Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 346
Filter
Add more filters

Publication year range
2.
Nat Immunol ; 22(5): 607-619, 2021 05.
Article in English | MEDLINE | ID: mdl-33833438

ABSTRACT

FOXP3 deficiency in mice and in patients with immune dysregulation polyendocrinopathy enteropathy X-linked (IPEX) syndrome results in fatal autoimmunity by altering regulatory T (Treg) cells. CD4+ T cells in patients with IPEX syndrome and Foxp3-deficient mice were analyzed by single-cell cytometry and RNA-sequencing, revealing heterogeneous Treg-like cells, some very similar to normal Treg cells, others more distant. Conventional T cells showed no widespread activation or helper T cell bias, but a monomorphic disease signature affected all CD4+ T cells. This signature proved to be cell extrinsic since it was extinguished in mixed bone marrow chimeric mice and heterozygous mothers of patients with IPEX syndrome. Normal Treg cells exerted dominant suppression, quenching the disease signature and revealing in mutant Treg-like cells a small cluster of genes regulated cell-intrinsically by FOXP3, including key homeostatic regulators. We propose a two-step pathogenesis model: cell-intrinsic downregulation of core FOXP3-dependent genes destabilizes Treg cells, de-repressing systemic mediators that imprint the disease signature on all T cells, furthering Treg cell dysfunction. Accordingly, interleukin-2 treatment improved the Treg-like compartment and survival.


Subject(s)
Diabetes Mellitus, Type 1/congenital , Diarrhea/genetics , Forkhead Transcription Factors/deficiency , Genetic Diseases, X-Linked/genetics , Immune System Diseases/congenital , T-Lymphocytes, Regulatory/immunology , Adolescent , Animals , Case-Control Studies , Child , Child, Preschool , Cohort Studies , Datasets as Topic , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Diarrhea/blood , Diarrhea/immunology , Disease Models, Animal , Flow Cytometry , Forkhead Transcription Factors/genetics , Genetic Diseases, X-Linked/blood , Genetic Diseases, X-Linked/immunology , Humans , Immune System Diseases/blood , Immune System Diseases/genetics , Immune System Diseases/immunology , Infant , Male , Mice , Mice, Transgenic , Mutation , RNA-Seq , Single-Cell Analysis , T-Lymphocytes, Regulatory/metabolism , Young Adult
3.
Nat Immunol ; 21(11): 1359-1370, 2020 11.
Article in English | MEDLINE | ID: mdl-32929274

ABSTRACT

Elucidating the mechanisms that sustain asthmatic inflammation is critical for precision therapies. We found that interleukin-6- and STAT3 transcription factor-dependent upregulation of Notch4 receptor on lung tissue regulatory T (Treg) cells is necessary for allergens and particulate matter pollutants to promote airway inflammation. Notch4 subverted Treg cells into the type 2 and type 17 helper (TH2 and TH17) effector T cells by Wnt and Hippo pathway-dependent mechanisms. Wnt activation induced growth and differentiation factor 15 expression in Treg cells, which activated group 2 innate lymphoid cells to provide a feed-forward mechanism for aggravated inflammation. Notch4, Wnt and Hippo were upregulated in circulating Treg cells of individuals with asthma as a function of disease severity, in association with reduced Treg cell-mediated suppression. Our studies thus identify Notch4-mediated immune tolerance subversion as a fundamental mechanism that licenses tissue inflammation in asthma.


Subject(s)
Asthma/etiology , Asthma/metabolism , Growth Differentiation Factor 15/metabolism , Receptor, Notch4/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Allergens/immunology , Analysis of Variance , Asthma/diagnosis , Biomarkers , Disease Susceptibility , Gene Expression , Hippo Signaling Pathway , Humans , Immune Tolerance , Immunophenotyping , Protein Serine-Threonine Kinases/metabolism , Severity of Illness Index , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Wnt Signaling Pathway
4.
Nat Immunol ; 20(9): 1208-1219, 2019 09.
Article in English | MEDLINE | ID: mdl-31384057

ABSTRACT

Regulatory T cells (Treg cells) deficient in the transcription factor Foxp3 lack suppressor function and manifest an effector T (Teff) cell-like phenotype. We demonstrate that Foxp3 deficiency dysregulates metabolic checkpoint kinase mammalian target of rapamycin (mTOR) complex 2 (mTORC2) signaling and gives rise to augmented aerobic glycolysis and oxidative phosphorylation. Specific deletion of the mTORC2 adaptor gene Rictor in Foxp3-deficient Treg cells ameliorated disease in a Foxo1 transcription factor-dependent manner. Rictor deficiency re-established a subset of Treg cell genetic circuits and suppressed the Teff cell-like glycolytic and respiratory programs, which contributed to immune dysregulation. Treatment of Treg cells from patients with FOXP3 deficiency with mTOR inhibitors similarly antagonized their Teff cell-like program and restored suppressive function. Thus, regulatory function can be re-established in Foxp3-deficient Treg cells by targeting their metabolic pathways, providing opportunities to restore tolerance in Treg cell disorders.


Subject(s)
Cellular Reprogramming/immunology , Forkhead Transcription Factors/genetics , Mechanistic Target of Rapamycin Complex 2/metabolism , Rapamycin-Insensitive Companion of mTOR Protein/genetics , T-Lymphocytes, Regulatory/immunology , Animals , Cells, Cultured , Female , Gene Expression Regulation , Glycolysis/physiology , Humans , Male , Mechanistic Target of Rapamycin Complex 2/antagonists & inhibitors , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidative Phosphorylation , Signal Transduction , T-Lymphocytes, Regulatory/cytology
5.
Immunity ; 55(11): 1978-1980, 2022 11 08.
Article in English | MEDLINE | ID: mdl-36351372

ABSTRACT

RORγt+ regulatory T (Treg) cells are critical toward maintaining gut immune tolerance. In recent studies published in Nature, Kedmi et al., Lyu et al., and Akagbosu et al. describe MHCII+RORγt+ antigen-presenting cells that mediate RORγt+ Treg cell differentiation but propose disparate identities for these cells.


Subject(s)
Nuclear Receptor Subfamily 1, Group F, Member 3 , Peripheral Tolerance , Embarrassment , T-Lymphocytes, Regulatory , Antigen-Presenting Cells , Th17 Cells , Forkhead Transcription Factors , Immune Tolerance
6.
Immunity ; 55(7): 1173-1184.e7, 2022 07 12.
Article in English | MEDLINE | ID: mdl-35700740

ABSTRACT

Regulatory T (Treg) cells expressing the transcription factor Foxp3 are an essential suppressive T cell lineage of dual origin: Foxp3 induction in thymocytes and mature CD4+ T cells gives rise to thymic (tTreg) and peripheral (pTreg) Treg cells, respectively. While tTreg cells suppress autoimmunity, pTreg cells enforce tolerance to food and commensal microbiota. However, the role of Foxp3 in pTreg cells and the mechanisms supporting their differentiation remain poorly understood. Here, we used genetic tracing to identify microbiota-induced pTreg cells and found that many of their distinguishing features were Foxp3 independent. Lineage-committed, microbiota-dependent pTreg-like cells persisted in the colon in the absence of Foxp3. While Foxp3 was critical for the suppression of a Th17 cell program, colitis, and mastocytosis, pTreg cells suppressed colonic effector T cell expansion in a Foxp3-independent manner. Thus, Foxp3 and the tolerogenic signals that precede and promote its expression independently confer distinct facets of pTreg functionality.


Subject(s)
Forkhead Transcription Factors , T-Lymphocytes, Regulatory , Forkhead Transcription Factors/metabolism , Immune Tolerance , Th17 Cells/metabolism , Thymocytes/metabolism
7.
Immunity ; 54(6): 1186-1199.e7, 2021 06 08.
Article in English | MEDLINE | ID: mdl-33915108

ABSTRACT

A cardinal feature of COVID-19 is lung inflammation and respiratory failure. In a prospective multi-country cohort of COVID-19 patients, we found that increased Notch4 expression on circulating regulatory T (Treg) cells was associated with disease severity, predicted mortality, and declined upon recovery. Deletion of Notch4 in Treg cells or therapy with anti-Notch4 antibodies in conventional and humanized mice normalized the dysregulated innate immunity and rescued disease morbidity and mortality induced by a synthetic analog of viral RNA or by influenza H1N1 virus. Mechanistically, Notch4 suppressed the induction by interleukin-18 of amphiregulin, a cytokine necessary for tissue repair. Protection by Notch4 inhibition was recapitulated by therapy with Amphiregulin and, reciprocally, abrogated by its antagonism. Amphiregulin declined in COVID-19 subjects as a function of disease severity and Notch4 expression. Thus, Notch4 expression on Treg cells dynamically restrains amphiregulin-dependent tissue repair to promote severe lung inflammation, with therapeutic implications for COVID-19 and related infections.


Subject(s)
Host-Pathogen Interactions , Immunity, Cellular , Pneumonia, Viral/etiology , Pneumonia, Viral/metabolism , Receptor, Notch4/metabolism , Signal Transduction , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Amphiregulin/pharmacology , Animals , Biomarkers , Cytokines/metabolism , Disease Models, Animal , Disease Susceptibility , Host-Pathogen Interactions/immunology , Humans , Immunohistochemistry , Immunomodulation/drug effects , Inflammation Mediators/metabolism , Influenza A virus/physiology , Lung/immunology , Lung/metabolism , Lung/pathology , Lung/virology , Mice , Mice, Transgenic , Pneumonia, Viral/pathology , Receptor, Notch4/antagonists & inhibitors , Receptor, Notch4/genetics , Severity of Illness Index
9.
Immunity ; 53(2): 277-289, 2020 08 18.
Article in English | MEDLINE | ID: mdl-32814026

ABSTRACT

The steep rise in food allergy (FA) has evoked environmental factors involved in disease pathogenesis, including the gut microbiota, diet, and their metabolites. Early introduction of solid foods synchronizes with the "weaning reaction," a time during which the microbiota imprints durable oral tolerance. Recent work has shown that children with FA manifest an early onset dysbiosis with the loss of Clostridiales species, which promotes the differentiation of ROR-γt+ regulatory T cells to suppress FA. This process can be reversed in pre-clinical mouse models by targeted bacteriotherapy. Here, we review the dominant tolerance mechanisms enforced by the microbiota to suppress FA and discuss therapeutic intervention strategies that act to recapitulate the early life window of opportunity in stemming the FA epidemic.


Subject(s)
Diet , Dysbiosis/microbiology , Food Hypersensitivity/immunology , Gastrointestinal Microbiome/physiology , Animals , Clostridiales/isolation & purification , Desensitization, Immunologic/methods , Humans , Immune Tolerance/immunology , Immunoglobulin E/immunology , Mice , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology
10.
Immunity ; 53(5): 971-984.e5, 2020 11 17.
Article in English | MEDLINE | ID: mdl-33176163

ABSTRACT

Regulatory T (Treg) cell identity is defined by the lineage-specifying transcription factor (TF) Foxp3. Here we examined mechanisms of Foxp3 function by leveraging naturally occurring genetic variation in wild-derived inbred mice, which enables the identification of DNA sequence motifs driving epigenetic features. Chromatin accessibility, TF binding, and gene expression patterns in resting and activated subsets of Treg cells, conventional CD4 T cells, and cells expressing a Foxp3 reporter null allele revealed that the majority of Foxp3-dependent changes occurred at sites not bound by Foxp3. Chromatin accessibility of these indirect Foxp3 targets depended on the presence of DNA binding motifs for other TFs, including TCF1. Foxp3 expression correlated with decreased TCF1 and reduced accessibility of TCF1-bound chromatin regions. Deleting one copy of the Tcf7 gene recapitulated Foxp3-dependent negative regulation of chromatin accessibility. Thus, Foxp3 defines Treg cell identity in a largely indirect manner by fine-tuning the activity of other major chromatin remodeling TFs such as TCF1.


Subject(s)
Forkhead Transcription Factors/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Animals , Autoimmune Diseases/etiology , Autoimmune Diseases/metabolism , Autoimmune Diseases/pathology , Autoimmunity/genetics , Binding Sites , Chromatin Assembly and Disassembly , Disease Models, Animal , Epigenesis, Genetic , Female , Forkhead Transcription Factors/genetics , Gene Expression Regulation , Immunohistochemistry , Male , Mice , Nucleotide Motifs , Organ Specificity/genetics , Organ Specificity/immunology , Protein Binding , Trans-Activators/metabolism
11.
Immunity ; 53(6): 1202-1214.e6, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33086036

ABSTRACT

The mechanisms by which regulatory T (Treg) cells differentially control allergic and autoimmune responses remain unclear. We show that Treg cells in food allergy (FA) had decreased expression of transforming growth factor beta 1 (TGF-ß1) because of interleukin-4 (IL-4)- and signal transducer and activator of transciription-6 (STAT6)-dependent inhibition of Tgfb1 transcription. These changes were modeled by Treg cell-specific Tgfb1 monoallelic inactivation, which induced allergic dysregulation by impairing microbiota-dependent retinoic acid receptor-related orphan receptor gamma t (ROR-γt)+ Treg cell differentiation. This dysregulation was rescued by treatment with Clostridiales species, which upregulated Tgfb1 expression in Treg cells. Biallelic deficiency precipitated fatal autoimmunity with intense autoantibody production and dysregulated T follicular helper and B cell responses. These results identify a privileged role of Treg cell-derived TGF-ß1 in regulating allergy and autoimmunity at distinct checkpoints in a Tgfb1 gene dose- and microbiota-dependent manner.


Subject(s)
Autoimmunity/immunology , Hypersensitivity/immunology , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta1/immunology , Adolescent , Animals , Autoimmunity/genetics , B-Lymphocytes/immunology , Cell Differentiation , Child , Child, Preschool , Food Hypersensitivity/immunology , Gene Dosage , Humans , Hypersensitivity/genetics , Immunoglobulin G/immunology , Infant , Mast Cells/immunology , Mice , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , T Follicular Helper Cells/immunology , T-Lymphocytes, Regulatory/metabolism , Transcription, Genetic , Transforming Growth Factor beta1/genetics , Young Adult
13.
Immunol Rev ; 326(1): 219-226, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39285835

ABSTRACT

The early development of the neonatal immune system is profoundly influenced by exposure to dietary and microbial antigens, which shapes mucosal tolerance. Successful oral tolerance induction is crucially dependent on microbially imprinted immune cells, most notably the RORγt+ regulatory T (Treg) and antigen presenting cells and is essential for preventing food allergy (FA). The development of FA can be envisioned to result from disruptions at key checkpoints (CKPTs) that govern oral tolerance induction. These include gut epithelial sensory and effector circuits that when dysregulated promote pro-allergic gut dysbiosis. They also include microbially imprinted immune regulatory circuits that are disrupted by dysbiosis and pro-allergic immune responses unleashed by the dysregulation of the aforementioned cascades. Understanding these checkpoints is essential for developing therapeutic strategies to restore immune homeostasis in FA.


Subject(s)
Food Hypersensitivity , Gastrointestinal Microbiome , Immune Tolerance , T-Lymphocytes, Regulatory , Humans , Food Hypersensitivity/immunology , Animals , Gastrointestinal Microbiome/immunology , T-Lymphocytes, Regulatory/immunology , Dysbiosis/immunology , Allergens/immunology
14.
Nat Immunol ; 16(11): 1162-73, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26437242

ABSTRACT

Receptors of the Notch family direct the differentiation of helper T cell subsets, but their influence on regulatory T cell (T(reg) cell) responses is obscure. We found here that lineage-specific deletion of components of the Notch pathway enhanced T(reg) cell-mediated suppression of type 1 helper T cell (T(H)1 cell) responses and protected against their T(H)1 skewing and apoptosis. In contrast, expression in T(reg) cells of a gain-of-function transgene encoding the Notch1 intracellular domain resulted in lymphoproliferation, exacerbated T(H)1 responses and autoimmunity. Cell-intrinsic canonical Notch signaling impaired T(reg) cell fitness and promoted the acquisition by T(reg) cells of a T(H)1 cell-like phenotype, whereas non-canonical Notch signaling dependent on the adaptor Rictor activated the kinase AKT-transcription factor Foxo1 axis and impaired the epigenetic stability of Foxp3. Our findings establish a critical role for Notch signaling in controlling peripheral T(reg) cell function.


Subject(s)
Peripheral Tolerance , Receptor, Notch1/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Carrier Proteins/genetics , Carrier Proteins/immunology , Epigenesis, Genetic , Female , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Graft vs Host Disease/immunology , Graft vs Host Disease/prevention & control , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mutation , Rapamycin-Insensitive Companion of mTOR Protein , Receptor, Notch1/deficiency , Receptor, Notch1/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Signal Transduction/immunology , Th1 Cells/immunology , Transcriptome
15.
Semin Immunol ; 70: 101847, 2023 11.
Article in English | MEDLINE | ID: mdl-37837939

ABSTRACT

Regulatory T (Treg) cells maintain immune tolerance to allergens at the environmental interfaces in the airways, skin and gut, marshalling in the process distinct immune regulatory circuits operative in the respective tissues. Treg cells are coordinately mobilized with allergic effector mechanisms in the context of a tissue-protective allergic inflammatory response against parasites, toxins and potentially harmful allergens, serving to both limit the inflammation and promote local tissue repair. Allergic diseases are associated with subverted Treg cell responses whereby a chronic allergic inflammatory environment can skew Treg cells toward pathogenic phenotypes that both perpetuate and aggravate disease. Interruption of Treg cell subversion in chronic allergic inflammatory conditions may thus provide novel therapeutic strategies by re-establishing effective immune regulation.


Subject(s)
Hypersensitivity , T-Lymphocytes, Regulatory , Humans , Hypersensitivity/therapy , Allergens , Inflammation/pathology , Immune Tolerance
17.
J Allergy Clin Immunol ; 153(1): 28-41, 2024 01.
Article in English | MEDLINE | ID: mdl-37778472

ABSTRACT

Regulatory T cells expressing the transcription factor forkhead box protein 3 mediate peripheral immune tolerance both to self-antigens and to the commensal flora. Their defective function due to inborn errors of immunity or acquired insults is associated with a broad range of autoimmune and immune dysregulatory diseases. Although their function in suppressing autoimmunity and enforcing commensalism is established, a broader role for regulatory T cells in tissue repair and metabolic regulation has emerged, enabled by unique programs of tissue adaptability and specialization. In this review, we focus on the myriad roles played by regulatory T cells in immunologic tolerance and host homeostasis and the potential to harness these cells in novel therapeutic approaches to human diseases.


Subject(s)
Autoimmune Diseases , Immune System Diseases , Humans , T-Lymphocytes, Regulatory , Immune Tolerance , Immune System Diseases/metabolism , Autoimmunity , Forkhead Transcription Factors
18.
Nat Immunol ; 13(6): 612-20, 2012 May 13.
Article in English | MEDLINE | ID: mdl-22581261

ABSTRACT

The adaptors DOCK8 and MyD88 have been linked to serological memory. Here we report that DOCK8-deficient patients had impaired antibody responses and considerably fewer CD27(+) memory B cells. B cell proliferation and immunoglobulin production driven by Toll-like receptor 9 (TLR9) were considerably lower in DOCK8-deficient B cells, but those driven by the costimulatory molecule CD40 were not. In contrast, TLR9-driven expression of AICDA (which encodes the cytidine deaminase AID), the immunoglobulin receptor CD23 and the costimulatory molecule CD86 and activation of the transcription factor NF-κB, the kinase p38 and the GTPase Rac1 were intact. DOCK8 associated constitutively with MyD88 and the tyrosine kinase Pyk2 in normal B cells. After ligation of TLR9, DOCK8 became tyrosine-phosphorylated by Pyk2, bound the Src-family kinase Lyn and linked TLR9 to a Src-kinase Syk-transcription factor STAT3 cascade essential for TLR9-driven B cell proliferation and differentiation. Thus, DOCK8 functions as an adaptor in a TLR9-MyD88 signaling pathway in B cells.


Subject(s)
B-Lymphocytes/immunology , Guanine Nucleotide Exchange Factors/immunology , Immunologic Memory/immunology , Myeloid Differentiation Factor 88/immunology , Toll-Like Receptor 9/immunology , Adolescent , Animals , Cell Differentiation/immunology , Child , Child, Preschool , Flow Cytometry , Focal Adhesion Kinase 2/immunology , Humans , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/immunology , Phosphorylation , STAT3 Transcription Factor/immunology , src-Family Kinases/immunology
19.
Allergy ; 2024 Oct 03.
Article in English | MEDLINE | ID: mdl-39361431

ABSTRACT

BACKGROUND: Immune dysregulation and SARS-CoV-2 plasma viremia have been implicated in fatal COVID-19 disease. However, how these two factors interact to shape disease outcomes is unclear. METHODS: We carried out viral and immunological phenotyping on a prospective cohort of 280 patients with COVID-19 presenting to acute care hospitals in Boston, Massachusetts and Genoa, Italy between June 1, 2020 and February 8, 2022. Disease severity, mortality, plasma viremia, and immune dysregulation were assessed. A mouse model of lethal H1N1 influenza infection was used to analyze the therapeutic potential of Notch4 and pyroptosis inhibition in disease outcome. RESULTS: Stratifying patients based on %Notch4+ Treg cells and/or the presence of plasma viremia identified four subgroups with different clinical trajectories and immune phenotypes. Patients with both high %Notch4+ Treg cells and viremia suffered the most disease severity and 90-day mortality compared to the other groups even after adjusting for baseline comorbidities. Increased Notch4 and plasma viremia impacted different arms of the immune response in SARS-CoV-2 infection. Increased Notch4 was associated with decreased Treg cell amphiregulin expression and suppressive function whereas plasma viremia was associated with increased monocyte cell pyroptosis. Combinatorial therapies using Notch4 blockade and pyroptosis inhibition induced stepwise protection against mortality in a mouse model of lethal H1N1 influenza infection. CONCLUSIONS: The clinical trajectory and survival outcome in hospitalized patients with COVID-19 is predicated on two cardinal factors in disease pathogenesis: viremia and Notch4+ Treg cells. Intervention strategies aimed at resetting the immune dysregulation in COVID-19 by antagonizing Notch4 and pyroptosis may be effective in severe cases of viral lung infection.

20.
Immunity ; 43(2): 289-303, 2015 Aug 18.
Article in English | MEDLINE | ID: mdl-26231118

ABSTRACT

Commensal microbiota promote mucosal tolerance in part by engaging regulatory T (Treg) cells via Toll-like receptors (TLRs). We report that Treg-cell-specific deletion of the TLR adaptor MyD88 resulted in deficiency of intestinal Treg cells, a reciprocal increase in T helper 17 (Th17) cells and heightened interleukin-17 (IL-17)-dependent inflammation in experimental colitis. It also precipitated dysbiosis with overgrowth of segmented filamentous bacteria (SFB) and increased microbial loads in deep tissues. The Th17 cell dysregulation and bacterial dysbiosis were linked to impaired anti-microbial intestinal IgA responses, related to defective MyD88 adaptor- and Stat3 transcription factor-dependent T follicular regulatory and helper cell differentiation in the Peyer's patches. These findings establish an essential role for MyD88-dependent microbial sensing by Treg cells in enforcing mucosal tolerance and maintaining commensalism by promoting intestinal Treg cell formation and anti-commensal IgA responses.


Subject(s)
Colitis/immunology , Escherichia coli Infections/immunology , Escherichia coli/immunology , Intestines/immunology , Myeloid Differentiation Factor 88/metabolism , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Animals , Antibodies, Bacterial/metabolism , Cell Differentiation , Cells, Cultured , Immune Tolerance , Immunity, Mucosal , Immunoglobulin A/metabolism , Intestines/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Myeloid Differentiation Factor 88/genetics , STAT3 Transcription Factor/metabolism , Symbiosis/immunology , Toll-Like Receptors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL