Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 156
Filter
1.
Annu Rev Immunol ; 36: 359-381, 2018 04 26.
Article in English | MEDLINE | ID: mdl-29400985

ABSTRACT

IgA is the dominant immunoglobulin isotype produced in mammals, largely secreted across the intestinal mucosal surface. Although induction of IgA has been a hallmark feature of microbiota colonization following colonization in germ-free animals, until recently appreciation of the function of IgA in host-microbial mutualism has depended mainly on indirect evidence of alterations in microbiota composition or penetration of microbes in the absence of somatic mutations in IgA (or compensatory IgM). Highly parallel sequencing techniques that enable high-resolution analysis of either microbial consortia or IgA sequence diversity are now giving us new perspectives on selective targeting of microbial taxa and the trajectory of IgA diversification according to induction mechanisms, between different individuals and over time. The prospects are to link the range of diversified IgA clonotypes to specific antigenic functions in modulating the microbiota composition, position and metabolism to ensure host mutualism.


Subject(s)
Gastrointestinal Microbiome/immunology , Immunoglobulin A/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Age Factors , Animals , Disease Susceptibility , Host-Pathogen Interactions/immunology , Humans , Intestinal Mucosa/metabolism , Protein Binding
2.
Nat Immunol ; 24(7): 1098-1109, 2023 07.
Article in English | MEDLINE | ID: mdl-37337103

ABSTRACT

Macrophages are involved in immune defense, organogenesis and tissue homeostasis. Macrophages contribute to the different phases of mammary gland remodeling during development, pregnancy and involution postlactation. Less is known about the dynamics of mammary gland macrophages in the lactation stage. Here, we describe a macrophage population present during lactation in mice. By multiparameter flow cytometry and single-cell RNA sequencing, we identified a lactation-induced CD11c+CX3CR1+Dectin-1+ macrophage population (liMac) that was distinct from the two resident F4/80hi and F4/80lo macrophage subsets present pregestationally. LiMacs were predominantly monocyte-derived and expanded by proliferation in situ concomitant with nursing. LiMacs developed independently of IL-34, but required CSF-1 signaling and were partly microbiota-dependent. Locally, they resided adjacent to the basal cells of the alveoli and extravasated into the milk. We found several macrophage subsets in human milk that resembled liMacs. Collectively, these findings reveal the emergence of unique macrophages in the mammary gland and milk during lactation.


Subject(s)
Lactation , Milk, Human , Pregnancy , Female , Mice , Humans , Animals , Macrophages , Mammary Glands, Animal
3.
Cell ; 181(6): 1202-1204, 2020 06 11.
Article in English | MEDLINE | ID: mdl-32497500

ABSTRACT

ROR-γt+ regulatory T cells (Tregs) of the colon can prevent excessive inflammation but also delay pathogen clearance. How these cells are regulated has remained elusive. In this issue of Cell, Ramanan et al. find that the set-point for ROR-γt+ Tregs is non-genetically maternally inherited during a critical time window after birth through immunoglobulin A present in breast milk.


Subject(s)
Gastrointestinal Microbiome , T-Lymphocytes, Regulatory , Female , Humans , Milk, Human , Nuclear Receptor Subfamily 1, Group F, Member 3 , Th17 Cells
4.
Cell ; 181(5): 1080-1096.e19, 2020 05 28.
Article in English | MEDLINE | ID: mdl-32380006

ABSTRACT

Environmental signals shape host physiology and fitness. Microbiota-derived cues are required to program conventional dendritic cells (cDCs) during the steady state so that they can promptly respond and initiate adaptive immune responses when encountering pathogens. However, the molecular underpinnings of microbiota-guided instructive programs are not well understood. Here, we report that the indigenous microbiota controls constitutive production of type I interferons (IFN-I) by plasmacytoid DCs. Using genome-wide analysis of transcriptional and epigenetic regulomes of cDCs from germ-free and IFN-I receptor (IFNAR)-deficient mice, we found that tonic IFNAR signaling instructs a specific epigenomic and metabolic basal state that poises cDCs for future pathogen combat. However, such beneficial biological function comes with a trade-off. Instructed cDCs can prime T cell responses against harmless peripheral antigens when removing roadblocks of peripheral tolerance. Our data provide fresh insights into the evolutionary trade-offs that come with successful adaptation of vertebrates to their microbial environment.


Subject(s)
Dendritic Cells/immunology , Interferon Type I/immunology , Microbiota/immunology , Adaptive Immunity/immunology , Adaptive Immunity/physiology , Animals , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/microbiology , Female , Male , Mice , Mice, Inbred C57BL , Microbiota/physiology , Receptor, Interferon alpha-beta/metabolism , Signal Transduction/immunology
5.
Immunity ; 56(8): 1712-1726, 2023 08 08.
Article in English | MEDLINE | ID: mdl-37557080

ABSTRACT

The enteric nervous system is largely autonomous, and the central nervous system is compartmentalized behind the blood-brain barrier. Yet the intestinal microbiota shapes gut function, local and systemic immune responses, and central nervous system functions including cognition and mood. In this review, we address how the gut microbiota can profoundly influence neural and immune networks. Although many of the interactions between these three systems originate in the intestinal mucosa, intestinal function and immunity are modulated by neural pathways that connect the gut and brain. Furthermore, a subset of microbe-derived penetrant molecules enters the brain and regulates central nervous system function. Understanding how these seemingly isolated entities communicate has the potential to open up new avenues for therapies and interventions.


Subject(s)
Enteric Nervous System , Gastrointestinal Microbiome , Microbiota , Central Nervous System , Brain
6.
Immunity ; 56(5): 1115-1131.e9, 2023 05 09.
Article in English | MEDLINE | ID: mdl-36917985

ABSTRACT

Intestinal IL-17-producing T helper (Th17) cells are dependent on adherent microbes in the gut for their development. However, how microbial adherence to intestinal epithelial cells (IECs) promotes Th17 cell differentiation remains enigmatic. Here, we found that Th17 cell-inducing gut bacteria generated an unfolded protein response (UPR) in IECs. Furthermore, subtilase cytotoxin expression or genetic removal of X-box binding protein 1 (Xbp1) in IECs caused a UPR and increased Th17 cells, even in antibiotic-treated or germ-free conditions. Mechanistically, UPR activation in IECs enhanced their production of both reactive oxygen species (ROS) and purine metabolites. Treating mice with N-acetyl-cysteine or allopurinol to reduce ROS production and xanthine, respectively, decreased Th17 cells that were associated with an elevated UPR. Th17-related genes also correlated with ER stress and the UPR in humans with inflammatory bowel disease. Overall, we identify a mechanism of intestinal Th17 cell differentiation that emerges from an IEC-associated UPR.


Subject(s)
Endoplasmic Reticulum Stress , Intestinal Mucosa , Th17 Cells , Endoplasmic Reticulum Stress/drug effects , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Th17 Cells/cytology , Th17 Cells/metabolism , Cell Differentiation , Humans , Animals , Mice , Mice, Transgenic , Anti-Bacterial Agents/pharmacology
7.
Cell ; 171(3): 655-667.e17, 2017 Oct 19.
Article in English | MEDLINE | ID: mdl-29053971

ABSTRACT

The gut microbiota contributes to the development of normal immunity but, when dysregulated, can promote autoimmunity through various non-antigen-specific effects on pathogenic and regulatory lymphocytes. Here, we show that an integrase expressed by several species of the gut microbial genus Bacteroides encodes a low-avidity mimotope of the pancreatic ß cell autoantigen islet-specific glucose-6-phosphatase-catalytic-subunit-related protein (IGRP206-214). Studies in germ-free mice monocolonized with integrase-competent, integrase-deficient, and integrase-transgenic Bacteroides demonstrate that the microbial epitope promotes the recruitment of diabetogenic CD8+ T cells to the gut. There, these effectors suppress colitis by targeting microbial antigen-loaded, antigen-presenting cells in an integrin ß7-, perforin-, and major histocompatibility complex class I-dependent manner. Like their murine counterparts, human peripheral blood T cells also recognize Bacteroides integrase. These data suggest that gut microbial antigen-specific cytotoxic T cells may have therapeutic value in inflammatory bowel disease and unearth molecular mimicry as a novel mechanism by which the gut microbiota can regulate normal immune homeostasis. PAPERCLIP.


Subject(s)
Autoantigens/immunology , Bacteroides/immunology , Colitis/immunology , Gastrointestinal Microbiome , Glucose-6-Phosphatase/immunology , Adult , Animals , Bacteroides/classification , Bacteroides/enzymology , Colitis/microbiology , Female , Glucose-6-Phosphatase/genetics , Humans , Lymphoid Tissue/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Middle Aged , Molecular Mimicry , T-Lymphocytes/immunology
8.
Immunity ; 55(10): 1753-1755, 2022 10 11.
Article in English | MEDLINE | ID: mdl-36223722

ABSTRACT

Immune cells generated in early life play pivotal roles for immune homeostasis in adulthood. Vergani et al. identify a population of early-life-origin IgA B cells that are maintained throughout life to achieve homeostasis in the adult gut.


Subject(s)
B-Lymphocytes , Origin of Life , Homeostasis , Immunoglobulin A
9.
Nature ; 613(7945): 639-649, 2023 01.
Article in English | MEDLINE | ID: mdl-36697862

ABSTRACT

Whether the human fetus and the prenatal intrauterine environment (amniotic fluid and placenta) are stably colonized by microbial communities in a healthy pregnancy remains a subject of debate. Here we evaluate recent studies that characterized microbial populations in human fetuses from the perspectives of reproductive biology, microbial ecology, bioinformatics, immunology, clinical microbiology and gnotobiology, and assess possible mechanisms by which the fetus might interact with microorganisms. Our analysis indicates that the detected microbial signals are likely the result of contamination during the clinical procedures to obtain fetal samples or during DNA extraction and DNA sequencing. Furthermore, the existence of live and replicating microbial populations in healthy fetal tissues is not compatible with fundamental concepts of immunology, clinical microbiology and the derivation of germ-free mammals. These conclusions are important to our understanding of human immune development and illustrate common pitfalls in the microbial analyses of many other low-biomass environments. The pursuit of a fetal microbiome serves as a cautionary example of the challenges of sequence-based microbiome studies when biomass is low or absent, and emphasizes the need for a trans-disciplinary approach that goes beyond contamination controls by also incorporating biological, ecological and mechanistic concepts.


Subject(s)
Biomass , DNA Contamination , Fetus , Microbiota , Animals , Female , Humans , Pregnancy , Amniotic Fluid/immunology , Amniotic Fluid/microbiology , Mammals , Microbiota/genetics , Placenta/immunology , Placenta/microbiology , Fetus/immunology , Fetus/microbiology , Reproducibility of Results
10.
Immunity ; 49(3): 545-559.e5, 2018 09 18.
Article in English | MEDLINE | ID: mdl-30193848

ABSTRACT

Although the mammalian microbiota is well contained within the intestine, it profoundly shapes development and metabolism of almost every host organ. We questioned the range and depth of microbial metabolite penetration into the host, and how this is modulated by intestinal immunity. Chemically identical microbial and host metabolites were distinguished by stable isotope tracing from 13C-labeled live non-replicating Escherichia coli, differentiating 12C host isotopes with high-resolution mass spectrometry. Hundreds of endogenous microbial compounds penetrated 23 host tissues and fluids after intestinal exposure: subsequent 12C host metabolome signatures included lipidemia, reduced glycolysis, and inflammation. Penetrant bacterial metabolites from the small intestine were rapidly cleared into the urine, whereas induced antibodies curtailed microbial metabolite exposure by accelerating intestinal bacterial transit into the colon where metabolite transport mechanisms are limiting. Pervasive penetration of microbial molecules can cause extensive host tissue responses: these are limited by immune and non-immune intestinal mucosal adaptations to the microbiota.


Subject(s)
Antibodies/metabolism , Gastrointestinal Microbiome/physiology , Glycolysis/immunology , Hyperlipidemias/immunology , Inflammation/immunology , Mammals/immunology , Animals , Antibodies/immunology , Carbon Radioisotopes/analysis , Host-Pathogen Interactions , Immunity , Immunoglobulin Heavy Chains/genetics , Mass Spectrometry , Mice , Mice, Inbred C57BL , Mice, Knockout
11.
Immunity ; 49(2): 342-352.e5, 2018 08 21.
Article in English | MEDLINE | ID: mdl-30097293

ABSTRACT

Interleukin-22 (IL-22)-producing group 3 innate lymphoid cells (ILC3) maintains gut homeostasis but can also promote inflammatory bowel disease (IBD). The regulation of ILC3-dependent colitis remains to be elucidated. Here we show that Foxp3+ regulatory T cells (Treg cells) prevented ILC3-mediated colitis in an IL-10-independent manner. Treg cells inhibited IL-23 and IL-1ß production from intestinal-resident CX3CR1+ macrophages but not CD103+ dendritic cells. Moreover, Treg cells restrained ILC3 production of IL-22 through suppression of CX3CR1+ macrophage production of IL-23 and IL-1ß. This suppression was contact dependent and was mediated by latent activation gene-3 (LAG-3)-an immune checkpoint receptor-expressed on Treg cells. Engagement of LAG-3 on MHC class II drove profound immunosuppression of CX3CR1+ tissue-resident macrophages. Our study reveals that the health of the intestinal mucosa is maintained by an axis driven by Treg cells communication with resident macrophages that withhold inflammatory stimuli required for ILC3 function.


Subject(s)
Antigens, CD/metabolism , CX3C Chemokine Receptor 1/metabolism , Colitis/immunology , Colitis/pathology , Interleukin-23 Subunit p19/immunology , Intestinal Mucosa/pathology , Macrophages/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cells, Cultured , Dendritic Cells/immunology , Forkhead Transcription Factors/metabolism , Histocompatibility Antigens Class II/immunology , Interleukin-10/immunology , Interleukin-1beta/immunology , Interleukins/immunology , Intestinal Mucosa/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes, Regulatory/transplantation , Lymphocyte Activation Gene 3 Protein , Interleukin-22
12.
Nature ; 598(7882): 657-661, 2021 10.
Article in English | MEDLINE | ID: mdl-34646015

ABSTRACT

Dimeric IgA secreted across mucous membranes in response to nonpathogenic taxa of the microbiota accounts for most antibody production in mammals. Diverse binding specificities can be detected within the polyclonal mucosal IgA antibody response1-10, but limited monoclonal hybridomas have been studied to relate antigen specificity or polyreactive binding to functional effects on microbial physiology in vivo11-17. Here we use recombinant dimeric monoclonal IgAs (mIgAs) to finely map the intestinal plasma cell response to microbial colonization with a single microorganism in mice. We identify a range of antigen-specific mIgA molecules targeting defined surface and nonsurface membrane antigens. Secretion of individual dimeric mIgAs targeting different antigens in vivo showed distinct alterations in the function and metabolism of intestinal bacteria, largely through specific binding. Even in cases in which the same microbial antigen is targeted, microbial metabolic alterations differed depending on IgA epitope specificity. By contrast, bacterial surface coating generally reduced motility and limited bile acid toxicity. The overall intestinal IgA response to a single microbe therefore contains parallel components with distinct effects on microbial carbon-source uptake, bacteriophage susceptibility, motility and membrane integrity.


Subject(s)
Immunoglobulin A, Secretory/immunology , Intestines/immunology , Microbiota/immunology , Plasma Cells/immunology , Animals , Antibodies, Monoclonal/immunology , Antigens, Bacterial/immunology , Escherichia coli , Germ-Free Life , Mice , Mice, Inbred C57BL , Porins/immunology
13.
Nature ; 584(7820): 274-278, 2020 08.
Article in English | MEDLINE | ID: mdl-32760003

ABSTRACT

Colonization by the microbiota causes a marked stimulation of B cells and induction of immunoglobulin, but mammals colonized with many taxa have highly complex and individualized immunoglobulin repertoires1,2. Here we use a simplified model of defined transient exposures to different microbial taxa in germ-free mice3 to deconstruct how the microbiota shapes the B cell pool and its functional responsiveness. We followed the development of the immunoglobulin repertoire in B cell populations, as well as single cells by deep sequencing. Microbial exposures at the intestinal mucosa generated oligoclonal responses that differed from those of germ-free mice, and from the diverse repertoire that was generated after intravenous systemic exposure to microbiota. The IgA repertoire-predominantly to cell-surface antigens-did not expand after dose escalation, whereas increased systemic exposure broadened the IgG repertoire to both microbial cytoplasmic and cell-surface antigens. These microbial exposures induced characteristic immunoglobulin heavy-chain repertoires in B cells, mainly at memory and plasma cell stages. Whereas sequential systemic exposure to different microbial taxa diversified the IgG repertoire and facilitated alternative specific responses, sequential mucosal exposure produced limited overlapping repertoires and the attrition of initial IgA binding specificities. This shows a contrast between a flexible response to systemic exposure with the need to avoid fatal sepsis, and a restricted response to mucosal exposure that reflects the generic nature of host-microbial mutualism in the mucosa.


Subject(s)
B-Lymphocytes/cytology , B-Lymphocytes/immunology , Immunity, Mucosal/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Symbiosis/immunology , Administration, Intravenous , Administration, Oral , Animals , Clostridiales/immunology , Clostridiales/isolation & purification , Escherichia coli/immunology , Escherichia coli/isolation & purification , Female , Germ-Free Life , Immunoglobulin A/chemistry , Immunoglobulin A/immunology , Immunoglobulin G/chemistry , Immunoglobulin G/immunology , Immunoglobulin Heavy Chains/immunology , Immunologic Memory/immunology , Male , Mice , Mice, Inbred C57BL , Plasma Cells/cytology , Plasma Cells/immunology , Repetition Priming
14.
Nature ; 578(7794): 284-289, 2020 02.
Article in English | MEDLINE | ID: mdl-32025031

ABSTRACT

Neural control of the function of visceral organs is essential for homeostasis and health. Intestinal peristalsis is critical for digestive physiology and host defence, and is often dysregulated in gastrointestinal disorders1. Luminal factors, such as diet and microbiota, regulate neurogenic programs of gut motility2-5, but the underlying molecular mechanisms remain unclear. Here we show that the transcription factor aryl hydrocarbon receptor (AHR) functions as a biosensor in intestinal neural circuits, linking their functional output to the microbial environment of the gut lumen. Using nuclear RNA sequencing of mouse enteric neurons that represent distinct intestinal segments and microbiota states, we demonstrate that the intrinsic neural networks of the colon exhibit unique transcriptional profiles that are controlled by the combined effects of host genetic programs and microbial colonization. Microbiota-induced expression of AHR in neurons of the distal gastrointestinal tract enables these neurons to respond to the luminal environment and to induce expression of neuron-specific effector mechanisms. Neuron-specific deletion of Ahr, or constitutive overexpression of its negative feedback regulator CYP1A1, results in reduced peristaltic activity of the colon, similar to that observed in microbiota-depleted mice. Finally, expression of Ahr in the enteric neurons of mice treated with antibiotics partially restores intestinal motility. Together, our experiments identify AHR signalling in enteric neurons as a regulatory node that integrates the luminal environment with the physiological output of intestinal neural circuits to maintain gut homeostasis and health.


Subject(s)
Gastrointestinal Microbiome/physiology , Intestines/physiology , Neurons/physiology , Peristalsis , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cytochrome P-450 CYP1A1/metabolism , Female , Germ-Free Life , Intestines/innervation , Ligands , Male , Mice , Neural Pathways , Receptors, Aryl Hydrocarbon/metabolism , Signal Transduction , Transcriptome/genetics
15.
EMBO J ; 40(23): e108605, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34622466

ABSTRACT

The immune cells of the central nervous system (CNS) comprise parenchymal microglia and at the CNS border regions meningeal, perivascular, and choroid plexus macrophages (collectively called CNS-associated macrophages, CAMs). While previous work has shown that microglial properties depend on environmental signals from the commensal microbiota, the effects of microbiota on CAMs are unknown. By combining several microbiota manipulation approaches, genetic mouse models, and single-cell RNA-sequencing, we have characterized CNS myeloid cell composition and function. Under steady-state conditions, the transcriptional profiles and numbers of choroid plexus macrophages were found to be tightly regulated by complex microbiota. In contrast, perivascular and meningeal macrophages were affected to a lesser extent. An acute perturbation through viral infection evoked an attenuated immune response of all CAMs in germ-free mice. We further assessed CAMs in a more chronic pathological state in 5xFAD mice, a model for Alzheimer's disease, and found enhanced amyloid beta uptake exclusively by perivascular macrophages in germ-free 5xFAD mice. Our results aid the understanding of distinct microbiota-CNS macrophage interactions during homeostasis and disease, which could potentially be targeted therapeutically.


Subject(s)
Alzheimer Disease/immunology , Bacteria/growth & development , Central Nervous System/immunology , Homeostasis , Macrophages/immunology , Myeloid Cells/immunology , Alzheimer Disease/genetics , Alzheimer Disease/microbiology , Alzheimer Disease/pathology , Animals , Bacteria/classification , Bacteria/metabolism , Central Nervous System/metabolism , Central Nervous System/microbiology , Central Nervous System/pathology , Female , Macrophages/metabolism , Macrophages/microbiology , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Microbiota , Myeloid Cells/metabolism , Myeloid Cells/microbiology , Myeloid Cells/pathology , Transcriptome
16.
Immunity ; 44(6): 1312-24, 2016 06 21.
Article in English | MEDLINE | ID: mdl-27212436

ABSTRACT

How systemic metabolic alterations during acute infections impact immune cell function remains poorly understood. We found that acetate accumulates in the serum within hours of systemic bacterial infections and that these increased acetate concentrations are required for optimal memory CD8(+) T cell function in vitro and in vivo. Mechanistically, upon uptake by memory CD8(+) T cells, stress levels of acetate expanded the cellular acetyl-coenzyme A pool via ATP citrate lyase and promoted acetylation of the enzyme GAPDH. This context-dependent post-translational modification enhanced GAPDH activity, catalyzing glycolysis and thus boosting rapid memory CD8(+) T cell responses. Accordingly, in a murine Listeria monocytogenes model, transfer of acetate-augmented memory CD8(+) T cells exerted superior immune control compared to control cells. Our results demonstrate that increased systemic acetate concentrations are functionally integrated by CD8(+) T cells and translate into increased glycolytic and functional capacity. The immune system thus directly relates systemic metabolism with immune alertness.


Subject(s)
Acetates/metabolism , CD8-Positive T-Lymphocytes/immunology , Immunologic Memory , Listeria monocytogenes/immunology , Listeriosis/immunology , ATP Citrate (pro-S)-Lyase/metabolism , Acetyl-CoA C-Acetyltransferase/metabolism , Animals , CD8-Positive T-Lymphocytes/transplantation , Cells, Cultured , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating) , Glycolysis , Immunity, Innate , Mice , Mice, Inbred C57BL , Mice, Transgenic , Protein Processing, Post-Translational , Stress, Physiological/immunology
17.
PLoS Biol ; 20(9): e3001743, 2022 09.
Article in English | MEDLINE | ID: mdl-36126044

ABSTRACT

The capacity of the intestinal microbiota to degrade otherwise indigestible diet components is known to greatly improve the recovery of energy from food. This has led to the hypothesis that increased digestive efficiency may underlie the contribution of the microbiota to obesity. OligoMM12-colonized gnotobiotic mice have a consistently higher fat mass than germ-free (GF) or fully colonized counterparts. We therefore investigated their food intake, digestion efficiency, energy expenditure, and respiratory quotient using a novel isolator-housed metabolic cage system, which allows long-term measurements without contamination risk. This demonstrated that microbiota-released calories are perfectly balanced by decreased food intake in fully colonized versus gnotobiotic OligoMM12 and GF mice fed a standard chow diet, i.e., microbiota-released calories can in fact be well integrated into appetite control. We also observed no significant difference in energy expenditure after normalization by lean mass between the different microbiota groups, suggesting that cumulative small differences in energy balance, or altered energy storage, must underlie fat accumulation in OligoMM12 mice. Consistent with altered energy storage, major differences were observed in the type of respiratory substrates used in metabolism over the circadian cycle: In GF mice, the respiratory exchange ratio (RER) was consistently lower than that of fully colonized mice at all times of day, indicative of more reliance on fat and less on glucose metabolism. Intriguingly, the RER of OligoMM12-colonized gnotobiotic mice phenocopied fully colonized mice during the dark (active/eating) phase but phenocopied GF mice during the light (fasting/resting) phase. Further, OligoMM12-colonized mice showed a GF-like drop in liver glycogen storage during the light phase and both liver and plasma metabolomes of OligoMM12 mice clustered closely with GF mice. This implies the existence of microbiota functions that are required to maintain normal host metabolism during the resting/fasting phase of circadian cycle and which are absent in the OligoMM12 consortium.


Subject(s)
Liver Glycogen , Microbiota , Animals , Germ-Free Life , Glucose , Mice , Obesity/metabolism
18.
PLoS Pathog ; 18(4): e1009854, 2022 04.
Article in English | MEDLINE | ID: mdl-35446919

ABSTRACT

Interactions between pathogens, host microbiota and the immune system influence many physiological and pathological processes. In the 20th century, widespread dermal vaccination with vaccinia virus (VACV) led to the eradication of smallpox but how VACV interacts with the microbiota and whether this influences the efficacy of vaccination are largely unknown. Here we report that intradermal vaccination with VACV induces a large increase in the number of commensal bacteria in infected tissue, which enhance recruitment of inflammatory cells, promote tissue damage and influence the host response. Treatment of vaccinated specific-pathogen-free (SPF) mice with antibiotic, or infection of genetically-matched germ-free (GF) animals caused smaller lesions without alteration in virus titre. Tissue damage correlated with enhanced neutrophil and T cell infiltration and levels of pro-inflammatory tissue cytokines and chemokines. One month after vaccination, GF and both groups of SPF mice had equal numbers of VACV-specific CD8+ T cells and were protected from disease induced by VACV challenge, despite lower levels of VACV-neutralising antibodies observed in GF animals. Thus, skin microbiota may provide an adjuvant-like stimulus during vaccination with VACV and influence the host response to vaccination.


Subject(s)
Smallpox , Vaccinia , Animals , Antibodies, Viral , Bacteria , Mice , Smallpox/prevention & control , Vaccination , Vaccinia virus
19.
Immunity ; 43(3): 416-8, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26377894

ABSTRACT

IgA is induced through T-cell-dependent and -independent pathways. In this issue, Bunker et al. (2015) now show that the T-cell-independent pathway is sufficient to coat most small intestinal microbes specifically, and Fransen et al. (2015) find that IgA coating promotes uptake of microbes into Peyer's patches and drives further induction in a positive-feedback loop.


Subject(s)
Adaptive Immunity/immunology , Antigens, Bacterial/immunology , Bacteria/immunology , Genetic Variation/immunology , Immunity, Humoral/immunology , Immunity, Innate/immunology , Immunoglobulin A/immunology , Intestine, Small/immunology , Microbiota/immunology , Animals , Humans
20.
Immunity ; 43(5): 998-1010, 2015 Nov 17.
Article in English | MEDLINE | ID: mdl-26522986

ABSTRACT

Intestinal helminths are potent regulators of their host's immune system and can ameliorate inflammatory diseases such as allergic asthma. In the present study we have assessed whether this anti-inflammatory activity was purely intrinsic to helminths, or whether it also involved crosstalk with the local microbiota. We report that chronic infection with the murine helminth Heligmosomoides polygyrus bakeri (Hpb) altered the intestinal habitat, allowing increased short chain fatty acid (SCFA) production. Transfer of the Hpb-modified microbiota alone was sufficient to mediate protection against allergic asthma. The helminth-induced anti-inflammatory cytokine secretion and regulatory T cell suppressor activity that mediated the protection required the G protein-coupled receptor (GPR)-41. A similar alteration in the metabolic potential of intestinal bacterial communities was observed with diverse parasitic and host species, suggesting that this represents an evolutionary conserved mechanism of host-microbe-helminth interactions.


Subject(s)
Gastrointestinal Microbiome/immunology , Helminths/immunology , Hypersensitivity/immunology , Inflammation/immunology , Inflammation/parasitology , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Adult , Aged , Animals , Asthma/immunology , Asthma/microbiology , Asthma/parasitology , Cytokines/immunology , Fatty Acids/immunology , Female , Humans , Hypersensitivity/microbiology , Hypersensitivity/parasitology , Inflammation/microbiology , Intestinal Mucosa/parasitology , Male , Mice , Mice, Inbred C57BL , Middle Aged , Nematospiroides dubius/immunology , Receptors, G-Protein-Coupled/immunology , Strongylida Infections/immunology , Strongylida Infections/microbiology , Strongylida Infections/parasitology
SELECTION OF CITATIONS
SEARCH DETAIL