US20220080003A1 - Lactic acid bacterial strains - Google Patents

Lactic acid bacterial strains Download PDF

Info

Publication number
US20220080003A1
US20220080003A1 US17/232,871 US202117232871A US2022080003A1 US 20220080003 A1 US20220080003 A1 US 20220080003A1 US 202117232871 A US202117232871 A US 202117232871A US 2022080003 A1 US2022080003 A1 US 2022080003A1
Authority
US
United States
Prior art keywords
lactobacillus
pharmaceutical composition
lab
lactic acid
strains
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
US17/232,871
Inventor
Denise Kelly
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.)
4D Pharma Research Ltd
Armistice Capital Master Fund Ltd
Original Assignee
4D Pharma Research 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 4D Pharma Research Ltd filed Critical 4D Pharma Research Ltd
Priority to US17/232,871 priority Critical patent/US20220080003A1/en
Assigned to 4D PHARMA RESEARCH LIMITED reassignment 4D PHARMA RESEARCH LIMITED CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: GT BIOLOGICS LIMITED
Assigned to GT BIOLOGICS LIMITED reassignment GT BIOLOGICS LIMITED ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: KELLY, DENISE
Assigned to OXFORD FINANCE LUXEMBOURG S.A R.L. reassignment OXFORD FINANCE LUXEMBOURG S.A R.L. INTELECTUAL PROPERTY SECURITY AGREEMENT Assignors: 4D Pharma Cork Limited, 4D PHARMA DELAWARE INC., 4D PHARMA PLC, 4D PHARMA RESEARCH LIMITED
Publication of US20220080003A1 publication Critical patent/US20220080003A1/en
Assigned to ARMISTICE CAPITAL MASTER FUND LTD. reassignment ARMISTICE CAPITAL MASTER FUND LTD. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: OXFORD FINANCE LUXEMBOURG S.A R.L.
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
    • 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
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23KFODDER
    • A23K10/00Animal feeding-stuffs
    • A23K10/10Animal feeding-stuffs obtained by microbiological or biochemical processes
    • A23K10/16Addition of microorganisms or extracts thereof, e.g. single-cell proteins, to feeding-stuff compositions
    • A23K10/18Addition of microorganisms or extracts thereof, e.g. single-cell proteins, to feeding-stuff compositions of live microorganisms
    • 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/10Laxatives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/12Antidiarrhoeals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/14Prodigestives, e.g. acids, enzymes, appetite stimulants, antidyspeptics, tonics, antiflatulents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • 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
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23VINDEXING SCHEME RELATING TO FOODS, FOODSTUFFS OR NON-ALCOHOLIC BEVERAGES AND LACTIC OR PROPIONIC ACID BACTERIA USED IN FOODSTUFFS OR FOOD PREPARATION
    • A23V2002/00Food compositions, function of food ingredients or processes for food or foodstuffs
    • 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
    • C12R2001/225Lactobacillus
    • 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
    • C12R2001/225Lactobacillus
    • C12R2001/23Lactobacillus acidophilus
    • 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
    • C12R2001/225Lactobacillus
    • C12R2001/25Lactobacillus plantarum
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the present invention relates to bacterial strains isolated from pigs. More specifically, the invention relates to the isolation of lactic acid bacteria from organically-reared pigs.
  • the claimed lactic acid bacteria have useful probiotic and therapeutic applications.
  • the composition of the microbial flora of pigs, their gut innate immune function and possible susceptibility to infection is greatly influenced by the environment in which they were reared during early life (Mulder et al, 2009). Outdoor-reared pigs generally have a more developed gut immune system, perform better and are healthier than indoor-reared counterparts. The outdoor environment dramatically influences microbial diversity of the gut and is associated with high levels of Firmicutes, in particular Lactic Acid Bacteria [LAB].
  • LAB comprise a clade of gram-positive, low-GC, acid-tolerant, generally non-sporulating, non-respiring bacteria that are associated with certain common metabolic and physiological characteristics.
  • LAB are rod-shaped bacilli or coccus that are characterized by an increased tolerance to a lower pH range.
  • LAB produce lactic acid as the major metabolic end-product of carbohydrate fermentation and are amongst the most important groups of microorganisms used in the food industry.
  • Lactobacilli are predominant in the gut flora of organically (outdoor) reared pigs. In contrast, the numbers of these bacteria are low in indoor-reared pigs and levels of potentially pathogenic phylotypes are high (Mulder et al, 2009). Furthermore, gut immune development and function of indoor-reared pigs is known to deviate from normal. In particular, expression of Type 1 interferon genes, Major Histocompatibility Complex class I and several chemokines are known to be increased (Mulder et al, 2009).
  • Lactic acid bacteria may modify the flora and gut structure and function in several ways (Cotter et al, 2005; Ohashi and Ushida, 2009). For example, they may compete with harmful bacteria for key nutrients or attachment sites on the gut, resulting in their exclusion. Alternatively, they can produce bioactive substances that aid or promote colonisation by beneficial bacteria or kill/interfere with the growth of potentially harmful or pathogenic bacteria. Alternatively, these bioactive factors can be immune-modulators that promote immune development and barrier integrity of the gut. Strains of LAB vary greatly in their biological activity. The present invention seeks to provide LAB strains that have therapeutically useful properties. More specifically, the invention seeks to provide LAB strains that are capable of promoting gut and immune development and health, thereby having considerable therapeutic potential as probiotics.
  • microbiota of out-door reared pigs contain LAB strains that produce potent and specific anti-microbial or cell-/immune-modulating bioactive factors.
  • a first aspect of the invention relates to a porcine lactic acid bacterial strain, wherein said bacterial strain is characterised by one or more of the following characteristics:
  • a second aspect relates to a composition
  • a composition comprising one or more lactic acid bacterial strains according to the invention and a pharmaceutically acceptable excipient, carrier or diluent.
  • a third aspect relates to a probiotic composition
  • a probiotic composition comprising one or more lactic acid bacterial strains according to the invention.
  • a fourth aspect relates to one or more lactic acid bacterial strains according to the invention for use in medicine.
  • a fifth aspect relates to one or more lactic acid bacterial strains according to the invention for use in treating an intestinal disorder in a subject.
  • a sixth aspect relates to the use of one or more lactic acid bacterial strains according to the invention in the preparation of a medicament for treating an intestinal disorder in a subject.
  • a seventh aspect relates to a method of treating an intestinal disorder in a subject, said method comprising administering to the subject a pharmaceutically effective amount of one or more lactic acid bacterial strains or composition according to the invention.
  • An eighth aspect of the invention relates to one or more lactic acid bacterial strains according to the invention for improving intestinal microbiota.
  • a ninth aspect of the invention relates to a method of improving intestinal microbiota in a subject, said method comprising administering to the subject one or more lactic acid bacterial strains or composition according to the invention.
  • a tenth aspect relates to a feedstuff comprising one or more lactic acid bacterial strains according to the invention.
  • An eleventh aspect relates to a food product comprising one or more lactic acid bacterial strains according to the invention.
  • a twelfth aspect relates to a dietary supplement comprising one or more lactic acid bacterial strains according to the invention.
  • a thirteenth aspect relates to a food additive comprising one or more lactic acid bacterial strains according to the invention.
  • a fourteenth aspect relates to a process for producing a probiotic, said process comprising culturing a lactic acid bacterial strain according to the invention.
  • a fifteenth aspect of the invention relates to a process for obtaining a porcine lactic acid bacterial strain, said process comprising obtaining faeces from an organically reared pig and extracting one or more porcine lactic acid bacterial strains from said faeces.
  • a sixteenth aspect of the invention relates to one or more porcine lactic acid bacterial strains obtained by, or obtainable by, the process described above.
  • the present invention relates to one or more porcine lactic acid bacterial strains.
  • the lactic acid bacterial strain is characterised by one or more of the following characteristics:
  • the term “porcine” means “of or pertaining to swine”, i.e. of or pertaining to any of several mammals of the family Suidae, especially the domesticated hog, Sus scrofa domesticus , or Sus domesticus when young or of comparatively small size.
  • the pig is less than 3 months old, preferably, less than 2 months old.
  • the porcine lactic acid bacterial strain is from an organically reared pig.
  • the pigs are reared free range, outside (with exposure to soil) and in the absence of antibiotics, growth promoters and/or growth enhancers.
  • the porcine lactic acid bacterial strain is from an outdoor reared pig.
  • the pigs are reared outside for at least 60% of their lives. More preferably, the animals are reared outside for at least 80% of their lives, more preferably, at least 90% of their lives, even more preferably still, 100% of their lives.
  • the lactic acid bacterial strain is selected from L. johnsonii, L. reuteri, L. plantarum, L. gasseri, L. pentosus, L. acidophilus, L. vaginalis and L. mucosae.
  • the lactic acid bacterial strain is selected from L. johnsonii, L. reuteri and L. plantarum.
  • the lactic acid bacterial strain is in the form of a live bacterial population, a lyophilized bacterial population, a non-viable bacterial preparation, or the cellular components thereof.
  • the bacterial strain is in the form of a non-viable bacterial preparation, it is selected from heat-killed bacteria, irradiated bacteria and lysed bacteria.
  • the lactic acid bacterial strain is in the form of a live bacterium, a dead bacterium, or the cellular components thereof.
  • the lactic acid bacterial strain is in isolated form.
  • isolated means isolated from its native environment.
  • the lactic acid bacterial strain is in biologically pure form.
  • biologically pure refers to a bacterial strain in the form of a laboratory culture that is substantially free from other species of organism.
  • the lactic acid bacterial strain is in the form of a culture of a single species of organism.
  • lactic acid bacterial strain also encompasses mutants of said lactic acid bacterial strain.
  • mutant includes derived bacterial strains having at least 93% homology, preferably at least 96% homology, more preferably 98% homology to the polynucleotide sequence of a referenced strain, but otherwise comprising mutations in other sequences in the bacterial genome. Mutants are obtainable by genetic engineering techniques inferring alteration of the genetic material of the strains of the invention or inferring a recombination of the genetic material of the strains of the invention with other molecules. Typically, in order to obtain such mutant strains, a person skilled in the art can use standard mutagenesis techniques such as UV radiation or exposure to mutagenic chemical products.
  • mutants includes natural or induced mutations comprising at least single base alterations including deletions, insertions, transversions, and other modifications known to those skilled in the art, including genetic modification introduced into a parent nucleotide or amino acid sequence whilst maintaining at least 50% homology to the parent sequence.
  • sequence comprising the mutation or mutations has at least 60%, more preferably at least 75%, more preferably still 85% homology with the parental sequence.
  • sequence “homology” can be determined using standard techniques known to those skilled in the art. For example, homology may be determined using the on-line homology algorithm “BLAST” program, publicly available at http)://www.ncbi.nlm.nih.gov/BLAST/.
  • lactic acid bacterial strain also encompasses homologues of the lactic acid bacterial strains.
  • the term “homologue” refers to a lactic acid bacterial strain having a nucleotide sequence having a degree of sequence identity or sequence homology with the nucleotide sequence of the parent lactic acid bacterial strain (hereinafter referred to as a “homologous sequence(s)”).
  • the term “homologous” means an entity having a certain homology with the subject nucleotide sequence.
  • the term “homology” can be equated with “identity”.
  • a homologous sequence is taken to include a nucleotide sequence which may be at least 50, 60, 70, 75, 80, 85 or 90% identical, preferably at least 95%, 97%, 98% or 99% identical to the nucleotide sequence of the parent lactic acid bacterial strain (the subject sequence).
  • Homology comparisons can be conducted by eye, or more usually, with the aid of readily available sequence comparison programs. These commercially available computer programs can calculate % homology between two or more sequences. % homology may be calculated over contiguous sequences, i.e. one sequence is aligned with the other sequence and each amino acid in one sequence is directly compared with the corresponding amino acid in the other sequence, one residue at a time. This is called an “ungapped” alignment. Typically, such ungapped alignments are performed only over a relatively short number of residues.
  • the degree of identity with regard to a nucleotide sequence is determined over at least 20 contiguous nucleotides, preferably over at least 30 contiguous nucleotides, preferably over at least 40 contiguous nucleotides, preferably over at least 50 contiguous nucleotides, preferably over at least 60 contiguous nucleotides, preferably over at least 100 contiguous nucleotides.
  • the degree of identity with regard to a nucleotide sequence may be determined over the whole sequence.
  • the traditional identification of bacteria on the basis of phenotypic characteristics is generally not as accurate as identification based on genotypic methods.
  • Comparison of the bacterial 16S rRNA gene sequence has emerged as a preferred genetic technique and allows for new strains to be identified by comparison of sequences with known bacterial DNA sequences using BLAST (http://blast.ncbi.nlm.nih.gov/Blast.cgi).
  • the 16S rRNA gene sequence is universal in bacteria, and so relationships can be measured across many different bacteria.
  • the comparison of the 16S rRNA sequence allows differentiation between organisms at the genus level across all major phyla of bacteria, in addition to classifying strains at multiple levels, including species and sub-species level.
  • the 16S rRNA gene sequence has been determined for a large number of strains.
  • GenBank the largest databank of nucleotide sequences, has over 20 million deposited sequences, of which over 90,000 are of 16S rRNA genes. This means that there are many previously deposited sequences against which to compare the sequence of an unknown strain.
  • the lactic acid bacterial strain has a 16S rRNA gene sequence selected from SEQ ID NOS 1-87, or a homologue or variant thereof.
  • Another embodiment of the invention relates to a lactic acid bacterial strain that comprises a 16S rRNA gene sequence selected from SEQ ID NOS 1-87, or a homologue or variant thereof. Preferred uses/methods apply to this aspect mutatis mutandis.
  • the term “homologue” is as defined hereinabove.
  • the term “variant” includes any variation wherein: (a) one or more nucleotides are substituted by another nucleotide or deleted, (b) the order of two or more nucleotides is reversed, (c) both (a) and (b) are present together.
  • the variants arise from one of (a), (b) or (c). More preferably, one or two nucleotides are substituted or deleted. Even more preferably, one nucleotide is substituted by another.
  • the lactic acid bacterial strain is characterised by the ability to exhibit antimicrobial activity against E. coli .
  • the observed antimicrobial activity is most likely by virtue of anti-microbial substances produced by the lactic acid bacterial strains of the invention, although nature of these anti-microbial substances has not been determined.
  • the ability to exhibit antimicrobial activity against E. coli can be determined by measuring inhibition of the growth of E. coli in an in vitro well diffusion assay. Further details of the well diffusion assay are set forth in the accompanying examples.
  • the assay is carried out using Escherichia coli K88 on MacConkey No 3 agar, incubating the plates for 16 hours at 37° C. More specifically, Escherichia coli K88 is added to the agar (1 ml of a 1:1000 dilution of an overnight culture of Escherichia coli K88 in 200 ml agar to give the equivalent of 106 CFU/ml). The agar is poured into petri dishes and allowed to set.
  • the plates are marked off into quadrants and an approximately 5 mm well cut out in each quadrant. An aliquot (60 ⁇ l) of conditioned media or MRS broth is added to the wells. The plates are covered and incubated for 16 hours at 37° C. They are photographed using a digital camera. Images are transferred to Photoshop, and the diameter of the well and zone of inhibition were determined using the measure tool.
  • the lactic acid bacterial strain of the invention exhibits ⁇ 20000 units of inhibition, more preferably 20000-40000 units, even more preferably 40000-60000 units, more preferably 60000-80000 units, more preferably 80000-100000 units of inhibition, even more preferably still >100000 units of inhibition.
  • the lactic acid bacterial strain is characterised by the ability to exhibit antimicrobial activity against S. enteritidis .
  • the observed antimicrobial activity is most likely by virtue of anti-microbial substances produced by the lactic acid bacterial strains of the invention, although nature of these anti-microbial substances has not been determined.
  • the ability to exhibit antimicrobial activity against S. enteritidis can be determined by measuring the ability to inhibit the growth of S. enteritidis in an in vitro well diffusion assay. Further details of the well diffusion assay are set forth in the accompanying examples. The assay is carried out using Salmonella enteritidis S1400 on XLD agar, incubating the plates for 16 hours at 37° C. XLD agar is prepared as per manufacturer's instructions and cooled to 45° C.
  • Salmonella enteritidis S1400 is added to the XLD agar (1 ml of a 1:1000 dilution of an overnight culture of Salmonella enteritidis S1400 in 200 ml agar to give the equivalent of 106 CFU/ml).
  • the XLD agar is poured into petri dishes and allowed to set. The plates are marked off into quadrants and an approximately 5 mm well cut out in each quadrant. An aliquot (60 ⁇ l) of conditioned media or MRS broth is added to the wells. The plates are covered and incubated for 16 hours at 37° C. and the data analysed as described above for the E. coli assay.
  • the lactic acid bacterial strain of the invention exhibits ⁇ 20000 units of inhibition, more preferably 20000-40000 units, even more preferably 40000-60000 units, more preferably 60000-80000 units, more preferably 80000-100000 units of inhibition, even more preferably still >100000 units of inhibition.
  • the ability to exhibit antimicrobial activity against S. enteritidis can be determined by measuring the ability to inhibit S. enteritidis in vivo in C3H/HeN or C57BI/6 mice. Further details of appropriate in vivo assays are set forth in the accompanying examples.
  • C3H/HeN and C57BI/6 mice are treated with a lactic acid bacterial strain according to the invention prior to and post-challenge with Salmonella enteritidis .
  • the mice are euthanased and dissected 6 (C57BI/6) or 10 (C3H/HeN) days post-infection and viable salmonella are detected in systemic tissues (e.g. the mesenteric lymph node, liver and spleen), in the intestine (e.g. caecum, colon) and in the faeces as compared to appropriate controls.
  • the in vivo activity of the lactic acid bacterial strain of the invention can also be measured by determining the level of myeloperoxidase [MPO], a marker for neutrophils, in the intestine of C3H/HeN mice treated with salmonella or salmonella plus LAB.
  • MPO in the intestine is greatly increased by salmonella infection, due to recruitment of neutrophils to the intestine part of the host response to infection.
  • Co-treatment with a lactic acid bacterial strain according to the invention reduces MPO activity in the intestine of salmonella -infected mice, indicating that the intestinal inflammatory responses to infection are lowered in these animals, relative to control experiments.
  • the lactic acid bacterial strain is characterised by the ability to suppress inflammation in IPEC cells induced by 12-O-tetradecaboylphorbol-13-acetate (PMA).
  • PMA 12-O-tetradecaboylphorbol-13-acetate
  • this refers to the ability of the lactic acid bacterial strain to block interleukin-8 (IL-8) gene expression triggered by PMA. More specifically, it can be determined by measuring the suppression of inflammation in IPEC-J2 cells induced by PMA when incubated for 2 hours at 37° C., 5% CO 2 , 95% humidity.
  • RNA and reverse transcription are carried out on a 7500 Fast Real-time PCR system operating with 7500 Fast System v 1.4.0 Sequence Detection Software version 1.4 (Applied Biosystem), using primers for porcine IL-8 and TNF- ⁇ (prepared by Sigma Aldrich).
  • the reaction mix is: 100 Power Sybergreen Master mix, 2.5 ⁇ l of forward primer, 2.5 ⁇ l of reverse primer and 5 ⁇ l of cDNA,
  • the Real Time PCR is then run according to the Standard 7500 protocol (95° C., 10 min, 1 cycle. 95° C., 15 sec, 40 cycles. 60° C., 1 min, 40 cycles. 95° C., 15 sec, 1 cycle. 60° C., 1 min, 1 cycle.
  • the lactic acid bacterial strain is characterised by the ability to block the attachment or invasion of IPEC cells by S. enteritidis .
  • This can be measured by the assay set forth in the accompanying examples.
  • monolayers of IPEC-J2 cells are grown to 3 days post-confluence in 24-well plates and synchronised by the addition of DTS media 24 hrs prior to use.
  • Overnight cultures of pig LAB (10 ml) are centrifuged and the bacteria re-suspended in phosphate buffered saline [PBS].
  • An aliquot (50 ⁇ l)) of LAB is added to the wells. The plates are incubated for 2 hours at 37° C., 5% CO 2 , 95% humidity.
  • S. enteritidis S1400 An overnight culture of Salmonella enterica serovar Enteritidis S1400 [ S. enteritidis S1400] is sub-cultured (0.5 ml in 10 ml) into Luria Bertani (LB) media and incubated aerobically for 2-3 hours at 37° C. until it reaches an optical density (560 nm) of 0.8 (a concentration equivalent to 1 ⁇ 108 CFU/ml). The culture is centrifuged and the bacteria re-suspended in PBS. An aliquot (50 ⁇ l) is added to the wells of IPEC-J2 cells. The plates are incubated for a further 2 hours at 37° C., 5% CO 2 , 95% humidity.
  • LB Luria Bertani
  • IPEC-J2 cell monolayers are washed with HBSS.
  • a solution (0.5 ml) of PBS containing Triton-X100 (10 ml/litre) is added to each well, the monolayer scraped off and dispersed.
  • Viable salmonella are estimated on XLD agar plates (incubated for 24 hours at 37° C.) by the Miles and Misra method. Lactic acid bacteria are determined by the same procedure (incubated anaerobically for 48 hours at 37° C.).
  • the lactic acid bacterial strain of the invention exhibits 0-20% inhibition of adherence/invasion, more preferably 20-40%, even more preferably 40-60%, more preferably still, 60-80%, even more preferably still, 80-100% inhibition of adherence/invasion as measured by the above assay.
  • the lactic acid bacterial strain is characterised by the ability to block the attachment or invasion of IPEC cells by E. coli . This can be measured by a similar assay to that described above for S. enteritidis , and as set forth in the accompanying examples.
  • the lactic acid bacterial strain of the invention exhibits 0-20% inhibition of adherence/invasion, more preferably 20-40%, even more preferably 40-60%, more preferably still, 60-80%, even more preferably still, 80-100% inhibition of adherence/invasion as measured by the above assay.
  • the lactic acid bacterial strain is characterised by the absence of antibiotic resistance to one or more antibiotics selected from the following: ampicillin; cefotaxime; chloramphenicol; erythromycin; gentamicin; tetracycline; vancomycin; metronizadole; nalidixic acid; and kanamycin.
  • antibiotic resistance can be determined by measuring the effect of various antibiotic-containing discs on an MRS agar plate culture of the lactic acid bacterial strain, when placed in an anaerobic jar and incubated for 24 hours at 37° C. Further details of the assay are set forth in the accompanying examples.
  • pig LAB [0.5 ml of a 1:100 dilution of an overnight culture] is spread onto the surface of an MRS agar plate and dried off.
  • the plates are marked off into 4 quadrants and in each quadrant is placed an antibiotic-containing disc [Ampicillin, 10 ⁇ g. Cefotaxime, 30 ⁇ g. Chloramphenicol, 10 ⁇ g. Erythromycin, 15 Gentamicin, 10 ⁇ g. Kanamycin, 30 ⁇ g. Metronizadole, 50 ⁇ g. Nalidixic acid, 30 ⁇ g. Tetracycline, 30 ⁇ g. Vancomycin, 30 ⁇ g].
  • the plates are covered, placed in an anaerobic jar and incubated for 24 hours at 37° C. The plates are photographed using a digital camera. Images are transferred to Photoshop, and the diameter of the zone of inhibition is determined using the measure tool. For each antibiotic, the exclusion area for the test strain is taken and divided with the maximum area of exclusion obtained for that antibiotic.
  • the LAB of the invention is characterised by the absence of resistance to the antibiotics ampicillin, cefotaxime, chloramphenicol, erythromycin, gentamicin, tetracycline, vancomycin, metronizadole, nalidixic acid and kanamycin. More preferably, the LAB of the invention is characterised by the absence of resistance to the antibiotics ampicillin, cefotaxime, chloramphenicol, erythromycin, gentamicin, tetracycline and vancomycin.
  • the lactic acid bacterial strain is characterised by the ability to exhibit heat stability when subjected to three cycles of heating, each cycle comprising heating at a temperature of 70° C. for a period of 15 minutes.
  • heat stability is measured by centrifuging an overnight culture (10 ml) of isolated pig LAB and resuspending the pellet in fresh MRS broth (10 ml). An aliquot (1 ml) is heated at 70° C. for 15 min and then plated out (0.5 ml) out on MRS agar and incubated in an anaerobic jar for 48 hours at 37° C.
  • the lactic acid bacterial strain has any two of the characterising features selected from the group consisting of (i), (ii), (iii), (iv), (v), (vi) and (vii) set forth above.
  • the lactic acid bacterial strain has any three of the characterising features selected from the group consisting of (i), (ii), (iii), (iv), (v), (vi) and (vii) set forth above.
  • the lactic acid bacterial strain has any four of the characterising features selected from the group consisting of (i), (ii), (iii), (iv), (v), (vi) and (vii) set forth above.
  • the lactic acid bacterial strain has any five of the characterising features selected from the group consisting of (i), (ii), (iii), (iv), (v), (vi) and (vii) set forth above.
  • the lactic acid bacterial strain has any six of the characterising features selected from the group consisting of (i), (ii), (iii), (iv), (v), (vi) and (vii) set forth above.
  • the lactic acid bacterial strain has all seven of the characterising features (i), (ii), (iii), (iv), (v), (vi) and (vii) set forth above.
  • the lactic acid bacterial strain is characterised by features (i) and (ii) above.
  • (B) the lactic acid bacterial strain characterised by features (iv) and (v) above.
  • the lactic acid bacterial strain is characterised by features (iv) and (v) above.
  • the lactic acid bacterial strain is characterised by features denoted (D) to (G) as follows:
  • the lactic acid bacterial strain is further characterised by feature (vi) in addition to those features recited in any one of embodiments (A) to (G) above.
  • the lactic acid bacterial strain is further characterised by feature (iii) in addition to those features recited in any one of embodiments (A) to (G) above.
  • the lactic acid bacterial strain is further characterised by feature (vii) in addition to those features recited in any one of embodiments (A) to (G) above.
  • One embodiment of the invention relates to a lactic acid bacterial strain isolated from the faeces of organically reared pigs and selected from the group consisting of strains deposited on 27 Jun. 2011 under the terms of the Budapest Treaty at National Collections of Industrial, Food and Marine Bacteria (NCIMB) at NCIMB Ltd, Ferguson Building, Craibstone Estate, Bucksburn, Aberdeen, UK, AB21 9YA, under the following accession numbers:
  • NCIMB 41846 Lactobacillus reuteri GGDK31;
  • NCIMB 41847 Lactobacillus plantarum/pentosus /paraplantarum GGDK161;
  • NCIMB 41848 Lactobacillus johnsonii/taiwanensis/acidophilus /gasseri GGDK255;
  • NCIMB 41849 Lactobacillus plantarum/pentosus/helveticus /paraplantarum GGDK258;
  • NCIMB 41850 Lactobacillus johnsonii GGDK266.
  • NCIMB 41846, NCIMB 41847, NCIMB 41848, NCIMB 41849 and NCIMB 41850 were made by Dr George Grant of the Rowett Institute of Nutrition and Health, University of Aberdeen, Greenburn Road, Aberdeen, AB21 9SB on behalf of the Applicant, GT Biologics Limited.
  • Another embodiment of the invention relates to a lactic acid bacterial strain isolated from the faeces of organically reared pigs and selected from the group consisting of strains deposited on 12 Jul. 2012 under the terms of the Budapest Treaty at National Collections of Industrial, Food and Marine Bacteria (NCIMB) at NCIMB Ltd, Ferguson Building, Craibstone Estate, Bucksburn, Aberdeen, UK, AB21 9YA, under the following accession numbers:
  • NCIMB 42008, NCIMB 42009, NCIMB 42010 and NCIMB 42011 and NCIMB 42012 were made by Professor Denise Kelly of GT Biologics Limited, c/o Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, Aberdeensshire, AB25 2ZD, UK, on behalf of the Applicant, GT Biologics Limited.
  • the invention also encompasses mutant strains, which can be obtained from said strains, and strains exhibiting a DNA-DNA homology of at least 70% and/or a 16S RNA identity of at least 99.5% with a strain selected from those deposited under the above accession numbers.
  • the term “16S rRNA identity” refers to the percentage identity with a known bacterial strain.
  • the lactic acid bacterial strain has a 16S rRNA identity of at least 85% or at least 90%, or at least 95, 96, 97, 98 or 99% with a strain selected from those deposited under the above accession numbers.
  • the lactic acid bacterial strain has a 16S rRNA identity of at least 99.5% with a strain selected from those deposited under the above accession numbers.
  • the term “DNA-DNA homology” refers to how closely related two or more separate strands of DNA are to each other, based on their nucleotide sequence. Typically, this is measured in terms of their % identity.
  • the lactic acid bacterial strain has a DNA-DNA homology of at least 70% with a strain selected from those deposited under the above accession numbers, more preferably, at least 80%, or at least 85%, more preferably still, at least 90, 95, 97, 98 or 99% homology with a strain selected from those deposited under the above accession numbers.
  • the lactic acid bacterial strain has a DNA-DNA homology of at least 70% and a 16S rRNA identity of at least 99.5% with a strain selected from those deposited under the above accession numbers.
  • compositions comprising one or more lactic acid bacterial strains as described above and a pharmaceutically acceptable excipient, carrier or diluent. Suitable excipients, diluents, carriers are described below.
  • the composition may be any composition, but is preferably a composition to be administered orally, enterally or rectally.
  • the composition may be an edible composition.
  • “Edible” means a material that is approved for human or animal consumption.
  • Another aspect of the invention relates to a probiotic composition
  • a probiotic composition comprising a lactic acid bacterial strain as described above.
  • Another aspect of the invention relates to combinations of two more lactic acid bacterial strains as described herein.
  • such combinations exhibit a synergistic functionality, for example, the combination is synergistic, i.e. the resultant effect is greater than the simple additive effects attributable to the individual lactic acid bacterial components in the combination.
  • One preferred embodiment of the invention relates to a combination of two, three, four or five different lactic acid bacteria, more preferably, two, three or four different lactic acid bacteria, more preferably, two or three different lactic acid bacteria.
  • the individual components of the combination may be present in any ratio.
  • the invention relates to a combination of two different lactic acid bacteria.
  • the two different lactic acid bacteria are present in a ratio of from 1/99.9 to 99.9/1 by weight, for example, 1/99 to 99/1 or 10/90 to 90/10, or 20/80 to 80/20, or 30/70 to 70/30 and the like.
  • the combination is a mixture of Lactobacillus johnsonii and Lactobacillus reuteri . Even more preferably, the combination is NCIMB 41850: Lactobacillus johnsonii and Lactobacillus reuteri GGDK266 as described above. Surprisingly, this particular combination of lactic acid bacteria unexpectedly gives rise to beneficial in vivo responses in early weaned pigs (see Examples).
  • the combination is a mixture of Lactobacillus plantarum and Lactobacillus reuteri . Even more preferably, the combination is NCIMB 41847: Lactobacillus plantarum/pentosus /paraplantarum and Lactobacillus reuteri GGDK161 as described above.
  • probiotic means microbial cell preparations or components of microbial cells with a beneficial effect on the health or well-being of the host.
  • the probiotic composition is an orally administrable composition of metabolically active, i.e., live and/or or lyophilized, or non-viable heat-killed, irradiated or lysed probiotic bacteria.
  • the probiotic composition may contain other ingredients.
  • the probiotic composition of the invention can be administered orally, i.e., in the form of a tablet, capsule or powder.
  • the probiotic composition of the invention may be administered orally as a food or nutritional product, such as milk or whey based fermented dairy product, or as a pharmaceutical product.
  • a suitable daily dose of the probiotic bacteria is from about 1 ⁇ 10 3 to about 1 ⁇ 10 11 colony forming units (CFU), more preferably from about 1 ⁇ 10 7 to about 1 ⁇ 10 10 CFU, more preferably, about 1 ⁇ 10 6 to about 1 ⁇ 10 10 CFU.
  • CFU colony forming units
  • the composition contains bacterial strains and/or their cellular components, as active ingredients, in an amount of from about 1 ⁇ 10 6 to about 1 ⁇ 10 12 CFU/g, respect to the weight of the composition, preferably from about 1 ⁇ 10 8 to about 1 ⁇ 10 10 CFU/g.
  • the dose may be of 1 g, 3 g, 5 g, and 10 g, by way of example.
  • a probiotic is optionally combined with at least one suitable prebiotic compound.
  • a prebiotic is usually a non-digestible carbohydrate such as an oligo- or polysaccharide, or a sugar alcohol which is not degraded or absorbed in the upper digestive tract.
  • Known prebiotics include commercial products such as inulin and transgalacto-oligosaccharides.
  • the composition of the present invention includes a prebiotic in an amount of from about 1 to about 30% by weight, respect to the total weight composition, preferably from 5 to 20% by weight.
  • Preferred carbohydrates are selected from: fructo-oligosaccharides (or FOS), short-chain fructo-oligosaccharides, inulin, isomalt-oligosaccharides, pectins, xylo-oligosaccharides (or XOS), chitosan-oligosaccharides (or COS), beta-glucans, arable gum modified and resistant starches, polydextrose, D-tagatose, acacia fibers, carob, oats, and citrus fibers.
  • Particularly preferred prebiotics are the short-chain fructo-oligosaccharides (for simplicity shown hereinbelow as FOSs-c.c); said FOSs-c.c. are not digestable glucides, generally obtained by the conversion of the beet sugar and including a saccharose molecule to which three glucose molecules are bonded.
  • a further aspect of the invention relates to a process for producing a probiotic, said process comprising culturing a lactic acid bacterial strain according to the invention.
  • the skilled person in the art will be familiar with standard techniques and conditions suitable for culturing a bacterial strain according to the invention.
  • a further aspect of the invention relates to a method of preparing one or more bacterial strains according to the invention, said method comprising the steps of:
  • Suitable agars include, for example, MRS or LAMVAB agar plates. However, other suitable agars can also be used, and would be familiar to the skilled person.
  • Suitable broths include, for example, MRS broth. However, other suitable broths can also be used, and would be familiar to the skilled person.
  • step (iii) involves incubating the agar for at least 72 hours at a temperature of about 37° C.
  • step (vi) involves incubating the seeded colonies for at least 48 hours at a temperature of about 37° C.
  • a further aspect of the invention relates to a process for obtaining a porcine lactic acid bacterial strain, said process comprising obtaining faeces from an organically reared pig and extracting one or more porcine lactic acid bacterial strains from said faeces.
  • the process comprises the steps of:
  • Another aspect of the invention relates to a porcine lactic acid bacterial strain obtained by, or obtainable by, the process described above.
  • Another aspect of the invention relates to one or more lactic acid bacterial strains as defined above for use in medicine.
  • Another aspect of the invention relates to one or more lactic acid bacterial strains as defined above for use in treating an intestinal disorder.
  • Another aspect of the invention relates to the use of one or more lactic acid bacterial strains or a composition as defined above in the preparation of a medicament for treating an intestinal disorder.
  • the term “medicament” as used herein encompasses medicaments for both human and animal usage in human and veterinary medicine.
  • the term “medicament” as used herein means any substance which provides a therapeutic and/or beneficial effect.
  • the term “medicament” as used herein is not necessarily limited to substances which need Marketing Approval, but may include substances which can be used in cosmetics, nutraceuticals, food (including feeds and beverages for example), probiotic cultures, and natural remedies.
  • the term “medicament” as used herein encompasses a product designed for incorporation in animal feed, for example livestock feed and/or pet food.
  • Another aspect of the invention relates to a method of treating an intestinal disorder in a subject, said method comprising administering to the subject a pharmaceutically effective amount of one or more lactic acid bacterial strains or a pharmaceutical composition or a probiotic composition as described above.
  • the intestinal disorder is selected from irritable bowel syndrome (IBS), inflammatory bowel disorder (IBD), functional dyspepsia, functional constipation, functional diarrhoea (including antibiotic associated diarrhoea, traveller's diarrhoea and pediatric diarrhoea), functional abdominal pain, functional bloating, Epigastric Pain Syndrome, Postprandial Distress Syndrome, Crohn's disease, ulcerative colitis, gastrointestinal reflux disease (GERD), allergies, atopic diseases e.g. atopic dermatitis, necrotising enterocolitis, other infections, and combinations thereof.
  • IBS irritable bowel syndrome
  • IBD inflammatory bowel disorder
  • functional dyspepsia including antibiotic associated diarrhoea, traveller's diarrhoea and pediatric diarrhoea
  • functional abdominal pain functional bloating
  • Epigastric Pain Syndrome Postprandial Distress Syndrome
  • Crohn's disease Crohn's disease
  • the intestinal disorder is IBS.
  • the precise pathophysiology of IBS remains to be elucidated. Recent studies have described mucosal inflammation and alterations in intestinal microbiota in IBS patients and a disease correlation with intestinal infections.
  • the disorder is salmonellosis.
  • Salmonellosis is a disease caused by various strains of salmonella that is characterized by fever and intestinal disorders.
  • Another aspect of the invention relates to one or more lactic acid bacterial strains as defined above for improving intestinal microbiota.
  • Another aspect of the invention relates to a method of improving intestinal microbiota in a subject, said method comprising administering to the subject a composition comprising one or more lactic acid bacterial strains or a pharmaceutical composition or a probiotic composition according to the invention.
  • compositions according to the invention are administered to a patient susceptible to, or otherwise at risk of, a particular disease in an amount that is sufficient to at least partially reduce the risk of developing a disease. Such an amount is defined to be “a prophylactic effective dose”.
  • a prophylactic effective dose The precise amounts depend on a number of patient specific factors such as the patient's state of health and weight.
  • the lactic acid bacterial strains and probiotic compositions according to the invention may also be used in animal nutrition (e.g. in pig nutrition), particularly in the early-weaned period and growing fattening period.
  • the probiotics are expected to enhance immune function reduce and prevent infectious diseases, beneficially alter the microbiota composition, and improve growth and performance of animals, for example, through increased feed conversion efficiency.
  • the term “animal” includes all animals including humans. Examples of animals are non-ruminants and ruminants. Ruminant animals include for example, sheep, goat, and cattle eg. cow as beef cattle and dairy cows. In a particular embodiment, the animal is a non-ruminant animal.
  • Non-ruminant animals include pet animals, eg horses, cats, and dogs; monogastric eg pigs or swine (including but not limited to, piglets growing pigs and sows); poultry such as turkeys, ducks, and chickens (including but not limited to broiler chicks, layers); fish (including but not limited to salmon, trout, tilapia, catfish and carp); and crustaceans (including but not limited to shrimp and prawn).
  • pet animals eg horses, cats, and dogs
  • monogastric eg pigs or swine including but not limited to, piglets growing pigs and sows
  • poultry such as turkeys, ducks, and chickens (including but not limited to broiler chicks, layers)
  • fish including but not limited to salmon, trout, tilapia, catfish and carp
  • crustaceans including but not limited to shrimp and prawn
  • a further aspect of the invention relates to food products, dietary supplements, nutraceuticals, nutritional formulae, drinks and medicaments containing one or more bacterial strains according to the invention.
  • the composition comprises additionally at least one other kind of other food grade bacterium, wherein the food grade bacterium is preferably selected from the group consisting of lactic acid bacteria, bifidobacteria, propionibacteria or mixtures thereof.
  • One aspect of the invention relates to a food product comprising one or more lactic acid bacterial strains according to the invention.
  • the term “food product” is intended to cover all consumable products that can be solid, jellied or liquid. Suitable food products may include, for example, functional food products, food compositions, pet food, livestock feed, health foods, feedstuffs and the like. In one preferred embodiment, the food product is a health food.
  • functional food product means food that is capable of providing not only a nutritional effect, but is also capable of delivering a further beneficial effect to the consumer. Accordingly, functional foods are ordinary foods that have components or ingredients (such as those described herein) incorporated into them that impart to the food a specific functional—e.g. medical or physiological benefit—other than a purely nutritional effect.
  • Examples of specific food products that are applicable to the present invention include milk-based products, ready to eat desserts, powders for re-constitution with, e.g., milk or water, chocolate milk drinks, malt drinks, ready-to-eat dishes, instant dishes or drinks for humans or food compositions representing a complete or a partial diet intended for pets or livestock.
  • the composition according to the present invention is a food product intended for humans, pets or livestock.
  • the composition may be intended for animals selected from the group consisting of dogs, cats, pigs, cattle, horses, goats, sheep or poultry.
  • the composition is a food product intended for adult species, in particular human adults.
  • milk-based product means any liquid or semi-solid milk or whey based product having a varying fat content.
  • the milk-based product can be, e.g., cow's milk, goat's milk, sheep's milk, skimmed milk, whole milk, milk recombined from powdered milk and whey without any processing, or a processed product, such as yoghurt, curdled milk, curd, sour milk, sour whole milk, butter milk and other sour milk products.
  • Another important group includes milk beverages, such as whey beverages, fermented milks, condensed milks, infant or baby milks; flavoured milks, ice cream; milk-containing food such as sweets.
  • One aspect of the invention relates to a feedstuff or animal feed comprising one or more bacterial strains according to the invention.
  • Feedstuff can be a food additive, a feed premixor an animal feed.
  • feedstuffs according to the invention include the following: animal feed additive comprising (a) porcine lactic acid bacteria according to the present invention (b) at least one fat soluble vitamin (c) at least one water soluble vitamin (d) at least one trace mineral and/or at least one macro mineral; an animal feed composition comprising a porcine lactic acid bacteria according to the present invention and a crude protein content of 50-88 g/kg feed.
  • the so-called premixes are examples of animal feed additives of the invention.
  • a premix designates a preferably uniform mixture of one or more micro-ingredients with diluent and/or carrier. Premixes are used to facilitate uniform dispersion of micro-ingredients in a larger mix.
  • feed-additive ingredients are coloring agents, e.g. carotenoids such as beta-carotene, astaxanthin, and lutein; aroma compounds; stabilisers; antimicrobial peptides; polyunsaturated fatty acids; reactive oxygen generating species; and/or at least one enzyme selected from amongst phytase (EC 3.1.3.8 or 3.1.3.26); xylanase (EC 3.2.1.8); galactanase (EC 3.2.1.89); alpha-galactosidase (EC 3.2.1.22); protease (EC 3.4.), phospholipase A1 (EC 3.1.1.32); phospholipase A2 (EC 3.1.1.4); lysophospholipase (EC 3.1.1.5); phospholipase C (EC 3.1.4.3); phospholipase D (EC 3.1.4.4); amylase such as, for example, alpha-amylase (EC 3.2.1.1); and/or
  • polyunsaturated fatty acids are C18, C20 and C22 polyunsaturated fatty acids, such as arachidonic acid, docosohexaenoic acid, eicosapentaenoic acid and gamma-linoleic acid.
  • reactive oxygen generating species are chemicals such as perborate, persulphate, or percarbonate; and enzymes such as an oxidase, an oxygenase or a syntethase.
  • Examples of fat-soluble vitamins are vitamin A, vitamin D3, vitamin E, and vitamin K, e.g. vitamin K3.
  • examples of water-soluble vitamins are vitamin B12, biotin and choline, vitamin B1, vitamin B2, vitamin B6, niacin, folic acid and panthothenate, e.g. Ca-D-panthothenate.
  • examples of trace minerals are manganese, zinc, iron, copper, iodine, selenium, and cobalt.
  • Examples of macro minerals are calcium, phosphorus and sodium.
  • the animal feed additive of the invention comprises at least one of the individual components specified in Table A of WO 01/58275. At least one means either of, one or more of, one, or two, or three, or four and so forth up to all thirteen, or up to all fifteen individual components. More specifically, this at least one individual component is included in the additive of the invention in such an amount as to provide an in-feed-concentration within the range indicated in column four, or column five, or column six of Table A of WO 01/58275.
  • Animal feed compositions or diets typically have a relatively high content of protein.
  • Poultry and pig diets can be characterized as indicated in Table B of WO 01/58275, columns 2-3.
  • Fish diets can be characterized as indicated in column 4 of this Table B.
  • WO 01/58275 corresponds to U.S. Ser. No. 09/779,334 which is hereby incorporated by reference.
  • An animal feed composition according to the invention typically has a crude protein content of 50-800 g/kg, and furthermore comprises a porcine lactic acid bacteria according to the present invention thereof as described and/or claimed herein.
  • the animal feed composition of the invention may have a content of metabolisable energy of 10-30 MJ/kg; and/or a content of calcium of 0.1-200 g/kg; and/or a content of available phosphorus of 0.1-200 g/kg; and/or a content of methionine of 0.1-100 g/kg; and/or a content of methionine plus cysteine of 0.1-150 g/kg; and/or a content of lysine of 0.5-50 g/kg.
  • the content of metabolisable energy, crude protein, calcium, phosphorus, methionine, methionine plus cysteine, and/or lysine is within any one of ranges 2, 3, 4 or 5 in Table B of WO 01/58275 (R. 2-5).
  • the nitrogen content is determined by the Kjeldahl method (A.O.A.C., 1984, Official Methods of Analysis 14th ed., Association of Official Analytical Chemists, Washington D.C.).
  • Metabolisable energy can be calculated on the basis of the NRC publication Nutrient requirements in swine, ninth revised edition 1988, subcommittee on swine nutrition, committee on animal nutrition, board of agriculture, national research council. National Academy Press, Washington, D.C., pp. 2-6, and the European Table of Energy Values for Poultry Feed-stuffs, Spelderholt centre for poultry research and extension, 7361 DA Beekbergen, The Netherlands. Grafisch bedrijf Ponsen & looijen by, Wageningen. ISBN 90-71463-12-5.
  • the dietary content of calcium, available phosphorus and amino acids in complete animal diets is calculated on the basis of feed tables such as Veevoedertabel 1997, gegevens over chemische samenstelling, verteerbaarheid en voederwaarde van voedermiddelen, Central Veevoederbureau, Runderweg 6, 8219 pk Lelystad. ISBN 90-72839-13-7.
  • the animal feed composition of the invention contains at least one vegetable protein or protein source. It may also contain animal protein, such as Meat and Bone Meal, and/or Fish Meal, typically in an amount of 0-25%.
  • vegetable proteins refers to any compound, composition, preparation or mixture that includes at least one protein derived from or originating from a vegetable, including modified proteins and protein-derivatives.
  • the protein content of the vegetable proteins is at least 10, 20, 30, 40, 50, or 60% (w/w).
  • Vegetable proteins may be derived from vegetable protein sources, such as legumes and cereals, for example materials from plants of the families Fabaceae (Leguminosae), Cruciferaceae, Chenopodiaceae, and Poaceae, such as soy bean meal, lupin meal and rapeseed meal.
  • the vegetable protein source is material from one or more plants of the family Fabaceae, e.g. soybean, lupine, pea, or bean.
  • Other examples of vegetable protein sources are rapeseed, sunflower seed, cotton seed, and cabbage.
  • Other examples of vegetable protein sources are cereals such as barley, wheat, rye, oat, maize (corn), rice, triticale, and sorghum.
  • Animal diets can e.g. be manufactured as mash feed (non pelleted) or pelleted feed.
  • the milled feed-stuffs are mixed and sufficient amounts of essential vitamins and minerals are added according to the specifications for the species in question.
  • a porcine lactic acid bacteria according to the present invention thereof can be added as solid or liquid formulations.
  • compositions of the present invention may be—or may be added to—food supplements, also referred to herein as dietary supplements or food additives.
  • dietary supplements also referred to herein as dietary supplements or food additives.
  • another aspect of the invention relates to a dietary supplement or food additive comprising one or more bacterial strains according to the invention.
  • Another embodiment of the invention relates to the use of a feedstuff as described above for improving animal growth performance as measured by daily weight gain and/or feed conversion ratio.
  • the invention relates to methods for using a feedstuff comprising one or more porcine lactic acid bacteria according to the present invention in animal feed for improving daily weight gain, improving the Feed Conversion Ratio (FCR) and/or for modulation of the gut microflora.
  • a feedstuff comprising one or more porcine lactic acid bacteria according to the present invention in animal feed for improving daily weight gain, improving the Feed Conversion Ratio (FCR) and/or for modulation of the gut microflora.
  • FCR Feed Conversion Ratio
  • the feedstuff comprising one or more porcine lactic acid bacteria according to the present invention improves animal feed digestibility, and/or maintains animal health by aiding in proper digestion and/or supporting immune system function.
  • the FCR may be determined on the basis of a piglet growth trial comprising a first treatment in which the feedstuff comprising a porcine lactic acid bacteria according to the present invention is added to the animal feed in a suitable concentration per kg feed, and a second treatment (control) with no addition of a porcine lactic acid bacteria according to the present invention to the animal feed.
  • the term Feed Conversion Ratio, or FCR is used synonymously with the term feed conversion.
  • the FCR is calculated as the feed intake in g/animal relative to the weight gain in g/animal. As it is generally known, an improved FCR is lower than the control FCR.
  • the FCR is improved (i.e., reduced) as compared to the control by at least 1.0%, preferably at least 1.5%, 1.6%, 1.7%, 1.8%, 1.9%, 2.0%, 2.1%, 2.2%, 2.3%, 2.4%, or at least 2.5%.
  • gut designates the gastrointestinal or digestive tract (also referred to as the alimentary canal) and it refers to the system of organs within multicellular animals which takes in food, digests it to extract energy and nutrients, and expels the remaining waste.
  • gut “microflora” refers to the natural microbial cultures residing in the gut and maintaining health by aiding in proper digestion and/or supporting immune system function.
  • module as used herein in connection with the gut microflora generally means to change, manipulate, alter, or adjust the function or status thereof in a healthy and normally functioning animal, i.e. a non-therapeutic use.
  • the invention also relates to compositions, more preferably pharmaceutical compositions, comprising a lactic acid bacterial strain according to the invention.
  • the lactic acid bacterial strains of the present invention are generally administered in admixture with a pharmaceutical carrier, excipient or diluent, particularly for human therapy.
  • the pharmaceutical compositions may be for human or animal usage in human and veterinary medicine.
  • Acceptable carriers or diluents for therapeutic use are well known in the pharmaceutical art, and are described, for example, in Remington's Pharmaceutical Sciences, Mack Publishing Co. (A. R. Gennaro edit. 1985).
  • suitable carriers include lactose, starch, glucose, methyl cellulose, magnesium stearate, mannitol, sorbitol and the like.
  • suitable diluents include ethanol, glycerol and water.
  • compositions may comprise as, or in addition to, the carrier, excipient or diluent any suitable binder(s), lubricant(s), suspending agent(s), coating agent(s), solubilising agent(s).
  • Suitable binders include starch, gelatin, natural sugars such as glucose, anhydrous lactose, free-flow lactose, beta-lactose, corn sweeteners, natural and synthetic gums, such as acacia, tragacanth or sodium alginate, carboxymethyl cellulose and polyethylene glycol.
  • Suitable lubricants include sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride and the like.
  • Preservatives, stabilizers, dyes and even flavoring agents may be provided in the pharmaceutical composition.
  • preservatives include sodium benzoate, sorbic acid and esters of p-hydroxybenzoic acid.
  • Antioxidants and suspending agents may be also used.
  • compositions of the present invention may be adapted for oral, rectal, vaginal, parenteral, intramuscular, intraperitoneal, intraarterial, intrathecal, intrabronchial, subcutaneous, intradermal, intravenous, nasal, buccal or sublingual routes of administration.
  • the compositions of the present invention are adapted for oral, rectal, vaginal, parenteral, nasal, buccal or sublingual routes of administration.
  • compositions of the present invention may also be in form of suppositories, pessaries, suspensions, emulsions, lotions, ointments, creams, gels, sprays, solutions or dusting powders.
  • transdermal administration is by use of a skin patch.
  • the active ingredient can be incorporated into a cream consisting of an aqueous emulsion of polyethylene glycols or liquid paraffin.
  • the lactic acid bacterial strain can also be incorporated into an ointment consisting of a white wax or white soft paraffin base together with such stabilisers and preservatives as may be required.
  • compositions may be formulated in unit dosage form, i.e., in the form of discrete portions containing a unit dose, or a multiple or sub-unit of a unit dose.
  • a person of ordinary skill in the art can easily determine an appropriate dose of one of the instant compositions to administer to a subject without undue experimentation.
  • a physician will determine the actual dosage which will be most suitable for an individual patient and it will depend on a variety of factors including the activity of the specific bacterial strain employed, the metabolic stability and length of action of that strain, the age, body weight, general health, sex, diet, mode and time of administration, rate of excretion, drug combination, the severity of the particular condition, and the individual undergoing therapy.
  • the dosages disclosed herein are exemplary of the average case. There can of course be individual instances where higher or lower dosage ranges are merited, and such are within the scope of this invention.
  • the usual effective daily dose in humans or in animals is from about 1 ⁇ 10 3 to about 1 ⁇ 10 11 , more preferably, from about 1 ⁇ 10 7 to about 1 ⁇ 10 11 , even more preferably, from about 1 ⁇ 10 6 to about 1 ⁇ 10 10 CFU.
  • compositions of the invention are administered in any combination, for example, two or more of the lactic acid bacteria may be administered in any combination or ratio.
  • compositions of the invention are administered in combination with one or more other active agents.
  • the compositions of the invention may be administered consecutively, simultaneously or sequentially with the one or more other active agents.
  • the LAB strains isolated (total of 436 individual colony picks) from faeces of organically-reared pigs were predominantly L. reuteri, L. johnsonii, L. gasseri, L. pentosus , strains with a small number of L. plantarum, L. acidophilus, L. vaginalis , a single L. mucosae and several uncultured strains.
  • Certain strains were selected on the basis of anti-microbial potency as determined in vitro. These bacteria were further screened for their ability to block adherence/invasion of intestinal pig epithelial cells (IPEC) by pathogens in vitro and their susceptibility to antibiotics.
  • IPEC intestinal pig epithelial cells
  • mice with pig LAB L. reuteri or L. mucosae greatly reduced the pathogenicity of S. enteritidis in acute (C57BI/6 mouse) and chronic (C3H/HeN mouse) forms of salmonellosis.
  • LAB strains were isolated and cultured from pig faeces using selective microbiological media. Individual bacterial colonies were isolated and 16S rRNA gene sequences were analysed to enable genotypic identification of bacterial strains. Phenotypic characteristic of potential probiotics was further determined following measurement of adherence, anti-bacterial and anti-inflammatory activities, antibiotic susceptibility and finally heat stability. Anti-bacterial activity of conditioned media derived from LAB was evaluated using well-diffusion assays to determine killing activity against the enteric pathogens Salmonella enteritidis and E. coli K88. The ability of LAB strains to block or interfere with S. enteritidis and E.
  • IPEC pig epithelial
  • PMA 12-O-Tetradecaboylphorbol-13-acetate
  • a ranking system based on scoring the biological properties of LAB was established and used for the selection of candidate LAB strains for probiotic evaluation in vivo.
  • the LAB (436 individual colony picks) isolated from faeces of organically-reared pigs were predominantly L. johnsonii or L. johnsonii -related and L. reuteri or L. reuteri -related with small numbers of L. plantarum -related and uncultured strains. This represented a much narrower range of porcine-associated LAB than reported by others (Martin et al, 2009; Yun et al, 2009; Lahteinen et al, 2010; Yao et al, 2011). However, in comparison to conventionally/intensively-reared pigs, out-door organically-reared pigs had high levels of LAB and more developed intestinal immune function (Mulder et al, 2009).
  • the present bacterial data indicate that L. johnsonii and L. reuteri strains are of particular importance in proper development of the gut and immune system in young pigs.
  • the inclusion of other lactic acid bacteria derived from the gut or faeces of organically-reared pigs, in particular, Lactobacillus delbrueckii and Lactobacillus amylovorous may enhance the immune homeostatic properties of Lactobacillus reuteri, Lactobacillus plantarum and Lactobacillus johnsonii.
  • the LAB may limit the access of pathogens to the epithelial layer by occupying binding-sites on the cell monolayer or by production of factors that interfere with attachment of the pathogen to the epithelial cells, such as blocking binding sites of surface adhesins (Ljungh and Wadstrom, 2006; Blandino et al, 2008; Williams, 2010).
  • Pig LAB may also block or suppress inflammatory gene (interleukin-8, IL-8)-expression triggered in IPEC cells by PMA.
  • inflammatory gene interleukin-8, IL-8
  • Individual cultures varied greatly in their ability to affect inflammation, but five strains (RINH vial 29, 30, 31 86 and 266) had potent anti-inflammatory properties.
  • Certain LAB strains are known to have immuno-modulatory or anti-inflammatory properties (Cotter et al, 2005; Blandino et al, 2008; Ohashi and Ushida, 2009; Elmadfa et al, 2010; Liu et al, 2010). The mechanisms involved remain unclear, but are likely to involve modulation of molecular signalling systems by bioactive factors produced by the LAB.
  • Antibiotic resistance is an increasing problem and can spread between bacteria by gene transfer (Korhonen et al, 2007; Gousia et al, 2011; Nicolau, 2011). Ideally, candidate probiotics should have little or no resistance to antibiotics to minimise the risk of transfer of resistance genes to the host flora.
  • Pig LAB 33 strains were screened for resistance to 10 individual antibiotics. One strain (RINH vial 266) was susceptible to all the tested antibiotics. Most were susceptible to ampicillin, cefotaxime, chloramphenicol, erythromycin, gentamicin, tetracycline and vancomycin. However, most exhibited resistance to metronizadole, nalidixic acid and to a lesser extent kanamycin. This relatively low incidence of antibiotic resistance amongst these LAB isolates may be linked to the environment in which the source piglets were reared [organic out-door reared] (Mulder et al, 2009).
  • L. johnsonii, L. reuteri and L. plantarum exhibited strain-specific general substrate reaction profiles, when assayed using an API CH 50 kit. However, most genotype strains exhibited fine differences in their substrate reactivity. This indicated that they were unique individual strains of the genotype.
  • the LAB can withstand being freeze dried to allow them to be handled and processed as probiotics. However, their viability can be greatly reduced during freezing and drying (Tomas et al, 2009; Strasser et al, 2009; Reddy et al, 2009). Skimmed milk powder, alone or in combination with simple sugars, is often used as a cryo-protectant to preserve the viability of the bacteria (Tomas et al, 2009; Strasser et al, 2009). In the present study, small losses in viability were evident on drying and storage of pig LAB in skimmed milk powder alone. Sucrose or lactose in combination with skimmed milk powder was slightly more protective. However, the product was hygroscopic and difficult to store or handle. It was therefore decided to dry and store pig LAB in skimmed milk powder.
  • Supplemental feeds for animal are often given as pellets, production of which involves high temperatures (De Angelis et al, 2006).
  • LAB to be added to animal feeds should therefore have a significant degree of heat-stability to minimise loss of viability during processing.
  • five LAB were subject to heating three times for 15 minutes at 70° C. All of the bacteria that were recovered after the third heat-treatment were viable and in most cases grew at rates similar to the native forms of the bacteria. Two of the bacteria retained the biological properties of the native non-heat-treated forms. However, one of the heat-treated strains had lost the ability to block attachment of pathogen to epithelial cells in vitro and another had greatly reduced blocking activity.
  • a further strain was unable to block PMA-induced inflammation in epithelial cells in vitro, although the native form was a potent suppressor of inflammation. Heat-treatment can thus differentially affect the biological properties of individual LAB. This needs to be taken into account when considering inclusion of LAB in pelleted animal feeds.
  • RINH vial 323 L. mucosae greatly reduced the ability of S. enteritidis to invade, spread to and proliferate in systemic tissues in acute (C57BI/6 mouse) and chronic (C3H/Hen mouse) salmonellosis .
  • RINH vial 31 [GGDK31] and RINH vial 32 were the most effective in this chronic model of salmonellosis .
  • These LAB have potential as novel probiotics to promote gut health or increase resistance to infection in vivo.
  • LAB By competing for nutrients, killing of pathogen or blocking of attachment sites, LAB could limit the numbers of salmonella in the large intestine reservoir. LAB may also prevent attachment to ileal mucosal cells, in a manner similar to that observed here with IPEC-J2 cells and with Caco-2 cells (Neeser et al, 2000) and thereby limit invasion.
  • LAB may directly modulate host responses to the infection, in particular suppression of inflammation.
  • gut damage and preserving barrier integrity Smith et al, 2008; Schreiber et al, 2009
  • the ability of salmonella to invade and spread would be greatly reduced.
  • FIG. 1 shows an assay of antibacterial activity of conditioned media from Lactic Acid Bacteria.
  • FIGS. 2A and 2B show inhibitory activity against S. enteritidis S1400 (expressed as area of inhibition in a well diffusion assay) of conditioned media of all individual LAB cultured from faeces of organically-reared pigs.
  • FIGS. 3A and 3B show inhibitory activity against E. coli K88 (expressed as area of inhibition in a well diffusion assay) of conditioned media of all individual LAB cultured from faeces of organically-reared pigs.
  • FIGS. 3C and 3D show inhibitory activity (expressed as area of inhibition in a well diffusion assay) of conditioned media of all individual LAB cultured from faeces of organically-reared pigs.
  • FIGS. 4A, 4B, and 4C shown inhibition of adherence by ( FIG. 4A ) S. enteritidis S1400; and ( FIG. 4B ) E. coli K88 to IPEC cells in culture by LAB cultured from faeces of organically-reared pigs; ( FIG. 4C ) comparison between inhibition of S. enteritidis S1400 and E. coli K88.
  • FIG. 5 shows an assay of the antibiotic susceptibility of Lactic Acid Bacteria using discs impregnated with a defined amount of antibiotic.
  • FIG. 6 shows an evaluation of substrate profile of LAB using an API CH 50 kit [49 substrates, pale colour indicated positive reaction, except 25 where positive reaction is black, dark colour indicates no reaction].
  • FIGS. 7A-7C show the ⁇ Ct ( FIG. 7A ), ratio ( FIG. 7B ) and fold-change ( FIG. 7C ) for IL-8 gene expression in IPEC cells treated with PMA and pig LAB.
  • FIGS. 8A and 8B show the stability of L. reuteri ( FIG. 8A ) and L. johnsonii ( FIG. 8B ) after freeze-drying in skimmed milk powder (SKP, (100 g/1), SKP+lactose (both 100 g/1), SKP+sucrose (both 100 g/1) or SKP (200 g/1).
  • FIGS. 9A-9D show the stability of isolated LAB to heat-treatment ( FIG. 9A ), the ratio ( FIG. 9B ) and fold-change ( FIG. 9C ) for IL-8 gene expression in IPEC cells treated with PMA and naive or heat-treated pig LAB; ( FIG. 9D ) Antibiotic susceptibility of native and heat-treated RINH vial 31.
  • FIG. 10 shows a protocol for the C3H/HeN mouse study to evaluate efficacy of vial 323 (L. mucosae) to counteract salmonella infection in vivo.
  • FIGS. 11A-11C show the distribution of S. enteritidis S1400 in tissues at 10 days post-infection in C3H/HeN mice that had or had not been co-treated with 323 ( L. mucosae , LM).
  • FIGS. 12A-12B show spleen weight (mg/100 g BW) and intestinal (ileal) myeloperoxidase ( ⁇ g) at 10 days post-infection in C3H/HeN mice that had or had not been co-treated with vial 323 (L. mucosae).
  • FIG. 13 shows a protocol for the C57 BI/6 mouse study to evaluate efficacy of vial 323 (L. mucosae) to counteract acute salmonella infection in vivo.
  • FIGS. 14A-14C shows the distribution of S. enteritidis S1400 in tissues at 6 days post-infection in C57BI/6 mice that had or had not been co-treated with RINH vial 323.
  • FIG. 15 shows spleen weight (mg/100 g BW) at 6 days post-infection in C57BI/6 mice that had or had not been co-treated with vial 323 ( L. mucosae ).
  • FIG. 16 shows a protocol for the C3H/HeN mouse study to evaluate efficacy of selected LAB from faeces of organically reared pigs to counteract salmonella infection in vivo.
  • FIGS. 17A & 17B show excretion of S. enteritidis in faeces at 7-8 days post-infection by C3H/HeN mice that had or had not been co-treated with selected LAB.
  • FIGS. 18A-18B show the distribution of S. enteritidis (Log 10 CFU/g) in caecum (18A[[b]]) and colon (18B) at 10 days post-infection of C3H/HeN mice that had or had not been co-treated with selected LAB.
  • FIGS. 19A-19C show the distribution of S. enteritidis (Log 10 CFU/g) in mesenteric lymph node (19A), liver (19B) and spleen (19C) at 10 days post-infection of C3H/HeN mice that had or had not been co-treated with selected LAB.
  • FIG. 20 shows the performance of pigs fed GGDK266 and GGDK31 versus a control (daily weight gain, DWG, in g/day) for days 0-7, 7-14 and 0-14.
  • FIG. 21 shows microbial diversity analysis using denaturing gel gradient electrophoresis (DGGE; Trial 1). DGGE using universal primers revealed no differences in overall microbial diversity between the treatments and placebo. Bands on the gel are visualised by silver staining.
  • FIG. 22 shows microbial diversity analysis using DGGE.
  • DGGE using lactic acid bacteria (LAB) specific primers revealed significant differences in LAB diversity between treatment with GGDK266 and placebo in both caecal and ileal samples. Bands on the gel are visualised by silver staining.
  • LAB lactic acid bacteria
  • FIG. 23 shows microbial diversity analysis using DGGE.
  • DGGE using lactic acid bacteria (LAB) specific primers revealed significant differences in LAB diversity between treatment with GGDK266 and placebo in ileal samples. Bands on the gel are visualised by silver staining.
  • LAB lactic acid bacteria
  • FIG. 24 shows microbial diversity analysis using DGGE.
  • DGGE using lactic acid bacteria (LAB) specific primers revealed significant differences in LAB diversity between treatment with GGDK266 and placebo in caecal samples. Bands on the gel are visualised by silver staining.
  • LAB lactic acid bacteria
  • FIG. 25 shows the gene ontology biological processes significantly down-regulated by oral administration of GGDK266.
  • FIG. 26 shows changes in immune response and response to stimuli in animals treated with GGDK266 versus animals treated with placebo (percent of genes versus a range of different GO annotations).
  • FIG. 27 shows the gene ontology biological processes significantly enriched by oral administration of GGDK266.
  • Pig faeces samples collected during the course of the study of outdoor- and indoor-reared pigs were used in these studies.
  • the culture collection was based primarily on LAB collected from frozen samples 411, 412 and 416, which were from outdoor-reared pigs with particularly high levels of LAB in their faeces.
  • MRS broth premix, agar and vancomycin, anaerobe gas packs and indicator and antibiotic discs were purchased from Oxoid, anaerobe catalyst from Fisher Scientific and cysteine-HCL, bromocresol green and skimmed milk powder from Sigma-Aldrich.
  • Pig colostrum carbohydrate fractions were prepared as part of the SMART 163 programme of D. Kelly.
  • DNA extraction kits were purchased from MP Biomedicals and PCR reagents and clean-up kits from Promega. API CH 50 kits were purchased from Biomerieux UK Ltd.
  • Standard media MRS broth and MRS agar were prepared according to the manufacturer's instructions.
  • LAMVAB agar was prepared according to the method of Jackson et al. (2002).
  • Agar plates were prepared immediately before use.
  • MRS broth was decanted (10 ml per tube) into sterile Hungate tubes under anaerobic conditions and stored at room temperature.
  • Carbohydrate-supplemented media SMART 163 ammonium sulphate precipitate of pig colostrum: precipitated at 0, 20, 25, 30, 35, 45, 50, 55 or 65% saturation or soluble at 65% saturation were weighed out in proportion to the amounts recovered from 15 ml or 50 ml of colostrum.
  • Carbohydrate fractions were each dispersed in 15 ml of MRS or LAMVAB agar, held at 45° C., and then individual plates were poured for each fraction. They were also dispersed in MRS broth (50 ml) and the supplemented broth decanted to eight (6 ml/tube) sterile Hungate tubes under anaerobic conditions.
  • mice Female C3H/HeN and C57BI/6 mice (5-6 weeks old) were purchased from Harlan UK. They were housed as groups or pairs in standard caging within HEPA-filtered flexifilm isolators situated in a class 2 containment facility. They had free access to a high quality rodent chow and sterile deionised water at all times and were allowed to acclimatise for 7 to 10 days prior to commencement of experiments.
  • the Rowett Institute of Nutrition and Health (RINH) is licensed under the UK Animals (Scientific Procedures) Act 1986.
  • Frozen stock An aliquot (0.7 ml) of each culture was drawn off with a sterile syringe and needle and dispensed into a plastic tube that was flushed with CO 2 and contained 0.3 ml glycerol and 2 mg L-cysteine. The tube was sealed with a plastic stopper, labelled, the contents mixed, frozen and stored at ⁇ 80° C.
  • Conditioned medium The remaining culture was transferred to a Corning 15 ml centrifuge tube, centrifuged at 1000 g ⁇ 5 min at room temperature, the supernatant decanted, aliquoted and frozen. The pellets were either extracted immediately for 16S rRNA gene analysis or frozen.
  • 16S rRNA gene analysis Bacterial DNA was extracted using a FastDNA® Spin kit for Soil in conjunction with a Fastprep 120 bead beater system, according to the protocol supplied with the kit. PCR was carried out (reaction mix: buffer, 104 dNTPs (2 mM), 5 ⁇ l. 27F Primer (20 pmol/2 ⁇ l. 1492R Primer (20 pmol/ ⁇ l)). 2 ⁇ l Go Taq Flexi Polymerase, 0.5 ⁇ l). MgCl2, 5 ⁇ l. H2O, 23.5 ⁇ l and 2 ⁇ l of extracted DNA) using MJ Research PTC-200 Peltier Thermal Cycler run through 35 cycles of 95° C. for 3 minutes, 95° C.
  • PCR product cleanup was done with a Wizard® SV Gel and PCR Clean-up kit (Promega), used according to the manufacturer's instructions.
  • 16S PCR products were sequenced using fully automated genetic analysers based on capillary electrophoresis technology (Genomics Section, RINH, UoA) using the reverse and forward primers 519R and 926F. Bacterial strains were identified by comparison of sequences with known bacterial DNA sequences using BLAST (http://blast.ncbi.nlm.nih.gov/Blast.cgi).
  • XLD agar was prepared as per manufacturer's instructions and cooled to 45° C. Salmonella enteritidis S1400 was added to the XLD agar [1 ml of a 1:1000 dilution of an overnight culture of salmonella in 200 ml XLD agar to give the equivalent of 106 CFU/ml]. The agar was poured into petri dishes and allowed to set. The plates were marked off into 4 quadrants and an approximately 5 mm well cut out in each quadrant. An aliquot (60 ⁇ l) of conditioned media or MRS broth was added to the wells. The plates were covered and incubated for 16 hours at 37° C. They were photographed using a digital camera. Images transferred to Photoshop, and the diameter of the well and zone of inhibition were determined using the measure tool. Values were calculated and stored on an Excel spreadsheet. The same procedure was used with Escherichia coli K88, except that MacConkey No 3 agar was used.
  • Pig LAB [0.5 ml of a 1:100 dilution of an overnight culture] was spread onto the surface of an MRS agar [90 mm] plate and dried off. The plates were marked off into 4 quadrants and in each quadrant was placed an antibiotic-containing disc [Ampicillin, 10 ⁇ g. Cefotaxime, 30 ⁇ g. Chloramphenicol, 10 ⁇ g. Erythromycin, 15 Gentamicin, 10 ⁇ g Kanamycin, 30 ⁇ g. Metronizadole, 50 ⁇ g. Nalidixic acid, 30 ⁇ g. Tetracycline, 30 ⁇ g. Vancomycin, 30 ⁇ g].
  • antibiotic-containing disc [Ampicillin, 10 ⁇ g. Cefotaxime, 30 ⁇ g. Chloramphenicol, 10 ⁇ g. Erythromycin, 15 Gentamicin, 10 ⁇ g Kanamycin, 30 ⁇ g. Metronizadole, 50 ⁇ g. Nalidixic acid, 30 ⁇ g. Tetracycl
  • the plates were covered, placed in an anaerobic jar and incubated for 24 hours at 37° C. They were photographed using a digital camera. Images transferred to Photoshop, and the diameter of the zone of inhibition was determined using the measure tool. Values were calculated and stored on an Excel spreadsheet.
  • IPEC-J2 cells Monolayers of IPEC-J2 cells were grown to 3 days post-confluence in 24-well plates and synchronised by the addition of DTS media 24 hrs prior to use. Overnight cultures of pig LAB (10 ml) were centrifuged [1000 g ⁇ 5 min at room temperature] and the bacteria re-suspended in 1 ml of phosphate buffered saline [PBS]. An aliquot (50 ⁇ l) of LAB was added to the wells. The plates were incubated for 2 hours at 37° C., 5% CO 2 , 95% humidity. An overnight culture of Salmonella enterica serovar Enteritidis S1400 [ S.
  • enteritidis S1400 was sub-cultured (0.5 ml in 10 ml) into Luria Bertani (LB) media and incubated aerobically for 2-3 hours at 37° C. until it reached an optical density (560 nm) of 0.8. This gave a concentration equivalent to 1 ⁇ 108 CFU/ml.
  • the culture was centrifuged [1000 g ⁇ 5 min at room temperature], the bacteria re-suspended in 10 ml of PBS. An aliquot (50 ⁇ l) was added to the wells of IPEC-J2 cells. Wells treated with PBS were used as controls. The plates were incubated for a further 2 hours at 37° C., 5% CO 2 , 95% humidity.
  • IPEC-J2 cell monolayers were washed 5 times with HBSS. A solution (0.5 ml) of PBS containing Triton-X100 (10 ml/litre) was added to each well, the monolayer scraped off and dispersed. Viable salmonella were estimated on XLD agar plates [incubated for 24 hours at 37° C.] by the Miles and Misra method [Robertson et al, 2003]. LAB were determined by the same procedure [incubated anaerobically for 48 hours at 37° C.].
  • IPEC-J2 cells Monolayers of IPEC-J2 cells were grown to 3 days post-confluence in 24-well plates and synchronised by the addition of DTS media 24 hrs prior to use. Overnight cultures of pig LAB (10 ml) were centrifuged [1000 g ⁇ 5 min at room temperature] and the bacteria re-suspended in 1 ml of PBS. An aliquot (50 ⁇ l) of LAB was added to each well [3 wells for each sample] along with 220 ng 12-O-Tetradecaboylphorbol-13-acetate [PMA] per well. PMA or PBS alone served as controls. The plates were incubated for 2 hours at 37° C., 5% CO 2 , 95% humidity.
  • RNA extraction was done using RNeasy® Mini kit in accordance with the manufacturer's protocols and reverse transcription with a high capacity cDNA Reverse Transcription Kit (Applied Biosystems). Real Time PCR was done on a 7500 Fast Real-time PCR system operating with 7500 Fast System v 1.4.0 Sequence Detection Software version 1.4 (Applied Biosystem).
  • IL-8 and TNF- ⁇ genes were analysed and compared to that of the ‘house-keeping’ gene ⁇ -actin. For comparison, values were given as the ratio of IL-8 and TNF- ⁇ per ⁇ -actin or fold-change.
  • Substrate reactivity The carbohydrate reactivity of individual LAB was determined using an API CH 50 kit (Biomerieux UK Ltd). Assays were done according to the manufacturer's instructions and reactions were recorded after incubation for 24 and 48 hours at 37° C. There are 50 capsules on an API CH 50 plate. These contain various potential substrates and negative controls. The range of substrates is as follows: Monosaccharides 16, Monosaccharides/alcohols 4, Disaccharides 8, Trisaccharides 2, Polysaccharides 3, Alcohols 6, Others 7. For each substrate group the number of positive reactions is counted. This is divided by the maximum possible to give the rank for that substrate group. The sum of all the substrate scores gives the overall ranking for the bacterium. High Ranking indicates broad spectrum of substrate reactivity
  • Heat-treatment of LAB A small amount of frozen faeces (100 mg) was dispersed in 5 ml of maximum recovery diluent (MRD). Sediment was allowed to settle out and the upper layer was decanted into eppendorf tubes (1 ml/tube). The tubes were heated at 50° C., 60° C. or 70° C. for 10 min. An aliquot (0.4 ml) of each was plated out on MRS agar and incubated in an anaerobic jar for 72 hours at 37° C. A small number of colonies were detected after heating at 70° C. Distinct colonies were picked off, seeded into Hungate tubes containing MRS broth and incubated for 48 hours at 37° C.
  • MRD maximum recovery diluent
  • Stability of freeze dried bacteria Overnight cultures of LAB were centrifuged (1000 g ⁇ 5 min at room temperature. Pellets were re-suspended in 2 ml sterile PBS and re-centrifuged. The subsequent pellets were then re-suspended in 5 ml of freezing solution [defatted skimmed milk powder (SKP), 100 g/1; SKP+lactose, both 100 g/1; SKP+sucrose, both 100 g/1; or SKP, 200 g/1]. The samples were frozen at ⁇ 20° C. (2-3 hours) and then stored at ⁇ 80° C. overnight. They were freeze-dried for 48 hours and dried material stored at room temperature. Viable bacteria in the samples were determined at 0 and approximately 40 and 80 days after completion of freeze drying. They were plated out on MRS agar and incubated anaerobically for 48 hours at 37° C.
  • the supernatant was discarded, tubes refilled with culture and re-centrifuged until all the bacteria had been recovered.
  • Each of the 6 tubes contained almost equal amounts of bacteria.
  • the bacteria in each tube were re-suspended in 40 ml of sterile PBS, re-centrifuged and the supernatant discarded.
  • the bacteria in each tube was re-suspended in 20 ml of SKM (100 g/1), frozen at ⁇ 20° C. (2-3 hours) and then overnight at ⁇ 80° C., freeze-dried for 48-72 hours and stored at 4° C.
  • one tube of freeze dried material was re-suspended in 20 ml of MRS broth, incubated at room temperature for 2 hours, diluted, plated out on MRS agar and incubated anaerobically for 48 hours at 37° C.
  • L. mucosae in vivo Study 1 Sixteen (6 week) old female C3H/HeN mice were dosed with an overnight culture of vial 323 ( L. mucosae; 50 ⁇ l; >109 CFU) at day ⁇ 7, ⁇ 4, ⁇ 2 and 0 and daily thereafter up to day +9. A further 16 mice (control) were given media. On day 0, eight mice (L. mucosae-treated) and eight control mice were given, by gavage, a single dose of Salmonella enteritidis S1400 (50 ⁇ l; ⁇ 108 CFU). In addition, eight mice ( L. mucosae -treated) and eight control mice were given a single dose of culture medium. Body weight and health score were monitored twice daily post- salmonella infection.
  • mice were euthanased (isoflurane overdose and exsanguination) and dissected at 10 days post- salmonella infection.
  • Stomach representative portions of jejunum and ileum, caecum plus contents, colon plus contents, spleen and liver and one kidney and the mesenteric lymph node were collected under near aseptic conditions for microbiology.
  • Representative portions of upper jejunum, mid jejunum, ileum, caecum and ascending and descending colon were placed in neutral buffered formalin or RNA-later and stored for future analysis.
  • L. mucosae in vivo Study 2 Five (6 week) old female C57BI/6 mice were dosed with an overnight culture of vial 323 ( L. mucosae; 50 ⁇ l; >109 CFU) at day ⁇ 7, ⁇ 4, ⁇ 2 and 0 and daily thereafter up to day+5. A further 5 mice were given media. On day 0, all ten mice were given, by gavage, a single dose of Salmonella enteritidis S1400 (50 ⁇ l; >107CFU). The mice were euthanased and dissected on day 6, according to the procedure for study 1.
  • Novel pig LAB in vivo Four (6 week) old female C3H/HeN mice were dosed with an overnight culture of RINH vial 31 ( L. reuteri; 50 ⁇ l; >109 CFU), four with RINH vial 32 ( L. reuteri ). Four with vial 323 ( L. mucosae ), four with RINH vial 46 ( L. reuteri ), four with RINH vial 47 ( L. reuteri ) and eight with MRS. This was done at day ⁇ 6, ⁇ 4, ⁇ 2 and 0 and daily thereafter up to day+9.
  • mice On day 0, all lactobacilli-treated mice and four control mice were given, by gavage, a single dose of Salmonella enteritidis S1400 (50 ⁇ l; ⁇ 108 CFU). In addition, the remaining four control mice were given a single dose of culture medium. The mice were euthanased and dissected on day 10, according to the procedure for study 1.
  • Tissues were homogenised [1:100 w/v] in MRD using a Janke-Kunkel Ultra-Turrax T25 tissue homogeniser at 20,000 rpm for 30 seconds, as were jejunal and ileal contents. Up to eight sequential dilutions (1: 10 v/v) of the primary homogenates were made, plated out onto XLD agar and MacConkey No. 3 agar and incubated overnight at 37° C. Viable counts were estimated as before [Robertson et al, 2003].
  • L. johnsonii and L. johnsonii -related strains [ L. johnsonii, L. johnsonii /gasseri, L. johnsonii/taiwanensis ] (240/436) and L. reuteri or L. reuteri -related [ L. reuteri, L. reuteri/pontis, L. reuteri/vaginalis, L. reuteri/acidophilus (169/436)].
  • L. reuteri, L. reuteri/pontis, L. reuteri/vaginalis, L. reuteri/acidophilus (169/436)
  • There were 7 L. plantarum/pentosus colonies 19 other species and 5 uncultured strains.
  • Conditioned media from isolated LAB were screened for anti-bacterial activity against Salmonella enteritidis S1400 using a well-diffusion assay ( FIG. 1 ).
  • Group 1 comprised of 14 strains (3.4% of total), Group 2 of 95 strains (22.8%), Group 3 of 99 strains (23.7%), Group 4 of 99 strains (23.7%), Group 5 of 86 strains (20.6%) and Group 6 of 24 strains (5.8%).
  • the latter group comprised of seventeen L. johnsonii and L. johnsonii -related, six L. reuteri or L. reuteri -related strains and one uncultured strain.
  • Conditioned media from LAB were also screened for anti- Escherichia coli K88 activity by the well diffusion assay.
  • Activity against E. coli K88, as with salmonella varied greatly between individual colonies of LAB ( FIG. 3A ). The range and variation in the activity was similar amongst the L. johnsonii and L. reuteri strains. In general, there was no direct correlation between the anti- salmonella and anti E. coli K88 activities for any individual LAB ( FIG. 3C, 3D ). However of the ten strains in E. coli K88 group 5 ( FIG. 3B ), seven had relatively high activities against both pathogens, two had high activity against E. coli K88 but moderate activity against salmonella and one was active primarily against E. coli K88.
  • the candidate LAB were screened for substrate reactivity using an API CH 50 kit (Table 5, 6, FIG. 6 ).
  • L. johnsonii, L. reuteri and L. plantarum each exhibited strain-specific general substrate reaction profiles.
  • most strains of each genotype exhibited fine differences in their substrate reactivity, indicative that they were unique individual strains.
  • FIGS. 7A-7C The ability of candidate LAB to block or suppress inflammatory responses triggered in IPEC cells by 12-O-Tetradecaboylphorbol-13-acetate [PMA] was tested ( FIGS. 7A-7C ; Table 7).
  • the candidate strains varied greatly in their capacity to block interleukin-8 (IL-8) gene-expression triggered by PMA.
  • Five strains (RINH vial 29, 30, 31 86 and 266) had potent anti-inflammatory effects.
  • FIGS. 8A and 8B The survival and viability of LAB after freeze drying in skimmed milk powder [SKP], SKP plus lactose or SKP plus sucrose was evaluated ( FIGS. 8A and 8B ).
  • FIG. 8A depicts the stability of L. reuteri
  • FIG. 8B depicts the stability of L. johnsonii . Small losses in viability were evident on storage for 42 and 84 days at room temperature of samples dried in SKP. This was less marked when skimmed milk powder and sugars were used in combination. However, the 24 latter preparations tended to be hygroscopic and difficult to maintain. Bulk preparations of GGDK266 and GGDK31 were therefore prepared by drying the bacteria in skimmed milk powder [100 g/1] (Table 8).
  • Suspensions of faeces from organically reared pigs were heat treated for varying periods of time at 50-70° C., plated out on MRS agar, colonies picked off and cultured in MRS broth [RINH vial 417-506].
  • the strain types recovered were variable and clostridium species formed a high proportion, the isolated strains remained sensitive to heat.
  • Isolated cultures of LAB were subject to heating three times for 15 minutes at 70° C. ( FIGS. 9A-9C ).
  • Viable bacteria decreased by 3-4 log orders after heat-treatment for the first time. However, the surviving bacteria had a degree of heat-resistance. With one exception, losses of viable bacteria were low when the bacteria were re-cultured and re-heated a further two times.
  • C3H/HeN mice develop a persistent but non-lethal, intestinal and systemic infection, which has many characteristics of the major form of human salmonellosis , when challenged with high levels of Salmonella enteritidis S1400.
  • C57BI/6 mice develop a severe primarily systemic, infection, reminiscent of acute infection in humans, when challenged with the same pathogen.
  • L. mucosae vial 323
  • C3H/HeN and C57BI/6 mice were treated with L. mucosae prior to and post-challenge with Salmonella enteritidis ( FIGS. 10, 13 ). The mice were euthanased and dissected 6 (C57BI/6) or 10 (C3H/HeN) days post-infection.
  • Intestinal myeloperoxidase [MPO] a marker for neutrophils, was determined in C3H/HeN mice treated with salmonella or salmonella plus RINH vial 323 ( L. mucosae ). MPO in the intestine was greatly increased by salmonella infection, due to recruitment of neutrophils to the intestine part of the host response to infection ( FIG. 12B [[ 12 b ]]), Co-treatment with RINH vial 323 ( L. mucosae ) reduced MPO activity in the intestine of salmonella -infected mice, indicating that the intestinal inflammatory responses to infection were lowered in these animals.
  • RINH vial 31, RINH vial 32, RINH vial 46 and RINH vial 47 All L. reuteri ; LR31, LR 32, LR 36 and LR47 respectively.
  • C3H/HeN mice were treated with these LAB or RINH vial 323 ( L. mucosae , LM] prior to and post-challenge with Salmonella enteritidis ( FIG. 16 ).
  • the mice were euthanased and dissected 10 days post-infection. Faecal excretion of S. enteritidis was reduced, if the mice had been co-treated with LAB ( FIG. 17A, 17B ).
  • LR31 and LR32 tended to have the greatest effects on faecal salmonella outputs.
  • Intestine Treatment with LR31, LR32, LM, LR46 or LR47 significantly reduced the numbers of salmonella in the caecum ( FIG. 18A ). Furthermore, LR31, LR32, LR46 and LR47 but not LM lowered salmonella numbers in the colon ( FIG. 18B ). The reductions tended to be greater with LR31 and LR32. In contrast to the large intestine, the LAB had no significant effects on numbers of salmonella in the small intestine.
  • the LAB strains isolated (total of 436 individual colony picks) from faeces of organically-reared pigs were predominantly L. reuteri, L. johnsonii, L. gasseri, L. pentosus , strains with a small number of L. plantarum, L. acidophilus, L. vaginalis , a single L. mucosae and several uncultured strains.
  • Most of the LAB produced substances that could inhibit the growth of S. enteritidis and/or E. coli K88 in vitro.
  • the potency of these anti-pathogen effects varied greatly between the individual bacterial strains.
  • a proportion of LAB had high activity against S. enteritidis but low activity against E. coli K88 and vice-versa, but the majority had similar activities against both pathogens.
  • IPEC intestinal pig epithelial cells
  • Heat stability is a useful feature for LAB to be used in pelleted animal foods.
  • Five heat-conditioned viable strains of isolated pig LAB were obtained.
  • the biological properties in vitro and probiotic potential of three of the strains were adversely affected by heat-treatment. Nonetheless, two of the bacteria retained the biological properties of their native non-heat-treated forms.
  • mice with these LAB greatly reduced the pathogenicity of S. enteritidis.
  • DNA is extracted from faecal or tissue samples utilizing the MP Bio FastDNATM spin kit for soil sample—116560000.
  • the DNA is then amplified using Muyzer primers, as it is essential to use primers with a GC Clamp to be run on the gel.
  • Muyzer primers as it is essential to use primers with a GC Clamp to be run on the gel.
  • specialised lactobacillus primers with a GC clamp were used.
  • the GC clam is as follows:
  • DGGE is a genetic analysis technique in which amplified PCR products are separated by the denaturants formamide and urea within the gel, based on the genetic sequence by as little as a single base difference. DGGE can be utilised to visualise the differences in microbial diversity between samples. DNA obtained from a range of samples can be used in DGGE e.g. tissue and faecal samples. Bands on the gel were visualised using silver staining.
  • tissue sample approximately 200 mg tissue sample was removed from RNAlater (Ambion) and lyzed in Trizol (Invitrogen) using a polytron homogenizer. The tissue was further homogenized by passing the lysate through a syringe fitted with a 19G needle 3-5 times. The samples were incubated for 5 min at RT to permit the complete dissociation of nucleoprotein complexes. Then, chloroform, isopropanol and ethanol steps were performed according to the manufacturer's instructions.
  • the tubes were shaken vigorously by hand for 10s, incubated at 4° C. for 10 min and centrifuged at 12,000 ⁇ g for 10 min at 4° C.
  • RNA precipitate was washed with ice-cold 75% ethanol, adding at least 1 mL of 75% ethanol per 1 mL of Trizol. The samples were vortexed and centrifuged at 7,400 ⁇ g for 5 min at 4° C. After air-drying the resultant RNA pellet, the RNA was resuspended in up to 100 ⁇ L RNase-free water. Total RNA was further extracted with the RNeasy kit (Qiagen) according to the manufacturer's instructions, including an RNase-free DNase I (Qiagen) digestion step.
  • RNeasy kit Qiagen
  • Cultured cells were homogenized by adding 350 ⁇ L Buffer RLT+1% ⁇ -mercaptoethanol. The cells were scraped off culture dishes with a filter tip and further homogenized by passing the lysate through a syringe fitted with a 19G needle 3-5 times. The cell lysate was then further processed using the RNeasy kit (Qiagen) according to the manufacturer's instructions, including an RNase-free DNase I (Qiagen) digestion step.
  • RNeasy kit Qiagen
  • RNA concentration and integrity was ascertained using a Nanodrop instrument and/or Agilent Bioanalyzer, and purified RNA was stored at ⁇ 70° C.
  • RNA was processed for Affymetrix GeneChips using the GeneChip 3′ IVT Express Kit (Affymetrix) according to the manufacturer's instructions.
  • aRNA quality was determined by Agilent 2100 Bioanalyzer. Hybridization to the GeneChip Mouse Genome 430 2.0 and GeneChip Human Genome U133 Plus 2.0 (Affymetrix) on a GeneChip Fluidics Station 450 (Affymetrix) was performed at the Institute of Medical Sciences Microarray Core Facility (University of Aberdeen, UK). Chips were scanned with an Affymetrix GeneChip Scanner 3000 (Affymetrix). Image quality analysis was performed using Gene Chip Operating Software (GCOS) (Affymetrix).
  • GCOS Gene Chip Operating Software
  • Piglets fed GGDK266 exhibited significantly improved daily weight gain (DWG) during the first week post-weaning relative to GGDK31 and placebo fed piglets.
  • DWG daily weight gain
  • DGGE using lactic acid bacteria (LAB) specific primers revealed significant differences in LAB diversity between treatment with GGDK 266 and placebo in both caecal and ileal samples (see FIG. 22 ).
  • DGGE using LAB specific primers revealed significant differences in LAB diversity between the treatment with GGDK266 and placebo in ileal samples (see FIG. 23 ).
  • DGGE using LAB specific primers revealed significant differences in LAB diversity between the treatment with 266 and placebo in caecal samples (see FIG. 24 ).
  • Results reveal that GGDK266 had a very specific and targeted effect on the immune system and the functional groups associated with response to stimuli (see FIG. 26 ).
  • GGDK266 promoted metabolic processes particularly in relation to nitrogen (see FIG. 27 ). Without wishing to be bound by theory, it is believed that these effects may explain the improved DWG in animals fed GGDK266.
  • Gene expression data revealed that a number of genes were significantly increased including antimicrobial peptides (eg. CSTA, BP1) and immune-regulatory genes (TIP).
  • GGDK266 reduced the expression of a diverse panel of genes involved in pro-inflammatory immunity (IFITM3, IL-16).
  • Lactobacillus plantarum Lactobacillus plantarum , pentosus , paraplantarum pentosus 12 Lactobacillus johnsonii , Lactobacillus johnsonii , gasseri , taiwanensis gasseri 16 Lactobacillus johnsonii , Lactobacillus johnsonii gasseri , taiwanensis 29 Lactobacillus reuteri , Lactobacillus reuteri pontis , vaginalis , frumenti 31 Lactobacillus reuteri Lactobacillus reuteri 86 Lactobacillus reuteri Lactobacillus reuteri 230 Lactobacillus johnsonii , Lactobacillus johnsonii taiwanensis , acidophilus 256 Lactobacillus johnsonii , Lactobacillus johnsonii , Lactobacillus
  • Lactobacillus johnsonii Lactobacillus johnsonii gasseri , taiwanensis 30 Lactobacillus reuteri , Lactobacillus reuteri pontis 32 Lactobacillus reuteri Lactobacillus reuteri 258 Lactobacillus plantarum , Lactobacillus plantarum , pentosus , helveticus pentosus , paraplantarum 260 Lactobacillus plantarum , Lactobacillus pentosus , pentosus , paraplantarum plantarum , paraplantarum 320 lactobacillus johnsonii Lactobacillus johnsonii NCC2822 F19785 364 lactobacillus johnsonii lactobacillus johnsonii 466 F10785 433 lactobacillus johnsonii lactobacillus johnsoni str.
  • Lactobacillus plantarum Lactobacillus plantarum , pentosus pentosus 218 Lactobacillus johnsonii , uncultured Firmicutes, taiwanensis Lactobacillus johnsonii 220 Lactobacillus johnsonii , uncultured Firmicutes, taiwanensis Lactobacillus johnsonii 356 lactobacillus johnsonii lactobacillus johnsonii NCC2822 F19785 363 lactobacillus johnsonii lactobacillus johnsonii 466 F10785 131 Lactobacillus reuteri Lactobacillus reuteri 434 Lactobacillus reuteri lactobacillus reuteri NM99-1 166 Lactobacillus johnsonii , Lactobacillus johnsonii taiwanensis , acidophilus 431
  • Candidate LAB strains selected on the basis of killing activity, capacity to block adherence of pathogen to IPEC cells, antibiotic susceptibility, substrate reactivity and ability to suppress inflammation (note 266 and 161 contain LR) RINH forward reverse Vial no.
  • SEQ ID NO: 70 GTGCCTAATACATGCAAGTCGAACGAACTCTGGTATTGATTGGTGCTTGCATCATGATTTACATTTGAGTGAGTGGCGAACTGGTGAGTAAC ACGTGGGAAACCTGCCCAGAAGCGGGGGATAACACCTGGAAACAGATGCTAATACCGCATAACAACTTGGACCGCATGGTCCGAGTTTGAAA GATGGCTTCGGCTATCACTTTTGGATGGTCCCGCGGCGTATTAGCTAGATGGTGAGGTAACGGCTCACCATGGCAATGATACGTAGCCGACC TGAGAGGGTAATCGGCCACATTGGGACTGAGACACGGCCCAAACTCCTACGGGAGGCAGCAGTAGGGAATCTTCCACAATGGACGAAAGTCT GATGGAGCAACGCCGCGTGAGTGAAGAAGGGTTTCGGCTCGTAAAACTCTGTTGTTAAAGAAGAACATATCTGAGAGTAACTGTTCAGGTAT TGACGGTATTTAACCAGAAAGCCACGGCTAACTAC

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Microbiology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Zoology (AREA)
  • Biotechnology (AREA)
  • Wood Science & Technology (AREA)
  • Genetics & Genomics (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Biochemistry (AREA)
  • Biomedical Technology (AREA)
  • Mycology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Polymers & Plastics (AREA)
  • Virology (AREA)
  • General Engineering & Computer Science (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Molecular Biology (AREA)
  • Food Science & Technology (AREA)
  • Epidemiology (AREA)
  • Nutrition Science (AREA)
  • Animal Husbandry (AREA)
  • Physiology (AREA)
  • Oncology (AREA)
  • Communicable Diseases (AREA)
  • Pain & Pain Management (AREA)
  • Rheumatology (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

A first aspect of the invention relates to a porcine lactic acid bacterial strain, wherein said bacterial strain is characterised by one or more of the following characteristics: (i) the ability to exhibit antimicrobial activity against E. coli; (ii) the ability to exhibit antimicrobial activity against S. enteritidis; (iii) the ability to suppress inflammation in IPEC cells induced by 12-O-tetradecaboylphorbol-13-acetate (PMA); (iv) the ability to block the attachment or invasion of IPEC cells by S. enteritidis; (v) the ability to block the attachment or invasion of IPEC cells by E. coli; (vi) the absence of antibiotic resistance to one or more antibiotics selected from the following: ampicillin; cefotaxime; chloramphenicol; erythromycin; gentamicin; tetracycline; vancomycin; metronizadole; nalidixic acid; and kanamycin; and (vii) the ability to exhibit heat stability when subjected to three cycles of heating, each cycle comprising heating at a temperature of 70° C. for a period of 15 minutes. Further aspects of the invention relate to compositions comprising said bacterial strains, and therapeutic uses of said bacterial strains.

Description

    CROSS REFERENCE
  • This application is a continuation of U.S. application Ser. No. 16/206,250, filed Nov. 30, 2018, now U.S. Pat. No. 11,013,773, issued May 25, 2021, which is a continuation of U.S. application Ser. No. 15/359,144, filed on Nov. 22, 2016, now U.S. Pat. No. 10,183,046, issued Jan. 22, 2019, which is a division of U.S. application Ser. No. 14/232,475, filed Oct. 17, 2014, now U.S. Pat. No. 9,539,293, issued Jan. 10, 2017, which is a national stage entry of PCT/GB2012/051686, filed Jul. 13, 2012, which claims the benefit of Great Britain Patent Application No. 1112091.2, filed Jul. 14, 2011, the entire contents of which are all incorporated herein by reference.
  • SEQUENCE LISTING
  • The instant application contains a Sequence Listing which has been submitted electronically in ASCII format and is hereby incorporated by reference in its entirety. Said ASCII copy, created on Apr. 16, 2021, is named 56708_704_302 SL.txt and is 83,040 bytes in size.
  • The present invention relates to bacterial strains isolated from pigs. More specifically, the invention relates to the isolation of lactic acid bacteria from organically-reared pigs. The claimed lactic acid bacteria have useful probiotic and therapeutic applications.
  • BACKGROUND TO THE INVENTION
  • The composition of the microbial flora of pigs, their gut innate immune function and possible susceptibility to infection is greatly influenced by the environment in which they were reared during early life (Mulder et al, 2009). Outdoor-reared pigs generally have a more developed gut immune system, perform better and are healthier than indoor-reared counterparts. The outdoor environment dramatically influences microbial diversity of the gut and is associated with high levels of Firmicutes, in particular Lactic Acid Bacteria [LAB].
  • LAB comprise a clade of gram-positive, low-GC, acid-tolerant, generally non-sporulating, non-respiring bacteria that are associated with certain common metabolic and physiological characteristics. LAB are rod-shaped bacilli or coccus that are characterized by an increased tolerance to a lower pH range. LAB produce lactic acid as the major metabolic end-product of carbohydrate fermentation and are amongst the most important groups of microorganisms used in the food industry.
  • Lactobacilli are predominant in the gut flora of organically (outdoor) reared pigs. In contrast, the numbers of these bacteria are low in indoor-reared pigs and levels of potentially pathogenic phylotypes are high (Mulder et al, 2009). Furthermore, gut immune development and function of indoor-reared pigs is known to deviate from normal. In particular, expression of Type 1 interferon genes, Major Histocompatibility Complex class I and several chemokines are known to be increased (Mulder et al, 2009).
  • Lactic acid bacteria may modify the flora and gut structure and function in several ways (Cotter et al, 2005; Ohashi and Ushida, 2009). For example, they may compete with harmful bacteria for key nutrients or attachment sites on the gut, resulting in their exclusion. Alternatively, they can produce bioactive substances that aid or promote colonisation by beneficial bacteria or kill/interfere with the growth of potentially harmful or pathogenic bacteria. Alternatively, these bioactive factors can be immune-modulators that promote immune development and barrier integrity of the gut. Strains of LAB vary greatly in their biological activity. The present invention seeks to provide LAB strains that have therapeutically useful properties. More specifically, the invention seeks to provide LAB strains that are capable of promoting gut and immune development and health, thereby having considerable therapeutic potential as probiotics.
  • STATEMENT OF INVENTION
  • The present applicant has shown that the microbiota of out-door reared pigs contain LAB strains that produce potent and specific anti-microbial or cell-/immune-modulating bioactive factors.
  • Aspects of the invention, together with preferred embodiments, are set forth in the accompanying claims.
  • A first aspect of the invention relates to a porcine lactic acid bacterial strain, wherein said bacterial strain is characterised by one or more of the following characteristics:
      • (i) the ability to exhibit antimicrobial activity against E. coli;
      • (ii) the ability to exhibit antimicrobial activity against S. enteritidis;
      • (iii) the ability to suppress inflammation in IPEC cells induced by 12-O-tetradecaboylphorbol-13-acetate (PMA);
      • (iv) the ability to block the attachment or invasion of IPEC cells by S. enteritidis;
      • (v) the ability to block the attachment or invasion of IPEC cells by E. coli;
      • (vi) the absence of antibiotic resistance to one or more antibiotics selected from the following: ampicillin; cefotaxime; chloramphenicol; erythromycin; gentamicin; tetracycline; vancomycin; metronizadole; nalidixic acid; and kanamycin; and
      • (vii) the ability to exhibit heat stability when subjected to three cycles of heating, each cycle comprising heating at a temperature of 70° C. for a period of 15 minutes.
  • A second aspect relates to a composition comprising one or more lactic acid bacterial strains according to the invention and a pharmaceutically acceptable excipient, carrier or diluent.
  • A third aspect relates to a probiotic composition comprising one or more lactic acid bacterial strains according to the invention.
  • A fourth aspect relates to one or more lactic acid bacterial strains according to the invention for use in medicine.
  • A fifth aspect relates to one or more lactic acid bacterial strains according to the invention for use in treating an intestinal disorder in a subject.
  • A sixth aspect relates to the use of one or more lactic acid bacterial strains according to the invention in the preparation of a medicament for treating an intestinal disorder in a subject.
  • A seventh aspect relates to a method of treating an intestinal disorder in a subject, said method comprising administering to the subject a pharmaceutically effective amount of one or more lactic acid bacterial strains or composition according to the invention.
  • An eighth aspect of the invention relates to one or more lactic acid bacterial strains according to the invention for improving intestinal microbiota.
  • A ninth aspect of the invention relates to a method of improving intestinal microbiota in a subject, said method comprising administering to the subject one or more lactic acid bacterial strains or composition according to the invention.
  • A tenth aspect relates to a feedstuff comprising one or more lactic acid bacterial strains according to the invention.
  • An eleventh aspect relates to a food product comprising one or more lactic acid bacterial strains according to the invention.
  • A twelfth aspect relates to a dietary supplement comprising one or more lactic acid bacterial strains according to the invention.
  • A thirteenth aspect relates to a food additive comprising one or more lactic acid bacterial strains according to the invention.
  • A fourteenth aspect relates to a process for producing a probiotic, said process comprising culturing a lactic acid bacterial strain according to the invention.
  • A fifteenth aspect of the invention relates to a process for obtaining a porcine lactic acid bacterial strain, said process comprising obtaining faeces from an organically reared pig and extracting one or more porcine lactic acid bacterial strains from said faeces.
  • A sixteenth aspect of the invention relates to one or more porcine lactic acid bacterial strains obtained by, or obtainable by, the process described above.
  • DETAILED DESCRIPTION OF THE INVENTION
  • As mentioned above, the present invention relates to one or more porcine lactic acid bacterial strains. The lactic acid bacterial strain is characterised by one or more of the following characteristics:
      • (i) the ability to exhibit antimicrobial activity against E. coli;
      • (ii) the ability to exhibit antimicrobial activity against S. enteritidis;
      • (iii) the ability to suppress inflammation in IPEC cells induced by 12-O-tetradecaboylphorbol-13-acetate (PMA);
      • (iv) the ability to block the attachment or invasion of IPEC cells by S. enteritidis;
      • (v) the ability to block the attachment or invasion of IPEC cells by E. coli;
      • (vi) the absence of antibiotic resistance to one or more antibiotics selected from the following: ampicillin; cefotaxime; chloramphenicol; erythromycin; gentamicin;
      • tetracycline; vancomycin; metronizadole; nalidixic acid; and kanamycin; and
      • (vii) the ability to exhibit heat stability when subjected to three cycles of heating, each cycle comprising heating at a temperature of 70° C. for a period of 15 minutes.
  • As used herein, the term “porcine” means “of or pertaining to swine”, i.e. of or pertaining to any of several mammals of the family Suidae, especially the domesticated hog, Sus scrofa domesticus, or Sus domesticus when young or of comparatively small size.
  • Preferably, the pig is less than 3 months old, preferably, less than 2 months old. Preferably, the porcine lactic acid bacterial strain is from an organically reared pig. In this regard, preferably, the pigs are reared free range, outside (with exposure to soil) and in the absence of antibiotics, growth promoters and/or growth enhancers.
  • Preferably, the porcine lactic acid bacterial strain is from an outdoor reared pig. Preferably, the pigs are reared outside for at least 60% of their lives. More preferably, the animals are reared outside for at least 80% of their lives, more preferably, at least 90% of their lives, even more preferably still, 100% of their lives.
  • In one preferred embodiment, the lactic acid bacterial strain is selected from L. johnsonii, L. reuteri, L. plantarum, L. gasseri, L. pentosus, L. acidophilus, L. vaginalis and L. mucosae.
  • In one preferred embodiment, the lactic acid bacterial strain is selected from L. johnsonii, L. reuteri and L. plantarum.
  • In another preferred embodiment, the lactic acid bacterial strain is in the form of a live bacterial population, a lyophilized bacterial population, a non-viable bacterial preparation, or the cellular components thereof. Preferably, where the bacterial strain is in the form of a non-viable bacterial preparation, it is selected from heat-killed bacteria, irradiated bacteria and lysed bacteria.
  • In one preferred embodiment, the lactic acid bacterial strain is in the form of a live bacterium, a dead bacterium, or the cellular components thereof.
  • In one preferred embodiment, the lactic acid bacterial strain is in isolated form. As used herein, the term “isolated” means isolated from its native environment.
  • In one preferred embodiment, the lactic acid bacterial strain is in biologically pure form. As used herein the term “biologically pure” refers to a bacterial strain in the form of a laboratory culture that is substantially free from other species of organism. Preferably, the lactic acid bacterial strain is in the form of a culture of a single species of organism.
  • As used herein, the term “lactic acid bacterial strain” also encompasses mutants of said lactic acid bacterial strain. As used herein, the term “mutant” includes derived bacterial strains having at least 93% homology, preferably at least 96% homology, more preferably 98% homology to the polynucleotide sequence of a referenced strain, but otherwise comprising mutations in other sequences in the bacterial genome. Mutants are obtainable by genetic engineering techniques inferring alteration of the genetic material of the strains of the invention or inferring a recombination of the genetic material of the strains of the invention with other molecules. Typically, in order to obtain such mutant strains, a person skilled in the art can use standard mutagenesis techniques such as UV radiation or exposure to mutagenic chemical products.
  • As used herein, the term “mutations” includes natural or induced mutations comprising at least single base alterations including deletions, insertions, transversions, and other modifications known to those skilled in the art, including genetic modification introduced into a parent nucleotide or amino acid sequence whilst maintaining at least 50% homology to the parent sequence. Preferably, the sequence comprising the mutation or mutations has at least 60%, more preferably at least 75%, more preferably still 85% homology with the parental sequence. As used herein, sequence “homology” can be determined using standard techniques known to those skilled in the art. For example, homology may be determined using the on-line homology algorithm “BLAST” program, publicly available at http)://www.ncbi.nlm.nih.gov/BLAST/.
  • As used herein, the term “lactic acid bacterial strain” also encompasses homologues of the lactic acid bacterial strains. As used herein the term “homologue” refers to a lactic acid bacterial strain having a nucleotide sequence having a degree of sequence identity or sequence homology with the nucleotide sequence of the parent lactic acid bacterial strain (hereinafter referred to as a “homologous sequence(s)”). Here, the term “homologous” means an entity having a certain homology with the subject nucleotide sequence. Here, the term “homology” can be equated with “identity”.
  • In the present context, a homologous sequence is taken to include a nucleotide sequence which may be at least 50, 60, 70, 75, 80, 85 or 90% identical, preferably at least 95%, 97%, 98% or 99% identical to the nucleotide sequence of the parent lactic acid bacterial strain (the subject sequence).
  • Homology comparisons can be conducted by eye, or more usually, with the aid of readily available sequence comparison programs. These commercially available computer programs can calculate % homology between two or more sequences. % homology may be calculated over contiguous sequences, i.e. one sequence is aligned with the other sequence and each amino acid in one sequence is directly compared with the corresponding amino acid in the other sequence, one residue at a time. This is called an “ungapped” alignment. Typically, such ungapped alignments are performed only over a relatively short number of residues.
  • Although this is a very simple and consistent method, it fails to take into consideration that, for example, in an otherwise identical pair of sequences, one insertion or deletion will cause the following amino acid residues to be put out of alignment, thus potentially resulting in a large reduction in % homology when a global alignment is performed.
  • Calculation of maximum % homology therefore firstly requires the production of an optimal alignment, taking into consideration gap penalties. A suitable computer program for carrying out such an alignment is the Vector NTI (Invitrogen Corp.). Examples of software that can perform sequence comparisons include, but are not limited to, the BLAST package (see Ausubel et al 1999 Short Protocols in Molecular Biology, 4th Ed—Chapter 18), BLAST 2 (see FEMS Microbiol Lett 1999 174(2): 247-50; FEMS Microbiol Lett 1999 177(1): 187-8), FASTA (Altschul et al 1990 J. Mol. Biol. 403-410) and AlignX for example. At least BLAST, BLAST 2 and FASTA are available for offline and online searching (see Ausubel et al 1999, pages 7-58 to 7-60).
  • Preferably, the degree of identity with regard to a nucleotide sequence is determined over at least 20 contiguous nucleotides, preferably over at least 30 contiguous nucleotides, preferably over at least 40 contiguous nucleotides, preferably over at least 50 contiguous nucleotides, preferably over at least 60 contiguous nucleotides, preferably over at least 100 contiguous nucleotides. Preferably, the degree of identity with regard to a nucleotide sequence may be determined over the whole sequence.
  • The traditional identification of bacteria on the basis of phenotypic characteristics is generally not as accurate as identification based on genotypic methods. Comparison of the bacterial 16S rRNA gene sequence has emerged as a preferred genetic technique and allows for new strains to be identified by comparison of sequences with known bacterial DNA sequences using BLAST (http://blast.ncbi.nlm.nih.gov/Blast.cgi). The 16S rRNA gene sequence is universal in bacteria, and so relationships can be measured across many different bacteria. In general, the comparison of the 16S rRNA sequence allows differentiation between organisms at the genus level across all major phyla of bacteria, in addition to classifying strains at multiple levels, including species and sub-species level. The 16S rRNA gene sequence has been determined for a large number of strains. GenBank, the largest databank of nucleotide sequences, has over 20 million deposited sequences, of which over 90,000 are of 16S rRNA genes. This means that there are many previously deposited sequences against which to compare the sequence of an unknown strain.
  • In one preferred embodiment, the lactic acid bacterial strain has a 16S rRNA gene sequence selected from SEQ ID NOS 1-87, or a homologue or variant thereof. Another embodiment of the invention relates to a lactic acid bacterial strain that comprises a 16S rRNA gene sequence selected from SEQ ID NOS 1-87, or a homologue or variant thereof. Preferred uses/methods apply to this aspect mutatis mutandis.
  • The term “homologue” is as defined hereinabove. As used herein, the term “variant” includes any variation wherein: (a) one or more nucleotides are substituted by another nucleotide or deleted, (b) the order of two or more nucleotides is reversed, (c) both (a) and (b) are present together. Preferably, the variants arise from one of (a), (b) or (c). More preferably, one or two nucleotides are substituted or deleted. Even more preferably, one nucleotide is substituted by another.
  • In one preferred embodiment of the invention, the lactic acid bacterial strain is characterised by the ability to exhibit antimicrobial activity against E. coli. The observed antimicrobial activity is most likely by virtue of anti-microbial substances produced by the lactic acid bacterial strains of the invention, although nature of these anti-microbial substances has not been determined.
  • In the context of the present invention, the ability to exhibit antimicrobial activity against E. coli can be determined by measuring inhibition of the growth of E. coli in an in vitro well diffusion assay. Further details of the well diffusion assay are set forth in the accompanying examples. The assay is carried out using Escherichia coli K88 on MacConkey No 3 agar, incubating the plates for 16 hours at 37° C. More specifically, Escherichia coli K88 is added to the agar (1 ml of a 1:1000 dilution of an overnight culture of Escherichia coli K88 in 200 ml agar to give the equivalent of 106 CFU/ml). The agar is poured into petri dishes and allowed to set. The plates are marked off into quadrants and an approximately 5 mm well cut out in each quadrant. An aliquot (60 μl) of conditioned media or MRS broth is added to the wells. The plates are covered and incubated for 16 hours at 37° C. They are photographed using a digital camera. Images are transferred to Photoshop, and the diameter of the well and zone of inhibition were determined using the measure tool.
  • In the context of killing E. coli in the above well diffusion assay, preferably the lactic acid bacterial strain of the invention exhibits <20000 units of inhibition, more preferably 20000-40000 units, even more preferably 40000-60000 units, more preferably 60000-80000 units, more preferably 80000-100000 units of inhibition, even more preferably still >100000 units of inhibition.
  • In one preferred embodiment of the invention, the lactic acid bacterial strain is characterised by the ability to exhibit antimicrobial activity against S. enteritidis. Again, the observed antimicrobial activity is most likely by virtue of anti-microbial substances produced by the lactic acid bacterial strains of the invention, although nature of these anti-microbial substances has not been determined.
  • In the context of the present invention, the ability to exhibit antimicrobial activity against S. enteritidis can be determined by measuring the ability to inhibit the growth of S. enteritidis in an in vitro well diffusion assay. Further details of the well diffusion assay are set forth in the accompanying examples. The assay is carried out using Salmonella enteritidis S1400 on XLD agar, incubating the plates for 16 hours at 37° C. XLD agar is prepared as per manufacturer's instructions and cooled to 45° C. Salmonella enteritidis S1400 is added to the XLD agar (1 ml of a 1:1000 dilution of an overnight culture of Salmonella enteritidis S1400 in 200 ml agar to give the equivalent of 106 CFU/ml). The XLD agar is poured into petri dishes and allowed to set. The plates are marked off into quadrants and an approximately 5 mm well cut out in each quadrant. An aliquot (60 μl) of conditioned media or MRS broth is added to the wells. The plates are covered and incubated for 16 hours at 37° C. and the data analysed as described above for the E. coli assay.
  • In the context of killing Salmonella enteritidis in the above well diffusion assay, preferably the lactic acid bacterial strain of the invention exhibits <20000 units of inhibition, more preferably 20000-40000 units, even more preferably 40000-60000 units, more preferably 60000-80000 units, more preferably 80000-100000 units of inhibition, even more preferably still >100000 units of inhibition.
  • In an alternative embodiment, the ability to exhibit antimicrobial activity against S. enteritidis can be determined by measuring the ability to inhibit S. enteritidis in vivo in C3H/HeN or C57BI/6 mice. Further details of appropriate in vivo assays are set forth in the accompanying examples.
  • Specifically, C3H/HeN and C57BI/6 mice are treated with a lactic acid bacterial strain according to the invention prior to and post-challenge with Salmonella enteritidis. The mice are euthanased and dissected 6 (C57BI/6) or 10 (C3H/HeN) days post-infection and viable salmonella are detected in systemic tissues (e.g. the mesenteric lymph node, liver and spleen), in the intestine (e.g. caecum, colon) and in the faeces as compared to appropriate controls. The in vivo activity of the lactic acid bacterial strain of the invention can also be measured by determining the level of myeloperoxidase [MPO], a marker for neutrophils, in the intestine of C3H/HeN mice treated with salmonella or salmonella plus LAB. MPO in the intestine is greatly increased by salmonella infection, due to recruitment of neutrophils to the intestine part of the host response to infection. Co-treatment with a lactic acid bacterial strain according to the invention reduces MPO activity in the intestine of salmonella-infected mice, indicating that the intestinal inflammatory responses to infection are lowered in these animals, relative to control experiments.
  • In one preferred embodiment of the invention, the lactic acid bacterial strain is characterised by the ability to suppress inflammation in IPEC cells induced by 12-O-tetradecaboylphorbol-13-acetate (PMA). In the context of the present invention, this refers to the ability of the lactic acid bacterial strain to block interleukin-8 (IL-8) gene expression triggered by PMA. More specifically, it can be determined by measuring the suppression of inflammation in IPEC-J2 cells induced by PMA when incubated for 2 hours at 37° C., 5% CO2, 95% humidity. Following RNA and reverse transcription, real time PCR is carried out on a 7500 Fast Real-time PCR system operating with 7500 Fast System v 1.4.0 Sequence Detection Software version 1.4 (Applied Biosystem), using primers for porcine IL-8 and TNF-α (prepared by Sigma Aldrich). The reaction mix is: 100 Power Sybergreen Master mix, 2.5 μl of forward primer, 2.5 μl of reverse primer and 5 μl of cDNA, The Real Time PCR is then run according to the Standard 7500 protocol (95° C., 10 min, 1 cycle. 95° C., 15 sec, 40 cycles. 60° C., 1 min, 40 cycles. 95° C., 15 sec, 1 cycle. 60° C., 1 min, 1 cycle. 95° C., 15 sec, 1 cycle. 60° C., 15 sec, 1 cycle). Expression of IL-8 and TNF-α genes are analysed and compared to that of the ‘house-keeping’ gene β-actin. For comparison, values are given as the ratio of IL-8 and TNF-α per β-actin or fold-change. Further details of this assay are set forth in the accompanying examples.
  • In one preferred embodiment of the invention, the lactic acid bacterial strain is characterised by the ability to block the attachment or invasion of IPEC cells by S. enteritidis. This can be measured by the assay set forth in the accompanying examples. Specifically, monolayers of IPEC-J2 cells are grown to 3 days post-confluence in 24-well plates and synchronised by the addition of DTS media 24 hrs prior to use. Overnight cultures of pig LAB (10 ml) are centrifuged and the bacteria re-suspended in phosphate buffered saline [PBS]. An aliquot (50 μl)) of LAB is added to the wells. The plates are incubated for 2 hours at 37° C., 5% CO2, 95% humidity. An overnight culture of Salmonella enterica serovar Enteritidis S1400 [S. enteritidis S1400] is sub-cultured (0.5 ml in 10 ml) into Luria Bertani (LB) media and incubated aerobically for 2-3 hours at 37° C. until it reaches an optical density (560 nm) of 0.8 (a concentration equivalent to 1×108 CFU/ml). The culture is centrifuged and the bacteria re-suspended in PBS. An aliquot (50 μl) is added to the wells of IPEC-J2 cells. The plates are incubated for a further 2 hours at 37° C., 5% CO2, 95% humidity. The IPEC-J2 cell monolayers are washed with HBSS. A solution (0.5 ml) of PBS containing Triton-X100 (10 ml/litre) is added to each well, the monolayer scraped off and dispersed. Viable salmonella are estimated on XLD agar plates (incubated for 24 hours at 37° C.) by the Miles and Misra method. Lactic acid bacteria are determined by the same procedure (incubated anaerobically for 48 hours at 37° C.).
  • Preferably, in the context of the adherence/invasion of IPEC cells by S. enteritidis the lactic acid bacterial strain of the invention exhibits 0-20% inhibition of adherence/invasion, more preferably 20-40%, even more preferably 40-60%, more preferably still, 60-80%, even more preferably still, 80-100% inhibition of adherence/invasion as measured by the above assay.
  • In one preferred embodiment of the invention, the lactic acid bacterial strain is characterised by the ability to block the attachment or invasion of IPEC cells by E. coli. This can be measured by a similar assay to that described above for S. enteritidis, and as set forth in the accompanying examples.
  • Preferably, in the context of the adherence/invasion of IPEC cells by E. coli K88 the lactic acid bacterial strain of the invention exhibits 0-20% inhibition of adherence/invasion, more preferably 20-40%, even more preferably 40-60%, more preferably still, 60-80%, even more preferably still, 80-100% inhibition of adherence/invasion as measured by the above assay.
  • In one preferred embodiment of the invention, the lactic acid bacterial strain is characterised by the absence of antibiotic resistance to one or more antibiotics selected from the following: ampicillin; cefotaxime; chloramphenicol; erythromycin; gentamicin; tetracycline; vancomycin; metronizadole; nalidixic acid; and kanamycin. In the context of the present invention, antibiotic resistance can be determined by measuring the effect of various antibiotic-containing discs on an MRS agar plate culture of the lactic acid bacterial strain, when placed in an anaerobic jar and incubated for 24 hours at 37° C. Further details of the assay are set forth in the accompanying examples. More specifically, pig LAB [0.5 ml of a 1:100 dilution of an overnight culture] is spread onto the surface of an MRS agar plate and dried off. The plates are marked off into 4 quadrants and in each quadrant is placed an antibiotic-containing disc [Ampicillin, 10 μg. Cefotaxime, 30 μg. Chloramphenicol, 10 μg. Erythromycin, 15 Gentamicin, 10 μg. Kanamycin, 30 μg. Metronizadole, 50 μg. Nalidixic acid, 30 μg. Tetracycline, 30 μg. Vancomycin, 30 μg]. The plates are covered, placed in an anaerobic jar and incubated for 24 hours at 37° C. The plates are photographed using a digital camera. Images are transferred to Photoshop, and the diameter of the zone of inhibition is determined using the measure tool. For each antibiotic, the exclusion area for the test strain is taken and divided with the maximum area of exclusion obtained for that antibiotic.
  • Preferably, the LAB of the invention is characterised by the absence of resistance to the antibiotics ampicillin, cefotaxime, chloramphenicol, erythromycin, gentamicin, tetracycline, vancomycin, metronizadole, nalidixic acid and kanamycin. More preferably, the LAB of the invention is characterised by the absence of resistance to the antibiotics ampicillin, cefotaxime, chloramphenicol, erythromycin, gentamicin, tetracycline and vancomycin.
  • In one preferred embodiment of the invention, the lactic acid bacterial strain is characterised by the ability to exhibit heat stability when subjected to three cycles of heating, each cycle comprising heating at a temperature of 70° C. for a period of 15 minutes. Further details of heat stability studies are set forth in the accompanying examples. More specifically, in the context of the present invention, heat stability is measured by centrifuging an overnight culture (10 ml) of isolated pig LAB and resuspending the pellet in fresh MRS broth (10 ml). An aliquot (1 ml) is heated at 70° C. for 15 min and then plated out (0.5 ml) out on MRS agar and incubated in an anaerobic jar for 48 hours at 37° C. A small number of colonies are detected, picked off, seeded into Hungate tubes containing MRS broth and incubated for 48 hours at 37° C. This culture is centrifuged, re-suspended in MRS broth, heated again at 70° C. for 15 min, plated out on MRS agar, incubated in an anaerobic jar for 48 hours at 37° C., picked off, seeded into Hungate tubes containing MRS broth and incubated for 48 hours at 37° C. This culture is centrifuged, re-suspended in MRS broth, re-heated at 70° C. for 15 min, plated out (0.5 ml) out on MRS agar, incubated in an anaerobic jar for 48 hours at 37° C., picked off, seeded into Hungate tubes containing MRS broth and incubated for 48 hours at 37° C.
  • In one preferred embodiment, the lactic acid bacterial strain has any two of the characterising features selected from the group consisting of (i), (ii), (iii), (iv), (v), (vi) and (vii) set forth above.
  • In one preferred embodiment, the lactic acid bacterial strain has any three of the characterising features selected from the group consisting of (i), (ii), (iii), (iv), (v), (vi) and (vii) set forth above.
  • In one preferred embodiment, the lactic acid bacterial strain has any four of the characterising features selected from the group consisting of (i), (ii), (iii), (iv), (v), (vi) and (vii) set forth above.
  • In one preferred embodiment, the lactic acid bacterial strain has any five of the characterising features selected from the group consisting of (i), (ii), (iii), (iv), (v), (vi) and (vii) set forth above.
  • In one preferred embodiment, the lactic acid bacterial strain has any six of the characterising features selected from the group consisting of (i), (ii), (iii), (iv), (v), (vi) and (vii) set forth above.
  • In one preferred embodiment, the lactic acid bacterial strain has all seven of the characterising features (i), (ii), (iii), (iv), (v), (vi) and (vii) set forth above.
  • In one particularly preferred embodiment, (A), the lactic acid bacterial strain is characterised by features (i) and (ii) above.
  • In one particularly preferred embodiment, (B), the lactic acid bacterial strain characterised by features (iv) and (v) above.
  • In one particularly preferred embodiment, (C), the lactic acid bacterial strain is characterised by features (iv) and (v) above.
  • In one particularly preferred embodiment, the lactic acid bacterial strain is characterised by features denoted (D) to (G) as follows:
      • (D) (i) and (iv); or
      • (E) (i) and (v); or
      • (F) (ii) and (iv); or
      • (G) (ii) and (v);
  • More preferably, the lactic acid bacterial strain is further characterised by feature (vi) in addition to those features recited in any one of embodiments (A) to (G) above.
  • Even more preferably, the lactic acid bacterial strain is further characterised by feature (iii) in addition to those features recited in any one of embodiments (A) to (G) above.
  • Even more preferably still, the lactic acid bacterial strain is further characterised by feature (vii) in addition to those features recited in any one of embodiments (A) to (G) above.
  • Biological Deposits
  • One embodiment of the invention relates to a lactic acid bacterial strain isolated from the faeces of organically reared pigs and selected from the group consisting of strains deposited on 27 Jun. 2011 under the terms of the Budapest Treaty at National Collections of Industrial, Food and Marine Bacteria (NCIMB) at NCIMB Ltd, Ferguson Building, Craibstone Estate, Bucksburn, Aberdeen, UK, AB21 9YA, under the following accession numbers:
  • NCIMB 41846: Lactobacillus reuteri GGDK31;
  • NCIMB 41847: Lactobacillus plantarum/pentosus/paraplantarum GGDK161;
  • NCIMB 41848: Lactobacillus johnsonii/taiwanensis/acidophilus/gasseri GGDK255;
  • NCIMB 41849: Lactobacillus plantarum/pentosus/helveticus/paraplantarum GGDK258;
  • NCIMB 41850: Lactobacillus johnsonii GGDK266.
  • The above deposits NCIMB 41846, NCIMB 41847, NCIMB 41848, NCIMB 41849 and NCIMB 41850, were made by Dr George Grant of the Rowett Institute of Nutrition and Health, University of Aberdeen, Greenburn Road, Aberdeen, AB21 9SB on behalf of the Applicant, GT Biologics Limited.
  • Subsequent studies by the Applicant revealed that the strain deposited as NCIMB 41847 was a mixture of Lactobacillus paraplantarum and Lactobacillus reuteri. Subsequent studies by the Applicant revealed that the strain deposited as NCIMB 41850 was a mixture of Lactobacillus johnsonii and Lactobacillus reuteri. Subsequent studies by the Applicant revealed that the strain deposited as NCIMB 41848 was Lactobacillus reuteri. Isolated strains for the respective components of strains NCIMB 41847 and NCIMB 41850 were subsequently deposited (see below).
  • Another embodiment of the invention relates to a lactic acid bacterial strain isolated from the faeces of organically reared pigs and selected from the group consisting of strains deposited on 12 Jul. 2012 under the terms of the Budapest Treaty at National Collections of Industrial, Food and Marine Bacteria (NCIMB) at NCIMB Ltd, Ferguson Building, Craibstone Estate, Bucksburn, Aberdeen, UK, AB21 9YA, under the following accession numbers:
  • NCI MB 42008 Lactobacillus johnsonii;
  • NCIMB 42009 Lactobacillus reuteri;
  • NCIMB 42010 Lactobacillus plantarum;
  • NCIMB 42011 Lactobacillus reuteri;
  • NCIMB 42012 Lactobacillus reuteri
  • The above deposits NCIMB 42008, NCIMB 42009, NCIMB 42010 and NCIMB 42011 and NCIMB 42012, were made by Professor Denise Kelly of GT Biologics Limited, c/o Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, Aberdeensshire, AB25 2ZD, UK, on behalf of the Applicant, GT Biologics Limited.
  • The invention also encompasses mutant strains, which can be obtained from said strains, and strains exhibiting a DNA-DNA homology of at least 70% and/or a 16S RNA identity of at least 99.5% with a strain selected from those deposited under the above accession numbers.
  • As used herein the term “16S rRNA identity” refers to the percentage identity with a known bacterial strain. In one preferred embodiment, the lactic acid bacterial strain has a 16S rRNA identity of at least 85% or at least 90%, or at least 95, 96, 97, 98 or 99% with a strain selected from those deposited under the above accession numbers. In one highly preferred embodiment, the lactic acid bacterial strain has a 16S rRNA identity of at least 99.5% with a strain selected from those deposited under the above accession numbers.
  • In the context of the present invention, the term “DNA-DNA homology” refers to how closely related two or more separate strands of DNA are to each other, based on their nucleotide sequence. Typically, this is measured in terms of their % identity. In one preferred embodiment, the lactic acid bacterial strain has a DNA-DNA homology of at least 70% with a strain selected from those deposited under the above accession numbers, more preferably, at least 80%, or at least 85%, more preferably still, at least 90, 95, 97, 98 or 99% homology with a strain selected from those deposited under the above accession numbers.
  • In one highly preferred embodiment, the lactic acid bacterial strain has a DNA-DNA homology of at least 70% and a 16S rRNA identity of at least 99.5% with a strain selected from those deposited under the above accession numbers.
  • Compositions
  • Another aspect of the invention relates to a composition comprising one or more lactic acid bacterial strains as described above and a pharmaceutically acceptable excipient, carrier or diluent. Suitable excipients, diluents, carriers are described below.
  • The composition may be any composition, but is preferably a composition to be administered orally, enterally or rectally. For example, the composition may be an edible composition. “Edible” means a material that is approved for human or animal consumption.
  • Another aspect of the invention relates to a probiotic composition comprising a lactic acid bacterial strain as described above.
  • Another aspect of the invention relates to combinations of two more lactic acid bacterial strains as described herein. In a particularly preferred embodiment, such combinations exhibit a synergistic functionality, for example, the combination is synergistic, i.e. the resultant effect is greater than the simple additive effects attributable to the individual lactic acid bacterial components in the combination.
  • One preferred embodiment of the invention relates to a combination of two, three, four or five different lactic acid bacteria, more preferably, two, three or four different lactic acid bacteria, more preferably, two or three different lactic acid bacteria. Where the invention relates to a combination of more than one lactic acid bacterial strain, the individual components of the combination may be present in any ratio.
  • More preferably still, the invention relates to a combination of two different lactic acid bacteria. Preferably, the two different lactic acid bacteria are present in a ratio of from 1/99.9 to 99.9/1 by weight, for example, 1/99 to 99/1 or 10/90 to 90/10, or 20/80 to 80/20, or 30/70 to 70/30 and the like.
  • In one highly preferred embodiment, the combination is a mixture of Lactobacillus johnsonii and Lactobacillus reuteri. Even more preferably, the combination is NCIMB 41850: Lactobacillus johnsonii and Lactobacillus reuteri GGDK266 as described above. Surprisingly, this particular combination of lactic acid bacteria unexpectedly gives rise to beneficial in vivo responses in early weaned pigs (see Examples).
  • In another highly preferred embodiment, the combination is a mixture of Lactobacillus plantarum and Lactobacillus reuteri. Even more preferably, the combination is NCIMB 41847: Lactobacillus plantarum/pentosus/paraplantarum and Lactobacillus reuteri GGDK161 as described above.
  • As used herein, the term “probiotic” means microbial cell preparations or components of microbial cells with a beneficial effect on the health or well-being of the host. (Salminen S, Ouwehand A. Benno Y. et al “Probiotics: how should they be defined” Trends Food Sci. Technol. 1999:10 107-10).
  • Preferably, the probiotic composition is an orally administrable composition of metabolically active, i.e., live and/or or lyophilized, or non-viable heat-killed, irradiated or lysed probiotic bacteria. The probiotic composition may contain other ingredients. The probiotic composition of the invention can be administered orally, i.e., in the form of a tablet, capsule or powder. Alternatively, the probiotic composition of the invention may be administered orally as a food or nutritional product, such as milk or whey based fermented dairy product, or as a pharmaceutical product.
  • A suitable daily dose of the probiotic bacteria is from about 1×103 to about 1×1011 colony forming units (CFU), more preferably from about 1×107 to about 1×1010 CFU, more preferably, about 1×106 to about 1×1010 CFU.
  • In one preferred embodiment, the composition contains bacterial strains and/or their cellular components, as active ingredients, in an amount of from about 1×106 to about 1×1012 CFU/g, respect to the weight of the composition, preferably from about 1×108 to about 1×1010 CFU/g. The dose may be of 1 g, 3 g, 5 g, and 10 g, by way of example.
  • Typically, a probiotic is optionally combined with at least one suitable prebiotic compound. A prebiotic is usually a non-digestible carbohydrate such as an oligo- or polysaccharide, or a sugar alcohol which is not degraded or absorbed in the upper digestive tract. Known prebiotics include commercial products such as inulin and transgalacto-oligosaccharides.
  • Preferably, the composition of the present invention includes a prebiotic in an amount of from about 1 to about 30% by weight, respect to the total weight composition, preferably from 5 to 20% by weight. Preferred carbohydrates are selected from: fructo-oligosaccharides (or FOS), short-chain fructo-oligosaccharides, inulin, isomalt-oligosaccharides, pectins, xylo-oligosaccharides (or XOS), chitosan-oligosaccharides (or COS), beta-glucans, arable gum modified and resistant starches, polydextrose, D-tagatose, acacia fibers, carob, oats, and citrus fibers. Particularly preferred prebiotics are the short-chain fructo-oligosaccharides (for simplicity shown hereinbelow as FOSs-c.c); said FOSs-c.c. are not digestable glucides, generally obtained by the conversion of the beet sugar and including a saccharose molecule to which three glucose molecules are bonded.
  • Preparation of Lactic Acid Bacteria
  • A further aspect of the invention relates to a process for producing a probiotic, said process comprising culturing a lactic acid bacterial strain according to the invention. The skilled person in the art will be familiar with standard techniques and conditions suitable for culturing a bacterial strain according to the invention.
  • A further aspect of the invention relates to a method of preparing one or more bacterial strains according to the invention, said method comprising the steps of:
      • (i) obtaining faeces from an organically reared pig;
      • (ii) freezing the faeces and dispersing in a suitable diluent;
      • (iii) applying the dispersed faeces obtained in step (ii) to a suitable agar, optionally in the presence of supplemental pig colostrum carbohydrates, and incubating under an anaerobic conditions;
      • (v) selecting off distinct colonies of bacteria formed during step (iv) and seeding into a suitable broth, optionally in the presence of supplemental pig colostrum carbohydrates;
      • (vi) incubating the seeded colonies obtained in step (v).
  • Suitable agars include, for example, MRS or LAMVAB agar plates. However, other suitable agars can also be used, and would be familiar to the skilled person.
  • Suitable broths include, for example, MRS broth. However, other suitable broths can also be used, and would be familiar to the skilled person.
  • Preferably, step (iii) involves incubating the agar for at least 72 hours at a temperature of about 37° C.
  • Preferably, step (vi) involves incubating the seeded colonies for at least 48 hours at a temperature of about 37° C.
  • A further aspect of the invention relates to a process for obtaining a porcine lactic acid bacterial strain, said process comprising obtaining faeces from an organically reared pig and extracting one or more porcine lactic acid bacterial strains from said faeces.
  • Preferably, the process comprises the steps of:
      • (i) obtaining faeces from an organically reared pig;
      • (ii) freezing the faeces and dispersing in a suitable diluent;
      • (iii) applying the dispersed faeces obtained in step (ii) to a suitable agar, optionally in the presence of supplemental pig colostrum carbohydrates, and incubating under an anaerobic conditions;
      • (v) selecting off distinct colonies of bacteria formed during step (iv) and seeding into a suitable broth, optionally in the presence of supplemental pig colostrum carbohydrates;
      • (vi) incubating the seeded colonies obtained in step (v).
  • Another aspect of the invention relates to a porcine lactic acid bacterial strain obtained by, or obtainable by, the process described above.
  • Therapeutic Applications
  • Another aspect of the invention relates to one or more lactic acid bacterial strains as defined above for use in medicine.
  • Another aspect of the invention relates to one or more lactic acid bacterial strains as defined above for use in treating an intestinal disorder.
  • Another aspect of the invention relates to the use of one or more lactic acid bacterial strains or a composition as defined above in the preparation of a medicament for treating an intestinal disorder.
  • The term “medicament” as used herein encompasses medicaments for both human and animal usage in human and veterinary medicine. In addition, the term “medicament” as used herein means any substance which provides a therapeutic and/or beneficial effect. The term “medicament” as used herein is not necessarily limited to substances which need Marketing Approval, but may include substances which can be used in cosmetics, nutraceuticals, food (including feeds and beverages for example), probiotic cultures, and natural remedies. In addition, the term “medicament” as used herein encompasses a product designed for incorporation in animal feed, for example livestock feed and/or pet food.
  • Another aspect of the invention relates to a method of treating an intestinal disorder in a subject, said method comprising administering to the subject a pharmaceutically effective amount of one or more lactic acid bacterial strains or a pharmaceutical composition or a probiotic composition as described above.
  • Preferably, the intestinal disorder is selected from irritable bowel syndrome (IBS), inflammatory bowel disorder (IBD), functional dyspepsia, functional constipation, functional diarrhoea (including antibiotic associated diarrhoea, traveller's diarrhoea and pediatric diarrhoea), functional abdominal pain, functional bloating, Epigastric Pain Syndrome, Postprandial Distress Syndrome, Crohn's disease, ulcerative colitis, gastrointestinal reflux disease (GERD), allergies, atopic diseases e.g. atopic dermatitis, necrotising enterocolitis, other infections, and combinations thereof.
  • In one preferred embodiment, the intestinal disorder is IBS. The precise pathophysiology of IBS remains to be elucidated. Recent studies have described mucosal inflammation and alterations in intestinal microbiota in IBS patients and a disease correlation with intestinal infections.
  • In one highly preferred embodiment, the disorder is salmonellosis. Salmonellosis is a disease caused by various strains of salmonella that is characterized by fever and intestinal disorders.
  • Another aspect of the invention relates to one or more lactic acid bacterial strains as defined above for improving intestinal microbiota.
  • Another aspect of the invention relates to a method of improving intestinal microbiota in a subject, said method comprising administering to the subject a composition comprising one or more lactic acid bacterial strains or a pharmaceutical composition or a probiotic composition according to the invention.
  • The lactic acid bacterial strains according to the invention may also be used in prophylactic applications. In prophylactic applications, compositions according to the invention are administered to a patient susceptible to, or otherwise at risk of, a particular disease in an amount that is sufficient to at least partially reduce the risk of developing a disease. Such an amount is defined to be “a prophylactic effective dose”. The precise amounts depend on a number of patient specific factors such as the patient's state of health and weight.
  • The lactic acid bacterial strains and probiotic compositions according to the invention may also be used in animal nutrition (e.g. in pig nutrition), particularly in the early-weaned period and growing fattening period. The probiotics are expected to enhance immune function reduce and prevent infectious diseases, beneficially alter the microbiota composition, and improve growth and performance of animals, for example, through increased feed conversion efficiency. The term “animal” includes all animals including humans. Examples of animals are non-ruminants and ruminants. Ruminant animals include for example, sheep, goat, and cattle eg. cow as beef cattle and dairy cows. In a particular embodiment, the animal is a non-ruminant animal. Non-ruminant animals include pet animals, eg horses, cats, and dogs; monogastric eg pigs or swine (including but not limited to, piglets growing pigs and sows); poultry such as turkeys, ducks, and chickens (including but not limited to broiler chicks, layers); fish (including but not limited to salmon, trout, tilapia, catfish and carp); and crustaceans (including but not limited to shrimp and prawn).
  • Feedstuffs/Products
  • A further aspect of the invention relates to food products, dietary supplements, nutraceuticals, nutritional formulae, drinks and medicaments containing one or more bacterial strains according to the invention.
  • In one preferred embodiment, the composition comprises additionally at least one other kind of other food grade bacterium, wherein the food grade bacterium is preferably selected from the group consisting of lactic acid bacteria, bifidobacteria, propionibacteria or mixtures thereof.
  • One aspect of the invention relates to a food product comprising one or more lactic acid bacterial strains according to the invention. The term “food product” is intended to cover all consumable products that can be solid, jellied or liquid. Suitable food products may include, for example, functional food products, food compositions, pet food, livestock feed, health foods, feedstuffs and the like. In one preferred embodiment, the food product is a health food.
  • As used herein, the term “functional food product” means food that is capable of providing not only a nutritional effect, but is also capable of delivering a further beneficial effect to the consumer. Accordingly, functional foods are ordinary foods that have components or ingredients (such as those described herein) incorporated into them that impart to the food a specific functional—e.g. medical or physiological benefit—other than a purely nutritional effect.
  • Examples of specific food products that are applicable to the present invention include milk-based products, ready to eat desserts, powders for re-constitution with, e.g., milk or water, chocolate milk drinks, malt drinks, ready-to-eat dishes, instant dishes or drinks for humans or food compositions representing a complete or a partial diet intended for pets or livestock.
  • In one preferred embodiment the composition according to the present invention is a food product intended for humans, pets or livestock. The composition may be intended for animals selected from the group consisting of dogs, cats, pigs, cattle, horses, goats, sheep or poultry. In a preferred embodiment, the composition is a food product intended for adult species, in particular human adults.
  • In the present invention, “milk-based product” means any liquid or semi-solid milk or whey based product having a varying fat content. The milk-based product can be, e.g., cow's milk, goat's milk, sheep's milk, skimmed milk, whole milk, milk recombined from powdered milk and whey without any processing, or a processed product, such as yoghurt, curdled milk, curd, sour milk, sour whole milk, butter milk and other sour milk products. Another important group includes milk beverages, such as whey beverages, fermented milks, condensed milks, infant or baby milks; flavoured milks, ice cream; milk-containing food such as sweets.
  • One aspect of the invention relates to a feedstuff or animal feed comprising one or more bacterial strains according to the invention.
  • Feedstuff can be a food additive, a feed premixor an animal feed. Particular examples of feedstuffs according to the invention include the following: animal feed additive comprising (a) porcine lactic acid bacteria according to the present invention (b) at least one fat soluble vitamin (c) at least one water soluble vitamin (d) at least one trace mineral and/or at least one macro mineral; an animal feed composition comprising a porcine lactic acid bacteria according to the present invention and a crude protein content of 50-88 g/kg feed. The so-called premixes are examples of animal feed additives of the invention. A premix designates a preferably uniform mixture of one or more micro-ingredients with diluent and/or carrier. Premixes are used to facilitate uniform dispersion of micro-ingredients in a larger mix.
  • Further, optional, feed-additive ingredients are coloring agents, e.g. carotenoids such as beta-carotene, astaxanthin, and lutein; aroma compounds; stabilisers; antimicrobial peptides; polyunsaturated fatty acids; reactive oxygen generating species; and/or at least one enzyme selected from amongst phytase (EC 3.1.3.8 or 3.1.3.26); xylanase (EC 3.2.1.8); galactanase (EC 3.2.1.89); alpha-galactosidase (EC 3.2.1.22); protease (EC 3.4.), phospholipase A1 (EC 3.1.1.32); phospholipase A2 (EC 3.1.1.4); lysophospholipase (EC 3.1.1.5); phospholipase C (EC 3.1.4.3); phospholipase D (EC 3.1.4.4); amylase such as, for example, alpha-amylase (EC 3.2.1.1); and/or beta-glucanase (EC 3.2.1.4 or EC 3.2.1.6).
  • Examples of polyunsaturated fatty acids are C18, C20 and C22 polyunsaturated fatty acids, such as arachidonic acid, docosohexaenoic acid, eicosapentaenoic acid and gamma-linoleic acid.
  • Examples of reactive oxygen generating species are chemicals such as perborate, persulphate, or percarbonate; and enzymes such as an oxidase, an oxygenase or a syntethase.
  • Usually fat- and water-soluble vitamins, as well as trace minerals form part of a so-called premix intended for addition to the feed, whereas macro minerals are usually separately added to the feed. Either of these composition types, when enriched with a porcine lactic acid bacteria according to the present invention, is an animal feed additive within the scope of the invention.
  • The following are non-exclusive lists of examples of these components: Examples of fat-soluble vitamins are vitamin A, vitamin D3, vitamin E, and vitamin K, e.g. vitamin K3. Examples of water-soluble vitamins are vitamin B12, biotin and choline, vitamin B1, vitamin B2, vitamin B6, niacin, folic acid and panthothenate, e.g. Ca-D-panthothenate. Examples of trace minerals are manganese, zinc, iron, copper, iodine, selenium, and cobalt. Examples of macro minerals are calcium, phosphorus and sodium.
  • The nutritional requirements of these components (exemplified with poultry and piglets/pigs) are listed in Table A of WO 01/58275. Nutritional requirement means that these components should be provided in the diet in the concentrations indicated.
  • In the alternative, the animal feed additive of the invention comprises at least one of the individual components specified in Table A of WO 01/58275. At least one means either of, one or more of, one, or two, or three, or four and so forth up to all thirteen, or up to all fifteen individual components. More specifically, this at least one individual component is included in the additive of the invention in such an amount as to provide an in-feed-concentration within the range indicated in column four, or column five, or column six of Table A of WO 01/58275.
  • Animal feed compositions or diets typically have a relatively high content of protein. Poultry and pig diets can be characterized as indicated in Table B of WO 01/58275, columns 2-3. Fish diets can be characterized as indicated in column 4 of this Table B.
  • Furthermore such fish diets usually have a crude fat content of 200-310 g/kg. WO 01/58275 corresponds to U.S. Ser. No. 09/779,334 which is hereby incorporated by reference.
  • An animal feed composition according to the invention typically has a crude protein content of 50-800 g/kg, and furthermore comprises a porcine lactic acid bacteria according to the present invention thereof as described and/or claimed herein.
  • Furthermore, or in the alternative (to the crude protein content indicated above), the animal feed composition of the invention may have a content of metabolisable energy of 10-30 MJ/kg; and/or a content of calcium of 0.1-200 g/kg; and/or a content of available phosphorus of 0.1-200 g/kg; and/or a content of methionine of 0.1-100 g/kg; and/or a content of methionine plus cysteine of 0.1-150 g/kg; and/or a content of lysine of 0.5-50 g/kg.
  • In certain preferred embodiments, the content of metabolisable energy, crude protein, calcium, phosphorus, methionine, methionine plus cysteine, and/or lysine is within any one of ranges 2, 3, 4 or 5 in Table B of WO 01/58275 (R. 2-5). Crude protein is calculated as nitrogen (N) multiplied by a factor 6.25, i.e. Crude protein (g/kg)=N (g/kg)×6.25. The nitrogen content is determined by the Kjeldahl method (A.O.A.C., 1984, Official Methods of Analysis 14th ed., Association of Official Analytical Chemists, Washington D.C.). Metabolisable energy can be calculated on the basis of the NRC publication Nutrient requirements in swine, ninth revised edition 1988, subcommittee on swine nutrition, committee on animal nutrition, board of agriculture, national research council. National Academy Press, Washington, D.C., pp. 2-6, and the European Table of Energy Values for Poultry Feed-stuffs, Spelderholt centre for poultry research and extension, 7361 DA Beekbergen, The Netherlands. Grafisch bedrijf Ponsen & looijen by, Wageningen. ISBN 90-71463-12-5.
  • The dietary content of calcium, available phosphorus and amino acids in complete animal diets is calculated on the basis of feed tables such as Veevoedertabel 1997, gegevens over chemische samenstelling, verteerbaarheid en voederwaarde van voedermiddelen, Central Veevoederbureau, Runderweg 6, 8219 pk Lelystad. ISBN 90-72839-13-7.
  • In one preferred embodiment, the animal feed composition of the invention contains at least one vegetable protein or protein source. It may also contain animal protein, such as Meat and Bone Meal, and/or Fish Meal, typically in an amount of 0-25%. The term vegetable proteins as used herein refers to any compound, composition, preparation or mixture that includes at least one protein derived from or originating from a vegetable, including modified proteins and protein-derivatives. In certain particularly preferred embodiments, the protein content of the vegetable proteins is at least 10, 20, 30, 40, 50, or 60% (w/w).
  • Vegetable proteins may be derived from vegetable protein sources, such as legumes and cereals, for example materials from plants of the families Fabaceae (Leguminosae), Cruciferaceae, Chenopodiaceae, and Poaceae, such as soy bean meal, lupin meal and rapeseed meal. In a particular embodiment, the vegetable protein source is material from one or more plants of the family Fabaceae, e.g. soybean, lupine, pea, or bean. Other examples of vegetable protein sources are rapeseed, sunflower seed, cotton seed, and cabbage. Other examples of vegetable protein sources are cereals such as barley, wheat, rye, oat, maize (corn), rice, triticale, and sorghum.
  • Animal diets can e.g. be manufactured as mash feed (non pelleted) or pelleted feed. Typically, the milled feed-stuffs are mixed and sufficient amounts of essential vitamins and minerals are added according to the specifications for the species in question. A porcine lactic acid bacteria according to the present invention thereof can be added as solid or liquid formulations.
  • The compositions of the present invention may be—or may be added to—food supplements, also referred to herein as dietary supplements or food additives. Thus, another aspect of the invention relates to a dietary supplement or food additive comprising one or more bacterial strains according to the invention.
  • Another embodiment of the invention relates to the use of a feedstuff as described above for improving animal growth performance as measured by daily weight gain and/or feed conversion ratio.
  • In a preferred embodiment, the invention relates to methods for using a feedstuff comprising one or more porcine lactic acid bacteria according to the present invention in animal feed for improving daily weight gain, improving the Feed Conversion Ratio (FCR) and/or for modulation of the gut microflora.
  • In alternative preferred embodiments, the feedstuff comprising one or more porcine lactic acid bacteria according to the present invention improves animal feed digestibility, and/or maintains animal health by aiding in proper digestion and/or supporting immune system function.
  • The FCR may be determined on the basis of a piglet growth trial comprising a first treatment in which the feedstuff comprising a porcine lactic acid bacteria according to the present invention is added to the animal feed in a suitable concentration per kg feed, and a second treatment (control) with no addition of a porcine lactic acid bacteria according to the present invention to the animal feed. In the present context, the term Feed Conversion Ratio, or FCR, is used synonymously with the term feed conversion. The FCR is calculated as the feed intake in g/animal relative to the weight gain in g/animal. As it is generally known, an improved FCR is lower than the control FCR. In particular embodiments, the FCR is improved (i.e., reduced) as compared to the control by at least 1.0%, preferably at least 1.5%, 1.6%, 1.7%, 1.8%, 1.9%, 2.0%, 2.1%, 2.2%, 2.3%, 2.4%, or at least 2.5%.
  • The term “gut” as used herein designates the gastrointestinal or digestive tract (also referred to as the alimentary canal) and it refers to the system of organs within multicellular animals which takes in food, digests it to extract energy and nutrients, and expels the remaining waste.
  • The term gut “microflora” as used herein refers to the natural microbial cultures residing in the gut and maintaining health by aiding in proper digestion and/or supporting immune system function.
  • The term “modulate” as used herein in connection with the gut microflora generally means to change, manipulate, alter, or adjust the function or status thereof in a healthy and normally functioning animal, i.e. a non-therapeutic use.
  • Diluents, Excipients and Carriers
  • As mentioned above, the invention also relates to compositions, more preferably pharmaceutical compositions, comprising a lactic acid bacterial strain according to the invention. The lactic acid bacterial strains of the present invention are generally administered in admixture with a pharmaceutical carrier, excipient or diluent, particularly for human therapy. The pharmaceutical compositions may be for human or animal usage in human and veterinary medicine.
  • Examples of such suitable excipients for the various different forms of pharmaceutical compositions described herein may be found in the “Handbook of Pharmaceutical Excipients, 2nd Edition, (1994), Edited by A Wade and P J Weller.
  • Acceptable carriers or diluents for therapeutic use are well known in the pharmaceutical art, and are described, for example, in Remington's Pharmaceutical Sciences, Mack Publishing Co. (A. R. Gennaro edit. 1985).
  • Examples of suitable carriers include lactose, starch, glucose, methyl cellulose, magnesium stearate, mannitol, sorbitol and the like. Examples of suitable diluents include ethanol, glycerol and water.
  • The choice of pharmaceutical carrier, excipient or diluent can be selected with regard to the intended route of administration and standard pharmaceutical practice. The pharmaceutical compositions may comprise as, or in addition to, the carrier, excipient or diluent any suitable binder(s), lubricant(s), suspending agent(s), coating agent(s), solubilising agent(s).
  • Examples of suitable binders include starch, gelatin, natural sugars such as glucose, anhydrous lactose, free-flow lactose, beta-lactose, corn sweeteners, natural and synthetic gums, such as acacia, tragacanth or sodium alginate, carboxymethyl cellulose and polyethylene glycol.
  • Examples of suitable lubricants include sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride and the like.
  • Preservatives, stabilizers, dyes and even flavoring agents may be provided in the pharmaceutical composition. Examples of preservatives include sodium benzoate, sorbic acid and esters of p-hydroxybenzoic acid. Antioxidants and suspending agents may be also used.
  • Administration
  • The compositions of the present invention may be adapted for oral, rectal, vaginal, parenteral, intramuscular, intraperitoneal, intraarterial, intrathecal, intrabronchial, subcutaneous, intradermal, intravenous, nasal, buccal or sublingual routes of administration. Preferably, the compositions of the present invention are adapted for oral, rectal, vaginal, parenteral, nasal, buccal or sublingual routes of administration.
  • For oral administration, particular use is made of compressed tablets, pills, tablets, gellules, drops, and capsules.
  • Other forms of administration comprise solutions or emulsions which may be injected intravenously, intraarterially, intrathecally, subcutaneously, intradermally, intraperitoneally or intramuscularly, and which are prepared from sterile or sterilisable solutions. The pharmaceutical compositions of the present invention may also be in form of suppositories, pessaries, suspensions, emulsions, lotions, ointments, creams, gels, sprays, solutions or dusting powders.
  • An alternative means of transdermal administration is by use of a skin patch. For example, the active ingredient can be incorporated into a cream consisting of an aqueous emulsion of polyethylene glycols or liquid paraffin. The lactic acid bacterial strain can also be incorporated into an ointment consisting of a white wax or white soft paraffin base together with such stabilisers and preservatives as may be required.
  • Compositions may be formulated in unit dosage form, i.e., in the form of discrete portions containing a unit dose, or a multiple or sub-unit of a unit dose.
  • Dosage
  • A person of ordinary skill in the art can easily determine an appropriate dose of one of the instant compositions to administer to a subject without undue experimentation. Typically, a physician will determine the actual dosage which will be most suitable for an individual patient and it will depend on a variety of factors including the activity of the specific bacterial strain employed, the metabolic stability and length of action of that strain, the age, body weight, general health, sex, diet, mode and time of administration, rate of excretion, drug combination, the severity of the particular condition, and the individual undergoing therapy. The dosages disclosed herein are exemplary of the average case. There can of course be individual instances where higher or lower dosage ranges are merited, and such are within the scope of this invention.
  • The usual effective daily dose in humans or in animals is from about 1×103 to about 1×1011, more preferably, from about 1×107 to about 1×1011, even more preferably, from about 1×106 to about 1×1010 CFU.
  • Combinations
  • In one preferred embodiment, the compositions of the invention are administered in any combination, for example, two or more of the lactic acid bacteria may be administered in any combination or ratio.
  • In another particularly preferred embodiment, the compositions of the invention are administered in combination with one or more other active agents. In such cases, the compositions of the invention may be administered consecutively, simultaneously or sequentially with the one or more other active agents.
  • Isolation and Characterisation of Bacterial Strains
  • The LAB strains isolated (total of 436 individual colony picks) from faeces of organically-reared pigs were predominantly L. reuteri, L. johnsonii, L. gasseri, L. pentosus, strains with a small number of L. plantarum, L. acidophilus, L. vaginalis, a single L. mucosae and several uncultured strains.
  • Most of the LAB produced substances that could inhibit the growth of S. enteritidis and/or E. coli K88 in vitro. The potency of these anti-pathogen effects varied greatly between the individual bacterial strains.
  • Certain strains were selected on the basis of anti-microbial potency as determined in vitro. These bacteria were further screened for their ability to block adherence/invasion of intestinal pig epithelial cells (IPEC) by pathogens in vitro and their susceptibility to antibiotics.
  • Certain strains were assayed for substrate range and specificity and their capacity to suppress inflammation in IPEC cells in vitro. From these, fourteen LAB (5 L. johnsonii, 6 L. reuteri and 3 L. plantarum) with favourable properties were identified. Two of these strains [GGDK266 and GGDK31] were prepared in bulk for in vivo evaluation in newly-weaned piglets. Other potentially important candidates were present amongst this set of 14 LAB.
  • Small losses in viability were evident on freeze drying and storage of LAB dried in skimmed milk powder. A combination of skimmed milk powder and simple sugars was slightly more effective, but difficult to maintain. Bulk preparations of GGDK266 and GGDK31 were freeze-dried and stored in this medium.
  • Five heat-conditioned cultures of LAB were obtained. However, the biological properties in vitro and probiotic potential of three strains were adversely affected by heat-treatment. Nonetheless, two of the bacteria retained the biological properties of the native non-heat-treated forms.
  • Oral treatment of mice with pig LAB (L. reuteri or L. mucosae) greatly reduced the pathogenicity of S. enteritidis in acute (C57BI/6 mouse) and chronic (C3H/HeN mouse) forms of salmonellosis.
  • The data indicate that LAB from organically-reared pigs have considerable potential as a source of novel and potent probiotics.
  • Studies carried out by the applicant involved isolating large numbers of LAB from organically-reared pigs and screening for potent probiotic LAB strains by assessing their biological potency and mode of action both in vitro and in vivo.
  • More specifically, experiments were undertaken to establish cultures of LAB derived from faeces of organically-reared pigs. The LAB strains were screened for anti-microbial activity against a number of pathogens in vitro. Experiments were undertaken to determine whether the LAB strains could block the attachment of pathogens to pig epithelial cells in vitro. Studies were also undertaken to evaluate the capacity of LAB to block inflammatory responses in pig epithelial cells in vitro. Strains demonstrating a good bioactive profile in vitro were selected and cultured in bulk for a large-scale study in vivo.
  • Further details on the experimental techniques are described in the accompanying examples section. In brief, LAB strains were isolated and cultured from pig faeces using selective microbiological media. Individual bacterial colonies were isolated and 16S rRNA gene sequences were analysed to enable genotypic identification of bacterial strains. Phenotypic characteristic of potential probiotics was further determined following measurement of adherence, anti-bacterial and anti-inflammatory activities, antibiotic susceptibility and finally heat stability. Anti-bacterial activity of conditioned media derived from LAB was evaluated using well-diffusion assays to determine killing activity against the enteric pathogens Salmonella enteritidis and E. coli K88. The ability of LAB strains to block or interfere with S. enteritidis and E. coli K88 adherence/invasion of pig epithelial (IPEC) was also evaluated, as was their capacity to suppress inflammation in IPEC cells induced by 12-O-Tetradecaboylphorbol-13-acetate [PMA]. In addition, the metabolic properties of LAB strains (API CH 50 kit) and their susceptibility to antibiotics was further determined. A ranking system, based on scoring the biological properties of LAB was established and used for the selection of candidate LAB strains for probiotic evaluation in vivo.
  • Further details on the results of the above experiments are described in the accompanying examples.
  • The LAB (436 individual colony picks) isolated from faeces of organically-reared pigs were predominantly L. johnsonii or L. johnsonii-related and L. reuteri or L. reuteri-related with small numbers of L. plantarum-related and uncultured strains. This represented a much narrower range of porcine-associated LAB than reported by others (Martin et al, 2009; Yun et al, 2009; Lahteinen et al, 2010; Yao et al, 2011). However, in comparison to conventionally/intensively-reared pigs, out-door organically-reared pigs had high levels of LAB and more developed intestinal immune function (Mulder et al, 2009). The present bacterial data indicate that L. johnsonii and L. reuteri strains are of particular importance in proper development of the gut and immune system in young pigs. In addition, the inclusion of other lactic acid bacteria derived from the gut or faeces of organically-reared pigs, in particular, Lactobacillus delbrueckii and Lactobacillus amylovorous may enhance the immune homeostatic properties of Lactobacillus reuteri, Lactobacillus plantarum and Lactobacillus johnsonii.
  • All of the isolated pig LAB produced substances that could kill or interfere with the growth of S. enteritidis in a well-diffusion assay and the majority killed or suppressed growth of E. coli K88. The potency of the anti-microbial activities varied greatly between individual colonies, irrespective of whether they were L. reuteri, L. johnsonii or L. plantarum. There was no general correlation between the anti-salmonella and anti-E. coli K88 potency of each of the LAB. LAB are known to produce a range of active factors, including organic acids, small anti-microbial compounds and anti-bacterial peptides (Cintas et al, 2001). The nature of these anti-microbial substances produced by LAB from organically-reared pigs has not been established.
  • Thirty three pig LAB strains, selected on the basis of anti-pathogen activity, were tested for the ability to block attachment/invasion of IPEC cells by S. enteritidis and E. coli K88. They were all able to dramatically reduce attachment/invasion of IPEC cells by salmonella. The majority could also block E. coli K88. As with pathogen killing, there was no general correlation between the abilities of the LAB to block salmonella and E. coli K88. Without wishing to be bound by theory, it is believe that the LAB may limit the access of pathogens to the epithelial layer by occupying binding-sites on the cell monolayer or by production of factors that interfere with attachment of the pathogen to the epithelial cells, such as blocking binding sites of surface adhesins (Ljungh and Wadstrom, 2006; Blandino et al, 2008; Williams, 2010).
  • Pig LAB may also block or suppress inflammatory gene (interleukin-8, IL-8)-expression triggered in IPEC cells by PMA. Individual cultures varied greatly in their ability to affect inflammation, but five strains ( RINH vial 29, 30, 31 86 and 266) had potent anti-inflammatory properties. Certain LAB strains are known to have immuno-modulatory or anti-inflammatory properties (Cotter et al, 2005; Blandino et al, 2008; Ohashi and Ushida, 2009; Elmadfa et al, 2010; Liu et al, 2010). The mechanisms involved remain unclear, but are likely to involve modulation of molecular signalling systems by bioactive factors produced by the LAB.
  • Antibiotic resistance is an increasing problem and can spread between bacteria by gene transfer (Korhonen et al, 2007; Gousia et al, 2011; Nicolau, 2011). Ideally, candidate probiotics should have little or no resistance to antibiotics to minimise the risk of transfer of resistance genes to the host flora. Pig LAB (33 strains) were screened for resistance to 10 individual antibiotics. One strain (RINH vial 266) was susceptible to all the tested antibiotics. Most were susceptible to ampicillin, cefotaxime, chloramphenicol, erythromycin, gentamicin, tetracycline and vancomycin. However, most exhibited resistance to metronizadole, nalidixic acid and to a lesser extent kanamycin. This relatively low incidence of antibiotic resistance amongst these LAB isolates may be linked to the environment in which the source piglets were reared [organic out-door reared] (Mulder et al, 2009).
  • L. johnsonii, L. reuteri and L. plantarum, as expected, exhibited strain-specific general substrate reaction profiles, when assayed using an API CH 50 kit. However, most genotype strains exhibited fine differences in their substrate reactivity. This indicated that they were unique individual strains of the genotype.
  • On the basis of their biological activities in vitro, fourteen LAB [4 L. plantarum-related, 3 L. johnsonii-related and 1 L. reuteri] were identified as having potential for testing in vivo. Two of these LAB strains [GGDK266 and GGDK31] were prepared in bulk.
  • Interestingly, 7 of the fourteen LAB ( RINH vials 85, 86, 131, 230, 255, 266) had been isolated from LAB-selective agars supplemented with carbohydrate fractions from pig colostrum. The growth and bioactivity profile of LAB is, in part, dependent on the carbohydrate substrate in which it is grown (Gopal et al, 2001; Tzortzis et al, 2004), The present data may indicate that some of the LAB are host-adapted and require certain pig-associated carbohydrates for optimal growth or bioactivity.
  • It is advantageous if the LAB can withstand being freeze dried to allow them to be handled and processed as probiotics. However, their viability can be greatly reduced during freezing and drying (Tomas et al, 2009; Strasser et al, 2009; Reddy et al, 2009). Skimmed milk powder, alone or in combination with simple sugars, is often used as a cryo-protectant to preserve the viability of the bacteria (Tomas et al, 2009; Strasser et al, 2009). In the present study, small losses in viability were evident on drying and storage of pig LAB in skimmed milk powder alone. Sucrose or lactose in combination with skimmed milk powder was slightly more protective. However, the product was hygroscopic and difficult to store or handle. It was therefore decided to dry and store pig LAB in skimmed milk powder.
  • Supplemental feeds for animal are often given as pellets, production of which involves high temperatures (De Angelis et al, 2006). LAB to be added to animal feeds should therefore have a significant degree of heat-stability to minimise loss of viability during processing. In the present study, five LAB were subject to heating three times for 15 minutes at 70° C. All of the bacteria that were recovered after the third heat-treatment were viable and in most cases grew at rates similar to the native forms of the bacteria. Two of the bacteria retained the biological properties of the native non-heat-treated forms. However, one of the heat-treated strains had lost the ability to block attachment of pathogen to epithelial cells in vitro and another had greatly reduced blocking activity. A further strain was unable to block PMA-induced inflammation in epithelial cells in vitro, although the native form was a potent suppressor of inflammation. Heat-treatment can thus differentially affect the biological properties of individual LAB. This needs to be taken into account when considering inclusion of LAB in pelleted animal feeds.
  • Experiments demonstrated that the pathogenicity of S. enteritidis was attenuated if mice were co-treated with LAB derived from organically-reared pigs. RINH vial 323 (L. mucosae) greatly reduced the ability of S. enteritidis to invade, spread to and proliferate in systemic tissues in acute (C57BI/6 mouse) and chronic (C3H/Hen mouse) salmonellosis. Furthermore, RINH vial 31 [GGDK31], RINH vial 32, RINH vial 46 or RINH vial 47 (all L. reuteri) reduced colonisation of the large intestine, invasion and systemic spread and proliferation in C3H/HeN mice by S. enteritidis. Overall, RINH vial 31 [GGDK31] and RINH vial 32 were the most effective in this chronic model of salmonellosis. These LAB have potential as novel probiotics to promote gut health or increase resistance to infection in vivo.
  • Infection by salmonella is a multi-factorial process (Naughton and Grant, 2005). S. enteritidis colonises the whole gastro-intestinal tract, moves through the mucus layer and attaches to the mucosa. The large intestine acts as a reservoir for the pathogen but invasion is primarily via M cells, present on the Peyer's patches of the ileum. Most invaded salmonella spread to the mesenteric lymph nodes and then out to the liver and spleen (Naughton and Grant, 2005). Without wishing to be bound by theory, it is believed that LAB could be blocking salmonella at various stages of the infection (Cintas et al, 2001; Cotter et al, 2005; Ohashi and Ushida, 2009). By competing for nutrients, killing of pathogen or blocking of attachment sites, LAB could limit the numbers of salmonella in the large intestine reservoir. LAB may also prevent attachment to ileal mucosal cells, in a manner similar to that observed here with IPEC-J2 cells and with Caco-2 cells (Neeser et al, 2000) and thereby limit invasion.
  • Alternatively, LAB may directly modulate host responses to the infection, in particular suppression of inflammation. By limiting gut damage and preserving barrier integrity (Smith et al, 2008; Schreiber et al, 2009), the ability of salmonella to invade and spread would be greatly reduced.
  • The present invention is further described by way of non-limiting example, and with reference to the following non-limiting figures, wherein:
  • FIG. 1 shows an assay of antibacterial activity of conditioned media from Lactic Acid Bacteria.
  • FIGS. 2A and 2B show inhibitory activity against S. enteritidis S1400 (expressed as area of inhibition in a well diffusion assay) of conditioned media of all individual LAB cultured from faeces of organically-reared pigs.
  • FIGS. 3A and 3B show inhibitory activity against E. coli K88 (expressed as area of inhibition in a well diffusion assay) of conditioned media of all individual LAB cultured from faeces of organically-reared pigs.
  • FIGS. 3C and 3D show inhibitory activity (expressed as area of inhibition in a well diffusion assay) of conditioned media of all individual LAB cultured from faeces of organically-reared pigs.
  • FIGS. 4A, 4B, and 4C shown inhibition of adherence by (FIG. 4A) S. enteritidis S1400; and (FIG. 4B) E. coli K88 to IPEC cells in culture by LAB cultured from faeces of organically-reared pigs; (FIG. 4C) comparison between inhibition of S. enteritidis S1400 and E. coli K88.
  • FIG. 5 shows an assay of the antibiotic susceptibility of Lactic Acid Bacteria using discs impregnated with a defined amount of antibiotic.
  • FIG. 6 shows an evaluation of substrate profile of LAB using an API CH 50 kit [49 substrates, pale colour indicated positive reaction, except 25 where positive reaction is black, dark colour indicates no reaction].
  • FIGS. 7A-7C show the ΔCt (FIG. 7A), ratio (FIG. 7B) and fold-change (FIG. 7C) for IL-8 gene expression in IPEC cells treated with PMA and pig LAB.
  • FIGS. 8A and 8B show the stability of L. reuteri (FIG. 8A) and L. johnsonii (FIG. 8B) after freeze-drying in skimmed milk powder (SKP, (100 g/1), SKP+lactose (both 100 g/1), SKP+sucrose (both 100 g/1) or SKP (200 g/1).
  • FIGS. 9A-9D show the stability of isolated LAB to heat-treatment (FIG. 9A), the ratio (FIG. 9B) and fold-change (FIG. 9C) for IL-8 gene expression in IPEC cells treated with PMA and naive or heat-treated pig LAB; (FIG. 9D) Antibiotic susceptibility of native and heat-treated RINH vial 31.
  • FIG. 10 shows a protocol for the C3H/HeN mouse study to evaluate efficacy of vial 323 (L. mucosae) to counteract salmonella infection in vivo.
  • FIGS. 11A-11C show the distribution of S. enteritidis S1400 in tissues at 10 days post-infection in C3H/HeN mice that had or had not been co-treated with 323 (L. mucosae, LM).
  • FIGS. 12A-12B show spleen weight (mg/100 g BW) and intestinal (ileal) myeloperoxidase (μg) at 10 days post-infection in C3H/HeN mice that had or had not been co-treated with vial 323 (L. mucosae).
  • FIG. 13 shows a protocol for the C57 BI/6 mouse study to evaluate efficacy of vial 323 (L. mucosae) to counteract acute salmonella infection in vivo.
  • FIGS. 14A-14C shows the distribution of S. enteritidis S1400 in tissues at 6 days post-infection in C57BI/6 mice that had or had not been co-treated with RINH vial 323.
  • FIG. 15 shows spleen weight (mg/100 g BW) at 6 days post-infection in C57BI/6 mice that had or had not been co-treated with vial 323 (L. mucosae).
  • FIG. 16 shows a protocol for the C3H/HeN mouse study to evaluate efficacy of selected LAB from faeces of organically reared pigs to counteract salmonella infection in vivo.
  • FIGS. 17A & 17B show excretion of S. enteritidis in faeces at 7-8 days post-infection by C3H/HeN mice that had or had not been co-treated with selected LAB.
  • FIGS. 18A-18B show the distribution of S. enteritidis (Log 10 CFU/g) in caecum (18A[[b]]) and colon (18B) at 10 days post-infection of C3H/HeN mice that had or had not been co-treated with selected LAB.
  • FIGS. 19A-19C show the distribution of S. enteritidis (Log 10 CFU/g) in mesenteric lymph node (19A), liver (19B) and spleen (19C) at 10 days post-infection of C3H/HeN mice that had or had not been co-treated with selected LAB.
  • FIG. 20 shows the performance of pigs fed GGDK266 and GGDK31 versus a control (daily weight gain, DWG, in g/day) for days 0-7, 7-14 and 0-14.
  • FIG. 21 shows microbial diversity analysis using denaturing gel gradient electrophoresis (DGGE; Trial 1). DGGE using universal primers revealed no differences in overall microbial diversity between the treatments and placebo. Bands on the gel are visualised by silver staining.
  • FIG. 22 shows microbial diversity analysis using DGGE. DGGE using lactic acid bacteria (LAB) specific primers revealed significant differences in LAB diversity between treatment with GGDK266 and placebo in both caecal and ileal samples. Bands on the gel are visualised by silver staining.
  • FIG. 23 shows microbial diversity analysis using DGGE. DGGE using lactic acid bacteria (LAB) specific primers revealed significant differences in LAB diversity between treatment with GGDK266 and placebo in ileal samples. Bands on the gel are visualised by silver staining.
  • FIG. 24 shows microbial diversity analysis using DGGE. DGGE using lactic acid bacteria (LAB) specific primers revealed significant differences in LAB diversity between treatment with GGDK266 and placebo in caecal samples. Bands on the gel are visualised by silver staining.
  • FIG. 25 shows the gene ontology biological processes significantly down-regulated by oral administration of GGDK266.
  • FIG. 26 shows changes in immune response and response to stimuli in animals treated with GGDK266 versus animals treated with placebo (percent of genes versus a range of different GO annotations).
  • FIG. 27 shows the gene ontology biological processes significantly enriched by oral administration of GGDK266.
  • EXAMPLES Materials and Methods
  • Materials: Pig faeces samples collected during the course of the study of outdoor- and indoor-reared pigs (Mulder et al, 2009) were used in these studies. The culture collection was based primarily on LAB collected from frozen samples 411, 412 and 416, which were from outdoor-reared pigs with particularly high levels of LAB in their faeces. MRS broth premix, agar and vancomycin, anaerobe gas packs and indicator and antibiotic discs were purchased from Oxoid, anaerobe catalyst from Fisher Scientific and cysteine-HCL, bromocresol green and skimmed milk powder from Sigma-Aldrich. Pig colostrum carbohydrate fractions were prepared as part of the SMART 163 programme of D. Kelly. DNA extraction kits were purchased from MP Biomedicals and PCR reagents and clean-up kits from Promega. API CH 50 kits were purchased from Biomerieux UK Ltd.
  • Standard media: MRS broth and MRS agar were prepared according to the manufacturer's instructions. LAMVAB agar was prepared according to the method of Jackson et al. (2002). Agar plates were prepared immediately before use. MRS broth was decanted (10 ml per tube) into sterile Hungate tubes under anaerobic conditions and stored at room temperature.
  • Carbohydrate-supplemented media: SMART 163 ammonium sulphate precipitate of pig colostrum: precipitated at 0, 20, 25, 30, 35, 45, 50, 55 or 65% saturation or soluble at 65% saturation were weighed out in proportion to the amounts recovered from 15 ml or 50 ml of colostrum. Carbohydrate fractions were each dispersed in 15 ml of MRS or LAMVAB agar, held at 45° C., and then individual plates were poured for each fraction. They were also dispersed in MRS broth (50 ml) and the supplemented broth decanted to eight (6 ml/tube) sterile Hungate tubes under anaerobic conditions.
  • Animals: Female C3H/HeN and C57BI/6 mice (5-6 weeks old) were purchased from Harlan UK. They were housed as groups or pairs in standard caging within HEPA-filtered flexifilm isolators situated in a class 2 containment facility. They had free access to a high quality rodent chow and sterile deionised water at all times and were allowed to acclimatise for 7 to 10 days prior to commencement of experiments. The Rowett Institute of Nutrition and Health (RINH) is licensed under the UK Animals (Scientific Procedures) Act 1986. Studies herein were carried out under the auspices of an approved Home Office Project Licence by staff holding the requisite Home Office Personal Licence (as defined and set out in the UK Animals (Scientific Procedures) Act 1986), and were reviewed and approved by the RINH Ethical Review Committee.
  • Methods
  • Culture of LAB: In initial studies, a small amount of frozen faeces (100 mg) was dispersed in 1 ml of maximum recovery diluent (MRD). Two further sequential ten-fold dilutions were made. All three suspensions were streaked out on MRS or LAMVAB agar plates. In later studies, the faeces sample was dispersed in 5 ml of MRD, further diluted (1:40) in MRD and 0.5 ml of this dilution spread over the surface of MRS or LAMVAB agar plates with or without supplemental pig colostrum carbohydrates. In all cases, the plates were incubated in an anaerobic jar for 72 hours at 37° C. Distinct colonies (at least 8 per plate) were picked off the agar plates and seeded into Hungate tubes containing MRS broth or where appropriate MRS broth containing pig colostral carbohydrates. The tubes were incubated for 48 hours at 37° C.
  • Frozen stock: An aliquot (0.7 ml) of each culture was drawn off with a sterile syringe and needle and dispensed into a plastic tube that was flushed with CO2 and contained 0.3 ml glycerol and 2 mg L-cysteine. The tube was sealed with a plastic stopper, labelled, the contents mixed, frozen and stored at −80° C.
  • Conditioned medium: The remaining culture was transferred to a Corning 15 ml centrifuge tube, centrifuged at 1000 g×5 min at room temperature, the supernatant decanted, aliquoted and frozen. The pellets were either extracted immediately for 16S rRNA gene analysis or frozen.
  • 16S rRNA gene analysis (Clarridge, 2004): Bacterial DNA was extracted using a FastDNA® Spin kit for Soil in conjunction with a Fastprep 120 bead beater system, according to the protocol supplied with the kit. PCR was carried out (reaction mix: buffer, 104 dNTPs (2 mM), 5 μl. 27F Primer (20 pmol/2 μl. 1492R Primer (20 pmol/μl)). 2 μl Go Taq Flexi Polymerase, 0.5 μl). MgCl2, 5 μl. H2O, 23.5 μl and 2 μl of extracted DNA) using MJ Research PTC-200 Peltier Thermal Cycler run through 35 cycles of 95° C. for 3 minutes, 95° C. for 30 seconds, 57° C. for 30 seconds and 72° C. for two minutes. Primer: 27F (F01) AGAGTTTGATCCTGGCTCAG; 1492R (RP2) ACGGCTACCTTGTTACGACTT. PCR product cleanup was done with a Wizard® SV Gel and PCR Clean-up kit (Promega), used according to the manufacturer's instructions. 16S PCR products were sequenced using fully automated genetic analysers based on capillary electrophoresis technology (Genomics Section, RINH, UoA) using the reverse and forward primers 519R and 926F. Bacterial strains were identified by comparison of sequences with known bacterial DNA sequences using BLAST (http://blast.ncbi.nlm.nih.gov/Blast.cgi).
  • Antibacterial activity: XLD agar was prepared as per manufacturer's instructions and cooled to 45° C. Salmonella enteritidis S1400 was added to the XLD agar [1 ml of a 1:1000 dilution of an overnight culture of salmonella in 200 ml XLD agar to give the equivalent of 106 CFU/ml]. The agar was poured into petri dishes and allowed to set. The plates were marked off into 4 quadrants and an approximately 5 mm well cut out in each quadrant. An aliquot (60 μl) of conditioned media or MRS broth was added to the wells. The plates were covered and incubated for 16 hours at 37° C. They were photographed using a digital camera. Images transferred to Photoshop, and the diameter of the well and zone of inhibition were determined using the measure tool. Values were calculated and stored on an Excel spreadsheet. The same procedure was used with Escherichia coli K88, except that MacConkey No 3 agar was used.
  • Antibiotic susceptibility: Pig LAB [0.5 ml of a 1:100 dilution of an overnight culture] was spread onto the surface of an MRS agar [90 mm] plate and dried off. The plates were marked off into 4 quadrants and in each quadrant was placed an antibiotic-containing disc [Ampicillin, 10 μg. Cefotaxime, 30 μg. Chloramphenicol, 10 μg. Erythromycin, 15 Gentamicin, 10 μg Kanamycin, 30 μg. Metronizadole, 50 μg. Nalidixic acid, 30 μg. Tetracycline, 30 μg. Vancomycin, 30 μg]. The plates were covered, placed in an anaerobic jar and incubated for 24 hours at 37° C. They were photographed using a digital camera. Images transferred to Photoshop, and the diameter of the zone of inhibition was determined using the measure tool. Values were calculated and stored on an Excel spreadsheet.
  • Prevention of adherence/invasion by salmonella in vitro: Monolayers of IPEC-J2 cells were grown to 3 days post-confluence in 24-well plates and synchronised by the addition of DTS media 24 hrs prior to use. Overnight cultures of pig LAB (10 ml) were centrifuged [1000 g×5 min at room temperature] and the bacteria re-suspended in 1 ml of phosphate buffered saline [PBS]. An aliquot (50 μl) of LAB was added to the wells. The plates were incubated for 2 hours at 37° C., 5% CO2, 95% humidity. An overnight culture of Salmonella enterica serovar Enteritidis S1400 [S. enteritidis S1400] was sub-cultured (0.5 ml in 10 ml) into Luria Bertani (LB) media and incubated aerobically for 2-3 hours at 37° C. until it reached an optical density (560 nm) of 0.8. This gave a concentration equivalent to 1×108 CFU/ml. The culture was centrifuged [1000 g×5 min at room temperature], the bacteria re-suspended in 10 ml of PBS. An aliquot (50 μl) was added to the wells of IPEC-J2 cells. Wells treated with PBS were used as controls. The plates were incubated for a further 2 hours at 37° C., 5% CO2, 95% humidity. The IPEC-J2 cell monolayers were washed 5 times with HBSS. A solution (0.5 ml) of PBS containing Triton-X100 (10 ml/litre) was added to each well, the monolayer scraped off and dispersed. Viable salmonella were estimated on XLD agar plates [incubated for 24 hours at 37° C.] by the Miles and Misra method [Robertson et al, 2003]. LAB were determined by the same procedure [incubated anaerobically for 48 hours at 37° C.].
  • Inhibition of inflammatory responses: Monolayers of IPEC-J2 cells were grown to 3 days post-confluence in 24-well plates and synchronised by the addition of DTS media 24 hrs prior to use. Overnight cultures of pig LAB (10 ml) were centrifuged [1000 g×5 min at room temperature] and the bacteria re-suspended in 1 ml of PBS. An aliquot (50 μl) of LAB was added to each well [3 wells for each sample] along with 220 ng 12-O-Tetradecaboylphorbol-13-acetate [PMA] per well. PMA or PBS alone served as controls. The plates were incubated for 2 hours at 37° C., 5% CO2, 95% humidity. Culture media was removed from the dishes and the cells washed twice with PBS. RLT buffer (0.5 ml) containing mercaptoethanol was added to each well, the cells scraped off and transferred to an eppendorf tube [for each sample scrapings from 3 wells were combined]. RNA extraction was done using RNeasy® Mini kit in accordance with the manufacturer's protocols and reverse transcription with a high capacity cDNA Reverse Transcription Kit (Applied Biosystems). Real Time PCR was done on a 7500 Fast Real-time PCR system operating with 7500 Fast System v 1.4.0 Sequence Detection Software version 1.4 (Applied Biosystem). Primers for porcine IL-8 and TNF-α [IPEC-J2, SY100604186-096 IL-8-2 Reverse, SY100604186-090 TNF1 a Reverse, SY100604186-095 IL-8 2 Forward, SY100604186-089 TN Fa 1 Forward, and SY100604186-093] were prepared by Sigma Aldrich. The reaction mix was: 10 μl Power Sybergreen Master mix, 2.5 μl of forward primer, 2.5 μl of reverse primer and 5 μl of cDNA, The Real Time PCR was then run according to the Standard 7500 protocol [95° C., 10 min, 1 cycle. 95° C., 15 sec, 40 cycles. 60° C., 1 min, 40 cycles. 95° C., 15 sec, 1 cycle. 60° C., 1 min, 1 cycle. 95° C., 15 sec, 1 cycle. 60° C., 15 sec, 1 cycle]. Expression of IL-8 and TNF-α genes were analysed and compared to that of the ‘house-keeping’ gene β-actin. For comparison, values were given as the ratio of IL-8 and TNF-α per β-actin or fold-change.
  • For example:
  • a. Calculate ΔCt (2h) for IL-8 [Ct IL-8 minus Ct β-actin]
  • b. Calculate ΔCt (2h) for PMA [Ct PMA minus Ct β-actin]
  • c. Divide ΔCt (IL-8) with ΔCt (PMA)
  • d. Round up value to whole number
  • Substrate reactivity: The carbohydrate reactivity of individual LAB was determined using an API CH 50 kit (Biomerieux UK Ltd). Assays were done according to the manufacturer's instructions and reactions were recorded after incubation for 24 and 48 hours at 37° C. There are 50 capules on an API CH 50 plate. These contain various potential substrates and negative controls. The range of substrates is as follows: Monosaccharides 16, Monosaccharides/alcohols 4, Disaccharides 8, Trisaccharides 2, Polysaccharides 3, Alcohols 6, Others 7. For each substrate group the number of positive reactions is counted. This is divided by the maximum possible to give the rank for that substrate group. The sum of all the substrate scores gives the overall ranking for the bacterium. High Ranking indicates broad spectrum of substrate reactivity
  • Heat-treatment of LAB: A small amount of frozen faeces (100 mg) was dispersed in 5 ml of maximum recovery diluent (MRD). Sediment was allowed to settle out and the upper layer was decanted into eppendorf tubes (1 ml/tube). The tubes were heated at 50° C., 60° C. or 70° C. for 10 min. An aliquot (0.4 ml) of each was plated out on MRS agar and incubated in an anaerobic jar for 72 hours at 37° C. A small number of colonies were detected after heating at 70° C. Distinct colonies were picked off, seeded into Hungate tubes containing MRS broth and incubated for 48 hours at 37° C.
  • In a second study, a small amount of frozen faeces (100 mg) was dispersed in 5 ml of maximum recovery diluent (MRD). Sediment was allowed to settle out and the upper layer was decanted into eppendorf tubes (1 ml/tube). The tubes were heated at 50° C. for 20 min, 50° C. for 20 min plus 60° C. for 20 min or 50° C. for 20 min plus 60° C. for 20 min plus 70° C. for 20 min. An aliquot (0.5 ml) of each was plated out on MRS agar and incubated in an anaerobic jar for 48 hours at 37° C. A small number of colonies were detected, picked off, seeded into Hungate tubes containing MRS broth and incubated for 48 hours at 37° C.
  • In the third study, an overnight culture (10 ml) of isolated pig LAB was centrifuged (1000 g×5 min at room temperature), the pellet re-suspended in fresh MRS broth (10 ml). An aliquot (1 ml) was heated at 70° C. for 15 min and then plated out (0.5 ml) out on MRS agar and incubated in an anaerobic jar for 48 hours at 37° C. A small number of colonies were detected, picked off, seeded into Hungate tubes containing MRS broth and incubated for 48 hours at 37° C. This culture was centrifuged, re-suspended in MRS broth, heated again at 70° C. for 15 min, plated out on MRS agar, incubated in an anaerobic jar for 48 hours at 37° C., picked off, seeded into Hungate tubes containing MRS broth and incubated for 48 hours at 37° C. As before, this culture was centrifuged, re-suspended in MRS broth, re-heated at 70° C. for 15 min, plated out (0.5 ml) out on MRS agar, incubated in an anaerobic jar for 48 hours at 37° C., picked off, seeded into Hungate tubes containing MRS broth and incubated for 48 hours at 37° C.
  • Stability of freeze dried bacteria: Overnight cultures of LAB were centrifuged (1000 g×5 min at room temperature. Pellets were re-suspended in 2 ml sterile PBS and re-centrifuged. The subsequent pellets were then re-suspended in 5 ml of freezing solution [defatted skimmed milk powder (SKP), 100 g/1; SKP+lactose, both 100 g/1; SKP+sucrose, both 100 g/1; or SKP, 200 g/1]. The samples were frozen at −20° C. (2-3 hours) and then stored at −80° C. overnight. They were freeze-dried for 48 hours and dried material stored at room temperature. Viable bacteria in the samples were determined at 0 and approximately 40 and 80 days after completion of freeze drying. They were plated out on MRS agar and incubated anaerobically for 48 hours at 37° C.
  • Bulk preparation of GGDK31 and GGDK266: Two 500 ml batches of MRS broth were prepared in 500 ml glass screw-top bottles, autoclaved and allowed to cool to room temperature (in proximity to gas flame) whilst being flushed with CO2. Four ml of a 24 hour culture of GGDK31 or GGDK266 was added to each bottles of MRS and the lids lightly closed. The bottles were placed in an anaerobic jar and incubated at 37° C. for 24 hours. The culture was centrifuged [1000 g×5 min at room temperature] in 6 sterile 50 ml centrifuge tubes. The supernatant was discarded, tubes refilled with culture and re-centrifuged until all the bacteria had been recovered. Each of the 6 tubes contained almost equal amounts of bacteria. The bacteria in each tube were re-suspended in 40 ml of sterile PBS, re-centrifuged and the supernatant discarded. The bacteria in each tube was re-suspended in 20 ml of SKM (100 g/1), frozen at −20° C. (2-3 hours) and then overnight at −80° C., freeze-dried for 48-72 hours and stored at 4° C. To evaluate viable bacteria in the sample, one tube of freeze dried material was re-suspended in 20 ml of MRS broth, incubated at room temperature for 2 hours, diluted, plated out on MRS agar and incubated anaerobically for 48 hours at 37° C.
  • L. mucosae in vivo Study 1: Sixteen (6 week) old female C3H/HeN mice were dosed with an overnight culture of vial 323 (L. mucosae; 50 μl; >109 CFU) at day −7, −4, −2 and 0 and daily thereafter up to day +9. A further 16 mice (control) were given media. On day 0, eight mice (L. mucosae-treated) and eight control mice were given, by gavage, a single dose of Salmonella enteritidis S1400 (50 μl;≥108 CFU). In addition, eight mice (L. mucosae-treated) and eight control mice were given a single dose of culture medium. Body weight and health score were monitored twice daily post-salmonella infection. The mice were euthanased (isoflurane overdose and exsanguination) and dissected at 10 days post-salmonella infection. Stomach, representative portions of jejunum and ileum, caecum plus contents, colon plus contents, spleen and liver and one kidney and the mesenteric lymph node were collected under near aseptic conditions for microbiology. Representative portions of upper jejunum, mid jejunum, ileum, caecum and ascending and descending colon were placed in neutral buffered formalin or RNA-later and stored for future analysis.
  • L. mucosae in vivo Study 2: Five (6 week) old female C57BI/6 mice were dosed with an overnight culture of vial 323 (L. mucosae; 50 μl; >109 CFU) at day −7, −4, −2 and 0 and daily thereafter up to day+5. A further 5 mice were given media. On day 0, all ten mice were given, by gavage, a single dose of Salmonella enteritidis S1400 (50 μl; >107CFU). The mice were euthanased and dissected on day 6, according to the procedure for study 1.
  • Novel pig LAB in vivo: Four (6 week) old female C3H/HeN mice were dosed with an overnight culture of RINH vial 31 (L. reuteri; 50 μl; >109 CFU), four with RINH vial 32 (L. reuteri). Four with vial 323 (L. mucosae), four with RINH vial 46 (L. reuteri), four with RINH vial 47 (L. reuteri) and eight with MRS. This was done at day −6, −4, −2 and 0 and daily thereafter up to day+9. On day 0, all lactobacilli-treated mice and four control mice were given, by gavage, a single dose of Salmonella enteritidis S1400 (50 μl;≥108 CFU). In addition, the remaining four control mice were given a single dose of culture medium. The mice were euthanased and dissected on day 10, according to the procedure for study 1.
  • Microbiology: Tissues were homogenised [1:100 w/v] in MRD using a Janke-Kunkel Ultra-Turrax T25 tissue homogeniser at 20,000 rpm for 30 seconds, as were jejunal and ileal contents. Up to eight sequential dilutions (1: 10 v/v) of the primary homogenates were made, plated out onto XLD agar and MacConkey No. 3 agar and incubated overnight at 37° C. Viable counts were estimated as before [Robertson et al, 2003].
  • Statistical analysis: Where appropriate data were initially assessed by one-way analysis of variance (ANOVA) regarding treatment outcome. If ANOVA indicated that there were significant differences (p<0.05) amongst all groups, the data was then analysed by the Tukey-Kramer Multiple Comparisons Test or the Kruskal-Wallis Multiple Comparisons Test as appropriate. This was done using the Instat Statistical Package (GraphPad Software Inc., San Diego, USA).
  • Based on the outputs from the multiple comparison tests, means in tables or graphs were marked with superscript letters. Means that differed significantly from each other (p<0.05) were allocated distinct superscript letters. Means that did not differ significantly from each other were allocated common superscript letters.
  • Results 1. Isolation of LAB
  • Faeces from organically-reared piglets were plated out on selective agars and were incubated under anaerobic conditions. From all studies, a total of 436 individual colonies of Lactic Acid Bacteria [LAB] were picked off, seeded into MRS broth and incubated under anaerobic conditions. Each culture was given a unique RINH vial number and an aliquot was frozen down in MRS media containing 30% glycerol and L-cysteine (˜2 mg/ml) and stored at −80° C. 16S rRNA gene analysis was done and bacterial strains were identified by comparison of sequences with known bacterial DNA sequences (Table 1).
  • The majority of the cultured LAB colonies were L. johnsonii and L. johnsonii-related strains [L. johnsonii, L. johnsonii/gasseri, L. johnsonii/taiwanensis] (240/436) and L. reuteri or L. reuteri-related [L. reuteri, L. reuteri/pontis, L. reuteri/vaginalis, L. reuteri/acidophilus (169/436)]. There were 7 L. plantarum/pentosus colonies, 19 other species and 5 uncultured strains.
  • 2. Anti-Salmonella Activity In Vitro
  • Conditioned media from isolated LAB were screened for anti-bacterial activity against Salmonella enteritidis S1400 using a well-diffusion assay (FIG. 1).
  • Conditioned media from individual colonies of LAB varied greatly in their activity against S. enteritidis (FIG. 2A). This was not strain dependent. The range of anti-salmonella activities amongst L. johnsonii was similar to that amongst L. reuteri. On an arbitrary basis, the cultures were separated into groupings on the basis of their capacity to inhibit salmonella in vitro (FIG. 2B [[2 b]]). Group 1 had <20000 units of inhibition, Group 2 20000-40000 units of inhibition, Group 3 40000-60000 units of inhibition, Group 4 60000-80000 units of inhibition, Group 5 80000-100000 units of inhibition and Group 6>>100000 units of inhibition (FIG. 2B Group 1 comprised of 14 strains (3.4% of total), Group 2 of 95 strains (22.8%), Group 3 of 99 strains (23.7%), Group 4 of 99 strains (23.7%), Group 5 of 86 strains (20.6%) and Group 6 of 24 strains (5.8%). The latter group comprised of seventeen L. johnsonii and L. johnsonii-related, six L. reuteri or L. reuteri-related strains and one uncultured strain.
  • 3. Anti-E. coli K88 Activity In Vitro
  • Conditioned media from LAB were also screened for anti-Escherichia coli K88 activity by the well diffusion assay. Activity against E. coli K88, as with salmonella, varied greatly between individual colonies of LAB (FIG. 3A). The range and variation in the activity was similar amongst the L. johnsonii and L. reuteri strains. In general, there was no direct correlation between the anti-salmonella and anti E. coli K88 activities for any individual LAB (FIG. 3C, 3D). However of the ten strains in E. coli K88 group 5 (FIG. 3B), seven had relatively high activities against both pathogens, two had high activity against E. coli K88 but moderate activity against salmonella and one was active primarily against E. coli K88.
  • 4. Initial Selection of Candidate LAB
  • Thirty-three strains were identified for further testing in vitro (Table 2).
  • These comprised 18 L. johnsonii and L. johnsonii-related strains, 11 L. reuteri or L. reuteri-related and 4 L. plantarum and L. plantarum-related strains (Table 2a).
  • 5. Attachment I invasion of Pig Intestinal Epithelial [IPEC-J2] Cells
  • The capacity of LAB to block adhesion/invasion of IPEC cells by S. enteritidis and E. coli K88 was evaluated (FIG. 4A, 4B,). The candidate LAB all greatly reduced attachment and invasion of IPEC cells by salmonella. Most of them were also very effective against E. coli K88. However, 3 of the strains had only limited effects on adhesion/invasion of IPEC cells by E. coli K88.
  • 6. Susceptibility of LAB to Antibiotics.
  • The susceptibility of the candidate LAB to a range of antibiotics was evaluated (Table 4, FIG. 5). All but one strain (RINH vial 266) exhibited some degree of resistance to individual antibiotics. All were susceptible to ampicillin (10 μg), cefotaxime (30 μg) and chloramphenicol (10 μg). The majority were susceptible to erythromycin (15 gentamicin (10 μg), tetracycline (30 μg) and vancomycin (30 μg). Most strains were resistant to metronizadole (50 μg) and nalidixic acid (30 μg) and to a lesser extent kanamycin (30 μg). 23
  • 7. Refined Selection of Candidate LAB
  • Twenty-three high ranking strains were identified for further testing in vitro.
  • 8. Substrate Specificity of LAB
  • The candidate LAB were screened for substrate reactivity using an API CH 50 kit (Table 5, 6, FIG. 6). L. johnsonii, L. reuteri and L. plantarum each exhibited strain-specific general substrate reaction profiles. In addition, most strains of each genotype exhibited fine differences in their substrate reactivity, indicative that they were unique individual strains.
  • 9. Suppression of Inflammation in Pig Intestinal Epithelial [IPEC-J2] Cells
  • The ability of candidate LAB to block or suppress inflammatory responses triggered in IPEC cells by 12-O-Tetradecaboylphorbol-13-acetate [PMA] was tested (FIGS. 7A-7C; Table 7). The candidate strains varied greatly in their capacity to block interleukin-8 (IL-8) gene-expression triggered by PMA. Five strains ( RINH vial 29, 30, 31 86 and 266) had potent anti-inflammatory effects.
  • 10. Final Selection of Candidate LAB
  • Fourteen strains were identified having killing and blocking activities against salmonella and E. coli K88, susceptibility to antibiotics carbohydrate reactivity and capacity to suppress inflammation in vitro. Seven of these were particularly preferred. The latter set comprised 4 L. plantarum-related, 3 L. johnsonii-related and one L. reuteri. Two of these LAB strains [GGDK266 and GGDK31] were prepared in bulk for evaluation in a trial with newly-weaned piglets (Table 8).
  • 11. Freeze Drying and Storage of LAB
  • The survival and viability of LAB after freeze drying in skimmed milk powder [SKP], SKP plus lactose or SKP plus sucrose was evaluated (FIGS. 8A and 8B). FIG. 8A depicts the stability of L. reuteri and FIG. 8B depicts the stability of L. johnsonii. Small losses in viability were evident on storage for 42 and 84 days at room temperature of samples dried in SKP. This was less marked when skimmed milk powder and sugars were used in combination. However, the 24 latter preparations tended to be hygroscopic and difficult to maintain. Bulk preparations of GGDK266 and GGDK31 were therefore prepared by drying the bacteria in skimmed milk powder [100 g/1] (Table 8).
  • 12. Heat-Treatment Studies
  • Suspensions of faeces from organically reared pigs were heat treated for varying periods of time at 50-70° C., plated out on MRS agar, colonies picked off and cultured in MRS broth [RINH vial 417-506]. The strain types recovered were variable and clostridium species formed a high proportion, the isolated strains remained sensitive to heat.
  • Isolated cultures of LAB were subject to heating three times for 15 minutes at 70° C. (FIGS. 9A-9C). Viable bacteria decreased by 3-4 log orders after heat-treatment for the first time. However, the surviving bacteria had a degree of heat-resistance. With one exception, losses of viable bacteria were low when the bacteria were re-cultured and re-heated a further two times.
  • Heat-treatment three times at 70° C. altered the biological activities of the strains (FIGS. 9A-9C). RINH vial 521 (vial 255 heat-treated) was not able to block attachment of pathogens to IPEC cells and the capacity of RINH vial 520 (vial 230 heat-treated) to prevent attachment was reduced. The ability of RINH vial 517 (vial 31 heat-treated) to abolish inflammatory responses triggered in IPEC cells was abolished. In contrast, the biological properties of RINH vial 518 (vial 85 heat-treated) and RINH vial 519 (vial 86 heat-treated) were similar to those of the native strains.
  • 13. Mouse Infection Studies
  • 13.1 L. mucosae (RINH Vial 323)
  • C3H/HeN mice develop a persistent but non-lethal, intestinal and systemic infection, which has many characteristics of the major form of human salmonellosis, when challenged with high levels of Salmonella enteritidis S1400. In contrast, C57BI/6 mice develop a severe primarily systemic, infection, reminiscent of acute infection in humans, when challenged with the same pathogen. To evaluate the capacity of L. mucosae (vial 323) to ameliorate salmonellosis, C3H/HeN and C57BI/6 mice were treated with L. mucosae prior to and post-challenge with Salmonella enteritidis (FIGS. 10, 13). The mice were euthanased and dissected 6 (C57BI/6) or 10 (C3H/HeN) days post-infection.
  • Systemic tissues: Oral treatment with L. mucosae limited the capacity of S. enteritidis to cause systemic infection both in C3H/HeN and C57BI/6 mice (FIG. 11A-11C; 14A-14C[[11a-c; 14a-c]]). High numbers of viable salmonella were detected in the mesenteric lymph node, liver and spleen of mice. In contrast, the numbers present in these tissues were greatly reduced if the mice had been co-treated with RINH vial 323 (L. mucosae). Salmonella infection caused enlargement of the spleen (FIG. 12A; 15). This tissue response was significantly reduced in mice treated with both RINH vial 323 (L. mucosae) and salmonella.
  • Intestine: Intestinal myeloperoxidase [MPO], a marker for neutrophils, was determined in C3H/HeN mice treated with salmonella or salmonella plus RINH vial 323 (L. mucosae). MPO in the intestine was greatly increased by salmonella infection, due to recruitment of neutrophils to the intestine part of the host response to infection (FIG. 12B[[12 b]]), Co-treatment with RINH vial 323 (L. mucosae) reduced MPO activity in the intestine of salmonella-infected mice, indicating that the intestinal inflammatory responses to infection were lowered in these animals.
  • 13.2 Novel pig LAB
  • Four LAB were selected: RINH vial 31, RINH vial 32, RINH vial 46 and RINH vial 47 (All L. reuteri; LR31, LR 32, LR 36 and LR47 respectively). To assess their efficacy to ameliorate a pathogen infection, C3H/HeN mice were treated with these LAB or RINH vial 323 (L. mucosae, LM] prior to and post-challenge with Salmonella enteritidis (FIG. 16). The mice were euthanased and dissected 10 days post-infection. Faecal excretion of S. enteritidis was reduced, if the mice had been co-treated with LAB (FIG. 17A, 17B). LR31 and LR32 tended to have the greatest effects on faecal salmonella outputs.
  • Intestine: Treatment with LR31, LR32, LM, LR46 or LR47 significantly reduced the numbers of salmonella in the caecum (FIG. 18A). Furthermore, LR31, LR32, LR46 and LR47 but not LM lowered salmonella numbers in the colon (FIG. 18B). The reductions tended to be greater with LR31 and LR32. In contrast to the large intestine, the LAB had no significant effects on numbers of salmonella in the small intestine.
  • Systemic tissues: Treatment with LR31, LR32, LM, LR46 or LR47 greatly reduced the numbers of salmonella detected in the spleen and liver (FIGS. 19A-19C). The reductions were more marked with LR31 and LR32 than with LM, LR46, or LR47. Salmonella numbers in the mesenteric lymph node were lowered following treatment with LR31, LR32 and LR46 but not with LM or LR47.
  • Discussion
  • The LAB strains isolated (total of 436 individual colony picks) from faeces of organically-reared pigs were predominantly L. reuteri, L. johnsonii, L. gasseri, L. pentosus, strains with a small number of L. plantarum, L. acidophilus, L. vaginalis, a single L. mucosae and several uncultured strains. Most of the LAB produced substances that could inhibit the growth of S. enteritidis and/or E. coli K88 in vitro. The potency of these anti-pathogen effects varied greatly between the individual bacterial strains. A proportion of LAB had high activity against S. enteritidis but low activity against E. coli K88 and vice-versa, but the majority had similar activities against both pathogens.
  • Thirty-three strains were selected on the basis of anti-microbial potency as determined in vitro. These bacteria were further screened for their ability to block adherence/invasion of intestinal pig epithelial cells (IPEC) by pathogens in vitro and their susceptibility to antibiotics.
  • Twenty-three strains were assayed for substrate range and specificity and their capacity to suppress inflammation in IPEC cells in vitro. From these, fourteen LAB (5 L. johnsonii, 6 L. reuteri and 3 L. plantarum) with particularly favourable properties were identified.
  • Two LAB strains [GGDK266 and GGDK31] were prepared in bulk for in vivo evaluation in newly-weaned piglets. Other potentially important candidate strains were present in this set of 14 LAB.
  • The survival and viability of LAB after freeze drying in various solutions was also evaluated. Small losses in viability were evident on prolonged storage of samples dried with skimmed milk powder. This was less marked when skimmed milk powder and sugars were used. However, the latter preparations were hygroscopic and were difficult to maintain. It was therefore decided to use a skimmed milk powder suspension for freeze drying and storage of LAB. The bulk preparations of GGDK266 and GGDK31 were freeze-dried in this medium.
  • Heat stability is a useful feature for LAB to be used in pelleted animal foods. Five heat-conditioned viable strains of isolated pig LAB were obtained. However, the biological properties in vitro and probiotic potential of three of the strains were adversely affected by heat-treatment. Nonetheless, two of the bacteria retained the biological properties of their native non-heat-treated forms.
  • Five pig LAB (L. reuteri [4] or L. mucosae [1]) were tested for ability to ameliorate salmonellosis in vivo. Treatment of mice with these LAB greatly reduced the pathogenicity of S. enteritidis.
  • 14. Evaluation of Oral Administration of Organic Lactobacilli Probiotic Strains on the modulation of the Gut Microbiota and Performance of early Weaned Pigs
  • In vivo trials were carried out on early weaned piglets to test the effect of two probiotic strains according to the invention, Lactobacilli strains GGDK266 and GGDK31.
  • Trial Design Animals:
      • 24 Large—White×Redon piglets
      • Early weaned (21 days old, ≈7-8 kg), born in a local farm
      • Weighted then distributed equally between the different group
      • 3 experimental treatments (n=8):
        • A—Basal diet+Placebo
        • B—Basal diet+probiotic GDDK 266−dose 10×1012
        • C—Basal diet+probiotic GDDK 31—dose 10×1012
      • Observation period: 14 days
    Diet:
  • Diets based on barley, wheat & soybean meal
      • Feed composition
  • Barley 36.5
    Wheat 21
    SBM 48 19
    Corn 10
    Soy oil 4
    Sugar 4
    Potato protein 2
    Premix 3.5
      • feed ad libitum in pelleted form
    Tissue Sampling and Measurements
    • Sampling: Day 0 Slaughter of 6 “naive” piglets for collection of the caecum Individual collection of faeces (if possible)
      • Day 7 individual collection of faeces during weight measurement
      • Day 14 Slaughter of 24 piglets for collection of:
  • Content (5 g): Tissus (10 cm):
    Gastric Jejunum
    Jejunum Ileum
    Ileum Caecum
    Caecum Lymphatic nodes
    (distal ileum level)
    • Storage: All samples were weighed, frozen in liquid nitrogen and stored at −80° C.
    • Performance: Daily Weight gain (DWG), Feed Intake (FI) and Feed Conversion Ratio (FCR)
      (1st step)
    • Analysis: Determination of the microbiota profile in the different gut content samples by
    • (2nd step) the molecular microbiology technique Denaturing gradient gel electrophoresis (DGGE).
      • Molecular analysis of gene expression data using pig affymetrix gene expression arrays to determine gene modulation patterns.
      • Determination of immunity markers in intestinal tissues
        Microbial analysis using Denaturing Gel Gradient Electrophoresis DGGE (Trial 1)
        DGGE methodology
  • DNA is extracted from faecal or tissue samples utilizing the MP Bio FastDNA™ spin kit for soil sample—116560000. The DNA is then amplified using Muyzer primers, as it is essential to use primers with a GC Clamp to be run on the gel. For samples of lactobacillus, specialised lactobacillus primers with a GC clamp were used.
  • Amplicon Annealing DGGE
    Target Primer Sequence Size temperature gradient
    Group Primer (5′-3′) (bp) (° C.) (%)
    All MF ATTACCGCGGCTGCTGG 233 55 35-70
    Bacteria MR-GCa GC-Clamp-
    CCTACGGGAGGCAGCAG
    LABs Lac1 AGCAGTAGGGAATCTTCCA 327 55 30-50
    Lac2-GCa GC-Clamp-
    ATTYCACCGCTACACATGc
  • Annotations:
  • aThe GC clam is as follows:
  • CGCCCGCCGCGCGCGGCGGGCGGGGCGGGGGCACGGGGGG
  • cY=C or T
  • PCR Program:
  • Time Temperature Cycles
    5 minutes 94° C. 1
    30 seconds 94° C. 35
    30 seconds 55° C.
    2 minutes 72° C.
    10 minutes 73° C. 1
  • DGGE is a genetic analysis technique in which amplified PCR products are separated by the denaturants formamide and urea within the gel, based on the genetic sequence by as little as a single base difference. DGGE can be utilised to visualise the differences in microbial diversity between samples. DNA obtained from a range of samples can be used in DGGE e.g. tissue and faecal samples. Bands on the gel were visualised using silver staining.
  • Molecular analysis and gene expression profiles of pig tissues
    RNA extraction and Affymetrix Microarray analysis
  • RNA was isolated from both animal tissue and cultured cells for use on Affymetrix GeneChips. For animal tissue, approximately 200 mg tissue sample was removed from RNAlater (Ambion) and lyzed in Trizol (Invitrogen) using a polytron homogenizer. The tissue was further homogenized by passing the lysate through a syringe fitted with a 19G needle 3-5 times. The samples were incubated for 5 min at RT to permit the complete dissociation of nucleoprotein complexes. Then, chloroform, isopropanol and ethanol steps were performed according to the manufacturer's instructions. Briefly, 0.2 mL of chloroform was added per 1 mL of Trizol, vortexed and incubated at RT for 5 min. The samples were centrifuged at 12,000×g for 15 min at 4° C. The resultant aqueous phase was transferred to a fresh tube, and the RNA was precipitated by the addition of 0.5 mL of isopropanol per 1 mL of Trizol. The tubes were shaken vigorously by hand for 10s, incubated at 4° C. for 10 min and centrifuged at 12,000×g for 10 min at 4° C.
  • The RNA precipitate was washed with ice-cold 75% ethanol, adding at least 1 mL of 75% ethanol per 1 mL of Trizol. The samples were vortexed and centrifuged at 7,400×g for 5 min at 4° C. After air-drying the resultant RNA pellet, the RNA was resuspended in up to 100 μL RNase-free water. Total RNA was further extracted with the RNeasy kit (Qiagen) according to the manufacturer's instructions, including an RNase-free DNase I (Qiagen) digestion step.
  • Cultured cells were homogenized by adding 350 μL Buffer RLT+1% β-mercaptoethanol. The cells were scraped off culture dishes with a filter tip and further homogenized by passing the lysate through a syringe fitted with a 19G needle 3-5 times. The cell lysate was then further processed using the RNeasy kit (Qiagen) according to the manufacturer's instructions, including an RNase-free DNase I (Qiagen) digestion step.
  • RNA concentration and integrity was ascertained using a Nanodrop instrument and/or Agilent Bioanalyzer, and purified RNA was stored at −70° C.
  • 250 ng RNA was processed for Affymetrix GeneChips using the GeneChip 3′ IVT Express Kit (Affymetrix) according to the manufacturer's instructions. aRNA quality was determined by Agilent 2100 Bioanalyzer. Hybridization to the GeneChip Mouse Genome 430 2.0 and GeneChip Human Genome U133 Plus 2.0 (Affymetrix) on a GeneChip Fluidics Station 450 (Affymetrix) was performed at the Institute of Medical Sciences Microarray Core Facility (University of Aberdeen, UK). Chips were scanned with an Affymetrix GeneChip Scanner 3000 (Affymetrix). Image quality analysis was performed using Gene Chip Operating Software (GCOS) (Affymetrix). Further quality analysis, normalization (gcRMA), statistical analysis and heatmap generation was performed with the freely available software packages R (http://www.r-project.org) and Bioconductor (http://www.bioconductor.org). Microarray data were submitted to the National Center for Biotechnology Information (NCBI) Gene Expression Omnibus (http://www.ncbi.nlm.nih.gov/geo).
  • Results Performance of Pigs Fed Probiotics GGDK266 and GGDK31
  • The results for pigs fed probiotics GGDK266 and GGDK31 are shown in FIG. 20. DWG (Daily weight gain), FI (food intake) and FCR (feed conversion ratio) are shown below:
  • GGDK266 DWG FI FCR
    D 0-d 7  +++ (*) + +
    D 7-d 14 = + +
    D 0-d 14 + + +
  • Piglets fed GGDK266 exhibited significantly improved daily weight gain (DWG) during the first week post-weaning relative to GGDK31 and placebo fed piglets.
  • Microbial diversity analysis using DGGE (Trial 1)
  • DGGE using universal primers revealed no differences in overall microbial diversity between the treatments and placebo (see FIG. 21).
  • DGGE using lactic acid bacteria (LAB) specific primers revealed significant differences in LAB diversity between treatment with GGDK 266 and placebo in both caecal and ileal samples (see FIG. 22).
  • DGGE using LAB specific primers revealed significant differences in LAB diversity between the treatment with GGDK266 and placebo in ileal samples (see FIG. 23).
  • DGGE using LAB specific primers revealed significant differences in LAB diversity between the treatment with 266 and placebo in caecal samples (see FIG. 24).
  • Overall the microbial diversity analysis revealed significant clustering of the LAB population in piglets fed GGDK266 indicating that the populations in individual animals on this treatment has a similar and stable microbiota.
  • Molecular analysis of ileal tissue samples: Affymetrix pig arrays
    Downregulated in GDK266 versus placebo
  • Gene ontology analysis of differentially expressed gene revealed that a significant reduction in immune system processes and pro-inflammatory activation in response to feeding young piglets probiotic GGDK266 relative to placebo (see FIG. 25).
  • Results reveal that GGDK266 had a very specific and targeted effect on the immune system and the functional groups associated with response to stimuli (see FIG. 26).
  • Upregulated in GGDK266 versus Placebo
  • In contrast to the effects on the immune system, GGDK266 promoted metabolic processes particularly in relation to nitrogen (see FIG. 27). Without wishing to be bound by theory, it is believed that these effects may explain the improved DWG in animals fed GGDK266.
  • Top differentially expressed genes between GGDK266 and Placebo
  • affy. id Gene Name Product FC p-value
    Ssc.645.1.S1_at CSTA Cystatin A 44.06 0.00000
    Ssc.11608.1.A1_at TIP_HUMAN T-cell immunomodulatory protein precursor 28.92 0.00030
    Ssc.10837.1.A1_at ROBO1 Roundabout homolog 1 precursor 13.35 0.00178
    Ssc.8960.1.A1_at BPI Bactericidal permeability-increasing protein 11.65 0.00476
    precursor
    Ssc.16234.1.S1_at TCN1 Transcobalamin 1 precursor 11.48 0.00023
    Ssc.1411.1.S1_at THBS4 Thrombospondin 4 precursor 8.92 0.00198
    Ssc.837.1.A1_at BPI Bactericidal permeability-increasing protein 4.55 0.00573
    precursor
    Ssc.30008.1.A1_at ESR1 Estrogen receptor 4.48 0.00053
    Ssc.13539.1.A1_at PLAGL1 Zinc finger protein PLAGL1 4.42 0.00881
    Ssc.26324.1.S1_at NP_981932 Iodotyrosine dehalogenase 1 protein 4.26 0.00200
    Ssc.29413.1.A1_at B3GALT2 UDP-Gal:betaGlcNAc beta 1,3- 4.00 0.00046
    galactosyltransferase 2
    Ssc.27410.1.S1_at MYCN N-myc proto-oncogene protein 3.80 0.00261
    Ssc.25176.1.A1_at GOLPH4 Golgi phosphoprotein 4 3.80 0.00009
    Ssc.15890.1.S1_at VNN1 Pantetheinase precursor 3.61 0.00271
    Ssc.23427.1.A1_at CYB561 Cytochrome b561 3.29 0.01512
    Ssc.16186.1.S1_at CD3E T-cell surface glycoprotein CD3 epsilon chain −2.62 0.00764
    precursor
    Ssc.22676.1.S1_at CXCR6 C-X-C chemokine receptor type 6 −2.63 0.01652
    Ssc.15565.1.S1_at LCP2 Lymphocyte cytosolic protein 2 −2.76 0.00024
    Ssc.18652.1.S1_at IL16 Interleukin-16 precursor −2.97 0.01132
    Ssc.181.1.S1_at TRGV9 T-cell receptor gamma chainV region PT- −3.04 0.01615
    gamma-1/2 precursor
    Ssc.23489.1.S1_at CD8A T-cell surface glycoprotein CD8 alpha chain −3.08 0.00071
    precursor
    Ssc.428.6.S1_a_at TCA_HUMAN T-cell receptor alpha chain C region −3.15 0.00027
    Ssc.10357.1.A1_at FMN2 Formin 2 −3.46 0.00308
    Ssc.27354.1.S1_at STXBP5 Tomosyn −3.88 0.02438
    Ssc.28909.3.A1_at TPH2 Tryptophan 5-hydoxylase 2 −4.36 0.00717
    Ssc.25976.1.S1_at GZMH Granzyme H precursor −5.46 0.00179
    Ssc.11070.1.S1_at IGHM Ig alpha-1 chain C region −9.07 0.00115
    Ssc.16566.1.S1_at LCT Lactase phlorizin hydrolase precursor −11.31 0.00328
    Ssc.13273.1.A1_at GCNT3 glucosaminyl (N-acetyl) transferase 3, mucin type −19.75 0.00016
    Ssc.11098.1.S1_at IFITM3 Interferon-induced transmembrane protein 3 −51.36 0.00044
  • Gene expression data revealed that a number of genes were significantly increased including antimicrobial peptides (eg. CSTA, BP1) and immune-regulatory genes (TIP). In contrast GGDK266 reduced the expression of a diverse panel of genes involved in pro-inflammatory immunity (IFITM3, IL-16).
  • CONCLUSIONS
      • Cellular and metabolic processes, particularly in relation to nitrogen, are increased in animals treated with GGDK266 relative to placebo.
      • Immune system processes are downregulated in animals treated with GGDK266 relative to placebo. Examples include T-cell markers CD3 and CD8, T cell receptor chains, chemokines/cytokines and IFN-related genes.
      • Animals administered with GGDK266 exhibited a stable population of lactic acid bacteria revealed by clustering of the bacterial profile of the individual induced by the administration of probiotic GGDK266.
      • FCR and performance were significantly improved during the first weeks of post-weaning life.
      • This improvement in growth performance correlated with the reduction in inflammatory immune responses and the increase in specific metabolic processing.
  • Various modifications and variations of the described aspects of the invention will be apparent to those skilled in the art without departing from the scope and spirit of the invention. Although the invention has been described in connection with specific preferred embodiments, it should be understood that the invention as claimed should not be unduly limited to such specific embodiments. Indeed, various modifications of the described modes of carrying out the invention which are obvious to those skilled in the relevant fields are intended to be within the scope of the following claims.
  • TABLE 1
    Summary of bacteria colonies selected from cultures
    of faeces from organically-reared pigs.
    Total number of cultured colony picks 443
    Media:
    LAMVAB agar 55
    LAMVAB agar + pig colostral carbohydrate 88
    MRS agar 29
    MRS agar + pig colostrum carbohydrate 176
    Glucose-free MRS agar + carbohydrate 57
    MRS agar after heat-treatment at up to 70° C. 38
    Main strains identified:
    Lactobacillus reuteri
    Lactobacillus johnsonii
    Lactobacillus plantarum
    Five isolated LAB were heated once, twice or three times
    at 70° C. for 15 min.
    Surviving bacteria were re-grown.
    In stock
    5 LAB heated once at 70° C.
    5 LAB heated twice at 70° C.
    5 LAB heated three times at 70° C.
  • TABLE 2
    Candidate LAB strains for further study selected on the basis of killing
    activity in well diffusion assays (note 266 and 161 contain LR)
    Pathogen killing (units)
    RINH Well diffusion assay
    Vial no. anti-SE anti-KSS
    85 LR 129886 60168
    255 LJ 101477 64390
    266 LJ 101335 60168
    436 LJ 81656 85010
    161 LP 77894 103346
    12 LJ 162709 42977
    16 LJ 117621 41365
    29 LR 174471 45720
    31 LR 116867 46907
    86 LR 98520 75147
    230 LJ 95705 64340
    256 LJ 94012 77459
    314 LJ 103497 48936
    361 LJ 100770 40254
    17 LJ 144765 23072
    30 LR 125463 36050
    32 LR 168892 32572
    258 LP 70724 68612
    260 LP 78197 68562
    320 LJ 66350 78044
    364 LJ 99137 55123
    433 LJ 95083 51461
    15 LP 77459 58669
    218 LJ 62329 50416
    220 LJ 68612 53834
    356 LJ 72986 55302
    363 LJ 79125 45555
    131 LR 42223 44108
    434 LR 10000 81656
    166 LJ 17064 79621
    431 LR 48657 31674
    47 LR 20722 34633
    46 LR 19867 34633
    LJ. L. johnsonii,
    LR. L. reuteri,
    LP. L. Plantarum
  • TABLE 2a
    Identification of candidate LAB strains (by 16S rRNA gene
    sequence) selected on the basis of killing activity in
    well diffusion assays (note 266 and 161 contain LR)
    RINH forward reverse
    Vial no. sequence sequence
    85 Lactobacillus reuteri Lactobacillus reuteri
    255 Lactobacillus johnsonii, Lactobacillus johnsonii,
    taiwanensis, acidophilus gasseri
    266 Lactobacillus johnsonii Lactobacillus johnsonii
    436 lactobacillus johnsonii Lactobacillus johnsonii
    str. 466 F19785
    161 Lactobacillus plantarum, Lactobacillus plantarum,
    pentosus, paraplantarum pentosus
    12 Lactobacillus johnsonii, Lactobacillus johnsonii,
    gasseri, taiwanensis gasseri
    16 Lactobacillus johnsonii, Lactobacillus johnsonii
    gasseri, taiwanensis
    29 Lactobacillus reuteri, Lactobacillus reuteri
    pontis, vaginalis,
    frumenti
    31 Lactobacillus reuteri Lactobacillus reuteri
    86 Lactobacillus reuteri Lactobacillus reuteri
    230 Lactobacillus johnsonii, Lactobacillus johnsonii
    taiwanensis, acidophilus
    256 Lactobacillus johnsonii, Lactobacillus johnsonii
    taiwanensis, acidophilus
    314 lactobacillus johnsonii uncultered bacterium
    BR0315
    361 lactobacillus johnsonii lactobacillus johnsonii
    str. NCC2822 F19785
    17 Lactobacillus johnsonii, Lactobacillus johnsonii
    gasseri, taiwanensis
    30 Lactobacillus reuteri, Lactobacillus reuteri
    pontis
    32 Lactobacillus reuteri Lactobacillus reuteri
    258 Lactobacillus plantarum, Lactobacillus plantarum,
    pentosus, helveticus pentosus, paraplantarum
    260 Lactobacillus plantarum, Lactobacillus pentosus,
    pentosus, paraplantarum plantarum, paraplantarum
    320 lactobacillus johnsonii Lactobacillus johnsonii
    NCC2822 F19785
    364 lactobacillus johnsonii lactobacillus johnsonii
    466 F10785
    433 lactobacillus johnsonii lactobacillus johnsonii
    str. CECT 289 F19785
    15 Lactobacillus plantarum, Lactobacillus plantarum,
    pentosus pentosus
    218 Lactobacillus johnsonii, uncultured Firmicutes,
    taiwanensis Lactobacillus johnsonii
    220 Lactobacillus johnsonii, uncultured Firmicutes,
    taiwanensis Lactobacillus johnsonii
    356 lactobacillus johnsonii lactobacillus johnsonii
    NCC2822 F19785
    363 lactobacillus johnsonii lactobacillus johnsonii
    466 F10785
    131 Lactobacillus reuteri Lactobacillus reuteri
    434 Lactobacillus reuteri lactobacillus reuteri
    NM99-1
    166 Lactobacillus johnsonii, Lactobacillus johnsonii
    taiwanensis, acidophilus
    431 lactobacillus reuteri lactobacillus reuteri
    str. Probio-16 JCM 1112
    47 Lactobacillus reuteri Lactobacillus reuteri
    46 Lactobacillus reuteri Lactobacillus reuteri
  • TABLE 3
    Candidate LAB strains for further study selected on the
    basis of killing activity in well diffusion assays and
    capacity to block adherence of pathogen to IPEC cells
    RINH Inhibition of adherence (%)
    Vial no. SE KSS
    85 88.31 87.93
    255 82.37 99.93
    266 88.03 98.09
    161 98.32 96.94
    12 96.89 99.92
    29 93.7 99.91
    31 98.64 99.75
    86 81 99.98
    256 82.47 99.92
    361 85.07 99.44
    17 84.56 99.66
    30 96.44 99.91
    32 87.74 99.86
    230 78.89 82.45
    258 96.37 86.5
    260 90.22 88.79
    314 79.68 94.2
    433 99.99 96.23
    16 87.68 45.38
    218 91.53 86.49
    363 85.61 99.93
    364 82.13 78.12
    15 79.19 99.52
    131 95.5 96.03
    220 91.04 78.6
    320 92.7 44.17
    356 82.15 78.4
    434 94.78 98.85
    436 99.97 1
    166 91.45 95.97
    431 96.35 86.47
    47 90.47 99.47
    46 83.51 99.7
  • TABLE 4
    Area of inhibition of LAB by defined amounts of antibitiotic (arbitrary units)
    ampicillin cefotaxime chloramphenicol erythromycin gentamicin kanamycin metronidazole nal. acid tetracycline vancomycin
    12 244011 340402 186699 13151 0 0 0 0 37668 22581
    15 277117 311725 204282 214008 0 0 0 0 0
    16 266033 294166 187805 64681 17000 7157 0 0 0 105209
    17 387224 400570 235430 277145 9193 0 0 0 50328 117741
    29 410335 444193 190293 114511 0 0 0 0 252497 11483
    30 292728 335927 77133 208117 31261 0 0 0 187805 31402
    31 334789 410966 165904 262226 38221 0 0 0 214037 24901
    32 404496 402291 247436 350238 71608 23786 0 0 261979 10691
    46 359232 402588 210421 251461 29550 0 0 0 21382 25069
    47 328283 410579 185515 270105 30342 0 0 0 211556 22231
    85 356114 369916 204992 309439 0 0 0 0 276800 3971
    86 250812 381270 183399 250805 41858 0 31264 0 16643 13355
    131 349955 473065 248521 123562 82466 14932 0 0 19354 7479
    161 338497 412977 258724 261133 51991 4536 29126 0 20435 5542
    166 268783 417393 185508 251607 61136 17671 0 0 24606 0
    218 209117 271547 148617 0 0 0 0 0 88668 122870
    220 209371 319970 165815 34230 58814 32572 0 0 34636 111666
    230 254614 335143 164405 51078 65717 45705 0 0 36644 41991
    255 330364 392169 217758 59224 56563 8486 0 0 29872 0
    256 456892 502325 228531 71258 93058 0 0 0 20955 42203
    258 401257 271932 195909 233326 28608 0 0 0 223143 0
    260 286400 364573 203796 33393 78821 78364 0 0 21757 62792
    266 287070 322869 198614 247085 54008 3079 6437 2737 48286 107882
    314 297057 332853 154830 44115 0 0 0 0 90259
    356 291920 339895 203692 62656 10472 5890 0 0 24194 8202
    361 320695 323713 201886 234140 0 0 0 0 91863
    363 275304 308159 193271 44491 86683 0 0 0 28212 18146
    364 288514 341651 194320 143978 45880 0 0 0 18322 103995
    431 339016 380459 226484 311725 74991 0 0 0 0 26302
    433 241710 203588 174124 63381 19139 0 0 0 19965 79034
    434 198112 261065 172223 68052 6049 0 0 0 60344 45863
    436 290458 287331 185812 142842 0 0 0 0 52279 61810
    Nal. Acid, naladixie acid.
  • TABLE 5
    Substrates in capsules of API CH 50 Kit
    Substrates in capules of API CH 50 kit
    1 glycerol polyol
    2 erythritol polyol
    3 D-arabinose monosaccharide
    4 L-arabinose monosaccharide
    5 D-ribose monosaccharide
    6 D-xylose monosaccharide
    7 L-xylose monosaccharide
    8 D-adonotol alcohol
    9 Methyl-βD-Xylopyranoside cyclic
    10 D-galactose monosaccharide
    11 D-glucose monosaccharide
    12 D-fructose monosaccharide
    13 D-mamose monosaccharide
    14 L-sorbose monosaccharide
    15 L-rhamose monosaccharide
    16 dulcitol monosaccharide/alcohol
    17 inositol polyol
    18 D-mamitol polyol
    19 D-sorbitol sugar/alcohol
    20 Methyl-αD-Mannopyranoside cyclic
    21 Methyl-αD-Glucopyranoside cyclic
    22 N-acetylglucosamine monosaccharide
    23 amygdalin glycoside
    24 arbutin glycoside
    25 esculin ferric citrate
    26 salicin glycoside
    27 D-cellobiose disaccharide
    28 D-maltose disaccharide
    29 D-lactose (bovine) disaccharide
    30 D-Melibiose disaccharide
    31 D-saccharose disaccharide
    32 D-trehalose disaccharide
    33 inulin polysaccharide
    34 D-melezitose trisaccharide
    35 D-rafinose trisaccharide
    36 amidon (starch) polysaccharide
    37 glycogen polysaccharide
    38 xylitol monosaccharide/alcohol
    39 gentobiose disaccharide
    40 D-turanose disaccharide
    41 D-lyxose monosaccharide
    42 D-tagatose monosaccharide
    43 D-fucose monosaccharide
    44 L-fucose monosaccharide
    45 D-arabitol monosaccharide/alcohol
    46 L-arabitol monosaccharide/alcohol
    47 potassium gluconate sequestrant
    48 potassium 2-ketogluconate sequestrant
    49 potassium 5-ketogluconate sequestrant
  • TABLE 6
    Substrate profile of LAB using an API CH 50 kit
    alcohol/
    monosaccharides monosaccharides disaccharides trisaccharides polysaccharides alcohols others
    17 0.4 0.0 0.8 0.5 0.3 0.0 0.4
    30 0.2 0.0 0.0 0.0 0.0 0.0 0.3
    31 0.3 0.0 0.6 0.5 0.0 0.0 0.6
    32 0.3 0.0 0.5 0.5 0.0 0.0 0.3
    46 0.2 0.0 0.5 0.5 0.0 0.0 0.4
    47 0.2 0.3 0.5 0.5 0.0 0.0 0.4
    85 0.1 0.0 0.3 0.0 0.0 0.0 0.4
    86 0.3 0.0 0.5 0.5 0.0 0.0 0.4
    131 0.4 0.0 1.0 1.0 0.3 0.3 0.9
    161 0.7 0.3 0.9 1.0 0.0 0.3 0.9
    166 0.4 0.0 0.6 0.0 0.7 0.0 0.3
    220 0.1 0.0 0.5 0.5 0.0 0.0 0.4
    230 0.3 0.0 0.8 0.5 0.0 0.0 0.4
    255 0.1 0.0 0.6 1.0 0.3 0.0 0.4
    256 0.2 0.0 0.5 1.0 0.0 0.2 0.6
    258 0.6 0.3 1.0 1.0 0.7 0.7 0.9
    260 0.4 0.3 0.9 1.0 1.0 0.0 0.6
    266 0.3 0.0 0.9 0.5 0.3 0.0 0.4
    320 0.3 0.0 0.5 0.5 0.3 0.0 0.3
    363 0.4 0.0 0.8 1.0 0.3 0.0 0.4
    364 0.4 0.0 0.8 0.5 0.3 0.0 0.4
    433 0.2 0.0 0.5 0.0 0.0 0.2 0.3
  • TABLE 7
    Candidate LAB strains selected on the basis of killing activity,
    capacity to block adherence of pathogen to IPEC cells, antibiotic
    susceptibility, substrate reactivity and ability to suppress
    inflammation (note 266 and 161 contain LR)
    RINH forward reverse
    Vial no. sequence sequence
    266 Lactobacillus johnsonii Lactobacillus johnsonii
    31 Lactobacillus reuteri Lactobacillus reuteri
    258 Lactobacillus plantarum, Lactobacillus plantarum,
    pentosus, helveticus pentosus, paraplantarum
    260 Lactobacillus plantarum, Lactobacillus pentosus,
    pentosus, paraplantarum plantarum, paraplantarum
    255 Lactobacillus johnsonii, Lactobacillus johnsonii,
    taiwanensis, acidophilus gasseri
    161 Lactobacillus plantarum, Lactobacillus plantarum,
    pentosus, paraplantarum pentosus
    256 Lactobacillus johnsonii, Lactobacillus johnsonii
    taiwanensis, acidophilus
    86 Lactobacillus reuteri Lactobacillus reuteri
    85 Lactobacillus reuteri Lactobacillus reuteri
    32 Lactobacillus reuteri Lactobacillus reuteri
    230 Lactobacillus johnsonii, Lactobacillus johnsonii
    taiwanensis, acidophilus
    131 Lactobacillus reuteri Lactobacillus reuteri
    30 Lactobacillus reuteri, Lactobacillus reuteri
    pontis
    364 lactobacillus johnsonii 466 lactobacillus johnsonii F10785
  • TABLE 8
    Identity for pig LAB strains selected for bulk
    preparation (note 266 and 161 contain LR)
    Bacteria Bacteria
    RINH Seq code identified Seq code identified
    vial no primer 926F by BLAST primer 519R by BLAST
    GGDK266
    266 S10CM218 Lactobacillus S10CM171 Lactobacillus
    johnsonii johnsonii
    GGDK31
    31 S10BL123 Lactobacillus S10BL141 Lactobacillus
    reuteri reuteri
  • 31 
    Figure US20220080003A1-20220317-P00899
     123 with 926F
    SEQ ID NO: 1
    GGTGGAGCATGTGGTTTAATTCGAAGCTACGCGAAGAACCTTACCAGGTCTTGACATCTTGCGCTAACCTTAGAGATAAG
    GCGTTCCCTTCGGGGACGCAATGACAGGTGGTGCATGGTCGTCGTCAGCTCGTGTCGTGAGATGTTGGGTTAAGTCCCGC
    AACGAGCGCAACCCTTGTTACTAGTTGCCAGCATTAAGTTGGGCACTCTAGTGAGACTGCCGGTGACAAACCGGAGGAAG
    GTGGGGACGACGTCAGATCATCATGCCCCTTATGACCTGGCCTACACACGTGCTACAATGGACGGTACAACGAGTCGCAA
    GCTCGCGAGAGTAAGCTAATCTCTTAAAGCCGTTCTCAGTTCGGACTGTAGGCTGCAACTCGCCTACACGAAGTCGGAAT
    CGCTAGTAATCGCGGATCAGCATGCCGCGGTGAATACGTTCCCGGGCCTTGTACACACCGCCCGTCACACCATGGGAGTT
    TGTAACGCCCAAAGTCGGTGGCCTAACCATTATGGAGGGAGCCGCCTAAGTGCGGGACAGATGACTGGGGTGAAGTCGTA
    ACAAGGTAGCCTGTATTTTCTTGCGGTTGTTCCCCCCCCNGGCGGGACTGCCTTACTCCTTTCACCNCCCGCGCCCCTGG
    AGGGGGCCGGAACCCCCCTCCCAACCCCCCTAACCCACCTCCTTCCTTTTAACCNGCT
    31 
    Figure US20220080003A1-20220317-P00899
     141 with 519R
    SEQ ID NO: 2
    GACTTTCTAGGTTGGATACCGTCACTGCGTGAACAGTTACTCTCACGCACGTTCTTCTCCAACAACAGAGCTTTACGAGC
    CGAAACCCTTCTTCACTCACGCGGTGTTGCTCCATCAGGCTTGCGCCCATTGCGGAAGATTCCCTAGTGCTGCCTCCCGT
    AGGAGTATGGACCGTGTCTCAGTTCCATTGTGGCCGATCAGTCTCTCAACTCGGCTATGCATCATCGCCTTGGTAAGCCG
    TTACCTTACCAACTAGCTAATGCACCGCAGGTCCATCCCAGAGTGATAGCCAAAGCCATCTTTCAAACAAAAGCCATGTG
    GACTTTCTTGTTATGCGGTATTAGCATCTGTTTCCAAATGTTATCCCCCGCTCCGGGGCAGGTTACCTACGTGTTACTCA
    CCCGTCCGCCACTCACTGGTGATCCATCGTCAATCAGGTGCAAGCACCATCAATCAGTTGGGCCAGTGCGTACGACTTGC
    ATGTATTAGGCACACCGCCGGCGTTCATCCTGAGCCATGATCAAACTCTANGCGTCAGTTTTACGGTCTCGGCTCGTTTC
    TCTGTTNTCTGACATCAACGTGCGTTACATTTGCGGTTTACGCATTGATTGTACTCCCTCCACATAGGTGGCGGCATACC
    CTTCGTGCTCCTCTACTCATCTCGTTCATTACAACTCGCTTTGTTACCTTCCCGGTGGGGTTCTCTACCTCCTTCGTTTT
    CTCTCACCTCATTCTCTCTCCCATCCTCTCNCTTTCCTCTTGCTC
    161 
    Figure US20220080003A1-20220317-P00899
     282 WITH 926F
    SEQ ID NO: 3
    GGTGGAGCATGTGGTTTAATTCGAAGCTACGCGAAGAACCTTACCAGGTCTTGACATACTATGCAAATCTAAGAGATTAG
    ACGTTCCCTTCGGGGACATGGATACAGGTGGTGCATGGTTGTAGTCAGCTCGTGTCGTGAGATGTTGGGTTAAGTCCCGC
    AACGAGCGCAACCCTTGTTATCAGTTGCCAGCATTAAGTTGGGCACTCTGGTGAGACTGCCGGTGACAAACCGGAGGAAG
    GTGGGGATGACGTCAAATCATCATGCCCCTTGATGACCTGGGCTAGACACGTGCTACAATGGATGGTACAACGAGTTGCG
    AACTCGCGAGAGTAAGCTAATCTCTTAAAGCCATTCTCAGTTACGGATGTGTAGGCTGCAACTCGCCATACATGAAGTCG
    GAATCGCTAGTAATCGCGGATACAGCATGCCGCGGTGAATACTGTTCCCGGGCCTATGTGACACACCGCCCGTCACACCA
    TGAGCAGTTTGTAATCACCCACACAGTCGGTGGGGTAACCTTTATAGGAACCAGCCGCCTACAGTGCGGGACCGATGATT
    ATGGGTGCACTCGTATCACTGTAACTTAAACCCTTGCGGCCGTACTCCCCAGGCGGAATGCTTAATACGTTACCTGCAAC
    CCTGAAGGGCGGAATCCCTCCAACGATTATCAAT
    161 
    Figure US20220080003A1-20220317-P00899
     300 WITH 519R
    SEQ ID NO: 4
    GTGGCTTTCTGGTTAAATACCGTCAATACCTGAACAGTTACTCTCAGATATGTTCTTCTTTAACAACAGAGTTTTACGAG
    CCGAAACCCTTCTTCACTCACGCGGCGTTGCTCCATCAGACTTTCGTCCATTGTGGAAGATTCCCTACTGCTGCCTCCCG
    TAGGAGTTTGGGCCGTGTCTCAGTCCCAATGTGGCCGATTACCCTCTCAGGTCGGCTACGTATCATTGCCATGGTGAGCC
    GTTACCCCACCATCTAGCTAATACGCCGCGGGACCATCCAAAAGTGATAGCCGAAGCCATCTTTCAAGCTCGGACCATGC
    GGTCCAAGTTGTTATGCGGTATTAGCATCTGTTTCCAGGTGTTATCCCCCGCTTCTGGGCAGGTTTCCCACGTGTTACTC
    ACCAGTTCGCCACTCACTCAAATGTAAATCATGATGAAGCACCAATCAATACCAAGTTCGTTCGACTTGCATGTATTA
    GGCACGCCGCCAGCGTTCGTCGCTGAGCCATGATCAAACTACTAAAGGCCCCCNATGCCTCCCACCCGCTTTGTTGCCGG
    GGCCCCCCGTTCCCATACCCCTTTTGGACGTTTTCCAGCCCCTTGGCGGGCCCTGTACCTCCCCCCAGGGCGGGGAATGC
    CTTAATTGCGTTNACCTTGCACCCCCTGAAGGGGCGGAATCCCTCCAACGATTACCT
    255 
    Figure US20220080003A1-20220317-P00899
     504 with 925F
    SEQ ID NO: 5
    GGTGGAGCATGTGGTTTAATTCGAAGCAACGCGAAGAACCTTACCAGGTCTTGACATCCAGTCGCATAACCTAAGAGATT
    AGGTGTTCCCTTCGGGGACGCTGAGACAGGTGGTGCATGGCTGTCGTCAGCTCGTGTCGTCACATGTTGGGTTAAGTCCC
    GCAACGAGCGCAACCCTTGTCATTAGTTGCCATCATTAAGTTGGGCACTCTAATGAGACTGCCGGTGACAAACCGGAGGA
    AGGTGGGGATGACGTCAAGATCATCATGCCCCTTATGACCTGGGCTACACACGTGCTAGAATGGACGGTACAACGAGATA
    GCGAACCTGCGAAGAGCTAAGCGGATCTCTTAAAGCCGTTCTCAGTTCGGACTGTAGGCTGCAACTCGCCTACACGAAGC
    TTGGAATCGCTAGTAATGGGGGATCAGCACTGCCGCGGTGAATACGTTCCCGGGCCTTGTACACACCGCCCGTCACACCA
    TGAGAGTCTGTAACTCCCAAAGTCGGTGGGATAACCTTCTATAGCGAGTGAGTCCGTTCGATGGGTAGGGACAAGATGAA
    TGAGCGGTGAAAGGTCGTTAAACCAAGGGTAGCAAGTAAGGATCCCTTTGGGGGTTTTATCTCCACGGGGGGGGTGTTTC
    TTTTCTGTCTTTA
    255 
    Figure US20220080003A1-20220317-P00899
     530 with 519R
    SEQ ID NO: 6
    ACTTTCTAGAGTTAGATGATACCGTTCAACATGACAGATGGCCACGTTTACTTACTCTCACTGACTACTGTTCTTTCATC
    TCACACAACAGAGCTTTACGAGCCGAAACCCTTCTTCACTCACGCGGCGTTGCTCCATCAGAGCTTTGCGTCCCATTGTG
    GAACATTCCCTACTGCTGCCTCCCGTAGGAGTATGGGCCGTGTCTCAGTCCCATTGTGGCCGATCAGTCTCTCAACTCGG
    CTATGCATCATCGCCTTGGTAAGCCGTTACCTTACCAACTAGCTAATGCACCGCAGGTCCATCCAAGAGTGATAGCCGAA
    CCATCTTTCACAACTCTAAACATGCTTGTAGTGTTGTTATTCCGGTATTAACATTCTGTTTCCAGGTTGTTATTCCCAGC
    TGATCTCGGGGCAGGGTTTACCCCAACGTTGGTTTACCTTCACCCCCGGTTNCGGCCCGGCTTCGNCCTTGGGTTAGTAC
    TNACGATTCTGCTATTATATACGATGGGCTAGACGACCAGCCTAACACAATTTCAATTTCGTNAAGTGTCGAGAGGNCCT
    ACGGTCGTCCCGTTAACGTGTAGNCNATTTGGCTTATTTGTTAAGTTGTCCANCGGGCCACCGACCCCCAGGGCCCGGTT
    GGTCCGGGTTTCCCCCATTGCAACGTCGCCAAAGTGCGGAAATTTCGAAAATACCCTTAACCAATGAAAAAAACATA
    258 
    Figure US20220080003A1-20220317-P00899
     414 with 
    Figure US20220080003A1-20220317-P00899
    F
    SEQ ID NO: 5
    GGTGGAGCATGTGGTTTAATTCGAAGCTACGCGAAGAACCTTACCAGGTCTTGACATACTATGCAAATCTAAGAGATTAG
    ACGTTCCCTTCGGGGACATGGATACAGGTGGTGCATGGTTGTCGTCAGCTCGTGTCGTGAGATGTTGGGGTTAAGTCCCT
    CAACGAGCGCAACCCTTATTATCAGTTGCCAGCATTAAGTTGGGCACTCTGGTGAGACTGCCGGTGACAAACCGGAGGAA
    GGTGGGGATGACGTCAAATCATCATGCCCCTTATGACCTGGGCTACACACGTGCTACAATGGATGGTACAACGAGTTGCG
    AACTCGCGAGAGTAAGCTAATCTCTTAAAGCCATTCTCAGTTGGGATTGTAGGCTGCAACTCGCCTACATGAAGTCGGAA
    TCGCTAGTAATCGCGGATCAGCATGCCGCGGTGAATACGTTCCCGGGCCTTGTACACACCGCCCGTCACACCATGAGAGT
    TTGTAACACCCAAAGTGGGTGGGGGTAACCTTTTTAGGAAACCAGCCCGCCCTAAAGGGTGGGGAACAAGAATGAATTAA
    GGGGGTTGAAAAGTTCCGTTAAACCAAAAGGGGTTAGCCCCNGNTNNGANNNNNNNNNGAC
    258 
    Figure US20220080003A1-20220317-P00899
     438 with 519R
    SEQ ID NO: 8
    GCTTTCTGGTTAAATACCGTCAATACCTGAACAGTTACTCTCAGATATGTGTCTTCTTTAACAACAGAGTTTTACGAGCC
    GAAACCCTTCTTCACTCACGCGGCGTTGCTCCATCAGACTTTCGTCCATTGTGGAAGATTCCCTACTGCTGCCTCCCGTA
    GGAGTTTGGGCCGTGTCTCAGTCCCAATGTGGCCGATTACCCTCTCAGGTCGGCTACGTATCATTGCCATGGTGAGCCGT
    TACCCCACCATCTAGCTAATACGCCGCGGGACCATCCAAAAGTGATAGCCGAAGCCATCTTTCAAGCTCGGACCATGCGG
    TCCAAGTTGTTATGGGGTATTAGCATCTGTTTCCAGGGTGTTATTCCCCCGCTTGGTGGGCAGGGTTTCCCACGTGTTAC
    TCACCAGTTCGCCACTCACTCAAATGTAAATCATGATGCAAGCACCAATCAATACCAGAGTTCGTTCGACTTGCATGTAT
    TAGGCACGCCGCCAGCGTTCGTCCTGAGCCATGATCAAACTCNGA
    Figure US20220080003A1-20220317-P00899
    indicates data missing or illegible when filed
  • S12KG200 GGDK 31-2 22F
    SEQ ID NO: 10
    TGCCTAATACATGCAAGTCGTACGCACTGGCCCAACTGATTGATGGTGCT
    TGCACCTGATTGACGATGGATCACCAGTGAGTGGCGGACGGGTGAGTAAC
    ACGTAGGTAACCTGCCCCGGAGCGGGGGATAACATTTGGAAACAGATGCT
    AATACCGCATAACAACAAAAGCCACATGGCTTTTGTTTGAAAGATGGCTT
    TGGCTATCACTCTGGGATGGACCTGCGGTGCATTAGCTAGTTGGTAAGGT
    AACGGCTTACCAAGGCGATGATGCATAGCCGAGTTGAGAGACTGATCGGC
    CACAATGGAACTGAGACACGGTCCATACTCCTACGGGAGGCAGCAGTAGG
    GAATCTTCCACAATGGGCGCAAGCCTGATGGAGCAACACCGCGTGAGTGA
    AGAAGGGTTTCGGCTCGTAAAGCTCTGTTGTTGGAGAAGAACGTGCGTGA
    GAGTAACTGTTCACGCAGTGACGGTATCCAACCAGAAAGTCACGGCTAAC
    TACGTGCCAGCAGCCGCGGTAATACGTAGGTGGCAAGCGTTATCCGGATT
    TATTGGGCGTAAAGCGAGCGCAGGCGGTTGCTTAGGTCTGATGTGAAAGC
    CTTCGGCTTAACCGAAGAAGTGCATCGGAAACCGGGCGACTTGAGTGCAG
    AAGAGGACAGTGGAACTC
    S12KG201 GGDK 31-1 519F
    SEQ ID NO: 11
    TCGGATTTATTGGGCGTAAAGCGAGCGCAGGCGGTTGCTTAGGTCTGATG
    TGAAAGCCTTCGGCTTAACCGAAGAAGTGCATCGGAAACCGGGCGACTTG
    AGTGCAGAAGAGGACAGTGGAACTCCATGTGTAGCGGTGGAATGCGTAGA
    TATATGGAAGAACACCAGTGGCGAAGGCGGCTGTCTGGTCTGCAACTGAC
    GCTGAGGCTCGAAAGCATGGGTAGCGAACAGGATTAGATACCCTGGTAGT
    CCATGCCGTAAACGATGAGTGCTAGGTGTTGGAGGGTTTCCGCCCTTCAG
    TGCCGGAGCTAACGCATTAAGCACTCCGCCTGGGGAGTACGACCGCAAGG
    TTGAAACTCAAAGGAATTGACGGGGGCCCGCACAAGCGGTGGAGCATGTG
    GTTTAATTCGAAGCTACGCGAAGAACCTTACCAGGTCTTGACATCTTGCG
    CTAACCTTAGAGATAAGGCGTTCCCTTCGGGGACGCAATGACAGGTGGTG
    CATGGTCGTCGTCAGCTCGTGTCGTGAGATGTTGGGTTAAGTCCCGCAAC
    GAGCGCAACCCTTGTTACTAGTTGCCAGCATTAAGTTGGGCACTCTAGTG
    AGACTGCCGGTGACAAACCGGAGGAAGGTGGGGACGACGTCAGATCATCA
    TGCCCTTATGACCTGGGCTA
    S12K202 GGDK 31-1 92GF
    SEQ ID NO: 12
    GAGCATGTGGTTTAATTCGAAGCTACGCGAAGAACCTTACCAGGTCTTGA
    CATCTTGCGCTAACCTTAGAGATAAGGCGTTCCCTTCGGGGACGCAATGA
    CAGGTGGTGCATGGTCGTCGTCAGCTCGTGTCGTGAGATGTTGGGTTAAG
    TCCCGCAACGAGCGCAACCCTTGTTACTAGTTGCCAGCATTAAGTTGGGC
    ACTCTAGTGAGACTGCCGGTGACAAACCGGAGGAAGGTGGGGACGACGTC
    AGATCATCATGCCCCTTATGACCTGGGCTACACACGTGCTACAATGGACG
    GTACAACGAGTCGCAAGCTCGCGAGAGTAAGCTAATCTCTTAAAGCCGTT
    CTCAGTTCGGACTGTAGGCTGCAACTCGCCTACACGAAGTCGGAATCGCT
    AGTAATCGCGGATCAGCATGCCGCGGTGAATACGTTCCCGGGCCTTGTAC
    ACACCGCCCGTCACACCATGGGAGTTTGTAACGCCCAAAGTCGGTGGCCT
    AACCATTATGGAGGGAGCCGCCTAAGGCGGGACAGATGACTGGGGTGAAG
    TCGTAACAAGGTAGCCGTA
    S12KG203 GGDK 31-1 926R
    SEQ ID NO: 13
    CTCCCCAGGCGGAGTGCTTAATGCGTTAGCTCTCCGGCACTGAAGGGCGG
    AAACCCTCCAACACCTAGCACTCATCGTTTACGGCATGGACTACCAGGGT
    ATCTAATCCTGTTCGCTACCCATGCTTTCGAGCCTCAGCGTCAGTTGCAG
    ACCAGACAGCCGCCTTCGCCACTGGTGTTCTTCCATATATCTACGCATTC
    CACCGCTACACATGGAGTTCCACTGTCCTCTTCTGCACTCAAGTCGCCCG
    GTTTCCGATGCACTTCTTCGGTTAAGCCGAAGGCTTTCACATCAGACCTA
    AGCAACCGCCTGCGCTCGCTTTACGCCCAATAAATCCGGATAACGCTTGC
    CACCTACGTATTACCGCGGCTGCTGGCACGTAGTTAGCCGTGACTTTCTG
    GTTGGATACCGTCACTGCGTGAACAGTTACTCTCACGCACGTTCTTCTCC
    AACAACAGAGCTTTACGAGCCGAAACCCTTCTTCACTCACGCGGTGTTGC
    TCCATCAGGCTTGCGCCCATTGTGGAAGATTCCCTACTGCTGCCTCCCGT
    AGGAGTATGGACCGTGTCTCAGTTCCATTGTGGCCGATCAGTCTCTCAAC
    TCGGCTATGCATCATCGCCTTGGTAAGCCGTTACCTTACCAACTAGCTAA
    TGCACCGCAGGTCCATCCCAGAGTGATAGCCAAAGCCATCTTTCAAACAA
    AAGCCATGTGGCTTTTGTTGTTATGC
    S12KG204 GGDK 31-1 519R
    SEQ ID NO: 14
    TTTCTGGTTGGATACCGTCACTGCGTGAACAGTTACTCTCACGCACGTTC
    TTCTCCAACAACAGAGCTTTACGAGTCCGAAACCCTTCTTCACTCACGCG
    GTGTTGCTCCATCAGGCTTGCGCCCATTGTGGAAGATTCCCTACTGCTGC
    CTCCCGTAGGAGTATGGACCGTGTCTCAGTTCCATTGTGGCCGATCAGTC
    TCTCAACTCGGCTATGCATCATCGCCTTGGTAAGCCGTTACCTTACCAAC
    TAGCTAATGCACCGCAGGTCCATCCCAGAGTGATAGCCAAAGCCATCTTT
    CAAACAAAAGCCATGTGGCTTTTGTTGTTATGCGGTATTAGCATCTGTTT
    CCAAATGTTATCCCCCGCTCCGGGGCAGGTTACCTACGTGTTACTCACCC
    GTCCGCCACTCACTGGTGATCCATCGTCAATCAGGTGCAAGCACCATCAA
    TCAGTTGGGCCAGTGCGTACGACTTGCATGTATTAGGCACACCGCCGGCG
    TTCATCCTGAGCCATGATCAAAC
    S12KG205 KKDK 31-1 RP2
    SEQ ID NO: 15
    CCGCCTTAGGCGGCTCCCTCCATAATGGTTAGGCCACCGACTTTGGGCGT
    TACAAACTCCCATGGTGTGACGGGCGGTGTGTACAAGGCCCGGGAACGTA
    TTCACCGCGGCATGCTGATCCGCGATTACTAGCGATTCCGACTTCGTGTA
    GGCGAGTTGCAGCCTACAGTCCGAACTGAGAACGGCTTTAAGAGATTAGC
    TTACTCTCGCGAGCTTGCGACTCGTTGTACCGTCCATTGTAGCACGTGTG
    TAGCCCAGGTCATAAGGGGCATGATGATCTGACGTCGTCCCCACCTTCCT
    CCGGTTTGTCACCGGCAGTCTCACTAGAGTGCCCAACTTAATGCTGGCAA
    CTAGTAACAAGGGTTGCGCTCGTTGCGGGACTTAACCCAACATCTCACGA
    CACGAGCTGACGACGACCATGCACCAACTGTCATTGCGTCCCCGAAGGGA
    ACGCCTTATCTCTAAGGTTAGCGCAAGATGTCAAGACCTGGTAAGGTTCT
    TCGCGTAGCTTCGAATTAAACCACATGCTCCACCGCTTGTGCGGGCCCCC
    GTCAATTCCTTTGAGTTTCCACCTTGCGGTCGTACTCCCCAGGCGGAGTG
    CTTAATGCGTTAGCTCCGGCACTGAAGGGCGGAAACCCTCCAACACCTAG
    CACTCATCGTTTACGGCATGGACTACCAGGG

    NCIMB 41847 GGDK161—contains both Lactobacillus plantarum and Lactobacillus reuteri Lactobacillus plantarum
  • S12KG218 GGDK 161-1 27F
    SEQ ID NO: 16
    GTGCCTAATACATGCAAGTCGAACGAACTCTGGTATTGATTGGTGCTTGC
    ATCATGATTTACATTTGAGTGAGTGGCGAACTGGTGAGTAACACGTGGGA
    AACCTGCCCAGAAGCGGGGGATAACACCTGGAAACAGATGCTAATACCGC
    ATAACAACTTGGACCGCATGGTCCGAGTTTGAAAGATGGCTTCGGCTATC
    ACTTTTGGATGGTCCCGCGGCGTATTAGCTAGATGGTGAGGTAACGGCTC
    ACCATGGCAATGATACGTAGCCGACCTGAGAGGGTAATCGGCCACATTGG
    GACTGAGACACGGCCCAAACTCCTACGGGAGGCAGCAGTAGGGAATCTTC
    CACAATGGACGAAAGTCTGATGGAGCAACGCCGCGTGAGTGAAGAAGGGT
    TTCAGGCTCGTAAAACTCTGTTGTTAAAGAAGAACATATCTGAGAGTAAC
    TGTTCAGGTATTGACGGTATTTAACCAGAAAGCCACGGCTAACTACGTGC
    CAGCAGCCGCGGTAATACGTAGGTGGCAAGCGTTGTCCGGATTTATTGGG
    CGTAAAGCGAGCGCAGGCGTTTTTTAAGTCTGATGTGAAAGCCTTCGGCT
    CAACCGAAGAAGTGCATCGGAAACTGGGAAGCTTGAGTGCAGAAGAGGAC
    AGTGGAACTCCATGTGTAGCGGTGAAATGCGT
    S12KG219 GGDK 161-1 S19F
    SEQ ID NO: 17
    CGGATTTATTGGGCGTAAAGCGAGCGCAGGCGGTTTTTTAAGTCTGATGT
    GAAAGCCTTCGGCTCAACCGAAGAAGTGCATCGGAAACTGGGAAACTTGA
    GTGCAGAAGAGGACAGTGGAACTCCATGTGTAGCGGTGAAATGCGTAGAT
    ATATGGAAGAACACCAGTGGCGAAGGCGGCTGTCTGGTCTGTAACTGACG
    CTGAGGCTCGAAAGTATGGGTAGCAAACAGGATTAGATACCCTGGTAGTC
    CATACCGTAAACGATGAATGCTAAGTGTTGGAGGGTTTCCGCCCTTCAGT
    GCTGCAGCTAACGCATTAAGCATTCCGCCTGGGGAGTACGGCCGCAAGGC
    TGAAACTCAAAGGAATTGACGGGGGCCCGCACAAGCGGTGGAGCATGTGG
    TTTAATTCGAAGCTACGCGAAGAACCTTACCAGGTCTTGACATACTATGC
    AAATCTAAGAGATTAGACGTTCCCTTCGGGGACATGGATACAGGTGGTGC
    ATGGTTGTCGTCAGCTCGTGTCGTGAGATGTTGGGTTAAGTCCCGCAACG
    AGCGCAACCCTTATTATCAGTTGCCAGCATTAAGTTGGGCACTCTGGTGA
    GACTGCCGGTGACAAACCGGA
    S12KG220 GGDK 161-1 926F
    SEQ ID NO: 18
    TGGAGCATGTGGTTTAATTCGAAGCTACGCGAAGAACCTTACCAGGTCTT
    GACATACTATGCAAATCTAAGAGATTAGACGTTCCCTTCGGGGACATGGA
    TACAGGTGGTGCATGGTTGTCGTCAGCTCGTGTCGTGAGATGTTGGGTTA
    AGTCCCGCAACGAGCGCAACCCTTATTATCAGTTGCCAGCATTAAGTTGG
    GCACTCTGGTGAGACTGCCGGTGACAAACCGGAGGAAGGTGGGGATGACG
    TCAAATCATCATGCCCCTTATGACCTGGGCTACACACGTGCTACAATGGA
    TGGTACAACGAGTTGCGAACTCGCGAGAGTAAGCTAATCTCTTAAAGCCA
    TTCTCAGTTCGGATTGTAGGCTGCAACTCGCCTACATGAAGTCGGAATCG
    CTAGTAATCGCGGATCAGCATGCCGCGGTGAATACGTTCCCGGGCCTTGT
    ACACACCGCCCGTCACACCATGAGAGTTTGTAACACCCAAAGTCGGTGGG
    GTAACCTTTTAGGAACCAGCCGCCTAAGGTGGGACAGATGATTAGGGTGA
    AGTCGTAACAAGGTAGCCCGTA
    S12KG221 GGDK 161-1 926R
    SEQ ID NO: 19
    ACTCCCCAGGCGGAATGCTTAATGCGTTAGCTGCAGCACTGAAGGGCGGA
    AACCCTCCAACACTTAGCATTCATCGTTTACGGTATGGACTACCAGGGTA
    TCTAATCCTGTTTGCTACCCATACTTTCGAGCCTCAGCGTCAGTTACAGA
    CCAGACAGCCGCCTTCGCCACTGGTGTTCTTCCATATATCTACGCATTTC
    ACCGCTACACATGGAGTTCCACTGTCCTCTTCTGCACTCAAGTTTCCCAG
    TTTCCGATGCACTTCTTCGGTTGAGCCGAAGGCTTTCACATCAGACTTAA
    AAAACCGCCTGCGCTCGCTTTACGCCCAATAAATCCGGACAACGCTTGCC
    ACCTACGTATTACCGCGGCTGCTGGCACGTAGTTAGCCGTGGCTTTCTGG
    TTAAATACCGTCAATACCTGAACAGTTACTCTCAGATATGTTCTTCTTTA
    ACAACAGAGTTTTACGAGCCGAAACCCTTCTTCACTCACGCGGCGTTGCT
    CCATCAGACTTTCGTCCATTGTGGAAGATTCCCTACTGCTGCCTCCCGTA
    GGAGTTTGGGCCGTGTCTCAGTCCCAATGTGGCCGATTACCCTCTCAGGT
    CGGCTACGTATCATTGCCATGGTGAGCCGTTACCCCACCATCTAGCTAAT
    ACGCCGCGGGACCATCCAAAAGTGATAGCCGAAGCCATCTTTCAAACTCG
    GACCATGCGGTCCAAGTTGT
    S12KG222 GGDK 161-1 519B
    SEQ ID NO: 20
    GCTTTCTGGTTAAATACCGTCAATACCTGAACAGTTACTCTCAGATATGT
    TCTTCTTTAACAACAGAGTTTTACGAGCCGAAACCCTTCTTCACTCACGC
    GGCGTTGCTCCATCAGACTTTCGTCCATTGTGGAAGATTCCCTACTGCTG
    CCTCCCGTAGGAGTTTGGGCCGTGTCTCAGTCCCAATGTGGCCGATTACC
    CTCTCAGGTCGGCTACGTATCATTGCCATGGTGAGCCGTTACCCCACCAT
    CTAGCTAATACGCCGCGGGACCATCCAAAAGTGATAGCCGAAGCCATCTT
    TCAAACTCGGACCATGCGGTCCAAGTTGTTATGCGGTATTAGCATCTGTT
    TCCAGGTGTTATCCCCCGCTTCTGGGCAGGTTTCCCACGTGTTACTCACC
    AGTTCGCCACTCACTCAAATGTAAATCATGATGCAAGCACCAATCAATAC
    CAAAGTTCGTTCGACTTGCATGTATTAGGCACGCCGCCAGCGTTCGTCCT
    GAGCCAGATCAAACTCTAA
    S12KG223 GGDK 161-1 RP2
    SEQ ID NO: 21
    CCACCTTAGGCGGCTGGTTCCTAAAAGGTTACCCCACCGACTTTGGGTGT
    TACAAACTCTCATGGTGTGACGGGCGGTGTGTACAAGGCCCGGGAACGTA
    TTCACCGCGGCATGCTGATCCGCGATTACTAGCGATTCCGACTTCATGTA
    GGCGAGTTGCAGCCTACAATCCGAACTGAGAATGGCTTTAAGAGATTAGC
    TTACTCTCGCGAGTTCGCAACTCGTTGTACCATCCATTGTAGCACGTGTG
    TAGCCCAGGTCATAAGGGGCATGATGATTTGACGTCATCCCCACCTTCCT
    CCGGTTTGTCACCGGCAGTCTCACCAGAGTGCCCAACTTAATGCTGGCAA
    CTGATAATAAGGGTTGCGCTCGTTGCGGGACTTAACCCAACATCTCACGA
    CACGAGCTGACGACAACCATGCACCACCTGTATCCATGTCCCCGAAGGGA
    ACGTCTAATCTCTTAGATTTGCATAGTATGTCAAGACCTGGTAAGGTTCT
    TCGCGTAGCTTCGAATTAAACCACATGCTCCACCGCTTGTGCGGGCCCCC
    GTCAATTCCTTTGAGTTTCAGCCTTGCGGCCGTACTCCCCAGGCGGAATG
    CTTAATGCGTTAGCTGCAGCACTGAAGGGCGGAAACCCTCCAACACTTAG
    CATTCATCGTTTACGGTATGGACTACCAGGGTATCTA

    NCIMB 41847 GGDK161—contains both Lactobacillus plantarum and Lactobacillus reuteri Lactobacillus reuteri
  • S12KG309 cGGDK 161-1 27F
    SEQ ID NO: 22
    ATGCTAGTCGTACGCACTGGCCCAACTGATTGATGGTGCTTGCACCTGAT
    TGACGATGGATCACCAGTGAGTGGCGGACGGGTGAGTAACACGTAGGTAA
    CCTGCCCCGGAGCGGGGGATAACATTTGGAAACAGATGCTAATACCGCAT
    AACAACAAAAGCCACATGGCTTTTGTTTGAAAGATGGCTTTGGCTATCAC
    TCTGGGATGGACCTGCGGTGCATTAGCTAGTTGGTAAGGTAACGGCTTAC
    CAAGGCGATGATGCATAGCCGAGTTGAGAGACTGATCGGCCACAATGGAA
    CTGAGACACGGTCCATACTCCTACGGGAGGCAGCAGTAGGGAATCTTCCA
    CAATGGGCGCAAGCCTGATGGAGCAACACCGCGTGAGTGAAGAAGGGTTT
    CGGCTCGTAAAGCTCTGTTGTTGGAGAAGAACGTGCGTGAGAGTAACTGT
    TCACGCAGTGACGGTATCCAACCAGAAAGTCACGGCTAACTACGTGCCAG
    CAGCCGCGGTAATACGTAGGTGGCAAGCGTTATCCGGATTTATTGGGCGT
    AAAGCGAGCGCAGGCGGTTGCTTAGGTCTGATGTGAAAGCCTTCGGCTTA
    ACCGAAGAAGTGCATCGGAAACCGGGCGACTTGAGTGCAGAAGAGGACAG
    TGGAAC
    S12KG310 cDGGDK 161-1 591F
    SEQ ID NO: 23
    TCCGGATTTATTGGGCGTAAAGCGAGCGCAGGCGGTTGCTTAGGTCTGAT
    GTGAAAGCCTTCGGCTTAACCGAAGAAGTGCATCGGAAACCGGGCGACTT
    GAGTGCAGAAGAGGACAGTGGAACTCCATGTGTAGCGGTGGAATGCGTAG
    ATATATGGAAGAACACCAGTGGCGAAGGCGGCTGTCTGGTCTGCAACTGA
    CGCTGAGGCTCGAAAGCATGGGTAGCGAACAGGATTAGATACCCTGGTAG
    TCCATGCCGTAAACGATGAGTGCTAGGTGTTGGAGGGTTTCCGCCCTTCA
    GTGCCGGAGCTAACGCATTAAGCACTCCGCCTGGGAGTACGACCGCAAGG
    TTGAAACTCAAAGGAATTGACGGGGGCCCGCACAAGCGGTGGAGCATGTG
    GTTTAATTCGAAGCTACGCGAAGAACCTTACCAGGTCTTGACATCTTGCG
    CTAACCTTAGAGATAAGGCGTTCCCTTCGGGGACGCAATGACAGGTGGTG
    CATGGTCGTCGTCAGCTCGTGTCGTGAGATGTTGGGTTAAGTCCCGCAAC
    GAGCGCAACCCTTGTTACTAGTTGCCAGCATTAAGTTGGGCACTCTAGTG
    AGACTGCCGGTGACAAACCGGAGGAAGGTGGGGACGACGTCAGATCATCA
    TGCCCCTTATGACCTGGGCTACACACGTGCTACAATGGACGGTACAACGA
    GTCGCAAGCTCGCGAGAG
    S12KG311 cGGDK 161-1 926F
    SEQ ID NO: 24
    GGAGCATGTGGTTTAATTCGAAGCTACGCGAAGAACCTTACCAGGTCTTG
    ACATCTTGCGCTAACCTTAGAGATAAGGCGTTCCCTTCGGGGACGCAATG
    ACAGGTGGTGCATGGTCGTCGTCAGCTCGTGTCGTGAGATGTTGGGTTAA
    GTCCCGCAACGAGCGCAACCCTTGTTACTAGTTGCCAGCATTAAGTTGGG
    CACTCTAGTGAGACTGCCGGTGACAAACCGGAGGAAGGTGGGGACGACGT
    CAGATCATCATGCCCCTTATGACCTGGGCTACACACGTGCTACAATGGAC
    GGTACAACGAGTCGCAAGCTCGCGAGAGTAAGCTAATCTCTTAAAGCCGT
    TCTCAGTTCGGACTGTAGGCTGCAACTCGCCTACACGAAGTCGGAATCGC
    TAGTAATCGCGGATCAGCATGCCGCGGTGAATACGTTCCCGGGCCTTGTA
    CACACCGCCCGTCACACCATGGGAGTTTGTAACGCCCAAAGTCGGTGGCC
    TAACCTTTATGGAGGGAGCCGCCTAAGGCGGGACAGATGACTGGGGTGAA
    GTCGTAACAAGGTAGCCGTA
    S12KG312 cGGDK 161-1 926A
    SEQ ID NO: 25
    TCCCCAGGCGGAGTGCTTAATGCGTTAGCTCCGGCACTGAAGGGCGGAAA
    CCCTCCAACACCTAGCACTCATCGTTTACGGCATGGACTACCAGGGTATC
    TAATCCTGTTCGCTACCCATGCTTTCGAGCCTCAGCGTCAGTTGCAGACC
    AGACAGCCGCCTTCGCCACTGGTGTTCTTCCATATATCTACGCATTCCAC
    CGCTACACATGGAGTTCCACTGTCCTCTTCTGCACTCAAGTCGCCCGGTT
    TCCGATGCACTTCTTCGGTTAAGCCGAAGGCTTTCACATCAGACCTAAGC
    AACCGCCTGCGCTCGCTTTACGCCCAATAAATCCGGATAACGCTTGCCAC
    CTACGTATTACCGCGGCTGCTGGCACGTAGTTAGCCGTGACTTTCTGGTT
    GGATACCGTCACTGCGTGAACAGTTACTCTCACGCACGTTCTTCTCCAAC
    AACAGAGCTTTACGAGCCGAAACCCTTCTTCACTCACGCGGTGTTGCTCC
    ATCAGGCTTGCGCCCATTGTGGAAGATTCCCTACTGCTGCCTCCCGTAGG
    AGTATGGACCGTGTCTCAGTTCCATTGTGGCCGATCAGTCTCTCAACTCG
    GCTATGCATCATCGCCTTGGTAAGCCGTTACCTTACCAACTAGCTAATGC
    ACCGCAGGTCCATCCCAGAGTGATAGCCAAAGCCATCTTTCAAACAAAAG
    CCATGTGGCTTTT
    S12KG313 cGGDK 161-1 519R
    SEQ ID NO: 26
    TTTCTGGTTGGATACCGTCACTGCGTGAACAGTTACTCTCACGCACGTTC
    TTCTCCAACAACAGAGCTTTACGAGCCGAAACCCTTCTTCACTCACGCGG
    TGTTGCTCCATCAGGCTTGCGCCCATTGTGGAAGATTCCCTACTGCTGCC
    TCCCGTAGGAGTATGGACCGTGTCTCAGTTCCATTGTGGCCGATCAGTCT
    CTCAACTCGGCTATGCATCATCGCCTTGGTAAGCCGTTACCTTACCAACT
    AGCTAATGCACCGCAGGTCCATCCCAGAGTGATAGCCAAAGCCATCTTTC
    AAACAAAAGCCATGTGGCTTTTGTTGTTATGCGGTATTAGCATCTGTTTC
    CAAATGTTATCCCCCGCTCCGGGGCAGGTTACCTACGTGTTACTCACCCG
    TCCGCCACTCACTGGTAATCCATCGTCAATCAGGTGCAAGCACCATCAAT
    CAGTTGGGCCAGTGCGTACGACTTGCATGTATTAGGCACACCGCCGGCGT
    TCATCCTGAGCCATGATCAAAC
    S12KG314 cGGDK 161-1 RP2
    SEQ ID NO: 27
    GCGGCTCCCTCCATAAAGGTTAGCGCCACCGACTTTGGGCGTTACAAACT
    CCCATGGTGTGACGGGCGGTGTGTACAAGGCCCGGGAACGTATTCACCGC
    GGCATGCTGATCCGCGATTACTAGCGATTCCGACTTCGTGTAGGCGAGTT
    GCAGCCTACAGTCCGAACTGAGAACGGCTTTAAGAGATTAGCTTACTCTC
    GCGAGCTTGCGACTCGTTGTACCGTCCATTGTAGCACGTGTGTAGCCCAG
    GTCATAAGGGGCATGATGATCTGACGTCGTCCCCACCTTCCTCCGGTTTG
    TCACCGGCAGTCTCACTAGAGTGCCCAACTTAATGCTGGCAACTAGTAAC
    AAGGGTTGCGCTCGTTGCGGGACTTAACCCAACATCTCACGACACGAGCT
    GACGACGACCATGCACCACCTGTCATTGCGTCCCCGAAGGGAACGCCTTA
    TCTCTAAGGTTAGCGCAAGATGTCAAGACCTGGTAAGGTTCTTCGCGTAG
    CTTCGAATTAAACCACATGCTCCACCGCTTGTGCGGGCCCCCGTCAATTC
    CTTTGAGTTTCAACCTTGCGGTCGTACTCCCCAGGCGGAGTGCTTAATGC
    GTTAGCTCCGGCACTGAAGGGCGGAAACCCTCCAACACCTAGCACTCATC
    GTTTACGGCAT

    NCIMB 41848 GGDK255—Lactobacillus reuteri
  • S12KG237 GGDK 255-1 27F
    SEQ ID NO: 28
    GTGTGCCTAATACATGCAAGTCGTACGCACTGGCCCAACTGATTGATGGT
    GCTTGCACCTGATTGACGATGGATCACCAGTGAGTGGCGGACGGGTGAGT
    AACACGTAGGTAACCTGCCCCGGAGCGGGGGATAACATTTGGAAACAGAT
    GCTAATACCGCATAACAACAAAAGCCACATGGCTTTTGTTTGAAAGATGG
    CTTTGGCTATCACTCTGGGATGGACCTGCGGTGCATTAGCTAGTTGGTAA
    GGTAACGGCTTACCAAGGCGATGATGCATAGCCGAGTTGAGAGACTGATC
    GGCCACAATGGAACTGAGACACGGTCCATACTCCTACGGGAGGCAGCAGT
    AGGGAATCTTCCACAATGGGCGCAAGCCTGATGGAGCAACACCGCGTGAG
    TGAAGAAGGGTTTCGGCTCGTAAAGCTCTGTTGTTGGAGAAGAACGTGCG
    TGAGAGTAACTGTTCACGCAGTGACGGTATCCAACCAGAAAGTCACGGCT
    AACTACGTGCCAGCAGCCGCGGTAATACGTAGGTGGCAAGCGTTATCCGG
    ATTTATTGGGCGTAAAGCGAGCGCAGGCGGTTGCTTAGGTCTGATGTGAA
    AGCCTTCGGCTTAACCGAAGAAGTGCATCGGAAACCGGGCGACTTGAGTG
    CAGAAGAGGACAGTGGAACTCCATGTGTAGCGGTGGAATGCGTAGATATA
    TGGAAGAACACCAGTG
    S12KG238 GGDK 225-1 519F
    SEQ ID NO: 29
    TCGGATTTATTGGGCGTAAAGCGAGCGCAGGCGGTTGCTTAGGTCTGATG
    TGAAAGCCTTCGGCTTAACCGAAGAAGTGCATCGGAAACCGGGCGACTTG
    AGTGCAGAAGAGGACAGTGGAACTCCATGTGTAGCGGTGGAATGCGTAGA
    TATATGGAAGAACACCAGTGGCGAAGGCGGCTGTCTGGTCTGCAACTGAC
    GCTGAGGCTCGAAAGCATGGGTAGCGAACAGGATTAGATACCCTGGTAGT
    CCATGCCGTAAACGATGAGTGCTAGGTGTTGGAGGGTTTCCGCCCTTCAG
    TGCCGGAGCTAACGCATTAAGCACTCCGCCTGGGGAGTACGACCGCAAGG
    TTGAAACTCAAAGGAATTGACGGGGGCCCGCACAAGCGGTGGAGCATGTG
    GTTTAATTCGAAGCTACGCGAAGAACCTTACCAGGTCTTGACATCTTGCG
    CTAACCTTAGAGATAAGGCGTTCCCTTCGGGGACGCAATGACAGGTGGTG
    CATGGTCGTCGTCAGCTCGTGTCGTGAGATGTTGGGTTAAGTCCCGCAAC
    GAGCGCAACCCTTGTTACTAGTTGCCAGCATTAAGTTGGGCACTCTAGTG
    AGACTGCCGGTGACAAACCGGAGGAAGGTGGGGACGACGTCAGATCATCA
    TGCCCCTTATGACCTGGGCTACACACGTGCTAC
    S12KG229 GGDK 225-1 926F
    SEQ ID NO: 30
    TGGAGCATGTGGTTTAATTCGAAGCTACGCGAAGAACCTTACCAGGTCTT
    GACATCTTGCGCTAACCTTAGAGATAAGGCGTTCCCTTCGGGGACGCAAT
    GACAGGTGGTGCATGGTCGTCGTCAGCTCGTGTCGTGAGATGTTGGGTTA
    AGTCCCGCAACGAGCGCAACCCTTGTTACTAGTTGCCAGCATTAAGTTGG
    GCACTCTAGTGAGACTGCCGGTGACAAACCGGAGGAAGGTGGGGACGACG
    TCAGATCATCATGCCCCTTATGACCTGGGCTACACACGTGCTACAATGGA
    CGGTACAACGAGTCGCAAGCTCGCGAGAGTAAGCTAATCTCTTAAAGCCG
    TTCTCAGTTCGGACTGTAGGCTGCAACTCGCCTACACGAAGTCGGAATCG
    CTAGTAATCGCGGATCAGCATGCCGCGGTGAATACGTTCCCGGGCCTTGT
    ACACACCGCCCGTCACACCATGGGAGTTTGTAACGCCCAAAGTCGGTGGC
    CTAACCTTTATGGAGGGAGCCGCCTAAGGCGGGACAGATGACTGGGGTGA
    AGTCGTAACAAGGTAGCCGTA
    S12KG240 GGDK 255-1 526R
    SEQ ID NO: 31
    TACTCCCCAGGCGGAGTGCTTAATGCGTTAGCTCCGGCACTGAAGGGCGG
    AAACCCTCCAACACCTAGCACTCATCGTTTACGGCATGGACTACCAGGGT
    ATCTAATCCTGTTCGCTACCCATGCTTTCGAGCCTCAGCGTCAGTTGCAG
    ACCAGACAGCCGCCTTCGCCACTGGTGTTCTTCCATATATCTACGCATTC
    CACCGCTACACATGGAGTTCCACTGTCCTCTTCTGCACTCAAGTCGCCCG
    GTTTCCGATGCACTTCTTCGGTTAAGCCGAAGGCTTTCACATCAGACCTA
    AGCAACCGCCTGCGCTCGCTTTACGCCCAATAAATCCGGATAACGCTTGC
    CACCTACGTATTACCGCGGCTGCTGGCACGTAGTTAGCCGTGACTTTCTG
    GTTGGATACCGTCACTGCGTGAACAGTTACTCTCACGCACGTTCTTCTCC
    AACAACAGAGCTTTACGAGCCGAAACCCTTCTTCACTCACGCGGTGTTGC
    TCCATCAGGCTTGCGCCCATTGTGGAAGATTCCCTACTGCTGCCTCCCGT
    AGGAGTATGGACCGTGTCTCAGTTCCATTGTGGCCGATCAGTCTCTCAAC
    TCGGCTATGCATCATCGCCTTGGTAAGCCGTTACCTTACCAACTAGCTAA
    TGCACCGCAGGTCCATCCCAGAGTGATAGCCAAAGCCATCTTTCAAACAA
    AAGCCATGTGGCTTTTG
    S12KG241 GGDK 255-1 519R
    SEQ ID NO: 32
    TTTCTGGTTGGATACCGTCACTGCGTGAACAGTTACTCTCACGCACGTTC
    TTCTCCAACAACAGAGCTTTACGAGCCGAAACCCTTCTTCACTCACGCGG
    TGTTGCTCCATCAGGCTTGCGCCCATTGTGGAAGATTCCCTACTGCTGCC
    TCCCGTAGGAGTATGGACCGTGTCTCAGTTCCATTGTGGCCGATCAGTCT
    CTCAACTCGGCTATGCATCATCGCCTTGGTAAGCCGTTACCTTACCAACT
    AGCTAATGCACCGCAGGTCCATCCCAGAGTGATAGCCAAAGCCATCTTTC
    AAACAAAAGCCATGTGGCTTTTGTTGTTATGCGGTATTAGCATCTGTTTC
    CAAATGTTATCCCCCGCTCCGGGGCAGGTTACCTACGTGTTACTCACCCG
    TCCGCCACTCACTGGTGATCCATCGTCAATCAGGTGCAAGCACCATCAAT
    CAGTTGGGCCAGTGCGTACGACTTGCATGTATTAGGCACACCGCCGGCGT
    CCATCCTGAGCCATGATCAAAC
    S12KG242 GGDK 255-1 RP2
    SEQ ID NO: 33
    CCGCCTTAGGCGGCTCCCTCCATAAAGGTTAGGCCACCGACTTTGGGCGT
    TACAAACTCCCATGGTGTGACGGGCGGTGTGTACAAGGCCCGGGAACGTA
    TTCACCGCGGCATGCTGATCCGCGATTACTAGCGATTCCGACTTCGTGTA
    GGCGAGTTGCAGCCTACAGTCCGAACTGAGAACGGCTTTAAGAGATTAGC
    TTACTCTCGCGAGCTTGCGACTCGTTGTACCGTCCATTGTAGCACGTGTG
    TAGCCCAGGTCATAAGGGGCATGATGATCTGACGTCGTCCCCACCTTCCT
    CCGGTTTGTCACCGGCAGTCTCACTAGAGTGCCCAACTTAATGCTGGCAA
    CTAGTAACAAGGGTTGCGCTCGTTGCGGGACTTAACCCAACATCTCACGA
    CACGAGCTGACGACGACCATGCACCACCTGTCATTGCGTCCCCGAAGGGA
    ACGCCTTATCTCTAAGGTTAGCGCAAGATGTCAAGACCTGGTAAGGTTCT
    TCGCGTAGCTTCGAATTAAACCACATGCTCCACCGCTTGTGCGGGCCCCC
    GTCAATTCCTTTGAGTTTCAACCTTGCGGTCGTACTCCCCAGGCGGAGTG
    CTTAATGCGTTAGCTCCGGCACTGAAGGGCGGAAACCCTCCAACACCTAG
    CACTCATCGTT

    NCIMB 41849 GGDK 258—Lactobacillus plantarum
  • S12KG267 GGDK 258-3 27F
    SEQ ID NO: 34
    GTGCCTAATACATGCAAGTCGAACGAACTCTGGTATTGATTGGTGCTTGC
    ATCATGATTTACATTTGAGTGAGTGGCGAACTGGTGAGTAACACGTGGGA
    AACCTGCCCAGAAGCGGGGGATAACACCTGGAAACAGATGCTAATACCGC
    ATAACAACTTGGACCGCATGGTCCGAGTTTGAAAGATGGCTTCGGCTATC
    ACTTTTGGATGGTCCCGCGGCGTATTAGCTAGATGGTGAGGTAACGGCTC
    ACCATGGCAATGATACGTAGCCGACCTGAGAGGGTAATCGGCCACATTGG
    GACTGAGACACGGCCCAAACTCCTACGGGAGGCAGCAGTAGGGAATCTTC
    CACAATGGACGAAAGTCTGATGGAGCAACGCCGCGTGAGTGAAGAAGGGT
    TTCGGCTCGTAAAACTCTGTTGTTAAAGAAGAACATATCTGAGAGTAACT
    GTTCAGGTATTGACGGTATTTAACCAGAAAGCCACGGCTAACTACGTGCC
    AGCAGCCGCGGTAATACGTAGGTGGCAAGCGTTGTCCGGATTTATTGGGC
    GTAAAGCGAGCGCAGGCGGTTTTTTAAGTCTGATGTGAAAGCCTTCGGCT
    CAACCGAAGAAGTGCATCGGAAACTGGGAAACTTGAGTGCAGAAGAGGAC
    AGTGGAACTC
    S12KG268 GGDK 256-3 591F
    SEQ ID NO: 35
    GGATTTATTGGGCGTAAAGCGAGCGCAGGCGGTTTTTTAAGTCTGATGTG
    AAAGCCTTCGGCTCAACCGAAGAAGTGCATCGGAAACTGGGAAACTTGAG
    TGCAGAAGAGGACAGTGGAACTCCATGTGTAGCGGTGAAATGCGTAGATA
    TATGGAAGAACACCAGTGGCGAAGGCGGCTGTCTGGTCTGTAACTGACGC
    TGAGGCTCGAAAGTATGGGTAGCAAACAGGATTAGATACCCTGGTAGTCC
    ATACCGTAAACGATGAATGCTAAGTGTTGGAGGGTTTCCGCCCTTCAGTG
    CTGCAGCTAACGCATTAAGCATTCCGCCTGGGGAGTACGGCCGCAAGGCT
    GAAACTCAAAGGAATTGACGGGGGCCCGCACAAGCGGTGGAGCATGTGGT
    TTAATTCGAAGCTACGCGAAGAACCTTACCAGGTCTTGACATACTATGCA
    AATCTAAGAGATTAGACGTTCCCTTCGGGGACATGGATACAGGTGGTGCA
    TGGTTGTCGTCAGCTCGTGTCGTGAGATGTTGGGTTAAGTCCCGCAACGA
    GCGCAACCCTTATTATCAGTTGCCAGCATTAAGTTGGGCACTCTGGTGAG
    ACTGCCGGTGACAAACCGGAGGAAGGTGGGGATGACGTCAAATCATCATG
    CCCCTTATGACCTGGGCTAC
    S12KG269 GGDK 258-8 926F
    SEQ ID NO: 36
    GTGGAGCATGTGGTTTAATTCGAAGCTACGCGAAGAACCTTACCAGGTCT
    TGACATACTATGCAAATCTAAGAGATTAGACGTTCCCTTCGGGGACATGG
    ATACAGGTGGTGCATGGTTGTCGTCAGCTCGTGTCGTGAGATGTTGGGTT
    AAGTCCCGCAACGAGCGCAACCCTTATTATCAGTTGCCAGCATTAAGTTG
    GGCACTCTGGTGAGACTGCCGGTGACAAACCGGAGGAAGGTGGGGATGAC
    GTCAAATCATCATGCCCCTTATGACCTGGGCTACACACGTGCTACAATGG
    ATGGTACAACGAGTTGCGAACTCGCGAGAGTAAGCTAATCTCTTAAAGCC
    ATTCTCAGTTCGGATTGTAGGCTGCAACTCGCCTACATGAAGTCGGAATC
    GCTAGTAATCGCGGATCAGCATGCCGCGGTGAATACGTTCCCGGGCCTTG
    TACACACCGCCCGTCACACCATGAGAGTTTGTAACACCCAAAGTCGGTGG
    GGTAACCTTTTAGGAACCAGCCGCCTAAGGTGGGACAGATGATTAGGGTG
    AAGTCGTAACAAGGTAGCCCGTA
    S12KG270 GGDK 258-3 926R
    SEQ ID NO: 37
    ACTCCCCAGGCGGAATGCTTAATGCGTTAGCTGCAGCACTGAAGGGCGGA
    AACCCTCCAACACTTAGCATTCATCGTTTACGGTATGGACTACCAGGGTA
    TCTAATCCTGTTTGCTACCCATACTTTCGAGCCTCAGCGTCAGTTACAGA
    CCAGACAGCCGCCTTCGCCACTGGTGTTCTTCCATATATCTACGCATTTC
    ACCGCTACACATGGAGTTCCACTGTCCTCTTCTGCACTCAAGTTTCCCAG
    TTTCCGATGCACTTCTTCGGTTGAGCCGAAGGCTTTCACATCAGACTTAA
    AAAACCGCCTGCGCTCGCTTTACGCCCAATAAATCCGGACAACGCTTGCC
    ACCTACGTATTACCGCGGCTGCTGGCACGTAGTTAGCCGTGGCTTTCTGG
    TTAAATACCGTCAATACCTGAACAGTTACTCTCAGATATGTTCTTCTTTA
    ACAACAGAGTTTTACGAGCCGAAACCCTTCTTCACTCACGCGGCGTTGCT
    CCATCAGACTTTCGTCCATTGTGGAAGATTCCCTACTGCTGCCTCCCGTA
    GGAGTTTGGGCCGTGTCTCAGTCCCAATGTGGCCGATTACCCTCTCAGGT
    CGGCTACGTATCATTGCCATGGTGAGCCGTTACCTCACCATCTAGCTAAT
    ACGCCGCGGGACCATCCAAAAGTGATAGCCGAAGCCATCTTTCAAACTCG
    GACCATGCGGTCCAAGTTGTTATGCGGTATTAGCATCTGTTTC
    S12KG271 GGDK 258-3 519R
    SEQ ID NO: 38
    TTTCTGGTTAAATACCGTCAATACCTGAACAGTTACTCTCAGATATGTTC
    TTCTTTAACAACAGAGTTTTACGAGCCGAAACCCTTCTTCACTCACGCGG
    CGTTGCTCCATCAGACTTTCGTCCATTGTGGAAGATTCCCTACTGCTGCC
    TCCCGTAGGAGTTTGGGCCGTGTCTCAGTCCCAATGTGGCCGATTACCCT
    CTCAGGTCGGCTACGTATCATTGCCATGGTGAGCCGTTACCTCACCATCT
    AGCTAATACGCCGCGGGACCATCCAAAAGTGATAGCCGAAGCCATCTTTC
    AAACTCGGACCATGCGGTCCAAGTTGTTATGCGGTATTAGCATCTGTTTC
    CAGGTGTTATCCCCCGCTTCTGGGCAGGTTTCCCACGTGTTACTCACCAG
    TTCGCCACTCACTCAAATGTAAATCATGATGCAAGCACCAATCAATACCA
    GAGTTCGTTCGACTTGCATGTATTAGGCACGCCGCCAGCGTTCGTCCTGA
    GCCATGATCAAAC
    S12KG272 GGDK 258-3 RP2
    SEQ ID NO: 39
    CCACCTTAGGCGGCTGGTTCCTAAAAGGTTACCCCACCGACTTTGGGTGT
    TACAAACTCTCATGGTGTGACGGGCGGTGTGTACAAGGCCCGGGAACGTA
    TTCACCGCGGCATGCTGATCCGCGATTACTAGCGATTCCGACTTCATGTA
    GGCGAGTTGCAGCCTACAATCCGAACTGAGAATGGCTTTAAGAGATTAGC
    TTACTCTCGCGAGTTCGCAACTCGTTGTACCATCCATTGTAGCACGTGTG
    TAGCCCAGGTCATAAGGGGCATGATGATTTGACGTCATCCCCACCTTCCT
    CCGGTTTGTCACCGGCAGTCTCACCAGAGTGCCCAACTTAATGCTGGCAA
    CTGATAATAAGGGTTGCGCTCGTTGCGGGACTTAACCCAACATCTCACGA
    CACGAGCTGACGACAACCATGCACCACCTGTATCCATGTCCCCGAAGGGA
    ACGTCTAATCTCTTAGATTTGCATAGTATGTCAAGACCTGGTAAGGTTCT
    TCGCGTAGCTTCGAATTAAACCACATGCTCCACCGCTTGTGCGGGCCCCC
    GTCAATTCCTTTGAGTTTCAGCCTTGCGGCCGTACTCCCCAGGCGGAATG
    CTTAATGCGTTAGCTGCAGCACTGAAGGGCGGAAACCCTCCAACACTTAG
    CATTCATCGTTTACGGTATGGACTACCAGGGTATCTAATCCTGTTTGCTA
    CCCATACTTTCGAGCCTCAGCGTCAGTTACAGACCAGACAGCCGCCT

    NCIMB 41850 GGDK 266—contains both Lactobacillus iohnsonii and Lactobacillus reuteri Lactobacillus iohnsonii
  • S12KG273 GGDK 266-1 27F-repeat
    SEQ ID NO: 40
    GTGCCTAATACATGCAAGTCGAGCGAGCTTGCCTAGATGATTTTAGTGCT
    TGCACTAAATGAAACTAGATACAAGCGAGCGGCGGACGGGTGAGTAACAC
    GTGGGTAACCTGCCCAAGAGACTGGGATAACACCTGGAAACAGATGCTAA
    TACCGGATAACAACACTAGACGCATGTCTAGAGTTTGAAAGATGGTTCTG
    CTATCACTCTTGGATGGACCTGCGGTGCATTAGCTAGTTGGTAAGGTAAC
    GGCTTACCAAGGCAATGATGCATAGCCGAGTTGAGAGACTGATCGGCCAC
    ATTGGGACTGAGACACGGCCCAAACTCCTACGGGAGGCAGCAGTAGGGAA
    TCTTCCACAATGGACGAAAGTCTGATGGAGCAACGCCGCGTGAGTGAAGA
    AGGGTTTCGGCTCGTAAAGCTCTGTTGGTAGTGAAGAAAGATAGAGGTAG
    TAACTGGCCTTTATTTGACGGTAATTACTTAGAAAGTCACGGCTAACTAC
    GTGCCAGCAGCCGCGGTAATACGTAGGTGGCAAGCGTTGTCCGGATTTAT
    TGGGCGTAAAGCGAGTGCAGGCGGTTCAATAAGTCTGATGTGAAAGCCTT
    CGGCTCAACCGGAGAAT
    S12KG274 GGDK 266-1 519F
    SEQ ID NO: 41
    TCCGGATTTATTGGGCGTAAAGCGAGTGCAGGCGGTTCAATAAGTCTGAT
    GTGAAAGCCTTCGGCTCAACCGGAGAATTGCATCAGAAACTGTTGAACTT
    GAGTGCAGAAGAGGAGAGTGGAACTCCATGTGTAGCGGTGGAATGCGTAG
    ATATATGGAAGAACACCAGTGGCGAAGGCGGCTCTCTGGTCTGCAACTGA
    CGCTGAGGCTCGAAAGCATGGGTAGCGAACAGGATTAGATACCCTGGTAG
    TCCATGCCGTAAACGATGAGTGCTAAGTGTTGGGAGGTTTCCGCCTCTCA
    GTGCTGCAGCTAACGCATTAAGCACTCCGCCTGGGGAGTACGACCGCAAG
    GTTGAAACTCAAAGGAATTGACGGGGGCCCGCACAAGCGGTGGAGCATGT
    GGTTTAATTCGAAGCAACGGGAAGAACCTTACCAGGTCTTGACATCCAGT
    GCAAACCTAAGAGATTAGGTGTTCCCTTCGGGGACGCTGAGACAGGTGGT
    GCATGGCTGTCGTCAGCTCGTGT
    S12KG275 GGDK 226-1 926F-repeat
    SEQ ID NO: 42
    GGAGCATGTGGTTTAATTCGAAGCAACGCGAAGAACCTTACCAGGTCTTG
    ACATCCAGTGCAAACCTAAGAGATTAGGTGTGTCCCTTCGGGGACGCTGA
    GACAGGTGGTGCATGGCTGTCGTCAGCTCGTGTCGTGAGATGTTGGGTTA
    AGTCCCGCAACGAGCGCAACCCTTGTCATTAGTTGCCATCATTAAGTTGG
    GCACTCTAATGAGACTGCCGGTGACAAACCGGAGGAAGGTGGGGATGACG
    TCAAGTCATCATGCCCCTTATGACCTGGGCTACACACGTGCTACAATGGA
    CGGTACAACGAGAAGCGAACCTGCGAAGGCAAGCGGATCTCTTAAAGCCG
    TTCTCAGTTCGGACTGTAGGCTGCAACTCGCCTACACGAAGCTGGAATCG
    CTAGTAATCGCGGATCAGCACGCCGCGGTGAATACGTTCCCGGGCCTTGT
    ACACACCGCCCGTCACACCATGAGAGTCTGTA
    S12KG276 GGDK 255-1 926R
    SEQ ID NO: 43
    ACTCCCCAGGCGGAGTGCTTAATGCGTTAGCTGCAGCACTGAGAGGCGGA
    AACCTCCCAACACTTAGCACTCATCGTTTACGGCATGGACTACCAGGGTA
    TCTAATCCTGTTCGCTACCCATGCTTTCGAGCCTCAGCGTCAGTTGCAGA
    CCAGAGAGCCGCCTTCGCCACTGGTGTTCTTCCATATATCTACGCATTCC
    ACCGCTACACATGGAGTTCCACTCTCCTCTTCTGCACTCAAGTTCAACAG
    TTTCTGATGCAATTCTCCGGTTGAGCCGAAGGCTTTCACATCAGACTTAT
    TGAACCGCCTGCACTCGCTTTACGCCCAATAAATCCGGACAACGCTTGCC
    ACCTACGTATTACCGCGGCTGCTGGCACGTAGTTAGCCGTGACTTTCTAA
    GTAATTACCGTCAAATAAAGGCCAGTTACTACCTCTATCTTTCTTCACTA
    CCAACAGAGCTTTACGAGCCGAAACCCTTCTTCACTCACGCGGCGTTGCT
    CCATCAGACTTTCGTCCATTGTGGAAGATTCCCTACTGCTGCCTCCCGTA
    GGAGTTTGGGCCGTGTCTCAGTCCCAATGTGGCCGATCAGTCTCTCAACT
    CGGCTATGCATCATTGCCTTGGTAAGCCGTTACCTTACCAACTAGCTAAT
    GCACCGCAGGTCCATCCAAGAGTGATAGCAGAACCATCTTTCAAACTCTA
    GCACATGCGTCTAGTGTTGT
    S12KG277 GGDK 266-1 519R
    SEQ ID NO: 44
    ACTTTCTAAGTAATTACCGTCAAATAAAGGCCAGTTACTACCTCTATCTT
    TCTTCACTACCAACAGAGCTTTACGAGCCGAAACCCTTCTTCACTCACGC
    GGCGTTGCTCCATCAGACTTTCGTCCATTGTGGAAGATTCCCTACTGCTG
    CCTCCCGTAGGAGTTTGGGCCGTGTCTCAGTCCCAATGTGGCCGATCAGT
    CTCTCAACTCGGCTATGCATCATTGCCTTGGTAAGCCGTTACCTTACCAA
    CTAGCTAATGCACCGCAGGTCCATCCAAGAGTGATAGCAGAACCATCTTT
    CAAACTCTAGACATGCGTCTAGTGTTGTTATCCGGTATTAGCATCTGTTT
    CCAGGTGTTATCCCAGTCTCTTGGGCAGGTTACCCACGTGTTACTCACCC
    GTCCGCCGCTCGCTTGTATCTAGTTTCATTTAGTGCAAGCACTAAAATCA
    TCTAGGCAAGCTCGCTCGACTTGCATGTATTAGGCACGCCGCCAGCGTTC
    GTCCTGAGCCATGATCAAACT
    S12KG278 GGDK 266-1 RP2
    SEQ ID NO: 45
    CTACCTTAGACGGCTGACTCCTATAAAGGTTATCCCACCGGCTTTGGGTG
    TTACAGACTCTCATGGTGTGACGGGCGGTGTGTACAAGGCCCGGGAACGT
    ATTCACCGCGGCGTGCTGATCCGCGATTACTAGCGATTCCAGCTTCGTGT
    AGGCGAGTTGCAGCCTACAGTCCGAACTGAGAACGGCTTTAAGAGATCCG
    CTTGCCTTCGCAGGTTCGCTTCTCGTTGTACCGTCCATTGTAGCACGTGT
    GTAGCCCAGGTCATAAGGGGCATGATGACTTGACGTCATCCCCACCTTCC
    TCCGGTTTGTCACCGGCAGTCTCATTAGAGTGCCCAACTTAATGATGGCA
    ACTAATGACAAGGGTTGCGCTCGTTGCGGGACTTAACCCAACATCTCACG
    ACACGAGCTGACGACAGCCATGCACCACCTGTCTCAGCGTCCCCGAAGGG
    AACACCTAATCTCTTAGGTTTGCACTGGATGTCAAGACCTGGTAAGGTTC
    TTCGCGTTGCTTCGAATTAAACCACATGCTCCACCGCTTGTGCGGGCCCC
    CGTCAATTCCTTTGAGTTTCAACCTTGCGGTCGTACACCCCAGGCGGAGT
    GCTTAATGCGTTAGCTGCAGCACTGAGAGGCGGAAACCTCCCAACACTTA
    GCACTCATCGTTTACGGCATGGACTACCAGGGTATCTAATCCTGTTCGCT
    ACCCATGCTTTCGAGCCTCAGCGTCAGTTGCAGACCAGAGAGCCGCCT

    NCIMB 41850 GGDK 266—contains both Lactobacillus iohnsonii and Lactobacillus reuteri Lactobacillus reuteri
  • S12KG279 GGDK-266-2 27F
    SEQ ID NO: 46
    GTGTGCCTAATACATGCAAGTCGTACGCACTGGCCCAACTGATTGATGGT
    GCTTGCACCTGATTGACGATGGATCACCAGTGAGTGGCGGACGGGTGAGT
    AACACGTAGGTAACCTGCCCCGGAGCGGGGGATAACATTTGGAAACAGAT
    GCTAATACCGCATAACAACAAAAGCCACATGGCTTTTGTTTGAAAGATGG
    CTTCGGCTATCACTCTGGGATGGACCTGCGGTGCATTAGCTAGTTGGTAA
    GGTAACGGCTTACCAAGGCGATGATGCATAGCCGAGTTGAGAGACTGATC
    GGCCACAATGGAACTGAGACACGGTCCATACTCCTACGGGAGGCAGCAGT
    AGGGAATCTTCCACAATGGGCGCAAGCCTGATGGAGCAACACCGCGTGAG
    TGAAGAAGGGTTTCGGCTCGTAAAGCTCTGTTGTTGGAGAAGAACGTGCG
    TGAGAGTAACTGTTCACGCAGTGACGGTATCCAACCAGAAAGTCACGGCT
    AACTACGTGCCAGCAGCCGCGGTAATACGTAGGTGGCAAGCGTTATCCGG
    ATTTATTGGGCGTAAAGCGAGCGCAGGCGGTTGCTTAGGTCTGATGTGAA
    AGCCTTCGGCTTAACCGAAGAAGTGCATCGGAAACCGGGCGACTTGAGTG
    CAGAAGAGGACAGTGGAACTCCATGTGTAGCGGTGGAATGCGTA
    S12KG280 GGDK-266-2 519F-repeat
    SEQ ID NO: 47
    CGGATTTATTGGGCGTAAAGCGAGCGCAGGCGGTTGCTTAGGTCTGATGT
    GAAAGCCTTCGGCTTAACCGAAGAAGTGCATCGGAAACCGGGCGACTTGA
    GTGCAGAAGAGGACAGTGGAACTCCATGTGTAGCGGTGGAATGCGTAGAT
    ATATGGAAGAACACCAGTGGCGAAGGCGGCTGTCTGGTCTGCAACTGACG
    CTGAGGCTCGAAAGCATGGGTAGCGAACAGGATTAGATACCCTGGTAGTC
    CATGCCGTAAACGATGAGTGCTAGGTGTTGGAGGGTTTCCGCCCTTCAGT
    GCCGGAGCTAACGCATTAAGCACTCCGCCTGGGGAGTACGACCGCAAGGT
    TGAAACTCAAAGGAATTGACGGGGGCCCGCACAAGCGGTGGAGCATGTGG
    TTTAATTCG
    S12KG281 GGDK-266-2 926F-repeat
    SEQ ID NO: 48
    GAGCATGTGGTTTAATTCGAAGCTACGCGAAGAACCTTACCAGGTCTTGA
    CATCTTGCGCTAACCTTAGAGATAAGGCGTTCCCTTCGGGGACGCAATGA
    CAGGTGGTGCATGGTCGTCGTCAGCTCGTGTCGTGAGATGTTGGGTTAAG
    TCCCGCAACGAGCGCAACCCTTGTTACTAGTTGCCAGCATTAAGTTGGGC
    ACTCTAGTGAGACTGCCGGTGACAAACCGGAGGAAGGTGGGGACGACGTC
    AGATCATCATGCCCCTTATGACCTGGGCTACACACGTGCTACAATGGACG
    GTACAACGAGTCGCAAGCTCGCGAGAGTAAGCTAATCTCTTAAAGCCGTT
    CTCAGTTCGGACTGTAGGCTGCAACTCGCCTACACGAAGTCGGAATCGCT
    AGTAATCGCGGATCAGCATGCCGCGGTGAATACGTTCCCGGGCCTTGTAC
    ACACCGCCCGTCACACC
    S12KG282 GGCK-266-2 926R-repeat
    SEQ ID NO: 49
    ACTCCCCAGGCGGAGTGCTTAATGCGTTAGCTCCGGCACTGAAGGGCGGA
    AACCCTCCAACACCTAGCACTCATCGTTTACGGCATGGACTACCAGGGTA
    TCTAATCCTGTTCGCTACCCATGCTTTCGAGCCTCAGCGTCAGTTGCAGA
    CCAGACAGCCGCCTTCGCCACTGGTGTTCTTCCATATATCTACGCATTCC
    ACCGCTACACATGGAGTTCCACTGTCCTCTTCTGCACTCAAGTCGCCCGG
    TTTCCGATGCACTTCTTCGGTTAAGCCGAAGGCTTTCACATCAGACCTAA
    GCAACCGCCTGCGCTCGCTTTACGCCCAATAAATCCGGATAACGCTTGCC
    ACCTACGTATTACCGCGGCTGCTGGCACGTAGTTAGCCGTGACTTTCTGG
    TTGGATACCGTCACTGCGTGAACAGTTACTCTCACGCACGTTCTTCTCCA
    ACAACAGAGCTTTACGAGCCGAAACCCTTCTTCACTCACGCGGTGTTGCT
    CCATCAGGCTTGCGCCCATTGTGGAAGATTCCCTACTGCTGCCTCCCGTA
    GGAGTATGGACCGTGTCTCAGTTCCATTGTGGCCGATCAGTCTCTCAACT
    CGGCTATGCATCATCGCTTGGTAACCGTTCACCTTACCAACTAGCTAATG
    CACCGCAGGT
    S12KG283 GGDK-266-2 591R
    SEQ ID NO: 50
    TTTCTGGTTGGATACCGTCACTGCGTGAACAGTTACTCTCACGCACGTTC
    TTCTCCAACAACAGAGCTTTACGAGCCGAAACCCTTCTTCACTCACGCGG
    TGTTGCTCCATCAGGCTTGCGCCCATTGTGGAAGATTCCCTACTGCTGCC
    TCCCGTAGGAGTATGGACCGTGTCTCAGTTCCATTGTGGCCGATCAGTCT
    CTCAACTCGGCTATGCATCATCGCCTTGGTAAGCCGTTACCTTACCAACT
    AGCTAATGCACCGCAGGTCCATCCCAGAGTGATAGCCAAAGCCATCTTTC
    AAACAAAAGCCATGTGGCTTTTGTTGTTATGCGGTATTAGCATCTGTTTC
    CAAATGTTATCCCCCGCTCCGGGGCAGGTTACCTACGTGTTACTCACCCG
    TCCGCCACTCACTGGTGATCCATCGTCAATCAGGTGCAAGCACCATCAAT
    CAGTTGGGCCAGTGCGTACGACTTGCATGTATTAGGCACACCGCCGGCGT
    TCATCCTGAGCCATGATCAAACTCT
    S12KG284 GGDK-266-2 RP2
    SEQ ID NO: 51
    TCCCGCCTTAGGCGGCTCCCTCCATAATGGTTAGGCCACCGACTTTGGGC
    GTTACAAACTCCCATGGTGTGACGGGCGGTGTGTACAAGGCCCGGGAACG
    TATTCACCGCGGCATGCTGATCCGCGATTACTAGCGATTCCGACTTCGTG
    TAGGCGAGTTGCAGCCTACAGTCCGAACTGAGAACGGCTTTAAGAGATTA
    GCTTACTCTCGCGAGCTTGCGACTCGTTGTACCGTCCATTGTAGCACGTG
    TGTAGCCCAGGTCATAAGGGGCATGATGATCTGACGTCGTCCCCACCTTC
    CTCCGGTTTGTCACCGGCAGTCTCACTAGAGTGCCCAACTTAATGCTGGC
    AACTAGTAACAAGGGTTGCGCTCGTTGCGGGACTTAACCCAACATCTCAC
    GACACGAGCTGACGACGACCATGCACCACCTGTCATTGCGTCCCCGAAGG
    GAACGCCTTATCTCTAAGGTTAGCGCAAGATGTCAAGACCTGGTAAGGTT
    CTTCGCGTAGCTTCGAATTAAACCACATGCTCCACCGCTTGTGCGGGCCC
    CCGTCAATTCCTTTGAGTTTCAACCTTGCGGTCGTACTCCCCAGGCGGAG
    TGCTTAATGCGTTAGCTCCGGCACTGAAGGGCGGAAACCCTCCAACACCT
    AGCACTCATCGTTTACGGCATGGACTACCAGGGTATCTAATCCTGTTCGC
    TACCCATGCTTTCGAGCCTCAGCGTCAGTTGCAGACCAGACAGCCGCCTT
    CGCCACTGGTG

    NCIMB 41850 GGDK 266—contains both Lactobacillus iohnsonii and Lactobacillus reuteri Lactobacillus reuteri
  • S12KG381 27F
    SEQ ID NO: 52
    GTGTCCTAATACATGCAAGTCGTACGCACTGGCCCAACTGATTGATGGTGCTTGCACCTGATTGACGATGGATCACCAGTGAGTGGCGGACG
    GGTGAGTAACACGTAGGTAACCTGCCCCGGAGCGGGGGATAACATTTGGAAACAGATGCTAATACCGCATAACAACAAAAGCCACATGGCTT
    TTGTTTGAAAGATGGCTTTGGCTATCACTCTGGGATGGACCTGCGGTGCATTAGCTAGTTGGTAAGGTAACGGCTTACCAAGGCGATGATGC
    ATAGCCGAGTTGAGAGACTGATCGGCCACAATGGAACTGAGACACGGTCCATACTCCTACGGGAGGCAGCAGTAGGGAATCTTCCACAATGG
    GCGCAAGCCTGATGGAGCAACACCGCGTGAGTGAAGAAGGGTTTCGGCTCGTAAAGCTCTGTTGTTGGAGAAGAACGTGCGTGAGAGTAACT
    GTTCACGCAGTGACGGTATCCAACCAGAAAGTCACGGCTAACTACGTGCCAGCAGCCGCGGTAATACGTAGGTGGCAAGCGTTATCCGGATT
    TATTGGGCGTAAAGCGAGCGCAGGCGGTTGCTTAGGTCTGATGTGAAAGCCTTCGGCTTAACCGAAGAAGTGCATCGGAAACCGGGCGACTT
    GAGTGC
    S12KG382 519F
    SEQ ID NO: 53
    TTATCCGGATTTATTGGGCGTAAAGCGAGCGCAGGCGGTTGCTTAGGTCTGATGTGAAAGCCTTCGGCTTAACCGAAGAAGTGCATCGGAAA
    CCGGGCAACTTGAGTGCAGAAGAGGACAGTGGAACTCCATGTGTAGCGGTGGAATGCGTAGATATATGGAAGAACACCAGTGGCGAAGGCGG
    CTGTCTGGTCTGCAACTGACGCTGAGGCTCGAAAGCATGGGTAGCGAACAGGATTAGATACCCTGGTAGTCCATGCCGTAAACGATGAGTGC
    TAGGTGTTGGAGGGTTTCCGCCCTTCAGTGCCGGAGCTAACGCATTAAGCACTCCGCCTGGGGAGTACGACCGCAAGGTTGAAACTCAAAGG
    AATTGACGGGGGCCCGCACAAGCGGTGGAGCATGTGGTTTAATTCGAAGCTACGCGAAGAACCTTACCAGGTCTTGACATCTTGCGCTAACC
    TTAGAGATAAGGCGTCCCTACGGGGACGCAATGACAGGTGGTGCATGGTCGTCGTCAGCTCGTGTCGTGAGATGTTGGGTTAAGTCCCGCAA
    CGAGCGCAACCCTTGTTACTAGTTGCCAGCATTAAGTTGGGCACTCTAGTGAGACTGCCGGTGACAAACCGGAGGAAGGTGGGGACGACGTC
    A
    S12KG383 926F
    SEQ ID NO: 54
    GGTGGAGCATGTGGTTTAATTCGAAGCTACGCGAAGAACCTTACCAGGTCTTGACATCTTGCGCTAACCTTAGAGATAAGGCGTTCCCTTCG
    GGGACGCAATGACAGGTGGTGCATGGTCGTCGTCAGCTCGTGTCGTGAGATGTTGGGTTAAGTCCCGCAACGAGCGCAACCCTTGTTACTAG
    TTGCCAGCATTAAGTTGGGCACTCTAGTGAGACTGCCGGTGACAAACCGGAGGAAGGTGGGGACGACGTCAGATCATCATGCCCCTTATGAC
    CTGGGCTACACACGTGCTACAATGGACGGTACAACGAGTCGCAAGCTCGCGAGAGTAAGCTAATCTCTTAAAGCCGTTCTCAGTTCGGACTG
    TAGGCTGCAACTCGCCTACACGAAGTCGGAATCGCTAGTAATCGCGGATCAGCATGCCGCGGTGAATACGTTCCCGGGCCTTGTACACACCG
    CCCGTCACACCATGGGAGTTTGTAACGCCCAAAGTCGGTGGCCTAACCATTATGGAGGGAGCCGCCTAAGGCGGGACAGATGACTGGGGTGA
    AGTCGTAACAAGGTAGCCGTA
    S12KG384 926R
    SEQ ID NO: 55
    TACTCCCCAGGCGGAGTGCTTAATGCGTGAGCTCCGGCACTGAAGGGCGGAAACCCTCCAACACCTAGCACTCATCGTTTACGGCATGGACT
    ACCAGGGTATCTAATCCTGTTCGCTACCCATGCTTTCGAGCCTCAGCGTCAGTTGCAGACCAGACAGCCGCCTTCGCCACTGGTGTTCTTCC
    ATATATCTACGCATTCCACCGCTACACATGGAGTTCCACTGTCCTCTTCTGCACTCAAGTCGCCCGGTTTCCGATGCACTTCTTCGGTTAAG
    CCGAAGGCTTTCACATCAGACCTAAGCAACCGCCTGCGCTCGCTTTACGCCCAATAAATCCGGATAACGCTTGCCACCTACGTATTACCGCG
    GCTGCTGGCACGTAGTTAGCCGTGACTTTCTGGTTGGATACCGTCACTGCGTGAACAGTTACTCTCACGCACGTTCTTCTCCAACAACAGAG
    CTTTACGAGCCGAAACCCTTCTTCACTCACGCGGTGTTGCTCCATCAGGCTTGCGCCCATTGTGGAAGATTCCCTACTGCTGCCTCCCGTAG
    GAGTATGGACCGTGTCTCAGTTCCATTGTGGCCGATCAGTCTCTCAACTCGGCTATGCATCATCGCCTTGGTAAGCCGTTACCTTACCAACT
    AGCTATGCACCGCAGGTCCATCCCAGAGTGATAGCCAAAGCCATCTTTCAAACAAAAGCC
    S12KG385 519R
    SEQ ID NO: 56
    GTGACTTTCTGGTTGGATACCGTCACTGCGTGAACAGTTACTCTCACGCACGTGCTTCTCCAACAACAGAGCTTTACGAGCCGAAACCCTTC
    TTCACTCACGCGGTGTTGCTCCATCAGGCTTCGCCCATTGTGGAAGATTCCCTACTGCTGCCTCCCGTAGGAGTATGGACCGTGTCTCAGTT
    CCATTGTGGCCGATCAGTCTCTCAACTCGGCTATGCATCATCGCCTTGGTAAGCCGTTACCTTACCAACTAGCTAATGCACCGCAGGTCCAT
    CCCAGAGTGATAGCCAAAGCCATCTTTCAAACAAAAGCCATGTGGCTTTTGTTGTTATGCGGTATTAGCATCTGTTTCCAAATGTTATCCCC
    CGCTCCGGGGCAGGTTACCTACGTGTTACTCACCCGTCCGCCACTCACTGGTGATCCATCGTCAATCAGGTGCAAGCACCATCAATCAGTTG
    GGCCAGTGCGTACGACTTGCATGTATTAGGCACACCGCCGGCGTTCATCCTGAGCCATGATCAAAC
    S12KG386 RP2
    SEQ ID NO: 57
    TCCCGCACTTAGGCGGCTCCCTCCATAATGGTTAGGCCACCGACTTTGGGCGTTACAAACTCCCATGGTGTGACGGGCGGTGTGTACAAGGC
    CCGGGAAGTATTCACCGCGGCATGCTGATCCGCGATTACTAGCGATTCCGACTTCGTGTAGGCGAGTTGCAGCCTACAGTCCGAACTGAGAA
    CGGCTTTAAGAGATTAGCTTACTCTCGCGAGCTTGCGACTCGTTGTACCGTCCATTGTAGCACGTGTGTAGCCCAGGTCATAAGGGGCATGA
    TGATCTGACGTCGTCCCCACCTTCCTCCGGTTTGTCACCGGCAGTCTCACTAGAGTGCCCAACTTAATGCTGGCAACTAGTAACAAGGGTTG
    CGCTCGTTGCGGGACTTAACCCAACATCTCACGACACGAGCTGACGACGACCATGCACCACCTGTCATTGCGTCCCCGAAGGGAACGCCTTA
    TCTCTAAGGTTAGCGCAAGATGTCAAGACCTGGTAAGGTTCTTCGCGTAGCTTCGAATTAACCACATGCTCCACCGCTTGTGCGGGCCCCCG
    TCAATTCCTTTGAGTTTCAACCTTGCGGTCGTACTCCCCAGGCGGAGTGCTTAATGCGTTAGCTCCGGCACTGAAGGGCGGAAACCCTCCAA
    CACCTAGCACTCATCGTTTACGGCATGGACTACCAGGGTATCTAATCCTGTTCGCTACCCATGCTTTCGAGCC
    NCIMB 42008 GGDK266a-L. johnsonii (sample 4a)
    S12KG399 27F
    SEQ ID NO: 58
    GCGTGCCTAATACATGCAAGTCGAGCGAGCTTGCCTAGATGATTTTAGTGCTTGCACTAAATGAAACTAGATACAAGCGAGCGGCGGACGGG
    TGAGTAACACGTGGGTAACCTGCCCAAGAGACTGGGATAACACCTGGAAACAGATGCTAATACCGGATAACAACACTAGACGCATGTCTAGA
    GTTTGAAAGATGGTTCTGCTATCCACTCTTGGATGGACCTGCGGTGCATTAGCTAGTTGGTAAGGTAACGGCTTACCAAGGCAATGATGCAT
    AGCCGAGTTGAGAGACTGATCGGCCACATTGGGACTGAGACACGGCCCAAACTCCTACGGGAGGCAGCAGTAGGGAATCTTCCACAATGGAC
    GAAAGTCTGATGGAGCAACGCCGCGTGAGTGAAGAAGGGTTTCGGCTCGTAAAGCTCTGTTGGTAGTGAAGAAAGATAGAGGTAGTAACTGG
    CCTTTATTTGACGGTAATTACTTAGAAAGTCACGGCTAACTACGTGCCAGCAGCCGCGGTAATACGTAGGTGGCAAGCGTTGTCCGGATTTA
    TTGGGCGTAAAGCGAGTGCAGGCGGTTCAATAAGTCTGATGTGAAAGCCTTCGGCTCAACCGGAGAATTGCATCAGAAACTGTTGAACTTGA
    GTGCAGAAGAGGAGAGTGGAACTCCATGTGTAGCGGTGGAATGCGTA
    S12KG400 519F
    SEQ ID NO: 59
    TGTCCGGATTTATTGGGCGTAAAGCGAGTGCAGGCGGTTCAATAAGTCTGATGTGAAAGCCTTCGGCTCAACCGGAGAATTGCATCAGAAAC
    TGTTGAACTTGAGTGCAGAAGAGGAGAGTGGAACTCCATGTGTAGCGGTGGAATGCGTAGATATATGGAAGAACACCAGTGGCGAAGGCGGC
    TCTCTGGTCTGCAACTGACGCTGAGGCTCGAAAGCATGGGTAGCGAACAGGATTAGATACCCTGGTAGTCCATGCCGTAAACGATGAGTGCT
    AAGTGTTGGGAGGTTTCCGCCTCTCAGTGCTGCAGCTAACGCATTAAGCACTCCGCCTGGGGAGTACGACCGCAAGGTTGAAACTCAAAGGA
    ATTGACGGGGGCCCGCACAAGCGGTGGGCATGTGGTTTAATTCGAAGCAACGCGAAGAACCTTACCAGGTCTTGACATCCAGTGCAAACCTA
    AGAGATTAGGTGTTCCCTTCGGGGACGCTGAGACAGGTGGTGCATGGCTGTCGTCAGCTCGTGTCGTGAGATGTTGGGTTAAGTCCCGCAAC
    GAGCGCAACCCTTGTCATTAGTTGCCATCATTAAGTTGGGCACTCTAATGAGACTGCCGGTGACAAACCGGAGGAAGGTGGGGAT
    S12KG401 926F
    SEQ ID NO: 60
    GGTGGAGCATGTGGTTTAATTCGAAGCAACGCGAAGAACCTTACCAGGTCTTGACATCCAGTGCAAACCTAAGAGATTAGGTGTTCCCTTCG
    GGGACGCTGAGACAGGTGGTGCATGGCTGTCGTCAGCTCGTGTCGTGAGATGTTGGGTTAAGTCCCGCAACGAGCGCAACCCTTGTCATTAG
    TTGCCATCATTAAGTTGGGCACTCTAATGAGACTGCCGGTGACAAACCGGAGGAAGGTGGGGATGACGTCAAGTCATCATGCCCTTATGACC
    TGGGCTACACACGTGCTACAATGGACGGTACAACGAGAAGCGAACCTGCGAAGGCAAGCGGATCTCTTAAAGCCGTTCTCAGTTCGGACTGT
    AGGCTGCAACTCGCCTACACGAAGCTGGAATCGCTAGTAATCGCGGATCAGCACGCCGCGGTGAATACGTTCCCGGGCCTTGTACACACCGC
    CCGTCACACCATGAGAGTCTGTAACACCCAAAGCCGGTGGGATAACCTTTATAGGAGTCAGCCGTCTAAGGTAGGACAGATGATTAGGGTGA
    AGTCGTAACAAGGTAG
    S12KG402 926R
    SEQ ID NO: 61
    TACTCCCCAGGCGGAGTGCTTAATGCGTTAGCTGCAGCACTGAGAGGCGGAAACCTCCCAACACTTAGCACTCATCGTTTACGGCATGGACT
    ACCAGGGTATCTAATCCTGTTCGCTACCCATGCTTTCGAGCCTCAGCGTCAGTTGCAGACCAGAGAGCCGCCTTCGCCACTGGTGTTCTTCC
    ATATATCTACGCATTCCACCGCTACACATGGAGTTCCACTCTCCTCTTCTGCACTCAAGTTCAACAGTTCTGATGCAATTCTCCGGTTGAGC
    CGAAGGCTTTCACATCAGACTTATTGAACCGCCTGCACTCGCTTTACGCCCAATAAATCCGGACAACGCTTGCCACCTACGTATTACCGCGG
    CTGCTGGCACGTTCACATCAGACTTATTGAACCGCCTGCACTCGCTTTACGCCCAATAAATCCGGACAACGCTTGCCACCTACGTATTACCG
    CGGCTGCTGGCACGTAGTTAGCCGTGACTTTCTAAGTAATTACCGTCAAATAAAGGCCAGTTACTACCTCTATCTTTCTTCACTACCAACAG
    AGCTTTACGAGCCGAAACCCTTCTTCACTCACGCGGCGTTGCTTCCATCAGACTTTCGTCCATTGTGGAAGATTCCCTACTGCTGCCTCCCG
    TAGGAGTTTGGGCCGTGTCTCAGTCCCAATGTGCCGATCAGTCTCTCAACTCGGCTATGCATCATTGCCTTGGTAAGCCGTTACCTTACCAA
    CTAGCTAATGCACCGCAGGTCCATCCAAGAGTGATAGACAGAACCATCTTTCAAACTCTAGACATGCGTCTAGTG
    S12KG403 519R
    SEQ ID NO: 62
    GTGACTTTCTAAGTAATTACCGTCAAATAAAGGCCAGTTACTACCTCTATCTTTCTTCACTACCAACAGAGCTTTACGAGCCGAAACCCTTC
    TTCACTCACGCGGCGTTGCTCCATCAGACTTTCGTCCATTGTGGAAGATTCCCTACTGCTGCCTCCCGTAGGAGTTTGGGCCGTGTCTCAGT
    CCCAATGTGGCCGATCAGTCTCTCAACTCGGCTATGCATCATTGCCTTGGTAAGCCGTTACCTTACCAACTAGCTAATGCACCGCAGGTCCA
    TCCAAGAGTGATAGCAGAACCATCTTTCAAACTCTAGACATGCGTCTAGTGTTGTTATCCGGTATTAGCATCTGTTTCCAGGTGTTATCCCA
    GTCTCTTGGGCAGGTTACCCACGTGTTACTCACCCGTCCGCCGCTCGCTTGTATCTAGTTTCATTTAGTGCAAGCACTAAAATCATCTAGGC
    AAGCTCGCTCGACTTGCATGTATTAGGCACGCCGCCAGCGTTCGTCCTGAGCCA
    S12KS404 RP2
    SEQ ID NO: 63
    TCCTACACTTAGACGGCTGACTCCTATAAAGGTTATCCCACCGGCTTTGGGTGTTACAGACTCTCATGGTGTGACGGGCGGTGTGTACAAGG
    CCCGGGAACGTATTCACCGCGGCGTGCTGATCCGCGATTACTAGCGATTCCAGTTCGTGTAGGCGAGTTGCAGCCTACAGTCCGAACTGAGA
    ACGGCTTTAAGAGATCCGCTTGCCTTCGCAGGTTCGCTTCTCGTTGTACCGTCCATTGTAGCACGTGTGTAGCCCAGGTCATAAGGGGCATG
    ATGACTTGACGTCATCCCCACCTTCCTCCGGTTTGTCACCGGCAGTCTCATTAGAGTGCCCAACTTAATGATGGCAACTAATGACAAGGGTT
    GCGCTCGTTGCGGGACTTAACCCAACATCTCACGACACGAGCTGACGACAGCCATGCACCACCTGTCTCAGCGTCCCCGAAGGGAACACCTA
    ATCTCTTAGGTTTGCACTGGATGTCAAGACCTGGTAAGGTTCTTCGCGTTGCTTCGAATTAAACCACATGCTCCACCGCTTGTGCGGGCCCC
    CGTCAATTCCTTTGAGTTTCAACCTTGCGGTCGTACTCCCCAGGCGGAGTGCTTAATGCGTTAGCTGCAGCACTGAGAGGCGGAAACCTCCC
    AACACTTAGCACTCATCGTTTACGGCATGGACTACCAGGGTATCTAATCCTGTTCGCTACCCATGC
    MCIMB 42009 GGDK286h-L. reuteri (sample 6e)
    S12KG411 37F
    SEQ ID NO: 64
    GTGTGCCTAATACATGCAAGTCGTACGCACTGGCCCAACTGATTGATGGTGCTTGCACCTGATTGACGATGGATCACCAGTGAGTGGCGGAC
    GGGTGAGTAACACGTAGGTAACCTGCCCCGGAGCGGGGGATAACATTTGGAAACAGATGCTAATACCGCATAACAACAAAAGCCACATGGCT
    TTTGTTTGAAAGATGGCTTTGGCTATCACTCTGGGATGGACCTGCGGTGCATTAGCTAGTTGGTAAGGTAACGGCTTACCAAGGCGATGATG
    CATAGCCGAGTTGAGAGACTGATCGGCCACAATGGAACTGAGACACGGTCCATACTCCTACGGAGGCAGCAGTAGGGAATCTTCCACAATGG
    GCGCAAGCCTGATGGAGCAACACCGCGTGAGTGAAGAAGGGTTTCGGCTCGTAAAGCTCTGTTGTTGGAGAAGAACGTGCGTGAGAGTAACT
    GTTCACGCAGTGACGGTATCCAACCAGAAAGTCACGGCTAACTACGTGCCAGCAGCCGCGGTAATACGTAGGTGGCAAGCGT
    S12KG412 519F
    SEQ ID NO: 65
    TATCCGGATTTATTGGGCGTAAAGCGAGCGCAGGCGGTTGCTTAGGTCTGATGTGAAAGCCTTCGGCTTAACCGAAGAAGTGCATCGGAAAC
    CGGGCGACTTGAGTGCAGAAGAGGACAGTGGAACTCCATGTGTAGCCGGTGGAATGCGTAGATATATGGAAGAACACCAGTGGCGAAGGCGG
    CTGTCTGGTCTGCAACTGACGCTGAGGCTCGAAAGCATGGGTAGCGAACAGGATTAGATACCCTGGTAGTCCATGCCGTAAACGATGAGTGC
    TAGGTGTTGGAGGGTTTCCGCCCTTCAGTGCCGGAGCTAACGCATTAAGCACTCCGCCTGGGGAGTACGACCGCAAGGTTGAAACTCAAAGG
    AATTGACGGGGGCCCGCACAAGCGGTGGAGCATGTGGTTTAATTCGAAGCTACGCGAAGAACCTTACCAGGTCTTGACATCTTGCGCTAACC
    TTAGAGATAAGGCGTTCCCTTCGGGACGCAATGACAGGTGGTGCATGGTCGTCGTCAGCTCGTGTCGTGAGATGTTGGGTTAAGTCCCGCAA
    CGAGCGCAACCCTTGTTACTAGTTGCCAGCATTAAGTTGGGCACTCTAGTGAGACTGCCGGTGACAAACCGGAGGA
    S12KG413 926F
    SEQ ID NO: 66
    GTGGAGCATGTGGTTTAATTCGAAGCTACGCGAAGAACCTTACCAGGTCTTGACATCTTGCGCTAACCTTAGAGATAAGGCGTTCCCTTCGG
    GGACGCAATGACAGGTGGTGCATGGTCGTCGTCAGCTCGTGTCGTGAGATGTTGGGTTAAGTCCCGCAACGAGCGCAACCCTTGTTACTAGT
    TGCCAGCATTAAGTTGGGCACTCTAGTGAGACTGCCGGTGACAAACCGGAGGAAGGTGGGGACGACGTCAGATCATCATGCCCCTTATGACC
    TGGGCTACACACGTGCTACAATGGACGGTACAACGAGTCGCAAGCTCGCGAGAGTAAGCTAATCTCTTAAAGCCGTTCTTCAGTTCGGACTG
    TAGGCTGCAACTCGCCTACACGAAGTCGGAATCGCTAGTAATCGCGGATCAGCATGCCGCGGTGAATACGTTCCCGGGCCTTGTACACACCG
    CCCGTCACACCATGGGAGTTTGTAACGCCCAAAGTCGGTGGCCTAACCATTATGGAGGGAGCCGCCTAAGGCGGGACAGATGACTGGGGTGA
    AGTCGT
    S12KG414 926R
    SEQ ID NO: 67
    TACTCCCCAGGCGGAGTGCTTAATGCGTTAGCTCCGGCACTGAAGGGCGGAAACCCTCCAACACCTAGCACTCATCGTTTACGGCATGGACT
    ACCAGGGTATCTAATCCTGTTCGCTACCCATGCTTTCGAGCCTCAGCGTCAGTTGCAGACCAGACAGCCGCCTTCGCCACTGGTGTTCTTCC
    ATATATCTACGCATTCCACCGCTACACATGGAGTTCCACTGTCCTCTTCTGCACTCAAGTCGCCCGGTTTCCGATGCACTTCTTCGGTTAAG
    CCGAAGGCTTTCACATCAGACCTAAGCAACCGCCTGCGCTCGCTTTACGCCCAATAAATCCGGATAACGCTTGCCACCTACGTATTACCGCG
    GCTGCTGGCACGTAGTTAGCCGTGACTTCTGGTTGGATACCGTCACTGCGTGAACAGTTACTCTCACGCACGTTCTTCTCCAACAACAGAGC
    TTTACGAGCCGAAACCCTTCTTCACTCACGCGGTGTTGCTCCATCAGGCTTGCGCCCATTGTGGAAGATTCCCTACTGCTGCCTCCCGTAGG
    AGTATGGACCGTGTCTCAGTTCCATTGTGGCCGATCAGTCTCTCAACTCGGCTATGCATCATCGCC
    S12KG415 519R
    SEQ ID NO: 68
    GTGACTTTCTGGTTGGATACCGTCACTGCGTGAACAGTTACTCTCACGCACGTTCTTCTCCAACAACAGAGCTTTACGAGCCGAAACCCTTC
    TTCACTCACGCGGTGTTGCTCCATCAGGCTTGCGCCCATTGTGGAAGATTCCCTACTGCTGCCTCCGTAGGAGTATGGACCGTGTCTCAGTT
    CCATTGTGGCCGATCAGTCTCTCAACTCGGCTATGCATCATCGCCTTGGTAAGCCGTTACCTTACCAACTAGCTAATGCACCGCAGGTCCAT
    CCCAGAGTGATAGCCAAAGCCATCTTTCAAACAAAAGCCATGTGGCTTTTGTTGTTATGCGGTATTAGCATCTGTTTCCAAATGTTATCCCC
    CGCTCCGGGGCAGGTTACCTACGTGTTACTCACCCGTCCGCCACTCACTGGTGATCCATCGTCAATCAGGTGCAAGCACCATCAATCAGTTG
    GCCAGTGCGTACGACTTGCATGTATTAGGCACACCGCCGGCGTTCAT
    S12KG416 RP2
    SEQ ID NO: 69
    TCCCGCCTTAGGCGGCTCCCTCCATAATGGTTAGGCCACCGACTTTGGGCGTTACAAACTCCCATGGTGTGACGGGCGGTGTGTACAAGGCC
    CGGGAACGTATTCACCGCGGCATGCTGATCCGCGATTACTAGCGATTCCGACTTCGTGTAGGCGAGTTGCAGCCTACAGTCCGAACTGAGAA
    CGGCTTTAAGAGATTAGCTTACTCTCGCGAGCTTGCGACTCGTTGTACCGTCCATTGTAGCACGTGTGTAGCCCAGGTCATAAGGGGCATGA
    TGATCTGACGTCGTCCCCACCTTCCTCCGGTTTGTCACCGGCAGTCTCACTAGAGTGCCCAACTTAATGCTGGCAACTAGTAACAAGGGTTG
    CGCTCGTTGCGGGACTTAACCCAACATCTCACGACACGAGCTGACGACGACCATGCACCACCTGTCATTGCGTCCCCGAAGGGAACGCCTTA
    TCTCTAAGGTTAGCGCAAGATGTCAAGACCTGGTAAGGTTCTTCGCGTAGCTTCGAATTAAACCACATGCTCCACCGCTTGTGCGGGCCCCC
    GTCAATTCTTTGAGTTTCAACCTTGCGGTCGTACTCC
    NCMIMB 42010 GGDK161a-L. plantarium (sample 7a)
    SEQ ID NO: 70
    GTGCCTAATACATGCAAGTCGAACGAACTCTGGTATTGATTGGTGCTTGCATCATGATTTACATTTGAGTGAGTGGCGAACTGGTGAGTAAC
    ACGTGGGAAACCTGCCCAGAAGCGGGGGATAACACCTGGAAACAGATGCTAATACCGCATAACAACTTGGACCGCATGGTCCGAGTTTGAAA
    GATGGCTTCGGCTATCACTTTTGGATGGTCCCGCGGCGTATTAGCTAGATGGTGAGGTAACGGCTCACCATGGCAATGATACGTAGCCGACC
    TGAGAGGGTAATCGGCCACATTGGGACTGAGACACGGCCCAAACTCCTACGGGAGGCAGCAGTAGGGAATCTTCCACAATGGACGAAAGTCT
    GATGGAGCAACGCCGCGTGAGTGAAGAAGGGTTTCGGCTCGTAAAACTCTGTTGTTAAAGAAGAACATATCTGAGAGTAACTGTTCAGGTAT
    TGACGGTATTTAACCAGAAAGCCACGGCTAACTACGTGCCAGCAGCCGCGGTAATACGTAGGTGGCAAGCGTTGTCCGGATTTATTGGGCGT
    AAAGCGAGCGCAGGCGGTTTTTAAGTCTGATGTGAAAGCCTTCGGCTCAACCGAAGAAGTGCATCGGAAACTGGGAAACTTGAGTGCAGAAG
    AGGACAGTGGAACTCATGTGT
    S12KG418 519F
    SEQ ID NO: 71
    TCCGGATTTATTGGGCGTAAAGCGAGCGCAGGCGGTTTTTTAAGTCTGATGTGAAAGCCTTCGGCTCAACCGAAGAAGTGCATCGGAAACTG
    GGAAACTTGAGTGCAGAAGAGGACAGTGGAACTCCATGTGTAGCGGTGAAATGCGTAGATATATGGAAGAACACCAGTGGCGAAGGCGGCTG
    TCTGGTCTGTAACTGACGCTGAGGCTCGAAAGTATGGGTAGCAAACAGGATTAGATACCCTGGTAGTCCATACCGTAAACGATGAATGCTAA
    GTGTTGGAGGGTTTCCGCCCTTCAGTGCTGCAGCTAACGCATTAAGCATTCCGCCTGGGGAGTACGGCCGCAAGGCTGAAACTCAAAGGAAT
    TGACGGGGGCCCGCACAAGCGGTGGAGCATGTGGTTTAATTCGAAGCTACGCGAAGAACCTTACCAGGTCTTGACATACTATGCAAATCTAA
    GAGATTAGACGTTCCCTTCGGGGACATGGATACAGGTGGTGCATGGTTGTCGTCAGCTCGTGTCGTGAGATGTTGGGTTAAGTCCCGCAACG
    AGCGCAACCCTTATTATCAGTTGCCAGCATTAAGTTGGGCACTCTGGTGAGACTGCCGGTGACAAACCGGAGGAAGGTGGGGATGACGTCAA
    ATCATCATGCCCCTTATGACCTGGGCTACACAC
    S12KG419 926F
    SEQ ID NO: 72
    GGTGGAGCATGTGGTTTAATTCGAAGCTACGCGAAGAACCTTACCAGGTCTTGACATACTATGCAAATCTAAGAGATTAGACGTTCCCTTCG
    GGGACATGGATACAGGTGGTGCATGGTTGTCGTCAGCTCGTGTCGTGAGATGTTGGGTTAAGTCCCGCAACGAGCGCAACCCTTATTATCAG
    TTGCCAGCATTAAGTTGGGCACTCTGGTGAGACTGCCGGTGACAAACCGGAGGAAGGTGGGGATGACGTCAAATCATCATGCCCCTTATGAC
    CTGGGCTACACACGTGCTACAATGGATGGTACAACGAGTTGCGAACTCGCGAGAGTAAGCTAATCTCTTAAAGCCATTCTCAGTTCGGATTG
    TAGGCTGCAACTCGCCTACATGAAGTCGGAATCGCTAGTAATCGCGGATCACCATGCCGCGGTGAATACGTTCCCGGGCCTTGTACACACCG
    CCCGTCACACCATGAGAGTTTGTAACACCCAAAGTCGGTGGGGTAACCTTTTAGGAACCAGCCGCCTAAGGTGGGACAGATGATTACGGTGA
    AGTCGTAACAAGGTAGCCCGTA
    S12KG420 526R
    SEQ ID NO: 73
    GTACTCCCCAGGCGGAATGCTTAATGCGTTAGCTGCAGCACTGAAGGGCGGAAACCCTCCAACACTTAGCATTCATCGTTTACGGTATGGAC
    TACCAGGGTATCTAATCCTGTTTGCTACCCATACTTTCGAGCCTCAGCGTCAGTTACAGACCAGACAGCCGCCTTCGCCACTGGTGTTCTTC
    CATATATCTACGCATTTCACCGCTACACATGGAGTTCCACTGTCCTCTTCTGCACTCAAGTTTCCCAGTTTCCGATGCACTTCTTCGGTTGA
    GCCGAAGGCTTTCACATCAGACTTAAAAAACCGCCTGCGCTCGCTTTACGCCCAATAAATCCGGACAACGCTTGCCACCTACGTATTACCGC
    GGCTGCTGGCACGTAGTTAGCCGTGGCTTTCTGGTTAAATACCGTCAATACCTGAACAGTTACTCTCAGATATGTTCTTCTTTAACAACAGA
    GTTTTACGAGCCGAAACCCTTCTTCACTCACGCGGCGTTGCTCCATCAGACTTTCGTCCATTGTGGAAGATTCCCTACTGCTGCCTCCCGTA
    GGAGTTTGGGCCGTGTCTCAGTCCCAATGTGGCCGATTACCCTCTCAGGTCGGCTACGTATCATTGCCATGGTGAGCCGTTACCTCACCATC
    TAGCTAATACGCCGCGGGACCATCCAAAAGTGATA
    S12KG421 519R
    SEQ ID NO: 74
    TGGCTTTCTGGTTAAATACCGTCAATACCTGAACAGTTACTCTCAGATATGTTCTTCTTTAACAACAGAGTTTTACGAGCCGAAACCCTTCT
    TCACTCACGCGGCGTTGCTCCATCAGACTTTCGTCCATTGTGGAAGATTCCCTACTGCTGCCTCCCGTAGGAGTTTGGGCCGTGTCTCAGTC
    CCAATGTGGCCGATTACCCTCTCAGGTCGGCTACGTATCATTGCCATGGTGAGCCGTTACCTCACCATCTAGCTAATACGCCGCGGGACCAT
    CTAAAAGTGATAGCCGAAGCCATCTTTCAAACTCGGACCATGCGGTCCAAGTTGTTATGCGGTATTAGCATCTGTTTCCAGGTGTTATCCCC
    CGCTTCTGGGCAGGTTTCCCACGTGTTACTCACCAGTTCGCCACTCACTCAAATGTAAATCATGATGCAAGCACCAATCAATACCAGAGTTC
    GTTCGACTTGCATGTATTAGGCACGCCGCCAGCGTTCGTCCTGAGCCATGATCAAACTCTA
    S12KG422 RP2
    SEQ ID NO: 75
    ACTTAGGCGGCTGGTTCCTAAAAGGTTACCCCACCGACTTTGGGTGTTACAAACTCTCATGGTGTGACGGGCGGTGTGTACAAGGCCCGGGA
    ACGTATTCACCGCGGCATGCTGATCCGCGATTACTAGCGATTCCGACTTCATGTAGGCGAGTTGCAGCCTACAATCCGAACTGAGAATGGCT
    TTAAGAGATTAGCTTACTCTCGCGAGTTCGCAACTCGTTGTACCATCCATTGTAGCACGTGTGTAGCCCAGGTCATAAGGGGCATGATGATT
    TGACGTCATCCCCACCTTCCTCCGGTTTGTCACCGGCAGTCTCACCAGAGTGCCCAACTTAATGCTGGCAACTGATAATAAGGATTGCGCTC
    GTTGCGGGACTTAACCCAACATCTCACGACACGAGCTGACGACAACCATGCACCACCTGTATCCATGTCCCCGAAGGGAACGTCTAATCTCT
    TAGATTTGCATAGTATGTCAAGACCTGGTAAGGTTCTTCGCGTAGCTTCGATTAAACCACATGCTCCACCGCTTGTGCGGGCCCCCGTCAAT
    TCCTTTGAGTTTCAGCCTTGCGGCCGTACTCCCCAGGCGGAATGCTTAATGCGTTAGCTGCAGCACTGAAGGGCGGAAACCCTCCA

    NCIMB 42011 GGDK161b—L.reuteri (sample 11a)
  • S12KG441 27F
    SEQ ID NO: 76
    TAATACATGCAAGTCGTACGCACTGGCCCAACTGATTGATGGTGCTTGCA
    CCTGATTGACGATGGATCACCAGTGAGTGGCGGACGGGTGAGTAACACGT
    AGGTAACCTGCCCCGGAGCGGGGGATAACATTTGGAAACAGATGCTAATA
    CCGCATAACAACAAAAGCCACATGGCTTTTGTTTGAAAGATGGCTTTGGC
    TATCACTCTGGGATGGACCTGCGGTGCATTAGCTAGTTGGTAAGGTAACG
    GCTTACCAAGGCGATGATGCATAGCCGAGTTGAGAGACTGATCGGCCACA
    ATGGAACTGAGACACGGTCCATACTCCTACGGGAGGGCAGCAGTAGGGAA
    TCTTCCACAATGGGCGCAAGCCTGATGGAGCAACACCGCGTGAGTGAAGA
    AGGGTTTCGGCTCGTAAAGCTCTGTTGTTGGAGAAGAACGTGCGTGAGAG
    TAACTGTTCACGCAGTGACGGTATCCAACCAGAAAGTCACGGCTAACTAC
    GTGCCAGCAGCCGCGGTAATACGTAGGTGGCAAGCGTGATCCGGATTTAT
    TGGGCGTAAAGCGAGCGCAGGCGGTTGCTTAGGTCTGATGTGAAAGCCTT
    CGGCTTAACCGAAGAAGTGCATCGGAGACGGGCGACTTGAGTGCA
    S12KG442 519F
    SEQ ID NO: 77
    TTATCCGGATTTATTGGGCGTAAAGCGAGCGCAGGCGGTTGCTTAGGTCT
    GATGTGAAAGCCTTCGGCTTAACCGAAGAAGTGCATCGGAAACCGGGCGA
    CTTGAGTGCAGAAGAGGACAGTGGAACTCCATGTGTAGCGGTGGAATGCG
    TAGATATATGGAAGAACACCAGTGGCGAAGGCGGCTGTCTGGTCTGCAAC
    TGACGCTGAGGCTCGAAAGCATGGGTAGCGAACAGGATTAGATACCCTGG
    TAGTCCATGCCGTAAACGATGAGTGCTAGGTGTTGGAGGGTTTCCGCCCT
    TCAGTGCCGGAGCTAACGCATTAAGCACTCCGCCTGGGGAGTACGACCGC
    AAGGTTGAAACGCAAAGGAATTGACGGGGGCCCGCACAAGCGGTGGAGCA
    TGTGGTTTAATTCGAAGCTACGCGAAGAACCTTACCAGGTCTTGACATCT
    TGCGCTAACCTTANAAGGCGTCCCCTTCGGGGACTCAATGACAGGTGGTG
    CATGGTT
    S12KG443 926F
    SEQ ID NO: 78
    GGTGGAGCATGTGGTTTAATTCGAAGCTACGCGAAGAACCTTACCAGGTC
    TTGACATCTTGCGCTAACCTTAGAGATAAGGCGTTCCCTTCGGGGACGCA
    ATGACAGGTGGTGCATGGTCGTCGTCAGCTCGTGTCGTGAGATGTTGGGT
    TAAGTCCCGCAACGAGCGCAACCCTTGTTACTAGTTGCCAGCATTAAGTT
    GGGCACTCTAGTGAGACTGCCGGTGACAAACCGGAGGAAGGTGGGGACGA
    CGTCAGATCATCATGCCCCTTATGACCTGGGCTACACACGTGCTACAATG
    GACGGTACAACGAGTCGCAAGCTCGCGAGAGTAAGCTAATYCTCTTAAAG
    CCGTTCTCAGTTCGGACTGTAGGCTGCAACTCGCCTACACGAAGTCGGAA
    TCGCTAGTAATCGCGGATCAGCATGCCGCGGTGAATACGTTCCCGGGCCT
    TGTACACACCGCCCGTCACACCATGGGAGTTTGTAACGCCCAAAGTCGGT
    GGCCTAACCTTTATGGAGGGAGCCGCCTAAGGCGGGACAGATGACTGGGG
    TGAAGTCGTAACAAGGTAG
    S12KG444 926R
    SEQ ID NO: 79
    No results
    S12KG445 519R
    SEQ ID NO: 80
    GTGACTTTCTGGTTGGATACCGTCACTGCGTGAACAGTTACTCTCACGCA
    CGTTCTTCTCCAACAACAGAGCTTTACGAGCCGAAACCCTTCTTCACTCA
    CGCGGTGTTGCTCCATCAGGCTTGCGCCCATTGTGGAAGATTCCCTACTG
    CTGCCTCCCGTAGGAGTATGGACCGTGTCTCAGTTCCATTGTGGCCGATC
    AGTCTCTCAACTCGGCTATGCATCATCGCCTTGGTAAGCCGTTACCTTAC
    CAACTAGCTAATGCACCGCAGGTCCATCCCAGAGTGATAGCCAAAGCCAT
    CTTTCAAACAAAAGCCATGTGGCTTTTGTTGTTATGCGGTATTAGCATCT
    GTTTCCAAATGTTATCCCCCGCTCCGGGGCAGGTTACCTACGTGTTACTC
    ACCCGTCCGCCACTCACTGGTAATCCATCGTCAATCAGGTGCAAGCACCA
    TCAATCAGTTGGGCCAGTGCGTACGACTTGCATGTATTAGGCACACCGCC
    GGCGTTCATCCTGAGCCA
    S12KG446 RP2
    SEQ ID NO: 81
    CTCCCTCCATAAAGGTTAGGCCACCGACTTTGGGCGTTACAAACTCCCAT
    GGTGTGACGGGCGGTGTGTACAAGGCCCGGGAACGTATTCACCGCGGCAT
    GCTGATCCGCGATTACTAGCGATTCCGACTTCGTGTAGGCGAGTTGCAGC
    CTACAGTCCGAACTGAGAACGGCTTTAAGAGATTAGCTTACTCTCGCGAG
    CTTGCGACTCGTTGTACCGTCCATTGTAGCACGTGTGTAGCCCAGGTCAT
    AAGGGGCATGATGATCTGACGTCGTCCCCACCTTCCTCCGGTTTGTCACC
    GGCAGTCTCACTAGAGTGCCCAACTTAATGCTGGCAACTACTAACAAGGG
    TTGCGCTCGTTGCGGGACTTAACCCAACATCTCACGACACGAGCTGACGA
    CGACCATGCACCACCTGTCATTGCGTCCCCGAAGGGAACGCCTTATCTCT
    AAGGTTAGCGCAAGATGTCAAGACCTGGTAAGGTTCTTCGCGTAGCTTCG
    AATTAAACCACATGCTCCACCGCTTGTGCGGGCCCCCGTCAATTCCTTTG
    AGTTTCAACCTTGGCGGTCGTACTCCCCAGGCGGAGTGCTTAATGCGTTA
    GCTCCGGCACTGAAGGGCGGAA

    NCIMB 42012 GGDKZ66c—L.reuteri (sample 1a)
  • 512KG381 27F
    SEQ ID NO: 82
    GTGTGCCTAATACATGCAAGTCGTACGCACTGGCCCAACTGATTGATGGT
    GCTTGCACCTGATTGACGATGGATCACCAGTGAGTGGCGGACGGGTGAGT
    AACACGTAGGTAACCTGCCCCGGAGCGGGGGATAACATTTGGAAACAGAT
    GCTAATACCGCATAACAACAAAAGCCACATGGCTTTTGTTTGAAAGATGG
    CTTTGGCTATCACTCTGGGATGGACCTGCGGTGCATTAGCTAGTTGGTAA
    GGTAACGGCTTACCAAGGCGATGATGCATAGCCGAGTTGAGAGACTGATC
    GGCCACAATGGAACTGAGACACGGTCCATACTCCTACGGGAGGCAGCAGT
    AGGGAATCTTCCACAATGGGCGCAAGCCTGATGGAGCAACACCGCGTGAG
    TGAAGAAGGGTTTCGGCTCGTAAAGCTCTGTTGTTGGAGAAGAACGTGCG
    TGAGAGTAACTGTTCACGCAGTGACGGTATCCAACCAGAAAGTCACGGCT
    AACTACGTGCCAGCAGCCGCGGTAATACGTAGGTGGCAAGCGTTATCCGG
    ATTTATTGGGCGTAAAGCGAGCGCAGGCGGTTGCTTAGGTCTGATGTGAA
    AGCCTTCGGCTTAACCGAAGAAGTGCATCGGAAACCGGGCGACTTGAGTG
    C
    S12KG382 519F
    SEQ ID NO: 83
    TTATCCGGATTTATTGGGCGTAAAGCGAGCGCAGGCGGTTGCTTAGGTCT
    GATGTGAAAGCCTTCGGCTTAACCGAAGAAGTGCATCGGAAACCGGGCAA
    CTTGAGTGCAGAAGAGGACAGTGGAACTCCATGTGTAGCGGTGGAATGCG
    TAGATATATGGAAGAACACCAGTGGCGAAGGCGGCTGTCTGGTCTGCAAC
    TGACGCTGAGGCTCGAAAGCATGGGTAGCGAACAGGATTAGATACCCTGG
    TAGTCCATGCCGTAAACGATGAGTGCTAGGTGTTGGAGGGTTTCCGCCCT
    TCAGTGCCGGAGCTAACGCATTAAGCACTCCGCCTGGGGAGTACGACCGC
    AAGGTTGAAACTCAAAGGAATTGACGGGGGCCCGCACAAGCGGTGGAGCA
    TGTGGTTTAATTCGAAGCTACGCGAAGAACCTTACCAGGTCTTGACATCT
    TGCGCTAACCTTAGAGATAAGGCGTCCCTTCGGGGACGCAATGACAGGTG
    GTGCATGGTCGTCGTCAGCTCGTGTCGTGAGATGTTGGGTTAAGTCCCGC
    AACGAGCGCAACCCTTGTTACTAGTTGCCAGCATTAAGTTGGGCACTCTA
    GTGAGACTGCCGGTGACAAACCGGAGGAAGGTGGAGGGACGACGTCA
    S12KG383 926F
    SEQ ID NO: 84
    GGTGGAGCATGTGGTTTAATTCGAAGCTACGCGAAGAACCTTACCAGGTC
    TTGACATCTTGCGCTAACCTTAGAGATAAGGCGTTCCCTTCGGGGACGCA
    ATGACAGGTGGTGCATGGTCGTCGTCAGCTGTGTCGTGAGATGTTGGGTT
    AAGTCCCGCAACGAGCGCAACCCTTGTTACTAGTTGCCAGCATTAAGTTG
    GGCACTCTAGTGAGACTGCCGGTGACAAACCGGAGGAAGGTGGGGACGAC
    GTCAGATCATCATGCCCCTTATGACCTGGGCTACACACGTGCTACAATGG
    ACGGTACAACGAGTCGCAAGCTCGCGAGAGTAAGCTAATCTCTTAAAGCC
    GTTCTCAGTTCGGACTGTAGGCTGCAACTCGCCTACACGAAGTCGGAATC
    GCTAGTAATCGCGGATCAGCATGCCGCGGTGAATACGTTCCCGGGCCTTG
    TACACACCGCCCGTCACACCATGGGAGTTTGTAACGCCCAAAGTCGGTGG
    CCTAACCATTATGGAGGGAGCCGCCTAAGGCGGGACAGATGACTGGGGTG
    AAGTCGTAACAAGGTAGCCGTA
    S12KG384 928R
    SEQ ID NO: 85
    TACTCCCCAGGCGGAGTGCTTAATGCGTGAGCTCCGGCACTGAAGGGCGG
    AAACCCTCCAACACCTAGCACTCATCGTTTACGGCATGGACTACCAGGGT
    ATCTAATCCTGTTCGCTACCCATGCTTTCGAGCCTCAGCGTCAGTTGCAG
    ACCAGACAGCCGCCTTCGCCACTGGTGTTCTTCCATATATCTACGCATTC
    CACCGCTACACATGGAGTTCCACTGTCCTCTTCTGCACTCAAGTCGCCCG
    GTTTCCGATGCACTTCTTCGGTTAAGCCGAAGGCTTTCACATCAGACCTA
    AGCAACCGCCTGCCGCTCGCTTTACGCCCAATAAATCCGGATAACGCTTG
    CCACCTACGTATTACCGCGGCTGCTGGCACGTAGTTAGCCGTGACTTTCT
    GGTTGGATACCGTCACTGCGTGAACAGTTACTCTCACGCACGTTCTTCTC
    CAACAACAGAGCTTTACTGAGCCGAAACCCTTCTTCACTCACGCGGTGTT
    GCTCCATCAGGCTTGCGCCCATTGTGGAAGATTCCCTACTGCTGCCTCCC
    GTAGGAGTATGGACCGTGTCTCAGTTCCATTGTGGCCGATCAGTCTCTCT
    CAACTCGGCTATGCATCATCGCCTTGGTAAGCCGTTACCTTACCAACTAG
    CTAATGCACCGCAGGTCCATCCCAGAGTGATAGCCAAAGCCATCTTTCAA
    ACAAAAGCC
    S12KG385 519R
    SEQ ID NO: 86
    GTGACTTTCTGGTTGGATACCGTCACTGCGTGAACAGTTACTCTCACGCA
    CGTGCTTCTCCAACAACAGAGCTTTACGAGCCGAAACCCTTCTTCACTCA
    CGCGGTGTTGCTCCATCAGGCTTGCGCCCATTGTGGAAGATTCCCTACTG
    CTGCCTCCCGTAGGAGTATGGACCGTGTCTCAGTTCCATTGTGGCCGATC
    AGTCTCTCAACTCGGCTATGCATCATCGCCTTGGTAAGCCGTTACCTTAC
    CAACTAGCTAATGCACCGCAGGTCCATCCCAGAGTGATAGCCAAAGCCAT
    CTTTCAAACAAAAGCCATGTGGCTTTTGTTGTTATGCGGTATTAGCATCT
    GTTTCCAAATGTTATCCCCCGCTCCGGGGCAGGTTACCTACGTGTTACTC
    ACCCGTCCGCCACTCACTGGTGATCCATCGTCAATCAGGTGCAAGCACCA
    TCAATCAGTTGGGCCAGTGCGTACGACTTGCATGTATTAGGCACACCGCC
    GGCGTTCATCCTGAGCCATGATCAAAC
    S12KG386 RP2
    SEQ ID NO: 87
    TCCCGCACTTAGGCGGCTCCCTCCATAATGGTTAGGCCACCGACTTTGGG
    CGTTACAAACTCCCATGGTGTGACGGCGGTGTGTACAAGGCCCGGGAACG
    TATTCACCGCGGCATGCTGATCCGCGATTACTAGCGATTCCGACTTCGTG
    TAGGCGAGTTGCAGCCTACAGTCCGAACTGAGAACGGCTTTAAGAGATTA
    GCTTACTCTCGCGAGCTTGCGACTCGTTGTACCGTCCATTGTAGCACGTG
    TGTAGCCCAGGTCATAAGGGGCATGATGATCTGACGTCGTCCCCACCTTC
    CTCCGGTTTGTCACCGGCAGTCTCACTAGAGTGCCCAACTTAATGCTGGC
    AACTAGTAACAAGGGTTGCGCTCGTTGCGGGACTTAACCCAACATCTCAC
    GACACGAGCTGACGACGACCATGCACCACCTGTCATTGCGTCCCCGAAGG
    GAACGCCTTATCTCTCTAAGGTTAGCGCAAGATGTCAAGACCTGGTAAGG
    TTCTTCGCGTAACTTCGAATTAAACCACATGTCCACCGCTTGTGCGGGCC
    CCCGTCAATTCCTTTGAGTTTCAACCTTGCGGTCGTACTCCCCAGGCGGA
    GTGCTTAATGCGTTAGCTCCGGCACTGAAGGGCGGAAACCCTCCAACACC
    TAGCACTCATCGTTTACGGCATGGACTACCAGGGTATCTAATCCTGTTCG
    CTACCCATGCTTTCGAGCC
  • REFERENCES
    • Blandino, G., Fazio, D., Di Marco, R. Probiotics: Overview of microbiological and immunological characteristics (2008). Expert Review of Anti-Infective Therapy, 6 (4), pp. 497-508.
    • Cintas L M, Casaus M P, Herranz C, Nes I F, Hernandez P E. Review: bacteriocins of lactic acid bacteria (2001). Food Sci Technol Int. 7(4):281-305.
    • Clarridge III, J. E. Impact of 16S rRNA gene sequence analysis for identification of bacteria on clinical microbiology and infectious diseases (2004). Clinical Microbiology Reviews, 17 (4), pp. 840-862.
    • Cotter, P. D., Hill, C, Ross, R. P. Food microbiology: Bacteriocins: Developing innate immunity for food (2005). Nature Reviews Microbiology, 3 (10), pp. 777-788.
    • De Angelis, M., Siragusa, S., Berloco, M., Caputo, L., Settanni, L., Alfonsi, G., Amerio, M., Grandi, A., Ragni, A., Gobbetti, M. Selection of potential probiotic lactobacilli from pig feces to be used as additives in pelleted feeding (2006). Research in Microbiology, 157 (8), pp. 792-801
    • Elmadfa, I., Klein, P., Meyer, A. L. Immune-stimulating effects of lactic acid bacteria in vivo and in vitro (2010). Proceedings of the Nutrition Society, 69 (3), pp. 416-420.
    • Gopal, P. K., Sullivan, P. A., Smart, J. B. Utilisation of galacto-oligosaccharides as selective substrates for growth by lactic acid bacteria including Bifidobacterium lactis DR10 and Lactobacillus rhamnosus DR20 (2001). International Dairy Journal, 11 (1-2), pp. 19-25.
    • Gousia, P., Economou, V., Sakkas, H., Leveidiotou, S., Papadopoulou, C. Antimicrobial resistance of major foodborne pathogens from major meat products (2011). Foodborne Pathogens and Disease, 8 (1), pp. 27-38.
    • Jackson M S, Bird A R, McOrist A L. Comparison of two selective media for the detection and enumeration of Lactobacilli in human faeces (2002). J Microbiol Methods. 51(3):313-21. 65
    • Korhonen, J. M., Sclivagnotis, Y., Wright, A. V. Characterization of dominant cultivable lactobacilli and their antibiotic resistance profiles from faecal samples of weaning piglets (2007). Journal of Applied Microbiology, 103 (6), pp. 2496-2503.
    • Lähteinen, T., Malinen, E., Koort, J. M. K., Mertaniemi-Hannus, U., Hankimo, T., Karikoski, N., Pakkanen, S., Laine, H., Sillanpää, H., Söderholm, H., Palva, A. Probiotic properties of Lactobacillus isolates originating from porcine intestine and feces (2010). Anaerobe, 16 (3), pp. 293-300
    • Liu, Y., Fatheree, N. Y., Mangalat, N., Rhoads, J. M. Human-derived probiotic Lactobacillus reuteri strains differentially reduce intestinal inflammation (2010). American Journal of Physiology—Gastrointestinal and Liver Physiology, 299 (5), pp. G1087-G1096.
    • Ljungh, A., Wadström, T. Lactic acid bacteria as probiotics (2006). Current Issues in Intestinal Microbiology, 7 (2), pp. 73-90.
    • Martin, R, Delgado, S, Maldonado, A, Jiménez, E, Olivares, M, Fernández, L, Sobrino, O J, Rodríguez, J M. Isolation of lactobacilli from sow milk and evaluation of their probiotic potential (2009). Journal of Dairy Research, 76 (4), pp. 418-425.
    • Mulder I E, Schmidt B, Stokes C R, Lewis M, Bailey M, Aminov R I, Prosser J I, Gill B P, Pluske J R, Mayer C D, Musk C C, Kelly D. Environmentally-acquired bacteria influence microbial diversity and natural innate immune responses at gut surfaces (2009). BMC Biol. 7:79.
    • Naughton P J; Grant G. (2005) Modelling of salmonellosis In: Microbial Ecology of the Growing Animal Holzapfel W H, Naughton P J. (Eds). London, Elsevier. pp. 235-257
    • Neeser, J.-R., Granato, D., Rouvet, M., Servin, A., Teneberg, S., Karlsson, K.-A. Lactobacillus johnsonii Lal shares carbohydrate-binding specificities with several enteropathogenic bacteria (2000). Glycobiology, 10 (11), pp. 1193-1199.
    • Nicolau, D. P. Current challenges in the management of the infected patient (2011). Current Opinion in Infectious Diseases, 24 (Suppl 1), pp. S1-S10.
    • Ohashi, Y., Ushida, K. Health-beneficial effects of probiotics: Its mode of action (2009). Animal Science Journal, 80 (4), pp. 361-371.
    • Reddy, K. B. P. K., Awasthi, S. P., Madhu, A. N., Prapulla, S. G. Role of cryoprotectants on the viability and functional properties of probiotic lactic acid bacteria during freeze drying (2009). Food Biotechnology, 23 (3), pp. 243-265.
    • Robertson, J. M. C., McKenzie, N. H., Duncan, M., Allen-Vercoe, E., Woodward, M. J., Flint, H. J., Grant, G. Lack of flagella disadvantages Salmonella enterica serovar Enteritidis during the early stages of infection in the rat (2003). Journal of Medical Microbiology, 52 (1), pp. 91-99. Schreiber, O., Petersson, J., Phillipson, M., Perry, M., Roos, S., Holm, L. Lactobacillus reuteri prevents colitis by reducing P-selectin-associated leukocyte- and platelet-endothelial cell interactions (2009). American Journal of Physiology—Gastrointestinal and Liver Physiology, 296 (3), pp. G534-G542.
    • Smith, C. L., Geier, M. S., Yazbeck, R., Torres, D. M., Butler, R. N., Howarth, G. S. Lactobacillus fermentum BR11 and fructo-oligosaccharide partially reduce jejunal inflammation in a model of intestinal mucositis in rats (2008). Nutrition and Cancer, 60 (6), pp. 757-767.
    • Strasser, S., Neureiter, M., Geppl, M., Braun, R., Danner, H. Influence of lyophilization, fluidized bed drying, addition of protectants, and storage on the viability of lactic acid bacteria (2009). Journal of Applied Microbiology, 107 (1), pp. 167-177.
    • Tomas, M. S. J., Bru, E., Martos, G., Nader-Macias, M. E. Stability of freeze-dried vaginal Lactobacillus strains in the presence of different lyoprotectors (2009). Canadian Journal of Microbiology, 55 (5), pp. 544-552.
    • Tzortzis, G., Baillon, M.-L. A., Gibson, G. R., Rastall, R. A. Modulation of anti-pathogenic activity in canine-derived Lactobacillus species by carbohydrate growth substrate (2004). Journal of Applied Microbiology, 96 (3), pp. 552-559.
    • Williams, N. T. Probiotics (2010). American Journal of Health-System Pharmacy, 67 (6), pp. 449-458.
    • Yao, W., Zhu Wei-yun, W.-Y., Smidt, H., Verstegen, M. W. A. Cultivation-Independent Analysis of the Development of the Lactobacillus spp. Community in the Intestinal Tract of Newborn Piglets (2011) Agricultural Sciences in China, 10 (3), pp. 438-447.
    • Yun, J. H., Lee, K. B., Sung, Y. K., Kim, E. B., Lee, H.-G., Choi, Y. J.
      Isolation and characterization of potential probiotic lactobacilli from pig feces (2009). Journal of Basic Microbiology, 49 (2), pp. 220-226.

Claims (21)

1.-31. (canceled)
32. A pharmaceutical composition comprising at least 103 colony forming units (CFU) of at least one Lactobacillus bacteria strain, and a pharmaceutically acceptable excipient, diluent, or carrier, wherein the at least one Lactobacillus bacteria strain comprises a 16S rRNA gene sequence with at least 95% sequence identity to a 16S rRNA gene sequence selected from the group consisting of SEQ ID NOs 10, 11, 13, and 14, as determined by a sequence alignment performed using BLAST, and wherein the pharmaceutical composition is a solid formulation.
33. The pharmaceutical composition of claim 32, wherein the pharmaceutical composition is lyophilized.
34. The pharmaceutical composition of claim 32, wherein the pharmaceutical composition is encapsulated in one or more capsules.
35. The pharmaceutical composition of claim 32, wherein the pharmaceutical composition further comprises a preservative or stabilizer.
36. The pharmaceutical composition of claim 35, wherein the preservative comprises sodium benzoate, sorbic acid, or esters of p-hydroxybenzoic acid.
37. The pharmaceutical composition of claim 32, wherein the pharmaceutical composition comprises from about 103 to about 1011 colony forming units (CFU) of the at least one Lactobacillus bacteria strain per gram of the pharmaceutical composition.
38. The pharmaceutical composition of claim 32, wherein the at least one Lactobacillus bacteria strain exhibits at least one characteristic selected from the group consisting of:
(i) antimicrobial activity against E. coli;
(ii) antimicrobial activity against S. enteritidis;
(iii) suppression of inflammation in intestinal pig epithelial cells (IPEC) induced by 12-O-tetradecaboylphorbol-13-acetate (PMA);
(iv) an ability to block the adherence or invasion of the IPEC by S. enteritidis;
(v) an ability to block the adherence or invasion of the IPEC by E. coli; and
(vi) absence of antibiotic resistance to an antibiotic selected from the group consisting of ampicillin, cefotaxime, chloramphenicol, erythromycin, gentamicin, tetracycline, vancomycin, metronizadole, nalidixic acid, and kanamycin.
39. The pharmaceutical composition of claim 38, wherein the at least one Lactobacillus bacteria strain exhibits at least two characteristics selected from the group of claim 38.
40. The pharmaceutical composition of claim 38, wherein the at least one Lactobacillus bacteria strain exhibits at least three characteristics selected from the group of claim 38.
41. The pharmaceutical composition of claim 32, wherein the at least one Lactobacillus bacteria strain is of a species selected from the group consisting of Lactobacillus johnsonii, Lactobacillus reuteri, Lactobacillus plantarum, Lactobacillus paraplantarum, Lactobacillus gasseri, Lactobacillus pentosus, Lactobacillus helveticus, Lactobacillus acidophilus, Lactobacillus vaginalis, Lactobacillus mucosae, and any combinations thereof.
42. A method of treating an intestinal disorder in a subject in need thereof, comprising administering to the subject a pharmaceutical composition comprising at least at least 103 colony forming units (CFU) of at least one Lactobacillus bacteria strain, and a pharmaceutically acceptable excipient, diluent, or carrier, wherein the at least one Lactobacillus bacteria strain comprises a 16S rRNA gene sequence with at least 93% sequence identity to a 16S rRNA gene sequence selected from the group consisting of SEQ ID NOs 10, 11, 13, and 14, as determined by sequence alignment performed using BLAST, and wherein the pharmaceutical composition is a solid formulation in unit dose form.
43. The method of claim 42, wherein the intestinal disorder is selected from the group consisting of salmonellosis, irritable bowel syndrome (IBS), inflammatory bowel disease (IBD), functional dyspepsia, functional constipation, functional diarrhea, functional abdominal pain, functional bloating, Epigastric Pain Syndrome, Postprandial Distress Syndrome, Crohn's disease, ulcerative colitis, gastroesophageal reflux disease (GERD), necrotizing enterocolitis, and any combination thereof.
44. The method of claim 42, wherein the administering is oral.
45. The method of claim 42, wherein the subject is a human.
46. The method of claim 42, wherein the at least one Lactobacillus bacteria strain is of a species selected from the group consisting of Lactobacillus johnsonii, Lactobacillus reuteri, Lactobacillus plantarum, Lactobacillus paraplantarum, Lactobacillus gasseri, Lactobacillus pentosus, Lactobacillus helveticus, Lactobacillus acidophilus, Lactobacillus vaginalis, Lactobacillus mucosae, and any combinations thereof.
47. A method of improving intestinal microbiota in a subject in need thereof, comprising administering to the subject a pharmaceutical composition comprising at least at least 103 colony forming units (CFU) of at least one Lactobacillus bacteria strain, and a pharmaceutically acceptable excipient, diluent, or carrier, wherein the at least one Lactobacillus bacteria strain comprises a 16S rRNA gene sequence with at least 95% sequence identity to a 16S rRNA gene sequence selected from the group consisting of SEQ ID NOs 10, 11, 13, and 14, as determined by sequence alignment performed using BLAST, and wherein the pharmaceutical composition is a solid formulation.
48. A method of improving growth in a subject in need thereof, comprising administering to the subject a pharmaceutical composition comprising at least at least 103 colony forming units (CFU) of at least one Lactobacillus bacteria strain, and a pharmaceutically acceptable excipient, diluent, or carrier, wherein the at least one Lactobacillus bacteria strain comprises a 16S rRNA gene sequence with at least 95% sequence identity to a 16S rRNA gene sequence selected from the group consisting of SEQ ID NO 41, as determined by sequence alignment performed using BLAST, and wherein the pharmaceutical composition is a solid formulation.
49. The method of claim 48, wherein the subject comprises an early-weaned animal.
50. The method of claim 48, wherein the subject comprises a sheep, a goat, a cow, a pig, a horse, a cat, a dog, a turkey, a duck, a chicken, a fish, or a crustacean.
51. The method of claim 48, wherein the improving growth comprises gaining weight.
US17/232,871 2011-07-14 2021-04-16 Lactic acid bacterial strains Abandoned US20220080003A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US17/232,871 US20220080003A1 (en) 2011-07-14 2021-04-16 Lactic acid bacterial strains

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
GB1112091.2 2011-07-14
GBGB1112091.2A GB201112091D0 (en) 2011-07-14 2011-07-14 Bacterial strains isolated from pigs
PCT/GB2012/051686 WO2013008039A2 (en) 2011-07-14 2012-07-13 Bacterial strains isolated from pigs
US201414232475A 2014-10-17 2014-10-17
US15/359,144 US10183046B2 (en) 2011-07-14 2016-11-22 Porcine lactic acid bacterial strains
US16/206,250 US11013773B2 (en) 2011-07-14 2018-11-30 Lactic acid bacterial strains
US17/232,871 US20220080003A1 (en) 2011-07-14 2021-04-16 Lactic acid bacterial strains

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US16/206,250 Continuation US11013773B2 (en) 2011-07-14 2018-11-30 Lactic acid bacterial strains

Publications (1)

Publication Number Publication Date
US20220080003A1 true US20220080003A1 (en) 2022-03-17

Family

ID=44586578

Family Applications (4)

Application Number Title Priority Date Filing Date
US14/232,475 Active US9539293B2 (en) 2011-07-14 2012-07-13 Porcine lactic acid bacterial strains
US15/359,144 Active US10183046B2 (en) 2011-07-14 2016-11-22 Porcine lactic acid bacterial strains
US16/206,250 Active US11013773B2 (en) 2011-07-14 2018-11-30 Lactic acid bacterial strains
US17/232,871 Abandoned US20220080003A1 (en) 2011-07-14 2021-04-16 Lactic acid bacterial strains

Family Applications Before (3)

Application Number Title Priority Date Filing Date
US14/232,475 Active US9539293B2 (en) 2011-07-14 2012-07-13 Porcine lactic acid bacterial strains
US15/359,144 Active US10183046B2 (en) 2011-07-14 2016-11-22 Porcine lactic acid bacterial strains
US16/206,250 Active US11013773B2 (en) 2011-07-14 2018-11-30 Lactic acid bacterial strains

Country Status (20)

Country Link
US (4) US9539293B2 (en)
EP (1) EP2732023B1 (en)
CN (2) CN104080903B (en)
BR (1) BR112014000835A2 (en)
CA (1) CA2841576A1 (en)
CY (1) CY1119911T1 (en)
DK (1) DK2732023T3 (en)
ES (1) ES2656037T3 (en)
GB (1) GB201112091D0 (en)
HR (1) HRP20180152T1 (en)
HU (1) HUE036181T2 (en)
LT (1) LT2732023T (en)
ME (1) ME02941B (en)
NO (1) NO2732023T3 (en)
PL (1) PL2732023T3 (en)
PT (1) PT2732023T (en)
RS (1) RS56853B1 (en)
RU (2) RU2677890C2 (en)
SI (1) SI2732023T1 (en)
WO (1) WO2013008039A2 (en)

Families Citing this family (58)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB201112091D0 (en) 2011-07-14 2011-08-31 Gt Biolog Ltd Bacterial strains isolated from pigs
GB201117313D0 (en) 2011-10-07 2011-11-16 Gt Biolog Ltd Bacterium for use in medicine
GB201306536D0 (en) 2013-04-10 2013-05-22 Gt Biolog Ltd Polypeptide and immune modulation
NO3193901T3 (en) 2014-12-23 2018-09-01
DK3065748T3 (en) 2014-12-23 2018-01-29 4D Pharma Res Ltd A BACTEROIDES THETAIOTAOMICRON STREAM AND ITS APPLICATION FOR REDUCING INFLAMMATION
SI3650033T1 (en) 2015-06-15 2022-05-31 4D Pharma Research Limited Compositions comprising bacterial strains
MA41060B1 (en) 2015-06-15 2019-11-29 4D Pharma Res Ltd Compositions comprising bacterial strains
MA41010B1 (en) 2015-06-15 2020-01-31 4D Pharma Res Ltd Compositions comprising bacterial strains
EP3636272A1 (en) 2015-06-15 2020-04-15 4D Pharma Research Limited Compositions comprising bacterial strains
TW202222339A (en) 2015-06-15 2022-06-16 英商4D製藥研究有限公司 Compositions comprising bacterial strains
MY187060A (en) * 2015-06-30 2021-08-28 Perfect China Co Ltd Bifidobacteria as probiotic foundation species of gut microbiota
GB201520497D0 (en) 2015-11-20 2016-01-06 4D Pharma Res Ltd Compositions comprising bacterial strains
MX2018006240A (en) 2015-11-20 2018-08-01 4D Pharma Res Ltd Compositions comprising bacterial strains.
GB201520631D0 (en) 2015-11-23 2016-01-06 4D Pharma Res Ltd Compositions comprising bacterial strains
GB201520638D0 (en) 2015-11-23 2016-01-06 4D Pharma Res Ltd Compositions comprising bacterial strains
CN105343133B (en) * 2015-12-08 2019-11-19 东北农业大学 A kind of compound probiotic that treating ulcerative colitis, drug and preparation method thereof
CN105343132B (en) * 2015-12-08 2019-04-26 东北农业大学 Composition, the drug and preparation method thereof for treating colitis
GB201612191D0 (en) 2016-07-13 2016-08-24 4D Pharma Plc Compositions comprising bacterial strains
SI3313423T1 (en) 2016-03-04 2019-07-31 4D Pharma Plc Compositions comprising bacterial blautia strains for treating visceral hypersensitivity
CN105779350B (en) * 2016-04-11 2019-10-15 北京科拓恒通生物技术股份有限公司 A kind of lactobacillus plantarum and application thereof of anti-Bacterium enteritidis infection
TW201821093A (en) 2016-07-13 2018-06-16 英商4D製藥有限公司 Compositions comprising bacterial strains
GB201621123D0 (en) 2016-12-12 2017-01-25 4D Pharma Plc Compositions comprising bacterial strains
CN106497854B (en) * 2017-01-19 2019-10-22 南京农业大学 Lactobacillus D8 and its application
DK3630136T3 (en) 2017-05-22 2021-05-25 4D Pharma Res Ltd COMPOSITIONS INCLUDING BACTERIA STRAINS
MA41708A (en) 2017-05-24 2020-04-08 4D Pharma Res Ltd COMPOSITIONS CONTAINING BACTERIAL STRAINS
PT3600363T (en) 2017-06-14 2021-02-03 4D Pharma Res Ltd Compositions comprising bacterial strains
LT3638271T (en) 2017-06-14 2021-01-11 4D Pharma Research Limited Compositions comprising bacterial strains
EP4104843A1 (en) 2017-06-14 2022-12-21 4D Pharma Research Limited Compositions comprising bacterial strains
KR102250597B1 (en) 2017-11-20 2021-05-11 경희대학교 산학협력단 Novel lactic acid bacteria and use thereof
WO2019141997A1 (en) 2018-01-19 2019-07-25 4D Pharma Research Limited Combination therapy for treating or preventing cancer
CN111902153A (en) 2018-01-19 2020-11-06 4D制药研究有限公司 Combination therapy for treating or preventing cancer
TW201934139A (en) 2018-01-19 2019-09-01 英商4D製藥研究有限公司 Combination therapy for treating or preventing cancer
EP3740218A1 (en) 2018-01-19 2020-11-25 4D Pharma Research Limited Combination therapy for treating or preventing cancer
CA3094297A1 (en) 2018-03-19 2019-09-26 4D Pharma Research Limited Compositions comprising bacterial strains
TW202014513A (en) 2018-05-11 2020-04-16 英商4D製藥研究有限公司 Compositions comprising bacterial strains
WO2019238969A1 (en) 2018-06-14 2019-12-19 4D Pharma Research Ltd Compositions comprising bacterial strains
LT3723777T (en) 2018-06-25 2021-06-10 4D Pharma Research Limited Compositions comprising bacterial strains
CA3106139A1 (en) 2018-07-16 2020-01-23 4D Pharma Research Limited Compositions comprising bacterial strains
TW202023590A (en) 2018-08-17 2020-07-01 英商4D製藥研究有限公司 Compositions comprising bacterial strains
US11590182B2 (en) 2018-09-10 2023-02-28 Ohio State Innovation Foundation Methods and compositions to modulate antibiotic resistance and gastrointestinal microbiota
WO2020058499A1 (en) 2018-09-20 2020-03-26 4D Pharma Research Limited Compositions comprising bacterial strains
TW202027767A (en) 2018-10-09 2020-08-01 英商4D製藥研究有限公司 Compositions comprising bacterial strains
WO2020079282A1 (en) 2018-10-19 2020-04-23 4D Pharma Research Ltd Compositions comprising bacterial strains
WO2020089488A1 (en) 2018-11-02 2020-05-07 4D Pharma Research Limited Compositions comprising bacterial strains
TW202038979A (en) 2018-12-12 2020-11-01 英商4D製藥研究有限公司 Compositions comprising bacterial strains
TW202038977A (en) 2018-12-12 2020-11-01 英商4D製藥研究有限公司 Compositions comprising bacterial strains
KR20210102311A (en) 2018-12-12 2021-08-19 4디 파마 리서치 리미티드 A composition for treating cancer comprising a parabacteroid bacterial strain
EP3917550B1 (en) 2019-05-10 2024-01-24 CJ Bioscience, Inc. Compositions comprising blautia hydrogenotrophica in the treatemnt of fibromyalgia
CA3145904A1 (en) 2019-07-05 2021-01-14 4D Pharma Research Limited Compositions comprising bacterial strains
JP2023503410A (en) 2019-11-20 2023-01-30 フォーディー ファーマ リサーチ リミテッド Compositions containing bacterial strains
CN110878266B (en) * 2019-11-21 2021-04-20 中国农业科学院兰州兽医研究所 Lactobacillus johnsonii and application thereof
EP3838281A1 (en) 2019-12-20 2021-06-23 4D Pharma Research Limited Compositions comprising bacterial strains
TW202140773A (en) 2020-01-27 2021-11-01 英商4D製藥研究有限公司 Compositions comprising bacterial strains
WO2021206106A1 (en) * 2020-04-08 2021-10-14 雪印メグミルク株式会社 Composition for improving intestinal bacterial flora
CN111549108B (en) * 2020-05-28 2023-08-04 浙江省农业科学院 Method for analyzing composition and functional difference of intestinal microorganisms of laying ducks by different residual feed intake
CN114214256B (en) * 2022-01-12 2022-06-28 哈尔滨美华生物技术股份有限公司 Lactobacillus gasseri for preventing and treating urogenital infection and application thereof
CN115786175B (en) * 2022-09-30 2023-12-08 广东省农业科学院动物卫生研究所 Lactobacillus mucosae and application thereof
CN116121122B (en) * 2022-12-08 2023-10-03 玫斯江苏宠物食品科技有限公司 Probiotic composite preparation for preventing and treating diarrhea of cats and application thereof

Family Cites Families (322)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US1008077A (en) 1908-12-28 1911-11-07 Bernard A Schroder Water-heater.
US1004601A (en) 1909-01-04 1911-10-03 James Steel Type-assembling device.
US1005857A (en) 1910-02-09 1911-10-17 Otis Elevator Co Alternating-current electromagnet.
US1018304A (en) 1910-10-13 1912-02-20 Roy C Douglas Fog-signal light.
US1008602A (en) 1911-02-14 1911-11-14 Golladay Lake Cereal-percolator.
NL154598B (en) 1970-11-10 1977-09-15 Organon Nv PROCEDURE FOR DETERMINING AND DETERMINING LOW MOLECULAR COMPOUNDS AND PROTEINS THAT CAN SPECIFICALLY BIND THESE COMPOUNDS AND TEST PACKAGING.
JPS5031249B1 (en) 1970-12-17 1975-10-08
US3817837A (en) 1971-05-14 1974-06-18 Syva Corp Enzyme amplification assay
JPS5710876B2 (en) 1974-04-08 1982-03-01
US3939350A (en) 1974-04-29 1976-02-17 Board Of Trustees Of The Leland Stanford Junior University Fluorescent immunoassay employing total reflection for activation
US3996345A (en) 1974-08-12 1976-12-07 Syva Company Fluorescence quenching with immunological pairs in immunoassays
US4277437A (en) 1978-04-05 1981-07-07 Syva Company Kit for carrying out chemically induced fluorescence immunoassay
US4275149A (en) 1978-11-24 1981-06-23 Syva Company Macromolecular environment control in specific receptor assays
US4366241A (en) 1980-08-07 1982-12-28 Syva Company Concentrating zone method in heterogeneous immunoassays
NL8300698A (en) 1983-02-24 1984-09-17 Univ Leiden METHOD FOR BUILDING FOREIGN DNA INTO THE NAME OF DIABIC LOBAL PLANTS; AGROBACTERIUM TUMEFACIENS BACTERIA AND METHOD FOR PRODUCTION THEREOF; PLANTS AND PLANT CELLS WITH CHANGED GENETIC PROPERTIES; PROCESS FOR PREPARING CHEMICAL AND / OR PHARMACEUTICAL PRODUCTS.
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US4683202A (en) 1985-03-28 1987-07-28 Cetus Corporation Process for amplifying nucleic acid sequences
DK122686D0 (en) 1986-03-17 1986-03-17 Novo Industri As PREPARATION OF PROTEINS
US5443826A (en) 1988-08-02 1995-08-22 Borody; Thomas J. Treatment of gastro-intestinal disorders with a fecal composition or a composition of bacteroides and E. Coli
KR100225087B1 (en) 1990-03-23 1999-10-15 한스 발터라벤 The expression of phytase in plants
AU639570B2 (en) 1990-05-09 1993-07-29 Novozymes A/S A cellulase preparation comprising an endoglucanase enzyme
GB9107305D0 (en) 1991-04-08 1991-05-22 Unilever Plc Probiotic
EP0581171B1 (en) 1992-07-20 1998-02-04 Kabushiki Kaisha Yakult Honsha Species-specific oligonucleotides for bifidobacteria and a method of detection using the same
CA2151154C (en) 1992-12-10 1999-01-26 William E. Hintz Production of heterologous proteins in filamentous fungi
US5741665A (en) 1994-05-10 1998-04-21 University Of Hawaii Light-regulated promoters for production of heterologous proteins in filamentous fungi
US5599795A (en) 1994-08-19 1997-02-04 Mccann; Michael Method for treatment of idiopathic inflammatory bowel disease (IIBD)
AUPM823094A0 (en) 1994-09-16 1994-10-13 Goodman Fielder Limited Probiotic compositions
RU2078815C1 (en) 1995-01-17 1997-05-10 Московский научно-исследовательский институт эпидемиологии и микробиологии им.Г.Н.Габричевского Strain of bacterium bifidobacterium breve used for preparing the bacterial curative-prophylaxis bifido-containing preparations
JPH08259450A (en) 1995-03-17 1996-10-08 Nichinichi Seiyaku Kk Enhancer for production of interferon
US6861053B1 (en) 1999-08-11 2005-03-01 Cedars-Sinai Medical Center Methods of diagnosing or treating irritable bowel syndrome and other disorders caused by small intestinal bacterial overgrowth
AUPN698495A0 (en) 1995-12-06 1996-01-04 Pharma Pacific Pty Ltd Improved therapeutic formulation and method
SE508045C2 (en) 1996-02-26 1998-08-17 Arla Ekonomisk Foerening Adhesion inhibitors, preparations containing the same and process for their preparation
KR20000064729A (en) 1996-03-20 2000-11-06 번스 필프 앤드 컴파니 리미티드 Alteration of microbial populations in the gastrointestinal tract
CN1120235C (en) 1996-03-27 2003-09-03 诺沃奇梅兹有限公司 Alkaline protease deficient filamentous fungi
US6033864A (en) 1996-04-12 2000-03-07 The Regents Of The University Of California Diagnosis, prevention and treatment of ulcerative colitis, and clinical subtypes thereof, using microbial UC pANCA antigens
WO1998043081A1 (en) 1997-03-26 1998-10-01 Ligand Pharmaceuticals Incorporated Treatment of gastrointestinal disease with ppar modulators
SE511524C2 (en) 1997-06-02 1999-10-11 Essum Ab Lactobacillus casei rhamnosus strain and pharmaceutical preparation for the control of pathogenic intestinal bacteria
US5925657A (en) 1997-06-18 1999-07-20 The General Hospital Corporation Use of PPARγ agonists for inhibition of inflammatory cytokine production
AUPO758297A0 (en) 1997-06-27 1997-07-24 Rowe, James Baber Control of acidic gut syndrome
US5951977A (en) 1997-10-14 1999-09-14 The United States Of America, As Represented By The Secretary Of Agriculture Competitive exclusion culture for swine
IT1298918B1 (en) 1998-02-20 2000-02-07 Mendes Srl USE OF ARGININE DEIMINASE BACTERIA TO INDUCE APOPTOSIS AND / OR REDUCE AN INFLAMMATORY REACTION AND PHARMACEUTICAL COMPOSITIONS
DE19826928A1 (en) 1998-06-17 1999-12-23 Novartis Consumer Health Gmbh Medicines containing viable anaerobic bacteria that inhibit sulfate reduction by sulfate-reducing bacteria
ID29150A (en) 1999-01-15 2001-08-02 Entpr Ireland Cs USE OF LACTOBACILLUS SALIVARIUS
US7090973B1 (en) 1999-04-09 2006-08-15 Oscient Pharmaceuticals Corporation Nucleic acid sequences relating to Bacteroides fragilis for diagnostics and therapeutics
ATE252091T1 (en) 1999-08-27 2003-11-15 Lilly Co Eli BIARYL-OXA(THIA)ZOL DERIVATIVES AND THEIR USE AS PPARS MODULATORS
AU781415B2 (en) 2000-02-08 2005-05-19 Dsm Ip Assets B.V. Use of acid-stable proteases in animal feed
FR2808689B1 (en) 2000-05-11 2004-09-03 Agronomique Inst Nat Rech USE OF HYDROGENOTROPHIC ACETOGENIC STRAINS FOR THE PREVENTION OR TREATMENT OF DIGESTIVE DISORDERS
US20020013270A1 (en) 2000-06-05 2002-01-31 Bolte Ellen R. Method for treating a mental disorder
AUPQ899700A0 (en) 2000-07-25 2000-08-17 Borody, Thomas Julius Probiotic recolonisation therapy
DE10101793A1 (en) 2001-01-17 2002-08-01 Manfred Nilius Use of SLPI to treat inflammatory bowel disease
EP1227152A1 (en) 2001-01-30 2002-07-31 Société des Produits Nestlé S.A. Bacterial strain and genome of bifidobacterium
KR100437497B1 (en) * 2001-03-07 2004-06-25 주식회사 프로바이오닉 Acid-tolerant Lactobacillus reuteri Probio-16 suppressing the growth of pathogenic microorganisms and rotavirus, and Probiotics containing the same
US20050184033A1 (en) * 2001-03-21 2005-08-25 Johann Herrmann Utilization of a process gas mixture and method for laser beam welding
WO2002085933A1 (en) 2001-04-20 2002-10-31 The Institute For Systems Biology Toll-like receptor 5 ligands and methods of use
EP1260227A1 (en) * 2001-05-23 2002-11-27 Societe Des Produits Nestle S.A. Lipoteichoic acid from lactic acid bacteria and its use to modulate immune responses mediated by gram-negative bacteria, potential pathogenic gram-positive bacteria
PE20030284A1 (en) 2001-07-26 2003-05-01 Alimentary Health Ltd BIFIDOBACTERIUM STRAINS
AU2002341384A1 (en) 2001-09-05 2003-03-24 Actial Farmaceutica, Lda. Lactic acid bacteria comprising unmethylated cytosine-guanine dinucleotides for use in therapy
GB0127916D0 (en) 2001-11-21 2002-01-16 Rowett Res Inst Method
WO2003045317A2 (en) 2001-11-27 2003-06-05 Washington University Therapeutic protein and treatments
AU2002365279B2 (en) 2001-12-17 2009-08-13 Corixa Corporation Compositions and methods for the therapy and diagnosis of inflammatory bowel disease
US7101565B2 (en) 2002-02-05 2006-09-05 Corpak Medsystems, Inc. Probiotic/prebiotic composition and delivery method
DE10206995B4 (en) 2002-02-19 2014-01-02 Orthomol Pharmazeutische Vertriebs Gmbh Micronutrient combination product with pro- and prebiotics
JP2003261453A (en) 2002-03-11 2003-09-16 Nippon Berumu Kk Antitumor agent and radiation-protecting agent consisting of e. faecalis
PT1565547E (en) 2002-06-28 2008-11-03 Puleva Biotech Sa Probiotic strains, a process for the selection of them, compositions thereof, and their use
GB0307026D0 (en) 2003-03-27 2003-04-30 Rowett Res Inst Bacterial supplement
EP1481681A1 (en) 2003-05-30 2004-12-01 Claudio De Simone Lactic acid bacteria combination and compositions thereof
WO2005007834A1 (en) * 2003-07-23 2005-01-27 Probionic Corp. Acid tolerant probiotic lactobacillus plantarum probio-38 that can suppress the growth of pathogenic microorganism and tge coronavirus
US7485325B2 (en) 2003-08-06 2009-02-03 Gayle Dorothy Swain Animal food supplement compositions and methods of use
JP4683881B2 (en) 2003-08-27 2011-05-18 有限会社アーク技研 Antitumor active
US8192733B2 (en) 2003-08-29 2012-06-05 Cobb & Associates Probiotic composition useful for dietary augmentation and/or combating disease states and adverse physiological conditions
US20050163764A1 (en) 2003-09-22 2005-07-28 Yale University Treatment with agonists of toll-like receptors
GB0323039D0 (en) 2003-10-01 2003-11-05 Danisco Method
DK1675481T3 (en) 2003-10-24 2009-01-19 Nutricia Nv Synbiotic preparation for children
JP4850715B2 (en) 2003-12-17 2012-01-11 エヌ.ブイ.・ヌートリシア Lactic acid producing bacteria and lung function
ES2235642B2 (en) 2003-12-18 2006-03-01 Gat Formulation Gmbh CONTINUOUS MULTI-MICROENCAPSULATION PROCESS FOR THE IMPROVEMENT OF STABILITY AND STORAGE OF BIOLOGICALLY ACTIVE INGREDIENTS.
EP1727894A1 (en) 2004-03-22 2006-12-06 Government of the United States of America as represented by The Secretary of the Department of Health and Human Services Cellular and viral inactivation
US20080248068A1 (en) 2004-05-07 2008-10-09 Hans-Gustaf Ljunggren Use of Flagellin as an Adjuvant for Vaccine
ES2286558T5 (en) 2004-08-24 2013-10-15 N.V. Nutricia Nutritive composition comprising indigestible transgalactooligosaccharides and digestible galactose saccharides
US20060062774A1 (en) 2004-09-21 2006-03-23 The Procter & Gamble Company Compositions for maintaining and restoring normal urogenital flora
KR100468522B1 (en) 2004-10-12 2005-01-31 주식회사 프로바이오닉 2 -63new acid tolerant probiotic enterococcus faecium probio-63 that can suppress the growth of corona virus and porcine circovirus type 2
ITMI20042189A1 (en) 2004-11-16 2005-02-16 Anidral Srl COMPOSITION BASED ON PROBIOTIC BACTERIA AND ITS USE IN THE PREVENTION OF E-OR IN THE TREATMENT OF PATHOLOGIES AND-OR RESPIRATORY INFECTIONS AND IN THE IMPROVEMENT OF INTESTINAL FUNCTIONALITY
ES2702631T5 (en) 2005-02-28 2023-03-22 Nutricia Nv Nutritional composition with prebiotics and probiotics
US20090233888A1 (en) 2005-03-23 2009-09-17 Usc Stevens, University Of Southern California Treatment of disease conditions through modulation of hydrogen sulfide produced by small intestinal bacterial overgrowth
US20100048595A1 (en) 2005-03-23 2010-02-25 Washington University In St. Louis Use of archaea to modulate the nutrient harvesting functions of the gastrointestinal microbiota
JP2006265212A (en) 2005-03-25 2006-10-05 Institute Of Physical & Chemical Research Il-21 production-inducing agent
EP1714660A1 (en) 2005-04-21 2006-10-25 N.V. Nutricia Uronic acid and probiotics
EP1874917B1 (en) * 2005-04-26 2012-03-14 TEAGASC, The Agriculture and Food Development Authority Probiotic composition suitable for animals
US7572474B2 (en) 2005-06-01 2009-08-11 Mead Johnson Nutrition Company Method for simulating the functional attributes of human milk oligosaccharides in formula-fed infants
US8075934B2 (en) 2008-10-24 2011-12-13 Mead Johnson Nutrition Company Nutritional composition with improved digestibility
JP2007084533A (en) 2005-08-24 2007-04-05 Prima Meat Packers Ltd Immune response-modulating composition and food with the same as active ingredient
US7625704B2 (en) 2005-08-31 2009-12-01 Fred Hutchinson Cancer Research Center Methods and compositions for identifying bacteria associated with bacteria vaginosis
BRPI0615297A2 (en) 2005-09-01 2011-05-17 Schering Corp il-23 and il-17 antagonists to treat autoimmune inflammatory eye disease and their uses
WO2007035057A1 (en) 2005-09-23 2007-03-29 Gwangju Institute Of Science And Technology Composition for preventing or treating artritis comprising lactic acid bacteria and collangen as active ingredients
EP1776877A1 (en) 2005-10-21 2007-04-25 N.V. Nutricia Method for stimulating the intestinal flora
WO2007050656A2 (en) 2005-10-24 2007-05-03 Nestec S.A. Dietary fiber formulation and method of administration
JP2007116991A (en) 2005-10-28 2007-05-17 Eternal Light General Institute Inc Functional food
US8889149B2 (en) 2006-02-16 2014-11-18 Wayne State University Use of flagellin to prevent and treat gram negative bacterial infection
US20080260898A1 (en) 2006-03-17 2008-10-23 Marko Stojanovic Compositions comprising probiotic and sweetener components
JP5031249B2 (en) 2006-03-22 2012-09-19 学校法人北里研究所 Bacteria-containing composition having anti-inflammatory effect
EP2040724B1 (en) 2006-05-18 2011-10-05 Biobalance Llc Biotherapeutic compositions comprising probiotic escherichia coli and metronidazole and uses thereof
EP2021011A2 (en) 2006-05-26 2009-02-11 Nestec S.A. Methods of use and nutritional compositions of touchi extract
EP2046352A4 (en) 2006-06-06 2012-03-21 Univ Mcgill Fermented milk product and use thereof
US8691213B2 (en) 2006-08-04 2014-04-08 SHS International Protein free formula
WO2008031438A2 (en) 2006-09-13 2008-03-20 Region Hovedstaden V/Gentofte Hospital Treatment of asthma, eczema and/or allergy using non-pathogenic organisms
US20080069861A1 (en) 2006-09-19 2008-03-20 National Starch And Chemical Investment Holding Corporation Probiotic/Non-Probiotic Combinations
WO2008050209A1 (en) 2006-10-27 2008-05-02 Pfizer Products Inc. Hydroxypropyl methyl cellulose hard capsules and process of manufacture
US20080118473A1 (en) 2006-11-01 2008-05-22 The Procter & Gamble Company Methods of treating a respiratory condition comprising probiotic treatment
PL1920781T3 (en) 2006-11-10 2015-06-30 Glycotope Gmbh Compositions comprising a core-1 positive microorganism and their use for the treatment or prophylaxis of tumors
WO2008064489A1 (en) 2006-12-01 2008-06-05 Mcmaster University Probiotics to inhibit inflammation
DE102006062250A1 (en) 2006-12-22 2008-06-26 Roland Saur-Brosch Use of a composition of minerals and / or vitamins and optionally acetogenic and / or butyrogenic bacteria for oral or rectal administration for the treatment and prevention of abdominal discomfort
JP2008195635A (en) * 2007-02-09 2008-08-28 Crossfield Bio Inc Lactic acid bacteria preparation for horse
ES2388198T5 (en) 2007-02-28 2020-07-14 Mjn Us Holdings Llc Inactivated Lactobacillus Rhammosus GG to treat systemic inflammation in infants
EP2134835B1 (en) 2007-03-28 2014-10-15 Alimentary Health Limited Probiotic bifidobacterium strains
EP2134833B1 (en) 2007-03-28 2016-03-09 Alimentary Health Limited Probiotic bifidobacterium strain
WO2008134450A2 (en) * 2007-04-24 2008-11-06 Kemin Industries, Inc. Broad-spectrum antibacterial and antifungal activity of lactobacillus johnsonii d115
EP1997499A1 (en) 2007-05-31 2008-12-03 Puleva Biotech, S.A. Mammalian milk microorganisms, compositions containing them and their use for the treatment of mastitis
EP1997905A1 (en) 2007-06-01 2008-12-03 Friesland Brands B.V. Nucleic acid amplification
EP1997906A1 (en) 2007-06-01 2008-12-03 Friesland Brands B.V. Lactobacillus
EP1997907A1 (en) 2007-06-01 2008-12-03 Friesland Brands B.V. Bifidobacteria
WO2008153377A1 (en) 2007-06-15 2008-12-18 N.V. Nutricia Nutrition with non-viable bifidobacterium and non-digestible oligosaccharide
EP2522358B1 (en) 2007-06-27 2016-11-09 Laboratorios Ordesa, S.l. Peptides against rotavirus infection
HUP0700552A2 (en) 2007-08-27 2009-03-30 Janos Dr Feher Method and composition inhibiting inflammation
WO2009030254A1 (en) 2007-09-04 2009-03-12 Curevac Gmbh Complexes of rna and cationic peptides for transfection and for immunostimulation
US20100284979A1 (en) 2007-10-01 2010-11-11 University College Cork, National University Of Ireland, Cork Modulation of Tissue Fatty Acid Composition of a Host by Human Gut Bacteria
ES2431572T3 (en) 2007-10-20 2013-11-27 Université de Liège Bifidobacterial species
CA2740434C (en) 2007-10-26 2017-11-07 Brenda E. Moore Probiotic compositions and methods for inducing and supporting weight loss
EP2065048A1 (en) 2007-11-30 2009-06-03 Institut Pasteur Use of a L. casei strain, for the preparation of a composition for inhibiting mast cell activation
WO2009072889A1 (en) 2007-12-07 2009-06-11 N.V. Nutricia Bifidobacterium for dust mite allergy
WO2009079564A2 (en) 2007-12-17 2009-06-25 Emory University Immunogenic compositions and methods of use thereof
EP2268793A2 (en) 2008-02-06 2011-01-05 The Procter & Gamble Company Compositions methods and kits for enhancing immune response to a respiratory condition
WO2009128949A2 (en) 2008-04-18 2009-10-22 Vaxinnate Corporation Compositions of dengue viral proteins and methods of use
MX2008006546A (en) 2008-05-21 2009-11-23 Sigma Alimentos Sa De Cv Bifidobacteria that produces folic acid, food composition and use of said bifidobacteria.
CN102940652B (en) 2008-05-28 2015-03-25 青岛东海药业有限公司 Eubacterium biforme preparation and use thereof
CN101590081A (en) 2008-05-28 2009-12-02 青岛东海药业有限公司 Eubacterium ventriosum and Eubacterium biforme preparation and application thereof
US8586029B2 (en) 2008-06-04 2013-11-19 Trustees Of Dartmouth College Prevention or treatment of immune-relevant disease by modification of microfloral populations
EP2133088A3 (en) 2008-06-09 2010-01-27 Nestec S.A. Rooibos and inflammation
WO2009151315A1 (en) 2008-06-13 2009-12-17 N.V. Nutricia Nutritional composition for infants delivered via caesarean section
WO2009154463A2 (en) 2008-06-20 2009-12-23 Stichting Top Institute Food And Nutrition Butyrate as a medicament to improve visceral perception in humans
EP2138186A1 (en) 2008-06-24 2009-12-30 Nestec S.A. Probiotics, secretory IgA and inflammation
WO2010002241A1 (en) 2008-06-30 2010-01-07 N.V. Nutricia Nutritional composition for infants delivered via caesarean section
KR101017448B1 (en) 2008-09-18 2011-02-23 주식회사한국야쿠르트 Colorectal health promoting composition containing Bifidobacterium longum HY8004 as an effective factor
US20100074870A1 (en) 2008-09-19 2010-03-25 Bristol-Myers Squibb Company Probiotic infant products
US8137718B2 (en) 2008-09-19 2012-03-20 Mead Johnson Nutrition Company Probiotic infant products
KR101057357B1 (en) 2008-09-22 2011-08-17 광주과학기술원 Pharmaceutical and Food Compositions for Preventing or Treating Arthritis Comprising Lactic Acid Bacteria and Collagen as Active Ingredients
WO2010036876A2 (en) 2008-09-25 2010-04-01 New York University Compositions and methods for characterizing and restoring gastrointestinal, skin, and nasal microbiota
WO2010037408A1 (en) 2008-09-30 2010-04-08 Curevac Gmbh Composition comprising a complexed (m)rna and a naked mrna for providing or enhancing an immunostimulatory response in a mammal and uses thereof
EP2373182A1 (en) 2008-12-05 2011-10-12 Nestec S.A. Compositions for use in low-birth weight infants
RU2011129812A (en) 2008-12-19 2013-01-27 Нестек С.А. PREVENTION AND TREATMENT OF ROTAVIRAL DIARRHEA
IT1392672B1 (en) 2009-01-12 2012-03-16 Wyeth Consumer Healthcare S P A COMPOSITIONS INCLUDING PROBIOTIC COMPONENTS AND PREBIOTICS AND MINERAL SALTS, WITH LACTOFERRINA
EP2772269A3 (en) 2009-03-05 2015-01-14 Abbvie Inc. IL-17 binding proteins
JP5710876B2 (en) 2009-03-26 2015-04-30 クロスフィールドバイオ株式会社 Novel Bifidobacterium and its utilization
US8816067B2 (en) 2009-05-07 2014-08-26 Tate & Lyle Ingredients France SAS Compositions and methods for making alpha-(1,2)-branched alpha-(1,6) oligodextrans
EP2251022A1 (en) 2009-05-11 2010-11-17 Nestec S.A. Non-replicating micro-organisms and their immune boosting effect
EP2251020A1 (en) 2009-05-11 2010-11-17 Nestec S.A. Short-time high temperature treatment generates microbial preparations with anti-inflammatory profiles
CA2761444C (en) 2009-05-11 2018-04-24 Nestec S.A. Lactobacillus johnsonii la1 ncc533 (cncm i-1225) and immune disorders
KR20100128168A (en) 2009-05-27 2010-12-07 중앙대학교 산학협력단 Novel bacterial strains having excellent production potential of conjugated linoleic acid
US20100311686A1 (en) 2009-06-03 2010-12-09 Kasper Lloyd H Nutraceutical composition and methods for preventing or treating multiple sclerosis
WO2010143940A1 (en) 2009-06-12 2010-12-16 N.V. Nutricia Synergistic mixture of beta-galacto-oligosaccharides with beta-1,3 and beta-1,4/1,6 linkages
WO2010147714A1 (en) 2009-06-16 2010-12-23 The Trustees Of Columbia University In The City Of New York Autism-associated biomarkers and uses thereof
WO2011005756A1 (en) 2009-07-06 2011-01-13 Puretech Ventures, Llc Delivery of agents targeted to microbiota niches
AU2010285128B2 (en) 2009-08-18 2015-05-14 Société des Produits Nestlé S.A. A nutritional composition comprising Bifidobacterium longum strains and reducing food allergy symtoms, especially in infants and children
US20110053829A1 (en) 2009-09-03 2011-03-03 Curevac Gmbh Disulfide-linked polyethyleneglycol/peptide conjugates for the transfection of nucleic acids
WO2011036539A1 (en) 2009-09-23 2011-03-31 Borody Thomas J Therapy for enteric infections
EP2308498A1 (en) 2009-09-30 2011-04-13 Nestec S.A. Administration of Bifidobacterium breve during infancy to prevent inflammation later in life
EP2486143A1 (en) 2009-10-05 2012-08-15 AAK Patent B.V. Methods for diagnosing irritable bowel syndrome
EP2485742A4 (en) 2009-10-06 2013-03-20 Scott Dorfner Antibiotic formulations providing reduced gastrointentestinal side effects
PL2498789T3 (en) 2009-11-11 2017-01-31 Alimentary Health Limited Probiotic bifidobacterium strain
US20150104418A1 (en) 2014-12-18 2015-04-16 Microbios, Inc. Bacterial composition
FR2955774A1 (en) 2010-02-02 2011-08-05 Aragan PREPARATION FOR TREATING PONDERAL EXCES AND ASSOCIATED DISORDERS AND APPLICATIONS THEREOF
NL2004200C2 (en) 2010-02-05 2011-08-08 Friesland Brands Bv Use of sialyl oligosaccharides in weight management.
NL2004201C2 (en) 2010-02-05 2011-08-08 Friesland Brands Bv Use of sialyl oligosaccharides to modulate the immune system.
IT1398553B1 (en) 2010-03-08 2013-03-01 Probiotical Spa COMPOSITION INCLUDING PROBIOTIC BACTERIA FOR THE TREATMENT OF PATHOLOGIES ASSOCIATED WITH THE ALTERATION OF THE IMMUNE SYSTEM.
JP5737646B2 (en) 2010-03-24 2015-06-17 森下仁丹株式会社 Antiallergic agent
WO2011121379A1 (en) 2010-03-30 2011-10-06 Assistance Publique - Hopitaux De Paris Use of bifidobacteria for preventing allergy in breastfed infants
US8951512B2 (en) 2010-05-04 2015-02-10 New York University Methods for treating bone disorders by characterizing and restoring mammalian bacterial microbiota
WO2011149335A1 (en) 2010-05-25 2011-12-01 N.V. Nutricia Immune imprinting nutritional composition
CN102947441A (en) 2010-06-01 2013-02-27 穆尔研究企业有限责任公司 Cellular constituents from bacteroides, compositions thereof, and therapeutic methods employing bacteroides or cellular constituents thereof
WO2011151941A1 (en) 2010-06-04 2011-12-08 国立大学法人東京大学 Composition having activity of inducing proliferation or accumulation of regulatory t cell
TWI417054B (en) 2010-06-15 2013-12-01 Jen Shine Biotechnology Co Ltd Novel enterococcus faecium ljs-01 and its use for probiotic
EP2397145A1 (en) 2010-06-18 2011-12-21 Nestec S.A. L. johnsonii La1, B. longum NCC2705 and immune disorders
FR2962045B1 (en) 2010-07-05 2012-08-17 Bifinove MACROMOLECULAR COMPLEX OF BACTERIAL ORIGIN AND USE OF SAID MOLECULAR COMPLEX FOR PREVENTING AND TREATING INFLAMMATORY RHUMATISMS
TWI401086B (en) * 2010-07-20 2013-07-11 Univ China Medical Lactobacillus plantarum and uses thereof
WO2012024638A2 (en) 2010-08-20 2012-02-23 New York University Compositions and methods for treating obesity and related disorders by characterizing and restoring mammalian bacterial microbiota
KR101250463B1 (en) 2010-10-12 2013-04-15 대한민국 Oxygen tolerant Bifidobacterium longum from Korean neonate fecal samples and probiotic compositions produced by the same
CN102031235B (en) 2010-11-09 2012-07-25 中国农业大学 Enterococcus faecium ANSE228 and application thereof
EP2455092A1 (en) 2010-11-11 2012-05-23 Nestec S.A. Non-replicating probiotic micro-organisms protect against upper respiratory tract infections
US20120128644A1 (en) 2010-11-24 2012-05-24 Oragenics, Inc. Use of Bacteria to Treat and Prevent Respiratory Infections
CN102093967B (en) 2010-12-02 2013-01-30 中国农业科学院特产研究所 Mink source enterococcus faecium and application thereof
ES2389547B1 (en) 2010-12-07 2013-08-08 Consejo Superior De Investigaciones Científicas (Csic) BIFIDOBACTERIUM CECT 7765 AND ITS USE IN THE PREVENTION AND / OR TREATMENT OF OVERWEIGHT, OBESITY AND ASSOCIATED PATHOLOGIES.
KR20140030132A (en) 2011-01-10 2014-03-11 클리브랜드 바이오랩스, 아이엔씨. Use of toll-like receptor agonist for treating cancer
PL2481299T3 (en) 2011-01-31 2017-09-29 Synformulas Gmbh Bifidobacterium bifidum strains for application in gastrointestinal diseases
JP5840368B2 (en) 2011-02-02 2016-01-06 カルピス株式会社 Substances for preventing and improving arthritis
WO2012108830A1 (en) * 2011-02-09 2012-08-16 Lavivo Ab Synbiotic compositions for restoration and reconstitution of gut microbiota
ES2636439T3 (en) 2011-03-09 2017-10-05 Regents Of The University Of Minnesota Compositions and methods for colon microbiota transplantation
BRPI1100857A2 (en) 2011-03-18 2013-05-21 Alexandre Eduardo Nowill immunomodulatory agent and combinations thereof, their use and immunotherapeutic method for real time recontextualization, reprogramming and rebuilding of the immune system
WO2012140636A1 (en) 2011-04-11 2012-10-18 Alimentary Health Limited A probiotic formulation
WO2012142605A1 (en) 2011-04-15 2012-10-18 Samaritan Health Services Rapid recolonization deployment agent
BR112013026929A2 (en) 2011-04-20 2016-12-27 Mico Bio Inc composition and method for enhancing an immune response
WO2012158517A1 (en) 2011-05-13 2012-11-22 Glycosyn LLC The use of purified 2'-fucosyllactose, 3-fucosyllactose and lactodifucotetraose as prebiotics
KR20120133133A (en) 2011-05-30 2012-12-10 한국 한의학 연구원 Composition for Prevention or Treatment of Respiratory Disease Comprising Herbal Extract and Fermentation Product thereof with Lactic acid Bacteria
GB201110095D0 (en) 2011-06-15 2011-07-27 Danisco Method of treatment
JP2013005759A (en) 2011-06-24 2013-01-10 Kyodo Milk Industry Co Ltd Method for estimating intestinal bacterial flora of mouse
JP6222626B2 (en) 2011-07-07 2017-11-01 長岡香料株式会社 Fructose absorption inhibitor
GB201112091D0 (en) * 2011-07-14 2011-08-31 Gt Biolog Ltd Bacterial strains isolated from pigs
US20130022575A1 (en) 2011-07-19 2013-01-24 Microbial Rx Systems and methods of replacing intestinal flora
CN102304483A (en) 2011-08-12 2012-01-04 北京金泰得生物科技股份有限公司 Enterococcus faecium for feeding and applications thereof
KR101261872B1 (en) 2011-08-23 2013-05-14 대한민국 (식품의약품안전처장) A intestinal microbial enzyme mixture and it's preparation thereof
CA2848762C (en) 2011-09-14 2021-07-27 Queen's University At Kingston Method for treatment of disorders of the gastrointestinal system
GB201117313D0 (en) 2011-10-07 2011-11-16 Gt Biolog Ltd Bacterium for use in medicine
CA2850437A1 (en) 2011-10-11 2013-04-18 Achim Biotherapeutics Ab Composition comprising anaerobically cultivated human intestinal microbiota
CN103082292B (en) 2011-11-02 2015-03-04 深圳华大基因研究院 Use of Roseburia for the treatment and prevention of obesity-related diseases
CN102373172B (en) 2011-11-03 2013-03-20 北京龙科方舟生物工程技术有限公司 Enterococcus faecium and application thereof
CN104160014A (en) 2011-12-01 2014-11-19 国立大学法人东京大学 Human-derived bacteria that induce proliferation or accumulation of regulatory t cells
ES2408279B1 (en) 2011-12-15 2014-09-09 Universidad De Las Palmas De Gran Canaria PROBIOTIC LACTIC ACID BACTERIA
ITBG20120010A1 (en) 2012-02-24 2013-08-25 Milano Politecnico DEVICE FOR SURGICAL TRAINING
ITMI20120471A1 (en) 2012-03-26 2013-09-27 Giovanni Mogna COMPOSITION BASED ON BACTERIA BIFID BACTERIUM LONGUM STRIPS ABLE TO HELP THE EXTENSION OF LIFE
JP5792105B2 (en) 2012-03-27 2015-10-07 森永乳業株式会社 Method for producing lacto-N-biose I
JP6201982B2 (en) 2012-03-30 2017-09-27 味の素株式会社 Diabetes-inducing bacteria
WO2013154826A2 (en) 2012-04-11 2013-10-17 Nestec Sa Methods for diagnosing impending diarrhea
GB201206599D0 (en) 2012-04-13 2012-05-30 Univ Manchester Probiotic bacteria
EP2836218A4 (en) 2012-04-13 2015-10-21 Trustees Boston College Prebiotic compositions and methods of use
JP6436580B2 (en) 2012-06-04 2018-12-12 ガウラブ アグラーワル, Compositions and methods for treating Crohn's disease and related conditions and infections
WO2014001368A1 (en) 2012-06-25 2014-01-03 Orega Biotech Il-17 antagonist antibodies
ES2609654T3 (en) 2012-07-31 2017-04-21 Nestec S.A. Nutritive composition to promote the health of the musculoskeletal system of patients suffering from inflammatory bowel disease (IBD)
US20150216806A1 (en) 2012-08-29 2015-08-06 Salix Pharmaceuticals, Inc. Laxative compositions and methods for treating constipation and related gastrointestinal diseases and conditions
WO2014036182A2 (en) 2012-08-29 2014-03-06 California Institute Of Technology Diagnosis and treatment of autism spectrum disorder
EP2894985A4 (en) 2012-09-13 2016-09-28 Massachusetts Inst Technology Programmable drug delivery profiles of tumor-targeted bacteria
KR101473058B1 (en) 2012-09-19 2014-12-16 주식회사 쎌바이오텍 Composition for preventing or treating irritable bowel syndrome
CN103652322B (en) 2012-09-21 2016-02-10 临沂思科生物科技有限公司 A kind of preparation method of lactobacteria-containing composite probiotics feed additive
FR2997091B1 (en) 2012-10-22 2016-05-06 Fond Mediterranee Infection USE OF ANTIOXIDANT COMPOUND FOR THE CULTURE OF BACTERIA SENSITIVE TO OXYGEN TENSION
US9839657B2 (en) 2012-10-30 2017-12-12 Deerland Enzymes, Inc. Prebiotic compositions comprising one or more types of bacteriophage
WO2014067976A1 (en) 2012-10-30 2014-05-08 Nestec S.A. Compositions comprising microparticles and probiotics to deliver a synergistic immune effect
AU2013338774B2 (en) 2012-11-01 2017-03-02 Academisch Ziekenhuis Groningen Methods and compositions for stimulating beneficial bacteria in the gastrointestinal tract
WO2014075745A1 (en) 2012-11-19 2014-05-22 Université Catholique de Louvain Use of akkermansia for treating metabolic disorders
US8906668B2 (en) 2012-11-23 2014-12-09 Seres Health, Inc. Synergistic bacterial compositions and methods of production and use thereof
CA3212215A1 (en) 2012-11-23 2014-05-30 Seres Therapeutics, Inc. Synergistic bacterial compositions and methods of production and use thereof
EP2922555A4 (en) 2012-11-26 2016-06-15 Borody Thomas J Compositions for the restoration of a fecal microbiota and methods for making and using them
US20140193464A1 (en) 2013-01-08 2014-07-10 Imagilin Technology, Llc Effects of probiotics on humans and animals under environmental or biological changes
EP3904502A3 (en) 2013-02-04 2022-02-23 Seres Therapeutics, Inc. Compositions and methods
AU2014212004B2 (en) 2013-02-04 2018-09-20 Seres Therapeutics, Inc. Compositions for treating or preventing or reducing the severity of clostridium difficile related diseases
AU2014219048B2 (en) 2013-02-22 2018-12-13 The Regents Of The University Of California Compositions and methods for promoting growth of beneficial microbes to treat or prevent disease or prolong life
BR112015020819A2 (en) 2013-03-05 2017-07-18 Academisch Ziekenhuis Groningen use of faecalibacterium prausnitzii htf-f (dsm 26943) for suppression of inflammation
CA3101218A1 (en) 2013-03-14 2014-09-25 Therabiome, Llc Targeted gastrointestinal tract delivery of probiotic organisms and/or therapeutic agents
US20160040215A1 (en) 2013-03-14 2016-02-11 Seres Therapeutics, Inc. Methods for Pathogen Detection and Enrichment from Materials and Compositions
WO2014150094A1 (en) 2013-03-15 2014-09-25 University Of Florida Research Foundation, Inc. Butyrogenic bacteria as probiotics to treat clostridium difficile
CA2906921A1 (en) 2013-03-15 2014-09-18 Seres Therapeutics, Inc. Network-based microbial compositions and methods
CN103142656A (en) 2013-03-18 2013-06-12 广州知光生物科技有限公司 Application of bacteroides fragilis in preparing composition for preventing and treating colon cancer
CN103156888A (en) 2013-03-18 2013-06-19 广州知光生物科技有限公司 Application of bacteroides fragilis in preparation of composition for treating inflammatory bowel diseases
CN103146620A (en) 2013-03-25 2013-06-12 广州知光生物科技有限公司 Bacteroides fragilis with characteristics of probiotics
JP2014196260A (en) 2013-03-29 2014-10-16 公立大学法人奈良県立医科大学 Preventive or therapeutic composition of chronic obstructive pulmonary disease
GB201306536D0 (en) 2013-04-10 2013-05-22 Gt Biolog Ltd Polypeptide and immune modulation
WO2014182966A1 (en) 2013-05-10 2014-11-13 California Institute Of Technology Probiotic prevention and treatment of colon cancer
US9511099B2 (en) 2013-06-05 2016-12-06 Rebiotix, Inc. Microbiota restoration therapy (MRT), compositions and methods of manufacture
WO2014197562A1 (en) 2013-06-05 2014-12-11 Rebiotix, Inc. Microbiota restoration therapy (mrt), compositions and methods of manufacture
US20160120915A1 (en) 2013-06-10 2016-05-05 New York University Methods for manipulating immune responses by altering microbiota
WO2014200334A1 (en) 2013-06-14 2014-12-18 N.V. Nutricia Synbiotic composition for treatment of infections in allergic patients
WO2015003001A1 (en) 2013-07-01 2015-01-08 The Washington University Methods for identifying supplements that increase gut colonization by an isolated bacterial species, and compositions derived therefrom
WO2015003305A1 (en) 2013-07-08 2015-01-15 吉瑞高新科技股份有限公司 Electronic cigarette case
WO2015006355A2 (en) 2013-07-09 2015-01-15 Puretech Ventures, Llc Compositions containing combinations of bioactive molecules derived from microbiota for treatment of disease
WO2015013214A2 (en) 2013-07-21 2015-01-29 Whole Biome, Inc. Methods and systems for microbiome characterization, monitoring and treatment
EP3027058B1 (en) 2013-07-31 2020-07-15 Incredible Foods, Inc. Encapsulated functional food compositions
EP3033091B1 (en) 2013-08-16 2022-09-07 Versitech Limited Probiotic composition and use thereof in the prevention and treatment of hepatocellular carcinoma
CN103509741B (en) 2013-08-22 2015-02-18 河北农业大学 Blautia sp. AUH-JLD56 and application thereof in conversion of arctigenin
US10203329B2 (en) 2013-09-12 2019-02-12 The Johns Hopkins University Biofilm formation to define risk for colon cancer
RU2016116954A (en) 2013-10-18 2017-11-23 ИнноваЧайлдфуд АБ Balanced nutritional value combined food product for young children and young children and a method of manufacturing the specified product
PL229020B1 (en) 2013-11-13 2018-05-30 Inst Biotechnologii Surowic I Szczepionek Biomed Spolka Akcyjna New strain of Bifidobacterium breve
CA3203756A1 (en) 2013-11-25 2015-05-28 Seres Therapeutics, Inc. Synergistic bacterial compositions and methods of production and use thereof
EP3082431A4 (en) 2013-12-16 2017-11-15 Seres Therapeutics, Inc. Bacterial compositions and methods of use thereof for treatment of immune system disorders
CN103981115B (en) 2013-12-24 2018-10-26 北京大伟嘉生物技术股份有限公司 One plant height resistance enterococcus faecium and its application
CN103981117B (en) 2013-12-24 2018-10-26 北京大伟嘉生物技术股份有限公司 One plant height resistance enterococcus faecium and its cultural method and application
CN103820363B (en) 2014-01-27 2016-02-24 福建省农业科学院生物技术研究所 A kind of preparation and application of faecium bacterium powder
CN103865846B (en) 2014-02-27 2016-03-30 扬州绿保生物科技有限公司 A kind of faecium and preparation method thereof
CN103849590B (en) 2014-03-25 2016-07-06 上海交通大学 The one acidproof bifidobacterium breve BB8dpH of strain and application thereof
KR101683474B1 (en) 2014-03-26 2016-12-08 주식회사 쎌바이오텍 Composition for preventing or treating irritable bowel syndrome
US9783858B2 (en) 2014-04-02 2017-10-10 Northwestern University Altered microbiome of chronic pelvic pain
KR101583546B1 (en) 2014-04-09 2016-01-11 국립암센터 Method for prediction of reactivity to sorafenib treatment Using gene polymorphism
JP6617935B2 (en) 2014-04-10 2019-12-11 国立研究開発法人理化学研究所 Compositions and methods for the induction of Th17 cells
CN104195075B (en) 2014-08-14 2017-04-19 生合生物科技股份有限公司 Enterococcus faecium EF08 as well as feed additive and feed containing enterococcus faecium EF08
WO2015168534A1 (en) 2014-05-02 2015-11-05 Novogy, Inc. Therapeutic treatment of gastrointestinal microbial imbalances through competitive microbe displacement
SG11201608835VA (en) 2014-05-08 2016-11-29 Panoptes Pharma Ges M B H Compounds for treating ophthalmic diseases and disorders
CN106687130B (en) 2014-08-05 2020-01-21 深圳华大基因科技有限公司 Use of eubacterium for preventing and treating colorectal cancer-related diseases
EP3453396A1 (en) 2014-08-28 2019-03-13 Yale University Compositions and methods for treating an inflammatory disease or disorder
US20160058808A1 (en) 2014-09-03 2016-03-03 California Institute Of Technology Microbe-based modulation of serotonin biosynthesis
CN104546933A (en) 2014-09-30 2015-04-29 深圳华大基因科技有限公司 Application of bacteroides caccae in treatment or prevention of rheumatoid arthritis or related diseases thereof
CN104546934B (en) 2014-09-30 2019-04-09 深圳华大基因科技有限公司 Excrement pair bacteroid is treating or preventing the application in rheumatoid arthritis or its related disease
CN104546935A (en) 2014-09-30 2015-04-29 深圳华大基因科技有限公司 Application of bacteroides thetaiotaomicron in treating or preventing rheumatoid arthritis or related diseases thereof
CN104546940A (en) 2014-09-30 2015-04-29 深圳华大基因科技有限公司 Application of common bacteroides in treatment or prevention of rheumatoid arthritis or related diseases thereof
CN104546942A (en) 2014-09-30 2015-04-29 深圳华大基因科技有限公司 Application of bacteroides dorei in treating or preventing rheumatoid arthritis or related diseases thereof
CN104546932A (en) 2014-09-30 2015-04-29 深圳华大基因科技有限公司 Application of bacteroides ovatus in treating or preventing rheumatoid arthritis or related diseases thereof
US10046030B2 (en) 2014-10-07 2018-08-14 University Of Virginia Patent Foundation Compositions and methods for preventing and treating infection
AU2015335601A1 (en) 2014-10-24 2017-06-01 Evolve Biosystems Inc. Activated bifidobacteria and methods of use thereof
EP3212207A4 (en) 2014-10-30 2018-06-13 California Institute of Technology Compositions and methods comprising bacteria for improving behavior in neurodevelopmental disorders
US10124025B2 (en) 2014-10-30 2018-11-13 California Institute Of Technology Compositions and methods comprising bacteria for improving behavior in neurodevelopmental disorders
EP3881680A1 (en) 2014-10-31 2021-09-22 Pendulum Therapeutics, Inc. Methods and compositions relating to microbial treatment
CN104435000A (en) 2014-11-12 2015-03-25 江南大学 Application of lactic acid bacteria for treating bronchial asthma
JP6777643B2 (en) 2014-11-25 2020-10-28 メモリアル スローン ケタリング キャンサー センター Gut microbiota and GVHD
MA41020A (en) 2014-11-25 2017-10-03 Evelo Biosciences Inc PROBIOTIC AND PREBIOTIC COMPOSITIONS, AND THEIR METHODS OF USE FOR MODULATION OF THE MICROBIOME
NO3193901T3 (en) 2014-12-23 2018-09-01
DK3065748T3 (en) 2014-12-23 2018-01-29 4D Pharma Res Ltd A BACTEROIDES THETAIOTAOMICRON STREAM AND ITS APPLICATION FOR REDUCING INFLAMMATION
CN104560820B (en) 2014-12-30 2017-10-20 杭州师范大学 VREF KQ2.6 and application
KR20170128247A (en) 2015-01-23 2017-11-22 템플 유니버시티-오브 더 커먼웰쓰 시스템 오브 하이어 에듀케이션 Use of short-chain fatty acids in cancer prevention
CN105982919A (en) 2015-02-26 2016-10-05 王汉成 Biological retarder anti-cancer technology
WO2016139217A1 (en) 2015-03-04 2016-09-09 Ab-Biotics, S.A. Composition comprising anaerobically cultivated human intestinal microbiota
US20180078587A1 (en) 2015-03-18 2018-03-22 Trustees Of Tufts College Compositions and methods for preventing colorectal cancer
WO2016149687A1 (en) 2015-03-18 2016-09-22 Whole Biome, Inc. Methods and compositions relating to microbial treatment and diagnosis of skin disorders
CN107847530A (en) 2015-06-01 2018-03-27 芝加哥大学 By controlling symbiotic microorganism clump come treating cancer
MA41010B1 (en) 2015-06-15 2020-01-31 4D Pharma Res Ltd Compositions comprising bacterial strains
MA41060B1 (en) 2015-06-15 2019-11-29 4D Pharma Res Ltd Compositions comprising bacterial strains
TW202222339A (en) 2015-06-15 2022-06-16 英商4D製藥研究有限公司 Compositions comprising bacterial strains
EP3636272A1 (en) 2015-06-15 2020-04-15 4D Pharma Research Limited Compositions comprising bacterial strains
SI3650033T1 (en) 2015-06-15 2022-05-31 4D Pharma Research Limited Compositions comprising bacterial strains
CN105112333A (en) 2015-08-31 2015-12-02 江南大学 Bifidobacterium longum with good intestinal tract colonizing ability and screening method and application of bifidobacterium longum
MX2018006240A (en) 2015-11-20 2018-08-01 4D Pharma Res Ltd Compositions comprising bacterial strains.
GB201520497D0 (en) 2015-11-20 2016-01-06 4D Pharma Res Ltd Compositions comprising bacterial strains
GB201520638D0 (en) 2015-11-23 2016-01-06 4D Pharma Res Ltd Compositions comprising bacterial strains
GB201520631D0 (en) 2015-11-23 2016-01-06 4D Pharma Res Ltd Compositions comprising bacterial strains
AU2016361583B2 (en) 2015-11-25 2021-05-13 Memorial Sloan-Kettering Cancer Center Methods and compositions for reducing vancomycin-resistant enterococci infection or colonization
SI3313423T1 (en) 2016-03-04 2019-07-31 4D Pharma Plc Compositions comprising bacterial blautia strains for treating visceral hypersensitivity
TW201821093A (en) 2016-07-13 2018-06-16 英商4D製藥有限公司 Compositions comprising bacterial strains
GB201621123D0 (en) 2016-12-12 2017-01-25 4D Pharma Plc Compositions comprising bacterial strains
WO2018112363A1 (en) 2016-12-16 2018-06-21 Evelo Biosciences, Inc. Methods of treating cancer using parabacteroides
WO2018112365A2 (en) 2016-12-16 2018-06-21 Evelo Biosciences, Inc. Methods of treating colorectal cancer and melanoma using parabacteroides goldsteinii

Also Published As

Publication number Publication date
SI2732023T1 (en) 2018-03-30
US10183046B2 (en) 2019-01-22
HUE036181T2 (en) 2018-06-28
US11013773B2 (en) 2021-05-25
ME02941B (en) 2018-04-20
US20150050254A1 (en) 2015-02-19
RU2677890C2 (en) 2019-01-22
US9539293B2 (en) 2017-01-10
WO2013008039A2 (en) 2013-01-17
BR112014000835A2 (en) 2017-02-21
NO2732023T3 (en) 2018-04-21
CN109777746A (en) 2019-05-21
CN104080903A (en) 2014-10-01
CN104080903B (en) 2018-06-22
US20170173089A1 (en) 2017-06-22
PT2732023T (en) 2018-02-19
CY1119911T1 (en) 2018-06-27
DK2732023T3 (en) 2018-01-22
RS56853B1 (en) 2018-04-30
WO2013008039A3 (en) 2013-03-21
ES2656037T3 (en) 2018-02-22
GB201112091D0 (en) 2011-08-31
EP2732023A2 (en) 2014-05-21
RU2014105461A (en) 2015-08-20
LT2732023T (en) 2018-02-12
US20190216865A1 (en) 2019-07-18
PL2732023T3 (en) 2018-04-30
RU2019100532A (en) 2019-03-12
HRP20180152T1 (en) 2018-03-09
CA2841576A1 (en) 2013-01-17
EP2732023B1 (en) 2017-11-22

Similar Documents

Publication Publication Date Title
US20220080003A1 (en) Lactic acid bacterial strains
US11723933B2 (en) Composition of bacteroides thetaiotaomicron for immune modulation
Heinritz et al. Use of pigs as a potential model for research into dietary modulation of the human gut microbiota
JP7012377B2 (en) New bacterial species
Kumar et al. Probiotic potential of a Lactobacillus bacterium of canine faecal-origin and its impact on select gut health indices and immune response of dogs
Allaart et al. Predisposing factors and prevention of Clostridium perfringens-associated enteritis
EP2459203B1 (en) Lactic acid bacteria and bifidobacteria for treating endotoxemia
Argañaraz-Martínez et al. Physiological and functional characteristics of Propionibacterium strains of the poultry microbiota and relevance for the development of probiotic products
JP2015530401A (en) Probiotic composition and prebiotic composition
Dhama et al. Beneficial effects of probiotics and prebiotics in livestock and poultry: the current perspectives
Ihara et al. Retracted: Isolation, characterization, and effect of administration in vivo, a novel probiotic strain from pig feces
Modesto et al. Strategies to augment non-immune system based defence mechanisms against gastrointestinal diseases in pigs
Gupta et al. Selection criteria for probiotics and potential of cereal based food products as novel probiotic-carriers
Pedroso et al. The composition and role of the microbiota in chickens
de Almada et al. Paraprobiotics as potential agents for improving animal health
Sukegawa et al. Effects of oral administration of heat‐killed E nterococcus faecium strain NHRD IHARA in post‐weaning piglets
Gupta et al. Functional and probiotic characterization of Ligilactobacillus salivarius CPN60 isolated from calf faeces and its appraisal in rats
Tajima et al. Structure and function of a nonruminant gut: a porcine model
Kassa et al. Isolation, identification, and molecular characterization of probiotic bacteria from locally selected Ethiopian free range chickens gastrointestinal tract
TW201742920A (en) Immune modulation
Ajithdoss et al. Genomics of probiotic–host interactions
AU2018205072B2 (en) Immune modulation
TWI750342B (en) Composition comprising a bacterial strain and its use, process of producing and product
Heinritz Impact of diets differing in fat and fiber content on intestinal microbiota, microbial activity and metabolic markers in a pig model
Aminov Kiyoshi Tajima1 and Rustam Aminov2* National Agriculture and Food Research Organization, National Institute of Livestock

Legal Events

Date Code Title Description
AS Assignment

Owner name: 4D PHARMA RESEARCH LIMITED, UNITED KINGDOM

Free format text: CHANGE OF NAME;ASSIGNOR:GT BIOLOGICS LIMITED;REEL/FRAME:056079/0965

Effective date: 20150330

Owner name: GT BIOLOGICS LIMITED, UNITED KINGDOM

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:KELLY, DENISE;REEL/FRAME:056079/0935

Effective date: 20150327

AS Assignment

Owner name: OXFORD FINANCE LUXEMBOURG S.A R.L., LUXEMBOURG

Free format text: INTELECTUAL PROPERTY SECURITY AGREEMENT;ASSIGNORS:4D PHARMA PLC;4D PHARMA RESEARCH LIMITED;4D PHARMA CORK LIMITED;AND OTHERS;REEL/FRAME:057042/0715

Effective date: 20210729

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

AS Assignment

Owner name: ARMISTICE CAPITAL MASTER FUND LTD., NEW YORK

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:OXFORD FINANCE LUXEMBOURG S.A R.L.;REEL/FRAME:061806/0371

Effective date: 20221012

STCB Information on status: application discontinuation

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