WO2021221110A1 - Small intestinal bacterium inducing proliferation or activation of th1 cells and/or th17 cells - Google Patents

Small intestinal bacterium inducing proliferation or activation of th1 cells and/or th17 cells Download PDF

Info

Publication number
WO2021221110A1
WO2021221110A1 PCT/JP2021/017000 JP2021017000W WO2021221110A1 WO 2021221110 A1 WO2021221110 A1 WO 2021221110A1 JP 2021017000 W JP2021017000 W JP 2021017000W WO 2021221110 A1 WO2021221110 A1 WO 2021221110A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
bacterium
bacteria
disease
polynucleotide
Prior art date
Application number
PCT/JP2021/017000
Other languages
French (fr)
Japanese (ja)
Inventor
賢也 本田
学 永山
博徳 山本
Original Assignee
学校法人慶應義塾
国立研究開発法人理化学研究所
学校法人自治医科大学
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 学校法人慶應義塾, 国立研究開発法人理化学研究所, 学校法人自治医科大学 filed Critical 学校法人慶應義塾
Publication of WO2021221110A1 publication Critical patent/WO2021221110A1/en

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/195Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria
    • 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

Definitions

  • the present invention relates to a small intestinal bacterium that induces proliferation or activation of Th1 cells and / or Th17 cells (hereinafter also referred to as "Th1 cells and the like").
  • the present invention relates to a method for evaluating a disease caused by Th1 cells or the like using the bacterium as an index.
  • the present invention relates to a composition or a vaccine composition for inducing proliferation or activation of Th1 cells or the like containing the bacteria or the like as an active ingredient.
  • the present invention is for treating a composition for suppressing the proliferation or activation of Th1 cells or the like or a disease caused by the Th1 cells or the like, which contains a substance having an antibacterial action against the bacteria as an active ingredient.
  • the composition is for treating a composition for suppressing the proliferation or activation of Th1 cells or the like or a disease caused by the Th1 cells or the like, which contains a substance having an antibacterial action against the bacteria as an active ingredient.
  • the present invention relates to a method for screening a substance using the bacterium as an index. Furthermore, the present invention relates to a method for suppressing the proliferation or activation of Th1 cells or the like, or a method for treating a disease caused by Th1 cells or the like, which targets the bacterium.
  • CD Crohn's disease
  • Non-Patent Documents 4 and 5 The gut microbiota is a potent regulator of host immunity and has been reported to be associated with several diseases.
  • CD it is being shown that a change in the bacterial flora called intestinal bacterial flora symbiotic imbalance (disbiosis) affects the pathogenesis.
  • intestinal bacterial flora symbiotic imbalance affects the pathogenesis.
  • disbiosis intestinal bacterial flora symbiotic imbalance
  • a decrease in ⁇ -diversity of the microbial flora and a decrease in the abundance of Faecalibacterium prasnitzii have been frequently reported (Non-Patent Document 6).
  • An object of the present invention is to provide a composition for treating, improving or preventing a CD or the like that targets the identified bacterium, and a method for inspecting the CD or the like using the bacterium as an index.
  • a mucosal sample of jejunum and ileum was collected from a CD patient using a double-balloon endoscopy (DBE) system.
  • the DBE system includes a fiberscope and balloon pump system, which allows for more detailed examination of SI (distal jejunum and proximal ileum) compared to conventional push enteroscopy.
  • SI mucosal samples were collected from CD patients and non-CD patients by scraping or biopsy using such DBE, and comparative microbiome analysis was performed. As a result, it was revealed that the composition of the microbial flora of the SI mucosa in CD patients was different from that in non-CD patients, and that the population belonging to several families including Enterobacteriaceae, Ruminococcaceae and Bacteroidaceae was increasing. ..
  • Th1 cells in the intestinal tract was enhanced. Furthermore, the accumulation of Th17 cells was enhanced, though not as much as Th1 cells.
  • the E. The coli strain induced Th1 cells specifically for the strain and showed a high ability to induce intestinal inflammation.
  • the present inventors have found a small intestinal bacterium that induces proliferation or activation of Th1 cells and / or Th17 cells that can be involved in the onset of CD, etc., and have completed the present invention.
  • ⁇ 1> A small intestinal bacterium that induces proliferation or activation of Th1 cells and / or Th17 cells.
  • Bacterial bacterial group according to ⁇ 1> which is at least one bacterium selected from the following bacterial group: Bacteria having a polynucleotide having 95% or more identity to the DNA sequence set forth in SEQ ID NO: 1. Bacteria having a polynucleotide having 95% or more identity to the DNA sequence set forth in SEQ ID NO: 2. Bacteria having a polynucleotide having 95% or more identity to the DNA sequence set forth in SEQ ID NO: 3.
  • Bacteria to have ⁇ 3> Bacteria having a polynucleotide having 95% or more identity to the DNA sequence set forth in SEQ ID NO: 1 and 95% or more identity to the DNA sequence set forth in SEQ ID NO: 2.
  • the bacterium according to ⁇ 1> which is a combination with a bacterium having a polynucleotide.
  • a step of ingesting a test substance into a non-human germ-free animal (2) When the number or activity of Th1 cells and / or Th17 cells in the small intestine of the non-human animal is detected, and when suppression of proliferation or activation of the cells is detected in (3) step (2).
  • the step of determining that the test substance is a substance that suppresses the proliferation or activation of Th1 cells and / or Th17 cells in the small intestine. How to include.
  • ⁇ 5> A method for screening a substance that suppresses the proliferation or activation of Th1 cells and / or Th17 cells in the small intestine.
  • the test substance is a bacterium having a polynucleotide having a polynucleotide having 95% or more identity with respect to the DNA sequence set forth in SEQ ID NO: 1 and / or 95% or more with respect to the DNA sequence set forth in SEQ ID NO: 2.
  • Ingestion of non-human animals colonized with a polynucleotide having a polynucleotide having the same identity as When (2) the step of detecting the number of the bacteria in the small intestine of the non-human animal and (3) the suppression of the growth of the bacteria are detected in the step (2), the test substance is Th1 cells in the small intestine. And / or the step of determining that the substance suppresses the proliferation or activation of Th17 cells. How to include.
  • ⁇ 6> The method according to ⁇ 4> or ⁇ 5>, wherein the substance is a bacterium or a bacteriophage.
  • ⁇ 7> A method for evaluating a disease caused by Th1 cells and / or Th17 cells. (1) For bacteria having a polynucleotide having 95% or more identity with respect to the DNA sequence set forth in SEQ ID NO: 1 and / or the DNA sequence set forth in SEQ ID NO: 2 in the small intestinal mucosa of the subject.
  • Quantifying bacteria with polynucleotides having 95% or more identity (2) A step of comparing the value obtained by quantifying in step (1) with a corresponding value obtained by quantifying the bacteria in the mucosa of the small intestine of a human who does not suffer from the disease, and step (3) (3).
  • the quantitative value in the small intestinal mucosa of the subject is higher than the corresponding value, the step of determining that the subject is suffering from the disease is performed.
  • ⁇ 8> Induces proliferation or activation of Th1 cells and / or Th17 cells containing the bacterium according to any one of ⁇ 1> to ⁇ 3> or a physiologically active substance derived from the bacterium as an active ingredient.
  • composition to do The bacterium according to any one of ⁇ 1> to ⁇ 3> for producing a composition for inducing proliferation or activation of Th1 cells and / or Th17 cells, or the same. Regarding the use of bioactive substances derived from bacteria.
  • a vaccine composition comprising the bacterium according to any one of ⁇ 1> to ⁇ 3> or an antigen specific to the bacterium as an active ingredient.
  • the present invention also relates to the use of the bacterium according to any one of ⁇ 1> to ⁇ 3> or an antigen specific to the bacterium for producing a vaccine composition.
  • ⁇ 10> Suppresses the proliferation or activation of Th1 cells and / or Th17 cells containing a substance having an antibacterial action against the bacterium according to any one of ⁇ 1> to ⁇ 3> as an active ingredient.
  • Composition for. ⁇ 11> The composition according to ⁇ 10>, wherein the substance is a substance selected by the method according to any one of ⁇ 4> to ⁇ 6>.
  • the present invention also relates to the bacterium according to any one of ⁇ 1> to ⁇ 3> for producing a composition for suppressing the proliferation or activation of Th1 cells and / or Th17 cells.
  • the present invention relates to the use of a substance having an antibacterial activity, or a bacterium or bacteriophage obtained by the screening method according to any one of ⁇ 4> to ⁇ 6>.
  • ⁇ 12> The composition according to any one of ⁇ 9> to ⁇ 11>, which is a composition for treating, ameliorating or preventing a disease caused by Th1 cells and / or Th17 cells.
  • ⁇ 13> The composition according to ⁇ 12>, which is ingested by the subject determined to be suffering from the disease by the method according to ⁇ 7>.
  • ⁇ 14> The composition according to ⁇ 12> or ⁇ 13>, wherein the disease is Crohn's disease.
  • the present invention also comprises the production of compositions for treating, ameliorating or preventing said diseases.
  • the present invention relates to the use of a substance having a bacterium or a substance selected by the method according to any one of ⁇ 4> to ⁇ 6>.
  • ⁇ 16> The bacterium according to any one of ⁇ 1> to ⁇ 3> or an antigen specific to the bacterium is ingested by the subject, and the proliferation or activity of Th1 cells and / or Th17 cells in the subject.
  • ⁇ 17> A substance having an antibacterial action against the bacterium according to any one of ⁇ 1> to ⁇ 3> is ingested by a subject, and the proliferation or activity of Th1 cells and / or Th17 cells in the subject.
  • ⁇ 18> The method according to ⁇ 17>, wherein the substance is a substance selected by the method according to any one of ⁇ 4> to ⁇ 6>.
  • the present invention also relates to the bacterium according to any one of ⁇ 1> to ⁇ 3> for suppressing the proliferation or activation of Th1 cells and / or Th17 cells in a subject, or a bacterium specific to the bacterium.
  • the antigen the substance having an antibacterial action against the bacterium according to any one of ⁇ 1> to ⁇ 3>, or the method according to any one of ⁇ 4> to ⁇ 6>.
  • ⁇ 19> A disease caused by Th1 cells and / or Th17 cells in the subject by ingesting the bacterium according to any one of ⁇ 1> to ⁇ 3> or an antigen specific to the bacterium. How to treat, improve or prevent.
  • ⁇ 20> A disease caused by Th1 cells and / or Th17 cells in a subject by ingesting a substance having an antibacterial action against the bacterium according to any one of ⁇ 1> to ⁇ 3>. How to treat, improve or prevent.
  • ⁇ 21> The method according to ⁇ 20>, wherein the substance is a substance selected by the method according to any one of ⁇ 4> to ⁇ 6>.
  • ⁇ 22> The item according to any one of ⁇ 19> to ⁇ 21>, wherein the subject is the subject determined to have the disease by the method according to ⁇ 7>.
  • Method. ⁇ 23> The method according to any one of ⁇ 19> to ⁇ 22>, wherein the disease is Crohn's disease.
  • the present invention also comprises the bacterium according to any one of ⁇ 1> to ⁇ 3> or an antigen specific to the bacterium for treating, ameliorating or preventing the disease, ⁇ 1> to ⁇ 3.
  • the present invention by targeting a small intestinal bacterium that induces proliferation or activation of Th1 cells and / or Th17 cells, it becomes possible to suppress the proliferation or activation of the cells, and thus to the cells. It is possible to treat, improve or prevent the resulting disease. Further, according to the present invention, it is also possible to test for diseases caused by the cells by using the amount of bacteria in the small intestine as an index.
  • FIG. 6 is a schematic diagram outlining a method for obtaining a sample from the small intestine (SI) via double-balloon enteroscopy.
  • SI small intestine
  • CD Crohn's disease
  • non-CD 16S rRNA gene sequence analysis
  • Each graph shows the results of analysis of the number of OTUs (Operational Taxonomy Units), ACE (Abundance-based Coverage Estimator), Chao1 and Shannon index.
  • Each bar graph represents the average value of the group.
  • Each point represents an individual sample. Error bars represent standard deviation (SD).
  • SD standard deviation
  • the P value obtained by analyzing with the unpaired Student's t-test is added. It is a two-dimensional plot figure which shows the result of having analyzed the structural similarity of the bacterial flora between the SI sample derived from a CD patient and that derived from a non-CD patient.
  • FIG. 5 is a graph showing the relative abundance of bacterial taxa at the Family level of the SI flora.
  • the horizontal axis shows the results for each bacteriaceae (the left side shows the sample analysis results derived from non-CD patients, and the right side shows the sample analysis results derived from CD patients).
  • Each thick bar represents the average value of the group.
  • Each point represents an individual sample. Error bars represent SD.
  • FIG. 5 is a graph showing the relative abundance of bacterial taxa at the family level of the SI flora.
  • the horizontal axis shows the results for each bacteriological family (the left side shows the sample analysis results derived from non-CD patients, and the middle shows the sample analysis results of CD patients without intestinal strictures (trictures).
  • Each right side shows the results of sample analysis from a CD patient with intestinal stenosis).
  • Each thick bar represents the average value of the group.
  • Each point represents an individual sample. Error bars represent SD.
  • Each asterisk indicates that the value obtained by multiple t-test by the FDR approach is *** ⁇ 0.05; ** less than 0.01; *** ⁇ 0.001; *** ⁇ 0.0001.
  • 6 is a histogram of LDA scores showing the results of calculations for the differentially abundant taxa between CD patients (“CD” in the figure) and non-CD patients (“non-CD” in the figure). In the figure, in the LDA score, the analysis result for non-CD patients is shown on the negative side, and the analysis result for CD patients is shown on the positive side. It is a histogram of the misjudgment rate which shows the result calculated by comparing the sample derived from a CD patient (“CD” in the figure) with that of a non-CD patient (“non-CD” in the figure).
  • FDR farnesoid ⁇ 0.05
  • the analysis result for non-CD patients is shown on the minus side
  • the analysis result for CD patients is shown on the plus side.
  • the three types of computational analysis are linear discriminant analysis (LDA) (LEfSe) combined with effect size measurement, multiple t-test method (Multiple t-tests) using false judgment rate, and CD-related SI mucous membrane [SI of CD patients].
  • each asterisk indicates that the value obtained by the unpaired Student's t-test is * P ⁇ 0.05; ** P ⁇ 0.01.
  • E. coli in saliva (“Saliva” in the figure), SI and feces (“Faces” in the figure). colli and R.
  • It is a graph which shows the relative abundance of gnavus.
  • the horizontal axis shows the results for each sample (the left side shows the sample analysis result derived from a non-CD patient, and each right side shows the sample analysis result derived from a CD patient).
  • the vertical axis represents the relative abundance.
  • Each thick bar represents the average of the group.
  • Each point represents an individual sample. Error bars represent SD.
  • Each asterisk indicates that the value obtained by the unpaired Student's t-test is * P ⁇ 0.05; ** P ⁇ 0.01.
  • It is a band graph which shows the SI bacterial flora composition in each sample determined by 16S rRNA sequencing.
  • OTUs significantly concentrated in CD patients are marked in warm colors, those concentrated in non-CD patients (non-CD patients) are blue, and those with no significant difference are those. Shown in gray.
  • nine kinds of strains isolated from the SI sample of the CD patient are shown.
  • CD4 + TCR ⁇ + T cells CD4T cells
  • SLP lamina propria of the small intestine
  • CLP lamina propria of the colon
  • GF Aseptic (GF) C57BL / 6 (B6) mice were orally inoculated with a mixture of the above 9 types of CD concentrates (9-mix) and euthanized after 3 weeks.
  • GF + 9mix (“+9mix” in FIGS. 3B and 3D) indicates the analysis result of the mouse.
  • GF indicates the analysis result of GF mice not inoculated with the CD concentrate.
  • SPF indicates the analysis result of a specific pathogen-free mouse. It is a graph which shows the result of having analyzed the ratio of IFN- ⁇ + cell and IL-17 + cell about the CD4T cell of the small intestine laminalitis and the colon laminalitis. Each point represents an individual mouse. Each thick bar represents the average of the group.
  • FIG. 3 is a flow cytometric plot showing representative results of analyzing the expression of IFN- ⁇ , IL-17 and DR3 for CD4T cells in the colon lamina intestinal. The notation in the figure is the same as in FIG. 3A. It is a graph which shows the result of having analyzed the expression of IFN- ⁇ , IL-17 and DR3 about the CD4T cell of the colon lamina propria.
  • FIG. 3 is a flow cytometric plot showing representative results of analyzing the expression of IFN- ⁇ and IL-17 for CD4T cells in the colon lamina basement.
  • “GF + 131A1” and “GF + 35A1” are referred to as R.I. gnavus strain and E. coli.
  • the analysis result of the GF mouse which was orally inoculated with each of the colli strains is shown.
  • "GF + 2mix” shows the analysis results of GF mice that were orally inoculated with a mixture of these two strains. It is a graph which shows the result of having analyzed the expression of IFN- ⁇ and IL-17 about the CD4T cell of the colon lamina intestinal.
  • "GF” indicates the analysis result of GF mice not inoculated with the strain.
  • “+ 131A1” and “+ 35A1” are R.I. gnavus strain and E. coli.
  • the analysis result of the GF mouse which orally inoculated each of the colli strains is shown.
  • “+ 2-mix” indicates the analysis result of GF mice orally inoculated with a mixture of these two strains. Representative results of analysis of the proportions of IFN- ⁇ + cells and IL-17 + cells for CD4T cells in the lamina intestinal of the small intestine (“SILP” in the upper middle of the figure) and the lamina intestinal of the colon (“CLP” in the lower middle of the figure) are shown. It is a flow cytometry plot figure which shows. In GF mice, CD patient-derived E. E. coli strain (E. coli 35A1 strain, E. coli LF82 strain, or E. coli MG1655 strain) was orally inoculated and euthanized 3 weeks later.
  • E. E. coli strain E. coli 35A1 strain, E. coli LF82 strain, or E. coli MG1655 strain
  • the coli strain was established, followed by intraperitoneal injection of anti-mouse IL-10 receptor (IL-10R) antibody (1 mg / individual) weekly from day 1 to the end of the experiment to assess intestinal inflammation.
  • IL-10R anti-mouse IL-10 receptor
  • "GF + 35A1" and "GF” are E.I.
  • the analysis results of the mouse in which the colli 35A1 strain is colonized and the mouse in which the colli 35A1 strain is not colonized are shown.
  • the "+” and "-" of anti-IL10R indicate the presence or absence of anti-mouse IL-10R antibody injection, respectively.
  • Each E. It is a graph which shows the histological colitis score in the cecum of the GF mouse which established the coli strain. Each point represents an individual mouse. Each thick bar represents the average of the group.
  • CD shows the result of analyzing a sample derived from a Crohn's disease patient
  • non-CD shows the result of analyzing a sample derived from a non-Crohn's disease patient.
  • Each graph shows the results of analysis for the number of observed OTUs, ACE, Chao1 and Shannon index.
  • “Antegrada” and “Retrograda” indicate that they are the results of analysis of SI mucosal samples obtained by antegrade and prograde insertion, respectively.
  • Each bar graph represents the average value of the group. The P value obtained by analyzing with the unpaired Student's t-test is added.
  • the analysis results for each bacterial phylum are shown in order from the left, SI samples derived from non-CD patients obtained by anterograde insertion, and SIs derived from CD patients obtained by anterograde insertion.
  • the sample, the SI sample derived from a non-CD patient obtained by retrograde insertion, and the SI sample derived from a CD patient obtained by retrograde insertion are shown.
  • Each bar graph represents the average value of the group. Error bars represent SD.
  • FIG. 5 is a graph showing the relative abundance of bacterial taxa at the Family level in the mucosa of the small intestine. The notation in the figure is the same as that in FIG. 5C. It is a graph which shows the relative abundance of a bacterial taxon at the OTU level in the small intestinal mucosa. The notation in the figure is the same as that in FIG. 5C. It is a graph which shows the ⁇ diversity index of SI bacterial flora.
  • CD shows the result of analyzing a sample derived from a Crohn's disease patient
  • non-CD shows the result of analyzing a sample derived from a non-Crohn's disease patient.
  • Each graph shows the results of analysis for the number of observed OTUs, ACE, Chao1 and Shannon index.
  • Biopsy and “Scrape” indicate that they are the results of analysis of SI mucosal samples obtained by biopsy and scraping, respectively.
  • Each bar graph represents the average value of the group. The P value obtained by analyzing with the unpaired Student's t-test is added.
  • FIG. 5 is a graph showing the relative abundance of bacterial taxa at the Family level in the mucosa of the small intestine. The notation in the figure is the same as that in FIG. 5H. It is a graph which shows the relative abundance of a bacterial taxon at the OTU level in the small intestinal mucosa. The notation in the figure is the same as that in FIG. 5H.
  • FIG. 1 It is a graph which shows the relative abundance in a family-level bacterial taxon in the SI mucosal flora.
  • the horizontal axis shows the results for each bacteriological family (each left side shows the sample analysis results from non-CD patients, and each right side shows the sample analysis from CD patients receiving anti-TNF- ⁇ antibody therapy. The results are shown, and the results of sample analysis from other CD patients are shown in the middle).
  • Each thick bar represents the average of the group.
  • Each triangle represents an individual sample. Error bars represent 95% confidence intervals. For each asterisk, the value obtained by the multiple t-test using the misjudgment rate is *** ⁇ 0.05; *** ⁇ 0.01; *** ⁇ 0.001; *** ⁇ 0.0001. Show that.
  • each left side shows a sample analysis result (non-CD) derived from a non-CD patient
  • each right side shows a sample analysis result (CD) derived from a CD patient.
  • Each thick bar represents the average of the group.
  • Each point represents an individual sample. Error bars represent 95% confidence intervals.
  • the value obtained by the unpaired Student's t-test is *** ⁇ 0.05; *** ⁇ 0.01; *** ⁇ 0.001; *** ⁇ 0.0001.
  • the horizontal axis shows the results for each OTU (each left side shows the sample analysis result derived from a non-CD patient, and each right side shows the sample analysis result derived from a CD patient).
  • Each bar shows the average for the group. Error bars represent SD.
  • E. 3 is a graph showing the DNA concentration of the strain in SI, cecum (“Cecum” in the figure) and feces (“Feces” in the figure) of mice in which only the coli 35A1 strain has been established.
  • DNA was extracted from the fecal pellets of the mice and the lumen contents of the cecum and SI, and the DNA concentration of the fungus was determined by qPCR. Each point represents an individual mouse. Each thick bar represents the average of the group. Error bars represent SD. For each asterisk, the value obtained by one-way analysis of variance (one-way ANOVA) used in combination with Tukey's post-test is * P ⁇ 0.05; ** P ⁇ 0.01; *** P ⁇ 0. Indicates that it is 001. It is a graph which shows the ratio of IFN- ⁇ + cell in CD4T cell in the colon laminalitis. E. The E.
  • the coli 35A1 strain was administered to GF mice on the first day, and the anti-mouse IL-10R antibody (1 mg / individual) was intraperitoneally administered every week from the first day to the end of the experiment. The results of the mouse are shown in "GF + 35A1 + anti-IL10R". "GF” is E.I. The results of GF mice to which the colli 35A1 strain and the anti-mouse IL-10R antibody were not administered are shown, and "GF + 35A1" was not administered with the anti-mouse IL-10R antibody. The results of GF mice in which the colli 35A1 strain was established are shown, and "GF + anti-IL10R" is described in E. coli.
  • the results of the GF mouse to which the anti-mouse IL-10R antibody was administered without administering the coli 35A1 strain are shown. It is a graph which shows the result of having analyzed the histological Escherichia coli inflammation score in the proximal colon (Proximal colon) of a mouse.
  • the coli MG1655 strain was administered to GF mice on the first day, and the anti-mouse IL-10R antibody (1 mg / individual) was intraperitoneally administered every week from the first day to the end of the experiment.
  • mice The results of these mice are shown in "+ anti-IL10R + 35A1", “+ anti-IL10R + LF82" and “+ anti-IL10R + MG1655", respectively.
  • "GF” is E.I. The results of GF mice to which the colli strain and the anti-mouse IL-10R antibody were not administered are shown, and "GF + 35A1" was not administered with the anti-mouse IL-10R antibody. The results of GF mice in which the colli 35A1 strain was established are shown, and "+ anti-IL10R GF” is described in E. coli. The results of the GF mouse to which the anti-mouse IL-10R antibody was administered without the administration of the coli strain are shown. Each point represents an individual mouse.
  • Each thick bar represents the average of the group. Error bars represent SD. Each asterisk indicates that the values obtained by one-way ANOVA in combination with Tukey's post-test are * P ⁇ 0.05, ** P ⁇ 0.01, *** P ⁇ 0.001. .. It is a photograph showing a representative result of analysis of the distal colon of a mouse by hematoxylin and eosin staining. The notation in the figure is the same as in FIG. 11C. It is a graph which shows the result of having analyzed the histological Escherichia coli inflammation score in the distal colon of a mouse. The notation in the figure is the same as in FIG. 11D.
  • ⁇ Small intestinal bacteria of the present invention As shown in Examples below, nine strains of small intestinal bacteria isolated from patients with Crohn's disease were found to be significantly associated with Crohn's disease, and Th1 cells and / or Th17. It has become clear that it can induce proliferation or activation of cells (Th1 cells, etc.).
  • the present invention provides a small intestinal bacterium that induces proliferation or activation of Th1 cells and the like, and preferably has a DNA sequence encoding 16S rRNA (V1-V2 region) of these nine bacterial strains. It provides a bacterium having 95% or more identity.
  • small intestinal bacteria means bacteria that can colonize the human small intestine.
  • the site of the small intestine in which the bacterium is colonized is not particularly limited, and may be the ileum or the jejunum.
  • the tissue of the small intestine to which the bacteria colonize is not particularly limited, but is usually the mucous membrane of the small intestine.
  • Th1 cell is a subgroup of CD4 positive helper T cells (Th cells) and means a cell that enhances cell-mediated immunity.
  • Th1 cell activity refers to the production of Th1 cytokines (IFN- ⁇ , etc.) by the cells, the activation of cells such as macrophages and cytotoxic T cells (CTL) by the cytokines, and the cells due to the activation. It means that it includes enhancement of sexual immunity.
  • induction of proliferation or activation of Th1 cells means that induction of differentiation from naive T cells to Th1 cells leading to proliferation or activation of Th1 cells is also included. It also includes the accumulation (accumulation) of Th1 cells.
  • Th17 cells is a subgroup of CD4 positive helper T cells (Th cells) and means cells that induce inflammation.
  • the activity of Th17 cells means that the cells produce inflammatory cytokines (IL-17, IL-21, IL-22, TNF- ⁇ , etc.) and induce inflammation by the cytokines.
  • induction of Th17 cell proliferation or activation means to include induction of differentiation from naive T cells to Th17 cells, which leads to proliferation or activation of Th17 cells. It also includes the accumulation (accumulation) of Th17 cells.
  • the small intestinal bacterium of the present invention may be a single strain of bacteria or a mixture of bacterial strains composed of a plurality of strains of bacteria.
  • the bacterial strains When composed of a plurality of strains of bacteria, it is desirable that at least one of the bacterial strains has an activity of inducing proliferation or activation of Th1 cells or the like.
  • the plurality of strains of bacteria have an action of enhancing the activity of the bacterial strain having the inducing activity, even if the bacterial strain does not have the inducing activity.
  • Examples of the small intestinal bacterium of the present invention include a bacterium having a polynucleotide having 95% or more identity with respect to the DNA sequence set forth in any one of SEQ ID NOs: 1 to 9.
  • the DNA sequences shown in SEQ ID NOs: 1 to 1, respectively, are Eschericia coli (strain ID: 35A1), Ruminococcus gnavus (strain ID: 131A1), Bacteroides doriei (strain ID: 131H4), and Klebsiella, respectively, which are shown in Examples described later. pneumoniae (strain ID: 39F4), Streptococcus pasteurianus (strain ID: 133A7), Parabacteroides distasonis (strain ID: 134F1), Bacteroides fragilis (strain ID: 32E9), Erym : 131A11) 16SrRNA (V1-V2 region) encoding DNA sequence.
  • the "identity" with respect to the DNA sequence defined by each SEQ ID NO: is at least 95%, preferably 96% or more, more preferably 97% or more, still more preferably 98% or more, more preferably 99%. As mentioned above, it is particularly preferably 100%. Further, “identity” can be determined by using an alignment program such as BLAST. For example, nucleotide sequence identity includes identity between nucleotide sequences calculated using blastn, and more specifically, blastn with default parameters (ie, with default parameters). ) The calculated identity between the nucleotide sequences can be mentioned.
  • such a bacterium may be a bacterium having a nucleotide having at least 95% identity with respect to the DNA sequence defined by each SEQ ID NO:, and their species are not specified.
  • Bacteria having a polynucleotide having 95% or more identity with respect to the DNA sequence shown in SEQ ID NO: 1 are preferably bacteria belonging to Escherichia coli.
  • Bacteria having a polynucleotide having 95% or more identity with respect to the DNA sequence shown in SEQ ID NO: 2 are preferably bacteria belonging to Ruminococcus gnavus.
  • Bacteria having a polynucleotide having 95% or more identity with respect to the DNA sequence shown in SEQ ID NO: 3 are preferably bacteria belonging to Bacteroides doriei.
  • Bacteria having a polynucleotide having 95% or more identity with respect to the DNA sequence shown in SEQ ID NO: 4 are preferably bacteria belonging to Klebsiella pneumoniae.
  • Bacteria having a polynucleotide having 95% or more identity with respect to the DNA sequence shown in SEQ ID NO: 5 are preferably bacteria belonging to Streptococcus pasteurianus.
  • Bacteria having a polynucleotide having 95% or more identity with respect to the DNA sequence shown in SEQ ID NO: 6 are preferably bacteria belonging to Parabacteroides distasonis.
  • Bacteria having a polynucleotide having 95% or more identity with respect to the DNA sequence shown in SEQ ID NO: 7 are preferably bacteria belonging to Bacteroides fragilis.
  • Bacteria having a polynucleotide having 95% or more identity with respect to the DNA sequence shown in SEQ ID NO: 8 are preferably bacteria belonging to the Erysiperatoclastridium ramosom.
  • Bacteria having a polynucleotide having 95% or more identity with respect to the DNA sequence shown in SEQ ID NO: 9 are preferably bacteria belonging to Bacteroides uniformis.
  • the bacteria in the small intestine of the present invention may be at least one of the above nine types, but the proliferation or activation of Th1 cells and / or Th17 cells in the small intestine. From the viewpoint of being easier to induce, preferably with respect to a bacterium having a polynucleotide having 95% or more identity with respect to the DNA sequence set forth in SEQ ID NO: 1, and the DNA sequence set forth in SEQ ID NO: 2.
  • the small intestinal bacteria of the present invention may be 2 types, 3 types, 4 types, 5 types, 6 types, 7 types, 8 types or all combinations of the above 9 types, which will be described later.
  • the small intestinal bacteria of the present invention may be 2 types, 3 types, 4 types, 5 types, 6 types, 7 types, 8 types or all combinations of the above 9 types, which will be described later.
  • the same DNA sequence as shown in SEQ ID NO: 1 is used. It is a combination of a bacterium having a polynucleotide having sex and a bacterium having a polynucleotide having 95% or more identity with respect to the DNA sequence shown in SEQ ID NO: 2.
  • the present invention provides the first screening method shown below.
  • a method of screening for substances that suppress the proliferation or activation of Th1 cells and / or Th17 cells in the small intestine (1) A step of ingesting a test substance into a non-human germ-free animal, (2) When the number or activity of Th1 cells and / or Th17 cells in the small intestine of the non-human animal is detected, and when suppression of proliferation or activation of the cells is detected in (3) step (2).
  • a method including a step of determining that the test substance is a substance that suppresses the proliferation or activation of Th1 cells and / or Th17 cells in the small intestine.
  • the present inventors have clarified that the number or activity of Th1 cells and the like in the small intestine is enhanced by colonizing the above-mentioned small intestinal bacteria. ..
  • the present invention also provides the following second screening method.
  • the test substance is a bacterium having a polynucleotide having a polynucleotide having 95% or more identity with respect to the DNA sequence set forth in SEQ ID NO: 1 and / or 95% or more with respect to the DNA sequence set forth in SEQ ID NO: 2.
  • the test substance is Th1 cells in the small intestine. And / or a method comprising the step of determining that the substance suppresses the proliferation or activation of Th17 cells.
  • test substance used in these screening methods of the present invention is not particularly limited as long as it can contain a substance that suppresses the proliferation or activation of Th1 cells and / or Th17 cells in the small intestine, but for example. , Cells (bacteria, plant cells, animal cells, etc.), phages, and extracts, cultures (culture supernatants, etc.), secretory products, and metabolites thereof. Furthermore, it may be in the form of a library composed of a plurality of types of cells or phages.
  • synthetic small molecule compounds for example, synthetic small molecule compounds, antibodies, polypeptides, polynucleotides, lipids, sugars (monosaccharides, disaccharides, oligosaccharides, sugar chains, etc.), marine organisms, plant or animal-derived extracts, soil, and these substances.
  • synthetic small molecule compounds include a library composed of (chemical library, random peptide library, phage display library, etc.).
  • the "bacterium” used for the screening of the present invention is not particularly limited and may be a wild type or a mutant (genetical recombinant or the like), but it is established in the small intestine. Bacteria present in the small intestine of animals can be mentioned from the viewpoint that it is desirable to obtain them. Examples of such animals include humans and non-human animals (non-human animals) described below.
  • the test substance may be an isolated bacterium or a mixture of a plurality of types of bacteria. More specifically, the mixture includes a sample in the small intestine (for example, the contents of the small intestine of the animal, the small intestine lavage fluid of the animal, or a culture thereof).
  • the "bacteriophage” means a virus that infects and dissolves bacteria, and may be a lytic bacteriophage or a lysogenic bacteriophage, but a lytic bacteriophage is preferable. It is a phage. Further, it may be a mutant such as a genetically modified bacteriophage. Furthermore, the test bacteriophage may be an isolated bacteriophage or a mixture of multiple bacteriophages. More specifically, the mixture includes a sample in the small intestine (for example, the contents of the small intestine of the animal, the small intestine lavage fluid of the animal, or a culture thereof).
  • a phage against Escherichia coli is preferably used as a test bacteriophage. More specifically, T-type phages (T1, T2, T3, T4, T5, T6, T7, etc.), ⁇ X-174, ⁇ , ⁇ X80, Q ⁇ , P1 and the like can be mentioned.
  • a phage against Ruminococcus gnavus is preferably used as a test bacteriophage. More specifically, ⁇ Ra02, ⁇ Ra04 and the like can be mentioned.
  • Non-human germ-free animal to ingest the test substance means a non-human animal that is born and growing under sterile conditions.
  • Non-human animals include, but are not limited to, for example, mice, rats, monkeys, pigs, cows, horses, sheep, goats, chickens, ducks, ostriches, ducks, dogs, cats, rabbits, hamsters and the like. ..
  • a mouse is preferably used.
  • a non-human animal carrying the fungus may be used instead of the non-human germ-free animal.
  • Such non-human animals include, for example, specific pathogen-free (SPF) non-human animals.
  • a bacterium having a polynucleotide having a polynucleotide having 95% or more identity with respect to the DNA sequence set forth in SEQ ID NO: 1 and / or the DNA sequence set forth in SEQ ID NO: 2.
  • a non-human animal in which a bacterium having a polynucleotide having a polynucleotide having 95% or more of the same identity (hereinafter, also referred to as “a bacterium defined by SEQ ID NO: 1 etc.”) has been established is as shown in Examples described later.
  • it can be prepared by administering (for example, oral administration) the bacteria defined by SEQ ID NO: 1 and the like to the non-human germ-free animal and colonizing the animal.
  • step (1) there is no particular limitation on the method of "ingesting" the test substance to a non-human animal, and it is usually carried out by oral administration, but it is parenteral administration (for example, administration into the intestinal tract). May be good.
  • the non-human animal is a non-human animal in which the bacteria defined by SEQ ID NO: 1 etc. have been established, the ingestion of the test substance and the bacteria defined by SEQ ID NO: 1 etc. is simultaneous.
  • the test substance may be ingested by a non-human germ-free animal and then the animal specified by SEQ ID NO: 1 etc. may be ingested, and the bacteria defined by SEQ ID NO: 1 etc. may be ingested by non-human germ-free animals.
  • the test substance may be ingested by the animal after being ingested by the animal.
  • the "detection of the number of Th1 cells and / or Th17 cells" in the small intestine in the step (2) does not have to be the number (amount) of these cells itself, and reflects, for example, the amount (number) of the cells. This can be done by measuring the amount of cell-specific substances. Examples of such a substance include the cell-specific oligonucleotide (for example, a transcript specific to each helper T cell).
  • Th1 cell and / or Th17 cell activity can be performed by measuring the amount of cytokine produced by the bacterium.
  • cytokines produced by Th1 cells include Th1 cytokines such as IFN- ⁇ , and examples of cytokines produced by Th17 cells include inflammatory cytokines (IL-17, IL-21, IL-22, TNF-). ⁇ , etc.).
  • detection of the number of bacteria defined by SEQ ID NO: 1 etc. in the small intestine does not have to be the amount (number) of the bacteria itself, as in the detection of the cell number, for example, the amount of the bacteria.
  • This can be done by measuring the amount of the bacterium-specific substance that reflects the (number).
  • Such substances include the bacterium-specific oligonucleotide (for example, 16SrRNA gene and its transcript), the bacterium-specific constituent substance (peptides, nucleic acids, sugars, lipids, and complexes thereof that constitute the bacterium). Etc.), products of the bacterium (eg, secretory products of the bacterium, metabolites of the bacterium).
  • oligonucleotide for example, PCR (RT-PCR, real-time PCR, quantitative PCR), DNA microarray analysis method, Northern blotting, next-generation sequencing method (synthetic sequencing).
  • Method sequencing-by-synthesis, eg, sequencing by Illumina Solexa genome analyzer or Hiseq® 2000), pyrosequencing method (eg, sequencer GSLX or FLX by Roche Diagnostics (454)). Sequencing by (so-called 454 sequencing)), rigase reaction sequencing method (for example, SoliD® manufactured by Life Technology Co., Ltd. or sequencing by 5500 xl), T-RFLP method, bead array method, in sit hybridization. , Dot blot, RNase protection assay, mass analysis, genomic PCR, and southern blotting can be used for quantification.
  • antibodies such as ELISA method, immunoblotting, antibody array analysis method, immunohistochemical staining method, flow cytometry, imaging cytometry, radioimmunoassay, and immunoprecipitation method are used. It can be quantified using a method of detection (immunological method).
  • the value obtained by quantifying in this way may be a relative value as well as an absolute quantity.
  • the relative value include a quantitative ratio (so-called numerical value expressed in an arbitrary unit (AU)) based on the measuring method or measuring device used for detection.
  • the relative value may be, for example, a value (relative abundance, occupancy) indicating the proportion of the bacterium in the entire small intestinal bacterial flora.
  • a value calculated based on the amount of reference small intestinal bacteria may be used.
  • the "reference small intestinal bacterium” according to the present invention may be a bacterium that is stably present in the small intestine and has a small difference in amount between different biological samples.
  • the timing of detection is not particularly limited, and a person skilled in the art can appropriately adjust the timing according to the type of animal used.
  • “Inhibition” of proliferation or activation of Th1 cells or the like in step (3) means that these values in non-human animals fed with the test substance are compared with those in non-human animals not fed with the sample. This means that it is significantly reduced.
  • the "growth suppression" of the bacterium defined by SEQ ID NO: 1 etc. means that the number of bacteria defined by SEQ ID NO: 1 etc. in the non-human animal ingested the test substance ingests the sample. It means that it is significantly reduced compared to those in non-human animals that have not been fed.
  • Statistical analysis methods include, for example, t-test, Wilcoxon signed rank test, analysis of variance (ANOVA), Kruskal-Wallis test, Mann-Whitney U test (Wilcoxon rank sum test), odds ratio, hazard ratio, etc. Fisher's exact test, receiver operating characteristic analysis (ROC analysis), classification tree and decision tree analysis (CART analysis) can be mentioned. Normalized or standardized and normalized data can also be used for comparison.
  • the screening method of the present invention if it is not possible to select a bacterium or bacteriophage that suppresses the proliferation or activation of Th1 cells and / or Th17 cells in the small intestine by one execution, it is obtained.
  • the bacterium or bacteriophage can be isolated by ingesting a new non-human animal as the next test substance and performing the above-mentioned screening a plurality of times. can.
  • the present invention also provides a third screening method shown below, as shown in Examples described later.
  • a method of screening for bacteria that induce the proliferation or activation of Th1 cells and / or Th17 cells in the small intestine (1) The process of ingesting the test substance into non-human germ-free animals, (2) A step of detecting the number or activity of Th1 cells and / or Th17 cells in the small intestine of the non-human animal. (3) A method comprising a step of isolating bacteria from a sample in the small intestine of a non-human animal in which proliferation or activation of Th1 cells and / or Th17 cells was detected in step (2).
  • the present invention also provides a fourth screening method shown below.
  • a method of screening for bacteria that induce the proliferation or activation of Th1 cells and / or Th17 cells in the small intestine (1) A step of ingesting a physiologically active substance derived from a small intestinal bacterium that induces proliferation or activation of Th1 cells and / or Th17 cells into a non-human germ-free animal. (2) A step of detecting the number or activity of Th1 cells and / or Th17 cells in the small intestine of the non-human animal. (3) When proliferation or activation of Th1 cells and / or Th17 cells is detected in step (2), the physiologically active substance is used to proliferate or activate Th1 cells and / or Th17 cells in the small intestine. A method comprising a step of determining that the substance is a physiologically active substance to be induced.
  • test substance "non-human germ-free animal”, “ingestion”, “detection of helper T cell proliferation (number) or activity” and “isolation of bacteria” in the third and fourth screening methods. Is the same as the first and second screening methods described above. Further, the “physiologically active substance” is as described later.
  • the small intestinal bacterium of the present invention is associated with the etiology of diseases caused by Th1 cells and the like, such as Crohn's disease. Therefore, the present invention A method for assessing diseases caused by Th1 cells and / or Th17 cells.
  • the present invention A method for assessing diseases caused by Th1 cells and / or Th17 cells.
  • Quantifying bacteria with polynucleotides having 95% or more identity (2) A step of comparing the value obtained by quantifying in step (1) with a corresponding value obtained by quantifying the bacteria in the mucosa of the small intestine of a human who does not suffer from the disease, and step (3) (3).
  • the "disease caused by Th1 cells and / or Th17 cells” means a disease induced by proliferation or activation of Th1 cells and / or Th17 cells, and is an inflammatory bowel disease (Clone's disease (CD). ), Chronic inflammatory bowel diseases such as ulcerative bowel disease and inflammatory bowel disease), type 1 diabetes, rheumatoid arthritis, experimental immune encephalitis (EAE), multiple sclerosis, autoimmune diseases such as systemic erythematosus, Chronic inflammatory diseases can be mentioned, and among them, Crohn's disease is a suitable subject of the present invention.
  • CD inflammatory bowel disease
  • Chronic inflammatory bowel diseases such as ulcerative bowel disease and inflammatory bowel disease
  • type 1 diabetes rheumatoid arthritis
  • EAE experimental immune encephalitis
  • multiple sclerosis autoimmune diseases such as systemic erythematosus
  • Chronic inflammatory diseases can be mentioned, and among them, Crohn's disease is a suitable subject of the present invention.
  • Crohn's disease is classified into small intestine type, small intestine large intestine type, and large intestine type according to the site where the lesion is present, but in the present invention, small intestine type Crohn's disease can be a more suitable target.
  • Examples of the "subject" in the evaluation method of the present invention include humans suspected of having or recurring a disease caused by Th1 cells and / or Th17 cells.
  • step (1) The quantification of small intestinal bacteria of the present invention in step (1) can be carried out in the same manner as in step (2) of the above-mentioned "screening method".
  • the bacteria can be collected from the mucosa of the small intestine using, for example, a double-balloon endoscopy (DBE) system as shown in Examples described later.
  • DBE double-balloon endoscopy
  • the bacterium to be quantified in the step (1) is a bacterium having a polynucleotide having 95% or more identity with respect to the DNA sequence set forth in SEQ ID NO: 1 and / or the bacterium described in SEQ ID NO: 2.
  • it is a bacterium having a polynucleotide having 95% or more identity with respect to the DNA sequence, it may be at least one of the nine small intestinal bacteria of the present invention.
  • the “human who does not suffer from a disease induced by proliferation or activation of Th1 cells and / or Th17 cells” to be compared in step (2) is not particularly limited as long as it does not suffer from the disease. , Age, gender, nationality and medical history are preferably consistent with the subject.
  • “comparison” with the corresponding value obtained by quantifying the bacterium in the small intestinal mucosa of a human not suffering from the disease can be performed by a person skilled in the art in the same manner as in step (3) of the above-mentioned "screening method". For example, it can be appropriately performed based on the above statistical analysis method.
  • the quantitative value obtained in the step (1) is higher than the corresponding value, and the difference is considered to be statistically significant (for example, P ⁇ 0.05).
  • the quantitative value may be twice or more (preferably 5 times or more and 10 times or more) the corresponding value.
  • the “determination” in step (3) includes not only the presence or absence of the onset of the disease but also the risk of its onset. That is, when the quantitative value in the small intestinal mucosa of the subject is higher than the corresponding value, it is determined that the subject has developed the disease or is at high risk of developing the disease.
  • the method of the present invention includes a method of collecting data on the above-mentioned quantitative values for diagnosis by a doctor, a method of presenting the data to a doctor, a method of comparing and analyzing the quantitative values and corresponding values, and a method of comparing and analyzing the corresponding values of a patient. It can also be described as a method for assisting a doctor's diagnosis in consideration of clinical symptoms and / or other test results.
  • the present invention provides a composition used in the above-mentioned inspection method. More specifically, the test composition of the following aspects is provided.
  • a composition for examining a disease caused by Th1 cells and / or Th17 cells which comprises an antibody that specifically recognizes the small intestinal bacteria of the present invention.
  • a composition for examining a disease caused by Th1 cells and / or Th17 cells which comprises a polynucleotide for detecting a nucleotide sequence specific to the small intestinal bacterium of the present invention.
  • the "antibody that specifically recognizes the small intestinal bacterium of the present invention” may be a polyclonal antibody, a monoclonal antibody, or an antibody as long as the bacterium can be specifically recognized.
  • Functional fragments eg, Fab, Fab', F (ab') 2, variable region fragment (Fv), disulfide-bound Fv, single-stranded Fv (scFv), sc (Fv) 2, diabodies, multispecific antibodies , Or a polymer of these).
  • an immune animal is immunized with an antigen (polypeptide, polynucleotide, sugar chain, lipid, etc.
  • the monoclonal antibody can be produced by a hybridoma method or a recombinant DNA method.
  • an antibody to which a labeling substance is bound can be used.
  • detecting the labeling substance it is possible to directly measure the amount of antibody bound to the small intestinal bacterium of the present invention or a substance derived from the bacterium.
  • the labeling substance is not particularly limited as long as it can bind to an antibody and can be detected by a chemical or optical method.
  • a fluorescent dye (GFP or the like), an enzyme (HRP or the like), or a radioactive substance is used. Substances are mentioned.
  • the test composition of the present invention may contain other components permitted as a composition in addition to the antibody component.
  • Such other components include, for example, carriers, excipients, disintegrants, buffers, emulsifiers, suspensions, stabilizers, preservatives, preservatives, physiological salts, labeling substances, secondary antibodies. ..
  • a labeled substance for example, secondary antibody, protein G, protein A, etc.
  • the kit for testing for diseases caused by Th1 cells and the like can include an instruction manual for the kit.
  • test composition of the present invention can be combined with an apparatus for detecting the antibody of the present invention.
  • a device include a flow cytometry device and a microplate reader.
  • the "polynucleotide for detecting the nucleotide sequence specific to the bacterium of the present invention is not particularly limited as long as the sequence specific to the bacterium is detected, and for example, at least 15 nucleotides.
  • the polynucleotide of the present invention has a base sequence complementary to the nucleotide sequence of the small intestinal bacterium of the present invention.
  • “complementary” does not have to be completely complementary as long as it hybridizes.
  • These polynucleotides are usually 80% or more, preferably 90% or more, more preferably 95% or more (for example, 96% or more, 97% or more, 98% or more, 99% or more) with respect to the nucleotide sequence. Particularly preferably, it has 100% homology.
  • the "chain length" of the polynucleotide of the present invention is usually 15 to 100 nucleotides, preferably 17 to 30 nucleotides, and more preferably 20 to 25 nucleotides.
  • the "chain length" of the polynucleotide of the present invention is usually 15 to 1000 nucleotides, preferably 20 to 100 nucleotides.
  • the polynucleotide of the present invention may be DNA or RNA, and in part or all thereof, LNA (registered trademark, crosslinked nucleic acid), ENA (registered trademark, 2'-O, 4'- Nucleotides may be substituted with artificial nucleic acids such as C-Ethylene-bridged nucleic acids), GNA (glycerol nucleic acids), TNAs (treose nucleic acids), and PNAs (peptide nucleic acids).
  • LNA registered trademark, crosslinked nucleic acid
  • ENA registered trademark, 2'-O, 4'- Nucleotides
  • artificial nucleic acids such as C-Ethylene-bridged nucleic acids
  • GNA glycerol nucleic acids
  • TNAs treose nucleic acids
  • PNAs peptide nucleic acids
  • the polynucleotide of the present invention can be chemically synthesized using a commercially available nucleotide automatic synthesizer or the like. Further, as the polynucleotide used for the test of the present invention, a polynucleotide to which a labeling substance is bound can be used.
  • the labeling substance is not particularly limited as long as it can bind to a polynucleotide and can be detected by a chemical or optical method.
  • a fluorescent dye (DEAC, FITC, R6G, TexRed, Cy5, etc.)
  • Dyes such as DAB (chromogen)
  • enzymes radioactive substances in addition to fluorescent dyes.
  • test composition of the present invention may contain other pharmacologically acceptable components in addition to the above-mentioned polynucleotide.
  • examples of such other components include buffers, emulsifiers, suspending agents, stabilizers, preservatives, physiological salts and the like.
  • a sample and the present invention are combined by combining a substrate necessary for detecting a labeling substance added to a polynucleotide, a positive control or a negative control, and a buffer solution used for diluting or washing the sample.
  • a tube or plate used for the reaction with the polynucleotide of No. 1 can be combined, and a kit for testing a disease caused by Th1 cells can also be used.
  • the kit for testing for diseases caused by such Th1 cells can include instructions for use of the kit.
  • test composition of the present invention can be combined with an apparatus for detecting a nucleotide sequence specific to the small intestinal bacterium of the present invention.
  • a device for detecting a nucleotide sequence specific to the small intestinal bacterium of the present invention examples include a thermal cycler, a sequencer, and a microarray.
  • the present invention is a composition for inducing proliferation or activation of Th1 cells and / or Th17 cells, which comprises the small intestinal bacterium of the present invention or a physiologically active substance derived from the bacterium as an active ingredient, or a method thereof. Can be provided.
  • the small intestinal bacterium of the present invention which can be a pathogen of the disease, has an immunostimulatory action.
  • Th1 cells are CD4-positive helper T cells that produce IFN- ⁇ , play an important role in defense against pathogen infections such as Mycobacterium tuberculosis and Listeria monocytogenes, and monitor and eliminate cancerous cells. It also plays an important role in.
  • the presence of bacteria in the intestinal tract such as Bacteroides and Bifidobacterium and the induction of Th1 cells by such bacteria have been one of the immunotherapies for cancer, such as immune checkpoint inhibitors (anti-CTLA-4 antibody and anti-PD-L1).
  • the present invention can provide a composition for activating immunity or a method thereof.
  • the "immunity" activated or suppressed in the present invention includes not only mucosal immunity (intestinal immunity, etc.) but also systemic immunity. Moreover, not only cell-mediated immunity but also humoral immunity is included.
  • the "small intestinal bacterium of the present invention” contained as an active ingredient in the composition of the present invention is as described in the above ⁇ small intestinal bacterium of the present invention>, but it may be a live bacterium or a dead bacterium. It may be.
  • Examples of the "killed cell” include a heat-treated body, a fire-treated body, a steamed body, a radiation ( ⁇ -ray, etc.) irradiation-treated body, an enzyme-treated body, a drug (antibiotic, etc.) treated body, and a chemical substance (formaline, etc.). )
  • a processed body and an ultrasonic processed body can be mentioned.
  • the form of the small intestinal bacterium of the present invention is not particularly limited and may be either liquid or solid, but the form is in a dry form (for example, powder form, bacterial powder form, etc.) during production or storage. It is suitable from the viewpoint of easy handling.
  • the dried product can be prepared by drying a suspension in which the cells are dispersed in a solvent (water or the like).
  • the drying method is not particularly limited, and examples thereof include a spray drying method and a freeze drying method.
  • the sterilization method is not particularly limited, and the retort sterilization method, UHT sterilization method, air sterilization method, pressure sterilization method, high pressure steam sterilization method, dry heat sterilization method, distribution steam sterilization method, electromagnetic wave sterilization method, electron beam sterilization method, High-frequency sterilization method, radiation sterilization method, ultraviolet sterilization method, ethylene oxide gas sterilization method, hydrogen peroxide plasma sterilization method, chemical sterilization method (alcohol sterilization method, formalin fixation method, electrolyzed water treatment method) and the like can be mentioned.
  • surfactant treatment, grinding / crushing treatment, enzyme treatment, fractionation treatment, etc. can be performed before and after the sterilization treatment by heating or the like, or before and after the drying treatment.
  • surfactant treatment, grinding / crushing treatment, enzyme treatment, fractionation treatment, etc. can be performed.
  • the obtained product can also be contained in the composition of the present invention.
  • the bacterium may be modified (gene modification, etc.) according to the use of the contained active ingredient.
  • modifications are not particularly limited, and when the composition of the present invention is a vaccine adjuvant described later, modifications such that the action of inducing proliferation or activation of Th1 cells or the like is enhanced can be mentioned.
  • the active ingredient contained in the composition of the present invention may be a culture of small intestinal bacteria of the present invention.
  • the culture may be one containing the bacterium (culture solution containing the grown bacterium, solid medium, etc.), and the form of the culture is not particularly limited and may be either liquid or solid.
  • a dry form (for example, a dried culture product) is suitable for this form from the viewpoint of easy handling during production and storage. Those skilled in the art can appropriately prepare such a dried culture product or the like by using the above-mentioned drying method.
  • composition of the present invention may contain one strain of the small intestinal bacterium of the present invention, but may contain a plurality of types of strains.
  • the compositions can be used in combination, and as a result, when ingested or absorbed in combination (in the case of a combination composition), the plurality of strains can be present in two or more kinds of compositions. (For example, they can be in separate compositions).
  • the "physiologically active substance” is not particularly limited, but includes a substance contained in the bacterium, a secreted product of the bacterium, and a metabolite of the bacterium, and more specifically, the bacterium or the metabolite. Examples thereof include a polypeptide fraction, a polynucleotide fraction, a sugar chain fraction, a lipid fraction, and a low molecular weight metabolite fraction of the culture supernatant.
  • Such a physiologically active substance can be obtained from, for example, the above-mentioned screening method from the small intestinal bacterium of the present invention, the culture supernatant thereof, the small intestinal sample of a non-human animal in which the bacterium has settled, the human small intestine sample, and the like. As described above, it is possible to identify by purifying the active ingredient using the proliferation or activation of Th1 cells or the like as an index.
  • composition of the present invention can be in the form of a pharmaceutical composition, food or drink (including animal feed), or a reagent used for research purposes (eg, in vitro or in vivo experiments).
  • the active ingredient of the composition of the present invention induces the proliferation or activation of Th1 cells and the like, and activates immunity.
  • Pharmaceutical composition for treatment, prevention or improvement of infectious diseases such as tuberculosis pharmaceutical composition for enhancing anticancer activity as food and drink, and in combination with anticancer agent or immune checkpoint inhibitor , As a food and drink, and also as a pharmaceutical composition (vaccine adjuvant) for enhancing its immune response action when used in combination with a vaccine.
  • “treatment” also includes assistance for treatment.
  • composition of the present invention can be formulated by a known pharmaceutical method.
  • a known pharmaceutical method for example, capsules, tablets, pills, liquids, powders, granules, fine granules, film coatings, pellets, lozenges, sublinguals, chewing agents, buccal agents, pastes, syrups, suspensions, Oral and parenteral (eg, intestinal tract) as elixirs, emulsions, coatings, ointments, plasters, paps, transdermal formulations, lotions, inhalants, aerosols, injections, suppositories, etc.
  • carriers that are pharmacologically or acceptable as foods and drinks, specifically, sterile water, physiological saline, vegetable oils, solvents, bases, emulsifiers, suspending agents, surfactants, stabilizers, etc.
  • the composition of the present invention is used in the small intestine. It may be combined with a composition that enables efficient delivery to the inside and the like.
  • the composition capable of such delivery into the small intestine or the like is not particularly limited, and a known composition can be appropriately adopted, for example, a pH-sensitive composition, suppression of release to the small intestine or the like.
  • compositions cellulose-based polymers, acrylic acid polymers and copolymers, vinyl acid polymers and copolymers, etc.
  • bioadhesive compositions that specifically adhere to the small intestinal mucosa
  • protease inhibitor-containing compositions in the small intestine Examples thereof include compositions that are specifically degraded by an enzyme.
  • composition that induces proliferation or activation of Th1 cells or the like of the present invention is used as a pharmaceutical composition
  • a known substance antiviral agent, antiviral agent, used for treatment, prevention or amelioration of infectious diseases and cancers, etc.
  • Antibacterial agents, anticancer agents, immune checkpoint inhibitors, etc. may be further contained, or may be used in combination with such substances.
  • a vaccine adjuvant in addition to the antigen that is the active ingredient of the vaccine (for example, a bacterial or virus-specific antigen, a cancer-specific antigen), other known vaccine adjuvants and immunopotentiators should be used.
  • the compositions of the invention may be included or may be used in combination with those substances.
  • the food or drink may be, for example, a health food, a functional food, a food for specified health use, a food with a nutritional function, a food with a functional claim, a nutritional supplement, a food for the sick, or It can be an animal feed.
  • Functional foods are usually classified into four categories, probiotics, biogenics, prebiotics, and symbiotics, based on their mechanism of action. In the present invention, probiotics, biogenics, and symbiotics are classified. It can take the form of tics or prebiotics.
  • foods and drinks include fermented beverages, oil-containing products, soups, dairy beverages, soft drinks, tea beverages, alcoholic beverages, drinks, jelly-like beverages and other liquid foods; carbohydrate-containing foods; processed livestock foods. Processed marine foods; processed vegetable foods; semi-solid foods; fermented foods; confectionery; retort products; microwave-compatible foods and the like. Further, healthy foods and drinks prepared in the form of powder, granules, tablets, capsules, liquid, paste or jelly can also be mentioned.
  • the food and drink according to the present invention can be produced by a production technique known in the art. In the food and drink, ingredients (for example, nutrients, etc.) effective for improving or preventing infectious diseases and cancer may be added. In addition, it may be a multifunctional food or drink by combining it with other ingredients or other functional foods that exhibit functions other than the improvement.
  • compositions of the present invention induce the proliferation or activation of Th1 cells and the like, or treat, improve or prevent diseases such as infectious diseases and cancer. It may be labeled as being used for this purpose.
  • the act of "displaying” includes all acts that inform the consumer of the use, and constitutes an expression that can directly recognize the use or an expression that can recall or infer the use. It may be attached to the product itself, or it may be attached to the container, packaging material or package insert containing the composition. Further, “display” refers to information related to the composition of the present invention, such as leaflets, pamphlets, pops, catalogs, posters, books, storage media such as DVDs, advertisements such as electronic bulletin boards and the Internet, and the like. It may be something that displays / advertises that it is effective.
  • composition of the present invention may be in the form of a kit.
  • kits include, for example, the small intestinal bacterium of the present invention or a physiologically active substance derived from the bacterium, an antiviral agent, an antibacterial agent, an anticancer agent, an immune checkpoint inhibitor, an antigen, another vaccine adjuvant, and the like.
  • an antiviral agent for example, an antiviral agent, an antibacterial agent, an anticancer agent, an immune checkpoint inhibitor, an antigen, another vaccine adjuvant, and the like.
  • the present invention requires that a composition for inducing proliferation or activation of Th1 cells or the like, or an active ingredient thereof, the small intestinal bacterium of the present invention or a physiologically active substance derived from the bacterium, be ingested. It also provides a method of inducing proliferation or activation of Th1 cells or Th17 cells in a subject, or a method of activating immunity in the subject.
  • composition of the present invention or an active ingredient thereof can be usually used for humans, but may be animals other than humans, and various livestock, poultry, pets, laboratory animals and the like are targeted. be able to.
  • an ingestion target of the composition for inducing the proliferation or activation of Th1 cells and the like of the present invention or the active ingredient thereof
  • animals infected with viruses, bacteria, etc. regardless of their onset, are used.
  • an animal that has not been infected with a virus, a bacterium, or the like or is suspected of being infected with the virus may be allowed to ingest the composition or the like of the present invention.
  • the composition of the present invention can be suitably used for animals that carry viruses, bacteria, etc. that do not have the symptom.
  • the composition of the present invention can be suitably used not only for animals suffering from cancer, but also for animals suspected of suffering from cancer and animals after anticancer therapy.
  • the method of ingesting the composition or the like of the present invention is not particularly limited and may be oral administration or parenteral administration (for example, administration into the small intestine), but in the case of oral administration.
  • the subject of ingestion of the composition of the present invention should reduce the production of gastric acid by ingesting a proton pump inhibitor (PPI) or the like. , Or it is preferable to take antibiotics.
  • PPI proton pump inhibitor
  • the ingestion amount depends on the age, weight, disease symptom, health condition, type of composition (pharmaceuticals, foods and drinks, etc.), intake method, etc. of the subject. Those skilled in the art can appropriately select it.
  • the small intestinal bacterium of the present invention induces a disease caused by Th1 cells or the like such as Crohn's disease by inducing proliferation or activation of Th1 cells or the like. Therefore, the disease can be treated, ameliorated or prevented by removing the bacteria from the small intestine or the like by inducing an immune response by targeting the bacteria in the small intestine of the present invention.
  • the present invention can provide a small intestinal bacterium of the present invention, a vaccine composition containing an antigen specific to the bacterium as an active ingredient, or a method for inducing an immune response against the bacterium.
  • the "small intestinal bacterium of the present invention” contained as an active ingredient in the composition of the present invention is as described above, and may be a live bacterium or a dead bacterium (for the dead bacterium). As mentioned above).
  • the small intestinal bacterium of the present invention may be a bacterium that has been modified (gene modification or the like) depending on the use of the active ingredient contained therein. The modification is not particularly limited, and when the composition of the present invention is a vaccine composition, the action of inducing the proliferation or activation of Th1 cells or the like in the small intestine or the like is suppressed so as not to induce inflammation or the like. Modifications can be mentioned.
  • the active ingredient contained in the composition of the present invention may be a culture of the bacteria in the small intestine of the present invention (the culture is as described above).
  • the composition of the present invention may contain at least one small intestinal bacterium of the present invention, but a plurality of strains (for example, 2 types, 3 types, 4 types, 5 types, 6 types, 7 types). , 8 types or 9 types).
  • the compositions can be used in combination, and as a result, when ingested or absorbed in combination (in the case of a combination composition), the plurality of strains can be present in two or more kinds of compositions. ..
  • the "antigen specific to the small intestinal bacterium of the present invention” is a substance (polypeptide, polynucleotide, sugar chain, lipid, etc.) contained in the bacterium, and is antigenic or immunogenic. It means a substance to have.
  • immunogenicity means that a primary immune response or a memory immune response can be activated.
  • the “immune response” includes responses of CD4 positive (CD4 +) T lymphocytes, CD8 positive (CD8 +) T lymphocytes and B lymphocytes.
  • T lymphocytes such responses are proliferative and / or cytokines (eg, IL-2, IL-3, IL-4, IL-5, IL-6, IL-12, IL-13, IL. -15, TNF- ⁇ , IFN- ⁇ ) production type may be used.
  • these responses may result in the production of cytotoxic T lymphocytes (CTL).
  • CTL cytotoxic T lymphocytes
  • the B lymphocyte reaction may result in antibody production by the reacting B lymphocytes.
  • "antigenic" means that it can be recognized by an antibody molecule or an antigen-specific T cell receptor (TCR) on activated effector T cells (eg, cytokine-producing T cells, CTL, etc.). ..
  • the "antigen specific to the small intestinal bacterium of the present invention” is a substance that can be recognized and bound to the antibody that specifically recognizes the bacterium or the like, or an appropriate antigen-presenting cell (APC). ), And after binding to a suitable major histocompatibility complex (MHC) molecule, contains a substance that can be recognized and bound to the TCR on the effector T cells induced in response to the bacteria or the like. Means.
  • MHC major histocompatibility complex
  • such a bacterial-specific antigen can be obtained by a person skilled in the art using a known screening method (for example, Paul WE editing, Fundamental Immunology) using the reactivity with the antigen-specific antiserum and / or T lymphocyte as an index. , 1993, 3rd edition, pp. 243-247, Harlow and Lane, Antibodies, A Laboratory Manual, Cold Spring Harbor Laboratory, 1998).
  • the amino acid sequence of the bacterial-specific antigen (polypeptide) is analyzed using a computer program (for example, MHC-THREAD, EpiPredict, HLA-DR4 binding, ProPred, BIMAS, SVMHC, NetMHC, PREDICT, LpPep, SYFPEITHI, RankPe. ) Can also be inferred.
  • the vaccine composition of the present invention can be formulated by a known pharmaceutical method.
  • inhalants aerosols, injections, powders, granules, fine granules, liquids, capsules, tablets, pills, film coatings, pellets, lozenges, sublinguals, chewing agents, buccal agents, pastes.
  • Oral and parenteral eg, intestinal tract as agents, syrups, suspensions, elixirs, emulsions, coatings, ointments, ointments, paps, transdermal formulations, lotions, suppositories, etc.
  • carriers that are pharmacologically or acceptable as foods and drinks, specifically, sterile water, physiological saline, vegetable oils, solvents, bases, emulsifiers, suspending agents, surfactants, stabilizers, etc.
  • the vaccine composition of the present invention may contain a known vaccine adjuvant and immunopotentiator.
  • the vaccine adjuvant include aluminum hydroxide, KLH, MPL, QS21, complete Freund's adjuvant, incomplete Freund's adjuvant, aluminum phosphate, BCG, myoban, TLR agonist such as CpG DNA, or a combination thereof.
  • an auxiliary agent such as albumin, a wetting agent, and an emulsifier may be added.
  • examples of the immunopotentiator include various cytokines (for example, IL-12, IL-18, GM-CSF, IFN- ⁇ , IFN- ⁇ , IFN- ⁇ , Flt3 ligand).
  • the product (pharmaceutical product, vaccine) of the composition of the present invention or its description induces an immune response against a bacterium that induces proliferation or activation of Th1 cells or the like in the small intestine or the like, and treats a disease caused by Th1 cells or the like.
  • It may be labeled as being used for improvement or prevention.
  • "marked on a product or instruction manual” means that a label is attached to the main body, container, packaging, etc. of the product, or a manual, package insert, advertisement, or other printed matter that discloses product information. It means that the display is attached to.
  • composition of the present invention may be in the form of a kit.
  • a kit for example, the small intestinal bacterium of the present invention or an antigen, a vaccine adjuvant, an immunopotentiator or the like specific to the bacterium usually exists as two or more kinds of substances (compositions and the like). Examples thereof include an embodiment in which one composition can be prepared by mixing or the like before ingesting the subject.
  • the present invention also comprises ingesting a vaccine composition, a bacterium as an active ingredient thereof, or an antigen specific to the bacterium into a subject, or a method for inducing an immune response against the bacterium in the subject. It also provides a method for treating, ameliorating or preventing a disease caused by Th1 cells or the like in a subject.
  • composition of the present invention or an active ingredient thereof can be used for animals including humans, but the animals other than humans are not particularly limited, and various livestock, poultry, pets, laboratory animals and the like are targeted. Can be.
  • a subject determined to be suffering from the disease by the above-mentioned test method of the present invention may be a suitable target.
  • the vaccine composition of the present invention or an active ingredient thereof is a bacterium that induces proliferation or activation of Th1 cells or the like in the small intestine or the like regardless of the onset of a disease caused by Th1 cells or the like.
  • examples include animals that possess. From the viewpoint of prevention, an animal that does not possess or is suspected of possessing the bacterium may be allowed to ingest the composition of the present invention or the like.
  • the method of ingesting the composition or the like of the present invention is not particularly limited, and may be oral administration or parenteral administration.
  • the amount of the ingestion may be appropriately determined by those skilled in the art depending on the age, body weight, symptoms of the disease, health condition, dosage form of the composition, ingestion method, etc. You can choose.
  • composition for suppressing proliferation or activation of Th1 cells and the like of the present invention The small intestinal bacterium of the present invention induces proliferation or activation of Th1 cells and the like, and enhances the immune action, thereby inducing diseases caused by Th1 cells and the like such as Crohn's disease. Therefore, if the bacterium is removed from the small intestine or the like, the induction of Th1 cells or the like is suppressed, and the immune action is suppressed, which leads to the treatment of the disease or the like.
  • the present invention is a composition for suppressing proliferation or activation of Th1 cells and / or Th17 cells, which contains a substance having an antibacterial effect against small intestinal bacteria of the present invention as an active ingredient, and suppresses immunity.
  • a composition for treating, ameliorating or preventing a disease caused by Th1 cells and / or Th17 cells is provided.
  • treatment means complete recovery from the disease
  • improvement includes alleviation or improvement of the symptoms of the disease, suppression of its progression, and suppression of its recurrence.
  • prevention includes suppression of the onset of the disease, delay, and suppression of its recurrence.
  • the "antibacterial action” means an action of suppressing the growth of bacteria and / or a bactericidal action.
  • the "substance having an antibacterial action against the bacteria in the small intestine of the present invention” contained as an active ingredient in the composition of the present invention is not particularly limited as long as it has the action, but for example, an antibiotic or a lytic substance ( Bacteriophage, lytic enzyme, etc.), antibodies that specifically recognize the bacterium, and the like. Further, it may be a substance that binds to a physiologically active substance derived from the small intestinal bacterium of the present invention, and examples of such a substance include an antibody that binds to the physiologically active substance and a low molecular weight compound that binds to the physiologically active substance. Can be mentioned. Furthermore, it may be a substance selected by the screening method of the present invention described above.
  • the composition of the present invention may contain a plurality of types of substances having an antibacterial action against the small intestinal bacteria of the present invention.
  • the compositions can be used in combination, and as a result, when ingested or absorbed in combination (in the case of a combination composition), the plurality of substances can be present in two or more kinds of compositions. ..
  • composition of the present invention can be in the form of a pharmaceutical composition, food or drink (including animal feed), or a reagent used for research purposes (eg, in vitro or in vivo experiments). Further, the composition of the present invention may be in the form of a kit, if necessary.
  • the composition of the present invention is a pharmaceutical composition for treating, preventing or improving the above-mentioned diseases caused by Th1 cells and the like, eating and drinking. It is preferably used as a product.
  • composition of the present invention can be formulated by a known pharmaceutical method.
  • a known pharmaceutical method for example, capsules, tablets, pills, liquids, powders, granules, fine granules, film coatings, pellets, lozenges, sublinguals, chewing agents, buccal agents, pastes, syrups, suspensions, Oral and parenteral (eg, intestinal tract) as elixirs, emulsions, coatings, ointments, plasters, paps, transdermal formulations, lotions, inhalants, aerosols, injections, suppositories, etc.
  • carriers that are pharmacologically or acceptable as foods and drinks, specifically, sterile water, physiological saline, vegetable oils, solvents, bases, emulsifiers, suspending agents, surfactants, stabilizers, etc.
  • the composition of the present invention is used in the small intestine. It may be combined with a composition that enables efficient delivery to the inside and the like.
  • the composition capable of such delivery into the small intestine or the like is not particularly limited, and a known composition can be appropriately adopted, for example, a pH-sensitive composition, suppression of release to the small intestine or the like.
  • compositions cellulose-based polymers, acrylic acid polymers and copolymers, vinyl acid polymers and copolymers, etc.
  • bioadhesive compositions that specifically adhere to the small intestinal mucosa
  • protease inhibitor-containing compositions in the small intestine Examples thereof include compositions that are specifically degraded by an enzyme.
  • composition that suppresses the proliferation or activation or immunity of Th1 cells or the like of the present invention is used as a pharmaceutical composition
  • a known substance used for the treatment, prevention or improvement of diseases caused by Th1 cells or the like for example, an anti-inflammatory agent, an immunosuppressant
  • an anti-inflammatory agent, an immunosuppressant may be further contained, or may be used in combination with such a substance.
  • the food or drink may be, for example, a health food, a functional food, a food for specified health use, a food with a nutritional function, a food with a functional claim, a nutritional supplement, a food for the sick, or It can be an animal feed.
  • Functional foods are usually classified into four categories, probiotics, biogenics, prebiotics, and symbiotics, based on their mechanism of action. In the present invention, probiotics, biogenics, and symbiotics are classified. It can take the form of tics or prebiotics.
  • foods and drinks include fermented beverages, oil-containing products, soups, dairy beverages, soft drinks, tea beverages, alcoholic beverages, drinks, jelly-like beverages and other liquid foods; carbohydrate-containing foods; livestock. Processed foods; processed marine products; processed vegetable foods; semi-solid foods; fermented foods; confectionery; retort products; microwave-compatible foods and the like. Further, healthy foods and drinks prepared in the form of powder, granules, tablets, capsules, liquid, paste or jelly can also be mentioned.
  • the food and drink according to the present invention can be produced by a production technique known in the art.
  • an ingredient for example, nutrients, etc.
  • it may be a multifunctional food or drink by combining it with other ingredients or other functional foods that exhibit functions other than the improvement.
  • the product (pharmaceutical product, food or drink, reagent) of the composition of the present invention or a description thereof suppresses the proliferation or activation of Th1 cells or the like, suppresses immunity, or treats or improves a disease caused by Th1 cells or the like. Alternatively, it may be labeled as being used for prevention.
  • the act of "displaying” includes all acts that inform the consumer of the use, and constitutes an expression that can directly recognize the use or an expression that can recall or infer the use. It may be attached to the product itself, or it may be attached to the container, packaging material or package insert containing the composition. Further, “display” refers to information related to the composition of the present invention, such as leaflets, pamphlets, pops, catalogs, posters, books, storage media such as DVDs, advertisements such as electronic bulletin boards and the Internet, and the like. It may be something that displays / advertises that it is effective.
  • the present invention has an antibacterial action against the composition for suppressing the proliferation or activation of Th1 cells and the like, the composition for suppressing immunity, or the small intestinal bacterium of the present invention which is an active ingredient thereof.
  • composition of the present invention or an active ingredient thereof can be used for animals including humans, but the animals other than humans are not particularly limited, and various livestock, poultry, pets, laboratory animals and the like are targeted. Can be.
  • the subject according to the present invention may be a person suffering from a disease caused by Th1 cells or the like, but it is not necessary to be diagnosed with these diseases, for example, a person who may have suffered from these diseases, these. Some people feel that they have a disease. Furthermore, even a healthy person can take it on a daily basis for the purpose of preventing these diseases.
  • a subject determined to be suffering from the disease by the above-mentioned test method of the present invention may be a suitable target.
  • the method of ingesting the composition or the like of the present invention is not particularly limited and may be oral administration or parenteral administration (for example, administration into the small intestine), but in the case of oral administration.
  • the subject of ingestion of the composition of the present invention should reduce the production of gastric acid by ingesting a proton pump inhibitor (PPI) or the like. Is preferable.
  • PPI proton pump inhibitor
  • the ingestion amount depends on the age, weight, disease symptom, health condition, type of composition (pharmaceuticals, foods and drinks, etc.), intake method, etc. of the subject. Those skilled in the art can appropriately select it.
  • SI mucosal samples were biopsied or biopsied from 27 Crohn's disease (CD) patients and 17 non-CD patients who received ongoing clinical care using a double-balloon endoscopy (DBE) system. Collected by scraping. Patients who underwent retrograde endoscopy underwent intestinal lavage from the day before the examination. Midazolam and pethidine were given intravenously for sedation and timepidium bromide hydrate or glucagon was used to reduce intestinal peristalsis. Based on the patient's condition, DBE insertion pathway (antegrade or prograde), endoscope type (EN-580 T, EN-450 T 5 or EN-450 P 5 (Fujifilm, Tokyo)) and The sampling method was decided.
  • endoscopic biopsy forceps (Radial Jaw 4P, Boston Scientific, Massachusetts, USA) or endoscopy search net (Roth net, US endoscopy, Ohio, USA) US) was used and obtained from the central SI of each patient.
  • SI solution about 50 cc was aspirated through the endoscope before collecting the mucosal specimen.
  • saliva and stool were collected one day before endoscopy.
  • the biopsy sample is suspended in TE10 (10 mM Tris-HCl, 10 mM EDTA) buffer, stored at -80 ° C, the mucosal scrapes and intestinal juice samples are centrifuged, and the pellet is placed in TE10 buffer. It was dissolved and stored at ⁇ 80 ° C.
  • biopsy samples were suspended in 20% glycerol / PBS, frozen in liquid nitrogen and then stored at -80 ° C. Mucosal scraping samples were centrifuged, pellets were dissolved in 20% glycerol / PBS, frozen in liquid nitrogen and stored at -80 ° C.
  • Frozen samples were thawed and mixed with 800 ⁇ l TE10 buffer containing RNaseA (final concentration 100 ⁇ gml- 1 , Invitrogen, MA, USA) and lysozyme (final concentration 15 mgml- 1, Sigma, MO, USA). The suspension was incubated at 37 ° C. for 1 hour with gentle mixing. Purified acromopeptidase (Wako, Osaka, Japan) was added at a final concentration of 2,000 units ml-1 , and the sample was incubated for an additional 30 minutes at 37 ° C.
  • RNaseA final concentration 100 ⁇ gml- 1 , Invitrogen, MA, USA
  • lysozyme final concentration 15 mgml- 1, Sigma, MO, USA
  • PCR was performed using primer sets of 27Fmod 5'-AGRGTTTTGATYMTGCTCAG-3'(SEQ ID NO: 10) and 338R 5'-TGCTGCCTCGTGGT-3' (SEQ ID NO: 11) targeting the V1-V2 region of the 16S rRNA gene. ..
  • the amplicon produced from each sample (about 330 bp) was purified using APPure XP (Beckman Coulter, CA, USA). DNA was quantified using a Quant-iT Picogreen ds DNA assay kit (Invitrogen) and a TBS-380 mini fluorometer (Turner Biosystems, CA, USA).
  • 16S metagenomic sequencing was performed using MiSeq according to the Illumina protocol. Two opposite-end reads were merged using a fastq-join program based on overlapping sequences. Reads with an average quality value of ⁇ 25 and inaccurate matches with both universal primers were excluded. The filtered reads were used for further analysis after trimming both primer sequences. For each sample, reads that have passed through the 3,000 quality filters are sorted in descending order according to their quality value and have a 97% pair-wise-identity cutoff using UCLUST program version 5.2.32. Clustered into OTUs having (https://www.drive5.com).
  • Taxonomic attribution of each OTU was made using the GLSEARCH program based on similarity to RDP and the National Center for Biotechnology Information (NCBI) genomic database. Taxa with relative amount> 0.1% were considered positive to determine bacterial prevalence.
  • Frozen samples are thawed, serially diluted with PBS and seeded on non-selective and selective agar plates (for aerobic culture: trypticase soiagar, for anaerobic culture: EG, BHK, MRS, CM 0619 + SR 0107).
  • EG and BHK are non-selective media for anaerobic bacteria.
  • MRS is a selective isolation medium for lactic acid bacteria.
  • CM 0619 supplemented with SR 0107 is a selective isolation medium for non-spore-forming anaerobes.
  • strains Individual isolates in the culture collection were classified as "strains" if their 16S rRNA sequences showed> 98% identity.
  • the obtained strain sequence was compared with the sequence of the NCBI database and the OTU observed by 16S rRNA analysis, and the closely related species and the corresponding OTU were determined.
  • TSI slope medium BD, NJ, USA
  • LIM medium LIM medium
  • SIM medium Eiken Chemical, Tokyo, Japan
  • API 20E BioMerieux, Marcy-l'Etoile
  • each E.I. coli strain 35A1, LF82 and MG1655 were cultured overnight aerobically in TS broth 37 ° C., gavage aliquots of 200 ⁇ l containing 7 ⁇ 9 ⁇ 10 8 colony forming units (CFU) in GF mice bottom.
  • the CD-related AIEC strain LF82 used in this example was originally isolated from the ileal mucosa of a CD patient by Darfeile-Michaud et al. (Reference 16).
  • the gnavus 131A1 strain was anaerobically cultured in EG broth or on an EG plate. The colonies on the plate were rubbed and resuspended in EG broth.
  • coli 35A1 strain and R Seven strains other than gnavus 131A1 strain were anaerobically cultured in EG or HK medium. Then, equal amounts of bacterial suspension were mixed to prepare 2-mix or 9-mix. A mixture of isolates or a single bacterial suspension was orally administered to GF mice and colonization was evaluated by Gram stain of the fecal suspension.
  • mice administered a mixture of specific bacterial strains or a single bacterial suspension were maintained on a single notobiotic isolator with a 12 hour / 12 hour light / dark cycle.
  • GFB6 mice (7-14 weeks old) were analyzed 3 weeks after the first gavage. All animal experiments were conducted with the approval of the Keio University Animal Experiment Committee.
  • the GF and SPF mice are all B6 mice obtained from CLEA Japan (Tokyo, Japan).
  • Intestinal lymphocytes were isolated as follows. First, the intestine was opened vertically and washed with PBS to remove the luminal contents. All samples were placed in 20 ml of 5 mM EDTA-containing Hanks Balanced Salt Solution (HBSS) and incubated in a shaking water bath at 37 ° C. for 20 minutes to remove enterocytes (IEC). The muscle layer and adipose tissue were then manually removed using forceps.
  • HBSS Hanks Balanced Salt Solution
  • the remaining LP layer was cut into small pieces and in 10 ml RPMI 1640, 4% fetal bovine serum, 0.5 mgml -1 collagenase D (Roche), 0.5 mgml -1 dispase (Gibco) and 40 ⁇ gml -1 DNase I ( Roche) was contained and incubated in a shaking water bath at 37 ° C. for 45 minutes.
  • Digested tissue was washed with HBSS containing 10 ml 5 mM EDTA, resuspended in 5 ml 40% Percoll (GE Healthcare, Illinois, USA) and in a 15 ml Falcon tube, 2.5 ml 80%. Percoll was layered. Percoll gradient separation was performed by centrifugation at 900 xg for 30 minutes at 25 ° C. Fractions containing lymphocytes were collected from the interface between the two layers and washed with RPMI 1640 containing 10% FBS.
  • cytokine detection cells were stimulated with 50 ngml -1 PMA and 750 ngml -1 ionomycin (both purchased from Sigma) at 37 ° C. for 3.5 hours in the presence of GolgiStop (BD).
  • the cells were permeabilized and treated with anti-TCR ⁇ (BV605; BioLegend, California, USA), anti-CD4 antibody (BV510; BioLegend), anti-TCR ⁇ antibody (BV510; biolegend).
  • anti-TCR ⁇ BV605; BioLegend, California, USA
  • anti-CD4 antibody BV510; BioLegend
  • anti-TCR ⁇ antibody BV510; biolegend
  • CD4T cells were defined as CD4 + TCR ⁇ + cell population within the live cell gate.
  • a model of colitis mediated by anti-IL-10R antibody A mouse model of colitis induced by an anti-IL-10 receptor (IL-10R) antibody was prepared as described in Schiering et al. (Reference 17). That is, on the first day, E.I. The coli 35A1, LF82 or MG1655 strains were established, followed by weekly intraperitoneal injection of anti-mouse IL-10R antibody (1 mg / individual) (BioXcell, New Hampshire, USA) from day 1 to the end of the experiment. Mice were then analyzed 5 weeks after initial colonization.
  • IL-10R anti-IL-10 receptor
  • RNA isolation and qPCR Total RNA was isolated from the scraped colonic epithelium using TRIzol reagent (Invitrogen) according to the manufacturer's instructions. The cDNA was synthesized using Revertra Ace qPCR RT Master Mix (Toyobo, Osaka, Japan). Then, using cDNA as a template, the following primer set and Thunderbird SYBR qPCR Mix (Toyobo) were used, and qPCR was performed with LightCyclor480 (Roche).
  • SI mucosal samples were obtained from 27 CD patients and 17 non-CD patients (patients with diseases other than SI, hamartomatous diseases, etc., see Table 1 below) by biopsy or scraping during the DBE method.
  • Fig. 1A Of the CD patients, 23 had intestinal stenosis (Montreal classification B2 or B3) and 19 were treated with anti-tumor necrosis factor- ⁇ (TNF- ⁇ ) antibody (Table 1).
  • the insertion route and sampling method of DBE were determined by the endoscopist based on the patient's condition. If the antegrade (oral) insertion route was selected, the distal jejunal mucosa was harvested. For retrograde (transanal) insertion, the proximal ileal mucosa was collected. Small intestinal samples were obtained from the mucosa outside the active ulcer or stenosis.
  • the small intestinal mucosal flora of CD patients contains more taxa within Proteobacteria and Bacteroidetes at the portal level and more taxa within Enterobacteriae, Ruminococcaceae and Bacteroidetes at the family level compared to those of non-CD patients.
  • Was FIGS. 1D and 1E.
  • the Firmicutes and Streptococcaceae taxa were reduced in CD patients (FIGS. 1D and 1E).
  • Table 2 shows 18 types of bacteria that are abundant in CD patients. For each group, the most closely related species or strain, National Center for Biotechnology Information (NCBI) genome database percent similarity (%), 9-mix strain ID, average abundance (Avenance), abundance (Abundance) Prevalence) is shown. The abundance rate was calculated by dividing the number of subjects in which a relative amount of bacteria exceeding 0.1% was detected in the SI mucosal sample by the total number of subjects.
  • NCBI National Center for Biotechnology Information
  • E. Colli, Ruminococcus gnavus, Bacteroides dorie, Klebsiella pneumoniae, Streptococcus pasteurianus, Parabacteroides merdee, Parabacteroides merdee, Parabacteroides merdae, Parabacteroides colli and R. gnavus was found to be significantly associated with CD SI mucosa in all three analyzes for abundance (Fig. 2C).
  • E. colli and R. gnavus was concentrated in the CD sample regardless of the insertion route or sampling method (FIGS. 5C-5E and 5H-5J).
  • strain ID: 39F4 S.A. pasteurianus
  • strain ID: 133A7 P. et al. Disstasonis
  • strain ID: 134F1 Bacteroides fragilis
  • strain ID: 32E9 Bacteroides fragilis
  • Erysiperatoclastridium ramosum strain ID: 131A10
  • Bacteroides fragilis strain ID: 131A10
  • Bacteroides uniformis strain ID: 131A11
  • the gut microbiota is recognized as a potent regulator of the host immune system (Reference 14). Therefore, the present inventors analyzed the ability of GF mice to stimulate immune cells in vivo with respect to the selected 9 strains. That is, each of the 9 strains was individually cultured, and the mixture was mixed to prepare a bacterial mixture (9-mix), which was orally administered to GF C57BL / 6 (B6) mice by oral forced gastric administration (Fig. 2G). Then, the mice were bred in a notobioto vinyl isolator for 3 weeks, and the lymphocytes and colon laminalitis (LP) in SI were examined by flow cytometry (Fig. 10).
  • Th17 cells in addition to Th1 cells 2-mix and E.I.
  • the coli 35A1 strain induced a significant increase in the Th cell frequency to the same extent as 9-mix. However, it was not as good as that in SPF mice (comparison between FIGS. 3B and 3F).
  • E. coli strain. colli LF82 strain or E.I. E. et al., A non-pathogenic laboratory strain derived from colli K-12.
  • a mouse in which the coli MG1655 strain has been established has been produced (Reference 16). Therefore, the degree of induction of Th1 cells and Th17 cells in the intestines of these mice was detected, and E.I. Compared with those of colli 35A1 colonized mice.
  • Th1 cell-induced E.I. in the onset of enteritis such as CD Th1 cell-induced E.I. in the onset of enteritis such as CD.
  • a colitis model induced by an anti-IL-10R antibody was used (Reference 17). Specifically, GF wild-type B6 mice were treated with E. coli on the first day. The coli 35A1 strain, the LF82 strain, or the MG1655 strain was established. Subsequently, anti-IL-10R antibody was injected intraperitoneally weekly and the intestines of those mice were analyzed.
  • CD is associated with intestinal flora disbiosis (decreased bacterial diversity) characterized by decreased ⁇ -diversity and increased potential inflammatory bacteria (Reference 11). However, few bacterial strains have been shown to be isolated from the CD-related flora and activate the host immune system.
  • the present inventors evaluated the SI bacterial flora of CD patients and screened for immunostimulatory bacteria.
  • the screening we obtained SI samples from CD patients using the DBE method, which is different from standard colonoscopy and is thorough from distal and proximal SI. It enables various tests and bacterial isolation.
  • E.I. The coli LF82 strain is a clinically relevant strain previously isolated from CD patients.
  • E. Escherichia coli LF82 strain is called intestinal adhesive invasive Escherichia coli (AIEC) because it adheres to and invades intestinal epithelial cells (EC) and can replicate in macrophages (References 16, 18, and 19).
  • AIEC intestinal adhesive invasive Escherichia coli
  • it involves mucin degradation (Vat-AIEC protease), adhesion to CEACAM6 via type 1 pili (expressed on the apical surface of EC) and activation of Toll-like receptors (TLRs) 5 via flagella.
  • E. The coli 35A1 strain is an inflammatory E. coli associated with the SI mucosa of CD patients. Another example of colli is shown. Interestingly, E.I. The coli 35A1 strain was used in the induction of Th1 cells by E. coli. It was stronger than the coli LF82 strain. Bifidobacterium longum and K. Considering that the pneumoniae strain is also associated with intestinal Th1 cell induction (References 25 and 26), E. coli. The colli 35A1 strain is B.I. longum and K. It may have factors similar to those of pneumoniae.
  • R. gnavus is also associated with CD.
  • R. gnavus has been reported to be increased in CD and thereby contributes to its pathogenesis (Reference 27).
  • R. gnavus comprises at least two lineage groups, one of which is predominant in CD patients and produces inflammation-inducing polysaccharides that induce the production of TNF- ⁇ by dendritic cells via TLR4 (References 28, 29). ).
  • R. The immunostimulatory effect of the gnavus 131A1 strain was weak, but R. gnavus may promote colonization of other bacteria associated with CD pathogenesis. In fact, R.
  • gnavus is a mucin-degrading bacterium that degrades the mucin layer, thereby promoting the colonization of other bacteria in the vicinity of the epithelial cell layer.
  • R. gnavus produces sialic acid (Reference 30), which is described in E. coli. It may promote the growth and colonization of other sialic acid-utilizing pathogens, including coli (Reference 31).
  • E.I. colli or R. It is noteworthy that the induction of Th17 cells by 2-mix colonization was significantly enhanced compared to gnavus alone (Fig. 3F).
  • the present invention by targeting a small intestinal bacterium that induces proliferation or activation of Th1 cells and / or Th17 cells, it becomes possible to suppress the proliferation or activation of the cells. As a result, it becomes possible to treat, improve or prevent the disease caused by the cells. Further, according to the present invention, it is also possible to test for diseases caused by the cells by using the amount of bacteria in the small intestine as an index.
  • the present invention is extremely useful in the development, treatment, improvement, prevention and diagnosis of pharmaceuticals relating to diseases such as Crohn's disease caused by Th1 cells and the like.

Abstract

In order to provide a composition for treating, ameliorating or preventing Crohn's disease or the like, a test method for this disease, etc., the present inventors successfully isolated a small intestinal bacterium, said bacterium inducing the proliferation or activation of Th1 cells and/or Th17 cells relating to Crohn's disease, from a bacterial flora in the small intestinal mucosa of a patient with this disease.

Description

Th1細胞及び/又はTh17細胞の増殖又は活性化を誘導する小腸内細菌Small intestinal bacteria that induce proliferation or activation of Th1 and / or Th17 cells
 本発明は、Th1細胞及び/又はTh17細胞(以下「Th1細胞等」とも称する)の増殖又は活性化を誘導する小腸内細菌に関する。本発明は、前記細菌を指標とするTh1細胞等に起因する疾患の評価方法に関する。さらに本発明は、前記細菌等を有効成分とする、Th1細胞等の増殖又は活性化を誘導するための組成物又はワクチン組成物に関する。また本発明は、前記細菌に対して抗菌作用を有する物質を有効成分とする、Th1細胞等の増殖又は活性化を抑制するための組成物又はTh1細胞等に起因する疾患を治療等するための組成物に関する。本発明は、前記細菌を指標とする、前記物質のスクリーニング方法に関する。さらにまた、本発明は、前記細菌を標的とする、Th1細胞等の増殖又は活性化を抑制する方法又はTh1細胞等に起因する疾患を治療等する方法に関する。 The present invention relates to a small intestinal bacterium that induces proliferation or activation of Th1 cells and / or Th17 cells (hereinafter also referred to as "Th1 cells and the like"). The present invention relates to a method for evaluating a disease caused by Th1 cells or the like using the bacterium as an index. Furthermore, the present invention relates to a composition or a vaccine composition for inducing proliferation or activation of Th1 cells or the like containing the bacteria or the like as an active ingredient. Further, the present invention is for treating a composition for suppressing the proliferation or activation of Th1 cells or the like or a disease caused by the Th1 cells or the like, which contains a substance having an antibacterial action against the bacteria as an active ingredient. Regarding the composition. The present invention relates to a method for screening a substance using the bacterium as an index. Furthermore, the present invention relates to a method for suppressing the proliferation or activation of Th1 cells or the like, or a method for treating a disease caused by Th1 cells or the like, which targets the bacterium.
 クローン病(CD)は、主に回腸及び結腸を侵す衰弱性慢性胃腸疾患であり、潰瘍性大腸炎の特徴とは異なる縦走潰瘍、アフタ及び狭窄を特徴とする。CDについては、ゲノムワイド関連解析によって、NOD2、IL23R、ATG16L1及びTNFSF15の各々をコードする遺伝子を含む100以上のCD関連遺伝子が明らかになっている(非特許文献1~3)。また、CDの病因に関しては、宿主側の内因子に加えて、腸内細菌叢を含む外因性因子が関与する可能性がある。実際、CD患者の数は、食事と生活習慣の変化のため、日本を含む国々でおそらく劇的に増加している。 Crohn's disease (CD) is a debilitating chronic gastrointestinal disease that mainly affects the ileum and colon, and is characterized by longitudinal ulcers, aphthae, and stenosis that are different from those of ulcerative colitis. For CD, genome-wide association studies have revealed more than 100 CD-related genes, including genes encoding each of NOD2, IL23R, ATG16L1 and TNFSF15 (Non-Patent Documents 1 to 3). Moreover, regarding the etiology of CD, in addition to the intrinsic factor on the host side, an extrinsic factor including the intestinal flora may be involved. In fact, the number of CD patients has probably increased dramatically in countries, including Japan, due to dietary and lifestyle changes.
 腸内細菌叢は、宿主免疫の強力な調節因子であり、いくつかの疾患と関連することが報告されている(非特許文献4及び5)。また、CDに関しても、腸内細菌叢共生バランス失調(ディスバイオーシス)と呼ばれる細菌叢の変化が、その病因に影響することが示されつつある。例えば、CD患者の糞便では、微生物叢のα多様性の減少とFaecalibacterium prausnitzii存在量の減少が頻繁に報告されている(非特許文献6)。 The gut microbiota is a potent regulator of host immunity and has been reported to be associated with several diseases (Non-Patent Documents 4 and 5). Also, regarding CD, it is being shown that a change in the bacterial flora called intestinal bacterial flora symbiotic imbalance (disbiosis) affects the pathogenesis. For example, in the feces of CD patients, a decrease in α-diversity of the microbial flora and a decrease in the abundance of Faecalibacterium prasnitzii have been frequently reported (Non-Patent Document 6).
 本発明が解決しようとする課題は、CD等を引き起こし得る細菌を同定することを目的とする。ひいては、同定した細菌を標的とする、CD等を治療、改善又は予防するための組成物、及び当該細菌を指標とする、CD等の検査方法等を提供することを目的とする。 The problem to be solved by the present invention is to identify bacteria that can cause CDs and the like. An object of the present invention is to provide a composition for treating, improving or preventing a CD or the like that targets the identified bacterium, and a method for inspecting the CD or the like using the bacterium as an index.
 CDの病因に関する従前の研究において、その多くは糞便微生物叢に焦点が当てられていた。一方、本発明者らは、CD等の発症に関与する炎症惹起性細菌は、おそらく小腸(SI)粘膜に豊富に存在すると考え、当該粘膜の微生物叢に着目した。そして、前記目的を達成すべく、先ず、ダブルバルーン内視鏡検査(DBE)システムを用い、CD患者から空腸及び回腸の粘膜サンプルを採取した。なお、DBEシステムは、ファイバースコープとバルーンポンプシステムを含み、従来のプッシュ式小腸内視鏡検査と比較してSI(遠位空腸と近位回腸)のより詳細な検査を可能にする。 Most previous studies on the etiology of CD have focused on the fecal microflora. On the other hand, the present inventors considered that the pro-inflammatory bacteria involved in the onset of CD and the like are probably abundant in the small intestine (SI) mucosa, and focused on the microbial flora of the mucosa. Then, in order to achieve the above-mentioned purpose, first, a mucosal sample of jejunum and ileum was collected from a CD patient using a double-balloon endoscopy (DBE) system. The DBE system includes a fiberscope and balloon pump system, which allows for more detailed examination of SI (distal jejunum and proximal ileum) compared to conventional push enteroscopy.
 かかるDBEを用いた擦過又は生検により、CD患者及び非CD患者からSI粘膜サンプルを採取し、比較マイクロバイオーム解析を行なった。その結果、CD患者におけるSI粘膜の微生物叢の組成は非CD患者のそれとは異なり、Enterobacteriaceae、Ruminococcaceae及びBacteroidaceaeを含むいくつかの科(family)に属する集団が増加していることが明らかになった。 SI mucosal samples were collected from CD patients and non-CD patients by scraping or biopsy using such DBE, and comparative microbiome analysis was performed. As a result, it was revealed that the composition of the microbial flora of the SI mucosa in CD patients was different from that in non-CD patients, and that the population belonging to several families including Enterobacteriaceae, Ruminococcaceae and Bacteroidaceae was increasing. ..
 また、CD患者のSI粘膜微生物叢を嫌気性培養したところ、80個の細菌株を分離することができた。次いで、それらの中から、CDとの有意な関連性に基づき、異なる9種を各々代表する9株(Escherichia coli株、Ruminococcus gnavus株、Klebsiella pneumoniae株、Erysipelatoclostridium ramosum株、Bacteroides dorei株、Bacteroides fragilis株、Bacteroides uniformis株、Parabacteroides distasonis株及びStreptococcus pasteurianus株)を選抜した。 In addition, when the SI mucosal microbial flora of a CD patient was anaerobically cultured, 80 bacterial strains could be isolated. Next, from among them, 9 strains representing each of the 9 different species (Escherichia coli strain, Ruminococcus gnavius strain, Klebsiella pneumoniae strain, Erysiperatoclastride Bacteroides strain, Bacteroides fragrance strain, Bacteroides fragrance strain, Bacteroides fragrance strain, Bacteroides fragrance strain, Bacteroides fragrance strain, Bacteroides fragrance strain, Bacteroides fragrance strain, Bacteroides fragrance strain, Bacteroides fragrance strain, Bacteroides fragrance strain, Bacteroides fragrance strain , Bacteroides uniformis strain, Parabacteroides distasonis strain and Streptococcus pasteurianus strain) were selected.
 そして、これら細菌株を無菌(GF)マウスに経口投与し、定着させた結果、腸管におけるTh1細胞の集積が増強された。さらに、Th1細胞程ではないが、Th17細胞の集積も増強された。また、前記9株の中でも前記E.coli株は、菌株特異的にTh1細胞を誘導し、高い腸管炎症誘導能を示した。 Then, as a result of orally administering these bacterial strains to sterile (GF) mice and establishing them, the accumulation of Th1 cells in the intestinal tract was enhanced. Furthermore, the accumulation of Th17 cells was enhanced, though not as much as Th1 cells. In addition, among the 9 strains, the E. The coli strain induced Th1 cells specifically for the strain and showed a high ability to induce intestinal inflammation.
 このように、本発明者らは、CDの発症等に関与し得る、Th1細胞及び/又はTh17細胞の増殖又は活性化を誘導する小腸内細菌を見出し、本発明を完成するに至った。 As described above, the present inventors have found a small intestinal bacterium that induces proliferation or activation of Th1 cells and / or Th17 cells that can be involved in the onset of CD, etc., and have completed the present invention.
 すなわち、本発明は以下を提供するものである。
<1> Th1細胞及び/又はTh17細胞の増殖又は活性化を誘導する小腸内細菌。
<2> 下記細菌群から選択される少なくとも1の細菌である、<1>に記載の細菌
細菌群:
配列番号:1に記載のDNA配列に対して95%以上の同一性を有するポリヌクレオチドを有する細菌、
配列番号:2に記載のDNA配列に対して95%以上の同一性を有するポリヌクレオチドを有する細菌、
配列番号:3に記載のDNA配列に対して95%以上の同一性を有するポリヌクレオチドを有する細菌、
配列番号:4に記載のDNA配列に対して95%以上の同一性を有するポリヌクレオチドを有する細菌、
配列番号:5に記載のDNA配列に対して95%以上の同一性を有するポリヌクレオチドを有する細菌、
配列番号:6に記載のDNA配列に対して95%以上の同一性を有するポリヌクレオチドを有する細菌、
配列番号:7に記載のDNA配列に対して95%以上の同一性を有するポリヌクレオチドを有する細菌、
配列番号:8に記載のDNA配列に対して95%以上の同一性を有するポリヌクレオチドを有する細菌、及び
配列番号:9に記載のDNA配列に対して95%以上の同一性を有するポリヌクレオチドを有する細菌。
<3> 配列番号:1に記載のDNA配列に対して95%以上の同一性を有するポリヌクレオチドを有する細菌と、配列番号:2に記載のDNA配列に対して95%以上の同一性を有するポリヌクレオチドを有する細菌との組み合わせである、<1>に記載の細菌。
<4> 小腸内でTh1細胞及び/又はTh17細胞の増殖又は活性化を抑制する物質をスクリーニングする方法であって、
(1)被験物質を非ヒト無菌動物に摂取させる工程、
(2)該非ヒト動物の小腸内におけるTh1細胞及び/又はTh17細胞の数又は活性を検出する工程、及び
(3)工程(2)にて前記細胞の増殖又は活性化の抑制が検出された場合、前記被験物質は、小腸内でTh1細胞及び/又はTh17細胞の増殖又は活性化を抑制する物質であると判定する工程を、
含む方法。
<5> 小腸内でTh1細胞及び/又はTh17細胞の増殖又は活性化を抑制する物質をスクリーニングする方法であって、
(1)被験物質を、配列番号:1に記載のDNA配列に対して95%以上の同一性を有するポリヌクレオチドを有する細菌及び/又は配列番号:2に記載のDNA配列に対して95%以上の同一性を有するポリヌクレオチドを有する細菌を定着させた非ヒト動物に、摂取させる工程、
(2)該非ヒト動物の小腸内における前記細菌の数を検出する工程、及び
(3)工程(2)にて前記細菌の増殖抑制が検出された場合、前記被験物質は、小腸内でTh1細胞及び/又はTh17細胞の増殖又は活性化を抑制する物質であると判定する工程を、
含む方法。
<6> 前記物質が、細菌又はバクテリオファージである、<4>又は<5>に記載の方法。
<7> Th1細胞及び/又はTh17細胞に起因する疾患を評価する方法であって、
(1)被検者の小腸粘膜における、配列番号:1に記載のDNA配列に対して95%以上の同一性を有するポリヌクレオチドを有する細菌及び/又は配列番号:2に記載のDNA配列に対して95%以上の同一性を有するポリヌクレオチドを有する細菌を定量する工程、
(2)工程(1)で定量して得られた値を、前記疾患に罹患していないヒトの小腸粘膜において前記細菌を定量して得られる対応値と比較する工程、及び
(3)工程(2)における比較の結果、被検者の小腸粘膜中における前記定量値が、前記対応値よりも高い場合に、前記被検者は前記疾患に罹患していると判定する工程を、
含む方法。
<8> <1>~<3>のうちのいずれか一項に記載の細菌又は該細菌に由来する生理活性物質を有効成分として含む、Th1細胞及び/又はTh17細胞の増殖又は活性化を誘導するための組成物。
本発明はまた、Th1細胞及び/又はTh17細胞の増殖又は活性化を誘導するための組成物を製造するための、<1>~<3>のうちのいずれか一項に記載の細菌又は該細菌に由来する生理活性物質の使用に関する。
<9> <1>~<3>のうちのいずれか一項に記載の細菌又は該細菌に特異的な抗原を有効成分として含む、ワクチン組成物。
本発明はまた、ワクチン組成物を製造するための、<1>~<3>のうちのいずれか一項に記載の細菌又は該細菌に特異的な抗原の使用に関する。
<10> <1>~<3>のうちのいずれか一項に記載の細菌に対して抗菌作用を有する物質を有効成分として含む、Th1細胞及び/又はTh17細胞の増殖又は活性化を抑制するための組成物。
<11> 前記物質が、<4>~<6>のうちのいずれか一項に記載の方法によって選抜される物質である、<10>に記載の組成物。
本発明はまた、Th1細胞及び/又はTh17細胞の増殖又は活性化を抑制するための組成物を製造するための、<1>~<3>のうちのいずれか一項に記載の細菌に対して抗菌作用を有する物質、又は、<4>~<6>のうちのいずれか一項に記載のスクリーニング方法によって得られる細菌若しくはバクテリオファージの使用に関する。
<12> Th1細胞及び/又はTh17細胞に起因する疾患を治療、改善又は予防するための組成物である、<9>~<11>のうちのいずれか一項に記載の組成物。
<13> <7>に記載の方法によって前記疾患に罹患していると判定された前記被検者に摂取させることを特徴とする、<12>に記載の組成物。
<14> 前記疾患がクローン病である、<12>又は<13>に記載の組成物。
<15> 前記クローン病が小腸型クローン病である、<14>に記載の組成物。
本発明はまた、前記疾患を治療、改善又は予防するための組成物を製造するための、
<1>~<3>のうちのいずれか一項に記載の細菌若しくは該細菌に特異的な抗原、<1>~<3>のうちのいずれか一項に記載の細菌に対して抗菌作用を有する物質、又は、<4>~<6>のうちのいずれか一項に記載の方法によって選抜される物質の使用に関する。
<16> <1>~<3>のうちのいずれか一項に記載の細菌又は該細菌に特異的な抗原を、対象に摂取させ、該対象におけるTh1細胞及び/又はTh17細胞の増殖又は活性化を抑制する方法。
<17> <1>~<3>のうちのいずれか一項に記載の細菌に対して抗菌作用を有する物質を、対象に摂取させ、該対象におけるTh1細胞及び/又はTh17細胞の増殖又は活性化を抑制する方法。
<18> 前記物質が、<4>~<6>のうちのいずれか一項に記載の方法によって選抜される物質である、<17>に記載の方法。
本発明はまた、対象におけるTh1細胞及び/又はTh17細胞の増殖又は活性化を抑制するための、<1>~<3>のうちのいずれか一項に記載の細菌若しくは該細菌に特異的な抗原、<1>~<3>のうちのいずれか一項に記載の細菌に対して抗菌作用を有する物質、又は、<4>~<6>のうちのいずれか一項に記載の方法によって選抜される物質の使用に関する。
<19> <1>~<3>のうちのいずれか一項に記載の細菌又は該細菌に特異的な抗原を、対象に摂取させ、該対象におけるTh1細胞及び/又はTh17細胞に起因する疾患を治療、改善又は予防する方法。
<20> <1>~<3>のうちのいずれか一項に記載の細菌に対して抗菌作用を有する物質を、対象に摂取させ、該対象におけるTh1細胞及び/又はTh17細胞に起因する疾患を治療、改善又は予防する方法。
<21> 前記物質が、<4>~<6>のうちのいずれか一項に記載の方法によって選抜される物質である、<20>に記載の方法。
<22> 前記対象が、<7>に記載の方法によって前記疾患に罹患していると判定された前記被検者である、<19>~<21>のうちのいずれか一項に記載の方法。
<23> 前記疾患がクローン病である、<19>~<22>のうちのいずれか一項に記載の方法。
<24> 前記クローン病が小腸型クローン病である、<23>に記載の方法。
本発明はまた、前記疾患を治療、改善又は予防するための、<1>~<3>のうちのいずれか一項に記載の細菌若しくは該細菌に特異的な抗原、<1>~<3>のうちのいずれか一項に記載の細菌に対して抗菌作用を有する物質、又は、<4>~<6>のうちのいずれか一項に記載の方法によって選抜される物質の使用に関する。
That is, the present invention provides the following.
<1> A small intestinal bacterium that induces proliferation or activation of Th1 cells and / or Th17 cells.
<2> Bacterial bacterial group according to <1>, which is at least one bacterium selected from the following bacterial group:
Bacteria having a polynucleotide having 95% or more identity to the DNA sequence set forth in SEQ ID NO: 1.
Bacteria having a polynucleotide having 95% or more identity to the DNA sequence set forth in SEQ ID NO: 2.
Bacteria having a polynucleotide having 95% or more identity to the DNA sequence set forth in SEQ ID NO: 3.
Bacteria having a polynucleotide having 95% or more identity to the DNA sequence set forth in SEQ ID NO: 4.
Bacteria having a polynucleotide having 95% or more identity to the DNA sequence set forth in SEQ ID NO: 5.
Bacteria having a polynucleotide having 95% or more identity to the DNA sequence set forth in SEQ ID NO: 6.
Bacteria having a polynucleotide having 95% or more identity to the DNA sequence set forth in SEQ ID NO: 7.
Bacteria having a polynucleotide having 95% or more identity to the DNA sequence set forth in SEQ ID NO: 8 and a polynucleotide having 95% or more identity to the DNA sequence set forth in SEQ ID NO: 9. Bacteria to have.
<3> Bacteria having a polynucleotide having 95% or more identity to the DNA sequence set forth in SEQ ID NO: 1 and 95% or more identity to the DNA sequence set forth in SEQ ID NO: 2. The bacterium according to <1>, which is a combination with a bacterium having a polynucleotide.
<4> A method for screening a substance that suppresses the proliferation or activation of Th1 cells and / or Th17 cells in the small intestine.
(1) A step of ingesting a test substance into a non-human germ-free animal,
(2) When the number or activity of Th1 cells and / or Th17 cells in the small intestine of the non-human animal is detected, and when suppression of proliferation or activation of the cells is detected in (3) step (2). The step of determining that the test substance is a substance that suppresses the proliferation or activation of Th1 cells and / or Th17 cells in the small intestine.
How to include.
<5> A method for screening a substance that suppresses the proliferation or activation of Th1 cells and / or Th17 cells in the small intestine.
(1) The test substance is a bacterium having a polynucleotide having a polynucleotide having 95% or more identity with respect to the DNA sequence set forth in SEQ ID NO: 1 and / or 95% or more with respect to the DNA sequence set forth in SEQ ID NO: 2. Ingestion of non-human animals colonized with a polynucleotide having a polynucleotide having the same identity as
When (2) the step of detecting the number of the bacteria in the small intestine of the non-human animal and (3) the suppression of the growth of the bacteria are detected in the step (2), the test substance is Th1 cells in the small intestine. And / or the step of determining that the substance suppresses the proliferation or activation of Th17 cells.
How to include.
<6> The method according to <4> or <5>, wherein the substance is a bacterium or a bacteriophage.
<7> A method for evaluating a disease caused by Th1 cells and / or Th17 cells.
(1) For bacteria having a polynucleotide having 95% or more identity with respect to the DNA sequence set forth in SEQ ID NO: 1 and / or the DNA sequence set forth in SEQ ID NO: 2 in the small intestinal mucosa of the subject. Quantifying bacteria with polynucleotides having 95% or more identity,
(2) A step of comparing the value obtained by quantifying in step (1) with a corresponding value obtained by quantifying the bacteria in the mucosa of the small intestine of a human who does not suffer from the disease, and step (3) (3). As a result of the comparison in 2), when the quantitative value in the small intestinal mucosa of the subject is higher than the corresponding value, the step of determining that the subject is suffering from the disease is performed.
How to include.
<8> Induces proliferation or activation of Th1 cells and / or Th17 cells containing the bacterium according to any one of <1> to <3> or a physiologically active substance derived from the bacterium as an active ingredient. Composition to do.
The bacterium according to any one of <1> to <3> for producing a composition for inducing proliferation or activation of Th1 cells and / or Th17 cells, or the same. Regarding the use of bioactive substances derived from bacteria.
<9> A vaccine composition comprising the bacterium according to any one of <1> to <3> or an antigen specific to the bacterium as an active ingredient.
The present invention also relates to the use of the bacterium according to any one of <1> to <3> or an antigen specific to the bacterium for producing a vaccine composition.
<10> Suppresses the proliferation or activation of Th1 cells and / or Th17 cells containing a substance having an antibacterial action against the bacterium according to any one of <1> to <3> as an active ingredient. Composition for.
<11> The composition according to <10>, wherein the substance is a substance selected by the method according to any one of <4> to <6>.
The present invention also relates to the bacterium according to any one of <1> to <3> for producing a composition for suppressing the proliferation or activation of Th1 cells and / or Th17 cells. The present invention relates to the use of a substance having an antibacterial activity, or a bacterium or bacteriophage obtained by the screening method according to any one of <4> to <6>.
<12> The composition according to any one of <9> to <11>, which is a composition for treating, ameliorating or preventing a disease caused by Th1 cells and / or Th17 cells.
<13> The composition according to <12>, which is ingested by the subject determined to be suffering from the disease by the method according to <7>.
<14> The composition according to <12> or <13>, wherein the disease is Crohn's disease.
<15> The composition according to <14>, wherein the Crohn's disease is a small intestinal Crohn's disease.
The present invention also comprises the production of compositions for treating, ameliorating or preventing said diseases.
Antibacterial action against the bacterium according to any one of <1> to <3> or an antigen specific to the bacterium, and against the bacterium according to any one of <1> to <3>. The present invention relates to the use of a substance having a bacterium or a substance selected by the method according to any one of <4> to <6>.
<16> The bacterium according to any one of <1> to <3> or an antigen specific to the bacterium is ingested by the subject, and the proliferation or activity of Th1 cells and / or Th17 cells in the subject. A method of suppressing the formation of bacteria.
<17> A substance having an antibacterial action against the bacterium according to any one of <1> to <3> is ingested by a subject, and the proliferation or activity of Th1 cells and / or Th17 cells in the subject. A method of suppressing sterilization.
<18> The method according to <17>, wherein the substance is a substance selected by the method according to any one of <4> to <6>.
The present invention also relates to the bacterium according to any one of <1> to <3> for suppressing the proliferation or activation of Th1 cells and / or Th17 cells in a subject, or a bacterium specific to the bacterium. By the antigen, the substance having an antibacterial action against the bacterium according to any one of <1> to <3>, or the method according to any one of <4> to <6>. Regarding the use of selected substances.
<19> A disease caused by Th1 cells and / or Th17 cells in the subject by ingesting the bacterium according to any one of <1> to <3> or an antigen specific to the bacterium. How to treat, improve or prevent.
<20> A disease caused by Th1 cells and / or Th17 cells in a subject by ingesting a substance having an antibacterial action against the bacterium according to any one of <1> to <3>. How to treat, improve or prevent.
<21> The method according to <20>, wherein the substance is a substance selected by the method according to any one of <4> to <6>.
<22> The item according to any one of <19> to <21>, wherein the subject is the subject determined to have the disease by the method according to <7>. Method.
<23> The method according to any one of <19> to <22>, wherein the disease is Crohn's disease.
<24> The method according to <23>, wherein the Crohn's disease is a small intestinal Crohn's disease.
The present invention also comprises the bacterium according to any one of <1> to <3> or an antigen specific to the bacterium for treating, ameliorating or preventing the disease, <1> to <3. >, The substance having an antibacterial action against the bacterium according to any one of <4> to <6>, or the substance selected by the method according to any one of <4> to <6>.
 本発明によれば、Th1細胞及び/又はTh17細胞の増殖又は活性化を誘導する小腸内細菌を標的とすることによって、前記細胞の増殖又は活性化の抑制等が可能となり、ひいては、前記細胞に起因する疾患を治療、改善又は予防することが可能となる。また、本発明によれば、前記小腸内細菌量を指標として、前記細胞に起因する疾患を検査することも可能となる。 According to the present invention, by targeting a small intestinal bacterium that induces proliferation or activation of Th1 cells and / or Th17 cells, it becomes possible to suppress the proliferation or activation of the cells, and thus to the cells. It is possible to treat, improve or prevent the resulting disease. Further, according to the present invention, it is also possible to test for diseases caused by the cells by using the amount of bacteria in the small intestine as an index.
ダブルバルーン小腸内視鏡検査を介して小腸(SI)からサンプルを得るための方法の概要を示す、概略図である。クローン病(CD)患者27人、非CD患者27人を対象とし、得られたサンプルについて、16SrRNA遺伝子配列解析を実施した。FIG. 6 is a schematic diagram outlining a method for obtaining a sample from the small intestine (SI) via double-balloon enteroscopy. For 27 patients with Crohn's disease (CD) and 27 patients with non-CD, 16S rRNA gene sequence analysis was performed on the obtained samples. SI細菌叢のα多様性指数を示すグラフである。図中、「CD」はクローン病患者由来のサンプルを解析した結果を示し、「non-CD」は非クローン病患者由来のサンプルを解析した結果を示す。各グラフは、OTU(Operational Taxonomic Unit)の数、ACE(Abundance-based Coverage Estimator)、Chao1及びシャノン指数(Shannon index)について解析した結果を各々示す。各棒グラフは、グループの平均値を表す。各ポイントは、個々のサンプルを表す。エラーバーは、標準偏差(SD)を表す。対応がないスチューデントのt検定にて解析して、得られたP値を付記する。It is a graph which shows the α diversity index of SI bacterial flora. In the figure, "CD" indicates the result of analyzing a sample derived from a Crohn's disease patient, and "non-CD" indicates the result of analyzing a sample derived from a non-Crohn's disease patient. Each graph shows the results of analysis of the number of OTUs (Operational Taxonomy Units), ACE (Abundance-based Coverage Estimator), Chao1 and Shannon index. Each bar graph represents the average value of the group. Each point represents an individual sample. Error bars represent standard deviation (SD). The P value obtained by analyzing with the unpaired Student's t-test is added. CD患者由来のSIサンプルと非CD患者由来のそれとの菌叢の構造的類似度を解析した結果を示す、二次元プロット図である。構造的類似度の比較は、Bray-Curtis距離ベース非計量多次元スケーリング(NMDS)プロットで可視化し、CD患者由来のサンプル(図中「CD」)及び非CD患者由来のサンプル(図中「non-CD」)間に関し、置換多変量分散分析(PERMANOVA)解析にて検定を行なった。各ポイントは、個々のサンプルを表す。It is a two-dimensional plot figure which shows the result of having analyzed the structural similarity of the bacterial flora between the SI sample derived from a CD patient and that derived from a non-CD patient. Comparisons of structural similarity are visualized on Bray-Curtis distance-based non-metric multidimensional scaling (NMDS) plots, with samples from CD patients (“CD” in the figure) and samples from non-CD patients (“non” in the figure). -CD ") was tested by Substitution Multivariate Analysis of Variance (PERMANOVA) analysis. Each point represents an individual sample. SI細菌叢の門(Phylum)レベルにおける細菌分類群の相対的存在量(Relative abundance(%))を示す、グラフである。図中、横軸において、各細菌門についての結果を示す(各左側は、非CD患者由来のサンプル解析結果を示し、各右側はCD患者由来のサンプル解析結果を示す)。各太いバーは、グループの平均値を表す。各ポイントは、個々のサンプルを表す。エラーバーは、SDを表す。各アスタリスクは、誤判定率(FDR)アプローチによる多重t検定による値が、*<0.05;**0.01未満;***≦0.001;****<0.0001であることを示す。It is a graph which shows the relative abundance (Reactive amount (%)) of a bacterial taxon at the phylum level of the SI flora. In the figure, the horizontal axis shows the results for each bacterial phylum (each left side shows the sample analysis result derived from a non-CD patient, and each right side shows the sample analysis result derived from a CD patient). Each thick bar represents the average value of the group. Each point represents an individual sample. Error bars represent SD. For each asterisk, the value obtained by multiple t-test using the false positive rate (FDR) approach is *** 0.05; *** less than 0.01; *** ≤ 0.001; *** <0.0001. Is shown. SI細菌叢の科(Family)レベルにおける細菌分類群の相対的存在量を示す、グラフである。図中、横軸において、各細菌科についての結果を示す(各左側は、非CD患者由来のサンプル解析結果を示し、各右側はCD患者由来のサンプル解析結果を示す)。各太いバーは、グループの平均値を表す。各ポイントは、個々のサンプルを表す。エラーバーは、SDを表す。各アスタリスクは、FDRアプローチによる多重t検定による値が、*<0.05;**0.01未満;***≦0.001;****<0.0001であることを示す。FIG. 5 is a graph showing the relative abundance of bacterial taxa at the Family level of the SI flora. In the figure, the horizontal axis shows the results for each bacteriaceae (the left side shows the sample analysis results derived from non-CD patients, and the right side shows the sample analysis results derived from CD patients). Each thick bar represents the average value of the group. Each point represents an individual sample. Error bars represent SD. Each asterisk indicates that the value obtained by multiple t-test by the FDR approach is *** <0.05; ** less than 0.01; *** ≤ 0.001; *** <0.0001. SI細菌叢の科レベルにおける細菌分類群の相対的存在量を示す、グラフである。図中、横軸において、各細菌科についての結果を示す(各左側は、非CD患者由来のサンプル解析結果を示し、各真ん中は腸管狭窄(strictures)を伴わないCD患者のサンプル解析結果を示し、各右側は腸管狭窄を伴うCD患者由来のサンプル解析結果を示す)。各太いバーは、グループの平均値を表す。各ポイントは、個々のサンプルを表す。エラーバーは、SDを表す。各アスタリスクは、FDRアプローチによる多重t検定による値が、*<0.05;**0.01未満;***≦0.001;****<0.0001であることを示す。FIG. 5 is a graph showing the relative abundance of bacterial taxa at the family level of the SI flora. In the figure, the horizontal axis shows the results for each bacteriological family (the left side shows the sample analysis results derived from non-CD patients, and the middle shows the sample analysis results of CD patients without intestinal strictures (trictures). , Each right side shows the results of sample analysis from a CD patient with intestinal stenosis). Each thick bar represents the average value of the group. Each point represents an individual sample. Error bars represent SD. Each asterisk indicates that the value obtained by multiple t-test by the FDR approach is *** <0.05; ** less than 0.01; *** ≤ 0.001; *** <0.0001. CD患者(図中「CD」)及び非CD患者(図中「non-CD」)間の差次的に豊富な分類群について計算した結果を示す、LDAスコアのヒストグラムである。図中、LDAスコアにおいて、マイナス側に非CD患者についての解析結果が示され、プラス側にCD患者についての解析結果が示される。6 is a histogram of LDA scores showing the results of calculations for the differentially abundant taxa between CD patients (“CD” in the figure) and non-CD patients (“non-CD” in the figure). In the figure, in the LDA score, the analysis result for non-CD patients is shown on the negative side, and the analysis result for CD patients is shown on the positive side. CD患者由来のサンプル(図中「CD」)と非CD患者のそれ(図中「non-CD」)とを比較して計算した結果を示す、誤判定率のヒストグラムである。図中、有意な分類群(FDR≦0.05)のみを示した。FDRにおいて、マイナス側に非CD患者についての解析結果が示され、プラス側にCD患者についての解析結果が示される。It is a histogram of the misjudgment rate which shows the result calculated by comparing the sample derived from a CD patient (“CD” in the figure) with that of a non-CD patient (“non-CD” in the figure). In the figure, only significant taxa (FDR ≤ 0.05) are shown. In FDR, the analysis result for non-CD patients is shown on the minus side, and the analysis result for CD patients is shown on the plus side. 3種類の計算解析を組み合わせることにより同定した18種類のCD関連細菌を示すベン図である。3種類の計算解析とは、効果サイズ測定と組み合わせた線形判別分析(LDA)(LEfSe)と、誤判定率を用いた多重t検定法(Multiple t ests)と、CD関連SI粘膜[CD患者のSI粘膜中の平均存在量>1%かつ倍率変化(CD/non-CD)>2]において高存在量を示すOTUに対する配列データのマイニング(Abundande in CD>1%&Fold change>2)とである。It is a Venn diagram showing 18 kinds of CD-related bacteria identified by combining 3 kinds of computational analysis. The three types of computational analysis are linear discriminant analysis (LDA) (LEfSe) combined with effect size measurement, multiple t-test method (Multiple t-tests) using false judgment rate, and CD-related SI mucous membrane [SI of CD patients]. Mining of sequence data for OTU showing high abundance in mucosal average abundance> 1% and magnification change (CD / non-CD)> 2] (Abundande in CD> 1% & Fold change> 2). 粘膜サンプル(図中「Mucosa」)及び腸液(図中「Juice」)中のE.coli及びR.gnavusの相対的存在量を示すグラフである。図中、横軸において、各サンプルについての結果を示す(各左側は、非CD患者由来のサンプル解析結果を示し、各右側はCD患者由来のサンプル解析結果を示す)。各太いバーは、グループの平均を表す。各ポイントは、個々のサンプルを表す。エラーバーはSDを表す。各アスタリスクは、対応のないスチューデントのt検定によって得られた値が、*P<0.05;**P<0.01であることを表す。E. coli in mucosal samples (“Mucosa” in the figure) and intestinal juice (“Juice” in the figure). colli and R. It is a graph which shows the relative abundance of gnavus. In the figure, the horizontal axis shows the results for each sample (the left side shows the sample analysis result derived from a non-CD patient, and each right side shows the sample analysis result derived from a CD patient). Each thick bar represents the average of the group. Each point represents an individual sample. Error bars represent SD. Each asterisk indicates that the value obtained by the unpaired Student's t-test is * P <0.05; ** P <0.01. 唾液(図中「Saliva」)、SI及び糞便(図中「Faces」)中のE.coli及びR.gnavusの相対的存在量を示すグラフである。図中、横軸において、各サンプルについての結果を示す(各左側は、非CD患者由来のサンプル解析結果を示し、各右側はCD患者由来のサンプル解析結果を示す)。縦軸は、相対的存在量を表す。各太いバーは、グループの平均を表す。各ポイントは、個々のサンプルを表す。エラーバーはSDを表す。各アスタリスクは、対応のないスチューデントのt検定によって得られた値が、*P<0.05;**P<0.01であることを表す。E. coli in saliva (“Saliva” in the figure), SI and feces (“Faces” in the figure). colli and R. It is a graph which shows the relative abundance of gnavus. In the figure, the horizontal axis shows the results for each sample (the left side shows the sample analysis result derived from a non-CD patient, and each right side shows the sample analysis result derived from a CD patient). The vertical axis represents the relative abundance. Each thick bar represents the average of the group. Each point represents an individual sample. Error bars represent SD. Each asterisk indicates that the value obtained by the unpaired Student's t-test is * P <0.05; ** P <0.01. 16SrRNAシーケンシングにより決定した、各サンプルにおけるSI細菌叢組成を示す、帯グラフである。図中、カラー表示下、CD患者(CD patients)で有意に濃縮されたOTUは暖色でマークされ、非CD患者(non-CD patients)で濃縮されたものは青で、有意差のないものは灰色で示される。また、付記する表において、CD患者のSIサンプルから分離した9種類の株を示す。表中、National Center for Biotechnology Information(NCBI)のデータベースにおけるそれらの最近縁種(Closest species)、パーセンテージ類似性(%)及び菌株のID(Strain ID of 9 strains)を示す。It is a band graph which shows the SI bacterial flora composition in each sample determined by 16S rRNA sequencing. In the figure, under color display, OTUs significantly concentrated in CD patients (CD patients) are marked in warm colors, those concentrated in non-CD patients (non-CD patients) are blue, and those with no significant difference are those. Shown in gray. In addition, in the attached table, nine kinds of strains isolated from the SI sample of the CD patient are shown. In the table, their most recent species (Closest species), percentage similarity (%), and strain ID (Strine ID of 9 strains) in the database of National Center for Biotechnology Information (NCBI) are shown. 小腸固有層(図中上部の「SILP」)及び結腸固有層(図中下部の「CLP」)のCD4+TCRβ+T細胞(CD4T細胞)について、IFN-γ+細胞及びIL-17+細胞の割合を解析した、代表的な結果を示す、フローサイトメトリープロット図である。無菌(GF)C57BL/6(B6)マウスに、前記9種類のCD濃縮株(9-mix)の混合物を経口接種し、3週間後に安楽死させた。図中、「GF+9mix」(図3B及び3Dにおいては「+9mix」)は、当該マウスの解析結果を示す。「GF」は、CD濃縮株非接種のGFマウスの解析結果を示す。「SPF」は、特定病原体フリーマウスの解析結果を示す。The ratio of IFN-γ + cells and IL-17 + cells was analyzed for CD4 + TCRβ + T cells (CD4T cells) in the lamina propria of the small intestine (“SILP” in the upper middle of the figure) and the lamina propria of the colon (“CLP” in the lower middle of the figure). It is a flow cytometry plot figure which shows a typical result. Aseptic (GF) C57BL / 6 (B6) mice were orally inoculated with a mixture of the above 9 types of CD concentrates (9-mix) and euthanized after 3 weeks. In the figure, "GF + 9mix" ("+9mix" in FIGS. 3B and 3D) indicates the analysis result of the mouse. "GF" indicates the analysis result of GF mice not inoculated with the CD concentrate. "SPF" indicates the analysis result of a specific pathogen-free mouse. 小腸固有層及び結腸固有層のCD4T細胞について、IFN-γ+細胞及びIL-17+細胞の割合を解析した結果を示すグラフである。各ポイントは、個々のマウスを表す。各太いバーは、グループの平均を表す。エラーバーはSDを表す。各アスタリスクは、Tukeyの事後検定(post hoc test)による一方向ANOVAによって得られた値が、*P<0.05;**P<0.01;***P<0.001;****P<0.0001であることを表す。その他、図中の表記については、図3Aと同様である。It is a graph which shows the result of having analyzed the ratio of IFN-γ + cell and IL-17 + cell about the CD4T cell of the small intestine lamina propria and the colon lamina propria. Each point represents an individual mouse. Each thick bar represents the average of the group. Error bars represent SD. For each asterisk, the value obtained by one-way ANOVA by Tukey's post hoc test (post hoc test) is * P <0.05; ** P <0.01; *** P <0.001; ** ** Indicates that P <0.0001. Other notations in the figure are the same as those in FIG. 3A. 結腸固有層のCD4T細胞について、IFN-γ、IL-17及びDR3の発現を解析した、代表的な結果を示す、フローサイトメトリープロット図である。図中の表記については、図3Aと同様である。FIG. 3 is a flow cytometric plot showing representative results of analyzing the expression of IFN-γ, IL-17 and DR3 for CD4T cells in the colon lamina propria. The notation in the figure is the same as in FIG. 3A. 結腸固有層のCD4T細胞について、IFN-γ、IL-17及びDR3の発現を解析した結果を示す、グラフである。各ポイントは、個々のマウスを表す。各太いバーは、グループの平均を表す。エラーバーはSDを表す。各アスタリスクは、Bonferroniの事後検定による双方向ANOVAによって得られた値が、*P<0.05;**P<0.01;***P<0.001;****P<0.0001であることを表す。その他、図中の表記については、図3Cと同様である。It is a graph which shows the result of having analyzed the expression of IFN-γ, IL-17 and DR3 about the CD4T cell of the colon lamina propria. Each point represents an individual mouse. Each thick bar represents the average of the group. Error bars represent SD. For each asterisk, the value obtained by bidirectional ANOVA by Bonferroni's post-test is * P <0.05; ** P <0.01; *** P <0.001; *** P <0. Indicates that it is .0001. Other notations in the figure are the same as in FIG. 3C. 結腸固有層のCD4T細胞について、IFN-γ及びIL-17の発現を解析した、代表的な結果を示す、フローサイトメトリープロット図である。「GF+131A1」及び「GF+35A1」は、図2Fに示したR.gnavus株及びE.coli株を各々経口接種させたGFマウスの解析結果を示す。「GF+2mix」は、これら2菌株の混合物を経口接種させたGFマウスの解析結果を示す。FIG. 3 is a flow cytometric plot showing representative results of analyzing the expression of IFN-γ and IL-17 for CD4T cells in the colon lamina propria. “GF + 131A1” and “GF + 35A1” are referred to as R.I. gnavus strain and E. coli. The analysis result of the GF mouse which was orally inoculated with each of the colli strains is shown. "GF + 2mix" shows the analysis results of GF mice that were orally inoculated with a mixture of these two strains. 結腸固有層のCD4T細胞について、IFN-γ及びIL-17の発現を解析した結果を示す、グラフである。「GF」は菌株を接種させていないGFマウスの解析結果を示す。「+131A1」及び「+35A1」は、図2Fに示したR.gnavus株及びE.coli株を各々経口接種したGFマウスの解析結果を示す。「+2-mix」は、これら2菌株の混合物を経口接種したGFマウスの解析結果を示す。It is a graph which shows the result of having analyzed the expression of IFN-γ and IL-17 about the CD4T cell of the colon lamina propria. "GF" indicates the analysis result of GF mice not inoculated with the strain. “+ 131A1” and “+ 35A1” are R.I. gnavus strain and E. coli. The analysis result of the GF mouse which orally inoculated each of the colli strains is shown. “+ 2-mix” indicates the analysis result of GF mice orally inoculated with a mixture of these two strains. 小腸固有層(図中上部の「SILP」)及び結腸固有層(図中下部の「CLP」)のCD4T細胞について、IFN-γ+細胞及びIL-17+細胞の割合を解析した、代表的な結果を示す、フローサイトメトリープロット図である。GFマウスに、CD患者由来E.coli株(E.coli 35A1株、E.coli LF82株、又はE.coli MG1655株)を経口接種し、3週間後に安楽死させた。図中、「GF+35A1」、「GF+LF82」及び「GF+MG1655」(図4B、4C、4E及び4F)においては、「+35A1」、「+LF82」及び「+MG1655」は、これらマウスの解析結果を各々示す。「GF」は、E.coli株非接種のGFマウスの解析結果を示す。「SPF」は、特定病原体フリーマウスの解析結果を示す。Representative results of analysis of the proportions of IFN-γ + cells and IL-17 + cells for CD4T cells in the lamina propria of the small intestine (“SILP” in the upper middle of the figure) and the lamina propria of the colon (“CLP” in the lower middle of the figure) are shown. It is a flow cytometry plot figure which shows. In GF mice, CD patient-derived E. E. coli strain (E. coli 35A1 strain, E. coli LF82 strain, or E. coli MG1655 strain) was orally inoculated and euthanized 3 weeks later. In the figure, in "GF + 35A1", "GF + LF82" and "GF + MG1655" (FIGS. 4B, 4C, 4E and 4F), "+ 35A1", "+ LF82" and "+ MG1655" indicate the analysis results of these mice, respectively. "GF" is E.I. The analysis result of the GF mouse which did not inoculate the coli strain is shown. "SPF" indicates the analysis result of a specific pathogen-free mouse. 小腸固有層のCD4T細胞について、IFN-γ+細胞及びIL-17+細胞の割合を解析した結果を示す、グラフである。各ポイントは、個々のマウスを表す。各太いバーは、グループの平均を表す。エラーバーはSDを表す。各アスタリスクは、Tukeyの事後検定による一方向ANOVAによって得られた値が、*P<0.05;**P<0.01であることを示す。また「ns」は、有意差なし(P>0.05)であることを示す。It is a graph which shows the result of having analyzed the ratio of IFN-γ + cell and IL-17 + cell about the CD4T cell of the small intestine lamina propria. Each point represents an individual mouse. Each thick bar represents the average of the group. Error bars represent SD. Each asterisk indicates that the value obtained by one-way ANOVA by Tukey's post-test is * P <0.05; ** P <0.01. Further, "ns" indicates that there is no significant difference (P> 0.05). 結腸固有層のCD4T細胞について、IFN-γ+細胞及びIL-17+細胞の割合を解析した結果を示す、グラフである。図中の表記については、図4Bと同様である。It is a graph which shows the result of having analyzed the ratio of IFN-γ + cell and IL-17 + cell about the CD4T cell of the colon lamina propria. The notation in the figure is the same as in FIG. 4B. 腸炎症の発生と重症度を評価するために、ヘマトキシリン・エオジン染色して観察した、代表的な結果を示す、写真である。GFマウスに、1日目に各E.coli株を定着させ、続いて抗マウスIL-10受容体(IL-10R)抗体(1mg/個体)を1日目から実験終了まで毎週腹腔内注射し、腸炎症を評価した。図中、「GF+35A1」及び「GF」は、各々E.coli 35A1株を定着させたマウス及び定着させていないマウスの解析結果を示す。anti-IL10Rの「+」及び「-」は、抗マウスIL-10R抗体注射の有無を各々示す。It is a photograph showing a representative result observed by staining with hematoxylin and eosin to evaluate the occurrence and severity of intestinal inflammation. In GF mice, on the first day, each E.I. The coli strain was established, followed by intraperitoneal injection of anti-mouse IL-10 receptor (IL-10R) antibody (1 mg / individual) weekly from day 1 to the end of the experiment to assess intestinal inflammation. In the figure, "GF + 35A1" and "GF" are E.I. The analysis results of the mouse in which the colli 35A1 strain is colonized and the mouse in which the colli 35A1 strain is not colonized are shown. The "+" and "-" of anti-IL10R indicate the presence or absence of anti-mouse IL-10R antibody injection, respectively. 各E.coli株を定着させたGFマウスの盲腸における組織学的大腸炎スコアを示すグラフである。各ポイントは、個々のマウスを表す。各太いバーは、グループの平均を表す。エラーバーはSDを表す。各アスタリスクは、Tukeyの事後検定による一方向ANOVAによって得られた値が、*P<0.05;**P<0.01;***P<0.001;****P<0.0001であることを示す。「+ anti-IL10R」は、抗マウスIL-10R抗体を腹腔内投与したマウスであることを表す。Each E. It is a graph which shows the histological colitis score in the cecum of the GF mouse which established the coli strain. Each point represents an individual mouse. Each thick bar represents the average of the group. Error bars represent SD. For each asterisk, the value obtained by one-way ANOVA by Tukey's post-test is * P <0.05; ** P <0.01; *** P <0.001; *** P <0. Indicates that it is .0001. "+ Anti-IL10R" indicates that the mouse is intraperitoneally administered with an anti-mouse IL-10R antibody. 各E.coli株を定着させたGFマウスの結腸上皮擦過サンプルにおける、Tnf mRNAの相対的発現を示すグラフである。図中の表記については、図4Eと同様である。Each E. It is a graph which shows the relative expression of Tnf mRNA in the colon epithelial scraping sample of the GF mouse which colonized the coli strain. The notation in the figure is the same as in FIG. 4E. SI細菌叢のα多様性指数を示すグラフである。図中、CDはクローン病患者由来のサンプルを解析した結果を示し、non-CDは非クローン病患者由来のサンプルを解析した結果を示す。各グラフは、観測されたOTUの数、ACE、Chao1及びシャノン指数について解析した結果を各々示す。「Antegrada」及び「Retrograda」は各々、順行性挿入及び逆行性挿入にて得られたSI粘膜サンプルを解析した結果であることを示す。各棒グラフは、グループの平均値を表す。対応がないスチューデントのt検定にて解析して、得られたP値を付記する。It is a graph which shows the α diversity index of SI bacterial flora. In the figure, CD shows the result of analyzing a sample derived from a Crohn's disease patient, and non-CD shows the result of analyzing a sample derived from a non-Crohn's disease patient. Each graph shows the results of analysis for the number of observed OTUs, ACE, Chao1 and Shannon index. "Antegrada" and "Retrograda" indicate that they are the results of analysis of SI mucosal samples obtained by antegrade and prograde insertion, respectively. Each bar graph represents the average value of the group. The P value obtained by analyzing with the unpaired Student's t-test is added. CD患者由来のSIサンプルと非CD患者由来のそれとの菌叢の構造的類似度を解析した結果を示す、二次元プロット図である。構造的類似度の比較は、Bray-Curtis距離ベース非計量多次元スケーリング(NMDS)プロットで可視化した。各ポイントは個々のサンプルを示す。「CD antegrade」及び「CD retrograde」は、順行性挿入及び逆行性挿入にて得られたCD患者由来のSIサンプルを解析した結果を各々示す。「non-CD antegrade」及び「non-CD retrograde」は、順行性挿入及び逆行性挿入にて得られた非CD患者由来のSIサンプルを解析した結果を各々示す。It is a two-dimensional plot figure which shows the result of having analyzed the structural similarity of the bacterial flora between the SI sample derived from a CD patient and that derived from a non-CD patient. Structural similarity comparisons were visualized on Bray-Curtis distance-based non-scaling multidimensional scaling (NMDS) plots. Each point indicates an individual sample. "CD antegrade" and "CD retrograde" show the results of analysis of SI samples derived from CD patients obtained by antegrade and prograde insertion, respectively. "Non-CD antegrade" and "non-CD retrograde" show the results of analysis of SI samples derived from non-CD patients obtained by antegrade and prograde insertion, respectively. 小腸粘膜(SI mucosa)における、門(Phylum)レベルでの細菌分類群の相対的存在量を示す、グラフである。図中、横軸において、各細菌門に関する解析結果を、左から順に、順行性挿入にて得られた非CD患者由来のSIサンプル、順行性挿入にて得られたCD患者由来のSIサンプル、逆行性挿入にて得られた非CD患者由来のSIサンプル、及び、逆行性挿入にて得られたCD患者由来のSIサンプルについて、各々示す。各棒グラフは、グループの平均値を表す。エラーバーはSDを表す。各アスタリスクは、FDRアプローチによる多重t検定によって得られた値が、*<0.05;**0.01未満;***≦0.001;****<0.0001であることを各々示す。It is a graph which shows the relative abundance of the bacterial taxon at the phylum (Phylum) level in the small intestinal mucosa (SI mucosa). In the figure, on the horizontal axis, the analysis results for each bacterial phylum are shown in order from the left, SI samples derived from non-CD patients obtained by anterograde insertion, and SIs derived from CD patients obtained by anterograde insertion. The sample, the SI sample derived from a non-CD patient obtained by retrograde insertion, and the SI sample derived from a CD patient obtained by retrograde insertion are shown. Each bar graph represents the average value of the group. Error bars represent SD. For each asterisk, the value obtained by the multiple t-test by the FDR approach is *** 0.05; less than ** 0.01; *** ≤ 0.001; *** <0.0001. Each is shown. 小腸粘膜における、科(Family)レベルでの細菌分類群の相対的存在量を示す、グラフである。図中の表記は図5Cと同様である。FIG. 5 is a graph showing the relative abundance of bacterial taxa at the Family level in the mucosa of the small intestine. The notation in the figure is the same as that in FIG. 5C. 小腸粘膜における、OTUレベルでの細菌分類群の相対的存在量を示す、グラフである。図中の表記は図5Cと同様である。It is a graph which shows the relative abundance of a bacterial taxon at the OTU level in the small intestinal mucosa. The notation in the figure is the same as that in FIG. 5C. SI細菌叢のα多様性指数を示すグラフである。図中、CDはクローン病患者由来のサンプルを解析した結果を示し、non-CDは非クローン病患者由来のサンプルを解析した結果を示す。各グラフは、観測されたOTUの数、ACE、Chao1及びシャノン指数について解析した結果を各々示す。「Biopsy」及び「Scrape」は各々、生検及び擦過にて得られたSI粘膜サンプルを解析した結果であることを示す。各棒グラフは、グループの平均値を表す。対応がないスチューデントのt検定にて解析して、得られたP値を付記する。It is a graph which shows the α diversity index of SI bacterial flora. In the figure, CD shows the result of analyzing a sample derived from a Crohn's disease patient, and non-CD shows the result of analyzing a sample derived from a non-Crohn's disease patient. Each graph shows the results of analysis for the number of observed OTUs, ACE, Chao1 and Shannon index. "Biopsy" and "Scrape" indicate that they are the results of analysis of SI mucosal samples obtained by biopsy and scraping, respectively. Each bar graph represents the average value of the group. The P value obtained by analyzing with the unpaired Student's t-test is added. CD患者由来のSIサンプルと非CD患者由来のそれとの菌叢の構造的類似度を解析した結果を示す、二次元プロット図である。構造的類似度の比較は、Bray-Curtis距離ベース非計量多次元スケーリング(NMDS)プロットで可視化した。各ポイントは個々のサンプルを示す。「CD biopsy」及び「CD scrape」は、生検及び擦過にて得られたCD患者由来のSIサンプルを解析した結果を各々示す。「non-CD biopsy」及び「non-CD scrape」は、生検及び擦過にて得られた非CD患者由来のSIサンプルを解析した結果を各々示す。It is a two-dimensional plot figure which shows the result of having analyzed the structural similarity of the bacterial flora between the SI sample derived from a CD patient and that derived from a non-CD patient. Structural similarity comparisons were visualized on Bray-Curtis distance-based non-scaling multidimensional scaling (NMDS) plots. Each point indicates an individual sample. "CD biopsy" and "CD scrape" indicate the results of analysis of SI samples derived from CD patients obtained by biopsy and scraping, respectively. "Non-CD biopsy" and "non-CD scrape" show the results of analysis of SI samples derived from non-CD patients obtained by biopsy and scraping, respectively. 小腸粘膜(SI mucosa)における、門(Phylum)レベルでの細菌分類群の相対的存在量を示す、グラフである。図中、横軸において、各細菌門に関する解析結果を、左から順に、生検にて得られた非CD患者由来のSIサンプル、生検にて得られたCD患者由来のSIサンプル、擦過にて得られた非CD患者由来のSIサンプル、及び、擦過にて得られたCD患者由来のSIサンプルについて、各々示す。各棒グラフは、グループの平均値を表す。エラーバーはSDを表す。各アスタリスクは、FDRアプローチによる多重t検定によって得られた値が、*<0.05;**0.01未満;***≦0.001;****<0.0001であることを各々示す。It is a graph which shows the relative abundance of the bacterial taxon at the phylum (Phylum) level in the small intestinal mucosa (SI mucosa). In the figure, on the horizontal axis, the analysis results for each bacterial phylum are shown in order from left to right: SI sample from non-CD patient obtained by biopsy, SI sample from CD patient obtained by biopsy, and scraping. The SI sample derived from a non-CD patient obtained from the above and the SI sample derived from a CD patient obtained by scraping are shown. Each bar graph represents the average value of the group. Error bars represent SD. For each asterisk, the value obtained by the multiple t-test by the FDR approach is *** 0.05; less than ** 0.01; *** ≤ 0.001; *** <0.0001. Each is shown. 小腸粘膜における、科(Family)レベルでの細菌分類群の相対的存在量を示す、グラフである。図中の表記は図5Hと同様である。FIG. 5 is a graph showing the relative abundance of bacterial taxa at the Family level in the mucosa of the small intestine. The notation in the figure is the same as that in FIG. 5H. 小腸粘膜における、OTUレベルでの細菌分類群の相対的存在量を示す、グラフである。図中の表記は図5Hと同様である。It is a graph which shows the relative abundance of a bacterial taxon at the OTU level in the small intestinal mucosa. The notation in the figure is the same as that in FIG. 5H. CD患者(CD)及び非CD患者(non-CD)における、CD患者に豊富な18種の細菌の保有率(Prevalence(%))を示す、ヒストグラムである。保有率は、SI粘膜サンプルに0.1%を超える相対量の細菌が含まれる被験者数を被験者総数で割ることにより算出した。図中、横軸において、各細菌についての結果を示す(各左側は、非CD患者由来のサンプル解析結果を示し、各右側はCD患者由来のサンプル解析結果を示す)。各棒グラフはグループの平均を表す。エラーバーは95%信頼区間を表す。各アスタリスクは、カイ二乗検定によって得られた値が、*<0.05;**<0.01;***<0.001であることを示す。It is a histogram which shows the prevalence (prevalence (%)) of 18 kinds of bacteria abundant in CD patients in CD patients (CD) and non-CD patients (non-CD). The prevalence was calculated by dividing the number of subjects whose SI mucosal sample contained a relative amount of bacteria exceeding 0.1% by the total number of subjects. In the figure, the horizontal axis shows the results for each bacterium (each left side shows the sample analysis result derived from a non-CD patient, and each right side shows the sample analysis result derived from a CD patient). Each bar graph represents the average of the groups. Error bars represent 95% confidence intervals. Each asterisk indicates that the value obtained by the chi-square test is *** <0.05; ** <0.01; *** <0.001. SI粘膜細菌叢における、科レベルの細菌分類群での相対的存在量を示すグラフである。図中、横軸において、各細菌科についての結果を示す(各左側は、非CD患者由来のサンプル解析結果を示し、各右側は抗TNF-α抗体療法を受けているCD患者由来のサンプル解析結果を示し、真ん中はその他のCD患者由来のサンプル解析結果を示す)。各太いバーはグループの平均を表す。各三角は個々のサンプルを表す。エラーバーは95%信頼区間を表す。各アスタリスクは、誤判定率を用いた多重t検定によって得られた値が、*<0.05;**<0.01;***<0.001;****<0.0001であることを示す。It is a graph which shows the relative abundance in a family-level bacterial taxon in the SI mucosal flora. In the figure, the horizontal axis shows the results for each bacteriological family (each left side shows the sample analysis results from non-CD patients, and each right side shows the sample analysis from CD patients receiving anti-TNF-α antibody therapy. The results are shown, and the results of sample analysis from other CD patients are shown in the middle). Each thick bar represents the average of the group. Each triangle represents an individual sample. Error bars represent 95% confidence intervals. For each asterisk, the value obtained by the multiple t-test using the misjudgment rate is *** <0.05; *** <0.01; *** <0.001; *** <0.0001. Show that. SI粘膜、唾液及び糞便における菌叢の構造的類似度を解析した結果を示す、二次元プロット図である。構造的類似度の比較は、Bray-Curtis距離ベース非計量多次元スケーリング(NMDS)プロットで可視化し、解剖学的位置間に関し、置換多変量分散分析(PERMANOVA)解析にて検定を行なった。各ポイントは、個々のCD患者由来のサンプル(CD)を表す。各三角は、個々の非CD患者由来のサンプル(non-CD)を表す。カラー表示下、赤色は唾液サンプル(1.Saliva)を表し、青色はSI粘膜サンプル(2.SI)を表し、緑色は糞便サンプル(3.Feces)を表す。It is a two-dimensional plot figure which shows the result of having analyzed the structural similarity of the bacterial flora in SI mucosa, saliva and feces. Structural similarity comparisons were visualized on a Bray-Curtis distance-based non-scaling multidimensional scaling (NMDS) plot and tested for anatomical positions by a substituted multivariate analysis of variance (PERMANOVA) analysis. Each point represents a sample (CD) from an individual CD patient. Each triangle represents a sample (non-CD) from an individual non-CD patient. Under color display, red represents saliva sample (1. Saliva), blue represents SI mucosal sample (2. SI), and green represents fecal sample (3. Feces). SI細菌叢のα多様性指数を示すグラフである。図中、「CD」はクローン病患者由来のサンプルを解析した結果を示し、「non-CD」は非クローン病患者由来のサンプルを解析した結果を示す。各グラフは、OTUの数、ACE、Chao1及びシャノン指数について解析した結果を各々示す。「Saliva」及び「Feces」は各々、唾液サンプル及び糞便サンプルを解析した結果であることを示す。各棒グラフはグループの平均を表す。各ポイントは個々のサンプルを表す。エラーバーは95%信頼区間を表す。各アスタリスクは、対応のないスチューデントのt検定によって得られた値が、*<0.05;**<0.01;***<0.001;****<0.0001であることを示す。「ns」は有意差なし(P値>0.05)であることを表す。It is a graph which shows the α diversity index of SI bacterial flora. In the figure, "CD" indicates the result of analyzing a sample derived from a Crohn's disease patient, and "non-CD" indicates the result of analyzing a sample derived from a non-Crohn's disease patient. Each graph shows the results of analysis for the number of OTUs, ACE, Chao1 and Shannon index. "Saliva" and "Feces" indicate that they are the results of analysis of saliva sample and fecal sample, respectively. Each bar graph represents the average of the groups. Each point represents an individual sample. Error bars represent 95% confidence intervals. For each asterisk, the value obtained by the unpaired Student's t-test is *** <0.05; *** <0.01; *** <0.001; *** <0.0001. Is shown. “Ns” indicates that there is no significant difference (P value> 0.05). CD患者由来の糞便サンプル(図中「CD」)と非CD患者のそれ(図中「non-CD」)とを比較して計算した結果を示す、誤判定率のヒストグラムである。図中、有意な分類群(FDR≦0.05)のみを示した。FDRにおいて、マイナス側に非CD患者についての解析結果が示され、プラス側にCD患者についての解析結果が示される。It is a histogram of the misjudgment rate which shows the result calculated by comparing the stool sample derived from a CD patient (“CD” in the figure) with that of a non-CD patient (“non-CD” in the figure). In the figure, only significant taxa (FDR ≤ 0.05) are shown. In FDR, the analysis result for non-CD patients is shown on the minus side, and the analysis result for CD patients is shown on the plus side. 唾液、SI粘膜及び糞便中のF.prausnitziiの相対的存在量を示す、グラフである。図中、「Saliva」、「SI」及び「Feces」は、唾液、SI粘膜及び糞便を解析した結果を示す。また、各サンプルの解析結果において、各左側は非CD患者由来のサンプル解析結果(non-CD)を示し、各右側はCD患者由来のサンプル解析結果(CD)を示す。各太いバーはグループの平均を表す。各ポイントは個々のサンプルを表す。エラーバーは95%信頼区間を表す。各アスタリスクは、対応のないスチューデントのt検定によって得られた値が、*<0.05;**<0.01;***<0.001;****<0.0001であることを示す。F. in saliva, SI mucosa and feces. It is a graph which shows the relative abundance of plausnitzii. In the figure, "Saliva", "SI" and "Feces" indicate the results of analysis of saliva, SI mucosa and feces. In the analysis results of each sample, each left side shows a sample analysis result (non-CD) derived from a non-CD patient, and each right side shows a sample analysis result (CD) derived from a CD patient. Each thick bar represents the average of the group. Each point represents an individual sample. Error bars represent 95% confidence intervals. For each asterisk, the value obtained by the unpaired Student's t-test is *** <0.05; *** <0.01; *** <0.001; *** <0.0001. Is shown. SI粘膜(SI mucosa)における、OTUレベルの細菌分類群での相対的存在量を示すヒストグラムである。図中、横軸において、各OTUについての結果を示す(各左側は、非CD患者由来のサンプル解析結果を示し、各右側はCD患者由来のサンプル解析結果を示す)。各バーはグループの平均を示す。エラーバーはSDを表す。各アスタリスクは、FDRを用いた多重t検定によって得られた値が、*FDR<0.05;**FDR<0.01;***FDR<0.001;****FDR<0.0001であることを示す。It is a histogram which shows the relative abundance in the bacterial taxon of OTU level in SI mucosa (SI mucosa). In the figure, the horizontal axis shows the results for each OTU (each left side shows the sample analysis result derived from a non-CD patient, and each right side shows the sample analysis result derived from a CD patient). Each bar shows the average for the group. Error bars represent SD. For each asterisk, the value obtained by multiple t-test using FDR is * FDR <0.05; ** FDR <0.01; *** FDR <0.001; *** FDR <0. Indicates that it is 0001. 結腸粘膜(colonic mucosa)における、OTUレベルの細菌分類群での相対的存在量を示すヒストグラムである。図中の表記については、図7Aと同様である。It is a histogram which shows the relative abundance in the bacterial taxon of OTU level in the colon mucosa (colonic mucosa). The notation in the figure is the same as in FIG. 7A. 糞便(feces)における、OTUレベルの細菌分類群での相対的存在量を示すヒストグラムである。図中の表記については、図7Aと同様である。図7A~7Cに示した結果から、14のOTUだけが有意にCD患者のSI粘膜に豊富に存在していることが示唆される。9 is a histogram showing the relative abundance of OTU levels in a bacterial taxon in feces. The notation in the figure is the same as in FIG. 7A. The results shown in FIGS. 7A-7C suggest that only 14 OTUs are significantly abundant in the SI mucosa of CD patients. CD患者のSIサンプルから、CDと有意に関連する細菌株を選抜した工程を示す、概略図である。It is a schematic diagram which shows the process of selecting the bacterial strain significantly associated with CD from the SI sample of a CD patient. CD患者のSIサンプルから単離した80菌についての、16SrRNAシーケンス結果に基づき作成した、系統樹である。A phylogenetic tree prepared based on 16S rRNA sequencing results for 80 bacteria isolated from SI samples of CD patients. in vitro培養したE.coli 35A1株をグラム染色にて解析した代表的な結果を示す、写真である。スケールバーは20μmを表す。In vitro cultured E. It is a photograph which shows the typical result of analysis of the coli 35A1 strain by Gram stain. The scale bar represents 20 μm. 生化学実験(TSI斜面培地での培養試験、LIM/SIM培地での培養試験、API 20 E test strip)において、E.coli 35A1株等が、糖発酵、運動性及びインドール産生にて陽性であることを示す、写真である。In biochemical experiments (culture test on TSI slope medium, culture test on LIM / SIM medium, API 20 E test strip), E.I. It is a photograph showing that the coli 35A1 strain and the like are positive in sugar fermentation, motility and indole production. フローサイトメトリー解析の工程を示す、概略図である。先ず、A及びBに示すように、腸粘膜固有層からデブリを除くためにゲートをかけた。さらに、単一細胞にゲートをかけた。次に、Cに示すように、生細胞にGhost red 780でゲートをかけた。D及びEに示すように、TCRβ陽性細胞及びCD4陽性細胞にゲートをかけた。CD4T細胞は生細胞ゲート中のCD4+TCRβ+のポピュレーションとして定義した。It is the schematic which shows the process of the flow cytometry analysis. First, as shown in A and B, a gate was applied to remove debris from the lamina propria of the intestinal mucosa. In addition, single cells were gated. Next, as shown in C, live cells were gated with Ghost red 780. TCRβ-positive and CD4-positive cells were gated as shown in D and E. CD4T cells were defined as the population of CD4 + TCRβ + in living cell gates. E.coli 35A1株のみを定着させたマウスの、SI、盲腸(図中、「Cecum」)及び糞便(図中、「Feces」)における、当該菌株のDNA濃度を示すグラフである。前記マウスの、糞便ペレット、並びに、盲腸及びSIの管腔内容物から、DNAを抽出し、qPCRにて菌のDNA濃度を決定した。各ポイントは個々のマウスを表す。各太いバーは、グループの平均を表す。エラーバーはSDを表す。各アスタリスクは、Tukeyの事後検定と併用する一元配置分散分析(one-way ANOVA)によって得られた値が、*P<0.05;**P<0.01;***P<0.001であることを示す。E. 3 is a graph showing the DNA concentration of the strain in SI, cecum (“Cecum” in the figure) and feces (“Feces” in the figure) of mice in which only the coli 35A1 strain has been established. DNA was extracted from the fecal pellets of the mice and the lumen contents of the cecum and SI, and the DNA concentration of the fungus was determined by qPCR. Each point represents an individual mouse. Each thick bar represents the average of the group. Error bars represent SD. For each asterisk, the value obtained by one-way analysis of variance (one-way ANOVA) used in combination with Tukey's post-test is * P <0.05; ** P <0.01; *** P <0. Indicates that it is 001. 結腸粘膜固有層におけるCD4T細胞中のIFN-γ+細胞の割合を示すグラフである。E.coli 35A1株を単独で経口投与したGFマウス(図中「+35A1」)及び前記9-mixから当該E.coli株を除いたものを経口投与したGFマウス(図中「+8mix」)を解析した結果を示す。各ポイントは個々のマウスを表す。各太いバーは、グループの平均を表す。エラーバーはSDを表す。各アスタリスクは、対応のないスチューデントのt検定によって得られた値が、*P<0.05;**P<0.01;***P<0.001であることを示す。It is a graph which shows the ratio of IFN-γ + cell in CD4T cell in the colon lamina propria. E. The E. coli 35A1 strain was orally administered alone from the GF mouse (“+ 35A1” in the figure) and the 9-mix. The result of analysis of the GF mouse (“+8 mix” in the figure) to which the one excluding the coli strain was orally administered is shown. Each point represents an individual mouse. Each thick bar represents the average of the group. Error bars represent SD. Each asterisk indicates that the value obtained by the unpaired Student's t-test is * P <0.05; ** P <0.01; *** P <0.001. マウスの近位結腸をヘマトキシリン・エオジン染色にて解析した、代表的な結果を示す、写真である。E.coli 35A1株を、1日目にGFマウスに投与し、抗マウスIL-10R抗体(1mg/個体)を1日目から実験終了まで、毎週、腹腔内投与した。当該マウスの結果を「GF+35A1+anti-IL10R」に示す。「GF」はE.coli 35A1株及び抗マウスIL-10R抗体を投与しなかったGFマウスの結果を示し、「GF+35A1」は抗マウスIL-10R抗体を投与しなかった、E.coli 35A1株を定着させたGFマウスの結果を示し、「GF+anti-IL10R」は、E.coli 35A1株を投与せずに、抗マウスIL-10R抗体を投与したGFマウスの結果を示す。It is a photograph showing typical results of analysis of the proximal colon of a mouse by hematoxylin and eosin staining. E. The coli 35A1 strain was administered to GF mice on the first day, and the anti-mouse IL-10R antibody (1 mg / individual) was intraperitoneally administered every week from the first day to the end of the experiment. The results of the mouse are shown in "GF + 35A1 + anti-IL10R". "GF" is E.I. The results of GF mice to which the colli 35A1 strain and the anti-mouse IL-10R antibody were not administered are shown, and "GF + 35A1" was not administered with the anti-mouse IL-10R antibody. The results of GF mice in which the colli 35A1 strain was established are shown, and "GF + anti-IL10R" is described in E. coli. The results of the GF mouse to which the anti-mouse IL-10R antibody was administered without administering the coli 35A1 strain are shown. マウスの近位結腸(Proximal colon)における組織的大腸菌炎スコアを解析した結果を示す、グラフである。E.coli 35A1株、E.coli LF82株又はE.coli MG1655株を、1日目にGFマウスに投与し、抗マウスIL-10R抗体(1mg/個体)を1日目から実験終了まで、毎週、腹腔内投与した。これらのマウスの結果を「+anti-IL10R +35A1」、「+anti-IL10R +LF82」及び「+anti-IL10R +MG1655」に各々示す。「GF」はE.coli株及び抗マウスIL-10R抗体を投与しなかったGFマウスの結果を示し、「GF+35A1」は抗マウスIL-10R抗体を投与しなかった、E.coli 35A1株を定着させたGFマウスの結果を示し、「+anti-IL10R GF」は、E.coli株を投与せずに、抗マウスIL-10R抗体を投与したGFマウスの結果を示す。各ポイントは個々のマウスを表す。各太いバーは、グループの平均を表す。エラーバーはSDを表す。各アスタリスクは、Tukeyの事後検定と併用する一元配置分散分析によって得られた値が、*P<0.05、**P<0.01、***P<0.001であることを示す。It is a graph which shows the result of having analyzed the histological Escherichia coli inflammation score in the proximal colon (Proximal colon) of a mouse. E. colli 35A1 strain, E.I. colli LF82 strain or E.I. The coli MG1655 strain was administered to GF mice on the first day, and the anti-mouse IL-10R antibody (1 mg / individual) was intraperitoneally administered every week from the first day to the end of the experiment. The results of these mice are shown in "+ anti-IL10R + 35A1", "+ anti-IL10R + LF82" and "+ anti-IL10R + MG1655", respectively. "GF" is E.I. The results of GF mice to which the colli strain and the anti-mouse IL-10R antibody were not administered are shown, and "GF + 35A1" was not administered with the anti-mouse IL-10R antibody. The results of GF mice in which the colli 35A1 strain was established are shown, and "+ anti-IL10R GF" is described in E. coli. The results of the GF mouse to which the anti-mouse IL-10R antibody was administered without the administration of the coli strain are shown. Each point represents an individual mouse. Each thick bar represents the average of the group. Error bars represent SD. Each asterisk indicates that the values obtained by one-way ANOVA in combination with Tukey's post-test are * P <0.05, ** P <0.01, *** P <0.001. .. マウスの遠位結腸をヘマトキシリン・エオジン染色にて解析した、代表的な結果を示す、写真である。図中の表記については、図11Cと同様である。It is a photograph showing a representative result of analysis of the distal colon of a mouse by hematoxylin and eosin staining. The notation in the figure is the same as in FIG. 11C. マウスの遠位結腸(Distal colon)における組織的大腸菌炎スコアを解析した結果を示す、グラフである。図中の表記については、図11Dと同様である。It is a graph which shows the result of having analyzed the histological Escherichia coli inflammation score in the distal colon of a mouse. The notation in the figure is the same as in FIG. 11D.
 <本発明の小腸内細菌>
 後述の実施例において示すとおり、本発明者らによって、クローン病の患者から単離された9株の小腸内細菌は、クローン病との有意な関連性が認められ、さらにTh1細胞及び/又はTh17細胞(Th1細胞等)の増殖又は活性化を誘導し得ることが、明らかとなった。
<Small intestinal bacteria of the present invention>
As shown in Examples below, nine strains of small intestinal bacteria isolated from patients with Crohn's disease were found to be significantly associated with Crohn's disease, and Th1 cells and / or Th17. It has become clear that it can induce proliferation or activation of cells (Th1 cells, etc.).
 よって、本発明は、Th1細胞等の増殖又は活性化を誘導する小腸内細菌を提供するものであり、好ましくは、これら9種の細菌株の16SrRNA(V1-V2領域)をコードするDNA配列と95%以上の同一性を有する細菌を提供するものである。 Therefore, the present invention provides a small intestinal bacterium that induces proliferation or activation of Th1 cells and the like, and preferably has a DNA sequence encoding 16S rRNA (V1-V2 region) of these nine bacterial strains. It provides a bacterium having 95% or more identity.
 本発明において「小腸内細菌」としては、ヒトの小腸内に定着し得る細菌を意味する。ここで、当該細菌が定着する小腸の部位としては、特に制限はなく、回腸であってもよく、空腸であってもよい。また、前記細菌が定着する小腸の組織としては、特に制限はないが、通常、小腸の粘膜である。 In the present invention, "small intestinal bacteria" means bacteria that can colonize the human small intestine. Here, the site of the small intestine in which the bacterium is colonized is not particularly limited, and may be the ileum or the jejunum. The tissue of the small intestine to which the bacteria colonize is not particularly limited, but is usually the mucous membrane of the small intestine.
 本発明において、「Th1細胞」とは、CD4陽性のヘルパーT細胞(Th細胞)の亜群であり、細胞性免疫を増強させる細胞を意味する。また、「Th1細胞の活性」とは、該細胞によるTh1サイトカイン(IFN-γ等)の産生、該サイトカインによるマクロファージ、細胞傷害性T細胞(CTL)等の細胞の活性化、該活性化による細胞性免疫の増強を含む意味である。さらに、「Th1細胞の増殖又は活性化の誘導」とは、Th1細胞の増殖又は活性化に至る、ナイーブT細胞からTh1細胞への分化誘導も含む意味である。また、Th1細胞の集積(蓄積)も含まれる。 In the present invention, "Th1 cell" is a subgroup of CD4 positive helper T cells (Th cells) and means a cell that enhances cell-mediated immunity. “Th1 cell activity” refers to the production of Th1 cytokines (IFN-γ, etc.) by the cells, the activation of cells such as macrophages and cytotoxic T cells (CTL) by the cytokines, and the cells due to the activation. It means that it includes enhancement of sexual immunity. Further, "induction of proliferation or activation of Th1 cells" means that induction of differentiation from naive T cells to Th1 cells leading to proliferation or activation of Th1 cells is also included. It also includes the accumulation (accumulation) of Th1 cells.
 本発明において、「Th17細胞」とは、CD4陽性のヘルパーT細胞(Th細胞)の亜群であり、炎症を誘導する細胞を意味する。また、「Th17細胞の活性」とは、該細胞による炎症性サイトカイン(IL-17、IL-21、IL-22、TNF-α等)の産生、該サイトカインによる炎症の誘導を含む意味である。さらに、「Th17細胞の増殖又は活性化の誘導」とは、Th17細胞の増殖又は活性化に至る、ナイーブT細胞からTh17細胞への分化誘導も含む意味である。また、Th17細胞の集積(蓄積)も含まれる。 In the present invention, "Th17 cells" is a subgroup of CD4 positive helper T cells (Th cells) and means cells that induce inflammation. Further, "the activity of Th17 cells" means that the cells produce inflammatory cytokines (IL-17, IL-21, IL-22, TNF-α, etc.) and induce inflammation by the cytokines. Further, "induction of Th17 cell proliferation or activation" means to include induction of differentiation from naive T cells to Th17 cells, which leads to proliferation or activation of Th17 cells. It also includes the accumulation (accumulation) of Th17 cells.
 また、本発明の小腸内細菌は、1株の細菌であってもよく、複数株の細菌から構成される細菌株の混合物であってもよい。なお、複数株の細菌から構成される場合には、そのうちの少なくとも1の細菌株がTh1細胞等の増殖又は活性化を誘導する活性を有していることが望ましい。また、そのような場合、前記複数株の細菌には、前記誘導活性を有していない細菌株であっても、前記誘導活性を有している細菌株の当該活性を増強する作用を有する細菌株、前記誘導活性を有している細菌株の増殖を維持する作用を有する細菌株、前記誘導活性を阻害する細菌に対して当該阻害活性を抑制する作用を有する細菌株が含まれていてもよい。 Further, the small intestinal bacterium of the present invention may be a single strain of bacteria or a mixture of bacterial strains composed of a plurality of strains of bacteria. When composed of a plurality of strains of bacteria, it is desirable that at least one of the bacterial strains has an activity of inducing proliferation or activation of Th1 cells or the like. Further, in such a case, the plurality of strains of bacteria have an action of enhancing the activity of the bacterial strain having the inducing activity, even if the bacterial strain does not have the inducing activity. Even if a strain, a bacterial strain having an action of maintaining the growth of the bacterial strain having the inducible activity, and a bacterial strain having an action of suppressing the inhibitory activity against a bacterium that inhibits the inducible activity are included. good.
 かかる本発明の小腸内細菌として、配列番号:1~9のうちのいずれかに記載のDNA配列に対して95%以上の同一性を有するポリヌクレオチドを有する細菌が挙げられる。 Examples of the small intestinal bacterium of the present invention include a bacterium having a polynucleotide having 95% or more identity with respect to the DNA sequence set forth in any one of SEQ ID NOs: 1 to 9.
 配列番号:1~9に記載のDNA配列は各々、後述の実施例において示される、Escherichia coli(菌株ID:35A1)、Ruminococcus gnavus(菌株ID:131A1)、Bacteroides dorei(菌株ID:131H4)、Klebsiella pneumoniae(菌株ID:39F4)、Streptococcus pasteurianus(菌株ID:133A7)、Parabacteroides distasonis(菌株ID:134F1)、Bacteroides fragilis(菌株ID:32E9)、Erysipelatoclostridium ramosum(菌株ID:131A10)、及びBacteroides uniformis(菌株ID:131A11)の16SrRNA(V1-V2領域)をコードするDNA配列である。 The DNA sequences shown in SEQ ID NOs: 1 to 1, respectively, are Eschericia coli (strain ID: 35A1), Ruminococcus gnavus (strain ID: 131A1), Bacteroides doriei (strain ID: 131H4), and Klebsiella, respectively, which are shown in Examples described later. pneumoniae (strain ID: 39F4), Streptococcus pasteurianus (strain ID: 133A7), Parabacteroides distasonis (strain ID: 134F1), Bacteroides fragilis (strain ID: 32E9), Erym : 131A11) 16SrRNA (V1-V2 region) encoding DNA sequence.
 本発明において各配列番号によって規定されるDNA配列に対する「同一性」は、少なくとも95%であり、好ましくは96%以上、より好ましくは97%以上、さらに好ましくは98%以上、より好ましくは99%以上、特に好ましくは100%である。また、「同一性」は、BLAST等のアラインメントプログラムを利用して決定できる。例えば、ヌクレオチド配列の同一性とは、blastnを用いて算出されるヌクレオチド配列間の同一性が挙げられ、より具体的には、blastnをデフォルトのパラメータを用いて(すなわち初期設定のパラメータを用いて)算出されるヌクレオチド配列間の同一性が挙げられる。 In the present invention, the "identity" with respect to the DNA sequence defined by each SEQ ID NO: is at least 95%, preferably 96% or more, more preferably 97% or more, still more preferably 98% or more, more preferably 99%. As mentioned above, it is particularly preferably 100%. Further, "identity" can be determined by using an alignment program such as BLAST. For example, nucleotide sequence identity includes identity between nucleotide sequences calculated using blastn, and more specifically, blastn with default parameters (ie, with default parameters). ) The calculated identity between the nucleotide sequences can be mentioned.
 また、かかる細菌は、各配列番号によって規定されるDNA配列に対して少なくとも95%の同一性を有するヌクレオチドを保有する細菌であればよく、それらの種は特定されるものではないが、
配列番号:1に記載のDNA配列に対して95%以上の同一性を有するポリヌクレオチドを有する細菌は、好ましくは、Escherichia coliに属する細菌であり、
配列番号:2に記載のDNA配列に対して95%以上の同一性を有するポリヌクレオチドを有する細菌は、好ましくは、Ruminococcus gnavusに属する細菌であり、
配列番号:3に記載のDNA配列に対して95%以上の同一性を有するポリヌクレオチドを有する細菌は、好ましくは、Bacteroides doreiに属する細菌であり、
配列番号:4に記載のDNA配列に対して95%以上の同一性を有するポリヌクレオチドを有する細菌は、好ましくは、Klebsiella pneumoniaeに属する細菌であり、
配列番号:5に記載のDNA配列に対して95%以上の同一性を有するポリヌクレオチドを有する細菌は、好ましくは、Streptococcus pasteurianusに属する細菌であり、
配列番号:6に記載のDNA配列に対して95%以上の同一性を有するポリヌクレオチドを有する細菌は、好ましくは、Parabacteroides distasonisに属する細菌であり、
配列番号:7に記載のDNA配列に対して95%以上の同一性を有するポリヌクレオチドを有する細菌は、好ましくは、Bacteroides fragilisに属する細菌であり、
配列番号:8に記載のDNA配列に対して95%以上の同一性を有するポリヌクレオチドを有する細菌は、好ましくは、Erysipelatoclostridium ramosumに属する細菌であり、
配列番号:9に記載のDNA配列に対して95%以上の同一性を有するポリヌクレオチドを有する細菌は、好ましくは、Bacteroides uniformisに属する細菌である。
Further, such a bacterium may be a bacterium having a nucleotide having at least 95% identity with respect to the DNA sequence defined by each SEQ ID NO:, and their species are not specified.
Bacteria having a polynucleotide having 95% or more identity with respect to the DNA sequence shown in SEQ ID NO: 1 are preferably bacteria belonging to Escherichia coli.
Bacteria having a polynucleotide having 95% or more identity with respect to the DNA sequence shown in SEQ ID NO: 2 are preferably bacteria belonging to Ruminococcus gnavus.
Bacteria having a polynucleotide having 95% or more identity with respect to the DNA sequence shown in SEQ ID NO: 3 are preferably bacteria belonging to Bacteroides doriei.
Bacteria having a polynucleotide having 95% or more identity with respect to the DNA sequence shown in SEQ ID NO: 4 are preferably bacteria belonging to Klebsiella pneumoniae.
Bacteria having a polynucleotide having 95% or more identity with respect to the DNA sequence shown in SEQ ID NO: 5 are preferably bacteria belonging to Streptococcus pasteurianus.
Bacteria having a polynucleotide having 95% or more identity with respect to the DNA sequence shown in SEQ ID NO: 6 are preferably bacteria belonging to Parabacteroides distasonis.
Bacteria having a polynucleotide having 95% or more identity with respect to the DNA sequence shown in SEQ ID NO: 7 are preferably bacteria belonging to Bacteroides fragilis.
Bacteria having a polynucleotide having 95% or more identity with respect to the DNA sequence shown in SEQ ID NO: 8 are preferably bacteria belonging to the Erysiperatoclastridium ramosom.
Bacteria having a polynucleotide having 95% or more identity with respect to the DNA sequence shown in SEQ ID NO: 9 are preferably bacteria belonging to Bacteroides uniformis.
 これら配列番号によって特定される細菌に関し、本発明の小腸内細菌としては、前記9種のうちの少なくとも1の細菌であればよいが、小腸内においてTh1細胞及び/又はTh17細胞の増殖又は活性化をより誘導し易いという観点から、好ましくは、配列番号:1に記載のDNA配列に対して95%以上の同一性を有するポリヌクレオチドを有する細菌、配列番号:2に記載のDNA配列に対して95%以上の同一性を有するポリヌクレオチドを有する細菌であり、より好ましくは、配列番号:1に記載のDNA配列に対して95%以上の同一性を有するポリヌクレオチドを有する細菌である。 Regarding the bacteria specified by these SEQ ID NOs, the bacteria in the small intestine of the present invention may be at least one of the above nine types, but the proliferation or activation of Th1 cells and / or Th17 cells in the small intestine. From the viewpoint of being easier to induce, preferably with respect to a bacterium having a polynucleotide having 95% or more identity with respect to the DNA sequence set forth in SEQ ID NO: 1, and the DNA sequence set forth in SEQ ID NO: 2. A bacterium having a polynucleotide having 95% or more identity, and more preferably a bacterium having a polynucleotide having 95% or more identity to the DNA sequence set forth in SEQ ID NO: 1.
 また、本発明の小腸内細菌としては、前記9種のうちの2種、3種、4種、5種、6種、7種、8種又は全ての組み合わせであってもよいが、後述の実施例に示すとおり、組み合わせて用いることにより、Th17細胞の増殖又は活性化が相乗的に誘導され得るという観点から、好ましくは、配列番号:1に記載のDNA配列に対して95%以上の同一性を有するポリヌクレオチドを有する細菌と配列番号:2に記載のDNA配列に対して95%以上の同一性を有するポリヌクレオチドを有する細菌との組み合わせである。 Further, the small intestinal bacteria of the present invention may be 2 types, 3 types, 4 types, 5 types, 6 types, 7 types, 8 types or all combinations of the above 9 types, which will be described later. As shown in Examples, from the viewpoint that proliferation or activation of Th17 cells can be synergistically induced by using them in combination, preferably, 95% or more of the same DNA sequence as shown in SEQ ID NO: 1 is used. It is a combination of a bacterium having a polynucleotide having sex and a bacterium having a polynucleotide having 95% or more identity with respect to the DNA sequence shown in SEQ ID NO: 2.
 <本発明のスクリーニング方法>
 本発明は、以下に示す第1のスクリーニング方法を提供する。
<Screening method of the present invention>
The present invention provides the first screening method shown below.
 小腸内でTh1細胞及び/又はTh17細胞の増殖又は活性化を抑制する物質をスクリーニングする方法であって、
(1)被験物質を非ヒト無菌動物に摂取させる工程、
(2)該非ヒト動物の小腸内におけるTh1細胞及び/又はTh17細胞の数又は活性を検出する工程、及び
(3)工程(2)にて前記細胞の増殖又は活性化の抑制が検出された場合、前記被験物質は、小腸内でTh1細胞及び/又はTh17細胞の増殖又は活性化を抑制する物質であると判定する工程を含む方法。
A method of screening for substances that suppress the proliferation or activation of Th1 cells and / or Th17 cells in the small intestine.
(1) A step of ingesting a test substance into a non-human germ-free animal,
(2) When the number or activity of Th1 cells and / or Th17 cells in the small intestine of the non-human animal is detected, and when suppression of proliferation or activation of the cells is detected in (3) step (2). A method including a step of determining that the test substance is a substance that suppresses the proliferation or activation of Th1 cells and / or Th17 cells in the small intestine.
 さらに、後述の実施例に示すとおり、本発明者らによって、小腸内におけるTh1細胞等の数又は活性は、上述の小腸内細菌を定着させることによって増強されるものであることを明らかにしている。 Furthermore, as shown in Examples described later, the present inventors have clarified that the number or activity of Th1 cells and the like in the small intestine is enhanced by colonizing the above-mentioned small intestinal bacteria. ..
 したがって、本発明はまた、下記第2のスクリーニング方法も提供する。 Therefore, the present invention also provides the following second screening method.
 小腸内でTh1細胞及び/又はTh17細胞の増殖又は活性化を抑制する物質をスクリーニングする方法であって、
(1)被験物質を、配列番号:1に記載のDNA配列に対して95%以上の同一性を有するポリヌクレオチドを有する細菌及び/又は配列番号:2に記載のDNA配列に対して95%以上の同一性を有するポリヌクレオチドを有する細菌を定着させた非ヒト動物に、摂取させる工程、
(2)該非ヒト動物の小腸内における前記細菌の数を検出する工程、及び
(3)工程(2)にて前記細菌の増殖抑制が検出された場合、前記被験物質は、小腸内でTh1細胞及び/又はTh17細胞の増殖又は活性化を抑制する物質であると判定する工程を含む方法。
A method of screening for substances that suppress the proliferation or activation of Th1 cells and / or Th17 cells in the small intestine.
(1) The test substance is a bacterium having a polynucleotide having a polynucleotide having 95% or more identity with respect to the DNA sequence set forth in SEQ ID NO: 1 and / or 95% or more with respect to the DNA sequence set forth in SEQ ID NO: 2. Ingestion of non-human animals colonized with a polynucleotide having a polynucleotide having the same identity as
When (2) the step of detecting the number of the bacteria in the small intestine of the non-human animal and (3) the suppression of the growth of the bacteria are detected in the step (2), the test substance is Th1 cells in the small intestine. And / or a method comprising the step of determining that the substance suppresses the proliferation or activation of Th17 cells.
 これらの本発明のスクリーニング方法に供される「被験物質」としては、小腸内でTh1細胞及び/又はTh17細胞の増殖又は活性化を抑制する物質を含有し得る限り、特に制限はないが、例えば、細胞(細菌、植物細胞、動物細胞等)、ファージ、並びにこれらの抽出液、培養物(培養上清等)、分泌産物、及び代謝産物が挙げられる。さらに、複数種の細胞又はファージから構成されるライブラリーの形態であっても良い。また例えば、合成低分子化合物、抗体、ポリペプチド、ポリヌクレオチド、脂質、糖類(単糖、二糖、オリゴ糖、糖鎖等)、海洋生物、植物又は動物由来の抽出物、土壌、及びこれら物質から構成されるライブラリー(ケミカルライブラリー、ランダムペプチドライブラリー、ファージディスプレイライブラリー等)が挙げられる。 The "test substance" used in these screening methods of the present invention is not particularly limited as long as it can contain a substance that suppresses the proliferation or activation of Th1 cells and / or Th17 cells in the small intestine, but for example. , Cells (bacteria, plant cells, animal cells, etc.), phages, and extracts, cultures (culture supernatants, etc.), secretory products, and metabolites thereof. Furthermore, it may be in the form of a library composed of a plurality of types of cells or phages. Also, for example, synthetic small molecule compounds, antibodies, polypeptides, polynucleotides, lipids, sugars (monosaccharides, disaccharides, oligosaccharides, sugar chains, etc.), marine organisms, plant or animal-derived extracts, soil, and these substances. Examples thereof include a library composed of (chemical library, random peptide library, phage display library, etc.).
 なお、本発明のスクリーニングに供される「細菌」としては、特に制限はなく、野生型であってもよく、変異体(遺伝子組換え体等)であってもよいが、小腸内に定着し得ることが望ましいという観点から、動物の小腸内に存在する細菌が挙げられる。かかる動物としては、ヒト、又は後述のヒト以外の動物(非ヒト動物)が挙げられる。かかる被験物質は、単離された細菌であってもよく、また複数種の細菌の混合物であってもよい。混合物として、より具体的に小腸内試料(例えば、前記動物の小腸内容物、前記動物の小腸洗浄液、又はそれらの培養物)が挙げられる。 The "bacterium" used for the screening of the present invention is not particularly limited and may be a wild type or a mutant (genetical recombinant or the like), but it is established in the small intestine. Bacteria present in the small intestine of animals can be mentioned from the viewpoint that it is desirable to obtain them. Examples of such animals include humans and non-human animals (non-human animals) described below. The test substance may be an isolated bacterium or a mixture of a plurality of types of bacteria. More specifically, the mixture includes a sample in the small intestine (for example, the contents of the small intestine of the animal, the small intestine lavage fluid of the animal, or a culture thereof).
 本発明にかかる「バクテリオファージ」とは、細菌に感染し溶解するウイルスを意味し、溶菌性バクテリオファージであってもよく、溶原性バクテリオファージであってもよいが、好ましくは、溶菌性バクテリオファージである。また、遺伝子組換えされたバクテリオファージ等の変異体であってもよい。さらにまた、かかる被験バクテリオファージは、単離されたバクテリオファージであってもよく、また複数種のバクテリオファージの混合物であってもよい。混合物として、より具体的に小腸内試料(例えば、前記動物の小腸内容物、前記動物の小腸洗浄液、又はそれらの培養物)が挙げられる。 The "bacteriophage" according to the present invention means a virus that infects and dissolves bacteria, and may be a lytic bacteriophage or a lysogenic bacteriophage, but a lytic bacteriophage is preferable. It is a phage. Further, it may be a mutant such as a genetically modified bacteriophage. Furthermore, the test bacteriophage may be an isolated bacteriophage or a mixture of multiple bacteriophages. More specifically, the mixture includes a sample in the small intestine (for example, the contents of the small intestine of the animal, the small intestine lavage fluid of the animal, or a culture thereof).
 また、配列番号:1に記載のDNA配列に対して95%以上の同一性を有するポリヌクレオチドを有する細菌の増殖抑制を試みるという観点から、被験バクテリオファージとして、Escherichia coliに対するファージが好適に用いられ、より具体的には、T系ファージ(T1、T2、T3、T4、T5、T6、T7等)、φX-174、λ、φX80、Qβ、P1等が挙げられる。 Further, from the viewpoint of attempting to suppress the growth of bacteria having a polynucleotide having a polynucleotide having 95% or more identity with respect to the DNA sequence set forth in SEQ ID NO: 1, a phage against Escherichia coli is preferably used as a test bacteriophage. More specifically, T-type phages (T1, T2, T3, T4, T5, T6, T7, etc.), φX-174, λ, φX80, Qβ, P1 and the like can be mentioned.
 また、配列番号:2に記載のDNA配列に対して95%以上の同一性を有するポリヌクレオチドを有する細菌の増殖抑制を試みるという観点から、被験バクテリオファージとして、Ruminococcus gnavusに対するファージが好適に用いられ、より具体的には、φRa02、φRa04等が挙げられる。 Further, from the viewpoint of attempting to suppress the growth of a bacterium having a polynucleotide having a polynucleotide having 95% or more identity with respect to the DNA sequence shown in SEQ ID NO: 2, a phage against Ruminococcus gnavus is preferably used as a test bacteriophage. More specifically, φRa02, φRa04 and the like can be mentioned.
 被験物質を摂取させる「非ヒト無菌動物」は、無菌条件下で、出生及び生育している、ヒト以外の動物を意味する。ヒト以外の動物としては、例えば、マウス、ラット、サル、ブタ、ウシ、ウマ、ヒツジ、ヤギ、ニワトリ、カモ、ダチョウ、アヒル、イヌ、ネコ、ウサギ、ハムスター等が挙げられるが、これらに制限されない。また、これら動物においては、マウスが好適に用いられる。 "Non-human germ-free animal" to ingest the test substance means a non-human animal that is born and growing under sterile conditions. Non-human animals include, but are not limited to, for example, mice, rats, monkeys, pigs, cows, horses, sheep, goats, chickens, ducks, ostriches, ducks, dogs, cats, rabbits, hamsters and the like. .. Moreover, in these animals, a mouse is preferably used.
 なお、本発明の第1のスクリーニング方法においては、非ヒト無菌動物の代わりに、菌を保有している非ヒト動物を用いてもよい。かかる非ヒト動物としては、例えば、特定病原体フリー(SPF)の非ヒト動物が挙げられる。 In the first screening method of the present invention, a non-human animal carrying the fungus may be used instead of the non-human germ-free animal. Such non-human animals include, for example, specific pathogen-free (SPF) non-human animals.
 また、本発明の第2のスクリーニング方法において、「配列番号:1に記載のDNA配列に対して95%以上の同一性を有するポリヌクレオチドを有する細菌及び/又は配列番号:2に記載のDNA配列に対して95%以上の同一性を有するポリヌクレオチドを有する細菌(以下「配列番号:1等によって規定される細菌」とも称する)を定着させた非ヒト動物」は、後述の実施例に示すように、例えば、前記非ヒト無菌動物に、配列番号:1等によって規定される細菌を投与(例えば、経口投与)し、定着させることで調製することができる。 Further, in the second screening method of the present invention, "a bacterium having a polynucleotide having a polynucleotide having 95% or more identity with respect to the DNA sequence set forth in SEQ ID NO: 1 and / or the DNA sequence set forth in SEQ ID NO: 2". A non-human animal in which a bacterium having a polynucleotide having a polynucleotide having 95% or more of the same identity (hereinafter, also referred to as “a bacterium defined by SEQ ID NO: 1 etc.”) has been established is as shown in Examples described later. In addition, for example, it can be prepared by administering (for example, oral administration) the bacteria defined by SEQ ID NO: 1 and the like to the non-human germ-free animal and colonizing the animal.
 工程(1)において、被験物質を、非ヒト動物に「摂取」させる方法としては特に制限はなく、通常、経口投与によって行われるが、非経口投与(例えば、腸管内への投与)であってもよい。また、非ヒト動物が配列番号:1等によって規定される細菌を定着させた非ヒト動物である場合には、被験物質と配列番号:1等によって規定される細菌との摂取は、同時であってもよく、被験物質を非ヒト無菌動物に摂取させた後に配列番号:1等によって規定される細菌を該動物に摂取させてもよく、配列番号:1等によって規定される細菌を非ヒト無菌動物に摂取させた後に被験物質を該動物に摂取させてもよい。 In step (1), there is no particular limitation on the method of "ingesting" the test substance to a non-human animal, and it is usually carried out by oral administration, but it is parenteral administration (for example, administration into the intestinal tract). May be good. In addition, when the non-human animal is a non-human animal in which the bacteria defined by SEQ ID NO: 1 etc. have been established, the ingestion of the test substance and the bacteria defined by SEQ ID NO: 1 etc. is simultaneous. Alternatively, the test substance may be ingested by a non-human germ-free animal and then the animal specified by SEQ ID NO: 1 etc. may be ingested, and the bacteria defined by SEQ ID NO: 1 etc. may be ingested by non-human germ-free animals. The test substance may be ingested by the animal after being ingested by the animal.
 工程(2)における、小腸内における「Th1細胞及び/又はTh17細胞の数の検出」は、これら細胞の数(量)そのものではなくともよく、例えば、当該細胞の量(数)を反映する当該細胞特有の物質の量を測定することによって行なうことができる。かかる物質としては、前記細胞特異的なオリゴヌクレオチド(例えば、各ヘルパーT細胞特有の転写産物)が挙げられる。 The "detection of the number of Th1 cells and / or Th17 cells" in the small intestine in the step (2) does not have to be the number (amount) of these cells itself, and reflects, for example, the amount (number) of the cells. This can be done by measuring the amount of cell-specific substances. Examples of such a substance include the cell-specific oligonucleotide (for example, a transcript specific to each helper T cell).
 さらに、「Th1細胞及び/又はTh17細胞の活性の検出」は、当該細菌が産生するサイトカイン量を測定することによって行なうことができる。Th1細胞が産生するサイトカインとしては、IFN-γ等のTh1サイトカインが挙げられ、またTh17細胞が産生するサイトカインとしては、例えば、炎症性サイトカイン(IL-17、IL-21、IL-22、TNF-α等)が挙げられる。 Furthermore, "detection of Th1 cell and / or Th17 cell activity" can be performed by measuring the amount of cytokine produced by the bacterium. Examples of cytokines produced by Th1 cells include Th1 cytokines such as IFN-γ, and examples of cytokines produced by Th17 cells include inflammatory cytokines (IL-17, IL-21, IL-22, TNF-). α, etc.).
 また、小腸内における「配列番号:1等によって規定される細菌の数の検出」は、前記細胞数の検出同様に、当該細菌の量(数)そのものではなくともよく、例えば、前記細菌の量(数)を反映する当該細菌特有の物質の量を測定することによって行なうことができる。かかる物質としては、前記細菌特異的なオリゴヌクレオチド(例えば、16SrRNA遺伝子、及びその転写物)、前記細菌特有の構成物質(当該細菌を構成する、ペプチド、核酸、糖、脂質、及びそれらの複合体等)、当該細菌による生成物(例えば、当該細菌の分泌産物、当該細菌による代謝産物)が挙げられる。 Further, "detection of the number of bacteria defined by SEQ ID NO: 1 etc." in the small intestine does not have to be the amount (number) of the bacteria itself, as in the detection of the cell number, for example, the amount of the bacteria. This can be done by measuring the amount of the bacterium-specific substance that reflects the (number). Such substances include the bacterium-specific oligonucleotide (for example, 16SrRNA gene and its transcript), the bacterium-specific constituent substance (peptides, nucleic acids, sugars, lipids, and complexes thereof that constitute the bacterium). Etc.), products of the bacterium (eg, secretory products of the bacterium, metabolites of the bacterium).
 なお、上記検出において測定される物質がオリゴヌクレオチドである場合には、例えば、PCR(RT-PCR、リアルタイムPCR、定量PCR)、DNAマイクロアレイ解析法、ノーザンブロッティング、次世代シーケンシング法(合成シーケンシング法(sequencing-by-synthesis、例えば、イルミナ社製Solexaゲノムアナライザー又はHiseq(登録商標)2000によるシーケンシング)、パイロシーケンシング法(例えば、ロッシュ・ダイアグノステックス(454)社製のシーケンサーGSLX又はFLXによるシーケンシング(所謂454シーケンシング))、リガーゼ反応シーケンシング法(例えば、ライフテクノロジー社製のSoliD(登録商標)又は5500xlによるシーケンシング))、T-RFLP法、ビーズアレイ法、in situ ハイブリダイゼーション、ドットブロット、RNaseプロテクションアッセイ法、質量分析法、ゲノムPCR法、サザンブロッティングを用いて、定量することができる。 When the substance measured in the above detection is an oligonucleotide, for example, PCR (RT-PCR, real-time PCR, quantitative PCR), DNA microarray analysis method, Northern blotting, next-generation sequencing method (synthetic sequencing). Method (sequencing-by-synthesis, eg, sequencing by Illumina Solexa genome analyzer or Hiseq® 2000), pyrosequencing method (eg, sequencer GSLX or FLX by Roche Diagnostics (454)). Sequencing by (so-called 454 sequencing)), rigase reaction sequencing method (for example, SoliD® manufactured by Life Technology Co., Ltd. or sequencing by 5500 xl), T-RFLP method, bead array method, in sit hybridization. , Dot blot, RNase protection assay, mass analysis, genomic PCR, and southern blotting can be used for quantification.
 その他の物質(サイトカインを含む)については、例えば、ELISA法、イムノブロッティング、抗体アレイ解析法、免疫組織化学的染色法、フローサイトメトリー、イメージングサイトメトリー、ラジオイムノアッセイ、免疫沈降法等の抗体を用いて検出する方法(免疫学的手法)を用いて、定量することができる。 For other substances (including cytokines), for example, antibodies such as ELISA method, immunoblotting, antibody array analysis method, immunohistochemical staining method, flow cytometry, imaging cytometry, radioimmunoassay, and immunoprecipitation method are used. It can be quantified using a method of detection (immunological method).
 また、このように定量して得られる値は、絶対量のみならず、相対値であってもよい。相対値としては、例えば、検出に用いる測定方法又は測定装置に基づく量比(所謂、任意単位(AU)で表される数値)が挙げられる。また、相対値としては、例えば、小腸内細菌叢全体におけるその細菌の割合を示す値(相対存在量、占有率)であってもよい。また、参照小腸内細菌の量を基準として算出した値を用いてもよい。本発明にかかる「参照小腸内細菌」は、小腸内において安定して存在しており、また異なる生体サンプル間において、その量の差が小さい細菌であればよい。 Further, the value obtained by quantifying in this way may be a relative value as well as an absolute quantity. Examples of the relative value include a quantitative ratio (so-called numerical value expressed in an arbitrary unit (AU)) based on the measuring method or measuring device used for detection. The relative value may be, for example, a value (relative abundance, occupancy) indicating the proportion of the bacterium in the entire small intestinal bacterial flora. Alternatively, a value calculated based on the amount of reference small intestinal bacteria may be used. The "reference small intestinal bacterium" according to the present invention may be a bacterium that is stably present in the small intestine and has a small difference in amount between different biological samples.
 なお、検出のタイミングとしては、特に制限はなく、当業者であれば、用いる動物の種類等に応じ、適宜調整し得る。 The timing of detection is not particularly limited, and a person skilled in the art can appropriately adjust the timing according to the type of animal used.
 工程(3)における、Th1細胞等の増殖又は活性化の「抑制」とは、被験物質を摂取させた非ヒト動物におけるこれらの値が、当該試料を摂取させていない非ヒト動物におけるそれらと比較して、有意に低減されていることを意味する。 “Inhibition” of proliferation or activation of Th1 cells or the like in step (3) means that these values in non-human animals fed with the test substance are compared with those in non-human animals not fed with the sample. This means that it is significantly reduced.
 また同様に、配列番号:1等によって規定される細菌の「増殖抑制」とは、被験物質を摂取させた非ヒト動物における配列番号:1等によって規定される細菌の数が、当該試料を摂取させていない非ヒト動物におけるそれらと比較して、有意に低減されていることを意味する。 Similarly, the "growth suppression" of the bacterium defined by SEQ ID NO: 1 etc. means that the number of bacteria defined by SEQ ID NO: 1 etc. in the non-human animal ingested the test substance ingests the sample. It means that it is significantly reduced compared to those in non-human animals that have not been fed.
 かかる比較は、当業者であれば、上記検出方法に合った統計学的解析方法を適宜選択して行うことができる。統計学的解析方法としては、例えば、t検定、ウィルコクソンの符号順位検定、分散分析(ANOVA)、クラスカル・ウォリス検定、マン・ホイットニーのU検定(ウィルコクソンの順位和検定)、オッズ比、ハザード比、フィッシャーの正確検定、受信者動作特性解析(ROC解析)、分類木と決定木解析(CART解析)が挙げられる。また、比較の際には、正規化された又は標準化かつ正規化されたデータを用いることもできる。 Those skilled in the art can appropriately select and perform a statistical analysis method suitable for the above detection method. Statistical analysis methods include, for example, t-test, Wilcoxon signed rank test, analysis of variance (ANOVA), Kruskal-Wallis test, Mann-Whitney U test (Wilcoxon rank sum test), odds ratio, hazard ratio, etc. Fisher's exact test, receiver operating characteristic analysis (ROC analysis), classification tree and decision tree analysis (CART analysis) can be mentioned. Normalized or standardized and normalized data can also be used for comparison.
 なお、本発明のスクリーニング方法において、1回の実施により、小腸内でTh1細胞及び/又はTh17細胞の増殖又は活性化を抑制する細菌又はバクテリオファージを選抜することができなかった場合には、得られた該細菌又はバクテリオファージを含む小腸内試料を、次なる被験物質として、新たな非ヒト動物に摂取させ、前述のスクリーニングを複数回行うことにより、前記細菌又はバクテリオファージを単離することができる。 In the screening method of the present invention, if it is not possible to select a bacterium or bacteriophage that suppresses the proliferation or activation of Th1 cells and / or Th17 cells in the small intestine by one execution, it is obtained. The bacterium or bacteriophage can be isolated by ingesting a new non-human animal as the next test substance and performing the above-mentioned screening a plurality of times. can.
 このようにして前記抑制が検出された非ヒト動物の小腸内試料の調製、及び細菌の単離は、後述の実施例に示すとおり、当業者であれば、公知の嫌気培養、好気培養を適宜選択し、行なうことができる。バクテリオファージの単離培養も当業者であれば、公知の手法を用い行なうことができる。かかる公知の手法としては、例えば、モレキュラー・クローニング 第4版 第1章に記載の方法、カレントプロトコール、1991年1月発行、13巻、1号に記載の方法、農業生物資源ジーンバンクが開示する方法(https://www.gene.affrc.go.jp/manuals-micro_phage.php)が挙げられる。 As shown in Examples described later, those skilled in the art can use known anaerobic cultures and aerobic cultures for the preparation of samples in the small intestine of non-human animals in which the suppression is detected and the isolation of bacteria. It can be selected and carried out as appropriate. Isolation and culturing of bacteriophage can also be carried out by those skilled in the art using known methods. As such a known method, for example, the method described in Chapter 1 of Molecular Cloning 4th Edition, Current Protocol, published in January 1991, Vol. 13, No. 1, the method described in Agricultural and Biological Resources Genebank discloses. A method (https://www.gene.affrc.go.jp/manuals-micro_page.php) can be mentioned.
 また、本発明は、後述の実施例に示すとおり、以下に示す第3のスクリーニング方法も提供する。 The present invention also provides a third screening method shown below, as shown in Examples described later.
 小腸内でTh1細胞及び/又はTh17細胞の増殖又は活性化を誘導する細菌をスクリーニングする方法であって、
(1)被験物質を非ヒト無菌動物に摂取させる工程と、
(2)該非ヒト動物の小腸内におけるTh1細胞及び/又はTh17細胞の数又は活性を検出する工程と、
(3)工程(2)にてTh1細胞及び/又はTh17細胞の増殖又は活性化が検出された非ヒト動物の小腸内試料から細菌を単離する工程とを、含む方法。
A method of screening for bacteria that induce the proliferation or activation of Th1 cells and / or Th17 cells in the small intestine.
(1) The process of ingesting the test substance into non-human germ-free animals,
(2) A step of detecting the number or activity of Th1 cells and / or Th17 cells in the small intestine of the non-human animal.
(3) A method comprising a step of isolating bacteria from a sample in the small intestine of a non-human animal in which proliferation or activation of Th1 cells and / or Th17 cells was detected in step (2).
 さらにまた、本発明は、以下に示す第4のスクリーニング方法をも提供する。 Furthermore, the present invention also provides a fourth screening method shown below.
 小腸内でTh1細胞及び/又はTh17細胞の増殖又は活性化を誘導する細菌をスクリーニングする方法であって、
(1)Th1細胞及び/又はTh17細胞の増殖又は活性化を誘導する小腸内細菌に由来する生理活性物質を、非ヒト無菌動物に摂取させる工程と、
(2)該非ヒト動物の小腸内におけるTh1細胞及び/又はTh17細胞の数又は活性を検出する工程と、
(3)工程(2)にてTh1細胞及び/又はTh17細胞の増殖又は活性化が検出された場合に、前記生理活性物質を、小腸内でTh1細胞及び/又はTh17細胞の増殖又は活性化を誘導する生理活性物質であると判定する工程とを、含む方法。
A method of screening for bacteria that induce the proliferation or activation of Th1 cells and / or Th17 cells in the small intestine.
(1) A step of ingesting a physiologically active substance derived from a small intestinal bacterium that induces proliferation or activation of Th1 cells and / or Th17 cells into a non-human germ-free animal.
(2) A step of detecting the number or activity of Th1 cells and / or Th17 cells in the small intestine of the non-human animal.
(3) When proliferation or activation of Th1 cells and / or Th17 cells is detected in step (2), the physiologically active substance is used to proliferate or activate Th1 cells and / or Th17 cells in the small intestine. A method comprising a step of determining that the substance is a physiologically active substance to be induced.
 なお、第3及び第4のスクリーニング方法における、「被験物質」、「非ヒト無菌動物」、「摂取」、「ヘルパーT細胞の増殖(数)又は活性の検出」及び「細菌の単離」については、上述の第1及び第2のスクリーニング方法と同様である。また、「生理活性物質」については、後述のとおりである。 Regarding "test substance", "non-human germ-free animal", "ingestion", "detection of helper T cell proliferation (number) or activity" and "isolation of bacteria" in the third and fourth screening methods. Is the same as the first and second screening methods described above. Further, the "physiologically active substance" is as described later.
 <本発明の検査方法>
 後述の実施例に示すとおり、本発明の小腸内細菌は、クローン病等のTh1細胞等に起因する疾患の病因に関連するものである。よって、本発明は、
 Th1細胞及び/又はTh17細胞に起因する疾患を評価する方法であって、
(1)被検者の小腸粘膜における、配列番号:1に記載のDNA配列に対して95%以上の同一性を有するポリヌクレオチドを有する細菌及び/又は配列番号:2に記載のDNA配列に対して95%以上の同一性を有するポリヌクレオチドを有する細菌を定量する工程、
(2)工程(1)で定量して得られた値を、前記疾患に罹患していないヒトの小腸粘膜において前記細菌を定量して得られる対応値と比較する工程、及び
(3)工程(2)における比較の結果、被検者の小腸粘膜中における前記定量値が、前記対応値よりも高い場合に、前記被検者は前記疾患に罹患していると判定する工程を含む方法。
<Inspection method of the present invention>
As shown in Examples below, the small intestinal bacterium of the present invention is associated with the etiology of diseases caused by Th1 cells and the like, such as Crohn's disease. Therefore, the present invention
A method for assessing diseases caused by Th1 cells and / or Th17 cells.
(1) For bacteria having a polynucleotide having 95% or more identity with respect to the DNA sequence set forth in SEQ ID NO: 1 and / or the DNA sequence set forth in SEQ ID NO: 2 in the small intestinal mucosa of the subject. Quantifying bacteria with polynucleotides having 95% or more identity,
(2) A step of comparing the value obtained by quantifying in step (1) with a corresponding value obtained by quantifying the bacteria in the mucosa of the small intestine of a human who does not suffer from the disease, and step (3) (3). A method including a step of determining that the subject is suffering from the disease when the quantitative value in the small intestinal mucosa of the subject is higher than the corresponding value as a result of the comparison in 2).
 本発明にかかる「Th1細胞及び/又はTh17細胞に起因する疾患」としては、Th1細胞及び/又はTh17細胞の増殖又は活性化によって誘発された疾患を意味し、炎症性腸疾患(クローン病(CD)、潰瘍性大腸炎、炎症性腸疾患といった慢性炎症性腸疾患等)、1型糖尿病、関節リウマチ、実験的免疫性脳炎(EAE)、多発性硬化症、全身性エリテマトーデス等の自己免疫疾患、慢性炎症性疾患が挙げられるが、中でも、クローン病は本発明の好適な対象となる。また、クローン病は、その病変がある部位によって、小腸型、小腸大腸型、大腸型に分類されるが、本発明においては、小腸型クローン病が、より好適な対象となり得る。 The "disease caused by Th1 cells and / or Th17 cells" according to the present invention means a disease induced by proliferation or activation of Th1 cells and / or Th17 cells, and is an inflammatory bowel disease (Clone's disease (CD). ), Chronic inflammatory bowel diseases such as ulcerative bowel disease and inflammatory bowel disease), type 1 diabetes, rheumatoid arthritis, experimental immune encephalitis (EAE), multiple sclerosis, autoimmune diseases such as systemic erythematosus, Chronic inflammatory diseases can be mentioned, and among them, Crohn's disease is a suitable subject of the present invention. In addition, Crohn's disease is classified into small intestine type, small intestine large intestine type, and large intestine type according to the site where the lesion is present, but in the present invention, small intestine type Crohn's disease can be a more suitable target.
 本発明の評価方法における「被検者」としては、Th1細胞及び/又はTh17細胞に起因する疾患の罹患又は再発が疑われるヒトが挙げられる。 Examples of the "subject" in the evaluation method of the present invention include humans suspected of having or recurring a disease caused by Th1 cells and / or Th17 cells.
 工程(1)における、本発明の小腸内細菌の定量については、上記「スクリーニング方法」の工程(2)同様にして行なうことができる。なお、小腸粘膜からの当該細菌の採取は、例えば、後述の実施例に示すとおり、ダブルバルーン内視鏡検査(DBE)システムを用いて行なうことができる。 The quantification of small intestinal bacteria of the present invention in step (1) can be carried out in the same manner as in step (2) of the above-mentioned "screening method". The bacteria can be collected from the mucosa of the small intestine using, for example, a double-balloon endoscopy (DBE) system as shown in Examples described later.
 また、工程(1)において定量の対象となる細菌は、配列番号:1に記載のDNA配列に対して95%以上の同一性を有するポリヌクレオチドを有する細菌及び/又は配列番号:2に記載のDNA配列に対して95%以上の同一性を有するポリヌクレオチドを有する細菌であるが、上記本発明の9種の小腸内細菌のうちの少なくとも1の細菌であってもよい。かかる場合、9種の小腸内細菌のうち、2種、3種、4種、5種、6種、7種、8種又は9種の細菌を定量対象とすることができるが、Th1細胞及び/又はTh17細胞に起因する疾患(特に、クローン病)の罹患等をより精度高く判定することができるという観点から、9種全てを定量対象とすることが好ましい。 In addition, the bacterium to be quantified in the step (1) is a bacterium having a polynucleotide having 95% or more identity with respect to the DNA sequence set forth in SEQ ID NO: 1 and / or the bacterium described in SEQ ID NO: 2. Although it is a bacterium having a polynucleotide having 95% or more identity with respect to the DNA sequence, it may be at least one of the nine small intestinal bacteria of the present invention. In such a case, out of 9 kinds of small intestinal bacteria, 2 kinds, 3 kinds, 4 kinds, 5 kinds, 6 kinds, 7 kinds, 8 kinds or 9 kinds of bacteria can be quantified, but Th1 cells and Th1 cells and / Or From the viewpoint that the morbidity of diseases caused by Th17 cells (particularly, Crohn's disease) can be determined with higher accuracy, it is preferable to quantify all 9 species.
 工程(2)において比較対象となる「Th1細胞及び/又はTh17細胞の増殖又は活性化によって誘発された疾患を罹患していないヒト」は、当該疾患を罹患していない限り、特に制限はないが、年齢、性別、国籍及び病歴のうちの少なくとも1の特徴が被検者と一致していることが好ましい。 The “human who does not suffer from a disease induced by proliferation or activation of Th1 cells and / or Th17 cells” to be compared in step (2) is not particularly limited as long as it does not suffer from the disease. , Age, gender, nationality and medical history are preferably consistent with the subject.
 また、前記疾患に罹患していないヒトの小腸粘膜において前記細菌を定量して得られる対応値との「比較」は、上記「スクリーニング方法」の工程(3)と同様にして、当業者であれば上記統計学的解析方法に基づき適宜行うことができる。例えば、工程(1)において得られた定量値が対応値より高く、その差が統計的に有意と認められること(例えば、P<0.05)が挙げられる。また、例えば、前記定量値が対応値の2倍以上(好ましくは、5倍以上、10倍以上)であることも挙げられる。 Further, "comparison" with the corresponding value obtained by quantifying the bacterium in the small intestinal mucosa of a human not suffering from the disease can be performed by a person skilled in the art in the same manner as in step (3) of the above-mentioned "screening method". For example, it can be appropriately performed based on the above statistical analysis method. For example, the quantitative value obtained in the step (1) is higher than the corresponding value, and the difference is considered to be statistically significant (for example, P <0.05). Further, for example, the quantitative value may be twice or more (preferably 5 times or more and 10 times or more) the corresponding value.
 工程(3)における「判定」には、前記疾患の発症の有無のみならず、その発症のリスクも含まれる。すなわち、被検者の小腸粘膜中における前記定量値が、前記対応値よりも高い場合に、前記被検者は前記疾患を発症している、またはその発症のリスクが高いと判定される。 The "determination" in step (3) includes not only the presence or absence of the onset of the disease but also the risk of its onset. That is, when the quantitative value in the small intestinal mucosa of the subject is higher than the corresponding value, it is determined that the subject has developed the disease or is at high risk of developing the disease.
 また、前記判定は、通常、医師(医師の指示を受けた者も含む)によって行われるが、上述の定量値等に関するデータは、医師による治療のタイミング等の判断も含めた診断に役立つものである。よって、本発明の方法は、医師による診断のために上述の定量値に関するデータを収集する方法、当該データを医師に提示する方法、前記定量値と対応値とを比較し分析する方法、患者の臨床症状及び/又は他の検査結果を考慮した上で医師による診断を補助するための方法とも表現し得る。 In addition, the determination is usually made by a doctor (including a person who has been instructed by a doctor), but the data on the above-mentioned quantitative values and the like are useful for diagnosis including the judgment of the timing of treatment by the doctor. be. Therefore, the method of the present invention includes a method of collecting data on the above-mentioned quantitative values for diagnosis by a doctor, a method of presenting the data to a doctor, a method of comparing and analyzing the quantitative values and corresponding values, and a method of comparing and analyzing the corresponding values of a patient. It can also be described as a method for assisting a doctor's diagnosis in consideration of clinical symptoms and / or other test results.
 <本発明の検査用組成物>
 本発明は、上述の検査方法に用いられる組成物を提供する。より具体的には以下に示す態様の検査用組成物を提供する。
<Inspection composition of the present invention>
The present invention provides a composition used in the above-mentioned inspection method. More specifically, the test composition of the following aspects is provided.
 本発明の小腸内細菌を特異的に認識する抗体を含む、Th1細胞及び/又はTh17細胞に起因する疾患を検査するための組成物。 A composition for examining a disease caused by Th1 cells and / or Th17 cells, which comprises an antibody that specifically recognizes the small intestinal bacteria of the present invention.
 本発明の小腸内細菌に特異的なヌクレオチド配列を検出するためのポリヌクレオチドを含む、Th1細胞及び/又はTh17細胞に起因する疾患を検査するための組成物。 A composition for examining a disease caused by Th1 cells and / or Th17 cells, which comprises a polynucleotide for detecting a nucleotide sequence specific to the small intestinal bacterium of the present invention.
 本発明において、「本発明の小腸内細菌を特異的に認識する抗体」は、該細菌を特異的に認識し得る限り、ポリクローナル抗体であっても、モノクローナル抗体であってもよく、また抗体の機能的断片(例えば、Fab、Fab’、F(ab’)2、可変領域断片(Fv)、ジスルフィド結合Fv、一本鎖Fv(scFv)、sc(Fv)2、ダイアボディー、多特異性抗体、又はこれらの重合体)であってもよい。本発明の抗体は、ポリクローナル抗体であれば、抗原(本発明の小腸内細菌に由来するポリペプチド、ポリヌクレオチド、糖鎖、脂質等)で免疫動物を免疫し、その抗血清から、従来の手段(例えば、塩析、遠心分離、透析、カラムクロマトグラフィーなど)によって、精製して取得することができる。また、モノクローナル抗体は、ハイブリドーマ法や組換えDNA法によって作製することができる。 In the present invention, the "antibody that specifically recognizes the small intestinal bacterium of the present invention" may be a polyclonal antibody, a monoclonal antibody, or an antibody as long as the bacterium can be specifically recognized. Functional fragments (eg, Fab, Fab', F (ab') 2, variable region fragment (Fv), disulfide-bound Fv, single-stranded Fv (scFv), sc (Fv) 2, diabodies, multispecific antibodies , Or a polymer of these). If the antibody of the present invention is a polyclonal antibody, an immune animal is immunized with an antigen (polypeptide, polynucleotide, sugar chain, lipid, etc. derived from the small intestinal bacterium of the present invention), and the antiserum thereof is used as a conventional means. It can be purified and obtained by (for example, salting out, centrifugation, dialysis, column chromatography, etc.). In addition, the monoclonal antibody can be produced by a hybridoma method or a recombinant DNA method.
 また、本発明の検査に用いる抗体としては、標識物質を結合させた抗体を使用することができる。当該標識物質を検出することにより、本発明の小腸内細菌又は該細菌に由来する物質に結合した抗体量を直接測定することが可能である。標識物質としては、抗体に結合することができ、化学的又は光学的方法に検出できるものであれば特に制限されることはなく、例えば、蛍光色素(GFP等)、酵素(HRP等)、放射性物質が挙げられる。 Further, as the antibody used for the test of the present invention, an antibody to which a labeling substance is bound can be used. By detecting the labeling substance, it is possible to directly measure the amount of antibody bound to the small intestinal bacterium of the present invention or a substance derived from the bacterium. The labeling substance is not particularly limited as long as it can bind to an antibody and can be detected by a chemical or optical method. For example, a fluorescent dye (GFP or the like), an enzyme (HRP or the like), or a radioactive substance is used. Substances are mentioned.
 本発明の検査用組成物には、抗体成分の他、組成物として許容される他の成分を含むことができる。このような他の成分としては、例えば、担体、賦形剤、崩壊剤、緩衝剤、乳化剤、懸濁剤、安定剤、保存剤、防腐剤、生理食塩、標識物質、二次抗体が挙げられる。また、上記検査用組成物の他、標識物質の検出に必要な基質、陽性対照や陰性対照、あるいはサンプルの希釈や洗浄に用いる緩衝液、サンプルと本発明の抗体との反応に用いるチューブ又はプレート等を組み合わせることができ、Th1細胞等に起因する疾患の検査用キットとすることもできる。また、標識されていない抗体を抗体標品とした場合には、当該抗体に結合する物質(例えば、二次抗体、プロテインG、プロテインA等)を標識化したものを組み合わせることができる。また、かかるTh1細胞等に起因する疾患の検査用キットには、当該キットの使用説明書を含めることができる。 The test composition of the present invention may contain other components permitted as a composition in addition to the antibody component. Such other components include, for example, carriers, excipients, disintegrants, buffers, emulsifiers, suspensions, stabilizers, preservatives, preservatives, physiological salts, labeling substances, secondary antibodies. .. In addition to the above test composition, a substrate necessary for detecting a labeling substance, a positive control or a negative control, a buffer solution used for diluting or washing the sample, and a tube or plate used for the reaction between the sample and the antibody of the present invention. Etc. can be combined, and a kit for testing a disease caused by Th1 cells or the like can be obtained. When an unlabeled antibody is used as an antibody preparation, a labeled substance (for example, secondary antibody, protein G, protein A, etc.) that binds to the antibody can be combined. In addition, the kit for testing for diseases caused by Th1 cells and the like can include an instruction manual for the kit.
 さらに、本発明の検査用組成物には、本発明の抗体を検出するための装置を組み合わせることもできる。かかる装置としては、例えば、フローサイトメトリー装置、マイクロプレートリーダーが挙げられる。 Further, the test composition of the present invention can be combined with an apparatus for detecting the antibody of the present invention. Examples of such a device include a flow cytometry device and a microplate reader.
 本発明において、「本発明の小腸内細菌に特異的なヌクレオチド配列を検出するためのポリヌクレオチド」としては、該細菌に特異的な配列を検出する限り、特に制限はなく、例えば、少なくとも15ヌクレオチドの鎖長を有する、下記(a)~(b)に記載のいずれかであるポリヌクレオチドが、挙げられる。
(a)前記特異的なヌクレオチド配列を挟み込むように設計された一対のプライマーであるポリヌクレオチド
(b)前記特異的なヌクレオチド配列を含むヌクレオチド配列にハイブリダイズするプライマー又はプローブであるポリヌクレオチド。
In the present invention, the "polynucleotide for detecting the nucleotide sequence specific to the bacterium of the present invention" is not particularly limited as long as the sequence specific to the bacterium is detected, and for example, at least 15 nucleotides. Examples of the polynucleotide having the chain length of any of the following (a) to (b).
(A) A polynucleotide that is a pair of primers designed to sandwich the specific nucleotide sequence (b) A polynucleotide that is a primer or probe that hybridizes to a nucleotide sequence containing the specific nucleotide sequence.
 本発明のポリヌクレオチドは、本発明の小腸内細菌のヌクレオチド配列に相補的な塩基配列を有する。ここで「相補的」とは、ハイブリダイズする限り、完全に相補的でなくともよい。これらポリヌクレオチドは、前記ヌクレオチド配列に対して、通常、80%以上、好ましくは90%以上、より好ましくは95%以上(例えば、96%以上、97%以上、98%以上、99%以上)、特に好ましくは100%の相同性を有する。 The polynucleotide of the present invention has a base sequence complementary to the nucleotide sequence of the small intestinal bacterium of the present invention. Here, "complementary" does not have to be completely complementary as long as it hybridizes. These polynucleotides are usually 80% or more, preferably 90% or more, more preferably 95% or more (for example, 96% or more, 97% or more, 98% or more, 99% or more) with respect to the nucleotide sequence. Particularly preferably, it has 100% homology.
 本発明のポリヌクレオチドにおける「鎖長」として、プライマーとして用いる場合に、通常15~100ヌクレオチドであり、好ましくは17~30ヌクレオチドであり、より好ましくは20~25ヌクレオチドである。また、プローブとして用いる場合には、通常15~1000ヌクレオチドであり、好ましくは20~100ヌクレオチドである。 When used as a primer, the "chain length" of the polynucleotide of the present invention is usually 15 to 100 nucleotides, preferably 17 to 30 nucleotides, and more preferably 20 to 25 nucleotides. When used as a probe, it is usually 15 to 1000 nucleotides, preferably 20 to 100 nucleotides.
 本発明のポリヌクレオチドは、DNAであってもRNAであってもよく、またその一部又は全部において、LNA(登録商標、架橋化核酸)、ENA(登録商標、2’-O,4’-C-Ethylene-bridged nucleic acids)、GNA(グリセロール核酸)、TNA(トレオ―ス核酸)、PNA(ペプチド核酸)等の人工核酸によって、ヌクレオチドが置換されているものであってもよい。 The polynucleotide of the present invention may be DNA or RNA, and in part or all thereof, LNA (registered trademark, crosslinked nucleic acid), ENA (registered trademark, 2'-O, 4'- Nucleotides may be substituted with artificial nucleic acids such as C-Ethylene-bridged nucleic acids), GNA (glycerol nucleic acids), TNAs (treose nucleic acids), and PNAs (peptide nucleic acids).
 なお、本発明のポリヌクレオチドは、市販のヌクレオチド自動合成機等を用いて化学的に合成することができる。また、本発明の検査に用いるポリヌクレオチドとしては、標識物質を結合させたポリヌクレオチドを使用することができる。標識物質としては、ポリヌクレオチドに結合することができ、化学的又は光学的方法に検出できるものであれば特に制限されることはなく、例えば、蛍光色素(DEAC、FITC、R6G、TexRed、Cy5等)、蛍光色素以外にDAB等の色素(chromogen)、酵素、放射性物質が挙げられる。 The polynucleotide of the present invention can be chemically synthesized using a commercially available nucleotide automatic synthesizer or the like. Further, as the polynucleotide used for the test of the present invention, a polynucleotide to which a labeling substance is bound can be used. The labeling substance is not particularly limited as long as it can bind to a polynucleotide and can be detected by a chemical or optical method. For example, a fluorescent dye (DEAC, FITC, R6G, TexRed, Cy5, etc.) ), Dyes such as DAB (chromogen), enzymes, radioactive substances in addition to fluorescent dyes.
 本発明の検査用組成物には、前述のポリヌクレオチドの他、薬理学上許容される他の成分を含むことができる。このような他の成分としては、例えば、緩衝剤、乳化剤、懸濁剤、安定剤、防腐剤、生理食塩等が挙げられる。 The test composition of the present invention may contain other pharmacologically acceptable components in addition to the above-mentioned polynucleotide. Examples of such other components include buffers, emulsifiers, suspending agents, stabilizers, preservatives, physiological salts and the like.
 また、上記検査用組成物の他、ポリヌクレオチドに付加した標識物質の検出に必要な基質、陽性対照や陰性対照、サンプルの希釈や洗浄に用いる緩衝液等の標品を組み合わせ、サンプルと本発明のポリヌクレオチドとの反応に用いるチューブ又はプレート等を組み合わせることができ、Th1細胞に起因する疾患の検査用キットとすることもできる。さらに、かかるTh1細胞に起因する疾患の検査用キットには、当該キットの使用説明書を含めることができる。 Further, in addition to the above test composition, a sample and the present invention are combined by combining a substrate necessary for detecting a labeling substance added to a polynucleotide, a positive control or a negative control, and a buffer solution used for diluting or washing the sample. A tube or plate used for the reaction with the polynucleotide of No. 1 can be combined, and a kit for testing a disease caused by Th1 cells can also be used. In addition, the kit for testing for diseases caused by such Th1 cells can include instructions for use of the kit.
 また、本発明の検査用組成物には、本発明の小腸内細菌に特異的なヌクレオチド配列を検出するための装置を組み合わせることもできる。かかる装置としては、例えば、サーマルサイクラー、シーケンサー、マイクロアレイが挙げられる。 Further, the test composition of the present invention can be combined with an apparatus for detecting a nucleotide sequence specific to the small intestinal bacterium of the present invention. Examples of such a device include a thermal cycler, a sequencer, and a microarray.
 <本発明のTh1細胞等を誘導するための組成物>
 後述の実施例に示すとおり、本発明の小腸内細菌を定着させることによって、Th1細胞及び/又はTh17細胞の増殖又は活性化を誘導することが可能となる。
<Composition for Inducing Th1 Cells, etc. of the Present Invention>
As shown in Examples described later, colonization of the small intestinal bacterium of the present invention makes it possible to induce proliferation or activation of Th1 cells and / or Th17 cells.
 したがって、本発明は、本発明の小腸内細菌又は該細菌に由来する生理活性物質を有効成分として含む、Th1細胞及び/又はTh17細胞の増殖若しくは活性化を誘導するための組成物、又はその方法を提供することができる。 Therefore, the present invention is a composition for inducing proliferation or activation of Th1 cells and / or Th17 cells, which comprises the small intestinal bacterium of the present invention or a physiologically active substance derived from the bacterium as an active ingredient, or a method thereof. Can be provided.
 また、クローン病等は、免疫作用の亢進に基づく疾患(自己免疫疾患)であることから、その病因となり得る本発明の小腸内細菌は免疫賦活作用を有するものである。さらにまた、Th1細胞は、IFN-γを産生するCD4陽性ヘルパーT細胞であり、結核菌やリステリア菌等の病原体感染防御に重要な役割を担っているとともに、がん化した細胞の監視、排除においても重要な働きをしている。近年では、BacteroidesやBifidobacterium等の腸管内における細菌の存在とかかる細菌によるTh1細胞の誘導とが、がんに対する免疫療法のひとつである免疫チェックポイント阻害剤(抗CTLA-4抗体や抗PD-L1抗体等)の抗腫瘍効果に影響を与えていることも明らかになっている。したがって、本発明は、免疫を賦活する組成物又はその方法を提供することができる。なお、本発明において賦活又は抑制される「免疫」には、粘膜免疫(腸管免疫等)のみならず、全身免疫も含まれる。また、細胞性免疫のみならず、液性免疫も含まれる。 Further, since Crohn's disease and the like are diseases based on enhanced immune action (autoimmune diseases), the small intestinal bacterium of the present invention, which can be a pathogen of the disease, has an immunostimulatory action. Furthermore, Th1 cells are CD4-positive helper T cells that produce IFN-γ, play an important role in defense against pathogen infections such as Mycobacterium tuberculosis and Listeria monocytogenes, and monitor and eliminate cancerous cells. It also plays an important role in. In recent years, the presence of bacteria in the intestinal tract such as Bacteroides and Bifidobacterium and the induction of Th1 cells by such bacteria have been one of the immunotherapies for cancer, such as immune checkpoint inhibitors (anti-CTLA-4 antibody and anti-PD-L1). It has also been clarified that it affects the antitumor effect of (antibodies, etc.). Therefore, the present invention can provide a composition for activating immunity or a method thereof. The "immunity" activated or suppressed in the present invention includes not only mucosal immunity (intestinal immunity, etc.) but also systemic immunity. Moreover, not only cell-mediated immunity but also humoral immunity is included.
 本発明の組成物に有効成分として含まれる「本発明の小腸内細菌」については、上記<本発明の小腸内細菌>に記載のとおりであるが、生菌であってもよく、死菌体であってもよい。「死菌体」としては、例えば、加熱処理体、焼成処理体、蒸煮処理体、放射線(γ線等)照射処理体、酵素処理体、薬物(抗生物質等)処理体、化学物質(ホルマリン等)処理体、超音波処理体が挙げられる。また、本発明の小腸内細菌の形態は特に限定されず、液体又は固体の何れでもよいが、当該形態は乾燥形態(例えば、粉体状、菌末状等)が、製造時や保管時の取扱いが容易である観点から好適である。乾燥物は、菌体を溶媒(水等)に分散させた懸濁液を乾燥させることによって調製することができる。乾燥方法は、特に制限はなく、例えば、噴霧乾燥(スプレードライ)法、凍結乾燥法が挙げられる。滅菌方法は、特に制限はなく、レトルト殺菌法、UHT殺菌法、空気殺菌法、加圧殺菌法、高圧蒸気滅菌法、乾熱滅菌法、流通蒸気消毒法、電磁波殺菌法、電子線滅菌法、高周波滅菌法、放射線滅菌法、紫外線殺菌法、酸化エチレンガス滅菌法、過酸化水素プラズマ滅菌法、化学的殺菌法(アルコール殺菌法、ホルマリン固定法、電解水処理法)等が挙げられる。また、場合によっては、加熱等による殺菌処理の前後、あるいは、乾燥処理の前後に、界面活性剤処理、磨砕・粉砕処理、酵素処理、分画処理等を行うこともでき、これらの処理により得られるものも、本発明の組成物に含有し得る。 The "small intestinal bacterium of the present invention" contained as an active ingredient in the composition of the present invention is as described in the above <small intestinal bacterium of the present invention>, but it may be a live bacterium or a dead bacterium. It may be. Examples of the "killed cell" include a heat-treated body, a fire-treated body, a steamed body, a radiation (γ-ray, etc.) irradiation-treated body, an enzyme-treated body, a drug (antibiotic, etc.) treated body, and a chemical substance (formaline, etc.). ) A processed body and an ultrasonic processed body can be mentioned. The form of the small intestinal bacterium of the present invention is not particularly limited and may be either liquid or solid, but the form is in a dry form (for example, powder form, bacterial powder form, etc.) during production or storage. It is suitable from the viewpoint of easy handling. The dried product can be prepared by drying a suspension in which the cells are dispersed in a solvent (water or the like). The drying method is not particularly limited, and examples thereof include a spray drying method and a freeze drying method. The sterilization method is not particularly limited, and the retort sterilization method, UHT sterilization method, air sterilization method, pressure sterilization method, high pressure steam sterilization method, dry heat sterilization method, distribution steam sterilization method, electromagnetic wave sterilization method, electron beam sterilization method, High-frequency sterilization method, radiation sterilization method, ultraviolet sterilization method, ethylene oxide gas sterilization method, hydrogen peroxide plasma sterilization method, chemical sterilization method (alcohol sterilization method, formalin fixation method, electrolyzed water treatment method) and the like can be mentioned. In some cases, before and after the sterilization treatment by heating or the like, or before and after the drying treatment, surfactant treatment, grinding / crushing treatment, enzyme treatment, fractionation treatment, etc. can be performed. The obtained product can also be contained in the composition of the present invention.
 また、含まれる有効成分の用途に応じ、改変(遺伝子改変等)が施されている細菌であってもよい。かかる改変としては特に制限はなく、本発明の組成物が後述のワクチンアジュバントである場合には、Th1細胞等の増殖又は活性化を誘導する作用が増強されるような改変が挙げられる。 Further, the bacterium may be modified (gene modification, etc.) according to the use of the contained active ingredient. Such modifications are not particularly limited, and when the composition of the present invention is a vaccine adjuvant described later, modifications such that the action of inducing proliferation or activation of Th1 cells or the like is enhanced can be mentioned.
 また、本発明の組成物が含有する有効成分は、本発明の小腸内細菌の培養物であってもよい。培養物は、当該細菌を含むもの(増殖した当該細菌を含む培養液、固形培地等)であってもよく、当該培養物の形態は、特に限定されず、液体又は固体の何れでもよいが、当該形態は乾燥形態(例えば培養乾燥物等)が、製造時や保管時の取扱いが容易な観点から、好適である。なお、かかる培養乾燥物等も、当業者であれば、上述の乾燥方法を用い、適宜調製し得る。 Further, the active ingredient contained in the composition of the present invention may be a culture of small intestinal bacteria of the present invention. The culture may be one containing the bacterium (culture solution containing the grown bacterium, solid medium, etc.), and the form of the culture is not particularly limited and may be either liquid or solid. A dry form (for example, a dried culture product) is suitable for this form from the viewpoint of easy handling during production and storage. Those skilled in the art can appropriately prepare such a dried culture product or the like by using the above-mentioned drying method.
 また、本発明の組成物は、本発明の小腸内細菌を1株含んでいればよいが、複数種の株を含むものであってもよい。または組成物を複合して用いることができ、結果として併用して摂取又は吸収される場合(併用組成物の場合)、該複数の菌株は2種以上の組成物の中に存在することもできる(例えば、それぞれ別々の組成物中に存在することもできる)。 Further, the composition of the present invention may contain one strain of the small intestinal bacterium of the present invention, but may contain a plurality of types of strains. Alternatively, the compositions can be used in combination, and as a result, when ingested or absorbed in combination (in the case of a combination composition), the plurality of strains can be present in two or more kinds of compositions. (For example, they can be in separate compositions).
 また、その「生理活性物質」としては、特に制限はされないが、前記細菌に含まれる物質、前記細菌の分泌産物、前記細菌による代謝産物を含む意味であり、より具体的には、前記細菌又はその培養上清のポリペプチド画分、ポリヌクレオチド画分、糖鎖画分、脂質画分、低分子代謝産物画分が挙げられる。なお、このような生理活性物質は、本発明の小腸内細菌、その培養上清、該細菌が定着した非ヒト動物の小腸内サンプル、ヒトの小腸内サンプル等から、例えば、上述のスクリーニング方法のように、Th1細胞等の増殖又は活性化を指標として、活性成分を精製することにより同定することが可能である。 The "physiologically active substance" is not particularly limited, but includes a substance contained in the bacterium, a secreted product of the bacterium, and a metabolite of the bacterium, and more specifically, the bacterium or the metabolite. Examples thereof include a polypeptide fraction, a polynucleotide fraction, a sugar chain fraction, a lipid fraction, and a low molecular weight metabolite fraction of the culture supernatant. Such a physiologically active substance can be obtained from, for example, the above-mentioned screening method from the small intestinal bacterium of the present invention, the culture supernatant thereof, the small intestinal sample of a non-human animal in which the bacterium has settled, the human small intestine sample, and the like. As described above, it is possible to identify by purifying the active ingredient using the proliferation or activation of Th1 cells or the like as an index.
 本発明の組成物は、医薬組成物、飲食品(動物用飼料を含む)、あるいは研究目的(例えば、インビトロやインビボの実験)に用いられる試薬の形態であり得る。 The composition of the present invention can be in the form of a pharmaceutical composition, food or drink (including animal feed), or a reagent used for research purposes (eg, in vitro or in vivo experiments).
 上述の通り、本発明の組成物の有効成分である、本発明の小腸内細菌又は該細菌に由来する生理活性物質は、Th1細胞等の増殖又は活性化を誘導し、また免疫を賦活するため、結核等の感染症の治療、予防又は改善のための医薬組成物、飲食品として、また抗がん剤又は免疫チェックポイント阻害剤との併用による抗がん作用を増強するための医薬組成物、飲食品として、さらに、ワクチンと併用することにより、その免疫応答作用を増強するための医薬組成物(ワクチンアジュバント)としても、好適に用いられる。なお、本発明において「治療」には、治療の補助も含まれる。 As described above, the active ingredient of the composition of the present invention, the small intestinal bacterium of the present invention or a physiologically active substance derived from the bacterium, induces the proliferation or activation of Th1 cells and the like, and activates immunity. , Pharmaceutical composition for treatment, prevention or improvement of infectious diseases such as tuberculosis, pharmaceutical composition for enhancing anticancer activity as food and drink, and in combination with anticancer agent or immune checkpoint inhibitor , As a food and drink, and also as a pharmaceutical composition (vaccine adjuvant) for enhancing its immune response action when used in combination with a vaccine. In addition, in the present invention, "treatment" also includes assistance for treatment.
 本発明の組成物は、公知の製剤学的方法により製剤化することができる。例えば、カプセル剤、錠剤、丸剤、液剤、散剤、顆粒剤、細粒剤、フィルムコーティング剤、ペレット剤、トローチ剤、舌下剤、咀嚼剤、バッカル剤、ペースト剤、シロップ剤、懸濁剤、エリキシル剤、乳剤、塗布剤、軟膏剤、硬膏剤、パップ剤、経皮吸収型製剤、ローション剤、吸引剤、エアゾール剤、注射剤、坐剤等として、経口的、非経口的(例えば、腸管内、筋肉内、静脈内、気管内、鼻内、経皮、皮内、皮下、眼内、膣、腹腔内、直腸若しくは吸入)、又はこれらの複数の組み合わせからなる経路による投与用に使用することができる。 The composition of the present invention can be formulated by a known pharmaceutical method. For example, capsules, tablets, pills, liquids, powders, granules, fine granules, film coatings, pellets, lozenges, sublinguals, chewing agents, buccal agents, pastes, syrups, suspensions, Oral and parenteral (eg, intestinal tract) as elixirs, emulsions, coatings, ointments, plasters, paps, transdermal formulations, lotions, inhalants, aerosols, injections, suppositories, etc. Intramuscular, intravenous, intravenous, intratracheal, intranasal, transdermal, intradermal, subcutaneous, intraocular, vaginal, intraperitoneal, rectal or inhalation), or for administration by a route consisting of multiple combinations thereof. be able to.
 これら製剤化においては、薬理学上若しくは飲食品として許容される担体、具体的には、滅菌水や生理食塩水、植物油、溶剤、基剤、乳化剤、懸濁剤、界面活性剤、安定剤、香味剤、芳香剤、賦形剤、ベヒクル、防腐剤、結合剤、希釈剤、等張化剤、無痛化剤、増量剤、崩壊剤、緩衝剤、コーティング剤、滑沢剤、着色剤、甘味剤、粘稠剤、矯味矯臭剤、溶解補助剤あるいはその他の添加剤等と適宜組み合わせることができる。 In these formulations, carriers that are pharmacologically or acceptable as foods and drinks, specifically, sterile water, physiological saline, vegetable oils, solvents, bases, emulsifiers, suspending agents, surfactants, stabilizers, etc. Flavors, fragrances, excipients, vehicles, preservatives, binders, diluents, isotonic agents, soothing agents, bulking agents, disintegrants, buffers, coating agents, lubricants, colorants, sweetness It can be appropriately combined with an agent, a thickener, a flavoring agent, a solubilizing agent, or other additives.
 また、これら製剤化においては、小腸内等におけるTh1細胞等の増殖又は活性化をより効率的に誘導する等の観点から、特に経口投与を目的とする製剤においては、本発明の組成物を小腸内等に効率良く送達することを可能にする組成物と組み合わせてもよい。このような小腸内等への送達を可能とする組成物については特に制限されることなく、公知の組成物を適宜採用することができ、例えば、pH感受性組成物、小腸等までの放出を抑制する組成物(セルロース系ポリマー、アクリル酸重合体及び共重合体、ビニル酸重合体及び共重合体等)、小腸粘膜特異的に接着する生体接着性組成物、プロテアーゼ阻害剤含有組成物、小腸内酵素によって特異的に分解される組成物が挙げられる。 Further, in these formulations, from the viewpoint of more efficiently inducing the proliferation or activation of Th1 cells or the like in the small intestine or the like, particularly in the formulation intended for oral administration, the composition of the present invention is used in the small intestine. It may be combined with a composition that enables efficient delivery to the inside and the like. The composition capable of such delivery into the small intestine or the like is not particularly limited, and a known composition can be appropriately adopted, for example, a pH-sensitive composition, suppression of release to the small intestine or the like. Compositions (cellulose-based polymers, acrylic acid polymers and copolymers, vinyl acid polymers and copolymers, etc.), bioadhesive compositions that specifically adhere to the small intestinal mucosa, protease inhibitor-containing compositions, in the small intestine Examples thereof include compositions that are specifically degraded by an enzyme.
 また、本発明のTh1細胞等の増殖又は活性化を誘導する組成物を医薬組成物として用いる場合には、感染症やがんの治療、予防又は改善に用いられる公知の物質(抗ウイルス剤、抗菌剤、抗がん剤、免疫チェックポイント阻害剤等)を更に含んでいてもよく、またかかる物質と併用してもよい。ワクチンアジュバントとして用いる場合には、ワクチンの有効成分である抗原(例えば、細菌又はウイルスに特異的な抗原、がん特異的な抗原)の他、他の公知のワクチンアジュバント、免疫増強剤を、本発明の組成物は含んでいてもよく、またそれら物質と併用してもよい。 Further, when the composition that induces proliferation or activation of Th1 cells or the like of the present invention is used as a pharmaceutical composition, a known substance (antiviral agent, antiviral agent, used for treatment, prevention or amelioration of infectious diseases and cancers, etc. Antibacterial agents, anticancer agents, immune checkpoint inhibitors, etc.) may be further contained, or may be used in combination with such substances. When used as a vaccine adjuvant, in addition to the antigen that is the active ingredient of the vaccine (for example, a bacterial or virus-specific antigen, a cancer-specific antigen), other known vaccine adjuvants and immunopotentiators should be used. The compositions of the invention may be included or may be used in combination with those substances.
 本発明の組成物を飲食品として用いる場合、当該飲食品は、例えば、健康食品、機能性食品、特定保健用食品、栄養機能食品、機能性表示食品、栄養補助食品、病者用食品、あるいは動物用飼料であり得る。なお、機能性食品は、通常、その作用メカニズムに基づき、プロバイオティクス、バイオジェニクス、プレバイオティクス、シンバイオティクスの4つに分類され、本発明においては、プロバイオティクス、バイオジェニクス、シンバイオティクス、プレバイオティクスの態様をとり得る。飲食品の具体例としては、発酵飲料、油分を含む製品、スープ類、乳飲料、清涼飲料水、茶飲料、アルコール飲料、ドリンク剤、ゼリー状飲料等の液状食品;炭水化物含有食品;畜産加工食品;水産加工食品;野菜加工食品;半固形状食品;発酵食品;菓子類;レトルト製品;電子レンジ対応食品等が挙げられる。さらには、粉末、穎粒、錠剤、カプセル剤、液状、ペースト状又はゼリー状に調製された健康飲食品も挙げられる。なお、本発明における飲食品の製造は、当該技術分野に公知の製造技術により実施することができる。当該飲食品においては、感染症やがんの改善又は予防に有効な成分(例えば、栄養素等)を添加してもよい。また、当該改善等以外の機能を発揮する他の成分あるいは他の機能性食品と組み合わせることによって、多機能性の飲食品としてもよい。 When the composition of the present invention is used as a food or drink, the food or drink may be, for example, a health food, a functional food, a food for specified health use, a food with a nutritional function, a food with a functional claim, a nutritional supplement, a food for the sick, or It can be an animal feed. Functional foods are usually classified into four categories, probiotics, biogenics, prebiotics, and symbiotics, based on their mechanism of action. In the present invention, probiotics, biogenics, and symbiotics are classified. It can take the form of tics or prebiotics. Specific examples of foods and drinks include fermented beverages, oil-containing products, soups, dairy beverages, soft drinks, tea beverages, alcoholic beverages, drinks, jelly-like beverages and other liquid foods; carbohydrate-containing foods; processed livestock foods. Processed marine foods; processed vegetable foods; semi-solid foods; fermented foods; confectionery; retort products; microwave-compatible foods and the like. Further, healthy foods and drinks prepared in the form of powder, granules, tablets, capsules, liquid, paste or jelly can also be mentioned. The food and drink according to the present invention can be produced by a production technique known in the art. In the food and drink, ingredients (for example, nutrients, etc.) effective for improving or preventing infectious diseases and cancer may be added. In addition, it may be a multifunctional food or drink by combining it with other ingredients or other functional foods that exhibit functions other than the improvement.
 本発明の組成物の製品(医薬品、飲食品、ワクチン、試薬)又はその説明書は、Th1細胞等の増殖若しくは活性化を誘導する、又は感染症、がん等の疾患を治療、改善若しくは予防するために用いられる旨の表示を付したものであり得る。 The products (pharmaceuticals, foods and drinks, vaccines, reagents) of the compositions of the present invention or their instructions induce the proliferation or activation of Th1 cells and the like, or treat, improve or prevent diseases such as infectious diseases and cancer. It may be labeled as being used for this purpose.
 また本発明で提供される飲食品組成物において、保健用途が表示された飲食品として提供・販売されることも可能である。「表示」行為には、需要者に対して前記用途を知らしめたる全ての行為が含まれ、前記用途を直接的に認識できる表現、あるいは前記用途を想起・類推させうるような表現を、組成物自体に付したものであってもよいし、組成物が含入する容器、包装材又は添付文書に付したものであってもよい。また「表示」は、本発明組成物の関連する情報として、チラシ、パンフレット、ポップ、カタログ、ポスター、書籍、DVD等の記憶媒体、電子掲示板やインターネット等の広告等で、本発明の組成物が効果的であることを表示・広告するものであってもよい。 It is also possible to provide and sell the food and drink composition provided in the present invention as a food and drink with a health use indication. The act of "displaying" includes all acts that inform the consumer of the use, and constitutes an expression that can directly recognize the use or an expression that can recall or infer the use. It may be attached to the product itself, or it may be attached to the container, packaging material or package insert containing the composition. Further, "display" refers to information related to the composition of the present invention, such as leaflets, pamphlets, pops, catalogs, posters, books, storage media such as DVDs, advertisements such as electronic bulletin boards and the Internet, and the like. It may be something that displays / advertises that it is effective.
 また、本発明の組成物は、キットの態様であってもよい。かかるキットとしては、例えば、本発明の小腸内細菌又は該細菌に由来する生理活性物質、抗ウイルス剤、抗菌剤、抗がん剤、免疫チェックポイント阻害剤、抗原、他のワクチンアジュバント等は、通常、2種以上の物質(組成物等)として存在しているが、対象に摂取させる前に、混合等して1の組成物に調製し得る態様が挙げられる。 Further, the composition of the present invention may be in the form of a kit. Such kits include, for example, the small intestinal bacterium of the present invention or a physiologically active substance derived from the bacterium, an antiviral agent, an antibacterial agent, an anticancer agent, an immune checkpoint inhibitor, an antigen, another vaccine adjuvant, and the like. Usually, it exists as two or more kinds of substances (compositions and the like), but there is an embodiment in which one composition can be prepared by mixing or the like before ingesting the subject.
 また、本発明は、Th1細胞等の増殖若しくは活性化を誘導するための組成物、又はその有効成分である本発明の小腸内細菌若しくは該細菌に由来する生理活性物質を対象に摂取させることを特徴とする、対象におけるTh1細胞又はTh17細胞の増殖若しくは活性化を誘導する方法、又は該対象における免疫を賦活する方法をも提供するものである。 In addition, the present invention requires that a composition for inducing proliferation or activation of Th1 cells or the like, or an active ingredient thereof, the small intestinal bacterium of the present invention or a physiologically active substance derived from the bacterium, be ingested. It also provides a method of inducing proliferation or activation of Th1 cells or Th17 cells in a subject, or a method of activating immunity in the subject.
 本発明の組成物又はその有効成分は、通常、ヒトを対象として使用することができるが、ヒト以外の動物であってもよく、種々の家畜、家禽、ペット、実験用動物等を対象とすることができる。 The composition of the present invention or an active ingredient thereof can be usually used for humans, but may be animals other than humans, and various livestock, poultry, pets, laboratory animals and the like are targeted. be able to.
 また、本発明のTh1細胞等の増殖若しくは活性化を誘導するための組成物、又はその有効成分の摂取対象としては、その発症の如何を問わず、ウイルス、細菌等に感染している動物が挙げられる。また予防の観点からは、ウイルス、細菌等に感染していない又はその感染の疑いのある動物に、本発明の組成物等を摂取させてもよい。さらに、再発予防の観点からは、その症状がでていないウイルス、細菌等を保因する動物にも、本発明の組成物等は好適に用いることができる。また同様に、がんを罹患している動物のみならず、がんの罹患の疑いのある動物、抗がん療法後の動物にも、本発明の組成物等は好適に用いることができる。 In addition, as an ingestion target of the composition for inducing the proliferation or activation of Th1 cells and the like of the present invention, or the active ingredient thereof, animals infected with viruses, bacteria, etc., regardless of their onset, are used. Can be mentioned. From the viewpoint of prevention, an animal that has not been infected with a virus, a bacterium, or the like or is suspected of being infected with the virus may be allowed to ingest the composition or the like of the present invention. Furthermore, from the viewpoint of preventing recurrence, the composition of the present invention can be suitably used for animals that carry viruses, bacteria, etc. that do not have the symptom. Similarly, the composition of the present invention can be suitably used not only for animals suffering from cancer, but also for animals suspected of suffering from cancer and animals after anticancer therapy.
 本発明の組成物等の摂取方法としては、特に制限はなく、経口投与であってもよく、また非経口投与(例えば、小腸内への投与)であってもよいが、経口投与である場合には、本発明の組成物等の効果をより向上させるという観点から、本発明の組成物等の摂取対象は、プロトンポンプ阻害剤(PPI)等の摂取により胃酸の産生を減少させておくこと、または抗生物質を摂取しておくことが好ましい。 The method of ingesting the composition or the like of the present invention is not particularly limited and may be oral administration or parenteral administration (for example, administration into the small intestine), but in the case of oral administration. In order to further improve the effect of the composition of the present invention, the subject of ingestion of the composition of the present invention should reduce the production of gastric acid by ingesting a proton pump inhibitor (PPI) or the like. , Or it is preferable to take antibiotics.
 また、本発明の組成物等を摂取させる場合、その摂取量は、対象の年齢、体重、疾患の症状、健康状態、組成物の種類(医薬品、飲食品等)、摂取方法等に応じて、当業者であれば適宜選択することができる。 In addition, when the composition or the like of the present invention is ingested, the ingestion amount depends on the age, weight, disease symptom, health condition, type of composition (pharmaceuticals, foods and drinks, etc.), intake method, etc. of the subject. Those skilled in the art can appropriately select it.
 <本発明のワクチン組成物等>
 本発明の小腸内細菌は、Th1細胞等の増殖又は活性化を誘導することによって、クローン病等のTh1細胞等に起因する疾患を誘発する。そのため、本発明の小腸内細菌を標的として免疫応答を誘導することによって、小腸内等から当該菌を除去すれば、前記疾患を治療、改善又は予防することができる。
<Vaccine composition of the present invention, etc.>
The small intestinal bacterium of the present invention induces a disease caused by Th1 cells or the like such as Crohn's disease by inducing proliferation or activation of Th1 cells or the like. Therefore, the disease can be treated, ameliorated or prevented by removing the bacteria from the small intestine or the like by inducing an immune response by targeting the bacteria in the small intestine of the present invention.
 したがって、本発明は、本発明の小腸内細菌若しくは該細菌に特異的な抗原を有効成分として含むワクチン組成物、又は前記細菌に対する免疫応答を誘導する方法を提供することができる。 Therefore, the present invention can provide a small intestinal bacterium of the present invention, a vaccine composition containing an antigen specific to the bacterium as an active ingredient, or a method for inducing an immune response against the bacterium.
 本発明の組成物に有効成分として含まれる「本発明の小腸内細菌」としては、上述の通りであり、生菌であってもよく、死菌体であってもよい(死菌体については上述のとおりである)。また、含まれる有効成分の用途に応じ、本発明の小腸内細菌は、改変(遺伝子改変等)が施されている細菌であってもよい。かかる改変としては特に制限はなく、本発明の組成物がワクチン組成物である場合には、小腸内等におけるTh1細胞等の増殖又は活性化を誘導する作用が抑制され、炎症等を誘発しないような改変が挙げられる。また、本発明の組成物が含有する有効成分は、本発明の小腸内細菌の培養物であってもよい(培養物については上述のとおりである)。 The "small intestinal bacterium of the present invention" contained as an active ingredient in the composition of the present invention is as described above, and may be a live bacterium or a dead bacterium (for the dead bacterium). As mentioned above). In addition, the small intestinal bacterium of the present invention may be a bacterium that has been modified (gene modification or the like) depending on the use of the active ingredient contained therein. The modification is not particularly limited, and when the composition of the present invention is a vaccine composition, the action of inducing the proliferation or activation of Th1 cells or the like in the small intestine or the like is suppressed so as not to induce inflammation or the like. Modifications can be mentioned. In addition, the active ingredient contained in the composition of the present invention may be a culture of the bacteria in the small intestine of the present invention (the culture is as described above).
 また、本発明の組成物は、本発明の小腸内細菌を少なくとも1種含んでいればよいが、複数種の株(例えば、2種、3種、4種、5種、6種、7種、8種又は9種)を含むものであってもよい。または組成物を複合して用いることができ、結果として併用して摂取または吸収される場合(併用組成物の場合)、該複数の菌株は2種以上の組成物の中に存在することもできる。 The composition of the present invention may contain at least one small intestinal bacterium of the present invention, but a plurality of strains (for example, 2 types, 3 types, 4 types, 5 types, 6 types, 7 types). , 8 types or 9 types). Alternatively, the compositions can be used in combination, and as a result, when ingested or absorbed in combination (in the case of a combination composition), the plurality of strains can be present in two or more kinds of compositions. ..
 本発明において、「本発明の小腸内細菌に特異的な抗原」とは、該細菌に含まれる物質(ポリペプチド、ポリヌクレオチド、糖鎖、脂質等)であり、かつ抗原性又は免疫原性を有する物質を意味する。ここで、「免疫原性」とは、一次免疫応答又は記憶免疫応答を活性化することができることを意味する。「免疫応答」には、CD4陽性(CD4+)Tリンパ球、CD8陽性(CD8+)Tリンパ球及びBリンパ球の応答が含まれる。Tリンパ球の場合、このような応答は、増殖型及び/又はサイトカイン(例えば、IL-2、IL-3、IL-4、IL-5、IL-6、IL-12、IL-13、IL-15、TNF-α、IFN-γ)産生型であってもよい。あるいはこれらの応答は、細胞傷害性Tリンパ球(CTL)の産生をもたらすものであってもよい。Bリンパ球反応は、反応するBリンパ球による抗体産生をもたらすものであってもよい。また、「抗原性」とは、抗体分子又は活性化されたエフェクターT細胞(例えば、サイトカイン産生T細胞、CTL等)上の抗原特異的T細胞受容体(TCR)により認識され得ることを意味する。このように、「本発明の小腸内細菌に特異的な抗原」は、該細菌等を特異的に認識する抗体によって認識され、これに結合することができる物質、又は適当な抗原提示細胞(APC)によるプロセッシング及び適当な主要組織適合遺伝子複合体(MHC)分子への結合後に、前記細菌等に反応して誘導されるエフェクターT細胞上のTCRによって認識されこれに結合することができる物質を含む意味である。 In the present invention, the "antigen specific to the small intestinal bacterium of the present invention" is a substance (polypeptide, polynucleotide, sugar chain, lipid, etc.) contained in the bacterium, and is antigenic or immunogenic. It means a substance to have. Here, "immunogenicity" means that a primary immune response or a memory immune response can be activated. The "immune response" includes responses of CD4 positive (CD4 +) T lymphocytes, CD8 positive (CD8 +) T lymphocytes and B lymphocytes. In the case of T lymphocytes, such responses are proliferative and / or cytokines (eg, IL-2, IL-3, IL-4, IL-5, IL-6, IL-12, IL-13, IL. -15, TNF-α, IFN-γ) production type may be used. Alternatively, these responses may result in the production of cytotoxic T lymphocytes (CTL). The B lymphocyte reaction may result in antibody production by the reacting B lymphocytes. Also, "antigenic" means that it can be recognized by an antibody molecule or an antigen-specific T cell receptor (TCR) on activated effector T cells (eg, cytokine-producing T cells, CTL, etc.). .. As described above, the "antigen specific to the small intestinal bacterium of the present invention" is a substance that can be recognized and bound to the antibody that specifically recognizes the bacterium or the like, or an appropriate antigen-presenting cell (APC). ), And after binding to a suitable major histocompatibility complex (MHC) molecule, contains a substance that can be recognized and bound to the TCR on the effector T cells induced in response to the bacteria or the like. Means.
 また、このような細菌特異的抗原は、当業者であれば、抗原特異的抗血清及び/又はTリンパ球との反応性を指標とした、公知のスクリーニング方法(例えば、Paul WE編集、Fundamental Immunology、1993年、第3版、243~247ページ、Harlow及びLane、Antibodies、A Laboratory Manual、Cold Spring Harbor Laboratory、1998年)によって同定することができる。また、細菌特異的抗原(ポリペプチド)のアミノ酸配列は、コンピュータープログラムを利用した分析(例えば、MHC-THREAD、EpiPredict、HLA-DR4 binding、ProPred、BIMAS、SVMHC、NetMHC、PREDICT、LpPep、SYFPEITHI、RankPep)によって推測することもできる。 In addition, such a bacterial-specific antigen can be obtained by a person skilled in the art using a known screening method (for example, Paul WE editing, Fundamental Immunology) using the reactivity with the antigen-specific antiserum and / or T lymphocyte as an index. , 1993, 3rd edition, pp. 243-247, Harlow and Lane, Antibodies, A Laboratory Manual, Cold Spring Harbor Laboratory, 1998). In addition, the amino acid sequence of the bacterial-specific antigen (polypeptide) is analyzed using a computer program (for example, MHC-THREAD, EpiPredict, HLA-DR4 binding, ProPred, BIMAS, SVMHC, NetMHC, PREDICT, LpPep, SYFPEITHI, RankPe. ) Can also be inferred.
 本発明のワクチン組成物は、公知の製剤学的方法により製剤化することができる。例えば、吸引剤、エアゾール剤、注射剤、散剤、顆粒剤、細粒剤、液剤、カプセル剤、錠剤、丸剤、フィルムコーティング剤、ペレット剤、トローチ剤、舌下剤、咀嚼剤、バッカル剤、ペースト剤、シロップ剤、懸濁剤、エリキシル剤、乳剤、塗布剤、軟膏剤、硬膏剤、パップ剤、経皮吸収型製剤、ローション剤、坐剤等として、経口的、非経口的(例えば、腸管内、筋肉内、静脈内、気管内、鼻内、経皮、皮内、皮下、眼内、膣、腹腔内、直腸若しくは吸入)、又はこれらの複数の組み合わせからなる経路による投与用に使用することができる。 The vaccine composition of the present invention can be formulated by a known pharmaceutical method. For example, inhalants, aerosols, injections, powders, granules, fine granules, liquids, capsules, tablets, pills, film coatings, pellets, lozenges, sublinguals, chewing agents, buccal agents, pastes. Oral and parenteral (eg, intestinal tract) as agents, syrups, suspensions, elixirs, emulsions, coatings, ointments, ointments, paps, transdermal formulations, lotions, suppositories, etc. Intramuscular, intravenous, intravenous, intratracheal, intranasal, transdermal, intradermal, subcutaneous, intraocular, vaginal, intraperitoneal, rectal or inhalation), or for administration by a route consisting of multiple combinations thereof. be able to.
 これら製剤化においては、薬理学上若しくは飲食品として許容される担体、具体的には、滅菌水や生理食塩水、植物油、溶剤、基剤、乳化剤、懸濁剤、界面活性剤、安定剤、香味剤、芳香剤、賦形剤、ベヒクル、防腐剤、結合剤、希釈剤、等張化剤、無痛化剤、増量剤、崩壊剤、緩衝剤、コーティング剤、滑沢剤、着色剤、甘味剤、粘稠剤、矯味矯臭剤、溶解補助剤あるいはその他の添加剤等と適宜組み合わせることができる。 In these formulations, carriers that are pharmacologically or acceptable as foods and drinks, specifically, sterile water, physiological saline, vegetable oils, solvents, bases, emulsifiers, suspending agents, surfactants, stabilizers, etc. Flavors, fragrances, excipients, vehicles, preservatives, binders, diluents, isotonic agents, soothing agents, bulking agents, disintegrants, buffers, coating agents, lubricants, colorants, sweetness It can be appropriately combined with an agent, a thickener, a flavoring agent, a solubilizing agent, or other additives.
 また、本発明のワクチン組成物は、公知のワクチンアジュバント、免疫増強剤を含むものであってもよい。ワクチンアジュバントとしては、水酸化アルミニウム、KLH、MPL、QS21、完全フロイントアジュバント、不完全フロイントアジュバント、リン酸アルミニウム、BCG、ミョウバン、CpG DNA等のTLRのアゴニスト等、又はこれらの組み合わせが挙げられる。さらに、必要に応じて、アルブミン、湿潤剤、乳化剤等の補助剤が添加されている態様であってもよい。また、免疫増強剤としては、各種サイトカイン(例えば、IL-12、IL-18、GM-CSF、IFN-γ、IFN-α、IFN-β、IFN-ω、Flt3リガンド)が挙げられる。 Further, the vaccine composition of the present invention may contain a known vaccine adjuvant and immunopotentiator. Examples of the vaccine adjuvant include aluminum hydroxide, KLH, MPL, QS21, complete Freund's adjuvant, incomplete Freund's adjuvant, aluminum phosphate, BCG, myoban, TLR agonist such as CpG DNA, or a combination thereof. Further, if necessary, an auxiliary agent such as albumin, a wetting agent, and an emulsifier may be added. In addition, examples of the immunopotentiator include various cytokines (for example, IL-12, IL-18, GM-CSF, IFN-γ, IFN-α, IFN-β, IFN-ω, Flt3 ligand).
 本発明の組成物の製品(医薬品、ワクチン)又はその説明書は、小腸内等でTh1細胞等の増殖又は活性化を誘導する細菌に対する免疫応答を誘導し、Th1細胞等に起因する疾患を治療、改善若しくは予防するために用いられる旨の表示を付したものであり得る。ここで「製品又は説明書に表示を付した」とは、製品の本体、容器、包装等に表示を付したこと、あるいは製品の情報を開示する説明書、添付文書、宣伝物、その他の印刷物等に表示を付したことを意味する。 The product (pharmaceutical product, vaccine) of the composition of the present invention or its description induces an immune response against a bacterium that induces proliferation or activation of Th1 cells or the like in the small intestine or the like, and treats a disease caused by Th1 cells or the like. , It may be labeled as being used for improvement or prevention. Here, "marked on a product or instruction manual" means that a label is attached to the main body, container, packaging, etc. of the product, or a manual, package insert, advertisement, or other printed matter that discloses product information. It means that the display is attached to.
 また、本発明の組成物は、キットの態様であってもよい。かかるキットとしては、例えば、本発明の小腸内細菌又は該細菌に特異的な抗原、ワクチンアジュバント、免疫増強剤等は、通常、2種以上の物質(組成物等)として存在しているが、対象に摂取させる前に、混合等して1の組成物に調製し得る態様が挙げられる。 Further, the composition of the present invention may be in the form of a kit. As such a kit, for example, the small intestinal bacterium of the present invention or an antigen, a vaccine adjuvant, an immunopotentiator or the like specific to the bacterium usually exists as two or more kinds of substances (compositions and the like). Examples thereof include an embodiment in which one composition can be prepared by mixing or the like before ingesting the subject.
 また、本発明は、ワクチン組成物、又はその有効成分である細菌若しくは該細菌に特異的な抗原を対象に摂取させることを特徴とする、対象における前記細菌に対する免疫応答を誘導する方法、又は該対象におけるTh1細胞等に起因する疾患を治療、改善若しくは予防するため方法をも提供するものである。 The present invention also comprises ingesting a vaccine composition, a bacterium as an active ingredient thereof, or an antigen specific to the bacterium into a subject, or a method for inducing an immune response against the bacterium in the subject. It also provides a method for treating, ameliorating or preventing a disease caused by Th1 cells or the like in a subject.
 本発明の組成物又はその有効成分は、ヒトを含む動物を対象として使用することができるが、ヒト以外の動物としては特に制限はなく、種々の家畜、家禽、ペット、実験用動物等を対象とすることができる。また、上述の本発明の検査方法によって、前記疾患に罹患していると判定された被検者は、好適な対象となり得る。 The composition of the present invention or an active ingredient thereof can be used for animals including humans, but the animals other than humans are not particularly limited, and various livestock, poultry, pets, laboratory animals and the like are targeted. Can be. In addition, a subject determined to be suffering from the disease by the above-mentioned test method of the present invention may be a suitable target.
 また、本発明のワクチン組成物、又はその有効成分の摂取対象としては、Th1細胞等に起因する疾患の発症の如何を問わず、小腸内等でTh1細胞等の増殖又は活性化を誘導する細菌を保有する動物が挙げられる。また予防の観点からは、該細菌を保有していない又はその保有の疑いのある動物に、本発明の組成物等を摂取させてもよい。 In addition, the vaccine composition of the present invention or an active ingredient thereof is a bacterium that induces proliferation or activation of Th1 cells or the like in the small intestine or the like regardless of the onset of a disease caused by Th1 cells or the like. Examples include animals that possess. From the viewpoint of prevention, an animal that does not possess or is suspected of possessing the bacterium may be allowed to ingest the composition of the present invention or the like.
 本発明の組成物等の摂取方法としては、特に制限はなく、経口投与であってもよく、また非経口投与であってもよい。また、本発明の組成物等を摂取させる場合、その摂取量は、対象の年齢、体重、疾患の症状、健康状態、組成物の剤型、摂取方法等に応じて、当業者であれば適宜選択することができる。 The method of ingesting the composition or the like of the present invention is not particularly limited, and may be oral administration or parenteral administration. In addition, when the composition or the like of the present invention is ingested, the amount of the ingestion may be appropriately determined by those skilled in the art depending on the age, body weight, symptoms of the disease, health condition, dosage form of the composition, ingestion method, etc. You can choose.
 <本発明のTh1細胞等の増殖又は活性化を抑制するための組成物等>
 本発明の小腸内細菌は、Th1細胞等の増殖又は活性化を誘導し、また免疫作用を増強することによって、ひいてはクローン病等のTh1細胞等に起因する疾患を誘発する。そのため、前記細菌を小腸内等より除去すれば、Th1細胞等の誘導が抑制され、また免疫作用が抑制されることによって、前記疾患の治療等に繋がる。
<Composition for suppressing proliferation or activation of Th1 cells and the like of the present invention>
The small intestinal bacterium of the present invention induces proliferation or activation of Th1 cells and the like, and enhances the immune action, thereby inducing diseases caused by Th1 cells and the like such as Crohn's disease. Therefore, if the bacterium is removed from the small intestine or the like, the induction of Th1 cells or the like is suppressed, and the immune action is suppressed, which leads to the treatment of the disease or the like.
 したがって、本発明は、本発明の小腸内細菌に対して抗菌作用を有する物質を、有効成分として含む、Th1細胞及び/又はTh17細胞の増殖若しくは活性化を抑制するための組成物、免疫を抑制するための組成物、又はTh1細胞及び/又はTh17細胞に起因する疾患を治療、改善若しくは予防するための組成物を提供する。 Therefore, the present invention is a composition for suppressing proliferation or activation of Th1 cells and / or Th17 cells, which contains a substance having an antibacterial effect against small intestinal bacteria of the present invention as an active ingredient, and suppresses immunity. To provide a composition for treating, ameliorating or preventing a disease caused by Th1 cells and / or Th17 cells.
 なお、本発明において、「治療」は、疾患からの完全な回復を意味し、「改善」には、前記疾患の症状の緩和又は改善、その進行の抑制、またその再発の抑制が含まれる。「予防」には、前記疾患の発症の抑制、遅延、及びその再発の抑制が含まれる。 In the present invention, "treatment" means complete recovery from the disease, and "improvement" includes alleviation or improvement of the symptoms of the disease, suppression of its progression, and suppression of its recurrence. "Prevention" includes suppression of the onset of the disease, delay, and suppression of its recurrence.
 本発明において「抗菌作用」としては、細菌の増殖を抑制する作用及び/又は殺菌作用を意味する。本発明の組成物に有効成分として含まれる「本発明の小腸内細菌に対して抗菌作用を有する物質」には、当該作用を有する限り、特に制限はないが、例えば、抗生物質、溶菌物質(バクテリオファージ、溶菌酵素等)、前記細菌を特異的に認識する抗体が挙げられる。また、本発明の小腸内細菌に由来する生理活性物質に結合する物質であってもよく、かかる物質としては、例えば、前記生理活性物質に結合する抗体、前記生理活性物質に結合する低分子化合物が挙げられる。さらに、上述の本発明のスクリーニング方法によって選抜される物質であってもよい。 In the present invention, the "antibacterial action" means an action of suppressing the growth of bacteria and / or a bactericidal action. The "substance having an antibacterial action against the bacteria in the small intestine of the present invention" contained as an active ingredient in the composition of the present invention is not particularly limited as long as it has the action, but for example, an antibiotic or a lytic substance ( Bacteriophage, lytic enzyme, etc.), antibodies that specifically recognize the bacterium, and the like. Further, it may be a substance that binds to a physiologically active substance derived from the small intestinal bacterium of the present invention, and examples of such a substance include an antibody that binds to the physiologically active substance and a low molecular weight compound that binds to the physiologically active substance. Can be mentioned. Furthermore, it may be a substance selected by the screening method of the present invention described above.
 本発明の組成物は、かかる本発明の小腸内細菌に対して抗菌作用を有する物質を複数種を含むものであってもよい。または組成物を複合して用いることができ、結果として併用して摂取又は吸収される場合(併用組成物の場合)、該複数の物質は2種以上の組成物の中に存在することもできる。 The composition of the present invention may contain a plurality of types of substances having an antibacterial action against the small intestinal bacteria of the present invention. Alternatively, the compositions can be used in combination, and as a result, when ingested or absorbed in combination (in the case of a combination composition), the plurality of substances can be present in two or more kinds of compositions. ..
 本発明の組成物は、医薬組成物、飲食品(動物用飼料を含む)、あるいは研究目的(例えば、インビトロやインビボの実験)に用いられる試薬の形態であり得る。また、本発明の組成物は、必要に応じ、キットの態様であってもよい。 The composition of the present invention can be in the form of a pharmaceutical composition, food or drink (including animal feed), or a reagent used for research purposes (eg, in vitro or in vivo experiments). Further, the composition of the present invention may be in the form of a kit, if necessary.
 上述の通り、本発明の組成物は、小腸内等におけるTh1細胞等の誘導及び免疫を抑制するため、上述のTh1細胞等に起因する疾患の治療、予防又は改善のための医薬組成物、飲食品として、好適に用いられる。 As described above, in order to suppress the induction and immunity of Th1 cells and the like in the small intestine and the like, the composition of the present invention is a pharmaceutical composition for treating, preventing or improving the above-mentioned diseases caused by Th1 cells and the like, eating and drinking. It is preferably used as a product.
 本発明の組成物は、公知の製剤学的方法により製剤化することができる。例えば、カプセル剤、錠剤、丸剤、液剤、散剤、顆粒剤、細粒剤、フィルムコーティング剤、ペレット剤、トローチ剤、舌下剤、咀嚼剤、バッカル剤、ペースト剤、シロップ剤、懸濁剤、エリキシル剤、乳剤、塗布剤、軟膏剤、硬膏剤、パップ剤、経皮吸収型製剤、ローション剤、吸引剤、エアゾール剤、注射剤、坐剤等として、経口的、非経口的(例えば、腸管内、筋肉内、静脈内、気管内、鼻内、経皮、皮内、皮下、眼内、膣、腹腔内、直腸若しくは吸入)、又はこれらの複数の組み合わせからなる経路による投与用に使用することができる。 The composition of the present invention can be formulated by a known pharmaceutical method. For example, capsules, tablets, pills, liquids, powders, granules, fine granules, film coatings, pellets, lozenges, sublinguals, chewing agents, buccal agents, pastes, syrups, suspensions, Oral and parenteral (eg, intestinal tract) as elixirs, emulsions, coatings, ointments, plasters, paps, transdermal formulations, lotions, inhalants, aerosols, injections, suppositories, etc. Intramuscular, intravenous, intravenous, intratracheal, intranasal, transdermal, intradermal, subcutaneous, intraocular, vaginal, intraperitoneal, rectal or inhalation), or for administration by a route consisting of multiple combinations thereof. be able to.
 これら製剤化においては、薬理学上若しくは飲食品として許容される担体、具体的には、滅菌水や生理食塩水、植物油、溶剤、基剤、乳化剤、懸濁剤、界面活性剤、安定剤、香味剤、芳香剤、賦形剤、ベヒクル、防腐剤、結合剤、希釈剤、等張化剤、無痛化剤、増量剤、崩壊剤、緩衝剤、コーティング剤、滑沢剤、着色剤、甘味剤、粘稠剤、矯味矯臭剤、溶解補助剤あるいはその他の添加剤等と適宜組み合わせることができる。 In these formulations, carriers that are pharmacologically or acceptable as foods and drinks, specifically, sterile water, physiological saline, vegetable oils, solvents, bases, emulsifiers, suspending agents, surfactants, stabilizers, etc. Flavors, fragrances, excipients, vehicles, preservatives, binders, diluents, isotonic agents, soothing agents, bulking agents, disintegrants, buffers, coating agents, lubricants, colorants, sweetness It can be appropriately combined with an agent, a thickener, a flavoring agent, a solubilizing agent, or other additives.
 また、これら製剤化においては、腸管内におけるTh1細胞の増殖又は活性化及び免疫をより効率的に抑制する等の観点から、特に経口投与を目的とする製剤においては、本発明の組成物を小腸内等に効率良く送達することを可能にする組成物と組み合わせてもよい。このような小腸内等への送達を可能とする組成物については特に制限されることなく、公知の組成物を適宜採用することができ、例えば、pH感受性組成物、小腸等までの放出を抑制する組成物(セルロース系ポリマー、アクリル酸重合体及び共重合体、ビニル酸重合体及び共重合体等)、小腸粘膜特異的に接着する生体接着性組成物、プロテアーゼ阻害剤含有組成物、小腸内酵素によって特異的に分解される組成物が挙げられる。 Further, in these formulations, from the viewpoint of more efficiently suppressing the proliferation or activation of Th1 cells in the intestinal tract and immunity, particularly in the formulation intended for oral administration, the composition of the present invention is used in the small intestine. It may be combined with a composition that enables efficient delivery to the inside and the like. The composition capable of such delivery into the small intestine or the like is not particularly limited, and a known composition can be appropriately adopted, for example, a pH-sensitive composition, suppression of release to the small intestine or the like. Compositions (cellulose-based polymers, acrylic acid polymers and copolymers, vinyl acid polymers and copolymers, etc.), bioadhesive compositions that specifically adhere to the small intestinal mucosa, protease inhibitor-containing compositions, in the small intestine Examples thereof include compositions that are specifically degraded by an enzyme.
 また、本発明のTh1細胞等の増殖若しくは活性化又は免疫を抑制する組成物を医薬組成物として用いる場合には、Th1細胞等に起因する疾患の治療、予防又は改善に用いられる公知の物質(例えば、抗炎症剤、免疫抑制剤)を更に含んでいてもよく、またかかる物質と併用してもよい。 Further, when the composition that suppresses the proliferation or activation or immunity of Th1 cells or the like of the present invention is used as a pharmaceutical composition, a known substance used for the treatment, prevention or improvement of diseases caused by Th1 cells or the like ( For example, an anti-inflammatory agent, an immunosuppressant) may be further contained, or may be used in combination with such a substance.
 本発明の組成物を飲食品として用いる場合、当該飲食品は、例えば、健康食品、機能性食品、特定保健用食品、栄養機能食品、機能性表示食品、栄養補助食品、病者用食品、あるいは動物用飼料であり得る。なお、機能性食品は、通常、その作用メカニズムに基づき、プロバイオティクス、バイオジェニクス、プレバイオティクス、シンバイオティクスの4つに分類され、本発明においては、プロバイオティクス、バイオジェニクス、シンバイオティクス、プレバイオティクスの態様をとり得る。また、飲食品の具体例としては、発酵飲料、油分を含む製品、スープ類、乳飲料、清涼飲料水、茶飲料、アルコール飲料、ドリンク剤、ゼリー状飲料等の液状食品;炭水化物含有食品;畜産加工食品;水産加工食品;野菜加工食品;半固形状食品;発酵食品;菓子類;レトルト製品;電子レンジ対応食品等が挙げられる。さらには、粉末、穎粒、錠剤、カプセル剤、液状、ペースト状又はゼリー状に調製された健康飲食品も挙げられる。なお、本発明における飲食品の製造は、当該技術分野に公知の製造技術により実施することができる。当該飲食品においては、Th1疾患に起因する疾患の改善又は予防に有効な成分(例えば、栄養素等)を添加してもよい。また、当該改善等以外の機能を発揮する他の成分あるいは他の機能性食品と組み合わせることによって、多機能性の飲食品としてもよい。 When the composition of the present invention is used as a food or drink, the food or drink may be, for example, a health food, a functional food, a food for specified health use, a food with a nutritional function, a food with a functional claim, a nutritional supplement, a food for the sick, or It can be an animal feed. Functional foods are usually classified into four categories, probiotics, biogenics, prebiotics, and symbiotics, based on their mechanism of action. In the present invention, probiotics, biogenics, and symbiotics are classified. It can take the form of tics or prebiotics. Specific examples of foods and drinks include fermented beverages, oil-containing products, soups, dairy beverages, soft drinks, tea beverages, alcoholic beverages, drinks, jelly-like beverages and other liquid foods; carbohydrate-containing foods; livestock. Processed foods; processed marine products; processed vegetable foods; semi-solid foods; fermented foods; confectionery; retort products; microwave-compatible foods and the like. Further, healthy foods and drinks prepared in the form of powder, granules, tablets, capsules, liquid, paste or jelly can also be mentioned. The food and drink according to the present invention can be produced by a production technique known in the art. In the food and drink, an ingredient (for example, nutrients, etc.) effective for improving or preventing a disease caused by Th1 disease may be added. In addition, it may be a multifunctional food or drink by combining it with other ingredients or other functional foods that exhibit functions other than the improvement.
 本発明の組成物の製品(医薬品、飲食品、試薬)又はその説明書は、Th1細胞等の増殖若しくは活性化を抑制する、免疫を抑制する、又はTh1細胞等に起因する疾患を治療、改善若しくは予防するために用いられる旨の表示を付したものであり得る。 The product (pharmaceutical product, food or drink, reagent) of the composition of the present invention or a description thereof suppresses the proliferation or activation of Th1 cells or the like, suppresses immunity, or treats or improves a disease caused by Th1 cells or the like. Alternatively, it may be labeled as being used for prevention.
 また本発明で提起される飲食品組成物において、保健用途が表示された飲食品として提供・販売されることも可能である。「表示」行為には、需要者に対して前記用途を知らしめたる全ての行為が含まれ、前記用途を直接的に認識できる表現、あるいは前記用途を想起・類推させうるような表現を、組成物自体に付したものであってもよいし、組成物が含入する容器、包装材又は添付文書に付したものであってもよい。また「表示」は、本発明組成物の関連する情報として、チラシ、パンフレット、ポップ、カタログ、ポスター、書籍、DVD等の記憶媒体、電子掲示板やインターネット等の広告等で、本発明の組成物が効果的であることを表示・広告するものであってもよい。 It is also possible to provide and sell the food and drink composition proposed in the present invention as a food and drink with a health use indication. The act of "displaying" includes all acts that inform the consumer of the use, and constitutes an expression that can directly recognize the use or an expression that can recall or infer the use. It may be attached to the product itself, or it may be attached to the container, packaging material or package insert containing the composition. Further, "display" refers to information related to the composition of the present invention, such as leaflets, pamphlets, pops, catalogs, posters, books, storage media such as DVDs, advertisements such as electronic bulletin boards and the Internet, and the like. It may be something that displays / advertises that it is effective.
 また、本発明は、Th1細胞等の増殖若しくは活性化を抑制するための組成物、免疫を抑制するための組成物、又はそれらの有効成分である本発明の小腸内細菌に対して抗菌作用を有する物質を対象に摂取させることを特徴とする、対象におけるTh1細胞等の増殖若しくは活性化を抑制する方法、該対象における免疫を抑制する方法、又は該対象におけるTh1細胞等に起因する疾患を治療、改善又は予防する方法をも提供するものである。 In addition, the present invention has an antibacterial action against the composition for suppressing the proliferation or activation of Th1 cells and the like, the composition for suppressing immunity, or the small intestinal bacterium of the present invention which is an active ingredient thereof. A method for suppressing the proliferation or activation of Th1 cells or the like in a subject, a method for suppressing immunity in the subject, or a disease caused by Th1 cells or the like in the subject, which is characterized by ingesting a substance having the substance. It also provides a way to improve or prevent.
 本発明の組成物又はその有効成分は、ヒトを含む動物を対象として使用することができるが、ヒト以外の動物としては特に制限はなく、種々の家畜、家禽、ペット、実験用動物等を対象とすることができる。また、本発明にかかる対象は、Th1細胞等に起因する疾患の罹患者であってもよいが、これら疾患と診断されている必要はなく、例えば、これら疾患に罹患したおそれがある者、これら疾患に罹患したと感じている者が挙げられる。さらに、健常者であっても、これら疾患の予防等を目的として日常的に摂取することができる。また、上述の本発明の検査方法によって、前記疾患に罹患していると判定された被検者は、好適な対象となり得る。 The composition of the present invention or an active ingredient thereof can be used for animals including humans, but the animals other than humans are not particularly limited, and various livestock, poultry, pets, laboratory animals and the like are targeted. Can be. Further, the subject according to the present invention may be a person suffering from a disease caused by Th1 cells or the like, but it is not necessary to be diagnosed with these diseases, for example, a person who may have suffered from these diseases, these. Some people feel that they have a disease. Furthermore, even a healthy person can take it on a daily basis for the purpose of preventing these diseases. In addition, a subject determined to be suffering from the disease by the above-mentioned test method of the present invention may be a suitable target.
 本発明の組成物等の摂取方法としては、特に制限はなく、経口投与であってもよく、また非経口投与(例えば、小腸内への投与)であってもよいが、経口投与である場合には、本発明の組成物等の効果をより向上させるという観点から、本発明の組成物等の摂取対象は、プロトンポンプ阻害剤(PPI)等の摂取により胃酸の産生を減少させておくことが好ましい。 The method of ingesting the composition or the like of the present invention is not particularly limited and may be oral administration or parenteral administration (for example, administration into the small intestine), but in the case of oral administration. In order to further improve the effect of the composition of the present invention, the subject of ingestion of the composition of the present invention should reduce the production of gastric acid by ingesting a proton pump inhibitor (PPI) or the like. Is preferable.
 また、本発明の組成物等を摂取させる場合、その摂取量は、対象の年齢、体重、疾患の症状、健康状態、組成物の種類(医薬品、飲食品等)、摂取方法等に応じて、当業者であれば適宜選択することができる。 In addition, when the composition or the like of the present invention is ingested, the ingestion amount depends on the age, weight, disease symptom, health condition, type of composition (pharmaceuticals, foods and drinks, etc.), intake method, etc. of the subject. Those skilled in the art can appropriately select it.
 以下、実施例に基づいて本発明をより具体的に説明するが、本発明は以下の実施例に限定されるものではない。なお、本実施例は以下に示す材料及び方法を用いて行なった。 Hereinafter, the present invention will be described in more detail based on Examples, but the present invention is not limited to the following Examples. This example was carried out using the materials and methods shown below.
 <材料及び方法>
 (患者とサンプル収集)
 小腸(SI)粘膜サンプルを、ダブルバルーン内視鏡検査(DBE)システムを用いて進行中の臨床ケアを受けた27人のクローン病(CD)患者及び17人の非CD患者から、生検又は擦過にて採取した。逆行性内視鏡検査を受けた患者は検査の前日から腸洗浄処置を受けた。鎮静のためミダゾラムとペチジンを静脈内投与し、腸ぜん動を減少させるためにチメピジウムブロミド水和物又はグルカゴンを使用した。患者の状態に基づいて、DBEの挿入経路(順行性又は逆行性)、内視鏡の種類(EN-580 T、EN-450 T 5又はEN-450 P 5(富士フイルム、東京))及びサンプリング方法を決定した。
<Materials and methods>
(Patient and sample collection)
Small intestinal (SI) mucosal samples were biopsied or biopsied from 27 Crohn's disease (CD) patients and 17 non-CD patients who received ongoing clinical care using a double-balloon endoscopy (DBE) system. Collected by scraping. Patients who underwent retrograde endoscopy underwent intestinal lavage from the day before the examination. Midazolam and pethidine were given intravenously for sedation and timepidium bromide hydrate or glucagon was used to reduce intestinal peristalsis. Based on the patient's condition, DBE insertion pathway (antegrade or prograde), endoscope type (EN-580 T, EN-450 T 5 or EN-450 P 5 (Fujifilm, Tokyo)) and The sampling method was decided.
 粘膜生検又は粘膜擦過物を、それぞれ内視鏡生検鉗子(米国マサチューセッツ州ボストン・サイエンティフィックのRadial Jaw 4 P)又は内視鏡検索ネット(Roth net、米国内視鏡検査、オハイオ州、米国)を用い、各患者の中央SIから得た。同時に、粘膜標本を採取する前に、内視鏡を通してSI液(約50cc)を吸引した。また、内視鏡検査の1日前に唾液と便を採取した。 For mucosal biopsy or mucosal scrapes, endoscopic biopsy forceps (Radial Jaw 4P, Boston Scientific, Massachusetts, USA) or endoscopy search net (Roth net, US endoscopy, Ohio, USA) US) was used and obtained from the central SI of each patient. At the same time, SI solution (about 50 cc) was aspirated through the endoscope before collecting the mucosal specimen. In addition, saliva and stool were collected one day before endoscopy.
 DNA抽出のために、生検サンプルをTE10(10mM Tris-HCl、10mM EDTA)緩衝液に懸濁し、-80℃で保存し、粘膜擦過物及び腸液サンプルを遠心分離し、ペレットをTE10緩衝液に溶解し、-80℃で保存した。細菌培養のために、生検サンプルを20%グリセロール/PBSに懸濁し、液体窒素中で凍結し、次いで-80℃で保存した。粘膜擦過サンプルを遠心分離し、ペレットを20%グリセロール/PBSに溶解し、液体窒素中で凍結し、そして-80℃で保存した。 For DNA extraction, the biopsy sample is suspended in TE10 (10 mM Tris-HCl, 10 mM EDTA) buffer, stored at -80 ° C, the mucosal scrapes and intestinal juice samples are centrifuged, and the pellet is placed in TE10 buffer. It was dissolved and stored at −80 ° C. For bacterial culture, biopsy samples were suspended in 20% glycerol / PBS, frozen in liquid nitrogen and then stored at -80 ° C. Mucosal scraping samples were centrifuged, pellets were dissolved in 20% glycerol / PBS, frozen in liquid nitrogen and stored at -80 ° C.
 (DNA抽出と16SrRNA微生物叢シーケンス)
 凍結サンプルを解凍し、RNaseA(最終濃度100μgml-1、Invitrogen、MA、USA)及びリゾチーム(終濃度15mgml-1、Sigma、MO、USA)を含有する800μlのTE10緩衝液と混合した。懸濁液を穏やかに混合しながら1時間37℃でインキュベートした。精製したアクロモペプチダーゼ(Wako、大阪、日本)を最終濃度2,000ユニットml-1で添加し、サンプルをさらに30分間37℃でインキュベートした。その後、ドデシル硫酸ナトリウム(終濃度1%)及びプロテイナーゼK(終濃度1mgml-1、スイス、バーゼル、ロシュ)を懸濁液に加え、混合物を1時間55℃でインキュベートし、フェノール:クロロホルム:イソアミルアルコール(25:24:1)を用いて高分子量DNAを抽出した。次いで、イソプロパノールで沈殿させ、75%エタノールで洗浄し、TE 50μl中に再懸濁した。
(DNA extraction and 16S rRNA microflora sequence)
Frozen samples were thawed and mixed with 800 μl TE10 buffer containing RNaseA (final concentration 100 μgml- 1 , Invitrogen, MA, USA) and lysozyme (final concentration 15 mgml- 1, Sigma, MO, USA). The suspension was incubated at 37 ° C. for 1 hour with gentle mixing. Purified acromopeptidase (Wako, Osaka, Japan) was added at a final concentration of 2,000 units ml-1 , and the sample was incubated for an additional 30 minutes at 37 ° C. Then sodium dodecyl sulfate (final concentration 1%) and proteinase K (final concentration 1 mgml-1, Switzerland, Basel, Roche) were added to the suspension and the mixture was incubated for 1 hour at 55 ° C. Phenol: chloroform: isoamyl alcohol. High molecular weight DNA was extracted using (25:24: 1). It was then precipitated with isopropanol, washed with 75% ethanol and resuspended in 50 μl TE.
 16SrRNA遺伝子のV1-V2領域を標的とする27Fmod 5’-AGRGTTTTGATYMTGCTCAG-3’(配列番号:10)及び338R 5’-TGCTGCCTCGTGAGT-3’ (配列番号:11)のプライマーセットを用いてPCRを実施した。 PCR was performed using primer sets of 27Fmod 5'-AGRGTTTTGATYMTGCTCAG-3'(SEQ ID NO: 10) and 338R 5'-TGCTGCCTCGTGGT-3' (SEQ ID NO: 11) targeting the V1-V2 region of the 16S rRNA gene. ..
 続いて、各サンプル(約330bp)から生成したアンプリコンを、AMPure XP(ベックマン・コールター、カリフォルニア州、米国)を用いて精製した。DNAはQuant-iT Picogreen dsDNAアッセイキット(Invitrogen)及びTBS-380ミニ蛍光光度計(Turner Biosystems、カリフォルニア州、米国)を用いて定量した。 Subsequently, the amplicon produced from each sample (about 330 bp) was purified using APPure XP (Beckman Coulter, CA, USA). DNA was quantified using a Quant-iT Picogreen ds DNA assay kit (Invitrogen) and a TBS-380 mini fluorometer (Turner Biosystems, CA, USA).
 16Sメタゲノムシーケンシングは、Illuminaプロトコルに従ってMiSeqを用いて行った。重複シーケンスに基づくfastq-joinプログラムを用いて、2つの対末端読み取りを併合した。平均クオリティーバリューが<25であり、両方のユニバーサルプライマーとの不正確な一致を有する読み取りを除外した。フィルターを通過したリードを、両方のプライマー配列をトリミングした後のさらなる分析のために使用した。各サンプルについて、3,000の品質フィルターを通過したリードは、それらの品質値に従って降順に並べ替え、UCLUSTプログラムバージョン5.2.32を使用して、97%のpair-wise-identityカットオフを有するOTUにクラスタ化した(https://www.drive5.com)。 16S metagenomic sequencing was performed using MiSeq according to the Illumina protocol. Two opposite-end reads were merged using a fastq-join program based on overlapping sequences. Reads with an average quality value of <25 and inaccurate matches with both universal primers were excluded. The filtered reads were used for further analysis after trimming both primer sequences. For each sample, reads that have passed through the 3,000 quality filters are sorted in descending order according to their quality value and have a 97% pair-wise-identity cutoff using UCLUST program version 5.2.32. Clustered into OTUs having (https://www.drive5.com).
 各OTUの分類学的帰属を、GLSEARCHプログラムを用いて、RDP及び米国バイオテクノロジー情報センター(NCBI)ゲノムデータベースとの類似性に基づいて行った。細菌保有率を決定するために、相対量>0.1%の分類群を陽性とみなした。 Taxonomic attribution of each OTU was made using the GLSEARCH program based on similarity to RDP and the National Center for Biotechnology Information (NCBI) genomic database. Taxa with relative amount> 0.1% were considered positive to determine bacterial prevalence.
 (細菌の培養と単離及びノトバイオート動物の作製)
 凍結サンプルを解凍し、PBSで連続希釈し、非選択的及び選択的寒天プレート(,好気培養用:トリプチカーゼソイアガー、嫌気培養用:EG、BHK、MRS、CM 0619+SR 0107)上に播種した(図8A)。EG及びBHKは嫌気性細菌の非選択的培地である。MRSは乳酸菌の選択的分離培地である。SR 0107を添加したCM 0619は非胞子形成嫌気性菌に対する選択的分離培地である。
(Culturing and isolation of bacteria and production of gnotobiotic animals)
Frozen samples are thawed, serially diluted with PBS and seeded on non-selective and selective agar plates (for aerobic culture: trypticase soiagar, for anaerobic culture: EG, BHK, MRS, CM 0619 + SR 0107). (Fig. 8A). EG and BHK are non-selective media for anaerobic bacteria. MRS is a selective isolation medium for lactic acid bacteria. CM 0619 supplemented with SR 0107 is a selective isolation medium for non-spore-forming anaerobes.
 37℃の好気条件で1日間、又は、嫌気性チャンバー(Coy Laboratory Products、ミシガン州、米国)にて、嫌気条件(80% N、10% H、10% CO)下、37℃で2~4日間培養した後、全部で572の個々のコロニーを採取し、それらの16SrRNA遺伝子をユニバーサルプライマー(27Fmod:5’-AGRGTTGATYMTGGCTCAG-3’(配列番号:10)、1492R:5’-GGYTACCTTGTTACGT-3’(配列番号:12))で増幅し、配列を決定した。これらの16SrRNA配列に基づく系統樹を近隣結合法を用いて構築した。培養コレクション中の個々の分離株は、それらの16SrRNA配列が>98%同一性を示す場合、「株」として分類した。また、得られた菌株配列を、NCBIデータベースの配列及び16SrRNA解析で観測されたOTUと比較し、近縁種及びそれらに対応するOTUを決定した。 37 ° C. for 1 day in an aerobic condition at 37 ° C. or in an anaerobic chamber (Coy Laboratory Products, Michigan, USA) under anaerobic conditions (80% N 2 , 10% H 2 , 10% CO 2 ). After culturing in 2-4 days, a total of 572 individual colonies were collected and their 16S rRNA genes were subjected to universal primers (27Fmod: 5'-AGRGTTGATYMTGGCTCAG-3'(SEQ ID NO: 10), 1492R: 5'-GGYTACCTTGTTTACGT. The sequence was determined by amplification with -3'(SEQ ID NO: 12)). A phylogenetic tree based on these 16S rRNA sequences was constructed using the neighbor-joining method. Individual isolates in the culture collection were classified as "strains" if their 16S rRNA sequences showed> 98% identity. In addition, the obtained strain sequence was compared with the sequence of the NCBI database and the OTU observed by 16S rRNA analysis, and the closely related species and the corresponding OTU were determined.
 腸内細菌科に属する分離細菌の同定は、TSI斜面培地(BD、ニュージャージー州、米国)、LIM培地、SIM培地(栄研化学、東京、日本)及びAPI 20 E(BioMerieux、Marcy-l’Etoile、フランス)を、各々の製造業者の指示に従って使用し、行った。また、標準プロトコルを用いてグラム染色を行った。 Identification of isolated bacteria belonging to Enterobacteriaceae is performed on TSI slope medium (BD, NJ, USA), LIM medium, SIM medium (Eiken Chemical, Tokyo, Japan) and API 20E (BioMerieux, Marcy-l'Etoile). , France) was used and performed according to the instructions of each manufacturer. In addition, Gram stain was performed using a standard protocol.
 ノトバイオートマウスを作製するために、各E.coli株(35A1、LF82及びMG1655)を37℃のTSブロス中で一晩好気的に培養し、7~9×10コロニー形成単位(CFU)を含む200μlのアリコートをGFマウスに強制経口投与した。なお、本実施例で用いたCD関連AIEC株 LF82は、元々、Darfeuille-MichaudらによりCD患者の回腸粘膜から単離されたものである(文献16)。R.gnavus 131A1株は、EGブロス中又はEGプレート上で嫌気培養した。プレート上のコロニーをこすり、EGブロスに再懸濁した。E.coli 35A1株及びR.gnavus 131A1株以外の7株はEG又はHK培地で嫌気培養した。その後、等量の細菌懸濁液を混合し、2-mix又は9-mixを調製した。分離株の混合物又は単一細菌懸濁液をGFマウスに経口投与し、糞便懸濁液のグラム染色によりコロニー形成を評価した。 In order to make notobiate mice, each E.I. coli strain (35A1, LF82 and MG1655) were cultured overnight aerobically in TS broth 37 ° C., gavage aliquots of 200μl containing 7 ~ 9 × 10 8 colony forming units (CFU) in GF mice bottom. The CD-related AIEC strain LF82 used in this example was originally isolated from the ileal mucosa of a CD patient by Darfeile-Michaud et al. (Reference 16). R. The gnavus 131A1 strain was anaerobically cultured in EG broth or on an EG plate. The colonies on the plate were rubbed and resuspended in EG broth. E. coli 35A1 strain and R. Seven strains other than gnavus 131A1 strain were anaerobically cultured in EG or HK medium. Then, equal amounts of bacterial suspension were mixed to prepare 2-mix or 9-mix. A mixture of isolates or a single bacterial suspension was orally administered to GF mice and colonization was evaluated by Gram stain of the fecal suspension.
 特定の細菌株の混合物又は単一の細菌懸濁液を投与した全てのマウスは、12時間/12時間の明/暗サイクルにて、単一のノトバイオティックアイソレーターで維持した。GF B6マウス(生後7~14週)は、最初の強制経口投与の3週間後に分析した。全ての動物実験は、慶應義塾大学動物実験委員会の承認を得て行なった。GF及びSPFマウスは全部、CLEA Japan(日本、東京)から得たB6マウスである。 All mice administered a mixture of specific bacterial strains or a single bacterial suspension were maintained on a single notobiotic isolator with a 12 hour / 12 hour light / dark cycle. GFB6 mice (7-14 weeks old) were analyzed 3 weeks after the first gavage. All animal experiments were conducted with the approval of the Keio University Animal Experiment Committee. The GF and SPF mice are all B6 mice obtained from CLEA Japan (Tokyo, Japan).
 (LP単核細胞の単離及びフローサイトメトリー)
 腸リンパ球は以下のようにして単離した。先ず、腸を縦に開き、管腔内容物を除去するためにPBSで洗浄した。全サンプルを、20mlの5mM EDTA含有Hanks平衡塩溶液(HBSS)に入れ、振とう水浴中にて、37℃で20分間インキュベートし、腸上皮細胞(IEC)を除去した。その後、筋層と脂肪組織を、鉗子を用いて手で除去した。残りのLP層を小片に切断し、10mlのRPMI 1640中で、4%のウシ胎児血清、0.5mgml-1コラゲナーゼD(Roche)、0.5mgml-1ディスパーゼ(Gibco)及び40μgml-1DNaseI(Roche)を含有し、振盪水浴中で、37℃で45分間インキュベートした。
(Isolation of LP mononuclear cells and flow cytometry)
Intestinal lymphocytes were isolated as follows. First, the intestine was opened vertically and washed with PBS to remove the luminal contents. All samples were placed in 20 ml of 5 mM EDTA-containing Hanks Balanced Salt Solution (HBSS) and incubated in a shaking water bath at 37 ° C. for 20 minutes to remove enterocytes (IEC). The muscle layer and adipose tissue were then manually removed using forceps. The remaining LP layer was cut into small pieces and in 10 ml RPMI 1640, 4% fetal bovine serum, 0.5 mgml -1 collagenase D (Roche), 0.5 mgml -1 dispase (Gibco) and 40 μgml -1 DNase I ( Roche) was contained and incubated in a shaking water bath at 37 ° C. for 45 minutes.
 消化された組織を、10mlの5mM EDTAを含むHBSSで洗浄し、5mlの40% Percoll(GE Healthcare、イリノイ州、米国)に再懸濁し、15mlのFalconチューブ中にて、2.5mlの80%Percollを重層した。900×g、30分間、25℃で遠心分離によりパーコール勾配分離を行った。二層の界面からリンパ球を含む画分を回収し、10% FBSを含むRPMI1640で洗浄した。 Digested tissue was washed with HBSS containing 10 ml 5 mM EDTA, resuspended in 5 ml 40% Percoll (GE Healthcare, Illinois, USA) and in a 15 ml Falcon tube, 2.5 ml 80%. Percoll was layered. Percoll gradient separation was performed by centrifugation at 900 xg for 30 minutes at 25 ° C. Fractions containing lymphocytes were collected from the interface between the two layers and washed with RPMI 1640 containing 10% FBS.
 サイトカイン検出のために、細胞を50ngml-1PMA及び750ngml-1イオノマイシン(両方ともSigmaから購入)で、GolgiStop(BD)の存在下、37℃で3.5時間刺激した。 For cytokine detection, cells were stimulated with 50 ngml -1 PMA and 750 ngml -1 ionomycin (both purchased from Sigma) at 37 ° C. for 3.5 hours in the presence of GolgiStop (BD).
 Ghost Dye780(Tonbo Biosciences、カリフォルニア州、米国)で標識した後、細胞を透過処理し、抗TCRβ(BV605;バイオレジェンド、カリフォルニア州、米国)、抗CD4抗体(BV510;バイオレジェンド)、抗TCRγδ抗体(BV421;バイオレジェンド)、抗IFN-γ抗体(ファンクション;バイオレジェンド)、抗IL-17A抗体(PerCP-Cy5.5;eBioscience、カリフォルニア州、米国)、抗TNF-α抗体(PE/Cy7;バイオレジェンド)、抗RORγt抗体(APC;eBioscience)、及び抗DR3抗体(PE;バイオレジェンド)、並びにFoxp3/転写因子染色バッファーキット(eBioscience)を用い、メーカーの指示に従って、免疫染色を行った。 After labeling with Ghost Dye780 (Tombo Biosciences, California, USA), the cells were permeabilized and treated with anti-TCRβ (BV605; BioLegend, California, USA), anti-CD4 antibody (BV510; BioLegend), anti-TCRγδ antibody (BV510; biolegend). BV421; BioLegend), Anti-IFN-γ antibody (Function; BioLegend), Anti-IL-17A antibody (PerCP-Cy5.5; eBioscience, California, USA), Anti-TNF-α antibody (PE / Cy7; BioLegend) ), Anti-RORγt antibody (APC; eBioscience), and anti-DR3 antibody (PE; BioLegend), and Foxp3 / transcription factor staining buffer kit (eBioscience) were used for immunostaining according to the manufacturer's instructions.
 セルソーター BD FACS Aria IIIuを用いて全データを収集し、Flowjoソフトウェア(TreeStar)にて全ての分析を行なった。なお、CD4T細胞は、生細胞ゲート内のCD4+TCRβ+細胞集団と定義した。 All data were collected using the cell sorter BD FACS Aria IIIu and all analysis was performed using Flowjo software (TreeStar). CD4T cells were defined as CD4 + TCRβ + cell population within the live cell gate.
 (抗IL-10R抗体を介した大腸炎モデル)
 抗IL-10受容体(IL-10R)抗体で誘発する大腸炎マウスモデルを、Schieringら(文献17)の記載の通りに作製した。すなわち、GF野生型B6マウスに、1日目にE.coli 35A1株、LF82株又はMG1655株を定着させ、続いて抗マウスIL-10R抗体(1mg/個体)(BioXcell、ニューハンプシャー州、米国)を1日目から実験の終了まで毎週腹腔内注射した。そして、最初の定着から5週間後にマウスを分析した。
(A model of colitis mediated by anti-IL-10R antibody)
A mouse model of colitis induced by an anti-IL-10 receptor (IL-10R) antibody was prepared as described in Schiering et al. (Reference 17). That is, on the first day, E.I. The coli 35A1, LF82 or MG1655 strains were established, followed by weekly intraperitoneal injection of anti-mouse IL-10R antibody (1 mg / individual) (BioXcell, New Hampshire, USA) from day 1 to the end of the experiment. Mice were then analyzed 5 weeks after initial colonization.
 (組織学的解析)
 腸炎症の発生と重症度を評価するために、盲腸と近位及び遠位結腸を4%パラホルムアルデヒドで固定し、パラフィンに包埋した。次いで、切片を作製し、ヘマトキシリン・エオジン染色を施し、観察した。炎症の程度は、以下の基準に従って盲検的に等級付けした。
炎症性細胞浸潤(スコア、0-4)、粘膜肥厚(スコア、0-4)、杯細胞枯渇(スコア、0-4)、陰窩膿瘍(スコア、0-4)及び構造破壊(スコア、0-4)。
最終的な組織学的スコアは、これらのパラメータのスコアの合計として定義した。
(Histological analysis)
To assess the onset and severity of enteritis, the cecum and proximal and distal colons were fixed with 4% paraformaldehyde and embedded in paraffin. Then, sections were prepared, stained with hematoxylin and eosin, and observed. The degree of inflammation was blindly graded according to the following criteria:
Inflammatory cell infiltration (score, 0-4), mucosal thickening (score, 0-4), goblet cell depletion (score, 0-4), crypt abscess (score, 0-4) and structural destruction (score, 0) -4).
The final histological score was defined as the sum of the scores for these parameters.
 (RNA単離及びqPCR)
 トータルRNAを、TRIzol試薬(Invitrogen)を用いて、製造業者のインストラクションに従って擦過結腸上皮から単離した。cDNAはReverTra Ace qPCR RT Master Mix(東洋紡、大阪、日本)を用いて合成した。そして、cDNAを鋳型とし、以下のプライマーセット及びThunderbird SYBR qPCR Mix(東洋紡)を用い、LightCycler480(Roche)にてqPCRを行なった。
Actb:5’-TATGCCAACACACACACACACGTGTC-3’(配列番号:13)及び5’-ACCGATCCACACAGAGTACTTG-3’(配列番号:14)
Tnfa:5’-TCATACCAGGAAAAGTCAACCTC-3’(配列番号:15)及び5’-GTATGGGCTCATACCAGGTTT-3’(配列番号:16)。
(RNA isolation and qPCR)
Total RNA was isolated from the scraped colonic epithelium using TRIzol reagent (Invitrogen) according to the manufacturer's instructions. The cDNA was synthesized using Revertra Ace qPCR RT Master Mix (Toyobo, Osaka, Japan). Then, using cDNA as a template, the following primer set and Thunderbird SYBR qPCR Mix (Toyobo) were used, and qPCR was performed with LightCyclor480 (Roche).
Actb: 5'-TATGCCAACACACACACACGTGTC-3'(SEQ ID NO: 13) and 5'-ACCGATCCACAGAGAGTACTTG-3'(SEQ ID NO: 14)
Tnfa: 5'-TCATACCAGGAAAAGTCACTC-3'(SEQ ID NO: 15) and 5'-GTATGGGGCTCATACCAGGTTT-3' (SEQ ID NO: 16).
 (統計学的解析)
 GraphPadPrism7ソフトウェア(GraphPad Software、カルフォルニア州、米国)を用いて統計解析を行った。特に、対応のないスチューデントのt検定(パラメトリック)を全ての2群間比較に用いた。多群比較には一元配置分散分析(ANOVA)と、それに続くTukeyの事後検定(パラメトリック)を用いた。カイ二乗検定を用いて、異なる群間の細菌存在率を比較した。2つのグループ化変数を持つデータに対しては、Bonferroniの事後検定に続く2方向ANOVAを用いた。Bray-Curtis距離ベースの非計量多次元スケーリング(NMDS)とペアワイズ多レベル比較による置換多変量分散分析(PERMANOVA)を、Rソフトウェア(バージョン3.6.1、veganパッケージ、phyloseq)を用いて実施した。2群間の多重比較は、FDR法とLEfSeによる多重t検定を用いて行なった(文献32)。P値<0.05を統計的に有意であるとした。
(Statistical analysis)
Statistical analysis was performed using GraphPad Prism7 software (GraphPad Software, CA, USA). In particular, the unpaired Student's t-test (parametric) was used for all two-group comparisons. One-way analysis of variance (ANOVA) followed by Tukey's post-test (parametric) was used for multigroup comparison. Bacterial abundance between different groups was compared using a chi-square test. For data with two grouping variables, a two-way ANOVA following Bonferroni's post-test was used. Bray-Curtis distance-based non-metric multidimensional scaling (NMDS) and pairwise multilevel comparison ANOVA (PERMANOVA) were performed using R software (version 3.6.1, vegan package, phyloseq). .. Multiple comparisons between the two groups were performed using the FDR method and the multiple t-test by LEfSe (Reference 32). A P value <0.05 was considered to be statistically significant.
 以上の材料及び方法を用いて行なった解析及び得られた結果を、以下に示す。 The analysis and results obtained using the above materials and methods are shown below.
 <CD患者のSI粘膜における特徴的な微生物叢構造>
 SI粘膜サンプルは、DBE法中の生検又は擦過により、27人のCD患者及び17人の非CD患者(SI以外の疾患、過誤腫性疾患等を有する患者等、下記表1参照)から得た(図1A)。CD患者のうち、23名の患者において腸狭窄(モントリオール分類B2又はB3)が認められ、19名は抗腫瘍壊死因子-α(TNF-α)抗体による治療を受けていた(表1)。DBEの挿入経路とサンプリング方法は、患者の状態に基づいて内視鏡専門医が決定した。順行性(経口)挿入経路を選択した場合、遠位空腸粘膜を採取した。逆行性(経肛門)挿入の場合、近位回腸粘膜を採取した。小腸サンプルは、活動性潰瘍又は狭窄の外側の粘膜から得た。

Figure JPOXMLDOC01-appb-T000001
<Characteristic microbial flora structure in SI mucosa of CD patients>
SI mucosal samples were obtained from 27 CD patients and 17 non-CD patients (patients with diseases other than SI, hamartomatous diseases, etc., see Table 1 below) by biopsy or scraping during the DBE method. (Fig. 1A). Of the CD patients, 23 had intestinal stenosis (Montreal classification B2 or B3) and 19 were treated with anti-tumor necrosis factor-α (TNF-α) antibody (Table 1). The insertion route and sampling method of DBE were determined by the endoscopist based on the patient's condition. If the antegrade (oral) insertion route was selected, the distal jejunal mucosa was harvested. For retrograde (transanal) insertion, the proximal ileal mucosa was collected. Small intestinal samples were obtained from the mucosa outside the active ulcer or stenosis.

Figure JPOXMLDOC01-appb-T000001
 次に、Illumina MiSeqプラットフォームを用い、16SrRNA遺伝子配列解析を行い、2,660の操作的分類単位(operational taxonomic unit;OTU)を44のSI粘膜サンプルから検出した。 Next, 16S rRNA gene sequence analysis was performed using the Illumina MiSeq platform, and 2,660 operational taxonomy units (OTUs) were detected in 44 SI mucosal samples.
 その結果、α多様性に有意な違いはなかったが(図1B)、Bray-Curtis距離に基づく非計量多次元尺度法(NMDS)プロットにより明らかになったように(図1C)、細菌叢組成はCD患者と対照(非CD患者)との間で有意に異なっていた。なお、CD患者と非CD患者との間で、細菌叢多様性に関し、挿入経路又はサンプリング手順による有意差はなかった(図5A及び5F)。 As a result, there was no significant difference in α-diversity (Fig. 1B), but as revealed by non-metric multidimensional scaling (NMDS) plots based on the Bray-Curtis distance (Fig. 1C), bacterial flora composition. Was significantly different between CD patients and controls (non-CD patients). There was no significant difference in bacterial flora diversity between CD and non-CD patients depending on the insertion route or sampling procedure (FIGS. 5A and 5F).
 CD患者の小腸粘膜細菌叢は、非CD患者のそれと比較して、門レベルでProteobacteria及びBacteroidetes内により多くの分類群を含み、科レベルでEnterobacteriaceae、Ruminococcaceae及びBacteroidaceae内により多くの分類群を含んでいた(図1D及び1E)。対照的に、Firmicutes門及びStreptococcaceae科の分類群はCD患者において減少していた(図1D及び1E)。 The small intestinal mucosal flora of CD patients contains more taxa within Proteobacteria and Bacteroidetes at the portal level and more taxa within Enterobacteriae, Ruminococcaceae and Bacteroidetes at the family level compared to those of non-CD patients. Was (FIGS. 1D and 1E). In contrast, the Firmicutes and Streptococcaceae taxa were reduced in CD patients (FIGS. 1D and 1E).
 <E.coliとRuminococcus gnavusはCD SI粘膜に濃縮されていた>
 CD患者と非CD患者の間の異なる細菌分類群を同定するため、以下の二つの比較に基づく統計学的方法を用いた。効果サイズ測定と組み合わせた線形判別分析(LDA)(LEfSe)及び誤判定率を用いた多重t検定(FDR)法である。
<E. E. coli and Ruminococcus gnavus were concentrated in the CD SI mucosa>
To identify different bacterial taxa between CD and non-CD patients, a statistical method based on the following two comparisons was used. Linear discriminant analysis (LDA) (LEfSe) combined with effect size measurement and multiple t-test (FDR) method using false discovery rate.
 その結果、LefSeでCD患者のSI粘膜において有意に高い存在量を有する6つのOTUが明らかになった(図2A)。また、多重t検定でCDに関連する14のOTUを同定することができた(図2B)。さらに、CD関連SI粘膜[CD患者のSI粘膜中の平均存在量>1%かつ倍率変化(CD/non-CD)>2]において高存在量を示すOTUに対する配列データをマイニングした結果、10のOTUがこれらの基準を満たした(図2C、下記表2)。これら三つの計算分析を組合せることで、18のCD関連OTUを同定した(図2C、下記表2)。 As a result, 6 OTUs with significantly higher abundance in the SI mucosa of CD patients were revealed in LefSe (Fig. 2A). In addition, 14 OTUs associated with CD could be identified by multiple t-test (Fig. 2B). Furthermore, as a result of mining the sequence data for OTU showing a high abundance in the CD-related SI mucosa [average abundance in SI mucosa of CD patients> 1% and magnification change (CD / non-CD)> 2], 10 OTU met these criteria (Fig. 2C, Table 2 below). By combining these three computational analyzes, 18 CD-related OTUs were identified (Fig. 2C, Table 2 below).
Figure JPOXMLDOC01-appb-T000002
Figure JPOXMLDOC01-appb-T000002
 なお、表2において、CD患者で豊富に存在する18種の菌を示す。各グループについて、最も近縁の種又は株、National Center for Biotechnology Information(NCBI)のゲノムデータベースのパーセント類似度(%)、9-mixの菌の株ID、平均存在量(Abundance)、存在率(Prevalence)を示す。存在率は、SI粘膜検体中に0.1%を超える相対量の細菌が検出された被験者数を被験者総数で除して算出した。 Table 2 shows 18 types of bacteria that are abundant in CD patients. For each group, the most closely related species or strain, National Center for Biotechnology Information (NCBI) genome database percent similarity (%), 9-mix strain ID, average abundance (Avenance), abundance (Abundance) Prevalence) is shown. The abundance rate was calculated by dividing the number of subjects in which a relative amount of bacteria exceeding 0.1% was detected in the SI mucosal sample by the total number of subjects.
 E.coli、Ruminococcus gnavus、Bacteroides dorei、Klebsiella pneumoniae、Streptococcus pasteurianus、Parabacteroides merdae、Parabacteroides distasonis及びRobinsoniella peoriensisと同一性を共有するOTUは、少なくとも2つの解析においてCDと有意に関連し、その中でE.coli及びR.gnavusは、存在量に関して3つ全ての解析において、CD SI粘膜との有意な関連性が認められた(図2C)。また、E.coli及びR.gnavusは、挿入経路やサンプル採取法に関係なくCDサンプル中に濃縮されていた(図5C~5E及び5H~5J)。加えて、E.coli及びR.gnavusと同一性を共有するこれらのOTUは、対照と比較し、CD患者において概して存在することが認められた(70%以上の患者で、0.1%を超える存在量(abundance)で検出された)(表2、図6A)。さらに、腸狭窄を伴うCD患者及び抗TNF-α抗体療法で治療したCD患者のSI粘膜細菌叢は、他のCD患者と比較してEnterobacteriaceae内により多くの分類群を含んでいた(図1F、図6B)。 E. Colli, Ruminococcus gnavus, Bacteroides dorie, Klebsiella pneumoniae, Streptococcus pasteurianus, Parabacteroides merdee, Parabacteroides merdee, Parabacteroides merdae, Parabacteroides colli and R. gnavus was found to be significantly associated with CD SI mucosa in all three analyzes for abundance (Fig. 2C). In addition, E. colli and R. gnavus was concentrated in the CD sample regardless of the insertion route or sampling method (FIGS. 5C-5E and 5H-5J). In addition, E. colli and R. These OTUs, which share the same identity as gnavus, were found to be generally present in CD patients compared to controls (more than 70% of patients were detected with an abundance greater than 0.1%). (Table 2, FIG. 6A). In addition, the SI mucosal flora of CD patients with intestinal stenosis and CD patients treated with anti-TNF-α antibody therapy contained more classification groups within Enterobacteriaceae compared to other CD patients (Fig. 1F, FIG. FIG. 6B).
 また、DBE検査の1日前に採取した唾液及び糞便の細菌叢と、CD患者及び非CD患者のSI粘膜における細菌叢とを比較した結果、Bray-Curtis距離に基づくNMDSプロットにおいて、CD患者又は非CD患者由来のSI粘膜、唾液及び糞便の細菌叢が互いに異なることが示された(図6C)。以前の報告(文献10、11)と同じく、CD患者の糞便細菌叢は非CD患者と比較して、α多様性が減少し(図6D)、Faecalibacterium prausnitziiの存在量が減少した(図6E及び6F)。糞便細菌叢とは異なり、SI粘膜におけるα多様性(図1B)及びF.prausnitzii(図6F)の存在量は群間で同様であった。 In addition, as a result of comparing the bacterial flora of saliva and feces collected one day before the DBE test with the bacterial flora in the SI mucosa of CD patients and non-CD patients, in the NMDS plot based on the Bray-Curtis distance, CD patients or non-CD patients It was shown that the SI mucosa, saliva and fecal bacterial flora from CD patients differed from each other (Fig. 6C). Similar to previous reports (References 10 and 11), the fecal flora of CD patients had reduced α-diversity (Fig. 6D) and the abundance of Faecalibacterium prasnitzii compared to non-CD patients (Fig. 6E and). 6F). Unlike the fecal flora, α-diversity in SI mucosa (Fig. 1B) and F. The abundance of plausnitzii (Fig. 6F) was similar between the groups.
 重要なことに、E.coli、R.gnavus及びK.pneumoniaeとCDとの間の顕著に強い関連が、糞便細菌叢よりもSI粘膜における細菌叢で観察された(図2E、図7A~7C)。また、粘膜サンプルの採取前に吸引したSI液を調べた結果、E.coli及びR.gnavusに関連するOTUが、粘膜サンプルと比較して液中に少ないことが見出された(図2D)。 Importantly, E. colli, R. gnavus and K. et al. A significantly stronger association between pneumoniae and CD was observed in the bacterial flora in the SI mucosa rather than in the fecal bacterial flora (FIGS. 2E, 7A-7C). In addition, as a result of examining the SI solution sucked before collecting the mucosal sample, E.I. colli and R. It was found that the OTU associated with gnavus was lower in the fluid compared to the mucosal sample (Fig. 2D).
 これらの結果から、E.coli及びR.gnavusは、SI粘膜層に存在するか、または上皮細胞に付着していることが示唆される。E.coli及びR.gnavusは、糞便細菌叢をプロファイリングした以前の大規模研究において、CDと関連性が示唆されていたが(文献12、13)、本発明者らによる上記結果は、SI粘膜微生物叢の分析が糞便細菌叢のそれよりも強力であり、より小さなサンプルサイズで、CDとの関連における潜在的病原性細菌の検出に十分であることを示唆した。 From these results, E. colli and R. It is suggested that gnavus is present in the SI mucosal layer or attached to epithelial cells. E. colli and R. Although gnavus was suggested to be associated with CD in a previous large study profiling the stool bacterial flora (References 12 and 13), the above results by the present inventors were based on the analysis of the SI mucosal microbial flora of stool. It was more potent than that of the flora and suggested that a smaller sample size was sufficient for the detection of potentially pathogenic bacteria in the context of CD.
 <CD関連E.coli株による腸管Th1細胞の誘導>
 CD患者のSI粘膜サンプルを、さらにいくつかの培地を用い、好気的及び嫌気的培養に供し、80の細菌株を異なる16SrRNA遺伝子配列(98%を超える配列同一性)で単離した(図8A及び8B)。そして、前述の統計解析(多重t検定及び/又はLEfSe、図2C)によるCDとの有意な関連に基づき、80分離株から、E.coli(菌株ID:35A1)、R.gnavus(菌株ID:131A1)、B.dorei(菌株ID:131H4)、K.pneumoniae(菌株ID:39F4)、S.pasteurianus(菌株ID:133A7)、P.distasonis(菌株ID:134F1)、Bacteroides fragilis(菌株ID:32E9)、Erysipelatoclostridium ramosum(菌株ID:131A10)、及びBacteroides uniformis(菌株ID:131A11)(図2F、表2)と、>97%の16SrRNA遺伝子配列類似性を有する9株を選択し、以下の解析に供した。なお、E.coliとしてのE.coli 35A1株の同一性は、いくつかの診断用同定培地及びAPI20Eシステムを用いた生化学的検査によってさらに確認した(図9A及び9B)。
<CD-related E. Induction of intestinal Th1 cells by coli strain>
SI mucosal samples of CD patients were subjected to aerobic and anaerobic cultures using several additional media and 80 bacterial strains were isolated with different 16S rRNA gene sequences (> 98% sequence identity) (Figure). 8A and 8B). Then, based on the significant association with CD by the above-mentioned statistical analysis (multiple t-test and / or LEfSe, FIG. 2C), from the 80 isolates, E.I. colli (strain ID: 35A1), R. gnavus (strain ID: 131A1), B.I. dorei (strain ID: 131H4), K. et al. pneumoniae (strain ID: 39F4), S.A. pasteurianus (strain ID: 133A7), P. et al. Disstasonis (strain ID: 134F1), Bacteroides fragilis (strain ID: 32E9), Erysiperatoclastridium ramosum (strain ID: 131A10), and Bacteroides fragilis (strain ID: 131A10), and Bacteroides uniformis (strain ID: 131A11) Nine strains with sequence similarity were selected and subjected to the following analysis. In addition, E. E. as a colli The identity of the coli 35A1 strain was further confirmed by biochemical examination using several diagnostic identification media and the API20E system (FIGS. 9A and 9B).
 腸内細菌叢は、宿主免疫系の強力な調節因子として認識されている(文献14)。そこで、本発明者らは、選択した9株に関し、GFマウスを用い、in vivoで免疫細胞を刺激する能力について解析した。すなわち、9株各々を個別に培養し、それらを混合して細菌混合物(9-mix)を調製し、それをGF C57BL/6(B6)マウスに経口強制胃内投与した(図2G)。そして、マウスをノトバイオートビニルアイソレータで3週間飼育し、SI中のリンパ球と結腸固有層(LP)をフローサイトメトリーで調べた(図10)。 The gut microbiota is recognized as a potent regulator of the host immune system (Reference 14). Therefore, the present inventors analyzed the ability of GF mice to stimulate immune cells in vivo with respect to the selected 9 strains. That is, each of the 9 strains was individually cultured, and the mixture was mixed to prepare a bacterial mixture (9-mix), which was orally administered to GF C57BL / 6 (B6) mice by oral forced gastric administration (Fig. 2G). Then, the mice were bred in a notobioto vinyl isolator for 3 weeks, and the lymphocytes and colon lamina propria (LP) in SI were examined by flow cytometry (Fig. 10).
 その結果、SIリンパ球に対する作用は弱かったが、インターフェロンγ+(IFN-γ+)CD4T細胞[ヘルパーT1(Th1)細胞]の顕著な集積が9-mixコロニー形成マウスの結腸LPで観察され、そのレベルは特定病原体フリー(SPF)マウスのそれと類似していた(図3A及び3B)。また、Th1細胞より低いが、インターロイキン-17+(IL-17+)CD4T細胞[Tヘルパー17(Th17)細胞]の割合は9-mixの定着(コロナイゼーション)により顕著に増加した(図3B)。さらに、誘導されたTh1細胞及びTh17細胞は、CD病因に関与するTNFSF15の受容体であるデスレセプター3(DR3、文献1及び15)を発現していた(図3C及び3D)。 As a result, although the action on SI lymphocytes was weak, a remarkable accumulation of interferon γ + (IFN-γ +) CD4T cells [helper T1 (Th1) cells] was observed in colon LP of 9-mix colonized mice, and the level thereof. Was similar to that of specific pathogen-free (SPF) mice (FIGS. 3A and 3B). In addition, although lower than Th1 cells, the proportion of interleukin-17 + (IL-17 +) CD4T cells [T helper 17 (Th17) cells] was significantly increased by colonization of 9-mix (Fig. 3B). Furthermore, the induced Th1 cells and Th17 cells expressed death receptor 3 (DR3, Documents 1 and 15), which is a receptor for TNFSF15 involved in CD pathogenesis (FIGS. 3C and 3D).
 9-mixの中で、E.coli 35A1株とR.gnavus 131A1株の混合物(2-mix)又はそれらのいずれかを個別に調べた(図3E及び3F)。これらの2つの株はCD粘膜に最も豊富に存在し優勢であった(図2A~2F、表2)。 In 9-mix, E.I. colli 35A1 strain and R. Mixtures (2-mix) of the gnavus 131A1 strain or any of them were individually examined (FIGS. 3E and 3F). These two strains were most abundant and predominant in the CD mucosa (FIGS. 2A-2F, Table 2).
 2-mix又はE.coli 35A1株によるGF B6マウスにおけるコロニー形成は、腸におけるTh1細胞集積を9-mixで誘導したのと同じレベルで誘導したが、R.gnavus 131A1株によるTh1誘導効果はわずかであった(図3E及び3F)。E.coli 35A1株は主に結腸と盲腸にてコロニーを形成し(図11A)、小腸と比較して結腸におけるより大きなTh1細胞誘導を反映したものとなった。 2-mix or E.I. Colonization of GFB6 mice with the colli 35A1 strain induced Th1 cell accumulation in the intestine at the same level as that induced by 9-mix, but R. The Th1 inducing effect of the gnavus 131A1 strain was slight (FIGS. 3E and 3F). E. The coli 35A1 strain colonized mainly in the colon and cecum (Fig. 11A), reflecting greater Th1 cell induction in the colon compared to the small intestine.
 また、E.coli 35A1株及び8-mix(E.coli 35A1株を含まない9-mix)によるコロニー形成の効果を比較した。単一コロニーにしたE.coli 35A1株のみによるコロニー形成(monocolonization)は、8-mixによるコロニー形成と比較して有意に高いTh1細胞集積をもたらし(図11B)、E.coli 35A1株が9-mixにより誘導されたTh1細胞集積における主要な寄与因子であることが示唆された。 Also, E. The effects of colonization by coli 35A1 strain and 8-mix (9-mix not containing E. coli 35A1 strain) were compared. E. made into a single colony. Colonization with only the colli 35A1 strain resulted in significantly higher Th1 cell accumulation compared to colonization with 8-mix (FIG. 11B). It was suggested that the coli 35A1 strain is a major contributor to 9-mix-induced Th1 cell accumulation.
 Th1細胞に加え、Th17細胞に関しても、2-mixとE.coli 35A1株は、9-mixと同程度に当該Th細胞頻度の有意な増加を誘導した。しかし、SPFマウスにおけるそれには及ばない程度であった(図3Bと3Fとで比較)。 For Th17 cells in addition to Th1 cells, 2-mix and E.I. The coli 35A1 strain induced a significant increase in the Th cell frequency to the same extent as 9-mix. However, it was not as good as that in SPF mice (comparison between FIGS. 3B and 3F).
 次に、Th1細胞及びTh17細胞に対するE.coli 35A1株の影響が、菌株特異的であるか、種特異的であるかを調べた。 Next, E.I. for Th1 cells and Th17 cells. It was investigated whether the influence of the coli 35A1 strain was strain-specific or species-specific.
 本発明者らは、以前にCD患者から分離した接着侵襲性E.coli株であるE.coli LF82株又はE.coli K-12由来の非病原性実験室株であるE.coli MG1655株を定着させたマウスを作製している(文献16)。そこで、これらマウスの腸内におけるTh1細胞及びTh17細胞の誘導の程度を検出し、E.coli 35A1定着マウスのそれらと比較した。 We have previously isolated an adhesively invasive E. coli from a CD patient. E. coli strain. colli LF82 strain or E.I. E. et al., A non-pathogenic laboratory strain derived from colli K-12. A mouse in which the coli MG1655 strain has been established has been produced (Reference 16). Therefore, the degree of induction of Th1 cells and Th17 cells in the intestines of these mice was detected, and E.I. Compared with those of colli 35A1 colonized mice.
 その結果、E.coli LF82株によるわずかなTh1及びTh17細胞誘導効果が、SI及び結腸において検出されたものの、E.coli 35A1株のそれらとは対照的に、結腸LPにおけるTh1細胞誘導能は、E.coli LF82株及びE.coli MG1655株のいずれにおいても低かった(図4A~4C)。よって、これらの結果から、腸管Th1細胞は、E.coli 35A1株によって株特異的に誘導されることが示唆された。 As a result, E.I. Although a slight Th1 and Th17 cell-inducing effect by the colli LF82 strain was detected in SI and colon, E. coli In contrast to those of the coli 35A1 strain, Th1 cell inducibility in colon LP was E. coli. colli LF82 strain and E.I. It was low in all of the coli MG1655 strains (FIGS. 4A-4C). Therefore, from these results, the intestinal Th1 cells were found to be E. coli. It was suggested that the coli 35A1 strain was induced in a strain-specific manner.
 CD等の腸炎発症における、Th1細胞誘導性E.coli 35A1株の役割を調べるために、抗IL-10R抗体で誘発した大腸炎モデルを用いた(文献17)。具体的には、GF野生型B6マウスに、1日目にE.coli 35A1株、LF82株、又はMG1655株を各々定着させた。続いて抗IL-10R抗体を毎週腹腔内に注射し、それらマウスの腸を解析した。 Th1 cell-induced E.I. in the onset of enteritis such as CD. In order to investigate the role of the coli 35A1 strain, a colitis model induced by an anti-IL-10R antibody was used (Reference 17). Specifically, GF wild-type B6 mice were treated with E. coli on the first day. The coli 35A1 strain, the LF82 strain, or the MG1655 strain was established. Subsequently, anti-IL-10R antibody was injected intraperitoneally weekly and the intestines of those mice were analyzed.
 その結果、図4D~4F及び11C~11Fに示すとおり、E.coli 35A1株の定着は、盲腸と近位結腸に強い腸炎症を誘導し、結腸上皮細胞画分におけるTNF-αの高発現を誘導した。すなわち、CD SI粘膜に関連するE.coliの菌株特異的炎症誘発特性が、より一層裏づけられた。 As a result, as shown in FIGS. 4D-4F and 11C-11F, E.I. Colonization of the coli 35A1 strain induced strong enteritis in the cecum and proximal colon and induced high expression of TNF-α in the colonic epithelial cell fraction. That is, E.I. The strain-specific pro-inflammatory properties of colli were further supported.
 CDは、α多様性の減少及び潜在的炎症性細菌の増加により特徴付けられる腸内細菌叢ディスバイオ―シス(細菌の多様性の低下)と関連している(文献11)。しかし、CD関連細菌叢から分離され、宿主免疫系を活性化することが示されている細菌株はわずかである。 CD is associated with intestinal flora disbiosis (decreased bacterial diversity) characterized by decreased α-diversity and increased potential inflammatory bacteria (Reference 11). However, few bacterial strains have been shown to be isolated from the CD-related flora and activate the host immune system.
 そこで、本発明者らは、上述のとおり、CD患者のSI細菌叢を評価し、免疫刺激細菌をスクリーニングした。なお、当該スクリーニングに際して、本発明者らは、DBE法を用いてCD患者からSIサンプルを得たが、これは標準的な回腸結腸鏡検査とは異なり、遠位及び近位SIからの徹底的な検査及び細菌分離を可能にするものである。 Therefore, as described above, the present inventors evaluated the SI bacterial flora of CD patients and screened for immunostimulatory bacteria. In the screening, we obtained SI samples from CD patients using the DBE method, which is different from standard colonoscopy and is thorough from distal and proximal SI. It enables various tests and bacterial isolation.
 そして、その結果、80の細菌分離株からなる培養コレクションを作製し、分離株の一つであるE.coli 35A1株が腸におけるTh1細胞を強く誘導することを見出した。なお、SI粘膜から単離したにもかかわらず、E.coli 35A1株は主に結腸にコロニーを形成し、SIよりも結腸において、より顕著なTh1細胞誘導が認められた。ヒトとマウスの間の腸環境の違いを考慮すると、ヒトSI由来の分離株がマウスの異なる解剖学的部位で効果を発揮することは驚くべきことではない。 Then, as a result, a culture collection consisting of 80 bacterial isolates was prepared, and E. coli, which is one of the isolates, was prepared. It was found that the coli 35A1 strain strongly induces Th1 cells in the intestine. Although isolated from the SI mucosa, E. The coli 35A1 strain mainly colonized the colon, and more prominent Th1 cell induction was observed in the colon than in SI. Given the differences in the intestinal environment between humans and mice, it is not surprising that human SI-derived isolates exert their effects at different anatomical sites in mice.
 また、上述の結果から、CD SI粘膜関連E.coliが実質的な炎症誘発特性を有する可能性が示唆される。ここで、E.coli LF82株はCD患者から以前に単離された臨床的に関連する株である。E.coli LF82株は腸上皮細胞(EC)に接着して侵入し、マクロファージ内で複製することができることから、腸管接着性侵入性大腸菌(AIEC)と呼ばれている(文献16、18、19)。さらに、ムチン分解(Vat-AIECプロテアーゼ)、1型線毛を介したCEACAM6への接着(ECの頂端表面に発現する)及び鞭毛を介したToll様受容体(TLR)5の活性化を含む、AIECのいくつかの毒性機構が報告されている(文献20~24)。しかし、E.coli 35A1株は、CD患者のSI粘膜に関連した炎症性E.coliの別の例を示す。また興味深いことに、E.coli 35A1株はTh1細胞の誘導においてE.coli LF82株よりも強力であった。Bifidobacterium longumとK.pneumoniae株も腸管Th1細胞誘導と関連している(文献25、26)ことを考慮すると、E.coli 35A1株は、B.longumとK.pneumoniaeが持つ因子と類似した因子を持つ可能性がある。 Also, from the above results, CD SI mucosa-related E.I. It is suggested that colli may have substantial pro-inflammatory properties. Here, E.I. The coli LF82 strain is a clinically relevant strain previously isolated from CD patients. E. Escherichia coli LF82 strain is called intestinal adhesive invasive Escherichia coli (AIEC) because it adheres to and invades intestinal epithelial cells (EC) and can replicate in macrophages (References 16, 18, and 19). In addition, it involves mucin degradation (Vat-AIEC protease), adhesion to CEACAM6 via type 1 pili (expressed on the apical surface of EC) and activation of Toll-like receptors (TLRs) 5 via flagella. Several toxicity mechanisms of AIEC have been reported (References 20-24). However, E. The coli 35A1 strain is an inflammatory E. coli associated with the SI mucosa of CD patients. Another example of colli is shown. Interestingly, E.I. The coli 35A1 strain was used in the induction of Th1 cells by E. coli. It was stronger than the coli LF82 strain. Bifidobacterium longum and K. Considering that the pneumoniae strain is also associated with intestinal Th1 cell induction (References 25 and 26), E. coli. The colli 35A1 strain is B.I. longum and K. It may have factors similar to those of pneumoniae.
 SI粘膜細菌叢の分析において、R.gnavusもCDと関連することが見出された。R.gnavusはCDにおいて増加していることが報告されており、それによりその病因に関与している(文献27)。R.gnavusは少なくとも2つの系統群を含み、そのうちの1つはCD患者に多く、TLR4を介して樹状細胞によるTNF-αの産生を誘導する炎症を惹起する多糖類を産生する(文献28、29)。R.gnavus 131A1株の免疫刺激効果は弱かったが、R.gnavusはCD病因と関係した他の細菌の定着を促進する可能性がある。実際、R.gnavusはムチン分解細菌であり、ムチン層を分解し、それによって上皮細胞層に近接する他の細菌の定着を促進する。さらに、R.gnavusはシアル酸を産生するが(文献30)、これはE.coliを含む他のシアル酸利用病原体の増殖と定着を促進する可能性がある(文献31)。ここで、E.coli又はR.gnavus単独と比較して、2-mix定着によるTh17細胞の誘導は顕著に増強されたことは注目に値する(図3F)。 In the analysis of SI mucosal bacterial flora, R. It was also found that gnavus is also associated with CD. R. gnavus has been reported to be increased in CD and thereby contributes to its pathogenesis (Reference 27). R. gnavus comprises at least two lineage groups, one of which is predominant in CD patients and produces inflammation-inducing polysaccharides that induce the production of TNF-α by dendritic cells via TLR4 (References 28, 29). ). R. The immunostimulatory effect of the gnavus 131A1 strain was weak, but R. gnavus may promote colonization of other bacteria associated with CD pathogenesis. In fact, R. gnavus is a mucin-degrading bacterium that degrades the mucin layer, thereby promoting the colonization of other bacteria in the vicinity of the epithelial cell layer. In addition, R. gnavus produces sialic acid (Reference 30), which is described in E. coli. It may promote the growth and colonization of other sialic acid-utilizing pathogens, including coli (Reference 31). Here, E.I. colli or R. It is noteworthy that the induction of Th17 cells by 2-mix colonization was significantly enhanced compared to gnavus alone (Fig. 3F).
 <本[実施例]にて引用する文献>
1. Yamazaki K.ら、Hum Mol Genet.、2005;14(22):3499~3506
2. Franke A.ら、Nat Genet.、2010;42(12):1118~1125
3. Liu JZ.ら、Nat Genet.、2015;47(9):979~986
4. Kostic AD.ら、Gastroenterology.、2014;146(6):1489~1499
5. Pascale A.ら、Endocrine.、2018;61(3):357~371
6. Sokol H.ら、Proc Natl Acad Sci USA.、2008;105(43):16731~16736
7. Yamamoto H.ら、J Gastroenterol.、2003;17(4):273~274
8. Yamamoto H.ら、Dige Endosc.、2015;27(3):331~337
9. Sunada K.ら、Inflamm Bowel Dis.、2016;22(2):380~386
10. Tamboli CP.ら、Gut.、2004;53(1):1~4
11. Nishida A.ら、Clin J Gastroenterol.、2018;11(1):1~10
12. Willing BP.ら、Gastroenterology.、2010;139(6):1844~54.e1.
13. Joossens M.ら、Gut.、2011;60(5):631~637
14. Sartor RB.ら、Gastroenterology.、2017;152(2):327~39.e4.
15. Siakavellas SI.ら、Inflamm Bowel Dis.、2015;21(10):2441~2452
16. Darfeuille-Michaud.ら、Gastroenterology.1998;115(6):1405~1413
17. Schiering C.ら、Nature.、2014;513(7519):564~568
18. Boudeau J.ら、Infect Immun.、1999;67(9):4499~4509
19. Darfeuille-Michaud A.ら、Gastroenterology.、2004;127(2):412~421
20. Barnich N.ら、J Clin Invest.、2007;117(6):1566~1574
21. Carvalho FA.ら、Inflamm Bowel Dis.、2008;14(8):1051~1060
22. Carvalho FA.ら、J Exp Med.、2009;206(10):2179~2189
23. Dreux N.ら、PLoS Pathog.、2013;9(1):e1003141
24. Gibold L.ら、Cell Microbiol.、2016;18(5):617~631
25. Geva-Zatorsky N.ら、Cell.、2017;168(5):928~943.e11.
26. Atarashi K.ら、Science.、2017;358(6361):359~365
27. Lloyd-Price J.ら、Nature.2019;569(7758):655~662
28. Hall AB.ら、Genome Med.、2017;9(1):103
29. Henke MT.ら、Proc Natl Acad Sci USA.、2019;116(26):12672~12677
30. Bell A.ら、Nat Microbiol.、2019;4(12):2393~2404
31. KM N.ら、Nature.、2013;502(7469):96~99
32. Segata Nら、Genome Biol.、2011;12(6):R60
<References cited in this [Example]>
1. 1. Yamazaki K.K. Et al., Hum Mol Genet. , 2005; 14 (22): 3499-3506
2. Franke A. Et al., Nat Genet. , 2010; 42 (12): 1118 to 1125
3. 3. Liu JZ. Et al., Nat Genet. , 2015; 47 (9): 979-986
4. Kostic AD. Et al., Gastroenterology. , 2014; 146 (6): 1489-1499
5. Pascale A. Et al., Endocrine. , 2018; 61 (3): 357-371
6. Sokol H. Et al., Proc Natl Acad Sci USA. 2008; 105 (43): 16731-16736
7. Yamamoto H. Et al., J Gastroenterol. , 2003; 17 (4): 273-274
8. Yamamoto H. Et al., Dige Endosc. , 2015; 27 (3): 331-337
9. Sunada K. Et al., Inflamm Bowel Dis. , 2016; 22 (2): 380-386
10. Tamboli CP. Et al., Gut. , 2004; 53 (1): 1-4
11. Nishida A. Et al., Clin J Gastroenterol. , 2018; 11 (1): 1-10
12. Willing BP. Et al., Gastroenterology. , 2010; 139 (6): 1844-54. e1.
13. Joossens M. Et al., Gut. , 2011; 60 (5): 631-637
14. Sartor RB. Et al., Gastroenterology. , 2017; 152 (2): 327-39. e4.
15. Siakavelas SI. Et al., Inflamm Bowel Dis. , 2015; 21 (10): 2441-2452
16. Darfeile-Michaud. Et al., Gastroenterology. 1998; 115 (6): 1405-1413
17. Schiering C. Et al., Nature. , 2014; 513 (7519): 564-568
18. Boudeau J.M. Et al., Infect Immun. , 1999; 67 (9): 4499-4509
19. Darfeile-Michaud A. Et al., Gastroenterology. , 2004; 127 (2): 412-421
20. Barnich N. Et al., J Clin Invest. , 2007; 117 (6): 1566 to 1574
21. Carvalho FA. Et al., Inflamm Bowel Dis. 2008; 14 (8): 1051 to 1060
22. Carvalho FA. Et al., J Exp Med. , 2009; 206 (10): 2179-2189
23. Dreux N. Et al., PLos Pathog. , 2013; 9 (1): e1003411
24. Gibold L. Et al., Cell Microbiol. , 2016; 18 (5): 617-631
25. Geva-Zatorsky N.M. Et al., Cell. , 2017; 168 (5): 928-943. e11.
26. Atarashi K. Et al., Science. , 2017; 358 (6361): 359-365
27. Lloyd-Price J. Et al., Nature. 2019; 569 (7758): 655-662
28. Hall AB. Et al., Genome Med. , 2017; 9 (1): 103
29. Henke MT. Et al., Proc Natl Acad Sci USA. , 2019; 116 (26): 12672-12677
30. Bell A. Et al., Nat Microbiol. , 2019; 4 (12): 2393-2404
31. KM N. Et al., Nature. , 2013; 502 (7469): 96-99
32. Segata N et al., Genome Biology. , 2011; 12 (6): R60
 以上説明したように、本発明によれば、Th1細胞及び/又はTh17細胞の増殖又は活性化を誘導する小腸内細菌を標的とすることによって、前記細胞の増殖又は活性化の抑制等が可能となり、ひいては、前記細胞に起因する疾患を治療、改善又は予防することが可能となる。また、本発明によれば、前記小腸内細菌量を指標として、前記細胞に起因する疾患を検査することも可能となる。 As described above, according to the present invention, by targeting a small intestinal bacterium that induces proliferation or activation of Th1 cells and / or Th17 cells, it becomes possible to suppress the proliferation or activation of the cells. As a result, it becomes possible to treat, improve or prevent the disease caused by the cells. Further, according to the present invention, it is also possible to test for diseases caused by the cells by using the amount of bacteria in the small intestine as an index.
 したがって、本発明は、Th1細胞等に起因するクローン病等の疾患に関する、医薬品の開発、治療、改善、予防及び診断において、極めて有用である。 Therefore, the present invention is extremely useful in the development, treatment, improvement, prevention and diagnosis of pharmaceuticals relating to diseases such as Crohn's disease caused by Th1 cells and the like.

Claims (24)

  1.  Th1細胞及び/又はTh17細胞の増殖又は活性化を誘導する小腸内細菌。 Small intestinal bacteria that induce proliferation or activation of Th1 cells and / or Th17 cells.
  2.  下記細菌群から選択される少なくとも1の細菌である、請求項1に記載の細菌
    細菌群:
    配列番号:1に記載のDNA配列に対して95%以上の同一性を有するポリヌクレオチドを有する細菌、
    配列番号:2に記載のDNA配列に対して95%以上の同一性を有するポリヌクレオチドを有する細菌、
    配列番号:3に記載のDNA配列に対して95%以上の同一性を有するポリヌクレオチドを有する細菌、
    配列番号:4に記載のDNA配列に対して95%以上の同一性を有するポリヌクレオチドを有する細菌、
    配列番号:5に記載のDNA配列に対して95%以上の同一性を有するポリヌクレオチドを有する細菌、
    配列番号:6に記載のDNA配列に対して95%以上の同一性を有するポリヌクレオチドを有する細菌、
    配列番号:7に記載のDNA配列に対して95%以上の同一性を有するポリヌクレオチドを有する細菌、
    配列番号:8に記載のDNA配列に対して95%以上の同一性を有するポリヌクレオチドを有する細菌、及び
    配列番号:9に記載のDNA配列に対して95%以上の同一性を有するポリヌクレオチドを有する細菌。
    The bacterium group according to claim 1, which is at least one bacterium selected from the following bacterium group:
    Bacteria having a polynucleotide having 95% or more identity to the DNA sequence set forth in SEQ ID NO: 1.
    Bacteria having a polynucleotide having 95% or more identity to the DNA sequence set forth in SEQ ID NO: 2.
    Bacteria having a polynucleotide having 95% or more identity to the DNA sequence set forth in SEQ ID NO: 3.
    Bacteria having a polynucleotide having 95% or more identity to the DNA sequence set forth in SEQ ID NO: 4.
    Bacteria having a polynucleotide having 95% or more identity to the DNA sequence set forth in SEQ ID NO: 5.
    Bacteria having a polynucleotide having 95% or more identity to the DNA sequence set forth in SEQ ID NO: 6.
    Bacteria having a polynucleotide having 95% or more identity to the DNA sequence set forth in SEQ ID NO: 7.
    Bacteria having a polynucleotide having 95% or more identity to the DNA sequence set forth in SEQ ID NO: 8 and a polynucleotide having 95% or more identity to the DNA sequence set forth in SEQ ID NO: 9. Bacteria to have.
  3.  配列番号:1に記載のDNA配列に対して95%以上の同一性を有するポリヌクレオチドを有する細菌と、配列番号:2に記載のDNA配列に対して95%以上の同一性を有するポリヌクレオチドを有する細菌との組み合わせである、請求項1に記載の細菌。 Bacteria having a polynucleotide having 95% or more identity to the DNA sequence set forth in SEQ ID NO: 1 and a polynucleotide having 95% or more identity to the DNA sequence set forth in SEQ ID NO: 2 The bacterium according to claim 1, which is a combination with the bacterium having.
  4.  小腸内でTh1細胞及び/又はTh17細胞の増殖又は活性化を抑制する物質をスクリーニングする方法であって、
    (1)被験物質を非ヒト無菌動物に摂取させる工程、
    (2)該非ヒト動物の小腸内におけるTh1細胞及び/又はTh17細胞の数又は活性を検出する工程、及び
    (3)工程(2)にて前記細胞の増殖又は活性化の抑制が検出された場合、前記被験物質は、小腸内でTh1細胞及び/又はTh17細胞の増殖又は活性化を抑制する物質であると判定する工程を、
    含む方法。
    A method of screening for substances that suppress the proliferation or activation of Th1 cells and / or Th17 cells in the small intestine.
    (1) A step of ingesting a test substance into a non-human germ-free animal,
    (2) When the number or activity of Th1 cells and / or Th17 cells in the small intestine of the non-human animal is detected, and when suppression of proliferation or activation of the cells is detected in (3) step (2). The step of determining that the test substance is a substance that suppresses the proliferation or activation of Th1 cells and / or Th17 cells in the small intestine.
    How to include.
  5.  小腸内でTh1細胞及び/又はTh17細胞の増殖又は活性化を抑制する物質をスクリーニングする方法であって、
    (1)被験物質を、配列番号:1に記載のDNA配列に対して95%以上の同一性を有するポリヌクレオチドを有する細菌及び/又は配列番号:2に記載のDNA配列に対して95%以上の同一性を有するポリヌクレオチドを有する細菌を定着させた非ヒト動物に、摂取させる工程、
    (2)該非ヒト動物の小腸内における前記細菌の数を検出する工程、及び
    (3)工程(2)にて前記細菌の増殖抑制が検出された場合、前記被験物質は、小腸内でTh1細胞及び/又はTh17細胞の増殖又は活性化を抑制する物質であると判定する工程を、
    含む方法。
    A method of screening for substances that suppress the proliferation or activation of Th1 cells and / or Th17 cells in the small intestine.
    (1) The test substance is a bacterium having a polynucleotide having a polynucleotide having 95% or more identity with respect to the DNA sequence set forth in SEQ ID NO: 1 and / or 95% or more with respect to the DNA sequence set forth in SEQ ID NO: 2. Ingestion of non-human animals colonized with a polynucleotide having a polynucleotide having the same identity as
    When (2) the step of detecting the number of the bacteria in the small intestine of the non-human animal and (3) the suppression of the growth of the bacteria are detected in the step (2), the test substance is Th1 cells in the small intestine. And / or the step of determining that the substance suppresses the proliferation or activation of Th17 cells.
    How to include.
  6.  前記物質が、細菌又はバクテリオファージである、請求項4又は5に記載の方法。 The method according to claim 4 or 5, wherein the substance is a bacterium or a bacteriophage.
  7.  Th1細胞及び/又はTh17細胞に起因する疾患を評価する方法であって、
    (1)被検者の小腸粘膜における、配列番号:1に記載のDNA配列に対して95%以上の同一性を有するポリヌクレオチドを有する細菌及び/又は配列番号:2に記載のDNA配列に対して95%以上の同一性を有するポリヌクレオチドを有する細菌を定量する工程、
    (2)工程(1)で定量して得られた値を、前記疾患に罹患していないヒトの小腸粘膜において前記細菌を定量して得られる対応値と比較する工程、及び
    (3)工程(2)における比較の結果、被検者の小腸粘膜中における前記定量値が、前記対応値よりも高い場合に、前記被検者は前記疾患に罹患していると判定する工程を、
    含む方法。
    A method for assessing diseases caused by Th1 cells and / or Th17 cells.
    (1) For bacteria having a polynucleotide having 95% or more identity with respect to the DNA sequence set forth in SEQ ID NO: 1 and / or the DNA sequence set forth in SEQ ID NO: 2 in the small intestinal mucosa of the subject. Quantifying bacteria with polynucleotides having 95% or more identity,
    (2) A step of comparing the value obtained by quantifying in step (1) with a corresponding value obtained by quantifying the bacteria in the mucosa of the small intestine of a human who does not suffer from the disease, and step (3) (3). As a result of the comparison in 2), when the quantitative value in the small intestinal mucosa of the subject is higher than the corresponding value, the step of determining that the subject is suffering from the disease is performed.
    How to include.
  8.  請求項1~3のうちのいずれか一項に記載の細菌又は該細菌に由来する生理活性物質を有効成分として含む、Th1細胞及び/又はTh17細胞の増殖又は活性化を誘導するための組成物。 A composition for inducing proliferation or activation of Th1 cells and / or Th17 cells, which comprises the bacterium according to any one of claims 1 to 3 or a physiologically active substance derived from the bacterium as an active ingredient. ..
  9.  請求項1~3のうちのいずれか一項に記載の細菌又は該細菌に特異的な抗原を有効成分として含む、ワクチン組成物。 A vaccine composition containing the bacterium according to any one of claims 1 to 3 or an antigen specific to the bacterium as an active ingredient.
  10.  請求項1~3のうちのいずれか一項に記載の細菌に対して抗菌作用を有する物質を有効成分として含む、Th1細胞及び/又はTh17細胞の増殖又は活性化を抑制するための組成物。 A composition for suppressing the proliferation or activation of Th1 cells and / or Th17 cells, which comprises a substance having an antibacterial action against the bacterium according to any one of claims 1 to 3 as an active ingredient.
  11.  前記物質が、請求項4~6のうちのいずれか一項に記載の方法によって選抜される物質である、請求項9に記載の組成物。 The composition according to claim 9, wherein the substance is a substance selected by the method according to any one of claims 4 to 6.
  12.  Th1細胞及び/又はTh17細胞に起因する疾患を治療、改善又は予防するための組成物である、請求項9~11のうちのいずれか一項に記載の組成物。 The composition according to any one of claims 9 to 11, which is a composition for treating, ameliorating or preventing a disease caused by Th1 cells and / or Th17 cells.
  13.  請求項7に記載の方法によって前記疾患に罹患していると判定された前記被検者に摂取させることを特徴とする、請求項12に記載の組成物。 The composition according to claim 12, wherein the subject determined to be suffering from the disease is ingested by the method according to claim 7.
  14.  前記疾患がクローン病である、請求項12又は13に記載の組成物。 The composition according to claim 12 or 13, wherein the disease is Crohn's disease.
  15.  前記クローン病が小腸型クローン病である、請求項14に記載の組成物。 The composition according to claim 14, wherein the Crohn's disease is a small intestinal Crohn's disease.
  16.  請求項1~3のうちのいずれか一項に記載の細菌又は該細菌に特異的な抗原を、対象に摂取させ、該対象におけるTh1細胞及び/又はTh17細胞の増殖又は活性化を抑制する方法。 A method for ingesting a bacterium according to any one of claims 1 to 3 or an antigen specific to the bacterium to suppress the proliferation or activation of Th1 cells and / or Th17 cells in the subject. ..
  17.  請求項1~3のうちのいずれか一項に記載の細菌に対して抗菌作用を有する物質を、対象に摂取させ、該対象におけるTh1細胞及び/又はTh17細胞の増殖又は活性化を抑制する方法。 A method for ingesting a substance having an antibacterial action against the bacterium according to any one of claims 1 to 3 and suppressing the proliferation or activation of Th1 cells and / or Th17 cells in the subject. ..
  18.  前記物質が、請求項4~6のうちのいずれか一項に記載の方法によって選抜される物質である、請求項17に記載の方法。 The method according to claim 17, wherein the substance is a substance selected by the method according to any one of claims 4 to 6.
  19.  請求項1~3のうちのいずれか一項に記載の細菌又は該細菌に特異的な抗原を、対象に摂取させ、該対象におけるTh1細胞及び/又はTh17細胞に起因する疾患を治療、改善又は予防する方法。 The bacterium according to any one of claims 1 to 3 or an antigen specific to the bacterium is ingested by a subject to treat, ameliorate, or treat a disease caused by Th1 cells and / or Th17 cells in the subject. How to prevent.
  20.  請求項1~3のうちのいずれか一項に記載の細菌に対して抗菌作用を有する物質を、対象に摂取させ、該対象におけるTh1細胞及び/又はTh17細胞に起因する疾患を治療、改善又は予防する方法。 A substance having an antibacterial action against the bacterium according to any one of claims 1 to 3 is ingested by a subject to treat, improve or treat a disease caused by Th1 cells and / or Th17 cells in the subject. How to prevent.
  21.  前記物質が、請求項4~6のうちのいずれか一項に記載の方法によって選抜される物質である、請求項20に記載の方法。 The method according to claim 20, wherein the substance is a substance selected by the method according to any one of claims 4 to 6.
  22.  前記対象が、請求項7に記載の方法によって前記疾患に罹患していると判定された前記被検者である、請求項19~21のうちのいずれか一項に記載の方法。 The method according to any one of claims 19 to 21, wherein the subject is the subject determined to be suffering from the disease by the method according to claim 7.
  23.  前記疾患がクローン病である、請求項19~22のうちのいずれか一項に記載の方法。 The method according to any one of claims 19 to 22, wherein the disease is Crohn's disease.
  24.  前記クローン病が小腸型クローン病である、請求項23に記載の方法。 The method according to claim 23, wherein the Crohn's disease is a small intestinal Crohn's disease.
PCT/JP2021/017000 2020-04-29 2021-04-28 Small intestinal bacterium inducing proliferation or activation of th1 cells and/or th17 cells WO2021221110A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US202063017268P 2020-04-29 2020-04-29
US63/017,268 2020-04-29
US202063046871P 2020-07-01 2020-07-01
US63/046,871 2020-07-01

Publications (1)

Publication Number Publication Date
WO2021221110A1 true WO2021221110A1 (en) 2021-11-04

Family

ID=78374088

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2021/017000 WO2021221110A1 (en) 2020-04-29 2021-04-28 Small intestinal bacterium inducing proliferation or activation of th1 cells and/or th17 cells

Country Status (1)

Country Link
WO (1) WO2021221110A1 (en)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2016509002A (en) * 2013-02-04 2016-03-24 セレス セラピューティクス インコーポレイテッド Compositions and methods for inhibition of pathogenic bacterial growth
JP2018502551A (en) * 2015-06-15 2018-02-01 フォーディー ファーマ リサーチ リミテッド4D Pharma Research Limited Composition comprising a bacterial strain
WO2019017389A1 (en) * 2017-07-18 2019-01-24 学校法人慶應義塾 Anti-bacterial composition against th1 cell-inducing bacteria
JP2019508479A (en) * 2016-03-14 2019-03-28 ホロバイオーム, インコーポレイテッド Modification of the intestinal microbiome to treat mental disorders or diseases of the central nervous system
JP2020503862A (en) * 2016-12-23 2020-02-06 学校法人慶應義塾 Compositions and methods for inducing CD8 + T cells

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2016509002A (en) * 2013-02-04 2016-03-24 セレス セラピューティクス インコーポレイテッド Compositions and methods for inhibition of pathogenic bacterial growth
JP2018502551A (en) * 2015-06-15 2018-02-01 フォーディー ファーマ リサーチ リミテッド4D Pharma Research Limited Composition comprising a bacterial strain
JP2019508479A (en) * 2016-03-14 2019-03-28 ホロバイオーム, インコーポレイテッド Modification of the intestinal microbiome to treat mental disorders or diseases of the central nervous system
JP2020503862A (en) * 2016-12-23 2020-02-06 学校法人慶應義塾 Compositions and methods for inducing CD8 + T cells
WO2019017389A1 (en) * 2017-07-18 2019-01-24 学校法人慶應義塾 Anti-bacterial composition against th1 cell-inducing bacteria

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
DATABASE Nucleotide ANONYMOUS: "Klebsiella pneumoniae strain M344-17 16S ribosomal RNA gene, partial sequence", XP055869910, Database accession no. MH680846.1 *
HENN, M. R . ET AL.: "Sequence 611 from patent US 9028841", GENBANK, 10 February 2016 (2016-02-10), Retrieved from the Internet <URL:https://www.ncbi.nlm.nih.gov/nuccore/hk693866> [retrieved on 20210701] *
LI, E. ET AL.: "Uncultured organism clone ELU0165- T391-S-NIPCRAMgANa 000329 small subunit ribosomal RNA gene, partial sequence", GENBANK, 30 July 2012 (2012-07-30), Retrieved from the Internet <URL:https://www.ncbi.nlm.nih.gov/nuccore/hq810989> [retrieved on 20210701] *

Similar Documents

Publication Publication Date Title
Jones et al. Osteomicrobiology: The influence of gut microbiota on bone in health and disease
Gaboriau-Routhiau et al. The key role of segmented filamentous bacteria in the coordinated maturation of gut helper T cell responses
US20160120915A1 (en) Methods for manipulating immune responses by altering microbiota
JP6201982B2 (en) Diabetes-inducing bacteria
JP7325106B2 (en) Lipopolysaccharide-regulating intestinal bacteria and uses thereof
JP7109794B2 (en) Antibacterial composition against Th1 cell-inducing bacteria
Lähteinen et al. Effect of Lactobacillus brevis ATCC 8287 as a feeding supplement on the performance and immune function of piglets
JP2022133363A (en) Bacteria that induce Th1 cells
JP7399400B2 (en) A composition for suppressing trypsin activity containing bacteria belonging to the genus Paraprevotella as an active ingredient
Wang et al. Microbiota and gut health: promising prospects for clinical trials from bench to bedside
WO2021221110A1 (en) Small intestinal bacterium inducing proliferation or activation of th1 cells and/or th17 cells
EP4076482A1 (en) Compositions for modulating gut microflora populations, enhancing drug potency and treating cancer, and methods for making and using same
Macfarlane et al. Manipulating the Indigenous Microbiota in Humans: Prebiotics, Probiotics, and Synbiotics
WO2020179868A1 (en) Antibacterial composition against drug-resistant bacteria or pro-inflammatory bacteria
US9885089B2 (en) Flagellum and anti-flagellum antibody levels in samples as a diagnostic biomarker and therapeutic target for metabolic syndrome
DK2580343T3 (en) APPLICATION OF ABO BLOOD TYPE IN A PROCEDURE FOR SPECIAL PREPARATION OF A BACTERIA COMPOSITION
JP2024518084A (en) Compositions and methods for treating disease
Al-Baroody Investigation Effect of Escherichia coli Producing Microcin B17 Isolated from Patients with Inflammatory Bowel Disease in Female Rats
Linden Time-Dependent and Microbiota-Dependent Effects of Interleukin-22 in a Colitis Mouse Model
JP2023508138A (en) Compositions and methods for treating inflammatory bowel disease
Du Role of bacteria and mucosal immune system and their interaction in the pathogenesis of inflammatory bowel disease
Class et al. Patent application title: METHODS FOR MANIPULATING IMMUNE RESPONSES BY ALTERING MICROBIOTA Inventors: Martin J. Blaser (New York, NY, US) Martin J. Blaser (New York, NY, US) Shingo Yamanishi (Bunkyo-Ku, JP) Laura M. Cox (Brooklyn, NY, US) Victoria E. Ruiz (Brooklyn, NY, US) Alexandra E. Livanos (New York, NY, US) Assignees: New York University
CN116615561A (en) Compositions comprising microorganisms for diagnosing or treating inflammatory diseases
Brawner Immunobiology of the Mucosal Response to Helicobacter pylori in Children
Mahendran Investigation of the possible role of Campylobacter concisus in inflammatory bowel disease

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 21796659

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 21796659

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: JP