Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Res Sq ; 2023 Oct 23.
Article in English | MEDLINE | ID: mdl-37961431

ABSTRACT

Persistent colonization and outgrowth of pathogenic organisms in the intestine may occur due to long-term antibiotic usage or inflammatory conditions, which perpetuate dysregulated immunity and tissue damage1,2. Gram-negative Enterobacteriaceae gut pathobionts are particularly recalcitrant to conventional antibiotic treatment3,4, though an emerging body of evidence suggests that manipulation of the commensal microbiota may be a practical alternative therapeutic strategy5-7. In this study, we rationally isolated and down-selected commensal bacterial consortia from healthy human stool samples capable of strongly and specifically suppressing intestinal Enterobacteriaceae. One of the elaborated consortia, consisting of 18 commensal strains, effectively controlled ecological niches by regulating gluconate availability, thereby reestablishing colonization resistance and alleviating antibiotic-resistant Klebsiella-driven intestinal inflammation in mice. Harnessing these microbial activities in the form of live bacterial therapeutics may represent a promising solution to combat the growing threat of proinflammatory, antimicrobial-resistant bacterial infection.

2.
J Exp Med ; 220(8)2023 08 07.
Article in English | MEDLINE | ID: mdl-37163450

ABSTRACT

Group 2 innate lymphoid cells (ILC2s) expressing IL-5 and IL-13 are localized at various mucosal tissues and play critical roles in the induction of type 2 inflammation, response to helminth infection, and tissue repair. Here, we reveal a unique ILC2 subset in the mouse intestine that constitutively expresses IL-4 together with GATA3, ST2, KLRG1, IL-17RB, and IL-5. In this subset, IL-4 expression is regulated by mechanisms similar to but distinct from those observed in T cells and is partly affected by IL-25 signaling. Although the absence of the microbiota had marginal effects, feeding mice with a vitamin B1-deficient diet compromised the number of intestinal IL-4+ ILC2s. The decrease in the number of IL-4+ ILC2s caused by the vitamin B1 deficiency was accompanied by a reduction in IL-25-producing tuft cells. Our findings reveal that dietary vitamin B1 plays a critical role in maintaining interaction between tuft cells and IL-4+ ILC2s, a previously uncharacterized immune cell population that may contribute to maintaining intestinal homeostasis.


Subject(s)
Diet , Intestinal Mucosa , Thiamine , Animals , Mice , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Thiamine/metabolism , Specific Pathogen-Free Organisms , Mice, Inbred C57BL , Interleukin-4/metabolism , Gastrointestinal Microbiome , Organoids/cytology , Organoids/immunology , Trinitrobenzenesulfonic Acid
3.
Cell ; 185(19): 3501-3519.e20, 2022 09 15.
Article in English | MEDLINE | ID: mdl-36041436

ABSTRACT

How intestinal microbes regulate metabolic syndrome is incompletely understood. We show that intestinal microbiota protects against development of obesity, metabolic syndrome, and pre-diabetic phenotypes by inducing commensal-specific Th17 cells. High-fat, high-sugar diet promoted metabolic disease by depleting Th17-inducing microbes, and recovery of commensal Th17 cells restored protection. Microbiota-induced Th17 cells afforded protection by regulating lipid absorption across intestinal epithelium in an IL-17-dependent manner. Diet-induced loss of protective Th17 cells was mediated by the presence of sugar. Eliminating sugar from high-fat diets protected mice from obesity and metabolic syndrome in a manner dependent on commensal-specific Th17 cells. Sugar and ILC3 promoted outgrowth of Faecalibaculum rodentium that displaced Th17-inducing microbiota. These results define dietary and microbiota factors posing risk for metabolic syndrome. They also define a microbiota-dependent mechanism for immuno-pathogenicity of dietary sugar and highlight an elaborate interaction between diet, microbiota, and intestinal immunity in regulation of metabolic disorders.


Subject(s)
Metabolic Syndrome , Microbiota , Animals , Diet, High-Fat , Dietary Sugars , Interleukin-17 , Intestinal Mucosa , Lipids , Mice , Mice, Inbred C57BL , Obesity , Th17 Cells
4.
Nature ; 599(7885): 458-464, 2021 11.
Article in English | MEDLINE | ID: mdl-34325466

ABSTRACT

Centenarians have a decreased susceptibility to ageing-associated illnesses, chronic inflammation and infectious diseases1-3. Here we show that centenarians have a distinct gut microbiome that is enriched in microorganisms that are capable of generating unique secondary bile acids, including various isoforms of lithocholic acid (LCA): iso-, 3-oxo-, allo-, 3-oxoallo- and isoallolithocholic acid. Among these bile acids, the biosynthetic pathway for isoalloLCA had not been described previously. By screening 68 bacterial isolates from the faecal microbiota of a centenarian, we identified Odoribacteraceae strains as effective producers of isoalloLCA both in vitro and in vivo. Furthermore, we found that the enzymes 5α-reductase (5AR) and 3ß-hydroxysteroid dehydrogenase (3ß-HSDH) were responsible for the production of isoalloLCA. IsoalloLCA exerted potent antimicrobial effects against Gram-positive (but not Gram-negative) multidrug-resistant pathogens, including Clostridioides difficile and Enterococcus faecium. These findings suggest that the metabolism of specific bile acids may be involved in reducing the risk of infection with pathobionts, thereby potentially contributing to the maintenance of intestinal homeostasis.


Subject(s)
Bacteria/metabolism , Biosynthetic Pathways , Centenarians , Gastrointestinal Microbiome , Lithocholic Acid/analogs & derivatives , Lithocholic Acid/biosynthesis , 3-Hydroxysteroid Dehydrogenases/metabolism , Aged, 80 and over , Animals , Anti-Bacterial Agents/biosynthesis , Anti-Bacterial Agents/metabolism , Bacteria/classification , Bacteria/enzymology , Bacteria/isolation & purification , Cholestenone 5 alpha-Reductase/metabolism , Feces/chemistry , Feces/microbiology , Female , Gram-Positive Bacteria/metabolism , Humans , Lithocholic Acid/metabolism , Male , Mice , Symbiosis
5.
Neurotherapeutics ; 18(2): 920-937, 2021 04.
Article in English | MEDLINE | ID: mdl-33829410

ABSTRACT

Gut microbiome studies in multiple sclerosis (MS) patients are unravelling some consistent but modest patterns of gut dysbiosis. Among these, a significant decrease of Clostridia cluster IV and XIVa has been reported. In the present study, we investigated the therapeutic effect of a previously selected mixture of human gut-derived 17 Clostridia strains, which belong to Clostridia clusters IV, XIVa, and XVIII, on the clinical outcome of experimental autoimmune encephalomyelitis (EAE). The observed clinical improvement was related to lower demyelination and astrocyte reactivity as well as a tendency to lower microglia reactivity/infiltrating macrophages and axonal damage in the central nervous system (CNS), and to an enhanced immunoregulatory response of regulatory T cells in the periphery. Transcriptome studies also highlighted increased antiinflammatory responses related to interferon beta in the periphery and lower immune responses in the CNS. Since Clostridia-treated mice were found to present higher levels of the immunomodulatory short-chain fatty acid (SCFA) butyrate in the serum, we studied if this clinical effect could be reproduced by butyrate administration alone. Further EAE experiments proved its preventive but slight therapeutic impact on CNS autoimmunity. Thus, this smaller therapeutic effect highlighted that the Clostridia-induced clinical effect was not exclusively related to the SCFA and could not be reproduced by butyrate administration alone. Although it is still unknown if these Clostridia strains will have the same effect on MS patients, gut dysbiosis in MS patients could be partially rebalanced by these commensal bacteria and their immunoregulatory properties could have a beneficial effect on MS clinical course.


Subject(s)
Butyrates/administration & dosage , Clostridiaceae/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/therapy , Gastrointestinal Microbiome/physiology , Animals , Dysbiosis/immunology , Dysbiosis/pathology , Dysbiosis/therapy , Encephalomyelitis, Autoimmune, Experimental/pathology , Fatty Acids, Volatile/administration & dosage , Female , Humans , Mice , Mice, Inbred C57BL , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology
6.
Cell Rep ; 35(4): 109052, 2021 04 27.
Article in English | MEDLINE | ID: mdl-33910010

ABSTRACT

Host-microbe interactions orchestrate skin homeostasis, the dysregulation of which has been implicated in chronic inflammatory conditions such as atopic dermatitis and psoriasis. Here, we show that Staphylococcus cohnii is a skin commensal capable of beneficially inhibiting skin inflammation. We find that Tmem79-/- mice spontaneously develop interleukin-17 (IL-17)-producing T-cell-driven skin inflammation. Comparative skin microbiome analysis reveals that the disease activity index is negatively associated with S. cohnii. Inoculation with S. cohnii strains isolated from either mouse or human skin microbiota significantly prevents and ameliorates dermatitis in Tmem79-/- mice without affecting pathobiont burden. S. cohnii colonization is accompanied by activation of host glucocorticoid-related pathways and induction of anti-inflammatory genes in the skin and is therefore effective at suppressing inflammation in diverse pathobiont-independent dermatitis models, including chemically induced, type 17, and type 2 immune-driven models. As such, S. cohnii strains have great potential as effective live biotherapeutics for skin inflammation.


Subject(s)
Inflammation/immunology , Skin/pathology , Staphylococcus/metabolism , Animals , Humans , Mice
7.
Gut Microbes ; 12(1): 1788898, 2020 11 09.
Article in English | MEDLINE | ID: mdl-32691669

ABSTRACT

Dysbiotic microbiota contributes to the pathogenesis of Crohn's disease (CD) by regulating the immune system. Although pro-inflammatory microbes are probably enriched in the small intestinal (SI) mucosa, most studies have focused on fecal microbiota. This study aimed to examine jejunal and ileal mucosal specimens from patients with CD via double-balloon enteroscopy. Comparative microbiome analysis revealed that the microbiota composition of CD SI mucosa differs from that of non-CD controls, with an increased population of several families, including Enterobacteriaceae, Ruminococcaceae, and Bacteroidaceae. Upon anaerobic culturing of the CD SI mucosa, 80 bacterial strains were isolated, from which 9 strains representing 9 distinct species (Escherichia coli, Ruminococcus gnavus, Klebsiella pneumoniae, Erysipelatoclostridium ramosum, Bacteroides dorei, B. fragilis, B. uniformis, Parabacteroides distasonis, and Streptococcus pasteurianus) were selected on the basis of their significant association with CD. The colonization of germ-free (GF) mice with the 9 strains enhanced the accumulation of TH1 cells and, to a lesser extent, TH17 cells in the intestine, among which an E. coli strain displayed high potential to induce TH1 cells and intestinal inflammation in a strain-specific manner. The present results indicate that the CD SI mucosa harbors unique pro-inflammatory microbiota, including TH1 cell-inducing E. coli, which could be a potential therapeutic target.


Subject(s)
Crohn Disease/microbiology , Escherichia coli/pathogenicity , Intestinal Mucosa/microbiology , Intestine, Small/microbiology , Th1 Cells/metabolism , Adult , Animals , Clostridiales/isolation & purification , Clostridiales/pathogenicity , Crohn Disease/immunology , Escherichia coli/isolation & purification , Female , Gastrointestinal Microbiome , Humans , Intestine, Small/immunology , Male , Mice , Middle Aged , Th17 Cells/metabolism
8.
Nature ; 565(7741): 600-605, 2019 01.
Article in English | MEDLINE | ID: mdl-30675064

ABSTRACT

There is a growing appreciation for the importance of the gut microbiota as a therapeutic target in various diseases. However, there are only a handful of known commensal strains that can potentially be used to manipulate host physiological functions. Here we isolate a consortium of 11 bacterial strains from healthy human donor faeces that is capable of robustly inducing interferon-γ-producing CD8 T cells in the intestine. These 11 strains act together to mediate the induction without causing inflammation in a manner that is dependent on CD103+ dendritic cells and major histocompatibility (MHC) class Ia molecules. Colonization of mice with the 11-strain mixture enhances both host resistance against Listeria monocytogenes infection and the therapeutic efficacy of immune checkpoint inhibitors in syngeneic tumour models. The 11 strains primarily represent rare, low-abundance components of the human microbiome, and thus have great potential as broadly effective biotherapeutics.


Subject(s)
Adenocarcinoma/immunology , Adenocarcinoma/therapy , Bacteria/classification , CD8-Positive T-Lymphocytes/immunology , Gastrointestinal Microbiome/immunology , Listeriosis/prevention & control , Symbiosis/immunology , Adenocarcinoma/pathology , Animals , Antigens, CD/metabolism , Bacteria/immunology , Bacteria/isolation & purification , CD8-Positive T-Lymphocytes/cytology , Cell Line, Tumor , Dendritic Cells/immunology , Feces/microbiology , Female , Healthy Volunteers , Histocompatibility Antigens Class I/immunology , Humans , Integrin alpha Chains/metabolism , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Listeria monocytogenes/immunology , Listeriosis/immunology , Listeriosis/microbiology , Male , Mice , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , Xenograft Model Antitumor Assays
9.
Elife ; 72018 04 17.
Article in English | MEDLINE | ID: mdl-29664397

ABSTRACT

Manipulation of the gut microbiota holds great promise for the treatment of diseases. However, a major challenge is the identification of therapeutically potent microbial consortia that colonize the host effectively while maximizing immunologic outcome. Here, we propose a novel workflow to select optimal immune-inducing consortia from microbiome compositicon and immune effectors measurements. Using published and newly generated microbial and regulatory T-cell (Treg) data from germ-free mice, we estimate the contributions of twelve Clostridia strains with known immune-modulating effect to Treg induction. Combining this with a longitudinal data-constrained ecological model, we predict the ability of every attainable and ecologically stable subconsortium in promoting Treg activation and rank them by the Treg Induction Score (TrIS). Experimental validation of selected consortia indicates a strong and statistically significant correlation between predicted TrIS and measured Treg. We argue that computational indexes, such as the TrIS, are valuable tools for the systematic selection of immune-modulating bacteriotherapeutics.


Subject(s)
Firmicutes/immunology , Host Microbial Interactions , Immunity, Cellular , Microbial Consortia , T-Lymphocytes, Regulatory/immunology , Animals , Computer Simulation , Lymphocyte Activation , Mice
10.
Science ; 358(6361): 359-365, 2017 10 20.
Article in English | MEDLINE | ID: mdl-29051379

ABSTRACT

Intestinal colonization by bacteria of oral origin has been correlated with several negative health outcomes, including inflammatory bowel disease. However, a causal role of oral bacteria ectopically colonizing the intestine remains unclear. Using gnotobiotic techniques, we show that strains of Klebsiella spp. isolated from the salivary microbiota are strong inducers of T helper 1 (TH1) cells when they colonize in the gut. These Klebsiella strains are resistant to multiple antibiotics, tend to colonize when the intestinal microbiota is dysbiotic, and elicit a severe gut inflammation in the context of a genetically susceptible host. Our findings suggest that the oral cavity may serve as a reservoir for potential intestinal pathobionts that can exacerbate intestinal disease.


Subject(s)
Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/microbiology , Intestines/immunology , Klebsiella/immunology , Microbiota/immunology , Mouth/microbiology , Th1 Cells/immunology , Animals , Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial , Dysbiosis/immunology , Dysbiosis/microbiology , Germ-Free Life , Intestines/microbiology , Klebsiella/drug effects , Klebsiella/isolation & purification , Klebsiella/pathogenicity , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Saliva/microbiology
11.
Sci Immunol ; 2(13)2017 Jul 21.
Article in English | MEDLINE | ID: mdl-28733471

ABSTRACT

Specific gut commensal bacteria improve host health by eliciting mutualistic regulatory T (Treg) cell responses. However, the bacteria that induce effector T (Teff) cells during inflammation are unclear. We addressed this by analyzing bacterial-reactive T cell receptor (TCR) transgenic cells and TCR repertoires in a murine colitis model. Unexpectedly, we found that mucosal-associated Helicobacter species triggered both Treg cell responses during homeostasis and Teff cell responses during colitis, as suggested by an increased overlap between the Teff/Treg TCR repertoires with colitis. Four of six Treg TCRs tested recognized mucosal-associated Helicobacter species in vitro and in vivo. By contrast, the marked expansion of luminal Bacteroides species seen during colitis did not trigger a commensurate Teff cell response. Unlike other Treg cell-inducing bacteria, Helicobacter species are known pathobionts and cause disease in immunodeficient mice. Thus, our study suggests a model in which mucosal bacteria elicit context-dependent Treg or Teff cell responses to facilitate intestinal tolerance or inflammation.

12.
J Lipid Res ; 58(3): 519-528, 2017 03.
Article in English | MEDLINE | ID: mdl-28130274

ABSTRACT

The gut microbiota were shown to play critical roles in the development of atherosclerosis, but the detailed mechanism is limited. The purpose of this study is to clarify the influence of gut microbiota on atherogenesis via lipid metabolism and systemic inflammation. Germ-free or conventionally raised (Conv) ApoE-deficient (ApoE-/-) mice were fed chow diet and euthanized at 20 weeks of age. We found that the lack of gut microbiota in ApoE-/- mice caused a significant increase in the plasma and hepatic cholesterol levels compared with Conv ApoE-/- mice. The absence of gut microbiota changed the bile acid composition in the ileum, which was associated with activation of the enterohepatic fibroblast growth factor 15, fibroblast growth factor receptor 4 axis, and reduction of cholesterol 7α-hydroxylase and hepatic bile acid synthesis, resulting in the accumulation of liver cholesterol content. However, we found that the lack of microbiota caused a significant reduction in atherosclerotic lesion formation compared with Conv ApoE-/- mice, which might be associated with the attenuation of lipopolysaccharide-mediated inflammatory responses. Our findings indicated that the gut microbiota affected both hypercholesterolemia and atherogenesis in mice.


Subject(s)
Apolipoproteins E/genetics , Atherosclerosis/microbiology , Cholesterol/metabolism , Inflammation/microbiology , Animals , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Bacteria/metabolism , Bacteria/pathogenicity , Cholesterol/genetics , Cholesterol 7-alpha-Hydroxylase/genetics , Diet , Disease Models, Animal , Fibroblast Growth Factors/genetics , Gastrointestinal Microbiome/genetics , Homeostasis , Humans , Ileum/metabolism , Ileum/microbiology , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Lipid Metabolism/genetics , Mice , Receptor, Fibroblast Growth Factor, Type 4/genetics
13.
Genome Biol ; 17(1): 121, 2016 06 03.
Article in English | MEDLINE | ID: mdl-27259475

ABSTRACT

Predicting dynamics of host-microbial ecosystems is crucial for the rational design of bacteriotherapies. We present MDSINE, a suite of algorithms for inferring dynamical systems models from microbiome time-series data and predicting temporal behaviors. Using simulated data, we demonstrate that MDSINE significantly outperforms the existing inference method. We then show MDSINE's utility on two new gnotobiotic mice datasets, investigating infection with Clostridium difficile and an immune-modulatory probiotic. Using these datasets, we demonstrate new capabilities, including accurate forecasting of microbial dynamics, prediction of stable sub-communities that inhibit pathogen growth, and identification of bacteria most crucial to community integrity in response to perturbations.


Subject(s)
Clostridioides difficile/genetics , Host-Pathogen Interactions/genetics , Microbiota/genetics , Models, Theoretical , Algorithms , Animals , Clostridioides difficile/growth & development , Clostridioides difficile/pathogenicity , Mice
14.
Nat Med ; 22(6): 679-84, 2016 06.
Article in English | MEDLINE | ID: mdl-27111280

ABSTRACT

CD4(+) T cells that express the forkhead box P3 (FOXP3) transcription factor function as regulatory T (Treg) cells and hinder effective immune responses against cancer cells. Abundant Treg cell infiltration into tumors is associated with poor clinical outcomes in various types of cancers. However, the role of Treg cells is controversial in colorectal cancers (CRCs), in which FOXP3(+) T cell infiltration indicated better prognosis in some studies. Here we show that CRCs, which are commonly infiltrated by suppression-competent FOXP3(hi) Treg cells, can be classified into two types by the degree of additional infiltration of FOXP3(lo) nonsuppressive T cells. The latter, which are distinguished from FOXP3(+) Treg cells by non-expression of the naive T cell marker CD45RA and instability of FOXP3, secreted inflammatory cytokines. Indeed, CRCs with abundant infiltration of FOXP3(lo) T cells showed significantly better prognosis than those with predominantly FOXP3(hi) Treg cell infiltration. Development of such inflammatory FOXP3(lo) non-Treg cells may depend on secretion of interleukin (IL)-12 and transforming growth factor (TGF)-ß by tissues and their presence was correlated with tumor invasion by intestinal bacteria, especially Fusobacterium nucleatum. Thus, functionally distinct subpopulations of tumor-infiltrating FOXP3(+) T cells contribute in opposing ways to determining CRC prognosis. Depletion of FOXP3(hi) Treg cells from tumor tissues, which would augment antitumor immunity, could thus be used as an effective treatment strategy for CRCs and other cancers, whereas strategies that locally increase the population of FOXP3(lo) non-Treg cells could be used to suppress or prevent tumor formation.


Subject(s)
Colorectal Neoplasms/immunology , Lymphocytes, Tumor-Infiltrating/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Adult , Aged , Aged, 80 and over , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Colorectal Neoplasms/surgery , Digestive System Surgical Procedures , Female , Forkhead Transcription Factors/metabolism , Humans , Interleukin-12 Subunit p35/immunology , Interleukin-12 Subunit p35/metabolism , Lymphocytes, Tumor-Infiltrating/metabolism , Male , Middle Aged , Neoplasm Staging , Prognosis , Survival Rate , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/metabolism , Transforming Growth Factor beta1/immunology , Transforming Growth Factor beta1/metabolism
15.
Nat Rev Immunol ; 16(5): 295-309, 2016 05.
Article in English | MEDLINE | ID: mdl-27087661

ABSTRACT

Gut-resident forkhead box P3 (FOXP3)(+)CD4(+) regulatory T cells (Treg cells) are distinct from those in other organs and have gut-specific phenotypes and functions. Whereas Treg cells in other organs have T cell receptors (TCRs) specific for self antigens, intestinal Treg cells have a distinct set of TCRs that are specific for intestinal antigens, and these cells have pivotal roles in the suppression of immune responses against harmless dietary antigens and commensal microorganisms. The differentiation, migration and maintenance of intestinal Treg cells are controlled by specific signals from the local environment. In particular, certain members of the microbiota continuously provide antigens and immunoregulatory small molecules that modulate intestinal Treg cells. Understanding the development and the maintenance of intestinal Treg cells provides important insights into disease-relevant host-microorganism interactions.


Subject(s)
Immunity, Mucosal , Peyer's Patches/cytology , Peyer's Patches/physiology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Animals , Cell Differentiation/genetics , Cell Differentiation/immunology , Cellular Microenvironment , Clonal Evolution/genetics , Clonal Evolution/immunology , Gastrointestinal Microbiome/immunology , Humans , Immune Tolerance , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/cytology
16.
Proc Natl Acad Sci U S A ; 112(41): 12770-5, 2015 Oct 13.
Article in English | MEDLINE | ID: mdl-26420876

ABSTRACT

The regulation of intestinal homeostasis by the immune system involves the dynamic interplay between gut commensal microbiota and resident immune cells. It is well known that a large and diverse lymphocyte antigen receptor repertoire enables the immune system to recognize and respond to a wide range of invading pathogens. There is also an emerging appreciation for a critical role the T-cell receptor (TCR) repertoire serves in the maintenance of peripheral tolerance by regulatory T cells (Tregs). Nevertheless, how the diversity of the TCR repertoire in Tregs affects intestinal homeostasis remains unknown. To address this question, we studied mice whose T cells express a restricted TCR repertoire. We observed the development of spontaneous colitis, accompanied by the induction of T-helper type 17 cells in the colon that is driven by gut commensal microbiota. We provide further evidence that a restricted TCR repertoire causes a loss of tolerogenicity to microbiota, accompanied by a paucity of peripherally derived, Helios(-) Tregs and hyperactivation of migratory dendritic cells. These results thus reveal a new facet of the TCR repertoire in which Tregs require a diverse TCR repitoire for intestinal homeostasis, suggesting an additional driving force in the evolutional significance of the TCR repertoire.


Subject(s)
Cell Movement/immunology , Colon/immunology , Microbiota/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Animals , Cell Movement/genetics , Colon/microbiology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , Dendritic Cells/immunology , Mice , Mice, Mutant Strains , Receptors, Antigen, T-Cell/genetics , Transcription Factors/genetics , Transcription Factors/immunology
17.
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
18.
Nature ; 500(7461): 232-6, 2013 Aug 08.
Article in English | MEDLINE | ID: mdl-23842501

ABSTRACT

Manipulation of the gut microbiota holds great promise for the treatment of inflammatory and allergic diseases. Although numerous probiotic microorganisms have been identified, there remains a compelling need to discover organisms that elicit more robust therapeutic responses, are compatible with the host, and can affect a specific arm of the host immune system in a well-controlled, physiological manner. Here we use a rational approach to isolate CD4(+)FOXP3(+) regulatory T (Treg)-cell-inducing bacterial strains from the human indigenous microbiota. Starting with a healthy human faecal sample, a sequence of selection steps was applied to obtain mice colonized with human microbiota enriched in Treg-cell-inducing species. From these mice, we isolated and selected 17 strains of bacteria on the basis of their high potency in enhancing Treg cell abundance and inducing important anti-inflammatory molecules--including interleukin-10 (IL-) and inducible T-cell co-stimulator (ICOS)--in Treg cells upon inoculation into germ-free mice. Genome sequencing revealed that the 17 strains fall within clusters IV, XIVa and XVIII of Clostridia, which lack prominent toxins and virulence factors. The 17 strains act as a community to provide bacterial antigens and a TGF-ß-rich environment to help expansion and differentiation of Treg cells. Oral administration of the combination of 17 strains to adult mice attenuated disease in models of colitis and allergic diarrhoea. Use of the isolated strains may allow for tailored therapeutic manipulation of human immune disorders.


Subject(s)
Clostridium/immunology , Metagenome/immunology , T-Lymphocytes, Regulatory/physiology , Adult , Animals , Cell Proliferation , Clostridium/classification , Clostridium/genetics , Colitis/microbiology , Colitis/pathology , Colon/immunology , Colon/microbiology , Disease Models, Animal , Feces/microbiology , Germ-Free Life , Humans , Inducible T-Cell Co-Stimulator Protein/metabolism , Interleukin-10/metabolism , Male , Metagenome/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, SCID , RNA, Ribosomal, 16S/genetics , Rats , Rats, Inbred F344 , T-Lymphocytes, Regulatory/cytology
19.
Semin Immunol ; 24(1): 50-7, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22172550

ABSTRACT

CD4+ regulatory T (Treg) cells expressing the transcription factor forkhead box P3 (Foxp3) play a critical role in maintaining immunological homeostasis. Treg cells are highly abundant in the mouse intestinal lamina propria, particularly in the colon. Recent studies using germ-free and gnotobiotic mice have revealed that specific components of the intestinal microbiota influence the number and function of Treg cells. Substantial changes in the composition of microbiota have been associated with inflammatory bowel disease. In this review, we will discuss recent findings that associate intestinal microbiota in mice with Treg responses and with the maintenance of intestinal immune homeostasis.


Subject(s)
Colon/immunology , Colon/microbiology , Homeostasis , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Metagenome , T-Lymphocytes, Regulatory/immunology , Animals , Humans , Immunity, Mucosal , Mice
20.
Science ; 331(6015): 337-41, 2011 Jan 21.
Article in English | MEDLINE | ID: mdl-21205640

ABSTRACT

CD4(+) T regulatory cells (T(regs)), which express the Foxp3 transcription factor, play a critical role in the maintenance of immune homeostasis. Here, we show that in mice, T(regs) were most abundant in the colonic mucosa. The spore-forming component of indigenous intestinal microbiota, particularly clusters IV and XIVa of the genus Clostridium, promoted T(reg) cell accumulation. Colonization of mice by a defined mix of Clostridium strains provided an environment rich in transforming growth factor-ß and affected Foxp3(+) T(reg) number and function in the colon. Oral inoculation of Clostridium during the early life of conventionally reared mice resulted in resistance to colitis and systemic immunoglobulin E responses in adult mice, suggesting a new therapeutic approach to autoimmunity and allergy.


Subject(s)
Clostridium/immunology , Colon/immunology , Colon/microbiology , Intestinal Mucosa/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Anti-Bacterial Agents/pharmacology , Cecum/microbiology , Cells, Cultured , Clostridium/growth & development , Colitis/immunology , Colitis/pathology , Colitis/prevention & control , Colon/metabolism , Feces/microbiology , Forkhead Transcription Factors/metabolism , Germ-Free Life , Immunity, Innate , Immunoglobulin E/biosynthesis , Interleukin-10/immunology , Interleukin-10/metabolism , Intestinal Mucosa/metabolism , Intestine, Small/immunology , Metagenome , Mice , Mice, Inbred A , Mice, Inbred BALB C , Receptors, Pattern Recognition/physiology , Specific Pathogen-Free Organisms , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Regulatory/metabolism , Transforming Growth Factor beta/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...