Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 126
Filter
Add more filters

Publication year range
1.
Cell ; 184(21): 5301-5303, 2021 10 14.
Article in English | MEDLINE | ID: mdl-34624223

ABSTRACT

The gut microbiota has been shown to promote the efficacy of cancer therapy through regulating adaptive immune responses. In this issue of Cell, Lam et al. provide new evidence demonstrating that specific gut bacteria also reprogram the innate immune tumor microenvironment to enhance the efficacy of cancer therapies.


Subject(s)
Microbiota , Neoplasms , Humans , Immunity , Monocytes , Neoplasms/therapy , Tumor Microenvironment
2.
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
3.
Immunity ; 55(7): 1250-1267.e12, 2022 07 12.
Article in English | MEDLINE | ID: mdl-35709757

ABSTRACT

The intestine harbors a large population of resident eosinophils, yet the function of intestinal eosinophils has not been explored. Flow cytometry and whole-mount imaging identified eosinophils residing in the lamina propria along the length of the intestine prior to postnatal microbial colonization. Microscopy, transcriptomic analysis, and mass spectrometry of intestinal tissue revealed villus blunting, altered extracellular matrix, decreased epithelial cell turnover, increased gastrointestinal motility, and decreased lipid absorption in eosinophil-deficient mice. Mechanistically, intestinal epithelial cells released IL-33 in a microbiota-dependent manner, which led to eosinophil activation. The colonization of germ-free mice demonstrated that eosinophil activation in response to microbes regulated villous size alterations, macrophage maturation, epithelial barrier integrity, and intestinal transit. Collectively, our findings demonstrate a critical role for eosinophils in facilitating the mutualistic interactions between the host and microbiota and provide a rationale for the functional significance of their early life recruitment in the small intestine.


Subject(s)
Communicable Diseases , Microbiota , Animals , Eosinophils , Homeostasis , Intestinal Mucosa , Intestine, Small , Mice
4.
Nat Immunol ; 19(10): 1100-1111, 2018 10.
Article in English | MEDLINE | ID: mdl-30250184

ABSTRACT

Females have an overall advantage over males in resisting Gram-negative bacteremias, thus hinting at sexual dimorphism of immunity during infections. Here, through intravital microscopy, we observed a sex-biased difference in the capture of blood-borne bacteria by liver macrophages, a process that is critical for the clearance of systemic infections. Complement opsonization was indispensable for the capture of enteropathogenic Escherichia coli (EPEC) in male mice; however, a faster complement component 3-independent process involving abundant preexisting antibodies to EPEC was detected in female mice. These antibodies were elicited predominantly in female mice at puberty in response to estrogen regardless of microbiota-colonization conditions. Estrogen-driven antibodies were maternally transferrable to offspring and conferred protection during infancy. These antibodies were conserved in humans and recognized specialized oligosaccharides integrated into the bacterial lipopolysaccharide and capsule. Thus, an estrogen-driven, innate antibody-mediated immunological strategy conferred protection to females and their offspring.


Subject(s)
Antibodies, Bacterial/immunology , Escherichia coli Infections/immunology , Immunity, Innate/immunology , Sex Characteristics , Animals , Enteropathogenic Escherichia coli , Estrogens/immunology , Female , Humans , Infant , Kupffer Cells/immunology , Male , Maternal-Fetal Exchange/immunology , Mice , Pregnancy
5.
Nat Immunol ; 17(12): 1388-1396, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27798617

ABSTRACT

Fibroblastic reticular cells (FRCs) of secondary lymphoid organs form distinct niches for interaction with hematopoietic cells. We found here that production of the cytokine IL-15 by FRCs was essential for the maintenance of group 1 innate lymphoid cells (ILCs) in Peyer's patches and mesenteric lymph nodes. Moreover, FRC-specific ablation of the innate immunological sensing adaptor MyD88 unleashed IL-15 production by FRCs during infection with an enteropathogenic virus, which led to hyperactivation of group 1 ILCs and substantially altered the differentiation of helper T cells. Accelerated clearance of virus by group 1 ILCs precipitated severe intestinal inflammatory disease with commensal dysbiosis, loss of intestinal barrier function and diminished resistance to colonization. In sum, FRCs act as an 'on-demand' immunological 'rheostat' by restraining activation of group 1 ILCs and thereby preventing immunopathological damage in the intestine.


Subject(s)
Citrobacter rodentium/immunology , Coronavirus Infections/immunology , Enterobacteriaceae Infections/immunology , Fibroblasts/immunology , Interleukin-15/metabolism , Lymphocytes/immunology , Murine hepatitis virus/immunology , Animals , Cells, Cultured , Immunity, Innate , Lymph Nodes/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Peyer's Patches/pathology , Th1 Cells/immunology , Toll-Like Receptor 7/genetics , Toll-Like Receptor 7/metabolism
6.
Immunity ; 51(1): 12-14, 2019 07 16.
Article in English | MEDLINE | ID: mdl-31315030

ABSTRACT

Appropriate priming of tolerogenic or effector immune responses is crucial for intestinal homeostasis. Recently in Nature, Esterházy et al. (2019) reveal how compartmentalization of lymphatic drainage to functionally distinct lymph nodes facilitates the simultaneous induction of tolerogenic and effector responses.


Subject(s)
Drainage , Lymph Nodes , Adaptive Immunity , Intestines
7.
Semin Immunol ; 73: 101885, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38788491

ABSTRACT

The gut microbiota is well known to possess immunomodulatory capacities, influencing a multitude of cellular signalling pathways to maintain host homeostasis. Although the formation of the immune system initiates before birth in a sterile environment, an emerging body of literature indicates that the neonatal immune system is influenced by a first wave of external stimuli that includes signals from the maternal microbiota. A second wave of stimulus begins after birth and must be tightly regulated during the neonatal period when colonization of the host occurs concomitantly with the maturation of the immune system, requiring a fine adjustment between establishing tolerance towards the commensal microbiota and preserving inflammatory responses against pathogenic invaders. Besides integrating cues from commensal microbes, the neonatal immune system must also regulate responses triggered by other environmental signals, such as dietary antigens, which become more complex with the introduction of solid food during the weaning period. This "window of opportunity" in early life is thought to be crucial for the proper development of the immune system, setting the tone of subsequent immune responses in adulthood and modulating the risk of developing chronic and metabolic inflammatory diseases. Here we review the importance of host-microbiota interactions for the development and maturation of the immune system, particularly in the early-life period, highlighting the known mechanisms involved in such communication. This discussion is focused on recent data demonstrating microbiota-mediated education of innate immune cells and its role in the development of lymphoid tissues.

8.
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
9.
Semin Immunol ; 67: 101755, 2023 05.
Article in English | MEDLINE | ID: mdl-36989542

ABSTRACT

Our microbiota has a critical role in shaping host immunity. Microbes that reside in the gut harbor a large metabolic arsenal to aid in physiological functions of the host. Microbial metabolites, which are products of microbial metabolism, such as short chain fatty acids (SCFA), purine metabolites, cyclic dinucleotides, tryptophan derivatives, and secondary bile acids, can tailor the host immune cell landscape in homeostasis and during cancer immunotherapy. The critical role of the microbiome in aiding immune checkpoint blockade therapies has become clearer over the past few years, with the most recent studies providing more detailed mechanistic insight on how microbes and their metabolites control the outcome of immunotherapy. This review summarizes recent studies on how microbial metabolites orchestrate immune responses during cancer immunotherapies.


Subject(s)
Gastrointestinal Microbiome , Microbiota , Neoplasms , Humans , Immunotherapy , Homeostasis , Neoplasms/therapy
10.
Immunity ; 46(4): 532-548, 2017 04 18.
Article in English | MEDLINE | ID: mdl-28423335

ABSTRACT

The intestinal immune system has the daunting task of protecting us from pathogenic insults while limiting inflammatory responses against the resident commensal microbiota and providing tolerance to food antigens. This role is particularly impressive when one considers the vast mucosal surface and changing landscape that the intestinal immune system must monitor. In this review, we highlight regional differences in the development and composition of the adaptive immune landscape of the intestine and the impact of local intrinsic and environmental factors that shape this process. To conclude, we review the evidence for a critical window of opportunity for early-life exposures that affect immune development and alter disease susceptibility later in life.


Subject(s)
Adaptive Immunity/immunology , Gastrointestinal Microbiome/immunology , Immune System/immunology , Intestinal Mucosa/immunology , Intestines/immunology , Animals , Gastrointestinal Microbiome/physiology , Host-Pathogen Interactions/immunology , Humans , Immune System/microbiology , Immune Tolerance/immunology , Intestinal Mucosa/microbiology , Intestines/microbiology , Models, Immunological
11.
Immunity ; 47(2): 339-348.e4, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28801232

ABSTRACT

The gut microbiota regulate susceptibility to multiple human diseases. The Nlrp6-ASC inflammasome is widely regarded as a hallmark host innate immune axis that shapes the gut microbiota composition. This notion stems from studies reporting dysbiosis in mice lacking these inflammasome components when compared with non-littermate wild-type animals. Here, we describe microbial analyses in inflammasome-deficient mice while minimizing non-genetic confounders using littermate-controlled Nlrp6-deficient mice and ex-germ-free littermate-controlled ASC-deficient mice that were all allowed to shape their gut microbiota naturally after birth. Careful microbial phylogenetic analyses of these cohorts failed to reveal regulation of the gut microbiota composition by the Nlrp6- and ASC-dependent inflammasomes. Our results obtained in two geographically separated animal facilities dismiss a generalizable impact of Nlrp6- and ASC-dependent inflammasomes on the composition of the commensal gut microbiota and highlight the necessity for littermate-controlled experimental design in assessing the influence of host immunity on gut microbial ecology.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Bacteria/genetics , Colitis/immunology , Dysbiosis/immunology , Gastrointestinal Microbiome/immunology , Inflammasomes/metabolism , Receptors, Cell Surface/metabolism , Animals , CARD Signaling Adaptor Proteins , Cells, Cultured , Colitis/chemically induced , Colitis/microbiology , Dysbiosis/microbiology , Female , Genetic Background , Immunity, Innate , Mice , Mice, Inbred C57BL , Mice, Knockout , Microbiota , RNA, Ribosomal, 16S/analysis , Receptors, Cell Surface/genetics , Sodium Dodecyl Sulfate
12.
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
13.
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
14.
Am J Respir Crit Care Med ; 207(1): 38-49, 2023 01 01.
Article in English | MEDLINE | ID: mdl-35926164

ABSTRACT

Rationale: Coronavirus disease 2019 (COVID-19) can lead to acute respiratory distress syndrome with fatal outcomes. Evidence suggests that dysregulated immune responses, including autoimmunity, are key pathogenic factors. Objectives: To assess whether IgA autoantibodies target lung-specific proteins and contribute to disease severity. Methods: We collected 147 blood, 9 lung tissue, and 36 BAL fluid samples from three tertiary hospitals in Switzerland and one in Germany. Severe COVID-19 was defined by the need to administer oxygen. We investigated the presence of IgA autoantibodies and their effects on pulmonary surfactant in COVID-19 using the following methods: immunofluorescence on tissue samples, immunoprecipitations followed by mass spectrometry on BAL fluid samples, enzyme-linked immunosorbent assays on blood samples, and surface tension measurements with medical surfactant. Measurements and Main Results: IgA autoantibodies targeting pulmonary surfactant proteins B and C were elevated in patients with severe COVID-19 but not in patients with influenza or bacterial pneumonia. Notably, pulmonary surfactant failed to reduce surface tension after incubation with either plasma or purified IgA from patients with severe COVID-19. Conclusions: Our data suggest that patients with severe COVID-19 harbor IgA autoantibodies against pulmonary surfactant proteins B and C and that these autoantibodies block the function of lung surfactant, potentially contributing to alveolar collapse and poor oxygenation.


Subject(s)
COVID-19 , Pulmonary Surfactants , Humans , Pulmonary Surfactants/metabolism , Bronchoalveolar Lavage Fluid/chemistry , Surface-Active Agents , Autoantibodies , Immunoglobulin A
15.
FASEB J ; 36(5): e22269, 2022 05.
Article in English | MEDLINE | ID: mdl-35344215

ABSTRACT

Dietary fiber promotes a healthy gut microbiome and shows promise in attenuating the unfavorable microbial changes resulting from a high-fat/sucrose (HFS) diet. High-fiber diets consisting of oligofructose alone (HFS/O) or in combination with ß-glucan (HFS/OB), resistant starch (HFS/OR), or ß-glucan and resistant starch (HFS/OBR) were fed to diet-induced obese rats for 8 weeks to determine if these fibers could attenuate the obese phenotype. Only the HFS/O group displayed a decrease in body weight and body fat, but all fiber interventions improved insulin sensitivity and cognitive function. The HFS/O diet was the least effective at improving cognitive function and only the HFS/OB group showed improvements in glucose tolerance, thus highlighting the differential effects of fiber types. Hippocampal cytokines (IL-6, IL-10) were more pronounced in the HFS/OB group which coincided with the most time spend in the open arms of the elevated plus maze. All fiber groups showed an increase in beneficial Bifidobacterium and Lactobacillus abundance while the HFS group showed higher abundance of Clostridium. Fecal microbiota transplant from fiber-treated rats into germ-free mice did not alter body composition in the mice but did result in a higher abundance of Bacteroides in the HFS/O and HFS/OB groups compared to HFS. The HFS/OB recipient mice also had higher insulin sensitivity compared to the other groups. This study highlights the influence of dietary fiber type on metabolic and cognitive outcomes suggesting that the type of supplementation (single or combined fibers) could be tailored to specific targeted outcomes.


Subject(s)
Insulin Resistance , beta-Glucans , Animals , Cognition , Diet, High-Fat/adverse effects , Dietary Fiber/pharmacology , Mice , Obesity/metabolism , Rats , Resistant Starch , Sucrose
16.
Immunity ; 41(5): 789-801, 2014 Nov 20.
Article in English | MEDLINE | ID: mdl-25464855

ABSTRACT

Microbial colonization of the gut induces the development of gut-associated lymphoid tissue (GALT). The molecular mechanisms that regulate GALT function and result in gut-commensal homeostasis are poorly defined. T follicular helper (Tfh) cells in Peyer's patches (PPs) promote high-affinity IgA responses. Here we found that the ATP-gated ionotropic P2X7 receptor controls Tfh cell numbers in PPs. Lack of P2X7 in Tfh cells enhanced germinal center reactions and high-affinity IgA secretion and binding to commensals. The ensuing depletion of mucosal bacteria resulted in reduced systemic translocation of microbial components, lowering B1 cell stimulation and serum IgM concentrations. Mice lacking P2X7 had increased susceptibility to polymicrobial sepsis, which was rescued by Tfh cell depletion or administration of purified IgM. Thus, regulation of Tfh cells by P2X7 activity is important for mucosal colonization, which in turn results in IgM serum concentrations necessary to protect the host from bacteremia.


Subject(s)
Intestinal Mucosa/immunology , Peyer's Patches/immunology , Receptors, Purinergic P2X7/immunology , Symbiosis/immunology , T-Lymphocytes, Helper-Inducer/immunology , Adenosine Triphosphate/metabolism , Animals , B-Lymphocytes/immunology , Bacteremia/immunology , Genetic Predisposition to Disease , Germinal Center/immunology , Humans , Immunoglobulin A/immunology , Immunoglobulin M/blood , Intestinal Mucosa/microbiology , Lymphocyte Depletion , Mice , Mice, Inbred C57BL , Mice, Knockout , Microbiota/immunology , Peyer's Patches/cytology , Receptors, Purinergic P2X7/genetics , Sepsis/immunology , Sepsis/microbiology
17.
Immunity ; 40(4): 608-20, 2014 Apr 17.
Article in English | MEDLINE | ID: mdl-24745335

ABSTRACT

Segmented filamentous bacterium (SFB) is a symbiont that drives postnatal maturation of gut adaptive immune responses. In contrast to nonpathogenic E. coli, SFB stimulated vigorous development of Peyer's patches germinal centers but paradoxically induced only a low frequency of specific immunoglobulin A (IgA)-secreting cells with delayed accumulation of somatic mutations. Moreover, blocking Peyer's patch development abolished IgA responses to E. coli, but not to SFB. Indeed, SFB stimulated the postnatal development of isolated lymphoid follicles and tertiary lymphoid tissue, which substituted for Peyer's patches as inductive sites for intestinal IgA and SFB-specific T helper 17 (Th17) cell responses. Strikingly, in mice depleted of gut organized lymphoid tissue, SFB still induced a substantial but nonspecific intestinal Th17 cell response. These results demonstrate that SFB has the remarkable capacity to induce and stimulate multiple types of intestinal lymphoid tissues that cooperate to generate potent IgA and Th17 cell responses displaying only limited target specificity.


Subject(s)
Clostridium Infections/immunology , Clostridium/immunology , Escherichia coli Infections/immunology , Escherichia coli/immunology , Immunoglobulin A/metabolism , Intestines/immunology , Plasma Cells/immunology , Th17 Cells/immunology , Animals , Antigens, Bacterial/immunology , Cell Communication , Cell Differentiation , Host-Pathogen Interactions , Lymphoid Tissue/immunology , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Peyer's Patches/immunology
18.
Int J Mol Sci ; 24(8)2023 Apr 15.
Article in English | MEDLINE | ID: mdl-37108484

ABSTRACT

Diet influences the pathogenesis and clinical course of inflammatory bowel disease (IBD). The Mediterranean diet (MD) is linked to reductions in inflammatory biomarkers and alterations in microbial taxa and metabolites associated with health. We aimed to identify features of the gut microbiome that mediate the relationship between the MD and fecal calprotectin (FCP) in ulcerative colitis (UC). Weighted gene co-expression network analysis (WGCNA) was used to identify modules of co-abundant microbial taxa and metabolites correlated with the MD and FCP. The features considered were gut microbial taxa, serum metabolites, dietary components, short-chain fatty acid and bile acid profiles in participants that experienced an increase (n = 13) or decrease in FCP (n = 16) over eight weeks. WGCNA revealed ten modules containing sixteen key features that acted as key mediators between the MD and FCP. Three taxa (Faecalibacterium prausnitzii, Dorea longicatena, Roseburia inulinivorans) and a cluster of four metabolites (benzyl alcohol, 3-hydroxyphenylacetate, 3-4-hydroxyphenylacetate and phenylacetate) demonstrated a strong mediating effect (ACME: -1.23, p = 0.004). This study identified a novel association between diet, inflammation and the gut microbiome, providing new insights into the underlying mechanisms of how a MD may influence IBD. See clinicaltrials.gov (NCT04474561).


Subject(s)
Colitis, Ulcerative , Diet, Mediterranean , Inflammatory Bowel Diseases , Humans , Colitis, Ulcerative/microbiology , Inflammatory Bowel Diseases/microbiology , Inflammation/genetics , Biomarkers , Feces/microbiology
19.
EMBO J ; 37(14)2018 07 13.
Article in English | MEDLINE | ID: mdl-29752423

ABSTRACT

Virtual memory T cells are foreign antigen-inexperienced T cells that have acquired memory-like phenotype and constitute 10-20% of all peripheral CD8+ T cells in mice. Their origin, biological roles, and relationship to naïve and foreign antigen-experienced memory T cells are incompletely understood. By analyzing T-cell receptor repertoires and using retrogenic monoclonal T-cell populations, we demonstrate that the virtual memory T-cell formation is a so far unappreciated cell fate decision checkpoint. We describe two molecular mechanisms driving the formation of virtual memory T cells. First, virtual memory T cells originate exclusively from strongly self-reactive T cells. Second, the stoichiometry of the CD8 interaction with Lck regulates the size of the virtual memory T-cell compartment via modulating the self-reactivity of individual T cells. Although virtual memory T cells descend from the highly self-reactive clones and acquire a partial memory program, they are not more potent in inducing experimental autoimmune diabetes than naïve T cells. These data underline the importance of the variable level of self-reactivity in polyclonal T cells for the generation of functional T-cell diversity.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cell Differentiation , Immunologic Memory , Receptors, Antigen, T-Cell/analysis , Animals , Homeostasis , Mice
SELECTION OF CITATIONS
SEARCH DETAIL