). 추출한 RNA는 cDNA 합성 시약 (Promega, A5000)을 이용하여 cDNA로 제작하였다. 합성된 cDNA는 실시간 중합효소 연쇄반응 장비(Applied Biosystems, Quantstudio3)를 이용하여 근육 분화 억제 연관 유전자인 MuRF1, Atrogin1, Hes1의 상대적인 mRNA 발현양을 비교하였다. To 12-week-old C57BL/6 mice, dexamethasone was administered at a concentration of 50 mg/kg daily for 12 days via intraperitoneal injection, and at the same time, L1 mutant ADAMTS1 was administered at 0.02 μg/μl (Low), 0.2 μg/μl (Mid), and 2 μg, respectively. It was administered by intraperitoneal injection by 100 μl at a concentration of /μl (High). After the administration of dexamethasone and L1 mutant ADAMTS1 was completed, the GA muscle tissue was excised at an autopsy. GA muscle tissue was subjected to homogenization using a pestle in a frozen state, and then RNA was extracted (Qiagen, 74104). The extracted RNA was prepared as cDNA using cDNA synthesis reagent (Promega, A5000). For the synthesized cDNA, the relative mRNA expression levels of MuRF1, Atrogin1, and Hes1, which are genes related to inhibition of muscle differentiation, were compared using real-time polymerase chain reaction equipment (Applied Biosystems, Quantstudio3).
도 19에서 확인할 수 있듯이, 덱사메타손 처리에 의한 근 감소 유도 동물 모델에 대해 L1 돌연변이 ADAMTS1을 처리한 경우, Low 용량군 및 High 용량군 모두에서 MuRF1, Atrogin1, Hes1 유전자의 발현이 유의하게 감소된 것을 확인하였다. 이는 L1 돌연변이 ADAMTS1이 근육 분화 억제 연관 유전자를 억제함으로써, 근육 분화를 증가시킬 수 있음을 보여준다.As can be seen in Figure 19, when L1 mutant ADAMTS1 was treated with respect to the dexamethasone treatment-induced muscle loss inducing animal model, the expression of MuRF1, Atrogin1, Hes1 genes was significantly reduced in both the Low dose group and the High dose group. did. This shows that the L1 mutant ADAMTS1 can increase muscle differentiation by suppressing the genes associated with the inhibition of muscle differentiation.
실시예 13: 항암제에 의한 근육 분화 억제 유도 세포(암 악액질(Cancer cachexia) 모델)에서의 근육 분화 증가 확인Example 13: Confirmation of increased muscle differentiation in cells (Cancer cachexia model) inducing muscle differentiation inhibition by anticancer agents
24 웰 세포 배양 플레이트에서 근아세포 세포주인 C2C12 세포주를 8일간 분화 유도하면서 50 μM 농도의 시스플라틴 항암제와 L1 ADAMTS1 돌연변이 재조합 단백질을 각각 0.1, 1, 10 ng/ml의 농도로 함께 처리하였다. 분화가 유도된 세포는 메탄올로 10분간 고정하였다. PBS로 3차례 충분히 세척해 준 뒤 2.5 mg/ml 농도로 메탄올에 용해되어 있는 제너 염색시약을 증류수와 1:1로 섞어 10분간 염색하였다. 염색이 끝난 플레이트는 증류수로 세척을 해 준 뒤 건조시켜 현미경으로 관찰하였다.In a 24-well cell culture plate, the C2C12 cell line, a myoblast cell line, was treated with a cisplatin anticancer drug at a concentration of 50 μM and a L1 ADAMTS1 mutant recombinant protein at a concentration of 0.1, 1, and 10 ng/ml, respectively, while inducing differentiation for 8 days. Differentiation-induced cells were fixed with methanol for 10 minutes. After sufficient washing with PBS three times, a Zener dyeing reagent dissolved in methanol at a concentration of 2.5 mg/ml was mixed with distilled water 1:1 and dyed for 10 minutes. The stained plate was washed with distilled water, dried and observed under a microscope.
도 20에서 확인할 수 있듯이, 시스플라틴을 처리하여 근육분화를 억제시킨 암 악액질(Cancer cachexia) 세포 모델에 대해서, L1 돌연변이 ADAMTS1을 처리한 경우, 감소되었던 마이오튜브의 길이는 용량 의존적으로 유의하게 증가하였다. As can be seen in FIG. 20 , for a cancer cachexia cell model in which muscle differentiation was inhibited by treatment with cisplatin, when L1 mutant ADAMTS1 was treated, the length of the myotube, which was reduced, was significantly increased in a dose-dependent manner. .
실시예 14: 프라이머리(primary) 근육 세포의 활성 증가 확인Example 14: Confirmation of increased activity of primary muscle cells
96 웰 플레이트에서 프라이머리 근육 세포를 24 시간 배양한 뒤 혈청이 함유되어 있지 않은 배지로 교환하여 24 시간을 추가 배양하였다. 그 다음 10 μM의 BrdU와함께 L1 ADAMTS1 돌연변이 단백질을 각각 0.01, 0.1, 1, 10, 100 ng/ml의 농도로 처리하여 24 시간 반응시켜주었다. 그 다음 BrdU 형광 측정 ELISA 키트 (Cell signaling technology, 6813S)를 이용하여 450 nm에서 흡광도를 측정하였다. 근육 세포 내에서 새롭게 합성되는 DNA의 양이 증가하면 측정되는 흡광도가 증가하고, 이는 근육 세포의 활성이 증가한 것으로 볼 수 있다. After culturing the primary muscle cells in a 96-well plate for 24 hours, the medium was replaced with a serum-free medium and further cultured for 24 hours. Then, the L1 ADAMTS1 mutant protein was treated with 10 μM of BrdU at concentrations of 0.01, 0.1, 1, 10, and 100 ng/ml, respectively, and reacted for 24 hours. Then, the absorbance was measured at 450 nm using a BrdU fluorescence measurement ELISA kit (Cell signaling technology, 6813S). When the amount of newly synthesized DNA in muscle cells increases, the measured absorbance increases, which can be seen as an increase in muscle cell activity.
도 21에서 확인할 수 있듯이, L1 돌연변이 ADAMTS1을 처리한 경우 프라이머리 근육세포의 활성이 용량 의존적으로 유의하게 증가됨을 확인하였다.As can be seen in FIG. 21 , it was confirmed that the activity of the primary muscle cells was significantly increased in a dose-dependent manner when the L1 mutant ADAMTS1 was treated.
이상으로 본 발명의 특정한 부분을 상세히 기술하였는 바, 당업계의 통상의 지식을 가진 자에게 있어서 이러한 구체적인 기술은 단지 바람직한 구현예일 뿐이며, 이에 본 발명의 범위가 제한되는 것이 아닌 점은 명백하다.As the specific parts of the present invention have been described in detail above, for those of ordinary skill in the art, these specific descriptions are only preferred embodiments, and it is clear that the scope of the present invention is not limited thereto.

Claims (12)

  1. 서열번호 1의 아미노산 서열로 이루어진 펩타이드의 295 내지 300번 위치의 아미노산으로 이루어진 군에서 선택된 1개 이상의 아미노산의 결실을 포함하는 L1 돌연변이 ADAMTS1 (A Disintegrin and Metalloprotease with ThromboSpondin motifs) 펩타이드.L1 mutant ADAMTS1 (A Disintegrin and Metalloprotease with ThromboSpondin motifs) peptide comprising a deletion of one or more amino acids selected from the group consisting of amino acids at positions 295 to 300 of the peptide consisting of the amino acid sequence of SEQ ID NO: 1.
  2. 제1항에 있어서, 상기 L1 돌연변이 펩타이드는 서열번호 1의 아미노산 서열로 이루어진 펩타이드의 295번 위치 히스티틴 (Histidine), 296번 위치 프롤린 (Proline), 297번 위치 세린 (Serine), 298번 위치 아이소루신 (Isoleucine), 299번 위치 아르기닌 (Arginine) 및 300번 위치 아스파라긴 (Asparagine)으로 이루어진 군에서 선택된 하나 이상의 위치의 아미노산의 결실을 포함하는 것인, 펩타이드.According to claim 1, wherein the L1 mutant peptide is a peptide consisting of the amino acid sequence of SEQ ID NO: 1, histidine at position 295, proline at position 296, proline at position 296, serine at position 297, iso at position 298 Leucine (Isoleucine), position 299 arginine (Arginine), and position 300 asparagine (Asparagine) that comprises a deletion of amino acids at one or more positions selected from the group consisting of, the peptide.
  3. 제1항에 있어서, 상기 L1 돌연변이 펩타이드는 서열번호 1의 아미노산 서열로 이루어진 펩타이드의 295 내지 300번 위치의 아미노산의 결실을 포함하는 것인, 펩타이드.The peptide of claim 1, wherein the L1 mutant peptide comprises a deletion of amino acids at positions 295 to 300 of the peptide consisting of the amino acid sequence of SEQ ID NO: 1.
  4. 제1항에 있어서, 상기 L1 돌연변이 펩타이드는 서열번호 1의 아미노산 서열로 이루어진 펩타이드의 301 내지 312번 위치의 아미노산으로 이루어진 군에서 선택된 1개 이상의 아미노산의 결실을 추가적으로 포함하는 것인, 펩타이드.The peptide of claim 1, wherein the L1 mutant peptide further comprises a deletion of one or more amino acids selected from the group consisting of amino acids at positions 301 to 312 of the peptide consisting of the amino acid sequence of SEQ ID NO: 1.
  5. 제1항에 있어서, 상기 L1 돌연변이 펩타이드는 서열번호 1의 아미노산 서열로 이루어진 펩타이드의 291 내지 294번 위치의 아미노산으로 이루어진 군에서 선택된 1개 이상의 아미노산의 결실을 추가적으로 포함하는 것인, 펩타이드.The peptide of claim 1, wherein the L1 mutant peptide further comprises a deletion of one or more amino acids selected from the group consisting of amino acids at positions 291 to 294 of the peptide consisting of the amino acid sequence of SEQ ID NO: 1.
  6. 제1항에 있어서, 상기 L1 돌연변이 펩타이드는 서열번호 1의 아미노산 서열로 이루어진 펩타이드의 413번 위치의 아스파르트산(Aspartic acid)부터 967번 위치의 세린(Serine)까지의 아미노산의 결실을 추가적으로 포함하는 것인, 펩타이드.According to claim 1, wherein the L1 mutant peptide further comprises a deletion of amino acids from aspartic acid at position 413 to serine at position 967 of the peptide consisting of the amino acid sequence of SEQ ID NO: 1 Phosphorus, peptide.
  7. 제1항에 있어서, 상기 L1 돌연변이 펩타이드는 서열번호 1의 아미노산 서열로 이루어진 펩타이드의 1 내지 252번 위치의 아미노산의 결실을 추가적으로 포함하는 것인, 펩타이드.The peptide of claim 1, wherein the L1 mutant peptide further comprises a deletion of amino acids at positions 1 to 252 of the peptide consisting of the amino acid sequence of SEQ ID NO: 1.
  8. 제7항에 있어서, 상기 L1 돌연변이 펩타이드는 서열번호 2의 아미노산 서열로 이루어진 펩타이드인 것인, 펩타이드.According to claim 7, wherein the L1 mutant peptide is a peptide consisting of the amino acid sequence of SEQ ID NO: 2, the peptide.
  9. 제1항 내지 제8항 중 어느 한 항의 펩타이드를 포함하는 근육질환 완화, 억제, 예방 또는 치료용 약제학적 조성물.A pharmaceutical composition for alleviating, inhibiting, preventing or treating muscle disease comprising the peptide of any one of claims 1 to 8.
  10. 제9항에 있어서, 상기 근육질환은 근감소증(Sarcopenia), 근위축증(Amyotrophy), 암악액질(Cancer cachexia), 근손상, 근육퇴행위축증, 심위축증, 긴장감퇴증(atony), 근이영양증(muscular dystrophy), 근육 퇴화증 및 근무력증으로 이루어진 군에서 선택된 것인, 근육질환 완화, 억제, 예방 또는 치료용 약제학적 조성물.10. The method of claim 9, wherein the muscle disease is sarcopenia, muscular atrophy (Amyotrophy), cancer cachexia (Cancer cachexia), muscle damage, muscular dystrophy, cardiac atrophy, dystonia (atony), muscular dystrophy (muscular dystrophy) , which will be selected from the group consisting of muscle degeneration and myasthenia gravis, a pharmaceutical composition for alleviating, inhibiting, preventing or treating muscle disease.
  11. 제1항 내지 제8항 중 어느 한 항의 펩타이드를 포함하는 비만 완화, 억제, 예방 또는 치료용 약제학적 조성물.A pharmaceutical composition for alleviating, suppressing, preventing or treating obesity comprising the peptide of any one of claims 1 to 8.
  12. 제1항 내지 제8항 중 어느 한 항의 펩타이드를 포함하는 당뇨병 완화, 억제, 예방 또는 치료용 약제학적 조성물.A pharmaceutical composition for alleviating, inhibiting, preventing or treating diabetes, comprising the peptide of any one of claims 1 to 8.
PCT/KR2021/018973 2020-12-14 2021-12-14 Composition for treating or preventing myopathy, obesity or diabetes WO2022131750A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
KR1020200174844A KR102486047B1 (en) 2020-12-14 2020-12-14 Compositions for treating or preventing muscle diseases, obesity or diabetes
KR10-2020-0174844 2020-12-14

Publications (1)

Publication Number Publication Date
WO2022131750A1 true WO2022131750A1 (en) 2022-06-23

Family

ID=82059316

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2021/018973 WO2022131750A1 (en) 2020-12-14 2021-12-14 Composition for treating or preventing myopathy, obesity or diabetes

Country Status (2)

Country Link
KR (1) KR102486047B1 (en)
WO (1) WO2022131750A1 (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020090373A1 (en) * 2000-03-22 2002-07-11 Leonard Buckbinder ADAMTS polypeptides, nucleic acids encoding them, and uses thereof
US20030032168A1 (en) * 1997-06-03 2003-02-13 Kunitaka Hirose Human ADAMTS-1 protein, gene encoding the same, pharmaceutical composition, and method for immunologically analyzing human ADAMTS-1 protein
WO2007147497A2 (en) * 2006-06-17 2007-12-27 Bayer Healthcare Ag Use of a disintegrin-like and metalloproteinase with thrombospondin type 1 motif (adamts1) as a therapeutic or diagnostic target

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9950043B2 (en) 2012-11-15 2018-04-24 The Board Of Trustees Of The Leland Stanford Junior University Modulation of muscle and adipocyte distribution and fate

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030032168A1 (en) * 1997-06-03 2003-02-13 Kunitaka Hirose Human ADAMTS-1 protein, gene encoding the same, pharmaceutical composition, and method for immunologically analyzing human ADAMTS-1 protein
US20020090373A1 (en) * 2000-03-22 2002-07-11 Leonard Buckbinder ADAMTS polypeptides, nucleic acids encoding them, and uses thereof
WO2007147497A2 (en) * 2006-06-17 2007-12-27 Bayer Healthcare Ag Use of a disintegrin-like and metalloproteinase with thrombospondin type 1 motif (adamts1) as a therapeutic or diagnostic target

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
DATABASE Protein 26 June 2022 (2022-06-26), ANONYMOUS : "A disintegrin and metalloproteinase with thrombospondin motifs 1 preproprotein [Homo sapiens]", XP055942839, retrieved from Genbank Database accession no. NP_008919 *
PETERS BAS J.M., RODIN ANDREI S., KLUNGEL OLAF H., STRICKER BRUNO H.CH., DE BOER ANTHONIUS, MAITLAND-VAN DER ZEE ANKE-HILSE: "Variants of ADAMTS1 modify the effectiveness of statins in reducing the risk of myocardial infarction", PHARMACOGENETICS AND GENOMICS, vol. 20, no. 12, 1 December 2010 (2010-12-01), US , pages 766 - 774, XP009537608, ISSN: 1744-6872, DOI: 10.1097/FPC.0b013e328340aded *

Also Published As

Publication number Publication date
KR20220084898A (en) 2022-06-21
KR102486047B1 (en) 2023-01-11

Similar Documents

Publication Publication Date Title
WO2022131756A1 (en) Composition for treating or preventing myopathy, obesity or diabetes
WO2018101708A1 (en) Pharmaceutical composition containing mitochondria
WO2019226015A1 (en) Composition for promoting myogenesis, containing processed ginseng extract
WO2018155997A1 (en) Use of erythropoietin-derived peptide through effect on cell damage prevention thereof
WO2020171285A1 (en) Peptide for preventing skin damage caused by air pollutants and for anti-aging, and use thereof
WO2022131750A1 (en) Composition for treating or preventing myopathy, obesity or diabetes
WO2020091463A1 (en) Pharmaceutical composition comprising isolated mitochondria for preventing or treating tendinopathy
WO2023022569A1 (en) Method for treating immune diseases by using calcineurin inhibitor and stem cells
WO2023027317A1 (en) Composition for treating muscle loss-related diseases comprising exosomes derived from tonsil mesenchymal stem cells
WO2022015106A1 (en) Composition for preventing or treating osteoarthritis, comprising mesenchymal stem cell expressing tumor necrosis factor-inducible gene 6
WO2021045567A1 (en) Composition for preventing or treating salivary gland disesaes using cell-derived vesicle
WO2021033995A1 (en) Composition comprising amomum tsaoko extract for prevention, alleviation, or treatment of sarcopenia-related disease
WO2021251790A1 (en) Deoxycholic acid-peptide conjugate having anti-obesity activity and use thereof
WO2019078381A1 (en) Pharmaceutical composition, food composition and food additive for preventing, alleviating or treating muscle loss, weakness and atrophy, containing, as active ingredient, enterococcus faecalis, culture liquid thereof or dead cells thereof
WO2015111971A1 (en) Pharmaceutical composition containing gpr119 ligand as active ingredient for preventing or treating non-alcoholic fatty liver disease
WO2024090922A1 (en) Pharmaceutical composition for preventing or treating cerebral nervous system diseases, comprising, as active ingredients, stem cell culture and extracellular vesicles isolated therefrom
WO2023219460A1 (en) Composition comprising lactobacillus fermentum strain and metabolism modulator for preventing or treating metabolic diseases by using combination therapy
WO2024122722A1 (en) Peptide for cartilage regeneration, and uses thereof
WO2024122718A1 (en) Peptide for cartilage regeneration, and uses thereof
WO2024122726A1 (en) Peptide for cartilage regeneration, and uses thereof
WO2012053758A2 (en) Pharmaceutical composition for treating or preventing alcoholic liver diseases, containing cilostazol as active ingredient
WO2024122720A1 (en) Peptide for cartilage regeneration, and uses thereof
WO2022235085A1 (en) Composition for promoting angiogenesis comprising extracellular vesicles derived from three-dimensional spheroid-type cell aggregate
WO2023085820A2 (en) Pharmaceutical composition for preventing or treating muscle diseases
WO2021215891A1 (en) Hepatic regeneration promoting composition comprising 6-o-trans-feruloyl catalpol as active ingredient

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: 21907054

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: 21907054

Country of ref document: EP

Kind code of ref document: A1