Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 19 de 19
1.
Commun Biol ; 7(1): 442, 2024 Apr 10.
Article En | MEDLINE | ID: mdl-38600349

Aryl hydrocarbon receptor (AHR) signalling integrates biological processes that sense and respond to environmental, dietary, and metabolic challenges to ensure tissue homeostasis. AHR is a transcription factor that is inactive in the cytosol but upon encounter with ligand translocates to the nucleus and drives the expression of AHR targets, including genes of the cytochrome P4501 family of enzymes such as Cyp1a1. To dynamically visualise AHR activity in vivo, we generated reporter mice in which firefly luciferase (Fluc) was non-disruptively targeted into the endogenous Cyp1a1 locus. Exposure of these animals to FICZ, 3-MC or to dietary I3C induced strong bioluminescence signal and Cyp1a1 expression in many organs including liver, lung and intestine. Longitudinal studies revealed that AHR activity was surprisingly long-lived in the lung, with sustained Cyp1a1 expression evident in discrete populations of cells including columnar epithelia around bronchioles. Our data link diet to lung physiology and also reveal the power of bespoke Cyp1a1-Fluc reporters to longitudinally monitor AHR activity in vivo.


Cytochrome P-450 CYP1A1 , Receptors, Aryl Hydrocarbon , Mice , Animals , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1A1/metabolism , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism , Luciferases/genetics , Liver/metabolism , Lung/metabolism
2.
Nature ; 621(7980): 821-829, 2023 Sep.
Article En | MEDLINE | ID: mdl-37586410

Endothelial cells line the blood and lymphatic vasculature, and act as an essential physical barrier, control nutrient transport, facilitate tissue immunosurveillance and coordinate angiogenesis and lymphangiogenesis1,2. In the intestine, dietary and microbial cues are particularly important in the regulation of organ homeostasis. However, whether enteric endothelial cells actively sense and integrate such signals is currently unknown. Here we show that the aryl hydrocarbon receptor (AHR) acts as a critical node for endothelial cell sensing of dietary metabolites in adult mice and human primary endothelial cells. We first established a comprehensive single-cell endothelial atlas of the mouse small intestine, uncovering the cellular complexity and functional heterogeneity of blood and lymphatic endothelial cells. Analyses of AHR-mediated responses at single-cell resolution identified tissue-protective transcriptional signatures and regulatory networks promoting cellular quiescence and vascular normalcy at steady state. Endothelial AHR deficiency in adult mice resulted in dysregulated inflammatory responses and the initiation of proliferative pathways. Furthermore, endothelial sensing of dietary AHR ligands was required for optimal protection against enteric infection. In human endothelial cells, AHR signalling promoted quiescence and restrained activation by inflammatory mediators. Together, our data provide a comprehensive dissection of the effect of environmental sensing across the spectrum of enteric endothelia, demonstrating that endothelial AHR signalling integrates dietary cues to maintain tissue homeostasis by promoting endothelial cell quiescence and vascular normalcy.


Endothelial Cells , Receptors, Aryl Hydrocarbon , Humans , Animals , Mice , Receptors, Aryl Hydrocarbon/metabolism , Endothelial Cells/metabolism , Intestines , Signal Transduction , Homeostasis , Ligands
5.
Immunity ; 49(2): 353-362.e5, 2018 08 21.
Article En | MEDLINE | ID: mdl-30119997

The epithelium and immune compartment in the intestine are constantly exposed to a fluctuating external environment. Defective communication between these compartments at this barrier surface underlies susceptibility to infections and chronic inflammation. Environmental factors play a significant, but mechanistically poorly understood, role in intestinal homeostasis. We found that regeneration of intestinal epithelial cells (IECs) upon injury through infection or chemical insults was profoundly influenced by the environmental sensor aryl hydrocarbon receptor (AHR). IEC-specific deletion of Ahr resulted in failure to control C. rodentium infection due to unrestricted intestinal stem cell (ISC) proliferation and impaired differentiation, culminating in malignant transformation. AHR activation by dietary ligands restored barrier homeostasis, protected the stem cell niche, and prevented tumorigenesis via transcriptional regulation of of Rnf43 and Znrf3, E3 ubiquitin ligases that inhibit Wnt-ß-catenin signaling and restrict ISC proliferation. Thus, activation of the AHR pathway in IECs guards the stem cell niche to maintain intestinal barrier integrity.


Basic Helix-Loop-Helix Transcription Factors/metabolism , Epithelial Cells/physiology , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Receptors, Aryl Hydrocarbon/metabolism , Stem Cells/cytology , Tight Junctions/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Carcinogenesis/pathology , Cell Differentiation/immunology , Cell Line , Cell Proliferation , Citrobacter rodentium/immunology , Enterobacteriaceae Infections/immunology , Enterobacteriaceae Infections/microbiology , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Aryl Hydrocarbon/genetics , Ubiquitin-Protein Ligases/biosynthesis , Ubiquitin-Protein Ligases/genetics , Wnt Signaling Pathway/physiology
6.
Biochem Pharmacol ; 151: 47-58, 2018 05.
Article En | MEDLINE | ID: mdl-29501585

The aryl hydrocarbon receptor (AHR) controls interleukin 22 production by T helper 17 cells (Th17). IL-22 contributes to intestinal homeostasis but has also been implicated in chronic inflammatory disorders and colorectal cancer, highlighting the need for appropriate regulation of IL-22 production. Upon activation, the AHR induces expression of cytochrome P4501 (CYP1) enzymes which in turn play an important feedback role that curtails the duration of AHR signaling by metabolizing AHR ligands. Recently we described how agents that inhibit CYP1 function potentiate AHR signaling by disrupting metabolic clearance of the endogenous ligand 6-formylindolo[3,2-b]carbazole (FICZ). In the present study, we investigated the immune-modulating effects of environmental pollutants such as polycyclic aromatic hydrocarbons on Th17 differentiation and IL-22 production. Using Th17 cells deficient in CYP1 enzymes (Cyp1a1/1a2/1b1-/-) we show that these chemicals potentiate AHR activation through inhibition of CYP1 enzymes which leads to increases in intracellular AHR agonists. Our findings demonstrate that IL-22 production by Th17 cells is profoundly enhanced by impaired CYP1-function and strongly suggest that chemicals able to modify CYP1 function or expression may disrupt AHR-mediated immune regulation by altering the levels of endogenous AHR agonist(s).


Cytochrome P450 Family 1/antagonists & inhibitors , Environmental Pollutants/toxicity , Interleukins/biosynthesis , Polycyclic Aromatic Hydrocarbons/toxicity , Receptors, Aryl Hydrocarbon/metabolism , Th17 Cells/drug effects , Animals , Carbazoles/metabolism , Cell Differentiation/drug effects , Cytochrome P450 Family 1/genetics , Dose-Response Relationship, Drug , Gene Expression/drug effects , Male , Mice, Inbred C57BL , Mice, Knockout , Receptors, Aryl Hydrocarbon/genetics , Th17 Cells/immunology , Th17 Cells/metabolism , Time Factors , Interleukin-22
7.
Nature ; 542(7640): 242-245, 2017 02 09.
Article En | MEDLINE | ID: mdl-28146477

The aryl hydrocarbon receptor (AHR) recognizes xenobiotics as well as natural compounds such as tryptophan metabolites, dietary components and microbiota-derived factors, and it is important for maintenance of homeostasis at mucosal surfaces. AHR activation induces cytochrome P4501 (CYP1) enzymes, which oxygenate AHR ligands, leading to their metabolic clearance and detoxification. Thus, CYP1 enzymes have an important feedback role that curtails the duration of AHR signalling, but it remains unclear whether they also regulate AHR ligand availability in vivo. Here we show that dysregulated expression of Cyp1a1 in mice depletes the reservoir of natural AHR ligands, generating a quasi AHR-deficient state. Constitutive expression of Cyp1a1 throughout the body or restricted specifically to intestinal epithelial cells resulted in loss of AHR-dependent type 3 innate lymphoid cells and T helper 17 cells and increased susceptibility to enteric infection. The deleterious effects of excessive AHR ligand degradation on intestinal immune functions could be counter-balanced by increasing the intake of AHR ligands in the diet. Thus, our data indicate that intestinal epithelial cells serve as gatekeepers for the supply of AHR ligands to the host and emphasize the importance of feedback control in modulating AHR pathway activation.


Feedback, Physiological , Intestines/immunology , Receptors, Aryl Hydrocarbon/metabolism , Signal Transduction , Animals , Citrobacter rodentium/immunology , Colon/cytology , Colon/immunology , Colon/metabolism , Colon/microbiology , Cytochrome P-450 CYP1A1/metabolism , Female , Immunity, Innate , Intestinal Mucosa/metabolism , Intestines/cytology , Intestines/microbiology , Ligands , Male , Mice , Th17 Cells/immunology
8.
Nat Commun ; 7: 11627, 2016 05 19.
Article En | MEDLINE | ID: mdl-27193261

IL-23 is a key driver of pathogenic Th17 cell responses. It has been suggested that the transcription factor T-bet is required to facilitate IL-23-driven pathogenic effector functions; however, the precise role of T-bet in intestinal T cell responses remains elusive. Here, we show that T-bet expression by T cells is not required for the induction of colitis or the differentiation of pathogenic Th17 cells but modifies qualitative features of the IL-23-driven colitogenic response by negatively regulating IL-23R expression. Consequently, absence of T-bet leads to unrestrained Th17 cell differentiation and activation characterized by high amounts of IL-17A and IL-22. The combined increase in IL-17A/IL-22 results in enhanced epithelial cell activation and inhibition of either IL-17A or IL-22 leads to disease amelioration. Our study identifies T-bet as a key modulator of IL-23-driven colitogenic responses in the intestine and has important implications for understanding of heterogeneity among inflammatory bowel disease patients.


CD4-Positive T-Lymphocytes/physiology , Colitis/immunology , Interleukin-23/metabolism , Intestines/immunology , T-Box Domain Proteins/metabolism , Animals , Disease Models, Animal , Female , Interleukin-17/metabolism , Interleukins/metabolism , Male , Mice, Inbred C57BL , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Receptors, Interleukin/metabolism , Interleukin-22
9.
ISME J ; 10(10): 2389-404, 2016 10.
Article En | MEDLINE | ID: mdl-27003245

The gut microbiome is significantly altered in inflammatory bowel diseases, but the basis of these changes is not well understood. We have combined metagenomic and metatranscriptomic profiling of the gut microbiome to assess modifications to both bacterial community structure and transcriptional activity in a mouse model of colitis. By using transcriptomic analysis of colonic tissue and luminal RNA derived from the host, we have also characterised how host transcription relates to the microbial transcriptional response in inflammation. In colitis, increased abundance and transcription of diverse microbial gene families involved in responses to nutrient deprivation, antimicrobial peptide production and oxidative stress support an adaptation of multiple commensal genera to withstand a diverse set of environmental stressors in the inflammatory environment. These data are supported by a transcriptional signature of activated macrophages and granulocytes in the gut lumen during colitis, a signature that includes the transcription of the key antimicrobial genes S100a8 and S100a9 (calprotectin). Genes involved in microbial resistance to oxidative stress, including Dps/ferritin, Fe-dependent peroxidase and glutathione S-transferase were identified as changing to a greater extent at the level of transcription than would be predicted by DNA abundance changes, implicating a role for increased oxygen tension and/or host-derived reactive oxygen species in driving transcriptional changes in commensal microbes.


Bacteria/genetics , Colitis/genetics , Colitis/microbiology , Gastrointestinal Microbiome , Animals , Bacteria/classification , Bacteria/isolation & purification , Bacteria/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Colitis/immunology , Female , Gene Expression Profiling , Humans , Metagenomics , Mice , Mice, Inbred C57BL
10.
Immunity ; 43(1): 187-99, 2015 Jul 21.
Article En | MEDLINE | ID: mdl-26200014

The role of intestinal eosinophils in immune homeostasis is enigmatic and the molecular signals that drive them from protective to tissue damaging are unknown. Most commonly associated with Th2 cell-mediated diseases, we describe a role for eosinophils as crucial effectors of the interleukin-23 (IL-23)-granulocyte macrophage colony-stimulating factor (GM-CSF) axis in colitis. Chronic intestinal inflammation was characterized by increased bone marrow eosinopoiesis and accumulation of activated intestinal eosinophils. IL-5 blockade or eosinophil depletion ameliorated colitis, implicating eosinophils in disease pathogenesis. GM-CSF was a potent activator of eosinophil effector functions and intestinal accumulation, and GM-CSF blockade inhibited chronic colitis. By contrast neutrophil accumulation was GM-CSF independent and dispensable for colitis. In addition to TNF secretion, release of eosinophil peroxidase promoted colitis identifying direct tissue-toxic mechanisms. Thus, eosinophils are key perpetrators of chronic inflammation and tissue damage in IL-23-mediated immune diseases and it suggests the GM-CSF-eosinophil axis as an attractive therapeutic target.


Colitis/immunology , Eosinophils/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Interleukin-23 Subunit p19/immunology , Animals , Cell Movement/immunology , Cytokine Receptor Common beta Subunit/genetics , Eosinophil Peroxidase/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Inflammation/immunology , Interleukin-5/antagonists & inhibitors , Intestines/cytology , Intestines/immunology , Intestines/pathology , Leukocyte Reduction Procedures , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/immunology , Tumor Necrosis Factors/metabolism
11.
Nature ; 513(7519): 564-568, 2014 Sep 25.
Article En | MEDLINE | ID: mdl-25043027

FOXP3(+) regulatory T cells (Treg cells) are abundant in the intestine, where they prevent dysregulated inflammatory responses to self and environmental stimuli. It is now appreciated that Treg cells acquire tissue-specific adaptations that facilitate their survival and function; however, key host factors controlling the Treg response in the intestine are poorly understood. The interleukin (IL)-1 family member IL-33 is constitutively expressed in epithelial cells at barrier sites, where it functions as an endogenous danger signal, or alarmin, in response to tissue damage. Recent studies in humans have described high levels of IL-33 in inflamed lesions of inflammatory bowel disease patients, suggesting a role for this cytokine in disease pathogenesis. In the intestine, both protective and pathological roles for IL-33 have been described in murine models of acute colitis, but its contribution to chronic inflammation remains ill defined. Here we show in mice that the IL-33 receptor ST2 is preferentially expressed on colonic Treg cells, where it promotes Treg function and adaptation to the inflammatory environment. IL-33 signalling in T cells stimulates Treg responses in several ways. First, it enhances transforming growth factor (TGF)-ß1-mediated differentiation of Treg cells and, second, it provides a necessary signal for Treg-cell accumulation and maintenance in inflamed tissues. Strikingly, IL-23, a key pro-inflammatory cytokine in the pathogenesis of inflammatory bowel disease, restrained Treg responses through inhibition of IL-33 responsiveness. These results demonstrate a hitherto unrecognized link between an endogenous mediator of tissue damage and a major anti-inflammatory pathway, and suggest that the balance between IL-33 and IL-23 may be a key controller of intestinal immune responses.


Interleukins/immunology , Intestines/cytology , Intestines/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Colitis/immunology , Colitis/pathology , Colon/cytology , Colon/immunology , Colon/pathology , Disease Models, Animal , Female , Immunity, Mucosal , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Interleukin-23/immunology , Interleukin-33 , Interleukins/antagonists & inhibitors , Interleukins/metabolism , Intestines/pathology , Male , Mice , Mice, Inbred C57BL , Receptors, Interleukin/metabolism , Signal Transduction/immunology , T-Lymphocytes, Regulatory/cytology , Thymus Gland/cytology , Transforming Growth Factor beta/metabolism
12.
J Immunol ; 190(7): 3235-45, 2013 Apr 01.
Article En | MEDLINE | ID: mdl-23455506

Suppressor of cytokine signaling (SOCS) proteins are key regulators of CD4(+) T cell differentiation, and in particular, we have recently shown that SOCS2 inhibits the development of Th2 cells and allergic immune responses. Interestingly, transcriptome analyses have identified SOCS2 as being preferentially expressed in both natural regulatory T cells (Tregs) and inducible Tregs (iTregs); however, the role of SOCS2 in Foxp3(+) Treg function or development has not been fully elucidated. In this study, we show that despite having no effect on natural Treg development or function, SOCS2 is highly expressed in iTregs and required for the stable expression of Foxp3 in iTregs in vitro and in vivo. Indeed, SOCS2-deficient CD4(+) T cells upregulated Foxp3 following in vitro TGF-ß stimulation, but failed to maintain stable expression of Foxp3. Moreover, in vivo generation of iTregs following OVA feeding was impaired in the absence of SOCS2 and could be rescued in the presence of IL-4 neutralizing Ab. Following IL-4 stimulation, SOCS2-deficient Foxp3(+) iTregs secreted elevated IFN-γ and IL-13 levels and displayed enhanced STAT6 phosphorylation. Therefore, we propose that SOCS2 regulates iTreg stability by downregulating IL-4 signaling. Moreover, SOCS2 is essential to maintain the anti-inflammatory phenotype of iTregs by preventing the secretion of proinflammatory cytokines. Collectively, these results suggest that SOCS2 may prevent IL-4-induced Foxp3(+) iTreg instability. Foxp3(+) iTregs are key regulators of immune responses at mucosal surfaces; therefore, this dual role of SOCS2 in both Th2 and Foxp3(+) iTregs reinforces SOCS2 as a potential therapeutic target for Th2-biased diseases.


Forkhead Transcription Factors/metabolism , Suppressor of Cytokine Signaling Proteins/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Animals , Forkhead Transcription Factors/genetics , Gene Expression Regulation , Interleukin-4/pharmacology , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice , Mice, Knockout , STAT6 Transcription Factor/metabolism , Suppressor of Cytokine Signaling Proteins/deficiency , Suppressor of Cytokine Signaling Proteins/genetics , T-Lymphocytes, Regulatory/drug effects
13.
Immunol Rev ; 252(1): 164-82, 2013 Mar.
Article En | MEDLINE | ID: mdl-23405904

Intestinal CD4(+) T cells are essential mediators of immune homeostasis and inflammation. Multiple subsets of CD4(+) T cells have been described in the intestine, which represents an important site for the generation and regulation of cells involved in immune responses both within and outside of the gastrointestinal tract. Recent advances have furthered our understanding of the biology of such cells in the intestine. Appreciation of the functional roles for effector and regulatory populations in health and disease has revealed potential translational targets for the treatment of intestinal diseases, including inflammatory bowel disease. Furthermore, the role of dietary and microbiota-derived factors in shaping the intestinal CD4(+) T-cell compartment is becoming increasingly understood. Here, we review recent advances in understanding the multifaceted roles of CD4(+) T cells in intestinal immunity.


CD4-Positive T-Lymphocytes/immunology , Inflammatory Bowel Diseases/immunology , Intestinal Mucosa/immunology , Intestines/immunology , T-Lymphocyte Subsets/immunology , CD4-Positive T-Lymphocytes/pathology , Cytokines/immunology , Homeostasis/immunology , Humans , Immunity, Mucosal , Inflammatory Bowel Diseases/pathology , Intestinal Mucosa/pathology , Intestines/pathology , Metagenome/immunology , T-Lymphocyte Subsets/pathology
14.
Immunity ; 37(6): 1116-29, 2012 Dec 14.
Article En | MEDLINE | ID: mdl-23200826

In interleukin-23 (IL-23)-dependent colitis, there is excessive accumulation of short-lived neutrophils and inflammatory monocytes in the intestine. It is unknown whether this reflects changes in mature cell populations or whether the IL-23-driven colitogenic T cell program regulates upstream hematopoietic stem and progenitor cells (HSPC). Here we have shown dysregulation of hematopoiesis in colitis mediated by inflammatory cytokines. First, there was an interferon-gamma-dependent accumulation of proliferating hematopoietic stem cells in the bone marrow and spleen. Second, there was a strong skew toward granulocyte-monocyte progenitor (GMP) production at the expense of erythroid and lymphoid progenitors. Extramedullary hematopoiesis was also evident, and granulocyte macrophage-colony stimulating factor (GM-CSF) blockade reduced the accumulation of splenic and colonic GMPs, resulting in amelioration of colitis. Importantly, transfer of GMPs exacerbated colitis. These data identify HSPCs as a major target of the IL-23-driven inflammatory axis suggesting therapeutic strategies for the treatment of inflammatory bowel disease.


Colitis/immunology , Colitis/metabolism , Hematopoietic Stem Cells/metabolism , Interleukin-23/physiology , Animals , Bone Marrow Cells/metabolism , Cell Lineage , Cell Proliferation , Colitis/chemically induced , Disease Models, Animal , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Hematopoiesis, Extramedullary , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Interferon-gamma/metabolism , Interferon-gamma/pharmacology , Interleukin-23/metabolism , Intestinal Mucosa/pathology , Intestinal Mucosa/physiology , Mice , Mice, Knockout , Spleen/physiology
15.
J Exp Med ; 209(9): 1595-609, 2012 Aug 27.
Article En | MEDLINE | ID: mdl-22891275

Although very high levels of interleukin (IL)-1ß are present in the intestines of patients suffering from inflammatory bowel diseases (IBD), little is known about the contribution of IL-1ß to intestinal pathology. Here, we used two complementary models of chronic intestinal inflammation to address the role of IL-1ß in driving innate and adaptive pathology in the intestine. We show that IL-1ß promotes innate immune pathology in Helicobacter hepaticus-triggered intestinal inflammation by augmenting the recruitment of granulocytes and the accumulation and activation of innate lymphoid cells (ILCs). Using a T cell transfer colitis model, we demonstrate a key role for T cell-specific IL-1 receptor (IL-1R) signals in the accumulation and survival of pathogenic CD4(+) T cells in the colon. Furthermore, we show that IL-1ß promotes Th17 responses from CD4(+) T cells and ILCs in the intestine, and we describe synergistic interactions between IL-1ß and IL-23 signals that sustain innate and adaptive inflammatory responses in the gut. These data identify multiple mechanisms through which IL-1ß promotes intestinal pathology and suggest that targeting IL-1ß may represent a useful therapeutic approach in IBD.


CD5 Antigens/metabolism , Colitis/immunology , Interleukin-17/metabolism , Interleukin-1beta/metabolism , Th17 Cells/metabolism , Animals , Cell Survival , Colitis/metabolism , Colitis/microbiology , Colon/immunology , Colon/microbiology , Colon/pathology , Granulocytes/immunology , Helicobacter Infections/immunology , Helicobacter Infections/metabolism , Helicobacter Infections/pathology , Helicobacter hepaticus/pathogenicity , Immunity, Innate , Interleukin-23/metabolism , Lymphocytes/immunology , Mice , Mice, Inbred C57BL , Receptors, Interleukin-1/metabolism , Receptors, Interleukin-1 Type I/metabolism , Th17 Cells/immunology
16.
Blood ; 117(20): 5413-24, 2011 May 19.
Article En | MEDLINE | ID: mdl-21450903

Mouse innate-like B cells are a heterogeneous collection of multifunctional cells that control infection, play housekeeping roles, contribute to adaptive immunity, and suppress inflammation. We show that, among leukocytes, chemokine internalization by the D6 receptor is a unique and universal feature of all known innate-like B-cell populations and, to our knowledge, the most effective unifying marker of these cells. Moreover, we identify novel D6(active) B1-cell subsets, including those we term B1d, which lack CD5 and CD11b but exhibit typical B1-cell properties, including spontaneous ex vivo production of IgM, IL-10, and anti-phosphorylcholine antibody. The unprecedented opportunity to examine D6 on primary cells has allowed us to clarify its ligand specificity and show that, consistent with a scavenging role, D6 internalizes chemokines but cannot induce Ca(2+) fluxes or chemotaxis. Unexpectedly, however, D6 can also suppress the function of CXCR5, a critical chemokine receptor in innate-like B-cell biology. This is associated with a reduction in B1 cells and circulating class-switched anti-phosphorylcholine antibody in D6-deficient mice. Therefore, in the present study, we identify a unifying marker of innate-like B cells, describe novel B1-cell subsets, reveal a dual role for D6, and provide the first evidence of defects in resting D6-deficient mice.


B-Lymphocyte Subsets/immunology , Immunity, Innate , Receptors, Chemokine/immunology , Animals , CD11b Antigen/metabolism , CD5 Antigens/metabolism , Calcium Signaling/immunology , Chemokines/metabolism , Chemotaxis/immunology , Female , Lymphoid Tissue/cytology , Lymphoid Tissue/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Omentum/cytology , Omentum/immunology , Peyer's Patches/cytology , Peyer's Patches/immunology , Receptors, Chemokine/deficiency , Receptors, Chemokine/genetics , Receptors, IgE/metabolism , Spleen/cytology , Spleen/immunology
17.
Immunity ; 33(2): 279-88, 2010 Aug 27.
Article En | MEDLINE | ID: mdl-20732640

Mutations in the IL23R gene are linked to inflammatory bowel disease susceptibility. Experimental models have shown that interleukin-23 (IL-23) orchestrates innate and T cell-dependent colitis; however, the cell populations it acts on to induce intestinal immune pathology are unknown. Here, using Il23r(-/-) T cells, we demonstrated that T cell reactivity to IL-23 was critical for development of intestinal pathology, but not for systemic inflammation. Through direct signaling into T cells, IL-23 drove intestinal T cell proliferation, promoted intestinal Th17 cell accumulation, and enhanced the emergence of an IL-17A(+)IFN-gamma(+) population of T cells. Furthermore, IL-23R signaling in intestinal T cells suppressed the differentiation of Foxp3(+) cells and T cell IL-10 production. Although Il23r(-/-) T cells displayed unimpaired Th1 cell differentiation, these cells showed impaired proliferation and failed to accumulate in the intestine. Together, these results highlight the multiple functions of IL-23 signaling in T cells that contribute to its colitogenic activity.


Colitis/immunology , Interleukin-23/immunology , T-Lymphocytes/immunology , Animals , Cell Proliferation , Cells, Cultured , Colitis/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Interleukin/deficiency , Receptors, Interleukin/immunology , T-Lymphocytes/cytology
18.
Cancer Res ; 70(15): 6161-70, 2010 Aug 01.
Article En | MEDLINE | ID: mdl-20631073

CD4(+) T cells play a central role in protective immunity. In a mouse tumor model, we previously found that tumor growth elicits natural CD4(+) T-cell responses, but impedes therapeutic vaccination. We show here that inhibition of vaccine-mediated naïve T-cell priming is due to the presence of a minor but distinct population of tumor-reactive CD4(+) T cells. These cells are generated in the tumor draining lymph nodes (LN), are capable of systemic redistribution, and act to limit the representation of antigen-bearing MHC II(+) antigen-presenting cells (APC) in contralateral LNs or when transferred to tumor-free mice. Surgical tumor resection, which lowers the representation of tumor primed CD4(+) T cells, restored to some extent vaccine-induced CD4(+) T-cell activation. Likewise, vaccination with artificial APCs (latex beads) or higher numbers of dendritic cells allowed comparable CD4(+) T-cell priming in tumor-free and tumor-bearing mice. Together, our results emphasize the ability of antigen-experienced CD4(+) T lymphocytes to interfere with therapeutic vaccination and highlight the need for alternative strategies able to surmount limitations imposed by ongoing immune responses.


Adenocarcinoma/immunology , CD4-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Dendritic Cells/immunology , Mammary Neoplasms, Experimental/immunology , Adenocarcinoma/therapy , Amino Acid Sequence , Animals , Antigen-Presenting Cells/immunology , Antigens, Protozoan/immunology , Cancer Vaccines/pharmacology , Female , Immunodominant Epitopes/immunology , Inbreeding , Lymph Nodes/immunology , Lymphocyte Activation , Male , Mammary Neoplasms, Experimental/therapy , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Molecular Sequence Data , Peptide Fragments/immunology , Protozoan Proteins/immunology , Treatment Failure
19.
J Immunol ; 179(5): 2899-909, 2007 Sep 01.
Article En | MEDLINE | ID: mdl-17709504

Dendritic cells loaded with tumor-derived peptides induce protective CTL responses and are under evaluation in clinical trails. We report in this study that prophylactic administration of dendritic cells loaded with a MHC class II-restricted peptide derived from a model tumor Ag (Leishmania receptor for activated C kinase (LACK)) confers protection against LACK-expressing TS/A tumors, whereas therapeutic vaccination fails to cure tumor-bearing mice. Although CD4+ T cell-directed dendritic cell vaccination primed effector-like (CD44(high)CD62L(low), IL-2(+), IFN-gamma(+)) and central memory-like lymphocytes (CD44(high)CD62L(high), only IL-2(+)) in tumor-free mice, this was not the case in tumor-bearing animals in which both priming and persistence of CD4+ T cell memory were suppressed. Suppression was specific for the tumor-associated Ag LACK, and did not depend on CD25+ T cells. Because T cell help is needed for protective immunity, we speculate that the ability of tumors to limit vaccine-induced CD4+ T cell memory could provide a partial explanation for the limited efficacy of current strategies.


Antigens, Protozoan/immunology , Dendritic Cells/immunology , Histocompatibility Antigens Class II/immunology , Neoplasms/prevention & control , Protozoan Proteins/immunology , Vaccination/methods , Animals , CD11b Antigen/analysis , CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/transplantation , Immunologic Memory , Mice , Mice, Inbred BALB C , Myeloid Cells/immunology , Neoplasms/therapy , Peptides/genetics , Peptides/immunology , Receptors, Chemokine/analysis
...