KR102161623B1 - Composition for Osteogenic Differentiation or Osteogenesis Comprising Inhibitor of CUEDC2 - Google Patents

Composition for Osteogenic Differentiation or Osteogenesis Comprising Inhibitor of CUEDC2 Download PDF

Info

Publication number
KR102161623B1
KR102161623B1 KR1020190022787A KR20190022787A KR102161623B1 KR 102161623 B1 KR102161623 B1 KR 102161623B1 KR 1020190022787 A KR1020190022787 A KR 1020190022787A KR 20190022787 A KR20190022787 A KR 20190022787A KR 102161623 B1 KR102161623 B1 KR 102161623B1
Authority
KR
South Korea
Prior art keywords
bone
cuedc2
protein
composition
differentiation
Prior art date
Application number
KR1020190022787A
Other languages
Korean (ko)
Other versions
KR20200104172A (en
Inventor
고정태
김정우
Original Assignee
전남대학교 산학협력단
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 전남대학교 산학협력단 filed Critical 전남대학교 산학협력단
Priority to KR1020190022787A priority Critical patent/KR102161623B1/en
Publication of KR20200104172A publication Critical patent/KR20200104172A/en
Application granted granted Critical
Publication of KR102161623B1 publication Critical patent/KR102161623B1/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • A61K38/1875Bone morphogenic factor; Osteogenins; Osteogenic factor; Bone-inducing factor
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/502Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing non-proliferative effects
    • G01N33/5023Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing non-proliferative effects on expression patterns
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/136Screening for pharmacological compounds
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/158Expression markers

Abstract

본 발명은 CUEDC2 억제제를 유효성분으로 포함하는 골분화 또는 골재생 촉진용 조성물 및 이를 포함하는 골질환의 예방 또는 치료용 약학 조성물, 골질환의 예방 또는 치료용 물질을 스크리닝하는 방법에 관한 것으로, 상기 CUEDC2 억제제는 골모세포의 분화 및 기질의 석회화를 유도하여 골을 재생시켜 골 손상 부위를 수복할 수 있다.The present invention relates to a composition for promoting bone differentiation or bone regeneration comprising a CUEDC2 inhibitor as an active ingredient, a pharmaceutical composition for preventing or treating bone diseases, and a method for screening a substance for preventing or treating bone diseases, the CUEDC2 inhibitors can induce differentiation of osteoblasts and calcification of the matrix to regenerate bone, thereby repairing bone damage.

Description

CUEDC2 억제제를 유효성분으로 포함하는 골분화 또는 골재생 촉진용 조성물{Composition for Osteogenic Differentiation or Osteogenesis Comprising Inhibitor of CUEDC2}Composition for promoting bone differentiation or bone regeneration comprising a CUEDC2 inhibitor as an active ingredient {Composition for Osteogenic Differentiation or Osteogenesis Comprising Inhibitor of CUEDC2}

본 발명은 CUEDC2의 발현 또는 활성을 억제하여 골모세포의 분화를 유도하는 조성물에 관한 것으로, 구체적으로 CUEDC2 억제제를 유효성분으로 포함하는 골분화 또는 골재생 촉진용 조성물 및 이를 포함하는 골질환의 예방 또는 치료용 약학 조성물, 골질환의 예방 또는 치료용 물질을 스크리닝하는 방법에 관한 것이다.The present invention relates to a composition for inducing the differentiation of osteoblasts by inhibiting the expression or activity of CUEDC2, and specifically, a composition for promoting bone differentiation or bone regeneration comprising the CUEDC2 inhibitor as an active ingredient, and prevention of bone disease comprising the same or It relates to a therapeutic pharmaceutical composition, a method for screening a substance for preventing or treating bone diseases.

뼈는 생체의 골격을 이루는 단단한 조직으로 인체의 연조직과 체중을 지탱해주며 내부 장기를 둘러싸서 외부의 충격으로부터 보호하고, 체내 칼슘이나 다른 필수 무기질인 인, 마그네슘 등과 같은 이온들의 저장고로서 이온의 양을 조절하고 유지하는 기능을 한다. 이러한 뼈는 골모 전구 세포(osteoprogenitor cell), 골모세포(osteoblast), 파골세포(osteoclast) 등 뼈 조직을 이루는 세포와 골 시알로단백질(bone sialoprotein), 오스테오칼신(osteocalcin), 콜라겐 I 타입(collagen I type) 등 뼈 기질을 이루는 기질 단백(matrix protein)으로 구성된다. 성장이 끝난 성인의 경우에는 오래된 뼈가 제거되고 새로운 뼈로 대체되는 생성 및 흡수 과정이 매우 역동적, 지속적으로 반복하여 뼈가 재생되는데, 이를 골재형성(bone remodeling)이라고 한다. 오래된 뼈를 제거하고 새로운 뼈로 대체하는 뼈의 순환(turnover)은 성장과 스트레스에 의해서 일어나는 뼈의 미세한 손상을 회복시키고 적절히 뼈의 기능을 유지하는데 필수적이라고 할 수 있다. Bone is a hard tissue that forms the skeleton of a living body, supports the soft tissue and weight of the human body, surrounds internal organs to protect from external shocks, and is a storage of ions such as calcium and other essential minerals such as phosphorus and magnesium in the body. It functions to regulate and maintain. These bones include cells that make up bone tissue such as osteoprogenitor cells, osteoblasts, and osteoclasts, and bone sialoprotein, osteocalcin, and collagen I type. ), etc. It is composed of matrix proteins that make up the bone matrix. In the case of grown-up adults, the process of generation and absorption in which old bones are removed and replaced with new bones is very dynamic, and bone is regenerated continuously, which is called bone remodeling. Bone turnover, which removes old bones and replaces them with new bones, is essential to recovering microscopic damage to bones caused by growth and stress, and maintaining proper bone function.

골재형성에는 크게 두 종류의 세포가 관여하는 것으로 알려져 있는데, 두 세포 중 하나는 뼈를 생성하는 골모세포이고, 다른 하나는 뼈를 파괴하는 파골세포이다. 골모세포는 신호분자나 단백질을 분비하여 세포 주위 기질의 석회화를 유도함으로써 뼈를 형성한다. 뼈의 작은 흠집이나 균열 등은 이러한 골모세포의 존재로 인해 원래의 조직 상태로 재생되므로 크게 문제가 되지 않는다. 그러나 교통사고, 낙상과 같은 외부적인 요인 또는 당뇨병과 같은 병인에 의해 광범위한 골조직에 손상을 입게 되면 골모세포에 의한 자연적 골재생이 불가능해지고 외과적 처치 및 골재생 촉진을 위한 적극적 치료과정을 받아야만 한다. It is known that two types of cells are largely involved in bone remodeling. One of the two cells is an osteoblast that produces bone, and the other is an osteoclast that destroys bone. Osteoblasts secrete signaling molecules or proteins to induce calcification of the matrix around the cells to form bone. Small scratches or cracks on the bone are not a big problem because they are regenerated to their original tissue state due to the presence of these osteoblasts. However, if extensive bone tissue is damaged by external factors such as traffic accidents, falls, or etiology such as diabetes, natural bone regeneration by osteoblasts becomes impossible, and surgical treatment and active treatment to promote bone regeneration must be performed.

현재의 골재생 치료법으로는 손상된 부위에 골 조직을 이식하거나 (자가골 이식, 이종골 이식, 합성골 이식), 또는 금속, 고분자, 세라믹으로 만들어진 인공뼈를 이식하는 방식이 있다. 그러나 자가골 조직 이식은 환자의 신체 손상이나 채취 양에서 한계를 가지고 있기 때문에 대부분 합성골의 성분 또는 인공뼈를 개인 맞춤형으로 개량하는 연구가 활발하게 진행되고 있다. 하지만 이러한 골대체제들은 원래의 골 조직과 성분이 다르고 주위 세포들이 골대체제 안쪽으로 들어가지 못하여 영양과 산소공급에 중요한 혈관이 생성되지 않는다. 최근에는 BMP2(bone morphogenetic protein 2)나 PTH(parathyroid hormone)와 같은 분비 단백과, Runx2(runt-related transcription factor 2)와 같은 전사인자 등을 이용하는 뼈 재생 연구가 알려지고 있다. Current bone regeneration treatments include transplanting bone tissue to the damaged area (autologous bone graft, xenograft, synthetic bone graft), or artificial bone made of metal, polymer, or ceramic. However, since autologous bone tissue transplantation has limitations in the amount of damage or collection of the patient's body, most of the researches on improving the components of synthetic bones or artificial bones customized to individuals are being actively conducted. However, these bone substitutes differ in composition from the original bone tissue, and the surrounding cells do not enter the bone substitute, so blood vessels important for nutrition and oxygen supply are not created. Recently, bone regeneration studies using secreted proteins such as BMP2 (bone morphogenetic protein 2) or PTH (parathyroid hormone), and transcription factors such as Runx2 (runt-related transcription factor 2) have been known.

한국등록특허 제1570832호Korean Patent No. 1570832 한국등록특허 제1762580호Korean Patent Registration No.1762580

상기한 문제점을 해결하기 위해, 본 발명은 CUEDC2의 발현 또는 활성 억제제를 유효성분으로 포함하는 골분화 또는 골재생 촉진용 조성물을 제공하는 것을 목적으로 한다.In order to solve the above problems, an object of the present invention is to provide a composition for promoting bone differentiation or bone regeneration comprising an inhibitor of CUEDC2 expression or activity as an active ingredient.

여기서, 상기 CUEDC2의 발현 또는 활성 억제제는 CUEDC2의 mRNA에 상보적으로 결합하는 안티센스 올리고뉴클레오티드, siRNA 및 shRNA로 이루어진 군에서 선택된 1종 이상일 수 있다. 이때, 상기 shRNA는 레트로바이러스 벡터 또는 아데노바이러스 벡터 시스템을 이용하는 것일 수 있다.Here, the expression or activity inhibitor of CUEDC2 may be at least one selected from the group consisting of antisense oligonucleotides, siRNAs, and shRNAs complementarily binding to CUEDC2 mRNA. In this case, the shRNA may be a retroviral vector or an adenovirus vector system.

상기 CUEDC2의 발현 또는 활성 억제제는 골형성 촉진, 골분화 촉진 또는 기질 석회화 유도의 작용을 갖는 것일 수 있다.The expression or activity inhibitor of CUEDC2 may be one having an action of promoting bone formation, promoting bone differentiation, or inducing matrix calcification.

상기 조성물은 인간 또는 쥐에서 유래된 골 시알로단백질(bone sialoprotein), 오스테오칼신(osteocalcin) 또는 BMP2(bone morphogenetic protein 2)를 더 포함하는 것일 수 있다.The composition may further include bone sialoprotein, osteocalcin, or bone morphogenetic protein 2 (BMP2) derived from human or rat.

또한, 본 발명은 상기 골분화 또는 골재생 촉진용 조성물을 유효성분으로 포함하는 골질환의 예방 또는 치료용 약학 조성물을 제공하는 것을 목적으로 한다.In addition, an object of the present invention is to provide a pharmaceutical composition for preventing or treating bone diseases comprising the composition for promoting bone differentiation or bone regeneration as an active ingredient.

여기서, 상기 골질환은 골결손, 골다공증, 골연화증, 골감소증, 골형성 부전증, 골형성 장애, 골관절염, 골괴사, 퇴행성 골질환, 골 관절염, 전이성 골암, 골종양, 치주질환, 골절 및 파제트병으로 이루어진 군으로부터 선택되는 것일 수 있다.Here, the bone disease is a group consisting of bone defects, osteoporosis, osteomalacia, osteopenia, bone insufficiency, bone formation disorders, osteoarthritis, bone necrosis, degenerative bone disease, osteoarthritis, metastatic bone cancer, bone tumors, periodontal disease, fractures and Paget's disease It may be selected from.

또한, 본 발명은 (a) CUEDC2 유전자 또는 CUEDC2 단백질을 포함하는 세포에 분석하고자 하는 시험물질을 접촉시키는 단계; (b) 상기 CUEDC2 유전자의 발현량, CUEDC2 단백질의 발현량 또는 CUEDC2 단백질의 활성도를 측정하는 단계; 및 (c) 상기 (b) 단계의 측정 결과에서 시험물질이 CUEDC2 유전자의 발현량, CUEDC2 단백질의 발현량 또는 CUEDC2 단백질의 활성도를 감소시킬 경우, 상기 시험물질을 골질환의 예방 또는 치료용 물질로 판정하는 단계를 포함하는 골질환의 예방 또는 치료용 물질을 스크리닝하는 방법을 제공하는 것을 목적으로 한다.In addition, the present invention comprises the steps of (a) contacting a test substance to be analyzed with a cell containing the CUEDC2 gene or CUEDC2 protein; (b) measuring the expression level of the CUEDC2 gene, the expression level of the CUEDC2 protein, or the activity of the CUEDC2 protein; And (c) in the measurement result of step (b), when the test material decreases the expression level of the CUEDC2 gene, the expression level of the CUEDC2 protein, or the activity of the CUEDC2 protein, the test material is used as a substance for preventing or treating bone diseases. An object of the present invention is to provide a method for screening a substance for preventing or treating bone diseases, including the step of determining.

여기서, 상기 시험물질은 안티센스 올리고뉴클레오티드, siRNA, shRNA, miRNA, 리보자임, DNAzyme, PNA, 항체, 앱타머, 천연 추출물 및 합성 화합물로 이루어진 군에서 선택되는 것일 수 있다.Here, the test substance may be selected from the group consisting of antisense oligonucleotide, siRNA, shRNA, miRNA, ribozyme, DNAzyme, PNA, antibody, aptamer, natural extract, and synthetic compound.

상기 (b) 단계의 측정은 역전사 중합효소 연쇄반응(reverse transcriptase-polymerase chain reaction), 실시간 중합효소 연쇄반응(real time-polymerase chain reaction), 웨스턴 블롯(western blot), 노던 블롯(northern blot), ELISA(enzyme linked immunosorbent assay), 방사선면역분석(radioimmunoassay), 방사 면역 확산법(radioimmunodiffusion), 면역침전분석법(immunoprecipitation assay) 및 면역조직화학적 분석(immunohistochemical analysis)으로 이루어진 군에서 선택되는 것일 수 있다.The measurement of step (b) is reverse transcriptase-polymerase chain reaction, real time-polymerase chain reaction, western blot, northern blot, ELISA (enzyme linked immunosorbent assay), radioimmunoassay, radioimmunodiffusion, immunoprecipitation assay, and immunohistochemical analysis (immunohistochemical analysis) It may be selected from the group consisting of.

<골분화 또는 골재생 촉진용 조성물><Composition for promoting bone differentiation or bone regeneration>

본 발명의 일 구체예에 따르면, 본 발명은 CUEDC2의 발현 또는 활성 억제제를 유효성분으로 포함하는 골분화 또는 골재생 촉진용 조성물을 제공한다.According to one embodiment of the present invention, the present invention provides a composition for promoting bone differentiation or bone regeneration, comprising an inhibitor of CUEDC2 expression or activity as an active ingredient.

CUEDC2(CUE domain-containing protein 2)는 기능이 명확하게 밝혀지지 않았으나, CUE 도메인을 포함하는 유비퀴틴화(ubiquitination)와 관련된 단백질로서 염증성 사이토카인 발현, 세포 증식, 활성산소 생성에 이르기까지 다양하게 작용하는 것으로 알려져 있다.CUEDC2 (CUE domain-containing protein 2) is a protein related to ubiquitination containing the CUE domain, although its function has not been clearly identified. It acts in a variety of ways from inflammatory cytokine expression, cell proliferation, and free radical production. It is known.

본 발명자들은 골모세포의 분화를 유도하는 새로운 생체분자를 개발하고자 노력한 결과, CUEDC2 유전자 또는 단백질의 발현을 억제함으로써 골모세포의 분화를 유도하고 기질의 석회화를 촉진하여 골질환을 예방 또는 치료할 수 있다는 것을 확인하였다. 이에 따라, 본 발명에서는 골모세포의 분화를 유도하고 기질의 석회화를 촉진하는데 CUEDC2의 발현 또는 활성을 저해하는 억제제를 적용하였다.As a result of trying to develop a new biomolecule that induces the differentiation of osteoblasts, the present inventors have found that it is possible to prevent or treat bone diseases by inducing the differentiation of osteoblasts and promoting calcification of the matrix by inhibiting the expression of the CUEDC2 gene or protein. Confirmed. Accordingly, in the present invention, an inhibitor that inhibits the expression or activity of CUEDC2 was applied to induce the differentiation of osteoblasts and promote calcification of the matrix.

상기 CUEDC2 유전자는 통상의 기술분야에 알려진 어떠한 서열로 구성되는 폴리뉴클레오티드를 포함할 수 있으며, 서열번호 1의 염기서열로 구성된 폴리뉴클레오티드일 수 있다. The CUEDC2 gene may include a polynucleotide composed of any sequence known in the art, and may be a polynucleotide composed of the nucleotide sequence of SEQ ID NO: 1.

상기 CUEDC2의 발현 또는 활성을 저해하는 억제제 (이하, 'CUEDC2 억제제'라 함)는 CUEDC2의 mRNA 발현 또는 단백질 활성을 감소시키는 물질을 통칭한다. 보다 구체적으로는, CUEDC2에 직접적으로 작용하거나 그의 리간드 또는 수용체에 간접적으로 작용하는 등의 방식을 통해 CUEDC2의 발현을 전사 수준에서 감소시키거나 그 활성을 방해함으로써 CUEDC2의 발현 또는 활성을 감소시키는 모든 물질을 포함할 수 있다. 상기 CUEDC2 발현을 저해하는 물질은 CUEDC2를 표적으로 하여 CUEDC2의 발현 또는 활성을 억제할 수 있는 화합물, 핵산, 펩티드, 바이러스 또는 상기 핵산을 포함하는 벡터 등 그 형태에 제한없이 사용 가능하다. CUEDC2 발현을 저해하는 물질의 예로는 CUEDC2의 mRNA에 특이적으로 결합하여 CUEDC2 mRNA의 발현을 저해하는 안티센스 올리고뉴클레오티드, siRNA, shRNA, miRNA, 리보자임(ribozyme), DNAzyme, PNA(peptide nucleic acids) 등을 들 수 있다. CUEDC2 활성을 저해하는 물질의 예로는 CUEDC2의 단백질에 특이적으로 결합하여 CUEDC2 단백질의 활성을 억제하는 항체 또는 그의 항원 결합 단편, 앱타머(aptamer), 화합물 등을 들 수 있다. 바람직하게는, 본 발명에 따른 CUEDC2 억제제는 CUEDC2의 mRNA에 상보적으로 결합하는 안티센스 올리고뉴클레오티드, siRNA 및 shRNA로 이루어진 군에서 선택된 1종 이상일 수 있다.The inhibitor that inhibits the expression or activity of CUEDC2 (hereinafter referred to as'CUEDC2 inhibitor') refers to a substance that reduces the mRNA expression or protein activity of CUEDC2. More specifically, any substance that reduces the expression or activity of CUEDC2 by reducing the expression of CUEDC2 at the transcriptional level or interfering with its activity through a method such as acting directly on CUEDC2 or indirectly acting on its ligand or receptor. It may include. The substance that inhibits CUEDC2 expression may be used without limitation in its form, such as a compound, nucleic acid, peptide, virus, or vector containing the nucleic acid capable of inhibiting the expression or activity of CUEDC2 by targeting CUEDC2. Examples of substances that inhibit CUEDC2 expression include antisense oligonucleotides, siRNA, shRNA, miRNA, ribozyme, DNAzyme, and peptide nucleic acids (PNA) that specifically bind to CUEDC2 mRNA and inhibit the expression of CUEDC2 mRNA. Can be mentioned. Examples of substances that inhibit CUEDC2 activity include antibodies or antigen-binding fragments thereof, aptamers, and compounds that specifically bind to CUEDC2 protein and inhibit the activity of CUEDC2 protein. Preferably, the CUEDC2 inhibitor according to the present invention may be at least one selected from the group consisting of antisense oligonucleotides, siRNAs, and shRNAs that bind complementarily to CUEDC2 mRNA.

상기 "안티센스 올리고뉴클레오티드(antisense oligonucleotide)"는 특정 mRNA의 서열에 상보적인 핵산 서열을 함유하고 있는 DNA, RNA 또는 이들의 유도체를 의미하는데, mRNA 내의 상보적인 서열에 결합하여 mRNA의 단백질로의 번역을 저해하는 효과를 나타낸다.The "antisense oligonucleotide" refers to DNA, RNA, or derivatives thereof that contain a nucleic acid sequence that is complementary to a specific mRNA sequence. By binding to a complementary sequence in the mRNA, translation of the mRNA into a protein is performed. It has an inhibitory effect.

상기 "siRNA(small interfering RNA)"는 특정 mRNA의 절단(cleavage)을 통하여 RNA 간섭을 유도할 수 있는 핵산 분자로서, 21 내지 25 뉴클레오티드 크기의 짧은 이중가닥 RNA를 말한다. 본 발명에 따른 siRNA는 센스 가닥(CUEDC2 mRNA 서열에 상응하는 서열)과 안티센스 가닥(CUEDC2 mRNA 서열에 상보적인 서열)이 서로 반대쪽에 위치하여 이중쇄를 이루는 구조를 가질 수 있으며, 자기-상보성(self-complementary) 센스 및 안티센스 가닥을 가지는 단일쇄 구조를 가질 수 있다. The "siRNA (small interfering RNA)" refers to a nucleic acid molecule capable of inducing RNA interference through cleavage of a specific mRNA, and refers to a short double-stranded RNA having a size of 21 to 25 nucleotides. The siRNA according to the present invention may have a structure in which a sense strand (a sequence corresponding to a CUEDC2 mRNA sequence) and an antisense strand (a sequence complementary to a CUEDC2 mRNA sequence) are located on opposite sides to form a double chain, and self-complementary (self -complementary) may have a single-chain structure with sense and antisense strands.

상기 "shRNA(short hairpin RNA)"는 siRNA와 같이 특정 mRNA에 상보적으로 결합하여 루프 영역에 의해 회문적으로 연결된 헤어핀 구조를 형성함으로써 그 mRNA를 절단하는 이중 가닥 RNA이다. siRNA 생합성시 높은 비용, 낮은 세포 형질감염 효율로 인한 RNA 간섭 효과의 단시간 유지 등의 siRNA의 단점을 극복하기 위해 개발된 것으로, shRNA는 RNA 중합효소 Ⅲ의 프로모터로부터 레트로바이러스 벡터(retroviral vector), 아데노바이러스(adenoviral vector), 렌티 바이러스(lentivirus vector), 플라스미드 발현 벡터(plasmid expression vector) 등의 시스템을 이용하여 이를 세포 내로 도입하여 발현시키는 방식을 이용한다. 바람직하게는, shRNA는 레트로바이러스 벡터 또는 아데노바이러스 벡터 시스템을 이용하는 것일 수 있다.The "shRNA (short hairpin RNA)" is a double-stranded RNA that binds complementarily to a specific mRNA like siRNA and cuts the mRNA by forming a hairpin structure palindromicly linked by a loop region. It was developed to overcome the shortcomings of siRNA such as high cost during siRNA biosynthesis and short-term maintenance of RNA interference effects due to low cell transfection efficiency. shRNA is a retroviral vector, adeno from the promoter of RNA polymerase III. A virus (adenoviral vector), a lentivirus (lentivirus vector), a plasmid expression vector (plasmid expression vector) system, such as using a system that is introduced into the cell and expressed. Preferably, the shRNA may be one using a retroviral vector or an adenovirus vector system.

본 발명에 따른 CUEDC2 억제제를 포함하는 골분화 또는 골재생 촉진용 조성물은 CUEDC2의 mRNA 또는 단백질의 발현을 감소시키고 발현된 단백질의 활성을 감소시킴으로써 우수한 골형성 촉진, 골분화 촉진 또는 석회화 유도 효과를 나타낼 수 있다. 이러한 효과를 더욱 향상시키기 위해, 상기 조성물은 CUEDC2 억제제와 함께 골형성 관련 단백질을 더 포함할 수 있다. 골형성 관련 단백질로는 골모세포의 분화 과정에서 Runx2, 오스테릭스(osterix)와 같은 전사인자(transcription factor)에 의해 발현이 증가되어 골모세포에 의한 골형성을 돕는 골 시알로단백질(bone sialoprotein) 또는 오스테오칼신(osteocalcin)이거나, 또는 골 형성을 유도하는 사이토카인인 BMP 패밀리(bone morphogenetic protein family)일 수 있다. 이러한 단백질은 인간 또는 쥐에서 유래된 것일 수 있다.The composition for promoting bone differentiation or bone regeneration comprising a CUEDC2 inhibitor according to the present invention reduces the expression of CUEDC2 mRNA or protein and reduces the activity of the expressed protein, thereby promoting excellent bone formation, promoting bone differentiation, or inducing calcification. I can. In order to further enhance this effect, the composition may further include a protein related to bone formation together with a CUEDC2 inhibitor. The bone formation-related protein is bone sialoprotein, which helps bone formation by osteoblasts by increasing expression by transcription factors such as Runx2 and Osterix during the differentiation of osteoblasts. It may be osteocalcin or may be a bone morphogenetic protein family, a cytokine that induces bone formation. These proteins can be derived from humans or mice.

<골질환의 예방 또는 치료용 약학 조성물><Pharmaceutical composition for preventing or treating bone disease>

본 발명의 다른 일 구체예에 따르면, 본 발명은 CUEDC2의 발현 또는 활성 억제제를 유효성분으로 포함하는 골질환의 예방 또는 치료용 약학 조성물을 제공한다.According to another embodiment of the present invention, the present invention provides a pharmaceutical composition for preventing or treating bone diseases comprising an inhibitor of expression or activity of CUEDC2 as an active ingredient.

상기 CUEDC2의 발현 또는 활성 억제제는 전술한 바와 동일하므로, 중복된 설명을 피하기 위해 생략한다.Since the CUEDC2 expression or activity inhibitor is the same as described above, it is omitted to avoid redundant description.

본 발명에 따른 약학 조성물은 골결손, 골다공증, 골연화증, 골감소증, 골형성 부전증, 골형성 장애, 골관절염, 골괴사, 퇴행성 골질환, 골 관절염, 전이성 골암, 골종양, 치주질환, 골절, 파제트병 등의 골질환을 예방하거나 치료하는데 사용될 수 있다.The pharmaceutical composition according to the present invention includes bone defects, osteoporosis, osteomalacia, osteopenia, bone insufficiency, bone formation disorders, osteoarthritis, bone necrosis, degenerative bone disease, osteoarthritis, metastatic bone cancer, bone tumors, periodontal disease, fractures, Paget's disease, etc. It can be used to prevent or treat bone disease.

이러한 약학 조성물은 약제학적으로 허용 가능한 첨가제를 더 포함할 수 있으며, 이때 약제학적으로 허용 가능한 첨가제로는 전분, 젤라틴화 전분, 미결정셀룰로오스, 유당, 포비돈, 콜로이달실리콘디옥사이드, 인산수소칼슘, 락토스, 만니톨, 엿, 아라비아고무, 전호화전분, 옥수수전분, 분말셀룰로오스, 히드록시프로필셀룰로오스, 오파드라이, 전분글리콜산나트륨, 카르나우바 납, 합성규산알루미늄, 스테아린산, 스테아린산마그네슘, 스테아린산알루미늄, 스테아린산칼슘, 백당, 덱스트로스, 소르비톨, 탈크 등이 사용될 수 있다. 상기 허용 가능한 첨가제는 상기 약학 조성물에 대해 0.1 중량부 내지 90 중량부 포함될 수 있다.Such pharmaceutical compositions may further include pharmaceutically acceptable additives, wherein pharmaceutically acceptable additives include starch, gelatinized starch, microcrystalline cellulose, lactose, povidone, colloidal silicon dioxide, calcium hydrogen phosphate, lactose, Mannitol, syrup, gum arabic, pregelatinized starch, corn starch, powdered cellulose, hydroxypropyl cellulose, Opadry, sodium starch glycolate, lead carnauba, synthetic aluminum silicate, stearic acid, magnesium stearate, aluminum stearate, calcium stearate, Sucrose, dextrose, sorbitol, talc, and the like can be used. The acceptable additive may be included in an amount of 0.1 to 90 parts by weight based on the pharmaceutical composition.

또한, 약학 조성물은 목적하는 방법에 따라 비경구 투여(일례로, 정맥 내, 피하, 복강 내 또는 국소에 적용)하거나 경구 투여할 수 있다. 비경구 투여를 위한 제제에는 멸균된 수용액, 비수성 용제, 현탁제, 유제, 동결건조 제제, 좌제 등이 포함된다. 경구 투여를 위한 액상 제제로는 현탁제, 내용액제, 유제, 시럽제 등이 해당되는데, 통상적으로 사용되는 단순 희석제인 물, 액체 파라핀 이외에 다양한 부형제, 예컨대 습윤제, 감미제, 방향제, 보존제 등이 함께 포함될 수 있다. 이때 투여량은 환자의 체중, 연령, 성별, 건강상태, 식이, 투여시간, 투여방법, 배설률, 질환의 중증도 등에 따라 그 범위가 다양하다. 본 발명에 따른 약학 조성물의 일일 투여량은 0.5 ㎎/kg 내지 70 ㎎/kg이며, 하루 일 회 내지 수회에 나누어 투여할 수 있다.In addition, the pharmaceutical composition may be parenterally administered (for example, intravenously, subcutaneously, intraperitoneally or topically applied) or orally according to a desired method. Formulations for parenteral administration include sterile aqueous solutions, non-aqueous solvents, suspensions, emulsions, lyophilized formulations, suppositories, and the like. Liquid formulations for oral administration include suspensions, liquid solutions, emulsions, syrups, etc., and various excipients, such as wetting agents, sweeteners, fragrances, preservatives, etc., may be included in addition to water and liquid paraffin, which are commonly used simple diluents. have. At this time, the dosage range varies according to the patient's weight, age, sex, health status, diet, administration time, administration method, excretion rate, and severity of disease. The daily dosage of the pharmaceutical composition according to the present invention is 0.5 mg/kg to 70 mg/kg, and may be administered once to several times a day.

<골질환의 예방 또는 치료용 물질을 스크리닝하는 방법><Method of screening substances for preventing or treating bone diseases>

본 발명의 다른 일 구체예에 따르면, 본 발명은 골질환의 예방 또는 치료용 물질을 스크리닝하는 방법을 제공한다. 구체적으로, 스크리닝 방법은 (a) CUEDC2 유전자 또는 CUEDC2 단백질을 포함하는 세포에 분석하고자 하는 시험물질을 접촉시키는 단계; (b) 상기 CUEDC2 유전자의 발현량, CUEDC2 단백질의 발현량 또는 CUEDC2 단백질의 활성도를 측정하는 단계; 및 (c) 상기 (b) 단계의 측정 결과에서 시험물질이 CUEDC2 유전자의 발현량, CUEDC2 단백질의 발현량 또는 CUEDC2 단백질의 활성도를 감소시킬 경우, 상기 시험물질을 골질환의 예방 또는 치료용 물질로 판정하는 단계로 진행된다. According to another embodiment of the present invention, the present invention provides a method for screening a substance for preventing or treating bone diseases. Specifically, the screening method includes the steps of: (a) contacting the cell containing the CUEDC2 gene or CUEDC2 protein with a test substance to be analyzed; (b) measuring the expression level of the CUEDC2 gene, the expression level of the CUEDC2 protein, or the activity of the CUEDC2 protein; And (c) in the measurement result of step (b), when the test material decreases the expression level of the CUEDC2 gene, the expression level of the CUEDC2 protein, or the activity of the CUEDC2 protein, the test material is used as a substance for preventing or treating bone diseases. It proceeds to the determining step.

상기 (a) 단계는 CUEDC2를 발현하는 세포에 분석하고자 하는 시험물질을 접촉시킨다. 상기 CUEDC2를 발현하는 세포는 CUEDC2를 내인적으로(endogenous) 발현하는 세포 또는 일시적으로(transiently) CUEDC2가 과발현된 세포를 포함하며, 골조직(osseous tissue) 유래 세포인 것이 바람직하다. 상기 "시험물질"은 CUEDC2의 발현에 영향을 미치는지 여부를 검사하기 위하여 스크리닝에서 이용되는 미지의 물질을 의미한다. 상기 시험물질은 안티센스 올리고뉴클레오티드, siRNA, shRNA, miRNA, 리보자임, DNAzyme, PNA, 항체, 앱타머, 천연 추출물, 합성 화합물 등일 수 있다.In step (a), the test substance to be analyzed is brought into contact with the cells expressing CUEDC2. The cells expressing CUEDC2 include cells that endogenous CUEDC2 or cells transiently overexpressing CUEDC2, and are preferably osseous tissue-derived cells. The "test substance" means an unknown substance used in screening to test whether it affects the expression of CUEDC2. The test substance may be an antisense oligonucleotide, siRNA, shRNA, miRNA, ribozyme, DNAzyme, PNA, antibody, aptamer, natural extract, synthetic compound, and the like.

상기 (b) 단계는 시험물질이 처리된 세포에서 CUEDC2 mRNA 또는 단백질의 발현량, CUEDC2 단백질의 활성도를 분석한다. 이때, 역전사 중합효소 연쇄반응(reverse transcriptase-polymerase chain reaction), 실시간 중합효소 연쇄반응(real time-polymerase chain reaction), 웨스턴 블롯(western blot), 노던 블롯(northern blot), ELISA(enzyme linked immunosorbent assay), 방사선면역분석(RIA: radioimmunoassay), 방사 면역 확산법(radioimmunodiffusion), 면역침전분석법(immunoprecipitation assay), 면역조직화학적 분석(immunohistochemical analysis) 등의 방법으로 통해 발현량 또는 활성도를 측정하여 분석할 수 있다.In the step (b), the expression level of CUEDC2 mRNA or protein and the activity of CUEDC2 protein are analyzed in the cells treated with the test substance. At this time, reverse transcriptase-polymerase chain reaction, real time-polymerase chain reaction, western blot, northern blot, enzyme linked immunosorbent assay (ELISA) ), radioimmunoassay (RIA), radioimmunodiffusion, immunoprecipitation assay, immunohistochemical analysis, etc., the expression level or activity can be measured and analyzed. .

상기 (c) 단계는 상기 측정 결과에 따라 시험물질이 CUEDC2 mRNA의 발현량, CUEDC2 단백질의 발현량 또는 CUEDC2 단백질의 활성도를 감소 또는 억제시킬 경우, 상기 시험물질을 골질환 치료제로 판단한다.In the step (c), when the test material reduces or inhibits the expression level of CUEDC2 mRNA, the expression level of CUEDC2 protein, or the activity of CUEDC2 protein according to the measurement result, the test material is determined as a treatment for bone disease.

본 발명에 따른 CUEDC2 억제제를 포함하는 조성물은 골모세포의 효율적인 분화를 유도함으로써 교통사고, 낙상 등에 의한 골 손실이 발생한 경우에 인공뼈로 손실된 뼈를 대체하는 기존 방식과는 다르게, 골모세포의 분화 및 기질의 석회화를 직접적으로 유도하여 뼈를 재생시켜 손실된 뼈를 수복할 수 있다. 이에 따라, 뼈 손상을 수복하는데 드는 비용과 기간을 현저히 줄일 수 있다. 한편, 보통의 인공뼈는 시술 부위의 예기치 못한 염증반응으로 인해 인공뼈와 환자의 뼈 사이의 골 유착이 제대로 이루어지지 않아 재수술을 요할 수 있으나, 본 발명에 따른 CUEDC2 억제제를 사용할 경우에는 이러한 문제가 발생하지 않는다. 또한, 임플란트 시술시 본 발명에 따른 CUEDC2 억제제를 포함하는 조성물을 처리함으로써 치조골의 불량골질 및 부족한 골량으로 인한 골 유착의 결여를 방지할 수 있다.The composition containing the CUEDC2 inhibitor according to the present invention induces the efficient differentiation of osteoblasts, unlike the conventional method of replacing the lost bone with artificial bone in case of bone loss due to a traffic accident, a fall, etc., differentiation of osteoblasts And it is possible to directly induce calcification of the substrate to regenerate the bone, thereby repairing the lost bone. Accordingly, it is possible to significantly reduce the cost and duration required to repair the bone damage. On the other hand, normal artificial bones may require reoperation because bone adhesion between the artificial bone and the patient's bone is not properly achieved due to an unexpected inflammatory reaction at the treatment site. However, this problem occurs when the CUEDC2 inhibitor according to the present invention is used. Does not occur. In addition, by treating the composition containing the CUEDC2 inhibitor according to the present invention during an implant procedure, it is possible to prevent the lack of bone adhesion due to poor bone quality and insufficient bone mass of the alveolar bone.

도 1은 본 발명의 일 구체예에 대한 실험 결과이다: (a) 골모세포 분화에 따른 CUEDC2, 골 시아로단백질(Bsp) 및 오스테오칼신(Oc) mRNA의 발현 정도를 qPCR로 측정한 그래프이다 (b) 골모세포 분화에 따른 CUEDC2, 골 시아로단백질(Bsp) 및 오스테오칼신(Oc) 단백질의 발현 정도를 웨스턴 블롯으로 측정한 사진이다.
도 2는 본 발명의 일 구체예에 대한 실험 결과로, CUEDC2 과발현시 골 시아로단백질(Bsp) 및 오스테오칼신(Oc) mRNA의 발현 정도를 qPCR로 측정한 그래프이다.
도 3은 본 발명의 일 구체예에 대한 실험 결과로, CUEDC2 과발현시 석회화 억제능을 확인하기 위해 (a) ALP 염색, (b) AR 염색 및 (c) 이소성 골형성 정도를 측정한 그래프 및 사진이다.
도 4는 본 발명의 일 구체예에 대한 실험 결과로, CUEDC2 발현을 감소시켰을 때 골 시아로단백질(Bsp) 및 오스테오칼신(Oc) mNRA의 발현 정도를 qPCR로 측정한 그래프이다.
도 5는 본 발명의 일 구체예에 대한 실험 결과로, CUEDC2의 발현을 감소시켰을 때 석회화 유도능을 확인하기 위해 (a) ALP 염색, (b) AR 염색 및 (c) 두개골 골형성 정도를 측정한 그래프 및 사진이다.
Figure 1 is an experimental result for one embodiment of the present invention: (a) a graph measuring the expression levels of CUEDC2, bone cyanoprotein (Bsp), and osteocalcin (Oc) mRNA according to osteoblast differentiation by qPCR (b ) This is a photograph of measuring the expression levels of CUEDC2, bone cyanoprotein (Bsp), and osteocalcin (Oc) proteins according to osteoblast differentiation by Western blot.
2 is an experimental result for an embodiment of the present invention, a graph measuring the expression level of bone cyanoprotein (Bsp) and osteocalcin (Oc) mRNA by qPCR when CUEDC2 is overexpressed.
3 is an experimental result for an embodiment of the present invention, graphs and photographs measuring the degree of (a) ALP staining, (b) AR staining, and (c) ectopic bone formation in order to confirm the ability to inhibit calcification upon overexpression of CUEDC2. .
4 is an experimental result for an embodiment of the present invention, when CUEDC2 expression is reduced, the expression level of bone cyanoprotein (Bsp) and osteocalcin (Oc) mNRA is measured by qPCR.
5 is an experimental result for an embodiment of the present invention, in order to confirm the ability to induce calcification when the expression of CUEDC2 is reduced, (a) ALP staining, (b) AR staining, and (c) measuring the degree of skull bone formation One is a graph and a picture.

이하, 첨부된 도면을 참조하며 본 발명에 따른 골분화 또는 골재생 촉진용 조성물을 보다 상세하게 설명한다. 그러나, 이러한 설명은 본 발명의 이해를 돕기 위하여 예시적으로 제시된 것일 뿐, 본 발명의 범위가 이러한 예시적인 설명에 의하여 제한되는 것은 아니다.Hereinafter, a composition for promoting bone differentiation or bone regeneration according to the present invention will be described in more detail with reference to the accompanying drawings. However, these descriptions are provided by way of example only to aid understanding of the present invention, and the scope of the present invention is not limited by these exemplary descriptions.

1. 재료 및 방법1. Materials and methods

1-1. 세포 배양 및 바이러스 감염1-1. Cell culture and viral infection

골모세포로 분화할 수 있는 MC3T3-E1 세포를 사용하였다. 세포는 10% 소태아혈청(fetal bovine serum; FBS), 페니실린(penicillin) 100 U/mL, 스트렙토마이신(streptomycin) 100 mg/mL를 포함한 α-MEM(α-minimal essential medium) 배지에서 배양하였다. MC3T3-E1 cells capable of differentiating into osteoblasts were used. Cells were cultured in α-MEM (α-minimal essential medium) medium containing 10% fetal bovine serum (FBS), 100 U/mL penicillin, and 100 mg/mL streptomycin.

위 세포에서 분화를 유도하기 위해, 세포를 1주에서 2주 동안 아스코르브산(ascorbic acid) 50 ㎍/mL, 5 mM 베타-글리세로포스페이트(β-glycerophosphate), BMP 250 ng/mL를 첨가한 배지에서 배양하였다. In order to induce differentiation in gastric cells, cells were cultured with 50 ㎍/mL of ascorbic acid, 5 mM beta-glycerophosphate, and 250 ng/mL BMP for 1 to 2 weeks. Cultured in.

바이러스 감염을 위해 골모세포를 10% FBS가 함유된 배지에서 24시간 동안 배양한 후 CUEDC2를 과발현시키는 adenovirus(ad)-CUEDC2 또는 CUEDC2의 발현을 낮추는 ad-shRNA(sh)CUEDC2가 첨가된 무혈청(serum-free) 배지로 교체하였다. 4시간 후 20% FBS를 첨가시키고, 20시간 후 골모세포로 분화할 수 있는 배지로 교체해 주었다. ad-GFP(green fluorescent protein)을 대조군으로 사용하였다.For viral infection, osteoblasts are cultured in a medium containing 10% FBS for 24 hours, and then adenovirus(ad)-CUEDC2 that overexpresses CUEDC2 or ad-shRNA(sh)CUEDC2 that reduces the expression of CUEDC2 is added to serum-free ( serum-free) medium. After 4 hours, 20% FBS was added, and after 20 hours, it was replaced with a medium capable of differentiating into osteoblasts. ad-GFP (green fluorescent protein) was used as a control.

1-2. RNA 분리 및 qPCR 분석1-2. RNA isolation and qPCR analysis

MC3T3-E1 세포를 골형성 배지 또는 일반적인 배지에서 지정된 시간 동안 배양 후 Trizol 시약을 이용하여 총 RNA(total RNA)를 추출하였다. After culturing MC3T3-E1 cells in an osteogenic medium or a general medium for a specified time, total RNA (total RNA) was extracted using Trizol reagent.

cDNA는 random primer (0.2 ㎍), MMLV (200 U), dNTP (0.5 mM), 그리고 RNAsin (40 U)을 이용하여 RNA 1.5 ㎍으로부터 합성하였다. PCR 수행 조건은 다음과 같다: 95 ℃에서 5분 후에 95 ℃, 55 ℃, 72 ℃ 각각 30초씩 40 사이클(cycle)을 반복 후 72 ℃에서 7분 동안 최종 연장함 cDNA was synthesized from 1.5 μg of RNA using random primer (0.2 μg), MMLV (200 U), dNTP (0.5 mM), and RNAsin (40 U). PCR performance conditions are as follows: After 5 minutes at 95 °C, 40 cycles of 95 °C, 55 °C, and 72 °C were repeated for 30 seconds each, and then finally extended at 72 °C for 7 minutes.

qPCR 분석을 하기 위해 StepOnePlusTM Real-Time PCR System을 이용하여 그 결과를 분석하였다. β-Actin은 내부 대조군으로 이용되었다.For qPCR analysis, the results were analyzed using the StepOnePlusTM Real-Time PCR System. β-Actin was used as an internal control.

1-3. 웨스턴 블롯 분석1-3. Western blot analysis

Lysis 완충용액을 배양용기에 첨가하고, 얼음 위에서 방치 후 세포 용해물(lysate)을 수확하여 4 ℃, 12,000 rpm에서 15분간 원심 분리하여 총 단백질이 들어있는 상층액을 얻었다. Lysis buffer was added to the culture vessel, and then left on ice to harvest cell lysates and centrifuged at 4° C. and 12,000 rpm for 15 minutes to obtain a supernatant containing total protein.

단백질 농도는 소혈청알부민(bovine serum albumin; BSA)으로 표준화하여 Lowry protein assay reagent를 사용하여 정량하였다. Protein concentration was normalized with bovine serum albumin (BSA) and quantified using a Lowry protein assay reagent.

단백질 샘플들은 10% 또는 8% SDS-PAGE 겔(gel)에서 전기영동하여 PVDF membrane으로 전이시켰다(transfer). 5%의 무지방 건조 밀크(skim milk)가 포함된 Tris 완충용액에서 전이된 단백질의 비특이적인 결합을 차단하고, 특이적 분석을 위해 CUEDC2, Runx2, Osx, Actin 항체(antibody)와 각각의 이차항체(secondary antibody)로 반응시켰다. Protein samples were transferred to a PVDF membrane by electrophoresis on a 10% or 8% SDS-PAGE gel. Blocks non-specific binding of transferred proteins in Tris buffer containing 5% fat-free dry milk (skim milk), and CUEDC2, Runx2, Osx, Actin antibodies and each secondary antibody for specific analysis (secondary antibody).

Enhanced chemiluminescence reagent와 LAS-4000 mini imager를 사용하여 각 단백질의 존재를 가시화하였다. The presence of each protein was visualized using an enhanced chemiluminescence reagent and a LAS-4000 mini imager.

1-4. 알칼라인 포스파타제(Alkaline phosphatase; ALP) 염색1-4. Alkaline phosphatase (ALP) staining

각 실험조건에서 5일간 배양된 MC3T3-E1 세포를 PBS로 세척한 후 4% 포르말린으로 15분간 고정하였다. 남은 포르말린을 증류수로 3번 세척하여 제거하였고, BCIP®/NBT Liquid Substrate System로 20분 동안 염색하여 관찰하였다. MC3T3-E1 cells cultured for 5 days in each experimental condition were washed with PBS and then fixed with 4% formalin for 15 minutes. The remaining formalin was washed 3 times with distilled water to remove, and observed by staining for 20 minutes with BCIP®/NBT Liquid Substrate System.

정량적인 분석을 위해 image J 프로그램을 이용하여 나타내었다.For quantitative analysis, it was shown using the image J program.

1-5. 알리자린 레드(Alizarin red; AR) 염색1-5. Alizarin red (AR) staining

각 실험조건에서 12일간 배양된 MC3T3-E1 세포를 PBS로 세척한 후 70% 에탄올로 1시간 동안 고정하였다. 남은 에탄올은 증류수로 3번 세척하여 제거하였고, 알리자린 레드 용액으로 10분 동안 염색하여 관찰하였다. MC3T3-E1 cells cultured for 12 days in each experimental condition were washed with PBS and then fixed with 70% ethanol for 1 hour. The remaining ethanol was removed by washing with distilled water three times, and stained with an alizarin red solution for 10 minutes and observed.

정량적인 분석을 위해 10% 세틸피리디늄 클로라이드(cetylpyridinium chloride)를 이용하여 염색된 세포를 탈색시킴 후 multiplate reader를 이용하여 540 nm 파장에서 흡광도를 측정하였다.For quantitative analysis, the stained cells were bleached using 10% cetylpyridinium chloride, and absorbance was measured at a wavelength of 540 nm using a multiplate reader.

1-6. 동물 수술 및 조직학적, 방사선학적 분석1-6. Animal surgery and histological and radiological analysis

실험에 사용한 동물은 쥐 C57BL/6로 ㈜다물사이언스에서 구입하였으며, 모든 동물실험은 전남대학교 동물실험윤리위원회의 규정에 준하여 시행되었다(동물실험 번호: CNU IACUC-YB-2017-73).The animals used in the experiment were rat C57BL/6, and were purchased from Damul Science, and all animal experiments were conducted in accordance with the regulations of the Animal Experimental Ethics Committee of Chonnam National University (Animal Experiment Number: CNU IACUC-YB-2017-73).

CUEDC2 유전자의 골 형성능에 대해 알아보고자 이소성 골형성 모델과 쥐 두개골 손상 모델을 이용하였다. In order to investigate the bone formation ability of CUEDC2 gene, an ectopic bone formation model and a rat skull injury model were used.

이소성 골형성 모델은 BMP2 3 ㎍와 ad-GFP 혼합, 또는 BMP2 3 ㎍와 ad-CUEDC2를 혼합한 것을 콜라겐 스펀지에 각각 주입한 후 생후 6주령의 C57BL/6 쥐 피하에 4주 동안 이식하였다.In the ectopic bone formation model, a mixture of 3 µg of BMP2 and ad-GFP, or a mixture of 3 µg of BMP2 and ad-CUEDC2, respectively, was injected into a collagen sponge, and then implanted subcutaneously in a 6-week-old C57BL/6 mouse for 4 weeks.

쥐 두개골 손상 모델은 생후 6주령의 C57BL/6 쥐를 마취한 후 5 mm 직경을 가진 트레핀버(trephine bur)로 쥐 두개골에 천공을 만들었고, 이후 ad-GFP 단독, BMP 2500 ng 와 ad-GFP 혼합, 또는 ad-shCUEDC2 단독, BMP 2500 ng 와 ad-shCUEDC2 혼합한 것을 콜라겐 스펀지에 각각 주입한 후 4주 동안 이식하였다. 이후, 모든 동물 실험 샘플을 수확(harvest)하여 4% 파라포름알데하이드에 고정하고, 10% EDTA에서 탈회시켜, 파라핀에 포매하여, 헤마톡실린-에오신으로 염색하여 조직학적으로 분석하였다.In the rat skull injury model, 6-week-old C57BL/6 mice were anesthetized and perforated in the rat skull with a 5 mm diameter trephine bur, after which ad-GFP alone, BMP 2500 ng and ad-GFP mixed. Alternatively, ad-shCUEDC2 alone, a mixture of 2500 ng of BMP and ad-shCUEDC2 was injected into a collagen sponge, respectively, and implanted for 4 weeks. Thereafter, all animal experimental samples were harvested, fixed in 4% paraformaldehyde, demineralized in 10% EDTA, embedded in paraffin, stained with hematoxylin-eosin, and analyzed histologically.

또한, 방사선학적 분석을 위해 동물 샘플을 고해상도 마이크로 전산화단층 촬영기 (Skyscan1172, Bruker)로 스캔하였다. 스캔 조건으로서 0.5 mm 두께의 알루미늄 필터를 사용하였고, X-선 발생기의 전압과 전류는 50 ㎸, 200 ㎂로 설정하였으며, 단위 픽셀의 크기는 30 ㎛로 설정하였다. 채득한 2차원 사진을 이용하여 CTvol 프로그램으로 골의 3차원 이미지를 얻었으며, CTAnalyzer 프로그램을 이용하여 골 부피(bone volume) 및 골 밀도(bone mineral density)를 분석하였다.In addition, animal samples were scanned with a high-resolution microcomputerized tomography (Skyscan1172, Bruker) for radiological analysis. An aluminum filter having a thickness of 0.5 mm was used as the scanning condition, the voltage and current of the X-ray generator were set to 50 ㎸ and 200 ㎂, and the size of the unit pixel was set to 30 µm. Three-dimensional images of bones were obtained using the CTvol program using the obtained two-dimensional photographs, and bone volume and bone mineral density were analyzed using the CTAnalyzer program.

정량 분석은 골 부피를 계측하여 각 군별로 재생된 뼈의 양 변화를 관찰하였으며, 정성 분석을 통해 골의 강도를 나타내는 골 밀도를 계측하여 각각의 대조군과 실험군의 골질을 비교 분석하였다.For quantitative analysis, bone volume was measured to observe the change in the amount of regenerated bone for each group, and bone density indicating bone strength was measured through qualitative analysis to compare and analyze the bone quality of each control group and the experimental group.

1-7. 통계 분석1-7. Statistical analysis

모든 실험 값은 평균과 표준 표차로 표시하였다. 결과 분석은 ANOVA Tukey test 또는 T test를 이용하였으며, 통계적 유의성은 p < 0.05 이내에서 검증하였다.All experimental values were expressed as mean and standard deviation. Results were analyzed using ANOVA Tukey test or T test, and statistical significance was verified within p <0.05.

2. 실험 결과2. Experiment result

2-1. 골모세포 분화에 따른 CUEDC2 유전자의 발현 변화 분석2-1. Analysis of changes in expression of CUEDC2 gene according to osteoblast differentiation

골모세포의 분화 과정 동안 CUEDC2의 발현 정도를 조사하기 위해, 골형성 배지에서 각각 2일, 4일, 6일 동안 배양한 후 qPCR, 웨스턴 블롯으로 확인하였다.In order to investigate the expression level of CUEDC2 during the differentiation process of osteoblasts, the cells were cultured for 2, 4, and 6 days in bone formation medium, respectively, and then confirmed by qPCR and Western blot.

실험 결과, 골모세포의 분화가 진행됨에 따라 골 시알로단백질(Bsp), 오스테오칼신(Oc), Runx2, 오스테릭스(Osx)와 같은 골모세포 마커(marker) 인자들이 증가하였고, 그와 동시에 CUEDC2의 mRNA 및 단백질 발현 정도가 감소하였다. 이는 CUEDC2가 골모세포 분화와 관련이 있음을 나타내는 것이다(도 1).As a result of the experiment, as osteoblast differentiation progressed, osteoblast marker factors such as bone sialoprotein (Bsp), osteocalcin (Oc), Runx2, and Osxia increased, and at the same time, the mRNA of CUEDC2 And the degree of protein expression decreased. This indicates that CUEDC2 is related to osteoblast differentiation (FIG. 1).

2-2. CUEDC2 과발현시 골모세포 분화 및 골형성 정도 분석2-2. Analysis of osteoblast differentiation and bone formation degree when CUEDC2 is overexpressed

CUEDC2가 골모세포 분화기능에 미치는 영향을 조사하기 위해, 야생형 CUEDC2 플라스미드 벡터 (ORIGENE 제조, CUEDC2 Mouse Tagged ORF Clone, #MR216274) 및 아데노바이러스(ad)-CUEDC2 (VECTOR BIOLABS 제조, Ad-GFP-m-CUEDC2, #ADV-256367)를 이용하여 CUEDC2를 과발현시킨 후 실험을 수행하였다.To investigate the effect of CUEDC2 on osteoblast differentiation function, wild-type CUEDC2 plasmid vector (manufactured by ORIGENE, CUEDC2 Mouse Tagged ORF Clone, #MR216274) and adenovirus (ad)-CUEDC2 (manufactured by VECTOR BIOLABS, Ad-GFP-m-) CUEDC2, #ADV-256367) was used to overexpress CUEDC2 and then an experiment was performed.

야생형 CUEDC2 플라스미드 벡터를 Lipofectamine 2000을 이용하여 농도별로 형질감염시킨 후, 3일 동안 골형성 배지에서 배양하여 골 시알로단백질(Bsp), 오스테오칼신(Oc) mRNA의 발현 정도를 qPCR로 평가하였다. 그 결과, CUEDC2의 과발현시 골모세포의 표지 유전자인 골 시알로단백질 및 오스테오칼신의 발현이 감소하였다(도 2). The wild-type CUEDC2 plasmid vector was transfected by concentration using Lipofectamine 2000, and then cultured in bone formation medium for 3 days to evaluate the expression level of bone sialoprotein (Bsp) and osteocalcin (Oc) mRNA by qPCR. As a result, upon overexpression of CUEDC2, the expression of bone sialoprotein and osteocalcin, which are marker genes for osteoblasts, decreased (FIG. 2).

골형성 분화의 초기 마커인 ALP 활성 또는 후기 마커인 무기질 침착정도를 분석하기 위해 세포에 아데노바이러스 CUEDC2(ad-CUEDC2)로 과발현시킨 후 각각 6일, 12일 동안 골형성 배지에서 배양하였다. 그 결과, ALP 염색 및 AR 염색 모두 CUEDC2 발현이 증가함에 따라 염색 정도가 감소되어 나타났다(도 3의 a, b). In order to analyze the ALP activity, which is an early marker of osteogenic differentiation, or the degree of mineral deposition, which is a late marker, cells were overexpressed with adenovirus CUEDC2 (ad-CUEDC2), and then cultured in osteogenic medium for 6 and 12 days, respectively. As a result, both ALP staining and AR staining showed that the degree of staining decreased as CUEDC2 expression increased (Fig. 3 a and b).

또한, CUEDC2가 직접적으로 뼈 재생에 미치는 영향을 평가하기 위하여 in vivo 이소성 골형성 실험을 수행하였다. 이를 위해 BMP2와 ad-GFP (대조군) 또는 BMP2와 ad-CUEDC2 (실험군)를 콜라겐 스펀지에 첨가하였고, 이를 쥐의 등에 이식하고 3주 후 골 형성정도를 마이크로전산화단층촬영(μ-CT) 및 조직염색을 통해 평가하였다. 그 결과, 대조군을 이식한 쥐에서는 전체적으로 일정한 형태의 골이 잘 형성되었으나, 실험군을 이식한 쥐에서는 부분적으로 골이 형성되었다. 실험군에서는 대조군에 비해 골의 부피, 미네랄의 밀도 및 골 면적 정도가 유의하게 감소되어 나타났다. 이를 통해 CUEDC2는 골모세포의 분화정도 뿐만 아니라 골 형성도 감소시킬 수 있음을 확인하였다(도 3의 c).In addition, in vivo ectopic bone formation experiments were performed to evaluate the effect of CUEDC2 on bone regeneration directly. To this end, BMP2 and ad-GFP (control group) or BMP2 and ad-CUEDC2 (experimental group) were added to a collagen sponge, which was implanted on the back of a mouse, and the bone formation level was measured by microcomputerized tomography (μ-CT) and tissue It was evaluated through staining. As a result, bones of a certain shape were well formed in the mice transplanted with the control group, but bones were partially formed in the mice transplanted with the experimental group. In the experimental group, the bone volume, mineral density, and bone area degree were significantly reduced compared to the control group. Through this, it was confirmed that CUEDC2 can reduce not only the degree of differentiation of osteoblasts, but also bone formation (Fig. 3c).

2-3. CUEDC2 발현 억제를 통한 골모세포 분화 및 뼈 재생 유도2-3. Induction of osteoblast differentiation and bone regeneration through inhibition of CUEDC2 expression

CUEDC2 발현 억제시 골 재생을 평가하기 위해, 하기 표 1의 si-RNA CUEDC2 (BIONEER 제조, #1344395) 및 ad-shCUEDC2 (VECTOR BIOLABS 제조, Ad-GFP-U6-m-CUEDC2-shRNA, #shADV-256367)를 이용하여 골모세포 분화 및 쥐 두개골 손상 모델 실험을 수행하였다. To evaluate bone regeneration upon inhibition of CUEDC2 expression, si-RNA CUEDC2 (manufactured by BIONEER, #1344395) and ad-shCUEDC2 (manufactured by VECTOR BIOLABS, Ad-GFP-U6-m-CUEDC2-shRNA, #shADV-) of Table 1 below. 256367) was used to perform osteoblast differentiation and rat skull injury model experiments.

염기서열 (5' > 3')Base sequence (5'> 3') si-RNA CUEDC2si-RNA CUEDC2 센스
(서열번호 2)
sense
(SEQ ID NO: 2)
GATACATCGACAACCAAGTGATACATCGACAACCAAGT
안티센스
(서열번호 3)
Antisense
(SEQ ID NO: 3)
ACTTGGTTGTCGATGTATCACTTGGTTGTCGATGTATC
ad-shCUEDC2
(서열번호 4)
ad-shCUEDC2
(SEQ ID NO: 4)
CCGGGACCATCAGAGGAGAACTTTGCTCGAGCAAAGTTCTCCTCTGATGGTCTTTTTGCCGGGACCATCAGAGGAGAACTTTGCTCGAGCAAAGTTCTCCTCTGATGGTCTTTTTG

si-RNA CUEDC2를 Lipofectamine RNAiMAX를 이용하여 농도별로 형질감염시킨 후 3일 동안 골형성 배지에서 배양하여 골 시알로단백질 및 오스테오칼신 mRNA의 발현 정도를 qPCR로 평가하였다. 그 결과, CUEDC2의 발현을 억제하였을 때 골모세포의 표지 유전자인 골 시알로단백질(Bsp) 및 오스테오칼신(Oc)의 발현이 BMP2 처리군에 비해 증가함을 확인하였다(도 4).si-RNA CUEDC2 was transfected for each concentration using Lipofectamine RNAiMAX and cultured in bone formation medium for 3 days to evaluate the expression levels of bone sialoprotein and osteocalcin mRNA by qPCR. As a result, it was confirmed that when the expression of CUEDC2 was suppressed, the expression of bone sialoprotein (Bsp) and osteocalcin (Oc), which are marker genes for osteoblasts, increased compared to the BMP2 treatment group (FIG. 4).

골형성 분화의 초기 마커인 ALP 활성 또는 후기 마커인 무기질 침착정도를 분석하기 위해 세포에 아데노바이러스 CUEDC2(ad-shCUEDC2)로 과발현시킨 후 각각 6일, 12일 동안 골형성 배지에서 배양하였다. 그 결과, ALP 활성 및 무기질 침착정도의 증가함을 관찰하였다(도 5의 a, b).In order to analyze the ALP activity, which is an early marker of osteogenic differentiation, or the degree of mineral deposition, which is a late marker, cells were overexpressed with adenovirus CUEDC2 (ad-shCUEDC2), and then cultured in osteogenic medium for 6 and 12 days, respectively. As a result, it was observed that the ALP activity and the degree of mineral deposition were increased (Fig. 5a, b).

CUEDC2의 발현 억제가 직접적으로 골 재생을 촉진할 수 있는지 평가하고자 BMP2와 ad-GFP (대조군) 또는 BMP2와 ad-CUEDC2 (실험군)를 콜라겐 스펀지에 처리하였고, 이를 쥐 두개골 손상 부위에 이식하였다. 그 결과, ad-shCUEDC2 단독 처리는 대조군에 비해 골이 재생되었으나, 통계적으로 유의성은 없었고, BMP2가 포함된 ad-shCUEDC2 실험군에는 재생된 골이 대조군에 비해 많이 생성된 것을 3차원 이미지를 통해 육안으로 확인할 수 있었고, 정량분석을 통해서도 마찬가지로 골 부피가 증가하는 것을 알 수 있었다(도 5). In order to evaluate whether the inhibition of CUEDC2 expression can directly promote bone regeneration, BMP2 and ad-GFP (control) or BMP2 and ad-CUEDC2 (experimental group) were treated with a collagen sponge, which was transplanted into a rat skull injury site. As a result, ad-shCUEDC2 treatment alone reproduced bones compared to the control group, but there was no statistical significance, and that the ad-shCUEDC2 experimental group containing BMP2 produced more bones than the control group with the naked eye through 3D images. It could be confirmed, and it was found that the bone volume was also increased through quantitative analysis (FIG. 5).

결론적으로, CUEDC2의 발현 억제는 골모세포의 분화를 유도하여 골 재생까지 영향을 미칠 수 있다는 것을 확인할 수 있었다.In conclusion, it was confirmed that the inhibition of CUEDC2 expression can affect bone regeneration by inducing osteoblast differentiation.

<110> INDUSTRY FOUNDATION OF CHONNAM NATIONAL UNIVERSITY <120> Composition for Osteogenic Differentiation or Osteogenesis Comprising Inhibitor of CUEDC2 <130> PN190048 <160> 4 <170> KoPatentIn 3.0 <210> 1 <211> 1932 <212> RNA <213> Artificial Sequence <220> <223> CUEDC2 <400> 1 gggccccgcc tttgatctcg ttggtggggc tagggggggt gataagaact gcaccgcgcg 60 ggacaagttg ctggcggcgc ctgacggagc aggcctgtct cgtcatgtga ctcaccctct 120 cccgaggcgg ctgctgcatc cctgtgtcct caggccagct cttctggagg cttatgagtg 180 ccctgctaac gaagacccag catggagttg gagaggatcg tcggctcagc cctcctcacc 240 ttcgttcagg cacacctgcc agaggcagac ctcagtggct tggatgaagt catcttctcc 300 tatgtgcttg gggtcctgga ggacctgggc ccctcaggac catcagagga gaactttgat 360 atggaggcct tcactgaaat gatggaggct tatgtgcctg gctttgccca catccccaga 420 ggtataatag gagacatgat gcagaagctc tcggtgcagc tgagcgatgc taggaacaaa 480 gagaacctgc acccacagag ctcctgtgtc caaggtcagg tgcccatttt tccagagacc 540 ccgaggcaag ctgaaaagct cgaagaagag agcaggcctc ccgctgctcc cgggaacacc 600 ctagatgagg cagctgcggc agaagaactg ccaggagtcg atgtgctcct ggaggtcttc 660 cctacctgct ctatggagca ggcccagtgg gtgctggcca aagctcgggg ggacttggag 720 gaagctgtgc acatgctggt agagggcaag gaagaggggc ctccaggctg ggacggccca 780 agtcaggact tgcccaggcg tctcagaggc ccccaaaagg atgacttgaa gtctttcatc 840 cttcagaagt acatgatggt ggacagggcg gaggaccaga agactcaccg acctatggct 900 cccaaggagg cccccaagaa gctgatccga tacatcgaca accaagtagt gagcaccaag 960 ggagagcgat tcaaagatgt ccggaaccct gaggccgagg agatgaaggc cacatacatc 1020 aacctcaagc cggccagaaa gtaccgcttc cactgagaca gtgctggatc ctgccgagca 1080 ctgctgatcc cagagggatg cagacaccct cccatccacc ctgtcccttc tggaggccct 1140 tgctctagtg ttctattctt ggggcttcct tcaagagcac agtaaaagtg gtccaaggaa 1200 ggtatatgct cattctgctc atttctgccc ccagggcagg gaaaaaacct gtctctctga 1260 ctagttccca gcttcccgtc ccctatgcac cacgcaagaa ggggtaaaaa gctctttatt 1320 tgaaattcca gggtgggaca ggggatttca caggcctgtc aaccacaggc ccccacagtt 1380 cagtttccct atggctcagg gtcaagcccg ggctacacag tccagctgtg ctcccacccc 1440 aaggaaagtt agtcagacct gtaggttgac aaagggtgcg aagcctgcca tgtcctggag 1500 tagcaccaga gcctggtgca ggggtggttc cacgtcgatg tccacaccac gcttccgcaa 1560 gcggctcagg ctcggttcag ccacatggtg gcagtgccgc acccgcagag agcgcagcgc 1620 cgggcagtac tcggccagtg tcctgaggga ggagtatggt gatgctaggt gctgaatctc 1680 acggtctgga gggccagccc tctaaacccg cagctggctc cagcaagttc tagctttccc 1740 tccagctcag ggtctgctct tttttactgt acctaaagga aggggccagg aaggaattac 1800 agactccagg ctcatttggg gtttctggtt cttagagagt accaatccat agttccttcc 1860 agtgtttcta gattacccac cttgttgcag aaatgccaga aatcaacttt atgtgacatt 1920 tctcagtgtg tc 1932 <210> 2 <211> 19 <212> RNA <213> Artificial Sequence <220> <223> si-RNA CUEDC2_sense <400> 2 gatacatcga caaccaagt 19 <210> 3 <211> 19 <212> RNA <213> Artificial Sequence <220> <223> si-RNA CUEDC2_antisense <400> 3 acttggttgt cgatgtatc 19 <210> 4 <211> 58 <212> RNA <213> Artificial Sequence <220> <223> ad-shCUEDC2 <400> 4 ccgggaccat cagaggagaa ctttgctcga gcaaagttct cctctgatgg tctttttg 58 <110> INDUSTRY FOUNDATION OF CHONNAM NATIONAL UNIVERSITY <120> Composition for Osteogenic Differentiation or Osteogenesis Comprising Inhibitor of CUEDC2 <130> PN190048 <160> 4 <170> KoPatentIn 3.0 <210> 1 <211> 1932 <212> RNA <213> Artificial Sequence <220> <223> CUEDC2 <400> 1 gggccccgcc tttgatctcg ttggtggggc tagggggggt gataagaact gcaccgcgcg 60 ggacaagttg ctggcggcgc ctgacggagc aggcctgtct cgtcatgtga ctcaccctct 120 cccgaggcgg ctgctgcatc cctgtgtcct caggccagct cttctggagg cttatgagtg 180 ccctgctaac gaagacccag catggagttg gagaggatcg tcggctcagc cctcctcacc 240 ttcgttcagg cacacctgcc agaggcagac ctcagtggct tggatgaagt catcttctcc 300 tatgtgcttg gggtcctgga ggacctgggc ccctcaggac catcagagga gaactttgat 360 atggaggcct tcactgaaat gatggaggct tatgtgcctg gctttgccca catccccaga 420 ggtataatag gagacatgat gcagaagctc tcggtgcagc tgagcgatgc taggaacaaa 480 gagaacctgc acccacagag ctcctgtgtc caaggtcagg tgcccatttt tccagagacc 540 ccgaggcaag ctgaaaagct cgaagaagag agcaggcctc ccgctgctcc cgggaacacc 600 ctagatgagg cagctgcggc agaagaactg ccaggagtcg atgtgctcct ggaggtcttc 660 cctacctgct ctatggagca ggcccagtgg gtgctggcca aagctcgggg ggacttggag 720 gaagctgtgc acatgctggt agagggcaag gaagaggggc ctccaggctg ggacggccca 780 agtcaggact tgcccaggcg tctcagaggc ccccaaaagg atgacttgaa gtctttcatc 840 cttcagaagt acatgatggt ggacagggcg gaggaccaga agactcaccg acctatggct 900 cccaaggagg cccccaagaa gctgatccga tacatcgaca accaagtagt gagcaccaag 960 ggagagcgat tcaaagatgt ccggaaccct gaggccgagg agatgaaggc cacatacatc 1020 aacctcaagc cggccagaaa gtaccgcttc cactgagaca gtgctggatc ctgccgagca 1080 ctgctgatcc cagagggatg cagacaccct cccatccacc ctgtcccttc tggaggccct 1140 tgctctagtg ttctattctt ggggcttcct tcaagagcac agtaaaagtg gtccaaggaa 1200 ggtatatgct cattctgctc atttctgccc ccagggcagg gaaaaaacct gtctctctga 1260 ctagttccca gcttcccgtc ccctatgcac cacgcaagaa ggggtaaaaa gctctttatt 1320 tgaaattcca gggtgggaca ggggatttca caggcctgtc aaccacaggc ccccacagtt 1380 cagtttccct atggctcagg gtcaagcccg ggctacacag tccagctgtg ctcccacccc 1440 aaggaaagtt agtcagacct gtaggttgac aaagggtgcg aagcctgcca tgtcctggag 1500 tagcaccaga gcctggtgca ggggtggttc cacgtcgatg tccacaccac gcttccgcaa 1560 gcggctcagg ctcggttcag ccacatggtg gcagtgccgc acccgcagag agcgcagcgc 1620 cgggcagtac tcggccagtg tcctgaggga ggagtatggt gatgctaggt gctgaatctc 1680 acggtctgga gggccagccc tctaaacccg cagctggctc cagcaagttc tagctttccc 1740 tccagctcag ggtctgctct tttttactgt acctaaagga aggggccagg aaggaattac 1800 agactccagg ctcatttggg gtttctggtt cttagagagt accaatccat agttccttcc 1860 agtgtttcta gattacccac cttgttgcag aaatgccaga aatcaacttt atgtgacatt 1920 tctcagtgtg tc 1932 <210> 2 <211> 19 <212> RNA <213> Artificial Sequence <220> <223> si-RNA CUEDC2_sense <400> 2 gatacatcga caaccaagt 19 <210> 3 <211> 19 <212> RNA <213> Artificial Sequence <220> <223> si-RNA CUEDC2_antisense <400> 3 acttggttgt cgatgtatc 19 <210> 4 <211> 58 <212> RNA <213> Artificial Sequence <220> <223> ad-shCUEDC2 <400> 4 ccgggaccat cagaggagaa ctttgctcga gcaaagttct cctctgatgg tctttttg 58

Claims (10)

CUEDC2(CUE domain-containing protein 2)의 발현 또는 활성 억제제를 유효성분으로 포함하며,
상기 CUEDC2의 발현 또는 활성 억제제는 CUEDC2의 mRNA에 상보적으로 결합하는 siRNA 또는 shRNA인 것인 골분화 또는 골재생 촉진용 조성물.
It contains an inhibitor of the expression or activity of CUEDC2 (CUE domain-containing protein 2) as an active ingredient,
The CUEDC2 expression or activity inhibitor is an siRNA or shRNA that complementarily binds to CUEDC2 mRNA. A composition for promoting bone differentiation or bone regeneration.
삭제delete 제1항에 있어서,
상기 shRNA는 레트로바이러스 벡터 또는 아데노바이러스 벡터 시스템을 이용하는 것인 골분화 또는 골재생 촉진용 조성물.
The method of claim 1,
The shRNA is a composition for promoting bone differentiation or bone regeneration using a retroviral vector or an adenovirus vector system.
제1항에 있어서,
상기 CUEDC2의 발현 또는 활성 억제제는 골형성 촉진, 골분화 촉진 또는 기질 석회화 유도의 작용을 갖는 것인 골분화 또는 골재생 촉진용 조성물.
The method of claim 1,
The expression or activity inhibitor of CUEDC2 is a composition for promoting bone differentiation or bone regeneration that has the action of promoting bone formation, promoting bone differentiation, or inducing matrix calcification.
제1항에 있어서,
상기 조성물은 인간 또는 쥐에서 유래된 골 시알로단백질(bone sialoprotein), 오스테오칼신(osteocalcin) 또는 BMP2(bone morphogenetic protein 2)를 더 포함하는 것인 골분화 또는 골재생 촉진용 조성물.
The method of claim 1,
The composition is a composition for promoting bone differentiation or bone regeneration further comprising a bone sialoprotein, osteocalcin or BMP2 (bone morphogenetic protein 2) derived from human or rat.
제1항, 제3항 내지 제5항 중 어느 한 항에 따른 골분화 또는 골재생 촉진용 조성물을 유효성분으로 포함하는 골질환의 예방 또는 치료용 약학 조성물.
A pharmaceutical composition for the prevention or treatment of bone diseases comprising the composition for promoting bone differentiation or bone regeneration according to any one of claims 1, 3 to 5 as an active ingredient.
제6항에 있어서,
상기 골질환은 골결손, 골다공증, 골연화증, 골감소증, 골형성 부전증, 골형성 장애, 골관절염, 골괴사, 퇴행성 골질환, 골 관절염, 전이성 골암, 골종양, 치주질환, 골절 및 파제트병으로 이루어진 군으로부터 선택되는 것인 골질환의 예방 또는 치료용 약학 조성물.
The method of claim 6,
The bone disease is selected from the group consisting of bone defect, osteoporosis, osteomalacia, osteopenia, bone insufficiency, bone dysplasia, osteoarthritis, bone necrosis, degenerative bone disease, osteoarthritis, metastatic bone cancer, bone tumor, periodontal disease, fracture and Paget's disease A pharmaceutical composition for the prevention or treatment of bone disease.
(a) CUEDC2 유전자 또는 CUEDC2 단백질을 포함하는 세포에 분석하고자 하는 시험물질을 접촉시키는 단계;
(b) 상기 CUEDC2 유전자의 발현량, CUEDC2 단백질의 발현량 또는 CUEDC2 단백질의 활성도를 측정하는 단계; 및
(c) 상기 (b) 단계의 측정 결과에서 시험물질이 CUEDC2 유전자의 발현량, CUEDC2 단백질의 발현량 또는 CUEDC2 단백질의 활성도를 감소시킬 경우, 상기 시험물질을 골질환의 예방 또는 치료용 물질로 판정하는 단계
를 포함하는 골질환의 예방 또는 치료용 물질을 스크리닝하는 방법.
(a) contacting a test substance to be analyzed with cells containing CUEDC2 gene or CUEDC2 protein;
(b) measuring the expression level of the CUEDC2 gene, the expression level of the CUEDC2 protein, or the activity of the CUEDC2 protein; And
(c) If the test substance decreases the expression level of the CUEDC2 gene, the expression level of the CUEDC2 protein, or the activity of the CUEDC2 protein in the measurement result of step (b), the test material is determined as a substance for preventing or treating bone diseases. Steps to
Method for screening a substance for the prevention or treatment of bone disease comprising a.
제8항에 있어서,
상기 시험물질은 안티센스 올리고뉴클레오티드, siRNA, shRNA, miRNA, 리보자임, DNAzyme, PNA, 항체, 앱타머, 천연 추출물 및 합성 화합물로 이루어진 군에서 선택되는 것인 골질환의 예방 또는 치료용 물질을 스크리닝하는 방법.
The method of claim 8,
The test substance is selected from the group consisting of antisense oligonucleotides, siRNA, shRNA, miRNA, ribozymes, DNAzyme, PNA, antibodies, aptamers, natural extracts, and synthetic compounds to screen substances for the prevention or treatment of bone diseases. Way.
제8항에 있어서,
상기 (b) 단계의 측정은 역전사 중합효소 연쇄반응(reverse transcriptase-polymerase chain reaction), 실시간 중합효소 연쇄반응(real time-polymerase chain reaction), 웨스턴 블롯(western blot), 노던 블롯(northern blot), ELISA(enzyme linked immunosorbent assay), 방사선면역분석(radioimmunoassay), 방사 면역 확산법(radioimmunodiffusion), 면역침전분석법(immunoprecipitation assay) 및 면역조직화학적 분석(immunohistochemical analysis)으로 이루어진 군에서 선택되는 것인 골질환의 예방 또는 치료용 물질을 스크리닝하는 방법.
The method of claim 8,
The measurement of step (b) is reverse transcriptase-polymerase chain reaction, real time-polymerase chain reaction, western blot, northern blot, Prevention of bone disease, selected from the group consisting of enzyme linked immunosorbent assay (ELISA), radioimmunoassay, radioimmunodiffusion, immunoprecipitation assay, and immunohistochemical analysis. Or a method of screening a therapeutic substance.
KR1020190022787A 2019-02-26 2019-02-26 Composition for Osteogenic Differentiation or Osteogenesis Comprising Inhibitor of CUEDC2 KR102161623B1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
KR1020190022787A KR102161623B1 (en) 2019-02-26 2019-02-26 Composition for Osteogenic Differentiation or Osteogenesis Comprising Inhibitor of CUEDC2

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
KR1020190022787A KR102161623B1 (en) 2019-02-26 2019-02-26 Composition for Osteogenic Differentiation or Osteogenesis Comprising Inhibitor of CUEDC2

Publications (2)

Publication Number Publication Date
KR20200104172A KR20200104172A (en) 2020-09-03
KR102161623B1 true KR102161623B1 (en) 2020-10-05

Family

ID=72450306

Family Applications (1)

Application Number Title Priority Date Filing Date
KR1020190022787A KR102161623B1 (en) 2019-02-26 2019-02-26 Composition for Osteogenic Differentiation or Osteogenesis Comprising Inhibitor of CUEDC2

Country Status (1)

Country Link
KR (1) KR102161623B1 (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115624654B (en) * 2022-08-17 2023-09-29 中国医科大学附属口腔医院 Application of berberine hydrochloride in barrier repair integrated alveolar bone defect bone increment technology and composite material thereof

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20140066604A1 (en) 2011-01-24 2014-03-06 Biomedical Analysis Center Of Academy Of Military Medical Sciences Molecular marker cuedc2 protein for prognostic determination of breast cancer endocrinology therapy

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DK2711023T3 (en) * 2011-05-13 2017-10-16 Nat Univ Corp Tokyo Medical & Dental Univ osteogenesis
KR101570832B1 (en) 2013-09-09 2015-11-20 주식회사 본셀바이오텍 Bone graft substitute using cuttlefish bone and method for preparing thereof
KR101762580B1 (en) 2015-09-26 2017-07-31 (주)에스겔 A method for preparing porous bone graft materials

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20140066604A1 (en) 2011-01-24 2014-03-06 Biomedical Analysis Center Of Academy Of Military Medical Sciences Molecular marker cuedc2 protein for prognostic determination of breast cancer endocrinology therapy

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
Nature medicine. 2011. Vol.17, No.6, pp.708-715.

Also Published As

Publication number Publication date
KR20200104172A (en) 2020-09-03

Similar Documents

Publication Publication Date Title
Hemming et al. EZH2 deletion in early mesenchyme compromises postnatal bone microarchitecture and structural integrity and accelerates remodeling
Littlewood-Evans et al. Localization of cathepsin K in human osteoclasts by in situ hybridization and immunohistochemistry
Stevenson et al. Expression of the ‘dead box’RNA helicase p68 is developmentally and growth regulated and correlates with organ differentiation/maturation in the fetus
JP5128273B2 (en) Methods, agents, and compound screening assays for inducing differentiation of undifferentiated mammalian cells into osteoblasts
Lin et al. AP2a enhanced the osteogenic differentiation of mesenchymal stem cells by inhibiting the formation of YAP/RUNX2 complex and BARX1 transcription
Li et al. Influences of hucMSC-exosomes on VEGF and BMP-2 expression in SNFH rats.
US20130195863A1 (en) Methods and Pharmaceutical Compositions for the Treatment of Bone Density Related Diseases
Li et al. GIT1 regulates angiogenic factor secretion in bone marrow mesenchymal stem cells via NF‐κB/Notch signalling to promote angiogenesis
KR102161623B1 (en) Composition for Osteogenic Differentiation or Osteogenesis Comprising Inhibitor of CUEDC2
JP2022188178A (en) Extracellular matrix-producing composition using mast4 gene and preparation method therefor
KR20110117982A (en) Compositions comprising nfat5 inhibitor as an active ingredient for preventing or treating of angiogenesis-related diseases
Lan et al. Targeted activation of androgen receptor signaling in the periosteum improves bone fracture repair
CN113925972B (en) Application of OTUB1 protein in treating osteoporosis
Li et al. Evidence for an important role of Smad-7 in intervertebral disc degeneration
KR102124435B1 (en) A composition for promoting osteogenesis containing peroxiredoxin-6 activity inhibitor
WO2022137964A1 (en) Pharmaceutical composition for preventing or treating cartilage/bone/joint diseases, and method for screening drug for preventing or treating cartilage/bone/joint diseases
KR101642202B1 (en) Composition comprising klf10 inhibitors for stimulating chondrogenesis or treatment of cartilage diseases, and method for stimulating chondrogenesis using the same
CN108949974B (en) Application of E3 ubiquitin ligase ASB3 in preparation of liver cancer treatment medicine
KR102124872B1 (en) Pharmaceutical composition for use in preventing or treating osteoporosis containing OVOL1 activator as an active ingredient
KR101713428B1 (en) Pharmaceutical composition for preventing or treating inflammatory bone disease comprising miR 218-2
KR101121987B1 (en) Anti-osteoclast mediated bone resorption siRNA vector for gene therapy
Liu et al. A tale of the good and bad: cell.
US20160331814A1 (en) Col 11A1 AND NOVEL FRAGMENT THEREOF FOR REGULATION OF BONE MINERALIZATION
KR102060230B1 (en) Composition for preventing and treating arthritis comprising a nucleolar orophate receptor RORα expression and activity modulator
US8367801B2 (en) Proteinaceous compounds

Legal Events

Date Code Title Description
E701 Decision to grant or registration of patent right
GRNT Written decision to grant