Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Cell ; 184(16): 4154-4167.e12, 2021 08 05.
Article in English | MEDLINE | ID: mdl-34324837

ABSTRACT

Environmental light cycles entrain circadian feeding behaviors in animals that produce rhythms in exposure to foodborne bacteria. Here, we show that the intestinal microbiota generates diurnal rhythms in innate immunity that synchronize with feeding rhythms to anticipate microbial exposure. Rhythmic expression of antimicrobial proteins was driven by daily rhythms in epithelial attachment by segmented filamentous bacteria (SFB), members of the mouse intestinal microbiota. Rhythmic SFB attachment was driven by the circadian clock through control of feeding rhythms. Mechanistically, rhythmic SFB attachment activated an immunological circuit involving group 3 innate lymphoid cells. This circuit triggered oscillations in epithelial STAT3 expression and activation that produced rhythmic antimicrobial protein expression and caused resistance to Salmonella Typhimurium infection to vary across the day-night cycle. Thus, host feeding rhythms synchronize with the microbiota to promote rhythms in intestinal innate immunity that anticipate exogenous microbial exposure.


Subject(s)
Circadian Clocks/physiology , Circadian Rhythm/physiology , Gastrointestinal Microbiome , Immunity, Innate , Animals , Antimicrobial Cationic Peptides/metabolism , Bacterial Adhesion , Cell Adhesion , Epithelial Cells/microbiology , Feeding Behavior , Intestine, Small/microbiology , Intestine, Small/ultrastructure , Lymphocytes/metabolism , Mice, Inbred C57BL , Muramidase/metabolism , Pancreatitis-Associated Proteins/metabolism , STAT3 Transcription Factor/metabolism , Salmonella Infections, Animal/microbiology , Signal Transduction
2.
Nature ; 553(7687): 208-211, 2018 01 11.
Article in English | MEDLINE | ID: mdl-29323293

ABSTRACT

Inflammatory diseases of the gastrointestinal tract are frequently associated with dysbiosis, characterized by changes in gut microbial communities that include an expansion of facultative anaerobic bacteria of the Enterobacteriaceae family (phylum Proteobacteria). Here we show that a dysbiotic expansion of Enterobacteriaceae during gut inflammation could be prevented by tungstate treatment, which selectively inhibited molybdenum-cofactor-dependent microbial respiratory pathways that are operational only during episodes of inflammation. By contrast, we found that tungstate treatment caused minimal changes in the microbiota composition under homeostatic conditions. Notably, tungstate-mediated microbiota editing reduced the severity of intestinal inflammation in mouse models of colitis. We conclude that precision editing of the microbiota composition by tungstate treatment ameliorates the adverse effects of dysbiosis in the inflamed gut.


Subject(s)
Colitis/drug therapy , Colitis/microbiology , Gastrointestinal Microbiome/drug effects , Intestines/drug effects , Intestines/microbiology , Anaerobiosis/drug effects , Animals , Cell Respiration/drug effects , Dysbiosis/drug therapy , Dysbiosis/microbiology , Enterobacteriaceae/drug effects , Enterobacteriaceae/growth & development , Enterobacteriaceae/metabolism , Female , Inflammation/drug therapy , Inflammation/microbiology , Inflammation/pathology , Intestinal Mucosa/drug effects , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Intestines/pathology , Male , Mice , Mice, Inbred C57BL , Molybdenum/metabolism , Tungsten Compounds/pharmacology , Tungsten Compounds/therapeutic use
3.
Proc Natl Acad Sci U S A ; 114(7): E1196-E1204, 2017 02 14.
Article in English | MEDLINE | ID: mdl-28137874

ABSTRACT

Class-switch recombination (CSR) alters the Ig isotype to diversify antibody effector functions. IgD CSR is a rare event, and its regulation is poorly understood. We report that deficiency of 53BP1, a DNA damage-response protein, caused age-dependent overproduction of secreted IgD resulting from increased IgD CSR exclusively within B cells of mucosa-associated lymphoid tissues. IgD overproduction was dependent on activation-induced cytidine deaminase, hematopoietic MyD88 expression, and an intact microbiome, against which circulating IgD, but not IgM, was reactive. IgD CSR occurred via both alternative nonhomologous end-joining and homologous recombination pathways. Microbiota-dependent IgD CSR also was detected in nasal-associated lymphoid tissue of WT mice. These results identify a pathway, present in WT mice and hyperactivated in 53BP1-deficient mice, by which microbiota signal via Toll-like receptors to elicit IgD CSR.


Subject(s)
Immunoglobulin Class Switching , Immunoglobulin D/immunology , Lymphoid Tissue/immunology , Microbiota/immunology , Mucous Membrane/immunology , Animals , Cytidine Deaminase/genetics , Cytidine Deaminase/immunology , Cytidine Deaminase/metabolism , DNA End-Joining Repair , Female , Immunoglobulin D/genetics , Immunoglobulin D/metabolism , Lymphoid Tissue/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Microbiota/genetics , Mucous Membrane/metabolism , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/immunology , Myeloid Differentiation Factor 88/metabolism , Recombination, Genetic , Tumor Suppressor p53-Binding Protein 1/deficiency , Tumor Suppressor p53-Binding Protein 1/genetics , Tumor Suppressor p53-Binding Protein 1/immunology
4.
Genes Dev ; 26(12): 1306-11, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22713870

ABSTRACT

For all newborn mammals, mother's milk is the perfect nourishment, crucial for their postnatal development. Here we report that, unexpectedly, maternal western diet consumption in mice causes the production of toxic milk that contains excessive long chain and saturated fatty acids, which triggers ceramide accumulation and inflammation in the nursing neonates, manifested as alopecia. This neonatal toxicity requires Toll-like-receptors (TLR), but not gut microbiota, because TLR2/4 deletion or TLR4 inhibition confers resistance, whereas germ-free mice remain sensitive. These findings unravel maternal western diet-induced inflammatory milk secretion as a novel aspect of the metabolic syndrome at the maternal offspring interface.


Subject(s)
Diet/adverse effects , Inflammation/pathology , Milk/toxicity , Mothers , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism , Western World , Animals , Animals, Newborn , Ceramides/metabolism , Fatty Acids/metabolism , Female , Gene Deletion , Germ-Free Life/drug effects , Lactation/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Milk/metabolism , Pregnancy , Toll-Like Receptor 4/antagonists & inhibitors
5.
Proc Natl Acad Sci U S A ; 108(42): 17390-5, 2011 Oct 18.
Article in English | MEDLINE | ID: mdl-21960441

ABSTRACT

RIG-I-like receptors (RLRs) activate host innate immune responses against virus infection through recruiting the mitochondrial adaptor protein MAVS (also known as IPS1, VISA, or CARDIF). Here we show that MAVS also plays a pivotal role in maintaining intestinal homeostasis. We found that MAVS knockout mice developed more severe mortality and morbidity than WT animals in an experimental model of colitis. Bone marrow transplantation experiments revealed that MAVS in cells of nonhematopoietic origin plays a dominant role in the protection against colitis. Importantly, RNA species derived from intestinal commensal bacteria activate the RIG-I-MAVS pathway to induce the production of multiple cytokines and antimicrobial peptides, including IFN-ß and RegIIIγ. These results unveil a previously unexplored role of MAVS in monitoring intestinal commensal bacteria and maintaining tissue homeostasis.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Colitis/immunology , Colitis/prevention & control , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Animals , Bone Marrow Transplantation/immunology , Colitis/chemically induced , DEAD Box Protein 58 , DEAD-box RNA Helicases/immunology , Dextran Sulfate/toxicity , Disease Models, Animal , Female , Immunity, Innate , Intestines/immunology , Intestines/microbiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/deficiency , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/immunology , RNA, Bacterial/immunology
6.
Proc Natl Acad Sci U S A ; 108(21): 8743-8, 2011 May 24.
Article in English | MEDLINE | ID: mdl-21555560

ABSTRACT

The mammalian gastrointestinal tract harbors thousands of bacterial species that include symbionts as well as potential pathogens. The immune responses that limit access of these bacteria to underlying tissue remain poorly defined. Here we show that γδ intraepithelial lymphocytes (γδ IEL) of the small intestine produce innate antimicrobial factors in response to resident bacterial "pathobionts" that penetrate the intestinal epithelium. γδ IEL activation was dependent on epithelial cell-intrinsic MyD88, suggesting that epithelial cells supply microbe-dependent cues to γδ IEL. Finally, γδ T cells protect against invasion of intestinal tissues by resident bacteria specifically during the first few hours after bacterial encounter, indicating that γδ IEL occupy a unique temporal niche among intestinal immune defenses. Thus, γδ IEL detect the presence of invading bacteria through cross-talk with neighboring epithelial cells and are an essential component of the hierarchy of immune defenses that maintain homeostasis with the intestinal microbiota.


Subject(s)
Homeostasis/immunology , Host-Pathogen Interactions/immunology , Intestinal Mucosa/immunology , Lymphocytes/immunology , Receptors, Antigen, T-Cell, gamma-delta/physiology , Animals , Bacteria/immunology , Cell Communication/immunology , Epithelial Cells , Immunity, Innate , Metagenome/immunology , Mice , Mice, Knockout
7.
Science ; 381(6660): 851-857, 2023 08 25.
Article in English | MEDLINE | ID: mdl-37616368

ABSTRACT

The intestinal microbiota regulates mammalian lipid absorption, metabolism, and storage. We report that the microbiota reprograms intestinal lipid metabolism in mice by repressing the expression of long noncoding RNA (lncRNA) Snhg9 (small nucleolar RNA host gene 9) in small intestinal epithelial cells. Snhg9 suppressed the activity of peroxisome proliferator-activated receptor γ (PPARγ)-a central regulator of lipid metabolism-by dissociating the PPARγ inhibitor sirtuin 1 from cell cycle and apoptosis protein 2 (CCAR2). Forced expression of Snhg9 in the intestinal epithelium of conventional mice impaired lipid absorption, reduced body fat, and protected against diet-induced obesity. The microbiota repressed Snhg9 expression through an immune relay encompassing myeloid cells and group 3 innate lymphoid cells. Our findings thus identify an unanticipated role for a lncRNA in microbial control of host metabolism.


Subject(s)
Gastrointestinal Microbiome , Intestines , Lipid Metabolism , PPAR gamma , RNA, Long Noncoding , Sirtuin 1 , Animals , Mice , Immunity, Innate , Lipid Metabolism/genetics , Lymphocytes/immunology , PPAR gamma/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Sirtuin 1/metabolism , Cell Cycle Proteins/metabolism , Apoptosis Regulatory Proteins/metabolism , Myeloid Cells/immunology , Intestines/metabolism , Intestines/microbiology , Adipose Tissue/microbiology , Humans
8.
J Immunol ; 182(5): 3047-54, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-19234201

ABSTRACT

The intestinal mucosal surface is in direct contact with a vast beneficial microbiota. The symbiotic nature of this relationship is threatened when the surface epithelium is injured, yet little is known about how mucosal surfaces maintain homeostasis with commensal microbes following damage. Gammadelta intraepithelial lymphocytes (gammadelta IEL) reside at the gut epithelial surface, where they stimulate mucosal healing following acute injury. A genome-wide analysis of the gammadelta IEL response to dextran sulfate sodium-induced colonic damage revealed induction of a complex transcriptional program, including coordinate regulation of cytoprotective, immunomodulatory, and antibacterial factors. Studies in germfree mice demonstrated that commensal microbiota regulate key components of this transcriptional program, thus revealing a dialogue between commensal bacteria and gammadelta IEL in injured epithelia. Analysis of TCRdelta-deficient mice indicated that gammadelta T cells are essential for controlling mucosal penetration of commensal bacteria immediately following dextran sulfate sodium-induced damage, suggesting that a key function of gammadelta IEL is to maintain host-microbial homeostasis following acute mucosal injury. Taken together, these findings disclose a reciprocal relationship between gammadelta T cells and intestinal microbiota that promotes beneficial host-microbial relationships in the intestine.


Subject(s)
Cell Communication/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Lymphocyte Subsets/immunology , Lymphocyte Subsets/microbiology , Receptors, Antigen, T-Cell, gamma-delta/biosynthesis , Animals , Colon/drug effects , Colon/immunology , Colon/microbiology , Colon/pathology , Dextran Sulfate/toxicity , Epithelial Cells/drug effects , Epithelial Cells/immunology , Epithelial Cells/microbiology , Epithelial Cells/pathology , Homeostasis/drug effects , Homeostasis/immunology , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Lymphocyte Subsets/drug effects , Lymphocyte Subsets/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Receptors, Antigen, T-Cell, gamma-delta/deficiency , Receptors, Antigen, T-Cell, gamma-delta/genetics
9.
Proc Natl Acad Sci U S A ; 105(52): 20858-63, 2008 Dec 30.
Article in English | MEDLINE | ID: mdl-19075245

ABSTRACT

The intestinal epithelium is in direct contact with a vast microbiota, yet little is known about how epithelial cells defend the host against the heavy bacterial load. To address this question we studied Paneth cells, a key small intestinal epithelial lineage. We found that Paneth cells directly sense enteric bacteria through cell-autonomous MyD88-dependent toll-like receptor (TLR) activation, triggering expression of multiple antimicrobial factors. Paneth cells were essential for controlling intestinal barrier penetration by commensal and pathogenic bacteria. Furthermore, Paneth cell-intrinsic MyD88 signaling limited bacterial penetration of host tissues, revealing a role for epithelial MyD88 in maintaining intestinal homeostasis. Our findings establish that gut epithelia actively sense enteric bacteria and play an essential role in maintaining host-microbial homeostasis at the mucosal interface.


Subject(s)
Bacteria/immunology , Bacterial Translocation/immunology , Host-Pathogen Interactions/immunology , Myeloid Differentiation Factor 88/immunology , Paneth Cells/immunology , Toll-Like Receptors/immunology , Animals , Anti-Infective Agents/immunology , Homeostasis/immunology , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Paneth Cells/microbiology , Toll-Like Receptors/genetics
10.
Science ; 374(6568): eabe6723, 2021 Nov 05.
Article in English | MEDLINE | ID: mdl-34735226

ABSTRACT

A diverse group of antimicrobial proteins (AMPs) helps protect the mammalian intestine from varied microbial challenges. We show that small proline-rich protein 2A (SPRR2A) is an intestinal antibacterial protein that is phylogenetically unrelated to previously discovered mammalian AMPs. In this study, SPRR2A was expressed in Paneth cells and goblet cells and selectively killed Gram-positive bacteria by disrupting their membranes. SPRR2A shaped intestinal microbiota composition, restricted bacterial association with the intestinal surface, and protected against Listeria monocytogenes infection. SPRR2A differed from other intestinal AMPs in that it was induced by type 2 cytokines produced during helminth infection. Moreover, SPRR2A protected against helminth-induced bacterial invasion of intestinal tissue. Thus, SPRR2A is a distinctive AMP triggered by type 2 immunity that protects the intestinal barrier during helminth infection.


Subject(s)
Cornified Envelope Proline-Rich Proteins/metabolism , Gastrointestinal Microbiome , Gram-Positive Bacteria/physiology , Intestinal Mucosa/metabolism , Intestines/microbiology , Nematospiroides dubius , Strongylida Infections/immunology , Animals , Bacterial Load , Cell Membrane/metabolism , Cell Membrane Permeability , Cornified Envelope Proline-Rich Proteins/genetics , Cytokines/metabolism , Disease Susceptibility , Goblet Cells/metabolism , Humans , Immunity, Innate , Intestinal Mucosa/microbiology , Listeria monocytogenes/physiology , Listeriosis/microbiology , Mice , Microbial Viability , Paneth Cells/metabolism , Pore Forming Cytotoxic Proteins/genetics , Pore Forming Cytotoxic Proteins/metabolism , Strongylida Infections/metabolism , Strongylida Infections/microbiology
11.
Cell Host Microbe ; 27(3): 376-388.e8, 2020 03 11.
Article in English | MEDLINE | ID: mdl-32075741

ABSTRACT

During short-lived perturbations, such as inflammation, the gut microbiota exhibits resilience and reverts to its original configuration. Although microbial access to the micronutrient iron is decreased during colitis, pathogens can scavenge iron by using siderophores. How commensal bacteria acquire iron during gut inflammation is incompletely understood. Curiously, the human commensal Bacteroides thetaiotaomicron does not produce siderophores but grows under iron-limiting conditions using enterobacterial siderophores. Using RNA-seq, we identify B. thetaiotaomicron genes that were upregulated during Salmonella-induced gut inflammation and were predicted to be involved in iron uptake. Mutants in the xusABC locus (BT2063-2065) were defective for xenosiderophore-mediated iron uptake in vitro. In the normal mouse gut, the XusABC system was dispensable, while a xusA mutant colonized poorly during colitis. This work identifies xenosiderophore utilization as a critical mechanism for B. thetaiotaomicron to sustain colonization during inflammation and suggests a mechanism of how interphylum iron metabolism contributes to gut microbiota resilience.


Subject(s)
Bacteroides thetaiotaomicron/metabolism , Colitis/microbiology , Enterobacteriaceae/genetics , Gastrointestinal Microbiome , Iron/metabolism , Siderophores/genetics , Animals , Bacteroides thetaiotaomicron/genetics , Female , Genes, Bacterial , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA-Seq , Symbiosis
12.
Science ; 365(6460): 1428-1434, 2019 09 27.
Article in English | MEDLINE | ID: mdl-31604271

ABSTRACT

Circadian rhythmicity is a defining feature of mammalian metabolism that synchronizes metabolic processes to day-night light cycles. Here, we show that the intestinal microbiota programs diurnal metabolic rhythms in the mouse small intestine through histone deacetylase 3 (HDAC3). The microbiota induced expression of intestinal epithelial HDAC3, which was recruited rhythmically to chromatin, and produced synchronized diurnal oscillations in histone acetylation, metabolic gene expression, and nutrient uptake. HDAC3 also functioned noncanonically to coactivate estrogen-related receptor α, inducing microbiota-dependent rhythmic transcription of the lipid transporter gene Cd36 and promoting lipid absorption and diet-induced obesity. Our findings reveal that HDAC3 integrates microbial and circadian cues for regulation of diurnal metabolic rhythms and pinpoint a key mechanism by which the microbiota controls host metabolism.


Subject(s)
Circadian Rhythm , Epithelial Cells/metabolism , Gastrointestinal Microbiome , Histone Deacetylases/metabolism , Intestine, Small/metabolism , Acetylation , Animals , CD36 Antigens/metabolism , Chromatin/metabolism , Colon , Diet, High-Fat , Germ-Free Life , Intestine, Small/cytology , Jet Lag Syndrome , Lipid Metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/metabolism , Receptors, Estrogen/metabolism , ERRalpha Estrogen-Related Receptor
13.
Cell Host Microbe ; 23(1): 54-64.e6, 2018 Jan 10.
Article in English | MEDLINE | ID: mdl-29276172

ABSTRACT

During Salmonella-induced gastroenteritis, mucosal inflammation creates a niche that favors the expansion of the pathogen population over the microbiota. Here, we show that Salmonella Typhimurium infection was accompanied by dysbiosis, decreased butyrate levels, and substantially elevated lactate levels in the gut lumen. Administration of a lactate dehydrogenase inhibitor blunted lactate production in germ-free mice, suggesting that lactate was predominantly of host origin. Depletion of butyrate-producing Clostridia, either through oral antibiotic treatment or as part of the pathogen-induced dysbiosis, triggered a switch in host cells from oxidative metabolism to lactate fermentation, increasing both lactate levels and Salmonella lactate utilization. Administration of tributyrin or a PPARγ agonist diminished host lactate production and abrogated the fitness advantage conferred on Salmonella by lactate utilization. We conclude that alterations of the gut microbiota, specifically a depletion of Clostridia, reprogram host metabolism to perform lactate fermentation, thus supporting Salmonella infection.


Subject(s)
Clostridium/growth & development , Dysbiosis/microbiology , Gastroenteritis/microbiology , Gastrointestinal Microbiome/drug effects , Intestinal Mucosa/microbiology , Lactic Acid/metabolism , Salmonella typhimurium/metabolism , Animals , Anti-Bacterial Agents/pharmacology , Butyric Acid/metabolism , Female , Fermentation , Gastroenteritis/pathology , L-Lactate Dehydrogenase/antagonists & inhibitors , Male , Mice , Mice, Inbred C57BL , PPAR gamma/agonists , Salmonella Infections/pathology , Salmonella typhimurium/growth & development , Triglycerides/pharmacology
14.
Cell Host Microbe ; 22(3): 291-301.e6, 2017 Sep 13.
Article in English | MEDLINE | ID: mdl-28844888

ABSTRACT

The mucosal inflammatory response induced by Salmonella serovar Typhimurium creates a favorable niche for this gut pathogen. Conventional wisdom holds that S. Typhimurium undergoes an incomplete tricarboxylic acid (TCA) cycle in the anaerobic mammalian gut. One change during S. Typhimurium-induced inflammation is the production of oxidized compounds by infiltrating neutrophils. We show that inflammation-derived electron acceptors induce a complete, oxidative TCA cycle in S. Typhimurium, allowing the bacteria to compete with the microbiota for colonization. A complete TCA cycle facilitates utilization of the microbiota-derived fermentation product succinate as a carbon source. S. Typhimurium succinate utilization genes contribute to efficient colonization in conventionally raised mice, but provide no growth advantage in germ-free mice. Mono-association of gnotobiotic mice with Bacteroides, a major succinate producer, restores succinate utilization in S. Typhimurium. Thus, oxidative central metabolism enables S. Typhimurium to utilize a variety of carbon sources, including microbiota-derived succinate.


Subject(s)
Bacteria/metabolism , Bacteroides/metabolism , Colitis/microbiology , Gastrointestinal Microbiome , Salmonella Infections/microbiology , Salmonella typhimurium/metabolism , Succinic Acid/metabolism , Animals , Bacteria/genetics , Bacteria/isolation & purification , Bacteroides/genetics , Bacteroides/isolation & purification , Citric Acid Cycle , Colitis/metabolism , Humans , Intestinal Mucosa/metabolism , Intestines/microbiology , Mice , Mice, Inbred CBA , Oxidative Stress , Salmonella Infections/metabolism , Salmonella typhimurium/genetics
15.
Cell Host Microbe ; 21(2): 208-219, 2017 Feb 08.
Article in English | MEDLINE | ID: mdl-28182951

ABSTRACT

Intestinal inflammation is frequently associated with an alteration of the gut microbiota, termed dysbiosis, which is characterized by a reduced abundance of obligate anaerobic bacteria and an expansion of facultative Proteobacteria such as commensal E. coli. The mechanisms enabling the outgrowth of Proteobacteria during inflammation are incompletely understood. Metagenomic sequencing revealed bacterial formate oxidation and aerobic respiration to be overrepresented metabolic pathways in a chemically induced murine model of colitis. Dysbiosis was accompanied by increased formate levels in the gut lumen. Formate was of microbial origin since no formate was detected in germ-free mice. Complementary studies using commensal E. coli strains as model organisms indicated that formate dehydrogenase and terminal oxidase genes provided a fitness advantage in murine models of colitis. In vivo, formate served as electron donor in conjunction with oxygen as the terminal electron acceptor. This work identifies bacterial formate oxidation and oxygen respiration as metabolic signatures for inflammation-associated dysbiosis.


Subject(s)
Dysbiosis/microbiology , Escherichia coli/metabolism , Formates/metabolism , Inflammation/microbiology , Animals , Colitis/microbiology , Disease Models, Animal , Female , Gastrointestinal Microbiome , Intestines/microbiology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Proteobacteria/metabolism
18.
Cell Host Microbe ; 6(2): 187-96, 2009 Aug 20.
Article in English | MEDLINE | ID: mdl-19683684

ABSTRACT

Toxoplasma gondii is a universally distributed pathogen that infects over one billion people worldwide. Host resistance to this protozoan parasite depends on a Th1 immune response with potent production of the cytokines interleukin-12 and interferon gamma. Although Toll-like receptor 11 (TLR11) plays a major role in controlling Th1 immunity to this pathogen in mice, this innate immune receptor is nonfunctional in humans, and the mechanisms of TLR11-independent sensing of T. gondii remain elusive. Here, we show that oral infection by T. gondii triggers a TLR11-independent but MyD88-dependent Th1 response that is impaired in TLR2xTLR4 double knockout and TLR9 single knockout mice. These mucosal innate and adaptive immune responses to T. gondii rely on the indirect stimulation of dendritic cells by normal gut microflora. Thus, our results reveal that gut commensal bacteria can serve as molecular adjuvants during parasitic infection, providing indirect immunostimulation that protects against T. gondii in the absence of TLR11.


Subject(s)
Bacteria/immunology , Dendritic Cells/immunology , Gastrointestinal Tract/microbiology , Toxoplasma/immunology , Toxoplasmosis, Animal/immunology , Animals , Cytokines/biosynthesis , Humans , Leukocytes, Mononuclear/immunology , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/deficiency , Myeloid Differentiation Factor 88/immunology , Toll-Like Receptor 2/deficiency , Toll-Like Receptor 2/immunology , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/immunology , Toll-Like Receptor 9/deficiency , Toll-Like Receptor 9/immunology , Toxoplasmosis, Animal/parasitology , Toxoplasmosis, Animal/pathology
19.
Science ; 313(5790): 1126-30, 2006 Aug 25.
Article in English | MEDLINE | ID: mdl-16931762

ABSTRACT

The mammalian intestine harbors complex societies of beneficial bacteria that are maintained in the lumen with minimal penetration of mucosal surfaces. Microbial colonization of germ-free mice triggers epithelial expression of RegIIIgamma, a secreted C-type lectin. RegIIIgamma binds intestinal bacteria but lacks the complement recruitment domains present in other microbe-binding mammalian C-type lectins. We show that RegIIIgamma and its human counterpart, HIP/PAP, are directly antimicrobial proteins that bind their bacterial targets via interactions with peptidoglycan carbohydrate. We propose that these proteins represent an evolutionarily primitive form of lectin-mediated innate immunity, and that they reveal intestinal strategies for maintaining symbiotic host-microbial relationships.


Subject(s)
Antigens, Neoplasm/metabolism , Bacteria/immunology , Biomarkers, Tumor/metabolism , Immunity, Innate , Intestine, Small/microbiology , Lectins, C-Type/metabolism , Paneth Cells/metabolism , Peptidoglycan/metabolism , Proteins/metabolism , Animals , Antigens, Neoplasm/pharmacology , Bacteria/growth & development , Biomarkers, Tumor/pharmacology , Chitin/metabolism , Colony Count, Microbial , Germ-Free Life , Gram-Positive Bacteria/immunology , Gram-Positive Bacteria/metabolism , Homeostasis , Humans , Immunity, Mucosal , Ligands , Listeria monocytogenes/ultrastructure , Mice , Oligonucleotide Array Sequence Analysis , Pancreatitis-Associated Proteins , Paneth Cells/immunology , Peptidoglycan/chemistry , Protein Structure, Tertiary , Proteins/genetics , Proteins/pharmacology , Recombinant Proteins/metabolism , Secretory Vesicles/metabolism , Symbiosis
SELECTION OF CITATIONS
SEARCH DETAIL