WO2024039197A1 - Composition for preventing or treating neurodegenerative disease comprising compounds originating from aspergillus sp. sf-7402 - Google Patents

Composition for preventing or treating neurodegenerative disease comprising compounds originating from aspergillus sp. sf-7402 Download PDF

Info

Publication number
WO2024039197A1
WO2024039197A1 PCT/KR2023/012186 KR2023012186W WO2024039197A1 WO 2024039197 A1 WO2024039197 A1 WO 2024039197A1 KR 2023012186 W KR2023012186 W KR 2023012186W WO 2024039197 A1 WO2024039197 A1 WO 2024039197A1
Authority
WO
WIPO (PCT)
Prior art keywords
composition
disease
cells
lps
sterigmatocystin
Prior art date
Application number
PCT/KR2023/012186
Other languages
French (fr)
Korean (ko)
Inventor
임정한
김일찬
한세종
오현철
손재학
조동규
Original Assignee
한국해양과학기술원
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 한국해양과학기술원 filed Critical 한국해양과학기술원
Publication of WO2024039197A1 publication Critical patent/WO2024039197A1/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/35Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom
    • A61K31/352Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom condensed with carbocyclic rings, e.g. methantheline 
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K36/00Medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicines
    • A61K36/06Fungi, e.g. yeasts
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia

Definitions

  • the present invention relates to the Antarctic fungus Aspergillus sp.
  • Neuroinflammation an inflammatory response within the brain or spinal cord, is mediated by the production of cytokines, chemokines, reactive oxygen species, and second messengers.
  • the role of these inflammatory mediators in the central nervous system (CNS) has been investigated in various neurodegenerative diseases, including Parkinson’s disease (PD), Huntington’s disease (HD), and Alzheimer’s disease (AD).
  • PD Parkinson’s disease
  • HD Huntington’s disease
  • AD Alzheimer’s disease
  • Microglia play an active role in immune surveillance of the CNS by producing factors that affect surrounding astrocytes and neurons, especially in response to pathogen invasion and tissue damage. Rapid microglial activation reflects the tissue's response to injury to initiate wound healing and protect neurons from further damage. Therefore, activated microglia are known to have a neuroprotective effect.
  • inflammatory microglia produce pro-inflammatory molecules, including nitric oxide (NO), inducible nitric oxide synthase (iNOS), and cyclooxygenase (COX)-2.
  • NO nitric oxide
  • iNOS inducible nitric oxide synthase
  • COX cyclooxygenase
  • NF- ⁇ B has also been demonstrated to be a key signaling agent affecting cell permeability, endocytosis, and intracellular transport at the blood-brain barrier level. Therefore, NF- ⁇ B plays an important role in regulating the causes of diseases related to neuroinflammation (Shih, R.H.; Wang, C.Y.; Yang, C.M. NF-kappaB signaling pathways in neurological inflammation: A mini review. Front. Mol. Neurosci. 2015 , 8, 77.)
  • the purpose of the present invention is to provide a composition for preventing or treating neurodegenerative disease containing a xanthone compound.
  • the purpose of the present invention is to provide a method for preventing or treating neurodegenerative disease including a xanthone compound.
  • the purpose of the present invention is to provide a xanthone compound for use in the prevention or treatment of neurodegenerative disease.
  • the object of the present invention is to provide a method for producing the composition.
  • the present invention provides a composition for preventing or treating neurodegenerative disease, comprising a compound represented by the following structural formula 1 as an active ingredient:
  • R is alkoxy or hydrogen.
  • the present invention provides a method for preventing or treating neurodegenerative diseases, comprising administering a compound represented by structural formula 1 to a patient.
  • the present invention provides the use of the compound represented by Structural Formula 1 in the production of a composition for preventing or treating neurodegenerative diseases.
  • the present invention relates to the fungus Aspergillus sp. Culturing SF-7402 to extract metabolites; and separating xanthone compounds from metabolites.
  • Figure 1 shows the structure of the isolated metabolite.
  • Figure 2 shows the results of confirming the effect of compounds on cell viability (A) and nitrite content (B).
  • BV2 cells were incubated with various concentrations of compounds 1-4 for 24 hours. Cell viability was determined using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay.
  • BV2 cells were pretreated with the indicated concentrations of compounds for 3 h and stimulated with lipopolysaccharide (LPS) (0.5 ⁇ g/mL) for 24 h. Bars represent the mean standard deviation of three independent experiments. ***p ⁇ 0.001 compared to LPS treatment group.
  • LPS lipopolysaccharide
  • Figure 3 shows inducible nitric oxide synthase (iNOS) (A, C) and cyclooxygenase-2 (COX-2) induced in BV2 cells stimulated with lipopolysaccharide (LPS).
  • iNOS inducible nitric oxide synthase
  • COX-2 cyclooxygenase-2
  • B,D shows the results of confirming the protein expression level.
  • Cells were pretreated with the indicated concentrations of compounds 1 and 2 for 3 h and stimulated with LPS (0.5 g/mL) for 24 h. Representative blots from three independent experiments are shown. Immunoblots were quantified using ImageJ software. Band intensity was normalized to ⁇ -actin. *p ⁇ 0.05, **p ⁇ 0.01 compared to LPS treatment group.
  • Figure 4 shows the effects of the isolated compounds on TNF- ⁇ (A), IL-6 (B), and PGE2 (C) in BV2 cells stimulated with LPS.
  • Cells were pretreated with the indicated concentrations of compounds 1-4 for 3 h and stimulated with LPS (0.5 ⁇ g/mL) for 24 h. Bars represent the mean standard deviation of three independent experiments. *p ⁇ 0.05, **p ⁇ 0.01, ***p ⁇ 0.001 compared to LPS treatment group.
  • Figure 5 shows the effects of isolated compounds on the NF- ⁇ B (p65) pathway in BV2 cells (A-D).
  • Cells were pretreated with the indicated concentrations of compounds 1 and 2 for 3 hours and stimulated with lipopolysaccharide (LPS, 1 g/mL) for 1 hour. Representative blots from three independent experiments are shown. Immunoblots were quantified using ImageJ software. I ⁇ B- ⁇ intensity was normalized to ⁇ -actin. P65 intensity was normalized to PCNA. *p ⁇ 0.05, **p ⁇ 0.01, ***p ⁇ 0.001 compared to LPS treatment group.
  • LPS lipopolysaccharide
  • Cicolorin A (6,8-O-dimethylversicolorin A) was isolated. Their chemical structures were revealed by extensive spectroscopic analysis, HR-ESI-MS, and comparison with those reported in the literature. The anti-neuroinflammatory effects of the isolated metabolites were assessed by measuring the production of nitric oxide (NO), TNF- ⁇ , and IL-6 at non-cytotoxic concentrations in LPS-activated microglia.
  • 5-methoxysterigmatocystin and sterigmatocystin showed a significant effect on NO production and a moderate effect on suppressing TNF- ⁇ and IL-6 expression.
  • the underlying molecular mechanisms were investigated using Western blot analysis. Treatment with 5-methoxysterigmatocystin and sterigmatocystin inhibited NO production through downregulation of nitric oxide synthase (iNOS) expression induced in LPS-stimulated BV2 cells. Additionally, 5-methoxysterigmatocystin and sterigmatocystin reduced the nuclear translocation of NF- ⁇ B.
  • the present invention relates to a composition for preventing or treating neurodegenerative disease, which contains a xanthone compound represented by the following structural formula 1 as an active ingredient:
  • R is alkoxy or hydrogen.
  • the present invention relates to a method for preventing or treating neurodegenerative disease, comprising administering a xanthone compound represented by structural formula 1 to a patient.
  • the present invention relates to the use of a xanthone compound represented by structural formula 1 in the production of a composition for preventing or treating neurodegenerative diseases.
  • Neuroinflammation is an early protective mechanism, and inflammatory mediators play a role in restoring damaged neurons and glial cells during acute neuroinflammation. However, chronic neuroinflammation tends to cause more nerve damage and ultimate degradation. Finding treatments for neuroinflammation could potentially reduce the progression of neurodegenerative diseases.
  • Nitric oxide is formed enzymatically from arginine by three nitric oxide synthase (NOS) isoforms.
  • NOS nitric oxide synthase
  • iNOS Inducible NOS
  • LPS LPS
  • cytokines various cytokines.
  • iNOS is expressed in many cell types, including microglia. In a healthy brain, microglia do not express iNOS. Nevertheless, under ischemic, traumatic, neurotoxic, or inflammatory injury, they are activated to produce iNOS and release large amounts of NO. Afterwards, cell membrane structures can be damaged, affecting DNA transcription and protein synthesis, and directly damaging neurons.
  • the iNOS promoter contains several cis-acting elements, including NF- ⁇ B, AP-1, C/EBP ⁇ , and Stat, while the COX-2 promoter contains NF- ⁇ B, C/EBP ⁇ , and CRE cis-acting elements. Promoter activity varies depending on cell type and applied stimulus.
  • PGE 2 a product of COX-2 enzyme, is also regulated by NF- ⁇ B and plays an important role in neuroinflammation.
  • compounds 1 and 2 were unable to inhibit PGE 2 production, reflecting the same effect of compounds 1 and 2 on COX-2 expression ( Figure 4C).
  • the present invention includes, for example, 5-methoxysterigmatocystin in structural formula 1, where R is methoxy, or sterigmatocystin, where R is hydrogen. It can be included.
  • Chronic neuroinflammation is mainly regulated by brain microglia and immune cells. Chronic neuroinflammation is known to be the main reason for neuronal loss during PD and forms the basis of neurodegeneration.
  • NF- ⁇ B is a member of the Rel family of transcription factors.
  • RelA (p65) is one of the five members of the Rel family. Post-transcriptional modification of p65 plays an important role in pathogenesis. Post-transcriptional modification of p65 plays an important role in the initiation of neurodegenerative processes activated by ischemic injury and glutamate or ⁇ -amyloid toxicity. The most important I ⁇ B protein is considered to be I ⁇ B- ⁇ because it interacts with the nuclear localization signal of p65.
  • Sterigmatocystin is a xanthone containing a xanthone and a bisfuran group. Sterigmatocystin derivatives are reported to have numerous biological activities, including antitumor, anti-inflammatory, antiviral and antibacterial activities.
  • compound 2 which differs from compound 1 by the absence of a methoxy group at the C-5 position, has a greater effect on the expression of p65 protein and I ⁇ B- ⁇ phosphorylation at the indicated concentrations in LPS-stimulated BV2 cells. shown ( Figure 5B). This shows the important role of methoxy among the aromatic ring groups of sterigmatocystin.
  • the compounds are derived from the Antarctic fungus Aspergillus sp. It may have been separated from SF-7402.
  • Two xanthones, sterigmatocystin (1, 2) and two anthraquinones (3, 4), present in SF-7402 were identified. It was confirmed that sterigmatocystin exhibits excellent anti-neuroinflammatory effects by inhibiting the production of NO, TNF- ⁇ , and IL-6 and the expression of iNOS protein. These effects were mediated through activation of the NF- ⁇ B pathway in LPS-stimulated BV2 microglia.
  • the neurodegenerative diseases include, for example, Parkinson's disease, Alzheimer's disease (AD), stroke, dementia, muscular dystrophy (MD), multiple sclerosis (MS), amyotrophic lateral sclerosis (ALS), cystic fibrosis, and Angelman syndrome. , Liddle syndrome, Pick disease, Paget disease, cancer, or traumatic brain injury, but are not limited thereto.
  • composition of the present invention may be prepared to include one or more pharmaceutically acceptable carriers in addition to the active ingredients described above for administration.
  • the pharmaceutically acceptable carrier must be compatible with the active ingredient of the present invention, and may include saline solution, sterile water, Ringer's solution, buffered saline solution, dextrose solution, maltodextrin solution, glycerol, ethanol, and one or two or more of these ingredients. It can be used by mixing, and other common additives such as antioxidants, buffers, and bacteriostatic agents can be added as needed.
  • diluents, dispersants, surfactants, binders, and lubricants can be additionally added to formulate injectable formulations such as aqueous solutions, suspensions, emulsions, etc.
  • injectable formulations such as aqueous solutions, suspensions, emulsions, etc.
  • a stabilizer for freeze-drying may be added.
  • it can be preferably formulated according to each disease or ingredient using an appropriate method in the art or a method disclosed by Remington's Pharmaceutical Science (Mack Publishing company, Easton PA).
  • the pharmaceutical composition of the present invention is preferably administered parenterally, and the dosage varies depending on the patient's weight, age, gender, health status, diet, administration time, method, excretion rate, or severity of the disease. An expert in the technical field can easily determine this.
  • a single dosage of the composition according to the present invention may be 1 ⁇ g/kg to 100 mg/kg, preferably 5 ⁇ g/kg to 50 mg/kg, and is administered once a day or 1-3 times a week.
  • the dosage and administration interval are not limited to this.
  • It relates to a method for preventing or treating neurodegenerative diseases, including administration to animals according to the present invention.
  • the animal may be a human, a primate such as a monkey, a dog, a pig, a cow, a sheep, a goat, a mouse, or a rat, but is not limited thereto.
  • the animal may be, for example, a human.
  • the method of administering the composition can be determined by an expert in the art based on the patient's symptoms and the severity of the disease.
  • the pharmaceutical composition of the present invention can be administered parenterally.
  • the route of administration of the composition according to the present invention is not limited to these, but includes, for example, oral cavity, intravenous, intramuscular, intraarterial, intramedullary, intrathecal, intracardiac, transdermal, subcutaneous, intraperitoneal, intestinal, sublingual. Alternatively, topical administration is possible.
  • the pharmaceutical composition of the present invention can be administered to the central nervous system (CNS).
  • the pharmaceutical composition of the present invention can be administered by intrathecal injection.
  • the dosage of the composition according to the present invention varies depending on the patient's weight, age, gender, health condition, diet, administration time, method, excretion rate, or severity of the disease, and is easily understood by an expert in the art. You can decide. Additionally, the composition of the present invention can be formulated into a suitable dosage form for clinical administration using known techniques.
  • Dose means a specified amount of pharmaceutical agent given in a single administration or over a specified period of time. Doses may be administered as one, two or more boluses, tablets, or injections. For example, in certain embodiments where subcutaneous administration is desired, the desired dose requires a volume that is not readily accommodated by a single injection, and two or more injections may be used to achieve the desired dose. Medications are administered by infusion over a long period of time or continuously. Dosage may be stated as the amount of pharmaceutical agent per hour, day, week, or month.
  • HMQC Heteronuclear multiple quantum coherence
  • HMBC heteronuclear multiple bond correlation
  • Fungal strain SF-7402 was isolated from lichen collected at King George Island, Antarctica (62°1406.15’’S, 58°46’22.68’’W) in January 2017. 1 g of sample was ground with a mortar and pestle and mixed with sterile seawater (10 mL). A portion of the sample (0.1 mL) was processed using the spread plate method in potato dextrose agar (PDA) medium containing seawater and incubated at 25°C for 14 days. After subculturing the isolate several times, the final pure culture was selected and stored at -70°C. Fungal strain SF-7402 was identified using ribosomal RNA (rRNA) sequencing.
  • rRNA ribosomal RNA
  • GenBank search using the ITS gene of SF-7402 (GenBank accession number MZ267531), Aspergillus jensenii (NR_135444), A. creber (NR_135442), A. versicolor (NR_131277), and A. protuberus (NR_135353) were identified at 99.62%, respectively. The closest matches showing sequence homology of 99.62%, 99.43%, and 99.43% were selected. Therefore, the fungal strain SF-7402 is Aspergillus sp. It was deposited on 2022.06.08 as A voucher specimen (SF-7402) (KCTC 14992BP).
  • the fungal strain Aspergillus sp. SF-7402 was cultured in 20 Fernbach flasks each containing 650 mL of PDA medium containing 3% NaCl (v/v). Flasks were individually inoculated with 2 mL seed cultures of fungal strains and incubated at 25°C for 14 days. Fermentation culture media were combined and extracted with EtOAc (20 L). The combined EtOAc extracts were then filtered through filter paper and evaporated to dryness to obtain the crude extract (4.8 g).
  • the crude extract was fractionated by reversed-phase (RP) C 18 flash column chromatography ( 30 Five fractions of SF-7402-1 to SF-7402-5 were obtained by eluting with a stepwise gradient of MeOH (500 mL each). Fraction SF-7402-5 (568.8 mg) was applied to a SephadexLH -20 column (33 7402-5.4) was obtained. Fraction SF-7402-5.2 (521.6 mg) was chromatographed on a silica gel column (33 1, SF7402-5.2.10) was obtained.
  • RP reversed-phase
  • Tissue culture reagents such as Roswell Park Memorial Institute 1640 (RPMI1640) and fetal bovine serum, were purchased from Gibco BRL Co. (Grand Island, NY, USA). All chemicals were purchased from Sigma-Aldrich Chemical Co.
  • Major antibodies including anti-iNOS, anti-COX-2, anti-pI ⁇ B ⁇ , anti-actin, anti-p65 and anti-PCNA antibodies, were purchased from Santa Cruz Biotechnology (Santa Cruz, CA, USA). All primary antibodies were of rabbit origin.
  • Anti-rabbit secondary antibody was purchased from Millipore (Billerica, MA, USA).
  • Enzyme-linked immunosorbent assay (ELISA) kits for IL-6 and TNF- ⁇ were purchased from R&D Systems, Inc. (Minneapolis, MN, USA).
  • BV2 cells were cultured in RPMI1640 containing 1% antibiotic (penicillin-streptomycin) and 10% heat-inactivated FBS at 5 Mitochondrial reductase reduced the tetrazolium salt to 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide (MTT) into formazan crystals. This was used to measure the effect of compounds 1, 2, 3 and 4 on cell viability. To measure cell viability, 5 mg/mL MTT was treated with each cell suspension (1105 cells/mL) to form formazan for 4 hours. The formed formazan was dissolved in DMSO and the absorbance was measured at 540 nm.
  • MTT 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide
  • the concentration of nitrite in the conditioned medium was determined using a method based on the Griess reaction. After mixing equal amounts of cell culture medium and Griess reagent and reacting, the nitrite level was measured at 570 nm.
  • BV2 cells were seeded in a 24-well culture plate at a density of 2 ⁇ 10 5 cells/well. After incubation, the supernatant was recovered and used in a cytokine ELISA kit to measure the concentrations of IL-6 and TNF- ⁇ .
  • BV2 cells were seeded in a 24-well culture plate at a density of 2 ⁇ 10 5 cells/well. After incubation, the supernatant was collected. PGE 2 levels were then measured using a specific ELISA kit from R&D Systems, Inc. (Minneapolis, MN, USA).
  • Pelleted BV2 cells were washed with phosphate-buffered saline and dissolved in RIPA buffer.
  • Protein assay dye reagent concentrate obtained from Bio-Rad Laboratories (#5000006; Hercules, CA, USA) was mixed in sample loading buffer to quantify equal amounts of protein and separated by SDS-PAGE. The separated proteins were transferred to a nitrocellulose membrane. Non-specific binding to the membrane was blocked by incubation in a solution of skim milk. The membrane was incubated with primary antibody overnight at 4°C and then reacted with horseradish peroxidase-labeled secondary antibody (Millipore).
  • BV2 cells were seeded in 24-well culture plates at a density of 40 and 2. Cytoplasmic and nuclear fractions were separated using a nuclear extraction kit (Cayman, Ann Arbor, MI, USA). Each extracted fraction was dissolved according to the protocol provided by the manufacturer. The expression of the NF- ⁇ B (p65) pathway was then determined by Western blot analysis. Equivalent amounts of protein were quantified with protein assay dye reagent concentrate obtained from Bio-Rad Laboratories (#5000006; Hercules, CA, USA), mixed with sample loading buffer, and separated by SDS-PAGE. The separated proteins were then transferred to a nitrocellulose membrane.
  • Non-specific binding to the membrane was blocked by incubation in a solution of skim milk.
  • Membranes from cytoplasmic fraction samples were incubated with pI ⁇ B ⁇ antibody overnight at 4°C, whereas membranes from nuclear fraction samples were incubated with p65 antibody and then reacted with horseradish peroxidase-labeled secondary antibody (Millipore).
  • the survival rate of BV2 cells was measured after treatment for 24 hours at concentrations ranging from 10 to 40 ⁇ M using the MTT assay. As shown in Figure 2A, all four compounds (1-4) showed cytotoxicity to BV2 cells at concentrations of 20 and 40 ⁇ M. Therefore, additional experiments were performed using each compound in the non-toxic concentration range (2-10 ⁇ M).
  • NO a neurotransmitter second messenger molecule, mediates a variety of physiological and pathological processes in many organ systems, including the brain.
  • cell culture medium was collected and nitrite concentration was measured using the Griess reaction.
  • Example 3 Sterigmatocystin inhibits the production of LPS-induced inflammatory cytokines in BV2 cells.
  • Compound 1 reduced the expression of intracellular TNF- ⁇ and IL-6 by approximately 25% and 20%, respectively, at a concentration of 10 ⁇ M.
  • Compound 2 showed a strong effect, inhibiting TNF- ⁇ expression by more than 30% in activated BV2 cells at a concentration of 10 ⁇ M and inhibiting IL-6 expression by more than 30% at all concentrations tested (2, 5, and 10 M).
  • pretreatment with sterigmatocystin (1 and 2) could not eliminate PGE2 production in LPS-stimulated BV2 cells, which is consistent with the same effect of sterigmatocystin (1 and 2) on the expression of iNOS and COX-2. reflects.
  • Example 4 Sterigmatocystin inhibits LPS-induced activation of the NF-kB pathway in BV2 cells
  • BV2 cells were pretreated with the indicated concentrations of compounds 1 and 2 for 3 hours and then stimulated with LPS (1 ⁇ g/mL) for 1 hour. Cytoplasmic and nuclear fractions were extracted from cell lysates, and the protein level of p65 was increased in the nuclear fraction of the LPS treatment group. Pretreatment with compounds 1 and 2 reduced p65 expression in a concentration-dependent manner (Figure 5).
  • sterigmatocystin or a derivative thereof, a xanthone compound exhibits excellent anti-neuroinflammatory effects by inhibiting the production of NO, TNF- ⁇ , and IL-6 and the expression of iNOS protein. It was confirmed that this was mediated through activation of the NF- ⁇ B pathway in LPS-stimulated BV2 microglial cells. Through this, it can be effectively used in the prevention or treatment of neurodegenerative diseases, and provides a promising basis for the development of therapeutic agents for the treatment of neurodegenerative diseases.

Abstract

The present invention relates to a composition for preventing or treating neurodegenerative disease, the composition comprising a compound isolated from the Antarctic fungus Aspergillus sp. SF-7402 and, particularly, a composition for preventing or treating neurodegenerative disease, the composition comprising sterigmatocystin which is a xanthone compound, or a derivative thereof.

Description

ASPERGILLUS SP. SF-7402 유래 화합물을 포함하는 신경퇴행성 질환의 예방 또는 치료용 조성물 ASPERGILLUS SP. Composition for preventing or treating neurodegenerative diseases containing a compound derived from SF-7402
본 발명은 남극 진균 Aspergillus sp. SF-7402에서 분리된 화합물을 포함하는 신경퇴행성 질환(neurodegenerative disease)의 예방 또는 치료용 조성물로, 구체적으로 크산톤(xanthone) 화합물인 스테리그마토시스틴 (sterigmatocystin) 또는 이의 유도체를 포함하는 신경퇴행성 질환의 예방 또는 치료용 조성물에 관한 것이다.The present invention relates to the Antarctic fungus Aspergillus sp. A composition for preventing or treating neurodegenerative diseases containing a compound isolated from SF-7402, specifically, neurodegenerative diseases containing sterigmatocystin, a xanthone compound, or a derivative thereof. It relates to a composition for the prevention or treatment of.
뇌 또는 척수 내의 염증 반응인 신경염증은 사이토카인, 케모카인, 활성산소종 및 2차 메신저의 생산에 의해 매개된다. 중추신경계 (CNS)에서 이러한 염증성 중재자의 역할은 파킨슨병 (Parkinson’s disease: PD), 헌팅턴병 (Huntington’s disease: HD), 알츠하이머병 (Alzheimer’s disease: AD) 등 다양한 신경퇴행성질환에서 조사되었다. 미세아교세포 (microglia)는 특히 병원체의 침입과 조직 손상에 반응하여 주변 성상세포와 뉴런에 영향을 미치는 인자를 생산함으로써 CNS의 면역감시에 적극적인 역할을 한다. 신속한 미세아교세포의 활성화는 상처 치유를 시작하고 뉴런을 추가 손상으로부터 보호하기 위해 손상에 대한 생체 조직의 반응을 반영한다. 그러므로 활성화된 미세아교세포는 신경보호 작용을 하는 것으로 알려져 있다. 게다가, 일단 활성화되면, 염증성 미세아교세포는 일산화질소(NO), 유도형 일산화질소 신타아제(iNOS: inducible nitric oxide synthase), 시클로옥시게나제(COX: cyclooxygenase)-2를 포함하는 염증-유발성 중재자를 증가된 수준으로 분비하고, 활성화된 B 세포(NF-κB) 경로의 핵인자-κ 경쇄 인핸서의 활성화와 연관되어 있는 TNF-α 및 IL-6과 같은 전염증성 사이토카인을 증가된 수준으로 분비한다. 다양한 연구를 통해 NF-κB의 활성화가 산화 스트레스, 흥분 독성, 아밀로이드β 펩타이드 독성와 같은 다양한 손상으로부터 뉴런을 보호하는 것으로 나타났다. NF-κB는 또한 혈액 뇌 장벽 수준에서 세포 투과성, 엔도사이토시스 및 세포 내 수송에 영향을 미치는 주요 신호 전달 물질로도 입증되었다. 따라서 NF-κB는 신경염증 관련 질병 발생원인의 조절에 중요한 역할을 한다 (Shih, R.H.; Wang, C.Y.; Yang, C.M. NF-kappaB signaling pathways in neurological inflammation: A mini review. Front. Mol. Neurosci. 2015, 8, 77.)Neuroinflammation, an inflammatory response within the brain or spinal cord, is mediated by the production of cytokines, chemokines, reactive oxygen species, and second messengers. The role of these inflammatory mediators in the central nervous system (CNS) has been investigated in various neurodegenerative diseases, including Parkinson’s disease (PD), Huntington’s disease (HD), and Alzheimer’s disease (AD). Microglia play an active role in immune surveillance of the CNS by producing factors that affect surrounding astrocytes and neurons, especially in response to pathogen invasion and tissue damage. Rapid microglial activation reflects the tissue's response to injury to initiate wound healing and protect neurons from further damage. Therefore, activated microglia are known to have a neuroprotective effect. Furthermore, once activated, inflammatory microglia produce pro-inflammatory molecules, including nitric oxide (NO), inducible nitric oxide synthase (iNOS), and cyclooxygenase (COX)-2. Secretes increased levels of mediators and pro-inflammatory cytokines such as TNF-α and IL-6, which are associated with activation of the nuclear factor-κ light chain enhancer of the activated B cell (NF-κB) pathway. Secrete. Various studies have shown that activation of NF-κB protects neurons from various damages such as oxidative stress, excitotoxicity, and amyloid-β peptide toxicity. NF-κB has also been demonstrated to be a key signaling agent affecting cell permeability, endocytosis, and intracellular transport at the blood-brain barrier level. Therefore, NF-κB plays an important role in regulating the causes of diseases related to neuroinflammation (Shih, R.H.; Wang, C.Y.; Yang, C.M. NF-kappaB signaling pathways in neurological inflammation: A mini review. Front. Mol. Neurosci. 2015 , 8, 77.)
새로운 생물활성 천연물을 탐구하기 위해 연구자들은 최근 몇 년 동안 다양한 환경에서 새로운 공급원을 발견하기 위해 많은 노력을 기울였다. 박테리아, 방선균, 진균 등 다양한 동물군과 미생물이 서식하는 극지는 항균, 항종양, 항바이러스, 항염증 작용이 있는 다양한 2차 대사산물의 풍부한 공급원으로 주목받고 있다. 강한 태양 방사선을 동반한 추운, 건조한 기후를 가진 남극 대륙은 주목받고 있는 많은 독특한 미생물 자원을 키웠다.To explore new bioactive natural products, researchers have made great efforts in recent years to discover new sources in various environments. The polar regions, where various animal groups and microorganisms such as bacteria, actinomycetes, and fungi live, are attracting attention as a rich source of various secondary metabolites with antibacterial, antitumor, antiviral, and anti-inflammatory effects. Antarctica, with its cold, dry climate accompanied by strong solar radiation, has fostered many unique microbial resources that are attracting attention.
남극의 미생물, 특히 진균은 새로운 2차 대사산물의 풍부한 공급원임이 입증되었다. 남극균 유래의 대사산물은 강력한 항염증 효과를 나타내는 폴리케타이드 (polyketide), 펩티드, 알칼로이드, 테르페노이드로 분류되고, 항암, 항균 및 단백질 티로신 포스파타제 1B (PTP1B) 억제 활성 등을 나타낸다.Antarctic microorganisms, especially fungi, have proven to be a rich source of new secondary metabolites. Metabolites derived from Antarctic bacteria are classified into polyketides, peptides, alkaloids, and terpenoids, which exhibit strong anti-inflammatory effects, and exhibit anticancer, antibacterial, and protein tyrosine phosphatase 1B (PTP1B) inhibitory activities.
이러한 기술적 배경하에서, 본 출원의 발명자들은 남극 진균 Aspergillus sp. SF-7402의 대사산물 중 화합물 예를 들어, 스테리그마토시스틴 또는 이의 유도체를 분리하고, 이러한 화합물이 신경퇴행성 질환의 예방 또는 치료에 사용될 수 있음을 확인하고, 본 발명을 완성하였다.Under this technical background, the inventors of the present application have developed the Antarctic fungus Aspergillus sp. Among the metabolites of SF-7402, compounds such as sterigmatocystin or derivatives thereof were isolated, and it was confirmed that these compounds could be used for the prevention or treatment of neurodegenerative diseases, and the present invention was completed.
발명의 요약Summary of the Invention
본 발명의 목적은 크산톤(xanthone) 화합물을 포함하는 신경퇴행성 질환 (neurodegenerative disease)의 예방 또는 치료용 조성물을 제공하는 데 있다.The purpose of the present invention is to provide a composition for preventing or treating neurodegenerative disease containing a xanthone compound.
본 발명의 목적은 크산톤(xanthone) 화합물을 포함하는 신경퇴행성 질환 (neurodegenerative disease)의 예방 또는 치료방법을 제공하는데 있다. The purpose of the present invention is to provide a method for preventing or treating neurodegenerative disease including a xanthone compound.
본 발명의 목적은 크산톤(xanthone) 화합물을 신경퇴행성 질환 (neurodegenerative disease)의 예방 또는 치료에 사용하기 위한 용도를 제공하는 데 있다.The purpose of the present invention is to provide a xanthone compound for use in the prevention or treatment of neurodegenerative disease.
본 발명의 목적은 상기 조성물을 제조하는 방법을 제공하는 데 있다.The object of the present invention is to provide a method for producing the composition.
상기 목적을 달성하기 위하여, 본 발명은 다음의 구조식 1로 표시되는 화합물을 유효성분으로 포함하는 신경퇴행성 질환 (neurodegenerative disease)의 예방 또는 치료용 조성물을 제공한다:In order to achieve the above object, the present invention provides a composition for preventing or treating neurodegenerative disease, comprising a compound represented by the following structural formula 1 as an active ingredient:
Figure PCTKR2023012186-appb-img-000001
Figure PCTKR2023012186-appb-img-000001
상기 구조식 1에서 R은 알콕시 또는 수소이다. In structural formula 1, R is alkoxy or hydrogen.
본 발명은 구조식 1로 표시되는 화합물을 환자에 투여하는 단계를 포함하는 신경퇴행성 질환의 예방 또는 치료방법을 제공한다.The present invention provides a method for preventing or treating neurodegenerative diseases, comprising administering a compound represented by structural formula 1 to a patient.
본 발명은 구조식 1로 표시되는 화합물을 신경퇴행성 질환의 예방 또는 치료용 조성물 제조에 사용하기 위한 용도를 제공한다.The present invention provides the use of the compound represented by Structural Formula 1 in the production of a composition for preventing or treating neurodegenerative diseases.
본 발명은 진균 Aspergillus sp. SF-7402를 배양하여 대사물질을 추출하는 단계; 및 대사물질 중 크산톤(xanthone) 화합물을 분리하는 단계를 포함하는, 상기 조성물을 제조하는 방법을 제공한다. The present invention relates to the fungus Aspergillus sp. Culturing SF-7402 to extract metabolites; and separating xanthone compounds from metabolites.
도 1은 분리된 대사산물의 구조를 나타낸 것이다. Figure 1 shows the structure of the isolated metabolite.
도 2는 세포 생존율(A)과 아질산염 함량(B)에 대한 화합물의 영향을 확인한 결과를 나타낸 것이다. BV2 세포를 다양한 농도의 화합물 1-4와 함께 24시간 동안 배양하였다. 세포 생존율은 3-(4,5-디메틸티아졸-2-일)-2,5-디페닐테트라졸륨 브로마이드 검정을 사용하여 결정하였다. BV2 세포는 표시된 농도의 화합물로 3시간 동안 전처리되었고, 리포폴리사카라이드(LPS) (0.5μg/mL)로 24시간 동안 자극되었다. 막대는 세 번의 독립적인 실험의 평균 표준 편차를 나타낸다. ***LPS 처리군에 비해 p<0.001.Figure 2 shows the results of confirming the effect of compounds on cell viability (A) and nitrite content (B). BV2 cells were incubated with various concentrations of compounds 1-4 for 24 hours. Cell viability was determined using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. BV2 cells were pretreated with the indicated concentrations of compounds for 3 h and stimulated with lipopolysaccharide (LPS) (0.5 μg/mL) for 24 h. Bars represent the mean standard deviation of three independent experiments. ***p<0.001 compared to LPS treatment group.
도 3은 리포폴리사카라이드(LPS)로 자극된 BV2 세포에서 유도된 일산화질소 신타아제(iNOS: inducible nitric oxide synthase)(A,C) 및 시클로옥시게나제-2(COX-2: cyclooxygenase-2)(B,D)의 단백질 발현 수준을 확인한 결과를 나타낸 것이다. 세포를 표시된 농도의 화합물 1 및 2로 3시간 동안 전처리하고, LPS(0.5 g/mL)로 24시간 동안 자극하였다. 세 개의 독립적인 실험에서 대표적인 블롯을 표시하였다. 면역블롯은 ImageJ 소프트웨어를 사용하여 정량화하였다. 밴드 강도는 β-액틴으로 정규화되었다. *p<0.05, **LPS 처리군에 비해 p<0.01.Figure 3 shows inducible nitric oxide synthase (iNOS) (A, C) and cyclooxygenase-2 (COX-2) induced in BV2 cells stimulated with lipopolysaccharide (LPS). )(B,D) shows the results of confirming the protein expression level. Cells were pretreated with the indicated concentrations of compounds 1 and 2 for 3 h and stimulated with LPS (0.5 g/mL) for 24 h. Representative blots from three independent experiments are shown. Immunoblots were quantified using ImageJ software. Band intensity was normalized to β-actin. *p<0.05, **p<0.01 compared to LPS treatment group.
도 4는 LPS로 자극된 BV2 세포에서 TNF-α (A), IL-6 (B), PGE2 (C)에 분리된 화합물의 효과를 나타낸 것이다. 세포를 지시된 농도의 화합물 1-4로 3시간 동안 전처리하고, LPS(0.5μg/mL)로 24시간 동안 자극하였다. 막대는 세 번의 독립적인 실험의 평균 표준 편차를 나타낸다. * LPS 처리군에 비해 p<0.05, **p<0.01, ***p<0.001.Figure 4 shows the effects of the isolated compounds on TNF-α (A), IL-6 (B), and PGE2 (C) in BV2 cells stimulated with LPS. Cells were pretreated with the indicated concentrations of compounds 1-4 for 3 h and stimulated with LPS (0.5 μg/mL) for 24 h. Bars represent the mean standard deviation of three independent experiments. *p<0.05, **p<0.01, ***p<0.001 compared to LPS treatment group.
도 5는 분리된 화합물이 BV2 세포 중 NF-κB (p65) 경로에 미치는 영향을 나타낸 것이다 (A-D). 세포를 지시된 농도의 화합물 1 및 2로 3시간 동안 전처리하고, 리포폴리사카라이드(LPS, 1 g/mL)로 1시간 동안 자극하였다. 세 개의 독립적인 실험에서 대표적인 블롯이 표시된다. 면역블롯은 ImageJ 소프트웨어를 사용하여 정량화하였다. IκB-α 강도는 β-액틴으로 정규화되었다. P65 강도는 PCNA로 정규화되었다. * LPS 처리군에 비해 p<0.05, **p<0.01, ***p<0.001.Figure 5 shows the effects of isolated compounds on the NF-κB (p65) pathway in BV2 cells (A-D). Cells were pretreated with the indicated concentrations of compounds 1 and 2 for 3 hours and stimulated with lipopolysaccharide (LPS, 1 g/mL) for 1 hour. Representative blots from three independent experiments are shown. Immunoblots were quantified using ImageJ software. IκB-α intensity was normalized to β-actin. P65 intensity was normalized to PCNA. *p<0.05, **p<0.01, ***p<0.001 compared to LPS treatment group.
발명을 실시하기 위한 구체적인 내용Specific details for carrying out the invention
다른 식으로 정의되지 않는 한, 본 명세서에서 사용된 모든 기술적 및 과학적 용어들은 본 발명이 속하는 기술분야에서 숙련된 전문가에 의해서 통상적으로 이해되는 것과 동일한 의미를 갖는다. 일반적으로, 본 명세서에서 사용된 명명법은 본 기술분야에서 잘 알려져 있고 통상적으로 사용되는 것이다.Unless otherwise defined, all technical and scientific terms used in this specification have the same meaning as commonly understood by a person skilled in the art to which the present invention pertains. In general, the nomenclature used herein is well known and commonly used in the art.
미세아교세포 (microglia)는 중추 신경계 (CNS: central nervous system)의 면역 방어와 조직 복구에 중요한 역할을 한다. 미세아교세포의 활성화와 이에 의한 신경 염증은 신경 퇴행성 질환의 병인에 중요한 역할을 한다. 최근 신경 퇴행성 질환에서 염증 완화 전략이 점점 더 주목을 받고 있다. 리포폴리사카라이드(LPS)로 자극된 BV2 세포에서 남극 진균 Aspergillus sp. SF-7402 유래 화합물의 항-신경염증 가능성을 발견하고 평가했다.Microglia play an important role in immune defense and tissue repair in the central nervous system (CNS). Activation of microglia and resulting neuroinflammation play an important role in the pathogenesis of neurodegenerative diseases. Recently, inflammation-alleviating strategies have received increasing attention in neurodegenerative diseases. In BV2 cells stimulated with lipopolysaccharide (LPS), the Antarctic fungus Aspergillus sp. The anti-neuroinflammatory potential of SF-7402 derived compounds was discovered and evaluated.
화학적 조사를 통해, 진균으로부터 4개의 대사물, 즉 5-메톡시스테리그마토시스틴(5-methoxysterigmatocystin), 스테리그마토시스틴(sterigmatocystin), 아베르신(aversin), 및 6,8-O-디메틸베르시콜로린 A(6,8-O-dimethylversicolorin A)가 분리되었다. 이들의 화학구조는 광범위한 분광분석과 HR-ESI-MS 및 문헌에서 보고된 것과의 비교에 의해 밝혀졌다. 분리된 대사산물의 항-신경 염증 효과는 LPS 활성화 microglia에서 비세포 독성 농도로 일산화질소 (NO), TNF -α 및 IL-6의 생산을 측정함으로써 평가되었다. 5-메톡시스테리그마토시스틴(5-methoxysterigmatocystin), 스테리그마토시스틴(sterigmatocystin)은 NO 생산에 유의한 효과를 나타내었고, TNF -α 및 IL-6 발현 억제에 중등도의 효과를 보였다. 근본적인 분자 메커니즘은 웨스턴 블롯 분석을 사용하여 조사되었다. 5-메톡시스테리그마토시스틴(5-methoxysterigmatocystin), 스테리그마토시스틴(sterigmatocystin) 처리는 LPS 자극 BV2 세포에서 유도된 일산화질소 합성효소(iNOS) 발현의 하향조절을 통해 NO 생산을 억제하였다. 또한, 5-메톡시스테리그마토시스틴(5-methoxysterigmatocystin), 스테리그마토시스틴(sterigmatocystin)은 NF-κB의 핵전이를 감소시켰다. 이러한 결과는 진균 균주 Aspergillus sp.에 존재하는 스테리그마토시스틴이 신경 염증성 질환의 치료를 위한 유망한 후보임을 제시한다.Through chemical investigation, four metabolites were identified from the fungus, namely 5-methoxysterigmatocystin, sterigmatocystin, aversin, and 6,8-O-dimethylver. Cicolorin A (6,8-O-dimethylversicolorin A) was isolated. Their chemical structures were revealed by extensive spectroscopic analysis, HR-ESI-MS, and comparison with those reported in the literature. The anti-neuroinflammatory effects of the isolated metabolites were assessed by measuring the production of nitric oxide (NO), TNF-α, and IL-6 at non-cytotoxic concentrations in LPS-activated microglia. 5-methoxysterigmatocystin and sterigmatocystin showed a significant effect on NO production and a moderate effect on suppressing TNF-α and IL-6 expression. The underlying molecular mechanisms were investigated using Western blot analysis. Treatment with 5-methoxysterigmatocystin and sterigmatocystin inhibited NO production through downregulation of nitric oxide synthase (iNOS) expression induced in LPS-stimulated BV2 cells. Additionally, 5-methoxysterigmatocystin and sterigmatocystin reduced the nuclear translocation of NF-κB. These results suggest that sterigmatocystin present in the fungal strain Aspergillus sp. is a promising candidate for the treatment of neuroinflammatory diseases.
이를 바탕으로, 본 발명은 다음의 구조식 1로 표시되는 크산톤(xanthone) 화합물을 유효성분으로 포함하는 신경퇴행성 질환 (neurodegenerative disease)의 예방 또는 치료용 조성물에 관한 것이다:Based on this, the present invention relates to a composition for preventing or treating neurodegenerative disease, which contains a xanthone compound represented by the following structural formula 1 as an active ingredient:
Figure PCTKR2023012186-appb-img-000002
Figure PCTKR2023012186-appb-img-000002
상기 구조식 1에서 R은 알콕시 또는 수소이다.In structural formula 1, R is alkoxy or hydrogen.
본 발명은 구조식 1로 표시되는 크산톤(xanthone) 화합물을 환자에 투여하는 단계를 포함하는 신경퇴행성 질환 (neurodegenerative disease)의 예방 또는 치료방법에 관한 것이다. The present invention relates to a method for preventing or treating neurodegenerative disease, comprising administering a xanthone compound represented by structural formula 1 to a patient.
본 발명은 구조식 1로 표시되는 크산톤(xanthone) 화합물을 신경퇴행성 질환의 예방 또는 치료용 조성물 제조에 사용하기 위한 용도에 관한 것이다.The present invention relates to the use of a xanthone compound represented by structural formula 1 in the production of a composition for preventing or treating neurodegenerative diseases.
신경염증 (Neuroinflammation)은 초기 보호 메커니즘이며 염증성 매개자가 급성 신경염증 중에 손상된 신경세포와 신경교세포를 회복시키는 역할을 한다. 그러나, 만성 신경염증은 더 많은 신경 손상과 궁극적인 열화 (degradation)를 일으키는 경향이 있다. 신경염증의 치료제를 찾는 것은 신경퇴행성 질환의 진행을 잠재적으로 감소시킬 수 있다.Neuroinflammation is an early protective mechanism, and inflammatory mediators play a role in restoring damaged neurons and glial cells during acute neuroinflammation. However, chronic neuroinflammation tends to cause more nerve damage and ultimate degradation. Finding treatments for neuroinflammation could potentially reduce the progression of neurodegenerative diseases.
남극의 미생물, 특히 진균은 새로운 2차 대사산물의 풍부한 공급원임이 입증되었다. 남극균 유래의 대사산물은 강력한 항염증 효과를 나타내는 폴리케타이드 (polyketide), 펩티드, 알칼로이드, 테르페노이드로 분류되고, 항암, 항균 및 단백질 티로신 포스파타제 1B (PTP1B) 억제 활성 등을 나타낸다. BV2 세포를 사용하여 Aspergillus sp. SF-7402 (KCTC 14992BP)에서 분리된 4개의 대사산물의 항신경염증 효과를 조사하였고, 신경염증의 치료를 위한 새로운 전략을 제공할 수 있다.Antarctic microorganisms, especially fungi, have proven to be a rich source of new secondary metabolites. Metabolites derived from Antarctic bacteria are classified into polyketides, peptides, alkaloids, and terpenoids, which exhibit strong anti-inflammatory effects, and exhibit anticancer, antibacterial, and protein tyrosine phosphatase 1B (PTP1B) inhibitory activities. Using BV2 cells, Aspergillus sp. The anti-neuroinflammatory effects of four metabolites isolated from SF-7402 (KCTC 14992BP) were investigated, and may provide a new strategy for the treatment of neuroinflammation.
남극균으로부터 분리된 생물활성 대사물질을 연구하기 위한 지속적인 시도에 의해 진균 균주 Aspergillus sp. SF-7402로부터 4개의 2차 대사산물(1-4)이 분리되었다. 분리된 대사물은 5-메톡시스테리그마토시스틴(5-methoxysterigmatocystin: 1), 스테리그마토시스틴(sterigmatocystin: 2), 아베르신(aversin: 3), 및 6,8-O-디메틸베르시콜로린 A(6,8-O-dimethylversicolorin A: 4)로 확인되었다. 이전 연구에서, 5-메톡시스테리그마토시스틴은 코발트 프로토포르피린(CoPP: cobalt protoporphyrin) 발현에 의해 유도된 HO (heme oxygenase)-1의 발현에 영향을 주지 않았다. 그러나, LPS로 자극된 BV2 세포에서 5-메톡시스테리그마토시스틴의 치료 메커니즘에 대한 명확한 연구는 없었다. In ongoing attempts to study bioactive metabolites isolated from Antarctic fungi, the fungal strain Aspergillus sp. Four secondary metabolites (1-4) were isolated from SF-7402. The isolated metabolites were 5-methoxysterigmatocystin (1), sterigmatocystin (2), aversin (3), and 6,8-O-dimethylversicolo. It was identified as Lin A (6,8-O-dimethylversicolorin A: 4). In a previous study, 5-methoxysterigmatocystin did not affect the expression of heme oxygenase (HO)-1 induced by cobalt protoporphyrin (CoPP) expression. However, there has been no clear study on the therapeutic mechanism of 5-methoxysterigmatocystin in BV2 cells stimulated with LPS.
스테리그마토시스틴(sterigmatocystin: 2), 아베르신(aversin: 3), 및 6,8-O-디메틸베르시콜로린 A(6,8-O-dimethylversicolorin A: 4)의 항신경염증 효과는 보고되지 않았다. 여기에서는 분리된 대사산물 1-4의 구조적 특성 및 LPS로 자극된 BV2 미세아교세포의 항신경염증성 활성에 대해 설명한다.The anti-neuroinflammatory effects of sterigmatocystin (2), aversin (3), and 6,8-O-dimethylversicolorin A (6,8-O-dimethylversicolorin A: 4) have been reported. It didn't work. Here, we describe the structural properties of the isolated metabolites 1–4 and their anti-neuroinflammatory activity in LPS-stimulated BV2 microglia.
일산화질소는 3개의 일산화질소 합성효소(NOS) 아이소폼에 의해 아르기닌으로부터 효소적으로 형성된다. NOS 패밀리의 이소형인 유도성 NOS(iNOS)는 LPS 및 다양한 사이토카인에 의해 유도될 수 있다. iNOS는 미세아교세포를 포함한 많은 세포 유형에서 발현된다. 건강한 뇌에서, 미세아교세포는 iNOS를 발현하지 않는다. 그럼에도 불구하고, 허혈성, 외상성, 신경독성, 또는 염증성 손상 하에서, 이들은 활성화되어 iNOS를 생성하고, 대량의 NO를 방출한다. 그 후, 세포막 구조는 손상될 수 있고 DNA 전사와 단백질 합성에 영향을 미치며 뉴런에 직접 손상을 준다.Nitric oxide is formed enzymatically from arginine by three nitric oxide synthase (NOS) isoforms. Inducible NOS (iNOS), an isoform of the NOS family, can be induced by LPS and various cytokines. iNOS is expressed in many cell types, including microglia. In a healthy brain, microglia do not express iNOS. Nevertheless, under ischemic, traumatic, neurotoxic, or inflammatory injury, they are activated to produce iNOS and release large amounts of NO. Afterwards, cell membrane structures can be damaged, affecting DNA transcription and protein synthesis, and directly damaging neurons.
NO는 세포 내의 Fe2+ 농도를 변화시킴으로써 신경퇴행을 일으킨다. 따라서, 미세아교세포에서 iNOS의 넉다운은 NO의 생산을 억제하여 CNS 변성을 감소시킨다. 2개의 크산톤, 5-메톡시스테리그마토시스틴 및 스테리그마토시스틴이 NO 생산을 유의하게 억제하였음을 보여주었다(도 2B). 이어서, BV2 세포에서 iNOS 및 COX-2 단백질의 LPS 유도 발현에 대한 스테리그마토시스틴(1 및 2)의 효과를 웨스턴 블롯 분석에 의해 조사하였다. 두 스테리그마토시스틴 (1 및 2)은 LPS 유도 iNOS 발현을 억제했지만 COX-2 발현에는 영향을 미치지 않았다(도 3). COX-2 및 iNOS 발현의 신호전달 경로는 복잡하지만, 이러한 상이한 효과는 다양한 전사 인자에 대한 iNOS 및 COX-2 프로모터의 의존성에 기인할 수 있다. iNOS 프로모터는 NF-κB, AP-1, C/EBPβ, Stat 등의 여러 시스 작용 요소를 포함하는 반면, COX-2 프로모터는 NF-κB, C/EBPβ, CRE 시스 작용 요소를 포함한다. 프로모터 활성은 세포 유형과 적용된 자극에 따라 다르다.NO causes neurodegeneration by changing intracellular Fe2+ concentration. Therefore, knockdown of iNOS in microglia reduces CNS degeneration by suppressing NO production. Two xanthones, 5-methoxysterigmatocystin and sterigmatocystin, showed significant inhibition of NO production (Figure 2B). Subsequently, the effect of sterigmatocystin (1 and 2) on LPS-induced expression of iNOS and COX-2 proteins in BV2 cells was examined by Western blot analysis. Both sterigmatocystins (1 and 2) inhibited LPS-induced iNOS expression but had no effect on COX-2 expression ( Fig. 3 ). Although the signaling pathways of COX-2 and iNOS expression are complex, these different effects may be due to the dependence of the iNOS and COX-2 promoters on various transcription factors. The iNOS promoter contains several cis-acting elements, including NF-κB, AP-1, C/EBPβ, and Stat, while the COX-2 promoter contains NF-κB, C/EBPβ, and CRE cis-acting elements. Promoter activity varies depending on cell type and applied stimulus.
이러한 개념을 뒷받침하기 위해, NF-κB의 활성화에 결정적으로 의존하는 것으로 알려진 TNF-α 및 IL-6과 같은 다른 염증 유발 사이토카인의 발현은 화합물 1 및 2에 의해 유의하게 억제되었다 (도 4A, B). 활성화된 미세아교세포에서 방출된 면역 매개체 중 NO 및 TNF-α 및 IL-6을 포함한 전염증성 사이토카인은 신경염증의 발병에 중요한 중재자이다. 다수의 증거들이 TNF-α 및 IL-6과 같은 사이토카인이 미세아교세포의 활성화를 나타내는 지표가 될 수 있음을 보여준다. 뇌세포에서 방출되는 TNF-α는 병리학적 조건 하에서 신경염증 반응의 중심에 있다. IL-6은 염증성 상해에 대한 급성 신경염증 반응을 일으키는 요인 중 하나이다. COX-2 효소의 산물인 PGE2도 NF-κB에 의해 조절되어 신경염증에 중요한 역할을 한다. 그러나, 화합물 1 및 2는 PGE2 생산을 억제할 수 없었고, COX-2 발현에 대한 화합물 1 및 2의 동일한 효과를 반영한다 (도 4C). 이러한 결과는 LPS 자극 BV2 세포에서 화합물 1 및 2의 조절 효과가 iNOS 프로모터에 비해 COX-2 프로모터에 대한 의존성이 더 적음을 시사한다.In support of this notion, the expression of other proinflammatory cytokines, such as TNF-α and IL-6, which are known to be critically dependent on the activation of NF-κB, were significantly inhibited by compounds 1 and 2 (Figure 4A, B). Among the immune mediators released from activated microglia, NO and proinflammatory cytokines, including TNF-α and IL-6, are important mediators in the pathogenesis of neuroinflammation. A large body of evidence shows that cytokines such as TNF-α and IL-6 can be indicators of microglial activation. TNF-α released from brain cells is at the center of neuroinflammatory responses under pathological conditions. IL-6 is one of the factors responsible for the acute neuroinflammatory response to inflammatory injury. PGE 2, a product of COX-2 enzyme, is also regulated by NF-κB and plays an important role in neuroinflammation. However, compounds 1 and 2 were unable to inhibit PGE 2 production, reflecting the same effect of compounds 1 and 2 on COX-2 expression (Figure 4C). These results suggest that the regulatory effects of compounds 1 and 2 in LPS-stimulated BV2 cells are less dependent on the COX-2 promoter compared to the iNOS promoter.
이러한 결과를 바탕으로, 본 발명은 예를 들어 상기 구조식 1 중 R이 메톡시인 5-메톡시스테리그마토시스틴(5-methoxysterigmatocystin)을 포함하거나, R은 수소인 스테리그마토시스틴(sterigmatocystin)을 포함할 수 있다. Based on these results, the present invention includes, for example, 5-methoxysterigmatocystin in structural formula 1, where R is methoxy, or sterigmatocystin, where R is hydrogen. It can be included.
만성 신경염증은 주로 뇌의 미세아교세포와 면역세포에 의해 조절된다. 만성 신경염증이 PD 중 뉴런 손실의 주된 이유로 알려져 있고 신경퇴행의 기초를 형성한다. NF-κB는 전사 인자의 Rel 패밀리 멤버이다. RelA (p65)는 Rel 패밀리 다섯 멤버 중 하나이다. p65의 전사후 변형은 발병에 중요한 역할을 한다. p65의 전사후 변형은 허혈성 상해 및 글루탐산 또는 β-아밀로이드 독성에 의해 활성화되는 신경퇴행 과정의 개시에 중요한 역할을 한다. p65의 핵 지역화 신호와 상호작용하기 때문에 가장 중요한 IκB 단백질은 IκB-α로 고려된다. 화합물 1 및 2 전처리는 농도 의존적 방식으로 p65 발현을 감소시킨다 (도 5). 세포질 분획 중 IκB-α 인산화는 LPS 자극에 의해 증가되나, 화합물 1 및 2는 농도의존적 방식으로 반응을 억제시킨다 (도 5). 이러한 결과는 스테리그마토시스틴이 NF-κB 억제에 의해 항-신경염증성 효과를 나타냄을 확인할 수 있다. Chronic neuroinflammation is mainly regulated by brain microglia and immune cells. Chronic neuroinflammation is known to be the main reason for neuronal loss during PD and forms the basis of neurodegeneration. NF-κB is a member of the Rel family of transcription factors. RelA (p65) is one of the five members of the Rel family. Post-transcriptional modification of p65 plays an important role in pathogenesis. Post-transcriptional modification of p65 plays an important role in the initiation of neurodegenerative processes activated by ischemic injury and glutamate or β-amyloid toxicity. The most important IκB protein is considered to be IκB-α because it interacts with the nuclear localization signal of p65. Compound 1 and 2 pretreatment reduced p65 expression in a concentration-dependent manner (Figure 5). IκB-α phosphorylation in the cytoplasmic fraction was increased by LPS stimulation, but compounds 1 and 2 inhibited the response in a concentration-dependent manner (Figure 5). These results confirm that sterigmatocystin exhibits an anti-neuroinflammatory effect by inhibiting NF-κB.
스테리그마토시스틴은 크산톤 (xanthone)과 비스푸란기(bisfuran group)를 포함하는 크산톤이다. 스테리그마토시스틴 유도체는 항종양, 항염증, 항바이러스 및 항균 활성을 포함한 다수의 생물학적 활성을 갖는 것으로 보고되어 있다. 단리된 스테리그마토시스틴 중에서, C-5 위치에 메톡시기가 없는 것에 의해 화합물 1과 다른 화합물 2는 LPS로 자극된 BV2 세포 중 지정된 농도에서 p65 단백질의 발현 및 IκB-α 인산화에 더 큰 효과를 나타낸다 (도 5B). 이 점은 스테리그마토시스틴의 방향족 고리 그룹 중 메톡시의 중요한 역할을 보여준다.Sterigmatocystin is a xanthone containing a xanthone and a bisfuran group. Sterigmatocystin derivatives are reported to have numerous biological activities, including antitumor, anti-inflammatory, antiviral and antibacterial activities. Among the isolated sterigmatocystins, compound 2, which differs from compound 1 by the absence of a methoxy group at the C-5 position, has a greater effect on the expression of p65 protein and IκB-α phosphorylation at the indicated concentrations in LPS-stimulated BV2 cells. shown (Figure 5B). This shows the important role of methoxy among the aromatic ring groups of sterigmatocystin.
여기서, 상기 화합물들은 남극 진균 Aspergillus sp. SF-7402에서 분리된 것일 수 있다. 남극 균주 Aspergillus sp. SF-7402에 존재하는 2개의 크산톤, 스테리그마토시스틴(1, 2) 및 2개의 안트라퀴논(3, 4)이 확인되었다. 스테리그마토시스틴이 NO, TNF-α, IL-6의 생산 및 iNOS 단백질의 발현을 억제하여 우수한 항-신경염증 효과를 나타냄을 확인하였다. 이러한 효과는 LPS 자극 BV2 미세아교세포에서 NF-κB 경로의 활성화를 통해 매개되었다.Here, the compounds are derived from the Antarctic fungus Aspergillus sp. It may have been separated from SF-7402. The Antarctic strain Aspergillus sp. Two xanthones, sterigmatocystin (1, 2) and two anthraquinones (3, 4), present in SF-7402 were identified. It was confirmed that sterigmatocystin exhibits excellent anti-neuroinflammatory effects by inhibiting the production of NO, TNF-α, and IL-6 and the expression of iNOS protein. These effects were mediated through activation of the NF-κB pathway in LPS-stimulated BV2 microglia.
상기 신경퇴행성 질환은 예를 들어, 파킨슨 질환, 알츠하이머 질환 (AD), 뇌졸중, 치매, 근위축증 (MD), 다발성 경화증 (MS), 근위축성측색경화증 (ALS), 낭성 섬유증, 앙겔만(Angelman) 증후군, 리들(Liddle) 증후군, 픽 질환, 파젯 질환, 암 또는 외상 뇌 손상 등일 수 있으나, 이에 제한되는 것은 아니다.The neurodegenerative diseases include, for example, Parkinson's disease, Alzheimer's disease (AD), stroke, dementia, muscular dystrophy (MD), multiple sclerosis (MS), amyotrophic lateral sclerosis (ALS), cystic fibrosis, and Angelman syndrome. , Liddle syndrome, Pick disease, Paget disease, cancer, or traumatic brain injury, but are not limited thereto.
본 발명의 조성물에는 투여를 위하여 상기 기재된 유효성분 이외에 추가로 약학적으로 허용되는 가능한 담체를 1종 이상 포함하여 제조될 수 있다. 약제학적으로 허용 가능한 담체는 본 발명의 유효성분과 양립 가능하여야 하며, 식염수, 멸균 수, 링거액, 완충 식염수, 덱스트로즈 용액, 말토덱스트린 용액, 글리세롤, 에탄올 및 이들 성분 중 한 성분 또는 둘 이상의 성분을 혼합하여 사용할 수 있고, 필요에 따라 항산화제, 완충액, 정균제 등 다른 통상의 첨가제를 첨가할 수 있다. The composition of the present invention may be prepared to include one or more pharmaceutically acceptable carriers in addition to the active ingredients described above for administration. The pharmaceutically acceptable carrier must be compatible with the active ingredient of the present invention, and may include saline solution, sterile water, Ringer's solution, buffered saline solution, dextrose solution, maltodextrin solution, glycerol, ethanol, and one or two or more of these ingredients. It can be used by mixing, and other common additives such as antioxidants, buffers, and bacteriostatic agents can be added as needed.
또한, 희석제, 분산제, 계면활성제, 결합제 및 윤활제를 부가적으로 첨가하여 수용액, 현탁 액, 유탁액 등과 같은 주사용 제형으로 제제화 할 수 있다. 특히, 동결건조(lyophilized)된 형태의 제형으로 제제화하여 제공하는 것이 바람직하다. 동결건조 제형 제조를 위해서 본 발명이 속하는 기술분야에서 통상적으로 알려져 있는 방 법이 사용될 수 있으며, 동결건조를 위한 안정화제가 추가될 수도 있다. 더 나아가 당 분야의 적정한 방법으로 또는 레밍톤 약학 과학(Remington's pharmaceutical Science, Mack Publishing company, Easton PA)에 개시되어 있는 방법을 이용하여 각 질병에 따라 또는 성분에 따라 바람직하게 제제화 할 수 있다.In addition, diluents, dispersants, surfactants, binders, and lubricants can be additionally added to formulate injectable formulations such as aqueous solutions, suspensions, emulsions, etc. In particular, it is preferable to formulate and provide it in a lyophilized form. To prepare a freeze-dried formulation, methods commonly known in the art to which the present invention pertains may be used, and a stabilizer for freeze-drying may be added. Furthermore, it can be preferably formulated according to each disease or ingredient using an appropriate method in the art or a method disclosed by Remington's Pharmaceutical Science (Mack Publishing company, Easton PA).
본 발명의 약학 조성물은 비경구 투여가 바람직하고, 투여량은 환자의 체중, 연령, 성별, 건강상태, 식이, 투여시간, 방법, 배설율 또는 질병의 중증도 등에 따 라 그 범위가 다양하며, 본 기술분야의 통상의 전문가가 용이하게 결정할 수 있다. The pharmaceutical composition of the present invention is preferably administered parenterally, and the dosage varies depending on the patient's weight, age, gender, health status, diet, administration time, method, excretion rate, or severity of the disease. An expert in the technical field can easily determine this.
본 발명에 따른 조성물의 1회 투여량은 1μg/kg 내지 100 ㎎/㎏일 수 있으며, 바람직하게는 5μg/kg 내지 50 ㎎/㎏이고, 일일 1회 또는 주 1-3회 투여하는 것을 특징으로 할 수 있으나, 투여량과 투여간격이 이에 한정되는 것은 아니다.A single dosage of the composition according to the present invention may be 1 μg/kg to 100 mg/kg, preferably 5 μg/kg to 50 mg/kg, and is administered once a day or 1-3 times a week. However, the dosage and administration interval are not limited to this.
본 발명에 따른 동물에게 투여하는 것을 포함하는, 신경퇴행성 질환의 예방 또는 치료방법에 관한 것이다. It relates to a method for preventing or treating neurodegenerative diseases, including administration to animals according to the present invention.
상기 동물은 인간, 원숭이 등의 영장류 개, 돼지, 소, 양, 염소, 마우스,래트일 수 있으며, 이에 제한되는 것은 아니다. 상기 동물은 예를 들어 인간일 수 있다. The animal may be a human, a primate such as a monkey, a dog, a pig, a cow, a sheep, a goat, a mouse, or a rat, but is not limited thereto. The animal may be, for example, a human.
상기 조성물의 투여방법은 통상의 환자의 증후와 질병의 심각도에 기초하여 본 기술분야의 통상의 전문가가 결정할 수 있다. The method of administering the composition can be determined by an expert in the art based on the patient's symptoms and the severity of the disease.
본 발명의 약학 조성물은 비경구 투여가 가능하다. 본 발명에 따른 조성물의 투여경로는 이들로 한정되는 것은 아니지만, 예를 들면, 구강, 정맥 내, 근육 내, 동맥 내, 골수 내, 경막 내, 심장 내, 경피, 피하, 복강 내, 장관, 설하 또는 국소 투여가 가능하다. 본 발명의 약학 조성물은 CNS (central nervous system)에 투여될 수 있다. 본 발명의 약학 조성물은 수막공간내 주사 (intrathecal injection)에 의해 투여될 수 있다.The pharmaceutical composition of the present invention can be administered parenterally. The route of administration of the composition according to the present invention is not limited to these, but includes, for example, oral cavity, intravenous, intramuscular, intraarterial, intramedullary, intrathecal, intracardiac, transdermal, subcutaneous, intraperitoneal, intestinal, sublingual. Alternatively, topical administration is possible. The pharmaceutical composition of the present invention can be administered to the central nervous system (CNS). The pharmaceutical composition of the present invention can be administered by intrathecal injection.
본 발명에 따른 조성물의 투여량은 환자의 체중, 연령, 성별, 건강상태, 식이, 투여시간, 방법, 배설율 또는 질병의 중증도 등에 따라 그 범위가 다양하며, 본 기술분야의 통상의 전문가가 용이하게 결정할 수 있다. 또한, 임상 투여를 위해 공지의 기술을 이용하여 본 발명의 조성물을 적합한 제형으로 제제화할 수 있다.The dosage of the composition according to the present invention varies depending on the patient's weight, age, gender, health condition, diet, administration time, method, excretion rate, or severity of the disease, and is easily understood by an expert in the art. You can decide. Additionally, the composition of the present invention can be formulated into a suitable dosage form for clinical administration using known techniques.
"용량"은 단일 투여에서, 또는 명시된 기간 동안에 제공된 약제학적 제제의 명시된 양을 의미한다. 용량은 하나, 둘 또는 그 이상의 볼러스 (boluses), 정제, 또는 주사제로 투여될 수 있다. 예를 들어, 피하 투여를 원하는 특정 구현예에서, 원하는 용량은 단일 주사에 의해서 쉽게 수용되지 않는 용적을 필요로 하며, 두 개 또는 그 이상의 주사제를 사용하여 원하는 용량을 달성할 수 있다. 약제는 장시간에 걸쳐 또는 연속적으로 주입에 의해 투여된다. 용량은 시간, 일, 주, 또는 개월 당 약제학적 제제의 양으로 언급될 수 있다.“Dose” means a specified amount of pharmaceutical agent given in a single administration or over a specified period of time. Doses may be administered as one, two or more boluses, tablets, or injections. For example, in certain embodiments where subcutaneous administration is desired, the desired dose requires a volume that is not readily accommodated by a single injection, and two or more injections may be used to achieve the desired dose. Medications are administered by infusion over a long period of time or continuously. Dosage may be stated as the amount of pharmaceutical agent per hour, day, week, or month.
실시예Example
이하, 실시예를 통하여 본 발명을 더욱 상세히 설명하고자 한다. 이들 실시예는 오로지 본 발명을 예시하기 위한 것으로서, 본 발명의 범위가 이들 실시예에 의해 제한되는 것으로 해석되지는 않는 것은 당업계에서 통상의 지식을 가진 자에게 있어서 자명할 것이다.Hereinafter, the present invention will be described in more detail through examples. These examples are only for illustrating the present invention, and it will be apparent to those skilled in the art that the scope of the present invention is not to be construed as limited by these examples.
재료 및 방법Materials and Methods
1. 일반적 실험 방법1. General experimental method
Jasco P-2000 디지털 선광계(JASCO Corp., Tokyo, Japan)를 사용하여 선광도를 기록했다. HR-ESI-MS 데이터는 ESI (electrospray ionization) 4중극 비행 시간형(Q-TOF) 탠덤 질량 분석(MS/MS) 시스템(ABSCRIEX Triple)을 사용하여 획득하였다. 핵자기 공명(NMR) 스펙트럼(1D 및 2D)은 JEOL JNM ECP-400 분광계(JEOL Ltd., Tokyo, Japan)를 사용하여 CDCl3(δH/δC =7.26/77.16)로 기록되고 화학적 이동은 잔류 용매의 피크를 기준으로 참조하였다. 이핵 다중중량 코히어런스(HMQC: Heteronuclear multiple quantum coherence) 및 이핵 다중 결합 상관(HMBC: heteronuclear multiple bond correlation) 실험은 1JCH=140Hz 및 nJCH=8Hz에 대하여 최적화되었다. 박층 크로마토그래피(TLC: Thin-layer chromatography)는 Kieselgel 60 F254(1.05715; Merck, Darmstadt, Germany) 또는 RP-18 F254s(Merck, Darmstadt, Germany) 플레이트에서 수행하였다. 스팟은 플레이트에 10% 황산 수용액 (H2SO4)을 분무한 다음 가열하여 시각화하였다. 컬럼 크로마토그래피는 실리카겔(Kieselgel 60, 70-230 메쉬 및 230-400 메쉬 Merck, Darmstadt, Germany) 및 YMC 옥타데실 기능기화 실리카겔 (C18)로 수행하였다. 고속 액체 크로마토그래피(HPLC)는 유속 5 mL/min로 분취 C18 컬럼(10 X 250 mm, 입자 직경 5μm)으로 실시하였rh, 화합물은 210 및 254 nm에서의 자외선(UV) 흡수에 의해 검출되었다.Optical rotation was recorded using a Jasco P-2000 digital polarimeter (JASCO Corp., Tokyo, Japan). HR-ESI-MS data were acquired using an electrospray ionization (ESI) quadrupole time-of-flight (Q-TOF) tandem mass spectrometry (MS/MS) system (ABSCRIEX Triple). Nuclear magnetic resonance (NMR) spectra (1D and 2D) were recorded in CDCl3 (δH/δC =7.26/77.16) using a JEOL JNM ECP-400 spectrometer (JEOL Ltd., Tokyo, Japan), and chemical shifts were recorded as those of residual solvent. Reference was made to the peak. Heteronuclear multiple quantum coherence (HMQC) and heteronuclear multiple bond correlation (HMBC) experiments were optimized for 1 JCH=140Hz and n JCH=8Hz. Thin-layer chromatography (TLC) was performed on Kieselgel 60 F 254 (1.05715; Merck, Darmstadt, Germany) or RP-18 F 254s (Merck, Darmstadt, Germany) plates. The spots were visualized by spraying a 10% aqueous solution of sulfuric acid (H 2 SO 4 ) on the plate and then heating it. Column chromatography was performed with silica gel (Kieselgel 60, 70-230 mesh and 230-400 mesh Merck, Darmstadt, Germany) and YMC octadecyl functionalized silica gel (C 18 ). High-performance liquid chromatography (HPLC) was performed on a preparative C 18 column (10 .
2. 진균 재료 및 배양2. Fungal material and culture
진균 균주 SF-7402는 2017년 1월 남극의 King George Island (62°1406.15’’S, 58°46’22.68’’W)에서 수집된 이끼류로부터 분리되었다. 샘플 1g은 모타 (mortar) 및 유봉으로 분쇄하고 멸균 해수(10 mL)와 혼합하였다. 샘플 일부 (0.1 mL)를 해수를 함유하는 감자 덱스트로스 아가 (potato dextrose agar : PDA) 배지 중에서 스프레드 플레이트 방법을 사용하여 처리하고, 25℃에서 14일 동안 인큐베이션하였다. 분리주를 여러 번 계대 배양 후 최종 순수 배양물을 선택하고, -70℃에 보관하였다. 진균 균주 SF-7402는 리보솜 RNA(rRNA) 서열 분석을 사용하여 확인되었다. SF-7402의 ITS 유전자를 이용한 GenBank 검색 (GenBank 수탁번호 MZ267531)을 통해 Aspergillus jensenii (NR_135444), A. creber (NR_135442), A. versicolor (NR_131277), 및 A. protuberus (NR_135353)을 각각 99.62%, 99.62%, 99.43%, 99.43%의 서열 상동성을 나타내는 가장 근접 매치로 선별하였다. 따라서, 진균 균주 SF-7402는 Aspergillus sp. A voucher specimen (SF-7402)으로 2022.06.08에 기탁하였다 (KCTC 14992BP). Fungal strain SF-7402 was isolated from lichen collected at King George Island, Antarctica (62°1406.15’’S, 58°46’22.68’’W) in January 2017. 1 g of sample was ground with a mortar and pestle and mixed with sterile seawater (10 mL). A portion of the sample (0.1 mL) was processed using the spread plate method in potato dextrose agar (PDA) medium containing seawater and incubated at 25°C for 14 days. After subculturing the isolate several times, the final pure culture was selected and stored at -70°C. Fungal strain SF-7402 was identified using ribosomal RNA (rRNA) sequencing. Through GenBank search using the ITS gene of SF-7402 (GenBank accession number MZ267531), Aspergillus jensenii (NR_135444), A. creber (NR_135442), A. versicolor (NR_131277), and A. protuberus (NR_135353) were identified at 99.62%, respectively. The closest matches showing sequence homology of 99.62%, 99.43%, and 99.43% were selected. Therefore, the fungal strain SF-7402 is Aspergillus sp. It was deposited on 2022.06.08 as A voucher specimen (SF-7402) (KCTC 14992BP).
3. 대사물질의 추출 및 분리3. Extraction and separation of metabolites
진균 균주 Aspergillus sp. SF-7402를 각각 3% NaCl(v/v)을 포함하는 650 mL의 PDA 배지를 포함하는 20개의 Fernbach 플라스크에서 배양하였다. 플라스크에 진균 균주의 2 mL 시드 배양물을 개별적으로 접종하고, 25℃에서 14일 동안 인큐베이션하였다. 발효 배양 배지를 합하고 EtOAc (20 L)로 추출하였다. 이어서, 합한 EtOAc 추출물을 여과지로 여과하고, 증발 건조시켜 조 추출물(4.8 g)을 얻었다. 조 추출물을 역상 (RP: reversed-phase) C18 플래시 컬럼 크로마토그래피 (30 X 4.5 cm)로 분획하고, 20%, 40%, 60%, 80% 및 100% (v/v) H2O (각 500 mL) 중 MeOH의 단계적 구배로 용출하여 SF-7402-1에서 SF-7402-5의 5 가지 분획을 얻었다. 분획 SF-7402-5(568.8 mg)를 CHCl3-MeOH(3:1, v/v)를 사용하여 SephadexLH-20 컬럼(33X3 cm)에 제공하여 4개의 분획(SF-7402-5.1에서 SF-7402-5.4)을 얻었다. 분획 SF-7402-5.2(521.6 mg)를 실리카겔 컬럼(33 X 2 cm)으로 크로마토그래피하고, n-헥산-아세톤(7:1, v/v)으로 용출하여 10개의 서브 분획(SF7402-5.2.1, SF7402-5.2.10)을 얻었다. 마지막으로, 서브 분획 SF-7402-5.2.7(153.9 mg)을 C18 prep HPLC(H2O 중 45-65% CH3CN, 45분 이상)를 사용하여 분리하고, 화합물 1(25.3 mg, tR=30분), 2 (15.8 mg, tR = 34분), 3 (9.5 mg, tR = 39분) 및 4 (1.1 mg, tR = 43분)를 얻었다.The fungal strain Aspergillus sp. SF-7402 was cultured in 20 Fernbach flasks each containing 650 mL of PDA medium containing 3% NaCl (v/v). Flasks were individually inoculated with 2 mL seed cultures of fungal strains and incubated at 25°C for 14 days. Fermentation culture media were combined and extracted with EtOAc (20 L). The combined EtOAc extracts were then filtered through filter paper and evaporated to dryness to obtain the crude extract (4.8 g). The crude extract was fractionated by reversed-phase (RP) C 18 flash column chromatography ( 30 Five fractions of SF-7402-1 to SF-7402-5 were obtained by eluting with a stepwise gradient of MeOH (500 mL each). Fraction SF-7402-5 (568.8 mg) was applied to a SephadexLH -20 column (33 7402-5.4) was obtained. Fraction SF-7402-5.2 (521.6 mg) was chromatographed on a silica gel column (33 1, SF7402-5.2.10) was obtained. Finally, subfraction SF-7402-5.2.7 (153.9 mg) was separated using C 18 prep HPLC (45-65% CH 3 CN in H 2 O, over 45 min) and compound 1 (25.3 mg, tR = 30 min), 2 (15.8 mg, tR = 34 min), 3 (9.5 mg, tR = 39 min) and 4 (1.1 mg, tR = 43 min).
Figure PCTKR2023012186-appb-img-000003
Figure PCTKR2023012186-appb-img-000003
Figure PCTKR2023012186-appb-img-000004
Figure PCTKR2023012186-appb-img-000004
Figure PCTKR2023012186-appb-img-000005
Figure PCTKR2023012186-appb-img-000005
Figure PCTKR2023012186-appb-img-000006
Figure PCTKR2023012186-appb-img-000006
4. 세포 배양 및 제제4. Cell culture and preparation
Roswell Park Memorial Institute 1640(RPMI1640) 및 소 태아 혈청과 같은 조직 배양 시약은 Gibco BRL Co.(Grand Island, NY, USA)에서 구입했다. 모든 화학물질은 Sigma-Aldrich Chemical Co에서 구입하였다. 주요 항체 예를 들어, 항-iNOS, 항-COX-2, 항-pIκBα, 항-액틴, 항-p65 및 항-PCNA 항체는 Santa Cruz Biotechnology (Santa Cruz, CA, USA)로부터 구입하였다. 1차 항체는 모두 토끼 유래였다. 항토끼 2차 항체는 Millipore(Billerica, MA, USA)로부터 구입하였다. IL-6 및 TNF-α를 위한 효소 결합 면역흡착 측정법(ELISA) 키트는 R&D Systems, Inc.(미네아폴리스, 미네소타주, 미국)로부터 구입하였다.Tissue culture reagents, such as Roswell Park Memorial Institute 1640 (RPMI1640) and fetal bovine serum, were purchased from Gibco BRL Co. (Grand Island, NY, USA). All chemicals were purchased from Sigma-Aldrich Chemical Co. Major antibodies, including anti-iNOS, anti-COX-2, anti-pIκBα, anti-actin, anti-p65 and anti-PCNA antibodies, were purchased from Santa Cruz Biotechnology (Santa Cruz, CA, USA). All primary antibodies were of rabbit origin. Anti-rabbit secondary antibody was purchased from Millipore (Billerica, MA, USA). Enzyme-linked immunosorbent assay (ELISA) kits for IL-6 and TNF-α were purchased from R&D Systems, Inc. (Minneapolis, MN, USA).
5. 세포 생존도 어세이5. Cell viability assay
BV2 세포는 1% 항생제(페니실린-스트렙토마이신)와 10% 열-불활성화 FBS를 포함한 RPMI1640에서 5 X 105 세포/mL, 37°C, 가습 5% CO2, 95% 공기 분위기에서 배양하였다. 미토콘드리아 환원 효소는 테트라졸륨 염을 3-[4,5-디메틸티아졸-2-일]-2,5-디페닐테트라졸륨 브로마이드(MTT)를 포르마잔 결정으로 환원하였다. 이를 사용하여 세포 생존율에 대한 화합물 1, 2, 3 및 4의 효과를 측정하였다. 세포 생존율을 측정하기 위해 5 mg/mL MTT를 각 세포 현탁액(1105 세포/mL)으로 처리하여 4시간 동안 포르마잔을 형성하였다. 형성된 포르마잔을 DMSO에 용해하고 흡광도는 540nm에서 측정하였다. BV2 cells were cultured in RPMI1640 containing 1% antibiotic (penicillin-streptomycin) and 10% heat-inactivated FBS at 5 Mitochondrial reductase reduced the tetrazolium salt to 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide (MTT) into formazan crystals. This was used to measure the effect of compounds 1, 2, 3 and 4 on cell viability. To measure cell viability, 5 mg/mL MTT was treated with each cell suspension (1105 cells/mL) to form formazan for 4 hours. The formed formazan was dissolved in DMSO and the absorbance was measured at 540 nm.
6. NO 생성 측정6. Measurement of NO production
조건화 배지 중 아질산염의 농도는 Griess 반응에 기초한 방법을 사용하여 결정되었다. 세포 배양액과 Griess 시약을 동량 혼합하고 반응시킨 후, 570nm에서 아질산염 수준을 측정하였다.The concentration of nitrite in the conditioned medium was determined using a method based on the Griess reaction. After mixing equal amounts of cell culture medium and Griess reagent and reacting, the nitrite level was measured at 570 nm.
7. IL-6 및 TNF -α 생산 어세이7. IL-6 and TNF-α production assay
배양 배지를 수집하여 상업용 키트(BioLegend, San Diego, CA, USA)를 사용하여 각 샘플에 존재하는 IL-6 및 TNF-α의 수준을 결정하였다. 분석은 제조자의 지시에 따라 수행되었다. BV2 세포를 24 웰 배양 플레이트에 2 X 105 세포/웰의 밀도로 접종하였다. 인큐베이션 후, 상청액을 회수하고 사이토카인 ELISA 키트에서 사용하여 IL-6 및 TNF-α의 농도를 측정하였다.Culture media were collected and the levels of IL-6 and TNF-α present in each sample were determined using commercial kits (BioLegend, San Diego, CA, USA). The analysis was performed according to the manufacturer's instructions. BV2 cells were seeded in a 24-well culture plate at a density of 2×10 5 cells/well. After incubation, the supernatant was recovered and used in a cytokine ELISA kit to measure the concentrations of IL-6 and TNF-α.
8. PGE2 (Prostaglandin E2) 레벨의 결정8. Determination of PGE 2 (Prostaglandin E2) levels
각 샘플에 존재하는 PGE2의 수준을 결정하기 위해 배지를 수집하였다. 분석은 제조자의 지시에 따라 수행되었다. BV2 세포를 24 웰 배양 플레이트에 2 X 105 세포/웰의 밀도로 접종하였다. 인큐베이션 후, 상청액을 수집하였다. 이후 R&D Systems, Inc.(Minneapolis, MN, USA)의 특정 ELISA 키트를 사용하여 PGE2 수준을 측정했다.Media was collected to determine the level of PGE2 present in each sample. The analysis was performed according to the manufacturer's instructions. BV2 cells were seeded in a 24-well culture plate at a density of 2×10 5 cells/well. After incubation, the supernatant was collected. PGE 2 levels were then measured using a specific ELISA kit from R&D Systems, Inc. (Minneapolis, MN, USA).
9. iNOS 및 COX-2 단백질 발현 분석9. iNOS and COX-2 protein expression analysis
펠렛화된 BV2 세포를 인산염 완충 식염수로 세척하고 RIPA 완충액에 용해시켰다. Bio-Rad Laboratories(#5000006; Hercules, CA, USA)로부터 입수한 단백질 어세이 염료 시약 농축물을 샘플 로딩 버퍼에 혼합하여 동량의 단백질을 정량하고, SDS-PAGE로 분리하였다. 분리된 단백질을 니트로셀룰로오스 멤브레인으로 이동시켰다. 막에 대한 비특이적 결합은 탈지 우유의 용액 중 인큐베이션하여 차단되었다. 멤브레인을 1차 항체와 4℃에서 밤새 인큐베이션한 후, 호스래디쉬 퍼옥시다제 표지된 2차 항체(Millipore)와 반응시켰다.Pelleted BV2 cells were washed with phosphate-buffered saline and dissolved in RIPA buffer. Protein assay dye reagent concentrate obtained from Bio-Rad Laboratories (#5000006; Hercules, CA, USA) was mixed in sample loading buffer to quantify equal amounts of protein and separated by SDS-PAGE. The separated proteins were transferred to a nitrocellulose membrane. Non-specific binding to the membrane was blocked by incubation in a solution of skim milk. The membrane was incubated with primary antibody overnight at 4°C and then reacted with horseradish peroxidase-labeled secondary antibody (Millipore).
10. NF-κB (p65) 발현 분석10. NF-κB (p65) expression analysis
NF-κB의 국소화를 연구하기 위해, BV2 세포를 24-웰 배양 플레이트에 40 X 105 세포/웰의 밀도로 시딩하고, 1시간 동안 LPS 자극(0.5 μg/mL)) 전에 3시간 동안 화합물 1 및 2의 농축물로 처리했다. 핵 추출 키트(Cayman, Ann Arbor, MI, USA)를 사용하여 세포질 분획과 핵 분획을 분리하였다. 추출된 각 분획은 제조자가 제공하는 프로토콜에 따라 용해되었다. 이어서, NF-κB (p65) 경로의 발현을 웨스턴 블롯 분석에 의해 결정하였다. 등량의 단백질을 Bio-Rad Laboratories(#5000006; Hercules, CA, USA)로부터 입수한 단백질 검정 염료 시약 농축물로 정량하고, 샘플 로딩 완충액으로 혼합하고, SDS-PAGE로 분리하였다. 이어서, 분리된 단백질을 니트로셀룰로오스 멤브레인으로 이동시켰다. 멤브레인에 대한 비특이적 결합은 탈지 우유의 용액 중에서 인큐베이션하여 차단하였다. 세포질 분획 샘플의 막은 pIκBα 항체와 4℃에서 하룻밤 인큐베이션한 반면, 핵 분획 샘플의 막은 p65 항체와 함께 인큐베이션한 다음, 호스래디쉬 퍼옥시다아제 표지 2차 항체(Millipore)와 반응시켰다.To study the localization of NF-κB, BV2 cells were seeded in 24-well culture plates at a density of 40 and 2. Cytoplasmic and nuclear fractions were separated using a nuclear extraction kit (Cayman, Ann Arbor, MI, USA). Each extracted fraction was dissolved according to the protocol provided by the manufacturer. The expression of the NF-κB (p65) pathway was then determined by Western blot analysis. Equivalent amounts of protein were quantified with protein assay dye reagent concentrate obtained from Bio-Rad Laboratories (#5000006; Hercules, CA, USA), mixed with sample loading buffer, and separated by SDS-PAGE. The separated proteins were then transferred to a nitrocellulose membrane. Non-specific binding to the membrane was blocked by incubation in a solution of skim milk. Membranes from cytoplasmic fraction samples were incubated with pIκBα antibody overnight at 4°C, whereas membranes from nuclear fraction samples were incubated with p65 antibody and then reacted with horseradish peroxidase-labeled secondary antibody (Millipore).
11. 통계 분석11. Statistical analysis
데이터는 세 번의 독립적인 실험의 평균±표준 편차로 표현된다. 중앙 집중식 분산 분석과 이어지는 Tukey의 다중 비교 검정을 사용하여 세 그룹을 비교했다. 통계 분석은 GraphPad Prism 소프트웨어 버전 5.01(GraphPad Software Inc., 미국 캘리포니아주 샌디에고)을 사용하여 수행하였다.Data are expressed as the mean ± standard deviation of three independent experiments. The three groups were compared using centralized analysis of variance followed by Tukey's multiple comparison test. Statistical analyzes were performed using GraphPad Prism software version 5.01 (GraphPad Software Inc., San Diego, CA, USA).
실시예 1. 구조 확인 및 BV2 세포 생존에 대한 분리된 대사 산물의 영향 확인Example 1. Confirmation of structure and impact of isolated metabolites on BV2 cell survival
항-신경염증 효과를 일으키는 생물활성 대사물질을 확인하기 위해, 진균 균주 Aspergillus sp. SF-7402에서 4개의 화합물(1-4)을 분리하기 위해 효율적인 크로마토그래피 분리 기술을 채택하였다 (도 1). 스펙트럼 데이터와 문헌에서 보고된 값을 비교하여 5-메톡시스테리그마토시스틴(1), 스테리그마토시스틴(2), 아베르신(3), 6,8-O-디메틸베르시콜로린 A(4)로 특징지었다.To identify bioactive metabolites that cause anti-neuroinflammatory effects, the fungal strain Aspergillus sp. An efficient chromatographic separation technique was adopted to separate the four compounds (1-4) in SF-7402 (Figure 1). By comparing the spectral data with values reported in the literature, 5-methoxysterigmatocystin (1), sterigmatocystin (2), aversin (3), and 6,8-O-dimethylversicolorin A( It was characterized as 4).
분리된 대사산물의 독성 효과를 피하기 위해, MTT 어세이를 사용하여 10-40μM 범위의 농도로 24시간 처리한 후 BV2 세포의 생존율을 측정하였다. 도 2A에 나타낸 바와 같이, 4개의 화합물(1-4) 모두 20 및 40 μM 농도에서 BV2 세포에 세포독성을 보였다. 따라서, 추가 실험은 비독성 농도 범위(2-10μM)에서 각 화합물을 사용하여 수행하였다. 신경전달물질인 2차 메신저 분자인 NO는 뇌를 포함한 많은 장기 시스템에서 다양한 생리학적 및 병리학적 과정을 중재한다. 분리된 대사산물(1-4)이 LPS에 의해 유도된 BV2 세포에서 NO 생산에 미치는 억제 효과를 조사하기 위해, 세포 배양 배지를 채취하고 Griess 반응을 사용하여 아질산염 농도를 측정하였다. 결과는 2개의 크산톤, 5-메톡시스테리그마토시스틴 (1), 스테리그마토시스틴(2)은 LPS 유도 BV2 세포에서 NO 생산을 유의하게 억제한 반면, 2개의 안트라퀴논인 아베르신 (3) 및 6,8-O-dimethylversicolorin A (4)는 NO 생산을 억제하지 않았다 (도 2B).To avoid toxic effects of the isolated metabolites, the survival rate of BV2 cells was measured after treatment for 24 hours at concentrations ranging from 10 to 40 μM using the MTT assay. As shown in Figure 2A, all four compounds (1-4) showed cytotoxicity to BV2 cells at concentrations of 20 and 40 μM. Therefore, additional experiments were performed using each compound in the non-toxic concentration range (2-10 μM). NO, a neurotransmitter second messenger molecule, mediates a variety of physiological and pathological processes in many organ systems, including the brain. To investigate the inhibitory effect of the isolated metabolites (1-4) on NO production in LPS-induced BV2 cells, cell culture medium was collected and nitrite concentration was measured using the Griess reaction. The results showed that two xanthones, 5-methoxysterigmatocystin (1) and sterigmatocystin (2), significantly inhibited NO production in LPS-induced BV2 cells, whereas two anthraquinones, aversin (3), significantly inhibited NO production in LPS-induced BV2 cells. ) and 6,8-O-dimethylversicolorin A (4) did not inhibit NO production (Figure 2B).
실시예 2. 스테리그마토시스틴의 BV2 세포에서 iNOS 및 COX-2 단백질 LPS 유도 발현 억제 확인Example 2. Confirmation of inhibition of LPS-induced expression of iNOS and COX-2 proteins in BV2 cells by sterigmatocystin
스테리그마토시스틴 (1 및 2)이 BV2 세포에서 iNOS 및 COX-2 단백질의 LPS 유도 발현에 대한 영향을 웨스턴 블롯 분석에 의해 조사하였다.The effect of sterigmatocystin (1 and 2) on LPS-induced expression of iNOS and COX-2 proteins in BV2 cells was investigated by Western blot analysis.
두가지 스테리그마토시스틴(1 및 2)은 LPS 유도성 iNOS 발현을 억제하는 것으로 밝혀졌지만, COX-2 발현에는 영향을 미치지 않았다(도 3). 크산톤 골격을 갖는 스테리그마토시스틴이 LPS 자극 BV2 세포에서 선택적 염증 억제를 위한 유망한 화합물임을 시사한다.Two sterigmatocystins (1 and 2) were found to inhibit LPS-induced iNOS expression, but had no effect on COX-2 expression (Figure 3). This suggests that sterigmatocystin with a xanthone skeleton is a promising compound for selective inflammation inhibition in LPS-stimulated BV2 cells.
실시예 3. 스테리그마토시스틴은 BV2 세포에서 LPS 유도 염증성 사이토카인의 생산 억제Example 3. Sterigmatocystin inhibits the production of LPS-induced inflammatory cytokines in BV2 cells.
전염증성 사이토카인 (pro-inflammatory cytokine)인 TNF-α, IL-6 및 PGE2의 발현에 대한 억제 효과를 평가하기 위해, LPS 자극된 BV2 세포 (LPS stimulated BV2 cell)의 치료에 단리된 대사물질(1-4)을 사용하였다. 실험은 ELISA 키트를 사용하여 수행하였다. 도 4에서와 같이, 스테리그마토시스틴 (1과 2)만이 TNF-α, IL-6 발현에 대하여 중등도의 효과적 저해능을 보인 반면, 다른 화합물은 발현을 저해하지 않았다.To evaluate the inhibitory effect on the expression of pro-inflammatory cytokines TNF-α, IL-6 and PGE 2 , isolated metabolites were treated with LPS stimulated BV2 cells. (1-4) was used. The experiment was performed using an ELISA kit. As shown in Figure 4, only sterigmatocystin (1 and 2) showed moderately effective inhibition of TNF-α and IL-6 expression, while other compounds did not inhibit expression.
화합물 1은 10μM 농도에서 세포 내 TNF-α 및 IL-6의 발현을 각각 약 25% 및 20% 감소시켰다. 화합물 2는 10μM 농도에서 활성화된 BV2 세포 중 TNF-α 발현을 30% 넘게 억제, 테스트한 모든 농도(2, 5 및 10 M)에서 IL-6 발현을 30% 넘게 억제하는 강력한 효과를 보였다. 그러나 스테리그마토시스틴 (1과 2)의 전처리는 LPS로 자극된 BV2 세포에서 PGE2 생산을 제거할 수 없었고, 이것은 iNOS 및 COX-2의 발현에 대한 스테리그마토시스틴(1 및 2)의 동일한 효과를 반영한다. 이러한 데이터는 전염증성 사이토카인 발현 억제에서 스테리그마토시스틴 (1과 2)의 역할을 보여준다. Compound 1 reduced the expression of intracellular TNF-α and IL-6 by approximately 25% and 20%, respectively, at a concentration of 10 μM. Compound 2 showed a strong effect, inhibiting TNF-α expression by more than 30% in activated BV2 cells at a concentration of 10 μM and inhibiting IL-6 expression by more than 30% at all concentrations tested (2, 5, and 10 M). However, pretreatment with sterigmatocystin (1 and 2) could not eliminate PGE2 production in LPS-stimulated BV2 cells, which is consistent with the same effect of sterigmatocystin (1 and 2) on the expression of iNOS and COX-2. reflects. These data demonstrate a role for sterigmatocystins (1 and 2) in suppressing proinflammatory cytokine expression.
실시예 4. 스테리그마토시스틴은 BV2 세포에서 LPS 유도 NF-kB 경로의 활성화를 억제함Example 4. Sterigmatocystin inhibits LPS-induced activation of the NF-kB pathway in BV2 cells
활성화 미세아교세포 (microglia)에서 NO, TNF-α 및 IL-6의 비정상적인 조절은 NF-kB (p65) 경로의 활성화와 관련되어 있기 때문에 스테리그마토시스틴 (1 및 2)은 NF-kB 활성화를 업 레귤레이션할지 여부를 더 평가했다. BV2 세포를 표시된 농도의 화합물 1 및 2로 3시간 동안 전처리한 후 LPS(1 μg/mL)로 1시간 자극하였다. 세포질 및 핵 분획은 세포 용해물로부터 추출되었고, p65의 단백질 수준은 LPS 처리 그룹의 핵 분획에서 증가하였다. 화합물 1 및 2에 의한 전처리는 농도 의존적 방식으로 p65 발현을 감소시켰다 (도 5). 세포질 분획에서 IκB-α의 인산화도 LPS 자극에 의해 증가되었다. 그러나, 화합물 1 및 2는 농도 의존적 방식으로 이 반응을 억제하였다(도 5). 이러한 결과는 스테리그마토시스틴이 NF-κB 경로의 활성화를 억제함으로써 항신경염증 효과를 나타낸다는 것을 시사한다.Because abnormal regulation of NO, TNF-α, and IL-6 in activated microglia is associated with activation of the NF-kB (p65) pathway, sterigmatocystins (1 and 2) promote NF-kB activation. We further evaluated whether to up-regulate. BV2 cells were pretreated with the indicated concentrations of compounds 1 and 2 for 3 hours and then stimulated with LPS (1 μg/mL) for 1 hour. Cytoplasmic and nuclear fractions were extracted from cell lysates, and the protein level of p65 was increased in the nuclear fraction of the LPS treatment group. Pretreatment with compounds 1 and 2 reduced p65 expression in a concentration-dependent manner (Figure 5). Phosphorylation of IκB-α in the cytosolic fraction was also increased by LPS stimulation. However, compounds 1 and 2 inhibited this reaction in a concentration-dependent manner (Figure 5). These results suggest that sterigmatocystin exerts an anti-neuroinflammatory effect by inhibiting the activation of the NF-κB pathway.
본 발명에 따르면, 크산톤 (xanthone) 화합물인 스테리그마토시스틴 또는 이의 유도체가 NO, TNF-α, IL-6의 생산 및 iNOS 단백질의 발현을 억제하여 우수한 항-신경염증 효과를 나타내고 이러한 효과는 LPS 자극 BV2 미세아교세포에서 NF-κB 경로의 활성화를 통해 매개됨을 확인하였다. 이를 통해, 신경퇴행성 질환의 예방 또는 치료에 유효하게 사용될 수 있으며, 신경퇴행성 질환의 치료를 위한 치료제의 개발을 위한 유망한 기초를 제공한다.According to the present invention, sterigmatocystin or a derivative thereof, a xanthone compound, exhibits excellent anti-neuroinflammatory effects by inhibiting the production of NO, TNF-α, and IL-6 and the expression of iNOS protein. It was confirmed that this was mediated through activation of the NF-κB pathway in LPS-stimulated BV2 microglial cells. Through this, it can be effectively used in the prevention or treatment of neurodegenerative diseases, and provides a promising basis for the development of therapeutic agents for the treatment of neurodegenerative diseases.
이상으로 본 발명의 내용의 특정한 부분을 상세히 기술하였는바, 당업계의 통상의 지식을 가진 자에게 있어서, 이러한 구체적 기술은 단지 바람직한 실시양태일 뿐이며, 이에 의해 본 발명의 범위가 제한되는 것이 아닌 점은 명백할 것이다. 따라서, 본 발명의 실질적인 범위는 첨부된 청구항들과 그것들의 등가물에 의하여 정의된다고 할 것이다.As above, specific parts of the content of the present invention have been described in detail, and for those skilled in the art, it is clear that these specific techniques are merely preferred embodiments and do not limit the scope of the present invention. It will be obvious. Accordingly, the substantial scope of the present invention will be defined by the appended claims and their equivalents.
Figure PCTKR2023012186-appb-img-000007
Figure PCTKR2023012186-appb-img-000007

Claims (6)

  1. 다음의 구조식 1로 표시되는 크산톤(xanthone) 화합물을 유효성분으로 포함하는 신경퇴행성 질환 (neurodegenerative disease)의 예방 또는 치료용 조성물:A composition for preventing or treating neurodegenerative disease comprising a xanthone compound represented by the following structural formula 1 as an active ingredient:
    Figure PCTKR2023012186-appb-img-000008
    Figure PCTKR2023012186-appb-img-000008
    상기 구조식 1에서 R은 알콕시 또는 수소이다. In structural formula 1, R is alkoxy or hydrogen.
  2. 제1항에 있어서, 상기 R은 메톡시인 5-메톡시스테리그마토시스틴(5-methoxysterigmatocystin)을 포함하는 조성물.The composition of claim 1, wherein R is methoxy, comprising 5-methoxysterigmatocystin.
  3. 제1항에 있어서, 상기 R은 수소인 스테리그마토시스틴(sterigmatocystin)을 포함하는 조성물. The composition of claim 1, wherein R is hydrogen and contains sterigmatocystin.
  4. 제1항에 있어서, 상기 화합물은 남극 진균 Aspergillus sp. SF-7402에서 유래된 것을 특징으로 하는 조성물.The method of claim 1, wherein the compound is an Antarctic fungus Aspergillus sp. A composition characterized in that it is derived from SF-7402.
  5. 제1항에 있어서, 상기 신경퇴행성 질환은 파킨슨 질환, 알츠하이머 질환 (AD), 뇌졸중, 치매, 근위축증 (MD), 다발성 경화증 (MS), 근위축성측색경화증 (ALS), 낭성 섬유증, 앙겔만(Angelman) 증후군, 리들(Liddle) 증후군, 픽 질환, 파젯 질환 또는 외상 뇌 손상인, 조성물.The method of claim 1, wherein the neurodegenerative disease includes Parkinson's disease, Alzheimer's disease (AD), stroke, dementia, muscular dystrophy (MD), multiple sclerosis (MS), amyotrophic lateral sclerosis (ALS), cystic fibrosis, and Angelman's disease. ) syndrome, Liddle syndrome, Pick's disease, Paget's disease or traumatic brain injury.
  6. 진균 Aspergillus sp. SF-7402를 배양하여 대사물질을 추출하는 단계; 및The fungus Aspergillus sp. Culturing SF-7402 to extract metabolites; and
    대사물질 중 크산톤(xanthone) 화합물을 분리하는 단계를 포함하는, 제1항의 조성물을 제조하는 방법.A method for producing the composition of claim 1, comprising the step of separating xanthone compounds from metabolites.
PCT/KR2023/012186 2022-08-18 2023-08-17 Composition for preventing or treating neurodegenerative disease comprising compounds originating from aspergillus sp. sf-7402 WO2024039197A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
KR1020220103406A KR20240025326A (en) 2022-08-18 2022-08-18 Composition for Preventing or Treating Neurodegenerative Disease Comprising Compounds from Aspergillus sp. SF-7402
KR10-2022-0103406 2022-08-18

Publications (1)

Publication Number Publication Date
WO2024039197A1 true WO2024039197A1 (en) 2024-02-22

Family

ID=89942016

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2023/012186 WO2024039197A1 (en) 2022-08-18 2023-08-17 Composition for preventing or treating neurodegenerative disease comprising compounds originating from aspergillus sp. sf-7402

Country Status (2)

Country Link
KR (1) KR20240025326A (en)
WO (1) WO2024039197A1 (en)

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR102236079B1 (en) * 2019-11-13 2021-04-05 건국대학교 산학협력단 A Method for Modulating Metabolism of Aspergillus

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR102236079B1 (en) * 2019-11-13 2021-04-05 건국대학교 산학협력단 A Method for Modulating Metabolism of Aspergillus

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
CAO THAO QUYEN, LIU ZHIMING, DONG LINSHA, LEE HWAN, KO WONMIN, VINH LE BA, TUAN NGUYEN QUOC, KIM YOUN-CHUL, SOHN JAE HAK, YIM JOUN: "Identification of Potential Anti-Neuroinflammatory Inhibitors from Antarctic Fungal Strain Aspergillus sp. SF-7402 via Regulating the NF-κB Signaling Pathway in Microglia", MOLECULES, SPRINGER VERLAG, BERLIN, DE, vol. 27, no. 9, 29 May 2022 (2022-05-29), DE , pages 2851, XP093139623, ISSN: 1433-1373, DOI: 10.3390/molecules27092851 *
KIM KWAN-WOO, KIM HYE JIN, SOHN JAE HAK, YIM JOUNG HAN, KIM YOUN-CHUL, OH HYUNCHEOL: "Anti-neuroinflammatory effect of 6,8,1′-tri- O -methylaverantin, a metabolite from a marine-derived fungal strain Aspergillus sp., via upregulation of heme oxygenase-1 in lipopolysaccharide-activated microglia", NEUROCHEMISTRY INTERNATIONAL, ELSEVIER, AMSTERDAM, NL, vol. 113, 1 February 2018 (2018-02-01), AMSTERDAM, NL , pages 8 - 22, XP093139625, ISSN: 0197-0186, DOI: 10.1016/j.neuint.2017.11.010 *
PARANJAPE SMITA, CHIANG YI-MING, SANCHEZ JAMES, ENTWISTLE RUTH, WANG CLAY, OAKLEY BERL, GAMBLIN T.: "Inhibition of Tau Aggregation by Three Aspergillus nidulans Secondary Metabolites: 2,ω-Dihydroxyemodin, Asperthecin, and Asperbenzaldehyde", PLANTA MEDICA, THIEME VERLAG, DE, vol. 80, no. 01, 10 January 2014 (2014-01-10), DE , pages 77 - 85, XP093139628, ISSN: 0032-0943, DOI: 10.1055/s-0033-1360180 *
RABIE C J, STEYN M, VAN SCHALKWYK G C: "New species of Aspergillus producing sterigmatocystin", APPLIED AND ENVIRONMENTAL MICROBIOLOGY, AMERICAN SOCIETY FOR MICROBIOLOGY, US, vol. 33, no. 5, 1 May 1977 (1977-05-01), US , pages 1023 - 1025, XP093139632, ISSN: 0099-2240, DOI: 10.1128/aem.33.5.1023-1025.1977 *

Also Published As

Publication number Publication date
KR20240025326A (en) 2024-02-27

Similar Documents

Publication Publication Date Title
Batkhuu et al. Suppression of NO production in activated macrophages in vitro and ex vivo by neoandrographolide isolated from Andrographis paniculata
Ueda et al. New sesquiterpene from Vietnamese agarwood and its induction effect on brain-derived neurotrophic factor mRNA expression in vitro
WO2011152671A9 (en) Pharmaceutical composition for the prevention or treatment of inflammatory diseases or immune diseases containing ramalin
Hu et al. Synthesis and biological evaluation of clovamide analogues as potent anti-neuroinflammatory agents in vitro and in vivo
El-Sedawy et al. Metabolism of swertiamarin from Swertia japonica by human intestinal bacteria
US5498405A (en) Indole alkaloids useful as UV protective and anti-inflammatory agents
Jung et al. Lignans from Saururus chinensis exhibit anti-inflammatory activity by influencing the Nrf2/HO-1 activation pathway
US10017489B2 (en) Method for preparing benzopyran compound and application thereof in treating pulmonary fibrosis
US6838443B2 (en) Antihelminthic anthraquinones and method of use thereof
WO2024039197A1 (en) Composition for preventing or treating neurodegenerative disease comprising compounds originating from aspergillus sp. sf-7402
Carraz et al. Isolation and antimalarial activity of new morphinan alkaloids on Plasmodium yoelii liver stage
US7854946B1 (en) Anti-inflammatory and anti-HIV compositions and methods of use
KR980008225A (en) Anticancer drugs containing dexacinol angelate as an active ingredient
KR0176413B1 (en) Decursin as an anti-tumour agent
US20060105967A1 (en) Flavone derivatives as TNFalpha inhibitors or antagonists
Hashim et al. Phenylpropanoid-conjugated iridoid glucosides from leaves of Morinda morindoides
CN113717105B (en) Diterpene alkaloid compound and extraction method and application thereof
Forget et al. Isolation of a jadomycin incorporating l-ornithine, analysis of antimicrobial activity and jadomycin reactive oxygen species (ROS) generation in MDA-MB-231 breast cancer cells
Nandjou Kenfack et al. Antisalmonellal activities of extracts, fractions, compounds and semi-synthetic flavonoid derivatives from Tristemma hirtum p. beauv (Melastomataceae)
Zhou et al. HPLC-PDA-Guided isolation of glucosyloxybenzyl 2-isobutylmalates from the pseudobulbs of Bletilla striata with neuroprotective and antimicrobial activities
KR102102680B1 (en) Pharmaceutical composition for preventing or treating gastrointestinal disorder comprising as an active ingredient a compound isolated from Bacillus amyloliquefaciens RWL-1 culture medium
Zhao et al. Cyclic pentapeptides with anti-inflammatory, cytotoxic or α-glucosidase inhibitory activities from Basidiobolus meristosporus
KR101712184B1 (en) Dimeric Sesquiterpene Compound as Nrf2 Activator and Phamaceutical Composition Conprising the Same
KR101987854B1 (en) Cyclic hexapeptide-based compounds and uses thereof
WO2017078353A1 (en) Composition including stipitalide as active ingredient

Legal Events

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

Ref document number: 23855163

Country of ref document: EP

Kind code of ref document: A1