WO2021040186A1 - Akkermansia muciniphila eb-amdk27 strain and use thereof - Google Patents

Akkermansia muciniphila eb-amdk27 strain and use thereof Download PDF

Info

Publication number
WO2021040186A1
WO2021040186A1 PCT/KR2020/006176 KR2020006176W WO2021040186A1 WO 2021040186 A1 WO2021040186 A1 WO 2021040186A1 KR 2020006176 W KR2020006176 W KR 2020006176W WO 2021040186 A1 WO2021040186 A1 WO 2021040186A1
Authority
WO
WIPO (PCT)
Prior art keywords
strain
akkermansia muciniphila
amdk27
disease
group
Prior art date
Application number
PCT/KR2020/006176
Other languages
French (fr)
Inventor
Jae-Gu SEO
Joo-Hyun SHIN
Do-Kyung Lee
Original Assignee
Enterobiome Inc.
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 Enterobiome Inc. filed Critical Enterobiome Inc.
Priority to EP20824072.1A priority Critical patent/EP3802787A4/en
Priority to US17/258,510 priority patent/US11439670B2/en
Priority to JP2021503148A priority patent/JP7055520B2/en
Priority to AU2020339348A priority patent/AU2020339348B2/en
Publication of WO2021040186A1 publication Critical patent/WO2021040186A1/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • 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
    • C12N1/20Bacteria; Culture media therefor
    • 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
    • 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
    • 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
    • A23L33/30Dietetic or nutritional methods, e.g. for losing weight
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • 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]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • 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
    • C12N1/20Bacteria; Culture media therefor
    • C12N1/205Bacterial isolates
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23VINDEXING SCHEME RELATING TO FOODS, FOODSTUFFS OR NON-ALCOHOLIC BEVERAGES AND LACTIC OR PROPIONIC ACID BACTERIA USED IN FOODSTUFFS OR FOOD PREPARATION
    • A23V2002/00Food compositions, function of food ingredients or processes for food or foodstuffs
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23VINDEXING SCHEME RELATING TO FOODS, FOODSTUFFS OR NON-ALCOHOLIC BEVERAGES AND LACTIC OR PROPIONIC ACID BACTERIA USED IN FOODSTUFFS OR FOOD PREPARATION
    • A23V2200/00Function of food ingredients
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23VINDEXING SCHEME RELATING TO FOODS, FOODSTUFFS OR NON-ALCOHOLIC BEVERAGES AND LACTIC OR PROPIONIC ACID BACTERIA USED IN FOODSTUFFS OR FOOD PREPARATION
    • A23V2200/00Function of food ingredients
    • A23V2200/30Foods, ingredients or supplements having a functional effect on health
    • A23V2200/32Foods, ingredients or supplements having a functional effect on health having an effect on the health of the digestive tract
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23VINDEXING SCHEME RELATING TO FOODS, FOODSTUFFS OR NON-ALCOHOLIC BEVERAGES AND LACTIC OR PROPIONIC ACID BACTERIA USED IN FOODSTUFFS OR FOOD PREPARATION
    • A23V2200/00Function of food ingredients
    • A23V2200/30Foods, ingredients or supplements having a functional effect on health
    • A23V2200/324Foods, ingredients or supplements having a functional effect on health having an effect on the immune system
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23VINDEXING SCHEME RELATING TO FOODS, FOODSTUFFS OR NON-ALCOHOLIC BEVERAGES AND LACTIC OR PROPIONIC ACID BACTERIA USED IN FOODSTUFFS OR FOOD PREPARATION
    • A23V2200/00Function of food ingredients
    • A23V2200/30Foods, ingredients or supplements having a functional effect on health
    • A23V2200/3262Foods, ingredients or supplements having a functional effect on health having an effect on blood cholesterol
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23VINDEXING SCHEME RELATING TO FOODS, FOODSTUFFS OR NON-ALCOHOLIC BEVERAGES AND LACTIC OR PROPIONIC ACID BACTERIA USED IN FOODSTUFFS OR FOOD PREPARATION
    • A23V2200/00Function of food ingredients
    • A23V2200/30Foods, ingredients or supplements having a functional effect on health
    • A23V2200/328Foods, ingredients or supplements having a functional effect on health having effect on glycaemic control and diabetes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12RINDEXING SCHEME ASSOCIATED WITH SUBCLASSES C12C - C12Q, RELATING TO MICROORGANISMS
    • C12R2001/00Microorganisms ; Processes using microorganisms
    • C12R2001/01Bacteria or Actinomycetales ; using bacteria or Actinomycetales

Definitions

  • the present invention relates to a novel Akkermansia muciniphila strain and the use thereof, and more particularly to a novel Akkermansia muciniphila strain which has the effect of preventing or treating inflammatory disease or metabolic disease, and a pharmaceutical composition for the prevention or treatment of inflammatory disease or metabolic disease, which contains the Akkermansia muciniphila strain.
  • Inflammatory diseases refer to diseases that are caused by inflammatory cytokines, such as tumor necrosis factor- ⁇ (TNF- ⁇ ), interleukin-1 (IL-1), IL-6, prostaglandin, leukotriene or nitric oxide (NO), which are secreted from immune cells such as macrophages due to an exaggerated reaction of the human body's immune system to harmful stimuli such as inflammatory agents or radiation.
  • TNF- ⁇ tumor necrosis factor- ⁇
  • IL-1 interleukin-1
  • IL-6 interleukin-6
  • prostaglandin prostaglandin
  • leukotriene leukotriene
  • NO nitric oxide
  • Inflammatory diseases such as inflammatory bowel disease (IBD) are a family of chronic, recurrent, and tissue-destructive diseases, characterized by dysfunction of mucosal T cells, abnormal cytokine production and cellular inflammation, which cause mucosal damage. Inflammatory diseases are serious diseases that dramatically decrease the quality of life, but the causes thereof still remain unclear.
  • IBD inflammatory bowel disease
  • one of the current treatments is a method of removing colon ulcers by colon resection, but this method may lower the quality of life and increase the risk of complications.
  • other medical approaches include a method of controlling inflammation by suppressing the immune system using an immunomodulator, but the immunomodulator may induce an immune-lowering condition that makes the patient susceptible to various other diseases.
  • Korean Patent Application Publication No. 2011-0095929 discloses a Lactobacillus reuteri strain which interferes with in vivo lipid absorption
  • Korean Patent No. 0996577 discloses a Lactobacillus curvatus strain which lowers blood cholesterol levels and inhibits obesity
  • Korean Patent Application Publication No. 2011-0010015 discloses a Lactobacillus johnsonii which inhibits obesity while lowering blood cholesterol levels
  • Korean Patent Application Publication No. 2019-0034796 discloses a Lactobacillus plantarum strain which has antioxidant and anti-inflammatory effects.
  • the probiotics as described above have insignificant effects on the amelioration of inflammatory disease or metabolic disease, and hence have a problem in that they are difficult to use as new preventive and therapeutic tools.
  • the present invention is intended to overcome the above-described limitation of the conventional art, and an object of the present invention is to provide a next-generation probiotic strain which inhibits inflammatory response by reducing or inhibiting the release of one or more inflammatory factors, such as tumor necrosis factor- ⁇ (TNF- ⁇ ), IL-8, IL-6 and IL-1 ⁇ , and which is effective in preventing or treating inflammatory disease, obesity, obesity complications or metabolic disease by inhibiting adipocyte differentiation.
  • TNF- ⁇ tumor necrosis factor- ⁇
  • IL-8 interleukin-6
  • IL-1 ⁇ interleukin-1 ⁇
  • Another object of the present invention is to provide a pharmaceutical composition effective for the prevention or treatment of inflammatory disease or metabolic disease, which contains an Akkermansia muciniphila strain, which is the next-generation probiotic strain.
  • Another object of the present invention is to provide a pharmaceutical composition for suppressing appetite, which contains the Akkermansia muciniphila strain.
  • Still another object of the present invention is to provide a food useful for ameliorating inflammatory disease or metabolic disease, which contains the Akkermansia muciniphila strain.
  • One aspect of the present invention for achieving the above objects is directed to an Akkermansia muciniphila EB-AMDK27 strain (accession number KCTC 13758BP).
  • Another aspect of the present invention for achieving the above objects is directed to a pharmaceutical composition for preventing or treating inflammatory disease or metabolic disease, which contains the Akkermansia muciniphila EB-AMDK27 strain (accession number KCTC 13758BP), a culture thereof, or a dried product thereof.
  • Another aspect of the present invention for achieving the above objects is directed to a pharmaceutical composition for suppressing appetite, which contains the Akkermansia muciniphila EB-AMDK27 strain (accession number KCTC 13758BP), a culture thereof, or a dried product thereof.
  • a pharmaceutical composition for suppressing appetite which contains the Akkermansia muciniphila EB-AMDK27 strain (accession number KCTC 13758BP), a culture thereof, or a dried product thereof.
  • Still another aspect of the present invention for achieving the above objects is directed to a food for preventing or ameliorating inflammatory disease or metabolic disease, which contains the Akkermansia muciniphila EB-AMDK27 strain (accession number KCTC 13758BP), a culture thereof, or a dried product thereof.
  • the novel Akkermansia muciniphila EB-AMDK27 strain of the present invention inhibits inflammatory response by reducing or inhibiting the release of inflammatory factors, and is effective in treating inflammatory disease, particularly bowel inflammation, by maintaining the stability of intestinal microbiota.
  • the pharmaceutical composition containing, as an active ingredient, the Akkermansia muciniphila EB-AMDK27 strain of the present invention may be used as a pharmaceutical composition or a functional food composition for treating and/or preventing inflammatory disease or metabolic disease.
  • novel Akkermansia muciniphila EB-AMDK27 strain of the present invention and the pharmaceutical composition containing the same may inhibit body weight gain and body fat accumulation, lower insulin resistance, lower total blood cholesterol levels, reduce the level of blood GPT that is a hepatotoxicity indicator, and reduce the levels of inflammatory cytokines, including IL-8, TNF- ⁇ , IL-6 and IL-1 ⁇ .
  • they may be useful for preventing or treating diabetes, obesity, insulin resistance, fatty liver, hyperlipidemia, or metabolic disease, which is associated with these factors.
  • the pharmaceutical composition of the present invention exhibits the effect of substantially inhibiting lipid accumulation by containing, as an active ingredient, the Akkermansia muciniphila EB-AMDK27 strain that reduces intracellular lipid accumulation, reduces expression of the adipocyte differentiation-related factor PPAR ⁇ , and also reduces the mRNA expression of CEBP ⁇ , aP2, CD36, ACC1, LPL (lipoprotein lipase), LDLR or FAS.
  • composition for suppressing appetite containing the Akkermansia muciniphila EB-AMDK27 strain (KCTC 13758BP) of the present invention may suppress appetite by stimulating the secretion of appetite suppressant hormones.
  • FIG. 1 shows micrographs of the Akkermansia muciniphila EB-AMDK27 strain (KCTC 13758BP) of the present invention and the type strain Akkermansia muciniphila ATCC BAA-835;
  • FIG. 2 shows the results of PCR analysis of the Akkermansia muciniphila EB-AMDK27 strain of the present invention and the Akkermansia muciniphila ATCC BAA-835 strain;
  • FIG. 3 shows the results of testing the hemolytic activities of the Akkermansia muciniphila EB-AMDK27 strain of the present invention and the Akkermansia muciniphila ATCC BAA-835 strain;
  • FIG. 4 shows the results of RAPD (Random Amplified Polymorphic DNA) analysis of the Akkermansia muciniphila EB-AMDK27 strain of the present invention and the Akkermansia muciniphila ATCC BAA-835 strain;
  • RAPD Random Amplified Polymorphic DNA
  • FIG. 5 shows the phylogenetic relationship between the Akkermansia muciniphila EB-AMDK27 strain of the present invention and other Akkermansia muciniphila strains;
  • FIG. 6 depicts graphs showing the relative mRNA expression levels of the cytokines IL-6, IL-8, IL-1 ⁇ and TNF- ⁇ , which demonstrate the anti-inflammatory effect of the Akkermansia muciniphila EB-AMDK27 strain of the present invention
  • FIG. 7 depicts photographs and a graph, which show the degree of lipid accumulation in 3T3-L1 cells after treatment with the Akkermansia muciniphila EB-AMDK27 strain of the present invention
  • FIG. 8 depicts graphs showing the relative mRNA expression levels of PPAR ⁇ , CEBP ⁇ , aP2, CD36, ACC1, LPL (lipoprotein lipase), LDLR and FAS in preadipocytes after treatment with the Akkermansia muciniphila EB-AMDK27 strain of the present invention
  • FIG. 9 shows the results of analyzing changes in the body weight of a group administered with the Akkermansia muciniphila EB-AMDK27 strain of the present invention, a control group (DM), and a group administered with the Akkermansia muciniphila ATCC BAA-835 strain;
  • FIG. 10 shows the results of analyzing changes in the weight of subcutaneous fat, epididymal fat and mesenteric fat of a group administered with the Akkermansia muciniphila EB-AMDK27 strain of the present invention, a control group (DM), and a group administered with the Akkermansia muciniphila ATCC BAA-835 strain;
  • FIG. 11 shows the results of analyzing changes in the glucose tolerance of a group administered with the Akkermansia muciniphila EB-AMDK27 strain of the present invention, a control group (DM), and a group administered with the Akkermansia muciniphila ATCC BAA-835 strain;
  • FIG. 12 shows the results of measuring changes in the blood insulin, cholesterol and glutamic pyruvic transaminase (GPT) concentrations in a group administered with the Akkermansia muciniphila EB-AMDK27 strain of the present invention, a control group (DM), and a group administered with the Akkermansia muciniphila ATCC BAA-835 strain, by ELISA;
  • GPT glutamic pyruvic transaminase
  • FIG. 13 shows Alcian blue staining images of a normal group, a high-fat diet group (HFD), a group administered with Garcinia cambogia (GC), a group administered with the Akkermansia muciniphila ATCC BAA-835 strain (BAA-835), and a group administered with the Akkermansia muciniphila EB-AMDK27 strain of the present invention (EB-AMDK27), and depicts a graph showing the positive area in each group, obtained by quantifying the images;
  • HFD high-fat diet group
  • GC Garcinia cambogia
  • BAA-835 Akkermansia muciniphila ATCC BAA-835
  • EB-AMDK27 a group administered with the Akkermansia muciniphila EB-AMDK27 strain of the present invention
  • FIG. 14 shows changes in the expression of TLR4, TLR2, GLP-1, PYY, IL-6, TNF- ⁇ , MCP-1, IL-10, ZO-1 and occluding in the enterocytes of mice of each test group;
  • FIGS. 15 and 16 are optical micrographs of hepatocytes and lipid droplets, obtained after H & E staining, and are graphs showing liver steatosis scores in each group;
  • FIG. 17 depicts photographs and a graph, which show the degree of lipid accumulation in the mesenteric adipose tissue of each test animal after treatment with the Akkermansia muciniphila EB-AMDK27 strain of the present invention
  • FIG. 18 shows a comparison of linear discriminant analysis results between test groups
  • FIG. 19 depicts graphs showing the relative abundance of intestinal microbiomes following administration of the Akkermansia muciniphila ATCC BAA-835 strain (BAA-835) and the Akkermansia muciniphila EB-AMDK27 strain of the present invention (EB-AMDK27) in one Example of the present invention.
  • inflammatory disease refers to a disease that is caused by an inflammatory reaction in the mammalian body.
  • Representative examples of inflammatory disease include: respiratory diseases such as asthma, chronic obstructive pulmonary disease, and rhinitis; skin diseases such as atopic dermatitis; digestive diseases such as gastritis and inflammatory enteritis; arteriosclerosis, sepsis, inflammatory joint disease, inflammatory brain disease, and the like.
  • treating refers to reversing or alleviating inflammatory disease, metabolic disease or one or more symptoms of the disease, or inhibiting the progress thereof, unless stated otherwise.
  • prevention is intended to include reducing the likelihood of developing inflammatory disease or metabolic disease.
  • cytokine refers to a secreted protein that affects the functions of other cells. Particularly, it relates to the modulation of interactions between cells of the immune system or cells involved in the inflammatory response.
  • cytokines include, but are not necessarily limited to, interleukin 1 (IL-1), preferably interleukin IL-1 ⁇ , interleukin-6 (IL-6), interleukin 8 (IL-8), and tumor necrosis factor- ⁇ (TNF- ⁇ ).
  • IL-1 interleukin 1
  • IL-6 interleukin-6
  • IL-8 interleukin 8
  • TNF- ⁇ tumor necrosis factor- ⁇
  • appetite suppressing appetite refers to any action that suppresses or delays appetite by administering the strain or composition to stimulate the secretion of diet-related hormones (e.g., appetite suppressant hormones).
  • One aspect of the present invention is directed to a next-generation probiotic strain which is an Akkermansia muciniphila EB-AMDK27 strain (accession number KCTC 13758BP).
  • the strain has the 16s rRNA gene of SEQ ID NO: 1.
  • the Akkermansia muciniphila EB-AMDK27 strain of the present invention is a monococcus or diplococcus isolated from a health Korean's feces, which is an elliptical cell of 0.5 to 1 ⁇ m in size. Also, it is a mucus-degrading bacterium which is anaerobic, non-motile, and gram-negative. It does not form an endospore.
  • the Akkermansia muciniphila EB-AMDK27 strain is capable of producing several mucolytic enzymes, and thus may use mucus as carbon and nitrogen sources.
  • the Akkermansia muciniphila EB-AMDK27 strain can metabolize various carbon sources, including galactose, N-acetylglucosamine, and lactose, and produces, as main metabolites, short-chain fatty acids such as propionic acid and acetic acid.
  • the Akkermansia muciniphila EB-AMDK27 strain is capable of exhibiting an anti-inflammatory effect by inhibiting the expression of inflammatory cytokines, specifically IL-8, IL-6, IL-1 ⁇ , and TNF- ⁇ .
  • Tight junctions formed between epithelial cells are particularly important for the barrier function of epithelial cells.
  • the tight junctions are a type of cell-cell junction, and are so strong that it appears under an electron microscope that the cell membranes of two adjacent cells are fused.
  • the barrier function is a function that blocks foreign substances from passing between epithelial cells, and is important in both blood vessels and digestive tracts.
  • Constituent molecules that are involved in formation of the tight junctions typically include a membrane protein called occludin and cytoplasmic ZO proteins as well as proteins such as inulin are also involved in the formation of the tight junctions. Through the interaction between these proteins, the structure and function of the tight junctions are completed.
  • the Akkermansia muciniphila EB-AMDK27 strain increases the expression of the tight junction proteins ZO-1 (zonular occludens-1, p ⁇ 0.01) and occludin and blocks inflammatory proteins from migrating into tissues, thereby promoting mucous regeneration and alleviating inflammation of colonic mucosa.
  • ZO-1 zonular occludens-1, p ⁇ 0.01
  • obesity When obesity is induced, it causes abnormalities in visceral adipose tissue, excessive secretion of tumor necrosis factor, infiltration of immune cells such as macrophages into adipose tissue, and increased expression of inflammatory cytokines. This results in chronic inflammation of adipose tissue, and the chronic inflammatory response reduces insulin sensitivity and induces glucose tolerance, leading to diabetes. Therefore, suppression of adipose tissue hypertrophy and inflammatory response can exhibit an anti-obesity effect, and as a result, can also be useful against anti-metabolic diseases.
  • the novel Akkermansia muciniphila EB-AMDK27 strain of the present invention may inhibit body weight gain and body fat accumulation, lower insulin resistance, lower total blood cholesterol levels, reduce the level of blood glutamic pyruvic transaminase (GPT) that is a hepatotoxicity indicator, and reduce the levels of inflammatory cytokines.
  • GPT blood glutamic pyruvic transaminase
  • it may be useful for preventing or treating diabetes, obesity, obesity-related disease, insulin resistance, fatty liver, hyperlipidemia, or metabolic disease, which is associated with these factors.
  • the metabolic disease may be a disease in which various metabolic diseases such as diabetes and obesity appear simultaneously in one person.
  • the Akkermansia muciniphila EB-AMDK27 strain of the present invention has therapeutic efficacy against both inflammatory disease and metabolic disease, and thus can provide the remarkable effect of comprehensively treating various diseases that are highly correlated with inflammation or obesity.
  • the novel Akkermansia muciniphila EB-AMDK27 strain of the present invention and a pharmaceutical composition containing the same exhibit the effect of substantially inhibiting lipid accumulation by containing, as an active ingredient, the Akkermansia muciniphila EB-AMDK27 strain that reduces intracellular lipid accumulation, reduces expression of the adipocyte differentiation-related factor PPAR ⁇ and also reduces the mRNA expression of CEBP ⁇ , aP2, CD36, ACC1, LPL (lipoprotein lipase), LDLR, or FAS.
  • Another aspect of the present invention is directed to a pharmaceutical composition for preventing or treating inflammatory disease or metabolic disease, which contains an Akkermansia muciniphila EB-AMDK27 strain (accession number KCTC 13758BP), a culture thereof, or a dried product thereof.
  • an Akkermansia muciniphila EB-AMDK27 strain accession number KCTC 13758BP
  • the pharmaceutical composition of the present invention may contain a probiotic form of the Akkermansia muciniphila EB-AMDK27 strain or a pasteurized form of the Akkermansia muciniphila EB-AMDK27 strain.
  • Pasteurization of the Akkermansia muciniphila EB-AMDK27 strain refers to heating at temperature equal to or higher than 50°C and lower than 100°C for 10 minutes or more.
  • the strain may be pasteurized at a temperature of 70°C for 30 minutes.
  • a pasteurized form of the Akkermansia muciniphila EB-AMDK27 strain can reduce body fat accumulation to a greater extent compared to a probiotic form of the strain.
  • the pasteurized form of the Akkermansia muciniphila EB-AMDK27 strain is more effective than the probiotic form has not been accurately identified, it can be presumed that when the Akkermansia muciniphila EB-AMDK27 strain is pasteurized, the cell wall components (such as Amuc_1100) or membrane proteins of the strain enhance metabolic benefits in a host.
  • the beneficial effect of the strain of the present invention is presumed to be related to a gene cluster (Amuc_1098-Amuc_1102) containing Amuc_1100 and to be attributed to polypeptides that interact with the signaling pathway of toll-like receptor 2 ("TLR2"), which is present on the surface of immune cells located near the barrier of intestinal mucosa and modulates intestinal homeostasis and host metabolism.
  • TLR2 toll-like receptor 2
  • the Amuc-1100 polypeptide is expected to maintain the integrity of the barrier of the intestinal mucosa and interact with TLR2 present on the surface of immune cells to modulate or promote the TLR2 signaling pathway, thereby promoting the secretion of cytokines (e.g., IL- 6, IL-8, and IL-10) from immune cells.
  • cytokines e.g., IL- 6, IL-8, and IL-10
  • Prolipoprotein diacylglyceryl transferase gene (Amuc_1104) is located in close proximity to the gene cluster (Amuc_1098-Amuc_1102).
  • Amuc_1100 can be stably maintained under the temperature condition used during pasteurization, thus contributing to the effect of the pasteurized strain.
  • the pharmaceutical composition of the present invention is effective for the treatment or prevention of inflammatory diseases, particularly inflammatory bowel disease.
  • inflammatory bowel disease examples include Crohn's disease, ulcerative colitis, intestinal Behcet's disease, simple ulcer, radiation enteritis, and ischemic enteritis.
  • the pharmaceutical composition is effective against Crohn's disease or ulcerative colitis.
  • the metabolic disease in the present invention is preferably obesity, insulin resistance, fatty liver, hyperlipidemia, or complications thereof, but is not necessarily limited thereto.
  • the pharmaceutical composition of the present invention is effective for preventing or treating inflammatory bowel disease (IBD), insulin resistance, or dyslipidemia, or for reducing cholesterol levels or body weight.
  • IBD inflammatory bowel disease
  • the pharmaceutical composition of the present invention contains, as an active ingredient, the Akkermansia muciniphila EB-AMDK27 strain at a concentration of 10 8 to 10 12 CFU/g of the composition, or a culture containing viable cells of the Akkermansia muciniphila EB-AMDK27 strain at the same concentration above.
  • the Akkermansia muciniphila EB-AMDK27 strain of the present invention may be recovered by a separation process such as centrifugation, and prepared as a probiotic by drying, for example, freeze-drying, for use.
  • the Akkermansia muciniphila EB-AMDK27 strain of the present invention is oxygen-sensitive, and hence is preferably cultured under anaerobic conditions (90% nitrogen, 5% hydrogen, and 5% carbon dioxide).
  • Components of the liquid medium during culture can affect the growth of the strain and the production of active ingredients. Thus, it is necessary to establish the components and their conditions of the liquid medium, which are optimal for culturing the novel Akkermansia muciniphila EB-AMDK27 strain of the present invention.
  • the liquid medium may contain, as a carbon source, one or more selected from the group consisting of glucose, lactose, galactose, N-acetylglucosamine, mannose, 1-fucose, lactate, formate, acetate, propionate, 1,2-propenediol, and butyrate, but is not limited thereto.
  • it may contain glucose and N-acetylglucosamine.
  • the liquid medium may contain, as a nitrogen source, one or more selected from the group consisting of tryptone, peptone, soy peptone, L-glutamic acid, and ammonium, but is not limited thereto.
  • the liquid medium may contain, as trace elements, one or more selected from the group consisting of KH 2 PH 4 , Na 2 HPO 4 , NaCl, MgCl 2 , CaCl 2 , FeCl 2 , ZnCl 2 , CuCl 2 , MnCl 2 , CoCl 2 , NiCl 2 , Na 2 SeO 3 , Na 2 WO 4 and Na 2 MoO 4 , but is not limited thereto.
  • the liquid medium may have a pH of 6.8 to 7.2, preferably a pH of 7.0.
  • the pH can affect the activity of protein by changing the charge of the amino or carboxyl group of the amino acid, which is a unit of the enzymatic protein important for cell metabolism.
  • changes in the pH in the external environment can affect the ionization of microbial nutrients, which can affect nutrient intake of the microorganism.
  • the liquid medium is preferably a medium containing glucose, N-acetylglucosamine, threonine, soypeptone, or any combination thereof.
  • composition of the present invention may further contain pharmaceutically acceptable carriers and/or excipients, in addition to the active ingredient.
  • the composition may be formulated with various additives, such as a binder, a disintegrant, a coating agent, and a lubricant, which are commonly used in the pharmaceutical industry.
  • composition of the present invention may be formulated in the form of powder, granule, tablet, capsule, or liquid by mixing the Akkermansia muciniphila EB-AMDK27 strain of the present invention with a suitable carrier, excipient, auxiliary active ingredient, etc.
  • the composition of the present invention may be formulated as a product for oral administration.
  • the composition of the present invention may be productized by enteric coating using any known method so that it can pass through the stomach and then reach the small intestine in which the active ingredient microorganism can be rapidly released into the intestines.
  • Excipients that may be used in the present invention include: sugars such as sucrose, lactose, mannitol, or glucose; and starches such as corn starch potato starch, rice starch, or partially pregelatinized starch. Binders that may be used in the present invention include polysaccharides such as dextrin, sodium alginate, carrageenan, guar gum, acacia, and agar; naturally-occurring macromolecular substances such as tragacanth, gelatin, and gluten; cellulose derivatives such as hydroxypropylcellulose, methylcellulose, hydroxypropylmethylcellulose, ethyl cellulose, hydroxypropyl ethyl cellulose, and sodium carboxymethyl cellulose; and polymers such as polyvinylpyrrolidone, polyvinyl alcohol, polyvinyl acetate, polyethylene glycol, polyacrylic acid, polymethacrylic acid, and vinyl acetate resin.
  • sugars such as sucrose, lactose,
  • Disintegrants that may be used in the present invention include: cellulose derivatives such as carboxymethylcellulose, calcium carboxymethylcellulose, low-substituted hydroxypropylcellulose, and cellulose derivatives; and starches such as sodium carboxymethyl starch, hydroxypropyl starch, corn starch, potato starch, rice starch, and partially pregelatinized starch.
  • cellulose derivatives such as carboxymethylcellulose, calcium carboxymethylcellulose, low-substituted hydroxypropylcellulose, and cellulose derivatives
  • starches such as sodium carboxymethyl starch, hydroxypropyl starch, corn starch, potato starch, rice starch, and partially pregelatinized starch.
  • lubricants examples include talc, stearic acid, calcium stearate, magnesium stearate, colloidal silica, hydrous silicon dioxide, and various types of waxes and hydrogenated oils.
  • Coating agents that may be used in the present invention include, but are not necessarily limited to, water-insoluble copolymers such as a dimethylaminoethyl methacrylate-methacrylic acid copolymer, a polyvinylacetal diethylaminoacetate, an ethylacrylate-methacrylic acid copolymer, an ethylacrylate-methylmethacrylate-chlorotrimethylammonium ethylmethacrylate copolymer, and ethyl cellulose; enteric polymers such as a methacrylic acid-ethyl acrylate copolymer, hydroxypropyl methyl cellulose phthalate, hydroxypropyl methyl cellulose acetate succinate; and water-soluble polymers such as methyl cellulose, hydroxypropyl methyl cellulose, polyvinylpyrrolidone, and polyethylene glycol.
  • water-insoluble copolymers such as a dimethylaminoethyl me
  • the dose of the strain as an active ingredient in the composition for preventing or treating inflammatory disease or metabolic disease according to the present invention may be determined in consideration of various factors, including the type of disease, the patient's age, body weight, sex and medical condition, the severity of the condition, and the route of administration.
  • the dose regime can vary widely, but can be routinely determined using standard methods.
  • For an adult patient generally 1 ⁇ 10 6 or more viable cells or pasteurized cells, preferably 1 ⁇ 10 8 to 1 ⁇ 10 12 viable cells or pasteurized cells may be may be administered once or several times as needed.
  • the exact formulation, route of administration, and dosage of the pharmaceutical composition disclosed herein can be determined by a physician by taking into account the patient's condition.
  • Still another aspect of the present invention is directed to a pharmaceutical composition for suppressing appetite, which contains an Akkermansia muciniphila EB-AMDK27 strain (accession number KCTC 13758BP), a culture thereof, or a dried product thereof.
  • an Akkermansia muciniphila EB-AMDK27 strain accession number KCTC 13758BP
  • KCTC 13758BP accession number KCTC 13758BP
  • Glucagon-like peptide-1 GLP-1
  • PYY peptide YY
  • GLP-1 glucagon like peptide 1
  • PYY peptide YY
  • Still another aspect of the present invention is directed to a food for preventing or ameliorating inflammatory disease or metabolic disease, which contains an Akkermansia muciniphila EB-AMDK27 strain (accession number KCTC 13758BP), a culture thereof, or a dried product thereof.
  • a food containing the Akkermansia muciniphila EB-AMDK27 strain of the present invention may be taken as a food or nutritional product, such as milk or dairy products, or as a food supplement or health functional food.
  • examples of the food product include, but are not necessarily to, foods such as dairy products, beverages, juices, soups, or foods for children.
  • the isolated strain would be an Akkermansia muciniphila strain
  • the isolated strain was observed under a microscope, and the results are shown in FIG. 1.
  • PCR analysis was performed using the AM-specific primers shown in Table 1 below, and the results of the analysis are shown in FIG. 2.
  • FIG. 1 A shows an Akkermansia muciniphila ATCC BAA-835 strain, and B is a 1,000 ⁇ magnification micrograph of an Akkermansia muciniphila EB-AMDK27 strain.
  • M represents a DNA size marker
  • lane 1 represents a positive control (ATCC BAA-835)
  • lane 2 represents the Akkermansia muciniphila EB-AMDK27 strain
  • lane 3 represents a negative control (distilled water).
  • the strain was cultured using an API50CH kit (Biomerieux, France), and then whether the strain would grow using each carbohydrate was compared with the type strain (ATCC BAA-835). The results of the comparison are shown in Table 2 below.
  • the Akkermansia muciniphila EB-AMDK27 strain of the present invention differed from the type strain (ATCC BAA-835) with respect to the utilization of ribose, D-fructose, D-mannose, D-lactose and L-fucose.
  • Genomic statistics Strains ATCC BAA-835 EB-AMDK27 Accseeion No. CP001071 CP027003 Assembly level Complete Complete Seq. category Chromosome Chromosome Total size (Mb) 2.6641 2.7342 GC (%) 55.8 55.4 Protein 2,246 2,289 Gene 2,321 2,382 CDS 2,257 2,318 Coding 2,246 2,289 rRNA 9 9 tRNA 52 52 Other RNA 3 3 Pseudogene 11 29 Symmetrical identity (%) - N/A *Symmetrical identity is relative to ATCC BAA-835
  • the Amuc_1100 polypeptide is associated with therapeutic activity against anti-inflammatory or metabolic disease and is a heat-stable protein acting on the Toll-like receptor (TLR) 2 (Plovier et al., 2017).
  • TLR Toll-like receptor
  • the Akkermansia muciniphila EB-AMDK27 strain of the present invention differed from the Akkermansia muciniphila ATCC BAA-835 strain with respect to the Amuc_1100 related gene.
  • the minimum inhibitory concentrations (MICs) of antibiotics for anaerobic bacteria (Piperacillin-Tazobactam (PTZ), Ceftizoxime (CTZ), Chloramphenicol (CHL), Clindamycin (CLI), Meropenem (MEM), Moxifloxacin (MXF), Metronidazole (MTZ), and Ciprofloxacin (CIP)) against the isolated strain were determined by broth microdilution according to the guideline of Clinical & Laboratory Standard Institute (CLSI, 2017), and the results are shown in Table 5 below.
  • MICs minimum inhibitory concentrations
  • the Akkermansia muciniphila EB-AMDK27 strain of the present invention showed susceptibility to all the antibiotics excluding moxifloxacin and ciprofloxacin, which were fluoroquinolone based antibiotics. Therefore, it can be confirmed that the Akkermansia muciniphila strain according to the present invention is a safe strain that is not resistant to most antibiotics.
  • the strain was cultured using a blood agar medium prepared by adding 5% w/v defibrinated sheep blood to tryptic soy agar (17.0 g/L pancreatic digest of casein, 3.0 g/L pancreatic digest of soybean, 2.5 g/L dextrose, 5.0 g/L sodium chloride, 2.5 g/L potassium phosphate, and 15 g/L agar). The results of the culture are shown in FIG. 3.
  • RAPD random amplified polymorphic DNA
  • Genomic DNA was extracted from the isolated strain, and then PCR-RAPD (MyCycler, BIO-RAD, USA) was performed using the extracted genomic DNA as a template and the primers shown in Table 6 below.
  • the PCR product was electrophoresed on 1% agarose gel for 1 hour and 30 minutes, and DNA fragmentation patterns were compared on a UV perforator. The results of the analysis are shown in FIG. 4.
  • the RAPD band pattern of the Akkermansia muciniphila EB-AMDK27 strain of the present invention was analyzed comparatively with that of the Akkermansia muciniphila type strain (ATCC BAA-835), and as a result, it showed a different band pattern. It is known that the RAPD band patterns of Akkermansia muciniphila species are different from each other when the species are different. Thus, it was confirmed that the Akkermansia muciniphila strain isolated in the present invention was different from the type strain.
  • the 16S rRNA gene was amplified using the 27F and 1541R primers shown in Table 7 below, and then sequenced using a 3730xl DNA analyzer.
  • the full-length 16S rRNA gene sequence of the Akkermansia muciniphila EB-AMD 27 strain is set forth in SEQ ID NO: 1.
  • the DNA sequence was analyzed using the DNA Star program and the Cluster V program to determine homology to the type strain.
  • a phylogenetic tree was prepared using the 16S rRNA gene sequences of the Akkermansia muciniphila strain of the present invention and other strains of the same species already published. The prepared phylogenetic tree is shown in FIG. 5.
  • the evolutionary relationship between the 16s rRNA gene sequences was analyzed through the phylogenetic tree, and as a result, it was confirmed that the Akkermansia muciniphila EB-AMDK27 strain was tied to the same group to which the Akkermansia muciniphila type strain (ATCC BAA-835) was tied. This suggests that the Akkermansia muciniphila EB-AMDK27 strain genetically belongs to Akkermansia muciniphila species.
  • the Akkermansia muciniphila strain isolated from human feces, through the biochemical method (API) and molecular biological methods (16s rRNA sequencing, RAPD, and full-length screening) using the Akkermansia muciniphila type strain (ATCC BAA-835) as a control, it was finally confirmed to be a strain belonging to A. muciniphila species.
  • the isolated Akkermansia muciniphila EB-AMDK27 strain was to be a safe strain that can function as probiotics.
  • the isolated strain was named Akkermansia muciniphila EB-AMDK27 strain and deposited with the Korean Collection for Type Cultures (KCTC), the Korea Research Institute of Bioscience and Biotechnology, under accession number KCTC 12398BP.
  • KCTC Korean Collection for Type Cultures
  • a culture of the Akkermansia muciniphila EB-AMDK27 strain was centrifuged at 12,000x g and 4°C for 5 minutes, and the cells harvested, suspended in PBS, and adjusted to an OD value of 0.25 ⁇ 0.03 (8 log CFU/mL). Next, the cells were pasteurized at 70°C for 30 minutes and stored in a cryogenic freezer until use.
  • cytokines and other immunomodulators were involved in the regulation of the inflammatory response in inflammatory bowel disease, the present inventors investigated whether the expression of these genes would be affected by administration of the strain of the present invention.
  • human colonic epithelial HT-29 cells ATCC ® HTB-38 TM , USA
  • the cells were cultured in an incubator (NUAIRE, USA) at 37°C under 5% CO 2 using, as basal culture medium, McCoy's 5A modified medium (Gibco, USA) supplemented with 10% FBS (fetal bovine serum, Hyclone, USA) and 10 ⁇ g/ml gentamicin.
  • inflammatory cytokines was analyzed using SYBR Green TOPreal TM qPCR 2X PreMIX (Enzynomics, Korea), and GAPDH was used as an internal standard. PCR was performed under the following conditions: pre-incubation (for UDG) of 4 min at 50°C, 10 min at 95°C, and 40 cycles, each consisting of 15 sec at 95°C and 1 min at 60°C. Data were analyzed by delta CT method using the program built in QuantStudio Design & Analysis Software v1.4.3, and the results are shown in FIG. 6.
  • FIG. 6 it was shown that treatment with the Akkermansia muciniphila EB-AMDK27 strain of the present invention decreased the mRNA expression of pro-inflammatory cytokines (such as IL-8, TNF- ⁇ , IL-6 and IL-1 ⁇ ), which was increased by the inflammation inducer LPS. This suggests that the strain of the present invention can be effective for the treatment and prevention of inflammatory disease.
  • pro-inflammatory cytokines such as IL-8, TNF- ⁇ , IL-6 and IL-1 ⁇
  • FIG. 7 depicts photographs and a graph, which show the results of measuring lipid accumulation following treatment with the strain of the present invention.
  • Oil Red-O staining is a method of measuring lipid accumulation in differentiated 3T3-L1 cells by staining the cells with Oil Red-O reagent.
  • Mouse preadipocyte 3T3-L1 cells (Korean Cell Line Bank, KOREA) were cultured in an incubator (NUAIRE, USA) at 37°C under 5% CO 2 using, as basal culture medium, Dulbecco's Modified Eagle's Medium (DMEM) supplemented with 10% FBS (fetal bovine serum, Hyclone, USA) and 1% penicillin/streptomycin.
  • DMEM Dulbecco's Modified Eagle's Medium
  • Adipose differentiation of the preadipocyte 3T3-L1 cells was induced by treatment with insulin (1 ⁇ g/ml), IBMX (0.5 mM) and dexamethasone (1 ⁇ M) for 10 days, and the cells were treated with the Akkermansia muciniphila EB-AMDK27 strain. Then, the culture was washed three times with PBS to remove the medium. 10% formalin (Sigma, USA) was added and Oil red O (Sigma, USA) solution was allowed to react with the cells for 1 hour, followed by washing with distilled water, thereby staining the lipid droplets.
  • FIG. 7(A) The lipid droplet sample stained with the Oil red O solution was lysed with isopropanol, and the absorbance at 500 nm was measured using a spectrophotometer (Epoch, BioTek, USA). The results of the measurement are shown in FIG. 7(B).
  • PPAR ⁇ peroxisome proliferator-activated receptor gamma
  • CEBP ⁇ CCAAT/enhancer binding protein alpha
  • adipocyte protein 2 aP2
  • CD36 cluster differentiating 36
  • FAS fatty acid synthase
  • ACC1 acetyl-coenzyme A-carboxylase
  • LPL lipoprotein lipase
  • LDLR low-density lipoprotein receptor
  • PCR reaction was performed using the Quant Studio 3 real time PCR system (Applied Biosystems, USA) under the following conditions: pre-incubation (for UDG) of 4 min at 50°C, 10 min at 95°C, and 40 cycles, each consisting of 15 sec at 95°C and 1 min at 60°C. Data were analyzed by delta CT using the program built in QuantStudio Design & Analysis Software v1.4.3.
  • FIG. 8 depicts graphs showing the results of measuring the mRNA expression of genes associated with preadipocyte-to-adipocyte differentiation and transcription.
  • PPAR ⁇ Peroxisome proliferator-activated receptor ⁇
  • CEBP ⁇ CCAAT/enhancer binding protein ⁇
  • aP2 ⁇ activator protein 2 alpha
  • adipocyte differentiation medium DM
  • the degree of lipid accumulation in micrographs of the lipid droplets and in the cells significantly decreased in the group treated with DM + the Akkermansia muciniphila BAA-835 strain or DM + the pasteurized Akkermansia muciniphila EB-AMDK27 strain (10% v/v) compared to that in the control group (p ⁇ 0.001), and it also significantly decreased in the group treated with the pasteurized Akkermansia muciniphila EB-AMDK27 strain compared to that in the positive control group (ATCC BAA-835) (P ⁇ 0.01).
  • DM adipocyte differentiation medium
  • the expression of PPAR ⁇ (p ⁇ 0.001), CEBP ⁇ (p ⁇ 0.001), CD36 (p ⁇ 0.01) and LPL (p ⁇ 0.01) significantly decreased in the group treated with the Akkermansia muciniphila BAA-835 strain compared to the group treated with the BAA-835 strain. That is, it could be confirmed that the Akkermansia muciniphila EB-AMDK27 strain of the present invention had a much better effect on the inhibition of adipocyte differentiation of 3T3-L1 cells than the Akkermansia muciniphila BAA-835 strain.
  • mice 8-week-old male C57BL mice (6 mice per group) were purchased and acclimated for one week, and then bred and housed for 8 weeks. During breeding, the animals were acclimated for one week at a temperature of 22°C and a relative humidity of 40 to 60% with 12-hr light/12-hr dark cycles.
  • the animals were fed with high-fat diet (60 kcal% fat; Research Diets Inc., NJ, USA) and allowed to freely access drinking water.
  • high-fat diet 60 kcal% fat; Research Diets Inc., NJ, USA
  • the experimental animals were divided into five groups as follows:
  • GC Experimental group III
  • EB-AMDK27 group fed with high fat diet for obesity induction and administered with Akkermansia muciniphila EB-AMDK27 strain (a probiotic form).
  • mice Normal mice were fed with 10% fat diet.
  • Garcinia Cambogia (GC, 60% HCA 1000 mg/kg), known as a food functional food useful for reducing body weight, and the Akkermansia muciniphila BAA-835 strain, was administered.
  • PBS (25% glycerol and 0.05% cysteine/PBS) was orally administered every day in the same amount as that of the high-fat diet or Garcinia Cambogia in order to exclude the effect of administration-induced stress or the like.
  • mice administered with the Akkermansia muciniphila EB-AMDK27 strain showed a significant reduction in body weight compared to the mice fed with high-fat diet (HFD) (P ⁇ 0.05).
  • HFD high-fat diet
  • the food intake of the group administered with the Akkermansia muciniphila EB-AMDK27 strain significantly decreased compared to that of the high-fat diet group. From these results, it can be seen that the Akkermansia muciniphila EB-AMDK27 strain of the present invention induces continued decreases in food intake and body weight.
  • the weights of liver, spleen, subcutaneous fat, epididymal fat and mesenteric fat all significantly increased in the group fed with high-fat diet (HFD) compared to the normal group, and the weights of liver, spleen, subcutaneous fat, epididymal fat and mesenteric fat significantly decreased in the group administered with the Akkermansia muciniphila EB-AMDK27 strain.
  • HFD high-fat diet
  • the weights of liver, spleen, subcutaneous fat, epididymal fat and mesenteric fat significantly decreased in the group administered with the Akkermansia muciniphila EB-AMDK27 strain.
  • the group administered with the Akkermansia muciniphila EB-AMDK27 strain showed a more significant decrease in subcutaneous fat weight compared to the group administered with Garcinia Cambogia or the Akkermansia muciniphila BAA-835 strain (P ⁇ 0.01).
  • epididymal fat only the group administered with the Akkermansia
  • the group administered with the Akkermansia muciniphila EB-AMDK27 strain immediately before glucose administration showed the greatest decrease in the blood glucose level among the groups administered, but this decrease was not significant.
  • TC total cholesterol
  • GPT liver function indicator
  • the insulin concentration was measured using an insulin ELISA kit (Morinaga, Japan), and the insulin resistance index (HOMA-IR index) was calculated using the following equation:
  • TC Total cholesterol
  • GPT glutamic pyruvic transaminase
  • the results of measurement of the insulin concentration showed that the insulin concentration and insulin resistance index (HOMA-IR) that increased in the high-fat diet group significantly decreased in all the groups administered.
  • the cholesterol level and insulin resistance index that increased in the high-fat diet group significantly decreased only in the group administered with the Akkermansia muciniphila EB-AMDK27 strain (P ⁇ 0.05).
  • the GPT concentration indicative of the degree of liver damage significantly decreased in all the groups administered.
  • the blood insulin, cholesterol and GPT concentrations that decreased due to administration of the Akkermansia muciniphila EB-AMDK27 strain are closely related to the weight loss effect. Taking the above results together, it can be seen that the strain and pharmaceutical composition of the present invention can be effectively used to improve carbohydrate and lipid metabolisms for the treatment of obesity.
  • the Akkermansia muciniphila EB-AMDK27 strain of the present invention has the effect of treating damage to colonic mucosa, colon tissues from the normal group, the high-fat diet group (HFD), the group administered with Garcinia Cambogia (GC), the group administered with the Akkermansia muciniphila ATCC BAA-835 strain (BAA-835), and the group administered with the Akkermansia muciniphila EB-AMDK27 strain (EB-AMDK27) were stained with Alcian blue staining and imaged (FIG. 13). In addition, the results obtained by quantifying the positive area (%) in each group on the basis of these results are graphically shown in FIG. 13.
  • the mucosa structure in the colon tissue from the group administered with the Akkermansia muciniphila EB-AMDK27 strain of the present invention was maintained and was similar to that in the colon tissue from the normal group.
  • the goblet cells that decreased in the colon tissue from the high-fat diet group (HFD) significantly increased in the group administered with the Akkermansia muciniphila EB-AMDK27 strain (p ⁇ 0.05).
  • the expression of the inflammatory mediators TLR2 (toll-like receptor 2, p ⁇ 0.05), TLR4 (p ⁇ 0.05), IL-6 (p ⁇ 0.05), TNF- ⁇ (p ⁇ 0.01) and MCP-1 (monocyte chemoattractant protein-1, p ⁇ 0.05), which increased in the high-fat diet group, significantly decreased in the group administered with the Akkermansia muciniphila EB-AMDK27 strain (TLR2 (p ⁇ 0.05), TLR4 (p ⁇ 0.05), IL-6 (p ⁇ 0.05), TNF- ⁇ (p ⁇ 0.01), and MCP-1 (p ⁇ 0.05)).
  • TLR2, TLR4 and TNF- ⁇ also decreased in the group administered with the Akkermansia muciniphila BAA-835.
  • the expression of the anti-inflammatory cytokine IL-10 (p ⁇ 0.05) significantly increased only in the group administered with the Akkermansia muciniphila EB-AMDK27 strain compared to the high-fat diet group.
  • the expression of the appetite suppressant hormone GLP-1 (glucagon like peptide-1) significantly decreased in the high-fat diet group (p ⁇ 0.01), but significantly increased in the group administered with the Akkermansia muciniphila EB-AMDK27 strain compared to the high-fat diet group (p ⁇ 0.01).
  • Visceral adipose tissue was extracted from the mice of each experimental group, and then weighed. A portion of the visceral adipose tissue was taken and fixed in 10% buffered formalin. Next, it was embedded in paraffin and sectioned to a thickness of 4 mm using a microtome (Reichert-Jung 2050, ALT, USA). The sections were mounted on glass slides, and then stained by H & E (Hematoxylin & Eosin) and ORO (Oil Red O) staining. Next, the shape and state of hepatocytes and lipid droplets were observed under an optical microscope at 200x to 400x magnification. The results are shown in FIGS. 15 and 16.
  • FIG. 17 Mesenteric fat tissue was isolated from the mice of each experimental group and analyzed by H & E staining, and the results of the analysis are shown in FIG. 17.
  • the diameter of adipocytes in the high-fat diet group increased compared to that in the normal group (p ⁇ 0.001).
  • the diameter of adipocytes was observed to decrease in the group administered with Garcinia Cambogia, the group administered with the Akkermansia muciniphila BAA-835 strain, and the group administered with the Akkermansia muciniphila EB-AMDK27 strain, compared to the high-fat diet group (p ⁇ 0.001).
  • the diameter of adipocytes in the group administered with the Akkermansia muciniphila EB-AMDK27 strain of the present invention significantly decreased compared to those in the group administered with the Garcinia Cambogia and the group administered with the Akkermansia muciniphila BAA-835 strain.
  • feces were collected from the mice of each group, and genomic DNA was extracted therefrom using a QIAamp DNA stool mini kit (Qiagen, USA).
  • QIAamp DNA stool mini kit Qiagen, USA
  • the position of the band of the genomic DNA was determined by electrophoresis on 1.5% agarose gel (QA-Agarose, Q-biogene, USA) in 1X TAE buffer using an electrophoresis apparatus at 100 V for 25 minutes.
  • the purity of DNA was measured at A260/A280 using a spectrophotometer.
  • PCR was performed using specially constructed barcode primers.
  • the amplified PCR product was sequenced using an Ion Torrent Next-Generation sequencing platform which is a next-generation sequencing (NGS) method, and the microbiota was analyzed. After reads with low quality scores or a very small number of reads were removed, the resulting microbial sequences are imported into the Greengenes database, and OUTs (operational taxonomic units) were assigned.
  • NGS next-generation sequencing
  • the microbiota between clusters was analyzed using the Quantitative insights into microbial ecology (QIIME) 1.9.0 which is a microbiome data integration analysis tool.
  • QIIME Quantitative insights into microbial ecology
  • PCA analysis was performed based on the results obtained for the normal group, the high-fat diet group, the group administered with the Akkermansia muciniphila BAA-835 strain, and the group administered with the Akkermansia muciniphila EB-AMDK27 strain.
  • the alpha-diversity was analyzed and presented by OTUs and Chao1, and the beta-diversity was analyzed by UniFracbased principal coordinates analysis (PCoA). Analysis of linear discriminant analysis effect size (LEfSe) was conducted through an online program ( http://huttenhower.sph.harvard.edu/galaxy ).
  • the group administered with the Akkermansia muciniphila EB-AMDK27 of the present invention showed relatively high abundances of Bacteroidaceae and Peptococcaceae .
  • Bacteroidales, Clostridiales and Lactobacillale orders at the order level and Lachnospiraceae , Rikenellaceae, Ruminococcaceae, Prevotellaceae and Streptococcaceae families at the family level were restored to the levels of the normal group microorganisms in the group administered with the EB-AMDK27 strain.
  • the strain and pharmaceutical composition of the present invention can be effectively used to improve carbohydrate and lipid metabolisms for the treatment of obesity.
  • Depository authority Korean Collection for Type Cultures

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Medicinal Chemistry (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Mycology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Zoology (AREA)
  • Microbiology (AREA)
  • Biotechnology (AREA)
  • Wood Science & Technology (AREA)
  • Genetics & Genomics (AREA)
  • Food Science & Technology (AREA)
  • Nutrition Science (AREA)
  • Polymers & Plastics (AREA)
  • Diabetes (AREA)
  • Hematology (AREA)
  • Obesity (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Virology (AREA)
  • Biomedical Technology (AREA)
  • Epidemiology (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • Pain & Pain Management (AREA)
  • Rheumatology (AREA)
  • Endocrinology (AREA)
  • Child & Adolescent Psychology (AREA)
  • Emergency Medicine (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)

Abstract

Disclosed are a novel Akkermansia muciniphila EB-AMDK27 strain having the effect of preventing or treating inflammatory disease or metabolic disease, and a pharmaceutical composition effective for preventing or treating inflammatory disease or metabolic disease, which contains the strain, a culture thereof, or a dried product thereof. While traditional probiotics generally have insufficient therapeutic effects on inflammatory or metabolic diseases, the disclosed next-generation probiotic strain has an excellent effect on the prevention or treatment of inflammatory disease and/or metabolic disease so that it may be used as a new prophylactic and therapeutic tool.

Description

AKKERMANSIA MUCINIPHILA EB-AMDK27 STRAIN AND USE THEREOF
The present invention relates to a novel Akkermansia muciniphila strain and the use thereof, and more particularly to a novel Akkermansia muciniphila strain which has the effect of preventing or treating inflammatory disease or metabolic disease, and a pharmaceutical composition for the prevention or treatment of inflammatory disease or metabolic disease, which contains the Akkermansia muciniphila strain.
Inflammatory diseases refer to diseases that are caused by inflammatory cytokines, such as tumor necrosis factor-α (TNF-α), interleukin-1 (IL-1), IL-6, prostaglandin, leukotriene or nitric oxide (NO), which are secreted from immune cells such as macrophages due to an exaggerated reaction of the human body's immune system to harmful stimuli such as inflammatory agents or radiation.
Inflammatory diseases, such as inflammatory bowel disease (IBD), are a family of chronic, recurrent, and tissue-destructive diseases, characterized by dysfunction of mucosal T cells, abnormal cytokine production and cellular inflammation, which cause mucosal damage. Inflammatory diseases are serious diseases that dramatically decrease the quality of life, but the causes thereof still remain unclear.
When inflammatory bowel disease is taken as an example, one of the current treatments is a method of removing colon ulcers by colon resection, but this method may lower the quality of life and increase the risk of complications.
Meanwhile, other medical approaches include a method of controlling inflammation by suppressing the immune system using an immunomodulator, but the immunomodulator may induce an immune-lowering condition that makes the patient susceptible to various other diseases.
In order to overcome the adverse effects caused by conventional drugs or immunomodulators, studies using probiotics alone or in combination with antibiotics have been conducted. As research on the role of intestinal bacteria in promoting health has been actively conducted, interest in lactic acid bacteria preparations has increased.
As technology for treating inflammation or obesity using probiotics, Korean Patent Application Publication No. 2011-0095929 discloses a Lactobacillus reuteri strain which interferes with in vivo lipid absorption, Korean Patent No. 0996577 discloses a Lactobacillus curvatus strain which lowers blood cholesterol levels and inhibits obesity, Korean Patent Application Publication No. 2011-0010015 discloses a Lactobacillus johnsonii which inhibits obesity while lowering blood cholesterol levels, and Korean Patent Application Publication No. 2019-0034796 discloses a Lactobacillus plantarum strain which has antioxidant and anti-inflammatory effects.
However, the probiotics as described above have insignificant effects on the amelioration of inflammatory disease or metabolic disease, and hence have a problem in that they are difficult to use as new preventive and therapeutic tools.
The present invention is intended to overcome the above-described limitation of the conventional art, and an object of the present invention is to provide a next-generation probiotic strain which inhibits inflammatory response by reducing or inhibiting the release of one or more inflammatory factors, such as tumor necrosis factor-α (TNF-α), IL-8, IL-6 and IL-1β, and which is effective in preventing or treating inflammatory disease, obesity, obesity complications or metabolic disease by inhibiting adipocyte differentiation.
Another object of the present invention is to provide a pharmaceutical composition effective for the prevention or treatment of inflammatory disease or metabolic disease, which contains an Akkermansia muciniphila strain, which is the next-generation probiotic strain.
Another object of the present invention is to provide a pharmaceutical composition for suppressing appetite, which contains the Akkermansia muciniphila strain.
Still another object of the present invention is to provide a food useful for ameliorating inflammatory disease or metabolic disease, which contains the Akkermansia muciniphila strain.
One aspect of the present invention for achieving the above objects is directed to an Akkermansia muciniphila EB-AMDK27 strain (accession number KCTC 13758BP).
Another aspect of the present invention for achieving the above objects is directed to a pharmaceutical composition for preventing or treating inflammatory disease or metabolic disease, which contains the Akkermansia muciniphila EB-AMDK27 strain (accession number KCTC 13758BP), a culture thereof, or a dried product thereof.
Another aspect of the present invention for achieving the above objects is directed to a pharmaceutical composition for suppressing appetite, which contains the Akkermansia muciniphila EB-AMDK27 strain (accession number KCTC 13758BP), a culture thereof, or a dried product thereof.
Still another aspect of the present invention for achieving the above objects is directed to a food for preventing or ameliorating inflammatory disease or metabolic disease, which contains the Akkermansia muciniphila EB-AMDK27 strain (accession number KCTC 13758BP), a culture thereof, or a dried product thereof.
The novel Akkermansia muciniphila EB-AMDK27 strain of the present invention inhibits inflammatory response by reducing or inhibiting the release of inflammatory factors, and is effective in treating inflammatory disease, particularly bowel inflammation, by maintaining the stability of intestinal microbiota.
The pharmaceutical composition containing, as an active ingredient, the Akkermansia muciniphila EB-AMDK27 strain of the present invention may be used as a pharmaceutical composition or a functional food composition for treating and/or preventing inflammatory disease or metabolic disease.
The novel Akkermansia muciniphila EB-AMDK27 strain of the present invention and the pharmaceutical composition containing the same may inhibit body weight gain and body fat accumulation, lower insulin resistance, lower total blood cholesterol levels, reduce the level of blood GPT that is a hepatotoxicity indicator, and reduce the levels of inflammatory cytokines, including IL-8, TNF-α, IL-6 and IL-1β. Thus, they may be useful for preventing or treating diabetes, obesity, insulin resistance, fatty liver, hyperlipidemia, or metabolic disease, which is associated with these factors.
In particular, the pharmaceutical composition of the present invention exhibits the effect of substantially inhibiting lipid accumulation by containing, as an active ingredient, the Akkermansia muciniphila EB-AMDK27 strain that reduces intracellular lipid accumulation, reduces expression of the adipocyte differentiation-related factor PPARγ, and also reduces the mRNA expression of CEBPα, aP2, CD36, ACC1, LPL (lipoprotein lipase), LDLR or FAS.
The composition for suppressing appetite containing the Akkermansia muciniphila EB-AMDK27 strain (KCTC 13758BP) of the present invention may suppress appetite by stimulating the secretion of appetite suppressant hormones.
FIG. 1 shows micrographs of the Akkermansia muciniphila EB-AMDK27 strain (KCTC 13758BP) of the present invention and the type strain Akkermansia muciniphila ATCC BAA-835;
FIG. 2 shows the results of PCR analysis of the Akkermansia muciniphila EB-AMDK27 strain of the present invention and the Akkermansia muciniphila ATCC BAA-835 strain;
FIG. 3 shows the results of testing the hemolytic activities of the Akkermansia muciniphila EB-AMDK27 strain of the present invention and the Akkermansia muciniphila ATCC BAA-835 strain;
FIG. 4 shows the results of RAPD (Random Amplified Polymorphic DNA) analysis of the Akkermansia muciniphila EB-AMDK27 strain of the present invention and the Akkermansia muciniphila ATCC BAA-835 strain;
FIG. 5 shows the phylogenetic relationship between the Akkermansia muciniphila EB-AMDK27 strain of the present invention and other Akkermansia muciniphila strains;
FIG. 6 depicts graphs showing the relative mRNA expression levels of the cytokines IL-6, IL-8, IL-1β and TNF-α, which demonstrate the anti-inflammatory effect of the Akkermansia muciniphila EB-AMDK27 strain of the present invention;
FIG. 7 depicts photographs and a graph, which show the degree of lipid accumulation in 3T3-L1 cells after treatment with the Akkermansia muciniphila EB-AMDK27 strain of the present invention;
FIG. 8 depicts graphs showing the relative mRNA expression levels of PPARγ, CEBPα, aP2, CD36, ACC1, LPL (lipoprotein lipase), LDLR and FAS in preadipocytes after treatment with the Akkermansia muciniphila EB-AMDK27 strain of the present invention;
FIG. 9 shows the results of analyzing changes in the body weight of a group administered with the Akkermansia muciniphila EB-AMDK27 strain of the present invention, a control group (DM), and a group administered with the Akkermansia muciniphila ATCC BAA-835 strain;
FIG. 10 shows the results of analyzing changes in the weight of subcutaneous fat, epididymal fat and mesenteric fat of a group administered with the Akkermansia muciniphila EB-AMDK27 strain of the present invention, a control group (DM), and a group administered with the Akkermansia muciniphila ATCC BAA-835 strain;
FIG. 11 shows the results of analyzing changes in the glucose tolerance of a group administered with the Akkermansia muciniphila EB-AMDK27 strain of the present invention, a control group (DM), and a group administered with the Akkermansia muciniphila ATCC BAA-835 strain;
FIG. 12 shows the results of measuring changes in the blood insulin, cholesterol and glutamic pyruvic transaminase (GPT) concentrations in a group administered with the Akkermansia muciniphila EB-AMDK27 strain of the present invention, a control group (DM), and a group administered with the Akkermansia muciniphila ATCC BAA-835 strain, by ELISA;
FIG. 13 shows Alcian blue staining images of a normal group, a high-fat diet group (HFD), a group administered with Garcinia cambogia (GC), a group administered with the Akkermansia muciniphila ATCC BAA-835 strain (BAA-835), and a group administered with the Akkermansia muciniphila EB-AMDK27 strain of the present invention (EB-AMDK27), and depicts a graph showing the positive area in each group, obtained by quantifying the images;
FIG. 14 shows changes in the expression of TLR4, TLR2, GLP-1, PYY, IL-6, TNF-α, MCP-1, IL-10, ZO-1 and occluding in the enterocytes of mice of each test group;
FIGS. 15 and 16 are optical micrographs of hepatocytes and lipid droplets, obtained after H & E staining, and are graphs showing liver steatosis scores in each group;
FIG. 17 depicts photographs and a graph, which show the degree of lipid accumulation in the mesenteric adipose tissue of each test animal after treatment with the Akkermansia muciniphila EB-AMDK27 strain of the present invention;
FIG. 18 shows a comparison of linear discriminant analysis results between test groups; and
FIG. 19 depicts graphs showing the relative abundance of intestinal microbiomes following administration of the Akkermansia muciniphila ATCC BAA-835 strain (BAA-835) and the Akkermansia muciniphila EB-AMDK27 strain of the present invention (EB-AMDK27) in one Example of the present invention.
The present invention will be described in more detail below with reference to the accompanying drawings.
As used in the claims, the term "comprises/comprising" or "contains/containing" is not intended to exclude the presence of one or more other species or steps.
As used herein, the term "inflammatory disease" refers to a disease that is caused by an inflammatory reaction in the mammalian body. Representative examples of inflammatory disease include: respiratory diseases such as asthma, chronic obstructive pulmonary disease, and rhinitis; skin diseases such as atopic dermatitis; digestive diseases such as gastritis and inflammatory enteritis; arteriosclerosis, sepsis, inflammatory joint disease, inflammatory brain disease, and the like.
As used herein, the term "treating" or "treatment" refers to reversing or alleviating inflammatory disease, metabolic disease or one or more symptoms of the disease, or inhibiting the progress thereof, unless stated otherwise. As used herein, the term "preventing" or "prevention" is intended to include reducing the likelihood of developing inflammatory disease or metabolic disease.
As used herein, the term "cytokine" refers to a secreted protein that affects the functions of other cells. Particularly, it relates to the modulation of interactions between cells of the immune system or cells involved in the inflammatory response. Examples of cytokines include, but are not necessarily limited to, interleukin 1 (IL-1), preferably interleukin IL-1β, interleukin-6 (IL-6), interleukin 8 (IL-8), and tumor necrosis factor-α (TNF-α).
As used herein, the term "suppressing appetite" refers to any action that suppresses or delays appetite by administering the strain or composition to stimulate the secretion of diet-related hormones (e.g., appetite suppressant hormones).
One aspect of the present invention is directed to a next-generation probiotic strain which is an Akkermansia muciniphila EB-AMDK27 strain (accession number KCTC 13758BP).
The strain has the 16s rRNA gene of SEQ ID NO: 1.
The Akkermansia muciniphila EB-AMDK27 strain of the present invention is a monococcus or diplococcus isolated from a health Korean's feces, which is an elliptical cell of 0.5 to 1 μm in size. Also, it is a mucus-degrading bacterium which is anaerobic, non-motile, and gram-negative. It does not form an endospore. The Akkermansia muciniphila EB-AMDK27 strain is capable of producing several mucolytic enzymes, and thus may use mucus as carbon and nitrogen sources. The Akkermansia muciniphila EB-AMDK27 strain can metabolize various carbon sources, including galactose, N-acetylglucosamine, and lactose, and produces, as main metabolites, short-chain fatty acids such as propionic acid and acetic acid.
According to one embodiment of the present invention, the Akkermansia muciniphila EB-AMDK27 strain is capable of exhibiting an anti-inflammatory effect by inhibiting the expression of inflammatory cytokines, specifically IL-8, IL-6, IL-1β, and TNF-α.
Tight junctions formed between epithelial cells are particularly important for the barrier function of epithelial cells. The tight junctions are a type of cell-cell junction, and are so strong that it appears under an electron microscope that the cell membranes of two adjacent cells are fused. The barrier function is a function that blocks foreign substances from passing between epithelial cells, and is important in both blood vessels and digestive tracts. Constituent molecules that are involved in formation of the tight junctions typically include a membrane protein called occludin and cytoplasmic ZO proteins as well as proteins such as inulin are also involved in the formation of the tight junctions. Through the interaction between these proteins, the structure and function of the tight junctions are completed. The Akkermansia muciniphila EB-AMDK27 strain according to one embodiment of the present invention increases the expression of the tight junction proteins ZO-1 (zonular occludens-1, p<0.01) and occludin and blocks inflammatory proteins from migrating into tissues, thereby promoting mucous regeneration and alleviating inflammation of colonic mucosa.
When obesity is induced, it causes abnormalities in visceral adipose tissue, excessive secretion of tumor necrosis factor, infiltration of immune cells such as macrophages into adipose tissue, and increased expression of inflammatory cytokines. This results in chronic inflammation of adipose tissue, and the chronic inflammatory response reduces insulin sensitivity and induces glucose tolerance, leading to diabetes. Therefore, suppression of adipose tissue hypertrophy and inflammatory response can exhibit an anti-obesity effect, and as a result, can also be useful against anti-metabolic diseases.
The novel Akkermansia muciniphila EB-AMDK27 strain of the present invention may inhibit body weight gain and body fat accumulation, lower insulin resistance, lower total blood cholesterol levels, reduce the level of blood glutamic pyruvic transaminase (GPT) that is a hepatotoxicity indicator, and reduce the levels of inflammatory cytokines. Thus, it may be useful for preventing or treating diabetes, obesity, obesity-related disease, insulin resistance, fatty liver, hyperlipidemia, or metabolic disease, which is associated with these factors. In particular, the metabolic disease may be a disease in which various metabolic diseases such as diabetes and obesity appear simultaneously in one person. The Akkermansia muciniphila EB-AMDK27 strain of the present invention has therapeutic efficacy against both inflammatory disease and metabolic disease, and thus can provide the remarkable effect of comprehensively treating various diseases that are highly correlated with inflammation or obesity.
In particular, the novel Akkermansia muciniphila EB-AMDK27 strain of the present invention and a pharmaceutical composition containing the same exhibit the effect of substantially inhibiting lipid accumulation by containing, as an active ingredient, the Akkermansia muciniphila EB-AMDK27 strain that reduces intracellular lipid accumulation, reduces expression of the adipocyte differentiation-related factor PPARγ and also reduces the mRNA expression of CEBPα, aP2, CD36, ACC1, LPL (lipoprotein lipase), LDLR, or FAS.
Another aspect of the present invention is directed to a pharmaceutical composition for preventing or treating inflammatory disease or metabolic disease, which contains an Akkermansia muciniphila EB-AMDK27 strain (accession number KCTC 13758BP), a culture thereof, or a dried product thereof.
The pharmaceutical composition of the present invention may contain a probiotic form of the Akkermansia muciniphila EB-AMDK27 strain or a pasteurized form of the Akkermansia muciniphila EB-AMDK27 strain. Pasteurization of the Akkermansia muciniphila EB-AMDK27 strain refers to heating at temperature equal to or higher than 50℃ and lower than 100℃ for 10 minutes or more. For example, the strain may be pasteurized at a temperature of 70℃ for 30 minutes. A pasteurized form of the Akkermansia muciniphila EB-AMDK27 strain can reduce body fat accumulation to a greater extent compared to a probiotic form of the strain.
Although the reason why the pasteurized form of the Akkermansia muciniphila EB-AMDK27 strain is more effective than the probiotic form has not been accurately identified, it can be presumed that when the Akkermansia muciniphila EB-AMDK27 strain is pasteurized, the cell wall components (such as Amuc_1100) or membrane proteins of the strain enhance metabolic benefits in a host.
The beneficial effect of the strain of the present invention is presumed to be related to a gene cluster (Amuc_1098-Amuc_1102) containing Amuc_1100 and to be attributed to polypeptides that interact with the signaling pathway of toll-like receptor 2 ("TLR2"), which is present on the surface of immune cells located near the barrier of intestinal mucosa and modulates intestinal homeostasis and host metabolism. For example, the Amuc-1100 polypeptide is expected to maintain the integrity of the barrier of the intestinal mucosa and interact with TLR2 present on the surface of immune cells to modulate or promote the TLR2 signaling pathway, thereby promoting the secretion of cytokines (e.g., IL- 6, IL-8, and IL-10) from immune cells. Prolipoprotein diacylglyceryl transferase gene (Amuc_1104) is located in close proximity to the gene cluster (Amuc_1098-Amuc_1102). In addition, Amuc_1100 can be stably maintained under the temperature condition used during pasteurization, thus contributing to the effect of the pasteurized strain.
The pharmaceutical composition of the present invention is effective for the treatment or prevention of inflammatory diseases, particularly inflammatory bowel disease. Examples of such inflammatory bowel disease include Crohn's disease, ulcerative colitis, intestinal Behcet's disease, simple ulcer, radiation enteritis, and ischemic enteritis. In particular, the pharmaceutical composition is effective against Crohn's disease or ulcerative colitis.
The metabolic disease in the present invention is preferably obesity, insulin resistance, fatty liver, hyperlipidemia, or complications thereof, but is not necessarily limited thereto. For example, the pharmaceutical composition of the present invention is effective for preventing or treating inflammatory bowel disease (IBD), insulin resistance, or dyslipidemia, or for reducing cholesterol levels or body weight.
The pharmaceutical composition of the present invention contains, as an active ingredient, the Akkermansia muciniphila EB-AMDK27 strain at a concentration of 10 8 to 10 12 CFU/g of the composition, or a culture containing viable cells of the Akkermansia muciniphila EB-AMDK27 strain at the same concentration above.
The Akkermansia muciniphila EB-AMDK27 strain of the present invention may be recovered by a separation process such as centrifugation, and prepared as a probiotic by drying, for example, freeze-drying, for use.
The Akkermansia muciniphila EB-AMDK27 strain of the present invention is oxygen-sensitive, and hence is preferably cultured under anaerobic conditions (90% nitrogen, 5% hydrogen, and 5% carbon dioxide).
Components of the liquid medium during culture can affect the growth of the strain and the production of active ingredients. Thus, it is necessary to establish the components and their conditions of the liquid medium, which are optimal for culturing the novel Akkermansia muciniphila EB-AMDK27 strain of the present invention.
The liquid medium may contain, as a carbon source, one or more selected from the group consisting of glucose, lactose, galactose, N-acetylglucosamine, mannose, 1-fucose, lactate, formate, acetate, propionate, 1,2-propenediol, and butyrate, but is not limited thereto. Preferably, it may contain glucose and N-acetylglucosamine. The liquid medium may contain, as a nitrogen source, one or more selected from the group consisting of tryptone, peptone, soy peptone, L-glutamic acid, and ammonium, but is not limited thereto.
The liquid medium may contain, as trace elements, one or more selected from the group consisting of KH 2PH 4, Na 2HPO 4, NaCl, MgCl 2, CaCl 2, FeCl 2, ZnCl 2, CuCl 2, MnCl 2, CoCl 2, NiCl 2, Na 2SeO 3, Na 2WO 4 and Na 2MoO 4, but is not limited thereto.
The liquid medium may have a pH of 6.8 to 7.2, preferably a pH of 7.0. The pH can affect the activity of protein by changing the charge of the amino or carboxyl group of the amino acid, which is a unit of the enzymatic protein important for cell metabolism. In addition, changes in the pH in the external environment can affect the ionization of microbial nutrients, which can affect nutrient intake of the microorganism.
The liquid medium is preferably a medium containing glucose, N-acetylglucosamine, threonine, soypeptone, or any combination thereof.
The composition of the present invention may further contain pharmaceutically acceptable carriers and/or excipients, in addition to the active ingredient. In addition, the composition may be formulated with various additives, such as a binder, a disintegrant, a coating agent, and a lubricant, which are commonly used in the pharmaceutical industry.
The composition of the present invention may be formulated in the form of powder, granule, tablet, capsule, or liquid by mixing the Akkermansia muciniphila EB-AMDK27 strain of the present invention with a suitable carrier, excipient, auxiliary active ingredient, etc. The composition of the present invention may be formulated as a product for oral administration. In addition, the composition of the present invention may be productized by enteric coating using any known method so that it can pass through the stomach and then reach the small intestine in which the active ingredient microorganism can be rapidly released into the intestines.
Excipients that may be used in the present invention include: sugars such as sucrose, lactose, mannitol, or glucose; and starches such as corn starch potato starch, rice starch, or partially pregelatinized starch. Binders that may be used in the present invention include polysaccharides such as dextrin, sodium alginate, carrageenan, guar gum, acacia, and agar; naturally-occurring macromolecular substances such as tragacanth, gelatin, and gluten; cellulose derivatives such as hydroxypropylcellulose, methylcellulose, hydroxypropylmethylcellulose, ethyl cellulose, hydroxypropyl ethyl cellulose, and sodium carboxymethyl cellulose; and polymers such as polyvinylpyrrolidone, polyvinyl alcohol, polyvinyl acetate, polyethylene glycol, polyacrylic acid, polymethacrylic acid, and vinyl acetate resin.
Disintegrants that may be used in the present invention include: cellulose derivatives such as carboxymethylcellulose, calcium carboxymethylcellulose, low-substituted hydroxypropylcellulose, and cellulose derivatives; and starches such as sodium carboxymethyl starch, hydroxypropyl starch, corn starch, potato starch, rice starch, and partially pregelatinized starch.
Examples of lubricants that may be used in the present invention include talc, stearic acid, calcium stearate, magnesium stearate, colloidal silica, hydrous silicon dioxide, and various types of waxes and hydrogenated oils.
Coating agents that may be used in the present invention include, but are not necessarily limited to, water-insoluble copolymers such as a dimethylaminoethyl methacrylate-methacrylic acid copolymer, a polyvinylacetal diethylaminoacetate, an ethylacrylate-methacrylic acid copolymer, an ethylacrylate-methylmethacrylate-chlorotrimethylammonium ethylmethacrylate copolymer, and ethyl cellulose; enteric polymers such as a methacrylic acid-ethyl acrylate copolymer, hydroxypropyl methyl cellulose phthalate, hydroxypropyl methyl cellulose acetate succinate; and water-soluble polymers such as methyl cellulose, hydroxypropyl methyl cellulose, polyvinylpyrrolidone, and polyethylene glycol.
The dose of the strain as an active ingredient in the composition for preventing or treating inflammatory disease or metabolic disease according to the present invention may be determined in consideration of various factors, including the type of disease, the patient's age, body weight, sex and medical condition, the severity of the condition, and the route of administration. Thus, the dose regime can vary widely, but can be routinely determined using standard methods. For an adult patient, generally 1 × 10 6 or more viable cells or pasteurized cells, preferably 1 × 10 8 to 1 × 10 12 viable cells or pasteurized cells may be may be administered once or several times as needed. The exact formulation, route of administration, and dosage of the pharmaceutical composition disclosed herein can be determined by a physician by taking into account the patient's condition.
Still another aspect of the present invention is directed to a pharmaceutical composition for suppressing appetite, which contains an Akkermansia muciniphila EB-AMDK27 strain (accession number KCTC 13758BP), a culture thereof, or a dried product thereof.
Glucagon-like peptide-1 (GLP-1) and peptide YY (PYY) are appetite suppressant hormones that are accompanied by decreased food intake (Wren and Bloom, Gastroenterology 132:2116-2130 (2007)). According to one embodiment of the present invention, the Akkermansia muciniphila EB-AMDK27 strain can suppress appetite by increasing the expression and secretion of the appetite suppressant hormones GLP-1 (glucagon like peptide 1) and PYY (peptide YY).
Still another aspect of the present invention is directed to a food for preventing or ameliorating inflammatory disease or metabolic disease, which contains an Akkermansia muciniphila EB-AMDK27 strain (accession number KCTC 13758BP), a culture thereof, or a dried product thereof. A food containing the Akkermansia muciniphila EB-AMDK27 strain of the present invention may be taken as a food or nutritional product, such as milk or dairy products, or as a food supplement or health functional food. In one embodiment of the present invention, examples of the food product include, but are not necessarily to, foods such as dairy products, beverages, juices, soups, or foods for children.
The present invention will be described in more detail below with reference to examples. However, these examples are merely to illustrate the present invention, and the scope of the present invention is not limited to these examples.
Examples
Example 1: Isolation and Identification of Akkermansia muciniphila Strain
1.1. Isolation and Identification of Strain
In order to isolate Akkermansia muciniphila from the feces of a healthy Korean (male, 45 years old, BMI: 23.1), according to the method of Derrien, selective culture was performed using mucin medium (0.4 g/L monopotassium phosphate, 0.53 g/L sodium dichlorophosphate, 0.3 g/L sodium chloride, 0.3 g/L aluminum chloride, 0.1 g/L magnesium chloride, 0.11 g/L calcium chloride, 4.0 g/L sodium bicarbonate, 1 mL acidic trace element solution, 1 mL alkaline trace element solution, 1 mL vitamin solution, 2.5 g/L porcine gastric fluid (Type III)), and 0.25 g/L sodium sulfide nonahydrate), and then a strain was isolated (Derrien et al., 2004).
In order to confirm that the isolated strain would be an Akkermansia muciniphila strain, the isolated strain was observed under a microscope, and the results are shown in FIG. 1. In addition, PCR analysis was performed using the AM-specific primers shown in Table 1 below, and the results of the analysis are shown in FIG. 2.
In FIG. 1, A shows an Akkermansia muciniphila ATCC BAA-835 strain, and B is a 1,000× magnification micrograph of an Akkermansia muciniphila EB-AMDK27 strain. In FIG. 2, M represents a DNA size marker, lane 1 represents a positive control (ATCC BAA-835), lane 2 represents the Akkermansia muciniphila EB-AMDK27 strain, and lane 3 represents a negative control (distilled water).
Designation Direction Sequence (5'→3') Amplicon size
AM1 Forward CAG CAC GTG AAG GTG GGG AC 327 bp
AM2 Reverse CCT TGC GGT TGG CTT CAG AT
1.2. Analysis of Utilization of Carbohydrates by Isolated Akkermansia muciniphila Strain
In order to examine the utilization of carbohydrates by the isolated Akkermansia muciniphila strain of the present invention, the strain was cultured using an API50CH kit (Biomerieux, France), and then whether the strain would grow using each carbohydrate was compared with the type strain (ATCC BAA-835). The results of the comparison are shown in Table 2 below.
No. Carbohydrates ATCC BAA-835 EB-AMDK27 No Carbohydrates ATCC BAA-835 EB-AMDK27
0 Negative control - - 25 Esculine - -
1 Glycerol - - 26 Salicine - -
2 Erythritol - - 27 D-Cellobiose - -
3 D-Arabinose w w 28 D-Maltose - -
4 L-Arabinose w w 29 D-Lactose (bovine origin) + w
5 Ribose + w 30 D-Melibiose - -
6 D-Xylose w w 31 D-Saccharose (sucrose) - -
(sucrose)
7 L-Xylose w w 32 D-Trehalose - -
8 Adonitol - - 33 Inuline - -
9 α-Methyl-xyloside - - 34 D-Melezitose - -
10 D-Galactose w w 35 D-Raffinose - -
11 D-Glucose + Mucin + + 36 Amidon (starch) - -
12 D-Fructose w - 37 Glycogene - -
13 D-Mannose + w 38 Xylitol - -
14 L-Sorbose - - 39 Gentiobiose - -
15 L-Rhamnose - - 40 D-Turanose - -
16 Dulcitol - - 41 D-Lyxose w w
17 Inositol - - 42 D-Tagatose - -
18 D-Mannitol - - 43 D-Fucose - -
19 D-Sorbitol - - 44 L-Fucose + w
20 Methyl-α D-mannopyranoside - - 45 D-Arabitol - -
21 Methyl-α D-glucopyranoside - - 46 L-Arabitol - -
22 N-Acetylglucosamine + + 47 Potassium Gluconate - -
23 Amygdaline - - 48 Potassium 2-Ketogluconate - -
24 Arbutine - - 49 Potassium 5-Ketogluconate w w
+ : growth, w : weak growth, - : no growth,
As can be seen in Table 2 above, it was confirmed that the Akkermansia muciniphila EB-AMDK27 strain of the present invention differed from the type strain (ATCC BAA-835) with respect to the utilization of ribose, D-fructose, D-mannose, D-lactose and L-fucose.
1.3. Whole Genome Sequencing
In order to analyze variations between the Akkermansia muciniphila EB-AMDK27 strain isolated as described above and the Akkermansia muciniphila ATCC BAA-835 at the genomic level, whole genome sequencing of the strain was performed using the PacBio technique and compared with that of the type strain. The results of the analysis are shown in in Table 3 below.
Genomic statistics Strains
ATCC BAA-835 EB-AMDK27
Accseeion No. CP001071 CP027003
Assembly level Complete Complete
Seq. category Chromosome Chromosome
Total size (Mb) 2.6641 2.7342
GC (%) 55.8 55.4
Protein 2,246 2,289
Gene 2,321 2,382
CDS 2,257 2,318
Coding 2,246 2,289
rRNA 9 9
tRNA 52 52
Other RNA 3 3
Pseudogene 11 29
Symmetrical identity (%) - N/A
*Symmetrical identity is relative to ATCC BAA-835
Genes Strains
ATCC BAA-835 EB-AMDK27
Amuc_1098 Sequence length 901 900
Align identity (%) 100 99.4
Mismatch No. 0 4
Insertion No. 0 0
Deletion No. 0 1
Amuc_1099 Sequence length 337 337
Align identity (%) 100 99.7
Mismatch No. 0 1
Insertion No. 0 0
Deletion No. 0 0
Amuc_1100 Sequence length 316 316
Align identity (%) 100 98.4
Mismatch No. 0 5
Insertion No. 0 0
Deletion No. 0 0
Amuc_1101 Sequence length 612 612
Align identity (%) 100 99
Mismatch No. 0 6
Insertion No. 0 0
Deletion No. 0 0
Amuc_1102 Sequence length 237 237
Align identity (%) 100 98.7
Mismatch No. 0 3
Insertion No. 0 0
Deletion No. 0 0
Gene_cluster Sequence length 2403 2402
Align identity (%) 100 99.2
Mismatch No. 0 19
Insertion No. 0 0
Deletion No. 0 1
*Gene_cluster corresponds to the length of five genes
As can be seen in Table 3 above, the whole genome statistics of the Akkermansia muciniphila EB-AMDK27 strain of the present invention differed from those of the Akkermansia muciniphila ATCC BAA-835 strain.
Specifically, it is known that the Amuc_1100 polypeptide is associated with therapeutic activity against anti-inflammatory or metabolic disease and is a heat-stable protein acting on the Toll-like receptor (TLR) 2 (Plovier et al., 2017). As shown in Table 4 above, the Akkermansia muciniphila EB-AMDK27 strain of the present invention differed from the Akkermansia muciniphila ATCC BAA-835 strain with respect to the Amuc_1100 related gene.
1.4. Analysis of Antimicrobial Susceptibility of Isolated Strain
In order to examine the antimicrobial susceptibility of the Akkermansia muciniphila strain isolated in the Example as described above, the minimum inhibitory concentrations (MICs) of antibiotics for anaerobic bacteria (Piperacillin-Tazobactam (PTZ), Ceftizoxime (CTZ), Chloramphenicol (CHL), Clindamycin (CLI), Meropenem (MEM), Moxifloxacin (MXF), Metronidazole (MTZ), and Ciprofloxacin (CIP)) against the isolated strain were determined by broth microdilution according to the guideline of Clinical & Laboratory Standard Institute (CLSI, 2017), and the results are shown in Table 5 below.
Antibiotics MIC a Breakpoints (μg/mL) QC Test strain
S I R ATCC 29741 b EB-AMDK27
PTZ ≤32/4 64/4 ≥128/4 8/4 ≤0.5/4 (S)
CTZ ≤32 64 ≥128 16 2 (S)
CHL ≤8 16 ≥32 8 4 (S)
CLI ≤2 4 ≥8 4 ≤0.125 (S)
MEM ≤4 8 ≥16 0.5 0.5 (S)
MXF ≤2 4 ≥8 8 >32 (R)
MTZ ≤8 16 ≥32 2 0.25 (S)
CIP ≤1 2 ≥4 >32 >32 (R)
PTZ : Piperacillin-tazobactam, CTZ : ceftizoxime (3 rd gen), CHL : chloramphenicol, CLI : clindamycin, MEM : meropenem, MXF : moxifloxacin (4 th gen), MTZ : metronidazole, CIP : ciprofloxacin (2 nd gen), aMIC : Minimum inhibitory concentration bBacteroides thetiotaomicron ATCC 29741
As can be seen in Table 5 above, the Akkermansia muciniphila EB-AMDK27 strain of the present invention showed susceptibility to all the antibiotics excluding moxifloxacin and ciprofloxacin, which were fluoroquinolone based antibiotics. Therefore, it can be confirmed that the Akkermansia muciniphila strain according to the present invention is a safe strain that is not resistant to most antibiotics.
1.5. Analysis of Hemolytic Activity of Isolated Strain
In order to verify the safety of the Akkermansia muciniphila EB-AMDK27 strain isolated as described above, whether the strain would have hemolytic activity was evaluated. To this end, the strain was cultured using a blood agar medium prepared by adding 5% w/v defibrinated sheep blood to tryptic soy agar (17.0 g/L pancreatic digest of casein, 3.0 g/L pancreatic digest of soybean, 2.5 g/L dextrose, 5.0 g/L sodium chloride, 2.5 g/L potassium phosphate, and 15 g/L agar). The results of the culture are shown in FIG. 3.
As can be seen through FIG. 3, it was confirmed that β-hemolysis associated with pathogenicity was not observed in the Akkermansia muciniphila EB-AMDK27 strain of the present invention, indicating that the strain of the present invention is a safe strain having no hemolytic activity.
1.6. Random Amplified Polymorphic DNA (RAPD) Analysis
In order to verify that the strain isolated in this Example is different from the type strain, random amplified polymorphic DNA (RAPD)-finger printing was performed.
Genomic DNA was extracted from the isolated strain, and then PCR-RAPD (MyCycler, BIO-RAD, USA) was performed using the extracted genomic DNA as a template and the primers shown in Table 6 below. The PCR product was electrophoresed on 1% agarose gel for 1 hour and 30 minutes, and DNA fragmentation patterns were compared on a UV perforator. The results of the analysis are shown in FIG. 4.
Designation Direction Sequence (5'→3') SEQ ID NO
ERIC-1 Forward ATG TAA GCT CCT GGG GAT TCA C SEQ ID NO: 2
ERIC-2 Reverse AAG TAA GTG ACT GGG GTG AGC G SEQ ID NO: 3
(GTG) 5 Forward/Reverse GTG GTG GTG GTG GTG SEQ ID NO: 4
As can be seen in FIG. 4, the RAPD band pattern of the Akkermansia muciniphila EB-AMDK27 strain of the present invention was analyzed comparatively with that of the Akkermansia muciniphila type strain (ATCC BAA-835), and as a result, it showed a different band pattern. It is known that the RAPD band patterns of Akkermansia muciniphila species are different from each other when the species are different. Thus, it was confirmed that the Akkermansia muciniphila strain isolated in the present invention was different from the type strain.
1.7. Phylogenetic Tree Analysis using Full-Length 16S rRNA Gene Sequence
In order to identify the Akkermansia muciniphila strain isolated in Example 1, 16S rRNA sequencing was performed.
For full-length 16S rRNA gene sequencing of the Akkermansia muciniphila strain isolated in Example 1, the 16S rRNA gene was amplified using the 27F and 1541R primers shown in Table 7 below, and then sequenced using a 3730xl DNA analyzer. The full-length 16S rRNA gene sequence of the Akkermansia muciniphila EB-AMD 27 strain is set forth in SEQ ID NO: 1. The DNA sequence was analyzed using the DNA Star program and the Cluster V program to determine homology to the type strain. A phylogenetic tree was prepared using the 16S rRNA gene sequences of the Akkermansia muciniphila strain of the present invention and other strains of the same species already published. The prepared phylogenetic tree is shown in FIG. 5.
Designation Direction Sequence (5'→3') Amplicon size
27F Forward AGA GTT TGA TCM TGG CTC AG (SEQ ID NO: 5) 1,505 bp
1541R Reverse AAG GAG GTG ATC CAG CCG CA (SEQ ID NO: 6)
As shown in FIG. 5, the evolutionary relationship between the 16s rRNA gene sequences was analyzed through the phylogenetic tree, and as a result, it was confirmed that the Akkermansia muciniphila EB-AMDK27 strain was tied to the same group to which the Akkermansia muciniphila type strain (ATCC BAA-835) was tied. This suggests that the Akkermansia muciniphila EB-AMDK27 strain genetically belongs to Akkermansia muciniphila species.
As a result of analyzing the Akkermansia muciniphila strain, isolated from human feces, through the biochemical method (API) and molecular biological methods (16s rRNA sequencing, RAPD, and full-length screening) using the Akkermansia muciniphila type strain (ATCC BAA-835) as a control, it was finally confirmed to be a strain belonging to A. muciniphila species. In addition, through the antibiotic resistance test, the isolated Akkermansia muciniphila EB-AMDK27 strain was to be a safe strain that can function as probiotics. Based on these results, the isolated strain was named Akkermansia muciniphila EB-AMDK27 strain and deposited with the Korean Collection for Type Cultures (KCTC), the Korea Research Institute of Bioscience and Biotechnology, under accession number KCTC 12398BP.
Example 2: Evaluation of Anti-Inflammatory Effect of Akkermansia muciniphila Strain
2.1. Production of Pasteurized Cells for Evaluation of Anti-Inflammatory Effect
A culture of the Akkermansia muciniphila EB-AMDK27 strain was centrifuged at 12,000x g and 4℃ for 5 minutes, and the cells harvested, suspended in PBS, and adjusted to an OD value of 0.25 ± 0.03 (8 log CFU/mL). Next, the cells were pasteurized at 70℃ for 30 minutes and stored in a cryogenic freezer until use.
2.2. Evaluation of Anti-Inflammatory Effect in HT-29 Intestinal Epithelial Cells
Since cytokines and other immunomodulators were involved in the regulation of the inflammatory response in inflammatory bowel disease, the present inventors investigated whether the expression of these genes would be affected by administration of the strain of the present invention. For an in vitro test for evaluation of anti-inflammatory efficacy, human colonic epithelial HT-29 cells (ATCC ® HTB-38 TM, USA) were cultured. The cells were cultured in an incubator (NUAIRE, USA) at 37℃ under 5% CO 2 using, as basal culture medium, McCoy's 5A modified medium (Gibco, USA) supplemented with 10% FBS (fetal bovine serum, Hyclone, USA) and 10 μg/ml gentamicin. In order to examine whether the Akkermansia muciniphila EB-AMDK27 strain would inhibit LPS-induced expression of the pro-inflammatory cytokines IL-8, TNF-α, IL-6 and IL-1β genes in HT-29 cells, real-time PCR was performed using the primers shown in Table 8 below.
Total RNA was extracted using TRI reagent (Sigma, USA), and for cDNA synthesis, 1 μg of the RNA was synthesized into cDNA using an M-MLV cDNA synthesis kit (Enzynomics, Korea). Real-time PCR was performed using a Quant Studio 3 real time PCR system (Applied Biosystems, USA).
The expression of inflammatory cytokines was analyzed using SYBR Green TOPreal TM qPCR 2X PreMIX (Enzynomics, Korea), and GAPDH was used as an internal standard. PCR was performed under the following conditions: pre-incubation (for UDG) of 4 min at 50℃, 10 min at 95℃, and 40 cycles, each consisting of 15 sec at 95℃ and 1 min at 60℃. Data were analyzed by delta CT method using the program built in QuantStudio Design & Analysis Software v1.4.3, and the results are shown in FIG. 6.
The results obtained by all the experiments in this Example were calculated as the mean and standard deviation of each group using the statistical program GraphPad Prism 7 (GraphPad software Inc., USA). The difference between the groups was analyzed using one-way ANOVA and Tukey's test, and a p value of 0.05 or less was considered statistically significant. AUC (area under curve) was calculated from some results. From some data, AUC (area under curve) was calculated.
Targets Primer sequences SEQ ID NOs.
GAPDH F: 5'- GAC ATC AAG AAG GTG GTG AAG CAG-3' SEQ ID NO: 7
R: 5'- ATA CCA GGA AAT GAG CTT GAC AAA-3' SEQ ID NO: 8
IL-8 F: 5'- TTT TGC CAA GGA GTG CTA AAG A-3' SEQ ID NO: 9
R: 5'- AAC CCT CTG CAC CCA GTT TTC -3' SEQ ID NO: 10
TNF-α F: 5'- AGC CCA TGT TGT AGC AAA CC-3' SEQ ID NO: 11
R: 5'- TGA GGT ACA GGC CCT CTG AT-3' SEQ ID NO: 12
IL-6 F: 5'- AAA GAG GCA CTG GCA GAA AA-3' SEQ ID NO: 13
R: 5'- TTT CAC CAG GCA AGT CTC CT-3' SEQ ID NO: 14
IL-1β F: 5'- CCG ACC ACC ACT ACA GCA AG-3' SEQ ID NO: 15
R: 5'- GGG CAG GGA ACC AGC ATC TT-3' SEQ ID NO: 16
Referring to FIG. 6, it was shown that treatment with the Akkermansia muciniphila EB-AMDK27 strain of the present invention decreased the mRNA expression of pro-inflammatory cytokines (such as IL-8, TNF-α, IL-6 and IL-1β), which was increased by the inflammation inducer LPS. This suggests that the strain of the present invention can be effective for the treatment and prevention of inflammatory disease.
Example 3: Evaluation of Lipid Accumulation Inhibitory Effect of Akkermansia muciniphila Strain
Whether the expression of biomarkers associated with lipid accumulation and obesity would be influenced by administration of the strain of the present invention was examined. FIG. 7 depicts photographs and a graph, which show the results of measuring lipid accumulation following treatment with the strain of the present invention.
3.1. Oil Red-O Staining of Cells
In order to evaluate the effect of the Akkermansia muciniphila EB-AMDK27 strain of the present invention on lipid accumulation during adipose differentiation and adipose production in 3T3-L1 cells, Oil Red-O staining was performed (Jeon T. et al., Red yeast rice extracts suppress adipogenesis by down-regulating adipogenic transcription factors and gene expression in 3T3-L1, Life Sci., 12;75(26), pp.3195-3203, 2004).
Oil Red-O staining is a method of measuring lipid accumulation in differentiated 3T3-L1 cells by staining the cells with Oil Red-O reagent. Mouse preadipocyte 3T3-L1 cells (Korean Cell Line Bank, KOREA) were cultured in an incubator (NUAIRE, USA) at 37℃ under 5% CO 2 using, as basal culture medium, Dulbecco's Modified Eagle's Medium (DMEM) supplemented with 10% FBS (fetal bovine serum, Hyclone, USA) and 1% penicillin/streptomycin. Adipose differentiation of the preadipocyte 3T3-L1 cells was induced by treatment with insulin (1 μg/ml), IBMX (0.5 mM) and dexamethasone (1 μM) for 10 days, and the cells were treated with the Akkermansia muciniphila EB-AMDK27 strain. Then, the culture was washed three times with PBS to remove the medium. 10% formalin (Sigma, USA) was added and Oil red O (Sigma, USA) solution was allowed to react with the cells for 1 hour, followed by washing with distilled water, thereby staining the lipid droplets.
After the completion of cell staining, the cells were washed three times with 40% isopropanol (Duksan, KOREA), and then dried. The size of lipid droplets in the cells was observed under an optical microscope, and the results are shown in FIG. 7(A). The lipid droplet sample stained with the Oil red O solution was lysed with isopropanol, and the absorbance at 500 nm was measured using a spectrophotometer (Epoch, BioTek, USA). The results of the measurement are shown in FIG. 7(B).
As a result, as shown in FIG. 7(B), it was confirmed that when the 3T3-L1 cells were treated with the Akkermansia muciniphila EB-AMDK27 strain of the present invention during differentiation of the cells, adipose accumulation in the treated cells was significantly inhibited compared to that in the control group administered with the Akkermansia muciniphila BAA-835 strain.
3.2. Evaluation of Effect against Biomarker Gene Expression
In order to evaluate the effect of the strain of the present invention on the inhibition of adipocyte differentiation, the mRNA expression of the following genes was analyzed by performing real-time PCR using the gene-specific primers shown in Table 9 below: PPARγ (peroxisome proliferator-activated receptor gamma), CEBPα (CCAAT/enhancer binding protein alpha), adipocyte protein 2 (aP2), CD36 (cluster differentiating 36), FAS (fatty acid synthase), ACC1 (acetyl-coenzyme A-carboxylase), LPL (lipoprotein lipase), and LDLR (low-density lipoprotein receptor).
Specifically, total RNA was extracted from cell monolayers using TRI reagent (Sigma, USA) according to the manufacturer's instruction, and cDNA was synthesized from 1 μg of the total RNA using an M-MLV cDNA synthesis kit (Enzynomics, Korea). PCR reaction was performed using the Quant Studio 3 real time PCR system (Applied Biosystems, USA) under the following conditions: pre-incubation (for UDG) of 4 min at 50℃, 10 min at 95℃, and 40 cycles, each consisting of 15 sec at 95℃ and 1 min at 60℃. Data were analyzed by delta CT using the program built in QuantStudio Design & Analysis Software v1.4.3.
Targets Primer sequences SEQ ID NOs.
GAPDH F: 5'-GAC ATC AAG AAG GTG GTG AAG CAG-3' SEQ ID NO: 17
R: 5'-ATA CCA GGA AAT GAG CTT GAC AAA-3' SEQ ID NO: 18
PPARγ F: 5'-CAA GAA TAC CAA AGT GCG ATC AA-3' SEQ ID NO: 19
R: 5'-GAG CTG GGT CTT TTC AGA ATA ATA AG-3' SEQ ID NO: 20
CEBPα F: 5'-AGC AAC GAG TAC CGG GTA CG-3' SEQ ID NO: 21
R: 5'-TGT TTG GCT TTA TCT CGG CTC-3' SEQ ID NO: 22
aP2 F: 5'-AGT GAA AAC TTC GAT GAT TAC ATG AA-3' SEQ ID NO: 23
R: 5'-GCC TGC CAC TTT CCT TGT G-3' SEQ ID NO: 24
CD36 F: 5'-TTG TAC CTA TAC TGT GGC TAA ATG AGA-3' SEQ ID NO: 25
R: 5'-CTT GTG TTT TGA ACA TTT CTG CTT-3' SEQ ID NO: 26
FAS F: 5'-AGG GGT CGA CCT GGT CCT CA-3' SEQ ID NO: 27
R: 5'-GCC ATG CCC AGA GGG TGG TT-3' SEQ ID NO: 28
ACC1 F: 5'-CCT CCG TCA GCT CAG ATA CA-3' SEQ ID NO: 29
R: 5'-TTT ACT AGG TGC AAG CCA GAC A-3' SEQ ID NO: 30
LPL F: 5'-TTG CCC TAA GGA CCC CTG AA-3' SEQ ID NO: 31
R: 5'-ACA GAG TCT GCT AAT CCA GGA AT-3' SEQ ID NO: 32
LDLR F: 5'-TGA CTC AGA CGA ACA AGG CTG-3' SEQ ID NO: 33
R: 5'-ATC TAG GCA ATC TCG GTC TCC-3' SEQ ID NO: 34
FIG. 8 depicts graphs showing the results of measuring the mRNA expression of genes associated with preadipocyte-to-adipocyte differentiation and transcription.
In general, the expression of PPARγ (Peroxisome proliferator-activated receptor γ), CEBPα (CCAAT/enhancer binding protein α), aP2α (activator protein 2 alpha), CD36, ACC1, LPL, LDLR, and FAS (fatty acid synthase) increases in the stage of differentiation to adipocytes.
Referring to FIG. 8, when the differentiation of 3T3-L1 cells to adipocytes was induced by treatment with adipocyte differentiation medium (DM), the degree of lipid accumulation in micrographs of the lipid droplets and in the cells significantly decreased in the group treated with DM + the Akkermansia muciniphila BAA-835 strain or DM + the pasteurized Akkermansia muciniphila EB-AMDK27 strain (10% v/v) compared to that in the control group (p<0.001), and it also significantly decreased in the group treated with the pasteurized Akkermansia muciniphila EB-AMDK27 strain compared to that in the positive control group (ATCC BAA-835) (P<0.01).
In addition, as shown in FIG. 8, when increased expression of PPARγ, CEBPα, aP2, CD36, ACC1, LPL, LDLR and FAS, which are genes involved in adipocyte differentiation, was calculated relative to 100% after the induction of adipocyte differentiation, it could be seen that the expression of PPARγ (p<0.001), CEBPα (p<0.001), aP2 (p<0.001), CD36 (p<0.001), ACC1 (p<0.01), LPL (p<0.001), LDLR (p<0.05), FAS (p<0.001) significantly decreased in the group treated with DM + the Akkermansia muciniphila BAA-835 strain, and the expression of PPARγ (p<0.001), CEBPα (p<0.001), aP2 (p<0.001), CD36 (p<0.001), ACC1 (p<0.001), LPL (p<0.001), LDLR (p<0.01) and FAS (p<0.001) significantly decreased in the group treated with DM + the EB-AMDK27 dead cells. In addition, the expression of PPARγ (p<0.001), CEBPα (p<0.001), CD36 (p<0.01) and LPL (p<0.01) significantly decreased in the group treated with the Akkermansia muciniphila BAA-835 strain compared to the group treated with the BAA-835 strain. That is, it could be confirmed that the Akkermansia muciniphila EB-AMDK27 strain of the present invention had a much better effect on the inhibition of adipocyte differentiation of 3T3-L1 cells than the Akkermansia muciniphila BAA-835 strain.
Example 4: Evaluation of Anti-Obesity Effect
4.1. Animal Models and Sampling
Animal experiments were performed in accordance with the Animal Use and Care Protocol of the Institutional Animal Care and Use Committee (IACUC). As experimental animals, 8-week-old male C57BL mice (6 mice per group) were purchased and acclimated for one week, and then bred and housed for 8 weeks. During breeding, the animals were acclimated for one week at a temperature of 22℃ and a relative humidity of 40 to 60% with 12-hr light/12-hr dark cycles.
To induce obesity, the animals were fed with high-fat diet (60 kcal% fat; Research Diets Inc., NJ, USA) and allowed to freely access drinking water.
The experimental animals were divided into five groups as follows:
- Experimental group I (Normal): normal diet group
- Experimental group II (HFD): obesity-induced group fed with high-fat diet
- Experimental group III (GC): group fed with high fat diet for obesity induction and administered with Garcinia cambogia
- Experimental group IV (BAA-835): group fed with high fat diet for obesity induction and administered with Akkermansia muciniphila BAA-835 type strain
- Experimental group V (EB-AMDK27): group fed with high fat diet for obesity induction and administered with Akkermansia muciniphila EB-AMDK27 strain (a probiotic form).
In the case of experimental group V, after the induction of obesity by high-fat diet, Akkermansia muciniphila EB-AMDK27 dead cells were orally administered every day at a concentration of 1 x 10 8 CFU/150 ㎕ PBS (25% glycerol and 0.05% cysteine/PBS).
Normal mice were fed with 10% fat diet. As negative controls, each of Garcinia Cambogia (GC, 60% HCA 1000 mg/kg), known as a food functional food useful for reducing body weight, and the Akkermansia muciniphila BAA-835 strain, was administered. To the normal group and the high-fat diet group (HFD), PBS (25% glycerol and 0.05% cysteine/PBS) was orally administered every day in the same amount as that of the high-fat diet or Garcinia Cambogia in order to exclude the effect of administration-induced stress or the like.
4.2. Changes in Body Weight and Feed Intake
At 12 weeks after obesity induction, the changes in body weight, food intake and calorie intake of each experimental group by administration of the Akkermansia muciniphila EB-AMDK27 strain were measured, and the results of the measurement are shown in FIG. 9.
Referring to FIG. 9, even in the group administered with Garcinia Cambogia or the Akkermansia muciniphila BAA-835 strain, a slight reduction in the body weight was observed, but a significant reduction in the body weight could not be seen.
In comparison with this, the mice administered with the Akkermansia muciniphila EB-AMDK27 strain showed a significant reduction in body weight compared to the mice fed with high-fat diet (HFD) (P<0.05). In addition, referring to FIG. 9, the food intake of the group administered with the Akkermansia muciniphila EB-AMDK27 strain significantly decreased compared to that of the high-fat diet group. From these results, it can be seen that the Akkermansia muciniphila EB-AMDK27 strain of the present invention induces continued decreases in food intake and body weight.
4.3. Changes in Tissue and Body Fat Weights
While obesity was induced for 12 weeks according to the method of 4.1. above, total body fat was measured using the EchoMRITM-500 Body Composition Analyzer in order to analyze the body composition of the mice administered with the strain of the present invention.
At the end of the experiment, subcutaneous fat, epididymal fat, mesenteric fat, liver and spleen were extracted under CO 2 anesthesia, washed with saline, dewatered, and then weighed. The results of the measurement are shown in FIG. 10.
Referring to FIG. 10, it could be seen that all the group administered with Garcinia Cambogia (GC), the group administered with the Akkermansia muciniphila BAA-835 strain, and the group administered with the Akkermansia muciniphila EB-AMDK27 strain showed significant decreases in fat weights compared to the high-fat diet group. These results suggest that the Akkermansia muciniphila strain of the present invention contributed to suppressing body weight gain by reducing lipid accumulation and inflammation in the spleen and liver cells.
As shown in FIG. 10, the weights of liver, spleen, subcutaneous fat, epididymal fat and mesenteric fat all significantly increased in the group fed with high-fat diet (HFD) compared to the normal group, and the weights of liver, spleen, subcutaneous fat, epididymal fat and mesenteric fat significantly decreased in the group administered with the Akkermansia muciniphila EB-AMDK27 strain. In particular, in the case of subcutaneous fat, the group administered with the Akkermansia muciniphila EB-AMDK27 strain showed a more significant decrease in subcutaneous fat weight compared to the group administered with Garcinia Cambogia or the Akkermansia muciniphila BAA-835 strain (P<0.01). In the case of epididymal fat, only the group administered with the Akkermansia muciniphila EB-AMDK27 strain showed a significant decrease in epididymal fat weight compared to the high-fat diet group (P<0.05).
These results suggest that administration of the Akkermansia muciniphila EB-AMDK27 strain has a better effect on the suppression of obesity induced by high-fat diet than administration of Garcinia Cambogia or the Akkermansia muciniphila BAA-835 strain.
In particular, these results show that the Akkermansia muciniphila EB-AMDK27 strain has a strong effect on the reduction of body fat, known to cause complications such as cardiovascular disease, rather than a simple effect on weight loss, and has a greater significance as an anti-obesity agent.
Example 5: Evaluation of Effect on Treatment or Prevention of Metabolic Disease
5.1. Oral Glucose Tolerance Test (OGTT)
In order to evaluate the effect of administration of the Akkermansia muciniphila EB-AMDK27 strain on glucose tolerance, at 12 weeks after the start of the experiment, 2 g/kg of glucose was orally administered to the mice after 18 hours of fasting. Immediately before and at 30, 60, 90 and 120 minutes after administration of glucose, blood was taken from the tail vein of each mouse, and the glucose level of the blood was measured with a blood glucose meter. The results of the measurement are shown in FIG. 11.
Referring to FIG. 11, the group administered with the Akkermansia muciniphila EB-AMDK27 strain immediately before glucose administration showed the greatest decrease in the blood glucose level among the groups administered, but this decrease was not significant.
30 minutes after glucose administration, the blood glucose levels in all the groups administered decreased compared to that of the high-fat diet group (P<0.001), and the blood glucose level in the group administered with the Akkermansia muciniphila EB-AMDK27 strain remarkably decreased compared to those of the other groups to a level that is insignificant compared to that of the normal group. These results show that oral administration of the Akkermansia muciniphila EB-AMDK27 strain can alleviate obesity-induced hyperlipidemia and enhance glucose tolerance.
5.2. Analysis of Blood Lipid Biochemical Indicators
After 18 hours of fasting, blood was taken from each experimental animal, and using the plasma separated from the blood, insulin concentration and insulin resistance index (HOMA-IR) levels were measured, and the concentration of total cholesterol (TC), a lipid content indicator, and the concentration of GPT, a liver function indicator, were measured.
Specifically, the insulin concentration was measured using an insulin ELISA kit (Morinaga, Japan), and the insulin resistance index (HOMA-IR index) was calculated using the following equation:
Fasting insulin concentration (mU/L) × fasting glucose concentration (mmol/L) / 22.5
Total cholesterol (TC) and glutamic pyruvic transaminase (GPT), which were lipid composition indicators, were quantified using the respective measurement kits all purchased from Asan Pharmaceutical Co., Ltd. (Korea). The results are shown in FIG. 12.
The results of measurement of the insulin concentration showed that the insulin concentration and insulin resistance index (HOMA-IR) that increased in the high-fat diet group significantly decreased in all the groups administered. In addition, it was observed that the cholesterol level and insulin resistance index that increased in the high-fat diet group significantly decreased only in the group administered with the Akkermansia muciniphila EB-AMDK27 strain (P<0.05). The GPT concentration indicative of the degree of liver damage significantly decreased in all the groups administered. The blood insulin, cholesterol and GPT concentrations that decreased due to administration of the Akkermansia muciniphila EB-AMDK27 strain are closely related to the weight loss effect. Taking the above results together, it can be seen that the strain and pharmaceutical composition of the present invention can be effectively used to improve carbohydrate and lipid metabolisms for the treatment of obesity.
5.3. Alleviation of Damage to Colonic Mucosa
5.3.1 Histological Evaluation of Colonic Mucosa
In order to examine whether the Akkermansia muciniphila EB-AMDK27 strain of the present invention has the effect of treating damage to colonic mucosa, colon tissues from the normal group, the high-fat diet group (HFD), the group administered with Garcinia Cambogia (GC), the group administered with the Akkermansia muciniphila ATCC BAA-835 strain (BAA-835), and the group administered with the Akkermansia muciniphila EB-AMDK27 strain (EB-AMDK27) were stained with Alcian blue staining and imaged (FIG. 13). In addition, the results obtained by quantifying the positive area (%) in each group on the basis of these results are graphically shown in FIG. 13.
As shown in FIG. 13, it was confirmed that the goblet cells and superficial epithelial tissue in the colon tissue from the high-fat diet group (HFD) were lost and a number of immune cells also infiltrated the colon tissue.
In contrast, it could be seen that the mucosa structure in the colon tissue from the group administered with the Akkermansia muciniphila EB-AMDK27 strain of the present invention was maintained and was similar to that in the colon tissue from the normal group. In addition, it could be seen that the goblet cells that decreased in the colon tissue from the high-fat diet group (HFD) significantly increased in the group administered with the Akkermansia muciniphila EB-AMDK27 strain (p<0.05).
5.3.2 Evaluation of Expression of Tight Junction Proteins
In order to evaluate the effect of the strain of the present invention on the expression of colonic immune-related hormones and tight junction hormones, PCR was performed in the same manner as Example 2.2 using the primers shown in Table 10 below, and the results are shown in FIG. 14.
Targets Primer sequences SEQ ID NOs.
GAPDH F: 5'-GAC ATC AAG AAG GTG GTG AAG CAG-3' SEQ ID NO: 35
R: 5'-ATA CCA GGA AAT GAG CTT GAC AAA-3' SEQ ID NO: 36
TLR2 F: 5'-AAG GAG GTG CGG ACT GTT TC-3' SEQ ID NO: 37
R: 5'-GAG CCA AAG AGC TCG TAG C-3' SEQ ID NO: 38
TLR4 F: 5'-CCT GAT GAC ATT CCT TCT TCA AC-3' SEQ ID NO: 39
R: 5'-TTG TTT CAA TTT CAC ACC TGG ATA AA-3' SEQ ID NO: 40
GLP-1 F: 5'-GGC ACA TTC ACC AGC GAC TAC-3' SEQ ID NO: 41
R: 5'-CAA TGG CGA CTT CTT CTG GG-3' SEQ ID NO: 42
PYY F: 5'-CGG CAG CGG TAT GGA AAA A-3' SEQ ID NO: 43
R: 5'-TGT GAA GAG CAG TTT GGA GAA CA-3' SEQ ID NO: 44
IL-6 F: 5'-CCT CTG GTC TTC TGG AGT ACC-3' SEQ ID NO: 45
R: 5'-ACT CCT TCT GTG ACT CCA GC-3' SEQ ID NO: 46
TNF-α F: 5'-GAC CCT CAC ACT CAG ATC ATC TTC T-3' SEQ ID NO: 47
R: 5'-CCA CTT GGT GGT TTG CTA CGA-3' SEQ ID NO: 48
MCP-1 F: 5'-AAG AGA TCA GGG AGT TTG CT-3' SEQ ID NO: 49
R: 5'-CTG CCT CCA TCA ACC ACT TT-3' SEQ ID NO: 50
IL-10 F: 5'-ATA ACT GCA CCC ACT TCC CA-3' SEQ ID NO: 51
R: 5'-GGG CAT CAC TTC TAC CAG GT-3' SEQ ID NO: 52
ZO-1 F: 5'-TTT TTG ACA GGG GGA GTG G-3' SEQ ID NO: 53
R: 5'-TGC TGC AGA GGT CAA AGT TCA AG-3' SEQ ID NO: 54
Occludin F: 5'-ATG TCC GGC CGA TGC TCT C-3' SEQ ID NO: 55
R: 5'-TTT GGC TGC TCT TGG GTC TGT AT-3' SEQ ID NO: 56
In addition, as shown in FIG. 14, the expression of the inflammatory mediators TLR2 (toll-like receptor 2, p<0.05), TLR4 (p<0.05), IL-6 (p<0.05), TNF-α (p<0.01) and MCP-1 (monocyte chemoattractant protein-1, p<0.05), which increased in the high-fat diet group, significantly decreased in the group administered with the Akkermansia muciniphila EB-AMDK27 strain (TLR2 (p<0.05), TLR4 (p<0.05), IL-6 (p<0.05), TNF-α (p<0.01), and MCP-1 (p<0.05)). In addition, it was observed that the expression of TLR2, TLR4 and TNF-α also decreased in the group administered with the Akkermansia muciniphila BAA-835. The expression of the anti-inflammatory cytokine IL-10 (p<0.05) significantly increased only in the group administered with the Akkermansia muciniphila EB-AMDK27 strain compared to the high-fat diet group.
Meanwhile, the expression of the appetite suppressant hormone GLP-1 (glucagon like peptide-1) significantly decreased in the high-fat diet group (p<0.01), but significantly increased in the group administered with the Akkermansia muciniphila EB-AMDK27 strain compared to the high-fat diet group (p<0.01). The expression of another appetite suppressant hormone PYY (peptide YY) significantly decreased in the high-fat diet group (p<0.05), but significantly increased in all the group administered with Garcinia Cambogia (p<0.05), the group administered with the Akkermansia muciniphila BAA-835 strain (p<0.01), and the group administered with the EB-AMDK27 strain (p<0.01), compared to the control group. The expression of the tight junction proteins ZO-1 (zonular occludens-1, p<0.01) and occludin (p<0.01) significantly decreased in the high-fat diet group. In contrast, in the group administered with the Akkermansia muciniphila EB-AMDK27 strain of the present invention, the expression of ZO-1 significantly increased (p<0.05), and the tight junction protein occludin also increased, which is expressed in colonic mucosa and maintains intestinal permeability at a suitable level. That is, it can be seen that administration of the Akkermansia muciniphila EB-AMDK27 strain of the present invention provides the effects of alleviating obesity-induced inflammation of colonic mucosa and improving intestinal permeability.
5.4. Amelioration of Hepatic Steatosis
Visceral adipose tissue was extracted from the mice of each experimental group, and then weighed. A portion of the visceral adipose tissue was taken and fixed in 10% buffered formalin. Next, it was embedded in paraffin and sectioned to a thickness of 4 mm using a microtome (Reichert-Jung 2050, ALT, USA). The sections were mounted on glass slides, and then stained by H & E (Hematoxylin & Eosin) and ORO (Oil Red O) staining. Next, the shape and state of hepatocytes and lipid droplets were observed under an optical microscope at 200x to 400x magnification. The results are shown in FIGS. 15 and 16.
Referring to FIGS. 15 and 16, worsened steatosis (p<0.05) and increased lipid droplets (p<0.001) in the high-fat diet group significantly decreased in the group administered with the Akkermansia muciniphila EB-AMDK27 strain (p<0.01 and p<0.001, respectively). It was observed that the decrease rate of lipid droplets in the group administered with the Akkermansia muciniphila EB-AMDK27 strain was more significant than that in the group administered with Garcinia Cambogia or the Akkermansia muciniphila BAA-835 strain, indicating that the Akkermansia muciniphila EB-AMDK27 strain showed the greatest improvement.
5.5. Change in Adipocyte Diameter
Mesenteric fat tissue was isolated from the mice of each experimental group and analyzed by H & E staining, and the results of the analysis are shown in FIG. 17. Referring to FIG. 17, the diameter of adipocytes in the high-fat diet group increased compared to that in the normal group (p<0.001). In contrast, the diameter of adipocytes was observed to decrease in the group administered with Garcinia Cambogia, the group administered with the Akkermansia muciniphila BAA-835 strain, and the group administered with the Akkermansia muciniphila EB-AMDK27 strain, compared to the high-fat diet group (p<0.001). In particular, the diameter of adipocytes in the group administered with the Akkermansia muciniphila EB-AMDK27 strain of the present invention significantly decreased compared to those in the group administered with the Garcinia Cambogia and the group administered with the Akkermansia muciniphila BAA-835 strain.
Example 6: Analysis of Microbiota in Feces
6.1. Changes in Microbial Diversity
For analysis of intestinal microorganisms, feces were collected from the mice of each group, and genomic DNA was extracted therefrom using a QIAamp DNA stool mini kit (Qiagen, USA). To confirm the extracted microbial genomic DNA, the position of the band of the genomic DNA was determined by electrophoresis on 1.5% agarose gel (QA-Agarose, Q-biogene, USA) in 1X TAE buffer using an electrophoresis apparatus at 100 V for 25 minutes. The purity of DNA was measured at A260/A280 using a spectrophotometer.
To amplify a specific portion of variable region 3 of 16S rDNA from the microbial DNA isolated from feces, PCR was performed using specially constructed barcode primers. The amplified PCR product was sequenced using an Ion Torrent Next-Generation sequencing platform which is a next-generation sequencing (NGS) method, and the microbiota was analyzed. After reads with low quality scores or a very small number of reads were removed, the resulting microbial sequences are imported into the Greengenes database, and OUTs (operational taxonomic units) were assigned.
Then, the microbiota between clusters was analyzed using the Quantitative insights into microbial ecology (QIIME) 1.9.0 which is a microbiome data integration analysis tool. To compare the alpha diversity between the groups administered with the microbial strains, PCA analysis was performed based on the results obtained for the normal group, the high-fat diet group, the group administered with the Akkermansia muciniphila BAA-835 strain, and the group administered with the Akkermansia muciniphila EB-AMDK27 strain. The alpha-diversity was analyzed and presented by OTUs and Chao1, and the beta-diversity was analyzed by UniFracbased principal coordinates analysis (PCoA). Analysis of linear discriminant analysis effect size (LEfSe) was conducted through an online program ( http://huttenhower.sph.harvard.edu/galaxy).
As a result of the analysis, there was a difference in the alpha diversity between the normal group and the high-fat diet group (FIG. 8(A)). The group administered with the Akkermansia muciniphila EB-AMDK27 strain showed some difference in diversity from the normal group, the high-fat diet group, and the group administered with the Akkermansia muciniphila BAA-835 strain.
6.2. Linear Discriminant Analysis (LDA)
In order to compare the beta diversity between the experimental groups, microorganisms having many differences in LDA scores between the groups were analyzed at the family levels (FIGS. 18 and 19). As a result, it can be seen that the normal group showed higher abundances of Muribaculaceae, Anaeroplasmataceae, Clostridiaceae and the like compared to other groups. The high-fat diet group showed higher abundances of Prevotellaceae, Streptococcaceae and Lactobacillace compared to other groups, and the group administered with the Akkermansia muciniphila BAA-835 strain showed higher abundances of Marinifilaceae and the like compared to other groups. The group administered with the Akkermansia muciniphila EB-AMDK27 of the present invention showed relatively high abundances of Bacteroidaceae and Peptococcaceae. As a result of the relative abundance analysis of the microorganisms, it was observed that changes in the high-fat diet group compared to the normal group were observed, and Bacteroidales, Clostridiales and Lactobacillale orders at the order level and Lachnospiraceae, Rikenellaceae, Ruminococcaceae, Prevotellaceae and Streptococcaceae families at the family level were restored to the levels of the normal group microorganisms in the group administered with the EB-AMDK27 strain.
Taking the above results together, it can be seen that the strain and pharmaceutical composition of the present invention can be effectively used to improve carbohydrate and lipid metabolisms for the treatment of obesity.
It will be apparent to those skilled in the art that the present invention may be modified or altered in various forms without departing from the spirit and scope thereof. The embodiments disclosed herein are only illustrative of preferred embodiments and are not intended to limit the scope of the present invention. The scope of protection of the present invention should be defined by the appended claims, and the above modifications and variations are intended to fall within the scope of protection of the present invention.
[Accession Number]
Depository authority: Korean Collection for Type Cultures
Accession number: KCTC 13758BP
Deposit date: November 30, 2018
Figure PCTKR2020006176-appb-img-000001

Claims (12)

  1. An Akkermansia muciniphila EB-AMDK27 strain (accession number KCTC 13758BP).
  2. The Akkermansia muciniphila EB-AMDK27 strain of claim 1, which has a 16s rRNA gene of SEQ ID NO: 1.
  3. The Akkermansia muciniphila EB-AMDK27 strain of claim 1, which has an activity of treating inflammatory disease or metabolic disease.
  4. A pharmaceutical composition for preventing or treating inflammatory disease or metabolic disease, which contains the strain according to any one of claims 1 to 3, a culture thereof, or a dried product thereof.
  5. The pharmaceutical composition of claim 4, which contains a probiotic form of the strain or a pasteurized form of the strain.
  6. The pharmaceutical composition of claim 4, which is used for preventing or treating inflammatory bowel disease (IBD), insulin resistance or dyslipidemia, or for reducing cholesterol levels or body weight.
  7. The pharmaceutical composition of claim 4, which contains, as an active ingredient, the Akkermansia muciniphila EB-AMDK27 strain at a concentration of 10 8 to 10 12 CFU/g of the composition, or a culture containing viable cells or pasteurized cells of the Akkermansia muciniphila EB-AMDK27 strain at a concentration of 10 8 to 10 12 CFU/g of the composition.
  8. The pharmaceutical composition of claim 4, which is effective for preventing or treating inflammatory bowel disease (IBD), insulin resistance or dyslipidemia, or for reducing cholesterol levels or body weight.
  9. The pharmaceutical composition of claim 4, which further contains a pharmaceutically acceptable carrier or excipient.
  10. A pharmaceutical composition for suppressing appetite, which contains the Akkermansia muciniphila EB-AMDK27 strain according to any one of claims 1 to 3, a culture thereof, or a dried product thereof.
  11. A composition for increasing secretion of appetite suppressant hormones GLP-1 (glucagon like peptide-1) and PYY (peptide YY), which contains the Akkermansia muciniphila EB-AMDK27 strain according to any one of claims 1 to 3, a culture thereof, or a dried product thereof.
  12. A food for preventing or ameliorating inflammatory disease or metabolic disease, which contains the Akkermansia muciniphila EB-AMDK27 strain according to any one of claims 1 to 3, a culture thereof, or a dried product thereof.
PCT/KR2020/006176 2019-08-23 2020-05-11 Akkermansia muciniphila eb-amdk27 strain and use thereof WO2021040186A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
EP20824072.1A EP3802787A4 (en) 2019-08-23 2020-05-11 Akkermansia muciniphila eb-amdk27 strain and use thereof
US17/258,510 US11439670B2 (en) 2019-08-23 2020-05-11 Akkermansia muciniphila EB-AMDK27 strain and use thereof
JP2021503148A JP7055520B2 (en) 2019-08-23 2020-05-11 Akkermansia muciniphila EB-AMDK27 strain and its use
AU2020339348A AU2020339348B2 (en) 2019-08-23 2020-05-11 Akkermansia muciniphila EB-AMDK27 strain and use thereof

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
KR10-2019-0103510 2019-08-23
KR20190103510 2019-08-23
KR1020200033881A KR102128289B1 (en) 2019-08-23 2020-03-19 NEW Akkermansia muciniphila EB-AMDK27 strain AND uses thereof
KR10-2020-0033881 2020-03-19

Publications (1)

Publication Number Publication Date
WO2021040186A1 true WO2021040186A1 (en) 2021-03-04

Family

ID=71121190

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2020/006176 WO2021040186A1 (en) 2019-08-23 2020-05-11 Akkermansia muciniphila eb-amdk27 strain and use thereof

Country Status (6)

Country Link
US (1) US11439670B2 (en)
EP (1) EP3802787A4 (en)
JP (1) JP7055520B2 (en)
KR (1) KR102128289B1 (en)
AU (1) AU2020339348B2 (en)
WO (1) WO2021040186A1 (en)

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR102185827B1 (en) * 2020-08-26 2020-12-03 주식회사 엔테로바이옴 Pharmaceutical composition for preventing or treating atopic disease comprising akkermansia muciniphila strain
US20230381246A1 (en) * 2020-10-19 2023-11-30 Duke University Genetically diverse akkermansia strains and methods of using same
CN113322202B (en) * 2021-05-31 2022-03-01 君维安(武汉)生命科技有限公司 Ackermanella, culture method and application thereof
CN114949004B (en) * 2022-06-08 2024-04-26 广州知易生物科技有限公司 Application of akkermansia muciniphila in preparation of composition for preventing and treating hyperlipidemia and composition containing akkermansia muciniphila
CN114832019A (en) * 2022-06-08 2022-08-02 广州知易生物科技有限公司 Application of akkermansia muciniphila in preparation of pharmaceutical composition for preventing and treating obesity, composition and application thereof
CN116200312B (en) * 2023-03-30 2023-08-22 广西爱生生命科技有限公司 Antioxidant, lipid-reducing and tumor growth-inhibiting Guangxi Akkera and product and application thereof

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR100996577B1 (en) 2009-04-01 2010-11-24 주식회사한국야쿠르트 Lactobacillus curvatus HY7601 having inhibitory activity against blood cholestrol and obesity, and product containing thereof as an effective factor
KR20110010015A (en) 2009-07-23 2011-01-31 삼성전기주식회사 Semiconductor package and method of manufacturing the same
KR20110095929A (en) 2005-07-26 2011-08-25 네스텍 에스.아. Anti-obesity agent and anti-obesity food
KR20130021920A (en) * 2011-08-24 2013-03-06 포항공과대학교 산학협력단 Composition comprising extracellular vesicles derived from akkermansia muciniphila and bacteroides acidifaciens as an active ingredient for treating or preventing inflammatory disease
KR20150093711A (en) * 2012-11-19 2015-08-18 위니베르시트카솔리끄드루뱅 Use of akkermansia for treating metabolic disorders
KR20150133646A (en) * 2014-05-20 2015-11-30 이화여자대학교 산학협력단 COMPOSITION COMPRISING EXTRACELLULAR VESICLES DERIVED FROM Akkermansia muciniphila AS AN ACTIVE INGREDIENT FOR TREATING OR PREVENTING METABOLIC DISEASE
KR101809172B1 (en) * 2016-07-11 2017-12-14 한국생명공학연구원 Composition for preventing, improving or treating metabolic disease comprising Akkermansia muciniphila strain or its culture broth cultivated in medium without mucin as effective component
KR20190034796A (en) 2017-09-25 2019-04-03 건국대학교 산학협력단 NOVEL STRAIN OF Lactobacillus plantarum AND COMPOSITION FOR PREVENTING OR TREATING OF INFLAMMATORY DISEASE USING THE SAME

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8906668B2 (en) * 2012-11-23 2014-12-09 Seres Health, Inc. Synergistic bacterial compositions and methods of production and use thereof
CN105106245A (en) * 2015-08-24 2015-12-02 上海交通大学医学院附属瑞金医院 Application of Akkermansia muciniphila BAA-835 bacterial strain
DK3626081T3 (en) * 2015-09-10 2022-02-28 Univ Catholique Louvain PASTEURIZED AKKERMANSIA TO PROMOTE WEIGHT LOSS
US10537597B2 (en) * 2015-10-05 2020-01-21 Schweizerisches Forschungsinstitut Fur Hochgebrigsklima Und Medizin In Davos Use of Akkermansia muciniphila for treating inflammatory conditions
JP6275931B1 (en) * 2016-03-14 2018-02-07 物産フードサイエンス株式会社 Intestinal butyric acid increasing agent and butyric acid-producing bacteria proliferating agent
EP3483256A4 (en) * 2016-07-11 2019-12-04 Korea Research Institute of Bioscience and Biotechnology Akkermansia muciniphila strain having effect of preventing or treating degenerative brain diseases or metabolic diseases, and use thereof
JP6967404B2 (en) * 2017-08-31 2021-11-17 雪印メグミルク株式会社 Composition for promoting Akkermansia mucinifira increase
JP7094537B2 (en) * 2018-02-15 2022-07-04 株式会社シトリアン Yuzu (Yuzu) -containing composition

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20110095929A (en) 2005-07-26 2011-08-25 네스텍 에스.아. Anti-obesity agent and anti-obesity food
KR100996577B1 (en) 2009-04-01 2010-11-24 주식회사한국야쿠르트 Lactobacillus curvatus HY7601 having inhibitory activity against blood cholestrol and obesity, and product containing thereof as an effective factor
KR20110010015A (en) 2009-07-23 2011-01-31 삼성전기주식회사 Semiconductor package and method of manufacturing the same
KR20130021920A (en) * 2011-08-24 2013-03-06 포항공과대학교 산학협력단 Composition comprising extracellular vesicles derived from akkermansia muciniphila and bacteroides acidifaciens as an active ingredient for treating or preventing inflammatory disease
KR20150093711A (en) * 2012-11-19 2015-08-18 위니베르시트카솔리끄드루뱅 Use of akkermansia for treating metabolic disorders
KR20150133646A (en) * 2014-05-20 2015-11-30 이화여자대학교 산학협력단 COMPOSITION COMPRISING EXTRACELLULAR VESICLES DERIVED FROM Akkermansia muciniphila AS AN ACTIVE INGREDIENT FOR TREATING OR PREVENTING METABOLIC DISEASE
KR101809172B1 (en) * 2016-07-11 2017-12-14 한국생명공학연구원 Composition for preventing, improving or treating metabolic disease comprising Akkermansia muciniphila strain or its culture broth cultivated in medium without mucin as effective component
KR20190034796A (en) 2017-09-25 2019-04-03 건국대학교 산학협력단 NOVEL STRAIN OF Lactobacillus plantarum AND COMPOSITION FOR PREVENTING OR TREATING OF INFLAMMATORY DISEASE USING THE SAME

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
DATABASE PROTEIN 11 December 2013 (2013-12-11), "carboxyl-terminal protease [Akkermansia muciniphila ATCC BAA-835]", XP055785809, retrieved from NCBI Database accession no. ACD05451 *
JEON T. ET AL.: "Red yeast rice extracts suppress adipogenesis by down-regulating adipogenic transcription factors and gene expression in 3T3-L1", LIFE SCI., vol. 75, no. 26, pages 3195 - 3203, XP004601760, DOI: 10.1016/j.lfs.2004.06.012
See also references of EP3802787A4
WRENBLOOM, GASTROENTEROLOGY, vol. 132, 2007, pages 2116 - 2130

Also Published As

Publication number Publication date
US20220133811A1 (en) 2022-05-05
US11439670B2 (en) 2022-09-13
EP3802787A4 (en) 2022-03-23
KR102128289B1 (en) 2020-06-30
AU2020339348B2 (en) 2023-04-13
AU2020339348A1 (en) 2021-08-05
JP7055520B2 (en) 2022-04-18
EP3802787A1 (en) 2021-04-14
JP2022502005A (en) 2022-01-11

Similar Documents

Publication Publication Date Title
AU2020336649B2 (en) Akkermansia muciniphila EB-AMDK19 strainand use thereof
AU2020339348B2 (en) Akkermansia muciniphila EB-AMDK27 strain and use thereof
WO2019216662A1 (en) Lactobacillus paracasei strain and use thereof
WO2020050460A1 (en) Pharmaceutical composition for preventing or treating inflammatory bowel diseases
WO2021261632A1 (en) Novel faecalibacterium prausnitzii strain eb-fpdk11 and use thereof
WO2020076136A2 (en) Akkermansia muciniphila strain and use thereof
WO2021194225A1 (en) Lactobacillus delbrueckii subsp. lactis ckdb001 strain, and composition for prevention, amelioration, or treatment of non-alcoholic fatty liver comprising same
WO2023055188A1 (en) Novel probiotics and use thereof
WO2020262755A1 (en) Novel probiotic composition for regulation of intestinal immunity
WO2018164468A1 (en) Bacillus amyloliquefaciens gf423 strain, and composition for providing antioxidant and anti-inflammatory activities or preventing or treating hyperlipidemia, including polypeptide produced by the same
AU2021212021B2 (en) Pharmaceutical composition for preventing or treating atopic disease containing Akkermansia muciniphila strain
WO2022039561A1 (en) Composition for treating or preventing clostridium difficile infection
WO2022035269A1 (en) Faecalibacterium prausnitzii strain and uses thereof
WO2023113541A1 (en) Pharmaceutical composition for preventing or treating cancer or inflammatory disease
WO2020226438A1 (en) Peptide for preventing or treating inflammatory bowel diseases
WO2023058801A1 (en) Composition for alleviating, preventing, or treating bowel disorder, comprising lactobacillus acidophilus kbl402 or kbl409 strain
WO2021261631A1 (en) Novel picalibacterium prosnich eb-fpdk9 strain and uses thereof
WO2021020923A1 (en) Composition and method for preventing, alleviating, or treating liver injury
WO2023158204A1 (en) Pharmaceutical composition for preventing or treating cancer
WO2023200203A1 (en) Novel microorganism and composition comprising same
WO2022265430A1 (en) Combination therapy use of lactobacillus fermentum strain and natural killer cells for preventing and treating metabolic diseases
WO2023167563A1 (en) Composition containing n-carbamyl-l-glutamic acid for treatment of inflammatory diseases
WO2023277638A1 (en) Composition comprising three lactobacillus sp. strains, and use thereof
WO2024038980A1 (en) Lactobacillus plantarum ku15120 strain or use thereof
WO2023277636A1 (en) Anti-aging composition containing lactobacillus sp. strain and medicinal herb material for combined therapy

Legal Events

Date Code Title Description
ENP Entry into the national phase

Ref document number: 2021503148

Country of ref document: JP

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2020824072

Country of ref document: EP

Effective date: 20201222

ENP Entry into the national phase

Ref document number: 2020339348

Country of ref document: AU

Date of ref document: 20200511

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE