US20130302380A1 - Agent for inducing interferon production containing lactic acid bacteria - Google Patents

Agent for inducing interferon production containing lactic acid bacteria Download PDF

Info

Publication number
US20130302380A1
US20130302380A1 US13/977,435 US201113977435A US2013302380A1 US 20130302380 A1 US20130302380 A1 US 20130302380A1 US 201113977435 A US201113977435 A US 201113977435A US 2013302380 A1 US2013302380 A1 US 2013302380A1
Authority
US
United States
Prior art keywords
ifn
lactic acid
agent
acid bacteria
lactococcus lactis
Prior art date
Legal status (The legal status 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 status listed.)
Abandoned
Application number
US13/977,435
Other languages
English (en)
Inventor
Daisuke Fujiwara
Kenta Jonai
Tetsu Sugimura
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Kirin Holdings Co Ltd
Original Assignee
Kirin Holdings Co Ltd
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 Kirin Holdings Co Ltd filed Critical Kirin Holdings Co Ltd
Assigned to KIRIN HOLDINGS KABUSHIKI KAISHA reassignment KIRIN HOLDINGS KABUSHIKI KAISHA ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: FUJIWARA, DAISUKE, JONAI, KENTA, SUGIMURA, TETSU
Publication of US20130302380A1 publication Critical patent/US20130302380A1/en
Priority to US15/367,649 priority Critical patent/US10220060B2/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/66Microorganisms or materials therefrom
    • A61K35/74Bacteria
    • A61K35/741Probiotics
    • A61K35/744Lactic acid bacteria, e.g. enterococci, pediococci, lactococci, streptococci or leuconostocs
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23CDAIRY PRODUCTS, e.g. MILK, BUTTER OR CHEESE; MILK OR CHEESE SUBSTITUTES; MAKING THEREOF
    • A23C19/00Cheese; Cheese preparations; Making thereof
    • A23C19/02Making cheese curd
    • A23C19/032Making cheese curd characterised by the use of specific microorganisms, or enzymes of microbial origin
    • A23C19/0323Making cheese curd characterised by the use of specific microorganisms, or enzymes of microbial origin using only lactic acid bacteria, e.g. Pediococcus and Leuconostoc species; Bifidobacteria; Microbial starters in general
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23CDAIRY PRODUCTS, e.g. MILK, BUTTER OR CHEESE; MILK OR CHEESE SUBSTITUTES; MAKING THEREOF
    • A23C19/00Cheese; Cheese preparations; Making thereof
    • A23C19/06Treating cheese curd after whey separation; Products obtained thereby
    • A23C19/061Addition of, or treatment with, microorganisms
    • A23C19/062Addition of, or treatment with, microorganisms using only lactic acid bacteria, e.g. pediococcus, leconostoc or bifidus sp., or propionic acid bacteria; Treatment with non-specified acidifying bacterial cultures
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23CDAIRY PRODUCTS, e.g. MILK, BUTTER OR CHEESE; MILK OR CHEESE SUBSTITUTES; MAKING THEREOF
    • A23C9/00Milk preparations; Milk powder or milk powder preparations
    • A23C9/12Fermented milk preparations; Treatment using microorganisms or enzymes
    • A23C9/123Fermented milk preparations; Treatment using microorganisms or enzymes using only microorganisms of the genus lactobacteriaceae; Yoghurt
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23CDAIRY PRODUCTS, e.g. MILK, BUTTER OR CHEESE; MILK OR CHEESE SUBSTITUTES; MAKING THEREOF
    • A23C9/00Milk preparations; Milk powder or milk powder preparations
    • A23C9/12Fermented milk preparations; Treatment using microorganisms or enzymes
    • A23C9/123Fermented milk preparations; Treatment using microorganisms or enzymes using only microorganisms of the genus lactobacteriaceae; Yoghurt
    • A23C9/1236Fermented milk preparations; Treatment using microorganisms or enzymes using only microorganisms of the genus lactobacteriaceae; Yoghurt using Leuconostoc, Pediococcus or Streptococcus sp. other than Streptococcus Thermophilus; Artificial sour buttermilk in general
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23LFOODS, FOODSTUFFS, OR NON-ALCOHOLIC BEVERAGES, NOT COVERED BY SUBCLASSES A21D OR A23B-A23J; THEIR PREPARATION OR TREATMENT, e.g. COOKING, MODIFICATION OF NUTRITIVE QUALITIES, PHYSICAL TREATMENT; PRESERVATION OF FOODS OR FOODSTUFFS, IN GENERAL
    • A23L33/00Modifying nutritive qualities of foods; Dietetic products; Preparation or treatment thereof
    • A23L33/10Modifying nutritive qualities of foods; Dietetic products; Preparation or treatment thereof using additives
    • A23L33/135Bacteria or derivatives thereof, e.g. probiotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/711Natural deoxyribonucleic acids, i.e. containing only 2'-deoxyriboses attached to adenine, guanine, cytosine or thymine and having 3'-5' phosphodiester links
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/66Microorganisms or materials therefrom
    • A61K35/74Bacteria
    • A61K35/741Probiotics
    • A61K35/744Lactic acid bacteria, e.g. enterococci, pediococci, lactococci, streptococci or leuconostocs
    • A61K35/747Lactobacilli, e.g. L. acidophilus or L. brevis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0053Mouth and digestive tract, i.e. intraoral and peroral administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • 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
    • 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
    • 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
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/74Vectors or expression systems specially adapted for prokaryotic hosts other than E. coli, e.g. Lactobacillus, Micromonospora
    • C12N15/746Vectors or expression systems specially adapted for prokaryotic hosts other than E. coli, e.g. Lactobacillus, Micromonospora for lactic acid bacteria (Streptococcus; Lactococcus; Lactobacillus; Pediococcus; Enterococcus; Leuconostoc; Propionibacterium; Bifidobacterium; Sporolactobacillus)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6809Methods for determination or identification of nucleic acids involving differential detection
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6863Cytokines, i.e. immune system proteins modifying a biological response such as cell growth proliferation or differentiation, e.g. TNF, CNF, GM-CSF, lymphotoxin, MIF or their receptors
    • G01N33/6866Interferon
    • 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
    • A23V2400/00Lactic or propionic acid bacteria
    • A23V2400/21Streptococcus, lactococcus
    • A23V2400/231Lactis
    • 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 an agent for inducing interferon production containing lactic acid bacteria and a pharmaceutical product and a food or drink product containing the agent for inducing interferon production.
  • Bacteria including lactic acid bacteria, are recognized and englobed by a group of immunocytes referred to as the innate immune system, and they cause physiological reactions, such as cytokine or chemokine production, changes in gene expression, and epigenetic gene modification.
  • Cells of the innate immune system can be roughly classified as macrophages, natural killer (NK) cells, and dendritic cells.
  • NK natural killer
  • dendritic cells In contrast to the long-term antigen-specific reactions of the acquired immune system, the reactions of the innate immune system are short-lasting, non-antigen-specific, inclusive reactions.
  • the innate immune system plays a key role in primary responses against infection with bacteria or viruses, and, in particular, dendritic cells are potent and critical constitutive cells.
  • Dendritic cells are highly flexible, a great number of different subspecies thereof exist, and these cells can be roughly classified into myeloid dendritic cells (mDCs), CD8+ dendritic cells (CD8+ DCs), and plasmacytoid dendritic cells (pDCs).
  • mDCs mainly release inflammatory cytokines, such as interleukin-12 (IL-12) and tumor necrosis factor- ⁇ (TNF- ⁇ ), upon infection with bacteria and induce activation of helper T cells (CD4+ T cells).
  • IL-12 interleukin-12
  • TNF- ⁇ tumor necrosis factor- ⁇
  • CD8+ DCs are high-power cells producing IL-12, which play a key role in induction of cytotoxic T lymphocytes (CTLs) upon virus infection or cross-priming of cancer antigens.
  • CTLs cytotoxic T lymphocytes
  • pDCs are major cells producing type I interferon (IFN) exhibiting growth-inhibiting activity against viruses in vivo, and they play a critical role in antiviral biophylaxis.
  • type I interferon include IFN- ⁇ and IFN- ⁇ .
  • TLRs Toll-like receptors
  • endosomal TLRs such as TLR3, TLR7, or TLR9.
  • viral double-stranded RNA, viral single-stranded RNA or an antiviral agent (imidazoquinoline), and non-methylated CpG DNA comprising cytosine and guanine joined by a phosphodiester bond are known as a TLR3 ligand, a TLR7 ligand, and a TLR9 ligand, respectively.
  • nucleic acids of bacteria or viruses are known to serve as ligands in the induction of type I interferon production.
  • IFN- ⁇ has been put into practical use as a therapeutic agent for hepatitis B, hepatitis C, chronic myeloid leukemia, multiple myeloma, renal cancer, and other diseases
  • IFN- ⁇ has been put into practical use as a therapeutic agent for multiple sclerosis, in addition to hepatitis B and hepatitis C.
  • pDC is considered to be the most important cell from the viewpoint of biophylaxis, and antiviral prophylaxis, in particular IFN- ⁇ is cytokine that is classified as type II interferon, and it is mainly produced by NK or Th1 cells, although antiviral effects thereof are weak.
  • IFN- ⁇ is considered to be enhancement of antiviral effects of IFN- ⁇ and IFN- ⁇ .
  • IFN- ⁇ is classified as type III interferon, such a cytokine has drawn attention recently because of its potent antiviral effects verified in recent years.
  • IFN- ⁇ is produced mainly by pDCs in an organism, as with the case of type I IFN.
  • a certain bacteria are known to activate pDC or produce IFN- ⁇ .
  • bacteria that are verified to activate pDC one of the food-poisoning bacteria, Staphylococcus aureus has been reported.
  • Staphylococcus aureus As bacteria that enhance IFN- ⁇ production in the blood, pathogenic bacteria, such as Chlamydia, Salmonella, Mycobacteria , and Listeria , are known. While some lactic acid bacteria have been reported to enhance IFN- ⁇ production (see Non-Patent Documents 1 and 2), the correlation thereof with pDC remains unknown, and screening has never been conducted using the capacity for IFN- ⁇ production or pDC activation as the indicator.
  • lactic acid bacteria have been reported to enhance IFN- ⁇ production (see Patent Documents 1 and 2 and Non-Patent Document 3) and to enhance IFN- ⁇ production (see Patent Document 3); however, the correlation thereof with pDC also remains unknown. While it has been reported that IFN- ⁇ has antiviral activity (see Non-Patent Documents 4 and 5) and IFN- ⁇ is mainly produced by pDCs (Non-Patent Document 6), the correlation of IFN- ⁇ with lactic acid bacteria remains unknown.
  • the present invention is intended to provide an IFN inducer capable of inducing IFN production comprising, as an active ingredient, lactic acid bacteria, an immunopotentiating agent or prophylactic agent against virus infection comprising such inducer, and a food or drink product comprising such inducer and having IFN inducing activity, immunopotentiating activity, or prophylactic activity against virus infection.
  • the present inventors constructed an assay system involving the use of pDC activation as the indicator and selected lactic acid bacteria that would potentiate the prophylactic activity against virus infection through ingestion thereof.
  • lactic acid bacteria would activate pDCs and induce interferon production from such pDCs. They further discovered that some lactic acid bacteria would be capable of exerting the effects in an organism even they were orally administered.
  • the present inventors discovered that the lactic acid bacteria could be used as agents for inducing IFN production, and such bacteria could also be used for prophylaxis against virus infection because of the immunopotentiating activity of an organism. This has led to the completion of the present invention.
  • the present invention is as follows.
  • An agent for inducing IFN production comprising, as an active ingredient, lactic acid bacteria capable of activating plasmacytoid dendritic cells (pDCs) and inducing IFN production or a cultured product thereof.
  • pDCs plasmacytoid dendritic cells
  • IFN is at least one member selected from the group consisting of IFN- ⁇ , IFN- ⁇ , and IFN- ⁇ .
  • [5] The agent for inducing IFN production according to any of [1] to [4], wherein, when the agent is orally administered, the lactic acid bacteria capable of activating plasmacytoid dendritic cells (pDCs) and inducing IFN production are highly tolerant to the gastric juice or intestinal juice and are capable of reaching the intestinal canal alive.
  • pDCs plasmacytoid dendritic cells
  • An immunopotentiating agent comprising the agent for inducing IFN production according to any of [1] to [6].
  • An agent for prevention or treatment of virus infection comprising the agent for inducing IFN production according to any of [1] to [6].
  • [10] The agent for prevention or treatment of virus infection according to [8], which is an oral preparation.
  • a food or drink product comprising the agent for inducing IFN production according to any of [1] to [6].
  • a method of screening for lactic acid bacteria capable of activating plasmacytoid dendritic cells (pDCs) and inducing IFN production comprising culturing the lactic acid bacteria with bone marrow cells and detecting activation of plasmacytoid dendritic cells (pDCs) and induction of IFN production,
  • the lactic acid bacteria are determined to be capable of activating plasmacytoid dendritic cells (pDCs) and inducing IFN production.
  • a host microorganism for a recombinant vaccine comprising the agent for inducing IFN production according to any of [1] to [6].
  • the agent for inducing IFN production comprising, as an active ingredient, the particular lactic acid bacteria of the present invention is capable of activating pDC and inducing production of interferon, such as IFN- ⁇ , in vitro and in vivo.
  • interferon such as IFN- ⁇
  • IFN- ⁇ interferon- ⁇
  • the IFN inducer comprising, as an active ingredient, lactic acid bacteria described above can be used as a pharmaceutical product for potentiating immune responses for prevention or treatment of virus infection. Further, such inducer can be used as a component of a food or drink product for potentiating immune responses useful for prevention of virus infection.
  • FIG. 1A shows a list of the tested lactic acid bacteria (Part 1).
  • FIG. 1B shows a list of the tested lactic acid bacteria (Part 2).
  • FIG. 1C shows a list of the tested lactic acid bacteria (Part 3).
  • FIG. 2A shows the number and ratio of lactic acid bacteria producing 50 pg/ml or more IFN- ⁇ , when compared rod-shaped bacteria with spherical-shaped bacteria.
  • FIG. 2B shows the number and ratio of lactic acid bacteria producing 100 pg/ml or more IFN- ⁇ , when compared rod-shaped bacteria with spherical-shaped bacteria.
  • FIG. 3 shows electron microscopy of Lactococcus lactis JCM5805 ( FIG. 3A ) and Lactococcus lactis JCM20101 ( FIG. 3B ).
  • FIG. 4 shows differences in lactic acid bacteria recognition (uptake) of pDCs.
  • FIG. 4A , FIG. 4B , and FIG. 4C show Lactococcus lactis JCM5805, Lactococcus lactis JCM21101, and Lactobacillus rhamnosus ATCC53103, respectively.
  • FIG. 5A shows the amount of IFN- ⁇ production from various lactic acid bacteria.
  • FIG. 5B shows the amount of IFN- ⁇ production from various lactic acid bacteria.
  • FIG. 5C shows the amount of IFN- ⁇ production from various lactic acid bacteria.
  • FIG. 5D shows the amount of IFN- ⁇ production from various lactic acid bacteria.
  • FIG. 6A shows the capacity for pDC activation of lactic acid bacteria having the capacity for inducing IFN- ⁇ production and the expression levels of MHCI, CD40, CD80, and CD86.
  • FIG. 6B shows the capacity for pDC activation of lactic acid bacteria having the capacity for inducing IFN- ⁇ production and the expression levels of OX40L, PDL-1, and ICOS-L.
  • FIG. 7 shows the capacity for stimulating IFN- ⁇ production of lactic acid bacteria in the presence of either or both pDCs and mDCs.
  • FIG. 8 shows photographs showing pDC configurations when Lactococcus lactis JCM5805, Lactococcus lactis JCM20101, and Lactobacillus rhamnosus ATCC53103 are added to the pDC monoculture system.
  • FIG. 8A shows the results concerning the control (to which no lactic acid bacteria were added)
  • FIG. 8B shows pDCs when Lactococcus lactis JCM5805 is added
  • FIG. 8C shows pDCs when Lactococcus lactis JCM20101 is added
  • FIG. 8D shows pDCs when Lactobacillus rhamnosus ATCC53103 is added.
  • FIG. 9A shows the amounts of IFN- ⁇ production when Lactococcus lactis JCM5805, Lactococcus lactis JCM20101, and Lactobacillus rhamnosus ATCC53103 are added to the pDC/mDC cells generated from TLR2 and TLR4 knockout mice.
  • FIG. 9B shows the amounts of IFN- ⁇ production when Lactococcus lactis JCM5805, Lactococcus lactis JCM20101, and Lactobacillus rhamnosus ATCC53103 are added to the pDC/mDC cells generated from TLR7, TLR9, and MyD88 knockout mice.
  • FIG. 10 shows the capacity for IFN- ⁇ activation of DNAs and RNAs of Lactococcus lactis JCM5805 and Lactococcus lactis JCM20101.
  • FIG. 11 shows a summary of the experimental design examining the effects of ingestion of Lactococcus lactis JCM5805 using healthy mice.
  • FIG. 12 shows changes in blood IFN- ⁇ levels of healthy mice that had ingested Lactococcus lactis JCM5805.
  • FIG. 13A shows changes in MHC class II levels in pDCs of the spleen of healthy mice that had ingested Lactococcus lactis JCM5805.
  • FIG. 13B shows changes in MHC class II levels in pDCs of the mesenteric lymph nodes of healthy mice that had ingested Lactococcus lactis JCM5805.
  • FIG. 13C shows changes in CD86 levels in pDCs of the spleen of healthy mice that had ingested Lactococcus lactis JCM5805.
  • FIG. 13D shows changes in CD86 levels in pDCs of the mesenteric lymph nodes of healthy mice that had ingested Lactococcus lactis JCM5805.
  • FIG. 14 shows a summary of a method for evaluation of Lactococcus lactis JCM5805 using the immunosuppression models.
  • FIG. 15A shows the effects of ingestion of Lactococcus lactis JCM5805 using the immunosuppression models with reference to changes in the body weights of mouse models.
  • FIG. 15B shows the effects of ingestion of Lactococcus lactis JCM5805 using the immunosuppression models with reference to changes in the blood IFN- ⁇ levels of mouse models.
  • FIG. 16A shows changes in MHC class II levels in pDCs of the immunosuppression mouse models that had ingested Lactococcus lactis JCM5805.
  • FIG. 16B shows changes in CD86 levels in pDCs of the immunosuppression mouse models that had ingested Lactococcus lactis JCM5805.
  • FIG. 16C shows the percentage of pDCs in the immunosuppression mouse models that had ingested Lactococcus lactis JCM5805.
  • FIG. 16D shows the results of flow cytometric analysis using lymphocytes of the immunosuppression mouse models that had ingested Lactococcus lactis JCM5805.
  • FIG. 17 shows Lactococcus lactis JCM5805 and a strain equivalent thereto (a strain derived from Lactococcus lactis JCM5805 and a strain from which Lactococcus lactis JCM5805 is derived).
  • FIG. 18 shows the capacity of live Lactococcus lactis JCM5805, Lactococcus lactis JCM20101, and Lactobacillus rhamnosus ATCC53103 for IFN- ⁇ activation.
  • FIG. 19A shows the percentage of pDCs determined by flow cytometric analysis of the purity of human pDCs isolated with MACS.
  • FIG. 19B shows the amount of IFN- ⁇ production detected via ELISA when Lactococcus lactis JCM5805 is added to human pDCs isolated with MACS.
  • FIG. 19C shows IFN- ⁇ 1, IFN- ⁇ , IFN- ⁇ 1, and GAPDH gene expression detected via RT-PCR when Lactococcus lactis JCM5805 is added to human pDCs isolated with MACS.
  • FIG. 20A shows a comparison of changes in MHC class II activity in pDCs of the group subjected to ingestion of yogurt containing Lactococcus lactis JCM5805 and of the placebo group.
  • FIG. 20B shows a comparison of changes in MHC class II activity in pDCs of a subject having high MHC class II activity in the group subjected to ingestion of yogurt containing Lactococcus lactis JCM5805 and in the placebo group.
  • FIG. 20C shows a comparison of changes in MHC class II activity in pDCs of a subject having low MHC class II activity in the group subjected to ingestion of yogurt containing Lactococcus lactis JCM5805 and in the placebo group.
  • FIG. 20D shows a comparison of changes in CD86 activity in pDCs of the group subjected to ingestion of yogurt containing Lactococcus lactis JCM5805 and of the placebo group.
  • FIG. 20E shows a comparison of changes in CD86 activity in pDCs of a subject having high MHC class II activity in the group subjected to ingestion of yogurt containing Lactococcus lactis JCM5805 and in the placebo group.
  • FIG. 20F shows a comparison of changes in CD86 activity in pDCs of a subject having low MHC class II activity in the group subjected to ingestion of yogurt containing Lactococcus lactis JCM5805 and in the placebo group.
  • FIG. 21 shows a comparison of amounts of IFN- ⁇ 1 gene transcription in PBMCs compared at week 0 and week 4 after the initiation of the test of a subject having low MHC class II activity in the group subjected to ingestion of yogurt containing Lactococcus lactis JCM5805 and in the placebo group.
  • FIG. 22 shows a comparison of amounts of IFN- ⁇ production influenced by CpG stimulation in PBMCs compared at week 0 and week 4 after the initiation of the test of a subject having low MHC class II activity in the group subjected to ingestion of yogurt containing Lactococcus lactis JCM5805 and in the placebo group.
  • FIG. 23 shows the results of comparison between the group subjected to ingestion of yogurt containing Lactococcus lactis JCM5805 and the placebo group regarding the development of cold symptoms during the period of test product ingestion examined every week.
  • the present invention relates to an agent for inducing IFN production comprising, as an active ingredient, lactic acid bacteria.
  • agent for inducing IFN production refers to induction of IFN production in vitro and in vivo.
  • Lactic acid bacteria that can be used as the agent for inducing IFN production in the present invention are capable of activating plasmacytoid dendritic cells (pDCs) and promoting IFN production of pDCs. Further, lactic acid bacteria that can be used as the agent for inducing IFN production in the present invention are capable of promoting expression of activation markers, such as CD80, CD86, and MHC class II, in pDCs. Whether or not candidate lactic acid bacteria have such properties may be determined by, for example, culturing candidate lactic acid bacteria in the presence of bone marrow cells generated from mammalians, such as mice, and detecting the occurrence of pDC activation and induction of production of IFN, such as IFN ⁇ and IFN ⁇ .
  • activation markers such as CD80, CD86, and MHC class II
  • IFN may be assayed by measuring the IFN concentration in a culture solution via, for example, ELISA.
  • Lactic acid bacteria that can be used as an agent for inducing IFN production in the present invention has property as described below.
  • mouse bone marrow cells from which erythrocytes have been removed are suspended to a concentration of 5 ⁇ 10 5 cells/ml in RPMI medium (SIGMA) containing 10% FCS and 2 ⁇ M ⁇ -mercaptoethanol, Flt-3L is added as a pDC inducing cytokine to a final concentration of 100 ng/ml to the resulting cell suspension, the resultant is cultured in a CO 2 incubator at 37° C.
  • the culture supernatant is collected 48 hours later, the IFN- ⁇ concentration in the culture supernatant is assayed via ELISA with the use of the IFN- ⁇ assay kit (PBL), and the determined IFN- ⁇ concentration is preferably 50 pg/ml or higher, and more preferably 100 pg/ml or higher.
  • spherical-shaped lactic acid bacteria that belong to the genera Lactococcus, Leuconostoc, Pediococcus , and Streptococcus are more preferable.
  • Particularly preferable strains are Lactococcus garvieae, Lactococcus lactis subsp. cremoris, Lactococcus lactis subsp. lactis, Lactococcus lactis subsp. hordniae, Leuconostoc lactis, Pediococcus damnosus , and Streptococcus thermophiles.
  • lactic acid bacteria include Lactococcus garvieae NBRC100934, Lactococcus lactis subsp. cremoris JCM16167, Lactococcus lactis subsp. cremoris NBRC100676, Lactococcus lactis subsp. hordniae JCM1180, Lactococcus lactis subsp. hordniae JCM11040, Lactococcus lactis subsp. lactis NBRC12007, Lactococcus lactis subsp. lactis NRIC1150, Lactococcus lactis subsp. lactis JCM5805, Lactococcus lactis subsp.
  • lactis JCM20101 Leuconostoc lactis NBRC12455, Leuconostoc lactis NRIC1540 , Pediococcus damnosus JCM5886, and Streptococcus thermophilus TA-45.
  • the capacity for inducing IFN- ⁇ production of Lactococcus lactis subsp. lactis JCM5805 and that of Lactococcus lactis subsp. lactis JCM20101 are particularly high.
  • use of Lactococcus lactis JCM5805 is particularly preferable.
  • Lactic acid bacteria that can be used as the agent for inducing IFN production in the present invention preferably exert activity of IFN induction in an organism when such bacteria are ingested orally.
  • Such lactic acid bacteria are highly tolerant to the gastric or intestinal juice.
  • such lactic acid bacteria have high-level tolerance to acids and are capable of reaching the intestinal canal alive.
  • Lactococcus lactis JCM5805 described above is orally ingested, it is capable of exerting a significant degree of activity for inducing IFN production in an organism.
  • the lactic acid bacteria described above can be obtained from the RIKEN BioResource Center (3-1-1 Koyadai, Tsukuba-shi, Ibaraki, Japan), the Biological Resource Center (NBRC) at the National Institute of Technology and Evaluation (http://www.nbrc.nite.go.jp), the Culture Collection Center, Tokyo University of Agriculture (http://nodaiweb.university.jp/nric/), and DANISCO.
  • bacterial strains equivalent to the strains such as Lactococcus garvieae NBRC100934, Lactococcus lactis subsp. cremoris JCM16167, Lactococcus lactis subsp.
  • lactis JCM20101, Leuconostoc lactis NBRC12455, Leuconostoc lactis NRIC1540, Pediococcus damnosus JCM5886, or Streptococcus thermophilus TA-45 can also be used.
  • Equivalent strains include strains derived from the bacterial strains mentioned above, the bacterial strains from which the strains mentioned above are derived, or offspring strains of such bacterial strains. Equivalent strains may be conserved in other institutions for culture collection.
  • FIG. 17 shows bacterial strains derived from Lactococcus lactis JCM5805 and bacterial strains from which Lactococcus lactis JCM5805 is derived.
  • Lactococcus lactis JCM5805 Bacterial strains equivalent to Lactococcus lactis JCM5805 shown in FIG. 17 can also be used as active ingredients of the agent for inducing IFN production of the present invention.
  • Lactococcus lactis JCM5805 also refers to such equivalent strains.
  • the agent for inducing IFN production of the present invention can induce any of type I interferon (type I IFN), type II interferon (type II IFN), or type III interferon (type III IFN).
  • Type I IFN is a cytokine that is effective against virus infection, and examples thereof include IFN- ⁇ 1, IFN- ⁇ 2, IFN- ⁇ 4, IFN- ⁇ 5, IFN- ⁇ 6, IFN- ⁇ 7, IFN- ⁇ 8, IFN- ⁇ 10, IFN- ⁇ 13, IFN- ⁇ 14, IFN- ⁇ 16, IFN- ⁇ 17, IFN- ⁇ 21, and IFN- ⁇ .
  • An example of type II IFN is IFN- ⁇
  • an example of type III IFN is IFN- ⁇ .
  • the agent for inducing IFN production of the present invention has activity of inducing production of type I IFN, in particular.
  • the agent for inducing IFN production of the present invention activates plasmacytoid dendritic cells (pDCs). When a plasmacytoid dendritic cell is activated, a cell process, which is characteristic of the activated dendritic cell, appears, and type I IFN and type III IFN are produced. At this time, lactic acid bacteria, which are active ingredients of the agent for inducing IFN production of the present invention, are incorporated into pDCs.
  • the agent for inducing IFN production of the present invention has the high capacity for inducing production of type I IFN and type III IFN, and the capacity for inducing production of IFN- ⁇ , which is type I IFN, is particularly high.
  • the agent for inducing IFN production of the present invention is also capable of inducing production of type II IFN, such as IFN- ⁇ , from NK cells or Th1 cells. Immune activity of an organism is enhanced via induction of IFN production.
  • lactic acid bacteria which are active ingredients of the agent for inducing IFN production of the present invention, are capable of inducing the expression of PDL-1.
  • PDL-1 is a programmed death ligand-1 (PD-1), and PDL-1 binds to PD-1 and induces regulatory T cells, so that PDL-1 can prevent an immune system from being excessively activated and suppress the autoimmune reactions.
  • the agent for inducing IFN production of the present invention is not only capable of inducing IFN production and activating the immune functions of an organism but also capable of suppressing excessive immune reactions and maintaining the balanced immune reactions in the organism.
  • the agent for inducing IFN production of the present invention can simultaneously induce production of type I IFN and type III IFN. Specifically, production of IFN- ⁇ , IFN- ⁇ , and IFN- ⁇ can be induced simultaneously.
  • the agent for inducing IFN production of the present invention promotes expression of CD80, CD86, and MHC class II in pDCs.
  • TLR9 is involved in the induction of IFN production as a receptor.
  • the agent for inducing IFN production of the present invention contains a culture product of the lactic acid bacteria above.
  • culture product refers to live bacteria, killed bacteria, fragmented live or killed bacteria, lyophilized live or killed bacteria, or a fragmented product, culture solution, or culture extract of such lyophilized bacteria.
  • the term also refers to part of lactic acid bacteria or treated lactic acid bacteria. Examples of treated lactic acid bacteria include products resulting from enzyme or thermal treatment of lactic acid bacteria and products recovered through ethanol precipitation of the products of enzyme or thermal treatment.
  • DNA or RNA of the above lactic acid bacteria is within the scope of the culture product of lactic acid bacteria. DNA or RNA of the above lactic acid bacteria is considered to be capable of activating pDCs and inducing IFN production.
  • Lactic acid bacteria can be cultured in accordance with a conventional technique using conventional media.
  • media examples include MRS, GAM, and LM17 media, and inorganic salts, vitamins, amino acids, antibiotics, sera, or other substances may be added thereto, according to need.
  • Culture may be carried out at 25° C. to 40° C. for several hours to several days.
  • lactic acid bacteria are harvested via centrifugation, filtration, or other means.
  • bacteria may be sterilized and inactivated with the use of an autoclave, for example.
  • Activity for inducing IFN production of lactic acid bacteria that can be used as active ingredients of the agent for inducing IFN production of the present invention can be assayed by culturing candidate bacteria, culturing IFN-producing cells in the presence of the culture product thereof, and detecting an increase in the amount of IFN produced by the IFN-producing cells.
  • lyophilized bacteria are used. The weight of bacteria in the lyophilization product is adjusted to 0.1 to 5 mg/ml, and the lyophilization product is then cultured with, for example, bone marrow cells. Origins of bone marrow cells are not particularly limited, and bone marrow cells derived from humans or bone marrow cells derived from non-human animals such as mice can be used.
  • the lactic acid bacteria can be determined to be usable as active ingredients of the agent for inducing IFN production of the present invention.
  • Activation of pDCs may be detected by, for example, assaying pDC activation markers, and examples of activation markers include CD80, CD86, and MHC class II. Such activation markers can be assayed via cell staining or flow cytometry using antibodies reacting with such markers.
  • Examples of IFN include type I IFNs such as IFN- ⁇ and IFN- ⁇ , type II IFNs such as IFN- ⁇ , and type III IFN such as IFN- ⁇ .
  • type I IFN and type III IFN are preferable, type I IFN is more preferable, and IFN- ⁇ is further preferable.
  • Induction of IFN production may be assayed by determining the amount of IFN in a medium in the culture system via, for example, ELISA.
  • the present invention includes a method for screening for lactic acid bacteria having activity of inducing IFN production and usable as active ingredients of the agent for inducing IFN production of the present invention.
  • the agent for inducing IFN production of the present invention can be used in the form of a pharmaceutical product that induces IFN production and enhances immune activity of an organism.
  • the agent for inducing IFN production can be used in the form of an immunopotentiating agent or immunostimulator.
  • Such pharmaceutical products can be used for preventive or therapeutic agents for diseases, which are already known to be associated with type I IFN.
  • diseases include: cancer, including renal cancer, multiple myeloma, chronic myeloid leukemia, hairy cell leukemia, gliosarcoma, medulloblastoma, astroglioma, malignant melanoma, mycosis fungoides, and adult T cell leukemia; virus infection, including subacute sclerosing panencephalitis, HTLV-1 associated myelopathy, hepatitis B, and hepatitis C; infection with bacteria, such as chlamydia (sexually transmitted disease), Mycobacteriaceae (tuberculosis), listeriosis (ichorrhemia), Staphylococcus (food poisoning), and Helicobacter (gastritis); and autoimmune diseases including multiple sclerosis.
  • cancer including renal cancer, multiple myeloma, chronic myeloid leukemia, hairy cell leukemia, gliosarcoma, medulloblastoma, astroglioma, mal
  • the pharmaceutical product is particularly useful as a prophylactic or therapeutic agent for virus infection. Since the function of inhibiting differentiation of osteoblasts into osteoclasts is known as activity of type I IFN, it can be used as a preventive or therapeutic agent for osteoporosis.
  • An antigen associated with a particular disease may be expressed in spherical-shaped lactic acid bacteria, which is the IFN inducer of the present invention, via genetic engineering, and the resultant may be used as a vaccine. Since the cell wall of lactic acid bacteria can protect antigens from gastric acid, such bacterial strains expressing foreign antigens are particularly preferable as host organisms of oral vaccines. In general, vaccines are classified as live vaccines, inactivated whole particle vaccines, or split vaccines. However, live vaccines pose a risk of potentiating the virus virulence, inactivated whole particle vaccines may evoke side effects because of the presence of impurities, and split vaccines with the highest safety are problematic in terms of efficacy.
  • target antigens are expressed in the spherical-shaped lactic acid bacteria having the effects of IFN induction according to the present invention, effects of an adjuvant can also be achieved, and it is thus very useful.
  • Dosage forms of the agent for inducing IFN production of the present invention are not particularly limited. Examples include powder, granules, tablets, syrup, injection preparations, drops, powdered drugs, suppositories, suspensions, and ointments.
  • the pharmaceutical product of the present invention may be administered orally or parenterally through intravenous injection, intramuscular injection, subcutaneous administration, rectal administration, or transdermal administration, with oral administration being preferable.
  • the agent for inducing IFN production may contain an excipient, a disintegrator, a binder, a lubricant, a colorant, or the like. Examples of excipients include glucose, lactose, corn starch, and sorbit.
  • disintegrators include starch, sodium alginate, powdered gelatin, calcium carbonate, calcium citrate, and dextrin.
  • binders include dimethylcellulose, polyvinyl alcohol, polyvinyl ether, methylcellulose, ethylcellulose, gum Arabic, gelatin, hydroxypropyl cellulose, and polyvinyl pyrrolidone.
  • lubricants include talc, magnesium stearate, polyethylene glycol, and hydrogenated vegetable oil.
  • a dose can be adequately determined in accordance with the age, body weight, or sexuality of a patient, a type of disease, severity of symptoms, or other conditions. The agent may be administered once or several separate times per day, and a culture product may be administered in an amount equivalent to 1 ⁇ 10 9 to 1 ⁇ 10 12 cells in a single instance. Alternatively, a dose may be 1 to 1,000 mg of lactic acid bacteria.
  • the agent for inducing IFN production of the present invention can be incorporated into a food or drink product.
  • the resulting food or drink product can be used for induction of IFN production, immunopotentiation, immunostimulation, prophylaxis against virus infection, or other purposes.
  • Target food or drink products are not particularly limited, provided that active ingredients for induction of IFN production are not inhibited. Examples include milk, dairy products, beverage, seasonings, alcoholic beverage, agricultural products, processed forest products, confectioneries, breads, cereals, noodles, seafood products, processed livestock products, oils and fats, processed oils and fats, prepared frozen foods, retort foods, ready-to-eat foods, and food materials.
  • fermented dairy products such as yogurt or cheese
  • drinks containing lactic acid bacteria is particularly suitable.
  • given amounts of killed lactic acid bacteria having activity of inducing IFN production may be added to fermented food or drink products.
  • such lactic acid bacteria may be used as starters to produce fermented food or drink products.
  • a fraction containing large quantities of nucleic acids of the lactic acid bacteria of the present invention can also be used as the agent for inducing IFN production.
  • a nucleic acid may be DNA, RNA, or a mixture thereof, with DNA being preferable.
  • Such fraction can be prepared by, for example, enzyme or thermal treatment (Patent Document 4) or recovery of a precipitate obtained with the aid of ethanol (Patent Document 5).
  • such fraction is referred to as a processed product of lactic acid bacteria. With the use of such fraction, a more effective health food or drink product with enriched active ingredients can be provided.
  • Examples of the food or drink product of the present invention include a health food or drink product, a food or drink product for specified health use, a food or drink product with nutrient function claims, and a dietary supplement food or drink product.
  • the term “food or drink product for specified health use” refers to a food or drink product that is to be ingested for specified healthcare objectives and has a labeling indicating that the objectives can be expected through ingestion thereof.
  • Such food or drink product may be provided with a labeling indicating that, for example, enhancement of body's immune functions, stimulation of body's immune functions, lowering of susceptibility to colds, enhancement of tolerance to infection with viruses such as influenza virus, norovirus, or rotavirus, or cancer prevention.
  • the lactic acid bacteria shown in FIG. 1A , FIG. 1B , and FIG. 1C were subjected to thermal treatment to prepare killed lactic acid bacteria.
  • the lactic acid bacteria mentioned above were purchased from microbial strain libraries in Japan and abroad.
  • the lactic acid bacteria were obtained from the Japan Collection of Microorganisms (JCM) of the RIKEN BioResource Center, the Culture Collection Center of the Institute of Fermentation, Osaka (IFO), the NODAI Culture Collection Center (NRIC) of Tokyo University of Agriculture, American Type Culture Collection (ATCC, U.S.A.), and DANISCO.
  • JCM Japan Collection of Microorganisms
  • IFO Culture Collection Center of the Institute of Fermentation
  • NRIC NODAI Culture Collection Center
  • ATCC U.S.A.
  • DANISCO DANISCO
  • Lactic acid bacteria were subjected to stationary culture in MRS, GAM, or LM17 medium at 30° C. or 37° C. for 24 to 48 hours.
  • the strains were harvested, washed three times with sterile water, and then disinfected in an autoclave at 100° C. for 30 minutes. Thereafter, the strains were lyophilized, and the concentration was adjusted to 1 mg/ml with PBS (Takara Bio).
  • the capacity of the lactic acid bacteria prepared in Example 1 for inducing IFN- ⁇ production was evaluated with reference to pDC activation.
  • Bone marrow cells were collected from the femoral bones of C57BL/6 mice in accordance with a conventional technique, and erythrocytes were removed therefrom. Subsequently, the collected bone marrow cells were suspended to a concentration of 5 ⁇ 10 5 cells/ml in RPMI medium (SIGMA) containing 10% FCS and 2 ⁇ M ⁇ -mercaptoethanol. Flt-3L (R&D Systems) was added as a pDC inducing cytokine to a final concentration of 100 ng/ml to the resulting cell suspension, and culture was conducted in a CO 2 incubator at 37° C. in the presence of 5% CO 2 .
  • SIGMA RPMI medium
  • Flt-3L R&D Systems
  • FIG. 2 shows strains evaluated to be capable of producing 50 pg/ml or more IFN- ⁇ via ELISA.
  • activity was observed in only 13 strains (i.e., Lactococcus garvieae NBRC100934, Lactococcus lactis subsp. cremoris JCM16167 , Lactococcus lactis subsp. cremoris NBRC100676, Lactococcus lactis subsp. hordniae JCM1180, Lactococcus lactis subsp. hordniae JCM11040, Lactococcus lactis subsp.
  • lactis NBRC12007 Lactococcus lactis subsp. lactis NRIC1150, Lactococcus lactis subsp. lactis JCM5805 , Lactococcus lactis subsp. lactis JCM20101, Leuconostoc lactis NBRC12455, Leuconostoc lactis NRIC1540, Pediococcus damnosus JCM5886, and Streptococcus thermophiles TA-45).
  • There were only 3 strains i.e., Lactococcus lactis subsp. lactis NRIC1150, Lactococcus lactis subsp.
  • lactis JCM5805 and Lactococcus lactis subsp. lactis JCM20101 that had been evaluated to be capable of producing 100 pg/ml or more IFN. Most bacteria did not have the capacity for inducing IFN- ⁇ production on pDCs. This indicates that such activity is not universal among various types of lactic acid bacteria.
  • the selected 3 strains inducing the production of IFN at a high level were spherical-shaped bacteria classified as Lactococcus lactis subsp. lactis .
  • the hit rate of spherical-shaped lactic acid bacteria is 34.29%, which is significantly higher than that of rod-shaped lactic acid bacteria (i.e., 0.00%).
  • the hit rate of spherical-shaped lactic acid bacteria is 8.57%, which is also higher than that of rod-shaped lactic acid bacteria (i.e., 0.00%).
  • Lactococcus lactis JCM5805 and JCM20101 strains exhibiting the capacity for inducing IFN- ⁇ production at a particularly significant level and, as a negative control, the rod-shaped Lactobacillus rhamnosus ATCC53103 strain were subjected to the following analysis.
  • FIG. 3 shows electron micrographs of the Lactococcus lactis JCM5805 and JCM20101 strains.
  • FIG. 3A shows the JCM5805 strain
  • FIG. 3B shows the JCM20101 strain.
  • These strains were oval-shaped bacteria with approximately 1- ⁇ m major axes and 0.5- ⁇ m minor axes. Since rod-shaped bacteria generally have approximately 1- ⁇ m minor axes and 3- ⁇ m major axes, it can be said that such bacteria are very small.
  • Lactococcus lactis JCM5805 and JCM20101 strains which were found to be pDC-activating lactic acid bacteria in Example 2, and Lactobacillus rhamnosus ATCC53103 as a negative control, an experiment was carried out to confirm that activity would depend on recognition of the bacteria by pDCs; i.e., uptake of the bacteria.
  • Example 2 bone marrow cells were cultured by laying a micro glass cover slip (Matsunami Glass Ind., Ltd.). Lactococcus lactis JCM5805, Lactococcus lactis JCM20101, and Lactobacillus rhamnosus ATCC53103 labeled with FITC(SIGMA) were added thereto, and culture was conducted in a CO 2 incubator at 37° C. in the presence of 5% CO, for 3 hours. Thereafter, the micro glass cover slip was collected. The cells were stained with anti-B220-PE-Cy5.5 (eBiosciencs), allowed to adhere to the glass slides (Matsunami Glass Ind., Ltd.), and then observed under a fluorescent microscope (Olympus Corporation).
  • eBiosciencs anti-B220-PE-Cy5.5
  • FIG. 4A , FIG. 4B , and FIG. 4C show Lactococcus lactis JCM5805, the Lactococcus lactis JCM21101 strain, and Lactobacillus rhamnosus ATCC53103, respectively.
  • B220-positive red cells are pDCs.
  • uptake of the lactic acid bacteria stained green into cells is observed, although Lactobacillus rhamnosus ATCC53103 is not incorporated into the cells. Therefore, whether or not activity occurs is considered to depend on the recognition of bacteria by pDCs.
  • the capacity of the lactic acid bacteria having the capacity for inducing IFN- ⁇ production for producing cytokines of other types was examined.
  • TLRLs i.e., Pam3CSK4 (TLR2L, 1 ⁇ g/ml, InvivoGen)
  • LPS TLR4L, 5 ng/ml, SIGMA-ALDRICH
  • CpG DNA TLR9L, 0.1 ⁇ M, InvivoGen
  • the culture supernatant was subjected to ELISA assays using the IFN- ⁇ assay kit (PBL), the IFN- ⁇ assay kit (PBL), the IFN- ⁇ assay kit (BD Pharmingen), and the IL-28/IFN- ⁇ assay kit (eBiosciencs).
  • FIG. 5A to FIG. 5D show the results concerning IFN- ⁇ , IFN- ⁇ , IFN- ⁇ , and IFN- ⁇ , respectively.
  • the capacity for IFN- ⁇ production was observed in Lactococcus lactis JCM5805 and in Lactococcus lactis JCM20101, and the titer thereof was equivalent to that of CpG DNA (ODN 1585) (i.e., TLR9L, 0.1 ⁇ M).
  • Concerning IFN- ⁇ , which is also type I IFN the capacity was observed selectively in the Lactococcus lactis JCM5805 and JCM20101 strains.
  • IFN- ⁇ which is type II IFN
  • all bacterial strains exerted the capacity for inducting production, although the degree thereof varied among bacterial species.
  • IFN- ⁇ which is type III IFN
  • induction was observed selectively in Lactococcus lactis JCM5805 and in Lactococcus lactis JCM20101.
  • IFN- ⁇ induced by IFN-stimulated genes
  • ISG IFN-stimulated genes
  • IFN- ⁇ is known to potentiate its antiviral effects in coordination with IFN- ⁇ (Non-Patent Document 5). Since the Lactococcus lactis JCM5805 and JCM20101 strains are capable of inducing production of all of type I, type II, and type III IFNs, these strains are considered to have very strong antiviral activity.
  • Example 3 The cells cultured in Example 3 were stained for 30 minutes at 4° C. with the use of anti-CD11b-APC-Cy7 antibody (BD Pharmingen), anti- ⁇ 220-PerCP antibody (BD Pharmingen), and anti-CD11c-PE-Cy7 antibody (eBiosciencs) for pDC gating, anti-MHC class II-FITC antibody (eBiosciencs), anti-CD40-FITC antibody (eBiosciencs), anti-CD80-APC antibody (eBiosciencs), and anti-CD86-APC antibody (eBiosciencs) as indicators for activation, and anti-OX40L-PE antibody (eBiosciencs), anti-PDL-1-PE antibody (eBiosciencs), and anti-ICOS-L-PE antibody (eBiosciencs) as inhibitory markers.
  • the cells were washed and then analyzed with the use of FACS Canto II (BD).
  • FIG. 6A shows MHCII, CD40, CD80, and CD86 expression levels
  • FIG. 6B shows OX40L, PDL-1, and ICOS-L expression levels.
  • the median fluorescent intensity values (MFI) attained without the addition of lactic acid bacteria are shown above, and the MFI values attained with the addition of lactic acid bacteria are shown below. According to the activation markers, enhancement was observed with the addition of any lactic acid bacteria.
  • PDL-1 which was confirmed to be induced upon stimulation by the Lactococcus lactis JCM5805 and JCM20101 strains in this test, is known to bind to PD-1 of the T cell to induce a regulatory T cell.
  • Lactococcus lactis JCM5805 and JCM20101 strains are considered to be capable of maintenance of the immune system in a well-balanced state while refraining from being excessively activated through PDL-1 expression, in addition to potentiation of the immune system through IFN- ⁇ production.
  • Example 2 the Lactococcus lactis JCM5805 and JCM20101 strains were selected on the basis of activity for inducing IFN- ⁇ production.
  • myeloid dendritic cells mDCs
  • pDCs myeloid dendritic cells
  • the pDC-mDC interactions are considered to be critical in an organism. For example, conversion of pDC into mDC upon virus infection has been reported. Accordingly, the effects of the addition of lactic acid bacteria in the pDC or mDC monoculture system, the pDC/mDC mixed culture system, and the mixed culture system in which pDCs were physically separated from mDCs were examined.
  • Transwell filter (Corning) was used as a semipermeable membrane, and the amount of lactic acid bacteria added was 10 ⁇ g/ml. Culture was conducted for 2 days and the amount of IFN- ⁇ in the culture supernatant was then assayed. As a positive control, CpG DNA (ODN1585), the capacity of which for pDC activation has been known, was used at 0.1 ⁇ M. Sorted pDCs were attached to glass slides (Matsunami glass Ind., Ltd) using Cytospin (Thermo Scientific), stained with Diff-Quick (Sysmex), and then observed under a microscope (Olympus Corporation).
  • FIG. 7 The results are shown in FIG. 7 .
  • the Lactococcus lactis JCM5805 and JCM20101 strains exhibited similar responses. Specifically, IFN- ⁇ production did not take place in the mDC monoculture system, a small quantity of IFN- ⁇ was induced in the pDC monoculture system, and a significant level of IFN- ⁇ production was observed in the pDC/mDC mixed culture system.
  • FIG. 8 shows photographs showing the configurations of pDCs when Lactococcus lactis JCM5805, Lactococcus lactis JCM20101, and Lactobacillus rhamnosus ATCC53103 are added to the pDC monoculture system.
  • FIG. 8A shows the results of a control (without the addition of lactic acid bacteria), FIG.
  • FIG. 8B shows pDCs when JCM5805 was added
  • FIG. 8C shows pDCs when JCM20101 was added
  • FIG. 8D shows pDCs when ATCC53103 was added.
  • the level of IFN- ⁇ production was drastically reduced to a level equivalent to that attained in the pDC monoculture when physical contact between pDCs and mDCs was blocked with a semipermeable membrane.
  • mDCs would be necessary in order to fully induce IFN- ⁇ production via activation of pDC, although pDCs are primary targets of lactic acid bacteria.
  • the mDC/pDC cross-talk was found to be mediated by a cell-to-cell contact instead of a humoral factor.
  • TLR2-, TLR4-, TLR7-, TLR9-, and MyD88-knockout mice (8- to 10-week-old, male) and wild-type C57BL/6 mice (8- to 10-week-old, male) were purchased from Charles River Laboratories.
  • pDCs and mDCs were induced from bone marrow cells of such mice in the same manner as in Example 2, and Lactococcus lactis JCM5805, Lactococcus lactis JCM20101, and Lactobacillus rhamnosus ATCC53103 were added.
  • TLR7L ssRNA40, 5 ⁇ g/ml, InvivoGen
  • the culture supernatant was collected 48 hours later, and the amount of IFN- ⁇ produced in the culture supernatant was assayed via ELISA.
  • FIG. 9A shows the results concerning TLR2-knockout mice and TLR4-knockout mice
  • FIG. 9B shows the results concerning TLR7-knockout mice, TLR9-knockout mice, and MyD88-knockout mice.
  • WT represents the results concerning wild-type mice. No changes in the capacity for IFN- ⁇ production of the Lactococcus lactis JCM5805 and JCM20101 strains were observed in TLR2- or TLR4-knockout mice, and the involvement thereof was accordingly denied.
  • TLR9 was verified to play a key role in IFN- ⁇ production by the Lactococcus lactis JCM5805 and JCM20101 strains.
  • TLR9 was found to be a recognition receptor for the Lactococcus lactis JCM5805 and JCM20101 strains. Identification of the ligands thereof was attempted.
  • DNA represented by CpG DNA is known as a TLR9 ligand.
  • Lactococcus lactis JCM5805 , Lactococcus lactis JCM20101, and Lactobacillus rhamnosus ATCC53103 were subjected to stationary culture. The strains were harvested and then washed three times with sterile water. A solution adjusted to comprise 50 mM Tris-HCl, 5 mM EDTA, and 6.7% sucrose (PH 8.0) was added thereto. Subsequently, N-acetylmuramidase (2.5 mg/ml, Seikagaku Kogyo) and lysozyme (50 mg/ml, Seikagaku Kogyo) were added, and the resultant was allowed to stand at 37° C. for 45 minutes.
  • N-acetylmuramidase 2.5 mg/ml, Seikagaku Kogyo
  • lysozyme 50 mg/ml, Seikagaku Kogyo
  • the supernatant was removed, RNase (Qiagen) was added thereto, and the resultant was allowed to stand at 37° C. for 60 minutes. Further, 5.0 M NaCl was added thereto, phenol, chloroform, and isoamyl alcohol (Wako Pure Chemicals Industries, Ltd.,) were added thereto, and the mixture was subjected to centrifugation. The supernatant was selectively removed, ethanol in an amount twice the amount of the supernatant was added thereto, and the resultant was then subjected to centrifugation. The supernatant was removed, 70% ethanol was added to the precipitate, and centrifugation was then carried out.
  • RNase Qiagen
  • Nuclease Free Water (Qiagen) was added to the precipitate from which the supernatant had been removed.
  • DNAs of Lactococcus lactis JCM5805, Lactococcus lactis JCM20101, and Lactobacillus rhamnosus ATCC53103 prepared in the manner described above were added to the pDC/mDC culture system at 0.1 ⁇ g/ml, 1 ⁇ g/ml, and 10 ⁇ g/ml, respectively.
  • the supernatant was collected 48 hours later, and the amount of IFN- ⁇ produced in the culture supernatant was assayed via ELISA.
  • the bacterial strains were used as controls.
  • Lactococcus lactis JCM5805 , Lactococcus lactis JCM20101, and Lactobacillus rhamnosus ATCC53103 were subjected to stationary culture. The strains were harvested and then washed three times with sterile water. RNAprotect Bacteria Reagent (Qiagen) was added thereto, and the resultant was allowed to stand at 37° C. for 5 minutes, followed by centrifugation. The supernatant was removed, lysozyme (5 mg/ml, Seikagaku Kogyo) was added, and the resultant was allowed to stand at 37° C. for 10 minutes.
  • RNAprotect Bacteria Reagent Qiagen
  • RNAs were prepared from Lactococcus lactis JCM5805, Lactococcus lactis JCM20101, and Lactobacillus rhamnosus ATCC53103 through DNase treatment (Qiagen) with the use of RNeasy Mini Kit (Qiagen).
  • Total RNAs were added to the culture system according to Example 3 at 0.1 ⁇ g/ml, 1 ⁇ g/ml, and 10 ⁇ g/ml, respectively.
  • the culture supernatant was collected 48 hours later, and the amount of IFN- ⁇ produced in the culture supernatant was assayed via ELISA.
  • the bacterial strains were used as controls.
  • DNA is the active substance of activity for inducing IFN- ⁇ production and DNA of an inactive strain has activity; and (2) since lactic acid bacteria that activate pDCs and induce IFN- ⁇ represented by the Lactococcus lactis JCM5805 and JCM20101 strains are recognized by pDCs as strains, activity is detected without performing DNA extraction.
  • a strain such as Lactobacillus rhamnosus ATCC53103 is inactive because it is not recognized by pDC; and (3) RNA of lactic acid bacteria becomes active besides DNA and it functions as TLRL, although it is very rare. If the examples mentioned above are taken into consideration, RNA of Lactococcus lactis JCM20101 is the RNA ligand for TLR9, the ligand of which has been known to be DNA, discovered for the first time.
  • Lactococcus lactis JCM5805 was designated as a representative example and examined regarding the immunostimulatory effects in vivo attained via oral administration.
  • mice Three groups of C57BL/6 mice (7-week-old, female) each consisting of 5 individuals were provided: a group to which standard feeds (AlN93G, Oriental Yeast Co., Ltd.) are administered; a group to which mixed feeds containing Lactococcus lactis JCM5805 are administered; and a group to which mixed feeds containing Lactobacillus rhamnosus ATCC53103 are administered.
  • the dose of lactic acid bacteria was adjusted to 10 mg per mouse per day. Blood sampling was carried out on day 0, day 3, and day 7 (at the time of anatomy), and the amount of IFN- ⁇ produced in the blood was assayed via ELISA.
  • splenic lymphocytes and mesenteric lymph node lymphocytes are prepared, these cells are suspended in HBSS (Gibco) containing 20 mM HEPES (Gibco), and the resulting cell suspension is superposed on 10%-FCS-containing RPMI medium (Sigma) comprising Histodenz (Sigma-Aldrich) dissolved to a final concentration of 15%.
  • low-density cell fractions After centrifugation, cells in the intermediate layer (i.e., low-density cell fractions) are recovered.) The low-density cell fractions were stained with anti-CD11b-APC-Cy7 antibody (BD Pharmingen), anti-mPDCA-1-APC antibody (Milteny Biotec), and anti-CD11c-PE-Cy7 antibody (eBiosciencs) for pDC gating and with anti-MHC class II-FITC antibody (eBiosciencs) and anti-CD86-PE antibody (eBiosciencs) as indicators for activation.
  • BD Pharmingen anti-mPDCA-1-APC antibody
  • eBiosciencs anti-CD11c-PE-Cy7 antibody
  • FIG. 11 shows a summary of a method for examining the effects of Lactococcus lactis JCM5805 ingestion using healthy mice.
  • FIG. 12 shows the results of blood IFN- ⁇ assays
  • FIG. 13A to FIG. 13D show the results of pDC activation
  • FIG. 13A and FIG. 13B show changes in MHC class II levels in pDCs of the spleen and the mesenteric lymph node, respectively
  • FIG. 13C and FIG. 13D show changes in CD86 levels in pDCs of the spleen and the mesenteric lymph node, respectively.
  • the blood IFN- ⁇ level did not increase at all in the group to which Lactobacillus rhamnosus ATCC53103 had been administered as in the case of the group to which the standard feeds had been administered.
  • Lactococcus lactis JCM5805 would stimulate pDCs in vivo, as well as in vitro, and it would be capable of inducing IFN- ⁇ production.
  • the lactic acid bacteria according to the present invention would be administered to persons with the weakened immune system or elderly people, as well as healthy persons.
  • the effects of Lactococcus lactis JCM5805 ingestion were examined using immunosuppression models.
  • Two groups of C57BL/6 mice (7-week-old, female) each consisting of 5 individuals were provided: a group to which standard feeds (AlN93G, Oriental Yeast Co., Ltd.) are administered; and a group to which mixed feeds containing Lactococcus lactis JCM5805 are administered.
  • the duration for administration of Lactococcus lactis JCM5805 was two weeks, the day on which administration of Lactococcus lactis JCM5805 was initiated was designated as “day ⁇ 7,” and an immunosuppressive agent (Cyclophosphamide, Sigma-Aldrich) was administered intraperitoneally at 200 mg/kg on day 0.
  • FIG. 14 shows a summary of a method for evaluation of JCM5805 using immunosuppression models.
  • FIG. 15A shows changes in body weights of immunosuppression mouse models and FIG. 15B shows changes in blood IFN- ⁇ levels.
  • FIG. 16A shows changes in MHC class II levels in pDCs and FIG. 16B shows changes in CD86 levels in pDCs.
  • FIG. 16C shows a percentage of pDCs and FIG. 16D shows the results of flow cytometric analysis.
  • a significant decrease was observed in body weights of mice of the group to which standard feeds had been administered after the administration of Cyclophosphamide.
  • body weight loss tended to be suppressed in the group to which Lactococcus lactis JCM5805 had been administered.
  • fermented milk set yogurt, stirred yogurt
  • natural cheese were prepared.
  • the “set yogurt” is also referred to as a “firm yoghurt” or “still yogurt,” which is subjected to fermentation in a container.
  • the “stirred yogurt” is also referred to as a “fermented yogurt” or “fluid yoghurt,” which is subjected to fermentation and then filled into a container.
  • raw milk e.g., milk or skim milk powder
  • highly-branched cyclic dextrin e.g., Cluster Dextrin
  • milk peptide a general-purpose product
  • yogurt flavor T. Hasegawa Co., Ltd.
  • thermophilus and Lc. lactis JCM5805
  • the resultant was filled into a container with a lid, followed by fermentation at 32° C. for about 6 to 7 hours.
  • the acidity of lactic acid reached 0.70
  • the resultant was cooled to 10° C. and stored.
  • Lactic acid bacteria count (Lc. lactis JCM5805): 10 7 cells/g or more
  • raw milk e.g., milk or skim milk powder
  • highly-branched cyclic dextrin e.g., Cluster Dextrin
  • Nihon Shokuhin Kako Co., Ltd. e.g., milk or skim milk powder
  • milk peptide a general-purpose product
  • yogurt flavor T. Hasegawa Co., Ltd.
  • Lactic acid bacteria count (Lc. lactis JCM5805): 10 7 cells/g or more
  • raw milk e.g., milk or skim milk powder
  • highly-branched cyclic dextrin e.g., Cluster Dextrin
  • Nihon Shokuhin Kako Co., Ltd. e.g., milk or skim milk powder
  • milk peptide a general-purpose product
  • yogurt flavor T. Hasegawa Co., Ltd.
  • the raw materials were mixed to prepare a dispersion, the resulting dispersion was heated to about 70° C., and the resultant was applied to a homogenizer at a homogenization pressure (15 to 17 MPa).
  • the resultant was heat-sterilized at 125° C., the resultant was cooled to about 35° C., and lactic acid bacteria were added thereto (bacterial species: Lc. lactis JCM5805), followed by fermentation at 32° C. for about 16 hours. Fermentation was terminated at pH 4.6, the product was cooled to about 20° C., and the resultant was filled into a container with stirring, followed by refrigeration at 10° C. or lower.
  • Lactic acid bacteria count ( Lc. lactis JCM5805): 10 7 cells/g or more
  • raw milk e.g., milk or skim milk powder
  • highly-branched cyclic dextrin e.g., Cluster Dextrin
  • Nihon Shokuhin Kako Co., Ltd. e.g., milk or skim milk powder
  • milk peptide a general-purpose product
  • yogurt flavor T. Hasegawa Co., Ltd.
  • Lactic acid bacteria count (Lc. lactis JCM5805): 10 7 cells/g or more
  • raw milk i.e., milk
  • rennet Standard Plus290, Christian Hansen
  • calcium chloride a general-purpose product
  • the moisture content of the product was adjusted with pressurization by applying the weight that is about 10 times greater than that of the curds packed into the mold.
  • Lactic acid bacteria count (Lc. lactis JCM5805): 10 7 cells/g or more
  • Lactococcus lactis JCM5805 , Lactococcus lactis JCM20101, and Lactobacillus rhamnosus ATCC53103 were subjected to stationary culture.
  • the strains were harvested, washed three times with sterile water, and then suspended in PBS.
  • the lactic acid bacteria count was determined using a particle size distribution measuring device (CDA-1000X, Sysmex Corporation), the cells were added to the pDC/mDC culture system at concentrations of 1 ⁇ 10 6 , 1 ⁇ 10 7 , and 1 ⁇ 10 8 cells, and culture was conducted in a CO 2 incubator for 48 hours.
  • the culture supernatant was recovered, and the amount of IFN- ⁇ produced in the culture supernatant was assayed.
  • lactic acid bacteria capable of acting on mouse pDC were found; however, whether or not such bacteria were capable of acting on human pDCs was unknown.
  • pDCs were isolated from human PBMCs with MACS, JCM5805 was designated as a representative sample, and activity thereof on human pDCs was inspected.
  • PBMCs were purchased from LONZA.
  • human pDCs were isolated with MACS (purity: 97%). Human pDCs (5 ⁇ 10 4 cells) were cultured on a 96-well flat-bottom plate (Corning). To the isolated human pDCs, IL-3 (R&D Systems) was added at 10 ng/ml as the survival factor. In order to inspect the purity of human pDCs, human pDCs were stained with anti-CD123-FITC (AC145) antibody and anti-BDCA4-APC (AD-17F6) antibody (Miltenyi Biotec) for human pDC gating and then analyzed using the FACS Canto II (BD).
  • BD FACS Canto II
  • Lactococcus lactis JCM5805 was added to a final concentration of 10 ⁇ g/ml, and culture was conducted in a CO 2 incubator for 24 hours. Human IFN- ⁇ levels were assayed using the Human IFN- ⁇ ELISA Kit (PBL Biomedical Laboratories).
  • RNAs were extracted using the RNeasy Mini Kit (Qiagen).
  • cDNA was synthesized from 200 ng of total RNA using the iScript cDNA Synthesis Kit (Bio-Rad), and IFN- ⁇ 1, IFN- ⁇ , IFN- ⁇ 1, and GAPDH genes were amplified via PCR using the synthesized cDNA as a template.
  • PCR was carried out using TaKaRa Ex Taq (TaKaRa) and the primers described in Non-Patent Document 7.
  • IFN- ⁇ 1, IFN- ⁇ , IFN- ⁇ 1, and GAPDH genes were subjected to the reaction at 94° C. for 1 minute, and a cycle of 94° C.
  • the PCR reaction solution was electrophoresed in accordance with a general technique, and development of amplified fragments and the density thereof were inspected.
  • FIG. 19 The results are shown in FIG. 19 .
  • FIG. 19A , FIG. 19B , and FIG. 19C show the purity of human pDCs isolated with MACS, the amount of IFN- ⁇ production detected at the protein level by ELISA, and IFN- ⁇ 1, IFN- ⁇ , IFN- ⁇ , and GAPDH gene expression levels detected by RT-PCR.
  • Lactococcus lactis JCM5805 With the addition of Lactococcus lactis JCM5805, induction of IFN- ⁇ production was observed at the protein level. Also, induction of IFN- ⁇ 1, IFN- ⁇ , and IFN- ⁇ gene expression was detected.
  • the above results demonstrate that Lactococcus lactis JCM5805 would also activate human pDCs.
  • Test product yogurt drink containing Lactococcus lactis JCM5805
  • Placebo product yogurt-like drink containing no lactic acid bacteria
  • This test was carried out aimed at examination of the influence of lactic acid bacteria on blood biomarkers associated with antiviral activity and on subjective evaluation by questionnaires concerning physical conditions of healthy, working, adult males and females who had continuously ingested yogurt drinks containing Lactococcus lactis JCM5805s for about 4 weeks, in comparison with the control experiment conducted with the use of yogurt-like drinks containing no lactic acid bacteria as placebos.
  • Test subjects were those who had no serious chronic disease, milk allergy, or other conditions, those who were evaluated to have no problem by a particular virus test, those who were capable of restricting intake of yogurt and cheese during the period of test product ingestion, and those who were not on steroid medications (internal or external use).
  • a daily dose was about 1 ⁇ 10 11 cfu of Lactococcus lactis JCM5805.
  • Test subjects were asked to drink a bottle of the test product (100 ml) before or after meal in the morning every day.
  • test product ingestion was about 4 weeks. Blood sampling was carried out three times: at a pre-test for grouping (1 month before the initiation of ingestion); at week 0 (the day before the initiation of ingestion); and at week 4 (the day after the termination of ingestion). Test subjects answered the questionnaires concerning physical conditions every day during the ingestion period.
  • PBMCs peripheral blood mononuclear cells
  • Blood biomarkers were analyzed by isolating PBMCs from the blood samples obtained on week 0 and week 4 and examining the isolated PBMCs.
  • PBMCs (1 ⁇ 10 6 cells) were stained with anti-CD123-FITC (AC145) (Miltenyi Biotec), anti-BDCA4-APC (AD-17F6) (Miltenyi Biotec), anti-CD86-PE (B7.2) (eBioscience), and anti-HLA-DR-PerCP (L243) (BD Biosciences) in accordance with a conventional technique.
  • HLA-DR MHC class II
  • CD86 fluorescent intensities of the cell populations detected in CD123 + /BDCA4 + were assayed with the use of FACS Canto II (BD), and the determined values were employed as the indicators for pDC activation.
  • IFN- ⁇ gene expression in the blood was detected by extracting total RNAs from 1 ⁇ 10 6 PBMCs using the RNeasy Mini Kit (Qiagen).
  • cDNA was synthesized from 100 ng of total RNA using the iScript cDNA Synthesis Kit (Bio-Rad), and the IFN- ⁇ 1 gene (the GAPDH gene as a reference) was analyzed via real-time PCR using the synthesized cDNA as a template.
  • Real-time PCR analysis was carried out using SYBR Premix Ex Taq (TaKaRa) and the primers described in Non-Patent Document 7. In accordance with general protocols, the samples were subjected to the reaction at 95° C. for 10 seconds, followed by a cycle of 95° C. for 10 seconds, 49° C. for 5 seconds, and 72° C. for 10 seconds repeated 50 times.
  • MHC class II When Streptococcus pyogenes or influenza virus was allowed to act on human pDCs in vitro, the MHC class II expression level was elevated with a good response, although no significant increase was observed in the CD86 expression level (Non-Patent Document 8). Accordingly, MHC class II was designated as a major activation marker, and 36 samples exhibiting values of average MHC class II activity ⁇ 2SD (18 samples from each group) were subjected to analyses of all biomarkers.
  • the samples were divided into those exhibiting values higher than the average MHC class II activity assayed at week 0 (hereafter referred to as “higher pDC activity”) and those exhibiting values lower than such average (hereafter referred to as “lower pDC activity”), and these samples were separately analyzed.
  • FIG. 20A and FIG. 20D show changes in MHC class II and CD86 activity in pDCs observed in the activity assays from week 0 to week 4. Changes in MHC class II and CD86 activity of the group that had ingested yogurt drinks containing Lactococcus lactis JCM5805 (hereafter, referred to as “the JCM5805 group”) were significantly higher than those of the group that had ingested yogurt-like drink containing no lactic acid bacteria (hereafter, referred to as “the placebo group”).
  • FIG. 20B and FIG. 20C The results of analyses separately conducted for the samples exhibiting higher pDC activity and for the samples exhibiting lower pDC activity and the changes in MHC class II activity are shown in FIG. 20B and FIG. 20C , respectively.
  • the results as mentioned above and the changes in CD86 activity are shown in FIG. 20E and FIG. 20F , respectively.
  • changes in MHC class II activity there was no significant difference in samples exhibiting higher pDC activity between the JCM5805 group and the placebo group. In the case of samples exhibiting lower pDC activity, however, such changes of the JCM5805 group were significantly higher than those of the placebo group.
  • changes in CD86 activity no significant difference was observed between the JCM5805 group and the placebo group, regardless of pDC activity levels.
  • FIG. 21 shows the results of analysis of IFN- ⁇ gene expression in the blood at lower pDC activity.
  • no significant changes were observed from week 0 to week 4 in the placebo group; however, a significant increase was observed in expression levels from week 0 to week 4 in the JCM5805 group.
  • no significant changes were observed from week 0 to week 4 in the placebo group and in the JCM5805 group (data not shown).
  • the results demonstrate that the amount of IFN- ⁇ gene transcription in the human blood is increased by ingestion of Lactococcus lactis JCM5805.
  • FIG. 22 shows the results of assays for the capacity for IFN- ⁇ production attained when the blood pDCs exhibiting lower pDC activity are stimulated with CpG DNA.
  • CpG DNA is a nucleic acid ligand targeting TLR9, and the virus recognition mechanism of pDC detects viral DNA or RNA by means of TLR9 or TLR7/8.
  • the virus recognition mechanism was wrongly stimulated by addition of a nucleic acid ligand (i.e., CpG DNA).
  • CpG DNA a nucleic acid ligand
  • the results of the experiments indicate that pDC activation induced by CpG DNA stimulation is potentiated in the JCM5805 group and it leads to an enhanced response at the time of virus infection.
  • FIG. 23 shows the results of the questionnaires concerning physical conditions.
  • the total number of days during which cold symptoms had developed and the total number of days during which cold symptoms did not develop were determined in every week, and the outcomes were subjected to the square test.
  • the total number of days during which the subjects in the JCM5805 group had developed cold symptoms was found to be significantly fewer, and the total number of days during which the subjects did not develop cold symptoms was found to be larger on week 4, compared with the placebo group. This indicates that continuous ingestion of Lactococcus lactis JCM5805 for 4 weeks leads the subjects to be less susceptible to colds.
  • Lactic acid bacteria capable of activating pDCs and inducing IFN production can be used for an immunostimulatory pharmaceutical product or food or drink product as the agent for inducing IFN production.

Landscapes

  • Life Sciences & Earth Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Microbiology (AREA)
  • General Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Molecular Biology (AREA)
  • Biotechnology (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Food Science & Technology (AREA)
  • Mycology (AREA)
  • Polymers & Plastics (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Biomedical Technology (AREA)
  • Biochemistry (AREA)
  • Immunology (AREA)
  • General Engineering & Computer Science (AREA)
  • Epidemiology (AREA)
  • Virology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • Nutrition Science (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Urology & Nephrology (AREA)
  • Biophysics (AREA)
  • Cell Biology (AREA)
  • Hematology (AREA)
  • General Physics & Mathematics (AREA)
US13/977,435 2010-12-28 2011-12-28 Agent for inducing interferon production containing lactic acid bacteria Abandoned US20130302380A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US15/367,649 US10220060B2 (en) 2010-12-28 2016-12-02 Agent for inducing interferon production containing lactic acid bacteria

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
JP2010293810 2010-12-28
JP2010-293810 2010-12-28
PCT/JP2011/080359 WO2012091081A1 (fr) 2010-12-28 2011-12-28 Agent induisant la production d'interférons contenant des bactéries à acide lactique

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2011/080359 A-371-Of-International WO2012091081A1 (fr) 2010-12-28 2011-12-28 Agent induisant la production d'interférons contenant des bactéries à acide lactique

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US14/263,306 Continuation US9549956B2 (en) 2010-12-28 2014-04-28 Agent for inducing interferon production containing lactic acid bacteria

Publications (1)

Publication Number Publication Date
US20130302380A1 true US20130302380A1 (en) 2013-11-14

Family

ID=46383169

Family Applications (5)

Application Number Title Priority Date Filing Date
US13/977,435 Abandoned US20130302380A1 (en) 2010-12-28 2011-12-28 Agent for inducing interferon production containing lactic acid bacteria
US14/263,306 Active US9549956B2 (en) 2010-12-28 2014-04-28 Agent for inducing interferon production containing lactic acid bacteria
US15/367,649 Active US10220060B2 (en) 2010-12-28 2016-12-02 Agent for inducing interferon production containing lactic acid bacteria
US16/290,581 Active 2032-02-25 US11224623B2 (en) 2010-12-28 2019-03-01 Agent for inducing interferon production containing lactic acid bacteria
US17/548,904 Active 2032-01-18 US11944657B2 (en) 2010-12-28 2021-12-13 Agent for inducing interferon production containing lactic acid bacteria

Family Applications After (4)

Application Number Title Priority Date Filing Date
US14/263,306 Active US9549956B2 (en) 2010-12-28 2014-04-28 Agent for inducing interferon production containing lactic acid bacteria
US15/367,649 Active US10220060B2 (en) 2010-12-28 2016-12-02 Agent for inducing interferon production containing lactic acid bacteria
US16/290,581 Active 2032-02-25 US11224623B2 (en) 2010-12-28 2019-03-01 Agent for inducing interferon production containing lactic acid bacteria
US17/548,904 Active 2032-01-18 US11944657B2 (en) 2010-12-28 2021-12-13 Agent for inducing interferon production containing lactic acid bacteria

Country Status (8)

Country Link
US (5) US20130302380A1 (fr)
EP (1) EP2659898B1 (fr)
JP (3) JP5950827B2 (fr)
AU (1) AU2011350554B2 (fr)
BR (1) BR112013016690A2 (fr)
ES (1) ES2668096T3 (fr)
NZ (1) NZ613335A (fr)
WO (1) WO2012091081A1 (fr)

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10525088B2 (en) 2014-05-30 2020-01-07 Kirin Holdings Kabushiki Kaisha Lactic acid bacterial immunopotentiating activity-increasing composition and method for increasing immunopotentiating activity of lactic acid bacteria
US10603377B2 (en) * 2015-10-26 2020-03-31 Kirin Holdings Kabushiki Kaisha Method for enhancing generation of antigen-specific cytotoxic T lymphocytes (CTL) and antibodies
CN111655272A (zh) * 2018-01-09 2020-09-11 中天生物科技股份有限公司 活化肿瘤浸润淋巴细胞(TILs)的方法
US10774306B2 (en) 2015-11-10 2020-09-15 Kirin Holdings Kabushiki Kaisha Method for increasing immunopotentiating activity of lactic acid bacteria
US10973908B1 (en) 2020-05-14 2021-04-13 David Gordon Bermudes Expression of SARS-CoV-2 spike protein receptor binding domain in attenuated salmonella as a vaccine
US20210128650A1 (en) * 2013-08-12 2021-05-06 University Of Guelph Antiviral methods and compositions comprising probiotic bacterial molecules
US11103543B2 (en) 2017-05-31 2021-08-31 Kirin Holdings Kabushiki Kaisha Composition for recovering from fatigue and/or preventing fatigue accumulation
US11389494B2 (en) 2016-09-09 2022-07-19 Kirin Holdings Kabushiki Kaisha Composition for suppressing or improving eye fatigue

Families Citing this family (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP6455920B2 (ja) * 2014-10-31 2019-01-23 キリン株式会社 微生物又はdnaの免疫賦活活性を予測する方法
US20190099456A1 (en) * 2016-04-04 2019-04-04 Kikkoman Corporation Composition for promoting interferon lambda production and production method therefor
JP6846376B2 (ja) * 2018-03-15 2021-03-24 キリンホールディングス株式会社 皮膚状態改善用組成物
JP2019167327A (ja) * 2018-03-26 2019-10-03 森永乳業株式会社 ビフィドバクテリウム属細菌を有効成分とする、iii型インターフェロン産生促進用組成物
CN108410777A (zh) * 2018-05-08 2018-08-17 西藏高原之宝牦牛乳业股份有限公司 一种嗜热链球菌及其制备方法
CN110468061B (zh) * 2018-05-11 2024-03-08 韩国亿诺生物有限公司 具有预防或治疗癌症的效果的新型菌株
JP6796299B2 (ja) * 2018-05-23 2020-12-09 キッコーマン株式会社 免疫賦活用組成物及びサイトカイン産生促進用組成物
CN112543641A (zh) * 2018-08-09 2021-03-23 龟甲万株式会社 FcεRI的表达抑制用组合物以及FcεRI的表达抑制方法
JP7316802B2 (ja) * 2019-02-19 2023-07-28 雪印メグミルク株式会社 糖吸収促進用組成物
KR102154254B1 (ko) * 2020-04-29 2020-09-09 (주)녹십자웰빙 우수한 면역기능 증진 효과를 갖는 신규한 유산균 및 이를 포함하는 식품조성물, 건강기능성 식품 조성물 및 프로바이오틱스
US20240066076A1 (en) 2021-03-11 2024-02-29 Miyarisan Pharmaceutical Co., Ltd. Interferon production promoter
US20240191177A1 (en) * 2021-04-28 2024-06-13 Kirin Holdings Kabushiki Kaisha Growth promoter for lactic acid cocci
KR102404016B1 (ko) * 2021-06-10 2022-06-02 한국식품연구원 락토코커스 락티스 아종 호르드니아에 균주 및 이의 퇴행성 뇌질환에 대한 예방, 개선 또는 치료 용도
AU2022375406A1 (en) * 2021-10-29 2024-05-16 Japan As Represented By Director General Of National Institute Of Infectious Diseases AGENT FOR INHIBITING PROLIFERATION OF SARS-CoV-2
WO2024117116A1 (fr) * 2022-11-29 2024-06-06 キリンホールディングス株式会社 Boisson sans sucre conditionnée

Family Cites Families (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH0755908B2 (ja) * 1992-11-24 1995-06-14 財団法人京都パストゥール研究所 免疫機能助長剤
JP2000262247A (ja) 1999-03-19 2000-09-26 Asama Kasei Kk 免疫調節剤、免疫調節食品および免疫調節飼料
JP2001046020A (ja) 1999-08-05 2001-02-20 Nichinichi Seiyaku Kk 花粉抗原による鼻および目症状を改善または予防する食品
JP4712289B2 (ja) * 2003-08-26 2011-06-29 株式会社エイ・エル・エイ 免疫促進用組成物
JP4589618B2 (ja) * 2003-11-28 2010-12-01 独立行政法人農業生物資源研究所 免疫調節性機能を誘導する乳酸菌類とその成分
JP5177728B2 (ja) * 2003-12-12 2013-04-10 株式会社明治 Nk細胞活性化剤
JP2006028047A (ja) 2004-07-14 2006-02-02 Kikkoman Corp インターロイキン12産生促進剤およびその製造法
JP2007117031A (ja) * 2005-10-31 2007-05-17 Nakagaki Gijutsushi Jimusho:Kk インターフェロンガンマ発現誘導作用を有する発酵乳、及び発酵乳製造用種菌
US8338161B2 (en) * 2007-02-13 2012-12-25 Morinaga Milk Industry Co., Ltd. Lactic acid bacteria
US20110256611A1 (en) 2007-04-03 2011-10-20 Tadaomi Kawashima Interferon beta production promoter and a method for producing thereof
JP2009005124A (ja) 2007-06-22 2009-01-08 Funai Electric Co Ltd デジタル放送記録装置
JP2009005123A (ja) 2007-06-22 2009-01-08 Panasonic Corp テレビジョン放送受信装置、およびテレビジョン放送受信方法
WO2009005124A1 (fr) 2007-07-04 2009-01-08 Kikkoman Corporation Arn double brin provenant d'une bactérie d'acide lactique
FI20075539L (fi) * 2007-07-12 2009-01-13 Valio Oy Maitohappobakteerit, joilla on proinflammatorisia ominaisuuksia
JP5247120B2 (ja) * 2007-11-02 2013-07-24 雪印メグミルク株式会社 L−オルニチン含有物の製造方法
JP4802216B2 (ja) * 2008-06-11 2011-10-26 森永乳業株式会社 ビフィドバクテリウム属菌含有組成物及びビフィドバクテリウム属菌含有組成物の製造方法
ES2409854T3 (es) 2008-06-11 2013-06-28 Morinaga Milk Industry Co., Ltd. Procedimiento para producir leche fermentada
JP2009296972A (ja) * 2008-06-16 2009-12-24 Snow Brand Milk Prod Co Ltd 酵素処理チーズ及びその製造方法
EP2206505B1 (fr) 2008-06-26 2013-07-31 Shinwa Pharmaceutical Co., Ltd. Bactérie d'acide lactique à l'échelle nano
FI20096058A0 (fi) * 2009-10-13 2009-10-13 Valio Oy Koostumuksia ja niihin liittyviä menetelmiä ja käyttöjä

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20210128650A1 (en) * 2013-08-12 2021-05-06 University Of Guelph Antiviral methods and compositions comprising probiotic bacterial molecules
US11857581B2 (en) * 2013-08-12 2024-01-02 Microsintesis Inc. Antiviral methods and compositions comprising probiotic bacterial molecules
US10525088B2 (en) 2014-05-30 2020-01-07 Kirin Holdings Kabushiki Kaisha Lactic acid bacterial immunopotentiating activity-increasing composition and method for increasing immunopotentiating activity of lactic acid bacteria
US10603377B2 (en) * 2015-10-26 2020-03-31 Kirin Holdings Kabushiki Kaisha Method for enhancing generation of antigen-specific cytotoxic T lymphocytes (CTL) and antibodies
US10774306B2 (en) 2015-11-10 2020-09-15 Kirin Holdings Kabushiki Kaisha Method for increasing immunopotentiating activity of lactic acid bacteria
US11389494B2 (en) 2016-09-09 2022-07-19 Kirin Holdings Kabushiki Kaisha Composition for suppressing or improving eye fatigue
US11890309B2 (en) 2016-09-09 2024-02-06 Kirin Holdings Kabushiki Kaisha Composition for suppressing or improving eye fatigue
US11103543B2 (en) 2017-05-31 2021-08-31 Kirin Holdings Kabushiki Kaisha Composition for recovering from fatigue and/or preventing fatigue accumulation
CN111655272A (zh) * 2018-01-09 2020-09-11 中天生物科技股份有限公司 活化肿瘤浸润淋巴细胞(TILs)的方法
EP3737395A4 (fr) * 2018-01-09 2021-09-29 Microbio Co., Ltd. Procédé d'activation de lymphocytes infiltrant les tumeurs (til)
US10973908B1 (en) 2020-05-14 2021-04-13 David Gordon Bermudes Expression of SARS-CoV-2 spike protein receptor binding domain in attenuated salmonella as a vaccine
US11406702B1 (en) 2020-05-14 2022-08-09 David Gordon Bermudes Expression of SARS-CoV-2 spike protein receptor binding domain in attenuated Salmonella as a vaccine

Also Published As

Publication number Publication date
AU2011350554A1 (en) 2013-07-25
US9549956B2 (en) 2017-01-24
US20190192584A1 (en) 2019-06-27
US10220060B2 (en) 2019-03-05
US20220105140A1 (en) 2022-04-07
US11944657B2 (en) 2024-04-02
JP6598824B2 (ja) 2019-10-30
US20170106028A1 (en) 2017-04-20
WO2012091081A1 (fr) 2012-07-05
JP2016073314A (ja) 2016-05-12
NZ613335A (en) 2015-05-29
JP6170190B2 (ja) 2017-07-26
JP5950827B2 (ja) 2016-07-13
ES2668096T3 (es) 2018-05-16
EP2659898A4 (fr) 2014-06-11
AU2011350554B2 (en) 2015-10-01
EP2659898B1 (fr) 2018-03-07
EP2659898A1 (fr) 2013-11-06
US20140234379A1 (en) 2014-08-21
JP2017201984A (ja) 2017-11-16
JPWO2012091081A1 (ja) 2014-06-05
AU2011350554A8 (en) 2013-09-05
US11224623B2 (en) 2022-01-18
WO2012091081A8 (fr) 2012-08-30
BR112013016690A2 (pt) 2016-10-04

Similar Documents

Publication Publication Date Title
US11944657B2 (en) Agent for inducing interferon production containing lactic acid bacteria
Yang et al. Oral administration of live Bifidobacterium substrains isolated from healthy centenarians enhanced immune function in BALB/c mice
Owen et al. Microbial activation of gut dendritic cells and the control of mucosal immunity
Fujie et al. Toll-like receptor-2-activating bifidobacteria strains differentially regulate inflammatory cytokines in the porcine intestinal epithelial cell culture system: finding new anti-inflammatory immunobiotics
JP2021518414A (ja) 細菌株を含む組成物
US11260085B2 (en) Salt-tolerant lactobacillus, method of culturing salt-tolerant lactobacillus, and immunostimulant
Yoshida et al. Lactobacillus gasseri OLL2809 and its RNA suppress proliferation of CD4+ T cells through a MyD88‐dependent signalling pathway
JP5690270B2 (ja) 乳酸菌由来のrnaを有効成分とする組成物
Ma et al. Live Mycobacterium leprae inhibits autophagy and apoptosis of infected macrophages and prevents engulfment of host cell by phagocytes
JP7358001B2 (ja) 乳酸菌、インターロイキン-22産生誘導剤、皮膚バリア機能増強剤
AU2015268629B2 (en) Interferon production-inducing agent containing lactic acid bacteria
Mattiola et al. Regulation of innate immune system function by the microbiome: consequences for tumor immunity and cancer immunotherapy
WO2022011271A2 (fr) Probiotiques destinés à être utilisés dans la prévention ou le traitement de maladies et/ou de symptômes associés à des coronavirus
Rolny et al. Lactobacillus delbrueckii subsp lactis CIDCA 133 modulates response of human epithelial and dendritic cells infected with Bacillus cereus.
JP2016113378A (ja) 濾胞性ヘルパーt細胞増加剤
JP7358002B2 (ja) バチルス属細菌、インターロイキン-22産生誘導剤、皮膚バリア機能増強剤
JP2018188436A (ja) 濾胞性制御性t細胞増加剤
Kandarina et al. Goat Milk Yoghurt by Using Lacto-B Culture Modulates the Production of Tumor Necrosis Factor-Alpha and Interleukin-10 in Malnourished Rats
Piraine et al. The beneficial role of Candida intermedia and Saccharomyces boulardii yeasts on the immune response of mice vaccinated with a SARS-CoV-2 experimental vaccine
KR20240006134A (ko) 마이코박테리움 파라고르도네를 포함하는 면역 증강용 조성물
Elawadli Investigations into the Effects of Lactobacilli on Murine Dendritic Cells
Tóth et al. THESIS FOR THE DEGREE OF DOCTOR OF PHILOSOPHY (PHD)

Legal Events

Date Code Title Description
AS Assignment

Owner name: KIRIN HOLDINGS KABUSHIKI KAISHA, JAPAN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:FUJIWARA, DAISUKE;JONAI, KENTA;SUGIMURA, TETSU;REEL/FRAME:031337/0027

Effective date: 20130501

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION