NZ619605B2 - Production and use of bacterial histamine - Google Patents

Production and use of bacterial histamine Download PDF

Info

Publication number
NZ619605B2
NZ619605B2 NZ619605A NZ61960512A NZ619605B2 NZ 619605 B2 NZ619605 B2 NZ 619605B2 NZ 619605 A NZ619605 A NZ 619605A NZ 61960512 A NZ61960512 A NZ 61960512A NZ 619605 B2 NZ619605 B2 NZ 619605B2
Authority
NZ
New Zealand
Prior art keywords
histamine
histidine
strain
use according
production
Prior art date
Application number
NZ619605A
Other versions
NZ619605A (en
Inventor
Eamonn Connolly
Carissa Michelle Thomas
James Versalovic
Original Assignee
Biogaia Ab
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US13/552,686 external-priority patent/US20130022586A1/en
Application filed by Biogaia Ab filed Critical Biogaia Ab
Publication of NZ619605A publication Critical patent/NZ619605A/en
Publication of NZ619605B2 publication Critical patent/NZ619605B2/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23LFOODS, FOODSTUFFS, OR NON-ALCOHOLIC BEVERAGES, NOT COVERED BY SUBCLASSES A21D OR A23B-A23J; THEIR PREPARATION OR TREATMENT, e.g. COOKING, MODIFICATION OF NUTRITIVE QUALITIES, PHYSICAL TREATMENT; PRESERVATION OF FOODS OR FOODSTUFFS, IN GENERAL
    • A23L33/00Modifying nutritive qualities of foods; Dietetic products; Preparation or treatment thereof
    • A23L33/10Modifying nutritive qualities of foods; Dietetic products; Preparation or treatment thereof using additives
    • A23L33/135Bacteria or derivatives thereof, e.g. probiotics
    • AHUMAN NECESSITIES
    • 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
    • A23L33/17Amino acids, peptides or proteins
    • A23L33/175Amino acids
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23VINDEXING SCHEME RELATING TO FOODS, FOODSTUFFS OR NON-ALCOHOLIC BEVERAGES AND LACTIC OR PROPIONIC ACID BACTERIA USED IN FOODSTUFFS OR FOOD PREPARATION
    • A23V2002/00Food compositions, function of food ingredients or processes for food or foodstuffs
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23VINDEXING SCHEME RELATING TO FOODS, FOODSTUFFS OR NON-ALCOHOLIC BEVERAGES AND LACTIC OR PROPIONIC ACID BACTERIA USED IN FOODSTUFFS OR FOOD PREPARATION
    • A23V2200/00Function of food ingredients
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23VINDEXING SCHEME RELATING TO FOODS, FOODSTUFFS OR NON-ALCOHOLIC BEVERAGES AND LACTIC OR PROPIONIC ACID BACTERIA USED IN FOODSTUFFS OR FOOD PREPARATION
    • A23V2200/00Function of food ingredients
    • A23V2200/30Foods, ingredients or supplements having a functional effect on health
    • A23V2200/324Foods, ingredients or supplements having a functional effect on health having an effect on the immune system
    • A23Y2220/71
    • 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
    • A61K2035/11Medicinal preparations comprising living procariotic cells
    • A61K2035/115Probiotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/417Imidazole-alkylamines, e.g. histamine, phentolamine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/4172Imidazole-alkanecarboxylic acids, e.g. histidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7004Monosaccharides having only carbon, hydrogen and oxygen atoms
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/66Microorganisms or materials therefrom
    • A61K35/74Bacteria
    • A61K35/741Probiotics
    • A61K35/744Lactic acid bacteria, e.g. enterococci, pediococci, lactococci, streptococci or leuconostocs
    • A61K35/747Lactobacilli, e.g. L. acidophilus or L. brevis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/02Bacterial antigens
    • A61K39/09Lactobacillales, e.g. aerococcus, enterococcus, lactobacillus, lactococcus, streptococcus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/16Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing nitrogen, e.g. nitro-, nitroso-, azo-compounds, nitriles, cyanates
    • A61K47/18Amines; Amides; Ureas; Quaternary ammonium compounds; Amino acids; Oligopeptides having up to five amino acids
    • A61K47/183Amino acids, e.g. glycine, EDTA or aspartame
    • 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
    • A61P1/02Stomatological preparations, e.g. drugs for caries, aphtae, periodontitis
    • 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
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/02Drugs for genital or sexual disorders; Contraceptives for disorders of the vagina
    • 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
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/02Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving viable microorganisms
    • C12Q1/04Determining presence or kind of microorganism; Use of selective media for testing antibiotics or bacteriocides; Compositions containing a chemical indicator therefor
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/195Assays involving biological materials from specific organisms or of a specific nature from bacteria
    • G01N2333/335Assays involving biological materials from specific organisms or of a specific nature from bacteria from Lactobacillus (G)
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/569Immunoassay; Biospecific binding assay; Materials therefor for microorganisms, e.g. protozoa, bacteria, viruses
    • G01N33/56911Bacteria

Abstract

Disclosed is a method of selecting a probiotic lactic acid bacterial strain for use in the local production of histamine in a mammal, wherein the method comprises screening bacteria for the presence of an active histidine operon and selecting a strain which has an active histidine operon and is capable of producing histamine. Also disclosed is the use of such selected bacteria for the manufacture of a medicament for the treatment and/or prophylaxis of inflammatory conditions. ble of producing histamine. Also disclosed is the use of such selected bacteria for the manufacture of a medicament for the treatment and/or prophylaxis of inflammatory conditions.

Description

PRODUCTION AND USE OF BACTERIAL HISTAMINE FIELD OF THE INVENTION This invention relates to a method of ing specific probiotic lactic acid bacteria producing histamine and the use of such strains to deliver beneficial s for the host.
OUND OF THE INVENTION The Food and Agricultural Organization of the United Nations define probiotics as “live microorganisms which when administered in adequate amounts confer a health benefit on the host”. Nowadays, a number of different bacteria are used as probiotics for example, -acid producing bacteria such as strains of Lacrobacillus and Bifidobacreria.
Lactic-acid producing ia are not only used for their beneficial effect on human or animal health, but they are also widely used in the food industry for fermentation processes. The effectiveness of probiotics is strain-specific, and each strain may contribute to host health through different mechanisms. Probiotics can prevent or inhibit the proliferation of pathogens, suppress production of Virulence factors by pathogens, or modulate the immune response in a pro-inflammatory or an anti—inflammatory way. Use of different strains of the probiotic lactic—acid ing bacteria Lactobacillus remeri is a ing therapy for the amelioration of ile colic, alleviation of eczema, reduction of episodes of workplace illness, and suppression of Helicobacrer pylori infection. L. reuteri is considered an indigenous organism of the human gastrointestinal tract and is t for example on the mucosa of the gastric corpus, gastric antium, duodenum, and ileum. See for example US.
Patent Nos. 678, 5,458,875, 5,534,253, 5,837,238, and 289.
When L rezrteri cells are grown under anaerobic conditions in the presence of glycerol, they produce the crobial nce known as reuterin (B-hydroxy propionaldehyde).
The onship between a host and its microbes is complex, and for some bacteria, this host:microbe relationship has been developing over many years of co—evolution. This appears to be especially true for Lactobacillus reuteri. Our knowledge of the mutualistic relationship between gut microbes and the human host is in its infancy, but already we are keenly aware that the gut microbiome plays an essential role in gut and immune system development, nutrition, and new links are being established between the gut microbiome and the brain. Dysbiosis, the perturbation of the normal gut microbiome, has been implicated in 2 2012/064351 wide range of disease processes ing those affecting the local gut environment. such as Inflammatory Bowel Disease (IBD) and Irritable Bowel Syndrome (IBS), and disease processes at sites distant to the gut, such as the metabolic syndrome. Significant therapeutic ial lies within the gut microbiome, and research is striving towards a future goal of altering the microbial community in order to prevent and/or treat distinct disease processes.
There is therefore a need to understand such specific ctions between microbes and man related to a specific disease or other situations influencing the health of the host so that the most riate probiotic strains can be selected and used to counteract such developments.
SUMMARY OF THE INVENTION The invention herein provides a specific method of locally ing histamine in mammals, especially humans, the local production of histamine includes but are not limited to production in the GI tract, genitourinary (GU) tract, oral cavity, in the lungs and airways, on the skin etc, of the mammalian body by selecting certain strains of lactic acid bacteria. The bacteria may be red together with certain amino acids and/or sugars, separately administered or already t at the active site.
A primary object of the t invention is to select strains that can y produce histamine in various locations, including the GI tract, GU tract, oral cavity, in the lungs and airways, on the skin etc, of the mammalian body.
It is a further object of the invention to provide products containing said strains.
It is a further object of the invention to combine the administration of ia with administration of ine, or histidine containing foods or compositions, to ensure local generation of histamine.
The present invention thus relates to a new method for selecting lactic acid bacterial cells which are useful as probiotics and in therapy. This new method involves the screening and selection for strains of lactic acid bacteria which have an active histidine operon and are capable of producing histamine. Surprisingly, the lactic acid bacterial strains selected by this method are useful as tics and in therapy, in particular in producing anti-inflammatory effects, by way of the local production of histamine. These effects of the ia are surprising, as discussed elsewhere herein, previously the presence in foodstuffs of bacteria producing histamine was actively avoided due to the recognised health risk, for example potential toxic effects, Thus, the administration to a mammal of a lactic acid bacteria capable of local tion of histamine, or indeed the screening and selection of lactic acid bacteria 3 2012/064351 for such capability of local production of histamine based on the presence of an active histidine operon and an y to produce histamine is r—intuitive to this teaching.
Indeed, probiotics have never before been reported to produce histamine.
Thus, at its broadest, the present invention provides a method of selecting a lactic acid bacterial strain for use in the local production of histamine in a mammal, wherein said method comprises screening bacteria for the presence of an active histidine operon and selecting a strain which has an active histidine operon and is e of producing histamine.
The histidine operon comprises three genes (the histidine/histamine antiporter, the histidine decarboxylase pyruvoyl type A (Hch), and histidine decarboxylase pyruvoyl type B (Hch)). It is believed that the activity of each of these genes is important for the present invention. Thus, in the screening methods of the invention, candidate bacteria are assessed for the presence of all three genes and strains positive for all three genes are ed. Any appropriate method can be used for the detection of the presence of all three genes, for e genetic methods such as PCR can be used. The production of good levels of histamine can also be an tor of the presence of all three genes and the presence of an active histidine operon. Thus, the ion method of the invention also involves the step of selecting a strain which is e of producing histamine. Strains with high tion levels of histamine are preferred. Thus, in preferred ments a strain is selected for its ability to produce histamine at a level of greater than 200, preferably greater than 250 or more 2O preferably greater than 300 pg/ml, for example a level of greater than 350, 400, 450, or 500 pg/ml. Such values generally refer to values of histamine measured in the supernatant of strains in culture.
Appropriate methods of ing levels of histamine production would be well known to a person skilled in the art. The method of mass spectrometry, more specifically triple quadrupole mass spectrometry, is exemplified herein and is preferred. r, equally ELISAs or immunoassays can be used to evaluate and quantify histamine production.
Thus, in some embodiments of the ion, the selection method will involve the step of detecting the amount or level of ine produced by a candidate strain. Because of the downstream uses of the strains which are selected by the methods of the invention, after histamine ing strains are selected or isolated, other embodiments will involve the further steps of culturing or propagating such strains, or possibly storing such strains for future uses.
Such further steps (and indeed the selection steps of the methods of the invention) will generally need to be carried out in an appropriate culture medium which supports histamine production. red e media will contain an appropriate carbon source which will support the production of histamine by said strain. In particularly preferred embodiments, the media will comprise glucose as a carbon source and preferably will not contain e, or at least will only comprise sucrose at such a level which will not significantly mise histamine production by the strain. Histidine or a histidine analog can also be provided, optionally together with s of other amino acids.
In preferred embodiments said strain is a strain of Lactobacillus i.
Once an appropriate strain has been selected using the method of the present invention it can then be used for the local tion of histamine in a mammal. Said strains thus also have to be capable of local production of histamine in a mammal.
Thus, a further aspect of the t invention provides a product comprising cells of a lactic acid bacterial strain obtainable by the selection method of the ion, wherein said lactic acid bacterial strain has an active histidine operon and is capable of producing histamine, for use in the local production of histamine in a mammal. As will be outlined elsewhere herein, preferred uses are in the treatment and/or prophylaxis of inflammatory conditions, or in the treatment and/or prophylaxis of conditions or diseases which will benefit from local histamine production. For example, such local production of histamine can result in an anti-inflammatory effect.
Alternative embodiments of the invention provide a lactic acid bacterial strain which is e of producing histamine for use in the local production of histamine in a mammal, n said lactic acid bacterial strain has an active histidine operon. Preferred features of this strain and its uses are described elsewhere herein.
Methods of treatment or methods for the local tion of histamine in a mammal, are also provided, said methods comprising the administration of a product comprising cells of a lactic acid bacterial strain obtainable by the selection method of the invention, or the administration of a lactic acid bacterial strain wherein said lactic acid bacterial strain has an active histidine operon and is capable of ing histamine, to said mammal in an amount effective to enable local production of histamine in said mammal. Preferred features of the strain and its therapeutic uses are described ere .
Also provided by the present invention is the use of a product comprising cells of a lactic acid bacterial strain obtainable by the selection method of the invention, n said lactic acid bacterial strain has an active histidine operon and is capable of producing histamine, in the manufacture of a composition or medicament for use in the local production of histamine in a mammal. Alternative embodiments provide the use of a lactic acid bacterial 5 2012/064351 strain, wherein said lactic acid bacterial strain has an active histidine operon and is capable of producing histamine, in the manufacture of a composition or medicament for use in the local production of histamine in a mammal. Preferred features of the strain and its therapeutic uses are described elsewhere herein.
BRIEF DESCRIPTION OF THE DRAWINGS Figure l — Quantification of histamine in HILIC—HPLC fractions. Tiiple quadrupole mass spectrometry was used to fy histamine present in a select range of HILIC-HPLC fractions. TNF—inhibitory fractions had the highest amounts of ine out of all the fractions ed.
Figure 2 — ed histamine and ine from L. reuteri 6475 inhibit TNF production via the histamine H2 receptor. A. Purified histamine significantly ted TNF production, an effect that is blocked by specific H2 receptor antagonists in a dose-dependent . Conditioned media (or supernatant) containing secreted factors from strain 6475 (including histamine) significantly inhibited TNF production, an effect that is partially blocked by specific H2 receptor antagonists. N=3, *p value < 0.05 compared to media l, “p value < 0.05 compared to histamine, Wp value < 0.05 compared to ATCC 6475 conditioned media (CM) B. The cell pellet wash from strain 6475 containing ine suppressed TNF production, an effect that was partially blocked by specific H2 receptor antagonists. Fraction B3, which contains relatively pure histamine, inhibited TNF production, an effect that was completely blocked by specific H2 receptor antagonists. N23, *p value < 0.05 compared to media control, Wp value <0.05 compared to ATCC 6475 cell pellet wash (CP), mp value < 0.05 compared to on B3.
Figure 3 — The histidine operon is important for the TNF-inhibitory phenotype of L. reuzeri 6475. A. The ine operon consists of three genes, the histidine/histamine antiporter, hch, and thB. B. Mutation in any one gene in the histidine operon results in a partial loss of TNF suppression by L. reuteri 6475. N29, *p value < 0.05 compared to media control, *p value < 0.05 compared to ATCC PTA 6475.
Figure 4 — L. reuteri 6475 significantly reduced weight loss induced by TNBS challenge, the figure represents data from two independent experiments, * , “1" p<0.01, W" p<0.001.
Figure 5 - L. reuteri 6475 significantly diminished macroscopic colon damage induced by TNBS challenge, the figure represents data from two independent experiments, ""p<0.05, **“"p<0.001.
Figure 6 - L. remeri 6475 icantly reduced SAA tration induced by TNBS challenge, the figure represents data from two independent experiments, p<0.05, W* p<0.001.
Figure 7 — thA mutant yielded diminished ability to attenuate colitis, the figure represents data from two independent experiments, p<0.05, *" p<0.01.
Figure 8 — thA mutant yielded diminished ability to attenuate colitis, the figure represents data from two independent experiments, * p<0.0l, W p<0.00l.
DETAILED DESCRIPTION OF THE INVENTION AND PREFERRED EMBODIMENTS F The inventors herein have found that a selected group of lactobacilli, including certain strains of Lactobacillus remeri locally produces histamine under specific growth conditions, and that such produced histamine will benefit the host by for example reducing inflammation, reducing certain cancers etc.
Histamine Histamine is an organic nitrogen compound involved in several health associated processes of a mammal, ing local immune responses as well as regulating physiological function in the gut and acting as a neurotransmitter. As part of an immune response to foreign pathogens, histamine is produced by basophils and mast cells. Histamine can be derived from the decarboxylation of the amino acid histidine, a reaction catalyzed by the enzyme L- histidine decarboxylase.
Bacteria are capable of producing histamine using ine decarboxylase enzymes unrelated to those found in eukaryotes. Up to now, such production of histamine by certain bacterial strains has been seen as a health risk rather than a possible benefit for humans. For example, oid poisoning, a form of non—infectious foodborne e, is due to histamine production by bacteria in spoiled food, particularly fish. Fermented foods and beverages naturally contain small quantities of histamine due to a similar conversion performed by ting bacteria or . Delivery of n controlled amounts of ine from selected ia can, surprisingly, give beneficial effects rather than detrimental effects as might be expected from the above ned studies.
Histamine receptors are a class of G protein—coupled receptors with histamine as their endogenous ligand. There are four known ine receptors; leeceptor (HIR), tor (HZR), ngeceptor (H3R) and H4receptor (H4R).
Vannier et a1. (Histamine sses Gene Expression and Synthesis of Tumor Necrosis Factor a via Histamine H2 Receptors; J Exp Med. 1991 July l;l74(l):281—4) showed that LIPS—induced synthesis of TNF-a in peripheral blood mononuclear cells was ssed by histamine and they r suggest that histamine e from mast cells may limit the extent of inflammatory and immune reactions by suppressing local ne sis in Hz receptor~bearing cells.
The anti-inflammatory activity of histamine has previously been disclosed by Wang et al. (Histamine Aizragorzizes Tumor Necrosis Factor (TNF) Signaling by Stimulating TNF Receptor Sheddingfrom the Cell e and Golgi Storage Pool; J. Biol. Chem. 278(24): 21751-21760), showing that histamine causes transient loss of surface TNFRl, increased TNFRI shedding, and mobilization of TNFRl molecules from the Golgi in cultured human endothelial cells. Histamine injection into human skin engrafted on immunodeficient mice caused shedding of TNFRl and diminished TNF-mediated ion of endothelial adhesion molecules.
Vannier et al. and Wang et al. did not n anything about using histamine producing bacterial strains as probiotics nor how to select the strains based on their histamine- producing abilities in order to assure certain health benefits for the host, such as anti— atory effects.
Ceplene, a pharmaceutical-grade form of histamine dihydrochloride, is used for the prevention of relapse in patients sed with acute d leukemia (AML). Ceplene is administered in conjunction with low doses of the immune—activating cytokine interleukin—2 (IL—2) in the post—remission phase of AML, i.e. when patients have completed the initial chemotherapy. Studies have shown that Ceplene/IL—Z can induce immune—mediated killing of leukemic cells. The ent, subcutaneous injections, is given in 3-week cycles by the patients at home for 18 months. The side effects of Ceplene include ent flush and headache. It would be advantageous for patients to receive locally ed histamine, when needed, instead of subcutaneous injections; this delivery strategy may be achieved by administering bacteiial-derived histamine to the patient using the strains selected according to this invention.
It is previously known that gram—negative bacteria form histamine for example in raw fish and meat following temperature abuse and that gram—positive bacteria cause histamine spoilage of fermented foods such as cheese, sausage, miso, soy sauce, beer and wine. The identification of ine—producing bacteria in foods has been difficult.
Also lociobacillus reuzeri has previously been associated with histamine production, Casas et al. (Validation of the Probiotic Concept: Lactobacillus reuteri Confers Broad— 8 2012/064351 spectrum Protection. against Disease in Humans and Animals; 2000, ISSN 0891-060X) reports that two s of L. reuteri in the hands of Straub et.al. (Z Lebensm Unters Forsch(1995) 201: 79- 82) has been shown to decarboxylate idine to form histamine and the authors warn against using such strains for the fermentation of food and as probiotics.
Trip et al. (Hch, a novel enzyme catalyzing maturation voyl-dependent histidine decarboxylase; Molecular Microbiology (2011) 79(4), 861-871) referring to three types of genetic organization of histidine decarboxylation loci among histamine-producing Gram—positive bacteria. The largest group is found in the lactic acid bacteria including L. hilgardii 0006, L. buchneri B30], L. reuteri F275 and T. halophilns. Lactobacillus hilgardii 0006 has been shown to e ine, in a study performed by Lucas et a1. (Histamine— Prodncing Pathway Encoded on an Unstable Plasmid in Lactobacillus hilgardii 0006; APPLIED AND ENVIRONMENTAL MICROBIOLOGY, Mar. 2005, Vol. 71, No. 3, p. 1417— 1424), they further say that histamine is a contaminant that appears in several ts during growth of undesirable bacteiia. Lucas et al. have performed a screening of a collection of wine lactic acid bacteria to identify the genes ed in the histamine—producing pathway of a gram-positive bacterium of wine.
A histamine producing strain of Lactobacillns bnchneri has been isolated from Swiss cheese that had been implicated in an outbreak of histamine poisoning (Summer et al.
Isolation ofhistamine-producing acillns bnchnerifrom Swiss cheese implicated in a food poisoning outbreak. ; d and Environmental Microbiology (1985), Vol. 50, Issue 4, p. 10944096).
-Enriquez et al. (Sequencing and Transcriptional Analysis of the Streptococcus philes Histamine Biosynthesis Gene Cluster: Factors That Afi‘ect Difl'erential hch Expression; APPLIED AND ENVIRONMENTAL MICROBIOLOGY, Sept. 2010, Vol 76, No. 18, p. 238) describe histamine—producing strains of Streptococcus thermophiles, a thermophilic starter used for the production of yogurt and certain cheese varieties. They further indicate that the presence of strains with the capacity to decarboxylate histidine could result in products containing histamine produced during manufacture or during storage before consumption and that this underlies the importance of using only histamine-negative strains in the manufacture of fermented dairy products.
Even if it has been previously known that some Lactobacillns can produce histamine it has certainly not been known that the ability to produce histamine is a key factor to assure certain health benefits for it’s host, for example anti—inflammatory properties of certain Lactobacillns strains. r is it known or obvious from the prior art that this can be used for screening and selection of certain probiotic strains of Lactobacillus.
Mast cells A mast cell (also known as mastocyte and labrocyte) is a resident cell of several types of tissues and contains many granules rich in histamine and heparin. Although best known for their role in allergy and anaphylaxis, mast cells play an important protective role as well, being for e intimately involved in wound healing and defense against pathogens.
Mast cells are present in most tissues characteristically surrounding blood vessels and nerves, and are especially prominent near the boundaries between the outside world and the internal milieu, such as the skin, mucosa of the lungs and digestive tract, as well as in the mouth, conjunctiva and nose.
In allergic reactions, mast cells remain inactive until an allergen binds to IgE y in association with the cell. Other membrane activation events can either prime mast cells for subsequent ulation or can act in synergy with FceRI signal transduction. Histamine from such granulation dilates post capillary venules, activates the endothelium, and increases blood vessel permeability. Histamine release leads to local edema (swelling), warmth, redness, and the attraction of other inflammatory cells to the site of release. It also irritates nerve endings (leading to itching or pain). Cutaneous signs of histamine release are the "ii are and wheal"-reacti0n. The bump and redness immediately following a to bite are a good example of this reaction, which occurs seconds after challenge of the mast cell by an allergen.
The other logic activities of mast cells are much less well-understood. Several lines of evidence suggest that mast cells may have a fairly fundamental role in innate immunity — they are e of ating a vast array of important cytokines and other inflammatory mediators such as TNFOL, they s multiple "pattern recognition receptors" thought to be involved in izing broad classes of pathogens, and mice without mast cells seem to be much more tible to a variety of infections.
Considering the toxicity of bacterial histamine in foods and the fact that it is recommended to avoid histamine-producing strains in fermented products (see further examples in Calles—Enriquez et al. cing and Transcriptional Analysis ofthe Streptococcus thermophiles Histamine Biosynthesis Gene Cluster: Factors That Aflecz Diflererztial hch Expression; APPLIED AND ENVIRONMENTAL IOLOGY, Sept. 2010, Vol. 76, No. 18, p. 6231-6238) it can not considered to be obvious to use certain selected acillus s for local production of histamine in treatment and/or prophylaxis of various diseases.
The relationship between a host and its microbes is complex, as it also is for certain of a mammalian’s own cell types, such as mast cells. This host—microbe relationship has been developing over many years of co-evolution, this includes the es production of various metabolites that can benefit the host nutritionally, immunologically etc, act as whole or part antagonists, t, de-sensitization etc. of specific receptors or other processes. There is therefore also a need to understand such specific interactions between microbes and man related to a specific disease or other situations influencing the health of the host so that the most appropriate probiotic strains can be selected and used to counteract such developments.
The ors herein have found that a selected group of lactobacilli, including certain strains of Lactobacillus reuteri y produces histamine under specific growth conditions.
And that such locally produced histamine, contrary to earlier belief, will benefit the host in multiple ways including reduction of inflammation, reducing certain cancers etc.
An other object of the invention is to provide products containing said strains together with a specific carbon , in order to have a synbiotic product.
Other objects and advantages will be more fully apparent from the following disclosure and appended claims.
Administering the lactic acid bacterial strains, selected accordingly to the present method, to a mammal will result in locally produced histamine that could be beneficial for several reasons.
A primary object of the t invention is to provide a method of ing lactic acid bacterial s assuring good anti-inflammatory effect. These strains could preferably be used for treatment and/or prophylaxis of inflammatory conditions, since the histidine operon and the production of histamine is essential for the anti—inflammatory capacity of certain lactic acid ium. ably the strains may be used for ent and/or prophylaxis of atory ses in the GI tract, GU tract, oral cavity, in the lungs and airways, on the skin etc, of the mammalian body, including but not limited to colitis, IBD, IBS, diverticulosis, gingivitis, vaginitis etc. It is previously known that histamine via the H2 receptor can reduce the gene expression of TNF-alpha. Further mast cells are capable of elaborating a vast array of important cytokines and other inflammatory ors such as TNF—alpha. However it is not usly known that the histidine operon, and local histamine production of such selected strains could be beneficial to the host and is for example a key factor in selected L. reuteri strains anti-inflammatory capacity. Neither is it previously known to use L. reuteri selected according to the present method in treatments requiring histamine.
Preferred products and strains for the treatment and/or prophylaxis of inflammatory conditions are acillus i, in particular lactobacillus reuteri 6475 (ATCC PTA 6475). In other embodiments of the invention the strain used is not lactobacillus reuteri 6475 (ATCC PTA 6475).
The therapeutic uses of the strains, products and compositions of the invention as defined herein generally result in the reduction or alleviation of the relevant disease or symptoms of disease, for e can result in a significant ion in inflammation levels in the mammal. For example, locally produced histamine may be activating H2 receptors on intestinal epithelial cells as well as immune cells to suppress host mucosal immunity, 6. g. via the inhibition of pro-inflammatory nes. Thus, the present invention allows for the conversion of a dietary component dine) to ine at the site of activity and local modulation of the host immune response (e.g. in the gut). It can be seen that such local production of histamine provided by the present invention can provide real advantages over for example oral ingestion or other forms of administration of histamine, ally given the fact that such oral ingestion would not be advocated due to the recognised toxic s and health risks.
In particular where inflammatory diseases of the intestine are concerned, the therapeutic uses of the s, products and compositions of the invention can result in significant reduction in ulceration and intestinal damage (e.g. colon damage) measured for example by a standard method such as a Wallace score, a significant reduction in weight loss or a significant reduction in inflammation of the intestine, e.g. the colon.
Such reduction or alleviation of disease or symptoms thereof can be measured by riate assay. Preferably the reduction or alleviation of disease or symptoms is statistically significant, ably with a probability value of <0.05. Such reduction or alleviation of disease or symptoms are generally determined compared to an appropriate control individual or population. for example a healthy mammal or an untreated or placebo treated mammal.
An appropriate mode of administration and formulation of the strains, etc., is chosen depending on the site where local production of histamine is desired. A preferred mode of administration is oral, however, equally for some treatments topical or some other form of local stration to the skin, rectum, vagina or gums will be riate, or intravenous or intramuscular ion will be appropriate.
Although the es herein demonstrate the use of strains of the invention and appropriate doses thereof to treat colitis, it will be appreciated that this is only one example of the inflammatory conditions which can be d in accordance with the present invention and that appropriate doses of the strains, products and compositions of the invention as defined herein can be chosen depending on the e to be treated, the mode of administration and the formulation concerned.
Dietary mixtures comprising histidine may be used to ensure the presence of histidine and thereby increase the efficacy of the bacteria. Histidine may be stered alone or together with the bacteria.
One possibility to ensure the bacteria’s supply of histidine is to eat histidine rich food, including but not limited to soy protein, cheese, egg, chicken and pork.
The histidine operon in bacteria has been shown to improve the growth capacity under conditions of low pH or energy source limitation s—Enriquez et al.) but the histidine operon has not been associated with anti-inflammatory es of certain L. reuteri strains. it is another object of the t invention to use the strains selected according to the present method in cancer therapy. Histamine in combination with IL—2 has been used for treatment of AML. Using the strains of the t invention will result in locally produced histamine that in combination with IL«2 could be used for treatment of AML.
Another object of the present invention is to use the selected strains in order to reduce food allergy, other allergic reactions or other autoimmune diseases. Systemic increases in histamine are as previously known a consequence of allergy by the granulation of mast cells.
When administering strains selected according to this invention to a recipient, the locally produced histamine will lead to a desensitization effect that will reduce y or other autoimmune diseases.
It is also an object of the present invention to use the histamine producing lactic acid bacteria strains to reduce the risk of er’s diarrhea. Patients treated with histamine blockers have an increased risk of g traveler’s diarrhea, this increased risk could be lized by administrating the lactic acid bacteria selected according to the present method.
Yet another object of the t invention is to use the selected strains in treatment of MS. Histamine has been proposed to be an important molecule for developing new treatments for MS and the strains ed accordingly to the present invention will provide the patient with ine.
Yet another object of the present invention is to use histamine producing ia as a nti—inflammatory treatment using available histidine and histidine analogs in the skin.
Since histidine is a substrate for urocanic acid which the skin produces by UV irradiation and the urocanic acid has anti—inflammatory properties on the skin.
W0 2013/01 1137 13 It is another object to use such selected strains to inhibit activation of ERKl/2 Another object of the invention is to t TNF alpha Another object of the invention is to reduce inflammation, y or systemically Another object of the invention is to enhance synaptic vesicle exocytosis by inhibiting ERKl/2 Another object of the invention is to promote human embryonic stem cell enewal by inhibiting ERKl/2 Another object of the invention is to induce macrophage ABCAl sion and cholesterol efflux by inhibiting ERKl/Z Another object of the invention is to reduce cardiac hypertrophy and heart failure by inhibiting ERK1/2 Another object of the invention is to reduce the proliferation of certain cancers including Leukemia (for example AML) or malignant melanoma. Thus, such cancers are preferred diseases which could be treated using the strains, products and compositions of the t invention.
Another object of the invention is to use selected strains to produce histamine under certain conditions as a neurotransmitter for example in GU tract interactions with the CNS, and also neural signaling in local pain. This role as a neurotransmitter can be extended to effects on intestinal motility (to treat constipation or diarrhea) and to pain signaling in the gut. 2O r object of the invention is to use selected strains for influencing the gut—brain axis as the selected LAB will produce histamine and affect al pain perception and signaling in enteric s system. Thus, it can be seen that the present invention can be used for the ent and/or prophylaxis of any disease which will benefit from local histamine production or the treatment and/or prophylaxis of any disease which can be treated with the local administration of histamine.
It is a further object of the invention to provide products containing said strains.
It is a further object of the invention to provide ts containing said strains together with a specific carbon source, in order to have a synbiotic product, which will through specific stimulation of the histamine—producing strain, e the effects.
It is a r object of the invention to provide products comprising the said strains together with ine, including histidine analogs or histidine containing products or composition. Preferably such a mixture is administered orally in a protective capsule for e of the content in the lower GI tract to ensure survival of both the histidine and the bacteria at the site of action.
W0 2013/011137 14 It is a further object of the invention to combine administration of said strains with a histidine rich diet.
A yet further aspect of the invention provides a product for the therapeutic uses as defined elsewhere , wherein said use further ses the administration of at least one further therapeutic or nutritional agent. In such embodiments, the further therapeutic agent can be any r agent which is useful in the treatment of the disease in question, for example is a r nflammatory agent or an immunotherapeutic agent such as for example a chemokine or cytokine (e. g. IL—2).
In preferred embodiments, said further agent comprises histidine or a histidine analog, an appropriate carbon source which supports the tion of ine by the bacterial strain, or a combination thereof.
Said further agents can be administered together with the strains of the ion or can be administered separately. In addition, said further agents can be administered at the same time as the strains of the ion or at different time points. Suitable administration regimes and timings can readily be determined by the skilled person depending on the further agent in question.
The present invention also provides a composition comprising: (i) a lactic acid bacteria] strain obtainable by the selection method of the invention (or a lactic acid bacterial strain capable of producing histamine as otherwise defined ), wherein said 2O lactic acid bacterial strain has an active histidine operon and is capable of producing histamine; and (ii) at least one additional component selected from the group consisting of an appropriate carbon source which supports the production of histamine by said strain, a source of histidine or histidine analog, and a combination thereof.
In ts, compositions and uses of the invention as described , preferably said histidine or histidine analog is in the form of a histidine or histidine analog containing foodstuff or food supplement, or said carbon source comprises e. Preferably said carbon source will not se sucrose, or at least will only comprise sucrose at such a level which will not significantly compromise histamine production by the strain. Optionally, sources of other amino acids can also be provided.
In alternative embodiments, the strains as defined in part (i) can be combined with a fuither component which is useful in the treatment of the disease in question, i.e. a further therapeutic agent, for example a further nflammatory agent or an immunotherapeutic agent such as for example a chemokine or cytokine (e.g. IL—2). wo 11137 15 Lacrobacillus reuterz’ is a heterofermentative lactic acid bacterial species that naturally inhabits the gut of humans and animals. Specific probiotic L. reureri strains potently suppress human TNFOL production while other probiotic L. reuteri strains enhance human TNFOt production.
The invention herein is made possible by mechanistic s of probiotic L. reuteri strain 6475 and other strains which has demonstrated their effect upon activated human d cells. L. i metabolites were isolated using HILIC—HPLC, and histamine was identified by NMR spectroscopy and mass spectrometry. Quantification of histamine by triple quadrupole MS revealed that L. reuteri strain 6475 produces relatively high concentrations of histamine when grown in a glucose-based minimal media. Previous transcriptomics studies had suggested that two genes in the L. reuteri histidine operon may play a role in TNF inhibition by strain 6475. ed mutagenesis of these genes revealed that each gene in the histidine operon, the histidine/histamine antiporter, Hch, and Hch, are important for the TNF—inhibitory phenotype of strain 6475. Mechanistic studies demonstrated that histamine is inhibiting TNF via ing through the H2 but not H1 receptor. Signaling through the H2 receptor increases intracellular CAMP, which activates PKA. PKA activity is necessary for TNF ssion by histamine. Histamine blocks tion of the K MAPK signaling pathway.
Histamine is better known for its pro—inflammatory effects in allergy and anaphylaxis, but l studies have demonstrated anti-inflammatory functions of histamine. In vitro studies have shown that histamine can inhibit production of pro—inflammatory cytokines, IL— 1. IL— 12, and TNF from LPS—stimulated human monocytes and macrophages and this effect is reversed by H2 receptor antagonists. Additionally, ine can ate production of the anti~inflammatory cytokine, IL—lO, via the H3 receptor. Signalling through the H2 receptor results in decreased expression of the CD14 receptor, a receptor involved in LPS recognition, on the surface of human monocytes. The TNF receptor is also affected by histamine.
Signalling through the H1 receptor induces shedding of both the TNFRl and the TNFR2. In vivo studies have also revealed an anti-inflammatory role for histamine. Treatment with 3O dimaprit, a specific H2 receptor t, d plasma TNF levels in mouse models of endotoxin shock (LPS challenge) and tis (LPS plus galactosamine challenge).
Histamine was protective in an LPS-induced liver injury mouse model, and these effects were attenuated in an H2 or knock—out mouse. In the gut, histamine may help protect against W0 2013/01 1137 16 bacterial infection. Signalling through the H2 receptor in Peyer’s patches helps prevent infection by Yersinia enterocalitica.
The effect of histamine can be ined by the expression of histamine receptors on the target cell. In T—cells. the effect of histamine is dependent on which histamine receptor is activated.
By signalling through the H1 receptor, histamine enhances THl~type responses but sses both THl and THZ responses via the H2 receptor. A study was peifonned g at histamine or expression in the human gastrointestinal tract. Many of the cell types examined expressed multiple histamine receptors. For example, immune cells, including macrophages, highly expressed the H1 and H2 receptor and demonstrated low expression of the H4 receptor. Increased mast cells and histamine have been implicated in the al hypersensitivity associated with IBS. The increased number and activity of mast cells near colonic mucosal innervation may result in ened abdominal pain perception. A study with ketotifen, a mast cell stabilizing agent, demonstrated an increased pain threshold in patients with IBS, decreased IBS symptoms, but no change in the number or activity (determined by ine and tryptase release) of mast cells in rectal biopsy tissue. The effects of ketotifen in improving IBS may not be the result of stabilizing mast cells, but could be attributed to its other role as an H1 receptor antagonist. If tion of the H1 receptor is associated with a pro—inflammatory response, blocking its ty with ketotifen can allow histamine produced either by mast cells or the gut iota, such as L. reuteri, to signaling through the H2 receptor only. As we have demonstrated, signaling via the H2 receptor can suppress TNF tion and cause an nflammatory effect. This ketotifen mechanism can be used for new therapies ing H1 receptor antagonists with general probiotic effect of a L. i strain.
Further changing the carbon source of the growth media from glucose to sucrose is sufficient to suppress the TNF—inhibitory phenotype of a selected strain, for example L. reuteri strain 6475. In addition, significant down—regulation of all three genes in the ine operon was observed with the sucrose growth condition.
The identification of histamine as an anti-inflammatory compound produced by selected probiotic Lacrobacillus strains will help determine therapeutic applications for such strains. Mechanistic studies linked the activation of the H2 receptor on THP—1 cells with histamine and the suppression of ERK activation. ERK activation is involved in many cellular ons besides TNF production. ERK activation is involved in proliferation, tumorigenesis, differentiation, and cell survival. The results suggest a role for ed strains such as L. reuteri 6475 in protecting against cancer by suppressing inflammation, cell proliferation, and apoptosis via inhibition of ERK activation. In addition, histamine is a known neurotransmitter. Production ofhistamine by ed strains can influence signalling in the enteric nervous system, impacting pain perception and gut motility. To ensure the production of histamine at the site of action it may be advantageously to provide the bacteria with histidine. Histidine may be administered together with the bacteria or alone, diets rich in histidine may increase the histamine production as well.
The present invention provides n strains of lactic acid bacteria and a method of selecting such strains and products sing such strains. The bacteria are selected using a screen for the ine operon, singly the ce of an active histidine operon has been shown to be essential for various beneficial effects such as the immunomodulatory properties of lactic acid bacterial s.
Other s and advantages of the present invention will become obvious to the reader and it is ed that these objects and advantages are within the scope of the present invention. The foregoing objects are to be read ctively with the object of at least providing the public with a useful alternative.
The invention will be further described with reference to the following non-limiting Examples: EXAMPLES Table 1. Bacterial strains used in this study Bacterial Strains Description Source L. reuteri ATCC PTA 6475 Isolate from Finnish mother’s milk BioGaia AB (Raleigh, L. reuteri ATCC PTA ional mutant This study 6475::JP577 L. reuteri ATCC PTA insertional mutant This study 6475:1229 L. reuteri ATCC PTA insertional mutant This study 6475:1230 L. reuteri ATCC PTA insertional mutant This study 6475:: 1231 Table 2. Transcriptomic analysis of the histidine operon in L. reuteri strain 6475 mutants Histidine decarboxylase, Histidine/Histamine Hch gene pyruvoyl type A (Hch) Antiporter -—_—— PocR/647S — - 2.4 0.10 1.3 0.66 *lnsertion mutants that lose the ability to inhibit TNF production compared to the wild—type strain 6475. CFAS: cyclopropane fatty acid synthase, THFSl: tetrahydrofolate synthase 1, THFS2: tetrahydrofolate synthase 2.
§Wild-type 6475 grown in LDMIIIS compared to wild—type 6475 grown in LDMIIIG. Wild— type 6475 grown in LDMHIS loses the ability to inhibit TNF production.
EXAMPLE 1: Production of histamine by selected lactobacillus Bacterial strains and e conditions All bacterial s used in this study are described in Table l. Lactobaciilus remeri ATCC PTA 6475 is an isolate from Finnish mother’s milk (available from ATCC, Manassas, VA, USA). L. remeri strains ATCC PTA 6475, ATCC 6475 JP577, ATCC 6475 1229, ATCC 6475 1230, and ATCC 6475 1231 will be ed to as strains 6475, JP577, 1229, 1230, and 1231, respectively, throughout this disclosure. L. reuteri s were cultured under anaerobic ions for 16—18 h in deMan, Rogosa, Sharpe media (Difco, Franklin Lakes, NJ), and inoculated into 2 L of a efined media, LDMIll (OD600 adjusted to 0.1), which has been described previously. The carbon source was either glucose, LDMlllG, or sucrose, LDMllIS.
The culture was grown for 24 h at 379C in an anaerobic workstation (MACS MG—SOO, Microbiology International, Frederick, MD) ed with a mixture of 10% C02, 10% H2, and 80% N2. Samples were taken at different times to follow the growth by measuring OD600.
At stationary phase (24 h), the cells were pelleted from the 2 L e (4000 x g, 10 min).
Cell pellets and bacteria cell~free supernatants were stored at —209C before r processing for HPLC separation and testing in a TNF inhibition bioassay.
Cell line and reagents W0 2013/01 1137 19 In vitro experiments were performed with THP—l cells (human toid cell line, ATCC, Manassas, VA) ined in RPMI (ATCC) and heat—inactivated fetal bovine serum (Invitrogen, Carlsbad, CA) at 379C, 5% C02. MEKl/Z, phospho-MEKI/Z, ERKl/Z, and phospho-ERKl/2 antibodies and MEK inhibitor U0126 were received from Cell Signaling Technology (Danvers, MA), and the B-Actin antibody was received from Abcam idge, MA). All other reagents were received from Sigma (St. Louis, MO) unless otheiwise stated.
HILIC—HPLC separation of cell wall associatedfactors Cell pellets (7 g) from strain 6475 grown in either LDMIIIG or LDMIIIS were washed with 30 mL ice cold 50% acetonitrile/O.l% trifluoroacetic acid (TFA). The cell suspension was centrifuged for 10 min, 4000 x g at 49C. Supernatants were filtered through polyvinylidene fluoride (PVDF) membrane filters (0.45pm pore size, Millipore, d, MA), lyophilized, and resuspended in 10 mL 0.1% formic acid. The resuspended sample was size fractionated with Amicon Ultra— 15 centrifugal filter units using ultracel-S membrane (Millipore, Bedford, MA). The e (9 mL) was dried down to l mL with a speed vacuum, and 0.75 mL was used for HILIC—HPLC. The sample was dissolved with 100% acetonitrile before running on a PolyLC Hydroxyethyl column with a gradient of 100-0% acetonitrile, 01% formic acid. The sample was tun for 25 min and 25 fractions (Al-Cl) were collected at mUmin/tube. Three milliliters from each fraction was lyophilized, ended in 3 mL 0.1% acetic acid, and lyophilized again for testing in a TNF inhibition bioassay TNF tion bioassay and TNF ELISA Bacterial tants (10 mL) from a 24 h LDMIH culture were filter—sterilized using PVDF membrane filters (0.22pm pore size, ore) and size fractionated as described above. One milliliter of the <3 kDa filtrate was speed vacuum dried and resuspended in RPMI media. These sed supernatant samples are termed conditioned media. All supernatants were normalized by volume to an OD600 = 1.0. Lyophilized fractions from the HILIC-HPLC separation were resuspended in 400 uL l0 mg/mL ammonium bicarbonate, speed vacuum dried, and resuspended in 400 uL RPMI media. Conditioned media and cell pellet wash fractions were tested for their ability to modulate TNF production in monocytoid cells. In brief, THP—1 cells (approximately 57th4 cells) were stimulated to produce TNF by the addition of 100 ng/mL Pam3Cys-SKKKK x 3 HCl (EMC Microcollections, gen, Germany) as usly described. tors ~ H2 receptor antagonists, ranitidine and cimetidine (104 ~ 1043 M), H1 recePtor antagonist, indomethacin (10-5 — 10‘6 M), MEK wo 2013/011137 20 inhibitor, U0126 (10 pM), and PKA inhibitor, H89 (N—[2-(p—bromocinnamy1amino)ethyl]—5— isoquinolinesulfonamide dihydrochlon'de) (10‘5 M) — were added to the THP—1 cells followed by L. reuteri conditioned media or cell pellet wash fractions (5% v/v), histamine (10'5 M), or dibutyryl CAMP (10'3 — 10'7 M). Plates were incubated at 37°C and 5% co2 for 3.5 h. THP-1 cells were ed (3000 x g, 5 min, 4°C), and quantitative ELISAs were used to determine TNF quantities in THP—l cell supernatants according to the manufacturer’s instmctions (R&D Systems, Minneapolis, MN).
L reateri 6475 TNF~inhibitory compomzd(s) were isolated using Hydrophilic ction Liquid Chromatography - High Performance Liquid Chromatography (HILIC—HPLC) Bacterial cell pellets were washed to remove compounds loosely associated with the cell surface. Components of the cell pellet wash were separated based on hydrophobicity using HILIC—HPLC, and the resulting 25 fractions were tested for retention of the TNF- inhibitory compound. L. i 6475 grown in a minimal media with glucose as the sole carbon source produces TNF-inhibitory factors that were retained in 3 separate HPLC fractions (B3, B5 and B6, data not shown). L. reareri 6475 grown with sucrose as the sole carbon sources loses the TNF-inhibitory phenotype and served as the negative control. None of the l-llLlC-HPLC ons from the 6475 sucrose cell pellet wash demonstrated significant TNF tion (data not .
Histamine was identified in a TNF-inhibitory HILIC—HPLCfraction by NMR spectroscopy and mass spectrometry TNF—inhibitory HPLC fraction B3 was analyzed by 1H NMR and compared to the neighboring non-TNF inhibitory fraction B4. A unique series of peaks with a chemical shift between 7.0—7.5 ppm, which is characteristic of an aromatic compound, were observed in fraction B3 but not fraction B4 (data not . This aromatic compound cluster was r analyzed with heteronuclear single quantum coherence (HSQC) 2—dimensional (2D) NMR in order to identify its components. The aromatic compounds consisted of tryptophan, phenylalanine, histamine, and one compound that was unidentifiable. Tryptophan and phenylalanine are components of the bacterial growth media while histamine is not. These results were confirmed using an additional 2D NMR , total correlation spectroscopy (TOCSY). Histamine is a ic amine that is produced from histidine via the histidine decarboxylase by some fermentative bacteria including lactobacilli. Histamine was also fied in fraction B3 using electrospray f—flight mass spectrometry (ESI TOF MS).
Histamine is not covalently modified based on its ntation pattern in MS/MS analysis.
Analysis of the corresponding B3 fraction of L. reuteri 6475 grown in a sucrose media with ESI TOF MS did not reveal any histamine. L. reateri 6475 grown in a e media produces histamine, which is present in a TNF—inhibitory HPLC fraction.
Histamine was quantified in select HILIC—HPLCfractions using triple quadrupole mass spectrometry Triple quadrupole mass spectrometry is an established, highly ive method of quantifying small molecular compounds. Histamine was quantified in a select range of HlLlC—HPLC fractions from L. reateri 6475 glucose (BZ—B7) and sucrose (B2-B9) as well as the bacterial culture atant. High levels of histamine (>300 ng/mL) correlated with the y of the HILIC-HPLC fractions to t TNF (Figure 1). Low levels of histamine were measured in most fractions examined, including those from 6475 sucrose (Figure l). The ability of histamine to inhibit TNF production appears to be concentration dependent.
Synthetic histamine and histamine produced by L. reateri 6475 inhibit TNF production via the H2 receptor Histamine can si gnificantly inhibit TNF production from TLRZ—activated human monocytoid cells (THP—l) e 4A). Histamine can signal through four different histamine ors, however, monocytoid cells express high levels of the H1 and H3 receptors only.
Previous studies have shown effects of histamine on TNF production via the H2 receptor. H1 and Hg receptor—specific antagonists were used to ine which receptor was mediating the effect of histamine on THP—1 cells. H2 receptor— specific antagonists, ranitidine and cimetidine, could block TNF—inhibition by histamine in a concentration dependent manner e 2A). Flow cytometry analysis with H2 receptor—specific antibodies revealed that THP— 1 cells highly express the H2 receptor (data not shown). An H1 receptor—specific antagonist, indomethacin, had no effect on hibition by histamine (Figure 2A). Histamine blocks TNF production from TLRZ—activated THP-l cells via signaling through the H2 or. L. reateri 6475 conditioned media containing histamine significantly inhibits TNF compared to the media control, and this effect is partially blocked by H2 receptor but not H1 receptor antagonists (Figure 2A). A partial block in TNF suppression indicates that histamine present in 6475 ioned media is signaling via the H2 receptor but that other hibitory factors that act through alternative mechanisms may also be present in the conditioned media.
The cell pellet wash containing histamine of strain 6475 also suppresses TNF production wo 2013/011137 22 (Figure 2B). As seen with 6475 conditioned media, H2 receptor antagonists lly block the effect of the 6475 cell pellet wash (Figure 2B), ting multiple immunomodulins are present in the unfractionated cell pellet wash. The effects of hibitory fraction B3, which contains high amounts of purified histamine, were completely blocked by the addition of H2 receptor antagonists (Figure ZB).
Selection of strains ing histamine [dentz'fication/selection of histamine ing bacteria Strains to be tested and possibly selected were cultured under anaerobic conditions for 16—18 h in deMan, , Sharpe media , Franklin Lakes, NJ), and inoculated into 2 L of a semi—defined media, LDMIII (OD600 adjusted to 0.1). The carbon source was glucose, LDMIIIG. Each culture was grown for 24 h at 379C in an anaerobic workstation (MACS MG— 500, Microbiology International, Frederick, MD) supplied with a mixture of 10% C03, 10% H2, and 80% N2. Samples were taken at different times to follow the growth by measuring OD600. At stationary phase (24 h), the cells were d for analysis using real-time PCR to test for the presence of the three genes, the histidine/histamine antiporter, Hch, and Hch.
For strains positive for the three genes, the levels of produced histamine is ined 2O by triple quadrupole mass spectrometry. The strains with highest production of histamine (>250 pg/ml) are selected. Histamine production can also be evaluated and quantified by ELISAs 0r immunoassays.
EXAMPLE 3 Demonstration of immunomodulation The histidine operon contributes to the INF-inhibitory phenotype ofL. remeri 6475 Three genes that appear to be part of an operon are involved in histamine production by Lt remeri 6475. These genes are the histidine/histamine antiporter, the histidine 3O decarboxylase pyruvoyl type A (Hch), and Hch (Figure 3A). Previous transcriptomics studies suggested that the histidine/histamine antiporter gene and Hale/i were potentially important for the TNF-inhibitory phenotype of strain 6475. All 3 genes are strongly downregulated in 6475 grown in a sucrose media (loses TNF inhibition) compared to 6475 grown in a glucose media (Table 2). In addition, at least 1 gene in the operon is down—regulated in 2 W0 2013/011137 23 ] mutants that lose TNF-inhibition (Table 2). These mutants were investigated previously, and even though the gene products didn’t have TNF—inhibitory properties, the genes appeared to be ant for the anti-inflammatory phenotype of 6475. In contrast, 2 mutants that do not lose TNF tion demonstrated no egulation of any of the genes in the ine operon (Table 2). Mutations were made in each of these 3 genes by inserting a premature stop codon into the gene sequence (strains 1229, 1230 and 1231). A mutation was also made in an unrelated gene, the rifampicin ance gene, to serve as a negative control (strain JP577). A mutation in just one of the genes in the histidine operon was sufficient to cause a partial loss of TNF—inhibition compared to the wild-type strain (Figure 3B), suggesting that each one of these genes is important for the TNF—inhibitory phenotype of L. reuteri 6475. A partial loss of activity suggests that other active immunomodulins are still being produced by L. reuteri 6475.
ERIC/2 activation is essentialfor TNF production by TLR2-stimulated monocyzoid cells ERKl/2 is activated by phosphorylation from upstream MAPKK, MEKl/Z, and has been shown usly to be important for TNF production. THP-1 cells were treated with a ic MEKl/2 inhibitor, U0126, for varying amounts of time prior to stimulation with a TLR2 agonist to suppress ERKl/Z tion. Treatment with U0126 for 30 min was sufficient to prevent TNF production (data not shown). ERK1/2 is activated following TLR2 stimulation and important for stimulating TNF production in our model system. ation of the H2 receptor results in increased CAMP within the cells The H2 receptor is a G protein linked receptor that can activate adenylate cyclase and increase intracellular CAMP. TNF can be inhibited at the level of transcription by CAMP and CAMP analogs. THP—1 cells were ated with a TLR2 agonist in the presence of media control, 6475 supernatant or histamine with or t an H2 receptor nist and intracellular levels of CAMP were measured. L. reuteri 6475 supernatant caused a small but significant increase in CAMP (data not shown). Treatment with an 1-]; antagonist blocked this . An increase in CAMP was also seen with histamine ent, and the effect was blocked by an H2 antagonist (data not shown). A synthetic analog of CAMP, dibutyryl CAMP (dCAMP), was added to TLR2-stimulated THP—1 cells and the effect on TNF production was monitored. The addition of chMP (10'5 —- 10'3 M) was sufficient to inhibit TNF production (data not shown). Stimulation of the histamine H2 receptor results in increased CAMP, which can block downstream TNF production in activated monocytoid cells. wo 2013/011137 24 Protein Kinase A (PK/l) ty is importantfor TNF inhibition by L. renteri 6475, ine, and chMP Increased concentration of CAMP can activate PKA and subsequently inhibit the downstream ERK MAPK signaling pathway. To determine if PKA activity was important for TNF suppression by histamine produced by strain 6475, activated THP-1 cells were treated with a specific PKA tor, H89, in the presence of 6475 atant, fraction B3, ine or varying concentrations of chMP. The addition of H89 partially blocked TNF inhibition by all of these normally TNF—inhibitory compounds (data not shown). PKA activity is important for suppression of TNF by histamine and chMP.
Signaling through the H2 receptor blocks activation ofMEKI/Z and ERKI/Z Previous studies have demonstrated that PKA can inhibit Ras/Raf activation of MEK and subsequently ERK MAPK signaling. Treatment of ted THP—1 cells with 6475 atant, histamine or U0126 blocks phosphorylation of both MEKl/2 and downstream ERKl/2 compared to the media l (data not shown). Treatment with an H2 receptor antagonist restores activation of both MEKl/2 and ERKl/Z (data not shown). There was no difference in MEKl/Z and ERKl/Z protein levels with any of the treatment options.
Histamine from strain 6475 inhibits activation of MEK and downstream ERK to result in decreased TNF production from TLRZ—stimulated myeloid cells.
These ments thus show that stimulation of the H2 receptor results in increased CAMP, activation of Protein Kinase A (PKA) and inhibition of the K MAPK signaling pathway. As described above, mechanistic studies were med to determine the effect of histamine on Mitogen Activated Protein Kinase (MAPK) signaling pathways.
Inhibition of the K signaling pathway with a MEK—specific inhibitor is ient to block TNF production. Treatment of activated THP~l cells with strain 6475 supernatant or histamine increased intracellular CAMP. The increase in CAMP was blocked by ranitidine, a specific H2 receptor antagonist. Treatment of TLR2-stimulated THP—l cells with a synthetic analog of CAMP, chMP, is sufficient to inhibit TNF production. Inhibition of PKA activity 3O partially blocks TNF suppression by previously TNF—inhibitory compounds 6475 conditioned media, fraction B3, histamine, and chMP. Treatment of ted THP—l cells with 6475 conditioned media, histamine, or U0126 suppressed tion of MEKl/Z, an effect that was blocked in the presence of ranitidine. Treatment of activated THP-l cells with 6475 ioned media, histamine, or U0126 suppressed activation of ERKl/Z, an effect that was blocked in the presence of ranitidine.
EXAMPLE 4 MEKJ/Z and ERKI/Z detection by western blot THP—1 cells were lysed in ice—cold lysis buffer consisting of 50 mM Tris, pH 7.4, 250 mM NaCl, 5 mM EDTA, 50 mM NaF, 1 mM , 1% v/v Nonidet P40, 0.2% v/v NaNg, and se and phosphatase inhibitors. s were incubated on ice for 30 min, ed every 10 min, and cleared by centrifugation at 13,000 x g for 10 min at 4°C. Protein concentrations were measured using the Quant-iTTM Protein Assay kit (lnvitrogen) and a Qubit fluorometer according to the manufacturer’s instructions. Equal s of proteins were loaded onto electrophoresis gels.
Analysis of ERKl/Z activation was performed using specific phospho-ERK1/2 antibodies. Cell extracts were loaded on a 10% SDS—polyacrylamide gel and transferred to polyvinylidene difluoride membranes (Bio—Rad, Hercules, CA). Membranes were blocked overnight at 4°C in blocking buffer (Li-Cor Biosciences, Lincoln, NE). After several washes, membranes were probed with ERKl/Z, phospho—ERKl/2 or B-Actin specific antibodies diluted in blocking buffer (Li-Cor) for 1 h at room temperature. After , nes were incubated with the appropriate horseradish peroxidase—conjugated secondary antibody for l h at room temperature, and blots were then ped using a chemiluminescent detection. Analysis of MEKl/2 activation was performed as described above except primary antibody incubation was overnight at 4°C.
EXAMPLE 5 hch mutant yield diminished ability to attenuate colitis Bacterial strains and culture Mutants were generated using RecT—mediated oligonucleotide recombineering. L. retiteri expressing RecT (strain OO) was used to construct mutations in rpoB (locus tag HMPREF0536_0828 (ZP_03961568)) and the target genes located in the histidine decarboxylase gene cluster 0536_1229 (ZP_03961969), HMPREF0536_1230 (ZP_0396l970) and HMPREF0536_1231 (ZP_03961971) to yield strains RPRB3002.
RPRB3004, RPRB3005 and 06, respectively. Mutations were verified by PCR, and W0 2013/011137 26 the integrity was med by sequence analysis.
L. reuteri ATCC PTA 6475 and histidine decarboxylase gene (hch) mutant were cultured in deMan, Rogosa, Sharpe (Difco, Franklin Lakes, NJ) at 37°C in an anaerobic workstation (MACS , Microbiology International, Frederick, MD) supplied with a mixture of % C02, 10% H2, and 80% N2. ation of L. reuteri cells and administration to mice A single colony of each of the L. reuteri strains was ated in 10 ml of MRS medium and grown at 37°C under anaerobic condition for 18—20 hours. Bacteria adjusted to OD600=0.03 were inoculated into 40ml of MRS to start the fermentation and grown at 37°C under anaerobic condition for 5.5 hrs (OD600z25, bacteria were in exponential phase at this time . The cells were gently pelleted (2500 x g, RT, 4 minutes) and resuspended in MRS at a tration of 25 x109 CPU/ml. As a media control, sterile MRS medium was used. Each 8— week old female BALB/c mouse received one dose of y prepared wild type L. reuteri 6475 or thA mutant or MRS (0.2 ml each time) everyday for seven days by orogastric gavage after 10 days of acclimatization. All mouse experiments were performed according to approved protocol (AN-4199; animal facility of Baylor College of Medicine). Mice (45 days old) were received from Harlan Laboratories (Houston, TX) and maintained under specific pathogen—free conditions in top cages (5 mice per cage) and had free access to distilled water and Harlan rodent chow 2918. Mice were divided into different groups randomly.
Induction of acute s using trinitrobenzene sulfonic acid (TNBS) rectal enema. Colitis was induced six hours before the sixth gavage. Mice were anesthetized by constant isoflurane inhalation. A 5% TNBS solution in water (Sigma-Aldrich, USA) was diluted with equal volume of absolute ethanol and administered at dose of 100 mg/kg body weight intrarectally.
Mice were kept head down in a vertical position for 2 minutes after enema to ensure complete retention of enema in the colon. Procedure control mice received 50% ethanol in PBS. Mice were weighed prior to TNBS administration and two days after TNBS stration. Then mice were sacrificed. Colonic inflammation and damage was determined by weight loss, macroscopic score and serum SAA tration.
Macroscopic assessment of colitis The colons were collected, opened longitudinally and images were recorded with a digital camera. Colonic inflammation and damage were determined according to the Wallace criteria (Morris et 31., 1989). Each colon was scored y. Statistics were performed using GraphPad Prism version 5.01 (GraphPad Software, La J011a, CA). Kruskal—Wallis test was used to detect a significant difference among all groups included in the analysis. Results were summarized as median and interquartile range. ement of serum amyloid protein A (SAA) as systemic inflammation marker Blood samples were collected by cardiac puncture, anti-coagulated and centrifuged for 10 minutes at 13,000 rpm to isolate plasma. Serum amyloid A (SAA) trations in plasma samples were measured using ELISA kits from ALPCO (Salem, NH) according to manufacturer instructions. SAA is an acute phase protein indicative of systemic inflammation in mice that ates with colitis severity.
Results L. reuteri 6475 protects mice against TNBS-induced acute colitis The anti-inflammatory effects of L. reuteri, 6475 were tested in a TNBS-induced mouse model of acute colitis. Mice that received L. reuteri 6475 by orogastric gavage every day were compared with mice that received the media control. Mice challenged with PBS instead of TNBS were also studied as colitis negative controls.
The figures 4—6 represent data from two independent experiments. Colitis negative controls that received PBS instead of TNBS ectally had very low weight loss (or even gained weight), rare colon damage and low serum SAA concentrations. Colitis ve mice that received MRS media and TNBS/ETOH developed a severe s terized by a large amount of weight loss, ulceration with inflammation in the colon and the major sites of damage extending greater than 1 cm, and significantly ed SAA concentrations in serum.
Orogastric gavagc with L. reuteri 6475 icantly reduced weight loss, macroscopic inflammation in the colon and serum SAA concentrations, showing that L. reuteri 6475 significantly ated colitis. thA mutant yields diminished y to attenuate colitis Using the same mouse model, we tested whether thA gene, which encodes histidine decarboxylase was required for the anti-inflammatory effects of L. reuteri 6475. 8—week old wo 2013/011137 28 2012/064351 female BALB/c mice were randomly divided into three groups which received wild type L. reuteii 6475 or thA mutant or MRS media respectively. The figures 7 and 8 represent data from two independent experiments. Again, orogastric gavage with L. reuteri 6475 significantly reduced weight loss and colon damage compared with media control group.
Mice received thA mutant significantly increased weight loss and macroscopic inflammation in the colon compared with mice that received wild type ia, showing that thA mutant yields diminished ability to attenuate colitis. 2012/064351 Print Out (Original in Electronic Form) (This sheet is not part of and does not count as a sheet of the international application) Form PCTIRO/134 (SAFE) Indications ng to Deposited Microorganism(s) or Other Biological Material (PCT Rule 13bis) 044 ed Using PCT Online Filing Version 3.5.000.225 MT/FOP 20020701/0.20.5.20 international ation No.
Applicant's or agent's tile reference 69 . 113613 1 The indications made below relate to the deposited microorganism(s) or other biological al referred to in the description on: 1-1 page 1 8 1-2 line 1 2 1-3 Identification of deposit 1'3'1 Name 0* deP°5iiaiY insmmi‘m ATCC American Type Culture Collection 12 Address of depositary institution 1 o 8 0 1 University Blvd .
I Manassas I Virginia 2 0110—22 0 9United States of America 1'3‘3 Dale°fdep°$i1 21 er 2004 (21.12.2004) 14 Accession Number ATCC PTA-64 75 1-5 Designated States for Which All designations Indications are Made FOR RECEIVING OFFICE USE ONLY This form was received with the international application: Yes (yes or no) Authorized officer Van Deursen, Theresia FOR INTERNATIONAL BUREAU USE ONLY This iorm was received by the international Bureau on: Authorized officer

Claims (24)

    What is claimed is:
  1. l. A method of selecting a probiotic lactic acid bacterial strain for use in the local production of histamine in a mammal, n said method comprises screening bacteria for the presence of an active histidine operon and selecting a strain which has an active histidine operon and is capable ofproducing histamine.
  2. 2. The method of claim 1, wherein said strain is selected for its ability to produce histamine at a level of greater than 250 pg/ml. 10
  3. 3. The method of claim 1 or claim 2, wherein said strain is Lactobacillus reuteri.
  4. 4. A t comprising cells of a lactic acid bacterial strain ed by the ion method according to any one of claims 1 to 3, wherein said lactic acid bacterial strain has an active histidine operon and is e of producing histamine, for use in the local production 15 of histamine in a mammal, wherein said use is in the treatment and/or prophylaxis of inflammatory conditions.
  5. 5. The product for use according to claim 4, n said mammal is a human. 20
  6. 6. The t for use according to claim 4 or claim 5, wherein the local production of histamine is in the GI tract, GU tract, oral cavity, lungs, airways, or on the skin of said mammal.
  7. 7. The product for use according to any one of claims 4 to 6, wherein the atory 25 condition is selected from the group consisting of Colitis, Inflammatory Bowel Disease, Irritable Bowel Syndrome, diverticulosis, gingivitis and vaginitis.
  8. 8. The product for use according to any one of claims 4 to 7, wherein said strain is Lacrobacillus reuterz'.
  9. 9. The product for use according to claim 8, wherein said strain is Lactobacillus reuteri 64 75.
  10. 10. The t for use according to any one of claims 4 to 9, wherein said use further comprises the administration of at least one further therapeutic or nutritional agent.
  11. 11. The t for use according to claim 10, wherein said further agent comprises histidine or a histidine analog, an appropriate carbon source which supports the production of histamine by said strain, or a combination thereof.
  12. 12. The product for use according to claim ll, wherein said histidine or histidine analog is in the form of a histidine or histidine analog containing foodstuff or food supplement, or 10 wherein said carbon source comprises e.
  13. 13. A composition comprising: (i) a lactic acid bacterial strain obtained by the selection method according to any one of claims 1 to 3, wherein said lactic acid bacterial strain has an active histidine operon and is 15 capable of ing ine; and (ii) at least one additional component which is a source of histidine or histidine analog, and optionally an appropriate carbon source which supports the production of histamine by said strain. 20
  14. 14. The ition according to claim 13, wherein said histidine or ine analog is in the form of a histidine or histidine analog containing foodstuff or food supplement, or wherein said carbon source comprises glucose.
  15. 15. Use of cells of a lactic acid bacterial strain obtained by the selection method according to 25 any one of claims 1 to 3, n said lactic acid bacterial strain has an active histidine operon and is capable of producing histamine locally in a mammal, in the manufacture of a medicament for the treatment and/or laxis of inflammatory ions.
  16. 16. The use according to claim 15, wherein said mammal is a human.
  17. 17. The use according to claim 15 or claim 16, wherein the local production of histamine is in the GI tract, GU tract, oral cavity, lungs, airways, or on the skin of said mammal.
  18. 18. The use according to any one of claims 15 to 17, n the inflammatory condition is selected from the group consisting of Colitis, Inflammatory Bowel Disease, Irritable Bowel me, diverticulosis, gingivitis and vaginitis.
  19. 19. The use according to any one of claims 15 to 18, wherein said strain is Lactobacillus reuteri.
  20. 20. The use according to claim 19, wherein said strain is Lactobacillus reuteri 6475. 10
  21. 21. The use according to any one of claims 15 to 20, wherein medicament is adapted for stration with at least one further therapeutic or nutritional agent.
  22. 22. The use according to claim 21, wherein said further agent comprises histidine or a histidine analog, an appropriate carbon source which ts the production of histamine by 15 said strain, or a combination thereof.
  23. 23. The use according to claim 22, wherein said histidine or histidine analog is in the form of a histidine or histidine analog containing foodstuff or food supplement, or wherein said carbon source ses glucose.
  24. 24. The method according to claim 1, substantially as herein described with reference to any one of the Examples and/or
NZ619605A 2011-07-21 2012-07-20 Production and use of bacterial histamine NZ619605B2 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US201161572742P 2011-07-21 2011-07-21
US61/572,742 2011-07-21
US13/552,686 2012-07-19
US13/552,686 US20130022586A1 (en) 2011-07-21 2012-07-19 Production and use of bacterial histamine
PCT/EP2012/064351 WO2013011137A1 (en) 2011-07-21 2012-07-20 Production and use of bacterial histamine

Publications (2)

Publication Number Publication Date
NZ619605A NZ619605A (en) 2015-10-30
NZ619605B2 true NZ619605B2 (en) 2016-02-02

Family

ID=

Similar Documents

Publication Publication Date Title
US10898529B2 (en) Production and use of bacterial histamine
AU2016322617B2 (en) Novel lactobacillus having various functions, and use thereof
EP3048165B1 (en) Novel strain of bifidobacterium animalis subsp. lactis cect 8145 and use thereof for the treatment and/or prevention of excess weight and obesity and associated diseases
US8936783B2 (en) Use of Lactobacillus for treatment of autoimmune diseases
van der Kleij et al. Protective effects of Lactobacillus reuteri and Bifidobacterium infantis in murine models for colitis do not involve the vagus nerve
JP6935632B2 (en) Faecalibacterium prausnitzi CNCM I-4573 strain for the treatment and prevention of gastroenteritis
KR101937364B1 (en) Novel lactic acid bacteria having immunoregulatory activities derived from human digestive tract and use thereof
KR101862051B1 (en) Novel lactic acid bacteria having immunoregulatory activities derived from human digestive tract and use thereof
US20120183504A1 (en) Composition and use of probiotic strain gm-263 (adr-1) in treating renal fibrosis in diabetes
CN108697742A (en) Composition for preventing and/or treating vitamin B12 deficiency disease and method
Wang et al. Modulation of intestinal barrier, inflammatory response, and gut microbiota by Pediococcus pentosaceus zy-B alleviates Vibrio parahaemolyticus infection in C57BL/6J mice
Meshkibaf et al. Preferential production of G-CSF by a protein-like Lactobacillus rhamnosus GR-1 secretory factor through activating TLR2-dependent signaling events without activation of JNKs
JP2020022488A (en) Novel lactic acid bacteria having various functionalities and application thereof
KR101247802B1 (en) Composition for preventing and treating inflammatory disease comprising piperine or pharmaceutically acceptable salt thereof as an active ingredient
NZ619605B2 (en) Production and use of bacterial histamine
Garcia-Gonzalez et al. Health-Promoting Role of Lactiplantibacillus plantarum Isolated from Fermented Foods. Microorganisms 2021, 9, 349
Casula Probiotic Lactobacillus strains attenuate oxysterols-induced alteration of human intestinal membrane permeability
JP2020015757A (en) Pharmaceutical composition and food composition for inducing satiation and extending satiety in object in need thereof
Merra et al. Oral Communications