Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Cell ; 163(2): 367-80, 2015 Oct 08.
Article in English | MEDLINE | ID: mdl-26411289

ABSTRACT

Intestinal Th17 cells are induced and accumulate in response to colonization with a subgroup of intestinal microbes such as segmented filamentous bacteria (SFB) and certain extracellular pathogens. Here, we show that adhesion of microbes to intestinal epithelial cells (ECs) is a critical cue for Th17 induction. Upon monocolonization of germ-free mice or rats with SFB indigenous to mice (M-SFB) or rats (R-SFB), M-SFB and R-SFB showed host-specific adhesion to small intestinal ECs, accompanied by host-specific induction of Th17 cells. Citrobacter rodentium and Escherichia coli O157 triggered similar Th17 responses, whereas adhesion-defective mutants of these microbes failed to do so. Moreover, a mixture of 20 bacterial strains, which were selected and isolated from fecal samples of a patient with ulcerative colitis on the basis of their ability to cause a robust induction of Th17 cells in the mouse colon, also exhibited EC-adhesive characteristics.


Subject(s)
Bacterial Adhesion , Citrobacter rodentium/physiology , Enterobacteriaceae Infections/immunology , Escherichia coli Infections/immunology , Escherichia coli O157/physiology , Intestinal Mucosa/immunology , Th17 Cells/immunology , Animals , Bacterial Infections/immunology , Epithelial Cells/immunology , Epithelial Cells/microbiology , Epithelial Cells/ultrastructure , Feces/microbiology , Humans , Immunoglobulin A/immunology , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Microscopy, Electron, Scanning , Rats , Rats, Inbred F344 , Species Specificity
2.
Immunology ; 146(1): 59-69, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25967857

ABSTRACT

To clarify the effect of secretory IgA (sIgA) deficiency on gut homeostasis, we examined intraepithelial lymphocytes (IELs) in the small intestine (SI) of polymeric immunoglobulin receptor-deficient (pIgR(-/-) ) mice. The pIgR(-/-) mice exhibited the accumulation of CD8αß(+) T-cell receptor (TCR)-αß(+) IELs (CD8αß(+) αß-IELs) after weaning, but no increase of CD8αß(+) γδ-IELs was detected in pIgR(-/-) TCR-ß(-/-) mice compared with pIgR(+/+) TCR-ß(-/-) mice. When 5-bromo-2'-deoxyuridine (BrdU) was given for 14 days, the proportion of BrdU-labelled cells in SI-IELs was not different between pIgR(+/+) mice and pIgR(-/-) mice. However, the proportion of BrdU-labelled CD8αß(+) -IELs became higher in pIgR(-/-) mice than pIgR(+/+) mice 10 days after discontinuing BrdU-labelling. Intravenously transferred splenic T cells migrated into the intraepithelial compartments of pIgR(+/+) TCR-ß(-/-) mice and pIgR(-/-) TCR-ß(-/-) mice to a similar extent. In contrast, in the case of injection of immature bone marrow cells, CD8αß(+) αß-IELs increased much more in the SI of pIgR(-/-) TCR-ß(-/-) mice than pIgR(+/+) TCR-ß(-/-) mice 8 weeks after the transfer. αß-IELs from pIgR(-/-) mice could produce more interferon-γ and interleukin-17 than those of pIgR(+/+) mice, and intestinal permeability tended to increase in the SI of pIgR(-/-) mice with aging. Taken together, these results indicate that activated CD8αß(+) αß-IELs preferentially accumulate in pIgR(-/-) mice through the enhanced differentiation of immature haematopoietic precursor cells, which may subsequently result in the disruption of epithelial integrity.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunoglobulin A, Secretory/genetics , Intestine, Small/immunology , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Polymeric Immunoglobulin/genetics , Adoptive Transfer , Animals , Bone Marrow Cells/immunology , Bromodeoxyuridine , CD8-Positive T-Lymphocytes/transplantation , Cell Differentiation/immunology , Cell Movement/immunology , Cells, Cultured , Immunoglobulin A, Secretory/immunology , Interferon-gamma/biosynthesis , Interleukin-17/biosynthesis , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Intestine, Small/cytology , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Staining and Labeling
3.
Proc Natl Acad Sci U S A ; 107(16): 7419-24, 2010 Apr 20.
Article in English | MEDLINE | ID: mdl-20360558

ABSTRACT

The indigenous bacteria create natural cohabitation niches together with mucosal Abs in the gastrointestinal (GI) tract. Here we report that opportunistic bacteria, largely Alcaligenes species, specifically inhabit host Peyer's patches (PPs) and isolated lymphoid follicles, with the associated preferential induction of antigen-specific mucosal IgA Abs in the GI tract. Alcaligenes were identified as the dominant bacteria on the interior of PPs from naïve, specific-pathogen-free but not from germ-free mice. Oral transfer of intratissue uncultured Alcaligenes into germ-free mice resulted in the presence of Alcaligenes inside the PPs of recipients. This result was further supported by the induction of antigen-specific Ab-producing cells in the mucosal (e.g., PPs) but not systemic compartment (e.g., spleen). The preferential presence of Alcaligenes inside PPs and the associated induction of intestinal secretory IgA Abs were also observed in both monkeys and humans. Localized mucosal Ab-mediated symbiotic immune responses were supported by Alcaligenes-stimulated CD11c(+) dendritic cells (DCs) producing the Ab-enhancing cytokines TGF-beta, B-cell-activating factor belonging to the TNF family, and IL-6 in PPs. These CD11c(+) DCs did not migrate beyond the draining mesenteric lymph nodes. In the absence of antigen-specific mucosal Abs, the presence of Alcaligenes in PPs was greatly diminished. Thus, indigenous opportunistic bacteria uniquely inhabit PPs, leading to PP-DCs-initiated, local antigen-specific Ab production; this may involve the creation of an optimal symbiotic environment on the interior of the PPs.


Subject(s)
Antibodies/chemistry , Bacteria/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Peyer's Patches/immunology , Animals , Humans , In Situ Hybridization, Fluorescence , Lymph Nodes/immunology , Lymphoid Tissue/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred DBA , Molecular Sequence Data , RNA, Ribosomal, 16S/metabolism , Spleen/immunology
4.
Digestion ; 84(2): 128-33, 2011.
Article in English | MEDLINE | ID: mdl-21525768

ABSTRACT

BACKGROUND/AIM: In previous studies, we described the beneficial effects of bifidobacteria-fermented milk in patients with ulcerative colitis (UC). Here, we examined the effects of a live Bifidobacterium breve strain Yakult, a probiotic contained in bifidobacteria-fermented milk, and galacto-oligosaccharide (GOS) as synbiotics in UC patients. METHODS: Forty-one patients with mild to moderate UC were assigned to two groups; one group was treated with the synbiotics and the other was not (control). The treatment group ingested 1 g of the probiotic powder (10(9) CFU/g) three times a day, and 5.5 g of GOS once a day for one year. At the start and the end of this study, colonoscopic index and the amount of myeloperoxidase in a lavage solution were used as disease activity indices. Bacterial counts in the feces at the start and the end of this study were also examined. RESULTS: After a one-year treatment with the synbiotics, the clinical status of the UC patients as assessed by colonoscopy, significantly improved. Furthermore, the amount of myeloperoxidase in the lavage also decreased in these patients after the synbiotic treatment. The synbiotics significantly reduced the fecal counts of Bacteroidaceae and fecal pH. CONCLUSION: Administration of live B. breve strain Yakult and GOS can improve the clinical condition of patients with UC. These results encouraged us to perform a large-scale randomized, placebo-controlled trial.


Subject(s)
Bifidobacterium , Colitis, Ulcerative/drug therapy , Oligosaccharides/therapeutic use , Prebiotics , Probiotics/therapeutic use , Adult , Colitis, Ulcerative/enzymology , Colitis, Ulcerative/microbiology , Colonoscopy , Feces/microbiology , Female , Humans , Male , Middle Aged , Peroxidase/analysis , Severity of Illness Index , Synbiotics
5.
FEMS Immunol Med Microbiol ; 52(1): 69-77, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17995961

ABSTRACT

Probiotics are used for the improvement of gut disorders. To explore the potential of probiotics, a gnotobiotic study using BALB/c mice to analyze epithelial gene expression was performed. Microarray analysis of probiotic strain-monoassociated mice showed that Lactobacillus casei Shirota and Bifidobacterium breve Yakult noticeably affected gene expression in the ileal and colonic epithelial cells, respectively, although to a smaller extent than segmented filamentous bacteria (SFB). Lactobacillus casei Shirota enhanced the gene expression involving defense/immune functions and lipid metabolism more strongly than B. breve Yakult. In the colon, expression of a chloride transporter was slightly enhanced, although downregulation of many genes, such as guanine nucleotide-binding protein, was evident in mice with B. breve Yakult compared with the ones with L. casei Shirota. SFB affected gene expression more strongly than the probiotic strains. In particular, alpha(1-2) fucosyltransferase and pancreatitis-associated protein were significantly enhanced only in SFB-monoassociated mice but not probiotic strain-monoassociated mice. Gene expression of SFB-monoassociated mice was either stimulated or repressed in a manner similar to or opposite that of conventional colonized mice. Taken together, probiotic strains of L. casei Shirota and B. breve Yakult differentially affect epithelial gene expression in the small intestine and colon, respectively.


Subject(s)
Bifidobacterium/physiology , Gene Expression Regulation , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Lacticaseibacillus casei/physiology , Probiotics , Animals , Colon/immunology , Colon/metabolism , Colon/microbiology , Colony Count, Microbial , Gene Expression Profiling , Ileum/immunology , Ileum/metabolism , Ileum/microbiology , Intestinal Mucosa/immunology , Lipid Metabolism/genetics , Mice , Mice, Inbred BALB C , Oligonucleotide Array Sequence Analysis , Pancreatitis-Associated Proteins
6.
PLoS One ; 11(8): e0160736, 2016.
Article in English | MEDLINE | ID: mdl-27500935

ABSTRACT

Psychological stress can exacerbate inflammatory bowel disease. However, the mechanisms underlying how psychological stress affects gut inflammation remain unclear. Here, we focused on the relationship between changes in the microbial community of mucosa-associated commensal bacteria (MACB) and mucosal immune responses induced by chronic psychological stress in a murine model of ulcerative colitis. Furthermore, we examined the effect of probiotic treatment on exacerbated colitis and MACB composition changes induced by chronic psychological stress. Repeated water avoidance stress (rWAS) in B6-Tcra-/- mice severely exacerbated colitis, which was evaluated by both colorectal tissue weight and histological score of colitis. rWAS treatment increased mRNA expression of UCN2 and IFN-γ in large intestinal lamina propria mononuclear cells (LI-LPMC). Interestingly, exacerbated colitis was associated with changes in the microbial community of MACB, specifically loss of bacterial species diversity and an increase in the component ratio of Clostridium, revealed by 16S rRNA gene amplicon analysis. Finally, the oral administration of a probiotic Lactobacillus strain was protective against the exacerbation of colitis and was associated with a change in the bacterial community of MACB in rWAS-exposed Tcra-/- mice. Taken together, these results suggested that loss of species diversity in MACB might play a key role in exacerbated colitis induced by chronic psychological stress. In addition, probiotic treatment may be used as a tool to preserve the diversity of bacterial species in MACB and alleviate gut inflammation induced by psychological stress.


Subject(s)
Colitis/etiology , Disease Models, Animal , Genes, T-Cell Receptor alpha/genetics , Intestinal Mucosa/microbiology , Lactobacillus/growth & development , Probiotics/administration & dosage , Stress, Psychological/complications , Animals , Chronic Disease , Colitis/psychology , Female , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , RNA, Ribosomal, 16S/genetics
7.
Microbes Infect ; 5(2): 115-22, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12650769

ABSTRACT

Indigenous Bacteroides strains are closely associated with the occurrence and exacerbation of ulcerative colitis (UC). In this study, we aimed to clarify the effect of Bifidobacterium strains, another major member of colonic bacteria, on the development of gut inflammation using gnotobiotic mouse models associated with Bacteroides strains isolated from UC patients. Dextran sulfate (DSS) administration induced inflammation in the large intestine, in particular of the cecum, in the gnotobiotic mice associated with three strains of Bacteroides vulgatus, judging from the myeloperoxidase activity, occult blood score, and IgG leakage into the intestinal contents. However, the severity of the inflammation was greatly reduced in the gnotobiotic mice associated with both B. vulgatus and Bifidobacterium strains. The severity of the cecal inflammation was well correlated with the concentration of succinic acid in the cecum. Bacteriologically, the density of B. vulgatus strain A (BV-A) greatly decreased and the predominant strain changed from BV-A to BV-B on additional association with Bifidobacterium strains. Among gnotobiotic mice associated with a single B. vulgatus strain, the severity of cecal inflammation in BV-A-associated mice was greater than that in BV-B-associated mice. Each Bifidobacterium strain produced compound(s) more effectively inhibiting the growth of BV-A than BV-B in in vitro culture. Taken together, these results suggest that the severity of DSS-induced gut inflammation is closely associated with a particular B. vulgatus strain, and that Bifidobacterium strains may repress exacerbation of intestinal inflammation through growth inhibition of the B. vulgatus strain.


Subject(s)
Antibiosis , Bacteroides/growth & development , Bifidobacterium/growth & development , Colitis, Ulcerative/microbiology , Germ-Free Life , Inflammation/prevention & control , Animals , Bacteroides/isolation & purification , Bacteroides Infections/microbiology , Cecum/immunology , Cecum/microbiology , Cecum/pathology , Colitis, Ulcerative/chemically induced , Dextran Sulfate/administration & dosage , Digestive System/immunology , Digestive System/microbiology , Digestive System/pathology , Disease Models, Animal , Female , Humans , Mice , Mice, Inbred BALB C
8.
Science ; 345(6202): 1254009, 2014 Sep 12.
Article in English | MEDLINE | ID: mdl-25214634

ABSTRACT

Fucosylation of intestinal epithelial cells, catalyzed by fucosyltransferase 2 (Fut2), is a major glycosylation mechanism of host-microbiota symbiosis. Commensal bacteria induce epithelial fucosylation, and epithelial fucose is used as a dietary carbohydrate by many of these bacteria. However, the molecular and cellular mechanisms that regulate the induction of epithelial fucosylation are unknown. Here, we show that type 3 innate lymphoid cells (ILC3) induced intestinal epithelial Fut2 expression and fucosylation in mice. This induction required the cytokines interleukin-22 and lymphotoxin in a commensal bacteria-dependent and -independent manner, respectively. Disruption of intestinal fucosylation led to increased susceptibility to infection by Salmonella typhimurium. Our data reveal a role for ILC3 in shaping the gut microenvironment through the regulation of epithelial glycosylation.


Subject(s)
Fucose/metabolism , Immunity, Innate , Intestinal Mucosa/immunology , Lymphocytes/immunology , Microbiota/immunology , Salmonella Infections/immunology , Salmonella typhimurium , Animals , Base Sequence , Disease Models, Animal , Fucosyltransferases/genetics , Fucosyltransferases/metabolism , Germ-Free Life , Glycosylation , Goblet Cells/enzymology , Goblet Cells/immunology , Goblet Cells/microbiology , Ileum/enzymology , Ileum/immunology , Ileum/microbiology , Interleukins/immunology , Intestinal Mucosa/enzymology , Intestinal Mucosa/microbiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Mutant Strains , Molecular Sequence Data , Paneth Cells/enzymology , Paneth Cells/immunology , Paneth Cells/microbiology , Salmonella Infections/microbiology , Interleukin-22 , Galactoside 2-alpha-L-fucosyltransferase
9.
World J Gastroenterol ; 19(8): 1200-9, 2013 Feb 28.
Article in English | MEDLINE | ID: mdl-23482518

ABSTRACT

AIM: To investigate the role of the pelvic nerve pathway in stress-induced acceleration of colorectal transit and defecation in rats. METHODS: Surgical transection of rectal nerves (rectal branches of the pelvic nerve), vagotomy (Vag) or adrenalectomy (Adx) were performed bilaterally in rats. Number of fecal pellet output of these rats was measured during 1-h water avoidance stress (WAS). To evaluate the colonic transit, rats were given phenol red through the catheter indwelled in the proximal colon and subjected to WAS. After WAS session, entire colon and rectum were isolated and distribution of phenol red was measured. Distal colonic and rectal transit was evaluated using glass bead. Rats were inserted the glass bead into the distal colon and evacuation rate of the bead was measured. Neural activation was assessed by immunohistochemical staining of c-Fos and PGP9.5 in colonic whole-mount preparations of longitudinal muscle myenteric plexus (LMMP). RESULTS: In the sham-operated rats (sham op), WAS significantly increased defecation and accelerated colorectal transit with marked elevation of plasma corticosterone level. Compared with sham-operated rats, increase in the excretion of fecal pellets during WAS was significantly reduced by rectal nerve transection (RNT) (sham op: 6.9 ± 0.8 vs RNT: 4.3 ± 0.6, P < 0.05) or Vag (sham op: 6.4 ± 0.8 vs Vag: 3.7 ± 1.1, P < 0.05), although corticosterone level remained elevated. Adx-rats significantly increased the defecation despite the lower corticosterone level. Distribution pattern of phenol red showed RNT inhibited distal colonic and rectal transit accelerated by WAS, while Vag inhibited proximal colonic transit. Suppression of distal colonic and rectal transit by RNT was further confirmed by the bead evacuation rate (sham op: 80.0% vs RNT: 53.8%). WAS significantly increased the number of c-Fos-immunoreactive neural cells in the LMMP of the proximal and distal colon, whereas c-Fos expression was decreased by RNT in the distal colon (sham op: 9.0 ± 2.0 vs RNT: 4.4 ± 1.0, P < 0.05) and decreased by Vag in the proximal colon. CONCLUSION: Pelvic nerve conveys WAS stimuli from the brain to the distal colon, and directly activate the myenteric neurons, followed by the increase of its motility.


Subject(s)
Colon/innervation , Defecation , Gastrointestinal Motility , Hypogastric Plexus/physiopathology , Parasympathetic Nervous System/physiopathology , Pelvis/innervation , Rectum/innervation , Stress, Psychological/physiopathology , Adrenalectomy , Animals , Biomarkers/metabolism , Disease Models, Animal , Efferent Pathways/physiopathology , Hypogastric Plexus/metabolism , Hypogastric Plexus/surgery , Male , Myenteric Plexus/metabolism , Myenteric Plexus/physiopathology , Parasympathetic Nervous System/surgery , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Sprague-Dawley , Stress, Psychological/complications , Time Factors , Vagotomy
10.
World J Gastroenterol ; 14(16): 2511-6, 2008 Apr 28.
Article in English | MEDLINE | ID: mdl-18442197

ABSTRACT

AIM: To determine the anti-inflammatory activity of probiotic Bifidobacteria in Bifidobacteria-fermented milk (BFM) which is effective against active ulcerative colitis (UC) and exacerbations of UC, and to explore the immunoregulatory mechanisms. METHODS: Peripheral blood mononuclear cells (PBMNC) from UC patients or HT-29 cells were co-cultured with heat-killed probiotic bacteria or culture supernatant of Bifidobacterium breve strain Yakult (BbrY) or Bifidobacterium bifidum strain Yakult (BbiY) to estimate the amount of IL-10 or IL-8 secreted. RESULTS: Both strains of probiotic Bifidobacteria contained in the BFM induced IL-10 production in PBMNC from UC patients, though BbrY was more effective than BbiY. Conditioned medium (CM) and DNA of both strains inhibited IL-8 secretion in HT-29 cells stimulated with TNF-alpha, whereas no such effect was observed with heat-killed bacteria. The inhibitory effect of CM derived from BbiY was greater than that of CM derived from BbrY. DNAs of the two strains had a comparable inhibitory activity against the secretion of IL-8. CM of BbiY induced a repression of IL-8 gene expression with a higher expression of IkappaB-zeta mRNA 4 h after culture of HT-29 cells compared to that in the absence of CM. CONCLUSION: Probiotic Bifidobacterium strains in BFM enhance IL-10 production in PBMNC and inhibit IL-8 secretion in intestinal epithelial cells, suggesting that BFM has anti-inflammatory effects against ulcerative colitis.


Subject(s)
Bifidobacterium/physiology , Colitis, Ulcerative/immunology , HT29 Cells/physiology , Interleukin-10/biosynthesis , Interleukin-8/antagonists & inhibitors , Leukocytes, Mononuclear/physiology , Probiotics , Cell Culture Techniques , Culture Media, Conditioned , Cytokines/metabolism , DNA, Bacterial/metabolism , Humans , Interleukin-8/metabolism , Leukocytes, Mononuclear/microbiology , Reverse Transcriptase Polymerase Chain Reaction , Tumor Necrosis Factor-alpha/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL