KR102635831B1 - Use for treating pulmonary fibrosis by inducing repolarization of M2 macrophage by CXCL11 - Google Patents

Use for treating pulmonary fibrosis by inducing repolarization of M2 macrophage by CXCL11 Download PDF

Info

Publication number
KR102635831B1
KR102635831B1 KR1020230148771A KR20230148771A KR102635831B1 KR 102635831 B1 KR102635831 B1 KR 102635831B1 KR 1020230148771 A KR1020230148771 A KR 1020230148771A KR 20230148771 A KR20230148771 A KR 20230148771A KR 102635831 B1 KR102635831 B1 KR 102635831B1
Authority
KR
South Korea
Prior art keywords
cxcl11
macrophages
pulmonary fibrosis
fibrosis
confirmed
Prior art date
Application number
KR1020230148771A
Other languages
Korean (ko)
Inventor
홍석호
송행석
박미라
김지영
Original Assignee
강원대학교 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 강원대학교 산학협력단 filed Critical 강원대학교 산학협력단
Priority to KR1020230148771A priority Critical patent/KR102635831B1/en
Application granted granted Critical
Publication of KR102635831B1 publication Critical patent/KR102635831B1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/195Chemokines, e.g. RANTES
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Epidemiology (AREA)
  • Pulmonology (AREA)
  • Zoology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Immunology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

본 발명은 CXCL11에 의한 M2 대식세포의 재분극 유도를 통한 폐섬유화증 치료 용도에 관한 것으로, 구체적으로, 블레오마이신 유도 폐섬유화 동물모델을 활용하여 M0, M1 및 M2 분비체(conditioned medium, CM) 주입을 통해 섬유화 치료 효과를 확인하였고, M1 대식세포 분비체를 주입한 마우스의 폐에서 콜라겐과 같은 섬유화 지표의 감소 및 정상군에 가까운 M1/M2 ratio가 변화되어 있는 것을 확인하였다. 또한, 블레오마이신 유도 폐섬유화 동물모델을 활용하여 human recombinant CXCL11 정맥 주입을 통해 섬유화 치료효과를 확인하였고, CXCL11을 투여한 마우스의 폐에서 콜라겐과 같은 섬유화 지표의 감소 및 정상군에 가까운 M1/M2 ratio가 변화되어 있는 것을 확인하였다. 즉, 본 발명에서는 폐 섬유화증 발병과정에서 CXCL11을 통한 M2 대식세포의 재분극을 유도하여 결과적으로 폐 섬유화증 치료의 가능한 표적 인자로 제시하고자 한다.The present invention relates to the use of pulmonary fibrosis treatment by inducing repolarization of M2 macrophages by CXCL11, and specifically, injection of M0, M1, and M2 secretions (conditioned medium, CM) using a bleomycin-induced pulmonary fibrosis animal model. The effectiveness of fibrosis treatment was confirmed, and a decrease in fibrosis indicators such as collagen and a change in the M1/M2 ratio close to the normal group were confirmed in the lungs of mice injected with M1 macrophage secretory body. In addition, using a bleomycin-induced pulmonary fibrosis animal model, the fibrosis treatment effect was confirmed through intravenous injection of human recombinant CXCL11. In the lungs of mice administered CXCL11, fibrosis indicators such as collagen were reduced and the M1/M2 ratio was close to that of the normal group. It was confirmed that has changed. In other words, the present invention seeks to induce repolarization of M2 macrophages through CXCL11 during the development of pulmonary fibrosis and consequently present it as a possible target factor for the treatment of pulmonary fibrosis.

Description

CXCL11에 의한 M2 대식세포의 재분극 유도를 통한 폐섬유화증 치료 용도{Use for treating pulmonary fibrosis by inducing repolarization of M2 macrophage by CXCL11}Use for treating pulmonary fibrosis by inducing repolarization of M2 macrophage by CXCL11}

본 발명은 CXCL11에 의한 M2 대식세포의 재분극 유도를 통한 폐섬유화증 치료 용도에 관한 것이다.The present invention relates to the use for treating pulmonary fibrosis through inducing repolarization of M2 macrophages by CXCL11.

대식세포는 극소 미세 환경에 따라 고전적으로 활성화된 M1 표현형 또는 대체적으로 활성화된 M2 표현형으로 분극화될 수 있다. 일반적으로 M1 대식세포는 폐포 상피 손상 후 상처 치유를 담당하고, M2 대식세포는 상처 치유 과정을 해결하거나 폐의 염증 반응을 종결하는 역할을 한다. 여러 연구에 따르면 섬유화 병변에서 활성화된 M2 대식세포는 콜라겐 합성과 침착을 강화하는 다량의 IL-10, TGFβ1과 같은 사이토카인을 생성하고, 이러한 전염증성 및 섬유화성 사이토카인은 부정적인 피드백을 통해 항염증성 사이토카인의 생성을 간접적으로 억제하여 섬유화를 더욱 촉진하는 것으로 알려져 있다.Macrophages can be polarized into either a classically activated M1 phenotype or an alternatively activated M2 phenotype depending on their microenvironment. Generally, M1 macrophages are responsible for wound healing after alveolar epithelial injury, while M2 macrophages are responsible for resolving the wound healing process or terminating the inflammatory response in the lung. Several studies have shown that activated M2 macrophages in fibrotic lesions produce large amounts of cytokines such as IL-10 and TGFβ1 that enhance collagen synthesis and deposition, and that these pro-inflammatory and pro-fibrotic cytokines exert anti-inflammatory properties through negative feedback. It is known to further promote fibrosis by indirectly suppressing the production of cytokines.

특발성 폐섬유화증(idiopathic pulmonary fibrosis, IPF)은 폐에 과도한 세포외 기질(ECM)이 축적되어 폐포 벽이 두꺼워지고 궁극적으로 폐포 구조가 파괴되어 호흡 부전으로 이어지는 질환으로, 진단 후 5년 생존률이 40% 미만에 불과하다. 현재까지 병인을 밝히기 위한 새로운 개념이 많이 정립되었지만, 불행히도 폐섬유증의 정확한 원인은 밝혀지지 않아 근본적 치료가 어려운 불치병이다. 현재 의학 기술에서 섬유화된 조직을 원 상태로 완전히 되돌려 놓는 기술은 없고, 섬유화의 진행을 억제하는 약물인 퍼페니돈(pirfenidone)과 닌텐다닙(nintendanib) 약제를 사용하고 있다. 이들 약물은 폐기능 저하를 지연시키지만 여전히 질병 진행 자체를 멈추게 하지는 못하고 또한 부작용이 심해 환자의 중도 복용 포기율이 높다. 특발성 폐섬유화증의 치료제 개발을 위해 많은 임상 연구가 진행중이나, 현재 임상에 돌입한 특발성 폐섬유화증에 효과가 보이는 약물은 콜라겐 생성에 영향을 주는 단백질의 작용을 감소시켜 콜라겐의 생성을 억제하거나 다양한 생리활성이 관여하는 수용체를 이용한 염증 및 섬유화를 막는 기전을 이용한 것으로 증상 개선에 초점이 맞춰져 있는데, 여전히 의료미충족 수요가 높아 새로운 치료제 개발이 시급한 상황이다.Idiopathic pulmonary fibrosis (IPF) is a disease in which excessive extracellular matrix (ECM) accumulates in the lungs, thickens the alveolar walls, and ultimately destroys the alveolar structure, leading to respiratory failure. The 5-year survival rate after diagnosis is 40%. It is only less than %. To date, many new concepts have been established to reveal the etiology, but unfortunately, the exact cause of pulmonary fibrosis is not known, making it an incurable disease that is difficult to fundamentally treat. Currently, there is no technology to completely return fibrotic tissue to its original state in medical technology, and drugs such as pirfenidone and nintendanib are used to suppress the progression of fibrosis. Although these drugs delay the decline in lung function, they still do not stop the disease progression, and the side effects are so severe that the rate of patients giving up taking the drug is high. Many clinical studies are in progress to develop treatments for idiopathic pulmonary fibrosis, but drugs that are currently in clinical trials and are shown to be effective for idiopathic pulmonary fibrosis inhibit collagen production by reducing the action of proteins that affect collagen production, or suppress collagen production in various ways. The focus is on improving symptoms by using a mechanism to prevent inflammation and fibrosis using receptors involved in physiological activity, but the development of new treatments is urgently needed due to the high unmet medical demand.

미국공개특허 US 2021-0330734 A1 (2021.10.28 공개)US published patent US 2021-0330734 A1 (published on October 28, 2021)

본 발명의 목적은 CXCL11을 유효성분으로 포함하는 폐섬유화증 치료용 약학 조성물을 제공하는 데에 있다.The purpose of the present invention is to provide a pharmaceutical composition for treating pulmonary fibrosis containing CXCL11 as an active ingredient.

또한, 본 발명의 다른 목적은 CXCL11을 유효성분으로 포함하는, 시험관(in vitro) 내 M2 대식세포의 M1 대식세포로의 재분극 유도용 시약 조성물 및 이를 이용한 M2 대식세포의 M1 대식세포로의 재분극 유도 방법을 제공하는 데에 있다.In addition, another object of the present invention is a reagent composition for inducing repolarization of M2 macrophages into M1 macrophages in vitro, comprising CXCL11 as an active ingredient, and inducing repolarization of M2 macrophages into M1 macrophages using the same. The purpose is to provide a method.

상기 목적을 달성하기 위하여, 본 발명은 CXCL11을 유효성분으로 포함하는 폐섬유화증 치료용 약학 조성물을 제공한다.In order to achieve the above object, the present invention provides a pharmaceutical composition for treating pulmonary fibrosis containing CXCL11 as an active ingredient.

또한, 본 발명은 CXCL11을 유효성분으로 포함하는, 시험관(in vitro) 내 M2 대식세포의 M1 대식세포로의 재분극 유도용 시약 조성물을 제공한다.Additionally, the present invention provides a reagent composition for inducing repolarization of M2 macrophages to M1 macrophages in vitro, comprising CXCL11 as an active ingredient.

또한, 본 발명은 시험관 내에서(in vitro) CXCL11을 대식세포에 처리하는 단계를 포함하는 M2 대식세포의 M1 대식세포로의 재분극 유도 방법을 제공한다.Additionally, the present invention provides a method of inducing repolarization of M2 macrophages into M1 macrophages, comprising treating macrophages with CXCL11 in vitro.

본 발명은 CXCL11에 의한 M2 대식세포의 재분극 유도를 통한 폐섬유화증 치료 용도에 관한 것으로, 구체적으로, 블레오마이신 유도 폐섬유화 동물모델을 활용하여 M0, M1 및 M2 분비체(conditioned medium, CM) 주입을 통해 섬유화 치료 효과를 확인하였고, M1 대식세포 분비체를 주입한 마우스의 폐에서 콜라겐과 같은 섬유화 지표의 감소 및 정상군에 가까운 M1/M2 ratio가 변화되어 있는 것을 확인하였다. 또한, 블레오마이신 유도 폐섬유화 동물모델을 활용하여 human recombinant CXCL11 정맥 주입을 통해 섬유화 치료효과를 확인하였고, CXCL11을 투여한 마우스의 폐에서 콜라겐과 같은 섬유화 지표의 감소 및 정상군에 가까운 M1/M2 ratio가 변화되어 있는 것을 확인하였다. 즉, 본 발명에서는 폐 섬유화증 발병과정에서 CXCL11을 통한 M2 대식세포의 재분극을 유도하여 결과적으로 폐 섬유화증 치료의 가능한 표적 인자로 제시하고자 한다.The present invention relates to the use of pulmonary fibrosis treatment by inducing repolarization of M2 macrophages by CXCL11, and specifically, injection of M0, M1, and M2 secretions (conditioned medium, CM) using a bleomycin-induced pulmonary fibrosis animal model. The effectiveness of fibrosis treatment was confirmed, and a decrease in fibrosis indicators such as collagen and a change in the M1/M2 ratio close to the normal group were confirmed in the lungs of mice injected with M1 macrophage secretory body. In addition, using a bleomycin-induced pulmonary fibrosis animal model, the fibrosis treatment effect was confirmed through intravenous injection of human recombinant CXCL11. In the lungs of mice administered CXCL11, fibrosis indicators such as collagen were reduced and the M1/M2 ratio was close to that of the normal group. It was confirmed that has changed. In other words, the present invention seeks to induce repolarization of M2 macrophages through CXCL11 during the development of pulmonary fibrosis and consequently present it as a possible target factor for the treatment of pulmonary fibrosis.

도 1은 M1 대식세포 분비체에 의한 섬유화 지표의 감소를 폐섬유화 동물모델에서 확인한 결과를 나타낸다.
도 2는 M1 대식세포 분비체에 의한 대식세포 극성조절을 폐섬유화 동물모델에서 확인한 결과를 나타낸다.
도 3은 M0, M1 및 M2 대식세포 분비체(conditioned medium, CM)의 human cytokine array를 통한 분석한 결과를 나타낸다.
도 4는 CXCL11에 의한 섬유화 지표의 감소를 폐섬유화 동물모델에서 확인한 결과를 나타낸다.
도 5는 CXCL11에 의한 대식세포 극성조절을 폐섬유화 동물모델에서 확인한 결과를 나타낸다.
Figure 1 shows the results of confirming the reduction of fibrosis indicators by M1 macrophage secretome in a pulmonary fibrosis animal model.
Figure 2 shows the results of confirming the regulation of macrophage polarity by the M1 macrophage secretome in an animal model of pulmonary fibrosis.
Figure 3 shows the results of analysis of M0, M1, and M2 macrophage secretome (conditioned medium, CM) using human cytokine array.
Figure 4 shows the results confirming the decrease in fibrosis indices by CXCL11 in a pulmonary fibrosis animal model.
Figure 5 shows the results of confirming macrophage polarity regulation by CXCL11 in a pulmonary fibrosis animal model.

폐에 세포 염증을 동반한 조직 손상은 섬유화 반응을 유도하여 섬유증의 발병 기전에 중요한 역할을 한다. 활성화된 대식세포는 일련의 항균 매개체 생산과 같은 일련의 선천적 면역 방어 전략을 사용하는데, 효율적인 염증 반응의 중요한 부작용은 조직 재생을 위한 복구 반응의 지속화에 따른 과도한 세포 외 기질 성분의 축적으로 인한 조직의 섬유화이다. 대체로 활성화된 M2 대식세포는 상처 치유에 중요한 역할을 하며 친섬유화 표현형을 획득하는데 이 표현형은 섬유화 면역 반응이 최고조에 달할 때 관찰되기 때문에 M2 대식세포가 섬유화의 중요한 유도자이자 조절자인 것으로 연구되고 있다. Tissue damage accompanied by cellular inflammation in the lung induces a fibrotic response and plays an important role in the pathogenesis of fibrosis. Activated macrophages employ a series of innate immune defense strategies, such as the production of a series of antibacterial mediators. An important side effect of an efficient inflammatory response is tissue damage due to the accumulation of excessive extracellular matrix components followed by the continuation of the repair response for tissue regeneration. It is fibrosis. In general, activated M2 macrophages play an important role in wound healing and acquire a pro-fibrotic phenotype. Since this phenotype is observed when the fibrotic immune response is at its peak, M2 macrophages are being studied as important inducers and regulators of fibrosis.

대식세포의 극성을 통한 폐 섬유화증 억제 연구는 활발히 진행되고 있고, 대식세포 유래 세포외 소포체 및 miRNA, peptide 등을 이용하여 M2 대식세포의 활성을 억제한다고 보고되고 있다. CXCL11 또한 폐섬유화 유도 마우스에 투여 시 폐의 혈관 신생 활성을 억제하고 콜라겐의 증가를 회복시켰다고 보고된 바 있으나, 이 연구는 섬유화 유도 약물인 bleomycin과 CXCL11의 동시처리가 진행되면서 치료의 개념이 아닌 예방에 가까운 연구가 진행되었다. 섬유화 억제 또한 본 발명에서 제시하고자 하는 M2 대식세포의 극성조절을 통한 기전 및 치료 잠재력은 아직 밝혀지지 않았다. 이미 진행 중인 폐 섬유화는 정상으로 되돌릴 수 없기 때문에 폐 섬유화 치료약물의 개발이 시급한 만큼, 본 발명은 폐 섬유화에 핵심적인 역할을 하는 면역세포인 M2 대식세포를 선택적으로 억제함으로써 부작용을 최소화하면서 치료를 극대화 시킬 수 있는 새로운 치료전략으로 활용될 수 있다.Research on suppressing pulmonary fibrosis through macrophage polarization is actively underway, and it has been reported that the activity of M2 macrophages is suppressed using macrophage-derived extracellular vesicles, miRNA, and peptides. It has been reported that CXCL11 also inhibited angiogenic activity in the lung and restored the increase in collagen when administered to mice inducing pulmonary fibrosis. However, this study showed that simultaneous treatment of bleomycin, a fibrosis-inducing drug, and CXCL11 was carried out, so that it was not a treatment concept, but rather a prevention concept. A similar study was conducted. In addition, the mechanism and therapeutic potential of suppressing fibrosis through controlling the polarity of M2 macrophages proposed in the present invention have not yet been revealed. Since pulmonary fibrosis that is already in progress cannot be returned to normal, the development of a treatment drug for pulmonary fibrosis is urgent. The present invention provides treatment while minimizing side effects by selectively inhibiting M2 macrophages, immune cells that play a key role in pulmonary fibrosis. It can be used as a new treatment strategy to maximize treatment.

본 발명은 CXCL11을 유효성분으로 포함하는 폐섬유화증 치료용 약학 조성물을 제공한다.The present invention provides a pharmaceutical composition for treating pulmonary fibrosis containing CXCL11 as an active ingredient.

바람직하게는, 상기 폐섬유화증은 특발성 폐섬유화증(idiopathic pulmonary fibrosis, IPF)일 수 있으나, 이에 한정되는 것은 아니다.Preferably, the pulmonary fibrosis may be idiopathic pulmonary fibrosis (IPF), but is not limited thereto.

바람직하게는, 상기 약학 조성물은 M2 대식세포의 감소 및 M1 대식세포의 증가를 유도할 수 있다. Preferably, the pharmaceutical composition can induce a decrease in M2 macrophages and an increase in M1 macrophages.

바람직하게는, 상기 약학 조성물은 M2 대식세포의 M1 대식세포로의 재분극을 유도할 수 있다. Preferably, the pharmaceutical composition is capable of inducing repolarization of M2 macrophages into M1 macrophages.

본 발명의 약학 조성물은 유효 성분 이외에 약제학적으로 적합하고 생리학적으로 허용되는 보조제를 사용하여 제조될 수 있으며, 상기 보조제로는 부형제, 붕해제, 감미제, 결합제, 피복제, 팽창제, 윤활제, 활택제 또는 향미제 등의 가용화제를 사용할 수 있다. 본 발명의 약학 조성물은 투여를 위해서 유효 성분 이외에 추가로 약제학적으로 허용 가능한 담체를 1 종 이상 포함하여 의약 조성물로 바람직하게 제제화할 수 있다. 액상 용액으로 제제화되는 조성물에 있어서 허용 가능한 약제학적 담체로는, 멸균 및 생체에 적합한 것으로서, 식염수, 멸균수, 링거액, 완충 식염수, 알부민 주사용액, 덱스트로즈 용액, 말토 덱스트린 용액, 글리세롤, 에탄올 및 이들 성분 중 1 성분 이상을 혼합하여 사용할 수 있으며, 필요에 따라 항산화제, 완충액, 정균제 등 다른 통상의 첨가제를 첨가할 수 있다. 또한 희석제, 분산제, 계면활성제, 결합제 및 윤활제를 부가적으로 첨가하여 수용액, 현탁액, 유탁액 등과 같은 주사용 제형, 환약, 캡슐, 과립 또는 정제로 제제화할 수 있다. The pharmaceutical composition of the present invention can be prepared using pharmaceutically suitable and physiologically acceptable auxiliaries in addition to the active ingredients, and the auxiliaries include excipients, disintegrants, sweeteners, binders, coating agents, swelling agents, lubricants, and glidants. Alternatively, solubilizers such as flavoring agents may be used. For administration, the pharmaceutical composition of the present invention can be preferably formulated as a pharmaceutical composition containing one or more pharmaceutically acceptable carriers in addition to the active ingredient. Acceptable pharmaceutical carriers for compositions formulated as liquid solutions include those that are sterile and biocompatible, such as saline solution, sterile water, Ringer's solution, buffered saline solution, albumin injection solution, dextrose solution, maltodextrin solution, glycerol, ethanol, and One or more of these ingredients can be mixed and used, and other common additives such as antioxidants, buffers, and bacteriostatic agents can be added as needed. In addition, diluents, dispersants, surfactants, binders, and lubricants can be additionally added to formulate injectable formulations such as aqueous solutions, suspensions, emulsions, etc., pills, capsules, granules, or tablets.

본 발명의 약학 조성물의 약제 제제 형태는 과립제, 산제, 피복정, 정제, 캡슐제, 좌제, 시럽, 즙, 현탁제, 유제, 점적제 또는 주사 가능한 액제 및 활성 화합물의 서방출형 제제 등이 될 수 있다. 본 발명의 의약 조성물은 정맥내, 동맥내, 복강내, 근육내, 동맥내, 복강내, 흉골내, 경피, 비측내, 흡입, 국소, 직장, 경구, 안구내 또는 피내 경로를 통해 통상적인 방식으로 투여할 수 있다. 본 발명의 의약 조성물의 유효성분의 유효량은 질환의 예방 또는 치료 요구되는 양을 의미한다. 따라서, 질환의 종류, 질환의 중증도, 조성물에 함유된 유효 성분 및 다른 성분의 종류 및 함량, 제형의 종류 및 환자의 연령, 체중, 일반 건강 상태, 성별 및 식이, 투여 시간, 투여 경로 및 조성물의 분비율, 치료 기간, 동시 사용되는 약물을 비롯한 다양한 인자에 따라 조절될 수 있다. The pharmaceutical preparation form of the pharmaceutical composition of the present invention may be granules, powders, coated tablets, tablets, capsules, suppositories, syrups, juices, suspensions, emulsions, drops or injectable solutions, and sustained-release preparations of the active compound. You can. The pharmaceutical composition of the present invention can be administered in any conventional manner via intravenous, intraarterial, intraperitoneal, intramuscular, intraarterial, intraperitoneal, intrasternal, transdermal, intranasal, inhalational, topical, rectal, oral, intraocular or intradermal routes. It can be administered. The effective amount of the active ingredient of the pharmaceutical composition of the present invention means the amount required for the prevention or treatment of disease. Therefore, the type of disease, the severity of the disease, the type and content of the active ingredient and other ingredients contained in the composition, the type of dosage form and the patient's age, weight, general health condition, gender and diet, administration time, administration route and composition. It can be adjusted depending on a variety of factors, including secretion rate, duration of treatment, and concurrent medications.

또한, 본 발명은 CXCL11을 유효성분으로 포함하는, 시험관(in vitro) 내 M2 대식세포의 M1 대식세포로의 재분극 유도용 시약 조성물을 제공한다. Additionally, the present invention provides a reagent composition for inducing repolarization of M2 macrophages to M1 macrophages in vitro, comprising CXCL11 as an active ingredient.

또한, 본 발명은 시험관 내에서(in vitro) CXCL11을 대식세포에 처리하는 단계를 포함하는 M2 대식세포의 M1 대식세포로의 재분극 유도 방법을 제공한다.Additionally, the present invention provides a method of inducing repolarization of M2 macrophages into M1 macrophages, comprising treating macrophages with CXCL11 in vitro.

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 skilled in the art that the scope of the present invention is not limited by these examples according to the gist of the present invention. .

<< 실험예Experiment example >>

하기의 실험예들은 본 발명에 따른 각각의 실시예에 공통적으로 적용되는 실험예를 제공하기 위한 것이다.The following experimental examples are intended to provide experimental examples commonly applied to each embodiment according to the present invention.

1. 실험동물1. Experimental animals

C57BL/6종 수컷 8주령 마우스를 두열바이오텍(서울, 한국)으로부터 구입하여 동물실험윤리위원회가 KW-211006-1 동물실험을 승인하였다. 마우스는 zoletil 50 (버박코리아, 한국) 및 Rumpun 근육이완 (바이엘 헬스케어, 20μl/kg, IP injection) 후 1mg/kg 블레오마이신을 기도 내 주입하여 폐 섬유화를 9일간 유도하였다. 폐 섬유화 유도 마우스에 대식세포 분비체 50ug/mouse를 블레오마이신 주입 후 7, 9, 11일에 정맥 투여하였다. 블레오마이신 주입 후 14일에 마우스를 희생시킨 후 폐 조직을 적출하여 효능 평가를 시행하였다.C57BL/strain 6 male 8-week-old mice were purchased from Duyeol Biotech (Seoul, Korea), and the Animal Experiment Ethics Committee approved KW-211006-1 animal testing. In mice, pulmonary fibrosis was induced for 9 days by intratracheal injection of 1mg/kg bleomycin after zoletil 50 (Vervac Korea, Korea) and Rumpun muscle relaxation (Bayer Healthcare, 20μl/kg, IP injection). Macrophage secretory 50ug/mouse was intravenously administered to mice induced with pulmonary fibrosis 7, 9, and 11 days after bleomycin injection. Mice were sacrificed 14 days after bleomycin injection, lung tissue was removed, and efficacy was evaluated.

2. 2. TrichromeTrichrome stain ( stain ( abcamabcam , , ab150686ab150686 ))

1) 탈파라핀 슬라이드를 50-60℃ Bouin's 용액에 담궈 60분 동안 반응하고, 10분 동안 식혔다.1) Deparaffinized slides were immersed in Bouin's solution at 50-60°C, reacted for 60 minutes, and cooled for 10 minutes.

2) 물로 3번 세척 후 Weigert's Iron Hematoxylin에 5분 동안 담갔다.2) After washing with water three times, it was soaked in Weigert's Iron Hematoxylin for 5 minutes.

3) 물로 3번 세척 후 Biebrich Scarlet / Acid Fuchsin 용액에 15분 동안 담갔다.3) After washing with water three times, it was soaked in Biebrich Scarlet / Acid Fuchsin solution for 15 minutes.

4) 물로 3번 세척 후 phosphomolybdic / phosphotungstic acid 용액에 10-15분 동안 반응시켰다.4) After washing with water three times, it was reacted in phosphomolybdic/phosphotungstic acid solution for 10-15 minutes.

5) Aniline Blue solution에 5-10 동안 슬라이드를 담갔다.5) The slide was soaked in Aniline Blue solution for 5-10.

6) 물로 3번 세척 후 acetic acid 용액에 3-5분 동안 담갔다.6) After washing with water three times, it was soaked in acetic acid solution for 3-5 minutes.

7) 물기 제거 후 마운트(mount)하였다.7) After removing the moisture, it was mounted.

3. sirius stain (3. sirius stain ( abcam,ab150681abcam,ab150681 ))

1) 탈파라핀 슬라이드를 picro-sirius red 용액에 60분 동안 반응시켰다.1) Deparaffinized slides were reacted in picro-sirius red solution for 60 minutes.

2) acetic acid 용액에 세척 후 absolute alcohol에 다시 세척하였다.2) After washing with acetic acid solution, it was washed again with absolute alcohol.

3) 물기 제거 후 마운트(mount)하였다.3) After removing the moisture, it was mounted.

4. 형광 염색4. Fluorescent staining

1) 탈파라핀 (Deparaffinization) 슬라이드를 Antigen retrieval 용액에 담그고, 121℃, 1분 동안 진행하였다. 증류수에 10분 동안 세척 후 PBS에 3분 동안 담그고 PBS-T(0.1% tween 20)에 3분 동안 세척하였다.1) Deparaffinization Slides were immersed in Antigen retrieval solution and carried out at 121°C for 1 minute. After washing in distilled water for 10 minutes, it was soaked in PBS for 3 minutes and washed in PBS-T (0.1% tween 20) for 3 minutes.

2) Hydrogen peroxide 과정이 끝난 슬라이드는 Peroxidase-blocking solution에 20분 동안 반응 후 PBS-T로 세척하였다. 10% Normal Goat Serum을 이용하여 blocking 후 collagen (abcam, ab34710, 1:400), α-SMA (Santa cruz, sc-53015,1:500), iNOS (NB300-605,NOVUS,1:400), CD206 (abcam,ab64693,1:800), F4/80 (Biorad,MCA497,1:400) 1차 항체에 4℃에서 하루 동안 반응시켰다.2) Slides that have completed the hydrogen peroxide process are After reacting in peroxidase-blocking solution for 20 minutes, it was washed with PBS-T. After blocking using 10% Normal Goat Serum, collagen (abcam, ab34710, 1:400), α-SMA (Santa cruz, sc-53015,1:500), iNOS (NB300-605,NOVUS,1:400), It was reacted with CD206 (abcam, ab64693, 1:800) and F4/80 (Biorad, MCA497, 1:400) primary antibodies at 4°C for one day.

3) PBS-T로 세척 후, Goat-anti-rabbit 488, 594 2차 항체를 상온에서 1시간 반응시켰다.3) After washing with PBS-T, Goat-anti-rabbit 488 and 594 secondary antibodies were reacted at room temperature for 1 hour.

4) PBS-T로 세척 후, Fluoreshield Mounting Medium with DAPI (Abcam, #ab104139)를 이용하여 마운트(mount)하였다.4) After washing with PBS-T, it was mounted using Fluoreshield Mounting Medium with DAPI (Abcam, #ab104139).

5. 단백질 분석 (SDS-PAGE)5. Protein analysis (SDS-PAGE)

Protein extraction solution(RIPA) 10ml(ELPIS BIOTECH, EBA-1149)과 100X protease inhibitor cocktail 100μl(Thermo scientific, 1860932) 100X EDTA solution 100μl(Thermo scientific, 1860851)으로 protein lysis buffer를 만들었으며, 300μl를 이용하여 폐조직 단백질을 분리하였다. 단백질 정량을 위해 BSA 1000μg/ml, 500μg/ml, 250μg/ml, 125μg/ml, 62.25μg/ml, 31.125μg/ml, 0μg/ml(D.W)로 standard를 만들었고, sample과 standard를 각각 20μl씩 96 well에 분주하였다. PierceTM BCA protein assay reagent A(Thermo scientific, 23228)와 B(Thermo scientific, 23224)를 각각 196μl, 4μl씩 standard well과 sample well에 넣고 36℃ 20분간 반응시켰다. 이후 micro plate spectrophotometer로 562nm에 해당되는 흡광도를 측정하여 단백질을 정량하였다. 정량된 단백질은 protein 5X sample buffer(ELPIS BIOTECH, EBA-1052), protein lysis buffer를 이용하여 20μg/10μl 농도로 만든 후 100℃ 10분간 heating 및 ice에 10분간 cooling하였다. 10% running gel, staking gel을 만들었으며 10μg의 샘플을 로딩하였다. Col1a1(Santa cruz, sc-293182,1:500) α-SMA(Santa cruz, sc-53015,1:500), actin (Santa cruz, sc-47778, 1:5000)의 protein level을 확인하였다.Protein lysis buffer was prepared with 10ml of protein extraction solution (RIPA) (ELPIS BIOTECH, EBA-1149), 100μl of 100X protease inhibitor cocktail (Thermo scientific, 1860932), and 100μl of 100X EDTA solution (Thermo scientific, 1860851). Tissue proteins were isolated. For protein quantification, standards were made with BSA 1000μg/ml, 500μg/ml, 250μg/ml, 125μg/ml, 62.25μg/ml, 31.125μg/ml, 0μg/ml (DW), and 20μl each of sample and standard were 96 It was busy in the well. Pierce TM BCA protein assay reagent A (Thermo scientific, 23228) and B (Thermo scientific, 23224) were added to standard wells and sample wells (196 μl and 4 μl, respectively) and reacted at 36°C for 20 minutes. Afterwards, the protein was quantified by measuring the absorbance at 562 nm using a micro plate spectrophotometer. The quantified protein was adjusted to a concentration of 20 μg/10 μl using protein 5X sample buffer (ELPIS BIOTECH, EBA-1052) and protein lysis buffer, then heated at 100°C for 10 minutes and cooled on ice for 10 minutes. A 10% running gel and staking gel were created, and 10 μg of sample was loaded. Protein levels of Col1a1 (Santa cruz, sc-293182, 1:500), α-SMA (Santa cruz, sc-53015, 1:500), and actin (Santa cruz, sc-47778, 1:5000) were confirmed.

6. Human 6.Human XLXL cytokinecytokine array (R&D systems, array (R&D systems, ARY022BARY022B ))

프로테옴 프로파일러 휴먼 XL 사이토카인 어레이는 멤브레인(membrane) 기반 면역분석법이다. 니트로셀룰로오스 멤브레인 이중으로 발견되는 포획 항체는 샘플에 존재하는 특정 표적 단백질에 결합하였다. 멤브레인에 결합된 단백질은 바이오틴화 검출 항체로 검출한 다음 화학 발광 검출 시약을 사용하여 시각화하였다. 발광되는 신호는 결합된 분석 물질의 양에 비례한다. 총 105가지 human 사이토카인과 케모카인을 검출할 수 있다.Proteome Profiler Human XL Cytokine Array is a membrane-based immunoassay. Capture antibodies found double on the nitrocellulose membrane bound to specific target proteins present in the sample. Proteins bound to the membrane were detected with a biotinylated detection antibody and then visualized using a chemiluminescent detection reagent. The signal emitted is proportional to the amount of bound analyte. A total of 105 human cytokines and chemokines can be detected.

1) 각 멤브레인을 별도의 well에 넣고 Array buffer를 well에 채운 후 rocking platform shaker에 1시간 동안 반응시켰다.1) Each membrane was placed in a separate well, filled with array buffer, and reacted on a rocking platform shaker for 1 hour.

2) blocking 후 Array buffer를 석션 후, 샘플을 well에 로딩하였다. 4℃가 유지되는 곳에 rocking platform shaker 상에서 하루 동안 반응시켰다.2) After blocking, the array buffer was suctioned and the sample was loaded into the well. The reaction was performed for one day on a rocking platform shaker where the temperature was maintained at 4°C.

3) 멤브레인을 개별 플라스틱 용기에 옮기고, 1X Wash Buffer로 10분 동안 세척하였다. 두 번 반복하였다.3) The membrane was transferred to an individual plastic container and washed with 1X Wash Buffer for 10 minutes. Repeated twice.

4) 단백질을 검출할 항체를 넣고 1시간 동안 rocking platform shaker 상에서 반응시켰다.4) Antibodies to detect proteins were added and reacted on a rocking platform shaker for 1 hour.

5) 1X Wash Buffer로 10분 동안 세척 후, 1X Streptavidin-HRP를 넣고 30분 동안 rocking platform shaker 상에서 반응시켰다.5) After washing with 1X Wash Buffer for 10 minutes, 1X Streptavidin-HRP was added and reacted on a rocking platform shaker for 30 minutes.

6) 1X Wash Buffer로 10분 동안 세척 후, Chemi Reagent Mix를 멤브레인에 골고루 퍼지도록 한 후, 1분간 반응시켰다.6) After washing with 1X Wash Buffer for 10 minutes, Chemi Reagent Mix was spread evenly on the membrane and reacted for 1 minute.

7) 멤브레인을 자동 방사선 촬영 필름에 넣고 1-10분 동안 노출시켰다.7) The membrane was placed in autoradiography film and exposed for 1-10 minutes.

7. 7. BMDMBMDM (Bone marrow-derived macrophage) 분화 및 M2 극성 변화(Bone marrow-derived macrophage) differentiation and M2 polarity changes.

1) 마우스의 골수를 PBS 플러시를 통해 수집하였다.1) The bone marrow of the mouse was collected through a PBS flush.

2) 분리한 골수 세포는 DMEM + 10% FBS + 1% 페니실린/스트렙토마이신 + 50ng/ml GM-CSF 배지에 배양해 대식세포(macrophage)로 분화시켰다.2) The isolated bone marrow cells were cultured in DMEM + 10% FBS + 1% penicillin/streptomycin + 50ng/ml GM-CSF medium and differentiated into macrophages.

3) 분화된 대식세포는 IL-4 20ng/ml에 12시간 노출시켜 M2 대식세포로 분화를 시켰다.3) Differentiated macrophages were exposed to 20ng/ml of IL-4 for 12 hours to differentiate into M2 macrophages.

4) 실험군은 다음과 같다.4) The experimental group is as follows.

- Naive : negative control- Naive: negative control

- IL4 : M2 대식세포- IL4: M2 macrophages

- I+L : IL4와 LPS에 동시 노출로 M1 대식 세포로 재분극을 유도하였다.- I+L: Simultaneous exposure to IL4 and LPS induced repolarization into M1 macrophages.

- I+FA : IL4 와 CXCL11 동시 노출로 M2 대식세포에 CXCL11에 의한 재분극을 유도하였다.- I+FA: Simultaneous exposure to IL4 and CXCL11 induced repolarization by CXCL11 in M2 macrophages.

- L : M1 대식세포 유도제로 LPS를 처리하여 M1 대식세포로 유도하였다.- L: M1 macrophages were induced by treating LPS as an M1 macrophage inducer.

위와 같은 실험군을 설정하여 12시간 동안 처리 후, M1 마커(iNOS, Socs3), M2 마커 (Arg1,Mrc3)를 mRNA 수준에서 확인하였다.The experimental group as above was set up and treated for 12 hours, and M1 markers (iNOS, Socs3) and M2 markers (Arg1, Mrc3) were confirmed at the mRNA level.

<< 실시예Example 1> M1 대식세포 분비체에 의한 섬유화 지표의 감소를 폐섬유화 동물모델에서 확인 1> A decrease in fibrosis indicators due to M1 macrophage secretion was confirmed in a pulmonary fibrosis animal model.

블레오마이신 유도 폐섬유화 동물모델을 활용하여 M0, M1 및 M2 분비체(conditioned medium, CM) 주입을 통해 섬유화 치료 효과를 확인하였다. 마우스의 폐조직에서 섬유화 지표 col1a1과 αSMA의 발현 확인 결과, M0와 M2 분비체 주입한 마우스 대비 M1 분비체를 주입한 마우스에서 섬유화 지표의 감소를 확인하였다(도 1A).Using a bleomycin-induced pulmonary fibrosis animal model, the fibrosis treatment effect was confirmed through injection of M0, M1, and M2 secretions (conditioned medium, CM). As a result of confirming the expression of fibrosis indicators col1a1 and αSMA in the lung tissue of mice, a decrease in fibrosis indicators was confirmed in mice injected with M1 secretion compared to mice injected with M0 and M2 secretion (Figure 1A).

단백질 발현 정도를 정량화하여 그래프로 나타낸 결과, M0와 M2 분비체 주입한 마우스 대비 M1 분비체를 주입한 마우스에서 섬유화 지표의 감소를 확인하였다(도 1B).As a result of quantifying the protein expression level and graphing it, a decrease in fibrosis index was confirmed in mice injected with M1 secretion compared to mice injected with M0 and M2 secretion (Figure 1B).

마우스의 폐조직 paraffin-slide에서 trichrome, sirius 염색을 통해 아교섬유(collagen fiber)의 축적 정도를 확인하고, 동일한 조직에서 collagen, αSMA의 발현을 형광 염색을 통하여 확인하였다. M0와 M2 분비체 주입한 마우스 대비 M1 분비체를 주입한 마우스에서 collagen fiber의 감소를 확인되고, 또한 형광염색으로 확인한 collagen, αSMA의 발현도 M1 분비체를 주입한 마우스에서 확연한 감소를 확인하였다(도 1C).The degree of accumulation of collagen fibers was confirmed through trichrome and sirius staining in paraffin-slides of mouse lung tissue, and the expression of collagen and αSMA in the same tissue was confirmed through fluorescence staining. A decrease in collagen fibers was confirmed in mice injected with M1 secretion compared to mice injected with M0 and M2 secretion, and the expression of collagen and αSMA, confirmed by fluorescent staining, was also confirmed to be significantly reduced in mice injected with M1 secretion ( Figure 1C).

M0와 M2 분비체 주입한 마우스 대비 M1 분비체를 주입한 마우스에서 섬유화 지표의 감소를 확인하였고, 발현 정도를 intensity로 정량화하여 그래프로 나타냈을 때, 동일하게 M1 분비체를 주입한 마우스에서 collagen, αSMA의 발현 감소를 확인하였다(도 1D).A decrease in fibrosis indicators was confirmed in mice injected with M1 secretion compared to mice injected with M0 and M2 secretion, and when the expression level was quantified by intensity and displayed in a graph, in mice injected with M1 secretion, collagen, A decrease in the expression of αSMA was confirmed (Figure 1D).

<< 실시예Example 2> M1 대식세포 분비체에 의한 대식세포 극성조절을 폐섬유화 동물모델에서 확인 2> Control of macrophage polarity by M1 macrophage secretome was confirmed in an animal model of pulmonary fibrosis

블레오마이신 유도 폐섬유화 동물모델을 활용하여 M0, M1 및 M2 분비체(conditioned medium, CM) 주입을 통해, M1과 M2 대식세포의 극성 확인하였다. 마우스의 폐조직 내 F4/80+iNOS(M1), F4/80+CD206(M2)를 형광 염색하여 확인했을 때 bleomycin 모델에서 증가한 F4/80+CD206(M2)는 M0와 M2 분비체 주입한 마우스 대비 M1 분비체를 주입한 마우스에서 F4/80+CD206(M2)의 감소를 보이고, bleomycin 모델에서 감소한 F4/80+iNOS(M1)은 M1 분비체를 주입한 마우스에서 증가를 보였다. 이는 M1 분비체의 M2 대식세포의 감소 및 M1 대식세포의 증가를 유도한다는 것을 확인하였다(도 2A).Using a bleomycin-induced pulmonary fibrosis animal model, the polarity of M1 and M2 macrophages was confirmed through injection of M0, M1, and M2 secretome (conditioned medium, CM). When F4/80+iNOS(M1) and F4/80+CD206(M2) in mouse lung tissue were confirmed by fluorescent staining, F4/80+CD206(M2) increased in the bleomycin model in mice injected with M0 and M2 secretions. In contrast, mice injected with M1 secretome showed a decrease in F4/80+CD206(M2), and F4/80+iNOS(M1), which was decreased in the bleomycin model, showed an increase in mice injected with M1 secretion. It was confirmed that this led to a decrease in M2 macrophages and an increase in M1 macrophages in the M1 secretome (Figure 2A).

M1, M2 마커의 발현을 field당 수를 count하여 ratio로 분석했을 때, M1 대식세포 분비체를 주입한 마우스에서 M1/M2 ratio는 정상에 가깝게 변화하는 것을 확인하였다. 이를 정량화하여 그래프로 나타냈다. 이는 M1 분비체는 정상군에 유사한 M1/M2 ratio의 변화를 유도한다는 것을 확인하였다(도 2B).When the expression of M1 and M2 markers was analyzed as a ratio by counting the number per field, it was confirmed that the M1/M2 ratio changed close to normal in mice injected with M1 macrophage secretion. This was quantified and displayed in a graph. This confirmed that the M1 secretome induces a change in the M1/M2 ratio similar to that in the normal group (Figure 2B).

<< 실시예Example 3> M0, M1 및 M2 대식세포 3> M0, M1 and M2 macrophages 분비체secretory body (conditioned medium, CM)의 human human in conditioned medium (CM) cytokinecytokine array를 통한 분석 Analysis through array

분비체 내 사이토카인 및 케모카인의 상대적 수준을 비교하여 항 섬유화 및 대식세포 재분극에 영향을 미치는 factor를 찾아내고자 실험을 진행하였다(도 3A).An experiment was conducted to find factors that affect anti-fibrosis and macrophage repolarization by comparing the relative levels of cytokines and chemokines in the secretome (Figure 3A).

M0와 M2 대식세포 분비체 대비 M1 대식세포 분비체에서 CXCL9, CXCL11 및 Pentraxin3의 발현이 증가되어 있음을 확인하였다. 발현 정도의 시각화된 검출양을 intensity로 표현하였다(도 3B).It was confirmed that the expression of CXCL9, CXCL11, and Pentraxin3 was increased in the M1 macrophage secretome compared to the M0 and M2 macrophage secretome. The visualized detection amount of the expression level was expressed as intensity (Figure 3B).

마우스에서 추출한 BMDM(Bone-marrow derived macrophage) 세포에서 CXCL11의 대식세포 극성조절을 확인 실험의 모식도를 도 3C에 나타냈다.A schematic diagram of an experiment confirming the regulation of macrophage polarity by CXCL11 in bone-marrow derived macrophage (BMDM) cells extracted from mice is shown in Figure 3C.

마우스 BMDM을 IL-4의 노출을 통해 M2 대식세포로 분화시킨 후, LPS(M1 극성 유도제)와 CXCL11을 처리하여 대식세포의 극성조절을 mRNA 수준에서 확인하였다. M1 마커 (iNOS, Socs3), M2 마커 (Arg1, Mrc1)를 통해 M2 대식세포의 재분극을 확인한 결과, M2 대식세포로 분화한 대식세포에 CXCL11을 처리하였을 때 M2 마커의 감소 및 M1 마커의 증가를 확인하였다. 이는 CXCL11은 M2 대식세포의 M1 대식세포로의 재분극을 유도함을 나타낸다(도 3D 및 도 3E).Mouse BMDM were differentiated into M2 macrophages through exposure to IL-4, and then treated with LPS (M1 polarity inducer) and CXCL11 to confirm macrophage polarity regulation at the mRNA level. As a result of confirming the repolarization of M2 macrophages through M1 markers (iNOS, Socs3) and M2 markers (Arg1, Mrc1), when CXCL11 was treated with macrophages differentiated into M2 macrophages, there was a decrease in M2 markers and an increase in M1 markers. Confirmed. This indicates that CXCL11 induces repolarization of M2 macrophages into M1 macrophages (Figures 3D and 3E).

<< 실시예Example 4> 4> CXCL11CXCL11 의한 섬유화 지표의 감소를 Reduction of fibrosis index by 폐섬유화pulmonary fibrosis 동물모델에서 확인 Confirmed in animal model

블레오마이신 유도 폐섬유화 동물모델을 활용하여 CXCL11 정맥 주입을 통해 항섬유화 효과를 확인하였다. 마우스의 폐조직에서 섬유화 지표 col1a1과 αSMA의 발현 확인 결과, CXCL11 주입한 마우스에서 섬유화 지표의 감소를 확인하였다(도 4A).Using a bleomycin-induced pulmonary fibrosis animal model, the anti-fibrotic effect was confirmed through intravenous injection of CXCL11. As a result of confirming the expression of fibrosis indicators col1a1 and αSMA in mouse lung tissue, a decrease in fibrosis indicators was confirmed in mice injected with CXCL11 (Figure 4A).

단백질 발현 정도를 정량화하여, 도 4B에 그래프로 나타냈다.The protein expression level was quantified and shown graphically in Figure 4B.

마우스의 폐조직 paraffin-slide에서 trichrome, sirius 염색을 통해 아교섬유(collagen fiber)의 축적 정도를 확인하고, 동일한 조직에서 collagen, αSMA의 발현을 형광 염색을 통하여 확인하였다. CXCL11을 주입한 마우스에서 collagen fiber의 감소 및 collagen, αSMA의 형광 발현 또한 감소한다는 것을 확인하였다(도 4C).The degree of accumulation of collagen fibers was confirmed through trichrome and sirius staining in paraffin-slides of mouse lung tissue, and the expression of collagen and αSMA in the same tissue was confirmed through fluorescence staining. It was confirmed that in mice injected with CXCL11, collagen fibers decreased and the fluorescence expression of collagen and αSMA also decreased (Figure 4C).

collagen, αSMA의 형광 발현 정도를 intensity로 정량화하여 그래프로 나타내었을 때, CXCL11에 의한 항 섬유화 효과를 확인하였다(도 4D).When the level of fluorescence expression of collagen and αSMA was quantified by intensity and displayed in a graph, the anti-fibrotic effect of CXCL11 was confirmed (Figure 4D).

<< 실시예Example 5> 5> CXCL11CXCL11 의한 대식세포 극성조절을 Control of macrophage polarity by 폐섬유화pulmonary fibrosis 동물모델에서 확인 Confirmed in animal model

블레오마이신 유도 폐섬유화 동물모델을 활용하여 CXCL11 주입을 통해, M1과 M2 대식세포의 극성을 확인하였다. 마우스의 폐조직 내 F4/80+iNOS(M1), F4/80+CD206(M2)를 형광 염색하여 확인했을 때 bleomycin 모델에서 증가한 F4/80+CD206(M2)는 CXCL11을 주입한 마우스에서 F4/80+CD206(M2)의 감소를 보이고, bleomycin 모델에서 감소한 F4/80+iNOS(M1)은 CXCL11을 주입한 마우스에서 증가를 보였다. 이는 CXCL11의 M2 대식세포의 감소 및 M1 대식세포의 증가를 유도한다는 것을 확인하였다(도 5A).Using a bleomycin-induced pulmonary fibrosis animal model, the polarity of M1 and M2 macrophages was confirmed through CXCL11 injection. When F4/80+iNOS(M1) and F4/80+CD206(M2) in mouse lung tissue were confirmed by fluorescent staining, F4/80+CD206(M2) increased in the bleomycin model, and F4/80+CD206(M2) was increased in mice injected with CXCL11. 80+CD206 (M2) showed a decrease, and F4/80+iNOS (M1), which was decreased in the bleomycin model, showed an increase in mice injected with CXCL11. It was confirmed that CXCL11 induced a decrease in M2 macrophages and an increase in M1 macrophages (Figure 5A).

M1, M2 마커의 발현을 field당 수를 count하여 ratio로 분석했을 때, CXCL11을 주입한 마우스에서 M1/M2 ratio는 정상에 가깝게 변화하는 것을 확인하였다. 이를 정량화하여 그래프로 나타냈다. 이는 CXCL11은 정상군에 유사한 M1/M2 ratio의 변화를 유도한다는 것을 확인하였다(도 5B).When the expression of M1 and M2 markers was analyzed by ratio by counting the number per field, it was confirmed that the M1/M2 ratio changed close to normal in mice injected with CXCL11. This was quantified and displayed in a graph. This confirmed that CXCL11 induced changes in the M1/M2 ratio similar to the normal group (Figure 5B).

이상으로 본 발명의 특정한 부분을 상세히 기술하였는 바, 당업계의 통상의 지식을 가진 자에게 있어서 이러한 구체적인 기술은 단지 바람직한 구현 예일 뿐이며, 이에 본 발명의 범위가 제한되는 것이 아닌 점은 명백하다. 따라서, 본 발명의 실질적인 범위는 첨부된 청구항과 그의 등가물에 의하여 정의된다고 할 것이다.As the specific parts of the present invention have been described in detail above, it is clear to those skilled in the art that these specific techniques are merely preferred implementation examples and do not limit the scope of the present invention. Accordingly, the substantial scope of the present invention will be defined by the appended claims and their equivalents.

Claims (6)

삭제delete 삭제delete CXCL11을 유효성분으로 포함하는, 폐섬유화 마우스 모델에서 M2 대식세포의 M1 대식세포로의 재분극 유도용 시약 조성물.A reagent composition for inducing repolarization of M2 macrophages into M1 macrophages in a pulmonary fibrosis mouse model, comprising CXCL11 as an active ingredient. 폐섬유화 마우스 모델에 CXCL11을 처리하는 단계를 포함하는 M2 대식세포의 M1 대식세포로의 재분극 유도 방법.A method for inducing repolarization of M2 macrophages into M1 macrophages, comprising treating a pulmonary fibrosis mouse model with CXCL11. CXCL11을 유효성분으로 포함하는, 시험관(in vitro) 내 M2 대식세포의 M1 대식세포로의 재분극 유도용 시약 조성물.A reagent composition for inducing repolarization of M2 macrophages into M1 macrophages in vitro, comprising CXCL11 as an active ingredient. 시험관 내에서(in vitro) CXCL11을 대식세포에 처리하는 단계를 포함하는 M2 대식세포의 M1 대식세포로의 재분극 유도 방법.A method of inducing repolarization of M2 macrophages into M1 macrophages, comprising treating macrophages with CXCL11 in vitro.
KR1020230148771A 2023-11-01 2023-11-01 Use for treating pulmonary fibrosis by inducing repolarization of M2 macrophage by CXCL11 KR102635831B1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
KR1020230148771A KR102635831B1 (en) 2023-11-01 2023-11-01 Use for treating pulmonary fibrosis by inducing repolarization of M2 macrophage by CXCL11

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
KR1020230148771A KR102635831B1 (en) 2023-11-01 2023-11-01 Use for treating pulmonary fibrosis by inducing repolarization of M2 macrophage by CXCL11

Publications (1)

Publication Number Publication Date
KR102635831B1 true KR102635831B1 (en) 2024-02-13

Family

ID=89899483

Family Applications (1)

Application Number Title Priority Date Filing Date
KR1020230148771A KR102635831B1 (en) 2023-11-01 2023-11-01 Use for treating pulmonary fibrosis by inducing repolarization of M2 macrophage by CXCL11

Country Status (1)

Country Link
KR (1) KR102635831B1 (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20200120624A (en) * 2018-02-12 2020-10-21 코디악 바이오사이언시즈, 인크. Methods and compositions for polarization of macrophages
US20210330734A1 (en) 2019-01-09 2021-10-28 Epitracker, Inc. Compositions and methods for diagnosis and treatment of neurodegenerative diseases

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20200120624A (en) * 2018-02-12 2020-10-21 코디악 바이오사이언시즈, 인크. Methods and compositions for polarization of macrophages
US20210330734A1 (en) 2019-01-09 2021-10-28 Epitracker, Inc. Compositions and methods for diagnosis and treatment of neurodegenerative diseases

Similar Documents

Publication Publication Date Title
Armour et al. Cellular and molecular pathology of HTS: basis for treatment
Hou et al. HMGB1 contributes to allergen-induced airway remodeling in a murine model of chronic asthma by modulating airway inflammation and activating lung fibroblasts
Kearley et al. IL-9 governs allergen-induced mast cell numbers in the lung and chronic remodeling of the airways
Dolivo et al. Tryptophan metabolites kynurenine and serotonin regulate fibroblast activation and fibrosis
Nonaka et al. Distinct immunohistochemical localization of IL-4 in human inflamed airway tissues. IL-4 is localized to eosinophils in vivo and is released by peripheral blood eosinophils.
Gårdlund et al. Plasma levels of cytokines in primary septic shock in humans: correlation with disease severity
van der Poll et al. Regulation of interleukin 10 release by tumor necrosis factor in humans and chimpanzees.
Fan et al. Increased gastric production of interleukin-8 and tumour necrosis factor in patients with Helicobacter pylori infection.
Denis Proinflammatory cytokines in hypersensitivity pneumonitis.
Ito et al. Hepatocyte growth factor attenuates airway hyperresponsiveness, inflammation, and remodeling
Greeneltch et al. The opioid antagonist naltrexone blocks acute endotoxic shock by inhibiting tumor necrosis factor-α production
Yamashita et al. Glycyrrhizin ameliorates fibrosis, vasculopathy, and inflammation in animal models of systemic sclerosis
Montinaro et al. Extrarenal cytokines modulate the glomerular response to IgA immune complexes
Jankov et al. Gadolinium chloride inhibits pulmonary macrophage influx and prevents O2-induced pulmonary hypertension in the neonatal rat
JP6327626B2 (en) Inhibitors of leukocyte extracellular trap formation
Huang et al. Roxadustat attenuates experimental pulmonary fibrosis in vitro and in vivo
EP0731697B1 (en) Ssi tyrphostins and pharmaceutical compositions
US5543143A (en) Method for activating macrophages/monocytes
Luong et al. Inhibition of the Progression of Skin Inflammation, Fibrosis, and Vascular Injury by Blockade of the CX 3 CL 1/CX 3 CR 1 Pathway in Experimental Mouse Models of Systemic Sclerosis
Zhang et al. Reversing CXCL10 deficiency ameliorates kidney disease in diabetic mice
Mayoral et al. Decreased tumor necrosis factor production during the initial stages of infection correlates with survival during murine gram-negative sepsis
KR100571010B1 (en) Drugs containing as the active ingredient Midkine or inhibitors thereof
KR102635831B1 (en) Use for treating pulmonary fibrosis by inducing repolarization of M2 macrophage by CXCL11
US20220080044A1 (en) Method for preventing or treating peripheral arterial occlusive disease
JP2023521634A (en) Methods and compositions for treating or preventing inflammatory conditions

Legal Events

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