WO2021167309A1 - Asthma-suppressing composition containing aldehyde dehydrogenase - Google Patents

Asthma-suppressing composition containing aldehyde dehydrogenase Download PDF

Info

Publication number
WO2021167309A1
WO2021167309A1 PCT/KR2021/001934 KR2021001934W WO2021167309A1 WO 2021167309 A1 WO2021167309 A1 WO 2021167309A1 KR 2021001934 W KR2021001934 W KR 2021001934W WO 2021167309 A1 WO2021167309 A1 WO 2021167309A1
Authority
WO
WIPO (PCT)
Prior art keywords
asthma
saccharomyces cerevisiae
aldh
present
kwonp
Prior art date
Application number
PCT/KR2021/001934
Other languages
French (fr)
Korean (ko)
Inventor
권흥택
Original Assignee
주식회사 피코엔텍
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from KR1020210020186A external-priority patent/KR102460545B1/en
Application filed by 주식회사 피코엔텍 filed Critical 주식회사 피코엔텍
Publication of WO2021167309A1 publication Critical patent/WO2021167309A1/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23LFOODS, FOODSTUFFS, OR NON-ALCOHOLIC BEVERAGES, NOT COVERED BY SUBCLASSES A21D OR A23B-A23J; THEIR PREPARATION OR TREATMENT, e.g. COOKING, MODIFICATION OF NUTRITIVE QUALITIES, PHYSICAL TREATMENT; PRESERVATION OF FOODS OR FOODSTUFFS, IN GENERAL
    • A23L33/00Modifying nutritive qualities of foods; Dietetic products; Preparation or treatment thereof
    • A23L33/10Modifying nutritive qualities of foods; Dietetic products; Preparation or treatment thereof using additives
    • A23L33/14Yeasts or derivatives thereof
    • A23L33/145Extracts
    • 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
    • A61K36/062Ascomycota
    • A61K36/064Saccharomycetales, e.g. baker's yeast
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/44Oxidoreductases (1)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N1/00Microorganisms, e.g. protozoa; Compositions thereof; Processes of propagating, maintaining or preserving microorganisms or compositions thereof; Processes of preparing or isolating a composition containing a microorganism; Culture media therefor
    • C12N1/14Fungi; Culture media therefor
    • C12N1/16Yeasts; Culture media therefor
    • C12N1/18Baker's yeast; Brewer's yeast

Definitions

  • the present invention relates to an asthma inhibitory composition comprising an aldehyde dehydrogenase. More specifically, the present invention provides Saccharomyces cerevisiae KwonP-3 and Saccharomyces cerevisiae KwonP-3) with accession number KCTC13925BP (Saccharomyces cerevisiae KwonP-1), KCTC14122BP ( Saccharomyces cerevisiae KwonP-2), KCTC14123BP (Saccharomyces cerevisiae KwonP-3) isolated from strains.
  • KCTC13925BP Saccharomyces cerevisiae KwonP-1
  • KCTC14122BP Saccharomyces cerevisiae KwonP-2
  • KCTC14123BP Sacharomyces cerevisiae KwonP-3 isolated from strains.
  • Asthma is a chronic disease that causes respiratory distress symptoms due to bronchial hypersensitivity and intermittent airway constriction due to chronic inflammation in the airways. It is known that there is no cure yet.
  • the antibodies recognize it and activate the immune system, leading to an inflammatory response that causes inflammation, swelling, and constriction of the airways. do.
  • Th2 lymphocytes in asthmatic patients are activated to increase IL-4, IL-5, IL-9 or IL-13.
  • IL-4 promotes mucus production and B-cell proliferation, and causes dyspnea. It produces IgE, which activates mast cells that secrete inflammatory mediators such as histamine or leukotriene, and expresses VCAM-1, which delivers eosinophils to lung tissue.
  • IgE which activates mast cells that secrete inflammatory mediators such as histamine or leukotriene
  • VCAM-1 which delivers eosinophils to lung tissue.
  • typical respiratory symptoms of asthma such as shortness of breath, cough, sputum, and wheezing (yin)
  • atypical symptoms may also appear.
  • Asthma sufferers may also experience elevated heart rate, dry crackling, etc.
  • cyanosis due to lack of oxygen, strong chest pain, numbness in limbs or sweating in the palms of the palms may be felt just before loss of consciousness, which is common Severe asthma attacks that do not respond to treatment can be life-threatening, leading to respiratory arrest and death.
  • This bronchial asthma is a very common disease that is reported to be suffering from about 5 to 10% of the total population in most countries around the world, including Korea. The prevalence is increasing worldwide.
  • External factors include airborne pollutants, various allergens and industrial dust. These asthma-inducing substances produce IgE in an immune response, and the produced IgE blocks the nasal mucosa, skin, and airways, and binds to receptors on the surface of mast cells in the bronchi to cause inflammation.
  • 'Treg cells' regulatory T cells
  • Asthma also has genetic factors. Human chromosome 5, ⁇ 2-adrenoceptor gene, and human chromosome 11, 12, 14, and 16 are involved, and it is reported that changes in these genes cause an increase in IgE and hypersensitivity of the bronchial tubes.
  • bronchial asthma When the nasal mucosa or bronchial mucosa is stimulated by bronchial asthma triggers, an excessive immune response (allergic reaction) occurs and inflammation occurs in the airway mucosal tissue, and accordingly, the three major symptoms of bronchial asthma: cough, aerobic wheezing, and dyspnea. This causes bronchial asthma and rhinitis symptoms such as runny nose, sneezing, or stuffy nose.
  • ROS reactive oxygen species
  • Treatment methods for bronchial asthma include avoidance and desensitization to remove or avoid allergens that cause bronchial asthma, drug therapy to select drugs, or immunotherapy to immunize by injecting a small amount of an allergen. .
  • bronchial asthma used in the drug therapy
  • a bronchodilator as a ⁇ 2-agonist that improves symptoms by widening a constricted airway
  • a corticosteroid-based inhalant for symptom relief or an anti-allergic agent
  • some asthma treatment drugs such as water-soluble IL-4 receptor, water-soluble IL-13 and anti-IL-5 antibody using a mechanism to inhibit the action of Th2-type cytokines are being developed and sold.
  • asthma medications can be purchased only through a doctor's consultation and prescription, and since most of them are steroid-based drugs, there is a problem that side effects may occur when used for a long period of time.
  • aldehyde dehydrogenase extracted from yeast inhibits cell infiltration and interleukin-4 (interleukin-4).
  • -4, IL-4) and interleukin-13 IL-13
  • Interleukin 4 (hereinafter, abbreviated as IL-4) secreted from T cells is a cytokine of about 20 kD consisting of 129 amino acids, and is an important cytokine that increases B-cell differentiation factors and B-cell proliferation factors, especially IgE. .
  • IL-4 is initially secreted by basophils and additionally secreted by activated T-cells (TH2). It has been reported to contribute to B cell activation, IgE type conversion, and induction of Th2 differentiation.
  • IL-4 production by Th2 cells is also accelerated by IL-4, and the receptor for IL-4 is a 130 kD protein that binds two cysteines, IL-4R ⁇ + ⁇ b and IL-4R ⁇ +IL-13R ⁇ .
  • IL-4 has many biological functions, such as stimulating activated B cells and T cells and differentiating B cells into plasma cells, and is an important regulator of humoral immunity and adaptive immunity.
  • IL-4 is known to induce class switching from B progenitor cells to B cells capable of producing IgE and IgG4, as well as promote the production of MHC class II. It causes an increase in IgE, resulting in an immediate onset hypersensitivity allergic reaction. IL-4 acts synergistically with IL-13 and is the causative agent of IgE- or eosinophil-mediated inflammatory responses.
  • IgE antibody When an IgE antibody is produced by exposure to an antigen, IgE binds to the surface of mast cells or basophils, and when the IgE-bound mast cells are exposed to the same antigen a second time, degranulation of the mast cells or basophil intragranule occurs and vasoactive substances Phosphorus histamine, serotonin, chemotactic factors of eosinophils or neutrophils, leukotrienes, and prostaglandins are released.
  • the ALDH-containing composition of the present invention can be an asthma treatment with small side effects by inhibiting IgE increase through IL-4 or IL-13 inhibition.
  • Interleukin 13 is a protein consisting of 122 amino acids, and the receptor for IL-13 is CD132 ( ⁇ c).
  • IL-13 is functionally similar to IL-4 and is secreted from Th2 cells, dendritic cells, and mast cells. alleviate
  • the receptor for IL-13 is composed of IL-13R ⁇ and IL-4R ⁇ , the IL-13 receptor also responds to IL-4.
  • the receptor for Il-13 is expressed in human B cells, but not in mouse B cells. IL-13 also differentiates monocytes into dendritic cells. However, it has been reported that IL-13 gene inactive mice do not actively produce Th2 cells or IgE.
  • IL-13 inhibitors or IL-13 signaling blockers can also be used as therapeutic agents for immune diseases such as atopy or asthma.
  • the aldehyde dehydrogenation reaction is a reaction for changing acetaldehyde or various aldehydes present in human metabolites into organic acids that can be discharged as waste products.
  • the enzyme that promotes this aldehyde dehydrogenation reaction decomposes the aldehyde, which is toxic to cells, and relieves carbonyl stress applied to cells and tissues.
  • aldehyde dehydrogenases A series of proteins containing functional groups of enzymes involved in aldehyde dehydrogenation are called aldehyde dehydrogenases, and about 20 types of ALDH are known so far. Therefore, in the present specification, the scope of the term ALDH includes all of the 20 known types of ALDH, all fusion proteins containing all or part of the dehydrogenation functional groups of enzymes involved in aldehyde dehydrogenation, and enzymes involved in aldehyde dehydrogenation. All recombinant proteins containing some or all of the dehydrogenation functional groups are included.
  • This prior art describes in detail mitochondrial aldehyde dehydrogenase-2 (ALDH2), a tetrameric protein consisting of four identical subunits encoded in the nuclear genome and transported into the mitochondria, each of 500 amino acid residues. wherein the tetramer is a dimer of a dimer, wherein the interface between the monomers forming the dimer is different from the interface between the two dimers forming the tetramer, and each subunit has three major sites, i.e., an aldehyde Dehydrogenation catalytic sites, coenzyme or NAD+-binding sites, and oligomerization sites are disclosed.
  • ADH2 mitochondrial aldehyde dehydrogenase-2
  • ALDH2 related diseases and conditions include ischemic stress, chronic free-radical related diseases, acute free-radical related diseases, insensitivity to nitroglycerin (eg, in angina and heart failure), hypertension, diabetes. , osteoporosis, cancer, Fanconi anemia, Alzheimer's disease, Parkinson's disease, alcoholism, alcohol intolerance, alcohol addiction, alcohol abuse disease, alcohol intoxication, alcohol dependence, alcohol poisoning
  • modulators of ALDH2, agonists of ALDH2, or antagonists of ALDH2, including symptoms of alcohol consumption, and drug addiction include ischemic stress, chronic free-radical related diseases, acute free-radical related diseases, insensitivity to nitroglycerin (eg, in angina and heart failure), hypertension, diabetes. , osteoporosis, cancer, Fanconi anemia, Alzheimer's disease, Parkinson's disease, alcoholism, alcohol intolerance, alcohol addiction, alcohol abuse disease, alcohol intoxication, alcohol dependence, alcohol poisoning
  • An agonist of ALDH2 is also useful for reducing the level of a compound that generates an aldehyde substrate of ALDH2 in a subject upon ingestion, absorption, or inhalation of a compound that generates an aldehyde substrate of ALDH2.
  • ALDH2 antagonists are used as adjuncts to standard cancer treatment. It has also been disclosed that antagonists of ALDH2 are useful in treating or suppressing alcoholism and drug addiction. However, this prior art does not make any suggestion or mention about the possibility that ALDH, which is the subject of the present invention, may inhibit the action of interleukin-4 or interleukin-13.
  • Korean Patent Application No. 10-2020-0019858 describes Saccharomyces cerevisiae that simultaneously produces aldehyde dehydrogenase (hereinafter abbreviated as ALDH) and glutathione, which are used as active ingredients in the pharmaceutical composition and food composition for inhibiting asthma of the present invention.
  • ALDH aldehyde dehydrogenase
  • glutathione glutathione
  • a basic object of the present invention is to provide a food composition containing an aldehyde dehydrogenase having an effect of alleviating and suppressing asthma symptoms.
  • Another object of the present invention is a composition for treating asthma and atopic rhinitis containing, as an active ingredient, a fusion protein or a recombinant protein comprising an aldehyde dehydrogenase having an effect of preventing and treating asthma symptoms or an action site involved in an aldehyde dehydrogenation reaction. is to provide
  • Saccharomyces cerevisiae yeast accession number KCTC13925BP (Saccharomyces cerevisiae KwonP-1), KCTC14122BP ( Saccharomyces cerevisiae KwonP-2), KCTC14123BP ( Saccharomyces cerevisiae KwonP-3) aldehyde isolated from It is achieved by providing a composition which contains a dehydrogenase and exhibits an asthma prevention or alleviation effect.
  • the yeast used by the present inventors for the production of ALDH for asthma suppression is Saccharomyces cerevisiae yeast, which is generally widely used for fermenting beer or bread. About 250 types of Saccharomyces cerevisiae yeast known to date have been reported, and all of them can be appropriately used for the ALDH production of the present invention.
  • the asthma inhibitory composition of the present invention is a novel Saccharomyces cerevisiae yeast deposited by the present inventors with the international depository KCTC, deposit number KCTC13925BP (Saccharomyces cerevisiae KwonP-1), KCTC14122BP ( Saccharomyces cerevisiae KwonP-1) cerevisiae KwonP-2) or KCTC14123BP ( Saccharomyces cerevisiae KwonP-3) may be used as a raw material for the production of the ALDH-containing composition for inhibiting asthma of the present invention.
  • KCTC13925BP Sacharomyces cerevisiae KwonP-1
  • KCTC14122BP Saccharomyces cerevisiae KwonP-1
  • KCTC14123BP Saccharomyces cerevisiae KwonP-3
  • the ALDH-containing compositions according to the present invention are effective in treating and preventing inflammatory reactions caused by oxidative stress, such as bronchial inflammation, allergic rhinitis, and asthma.
  • ALDH is not only contained in easily purchased foods, but also has the advantage of having little side effects on the human body because it is extracted from edible yeast.
  • the yeast extract of Saccharomyces cerevisiae according to the present invention can be used for immunoglobulin E (IgE) and total number of inflammatory cells and lymphocytes, monocytes, neutrophils, and eosinophils in the bronchoalveolar lavage fluid of asthma-induced mice in an in vivo experiment. It has the activity of inhibiting the increase and the secretion of airway mucus, and reduces the Th2 cytokines IL-4 and IL-13 and the degree of airway inflammation.
  • IgE immunoglobulin E
  • composition of the present invention exhibits an effect of preventing or treating bronchial asthma, allergic asthma, atopic asthma, non-topic asthma, exercise-induced asthma, aspirin asthma, psychogenic asthma, or alveolar asthma.
  • the ALDH contained in the composition of the present invention reduces airway hypersensitivity, suppresses the amount of active oxygen in the airways, or inhibits the infiltration of inflammatory cells in the bronchial tubes, thereby reducing the number of inflammatory cells in the bronchus, reducing the concentration of IgE antibody in the serum, or IL-4, a Th2 cytokine and IL-13, and the effect of inhibiting the expression of apoptosis-related proteins (JNK, ERK, p38, BCL-2 family, caspase-3, NF- ⁇ B) in the mechanism of regulating hypersensitive respiratory symptoms indicates Accordingly, the ALDH-containing compositions of the present invention can be used as pharmaceuticals and health foods useful for the prevention, improvement or treatment of bronchial asthma.
  • 1 is a graph showing the inhibitory effect of interleukin-4 increase in RBL-2HE cell line by the ALDH-containing composition of the present invention.
  • FIG. 2 is a graph showing the inhibitory effect of interleukin-13 in the sera of control and experimental mice of the ALDH-containing composition of the present invention.
  • FIG. 3 is a graph showing the IgE reduction effect of the ALDH-containing composition of the present invention in the sera of control and experimental mice.
  • ALDH-containing pharmaceutical composition or food composition of the present invention wild Saccharomyces yeast, Saccharomyces cerevisiae KwonP1 (KCTC13925BP), Saccharomyces cerevisiae KwonP2 (KCTC14122BP) or Saccharomyces cerevisiae Dry powder, lysate, or ALDH-containing extract powder of KwonP3 (KCTC14123BP) is included as an active ingredient.
  • Saccharomyces cerevisiae strain dry powder contained as an active ingredient in the composition of the present invention a method for preparing strains used for preparing ALDH-containing extract powder, lysate, Korea cited in the specification of the present invention It is described in detail in the specification of patent application No. 10-2020-00198858.
  • the ALDH-containing composition of the present invention may further include a pharmaceutically acceptable carrier.
  • the pharmaceutically acceptable carrier includes, but is not limited to, saline, sterile water, buffered saline, cyclodextrin, dextrose solution, maltodextrin solution, glycerol, ethanol, and the like.
  • the ALDH-containing composition of the present invention may contain glutathione derived from Saccharomyces cerevisiae, and may be formulated as an aqueous solution, suspension, emulsion, etc. by additionally adding a diluent, dispersant, surfactant, binder, lubricant, etc. , pills, capsules, granules or tablets.
  • the pharmaceutical composition of the present invention is administered in a pharmaceutically effective amount.
  • the effective amount level can be determined according to the patient's disease type, severity, drug activity, sensitivity to drug, administration time, administration route and excretion rate, duration of treatment, factors including concurrent drugs, and factors well known to those skilled in the art.
  • the food composition of the present invention is characterized in that it is formulated with one selected from the group consisting of tablets, pills, powders, granules, powders, capsules, and liquid formulations, including one or more of carriers, diluents, excipients, and additives.
  • Additives that may be further included in the food composition of the present invention include natural carbohydrates, flavoring agents, nutrients, vitamins, synthetic flavoring agents, natural flavoring agents, colorants, fillers, alginic acid and salts thereof, organic acids, protective colloidal thickeners, pH Regulators, stabilizers, preservatives, antioxidants, glycerin, alcohols and the like can be used.
  • Example 1-1 Yeast fermentation process in Saccharomyces cerevisiae containing ALDH
  • Saccharomyces cerevisiae yeast (KCTC139525BP) containing ALDH was fermented and cultured for 24 hours in an incubator at 160 rpm and 30 ° C using YPD medium (yeast extract, peptone, and glucose containing medium) in a 200 mL flask, This culture was carried out for 72 hours through a 5 L fermenter (Marado-05D-PS, CNS, Korea). After completion of the culture, the yeast was centrifuged using a high-speed centrifuge (Supra R22, Hanil, Korea).
  • YPD medium yeast extract, peptone, and glucose containing medium
  • Example 1-2 Preparation of ALDH-containing composition of the present invention
  • the centrifuged ALDH-containing yeast was frozen in a cryogenic freezer (CLN-52U, Nihon freezer, Japan) for 2 days, and then freeze-dried for 2 days in a freeze dryer (FDU-7006, Operon, Korea).
  • a freeze dryer FDU-7006, Operon, Korea.
  • PBS phosphate-buffered saline
  • 0.5 mm glass beads for cell disruption 11079105, Biospec 10 g was put into the bead homogenizer (Mixer Mill MM400, Retsch, Germany) to disrupt yeast over a total of 3 times for 2 minutes each.
  • After centrifugation using a high-speed centrifuge (Supra R22, Hanil, Korea), only the supernatant was separated and freeze-dried for 2 days with a freeze dryer (FDU-7006, Operon, Korea).
  • OVA Ovlabumin; mouse inflammation inducer
  • aluminum hydroxide immune response enhancer
  • the powder of the ALDH-containing composition of the present invention was dissolved in physiological saline from the time point after a week had elapsed and oral administration was carried out. caused It was conducted once a day for a total of 5 days.
  • the powder of the composition containing ALDH of the present invention was administered at 6 mg/kg/day, 30 mg/kg/day, and 150 mg/kg/day.
  • the powder of the ALDH-containing composition of the present invention was administered in the morning, and 20 ug/head/day of ovalbumin was administered by inhalation using a nebulizer in the afternoon. And this operation was repeated for 5 days. At this time, the powder administration of the ALDH-containing composition of the present invention was administered at different doses in 3 groups of 6 mg/kg, 30 mg/kg, and 150 mg/kg.
  • Dexamethasone (steroid) administration group was 1mg/kg/day per mouse.
  • control is a normal group to which ovalbumin, an asthma inducer, dexamethasone, a control drug for asthma treatment, or the powder of the composition containing ALDH of the present invention was not administered as a control group.
  • the OVA group was a group in which ovalbumin was administered intraperitoneally on the 1st and 8th days of the experiment and by inhalation using a nebulizer on the 14th day to induce asthma by the above method.
  • ovalbumin was administered intraperitoneally on the 1st and 8th days as described above, and from the 14th day onwards, dexamethasone (steroid) was administered 1mg/kg/day per mouse in the morning and repeated for 5 days from the 14th day, and ovalbumin 20ug/day in the afternoon.
  • the group was administered head/day by inhalation using a nebulizer.
  • the ALDH 6mg/kg group is a group in which 6mg/kg of the ALDH-containing composition powder of the present invention is administered in the morning repeatedly for 5 days from the 14th day, and 20ug/head/day of ovalbumin is administered by inhalation using a nebulizer in the afternoon, and ALDH
  • the 30mg/kg group is a group in which 30mg/kg of the ALDH-containing composition powder of the present invention is administered in the morning repeatedly for 5 days from the 14th day, and ovalbumin 20ug/head/day is administered by inhalation using a nebulizer in the afternoon
  • the /kg group is a group in which 150 mg/kg of the ALDH-containing composition powder of the present invention is administered in the morning repeatedly for 5 days from the 14th day, and 20 ug/head/day of ovalbumin is administered by inhalation using a nebulizer in the afternoon.
  • mice were sacrificed and Bronchioalveolar fluid (BALF) components were analyzed in the lung bronchi to measure the number of WBCs (white blood cells), and cytokines such as IL-4 and IL-13 were analyzed by enzyme-linked immunoassay (ELISA) method. was used for measurement.
  • BALF Bronchioalveolar fluid
  • nebulizer system After oral administration with the nebulizer system for 5 days, an autopsy was performed the next day. There were a total of 8 animals in one group, and blood was collected from all 8 mice. Bronchial Alveolar Lavage Fluid (BALF) was collected from 4 animals in each group, and the lungs were removed from the remaining 4 animals. The extracted lungs were divided into thirds. One was stored in 10% formalin to examine histopathology, and the other two were stored at -80°C to view cytokines.
  • BALF Bronchial Alveolar Lavage Fluid
  • the BALF obtained from the animal was centrifuged at 4°C at 3000 rpm for 5 minutes. Remove the supernatant and mix with PBS buffer to the remaining pellet. Thereafter, blood cell analysis was performed using a hemacytometer (Hemavet 950FS, Drew Scientific Inc, Korea).
  • the remaining BALF used for blood cell analysis was centrifuged at 4°C and 3,000 rpm for 5 minutes. After that, the supernatant was discarded, PBS buffer was added, and centrifugation was performed at 800 rpm for 5 minutes using a Cyto-Tek machine. Then, it was stained with Diff Qucik reagent.
  • BALF Bronchial Alveolar Lavage Fluid
  • Diff Qucik reagent is used using a hemocytometer (Hemavet 950FS, Drew Scientific Inc, Korea). Inflammatory cell count, neutrophil and mononuclear leukocytes were analyzed by staining using
  • An IgE ELISA set was used, and Capture Ab diluted in Coating Buffer was added to a micro well and incubated at 4 °C overnight. After washing 3 times, add assay diluent to each well and incubate at room temperature. After washing 3 times, add standard and sample diluted with assay diluent to each well and incubate at room temperature. After washing 5 times, put Working Detector in each well and incubate at room temperature. After washing 7 times, add Substrate Solution to each well and incubate in dark conditions at room temperature. Stop solution in each well was measured at 450 nm within 30 minutes.
  • Paraffin removal and hydration were carried out, and after dehydration, they were washed with tap water for 5 minutes, dried well, and then stained with heamatoxylin solution for 2 minutes. After staining, clarification and dehydration were performed, and tissue encapsulation was performed.
  • tissue encapsulation was performed.
  • Enzyme-Linked Immunosorbent Assay Add Capture Ab diluted in Coating Buffer to micro-wells and incubate at 4°C overnight. After washing 3 times, add assay diluent to each well and incubate at room temperature. After washing 3 times, add standard sample diluted with Assay Diluent to each well and incubate at room temperature. After washing 5 times, put Working Detector in each well and incubate at room temperature. After washing 7 times, add Substrate Solution to each well and incubate in dark conditions at room temperature. Stop solution in each well was measured at 450 nm within 30 minutes. Cytokine ELISA analysis was Th1 (IL-6, IFN- ⁇ ), Th2 (IL-4, IL-5, IL-13), Th17 (IL-6, TNF- ⁇ ) were analyzed.
  • Th1 IL-6, IFN- ⁇
  • Th2 IL-4, IL-5, IL-13
  • Th17 IL-6, TNF- ⁇
  • RT-PCR Real-time polymerase chain reaction
  • OVA ovalbumin
  • Al hydroxide immunophilicity enhancer
  • Example 5-1 Administration of the ALDH-containing composition of the present invention
  • Lyophilized ALDH-containing yeast lysate was dissolved in phosphate-buffered saline (PBS) to a concentration of 6 mg/kg, 30 mg/kg, and 150 mg/kg based on 560 ⁇ L, respectively, and then vortexer (F202A0175, VELP scientific, Italy) for 1 minute (C. Kla an. et al., J. Immunol, 1;199(1):48-61, 2017).
  • the positive control material, dexamethasone (Sinilpharm, Korea) was dissolved in physiological saline (Cleancle, JW Pharmaceutical, Korea) to a concentration of 1 mg/kg and mixed with a vortexer (F202A0175, VELP scientific, Italy) for 1 minute (C. Kla combined. et al., J. Immunol, 1;199(1):48-61, 2017).
  • Example 5-2 Administration of the ALDH-containing composition of the present invention to an asthma-induced mouse model
  • Example 6-1 Mouse autopsy and collection of blood and lung tissue
  • OVA spraying and oral administration using an ultrasonic nebulizer were performed for 5 days, and an autopsy was performed the next day.
  • Blood was collected from all 8 animals per group using a 1 mL syringe (Kovax-syringe 1 mL 26G 1/2, Koreavaccine, Korea), and lungs were removed from 4 mice.
  • the excised lungs were divided into thirds, one stored in 10% formalin to examine histopathology, and the other two pieces were stored at -80°C for interleukin measurement.
  • Example 6-2 Collection of bronchoalveolar lavage fluid
  • Example 7-2 In vitro efficacy of the ALDH-containing composition of the present invention for asthma
  • the RBL-2HE cell line was pre-treated with the ALDH-containing composition of the present invention at each concentration for 20 minutes, and an intracellular inflammatory state was induced by adding IgE and BSA to the cell line.
  • the inhibitory effect of the ALDH-containing composition of the present invention was measured by measuring the expression level of IL-4, a major marker of the asthma induction model, and the results are shown in FIG. 1 . Compared to the IL-4 expression level of the asthma-induced cell line, 13%, 34%, and 35% decrease in the IL-4 expression level was confirmed in the cells treated with the ALDH-containing composition of the present invention for each concentration.
  • the inhibitory effect of the ALDH-containing composition of the present invention on the IL-4 expression level was confirmed, and it was also confirmed that the IL-4 inhibitory effect increased depending on the dosage.
  • interleukin-13 showed up to about 80% reduction in expression at concentrations of 150 mg/Kg and 300 mg/Kg, which are the doses of the ALDH-containing composition of the present invention. (FIG. 2)
  • Example 7-3 IgE reduction effect of the ALDH-containing composition of the present invention in asthma-induced mice
  • the blood in a 1.5 mL tube was centrifuged at 12,000 rpm and 4° C. for 20 minutes, and only the supernatant was separated and used in the experiment for IgE measurement.
  • an IgE ELISA set (555248, BD bioscience, USA) was used, and the analysis method is the same as in Example 5-1.
  • the analysis results are shown in FIG. 3, and the IgE reduction effect was confirmed in the group administered with the ALDH-containing composition of the present invention compared to the asthma-induced mice.
  • mice Female and male ICR mice (7 weeks old) were received and acclimatized for 7 days. During the acclimatization period, general symptoms were observed, and only healthy animals were used for the test. Feed and water were ingested ad libitum, and group separation was performed so that there were 5 males and 5 females in each group based on the average body weight of about 20 g the day before oral administration.
  • the test substance was prepared by dissolving in physiological saline so that the doses of the test animals were 0, 750, 3000, and 5000 mg/Kg, respectively, based on the ALDH-containing composition of the present invention.
  • the standard of administration dose complied with the Korea National Toxicology Program (KNTP) toxicity test manual of the Ministry of Food and Drug Safety, and the maximum applied dose 5000mg/Kg guided by the KNTP manual was applied as the maximum concentration of this experiment.
  • Samples prepared for each group were orally administered once to each test animal, and physiological saline was administered to the normal group (G1).
  • Symptoms were observed at least once a day from the date of acquisition to the day of autopsy for all animals in the test group, and symptoms were observed for 7 days after oral administration. After the type of symptom observation, an autopsy was performed, and changes in each organ were visually observed at the time of autopsy.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • Mycology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Organic Chemistry (AREA)
  • Biotechnology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Microbiology (AREA)
  • Natural Medicines & Medicinal Plants (AREA)
  • Epidemiology (AREA)
  • Dermatology (AREA)
  • Botany (AREA)
  • Zoology (AREA)
  • Genetics & Genomics (AREA)
  • Wood Science & Technology (AREA)
  • Pulmonology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Biomedical Technology (AREA)
  • Virology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Alternative & Traditional Medicine (AREA)
  • Medical Informatics (AREA)
  • Nutrition Science (AREA)
  • Food Science & Technology (AREA)
  • Polymers & Plastics (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)

Abstract

[Abstract] The present invention relates to a food or pharmaceutical composition containing a yeast extract which exhibits an effect of suppressing symptoms of bronchial asthma, allergic asthma, atopic asthma, exercise-induced asthma, cardiac asthma or alveolar asthma. More specifically, the present invention relates to an asthma-suppressing food or pharmaceutical composition containing aldehyde dehydrogenase which is isolated from strains of Saccharomyces cerevisio or accession number KCTC13925BP (Saccharomyces cerevisiae KwonP-1), KCTC14122BP (Saccharomyces cerevisiae KwonP-2), KCTC14123BP (Saccharomyces cerevisiae KwonP-3). [Representative drawing] figure 1

Description

알데히드탈수소 효소를 함유하는 천식 억제용 조성물Composition for Inhibiting Asthma Containing Aldehyde Dehydrogenase
본 발명은 알데히드 탈수소효소를 함유하는 천식 억제 조성물에 관한 것이다. 보다 상세하게는, 본 발명은 싸카로마이세스세레비지오 그리고 기탁번호 KCTC13925BP ( Saccharomyces cerevisiae KwonP-1), KCTC14122BP ( Saccharomyces cerevisiae KwonP-2), KCTC14123BP( Saccharomyces cerevisiae KwonP-3) 균주들로 부터 분리된 알데히드 탈수소 효소를 함유하는 천식 억제용 조성물에 대한 것이다. The present invention relates to an asthma inhibitory composition comprising an aldehyde dehydrogenase. More specifically, the present invention provides Saccharomyces cerevisiae KwonP-3 and Saccharomyces cerevisiae KwonP-3) with accession number KCTC13925BP (Saccharomyces cerevisiae KwonP-1), KCTC14122BP ( Saccharomyces cerevisiae KwonP-2), KCTC14123BP (Saccharomyces cerevisiae KwonP-3) isolated from strains. To a composition for inhibiting asthma containing a dehydrogenase.
천식은 기도에 발생하는 만성 염증으로 인하여 기관지가 과민해지고 간헐적인 기도 수축이 발생하여 호흡곤란 증상을 유발하는 만성질병으로서, 아직까지는 완치방법이 없는 것으로 알려지고 있다. 천식 환자가 알러지 유발 물질을 흡입하면 항체들이 인지하고 면역계를 활성화시켜 염증 반응이 진행되어 기도의 염증과 부종, 기도의 수축을 야기하고 점액이 방출되면서 기관지가 좁아지면 이로 인해 다양한 천식의 증상들이 발현된다.Asthma is a chronic disease that causes respiratory distress symptoms due to bronchial hypersensitivity and intermittent airway constriction due to chronic inflammation in the airways. It is known that there is no cure yet. When an asthma patient inhales an allergen, the antibodies recognize it and activate the immune system, leading to an inflammatory response that causes inflammation, swelling, and constriction of the airways. do.
천식 환자의 Th2 임파구가 활성화되어 IL-4, IL-5, IL-9 또는 IL-13을 증가시키는데 이들 중 IL-4는 점액 생성과 B-세포 증식을 촉진하고, 호흡곤란을 야기한다. 히스타민이나 류코트리엔과 같은 염증 매개 물질을 분비하는 비만세포(mast cell)를 활성화시키는 IgE를 생성하고 폐조직으로 eosinophil을 전달하는 VCAM-1을 발현시키게 된다. 호흡곤란, 기침, 가래, 천명(음) 등 전형적인 천식의 호흡기증상 외에도 비전형적인 증상이 나타나기도 한다. Th2 lymphocytes in asthmatic patients are activated to increase IL-4, IL-5, IL-9 or IL-13. Among them, IL-4 promotes mucus production and B-cell proliferation, and causes dyspnea. It produces IgE, which activates mast cells that secrete inflammatory mediators such as histamine or leukotriene, and expresses VCAM-1, which delivers eosinophils to lung tissue. In addition to the typical respiratory symptoms of asthma such as shortness of breath, cough, sputum, and wheezing (yin), atypical symptoms may also appear.
천식 환자는 심박수 상승, 건성 수포음 등도 나타날 수 있고, 매우 심각한 천식 발작 동안에는 산소부족으로 청색증이 오고, 강한 흉통이 의식을 잃기 직전에 팔다리의 무감각을 느끼거나 손바닥에 땀이 나는 것을 느낄 수 있으며, 일반적인 치료에 반응하지 않는 심각한 천식 발작은 생명을 위협할 수 있으며, 호흡정지와 사망에 이를 수도 있다.Asthma sufferers may also experience elevated heart rate, dry crackling, etc. During very severe asthma attacks, cyanosis due to lack of oxygen, strong chest pain, numbness in limbs or sweating in the palms of the palms may be felt just before loss of consciousness, which is common Severe asthma attacks that do not respond to treatment can be life-threatening, leading to respiratory arrest and death.
이러한 기관지 천식은 우리나라를 포함하여 세계적으로 대다수의 국가들에서 전체 인구 중 약 5 내지 10%의 인구가 앓고 있는 것으로 보고될 만큼 매우 흔한 질병으로서, 최근 들어 대기등 환경오염의 속도가 빨라짐에 따라 그 유병률이 세계적으로 더욱 증가되고 있다. This bronchial asthma is a very common disease that is reported to be suffering from about 5 to 10% of the total population in most countries around the world, including Korea. The prevalence is increasing worldwide.
미국 국립 심폐혈관연구소와 유럽 호흡기 학회에 의하면, 천식에 관한 연간 비용은 미국에서 161억 달러, EU에서 163억 달러로 예측되며, 천식질환의 특성상 장기간 약물복용이 불가피하지만, 종래의 스테로이드제나 평활근 이완제와 같은 천식치료제들은 면역능 저하, 감염, 심장에 대한 작용 등의 많은 부작용을 나타낸다. 천식의 치료에 사용되는 경구용 스테로이드제는 전신에 흡수되므로 혈압 상승, 골다공증, 위궤양 등과 같은 부작용이 생길 수 있다. 호흡기 흡입용 스테로이드제 역시 목이 쉬는 부작용이 있다.According to the US National Institute of Cardiopulmonary Vascular Research and the European Respiratory Society, the annual cost of asthma is estimated to be 16.1 billion dollars in the US and 16.3 billion dollars in the EU. Asthma drugs, such as, show many side effects, such as decreased immunity, infection, and action on the heart. Oral steroids used for the treatment of asthma are absorbed systemically, so side effects such as elevated blood pressure, osteoporosis, and gastric ulcer may occur. Respiratory inhaled steroids also have the side effect of choking.
따라서 부작용이 작고, 근원적 치료 효과를 갖는 새로운 천식증 치료 약물이나 식품 조성물의 개발이 시급히 요구되고, 특히 종래의 천식증에 사용되는 스테로이드계 약물이 갖는 부작용을 해소할 수 있는 천식치료제 개발이 요구되고 있다.Therefore, there is an urgent need to develop a new asthma treatment drug or food composition with small side effects and a fundamental therapeutic effect. In particular, there is a need to develop an asthma treatment agent that can solve the side effects of steroid drugs used in conventional asthma. have.
우리나라의 천식 및 만성 폐쇄성 기도 질환으로 인한 입원률 및 평균 입원기간이 다른 OECD 국가들에 비해 2배 이상 높은 것으로 나타났고, 2003년 천식의 사회적 비용과 환자의 생활에 미치는 영향 조사 결과, 국내 인구 10만명당 천식 환자수는 4.19%로 이에 따른 직접 비용은 9,620억원, 간접비용을 포함한 사회적 비용은 2조 484억 원(국내총생산 (GDP)의 0.26%)에 달하였다. The hospitalization rate and average length of hospitalization for asthma and chronic obstructive airway disease in Korea are more than double that of other OECD countries. Asthma patients accounted for 4.19%, resulting in a direct cost of 962 billion won and social costs including indirect costs of 2.484 trillion won (0.26% of gross domestic product).
이러한 기관지 천식 발작을 일으키는 요인들을 외부적인 요인과 내부적인 요인이 있다. 외부적인 요인에는 대기 중의 오염물질, 각종 알러젠 및 산업 분진이 포함된다. 이들 천식 유발 원인 물질은 면역 반응에서 IgE를 생성시키고, 생성된 IgE는 코 점막, 피부 및 기도를 막고 기관지내의 비만세포 (mast cell) 표면의 수용체에 결합하여 염증을 일으킨다. There are external factors and internal factors that cause these bronchial asthma attacks. External factors include airborne pollutants, various allergens and industrial dust. These asthma-inducing substances produce IgE in an immune response, and the produced IgE blocks the nasal mucosa, skin, and airways, and binds to receptors on the surface of mast cells in the bronchi to cause inflammation.
천식의 발병 여부는 기도과민성, 혈장 내의 많은 IgE 양, 그리고 폐와 기도로 유입되는 면역세포의 수 증가로 알 수있으며, 이러한 특징들은 모두 Th2 세포 분화와 관련되어 있다. 또한 조절 T 세포(regulatory T cells, 이하'Treg 세포'라 함)는 다른 종류의 T 세포의 분화를 억제하여 기도 염증을 감소시키고, 기도 관용을 유발하는 것으로 정의된다.The onset of asthma can be known by airway hypersensitivity, large amount of IgE in plasma, and increase in the number of immune cells flowing into the lungs and airways, all of which are related to Th2 cell differentiation. In addition, regulatory T cells (hereinafter referred to as 'Treg cells') are defined as suppressing the differentiation of other types of T cells to reduce airway inflammation and induce airway tolerance.
천식증 요인으로는 유전적인 요인도 있다. 사람의 5 번째 염색체, β2-아드레노리셉터 유전자, 사람의 11, 12, 14, 16 번째 염색체가 관여하며, 이들 유전자 내의 변화가 IgE의 증가와 기관지의 과민반응을 일으키는 것으로 보고되고 있다.Asthma also has genetic factors. Human chromosome 5, β2-adrenoceptor gene, and human chromosome 11, 12, 14, and 16 are involved, and it is reported that changes in these genes cause an increase in IgE and hypersensitivity of the bronchial tubes.
기관지 천식 유발인자에 의해 비 점막 또는 기관지 점막이 자극되면, 과도한 면역 반응(알레르기 반응)을 보이고, 기도 점막조직에 염증이 발생하며, 이에 따라 기침, 호기성 천명음, 및 호흡곤란의 3대 기관지 천식 증상이 발생하고, 기관지 천식과 콧물, 재채기, 또는 코 막힘과 같은 비염 증상도 발생하게 된다.When the nasal mucosa or bronchial mucosa is stimulated by bronchial asthma triggers, an excessive immune response (allergic reaction) occurs and inflammation occurs in the airway mucosal tissue, and accordingly, the three major symptoms of bronchial asthma: cough, aerobic wheezing, and dyspnea. This causes bronchial asthma and rhinitis symptoms such as runny nose, sneezing, or stuffy nose.
한편, 세포의 대사과정에서 발생하는 활성산소종(reactive oxygen species, ROS)은 과도하게 생성될 경우, 면역계통의 이상을 유발한다. 활성산소종은 자가면역질환의 발병 원인 중 하나로서 인터루킨-4(IL-4)와 인터루킨-13(IL-13)의 발현을 증가시키고 이로 인한 산화적 스트레스가 폐에서 평활근의 수축, 혈관의 투과성 증가, 약물 반응 수용체의 손상, 점액 분비의 증가, 염증성 매개자의 분비, 기도 상피 세포의 손상 등의 작용을 하여, 천식을 포함한 다양한 폐 손상 질환의 병인에 있어 중요한 역할을 한다.On the other hand, when reactive oxygen species (ROS) generated in the metabolic process of cells are excessively generated, it causes abnormalities in the immune system. Reactive oxygen species is one of the causes of autoimmune diseases, and increases the expression of interleukin-4 (IL-4) and interleukin-13 (IL-13), and the resulting oxidative stress causes contraction of smooth muscle in the lung and permeability of blood vessels. It plays an important role in the pathogenesis of various lung injury diseases, including asthma, through actions such as increase, damage to drug-responsive receptors, increase in mucus secretion, secretion of inflammatory mediators, and damage to airway epithelial cells.
기관지 천식의 치료 방법으로는 기관지 천식의 원인물질 즉 알러젠을 제거하거나 회피하는 회피요법과 탈감작 용법, 약제를 선택하는 약물요법, 또는 소량의 알레르기 항원(allergen)을 주사하여 면역시키는 면역요법 등이 있다. Treatment methods for bronchial asthma include avoidance and desensitization to remove or avoid allergens that cause bronchial asthma, drug therapy to select drugs, or immunotherapy to immunize by injecting a small amount of an allergen. .
상기 약물요법에 사용되는 기관지 천식 치료제로는 수축된 기도를 넓혀 증상을 호전시키는 β2-효능제로서의 기관지 확장제, 증상완화를 위한 코르티코스테로이드 계열의 흡입제, 또는 항알레르기 제제 등이 있다. 그밖에 Th2 형태의 사이토카인의 작용을 억제하는 기전을 이용한 수용성 IL-4 수용체, 수용성 IL-13 및 항 IL-5 항체 등의 천식치료제가 일부 개발 판매중이다.As a treatment for bronchial asthma used in the drug therapy, there are a bronchodilator as a β2-agonist that improves symptoms by widening a constricted airway, a corticosteroid-based inhalant for symptom relief, or an anti-allergic agent. In addition, some asthma treatment drugs such as water-soluble IL-4 receptor, water-soluble IL-13 and anti-IL-5 antibody using a mechanism to inhibit the action of Th2-type cytokines are being developed and sold.
그런데, 많은 천식 치료제가 의사의 진료와 처방을 통해서만 구매가 가능하며, 그나마 대부분 스테로이드(Steroid)계 약물이기 때문에 장기간 사용 시 부작용이 초래될 수 있다는 문제점이 있다.However, many asthma medications can be purchased only through a doctor's consultation and prescription, and since most of them are steroid-based drugs, there is a problem that side effects may occur when used for a long period of time.
그러나, 현재까지 활성산소종의 조절을 통해 폐 손상 질환을 치료하고자 하는 많은 시도가 이루어져 왔음에도 불구하고, 활성산소종 조절 수준에 비해 그 효과가 기대에 미치지 못하거나 생체 내 실험(in vivo)에서의 효과가 일정하지 않았다. However, even though many attempts have been made to treat lung injury diseases through the regulation of reactive oxygen species, the effect does not meet expectations compared to the level of reactive oxygen species control, or in vivo experiments. effect was not consistent.
지금까지 시도되었던 대부분의 활성산소종 저해 방식은 NADPH oxidase에 의한 활성산소종의 생성을 표적으로 하였고, 미토콘드리아에서의 활성산소종 생성의 조절은 이루어지지 않았으므로, 이는 미토콘드리아의 활성산소종 생성을 조절하지 못하였기 때문이라는 가설이 대두되었다.Most of the methods of inhibiting reactive oxygen species that have been attempted so far targeted the generation of reactive oxygen species by NADPH oxidase. It was hypothesized that it could not be done.
이에, 본 발명자들은 생체내의 부작용 및 안전성 등을 고려하여 새로운 천식 치료제를 개발하기 위하여 연구하던 중, 효모로부터 추출된 알데히드탈수소효소(이하 ALDH하고 약칭함)가 세포 침윤을 억제하고 인터루킨-4(interleukin-4, IL-4)와 인터루킨-13(IL-13)의 발현을 억제하므로 천식의 예방 또는 치료에 유용하게 사용될 수 있음을 확인하고 본 발명을 완성하게 되었다.Therefore, while the present inventors were studying to develop a new asthma treatment in consideration of side effects and safety in vivo, aldehyde dehydrogenase (hereinafter abbreviated as ALDH) extracted from yeast inhibits cell infiltration and interleukin-4 (interleukin-4). -4, IL-4) and interleukin-13 (IL-13), it was confirmed that it can be usefully used for the prevention or treatment of asthma, and thus the present invention has been completed.
T세포에서 분비되는 인터류킨4(이하, IL-4로 약칭함)는 129개의 아미노산으로 이루어지는 약20kD의 사이토카인으로서, B-세포 분화 인자 그리고 B세포 증식 인자, 특히 IgE를 증가시키는 중요한 사이토카인이다. IL-4는 최초로 호염구에서 분비되고 활성화된 T-세포(TH2)에서 추가적으로 분비된다. B세포 활성화,IgE 종류변환, Th2분화유도에 기여하는 것으로 보고되었다. Interleukin 4 (hereinafter, abbreviated as IL-4) secreted from T cells is a cytokine of about 20 kD consisting of 129 amino acids, and is an important cytokine that increases B-cell differentiation factors and B-cell proliferation factors, especially IgE. . IL-4 is initially secreted by basophils and additionally secreted by activated T-cells (TH2). It has been reported to contribute to B cell activation, IgE type conversion, and induction of Th2 differentiation.
Th2 세포에 의한 IL-4의 생산은 IL-4에 의해 빨라지기도 하며, IL-4의 수용체는 두개의 시스테인이 결합하는 130kD의 단백질이고, IL-4Rα+γc형과 IL-4Rα+IL-13Rα이 존재한다.IL-4는 활성화된 B세포와 T세포를 자극하거나, B세포를 형질세포로 분화시키는 등 많은 생물학적 기능을 갖고 있으며, 체액성 면역과 후천성 면역의 중요한 조절인자이다. IL-4 production by Th2 cells is also accelerated by IL-4, and the receptor for IL-4 is a 130 kD protein that binds two cysteines, IL-4Rα+γb and IL-4Rα+IL-13Rα. IL-4 has many biological functions, such as stimulating activated B cells and T cells and differentiating B cells into plasma cells, and is an important regulator of humoral immunity and adaptive immunity.
또한 IL-4는 B 전구세포에서 IgE 및 IgG4 생산이 가능한 B세포로 전환되는 종류변환(class swithching)을 유도할 뿐만 아니라, MHC클래스 II의 생산을 촉진한다고 알려져 있다.IL-4의 과량 분비는 IgE의 증가를 야기하여 즉시 발현되는 과민 알러지 반응을 일으킨다. IL-4는 IL-13과 상승 작용을 하고 IgE 또는 호산구 매개성 염증 반응의 원인 물질이다.In addition, IL-4 is known to induce class switching from B progenitor cells to B cells capable of producing IgE and IgG4, as well as promote the production of MHC class II. It causes an increase in IgE, resulting in an immediate onset hypersensitivity allergic reaction. IL-4 acts synergistically with IL-13 and is the causative agent of IgE- or eosinophil-mediated inflammatory responses.
따라서 IL-4 저해제를 여러가지 면역질환 치료제로 개발하는 다양한 시도가 이루어지고 있으며, 특히 아토피, 천식, 두드러기 치료제로의 개발이 다양하게 시도되고 있다. Therefore, various attempts are being made to develop IL-4 inhibitors as therapeutic agents for various immune diseases, and in particular, various attempts are being made to develop them as therapeutic agents for atopy, asthma, and urticaria.
항원에 노출되어 IgE 항체가 만들어지면 IgE는 비만세포 또는 호염구 표면에 결합하고, IgE가 결합된 비만세포가 동일한 항원에 2차 노출되면, 비만세포나 호염구내 과립의 탈과립 현상이 일어나면서 혈관 활성물질인 히스타민, 세로토닌, 호산구 또는 호중구의 화학주성인자, 류코트리엔, 프로스타글란딘이 방출된다. When an IgE antibody is produced by exposure to an antigen, IgE binds to the surface of mast cells or basophils, and when the IgE-bound mast cells are exposed to the same antigen a second time, degranulation of the mast cells or basophil intragranule occurs and vasoactive substances Phosphorus histamine, serotonin, chemotactic factors of eosinophils or neutrophils, leukotrienes, and prostaglandins are released.
본 발명의 ALDH 함유 조성물은 IL-4 또는 IL-13 저해를 통하여 IgE증가를 억제함으로써 부작용이 작은 천식치료제가 될 수 있다. 인터류킨13은 아미노산 122개로 이루어진 단백질로서 IL-13의 수용체는 CD132(γc)이다. IL-13은 IL-4와 기능적으로 유사하고 Th2세포, 수지상 세포, 비만세포에서 분비되며, Th2 세포의 생성 및 IgE 생성을 억저해하여 B세포 증식분화, Th1을 제어하고, 알레르기, 천식 증상을 완화시킨다.The ALDH-containing composition of the present invention can be an asthma treatment with small side effects by inhibiting IgE increase through IL-4 or IL-13 inhibition. Interleukin 13 is a protein consisting of 122 amino acids, and the receptor for IL-13 is CD132 (γc). IL-13 is functionally similar to IL-4 and is secreted from Th2 cells, dendritic cells, and mast cells. alleviate
IL-13에 대한 수용체는 IL-13Rα와 IL-4Rα로 구성되므로 IL-13수용체는 IL-4에도 반응한다. Il-13에 대한 수용체는 사람의 B세포에서는 발현되지만, 생쥐의 B세포에서는 발현되지 아니한다. IL-13은 모노사이트를 수지상 세포로 분화시키도 한다. 그런데 IL-13 유전자 불활성 생쥐에서는 Th2 세포 또는 IgE를 활발하게 생산하지 못하는 것으로 보고되고 있다. Since the receptor for IL-13 is composed of IL-13Rα and IL-4Rα, the IL-13 receptor also responds to IL-4. The receptor for Il-13 is expressed in human B cells, but not in mouse B cells. IL-13 also differentiates monocytes into dendritic cells. However, it has been reported that IL-13 gene inactive mice do not actively produce Th2 cells or IgE.
따라서, IL-13 생성을 저해하거나 작용을 차단하면 IgE의 생성이 억제되므로, IL-13 저해제나 IL-13 신호전달 차단제는 아토피나 천식 등의 면역질환 치료제로도 사용될 수 있다.Therefore, since the production of IgE is inhibited when IL-13 production is inhibited or its action is blocked, IL-13 inhibitors or IL-13 signaling blockers can also be used as therapeutic agents for immune diseases such as atopy or asthma.
한편, 알데히드탈수소 반응은 아세트알데히드나 인체 대사 산물에 존재하는 다양한 알데히드를 노폐물로 배출될 수 있는 유기산으로 변화시키는 반응이다. 이러한 알데히드탈수소 반응을 촉진하는 효소는 세포에 대한 독성을 나타내는 알데히드를 분해하여 세포와 조직에 가해지는 카보닐 스트레스를 완화시킨다.On the other hand, the aldehyde dehydrogenation reaction is a reaction for changing acetaldehyde or various aldehydes present in human metabolites into organic acids that can be discharged as waste products. The enzyme that promotes this aldehyde dehydrogenation reaction decomposes the aldehyde, which is toxic to cells, and relieves carbonyl stress applied to cells and tissues.
알데히드탈수소 반응에 개입하는 효소의 작용기를 함유하는 일련의 단백질들을 알데히드탈수소 효소라고 지칭하며 현재까지 약20여종의 ALDH가 알려져있다. 따라서 본명세서에서 ALDH라는 용어의 범위에는 현재까지 알려진 20여종의 ALDH 전부와 알데히드탈수소 반응에 개입하는 효소의 탈수소 반응 작용기를 전부 또는 일부분을 포함하는 모든 융합단백질, 그리고 알데히드탈수소 반응에 개입하는 효소의 탈수소 반응 작용기의 일부분 또는 전부를 포함하는 모든 재조합 단백질을 모두 포함한다.A series of proteins containing functional groups of enzymes involved in aldehyde dehydrogenation are called aldehyde dehydrogenases, and about 20 types of ALDH are known so far. Therefore, in the present specification, the scope of the term ALDH includes all of the 20 known types of ALDH, all fusion proteins containing all or part of the dehydrogenation functional groups of enzymes involved in aldehyde dehydrogenation, and enzymes involved in aldehyde dehydrogenation. All recombinant proteins containing some or all of the dehydrogenation functional groups are included.
한편, 국제공개번호 WO 2014/160185로 공개된 더 보드 오브 트러스티스 오브 더 리랜드 스탠포드 쥬니어 유니버시티의 국제출원번호PCT/US2014/025993에는 미토콘드리아 알데히드 탈수소효소-2 (ALDH2) 활성의 조절인자로서 기능하는 화합물들, 및 이러한 화합물들의 제조 방법들이 공개되어 있다. On the other hand, in International Application No. PCT/US2014/025993 of The Board of Trusts of the Leland Stanford Junior University, published as International Publication No. WO 2014/160185, mitochondrial aldehyde dehydrogenase-2 (ALDH2) functions as a regulator of activity. Compounds, and methods of making such compounds, are disclosed.
이건 선행기술에서는 미토콘드리아 알데히드 탈수소효소-2 (ALDH2)을 상세하게 기재하고 있으며, 핵 게놈에서 인코딩되어 미토콘드리아 내부로 이동하는 4개의 동일한 서브유닛으로 이루어지는 사합체 단백질이며, 각각은 500개의 아미노산 잔기들로 구성되고, 이러한 사합체는 이합체의 이합체로서, 이합체를 형성하는 단량체들 사이의 경계면은 사합체를 형성하는 2개의 이합체들 사이의 경계면과 상이하며, 각각의 서브유닛은 3개의 주요 부위, 즉 알데히드 탈수소반응 촉매 부위, 조효소 또는 NAD+-결합 부위, 및 올리고머화 부위를 공개하고 있다.This prior art describes in detail mitochondrial aldehyde dehydrogenase-2 (ALDH2), a tetrameric protein consisting of four identical subunits encoded in the nuclear genome and transported into the mitochondria, each of 500 amino acid residues. wherein the tetramer is a dimer of a dimer, wherein the interface between the monomers forming the dimer is different from the interface between the two dimers forming the tetramer, and each subunit has three major sites, i.e., an aldehyde Dehydrogenation catalytic sites, coenzyme or NAD+-binding sites, and oligomerization sites are disclosed.
이건 선행기술은 ALDH2와 관련된 질병 및 상태들에는 허혈성 스트레스, 만성 자유-라디칼 관련 질병들, 급성 자유-라디칼 관련질병들, 니트로글리세린에 대한 무감응성 (예컨대, 협심증 및 심장 마비에서), 고혈압, 당뇨병, 골다공증, 암, 판코니 빈혈, 알츠하이머 병, 파킨슨병, 알코올의존증(alcoholism), 알코올 불내성, 알코올 탐닉(alcoholaddiction), 알코올 남용 질환, 알코올도취(alcohol intoxication), 알코올 의존, 알코올 중독(alcohol poisoning),알코올 소비증상, 및 마약 중독이 포함하여, ALDH2의 조절인자, ALDH2의 작용제, 또는 ALDH2의 길항제를 개시한다. This prior art states that ALDH2 related diseases and conditions include ischemic stress, chronic free-radical related diseases, acute free-radical related diseases, insensitivity to nitroglycerin (eg, in angina and heart failure), hypertension, diabetes. , osteoporosis, cancer, Fanconi anemia, Alzheimer's disease, Parkinson's disease, alcoholism, alcohol intolerance, alcohol addiction, alcohol abuse disease, alcohol intoxication, alcohol dependence, alcohol poisoning Disclosed are modulators of ALDH2, agonists of ALDH2, or antagonists of ALDH2, including symptoms of alcohol consumption, and drug addiction.
또한 ALDH2의 작용제는 ALDH2의 알데히드 기질을 발생시키는 화합물을 섭취, 흡수 또는 흡입시 피험체에서 ALDH2의알데히드 기질을 발생시키는 화합물의 수준을 감소시키기에 유용하다. ALDH2 길항제는 표준 암 치료에 대한 보조제로서 사용된다. 또한 ALDH2의 길항제는 알코올의존증 및 마약 중독을 치료 또는 억제하는데 유용하다는 사실을 공개하고 있다. 그러나, 이건 선행기술은 본원 발명의 요지인 ALDH가인터류킨-4 또는 인터류킨-13의 작용을 저해하거나, 할 수도 있다는 가능성에 대한 어떠한 암시나 언급도 하지 아니하고 있다.An agonist of ALDH2 is also useful for reducing the level of a compound that generates an aldehyde substrate of ALDH2 in a subject upon ingestion, absorption, or inhalation of a compound that generates an aldehyde substrate of ALDH2. ALDH2 antagonists are used as adjuncts to standard cancer treatment. It has also been disclosed that antagonists of ALDH2 are useful in treating or suppressing alcoholism and drug addiction. However, this prior art does not make any suggestion or mention about the possibility that ALDH, which is the subject of the present invention, may inhibit the action of interleukin-4 or interleukin-13.
한편, 국제공개번호 WO 2015/127137로 공개된 아비브 테라퓨틱스, 인크.의 국제출원번호 PCT/US2015/016703호는 암의 치료를 위한 폴리사이클릭 아미드와 결합하는 미토콘드리아 알데히드 탈수소효소 2(ALDH2) 및 그의 용도를 개시하고 있다. 이건 선행 기술은 미토콘드리아 알데히드 탈수소효소-2(ALDH2)와 결합하는 식 I의 화합물 및 즉, 암, 판코니 빈혈및 알코올-관련 장애를 갖는 환자를 치료하기 위한 상기 화합물의 사용 방법을 제공한다. 그러나, 이건 선행기술 역시 본원 발명의 요지인 ALDH가 인터류킨-4 또는 인터류킨-13의 작용을 저해하여 천식 증상을 억제할 수 있다는 가능성에 대한 어떠한 암시나 언급도 하지 아니하고 있다.Meanwhile, International Application No. PCT/US2015/016703 of Aviv Therapeutics, Inc., published as International Publication No. WO 2015/127137, relates to mitochondrial aldehyde dehydrogenase 2 (ALDH2) binding to polycyclic amide for the treatment of cancer. and uses thereof. This prior art provides compounds of formula I that bind mitochondrial aldehyde dehydrogenase-2 (ALDH2) and methods of using said compounds to treat patients with ie cancer, Fanconi's anemia and alcohol-related disorders. However, this prior art also does not suggest or mention the possibility that ALDH, which is the subject of the present invention, can suppress the asthma symptoms by inhibiting the action of interleukin-4 or interleukin-13.
한국특허출원 10-2020-0019858에서는 본 발명의 천식 억제용 약제 조성물과 식품 조성물의 유효성분으로 사용되는, 알데히드탈수소 효소(이하 ALDH라고 약칭함)와 글루타치온을 동시에 생산하는 싸카로마이세스 쎄레비지애 균주 대하여 상세하게 기재하고 있다.Korean Patent Application No. 10-2020-0019858 describes Saccharomyces cerevisiae that simultaneously produces aldehyde dehydrogenase (hereinafter abbreviated as ALDH) and glutathione, which are used as active ingredients in the pharmaceutical composition and food composition for inhibiting asthma of the present invention. The strain is described in detail.
1. Pavord ID, Korn S, Howarth P, et al. Mepolizumab for severe eosinophilic asthma (DREAM): a multicentre, double-blind, placebo-controlled trial. Lancet 2012;380:651- 659.1. Pavord ID, Korn S, Howarth P, et al. Mepolizumab for severe eosinophilic asthma (DREAM): a multicentre, double-blind, placebo-controlled trial. Lancet 2012;380:651-659.
2. Pocket guide for asthma management and prevention : Global Initiative for Asthma 2019.2. Pocket guide for asthma management and prevention: Global Initiative for Asthma 2019.
3. Chunhu Gu, Yuan Xing, [...], and Heng Ma. Impaired Cardiac SIRT1 Activity by Carbonyl Stress Contributes to Aging-Related Ischemic Intolerance. PloS One 2013: 8(9)3. Chunhu Gu, Yuan Xing, [...], and Heng Ma. Impaired Cardiac SIRT1 Activity by Carbonyl Stress Contributes to Aging-Related Ischemic Intolerance. PloS One 2013: 8(9)
4. Yancey SW, Ortega HG, Keene ON, et al. Meta-analysis of asthma-related hospitalization in mepolizumab studies of severe eosinophilic asthma. J Allergy Clin Immunol 2017; 139:1167-1175.e2.4. Yancey SW, Ortega HG, Keene ON, et al. Meta-analysis of asthma-related hospitalization in mepolizumab studies of severe eosinophilic asthma. J Allergy Clin Immunol 2017; 139:1167-1175.e2.
5. Ortega HG, Liu MC, Pavord ID, et al. Mepolizumab treatment in patients with severe eosinophilic asthma. N Engl J Med 2014; 371:1198-1207. 5. Ortega HG, Liu MC, Pavord ID, et al. Mepolizumab treatment in patients with severe eosinophilic asthma. N Engl J Med 2014; 371:1198-1207.
본 발명의 기본적인 목적은 천식 증세를 완화하고 억제하는 효과를 나타내는 알데히드 탈수소 효소를 함유하는 식품 조성물을 제공하는 것이다.A basic object of the present invention is to provide a food composition containing an aldehyde dehydrogenase having an effect of alleviating and suppressing asthma symptoms.
본 발명의 또 다른 목적은 천식 증세를 예방하고 치료하는 효과를 나타내는 알데히드 탈수소 효소 또는 알데히드탈수소반응에 간여하는 작용 부위를 포함하는 융합단백질 또는 재조합단백질을 유효성분으로 함유하는 천식, 아토피성 비염 치료제 조성물을 제공하는 것이다. Another object of the present invention is a composition for treating asthma and atopic rhinitis containing, as an active ingredient, a fusion protein or a recombinant protein comprising an aldehyde dehydrogenase having an effect of preventing and treating asthma symptoms or an action site involved in an aldehyde dehydrogenation reaction. is to provide
이상과 같은 본 발명의 목적들은, 싸카로마이세스 세레비지에 효모, 기탁번호 KCTC13925BP( Saccharomyces cerevisiae KwonP-1), KCTC14122BP ( Saccharomyces cerevisiae KwonP-2), KCTC14123BP( Saccharomyces cerevisiae KwonP-3)에서 분리되는 알데히드 탈수소 효소를 함유하는 천식 예방 또는 완화 효과를 나타내는 조성물을 제공함으로써 달성된다.The objects of the present invention as described above, Saccharomyces cerevisiae yeast, accession number KCTC13925BP (Saccharomyces cerevisiae KwonP-1), KCTC14122BP ( Saccharomyces cerevisiae KwonP-2), KCTC14123BP ( Saccharomyces cerevisiae KwonP-3) aldehyde isolated from It is achieved by providing a composition which contains a dehydrogenase and exhibits an asthma prevention or alleviation effect.
본 발명자들이 천식 억제용 ALDH 생산을 위하여 사용한 효모는, 맥주나 빵을 발효시키기 위하여 일반적으로 널리 사용되고 있는 싸카로마이세스 세레비지에 효모이다. 현재까지 알려진 싸카로마이세스 세레비지에 효모는 250여종이 보고되고 있고, 이들은 모두 본 발명의 ALDH 생산에 적절하게 사용될 수 있다. The yeast used by the present inventors for the production of ALDH for asthma suppression is Saccharomyces cerevisiae yeast, which is generally widely used for fermenting beer or bread. About 250 types of Saccharomyces cerevisiae yeast known to date have been reported, and all of them can be appropriately used for the ALDH production of the present invention.
보다 바람직하게는, 본 발명의 천식 억제 조성물로는, 본 발명자들이 국제기탁기관 KCTC에 기탁한 신규한 싸카로마이세스 세레비지에 효모인, 기탁번호 KCTC13925BP( Saccharomyces cerevisiae KwonP-1), KCTC14122BP ( Saccharomyces cerevisiae KwonP-2) 또는 KCTC14123BP( Saccharomyces cerevisiae KwonP-3)가 본 발명의 천식 억제용 ALDH함유 조성물의 생산 원료로 사용될 수 있다. More preferably, the asthma inhibitory composition of the present invention is a novel Saccharomyces cerevisiae yeast deposited by the present inventors with the international depository KCTC, deposit number KCTC13925BP (Saccharomyces cerevisiae KwonP-1), KCTC14122BP ( Saccharomyces cerevisiae KwonP-1) cerevisiae KwonP-2) or KCTC14123BP ( Saccharomyces cerevisiae KwonP-3) may be used as a raw material for the production of the ALDH-containing composition for inhibiting asthma of the present invention.
본 발명에 따른 ALDH 함유 조성물들은, 기관지 염증, 알레르기성 비염, 천식 등과 같은 산화 스트레스에 의한 염증반응의 치료 및 예방에 효과적이다. 특히, ALDH는 쉽게 구입할 수 있는 식품에도 함유되어 있을 뿐만 아니라, 식용 효모에서 추출한 것이므로 인체에 대한 부작용이 작다는 장점이 있다.The ALDH-containing compositions according to the present invention are effective in treating and preventing inflammatory reactions caused by oxidative stress, such as bronchial inflammation, allergic rhinitis, and asthma. In particular, ALDH is not only contained in easily purchased foods, but also has the advantage of having little side effects on the human body because it is extracted from edible yeast.
본 발명에 따른 싸카로마이세스 세레비지에 효모 추출물은 생체 내 실험(in vivo)에서 천식 유발 마우스의 기관지 폐포 세척액에서 면역글로불린 E (IgE) 및 전체 염증 세포수 및 림파구, 단핵구, 호중구, 호산구의 증가를 억제시키는 활성, 기도 점액분비를 억제하는 활성을 가지고, Th2 사이토카인인 IL-4와 IL-13 및 기도 염증 정도를 감소시킨다. The yeast extract of Saccharomyces cerevisiae according to the present invention can be used for immunoglobulin E (IgE) and total number of inflammatory cells and lymphocytes, monocytes, neutrophils, and eosinophils in the bronchoalveolar lavage fluid of asthma-induced mice in an in vivo experiment. It has the activity of inhibiting the increase and the secretion of airway mucus, and reduces the Th2 cytokines IL-4 and IL-13 and the degree of airway inflammation.
본 발명의 조성물은, 기관지성 천식, 알러지성 천식, 아토피형 천식, 비아토피형 천식, 운동유발 천식, 아스피린 천식, 심인성 천식 또는 폐포성 천식 예방 또는 치료하는 효과를 나타낸다. The composition of the present invention exhibits an effect of preventing or treating bronchial asthma, allergic asthma, atopic asthma, non-topic asthma, exercise-induced asthma, aspirin asthma, psychogenic asthma, or alveolar asthma.
본 발명의 조성물에 함유된 ALDH는 기도과민성 경감, 기도 내 활성 산소 발생량을 억제 또는 기관지 내 염증세포 침윤 억제함으로써 기관지 내 염증세포의 수 감소, 혈청 내 IgE 항체의 농도 감소 또는 Th2 싸이토카인인 IL-4와 IL-13의 발현을 억제하는 효과를 나타내고, 과민성 호흡기 증상 조절의 기전에서 Apoptosis와 관련된 protein(JNK, ERK, p38 , BCL-2 family, caspase-3, NF-κB) 의 발현을 억제하는 효과를 나타낸다. 따라서, 본 발명의 ALDH함유 조성물들은 기관지 천식의 예방, 개선 또는 치료에 유용한 의약품 그리고 건강식품으로 사용될 수 있다. ALDH contained in the composition of the present invention reduces airway hypersensitivity, suppresses the amount of active oxygen in the airways, or inhibits the infiltration of inflammatory cells in the bronchial tubes, thereby reducing the number of inflammatory cells in the bronchus, reducing the concentration of IgE antibody in the serum, or IL-4, a Th2 cytokine and IL-13, and the effect of inhibiting the expression of apoptosis-related proteins (JNK, ERK, p38, BCL-2 family, caspase-3, NF-κB) in the mechanism of regulating hypersensitive respiratory symptoms indicates Accordingly, the ALDH-containing compositions of the present invention can be used as pharmaceuticals and health foods useful for the prevention, improvement or treatment of bronchial asthma.
도 1은 본 발명의 ALDH함유 조성물에 의한 RBL-2HE 세포주에서의 인터류킨-4 증가 억제 효과를 보여주는 그래프이다. 1 is a graph showing the inhibitory effect of interleukin-4 increase in RBL-2HE cell line by the ALDH-containing composition of the present invention.
도 2는 본 발명의 ALDH함유 조성물에 의한 대조군과 실험군 마우스 혈청에서 인터류킨-13 억제 효과를 보여주는 그래프이다.2 is a graph showing the inhibitory effect of interleukin-13 in the sera of control and experimental mice of the ALDH-containing composition of the present invention.
도 3은 본 발명의 ALDH함유 조성물에 의한 대조군과 실험군 마우스 혈청에서 IgE 감소 효과를 보여주는 그래프이다.3 is a graph showing the IgE reduction effect of the ALDH-containing composition of the present invention in the sera of control and experimental mice.
도 4는 본 발명의 ALDH함유 조성물에 의한 대조군 및 실험군 마우스에서의 림파구 수의 변화를 관찰한 결과이다. 4 is a result of observing the change in the number of lymphocytes in the mice of the control group and the experimental group by the ALDH-containing composition of the present invention.
도 5는 본 발명의 ALDH함유 조성물에 의한 대조군 및 실험군 마우스에서의 호산성 백혈구의 변화를 관찰한 결과이다. 5 is a result of observing changes in eosinophilic leukocytes in control and experimental mice by the ALDH-containing composition of the present invention.
도 6은 본 발명의 ALDH함유 조성물에 의한 대조군 및 실험군 마우스에서의 인터류킨-4의 억제를 보여주는 결과이다.6 is a result showing the inhibition of interleukin-4 in control and experimental mice by the ALDH-containing composition of the present invention.
이하, 실시예들을 통하여 본 발명을 더욱 상세히 설명하고자 한다. 이들 실시예는 오로지 본 발명을 예시하기 위한 것으로서, 본 발명의 보호범위가 이들 실시예에 의해 제한되는 것으로 해석되지 아니하는 것은 당업계에서 통상의 지식을 가진 자에게 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 protection scope of the present invention is not to be construed as being limited by these examples.
본 발명의 ALDH함유 약학 조성물 또는 식품 조성물은, 야생 싸카로마이세스 효모, 싸카로마이세스 세레비지애 KwonP1(KCTC13925BP), 싸카로마이세스 세레비지애 KwonP2(KCTC14122BP) 또는 싸카로마이세스 세레비지애 KwonP3(KCTC14123BP)의 건조 분말, 용해분말(lysate) 또는 ALDH함유 추출분말을 유효성분으로서 포함한다.ALDH-containing pharmaceutical composition or food composition of the present invention, wild Saccharomyces yeast, Saccharomyces cerevisiae KwonP1 (KCTC13925BP), Saccharomyces cerevisiae KwonP2 (KCTC14122BP) or Saccharomyces cerevisiae Dry powder, lysate, or ALDH-containing extract powder of KwonP3 (KCTC14123BP) is included as an active ingredient.
본 발명의 조성물에 유효성분으로서 함유되는, 사카로마이세스 세래비지애 균주 건조 분말, 용해분말(lasate) ALDH함유 추출분말의 제조에 사용되는 균주들의 제조 방법은, 본 발명의 명세서에서 인용하는 한국 특허 출원 제10-2020-00198858호의 명세서에 상세하게 기재되어 있다.Saccharomyces cerevisiae strain dry powder contained as an active ingredient in the composition of the present invention, a method for preparing strains used for preparing ALDH-containing extract powder, lysate, Korea cited in the specification of the present invention It is described in detail in the specification of patent application No. 10-2020-00198858.
본 발명의 ALDH함유 조성물은, 약학적으로 허용 가능한 담체를 추가로 더 포함할 수 있다. 상기 약학적으로 허용 가능한 담체에는,식염수, 멸균수, 완충 식염수, 사이클로덱스트린, 덱스트로즈 용액, 말토덱스트린 용액, 글리세롤, 에탄올등을 포함하지만 이에 한정되지 아니한다. The ALDH-containing composition of the present invention may further include a pharmaceutically acceptable carrier. The pharmaceutically acceptable carrier includes, but is not limited to, saline, sterile water, buffered saline, cyclodextrin, dextrose solution, maltodextrin solution, glycerol, ethanol, and the like.
본 발명의 ALDH함유 조성물에는, 싸카로마이세스 세레비지애에서 유래된 글루타치온이 함유될 수 있으며 희석제, 분산제, 계면활성제, 결합제, 윤활제 등을 부가적으로 첨가하여 수용액, 현탁액, 유탁액 등과 같은 제형, 환약, 캡슐, 과립 또는 정제로 제제화할 수 있다.The ALDH-containing composition of the present invention may contain glutathione derived from Saccharomyces cerevisiae, and may be formulated as an aqueous solution, suspension, emulsion, etc. by additionally adding a diluent, dispersant, surfactant, binder, lubricant, etc. , pills, capsules, granules or tablets.
본 발명의 약학 조성물은 약학적으로 유효한 양으로 투여한다. 유효량 수준은 환자의 질환 종류, 중증도, 약물의 활성, 약물에 대한 민감도, 투여 시간, 투여 경로 및 배출비율, 치료기간, 동시 사용되는 약물을 포함한 요소 및 당업자에게 잘 알려진 요소에 따라 결정될 수있다.The pharmaceutical composition of the present invention is administered in a pharmaceutically effective amount. The effective amount level can be determined according to the patient's disease type, severity, drug activity, sensitivity to drug, administration time, administration route and excretion rate, duration of treatment, factors including concurrent drugs, and factors well known to those skilled in the art.
본 발명의 식품 조성물은 담체, 희석제, 부형제, 및 첨가제 중하나 이상을 포함하여 정제, 환제, 산제, 과립제, 분말제, 캡슐제, 및 액제 제형으로 이루어진 군에서 선택된 하나로 제형된 것을 특징으로 한다. The food composition of the present invention is characterized in that it is formulated with one selected from the group consisting of tablets, pills, powders, granules, powders, capsules, and liquid formulations, including one or more of carriers, diluents, excipients, and additives.
상기 본 발명의 식품 조성물에 더 포함될 수 있는 첨가제로는, 천연 탄수화물, 향미제, 영양제, 비타민, 합성 풍미제, 천연풍미제, 착색제, 충진제, 알긴산 및 그의 염, 유기산, 보호성 콜로이드 증점제, pH 조절제, 안정화제, 방부제, 산화 방지제, 글리세린, 알코올 등이 사용될 수 있다. Additives that may be further included in the food composition of the present invention include natural carbohydrates, flavoring agents, nutrients, vitamins, synthetic flavoring agents, natural flavoring agents, colorants, fillers, alginic acid and salts thereof, organic acids, protective colloidal thickeners, pH Regulators, stabilizers, preservatives, antioxidants, glycerin, alcohols and the like can be used.
[실시예][Example]
본 발명의 ALDH함유 조성물 준비Preparation of the ALDH-containing composition of the present invention
실시예1-1: ALDH 함유 사카로마이세스세레비지에 효모 발효 과정Example 1-1: Yeast fermentation process in Saccharomyces cerevisiae containing ALDH
ALDH 함유 사카로마이세스세레비지에 효모(KCTC139525BP)를 200 mL flask에 YPD 배지(효모추출물, 펩톤, 글루코즈 함유 배지)를 사용하여 160 rpm, 30℃ 조건의 인큐베이터에서 24시간 동안 발효하여 배양했으며, 본 배양은 5 L 발효기 (Marado-05D-PS, CNS, Korea)을 통하여 72 시간 동안 진행하였다. 배양 종료 후 고속원심분리기(Supra R22, Hanil, Korea)를 이용하여 효모를 원심분리하였다. Saccharomyces cerevisiae yeast (KCTC139525BP) containing ALDH was fermented and cultured for 24 hours in an incubator at 160 rpm and 30 ° C using YPD medium (yeast extract, peptone, and glucose containing medium) in a 200 mL flask, This culture was carried out for 72 hours through a 5 L fermenter (Marado-05D-PS, CNS, Korea). After completion of the culture, the yeast was centrifuged using a high-speed centrifuge (Supra R22, Hanil, Korea).
실시예1-2: 본 발명의 ALDH함유 조성물 준비Example 1-2: Preparation of ALDH-containing composition of the present invention
원심분리된 ALDH 함유 효모를 초저온냉동고(CLN-52U, Nihon freezer, Japan)에서 2일간 냉동시킨 후 동결건조기(FDU-7006, Operon, Korea)로 2일간 동결건조를 진행하였다. 동결건조된 효모 파우더 3 g에 단백질분해효소억제제(A32955, Thermo fisher, USA)를 넣은 50 mL 인산완충식염수(phosphate-buffered saline, PBS)에 녹인 후, 0.5 mm 세포파쇄용유리구슬 (11079105, Biospec) 10 g을 넣고 비드호모나게이저(Mixer Mill MM400, Retsch, Germany)로 2분씩 총 3번에 걸쳐 효모 파쇄시켰다. 고속원심분리기(Supra R22, Hanil, Korea)를 이용하여 원심분리한 후 상등액만 분리하여 동결건조기(FDU-7006, Operon, Korea)로 2일간 동결건조를 진행하였다. The centrifuged ALDH-containing yeast was frozen in a cryogenic freezer (CLN-52U, Nihon freezer, Japan) for 2 days, and then freeze-dried for 2 days in a freeze dryer (FDU-7006, Operon, Korea). After dissolving 3 g of lyophilized yeast powder in 50 mL phosphate-buffered saline (PBS) containing a protease inhibitor (A32955, Thermo fisher, USA), 0.5 mm glass beads for cell disruption (11079105, Biospec) 10 g was put into the bead homogenizer (Mixer Mill MM400, Retsch, Germany) to disrupt yeast over a total of 3 times for 2 minutes each. After centrifugation using a high-speed centrifuge (Supra R22, Hanil, Korea), only the supernatant was separated and freeze-dried for 2 days with a freeze dryer (FDU-7006, Operon, Korea).
[실시예2][Example 2]
1. 천식 억제 유효성 평가를 위한 OVA를 이용한 천식 마우스 모델제작1. Asthma mouse model production using OVA for asthma inhibitory efficacy evaluation
OVA(Ovlabumin; 마우스 염증 유도제)와 aluminum hydroxide (면역반응증강제)를 생리식염수에 녹여 혼합물을 마우스 한 마리당 1 mL 주사기를 이용해 복강투여로 감작을 진행하고. 복강투여는 일주일에 한번 씩 총 2번에 진행한후 일주일이 지난 시점부터 본 발명의 ALDH함유 조성물 분말을 생리식염수에 녹여 경구 투여를 진행하였고 OVA를 생리식염수에 녹여 OVA용액을 nebulizer system을 이용하여 야기하였다. 하루에 한번 씩 총 5일 동안 진행하였다. 본 발명의 ALDH함유 조성물 분말은 6mg/kg/day, 30mg/kg/day, 150mg/kg/day로 투여하였다.Dissolve OVA (Ovlabumin; mouse inflammation inducer) and aluminum hydroxide (immune response enhancer) in physiological saline and sensitize the mixture by intraperitoneal administration using a 1 mL syringe per mouse. After intraperitoneal administration was performed twice a week, the powder of the ALDH-containing composition of the present invention was dissolved in physiological saline from the time point after a week had elapsed and oral administration was carried out. caused It was conducted once a day for a total of 5 days. The powder of the composition containing ALDH of the present invention was administered at 6 mg/kg/day, 30 mg/kg/day, and 150 mg/kg/day.
1) 대조군은 아무 것도 투여 안하고 실험군은 Balb C 마우스 각 군당 8마리를 복강(intra peritoneal)으로 ovalbumin을 첫 날 20ug/head 주사하였다.1) The control group did not administer anything, and the experimental group injected 20 ug/head of ovalbumin intraperitoneally to 8 Balb C mice in each group on the first day.
2) 8일째에 다시 상기의 방법으로 동일한 반복 하여 동일한 양을 주사하였다.2) On the 8th day, the same amount was injected again in the same manner as above.
3) 14일째부터는 매일 오전에 본 발명의 ALDH함유 조성물 분말을 투여하고 오후에는 ovalbumin 20ug/head/day를 nebulizer를 이용하여 흡입으로 투여하였다. 그리고 이러한 작업을 5일간 반복하였다. 이때 본 발명의 ALDH함유 조성물 분말 투여는 6mg/kg, 30mg/kg, 150mg/kg 3군으로 용량을 다르게 투여하였다. 3) From the 14th day, the powder of the ALDH-containing composition of the present invention was administered in the morning, and 20 ug/head/day of ovalbumin was administered by inhalation using a nebulizer in the afternoon. And this operation was repeated for 5 days. At this time, the powder administration of the ALDH-containing composition of the present invention was administered at different doses in 3 groups of 6 mg/kg, 30 mg/kg, and 150 mg/kg.
4) Dexamethasone(스테로이드) 투여군은 마우스 한 마리당 1mg/kg/day이었다.4) Dexamethasone (steroid) administration group was 1mg/kg/day per mouse.
이하의 각 도면에서, con.(control)은 대조군으로 천식 유도물질인 ovalbumin이나 천식치료용 대조약물인 dexamethasone 또는 본 발명의 ALDH함유 조성물 분말을 전혀 투여 하지 않은 정상군이다. OVA군은 상기의 방법에 의하여 천식을 유도하기 위하여 ovalbumin을 실험 1일째와 8일째는 복강으로 14일째에는 nebulizer를 이용해 흡입으로 투여한 군이다. Dex군은 상기의 방법으로 ovalbumin을 1일째와 8일째는 복강으로 투여하고 14일째부터는 5일간 반복하여 매일 오전에 dexamethasone(스테로이드)를 마우스 한 마리당 1mg/kg/day를 투여하고 오후에는 ovalbumin 20ug/head/day를 nebulizer를 이용하여 흡입으로 투여한 군이다. ALDH 6mg/kg 군은 14일째부터는 5일간 반복하여 매일 오전에 본 발명의 ALDH함유 조성물 분말 6mg/kg을 투여하고 오후에는 ovalbumin 20ug/head/day를 nebulizer를 이용하여 흡입으로 투여한 군이고, ALDH 30mg/kg 군은 14일째부터는 5일간 반복하여 매일 오전에 본 발명의 ALDH함유 조성물 분말 30mg/kg을 투여하고 오후에는 ovalbumin 20ug/head/day를 nebulizer를 이용하여 흡입으로 투여한 군이며, ALDH 150mg/kg 군은 14일째부터는 5일간 반복하여 매일 오전에 본 발명의 ALDH함유 조성물 분말 150mg/kg을 투여하고 오후에는 ovalbumin 20ug/head/day를 nebulizer를 이용하여 흡입으로 투여한 군이다.In each of the following figures, con. (control) is a normal group to which ovalbumin, an asthma inducer, dexamethasone, a control drug for asthma treatment, or the powder of the composition containing ALDH of the present invention was not administered as a control group. The OVA group was a group in which ovalbumin was administered intraperitoneally on the 1st and 8th days of the experiment and by inhalation using a nebulizer on the 14th day to induce asthma by the above method. In the Dex group, ovalbumin was administered intraperitoneally on the 1st and 8th days as described above, and from the 14th day onwards, dexamethasone (steroid) was administered 1mg/kg/day per mouse in the morning and repeated for 5 days from the 14th day, and ovalbumin 20ug/day in the afternoon. The group was administered head/day by inhalation using a nebulizer. The ALDH 6mg/kg group is a group in which 6mg/kg of the ALDH-containing composition powder of the present invention is administered in the morning repeatedly for 5 days from the 14th day, and 20ug/head/day of ovalbumin is administered by inhalation using a nebulizer in the afternoon, and ALDH The 30mg/kg group is a group in which 30mg/kg of the ALDH-containing composition powder of the present invention is administered in the morning repeatedly for 5 days from the 14th day, and ovalbumin 20ug/head/day is administered by inhalation using a nebulizer in the afternoon, and ALDH 150mg The /kg group is a group in which 150 mg/kg of the ALDH-containing composition powder of the present invention is administered in the morning repeatedly for 5 days from the 14th day, and 20 ug/head/day of ovalbumin is administered by inhalation using a nebulizer in the afternoon.
상기 과정이 끝난 후 마우스를 희생시켜 폐기관지내에서 Bronchioalveolar fluid(BALF) 성분을 분석하여 WBC(백혈구)의 수를 측정하였고 IL-4와 IL-13등의 cytokine을 enzyme-linked immunoassay(ELISA) 법을 활용하여 측정하였다.After the above procedure, mice were sacrificed and Bronchioalveolar fluid (BALF) components were analyzed in the lung bronchi to measure the number of WBCs (white blood cells), and cytokines such as IL-4 and IL-13 were analyzed by enzyme-linked immunoassay (ELISA) method. was used for measurement.
[실시예 3][Example 3]
천식억제 유효성 평가를 위한 생쥐 부검Autopsy of mice to evaluate asthma inhibitory efficacy
5일간 nebulizer system와 경구투여를 진행하고 그 다음날 부검을 실시하였다. 한 그룹에 총 8마리가 있으며 8마리 전부 혈액을 채취하며 각 그룹별로 4마리는 기관지 폐포 세척액(Bronchial Alveolar Lavage Fluid; BALF)을 나머지 4마리에서는 폐를 적출하였다. 적출한 폐는 3등분하였다. 하나는 조직병리를 보기위해 10% 포르말린에 보관하고, 나머지 2등분은 Cytokine을 보기위해 -80℃에 보관하였다.After oral administration with the nebulizer system for 5 days, an autopsy was performed the next day. There were a total of 8 animals in one group, and blood was collected from all 8 mice. Bronchial Alveolar Lavage Fluid (BALF) was collected from 4 animals in each group, and the lungs were removed from the remaining 4 animals. The extracted lungs were divided into thirds. One was stored in 10% formalin to examine histopathology, and the other two were stored at -80°C to view cytokines.
기관지 폐포 세척액은 동물마취제에 의해 마취시킨 후, 해부판에 고정 시켜 혈액을 채취 후 기도를 확보하고 카테터를 삽입하여 1 mL 주사기에 PBS 완충 용액을 넣고 다시 빨아들인다. 빨아들인 액은 1.5 mL tube에 보관하고 다른 주사기를 이용해 한 번 더 진행하였다.After anesthetizing the bronchoalveolar lavage solution with an animal anesthetic, fix it on the dissection plate to collect blood, secure an airway, insert a catheter, insert PBS buffer into a 1 mL syringe, and suck again. The sucked solution was stored in a 1.5 mL tube and proceeded once more using another syringe.
[실시예 4][Example 4]
결과 분석Results analysis
(1) BALF(기관지 폐포세척액) 의 혈구분석(1) Blood cell analysis of BALF (Bronchoalveolar Lavage Fluid)
동물에게 얻은 BALF를 4℃에서 3000 rpm으로 5분간 원심분리를 진행하였다. 상층액은 제거하고 남아있는 펠렛(pellet)에 PBS 완충액을 넣어 섞어준다. 그 후 혈구분석기(Hemavet 950FS, Drew Scientific Inc, Korea)를 활용하여 혈구분석을 실시하였다.The BALF obtained from the animal was centrifuged at 4°C at 3000 rpm for 5 minutes. Remove the supernatant and mix with PBS buffer to the remaining pellet. Thereafter, blood cell analysis was performed using a hemacytometer (Hemavet 950FS, Drew Scientific Inc, Korea).
(2) BALF(기관지 폐포세척액) 의 염증세포 수 확인(2) Check the number of inflammatory cells in BALF (Bronchoalveolar Lavage Fluid)
혈구분석에 사용하고 남은 BALF를 원심분리 4℃에서 3,000 rpm으로 5분간 원심분리를 진행하였다. 그 후 상층액을 버리고 PBS 완충액을 넣고 Cyto-Tek 기계를 이용해 800 rpm으로 5분간 원심분리를 진행하였다. 그후 Diff Qucik 시약을 이용해 염색하였다. 기관지 폐포 세척액에서의 서의 염증세포수와 호중성백혈구 그리고 단핵 백혈구의 변화를 관찰한 결과이다. 각 그룹 별로 8마리의 마우스의 혈액을 채취하며 각 그룹별로 4마리는 기관지 폐포 세척액(Bronchial Alveolar Lavage Fluid; BALF)을 채취하여 혈구분석기(Hemavet 950FS, Drew Scientific Inc, Korea)를 활용하여 Diff Qucik 시약을 이용해 염색하여 염증세포수와 호중성 백혈구 및 단핵 백혈구분석을 실시하였다. The remaining BALF used for blood cell analysis was centrifuged at 4°C and 3,000 rpm for 5 minutes. After that, the supernatant was discarded, PBS buffer was added, and centrifugation was performed at 800 rpm for 5 minutes using a Cyto-Tek machine. Then, it was stained with Diff Qucik reagent. These are the results of observation of changes in the number of inflammatory cells, neutrophils, and mononuclear leukocytes in bronchoalveolar lavage fluid. Blood is collected from 8 mice from each group, and Bronchial Alveolar Lavage Fluid (BALF) is collected from 4 mice from each group, and Diff Qucik reagent is used using a hemocytometer (Hemavet 950FS, Drew Scientific Inc, Korea). Inflammatory cell count, neutrophil and mononuclear leukocytes were analyzed by staining using
(3) Serum의 IgE 분석(3) Serum IgE analysis
IgE ELISA set를 사용하였으며 micro well에 Coating Buffer에 희석한 Capture Ab를 넣고 4 ℃에서 overnight. 3번의 washing 후 각 well에 Assay Diluent를 넣고 실온에서 incubation. 3번의 washing 후 각 well에 Assay Diluent로 희석한 standard, sample를 넣고 실온에서 incubation. 5번의 washing 후 각 well에 Working Detector를 넣고 실온에서 incubation하고 7번의 washing 후 각 well에 Substrate Solution을 넣고 실온의 암조건에서 incubation. 각 well에 Stop Solution을 30분 이내에 450 nm에서 측정하였다.An IgE ELISA set was used, and Capture Ab diluted in Coating Buffer was added to a micro well and incubated at 4 °C overnight. After washing 3 times, add assay diluent to each well and incubate at room temperature. After washing 3 times, add standard and sample diluted with assay diluent to each well and incubate at room temperature. After washing 5 times, put Working Detector in each well and incubate at room temperature. After washing 7 times, add Substrate Solution to each well and incubate in dark conditions at room temperature. Stop solution in each well was measured at 450 nm within 30 minutes.
(4) 조직병리학적 분석(H&E, PAS stain)(4) Histopathological analysis (H&E, PAS stain)
파라핀 제거 및 수화과정을 실시하였으며 탈수과정 후 수돗물에 5분 동안 수세를 실시하고 물기를 잘 닦아준 후에 heamatoxylin solution에 2분간 염색하였다. 염색 후 청명과정 및 탈수과정을 실시한 후에 조직봉입을 실시하였다. Paraffin removal and hydration were carried out, and after dehydration, they were washed with tap water for 5 minutes, dried well, and then stained with heamatoxylin solution for 2 minutes. After staining, clarification and dehydration were performed, and tissue encapsulation was performed.
파라핀 제거 및 수화과정 실시 후 10분 동안 0.5% periodic acid solution에 담가 조직을 산화시키고 산화 후 10분 동안 schiff 시약에 담가 반응시킨 후 2분간 hematoxylin solution으로 염색을 진행하였다. 청명과정 및 탈수과정을 실시한 후에 조직봉입을 실시하였다.After paraffin removal and hydration, the tissue was oxidized by immersion in 0.5% periodic acid solution for 10 minutes. After oxidation, the tissue was immersed in Schiff reagent for 10 minutes for reaction. After performing the clarification process and dehydration process, tissue encapsulation was performed.
(5) Cytokine 변화 분석(5) Cytokine change analysis
Enzyme-Linked Immunosorbent Assay (ELISA): micro well에 Coating Buffer에 희석한 Capture Ab를 넣고 4 ℃에서 overnight. 3번의 washing 후 각 well에 Assay Diluent를 넣고 실온에서 incubation. 3번의 washing 후 각 well에 Assay Diluent로 희석한 standard sample를 넣고 실온에서 incubation. 5번의 washing 후 각 well에 Working Detector를 넣고 실온에서 incubation하고 7번의 washing 후 각 well에 Substrate Solution을 넣고 실온의 암조건에서 incubation. 각 well에 Stop Solution을 30분 이내에 450 nm에서 측정하였다. Cytokine의 ELISA 분석은 Th1(IL-6, IFN-γ), Th2(IL-4, IL-5, IL-13), Th17(IL-6, TNF-α)을 분석하였다.Enzyme-Linked Immunosorbent Assay (ELISA): Add Capture Ab diluted in Coating Buffer to micro-wells and incubate at 4°C overnight. After washing 3 times, add assay diluent to each well and incubate at room temperature. After washing 3 times, add standard sample diluted with Assay Diluent to each well and incubate at room temperature. After washing 5 times, put Working Detector in each well and incubate at room temperature. After washing 7 times, add Substrate Solution to each well and incubate in dark conditions at room temperature. Stop solution in each well was measured at 450 nm within 30 minutes. Cytokine ELISA analysis was Th1 (IL-6, IFN-γ), Th2 (IL-4, IL-5, IL-13), Th17 (IL-6, TNF-α) were analyzed.
실시간-중합효소연쇄반응(RT-PCR): 과민성 호흡기 증상 조절과 관련된 인자들의 변화를 측정하기 위하여, 폐 조직으로부터 extraction kit를 이용하여 total RNA를 추출하고 real-time-PCR 방법을 통해서 mRNA의 변화를 관찰하였다.Real-time polymerase chain reaction (RT-PCR): To measure changes in factors related to the control of hypersensitivity respiratory symptoms, total RNA is extracted from lung tissue using an extraction kit and mRNA changes through real-time-PCR method was observed.
(6) 세포사멸(apoptosis) 변화 분석(6) apoptosis change analysis
과민성 호흡기 증상 조절의 기전을 확인하기 위해 Apoptosis와 관련된 protein(JNK, ERK, p38 , BCL-2 family, caspase-3, NF-κB 등)의 변화를 측정하였다.In order to confirm the mechanism of control of hypersensitive respiratory symptoms, changes in apoptosis-related proteins (JNK, ERK, p38, BCL-2 family, caspase-3, NF-κB, etc.) were measured.
Western blot : 폐 조직으로부터 protein을 추출하고 Western blot 방법을 통해서 protein의 변화를 관찰하였다. Extraction and qualification of protein을 통해 정량하고 gel을 만들고 sample를 넣어 전기영동을 진행하였다. membrane으로 transfer 진행하고 blocking 후 primary antibody 단계 진행 후 Secondary antibody 단계를 진행하였다. Washing 후 chemi-doc을 이용하여 현상하였다.Western blot: Protein was extracted from lung tissue and changes in protein were observed by Western blot method. It was quantified through extraction and qualification of protein, a gel was made, and a sample was added and electrophoresis was performed. After transferring to the membrane, blocking, the primary antibody step, and then the secondary antibody step. After washing, it was developed using chemi-doc.
실시예4-2: 시험 동물의 천식 유발 Example 4-2: Induction of Asthma in Test Animals
마우스는 난알부민(ovalbumin, OVA, 마우스 염증 유도제)(A5503, Sigma, USA)과 수산화 알루미늄(aluminum hydroxide, 면역반응증강제)(77161, Thermo scientific, USA)을 이용하여 감작시켰다. 실험 제 1일 및 8일에 1 mg의 수산화 알루미늄(aluminum hydroxide)에 녹인 OVA 20 μg (전체 용량 500 μL)을 마우스의 복강 내로 주사하여 감작시켰다(X. MA. et al., Evid Based Complement Alternat Med, 2014:1-7, 2014). 실험 제15일 부터 19일 동안 1일 1회 초음파 분무기(NE-U17, Omron, Japan)를 이용하여 인산완충식염수(phosphate-buffered saline, PBS)에 녹인 5% 난알부민을 실험동물의 코에 분무하여 알레르기 비염 및 천식 마우스 모델을 제작하였다. 대조군은 난알부민과 수산화 알루미늄을 이용한 감작만 진행하였다.Mice were sensitized with ovalbumin (OVA, mouse inflammation inducer) (A5503, Sigma, USA) and aluminum hydroxide (immune response enhancer) (77161, Thermo scientific, USA). On the first and eighth days of the experiment, 20 μg of OVA dissolved in 1 mg of aluminum hydroxide (total volume 500 μL) was intraperitoneally injected into mice to sensitize them (X. MA. et al., Evid Based Complement Alternat). Med, 2014:1-7, 2014). 5% egg albumin dissolved in phosphate-buffered saline (PBS) was sprayed into the nose of the experimental animal once a day from the 15th to the 19th day of the experiment using an ultrasonic nebulizer (NE-U17, Omron, Japan). Thus, allergic rhinitis and asthma mouse models were prepared. In the control group, only sensitization using egg albumin and aluminum hydroxide was performed.
[실시예 5][Example 5]
천식 유발 마우스 모델에 대한 본 발명의 ALDH함유 조성물 투여 Administration of the ALDH-containing composition of the present invention to an asthma-induced mouse model
실시예5-1: 본 발명의 ALDH함유 조성물 투여 Example 5-1: Administration of the ALDH-containing composition of the present invention
동결건조 된 ALDH 함유 효모 용해물을 각각 560μL 기준으로 6 mg/kg, 30mg/kg, 150mg/kg의 농도가 되도록 인산완충용액(Phosphae-buffered saline, PBS)에 녹인 후 vortexer(F202A0175, VELP scientific, Italy)로 1분간 혼합시켜주었다(C. Klaßen. et al., J. Immunol, 1;199(1):48-61, 2017). 양성대조물질인 dexamethasone(Sinilpharm, Korea)은 1 mg/kg의 농도가 되도록 생리식염수(Cleancle, JW Pharmaceutical, Korea)에 녹인 후 vortexer(F202A0175, VELP scientific, Italy)로 1분간 혼합시켜주었다(C. Klaßen. et al., J. Immunol, 1;199(1):48-61, 2017).Lyophilized ALDH-containing yeast lysate was dissolved in phosphate-buffered saline (PBS) to a concentration of 6 mg/kg, 30 mg/kg, and 150 mg/kg based on 560 μL, respectively, and then vortexer (F202A0175, VELP scientific, Italy) for 1 minute (C. Klaßen. et al., J. Immunol, 1;199(1):48-61, 2017). The positive control material, dexamethasone (Sinilpharm, Korea), was dissolved in physiological saline (Cleancle, JW Pharmaceutical, Korea) to a concentration of 1 mg/kg and mixed with a vortexer (F202A0175, VELP scientific, Italy) for 1 minute (C. Klaßen. et al., J. Immunol, 1;199(1):48-61, 2017).
실시예5-2: 천식 유발 마우스 모델에 대한 본 발명의 ALDH함유 조성물 투여Example 5-2: Administration of the ALDH-containing composition of the present invention to an asthma-induced mouse model
실시예5-1에서 준비한 시료를 1 mL 주사기(Kovax-syringe 1 mL 26G 1/2, Koreavaccine, Korea)에 경구투여용바늘을 꽂아 실험 제15일 내지 19일 동안 1일 1회 각 시료마다 560 μL를 경구투여하였다. 본 실험은 각 그룹당 8마리로 구성되었고, 대조군과 유도군은 PBS, 치료군은 dexamethasone, 실험군은 ALDH 함유 효모 용해물을 각각 경구투여하였다.Insert a needle for oral administration into a 1 mL syringe (Kovax-syringe 1 mL 26G 1/2, Koreavaccine, Korea) with the sample prepared in Example 5-1, once a day for the 15th to 19th days of the experiment, 560 for each sample μL was orally administered. This experiment consisted of 8 rats in each group. The control and induction groups were orally administered with PBS, the treatment group with dexamethasone, and the experimental group with ALDH-containing yeast lysate, respectively.
[실시예 6][Example 6]
부검 및 분석용 샘플 채취 Sampling for autopsy and analysis
실시예6-1: 마우스 부검 및 혈액, 폐조직 채취 Example 6-1: Mouse autopsy and collection of blood and lung tissue
5일간 초음파 분무기(NE-U17, Omron, Japan)를 이용한 OVA 분무 및 경구투여를 진행하고 그 다음날 부검을 실시하였다. 한 그룹당 8마리 전부 1 mL 주사기(Kovax-syringe 1 mL 26G 1/2, Koreavaccine, Korea)로 혈액을 채취하며 4마리는 폐를 적출하였다. 적출한 폐는 3등분 하여 하나는 조직병리를 보기 위해 10% 포르말린에 보관하고, 나머지 2등분은 인터류킨 측정을을 위해 -80℃에 보관하였다.OVA spraying and oral administration using an ultrasonic nebulizer (NE-U17, Omron, Japan) were performed for 5 days, and an autopsy was performed the next day. Blood was collected from all 8 animals per group using a 1 mL syringe (Kovax-syringe 1 mL 26G 1/2, Koreavaccine, Korea), and lungs were removed from 4 mice. The excised lungs were divided into thirds, one stored in 10% formalin to examine histopathology, and the other two pieces were stored at -80°C for interleukin measurement.
실시예 6-2: 기관지 폐포세척액 채취Example 6-2: Collection of bronchoalveolar lavage fluid
기관지 폐포 세척액은 동물마취제에 의해 마취시킨 후, 해부판에 고정 시켜 혈액을 채취 후 기도를 확보하고 카테터를 삽입하여 1 mL 주사기(Kovax-syringe 1 mL 26G 1/2, Koreavaccine, Korea)에 인산완충용액(Phosphate-buffered saline, PBS)을 넣고 빨아들였다. 채취한 폐포세척액은 1.5 mL tube에 보관하고 다른 주사기를 이용해 동일한 방법으로 반복 채취하였다.After anesthetizing the bronchoalveolar lavage solution with an animal anesthetic, fix it on a dissection plate to collect blood, secure an airway, insert a catheter, and insert a phosphate buffer into a 1 mL syringe (Kovax-syringe 1 mL 26G 1/2, Koreavaccine, Korea). A solution (Phosphate-buffered saline, PBS) was added and sucked. The collected alveolar lavage fluid was stored in a 1.5 mL tube and collected repeatedly in the same manner using another syringe.
[실시예 7][Example 7]
천식 유발 마우스에서 본 발명의 ALDH 함유 조성물에 의한 인터류킨-4, ,인터류킨-13 억제 효과 확인 Confirmation of interleukin-4, interleukin-13 inhibitory effect by the ALDH-containing composition of the present invention in asthma-induced mice
실시예 7-1 적출한 폐포 조직 샘플에서 인터류킨 4, 인터류킨 13의 함량 분석 Example 7-1 Analysis of Interleukin 4 and Interleukin 13 Contents in Extracted Alveolar Tissue Samples
Zirconia bead가 담긴 2 mL 튜브에 폐조직 샘플을 넣은 뒤, 단백질 분해효소 저해제를 넣은 RIPA lysis buffer를 넣어주었다. 비드 호모게나이저로 조직을 분쇄한 후, 원심분리기를 이용하여 13,000 rpm, 4℃의 조건에서 20분간 원심분리를 진행하고, 상층액만 새로운 E-tube에 옮겨담았다. 분석을 위해 IL-4, IL-13 ELISA set (555232, 554599, BD bioscience, USA)를 사용하였다. .측정은 Enzyme-Linked Immunosorbent Assay (ELISA)방법으로 수행하였다. 마이크로 웰(Micro well) 플레이트에 완충액에 희석한 Capture 항체를 넣고 4 ℃에서 16시간 방치 한후, 워싱(washing)한 후 시험 물질을 넣고 반응 시켜 최종 450 nm 파장에서의 흡광도를 ELISA reader (Multiskan sky, Thermo Fisher, USA)로 측정함으로서 샘플 조직내 인터류킨 4와 13의 양을 확인하였다.(도 1,2)After putting the lung tissue sample in a 2 mL tube containing Zirconia beads, RIPA lysis buffer containing a protease inhibitor was added. After pulverizing the tissue with a bead homogenizer, centrifugation was performed for 20 minutes at 13,000 rpm and 4°C using a centrifuge, and only the supernatant was transferred to a new E-tube. For the analysis, IL-4, IL-13 ELISA set (555232, 554599, BD bioscience, USA) was used. . Measurement was performed by Enzyme-Linked Immunosorbent Assay (ELISA) method. After adding the Capture antibody diluted in buffer to a micro-well plate and leaving it at 4 °C for 16 hours, wash (washing), add the test substance, and react to measure the absorbance at the final 450 nm wavelength with an ELISA reader (Multiskan sky, The amount of interleukin 4 and 13 in the sample tissue was confirmed by measuring with Thermo Fisher, USA) (FIGS. 1,2).
실시예 7-2 천식에 대한 본 발명의 ALDH 함유 조성물 in vitro efficacy Example 7-2 In vitro efficacy of the ALDH-containing composition of the present invention for asthma
RBL-2HE 세포주에 본 발명의 ALDH 함유 조성물을 각 농도에 따라 20분 동안 전 처리하고, 세포주에 IgE, BSA를 가하여 세포내 염증 상태를 유발시켰다. 천식 유발 모델의 주요 마커인 IL-4 발현정도를 측정하여 본 발명의 ALDH 함유 조성물에 의한 억제 효과를 측정하여 그 결과를 도1에 나타내었다. 천식 유도 세포주의 IL-4 발현량 대비, 본 발명의 ALDH 함유 조성물이 처리된 세포에서 각 농도별로 13%, 34%, 35%의 IL-4발현량 감소가 확인되었다. IL-4 발현량에 대한 본 발명의 ALDH 함유 조성물의 억제 효과 확인하였고 투려량에 의존하여 IL-4 억제 효과가 증가한다는 사실도 확인되었다. 또한 인터류킨-13은 본 발명의 ALDH 함유 조성물 투여량인 150mg/Kg, 300mg/Kg의 농도에서 약 80% 까지의 발현 감소 효과가 나타났다.(도2) The RBL-2HE cell line was pre-treated with the ALDH-containing composition of the present invention at each concentration for 20 minutes, and an intracellular inflammatory state was induced by adding IgE and BSA to the cell line. The inhibitory effect of the ALDH-containing composition of the present invention was measured by measuring the expression level of IL-4, a major marker of the asthma induction model, and the results are shown in FIG. 1 . Compared to the IL-4 expression level of the asthma-induced cell line, 13%, 34%, and 35% decrease in the IL-4 expression level was confirmed in the cells treated with the ALDH-containing composition of the present invention for each concentration. The inhibitory effect of the ALDH-containing composition of the present invention on the IL-4 expression level was confirmed, and it was also confirmed that the IL-4 inhibitory effect increased depending on the dosage. In addition, interleukin-13 showed up to about 80% reduction in expression at concentrations of 150 mg/Kg and 300 mg/Kg, which are the doses of the ALDH-containing composition of the present invention. (FIG. 2)
실시예 7-3. 천식 유발 마우스에서 본 발명의 ALDH 함유 조성물에 의한 IgE 감소 효과Example 7-3. IgE reduction effect of the ALDH-containing composition of the present invention in asthma-induced mice
1.5 mL 튜브에 담긴 혈액을 12,000 rpm, 4℃ 조건에서 20분간 원심분리를 진행하고 상층액만 분리하여 IgE 측정을 위한 실험에 사용하였다. 분석을 위해 IgE ELISA set (555248, BD bioscience, USA)를 사용하였으며 분석방법은 실시예 5-1과 동일하다. 분석결과는 도 3에 나타내었으며, 천식 유도 마우스 대비 본 발명의 ALDH 함유 조성물 투여군에서 IgE 감소 효과가 확인되었다. The blood in a 1.5 mL tube was centrifuged at 12,000 rpm and 4° C. for 20 minutes, and only the supernatant was separated and used in the experiment for IgE measurement. For the analysis, an IgE ELISA set (555248, BD bioscience, USA) was used, and the analysis method is the same as in Example 5-1. The analysis results are shown in FIG. 3, and the IgE reduction effect was confirmed in the group administered with the ALDH-containing composition of the present invention compared to the asthma-induced mice.
[실시예 8][Example 8]
본 발명의 ALDH함유 조성물의 독성 평가 Toxicity evaluation of the ALDH-containing composition of the present invention
실시예 8-1. 실험 동물의 준비Example 8-1. Preparation of laboratory animals
실험동물은 암컷, 수컷 ICR 마우스(7 주령)를 분양받아 7일간 순화시켰으며 순화 기간 중 일반증상을 관찰하여 건강한 동물만 시험에 사용하였다. 사료와 물은 자유 섭취시켰고 경구투여 전날 평균 체중 약 20g을 기준으로 각 군별 암,수 5수씩 총 10수가 되도록 군 분리를 진행하였다.As experimental animals, female and male ICR mice (7 weeks old) were received and acclimatized for 7 days. During the acclimatization period, general symptoms were observed, and only healthy animals were used for the test. Feed and water were ingested ad libitum, and group separation was performed so that there were 5 males and 5 females in each group based on the average body weight of about 20 g the day before oral administration.
실시예 8-2 본 발명의 ALDH함유 조성물의 투여 Example 8-2 Administration of the ALDH-containing composition of the present invention
시험 물질은 본 발명의 ALDH 함유 조성물 기준으로 실험동물의 투여용량이 각 0, 750, 3000, 5000mg/Kg이 되도록 생리식염수에 녹여 제조하였다. 투여 용량의 기준은 식약처의 Korea national Toxicology Program(KNTP) 독성 시험 매뉴얼을 준수하였으며, KNTP 매뉴얼에서 가이드하는 적용 최대 용량 5000mg/Kg을 본 실험의 최대 농도로 적용하였다. 각 군별로 준비된 시료를 시험동물에 대하여 각 1회 경구투여를 실시 하였으며, 정상군(G1)의 경우 생리식염수를 투여하였다. The test substance was prepared by dissolving in physiological saline so that the doses of the test animals were 0, 750, 3000, and 5000 mg/Kg, respectively, based on the ALDH-containing composition of the present invention. The standard of administration dose complied with the Korea National Toxicology Program (KNTP) toxicity test manual of the Ministry of Food and Drug Safety, and the maximum applied dose 5000mg/Kg guided by the KNTP manual was applied as the maximum concentration of this experiment. Samples prepared for each group were orally administered once to each test animal, and physiological saline was administered to the normal group (G1).
실시예 8-3. 관찰 및 부검 Example 8-3. Observation and autopsy
모든 시험군의 동물에 대하여 입수일부터 부검일까지 매일 1회 이상 증상관찰을 실시하였으며, 경구투여 후 7일 동안 증상을 관찰하였다. 증상 관찰 종류 후, 부검을 진행하였고 부검 시 육안으로 각 장기에 대한 변화를 관찰하였다.Symptoms were observed at least once a day from the date of acquisition to the day of autopsy for all animals in the test group, and symptoms were observed for 7 days after oral administration. After the type of symptom observation, an autopsy was performed, and changes in each organ were visually observed at the time of autopsy.
마우스를 사용하여 본 발명의 ALDH 함유 조성물 단회투여독성 시험을 실시한 결과, 본 발명의 ALDH함유 조성물 5000mg/kg까지의 농도에서 7일간 사망예를 관찰할 수 없었으며, 체중증가, 사료 섭취량 등의 특이점을 발견할 수 없었다. 또한 관찰 종료 후 진행한 부검 결과에서도 특이한 소견은 발견되지 아니하였다. As a result of a single dose toxicity test of the ALDH-containing composition of the present invention using mice, no mortality was observed for 7 days at a concentration of up to 5000 mg/kg of the ALDH-containing composition of the present invention. could not be found Also, no unusual findings were found in the autopsy results after the observation was completed.
[수탁번호][Accession Number]
기탁기관명 : 한국생명공학연구원Name of deposit institution: Korea Research Institute of Bioscience and Biotechnology
수탁번호 : KCTC13925BPAccession number: KCTC13925BP
수탁일자 : 20190822Deposit date: 20190822
[규칙 제91조에 의한 정정 06.03.2021] 
Figure WO-DOC-FIGURE-156
[Correction by Rule 91 06.03.2021]
Figure WO-DOC-FIGURE-156
기탁기관명 : 한국생명공학연구원Name of deposit institution: Korea Research Institute of Bioscience and Biotechnology
수탁번호 : KCTC14122BPAccession number: KCTC14122BP
수탁일자 : 20200130Deposit date: 20200130
[규칙 제91조에 의한 정정 06.03.2021] 
Figure WO-DOC-FIGURE-161
[Correction by Rule 91 06.03.2021]
Figure WO-DOC-FIGURE-161
기탁기관명 : 한국생명공학연구원Name of deposit institution: Korea Research Institute of Bioscience and Biotechnology
수탁번호 : KCTC14123BPAccession number: KCTC14123BP
수탁일자 : 20200130Deposit date: 20200130
[규칙 제91조에 의한 정정 06.03.2021] 
Figure WO-DOC-FIGURE-165
[Correction by Rule 91 06.03.2021]
Figure WO-DOC-FIGURE-165

Claims (5)

  1. 알데히드 탈수소 효소를 함유하는, 천식 억제용 식품조성물.Containing an aldehyde dehydrogenase, a food composition for suppressing asthma.
  2. 제1항에 있어서, 상기 알데히드 탈수소 효소가 싸카로마이세스세레비지오 효모에서 분리된 것임을 특징으로 하는, 천식 억제용 식품조성물.According to claim 1, wherein the aldehyde dehydrogenase is characterized in that the isolated from Saccharomyces cerevisiae yeast, asthma control food composition.
  3. 제1항 또는 제2항에 있어서, 상기 싸카로마이세스세레비지오 효모가 기탁번호 KCTC13925BP ( Saccharomyces cerevisiae KwonP-1), KCTC14122BP ( Saccharomyces cerevisiae KwonP-2), KCTC14123BP( Saccharomyces cerevisiae KwonP-3) 균주들로 선택되는 어느 하나 또는 이들의 혼합 균주인 것을 특징으로 하는, 천식 억제용 식품 조성물.According to claim 1 or 2, wherein the yeast Saccharomyces cerevisiae with accession number KCTC13925BP (Saccharomyces cerevisiae KwonP-1), KCTC14122BP ( Saccharomyces cerevisiae KwonP-2), KCTC14123BP ( Saccharomyces cerevisiae KwonP-3) strains A food composition for inhibiting asthma, characterized in that any one selected or a mixed strain thereof.
  4. 알데히드탈수소 효소(ALDH)를 함유하는 기관지 천식, 알러지성 천식, 아토피성 천식, 운동유발 천식, 심인성 천식 또는 폐포성 천식 증상을 억제하는 약제 조성물. A pharmaceutical composition for suppressing symptoms of bronchial asthma, allergic asthma, atopic asthma, exercise-induced asthma, psychogenic asthma, or alveolar asthma containing aldehyde dehydrogenase (ALDH).
  5. 제4항에 있어서, 상기 싸카로마이세스세레비지오 효모가 기탁번호 KCTC13925BP ( Saccharomyces cerevisiae KwonP-1), KCTC14122BP ( Saccharomyces cerevisiae KwonP-2), KCTC14123BP( Saccharomyces cerevisiae KwonP-3) 균주들로 선택되는 어느 하나 또는 이들의 혼합 균주인 것을 특징으로 하는, 천식 억제 약제 조성물.The method according to claim 4, wherein the Saccharomyces cerevisiae yeast is any one selected from accession number KCTC13925BP (Saccharomyces cerevisiae KwonP-1), KCTC14122BP ( Saccharomyces cerevisiae KwonP-2), KCTC14123BP ( Saccharomyces cerevisiae KwonP-3) strains. Or a mixture thereof, characterized in that, asthma inhibitory pharmaceutical composition.
PCT/KR2021/001934 2020-02-18 2021-02-16 Asthma-suppressing composition containing aldehyde dehydrogenase WO2021167309A1 (en)

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
KR10-2020-0019824 2020-02-18
KR10-2020-0019793 2020-02-18
KR20200019793 2020-02-18
KR20200019824 2020-02-18
KR10-2021-0020186 2021-02-15
KR1020210020186A KR102460545B1 (en) 2020-02-18 2021-02-15 Asthma Suppressant which Contains mutant saccharomyces cerevisiae

Publications (1)

Publication Number Publication Date
WO2021167309A1 true WO2021167309A1 (en) 2021-08-26

Family

ID=77391049

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2021/001934 WO2021167309A1 (en) 2020-02-18 2021-02-16 Asthma-suppressing composition containing aldehyde dehydrogenase

Country Status (1)

Country Link
WO (1) WO2021167309A1 (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011158689A1 (en) * 2010-06-19 2011-12-22 天野エンザイム株式会社 Agent for reducing acetaldehyde in oral cavity
JP2013247939A (en) * 2012-06-04 2013-12-12 Kiso Machi New yeast, and pharmaceuticals or foods and drinks containing the same
KR20180003344A (en) * 2016-06-30 2018-01-09 (주)아모레퍼시픽 Anti-inflammatory composition comprising extracellular vesicles derived from yeast

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011158689A1 (en) * 2010-06-19 2011-12-22 天野エンザイム株式会社 Agent for reducing acetaldehyde in oral cavity
JP2013247939A (en) * 2012-06-04 2013-12-12 Kiso Machi New yeast, and pharmaceuticals or foods and drinks containing the same
KR20180003344A (en) * 2016-06-30 2018-01-09 (주)아모레퍼시픽 Anti-inflammatory composition comprising extracellular vesicles derived from yeast

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
BAZEWICZ CHRISTOPHER G.: "Aldehyde dehydrogenase in regulatory T‐cell development, immunity and cancer", IMMUNOLOGY, vol. 156, 1 January 2018 (2018-01-01), pages 47 - 55, XP055840160 *
FONSECA VANESSA M.B., MILANI THAMIRES M.S., PRADO RAFAEL, BONATO VANIA L.D., RAMOS SIMONE G., MARTINS FLAVIANO S., VIANNA ELCIO O.: "Oral administration of Saccharomyces cerevisiae UFMG A-905 prevents allergic asthma in mice : UFMG A-905 and asthma", RESPIROLOGY, vol. 22, no. 5, 1 July 2017 (2017-07-01), pages 905 - 912, XP055817368, ISSN: 1323-7799, DOI: 10.1111/resp.12990 *

Similar Documents

Publication Publication Date Title
EP3576724B1 (en) Cannabinoid-containing complex mixtures for the treatment of mast cell-associated or basophil-mediated inflammatory disorders
Kim et al. Involvement of IL-10 in peroxisome proliferator-activated receptor γ-mediated anti-inflammatory response in asthma
Dong et al. Chlorogenic acid alleviates allergic inflammatory responses through regulating Th1/Th2 balance in ovalbumin-induced allergic rhinitis mice
Jan et al. Suppressive effects of cannabidiol on antigen-specific antibody production and functional activity of splenocytes in ovalbumin-sensitized BALB/c mice
Jung et al. Antioxidant and antiasthmatic effects of saucerneol D in a mouse model of airway inflammation
Song et al. Sulfuretin attenuates allergic airway inflammation in mice
Yuk et al. Effects of astilbic acid on airway hyperresponsiveness and inflammation in a mouse model of allergic asthma
Feng et al. Pudilan xiaoyan oral liquid alleviates LPS-induced respiratory injury through decreasing nitroxidative stress and blocking TLR4 activation along with NF-ΚB phosphorylation in mice
JP2021511316A (en) Compositions and Uses Containing SUMO Inhibitors
WO2021167309A1 (en) Asthma-suppressing composition containing aldehyde dehydrogenase
Liu et al. Glycyrrhizin ameliorates vascular endothelial cell senescence by inhibiting HMGB1 in HFD/STZ-induced diabetic rats and human umbilical vein endothelial cells
WO2019231211A1 (en) Composition for preventing or treating allergic diseases containing mixed extract of two or more among asiasarum root, platycodon root, and cinnamomi ramulus as active ingredients
Zhou et al. Isomaltulose alleviates acute colitis via modulating gut microbiota and the Treg/Th17 balance in mice
KR102460545B1 (en) Asthma Suppressant which Contains mutant saccharomyces cerevisiae
CN114732814B (en) Application of urolithin A in preventing and treating allergic rhinitis and allergic asthma
WO2012093754A1 (en) Composition comprising extracellular vesicles derived from inside a mammalian body and a use therefor
Chen et al. A20 restores impaired intestinal permeability and inhibits Th2 response in mice with colitis
Huang et al. Ophiopogonin D ameliorates non‑alcoholic fatty liver disease in high‑fat diet‑induced obese mice by improving lipid metabolism, oxidative stress and inflammatory response
WO2021167310A1 (en) Atopy inhibitory composition containing aldehyde dehydrogenase
Rayees et al. Sitopaladi, a poly-herbal formulation inhibits IgE mediated allergic reactions
US8722098B2 (en) Ecklonia cava extracts for alleviating or preventing asthmatic reactions
US11058718B2 (en) Method for treating non-alcoholic steatohepatitis (NASH) with the combination of polaprezinc and sodium selenite
WO2022220339A1 (en) Composition for prevention or treatment of coronavirus (sars-cov-2) infection, comprising galla rhois extract as active ingredient
WO2024107013A1 (en) Platform for screening drug for autoimmune disease accompanied with infectious disease
TWI688396B (en) Water extract for promotion on hyperlipidemia, atherogenesis and obesity

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: 21756794

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 21756794

Country of ref document: EP

Kind code of ref document: A1