Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 97
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.
Nat Immunol ; 24(3): 531-544, 2023 03.
Article in English | MEDLINE | ID: mdl-36658240

ABSTRACT

Immunoglobulin A (IgA) secretion by plasma cells, terminally differentiated B cells residing in the intestinal lamina propria, assures microbiome homeostasis and protects the host against enteric infections. Exposure to diet-derived and commensal-derived signals provides immune cells with organizing cues that instruct their effector function and dynamically shape intestinal immune responses at the mucosal barrier. Recent data have described metabolic and microbial inputs controlling T cell and innate lymphoid cell activation in the gut; however, whether IgA-secreting lamina propria plasma cells are tuned by local stimuli is completely unknown. Although antibody secretion is considered to be imprinted during B cell differentiation and therefore largely unaffected by environmental changes, a rapid modulation of IgA levels in response to intestinal fluctuations might be beneficial to the host. In the present study, we showed that dietary cholesterol absorption and commensal recognition by duodenal intestinal epithelial cells lead to the production of oxysterols, evolutionarily conserved lipids with immunomodulatory functions. Using conditional cholesterol 25-hydroxylase deleter mouse line we demonstrated that 7α,25-dihydroxycholesterol from epithelial cells is critical to restrain IgA secretion against commensal- and pathogen-derived antigens in the gut. Intestinal plasma cells sense oxysterols via the chemoattractant receptor GPR183 and couple their tissue positioning with IgA secretion. Our findings revealed a new mechanism linking dietary cholesterol and humoral immune responses centered around plasma cell localization for efficient mucosal protection.


Subject(s)
Immunity, Innate , Plasma Cells , Animals , Mice , Cholesterol, Dietary , Epithelial Cells , Immunoglobulin A , Intestinal Mucosa , Receptors, G-Protein-Coupled , Intestines
3.
Cell ; 147(3): 489-91, 2011 Oct 28.
Article in English | MEDLINE | ID: mdl-22036556

ABSTRACT

The aryl hydrocarbon receptor (AhR) is responsible for the toxic effects of environmental pollutants such as dioxin, but little is known about its normal physiological functions. Li et al. (2011) now show that specific dietary compounds present in cruciferous vegetables act through the AhR to promote intestinal immune function, revealing AhR as a critical link between diet and immunity.

4.
PLoS Pathog ; 19(10): e1011691, 2023 10.
Article in English | MEDLINE | ID: mdl-37847677

ABSTRACT

Even though gammaherpesvirus and parasitic infections are endemic in parts of the world, there is a lack of understanding about the outcome of coinfection. In humans, coinfections usually occur sequentially, with fluctuating order and timing in different hosts. However, experimental studies in mice generally do not address the variables of order and timing of coinfections. We sought to examine the variable of coinfection order in a system of gammaherpesvirus-helminth coinfection. Our previous work demonstrated that infection with the intestinal parasite, Heligmosomoides polygyrus, induced transient reactivation from latency of murine gammaherpesvirus-68 (MHV68). In this report, we reverse the order of coinfection, infecting with H. polygyrus first, followed by MHV68, and examined the effects of preexisting parasite infection on MHV68 acute and latent infection. We found that preexisting parasite infection increased the propensity of MHV68 to reactivate from latency. However, when we examined the mechanism for reactivation, we found that preexisting parasite infection increased the ability of MHV68 to reactivate in a vitamin A dependent manner, a distinct mechanism to what we found previously with parasite-induced reactivation after latency establishment. We determined that H. polygyrus infection increased both acute and latent MHV68 infection in a population of tissue resident macrophages, called large peritoneal macrophages. We demonstrate that this population of macrophages and vitamin A are required for increased acute and latent infection during parasite coinfection.


Subject(s)
Coinfection , Gammaherpesvirinae , Helminths , Herpesviridae Infections , Latent Infection , Parasitic Diseases , Humans , Animals , Mice , Virus Activation , Virus Latency/physiology , Vitamin A , B-Lymphocytes , Herpesviridae Infections/complications , Gammaherpesvirinae/physiology , Macrophages , Mice, Inbred C57BL
5.
Nat Immunol ; 14(7): 654-9, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23778792

ABSTRACT

The human body is colonized with a diverse resident microflora that includes viruses. Recent studies of metagenomes have begun to characterize the composition of the human 'virobiota' and its associated genes (the 'virome'), and have fostered the emerging field of host-virobiota interactions. In this Perspective, we explore how resident viruses interact with the immune system. We review recent findings that highlight the role of the immune system in shaping the composition of the virobiota and consider how resident viruses may impact host immunity. Finally, we discuss the implications of virobiota-immune system interactions for human health.


Subject(s)
Gastrointestinal Tract/virology , Mouth/virology , Nasal Cavity/virology , Respiratory System/virology , Skin/virology , Viruses/immunology , Gastrointestinal Tract/immunology , Host-Pathogen Interactions/immunology , Humans , Mouth/immunology , Nasal Cavity/immunology , Respiratory System/immunology , Skin/immunology , Viruses/genetics
6.
Nat Immunol ; 14(2): 101-5, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23334821

ABSTRACT

Leading scientists working on the microbiome gathered in an October 2012 meeting in Baeza, Spain, to discuss recent advances in the understanding of the role of the microbiota in immunity, pathogen colonization, metabolism and disease.


Subject(s)
Immunity , Metagenome/immunology , Animals , Bacteria/growth & development , Host-Pathogen Interactions , Humans , Spain
7.
Nat Immunol ; 14(2): 136-42, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23263554

ABSTRACT

Activation of Toll-like receptors (TLRs) by pathogens triggers cytokine production and T cell activation, immune defense mechanisms that are linked to immunopathology. Here we show that IFN-γ production by CD4(+) T(H)1 cells during mucosal responses to the protozoan parasite Toxoplasma gondii resulted in dysbiosis and the elimination of Paneth cells. Paneth cell death led to loss of antimicrobial peptides and occurred in conjunction with uncontrolled expansion of the Enterobacteriaceae family of Gram-negative bacteria. The expanded intestinal bacteria were required for the parasite-induced intestinal pathology. The investigation of cell type-specific factors regulating T(H)1 polarization during T. gondii infection identified the T cell-intrinsic TLR pathway as a major regulator of IFN-γ production in CD4(+) T cells responsible for Paneth cell death, dysbiosis and intestinal immunopathology.


Subject(s)
Enterobacteriaceae Infections/pathology , Enterobacteriaceae/growth & development , Paneth Cells/pathology , Signal Transduction/immunology , Th1 Cells/pathology , Toxoplasma/growth & development , Toxoplasmosis, Animal/pathology , Animals , CD4-Positive T-Lymphocytes , Cell Death , Enterobacteriaceae/immunology , Enterobacteriaceae Infections/complications , Enterobacteriaceae Infections/immunology , Enterobacteriaceae Infections/microbiology , Gene Expression Regulation , Host-Parasite Interactions , Host-Pathogen Interactions , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-12/genetics , Interleukin-12/immunology , Lymphocyte Activation , Mice , Mice, Transgenic , Paneth Cells/microbiology , Paneth Cells/parasitology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Th1 Cells/microbiology , Th1 Cells/parasitology , Toxoplasma/immunology , Toxoplasmosis, Animal/complications , Toxoplasmosis, Animal/immunology , Toxoplasmosis, Animal/parasitology , alpha-Defensins/deficiency
8.
Immunity ; 42(1): 28-39, 2015 Jan 20.
Article in English | MEDLINE | ID: mdl-25607457

ABSTRACT

The mammalian gastrointestinal tract is home to a dense community of resident bacteria and is also exposed to microorganisms from the external environment. The epithelial surface of the intestine plays a critical role in host protection by producing a diverse repertoire of antimicrobial proteins that directly kill or hinder the growth of microorganisms. Here we discuss the general principles that govern the mechanisms of action of epithelial antimicrobial proteins, regulation of antimicrobial protein expression and activity, and in vivo functions of intestinal antimicrobial proteins. We also consider how altered antimicrobial protein expression and function can contribute to disease and how these endogenous antibiotics might be harnessed for the benefit of human health.


Subject(s)
Anti-Infective Agents/metabolism , Antimicrobial Cationic Peptides/metabolism , Intestinal Mucosa/immunology , Intestines/immunology , Animals , Anti-Bacterial Agents , Anti-Infective Agents/immunology , Antimicrobial Cationic Peptides/genetics , Antimicrobial Cationic Peptides/immunology , Gene Expression Regulation , Humans , Intestines/microbiology
9.
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
10.
J Biol Chem ; 298(2): 101463, 2022 02.
Article in English | MEDLINE | ID: mdl-34864058

ABSTRACT

Interleukin (IL)-22 is a cytokine that plays a critical role in intestinal epithelial homeostasis. Its downstream functions are mediated through interaction with the heterodimeric IL-22 receptor and subsequent activation of signal transducer and activator of transcription 3 (STAT3). IL-22 signaling can induce transcription of genes necessary for intestinal epithelial cell proliferation, tissue regeneration, tight junction fortification, and antimicrobial production. Recent studies have also implicated IL-22 signaling in the regulation of intestinal epithelial fucosylation in mice. However, whether IL-22 regulates intestinal fucosylation in human intestinal epithelial cells and the molecular mechanisms that govern this process are unknown. Here, in experiments performed in human cell lines and human-derived enteroids, we show that IL-22 signaling regulates expression of the B3GNT7 transcript, which encodes a ß1-3-N-acetylglucosaminyltransferase that can participate in the synthesis of poly-N-acetyllactosamine (polyLacNAc) chains. Additionally, we find that IL-22 signaling regulates levels of the α1-3-fucosylated Lewis X (Lex) blood group antigen, and that this glycan epitope is primarily displayed on O-glycosylated intestinal epithelial glycoproteins. Moreover, we show that increased expression of B3GNT7 alone is sufficient to promote increased display of Lex-decorated carbohydrate glycan structures primarily on O-glycosylated intestinal epithelial glycoproteins. Together, these data identify B3GNT7 as an intermediary in IL-22-dependent induction of fucosylation of glycoproteins and uncover a novel role for B3GNT7 in intestinal glycosylation.


Subject(s)
Epithelial Cells , Glycoproteins , Interleukins , Intestinal Mucosa , N-Acetylglucosaminyltransferases , Epithelial Cells/metabolism , Glycoproteins/metabolism , Glycosylation , Humans , Interleukins/genetics , Interleukins/metabolism , Intestinal Mucosa/metabolism , N-Acetylglucosaminyltransferases/biosynthesis , N-Acetylglucosaminyltransferases/metabolism , Polysaccharides/metabolism , Interleukin-22
11.
Proc Natl Acad Sci U S A ; 116(38): 19077-19082, 2019 09 17.
Article in English | MEDLINE | ID: mdl-31484771

ABSTRACT

Serum amyloid A (SAA) proteins are strongly induced in the liver by systemic infection and in the intestine by bacterial colonization. In infected mice, SAA proteins circulate in association with the vitamin A derivative retinol, suggesting that SAAs transport retinol during infection. Here we illuminate a structural basis for the retinol-SAA interaction. In the bloodstream of infected mice, most SAA is complexed with high-density lipoprotein (HDL). However, we found that the majority of the circulating retinol was associated with the small fraction of SAA proteins that circulate without binding to HDL, thus identifying free SAA as the predominant retinol-binding form in vivo. We then determined the crystal structure of retinol-bound mouse SAA3 at a resolution of 2.2 Å. Retinol-bound SAA3 formed a novel asymmetric trimeric assembly that was generated by the hydrophobic packing of the conserved amphipathic helices α1 and α3. This hydrophobic packing created a retinol-binding pocket in the center of the trimer, which was confirmed by mutagenesis studies. Together, these findings illuminate the molecular basis for retinol transport by SAA proteins during infection.


Subject(s)
Salmonella typhimurium/metabolism , Serum Amyloid A Protein/chemistry , Serum Amyloid A Protein/metabolism , Typhoid Fever/metabolism , Vitamin A/metabolism , Vitamins/metabolism , Animals , Crystallography, X-Ray , Mice , Mice, Knockout , Models, Molecular , Mutation , Protein Conformation , Serum Amyloid A Protein/genetics , Typhoid Fever/virology
12.
Proc Natl Acad Sci U S A ; 116(22): 10911-10916, 2019 05 28.
Article in English | MEDLINE | ID: mdl-31097581

ABSTRACT

Vitamin A is a dietary component that is essential for the development of intestinal immunity. Vitamin A is absorbed and converted to its bioactive derivatives retinol and retinoic acid by the intestinal epithelium, yet little is known about how epithelial cells regulate vitamin A-dependent intestinal immunity. Here we show that epithelial cell expression of the transcription factor retinoic acid receptor ß (RARß) is essential for vitamin A-dependent intestinal immunity. Epithelial RARß activated vitamin A-dependent expression of serum amyloid A (SAA) proteins by binding directly to Saa promoters. In accordance with the known role of SAAs in regulating Th17 cell effector function, epithelial RARß promoted IL-17 production by intestinal Th17 cells. More broadly, epithelial RARß was required for the development of key vitamin A-dependent adaptive immune responses, including CD4+ T-cell homing to the intestine and the development of IgA-producing intestinal B cells. Our findings provide insight into how the intestinal epithelium senses dietary vitamin A status to regulate adaptive immunity, and highlight the role of epithelial cells in regulating intestinal immunity in response to diet.


Subject(s)
Immunity, Mucosal/physiology , Intestinal Mucosa/metabolism , Receptors, Retinoic Acid/metabolism , Serum Amyloid A Protein/metabolism , Vitamin A/metabolism , Animals , Cell Line , Gastrointestinal Microbiome/physiology , Hep G2 Cells , Humans , Mice , Receptors, Retinoic Acid/genetics , Serum Amyloid A Protein/genetics
13.
Immunity ; 34(3): 290-2, 2011 Mar 25.
Article in English | MEDLINE | ID: mdl-21435583

ABSTRACT

In this issue of Immunity, Hall et al. (2011) show that vitamin A and its metabolites play a central role in regulating adaptive immunity by promoting the development of both inflammatory and regulatory T cell responses.

14.
Nature ; 505(7481): 103-7, 2014 Jan 02.
Article in English | MEDLINE | ID: mdl-24256734

ABSTRACT

Human body-surface epithelia coexist in close association with complex bacterial communities and are protected by a variety of antibacterial proteins. C-type lectins of the RegIII family are bactericidal proteins that limit direct contact between bacteria and the intestinal epithelium and thus promote tolerance to the intestinal microbiota. RegIII lectins recognize their bacterial targets by binding peptidoglycan carbohydrate, but the mechanism by which they kill bacteria is unknown. Here we elucidate the mechanistic basis for RegIII bactericidal activity. We show that human RegIIIα (also known as HIP/PAP) binds membrane phospholipids and kills bacteria by forming a hexameric membrane-permeabilizing oligomeric pore. We derive a three-dimensional model of the RegIIIα pore by docking the RegIIIα crystal structure into a cryo-electron microscopic map of the pore complex, and show that the model accords with experimentally determined properties of the pore. Lipopolysaccharide inhibits RegIIIα pore-forming activity, explaining why RegIIIα is bactericidal for Gram-positive but not Gram-negative bacteria. Our findings identify C-type lectins as mediators of membrane attack in the mucosal immune system, and provide detailed insight into an antibacterial mechanism that promotes mutualism with the resident microbiota.


Subject(s)
Anti-Bacterial Agents/metabolism , Antigens, Neoplasm/metabolism , Biomarkers, Tumor/metabolism , Intestines/chemistry , Lectins, C-Type/metabolism , Porins/metabolism , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/immunology , Anti-Bacterial Agents/pharmacology , Antigens, Neoplasm/chemistry , Antigens, Neoplasm/immunology , Biomarkers, Tumor/antagonists & inhibitors , Biomarkers, Tumor/chemistry , Biomarkers, Tumor/immunology , Cell Membrane Permeability/drug effects , Cryoelectron Microscopy , Crystallography, X-Ray , Gram-Negative Bacteria/drug effects , Gram-Negative Bacteria/immunology , Gram-Negative Bacteria/metabolism , Humans , Immunity, Mucosal/drug effects , Immunity, Mucosal/immunology , Intestines/immunology , Intestines/microbiology , Lectins, C-Type/antagonists & inhibitors , Lectins, C-Type/chemistry , Lectins, C-Type/immunology , Lipopolysaccharides/pharmacology , Listeria monocytogenes/drug effects , Listeria monocytogenes/immunology , Listeria monocytogenes/metabolism , Microbial Viability/drug effects , Models, Molecular , Pancreatitis-Associated Proteins , Peptidoglycan/metabolism , Phospholipids/metabolism , Porins/antagonists & inhibitors , Porins/chemistry , Symbiosis
15.
Proc Natl Acad Sci U S A ; 114(42): 11027-11033, 2017 10 17.
Article in English | MEDLINE | ID: mdl-28973871

ABSTRACT

The mammalian intestine is colonized by trillions of bacteria that perform essential metabolic functions for their hosts. The mutualistic nature of this relationship depends on maintaining spatial segregation between these bacteria and the intestinal epithelial surface. This segregation is achieved in part by the presence of a dense mucus layer at the epithelial surface and by the production of antimicrobial proteins that are secreted by epithelial cells into the mucus layer. Here, we show that resistin-like molecule ß (RELMß) is a bactericidal protein that limits contact between Gram-negative bacteria and the colonic epithelial surface. Mouse and human RELMß selectively killed Gram-negative bacteria by forming size-selective pores that permeabilized bacterial membranes. In mice lacking RELMß, Proteobacteria were present in the inner mucus layer and invaded mucosal tissues. Another RELM family member, human resistin, was also bactericidal, suggesting that bactericidal activity is a conserved function of the RELM family. Our findings thus identify the RELM family as a unique family of bactericidal proteins and show that RELMß promotes host-bacterial mutualism by regulating the spatial segregation between the microbiota and the intestinal epithelium.


Subject(s)
Gastrointestinal Microbiome , Gram-Negative Bacteria , Hormones, Ectopic/physiology , Intestinal Mucosa/microbiology , Animals , Humans , Immunity, Innate , Intercellular Signaling Peptides and Proteins , Intestinal Mucosa/immunology , Lipid Metabolism , Mice , Resistin/physiology , Symbiosis
16.
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
17.
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
18.
Brain Behav Immun ; 74: 176-185, 2018 11.
Article in English | MEDLINE | ID: mdl-30195028

ABSTRACT

The mammalian nervous system communicates important information about the environment to the immune system, but the underlying mechanisms are largely unknown. Secondary lymphoid organs are highly innervated by sympathetic neurons that secrete norepinephrine (NE) as the primary neurotransmitter. Immune cells express adrenergic receptors, enabling the sympathetic nervous system to directly control immune function. NE is a potent immunosuppressive factor and markedly inhibits TNF-α secretion from innate cells in response to lipopolysaccharide (LPS). In this study, we demonstrate that NE blocks the secretion of a variety of proinflammatory cytokines by rapidly inducing IL-10 secretion from innate cells in response to multiple Toll-like receptor (TLR) signals. NE mediated these effects exclusively through the ß2-adrenergic receptor (ADRB2). Consequently, Adrb2-/- animals were more susceptible to L. monocytogenes infection and to intestinal inflammation in a dextran sodium sulfate (DSS) model of colitis. Further, Adrb2-/- animals rapidly succumbed to endotoxemia in response to a sub-lethal LPS challenge and exhibited elevated serum levels of TNF-α and reduced IL-10. LPS-mediated lethality in WT animals was rescued by administering a ß 2-specific agonist and in Adrb2-/- animals by exogenous IL-10. These findings reveal a critical role for ADRB2 signaling in controlling inflammation through the rapid induction of IL-10. Our findings provide a fundamental insight into how the sympathetic nervous system controls a critical facet of immune function through ADRB2 signaling.


Subject(s)
Interleukin-10/metabolism , Receptors, Adrenergic, beta-2/metabolism , Animals , Cytokines/metabolism , Immune System/drug effects , Inflammation/metabolism , Interleukin-6/metabolism , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Norepinephrine/metabolism , Norepinephrine/pharmacology , Norepinephrine/physiology , Signal Transduction , Sympathetic Nervous System/drug effects , Toll-Like Receptors/metabolism , Tumor Necrosis Factor-alpha/metabolism
19.
Immunity ; 31(3): 368-76, 2009 Sep 18.
Article in English | MEDLINE | ID: mdl-19766080

ABSTRACT

The mammalian intestinal mucosal surface is continuously exposed to a complex and dynamic community of microorganisms. These microbes establish symbiotic relationships with their hosts, making important contributions to metabolism and digestive efficiency. The intestinal epithelial surface is the primary interface between the vast microbiota and internal host tissues. Given the enormous numbers of enteric bacteria and the persistent threat of opportunistic invasion, it is crucial that mammalian hosts monitor and regulate microbial interactions with intestinal epithelial surfaces. Here we discuss recent insights into how the innate and adaptive arms of the immune system collaborate to maintain homeostasis at the luminal surface of the intestinal host-microbial interface. These findings are also yielding a better understanding of how symbiotic host-microbial relationships can break down in inflammatory bowel disease.


Subject(s)
Immunity, Innate , Immunity, Mucosal , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Adaptation, Biological , Animals , Epithelium/immunology , Epithelium/microbiology , Humans , Inflammatory Bowel Diseases/immunology
20.
J Immunol ; 197(1): 97-107, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27217583

ABSTRACT

Commensal microbiota are critical for the development of local immune responses. In this article, we show that gut microbiota can regulate CD4 T cell polarization during pulmonary fungal infections. Vancomycin drinking water significantly decreased lung Th17 cell numbers during acute infection, demonstrating that Gram-positive commensals contribute to systemic inflammation. We next tested a role for RegIIIγ, an IL-22-inducible antimicrobial protein with specificity for Gram-positive bacteria. Following infection, increased accumulation of Th17 cells in the lungs of RegIIIγ(-/-) and Il22(-/-) mice was associated with intestinal segmented filamentous bacteria (SFB) colonization. Although gastrointestinal delivery of rRegIIIγ decreased lung inflammatory gene expression and protected Il22(-/-) mice from weight loss during infection, it had no direct effect on SFB colonization, fungal clearance, or lung Th17 immunity. We further show that vancomycin only decreased lung IL-17 production in mice colonized with SFB. To determine the link between gut microbiota and lung immunity, serum-transfer experiments revealed that IL-1R ligands increase the accumulation of lung Th17 cells. These data suggest that intestinal microbiota, including SFB, can regulate pulmonary adaptive immune responses.


Subject(s)
Aspergillosis/immunology , Aspergillus fumigatus/immunology , Gastrointestinal Microbiome/immunology , Gram-Positive Bacteria/immunology , Lung/immunology , Th17 Cells/immunology , Animals , Anti-Bacterial Agents/therapeutic use , Aspergillosis/drug therapy , Aspergillosis/microbiology , Cells, Cultured , Immunity/drug effects , Interleukins/genetics , Interleukins/metabolism , Lung/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Pancreatitis-Associated Proteins , Proteins/genetics , Proteins/metabolism , Vancomycin/therapeutic use , Interleukin-22
SELECTION OF CITATIONS
SEARCH DETAIL