Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 151
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 187(4): 897-913.e18, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38280374

ABSTRACT

Canonically, the complement system is known for its rapid response to remove microbes in the bloodstream. However, relatively little is known about a functioning complement system on intestinal mucosal surfaces. Herein, we report the local synthesis of complement component 3 (C3) in the gut, primarily by stromal cells. C3 is expressed upon commensal colonization and is regulated by the composition of the microbiota in healthy humans and mice, leading to an individual host's specific luminal C3 levels. The absence of membrane attack complex (MAC) components in the gut ensures that C3 deposition does not result in the lysis of commensals. Pathogen infection triggers the immune system to recruit neutrophils to the infection site for pathogen clearance. Basal C3 levels directly correlate with protection against enteric infection. Our study reveals the gut complement system as an innate immune mechanism acting as a vigilant sentinel that combats pathogens and spares commensals.


Subject(s)
Complement C3 , Intestinal Mucosa , Microbiota , Animals , Humans , Mice , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Neutrophils , Complement C3/metabolism , Stromal Cells/metabolism
2.
Cell ; 185(22): 4190-4205.e25, 2022 10 27.
Article in English | MEDLINE | ID: mdl-36243004

ABSTRACT

Neuroepithelial crosstalk is critical for gut physiology. However, the mechanisms by which sensory neurons communicate with epithelial cells to mediate gut barrier protection at homeostasis and during inflammation are not well understood. Here, we find that Nav1.8+CGRP+ nociceptor neurons are juxtaposed with and signal to intestinal goblet cells to drive mucus secretion and gut protection. Nociceptor ablation led to decreased mucus thickness and dysbiosis, while chemogenetic nociceptor activation or capsaicin treatment induced mucus growth. Mouse and human goblet cells expressed Ramp1, receptor for the neuropeptide CGRP. Nociceptors signal via the CGRP-Ramp1 pathway to induce rapid goblet cell emptying and mucus secretion. Notably, commensal microbes activated nociceptors to control homeostatic CGRP release. In the absence of nociceptors or epithelial Ramp1, mice showed increased epithelial stress and susceptibility to colitis. Conversely, CGRP administration protected nociceptor-ablated mice against colitis. Our findings demonstrate a neuron-goblet cell axis that orchestrates gut mucosal barrier protection.


Subject(s)
Colitis , Goblet Cells , Mice , Humans , Animals , Goblet Cells/metabolism , Nociceptors/metabolism , Calcitonin Gene-Related Peptide/metabolism , Colitis/metabolism , Mucus/metabolism , Receptor Activity-Modifying Protein 1/metabolism
3.
Cell ; 183(5): 1312-1324.e10, 2020 11 25.
Article in English | MEDLINE | ID: mdl-33212011

ABSTRACT

Interferon (IFN)-Is are crucial mediators of antiviral immunity and homeostatic immune system regulation. However, the source of IFN-I signaling under homeostatic conditions is unclear. We discovered that commensal microbes regulate the IFN-I response through induction of IFN-ß by colonic DCs. Moreover, the mechanism by which a specific commensal microbe induces IFN-ß was identified. Outer membrane (OM)-associated glycolipids of gut commensal microbes belonging to the Bacteroidetes phylum induce expression of IFN-ß. Using Bacteroides fragilis and its OM-associated polysaccharide A, we determined that IFN-ß expression was induced via TLR4-TRIF signaling. Antiviral activity of this purified microbial molecule against infection with either vesicular stomatitis virus (VSV) or influenza was demonstrated to be dependent on the induction of IFN-ß. In a murine VSV infection model, commensal-induced IFN-ß regulated natural resistance to virus infection. Due to the physiological importance of IFN-Is, discovery of an IFN-ß-inducing microbial molecule represents a potential approach for the treatment of some human diseases.


Subject(s)
Immunity, Innate , Microbiota , Virus Diseases/microbiology , Animals , Bacteroides fragilis/physiology , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Cell Membrane/drug effects , Cell Membrane/metabolism , Colon/pathology , Colon/virology , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Female , Gene Expression Regulation/drug effects , Glycolipids/metabolism , Immunity, Innate/drug effects , Interferon-beta/blood , Interferon-beta/metabolism , Male , Mice, Inbred C57BL , Microbiota/drug effects , Polysaccharides, Bacterial/pharmacology , Toll-Like Receptor 4/metabolism , Vesiculovirus/physiology , Virus Diseases/genetics
4.
Cell ; 181(6): 1276-1290.e13, 2020 06 11.
Article in English | MEDLINE | ID: mdl-32402238

ABSTRACT

At the species level, immunity depends on the selection and transmission of protective components of the immune system. A microbe-induced population of RORγ-expressing regulatory T cells (Tregs) is essential in controlling gut inflammation. We uncovered a non-genetic, non-epigenetic, non-microbial mode of transmission of their homeostatic setpoint. RORγ+ Treg proportions varied between inbred mouse strains, a trait transmitted by the mother during a tight age window after birth but stable for life, resistant to many microbial or cellular perturbations, then further transferred by females for multiple generations. RORγ+ Treg proportions negatively correlated with IgA production and coating of gut commensals, traits also subject to maternal transmission, in an immunoglobulin- and RORγ+ Treg-dependent manner. We propose a model based on a double-negative feedback loop, vertically transmitted via the entero-mammary axis. This immunologic mode of multi-generational transmission may provide adaptability and modulate the genetic tuning of gut immune responses and inflammatory disease susceptibility.


Subject(s)
Digestive System/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Disease Susceptibility/immunology , Female , Gastrointestinal Microbiome/immunology , Homeostasis/immunology , Immunoglobulin A/immunology , Inflammation/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Inbred NOD , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology
5.
Immunity ; 57(4): 876-889.e11, 2024 Apr 09.
Article in English | MEDLINE | ID: mdl-38479384

ABSTRACT

Concentrations of the secondary bile acid, deoxycholic acid (DCA), are aberrantly elevated in colorectal cancer (CRC) patients, but the consequences remain poorly understood. Here, we screened a library of gut microbiota-derived metabolites and identified DCA as a negative regulator for CD8+ T cell effector function. Mechanistically, DCA suppressed CD8+ T cell responses by targeting plasma membrane Ca2+ ATPase (PMCA) to inhibit Ca2+-nuclear factor of activated T cells (NFAT)2 signaling. In CRC patients, CD8+ T cell effector function negatively correlated with both DCA concentration and expression of a bacterial DCA biosynthetic gene. Bacteria harboring DCA biosynthetic genes suppressed CD8+ T cells effector function and promoted tumor growth in mice. This effect was abolished by disrupting bile acid metabolism via bile acid chelation, genetic ablation of bacterial DCA biosynthetic pathway, or specific bacteriophage. Our study demonstrated causation between microbial DCA metabolism and anti-tumor CD8+ T cell response in CRC, suggesting potential directions for anti-tumor therapy.


Subject(s)
Colorectal Neoplasms , Gastrointestinal Microbiome , Humans , Mice , Animals , Bile Acids and Salts , Deoxycholic Acid/pharmacology , CD8-Positive T-Lymphocytes
6.
Annu Rev Immunol ; 28: 107-30, 2010.
Article in English | MEDLINE | ID: mdl-19968562

ABSTRACT

The capsular polysaccharides (CPSs) of most pathogenic bacteria are T cell-independent antigens whose conjugation to carrier proteins evokes a carbohydrate-specific response eliciting T cell help. However, certain bacterial CPSs, known as zwitterionic polysaccharides (ZPSs), activate the adaptive immune system through processing by antigen-presenting cells and presentation by the major histocompatibility complex class II pathway to CD4(+) T cells. This discovery was the first mechanistic insight into how carbohydrates-a class of biological molecules previously thought to be T cell independent-can in fact activate T cells. Through their ability to activate CD4(+) T cells, ZPSs direct the cellular and physical maturation of the developing immune system. In this review, we explore the still-enigmatic relations between CPSs and the adaptive immune machinery at the cellular and molecular levels, and we discuss how new insights into the biological impact of ZPSs expand our concepts of the role of carbohydrates in microbial interactions with the adaptive immune system.


Subject(s)
Adaptive Immunity , Antigens, Bacterial/immunology , Bacteria/immunology , Polysaccharides, Bacterial/immunology , Animals , Antigens, Bacterial/chemistry , Bacteria/chemistry , Bacterial Vaccines/immunology , Humans , Polysaccharides, Bacterial/chemistry , T-Lymphocytes/immunology
7.
Cell ; 168(6): 1135-1148.e12, 2017 03 09.
Article in English | MEDLINE | ID: mdl-28262351

ABSTRACT

Investigation of host-environment interactions in the gut would benefit from a culture system that maintained tissue architecture yet allowed tight experimental control. We devised a microfabricated organ culture system that viably preserves the normal multicellular composition of the mouse intestine, with luminal flow to control perturbations (e.g., microbes, drugs). It enables studying short-term responses of diverse gut components (immune, neuronal, etc.). We focused on the early response to bacteria that induce either Th17 or RORg+ T-regulatory (Treg) cells in vivo. Transcriptional responses partially reproduced in vivo signatures, but these microbes elicited diametrically opposite changes in expression of a neuronal-specific gene set, notably nociceptive neuropeptides. We demonstrated activation of sensory neurons by microbes, correlating with RORg+ Treg induction. Colonic RORg+ Treg frequencies increased in mice lacking TAC1 neuropeptide precursor and decreased in capsaicin-diet fed mice. Thus, differential engagement of the enteric nervous system may partake in bifurcating pro- or anti-inflammatory responses to microbes.


Subject(s)
Clostridium/growth & development , Intestines/growth & development , Intestines/microbiology , Organ Culture Techniques , Animals , Clostridium/classification , Clostridium/physiology , Intestines/cytology , Mice , Symbiosis
8.
Cell ; 168(5): 928-943.e11, 2017 02 23.
Article in English | MEDLINE | ID: mdl-28215708

ABSTRACT

Within the human gut reside diverse microbes coexisting with the host in a mutually advantageous relationship. Evidence has revealed the pivotal role of the gut microbiota in shaping the immune system. To date, only a few of these microbes have been shown to modulate specific immune parameters. Herein, we broadly identify the immunomodulatory effects of phylogenetically diverse human gut microbes. We monocolonized mice with each of 53 individual bacterial species and systematically analyzed host immunologic adaptation to colonization. Most microbes exerted several specialized, complementary, and redundant transcriptional and immunomodulatory effects. Surprisingly, these were independent of microbial phylogeny. Microbial diversity in the gut ensures robustness of the microbiota's ability to generate a consistent immunomodulatory impact, serving as a highly important epigenetic system. This study provides a foundation for investigation of gut microbiota-host mutualism, highlighting key players that could identify important therapeutics.


Subject(s)
Bacteria/classification , Gastrointestinal Microbiome , Gastrointestinal Tract/immunology , Gastrointestinal Tract/microbiology , Adaptive Immunity , Animals , Bacterial Physiological Phenomena , Gastrointestinal Tract/cytology , Gastrointestinal Tract/physiology , Germ-Free Life , Humans , Immunity, Innate , Mice , Mice, Inbred C57BL , Symbiosis
9.
Cell ; 167(5): 1161-1162, 2016 11 17.
Article in English | MEDLINE | ID: mdl-27863236

ABSTRACT

In this issue of Cell, Desai et al. compare how dietary fiber affects the gut microbiota and susceptibility to disease. They find that a fiber-free diet promotes mucus-degrading bacteria and susceptibility to Citrobacter rodentium infection.


Subject(s)
Citrobacter rodentium , Enterobacteriaceae Infections/microbiology , Bacteria , Diet , Disease Susceptibility , Humans
10.
Cell ; 156(1-2): 123-33, 2014 Jan 16.
Article in English | MEDLINE | ID: mdl-24439373

ABSTRACT

Coevolution of beneficial microorganisms with the mammalian intestine fundamentally shapes mammalian physiology. Here, we report that the intestinal microbe Bacteroides fragilis modifies the homeostasis of host invariant natural killer T (iNKT) cells by supplementing the host's endogenous lipid antigen milieu with unique inhibitory sphingolipids. The process occurs early in life and effectively impedes iNKT cell proliferation during neonatal development. Consequently, total colonic iNKT cell numbers are restricted into adulthood, and hosts are protected against experimental iNKT cell-mediated, oxazolone-induced colitis. In studies with neonatal mice lacking access to bacterial sphingolipids, we found that treatment with B. fragilis glycosphingolipids-exemplified by an isolated peak (MW = 717.6) called GSL-Bf717-reduces colonic iNKT cell numbers and confers protection against oxazolone-induced colitis in adulthood. Our results suggest that the distinctive inhibitory capacity of GSL-Bf717 and similar molecules may prove useful in the treatment of autoimmune and allergic disorders in which iNKT cell activation is destructive.


Subject(s)
Bacteroides fragilis/metabolism , Colitis/immunology , Glycosphingolipids/metabolism , Natural Killer T-Cells/immunology , Animals , Animals, Newborn , Cell Proliferation , Colitis/chemically induced , Colitis/prevention & control , Colon/growth & development , Colon/microbiology , Mice , Mice, Inbred C57BL , Natural Killer T-Cells/cytology , Oxazolone
11.
Nature ; 619(7971): 837-843, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37380774

ABSTRACT

The human gut microbiome constantly converts natural products derived from the host and diet into numerous bioactive metabolites1-3. Dietary fats are essential micronutrients that undergo lipolysis to release free fatty acids (FAs) for absorption in the small intestine4. Gut commensal bacteria modify some unsaturated FAs-for example, linoleic acid (LA)-into various intestinal FA isomers that regulate host metabolism and have anticarcinogenic properties5. However, little is known about how this diet-microorganism FA isomerization network affects the mucosal immune system of the host. Here we report that both dietary factors and microbial factors influence the level of gut LA isomers (conjugated LAs (CLAs)) and that CLAs in turn modulate a distinct population of CD4+ intraepithelial lymphocytes (IELs) that express CD8αα in the small intestine. Genetic abolition of FA isomerization pathways in individual gut symbionts significantly decreases the number of CD4+CD8αα+ IELs in gnotobiotic mice. Restoration of CLAs increases CD4+CD8αα+ IEL levels in the presence of the transcription factor hepatocyte nuclear factor 4γ (HNF4γ). Mechanistically, HNF4γ facilitates CD4+CD8αα+ IEL development by modulating interleukin-18 signalling. In mice, specific deletion of HNF4γ in T cells leads to early mortality from infection by intestinal pathogens. Our data reveal a new role for bacterial FA metabolic pathways in the control of host intraepithelial immunological homeostasis by modulating the relative number of CD4+ T cells that were CD4+CD8αα+.


Subject(s)
Fatty Acids , Gastrointestinal Microbiome , Intraepithelial Lymphocytes , Animals , Humans , Mice , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Fatty Acids/chemistry , Fatty Acids/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intraepithelial Lymphocytes/immunology , Intraepithelial Lymphocytes/metabolism , Isomerism , Mice, Inbred C57BL , Receptors, Antigen, T-Cell, alpha-beta/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Lipolysis , Linoleic Acid/metabolism , Immunity, Mucosal
12.
Nature ; 617(7960): 377-385, 2023 05.
Article in English | MEDLINE | ID: mdl-37138075

ABSTRACT

The gut microbiota is a crucial regulator of anti-tumour immunity during immune checkpoint inhibitor therapy. Several bacteria that promote an anti-tumour response to immune checkpoint inhibitors have been identified in mice1-6. Moreover, transplantation of faecal specimens from responders can improve the efficacy of anti-PD-1 therapy in patients with melanoma7,8. However, the increased efficacy from faecal transplants is variable and how gut bacteria promote anti-tumour immunity remains unclear. Here we show that the gut microbiome downregulates PD-L2 expression and its binding partner repulsive guidance molecule b (RGMb) to promote anti-tumour immunity and identify bacterial species that mediate this effect. PD-L1 and PD-L2 share PD-1 as a binding partner, but PD-L2 can also bind RGMb. We demonstrate that blockade of PD-L2-RGMb interactions can overcome microbiome-dependent resistance to PD-1 pathway inhibitors. Antibody-mediated blockade of the PD-L2-RGMb pathway or conditional deletion of RGMb in T cells combined with an anti-PD-1 or anti-PD-L1 antibody promotes anti-tumour responses in multiple mouse tumour models that do not respond to anti-PD-1 or anti-PD-L1 alone (germ-free mice, antibiotic-treated mice and even mice colonized with stool samples from a patient who did not respond to treatment). These studies identify downregulation of the PD-L2-RGMb pathway as a specific mechanism by which the gut microbiota can promote responses to PD-1 checkpoint blockade. The results also define a potentially effective immunological strategy for treating patients who do not respond to PD-1 cancer immunotherapy.


Subject(s)
Drug Resistance, Neoplasm , Immunotherapy , Melanoma , Microbiota , Animals , Humans , Mice , Cell Adhesion Molecules, Neuronal , Disease Models, Animal , Down-Regulation , Drug Resistance, Neoplasm/drug effects , Fecal Microbiota Transplantation , Germ-Free Life , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Melanoma/immunology , Melanoma/microbiology , Melanoma/therapy , Protein Binding/drug effects , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
13.
Cell ; 149(7): 1578-93, 2012 Jun 22.
Article in English | MEDLINE | ID: mdl-22726443

ABSTRACT

Gut microbial induction of host immune maturation exemplifies host-microbe mutualism. We colonized germ-free (GF) mice with mouse microbiota (MMb) or human microbiota (HMb) to determine whether small intestinal immune maturation depends on a coevolved host-specific microbiota. Gut bacterial numbers and phylum abundance were similar in MMb and HMb mice, but bacterial species differed, especially the Firmicutes. HMb mouse intestines had low levels of CD4(+) and CD8(+) T cells, few proliferating T cells, few dendritic cells, and low antimicrobial peptide expression--all characteristics of GF mice. Rat microbiota also failed to fully expand intestinal T cell numbers in mice. Colonizing GF or HMb mice with mouse-segmented filamentous bacteria (SFB) partially restored T cell numbers, suggesting that SFB and other MMb organisms are required for full immune maturation in mice. Importantly, MMb conferred better protection against Salmonella infection than HMb. A host-specific microbiota appears to be critical for a healthy immune system.


Subject(s)
Immunity, Innate , Intestines/immunology , Intestines/microbiology , Metagenome , Animals , Bacteria/classification , Bacteria/genetics , Bacteria/metabolism , Cell Proliferation , Female , Germ-Free Life , Humans , Male , Mice , Rats , Rats, Sprague-Dawley , Salmonella Infections/immunology , Species Specificity , Specific Pathogen-Free Organisms , Symbiosis , T-Lymphocytes/cytology , T-Lymphocytes/immunology
14.
Nature ; 600(7888): 302-307, 2021 12.
Article in English | MEDLINE | ID: mdl-34759313

ABSTRACT

Small molecules derived from symbiotic microbiota critically contribute to intestinal immune maturation and regulation1. However, little is known about the molecular mechanisms that control immune development in the host-microbiota environment. Here, using a targeted lipidomic analysis and synthetic approach, we carried out a multifaceted investigation of immunomodulatory α-galactosylceramides from the human symbiont Bacteroides fragilis (BfaGCs). The characteristic terminal branching of BfaGCs is the result of incorporation of branched-chain amino acids taken up in the host gut by B. fragilis. A B. fragilis knockout strain that cannot metabolize branched-chain amino acids showed reduced branching in BfaGCs, and mice monocolonized with this mutant strain had impaired colonic natural killer T (NKT) cell regulation, implying structure-specific immunomodulatory activity. The sphinganine chain branching of BfaGCs is a critical determinant of NKT cell activation, which induces specific immunomodulatory gene expression signatures and effector functions. Co-crystal structure and affinity analyses of CD1d-BfaGC-NKT cell receptor complexes confirmed the interaction of BfaGCs as CD1d-restricted ligands. We present a structural and molecular-level paradigm of immunomodulatory control by interactions of endobiotic metabolites with diet, microbiota and the immune system.


Subject(s)
Amino Acids, Branched-Chain/immunology , Amino Acids, Branched-Chain/metabolism , Bacteroides fragilis/metabolism , Galactosylceramides/immunology , Galactosylceramides/metabolism , Gastrointestinal Microbiome/immunology , Symbiosis/immunology , Amino Acids, Branched-Chain/chemistry , Animals , Antigens, CD1d/immunology , Bacteroides fragilis/genetics , Humans , Mice , Models, Animal , Models, Molecular , Natural Killer T-Cells/cytology , Natural Killer T-Cells/immunology , Receptors, Antigen, T-Cell/immunology , Signal Transduction/immunology
15.
Nature ; 577(7791): 543-548, 2020 01.
Article in English | MEDLINE | ID: mdl-31915378

ABSTRACT

Although maternal antibodies protect newborn babies from infection1,2, little is known about how protective antibodies are induced without prior pathogen exposure. Here we show that neonatal mice that lack the capacity to produce IgG are protected from infection with the enteric pathogen enterotoxigenic Escherichia coli by maternal natural IgG antibodies against the maternal microbiota when antibodies are delivered either across the placenta or through breast milk. By challenging pups that were fostered by either maternal antibody-sufficient or antibody-deficient dams, we found that IgG derived from breast milk was crucial for protection against mucosal disease induced by enterotoxigenic E. coli. IgG also provides protection against systemic infection by E. coli. Pups used the neonatal Fc receptor to transfer IgG from milk into serum. The maternal commensal microbiota can induce antibodies that recognize antigens expressed by enterotoxigenic E. coli and other Enterobacteriaceae species. Induction of maternal antibodies against a commensal Pantoea species confers protection against enterotoxigenic E. coli in pups. This role of the microbiota in eliciting protective antibodies to a specific neonatal pathogen represents an important host defence mechanism against infection in neonates.


Subject(s)
Antibodies/immunology , Enterotoxigenic Escherichia coli/immunology , Escherichia coli Infections/immunology , Escherichia coli Infections/prevention & control , Immunity, Maternally-Acquired/immunology , Infant, Newborn/immunology , Microbiota/immunology , Milk, Human/immunology , Animals , Antibodies/blood , Antibodies/metabolism , Breast Feeding , Cross Reactions/immunology , Escherichia coli Infections/microbiology , Female , Humans , Immunoglobulin G/blood , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Male , Mice , Mothers , Pantoea/immunology , Receptors, Fc/immunology , Receptors, Fc/metabolism , Symbiosis/immunology
16.
Nature ; 577(7790): 410-415, 2020 01.
Article in English | MEDLINE | ID: mdl-31875848

ABSTRACT

The metabolic pathways encoded by the human gut microbiome constantly interact with host gene products through numerous bioactive molecules1. Primary bile acids (BAs) are synthesized within hepatocytes and released into the duodenum to facilitate absorption of lipids or fat-soluble vitamins2. Some BAs (approximately 5%) escape into the colon, where gut commensal bacteria convert them into various intestinal BAs2 that are important hormones that regulate host cholesterol metabolism and energy balance via several nuclear receptors and/or G-protein-coupled receptors3,4. These receptors have pivotal roles in shaping host innate immune responses1,5. However, the effect of this host-microorganism biliary network on the adaptive immune system remains poorly characterized. Here we report that both dietary and microbial factors influence the composition of the gut BA pool and modulate an important population of colonic FOXP3+ regulatory T (Treg) cells expressing the transcription factor RORγ. Genetic abolition of BA metabolic pathways in individual gut symbionts significantly decreases this Treg cell population. Restoration of the intestinal BA pool increases colonic RORγ+ Treg cell counts and ameliorates host susceptibility to inflammatory colitis via BA nuclear receptors. Thus, a pan-genomic biliary network interaction between hosts and their bacterial symbionts can control host immunological homeostasis via the resulting metabolites.


Subject(s)
Bile Acids and Salts/metabolism , Gastrointestinal Microbiome , Homeostasis , Intestines/immunology , Intestines/microbiology , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Bile Acids and Salts/chemistry , Mice, Inbred C57BL , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics
17.
Nature ; 554(7692): 392, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29342144

ABSTRACT

This corrects the article DOI: 10.1038/nature25019.

19.
Nature ; 552(7684): 244-247, 2017 12 14.
Article in English | MEDLINE | ID: mdl-29211710

ABSTRACT

Microbiome-wide association studies have established that numerous diseases are associated with changes in the microbiota. These studies typically generate a long list of commensals implicated as biomarkers of disease, with no clear relevance to disease pathogenesis. If the field is to move beyond correlations and begin to address causation, an effective system is needed for refining this catalogue of differentially abundant microbes and to allow subsequent mechanistic studies. Here we demonstrate that triangulation of microbe-phenotype relationships is an effective method for reducing the noise inherent in microbiota studies and enabling identification of causal microbes. We found that gnotobiotic mice harbouring different microbial communities exhibited differential survival in a colitis model. Co-housing of these mice generated animals that had hybrid microbiotas and displayed intermediate susceptibility to colitis. Mapping of microbe-phenotype relationships in parental mouse strains and in mice with hybrid microbiotas identified the bacterial family Lachnospiraceae as a correlate for protection from disease. Using directed microbial culture techniques, we discovered Clostridium immunis, a previously unknown bacterial species from this family, that-when administered to colitis-prone mice-protected them against colitis-associated death. To demonstrate the generalizability of our approach, we used it to identify several commensal organisms that induce intestinal expression of an antimicrobial peptide. Thus, we have used microbe-phenotype triangulation to move beyond the standard correlative microbiome study and identify causal microbes for two completely distinct phenotypes. Identification of disease-modulating commensals by microbe-phenotype triangulation may be more broadly applicable to human microbiome studies.


Subject(s)
Clostridium/isolation & purification , Clostridium/physiology , Colitis/microbiology , Colitis/prevention & control , Gastrointestinal Microbiome , Phenotype , Animals , Body Weight , Cell Survival , Clostridium/genetics , Feces/microbiology , Gastrointestinal Microbiome/genetics , Germ-Free Life , Intestinal Mucosa/metabolism , Intestines/microbiology , Limosilactobacillus reuteri/genetics , Limosilactobacillus reuteri/physiology , Mice , Pancreatitis-Associated Proteins/metabolism , Ruminococcus/genetics , Ruminococcus/physiology , Symbiosis
SELECTION OF CITATIONS
SEARCH DETAIL