WO2024085623A1 - Composition de microbiome de surnageant de culture de fermentation de souche km2 de lactiplantibacillus plantarum présentant un effet anti-inflammatoire - Google Patents

Composition de microbiome de surnageant de culture de fermentation de souche km2 de lactiplantibacillus plantarum présentant un effet anti-inflammatoire Download PDF

Info

Publication number
WO2024085623A1
WO2024085623A1 PCT/KR2023/016089 KR2023016089W WO2024085623A1 WO 2024085623 A1 WO2024085623 A1 WO 2024085623A1 KR 2023016089 W KR2023016089 W KR 2023016089W WO 2024085623 A1 WO2024085623 A1 WO 2024085623A1
Authority
WO
WIPO (PCT)
Prior art keywords
group
clostridium
pharmaceutical composition
strain
lactiplantibacillus
Prior art date
Application number
PCT/KR2023/016089
Other languages
English (en)
Korean (ko)
Inventor
성문희
Original Assignee
국민바이오 주식회사
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 국민바이오 주식회사 filed Critical 국민바이오 주식회사
Publication of WO2024085623A1 publication Critical patent/WO2024085623A1/fr

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
    • A23L33/135Bacteria or derivatives thereof, e.g. probiotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/66Microorganisms or materials therefrom
    • A61K35/74Bacteria
    • A61K35/741Probiotics
    • A61K35/744Lactic acid bacteria, e.g. enterococci, pediococci, lactococci, streptococci or leuconostocs
    • A61K35/747Lactobacilli, e.g. L. acidophilus or L. brevis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]

Definitions

  • the present invention relates to a microbiome composition of the fermentation culture supernatant of Lactiplantibacillus plantarum KM2 ( Lactiplantibacillus plantarum KM2) strain, which has anti-inflammatory effects.
  • Inflammation is the expression of a normal, protective defense mechanism in the body that appears locally in response to tissue damage caused by physical trauma, harmful chemicals, infection by microorganisms, or irritating substances in the body's metabolites. This inflammation is triggered by various chemical mediators produced from damaged tissues and migrating cells, and these chemical mediators are known to come in many different types depending on the type of inflammatory process. In normal cases, the body neutralizes or eliminates pathogenic factors through an inflammatory response and regenerates damaged tissues to restore normal structure and function.
  • this inflammatory reaction occurs abnormally, it may progress to a disease such as chronic inflammation, and if inflammation is triggered inappropriately by harmless substances such as pollen or an autoimmune reaction such as asthma or rheumatoid arthritis, the defense reaction itself Rather, it damages tissues and causes various diseases.
  • steroidal and non-steroidal therapeutic agents are largely used as the most common preventive or therapeutic agents for inflammatory diseases, but most of them have the problem of side effects, and the development of inflammatory disease therapeutic agents that overcome the above problems is actively underway. am.
  • An object of the present invention is to provide a pharmaceutical composition for preventing or treating inflammatory diseases.
  • Another object of the present invention is to provide a health functional food composition for preventing or improving inflammatory diseases.
  • Another object of the present invention is to provide a method for preventing or treating inflammatory diseases.
  • the present invention prevents inflammatory diseases comprising the fermentation culture supernatant of Lactiplantibacillus plantarum strain, its concentrate, its dried product, its fermentation metabolite, or a mixture thereof as an active ingredient.
  • a pharmaceutical composition for treatment is provided.
  • the present invention provides a health functional food composition for preventing or improving inflammatory diseases comprising the fermentation culture supernatant of the above strain, its concentrate, its dried product, its fermentation metabolite, or a mixture thereof as an active ingredient.
  • the present invention compares to the normal group, Weizmannia coagulans , Welch's bacillus ( clostridium perfringens ), Clostridium botulinum , Clostridium sporogenis, Clostridium sporogenes
  • a method for preventing or treating an inflammatory disease comprising treating a group of patients with an inflammatory disease in which the number of Bifidobacterium pseudolongum strains is increased compared to a normal group with the pharmaceutical composition of claim 1.
  • the fermentation culture supernatant of the Lactiplantibacillus plantarum KM2 strain exhibits anti-inflammatory activity, such as suppressing nitric oxide activity and controlling the expression of inflammatory cytokines.
  • anti-inflammatory activity such as suppressing nitric oxide activity and controlling the expression of inflammatory cytokines.
  • Figure 1 shows the results of analyzing the cytotoxicity of the fermentation culture supernatant (hereinafter referred to as sample) of the Lactiplantibacillus plantarum KM2 strain in macrophages.
  • Figure 2 shows the results of analyzing the effect of samples on the production of nitric oxide (hereinafter referred to as NO) in macrophages.
  • FIG 3 shows the results of analyzing the effect of samples on the production of prostaglandin E2 (Prostaglandin E2; hereinafter referred to as PGE2) in macrophages.
  • Figure 4 shows the results of analyzing the effect of samples on the expression of iNOS (Inducible nitric oxide synthase) and COX-2 (Cyclooxygenase-2) in macrophages.
  • iNOS Inducible nitric oxide synthase
  • COX-2 Cyclooxygenase-2
  • Figure 5 shows the results of analyzing the effect of samples on the production of pro-inflammatory and anti-inflammatory cytokines in macrophages.
  • Figure 6 shows the results of analyzing the effect of samples on TEER (Trans-epithelial electrical resistance) in intestinal epithelial cells.
  • Figure 7 shows the results of analyzing the effect of samples on paracellular permeability in intestinal epithelial cells.
  • Figure 8 shows the results of analyzing the effect of samples on intestinal junction protein expression in macrophages.
  • Figure 9 shows the results of analyzing the effect of samples on body weight and disease activity in animal models.
  • Figure 10 shows the results of analyzing the effect of samples on colon length in an animal model.
  • Figure 11 shows the results of analyzing the effect of samples on inflammatory cytokine production in an animal model.
  • Figure 12 shows the results of analyzing the effect of samples on intestinal tissue in an animal model.
  • Figure 13 shows the results of analyzing the effect of samples on intestinal microorganisms in an animal model.
  • the present invention provides a pharmaceutical composition for preventing or treating inflammatory diseases comprising as an active ingredient a fermentation culture supernatant of Lactiplantibacillus plantarum strain, a concentrate thereof, a dried product thereof, a fermentation metabolite thereof, or a mixture thereof. do.
  • the strain may be Lactiplantibacillus plantarum KM2 ( Lactiplantibacillus plantarum KM2) deposited with the deposit number KCTC 14637BP.
  • the inflammatory diseases include ulcerative colitis, ulcerative duodenitis, Crohn's disease, irritable bowel syndrome, intestinal Behcet's disease, and bleeding rectal ulcers ( hemorrhagic rectal ulcer, pouchitis, enteritis, ischemic colitis, acne, extra-intestinal manifestations dermatitis, atopic dermatitis, allergic dermatitis, seborrheic dermatitis, papular urticaria, eczema, Asthma, conjunctivitis, periodontitis, rhinitis, otitis media, Ubuntuis, pharyngitis, tonsillitis, pneumonia, pancreatitis, gastritis, hemorrhoids, gout, ankylosing spondylitis, lupus, fibromyalgia, psoriasis, rheumatoid arthritis, osteoarthritis, osteoporosis, hepatitis, cystitis, nep
  • the pharmaceutical composition may further include dead cells or spores of the Lactiplantibacillus plantarum KM2 strain.
  • the pharmaceutical composition may be used against Weizmannia coagulans , Welch's bacillus ( clostridium perfringens) , Clostridium botulinum , Clostridium sporogenes , Clostridium baratii ( It is possible to control one or more intestinal microorganisms selected from the group consisting of clostridium baratii , clostridium butyricum , and bifidobacterium pseudolongum .
  • the pharmaceutical composition contains prostaglandin E2 (Prostaglandin E2), iNOS (Inducible nitric oxide synthase), COX-2 (Cyclooxygenase-2), IL-1 ⁇ (Interleukin-1 ⁇ ), TNF- ⁇ (tumor necrosis factor- ⁇ ), and Inhibit the production or expression of one or more selected from the group consisting of IL-6 (Interleukin-6), or the production or expression of one or more selected from the group consisting of ZO-1, Occludin, Claudin-1 and IL-10 (Interleukin-10) can promote, but is not limited to this.
  • the pharmaceutical composition of the present invention is prepared in unit dose form or in a multi-dose container by formulating it using a pharmaceutically acceptable carrier according to a method that can be easily performed by those skilled in the art. It can be manufactured by internalizing it.
  • the pharmaceutically acceptable carriers are those commonly used in preparation, and include lactose, dextrose, sucrose, sorbitol, mannitol, starch, gum acacia, calcium phosphate, alginate, gelatin, calcium silicate, microcrystalline cellulose, Includes, but is not limited to, polyvinylpyrrolidone, cellulose, water, syrup, methyl cellulose, methyl hydroxybenzoate, propylhydroxybenzoate, talc, magnesium stearate, mineral oil, etc.
  • the pharmaceutical composition of the present invention may further include lubricants, wetting agents, sweeteners, flavoring agents, emulsifiers, suspending agents, preservatives, etc.
  • the content of additives included in the pharmaceutical composition is not particularly limited and can be appropriately adjusted within the content range used in conventional formulations.
  • the pharmaceutical compositions include injectable formulations such as aqueous solutions, suspensions, emulsions, etc., pills, capsules, granules, tablets, creams, gels, patches, sprays, ointments, warning agents, lotions, liniment agents, paste agents, and cataplasmase agents. It may be formulated in the form of one or more external skin preparations selected from the group consisting of, but is not limited to this.
  • the pharmaceutical composition of the present invention may additionally contain pharmaceutically acceptable carriers and diluents for formulation.
  • the pharmaceutically acceptable carriers and diluents include excipients such as starch, sugar and mannitol, fillers and extenders such as calcium phosphate, cellulose derivatives such as carboxymethylcellulose, hydroxypropylcellulose, gelatin, alginate, polyvinyl pyrrolidone. It includes, but is not limited to, binders such as talc, calcium stearate, lubricants such as hydrogenated castor oil and polyethylene glycol, disintegrants such as povidone and crospovidone, and surfactants such as polysorbate, cetyl alcohol, glycerol, etc.
  • the pharmaceutically acceptable carrier and diluent may be biologically and physiologically friendly to the subject. Examples of diluents include, but are not limited to, saline, aqueous buffers, solvents, and/or dispersion media.
  • the pharmaceutical composition of the present invention can be administered orally or parenterally (for example, intravenously, subcutaneously, intraperitoneally, or topically) depending on the desired method.
  • parenterally for example, intravenously, subcutaneously, intraperitoneally, or topically
  • it can be formulated as tablets, troches, lozenges, aqueous suspensions, oily suspensions, powders, granules, emulsions, hard capsules, soft capsules, syrups, elixirs, etc.
  • parenteral administration it can be formulated as an injection, suppository, powder for respiratory inhalation, aerosol for spray, ointment, powder for application, oil, cream, etc.
  • the dosage of the pharmaceutical composition of the present invention is determined by the patient's condition, weight, age, gender, health, dietary constitution specificity, nature of the preparation, degree of disease, administration time of the composition, administration method, administration period or interval, excretion rate, and
  • the range may vary depending on the drug form and can be appropriately selected by a person skilled in the art. For example, it may range from about 0.1 to 10,000 mg/kg, but is not limited and may be administered once or in divided doses several times a day.
  • the pharmaceutical composition may be administered orally or parenterally (eg, intravenously, subcutaneously, intraperitoneally, or topically applied) depending on the desired method.
  • the pharmaceutically effective amount and effective dosage of the pharmaceutical composition of the present invention may vary depending on the formulation method, administration method, administration time, administration route, etc. of the pharmaceutical composition, and those skilled in the art will know that it is effective for the desired treatment. Dosage can be easily determined and prescribed.
  • the pharmaceutical composition of the present invention may be administered once a day, or may be administered in several divided doses.
  • the present invention provides a health function for preventing or improving inflammatory diseases comprising the fermentation culture supernatant of Lactiplantibacillus plantarum strain, its concentrate, its dried product, its fermentation metabolite, or a mixture thereof as an active ingredient.
  • a food composition is provided.
  • the strain may be Lactiplantibacillus plantarum KM2 ( Lactiplantibacillus plantarum KM2) deposited with the deposit number KCTC 14637BP.
  • the present invention can be generally used with commonly used foods.
  • the food composition of the present invention can be used as a health functional food.
  • health functional food refers to food manufactured and processed using raw materials or ingredients with functionality useful to the human body in accordance with the Health Functional Food Act, and “functionality” refers to food that is related to the structure and function of the human body. It means ingestion for the purpose of controlling nutrients or obtaining useful health effects such as physiological effects.
  • the health functional food composition may contain common food additives, and its suitability as a “food additive” is determined in accordance with the general provisions and general test methods of the food additive code approved by the Ministry of Food and Drug Safety, unless otherwise specified. The decision is made based on the specifications and standards for the item.
  • Items listed in the “Food Additives Code” include, for example, chemical compounds such as ketones, glycine, potassium citrate, nicotinic acid, and cinnamic acid; natural additives such as subchromic pigment, licorice extract, crystalline cellulose, high-liquid pigment, and guar gum; Examples include mixed preparations such as sodium L-glutamate preparations, noodle additive alkaline preparations, preservative preparations, and tar coloring preparations.
  • the food composition of the present invention can be manufactured and processed in the form of tablets, capsules, powders, granules, liquids, pills, etc.
  • hard capsules can be manufactured by mixing and filling the composition according to the present invention with additives such as excipients in a regular hard capsule
  • soft capsules can be manufactured by mixing and filling the composition according to the present invention. It can be manufactured by mixing with additives such as excipients and filling it with a capsule base such as gelatin.
  • the soft capsule may contain plasticizers such as glycerin or sorbitol, colorants, preservatives, etc., if necessary.
  • prevention refers to all actions that suppress or delay inflammatory diseases by administering the composition according to the present invention.
  • treatment refers to any action that improves or beneficially changes the symptoms of an inflammatory disease by administering the composition according to the present invention.
  • improvement refers to all actions that improve the bad state of an inflammatory disease by administering the composition according to the present invention.
  • the present invention compares to the normal group, Weizmannia coagulans , Welch's bacillus ( clostridium perfringens) , Clostridium botulinum , Clostridium sporogenes , Clos.
  • a group of patients with inflammatory diseases in which the number of at least one strain selected from the group consisting of clostridium baratii and clostridium butyricum is reduced;
  • a method for preventing or treating an inflammatory disease comprising treating the pharmaceutical composition to a group of patients with an inflammatory disease in which the number of Bifidobacterium pseudolongum strains is increased compared to a normal group.
  • the KM2 stock strain stored at -70°C was activated and subjected to primary seed culture in a test tube and flask, followed by 20L in a 50L fermenter. 2% (v/v) was inoculated into the working volume and secondary seed culture was performed for 6 hours. This culture was incubated for 12 hours by inoculating 2% (v/v) in a 350L working volume in a 500L fermenter, and glucose was additionally fed once at 6 hours of culture.
  • the cell slurry was removed by first centrifugation at 7200 rpm and 2 L/min in a disk centrifuge, and the supernatant was second centrifuged at 15000 rpm and 1.5 L/min in a tubular centrifuge to form a cell cake. It was removed and the supernatant was recovered. The recovered supernatant was filtered through a 0.2 ⁇ m sterilization filter to obtain a fermentation culture supernatant of the KM2 strain from which the bacterial cells were finally removed. 3% (w/v) of trehalose was added as an excipient to the sterilized fermentation culture supernatant and freeze-dried for 96 to 120 hours to obtain a powdered fermentation culture supernatant.
  • Raw 264.7 cells a mouse-derived macrophage cell line, were purchased from the Korean cell line bank (KCLB) and used. Cells were cultured in DMEM (Dulbecco's modified Eagle's medium) supplemented with 10% FBS (fetal bovine serum) and 1% penicillin/streptomycin (hereinafter referred to as P/S) at 37°C and 5% CO2 . Subculture was carried out once every two days in a conditioned incubator.
  • DMEM Dulbecco's modified Eagle's medium
  • FBS fetal bovine serum
  • P/S penicillin/streptomycin
  • IPEC-J2 cells porcine intestinal epithelial cells, were purchased from the German Collection of Microorganisms and Cell Cultures GmbH (DSMZ). Cells were cultured in DMEM/F-12 (Dulbecco's Modified Eagle's Medium-Nutrient-Mixture F-12 medium) medium supplemented with 10% FBS and 1% P/S for 2 days in an incubator under 5% CO 2 and 37°C conditions. Subcultured once a year.
  • DMEM/F-12 Dulbecco's Modified Eagle's Medium-Nutrient-Mixture F-12 medium
  • C57BL/6 mice were purchased from Orient Bio. Healthy animals were used in the experiment by confirming the mouse population, observing general symptoms, measuring body weight, and confirming the test report provided by the animal supplier. All animals were checked for any abnormalities and were given an acclimatization period of 7 days to adapt to the animal room environment. During the acclimation period, all animals were observed for general symptoms once a day, and the health status of the animals was evaluated by checking body weight changes on the end of acclimatization. To ensure that the average weight of each experimental group was equal, they were separated into 4 groups with 9 animals per group. The animal's tail was marked as an individual using five-color permanent marker, and an individual identification card was attached to the breeding box.
  • the animal model was raised at a temperature of 20-26°C, relative humidity of 30-70%, and light/dark cycle of 12 hours/day (9:00 AM to 9:00 PM), and was supplied with food and drinking water.
  • Solid feed for laboratory animals (Lab DFiet #5053 PMI Nutrition International, U.S.A.) was fed ad libitum. Animal testing was performed with approval from the Korea Food Industry Cluster Promotion Agency Animal Experiment Ethics Committee (IACUC-22-014).
  • the EZ-Cytox kit (DAEIL lab, Korea) was used to determine the cell absorbance based on the decrease in absorbance at each sample treatment concentration compared to the control group (sample untreated group; control). The concentration of samples that affected survival rates was determined.
  • Raw 264.7 and IPEC-J2 cells were each seeded in a 96 well plate at a density of 1 ⁇ 10 4 cells/well, cultured for 24 hours in an incubator under 5% CO 2 and 37°C, and then samples were processed by concentration. Cultured for 24 hours.
  • the culture medium was removed, 90 ⁇ L of DMEM and 10 ⁇ L of EZ-Cytox reagent were mixed and dispensed, and reacted for 1 hour in an incubator with 5% CO 2 and 37°C in the dark. Afterwards, absorbance was measured at 450 nm using a microplate reader, and cell viability was measured using Equation 1 below.
  • NO activity was measured using a NO (nitric oxide) assay kit (Promega Corp, USA, Cat.#G2930).
  • Raw 264.7 cells were seeded in a 24 -well plate at a density of 2.5 Using the added DMEM medium, 1 ⁇ g/ml LPS (Lipopolysaccharides, Sigma-Aldrich, USA) (derived from Escherichia coli 055:B5) and samples were diluted by concentration and cultured for 24 hours. Afterwards, the amount of NO produced during cell culture was measured.
  • LPS Lipopolysaccharides
  • the amount of NO produced was determined by mixing 50 ⁇ L of cell culture medium and 100 ⁇ L of Griess reagent (Sigma-Aldrich Co.) according to the kit protocol and reacting for 10 minutes at room temperature. The absorbance of the colored reaction solution was measured at 520 nm using a microplate reader. Nitrite concentration was calculated using the standard curve.
  • Raw 264.7 cells were seeded in a 24-well plate at a density of 2.5 ⁇ 10 5 cells/well and incubated in an incubator under 5% CO 2 and 37°C conditions. After culturing for 24 hours, 1 ⁇ g/ml of LPS (Lipopolysaccharides, Sigma-Aldrich, USA) (from Escherichia coli 055:B5) was added to the sample using DMEM medium supplemented with 1% FBS and 1% P/S. It was diluted and cultured for 24 hours.
  • LPS Lipopolysaccharides
  • each cell culture was taken and centrifuged at 13000 rpm for 10 minutes, and the PGE2 content of the supernatant obtained through centrifugation was measured. All samples were stored frozen at -20°C until quantification, and PGE2 was quantified using a mouse enzyme-linked immunosorbent assay (ELISA) kit (Cat.# KGE004B, R&D Systems Inc., Minneapolis, MN, USA). The R2 value of the standard curve was over 0.99.
  • ELISA mouse enzyme-linked immunosorbent assay
  • qPCR was performed.
  • RNA was precipitated using 100% isopropanol and washed with 75% ethanol.
  • the extracted RNA was dissolved in distilled water from which nucleic acid degrading enzymes were removed, and then the concentration was measured using a nanospectrophotometer.
  • cDNA was synthesized using Maxime RT pre-Mix (Oligo dt 15 Primer) (iNtRON Biotechnology, Korea) according to the method provided by the manufacturer.
  • KAPA SYBR fast qPCR Kit KAPA biosystems, USA
  • qPCR conditions were set to 1 cycle at 95°C for 5 minutes, 35 cycles at 96°C for 20 seconds, 60°C for 20 seconds, 72°C for 20 seconds, and 1 cycle at 72°C for 5 minutes.
  • Light Cycler 96 software provided by the manufacturer (Roche Applied Science) was used for data analysis. Quantitative results were compared with reference mRNA (GAPDH) using the 2 - ⁇ CT method (Livak & Schmittgen, 2001).
  • iNOS and COX-2 expression was significantly increased in the LPS-only treated group compared to the control group (LPS and sample untreated group), and iNOS and COX-2 expression was significantly increased in the sample-treated group compared to the LPS-only treated group. It decreased significantly in a concentration-dependent manner.
  • IL-1 ⁇ (NCBI Gene ID: 16176) (Interleukin-1 ⁇ , Cat.# MLB00C), TNF- ⁇ (NCBI Gene ID: 21926) (tumor necrosis factor- ⁇ , Cat.# MTA00B), IL-6 (NCBI Gene ID: 16193) (Interleukin-6, Cat.# DY406-05) and IL -10 (NCBI Gene ID: 16153) (Interleukin-10, Cat.# M1000B-1)
  • the amount of cytokine production was measured using an ELISA kit. The experiment was conducted using Raw 264.7 cell culture supernatant according to the kit protocol. The absorbance of the colored reaction solution was measured at 450 nm using a microplate reader. The concentration of each cytokine was calculated using the standard curve.
  • the production of IL-1 ⁇ , IL-6, and TNF- ⁇ was significantly increased in the LPS-only treatment group compared to the control group (LPS and sample untreated group), and the above-described production in the sample-treated group compared to the LPS-only treatment group.
  • the production of three types of cytokines was significantly reduced.
  • IL-10 production significantly increased in the LPS-only treatment group and the sample-treatment group, and the increase in IL-10 production was more significant in the sample-treatment group compared to the LPS-only treatment group.
  • TEER trans-epithelial electrical resistance
  • LPS Lipopolysaccharides, Sigma-Aldrich, USA
  • HBSS Hank's Balanced Salt Solution
  • TEER Miller-ERS equipment
  • the TEER value decreased over time in all experimental groups, and the decrease in TEER value was most significant in the LPS-only treatment group, and the TEER value decreased in the sample treatment group compared to the LPS-only treatment group. It was confirmed that it was suppressed.
  • IPEC-J2 cells were treated with 1 ⁇ g/mL of LPS for 48 hours, and then cells were permeated using 4 kDa FITC-dextran (Sigma-Aldrich, USA). Ambient permeability was measured.
  • FITC-dextran dissolved in HBSS was dispensed to the LPS-treated cells into the upper well at a concentration of 1 mg/ml, and incubated for 4 hours, followed by Tecan Reader (excitation, 492 nm; emission, 520 nm, Tecan Group Ltd., Switzerland). ) was used to measure paracellular permeability.
  • IPEC-J2 cells cultured with LPS for 48 hours were washed once with DPBS (Dulbecco's phosphate-buffered saline), and 100-150 ⁇ l of PRO-PREP reagent was added to each well. After dispensing, the cells were recovered in a tube using a scraper. The recovered cells were left on ice for 30 minutes and then centrifuged at 13000 rpm and 4°C for 5 minutes to obtain a supernatant.
  • DPBS Dynabecco's phosphate-buffered saline
  • the supernatant was quantified for protein using a Bradford assay, and 50 ⁇ g of protein was separated using 8% SDS-PAGE and then transferred to a PVDF membrane (Polyvinylidene Fluoride membrane). Afterwards, block for 1 hour using blocking buffer (4% nonfat dry milk, 10mM Tris, 100mM NaCl, and 0.1% Tween 20, pH 7.5) and wash with 1% Tween 20-PBS three times for 15 minutes each. did.
  • blocking buffer 4% nonfat dry milk, 10mM Tris, 100mM NaCl, and 0.1% Tween 20, pH 7.5
  • ZO-1 NCBI Gene ID: 100736682
  • Occludin NCBI Gene ID: 397236
  • Claudin-1 NCBI Gene ID: 100625166
  • ⁇ -actin antibodies primary antibodies
  • enteritis was induced by providing 3% DSS (Dextran sulfate sodium salt) as a negative water to the animal model of Experimental Example 3 above.
  • DSS Extran sulfate sodium salt
  • 50mg/kg of 5-ASA (5-aminosalicyclic acid) was used as a positive control group, and samples and 5-ASA were administered once daily from the start date of 3% DSS intake using a disposable syringe (1 ml) attached to a sonde for oral administration. It was orally administered into the stomach of an animal model for 5 weeks.
  • the experimental group was specifically set as follows.
  • Sample treatment group (DSS+KM2): 3% DSS intake + oral administration of 2g/kg sample
  • DAI score body weight and disease activity index score
  • DSS intake weight loss and occurrence of bloody stools significantly increased in the negative control group, positive control group, and sample treatment group, and disease activity increased compared to the control group, and in the positive control group and sample treatment group compared to the negative control group.
  • the animal model was sacrificed by fasting for 24 hours, dissected, blood was collected from the heart, and the appearance, abdominal cavity, and thoracic cavity were visually observed. Afterwards, the large intestine was removed and the length of the large intestine was measured.
  • the animal model was sacrificed, blood was collected from the heart, centrifuged immediately, and serum was separated. The separated serum was stored in an ultra-low temperature freezer at -80°C until analysis. did. Afterwards, the cytokine content was measured using a spectrophotometer and analysis kit.
  • the colonic tissue of the animal model was fixed with 10% formaldehyde, a paraffin block was prepared, and H&E (Hematoxylin & Eosin) staining was performed. Quantitative image analysis was performed after photographing the stained tissue slides. In addition, alcian blue staining was performed to analyze the degree of intestinal mucus secretion.
  • the structure and size of the epithelial tissue forming the colon surface changed irregularly in the negative control group (DSS) compared to the control group (CON), and the infiltration of inflammatory cells increased.
  • DSS negative control group
  • CON control group
  • DS+ASA positive control group
  • DSS+KM2 sample treatment group
  • the degree of blue staining decreased in the negative control group compared to the control group in which the mucosal layer was stained blue, whereas the degree of blue staining increased in the positive control group and sample treatment group compared to the negative control group.
  • the number of Clostridium butyricum strains increased significantly.
  • the number of Bifidobacterium pseudolongum strains significantly increased in the negative control group compared to the sample treatment group.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Mycology (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • Microbiology (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Epidemiology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pain & Pain Management (AREA)
  • Molecular Biology (AREA)
  • Rheumatology (AREA)
  • Nutrition Science (AREA)
  • Engineering & Computer Science (AREA)
  • Food Science & Technology (AREA)
  • Polymers & Plastics (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

La présente invention concerne une composition de microbiome d'un surnageant de culture de fermentation de souche KM2 de Lactiplantibacillus plantarum présentant un effet anti-inflammatoire. Il a été confirmé que le surnageant de culture de fermentation de la souche présente une activité anti-inflammatoire, telle que la suppression de l'activité d'oxyde nitrique et la régulation de l'expression des cytokines inflammatoires, et ainsi, le surnageant de culture de fermentation peut être utile en tant que composition pour prévenir, traiter ou atténuer des maladies inflammatoires.
PCT/KR2023/016089 2022-10-18 2023-10-18 Composition de microbiome de surnageant de culture de fermentation de souche km2 de lactiplantibacillus plantarum présentant un effet anti-inflammatoire WO2024085623A1 (fr)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
KR10-2022-0134158 2022-10-18
KR20220134158 2022-10-18
KR1020230138331A KR102654005B1 (ko) 2022-10-18 2023-10-17 항염증 효능이 있는 락티플랜티바실러스 플란타룸 km2 균주 발효 배양상등액의 마이크로바이옴 조성물
KR10-2023-0138331 2023-10-17

Publications (1)

Publication Number Publication Date
WO2024085623A1 true WO2024085623A1 (fr) 2024-04-25

Family

ID=90637746

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2023/016089 WO2024085623A1 (fr) 2022-10-18 2023-10-18 Composition de microbiome de surnageant de culture de fermentation de souche km2 de lactiplantibacillus plantarum présentant un effet anti-inflammatoire

Country Status (2)

Country Link
KR (1) KR102654005B1 (fr)
WO (1) WO2024085623A1 (fr)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20220039617A (ko) * 2020-09-21 2022-03-29 국민바이오 주식회사 숙성 육류에서 분리된 신규 유산균 및 이의 용도
KR20220135180A (ko) * 2021-03-26 2022-10-06 서울대학교산학협력단 유산균의 생리활성 효능 증진용 조성물

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20220039617A (ko) * 2020-09-21 2022-03-29 국민바이오 주식회사 숙성 육류에서 분리된 신규 유산균 및 이의 용도
KR20220135180A (ko) * 2021-03-26 2022-10-06 서울대학교산학협력단 유산균의 생리활성 효능 증진용 조성물

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
FENGLIAN MA: "Lactiplantibacillus plantarum-12 Alleviates Inflammation and Colon Cancer Symptoms in AOM/DSS-Treated Mice through Modulating the Intestinal Microbiome and Metabolome", NUTRIENTS, M D P I AG, CH, vol. 14, no. 9, 3 May 2022 (2022-05-03), CH , pages 1916, XP093160927, ISSN: 2072-6643, DOI: 10.3390/nu14091916 *
YANG SOO-YEON, CHAE SEUNG A, BANG WON YEONG, LEE MINJEE, BAN O-HYUN, KIM SOO-JUNG, JUNG YOUNG HOON, YANG JUNGWOO: "Anti-inflammatory potential of Lactiplantibacillus plantarum IDCC 3501 and its safety evaluation.", BRAZILIAN JOURNAL OF MICROBIOLOGY, SOCIEDADE BRASILEIRA DE MICROBIOLOGIA,BRAZILIAN SOCIETY FOR MICROBIOLOGY, BR, vol. 52, no. 4, 1 December 2021 (2021-12-01), BR , pages 2299 - 2306, XP009553994, ISSN: 1517-8382, DOI: 10.1007/s42770-021-00603-2 *

Also Published As

Publication number Publication date
KR102654005B1 (ko) 2024-04-04

Similar Documents

Publication Publication Date Title
CA3149515A1 (fr) Souche d' akkermansia muciniphila eb-amdk19 et son utilisation
WO2020116733A1 (fr) Nouvelle souche atg-f4 de lactobacillus reuteri possédant une fonction d'amélioration de la sécrétion de dopamine et composition pharmaceutique la comprenant pour la prévention ou le traitement de la psychopathie
WO2018030732A1 (fr) Nanovésicules dérivées de bactéries du genre bacillus et leur utilisation
WO2020091166A1 (fr) Nouvelles bactéries lactiques et leur utilisation
WO2018105926A1 (fr) Composition anti-allergique contenant un extrait d'herbe médicinale composite fermenté par des lactobacilles à titre de principe actif
WO2019004668A1 (fr) Nanovésicule dérivée de bactéries du genre proteus et son utilisation
WO2012093755A1 (fr) Composition comprenant des vésicules extracellulaires provenant d'une nourriture fermentée et utilisation de celle-ci
US20230407241A1 (en) A novel probiotic streptococcus salivarius strain and its uses
WO2021230581A1 (fr) Découverte d'une nouvelle akkermansia muciniphila ak32 et application correspondante pour la prévention ou le traitement d'une lésion intestinale
WO2019172598A1 (fr) Nanovésicules dérivées de bactéries lactobacillus sp., et leur utilisation
WO2024085623A1 (fr) Composition de microbiome de surnageant de culture de fermentation de souche km2 de lactiplantibacillus plantarum présentant un effet anti-inflammatoire
Parthasarathy et al. Trichuris suis excretory secretory products (ESP) elicit interleukin-6 (IL-6) and IL-10 secretion from intestinal epithelial cells (IPEC-1)
WO2019156449A1 (fr) Nanovésicules dérivées de bactéries du genre lactococcus et leur utilisation
TW201636035A (zh) 新穎副乾酪乳桿菌副乾酪亞種k66
WO2023058801A1 (fr) Composition pour soulager, prévenir ou traiter un trouble intestinal, comprenant une souche de lactobacillus acidophilus kbl402 ou kbl409
WO2022060199A1 (fr) Nouvelles bactéries lactiques isolées à partir de viande rassise, et leur utilisation
WO2021242057A1 (fr) Souche de lactobacillus sp., reticulum endoplasmique dérivé de cette dernière, et utilisation anti-inflammatoire et antibactérienne associée
KR102323673B1 (ko) HtrA 샤페론 프로바이오틱스의 제조방법 및 이를 통해 제조된 장 마이크로바이옴 조절 효능이 있는 염증성 장질환 치료용 조성물
WO2021242056A1 (fr) Souche de l'espèce bifidobacterium et vésicule extracellulaire dérivée de cette dernière, et leurs utilisations anti-inflammatoires et antibactériennes
WO2021071292A9 (fr) Composition utilisant une souche de lactobacillus plantarum ou autre et destinée au soulagement de maladies respiratoires
WO2019164230A1 (fr) Nano-vésicules dérivées de bactéries de l'espèce cupriavidus et utilisation associée
WO2012093754A1 (fr) Composition comprenant des vésicules extracellulaires provenant de l'intérieur d'un corps de mammifère et utilisation de celle-ci
CN117279652A (zh) 含黑参有效成分的预防、改善或治疗冠状病毒感染的组合物
WO2019151825A1 (fr) Nanovésicules issues de bactéries bifidobacterium, et leur utilisation
WO2023121334A1 (fr) Composition comprenant un extrait de cardamine flexuosa en tant que principe actif pour la prévention, l'atténuation ou le traitement d'une maladie respiratoire