Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Cell ; 180(1): 79-91.e16, 2020 01 09.
Article in English | MEDLINE | ID: mdl-31866067

ABSTRACT

Lymphoid cells that produce interleukin (IL)-17 cytokines protect barrier tissues from pathogenic microbes but are also prominent effectors of inflammation and autoimmune disease. T helper 17 (Th17) cells, defined by RORγt-dependent production of IL-17A and IL-17F, exert homeostatic functions in the gut upon microbiota-directed differentiation from naive CD4+ T cells. In the non-pathogenic setting, their cytokine production is regulated by serum amyloid A proteins (SAA1 and SAA2) secreted by adjacent intestinal epithelial cells. However, Th17 cell behaviors vary markedly according to their environment. Here, we show that SAAs additionally direct a pathogenic pro-inflammatory Th17 cell differentiation program, acting directly on T cells in collaboration with STAT3-activating cytokines. Using loss- and gain-of-function mouse models, we show that SAA1, SAA2, and SAA3 have distinct systemic and local functions in promoting Th17-mediated inflammatory diseases. These studies suggest that T cell signaling pathways modulated by the SAAs may be attractive targets for anti-inflammatory therapies.


Subject(s)
Irritable Bowel Syndrome/metabolism , Serum Amyloid A Protein/metabolism , Th17 Cells/metabolism , Adult , Animals , Autoimmune Diseases/metabolism , Cell Differentiation/immunology , Cytokines/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Female , Humans , Inflammation/metabolism , Interleukin-17/metabolism , Irritable Bowel Syndrome/blood , Male , Mice , Mice, Inbred C57BL , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Th1 Cells , Th17 Cells/immunology
2.
Immunity ; 55(11): 2027-2043.e9, 2022 11 08.
Article in English | MEDLINE | ID: mdl-36243007

ABSTRACT

T helper 17 (Th17) cells regulate mucosal barrier defenses but also promote multiple autoinflammatory diseases. Although many molecular determinants of Th17 cell differentiation have been elucidated, the transcriptional programs that sustain Th17 cells in vivo remain obscure. The transcription factor RORγt is critical for Th17 cell differentiation; however, it is not clear whether the closely related RORα, which is co-expressed in Th17 cells, has a distinct role. Here, we demonstrated that although dispensable for Th17 cell differentiation, RORα was necessary for optimal Th17 responses in peripheral tissues. The absence of RORα in T cells led to reductions in both RORγt expression and effector function among Th17 cells. Cooperative binding of RORα and RORγt to a previously unidentified Rorc cis-regulatory element was essential for Th17 lineage maintenance in vivo. These data point to a non-redundant role of RORα in Th17 lineage maintenance via reinforcement of the RORγt transcriptional program.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Nuclear Receptor Subfamily 1, Group F, Member 3 , Cell Differentiation , Encephalomyelitis, Autoimmune, Experimental/metabolism , Gene Expression Regulation , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Th17 Cells/metabolism , Transcription Factors/metabolism
3.
Immunity ; 54(8): 1745-1757.e7, 2021 08 10.
Article in English | MEDLINE | ID: mdl-34348118

ABSTRACT

Environmental enteric dysfunction (EED) is a gastrointestinal inflammatory disease caused by malnutrition and chronic infection. EED is associated with stunting in children and reduced efficacy of oral vaccines. To study the mechanisms of oral vaccine failure during EED, we developed a microbiota- and diet-dependent mouse EED model. Analysis of E. coli-labile toxin vaccine-specific CD4+ T cells in these mice revealed impaired CD4+ T cell responses in the small intestine and but not the lymph nodes. EED mice exhibited increased frequencies of small intestine-resident RORγT+FOXP3+ regulatory T (Treg) cells. Targeted deletion of RORγT from Treg cells restored small intestinal vaccine-specific CD4 T cell responses and vaccine-mediated protection upon challenge. However, ablation of RORγT+FOXP3+ Treg cells made mice more susceptible to EED-induced stunting. Our findings provide insight into the poor efficacy of oral vaccines in EED and highlight how RORγT+FOXP3+ Treg cells can regulate intestinal immunity while leaving systemic responses intact.


Subject(s)
Bacterial Toxins/immunology , Escherichia coli Vaccines/immunology , Gastrointestinal Diseases/immunology , Intestine, Small/immunology , T-Lymphocytes, Regulatory/immunology , Administration, Oral , Animals , Cell Line , Disease Models, Animal , Drosophila , Escherichia coli/immunology , Female , Forkhead Transcription Factors/metabolism , Gastrointestinal Diseases/microbiology , Gastrointestinal Diseases/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Vaccination
4.
Cell ; 163(2): 381-93, 2015 Oct 08.
Article in English | MEDLINE | ID: mdl-26411290

ABSTRACT

RORγt(+) Th17 cells are important for mucosal defenses but also contribute to autoimmune disease. They accumulate in the intestine in response to microbiota and produce IL-17 cytokines. Segmented filamentous bacteria (SFB) are Th17-inducing commensals that potentiate autoimmunity in mice. RORγt(+) T cells were induced in mesenteric lymph nodes early after SFB colonization and distributed across different segments of the gastrointestinal tract. However, robust IL-17A production was restricted to the ileum, where SFB makes direct contact with the epithelium and induces serum amyloid A proteins 1 and 2 (SAA1/2), which promote local IL-17A expression in RORγt(+) T cells. We identified an SFB-dependent role of type 3 innate lymphoid cells (ILC3), which secreted IL-22 that induced epithelial SAA production in a Stat3-dependent manner. This highlights the critical role of tissue microenvironment in activating effector functions of committed Th17 cells, which may have important implications for how these cells contribute to inflammatory disease.


Subject(s)
Gastrointestinal Microbiome , Interleukins/metabolism , Intestines/immunology , Receptors, Interleukin/metabolism , Serum Amyloid A Protein/metabolism , Th17 Cells/immunology , Animals , Immunity, Innate , Interleukins/immunology , Intestines/anatomy & histology , Intestines/microbiology , Lymphocytes/immunology , Mice , Mice, Inbred C57BL , Receptors, Interleukin/immunology , Signal Transduction , Interleukin-22
6.
Immunity ; 51(1): 185-197.e6, 2019 07 16.
Article in English | MEDLINE | ID: mdl-31278058

ABSTRACT

Innate lymphoid cells (ILCs) promote tissue homeostasis and immune defense but also contribute to inflammatory diseases. ILCs exhibit phenotypic and functional plasticity in response to environmental stimuli, yet the transcriptional regulatory networks (TRNs) that control ILC function are largely unknown. Here, we integrate gene expression and chromatin accessibility data to infer regulatory interactions between transcription factors (TFs) and genes within intestinal type 1, 2, and 3 ILC subsets. We predicted the "core" TFs driving ILC identities, organized TFs into cooperative modules controlling distinct gene programs, and validated roles for c-MAF and BCL6 as regulators affecting type 1 and type 3 ILC lineages. The ILC network revealed alternative-lineage-gene repression, a mechanism that may contribute to reported plasticity between ILC subsets. By connecting TFs to genes, the TRNs suggest means to selectively regulate ILC effector functions, while our network approach is broadly applicable to identifying regulators in other in vivo cell populations.


Subject(s)
Intestines/physiology , Lymphocyte Subsets/physiology , Lymphocytes/physiology , Animals , Cell Differentiation , Cell Lineage , Cell Plasticity , Chromatin Assembly and Disassembly , Epigenetic Repression , Gene Regulatory Networks , Immunity, Innate , Immunomodulation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Proto-Oncogene Proteins c-bcl-6/genetics , Proto-Oncogene Proteins c-maf/genetics , Transcriptome
8.
J Immunol ; 211(3): 389-402, 2023 08 01.
Article in English | MEDLINE | ID: mdl-37272847

ABSTRACT

The impact of endemic parasitic infection on vaccine efficacy is an important consideration for vaccine development and deployment. We have examined whether intestinal infection with the natural murine helminth Heligmosomoides polygyrus bakeri alters Ag-specific Ab and cellular immune responses to oral and parenteral vaccination in mice. Oral vaccination of mice with a clinically relevant, live, attenuated, recombinant Salmonella vaccine expressing chicken egg OVA (Salmonella-OVA) induced the accumulation of activated, OVA-specific T effector cells rather than OVA-specific regulatory T cells in the GALT. Intestinal helminth infection significantly reduced Th1-skewed Ab responses to oral vaccination with Salmonella-OVA. Activated, adoptively transferred, OVA-specific CD4+ T cells accumulated in draining mesenteric lymph nodes of vaccinated mice, regardless of their helminth infection status. However, helminth infection increased the frequencies of adoptively transferred OVA-specific CD4+ T cells producing IL-4 and IL-10 in the mesenteric lymph node. Ab responses to the oral Salmonella-OVA vaccine were reduced in helminth-free mice adoptively transferred with OVA-specific CD4+ T cells harvested from mice with intestinal helminth infection. Intestinal helminth infection also significantly reduced Th2-skewed Ab responses to parenteral vaccination with OVA adsorbed to alum. These findings suggest that vaccine-specific CD4+ T cells induced in the context of helminth infection retain durable immunomodulatory properties and may promote blunted Ab responses to vaccination. They also underscore the potential need to treat parasitic infection before mass vaccination campaigns in helminth-endemic areas.


Subject(s)
Helminthiasis , Intestinal Diseases, Parasitic , Mice , Animals , Vaccine Efficacy , CD4-Positive T-Lymphocytes , Vaccines, Synthetic , Ovalbumin , Mice, Inbred BALB C
9.
Nature ; 562(7725): 150, 2018 10.
Article in English | MEDLINE | ID: mdl-29973715

ABSTRACT

Change History: This Article has been retracted; see accompanying Retraction. Corrected online 20 January: In this Article, author Frank Rigo was incorrectly listed with a middle initial; this has been corrected in the online versions of the paper.

10.
Genome Res ; 29(3): 449-463, 2019 03.
Article in English | MEDLINE | ID: mdl-30696696

ABSTRACT

Transcriptional regulatory networks (TRNs) provide insight into cellular behavior by describing interactions between transcription factors (TFs) and their gene targets. The assay for transposase-accessible chromatin (ATAC)-seq, coupled with TF motif analysis, provides indirect evidence of chromatin binding for hundreds of TFs genome-wide. Here, we propose methods for TRN inference in a mammalian setting, using ATAC-seq data to improve gene expression modeling. We test our methods in the context of T Helper Cell Type 17 (Th17) differentiation, generating new ATAC-seq data to complement existing Th17 genomic resources. In this resource-rich mammalian setting, our extensive benchmarking provides quantitative, genome-scale evaluation of TRN inference, combining ATAC-seq and RNA-seq data. We refine and extend our previous Th17 TRN, using our new TRN inference methods to integrate all Th17 data (gene expression, ATAC-seq, TF knockouts, and ChIP-seq). We highlight newly discovered roles for individual TFs and groups of TFs ("TF-TF modules") in Th17 gene regulation. Given the popularity of ATAC-seq, which provides high-resolution with low sample input requirements, we anticipate that our methods will improve TRN inference in new mammalian systems, especially in vivo, for cells directly from humans and animal models.


Subject(s)
Chromatin/genetics , Gene Regulatory Networks , Th17 Cells/metabolism , Transcription Factors/metabolism , Cell Differentiation , Chromatin/chemistry , Chromatin Assembly and Disassembly , Humans , Protein Binding , Software , Th17 Cells/cytology
11.
Nature ; 528(7583): 517-22, 2015 Dec 24.
Article in English | MEDLINE | ID: mdl-26675721

ABSTRACT

T helper 17 (TH17) lymphocytes protect mucosal barriers from infections, but also contribute to multiple chronic inflammatory diseases. Their differentiation is controlled by RORγt, a ligand-regulated nuclear receptor. Here we identify the RNA helicase DEAD-box protein 5 (DDX5) as a RORγt partner that coordinates transcription of selective TH17 genes, and is required for TH17-mediated inflammatory pathologies. Surprisingly, the ability of DDX5 to interact with RORγt and coactivate its targets depends on intrinsic RNA helicase activity and binding of a conserved nuclear long noncoding RNA (lncRNA), Rmrp, which is mutated in patients with cartilage-hair hypoplasia. A targeted Rmrp gene mutation in mice, corresponding to a gene mutation in cartilage-hair hypoplasia patients, altered lncRNA chromatin occupancy, and reduced the DDX5-RORγt interaction and RORγt target gene transcription. Elucidation of the link between Rmrp and the DDX5-RORγt complex reveals a role for RNA helicases and lncRNAs in tissue-specific transcriptional regulation, and provides new opportunities for therapeutic intervention in TH17-dependent diseases.


Subject(s)
DEAD-box RNA Helicases/metabolism , RNA, Long Noncoding/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism , Animals , Chromatin/genetics , Chromatin/metabolism , DEAD-box RNA Helicases/genetics , Female , Gene Expression Regulation/genetics , Hair/abnormalities , Hirschsprung Disease/genetics , Humans , Immunologic Deficiency Syndromes/genetics , Inflammation/immunology , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , Mutation/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Organ Specificity , Osteochondrodysplasias/congenital , Osteochondrodysplasias/genetics , Primary Immunodeficiency Diseases , Protein Binding , RNA, Long Noncoding/genetics , Transcription, Genetic/genetics
12.
Immunity ; 35(1): 13-22, 2011 Jul 22.
Article in English | MEDLINE | ID: mdl-21777796

ABSTRACT

Vitamin A elicits a broad array of immune responses through its metabolite, retinoic acid (RA). Recent evidence indicates that loss of RA leads to impaired immunity, whereas excess RA can potentially promote inflammatory disorders. In this review, we discuss recent advances showcasing the crucial contributions of RA to both immunological tolerance and the elicitation of adaptive immune responses. Further, we provide a comprehensive overview of the cell types and factors that control the production of RA and discuss how host perturbations may affect the ability of this metabolite to control tolerance and immunity or to instigate pathology.


Subject(s)
Adaptive Immunity , Immune Tolerance , Infections/immunology , T-Lymphocytes, Regulatory/immunology , Tretinoin/immunology , Animals , Cell Movement , Humans , Immunity, Mucosal , Immunoglobulin A/immunology , Signal Transduction/immunology , Vitamin A/metabolism
13.
Immunity ; 34(3): 435-47, 2011 Mar 25.
Article in English | MEDLINE | ID: mdl-21419664

ABSTRACT

Vitamin A and its metabolite, retinoic acid (RA) are implicated in the regulation of immune homeostasis via the peripheral induction of regulatory T cells. Here we showed RA was also required to elicit proinflammatory CD4(+) helper T cell responses to infection and mucosal vaccination. Retinoic acid receptor alpha (RARα) was the critical mediator of these effects. Antagonism of RAR signaling and deficiency in RARα (Rara(-/-)) resulted in a cell-autonomous CD4(+) T cell activation defect, which impaired intermediate signaling events, including calcium mobilization. Altogether, these findings reveal a fundamental role for the RA-RARα axis in the development of both regulatory and inflammatory arms of adaptive immunity and establish nutritional status as a broad regulator of adaptive T cell responses.


Subject(s)
Adaptive Immunity/immunology , CD4-Positive T-Lymphocytes/immunology , Receptors, Retinoic Acid/immunology , Tretinoin/immunology , Animals , Female , Homeostasis/immunology , Male , Mice , Mice, Inbred C57BL , Retinoic Acid Receptor alpha , Signal Transduction , Toxoplasmosis/immunology
14.
Immunity ; 31(5): 772-86, 2009 Nov 20.
Article in English | MEDLINE | ID: mdl-19896394

ABSTRACT

Using a model of lethal oral infection with Toxoplasma gondii, we examined the fate of both induced and natural regulatory T (Treg) cells in the face of strong inflammatory responses occurring in a tolerogenic-prone environment. We found that during highly T helper 1 (Th1) cell-polarized mucosal immune responses, Treg cell numbers collapsed via multiple pathways, including blockade of Treg cell induction and disruption of endogenous Treg cell homeostasis. In particular, shutdown of interleukin 2 (IL-2) in the highly Th1 cell-polarized environment triggered by infection directly contributes to Treg cell incapacity to suppress effector responses and eventually leads to immunopathogenesis. Furthermore, we found that environmental cues provided by both local dendritic cells and effector T cells can induce the expression of T-bet transcription factor and IFN-gamma by Treg cells. These data reveal a mechanism for Th1 cell pathogenicity that extends beyond their proinflammatory program to limit Treg cell survival.


Subject(s)
Forkhead Transcription Factors/metabolism , T-Lymphocytes, Regulatory/immunology , Toxoplasma , Animals , Cell Proliferation , Interleukin-2/metabolism , Mice , Mice, Inbred C57BL , Phenotype , Toxoplasmosis/immunology
15.
Immunity ; 29(4): 637-49, 2008 Oct 17.
Article in English | MEDLINE | ID: mdl-18835196

ABSTRACT

The intestinal tract is in intimate contact with the commensal microflora. Nevertheless, how commensals communicate with cells to ensure immune homeostasis is still unclear. In this study, we found that gut flora DNA (gfDNA) plays a major role in intestinal homeostasis through Toll-like receptor 9 (TLR9) engagement. Tlr9(-/-) mice displayed increased frequencies of CD4(+)Foxp3(+) regulatory T (Treg) cells within intestinal effector sites and reduced constitutive IL-17- and IFN-gamma-producing effector T (Teff) cells. Complementing this, gfDNA limited lamina propria dendritic cell-induced Treg cell conversion in vitro. Further, Treg/Teff cell disequilibrium in Tlr9(-/-) mice led to impaired immune responses to oral infection and to oral vaccination. Impaired intestinal immune responses were recapitulated in mice treated with antibiotics and were reversible after reconstitution with gfDNA. Together, these data point to gfDNA as a natural adjuvant for priming intestinal responses via modulation of Treg/Teff cell equilibrium.


Subject(s)
DNA, Bacterial/immunology , Dendritic Cells/immunology , Intestinal Mucosa/immunology , Intestines/microbiology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes/immunology , Toll-Like Receptor 9/metabolism , Adjuvants, Immunologic , Animals , Anti-Bacterial Agents/pharmacology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , DNA, Bacterial/metabolism , Dendritic Cells/metabolism , Immunity, Mucosal , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Interleukin-17/biosynthesis , Interleukin-17/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mucous Membrane/immunology , Mucous Membrane/metabolism , Signal Transduction , T-Lymphocytes, Regulatory/metabolism , Toll-Like Receptor 9/immunology
16.
Immunity ; 29(5): 758-70, 2008 Nov 14.
Article in English | MEDLINE | ID: mdl-19006694

ABSTRACT

CD4(+)Foxp3(+) regulatory T (Treg) cells originate primarily from thymic differentiation, but conversion of mature T lymphocytes to Foxp3 positivity can be elicited by several means, including in vitro activation in the presence of TGF-beta. Retinoic acid (RA) increases TGF-beta-induced expression of Foxp3, through unknown molecular mechanisms. We showed here that, rather than enhancing TGF-beta signaling directly in naive CD4(+) T cells, RA negatively regulated an accompanying population of CD4(+) T cells with a CD44(hi) memory and effector phenotype. These memory cells actively inhibited the TGF-beta-induced conversion of naive CD4(+) T cells through the synthesis of a set of cytokines (IL-4, IL-21, IFN-gamma) whose expression was coordinately curtailed by RA. This indirect effect was evident in vivo and required the expression of the RA receptor alpha. Thus, cytokine-producing CD44(hi) cells actively restrain TGF-beta-mediated Foxp3 expression in naive T cells, and this balance can be shifted or fine-tuned by RA.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Forkhead Transcription Factors/metabolism , Receptors, Retinoic Acid/immunology , T-Lymphocytes, Regulatory/immunology , Tretinoin/metabolism , Animals , CD4-Positive T-Lymphocytes/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Forkhead Transcription Factors/immunology , Gene Expression Regulation , Gene Knockdown Techniques , Homeostasis , Hyaluronan Receptors/analysis , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-4/immunology , Interleukin-4/metabolism , Interleukins/immunology , Interleukins/metabolism , Mice , Mice, Inbred C57BL , Receptors, Retinoic Acid/deficiency , Receptors, Retinoic Acid/metabolism , Retinoic Acid Receptor alpha , Signal Transduction , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/metabolism , Transforming Growth Factor beta/immunology , Transforming Growth Factor beta/metabolism
18.
J Biol Chem ; 288(20): 14080-14086, 2013 May 17.
Article in English | MEDLINE | ID: mdl-23546877

ABSTRACT

Members of the Amt family of channels mediate the transport of ammonium. The form of ammonium, NH3 or NH4(+), carried by these proteins remains controversial, and the mechanism by which they select against K(+) ions is unclear. We describe here a set of Escherichia coli AmtB proteins carrying mutations at the conserved twin-histidine site within the conduction pore that have altered substrate specificity and now transport K(+). Subsequent work established that AmtB-mediated K(+) uptake occurred against a concentration gradient and was membrane potential-dependent. These findings indicate that the twin-histidine element serves as a filter to prevent K(+) conduction and strongly support the notion that Amt proteins transport cations (NH4(+) or, in mutant proteins, K(+)) rather than NH3 gas molecules through their conduction pores.


Subject(s)
Cation Transport Proteins/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Potassium/metabolism , Biological Transport , Cytoplasm/metabolism , Dose-Response Relationship, Drug , Hydrogen-Ion Concentration , Ions , Membrane Potentials , Mutation , Protein Conformation , Quaternary Ammonium Compounds/metabolism , Substrate Specificity
19.
Proc Natl Acad Sci U S A ; 108(32): 13270-4, 2011 Aug 09.
Article in English | MEDLINE | ID: mdl-21775672

ABSTRACT

In Escherichia coli, each subunit of the trimeric channel protein AmtB carries a hydrophobic pore for transport of NH(4)(+) across the cytoplasmic membrane. Positioned along this substrate conduction pathway are two conserved elements--a pair of hydrogen-bonded histidines (H168/H318) located within the pore itself and a set of aromatic residues (F107/W148/F215) at its periplasmic entrance--thought to be critical to AmtB function. Using site-directed mutagenesis and suppressor genetics, we examined the requirement for these elements in NH(4)(+) transport. This analysis shows that AmtB can accommodate, by either direct substitution or suppressor generation, acidic residues at one or both positions of the H168/H318 twin-histidine site while retaining near wild-type activity. Similarly, study of the F107/W148/F215 triad indicates that good-to-excellent AmtB function is preserved upon individual and simultaneous replacement of these aromatic amino acids with aliphatic residues. Our findings lead us to conclude that these elements and their component parts are not required for AmtB function, but instead serve to optimize its performance.


Subject(s)
Cation Transport Proteins/metabolism , Escherichia coli K12/metabolism , Escherichia coli Proteins/metabolism , Histidine/metabolism , Quaternary Ammonium Compounds/metabolism , Amino Acids/metabolism , Cation Transport Proteins/chemistry , Escherichia coli K12/drug effects , Escherichia coli K12/growth & development , Escherichia coli Proteins/chemistry , Mutant Proteins/metabolism , Protein Transport/drug effects , Quaternary Ammonium Compounds/pharmacology
20.
Immunol Rev ; 234(1): 305-16, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20193027

ABSTRACT

Each microenvironment is controlled by a specific set of regulatory elements that have to be finely and constantly tuned to maintain local homeostasis. These environments could be site specific, such as the gut environment, or induced by chronic exposure to microbes. Various populations of dendritic cells are central to the orchestration of this control. In this review, we discuss some new findings associating dendritic cells from defined compartments with the induction and control of regulatory T cells in the context of exposure to both commensal and pathogenic microbes.


Subject(s)
Bacteria/immunology , Cell Differentiation , Cell Lineage , Dendritic Cells/immunology , Intestines/immunology , Parasites/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cell Differentiation/immunology , Cell Lineage/immunology , Communicable Diseases/immunology , Communicable Diseases/microbiology , Communicable Diseases/parasitology , Dendritic Cells/microbiology , Dendritic Cells/parasitology , Forkhead Transcription Factors/immunology , Homeostasis , Host-Parasite Interactions , Host-Pathogen Interactions , Humans , Intestinal Diseases/immunology , Intestinal Diseases/microbiology , Intestinal Diseases/parasitology , Intestines/microbiology , Intestines/parasitology , Peyer's Patches/immunology , Peyer's Patches/microbiology , Peyer's Patches/parasitology , Signal Transduction , T-Lymphocytes, Regulatory/microbiology , T-Lymphocytes, Regulatory/parasitology
SELECTION OF CITATIONS
SEARCH DETAIL