Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
PLoS Pathog ; 20(6): e1012306, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38843309

ABSTRACT

Staphylococcus aureus is a human-adapted pathogen that replicates by asymptomatically colonizing its host. S. aureus is also the causative agent of purulent skin and soft tissue infections as well as bloodstream infections that result in the metastatic seeding of abscess lesions in all organ tissues. Prolonged colonization, infection, disease relapse, and recurrence point to the versatile capacity of S. aureus to bypass innate and adaptive immune defenses as well as the notion that some hosts fail to generate protective immune responses. Here, we find a genetic trait that provides protection against this pathogen. Mice lacking functional H2-O, the equivalent of human HLA-DO, inoculated with a mouse-adapted strain of S. aureus, efficiently decolonize the pathogen. Further, these decolonized animals resist subsequent bloodstream challenge with methicillin-resistant S. aureus. A genetic approach demonstrates that T-cell dependent B cell responses are required to control S. aureus colonization and infection in H2-O-deficient mice. Reduced bacterial burdens in these animals correlate with increased titers and enhanced phagocytic activity of S. aureus-specific antibodies. H2-O negatively regulates the loading of high affinity peptides on major histocompatibility class II (MHC-II) molecules. Thus, we hypothesize that immune responses against S. aureus are derepressed in mice lacking H2-O because more high affinity peptides are presented by MHC-II. We speculate that loss-of-function HLA-DO alleles may similarly control S. aureus replication in humans.


Subject(s)
Staphylococcal Infections , Staphylococcus aureus , Animals , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , Mice , Staphylococcus aureus/immunology , Mice, Knockout , Mice, Inbred C57BL , Histocompatibility Antigens Class II/immunology , Methicillin-Resistant Staphylococcus aureus/immunology , Humans
2.
Front Immunol ; 15: 1345467, 2024.
Article in English | MEDLINE | ID: mdl-38504980

ABSTRACT

The vast diversity of mammalian adaptive antigen receptors allows for robust and efficient immune responses against a wide number of pathogens. The antigen receptor repertoire is built during the recombination of B and T cell receptor (BCR, TCR) loci and hypermutation of BCR loci. V(D)J recombination rearranges these antigen receptor loci, which are organized as an array of separate V, (D), and J gene segments. Transcription activation at the recombining locus leads to changes in the local three-dimensional architecture, which subsequently contributes to which gene segments are utilized for recombination. The endogenous retrovirus (ERV) mouse mammary tumor provirus 8 (Mtv8) resides on mouse chromosome 6 interposed within the large array of light chain kappa V gene segments. As ERVs contribute to changes in genomic architecture by driving high levels of transcription of neighboring genes, it was suggested that Mtv8 could influence the BCR repertoire. We generated Mtv8-deficient mice to determine if the ERV influences V(D)J recombination to test this possibility. We find that Mtv8 does not influence the BCR repertoire.


Subject(s)
Receptors, Antigen, T-Cell , V(D)J Recombination , Animals , Mice , Immunoglobulins/genetics , Mammals , Receptors, Antigen, T-Cell/genetics , V(D)J Recombination/genetics
3.
bioRxiv ; 2023 Oct 12.
Article in English | MEDLINE | ID: mdl-37873087

ABSTRACT

Microbial impact on tumorigenesis of heritable cancers proximal to the gut is well documented. Whether the microbiota influences cancers arising from inborn mutations at sites distal to the gut is undetermined. Using two models of heritable cancer, we found the microbiota to be inconsequential for tumor development. However, the type of tumor that develops may be influenced by the microbiota. This work furthers our understanding of the microbial impact on tumor development.

4.
J Virol ; 97(8): e0068523, 2023 08 31.
Article in English | MEDLINE | ID: mdl-37578238

ABSTRACT

Differential responses to viral infections are influenced by the genetic makeup of the host. Studies of resistance to retroviruses in human populations are complicated due to the inability to conduct proof-of-principle studies. Inbred mouse lines, which have a range of susceptible phenotypes to retroviruses, are an ideal tool to identify and characterize mechanisms of resistance and define their genetic underpinnings. YBR/Ei mice become infected with Mouse Mammary Tumor Virus, a mucosally transmitted murine retrovirus, but eliminate the virus from their pedigrees. Virus elimination correlates with a lack of virus-specific neonatal oral tolerance, which is a major mechanism for blocking the anti-virus response in susceptible mice. Virus control is unrelated to virus-neutralizing antibodies, cytotoxic CD8+ T cells, NK cells, and NK T cells, which are the best characterized mechanisms of resistance to retroviruses. We identified a single, dominant locus that controls the resistance mechanism, which we provisionally named attenuation of virus titers (Avt) and mapped to the distal region of chromosome 18. IMPORTANCE Elucidation of the mechanism that mediates resistance to retroviruses is of fundamental importance to human health, as it will ultimately lead to knowledge of the genetic differences among individuals in susceptibility to microbial infections.


Subject(s)
Retroviridae Infections , Retroviridae , Mice , Animals , Humans , CD8-Positive T-Lymphocytes , Mice, Inbred Strains , T-Lymphocytes, Cytotoxic , Disease Susceptibility
5.
Viruses ; 15(2)2023 01 29.
Article in English | MEDLINE | ID: mdl-36851600

ABSTRACT

Both viruses and bacteria produce "pathogen associated molecular patterns" that may affect microbial pathogenesis and anti-microbial responses. Additionally, bacteria produce metabolites, while viruses could change the metabolic profiles of the infected cells. Here, we used an unbiased metabolomics approach to profile metabolites in spleens and blood of murine leukemia virus-infected mice monocolonized with Lactobacillus murinus to show that viral infection significantly changes the metabolite profile of monocolonized mice. We hypothesize that these changes could contribute to viral pathogenesis or to the host response against the virus and thus open a new avenue for future investigations.


Subject(s)
Retroviridae Infections , Animals , Mice , Bacteria , Metabolomics , Leukemia Virus, Murine , Spleen
6.
bioRxiv ; 2023 Jan 11.
Article in English | MEDLINE | ID: mdl-36711645

ABSTRACT

Both viruses and bacteria produce 'pathogen associated molecular patterns' that may affect microbial pathogenesis and anti-microbial responses. Additionally, bacteria produce metabolites while viruses could change metabolic profiles of the infected cells. Here, we used an unbiased metabolomics approach to profile metabolites in spleens and blood of Murine Leukemia Virus-infected mice monocolonized with Lactobacillus murinus to show that viral infection significantly changes the metabolite profile of monocolonized mice. We hypothesize that these changes could contribute to viral pathogenesis or to the host response against the virus and thus, open a new avenue for future investigations.

7.
Cell Rep ; 40(11): 111341, 2022 09 13.
Article in English | MEDLINE | ID: mdl-36103821

ABSTRACT

The influence of the microbiota on viral transmission and replication is well appreciated. However, its impact on retroviral pathogenesis outside of transmission/replication control remains unknown. Using murine leukemia virus (MuLV), we found that some commensal bacteria promoted the development of leukemia induced by this retrovirus. The promotion of leukemia development by commensals is due to suppression of the adaptive immune response through upregulation of several negative regulators of immunity. These negative regulators include Serpinb9b and Rnf128, which are associated with a poor prognosis of some spontaneous human cancers. Upregulation of Serpinb9b is mediated by sensing of bacteria by the NOD1/NOD2/RIPK2 pathway. This work describes a mechanism by which the microbiota enhances tumorigenesis within gut-distant organs and points at potential targets for cancer therapy.


Subject(s)
Leukemia , Retroviridae , Animals , Bacteria/metabolism , Carcinogenesis , Humans , Mice , Symbiosis
8.
J Immunol ; 207(12): 2944-2951, 2021 12 15.
Article in English | MEDLINE | ID: mdl-34810225

ABSTRACT

H2-O (human HLA-DO) is a relatively conserved nonclassical MHC class II (MHCII)-like molecule. H2-O interaction with human HLA-DM edits the repertoire of peptides presented to TCRs by MHCII. It was long hypothesized that human HLA-DM inhibition by H2-O provides protection from autoimmunity by preventing binding of the high-affinity self-peptides to MHCII. The available evidence supporting this hypothesis, however, was inconclusive. A possibility still remained that the effect of H2-O deficiency on autoimmunity could be better revealed by using H2-O-deficient mice that were already genetically predisposed to autoimmunity. In this study, we generated and used autoimmunity-prone mouse models for systemic lupus erythematosus and organ-specific autoimmunity (type 1 diabetes and multiple sclerosis) to definitively test whether H2-O prevents autoimmune pathology. Whereas our data failed to support any significance of H2-O in protection from autoimmunity, we found that it was critical for controlling a γ-herpesvirus, MHV68. Thus, we propose that H2-O editing of the MHCII peptide repertoire may have evolved as a safeguard against specific highly prevalent viral pathogens.


Subject(s)
Autoimmunity , HLA-D Antigens , Animals , Antigen Presentation , HLA-D Antigens/genetics , Histocompatibility Antigens Class II , Humans , Mice , Peptides , Receptors, Antigen, T-Cell
9.
J Immunol ; 207(1): 5-14, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34135064

ABSTRACT

Pathogen-derived peptides are loaded on MHC class II (MHCII) and presented to CD4+ T cells for their activation. Peptide loading of MHCII occurs in specialized endosomal compartments and is controlled by the nonclassical MHCII molecules H2-M and H2-O, which are both constitutive αß heterodimers. H2-M catalyzes MHCII peptide loading, whereas H2-O modulates H2-M activity by acting as an MHCII mimic. Recently, we discovered that the H2-Ob allele inherited by retrovirus-resistant I/LnJ mice results in nonfunctional H2-O. I/LnJ H2-O binds to but does not inhibit H2-M. Compared with H2-Oß from virus-susceptible mice, H2-Oß from I/LnJ mice has four unique amino acid substitutions, three in the Ig domain and one in the cytoplasmic tail. In this study we show that the three amino acids in the Ig domain of I/LnJ Oß are critical for the H2-O inhibitory activity of H2-M. Unexpectedly, we found that MHCII presentation was significantly different in Ag-presenting cells from two closely related mouse strains, B6J and B6N, which carry identical alleles of MHCII, H2-O, and H2-M. Using a positional cloning approach, we have identified two loci, polymorphic between B6J and B6N, that mediate the difference in MHCII presentation. Collectively, these studies reveal extra complexity in MHCII/H2-M/H-2O interactions that likely involve yet to be identified modulators of the pathway.


Subject(s)
Histocompatibility Antigens Class II/genetics , Immunoglobulins/genetics , Polymorphism, Genetic/genetics , Animals , Histocompatibility Antigens Class II/immunology , Immunoglobulins/immunology , Mice , Mice, Congenic , Mice, Inbred C57BL , Mice, Knockout
10.
J Virol ; 94(24)2020 11 23.
Article in English | MEDLINE | ID: mdl-32999021

ABSTRACT

Viruses, including retroviruses, can be passed from mothers to their progeny during birth and breastfeeding. It is assumed that newborns may develop immune tolerance to milk-transmitted pathogens similarly to food antigens. I/LnJ mice are uniquely resistant to retroviruses acquired as newborns or as adults as they produce virus-neutralizing antibodies (Abs). A loss-of-function allele of H2-Ob (Ob), originally mapped within the virus infectivity controller 1 (vic1) locus, is responsible for production of antiretrovirus Abs in I/LnJ mice. Importantly, Ob-deficient and vic1 I/LnJ congenic mice on other genetic backgrounds produce antivirus Abs when infected as adults, but not as newborns. We report here that I/LnJ mice carry an additional genetic locus, virus infectivity controller 2 (vic2), that abrogates neonatal immune tolerance to retroviruses. Further genetic analysis mapped the vic2 locus to the telomeric end of chromosome 15. Identification of the vic2 gene and understanding of the related signaling pathways would make blocking of neonatal immune tolerance to retroviruses an achievable goal.IMPORTANCE This work describes a previously unknown genetic mechanism that allows neonates to respond to infections as efficiently as adults.


Subject(s)
Immune Tolerance/genetics , Retroviridae Infections/immunology , Retroviridae/immunology , Animals , Antibodies, Neutralizing , Chromosome Mapping , Female , Mice , Mice, Congenic , Mice, Inbred BALB C , Mice, Inbred Strains , Retroviridae Infections/virology
11.
J Immunol ; 205(4): 923-935, 2020 08 15.
Article in English | MEDLINE | ID: mdl-32690655

ABSTRACT

HLA molecules of the MHC class II (MHCII) bind and present pathogen-derived peptides for CD4 T cell activation. Peptide loading of MHCII in the endosomes of cells is controlled by the interplay of the nonclassical MHCII molecules, HLA-DM (DM) and HLA-DO (DO). DM catalyzes peptide loading, whereas DO, an MHCII substrate mimic, prevents DM from interacting with MHCII, resulting in an altered MHCII-peptide repertoire and increased MHCII-CLIP. Although the two genes encoding DO (DOA and DOB) are considered nonpolymorphic, there are rare natural variants. Our previous work identified DOB variants that altered DO function. In this study, we show that natural variation in the DOA gene also impacts DO function. Using the 1000 Genomes Project database, we show that ∼98% of individuals express the canonical DOA*0101 allele, and the remaining individuals mostly express DOA*0102, which we found was a gain-of-function allele. Analysis of 25 natural occurring DOα variants, which included the common alleles, identified three null variants and one variant with reduced and nine with increased ability to modulate DM activity. Unexpectedly, several of the variants produced reduced DO protein levels yet efficiently inhibited DM activity. Finally, analysis of associated single-nucleotide polymorphisms genetically linked the DOA*0102 common allele, a gain-of-function variant, with human hepatitis B viral persistence. In contrast, we found that the DOα F114L null allele was linked with viral clearance. Collectively, these studies show that natural variation occurring in the human DOA gene impacts DO function and can be linked to specific outcomes of viral infections.


Subject(s)
HLA-D Antigens/genetics , Hepatitis B/genetics , Histocompatibility Antigens Class II/genetics , Polymorphism, Single Nucleotide/genetics , Alleles , Antigen Presentation/genetics , Cell Line, Tumor , HeLa Cells , Hepatitis B/virology , Humans , Peptides/genetics
12.
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
13.
Cell Rep ; 29(3): 541-550.e4, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31618625

ABSTRACT

Environmental influences (infections and diet) strongly affect a host's microbiota. However, host genetics may influence commensal communities, as suggested by the greater similarity between the microbiomes of identical twins compared to non-identical twins. Variability of human genomes and microbiomes complicates the understanding of polymorphic mechanisms regulating the commensal communities. Whereas animal studies allow genetic modifications, they are sensitive to influences known as "cage" or "legacy" effects. Here, we analyze ex-germ-free mice of various genetic backgrounds, including immunodeficient and major histocompatibility complex (MHC) congenic strains, receiving identical input microbiota. The host's polymorphic mechanisms affect the gut microbiome, and both innate (anti-microbial peptides, complement, pentraxins, and enzymes affecting microbial survival) and adaptive (MHC-dependent and MHC-independent) pathways influence the microbiota. In our experiments, polymorphic mechanisms regulate only a limited number of microbial lineages (independently of their abundance). Our comparative analyses suggest that some microbes may benefit from the specific immune responses that they elicit.


Subject(s)
Adaptive Immunity/genetics , Immunity, Innate/genetics , Polymorphism, Genetic , Animals , Bacteria/genetics , Bacteria/isolation & purification , Defensins/genetics , Defensins/metabolism , Gastrointestinal Microbiome , Gene Expression , Immunocompromised Host , Intestinal Mucosa/metabolism , Intestines/microbiology , Major Histocompatibility Complex/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Principal Component Analysis , RNA, Ribosomal, 16S/metabolism
14.
Annu Rev Virol ; 6(1): 525-546, 2019 09 29.
Article in English | MEDLINE | ID: mdl-31567067

ABSTRACT

Genetic alleles that contribute to enhanced susceptibility or resistance to viral infections and virally induced diseases have often been first identified in mice before humans due to the significant advantages of the murine system for genetic studies. Herein we review multiple discoveries that have revealed significant insights into virus-host interactions, all made using genetic mapping tools in mice. Factors that have been identified include innate and adaptive immunity genes that contribute to host defense against pathogenic viruses such as herpes viruses, flaviviruses, retroviruses, and coronaviruses. Understanding the genetic mechanisms that affect infectious disease outcomes will aid the development of personalized treatment and preventive strategies for pathogenic infections.


Subject(s)
Mice/genetics , Virus Diseases/genetics , Virus Physiological Phenomena , Adaptive Immunity , Animals , Genetic Predisposition to Disease , Humans , Immunity, Innate , Mice/immunology , Mice/virology , Virus Diseases/immunology , Virus Diseases/virology , Viruses/genetics
15.
J Virol ; 92(16)2018 08 15.
Article in English | MEDLINE | ID: mdl-29875252

ABSTRACT

An essential step in the development of effective antiviral humoral responses is cytokine-triggered class switch recombination resulting in the production of antibodies of a specific isotype. Most viral and parasitic infections in mice induce predominantly IgG2a-specific antibody responses that are stimulated by interferon gamma (IFN-γ). However, in some mice deficient in IFN-γ, class switching to IgG2a antibodies is relatively unaffected, indicating that another signal(s) can be generated upon viral or parasitic infections that trigger this response. Here, we found that a single recessive locus, provisionally called IFN-γ-independent IgG2a (Igii), confers the ability to produce IFN-γ-independent production of IgG2a antibodies upon retroviral infection. The Igii locus was mapped to chromosome 9 and was found to function in the radiation-resistant compartment. Thus, our data implicate nonhematopoietic cells in activation of antiviral antibody responses in the absence of IFN-γ.IMPORTANCE Understanding the signals that stimulate antibody production and class switch recombination to specific antibody isotypes is crucial for the development of novel vaccines and adjuvants. While an interferon gamma-mediated switch to the IgG2a isotype upon viral infection in mice has been well established, this investigation reveals a noncanonical, interferon gamma-independent pathway for antiretroviral antibody production and IgG2a class switch recombination that is controlled by a single recessive locus. Furthermore, this study indicates that the radiation-resistant compartment can direct antiviral antibody responses, suggesting that detection of infection by nonhematopoietic cells is involved is stimulating adaptive immunity.


Subject(s)
Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Immunoglobulin G/blood , Viruses/immunology , Animals , Chromosome Mapping , Interferon-gamma/deficiency , Mice , Mice, Knockout
16.
Immunity ; 47(2): 310-322.e7, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28813660

ABSTRACT

Select humans and animals control persistent viral infections via adaptive immune responses that include production of neutralizing antibodies. The precise genetic basis for the control remains enigmatic. Here, we report positional cloning of the gene responsible for production of retrovirus-neutralizing antibodies in mice of the I/LnJ strain. It encodes the beta subunit of the non-classical major histocompatibility complex class II (MHC-II)-like molecule H2-O, a negative regulator of antigen presentation. The recessive and functionally null I/LnJ H2-Ob allele supported the production of virus-neutralizing antibodies independently of the classical MHC haplotype. Subsequent bioinformatics and functional analyses of the human H2-Ob homolog, HLA-DOB, revealed both loss- and gain-of-function alleles, which could affect the ability of their carriers to control infections with human hepatitis B (HBV) and C (HCV) viruses. Thus, understanding of the previously unappreciated role of H2-O (HLA-DO) in immunity to infections may suggest new approaches in achieving neutralizing immunity to viruses.


Subject(s)
Antibodies, Neutralizing , HLA-D Antigens/metabolism , Histocompatibility Antigens Class II/metabolism , Immunity, Humoral , Mammary Tumor Virus, Mouse/immunology , Rauscher Virus/immunology , Retroviridae Infections/immunology , Animals , Antibodies, Neutralizing/metabolism , Antibodies, Viral/metabolism , Antigen Presentation/genetics , Computational Biology , Female , Genetic Predisposition to Disease , HLA-D Antigens/genetics , HeLa Cells , Hepatitis B/immunology , Hepatitis B/transmission , Hepatitis C/immunology , Hepatitis C/transmission , Histocompatibility Antigens Class II/genetics , Humans , Immunity, Humoral/genetics , Male , Mice , Mice, Inbred Strains , Mice, Knockout , Mutation/genetics , Polymorphism, Genetic , Retroviridae Infections/transmission
17.
ILAR J ; 57(1): 12-23, 2016.
Article in English | MEDLINE | ID: mdl-27034391

ABSTRACT

Mouse mammary tumor virus (MMTV), which was discovered as a milk-transmitted, infectious, cancer-inducing agent in the 1930s, has been used as an animal model for the study of retroviral infection and transmission, antiviral immune responses, and breast cancer and lymphoma biology. The main target cells for MMTV infection in vivo are cells of the immune system and mammary epithelial cells. Although the host mounts an immune response to the virus, MMTV has evolved multiple means of evading this response. MMTV causes mammary tumors when the provirus integrates into the mammary epithelial and lymphoid cell genome during viral replication and thereby activates cellular oncogene expression. Thus, tumor induction is a by-product of the infection cycle. A number of important oncogenes have been discovered by carrying out MMTV integration site analysis, some of which may play a role in human breast cancer.


Subject(s)
Mammary Tumor Virus, Mouse/physiology , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/virology , Disease Models, Animal , Female , Humans , Mice
18.
Cell Host Microbe ; 18(4): 456-62, 2015 Oct 14.
Article in English | MEDLINE | ID: mdl-26468748

ABSTRACT

The orally transmitted retrovirus mouse mammary tumor virus (MMTV) requires the intestinal microbiota for persistence. Virion-associated lipopolysaccharide (LPS) activates Toll-like receptor 4 (TLR4), stimulating production of the immunosuppressive cytokine IL-10 and MMTV evasion of host immunity. However, the mechanisms by which MMTV associates with LPS remain unknown. We find that the viral envelope contains the mammalian LPS-binding factors CD14, TLR4, and MD-2, which, in conjunction with LPS-binding protein (LBP), bind LPS to the virus and augment transmission. MMTV isolated from infected mice lacking these LBPs cannot engage LPS or stimulate TLR4 and have a transmission defect. Furthermore, MMTV incorporation of a weak agonist LPS from Bacteroides, a prevalent LPS source in the gut, significantly enhances the ability of this LPS to stimulate TLR4, suggesting that MMTV intensifies these immunostimulatory properties. Thus, an orally transmitted retrovirus can capture, modify, and exploit mammalian receptors for bacterial ligands to ensure successful transmission.


Subject(s)
Host-Pathogen Interactions , Lipopolysaccharide Receptors/metabolism , Mammary Tumor Virus, Mouse/physiology , Viral Envelope Proteins/metabolism , Animals , Immune Evasion , Immunosuppressive Agents/metabolism , Interleukin-10/metabolism , Lipopolysaccharides/metabolism , Mice , Protein Binding , Signal Transduction , Toll-Like Receptor 4/metabolism
20.
Nature ; 514(7524): 638-41, 2014 Oct 30.
Article in English | MEDLINE | ID: mdl-25274297

ABSTRACT

Systemic infection induces conserved physiological responses that include both resistance and 'tolerance of infection' mechanisms. Temporary anorexia associated with an infection is often beneficial, reallocating energy from food foraging towards resistance to infection or depriving pathogens of nutrients. However, it imposes a stress on intestinal commensals, as they also experience reduced substrate availability; this affects host fitness owing to the loss of caloric intake and colonization resistance (protection from additional infections). We hypothesized that the host might utilize internal resources to support the gut microbiota during the acute phase of the disease. Here we show that systemic exposure to Toll-like receptor (TLR) ligands causes rapid α(1,2)-fucosylation of small intestine epithelial cells (IECs) in mice, which requires the sensing of TLR agonists, as well as the production of interleukin (IL)-23 by dendritic cells, activation of innate lymphoid cells and expression of fucosyltransferase 2 (Fut2) by IL-22-stimulated IECs. Fucosylated proteins are shed into the lumen and fucose is liberated and metabolized by the gut microbiota, as shown by reporter bacteria and community-wide analysis of microbial gene expression. Fucose affects the expression of microbial metabolic pathways and reduces the expression of bacterial virulence genes. It also improves host tolerance of the mild pathogen Citrobacter rodentium. Thus, rapid IEC fucosylation appears to be a protective mechanism that utilizes the host's resources to maintain host-microbial interactions during pathogen-induced stress.


Subject(s)
Disease , Epithelium/metabolism , Epithelium/microbiology , Fucose/metabolism , Intestine, Small/metabolism , Intestine, Small/microbiology , Symbiosis , Animals , Anorexia/complications , Anorexia/microbiology , Bacteria/genetics , Bacteria/metabolism , Bacteria/pathogenicity , Citrobacter rodentium/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Eating , Fatty Acids/chemistry , Fatty Acids/metabolism , Female , Fucosyltransferases/metabolism , Gene Expression Regulation, Bacterial , Glycosylation , Immune Tolerance , Immunity, Innate , Interleukins/biosynthesis , Interleukins/immunology , Ligands , Male , Metabolic Networks and Pathways/genetics , Mice , Microbiota/physiology , Protective Factors , Toll-Like Receptors/agonists , Toll-Like Receptors/immunology , Toll-Like Receptors/metabolism , Virulence Factors/genetics , Interleukin-22 , Galactoside 2-alpha-L-fucosyltransferase
SELECTION OF CITATIONS
SEARCH DETAIL
...