Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Front Immunol ; 14: 1108895, 2023.
Article in English | MEDLINE | ID: mdl-37006253

ABSTRACT

Background: Eosinophilic oesophagitis (EoE) is a chronic food allergic disorder limited to oesophageal mucosa whose pathogenesis is still only partially understood. Moreover, its diagnosis and follow-up need repeated endoscopies due to absence of non-invasive validated biomarkers. In the present study, we aimed to deeply describe local immunological and molecular components of EoE in well-phenotyped children, and to identify potential circulating EoE-biomarkers. Methods: Blood and oesophageal biopsies were collected simultaneously from French children with EoE (n=17) and from control subjects (n=15). Untargeted transcriptomics analysis was performed on mRNA extracted from biopsies using microarrays. In parallel, we performed a comprehensive analysis of immune components on both cellular and soluble extracts obtained from both biopsies and blood, using flow cytometry. Finally, we performed non-targeted plasma metabolomics using liquid chromatography coupled to high-resolution mass spectrometry (LC-HRMS). Uni/multivariate supervised and non-supervised statistical analyses were then conducted to identify significant and discriminant components associated with EoE within local and/or systemic transcriptomics, immunologic and metabolomics datasets. As a proof of concept, we conducted multi-omics data integration to identify a plasmatic signature of EoE. Results: French children with EoE shared the same transcriptomic signature as US patients. Network visualization of differentially expressed (DE) genes highlighted the major dysregulation of innate and adaptive immune processes, but also of pathways involved in epithelial cells and barrier functions, and in perception of chemical stimuli. Immune analysis of biopsies highlighted EoE is associated with dysregulation of both type (T) 1, T2 and T3 innate and adaptive immunity, in a highly inflammatory milieu. Although an immune signature of EoE was found in blood, untargeted metabolomics more efficiently discriminated children with EoE from control subjects, with dysregulation of vitamin B6 and various amino acids metabolisms. Multi-blocks integration suggested that an EoE plasma signature may be identified by combining metabolomics and cytokines datasets. Conclusions: Our study strengthens the evidence that EoE results from alterations of the oesophageal epithelium associated with altered immune responses far beyond a simplistic T2 dysregulation. As a proof of concept, combining metabolomics and cytokines data may provide a set of potential plasma biomarkers for EoE diagnosis, which needs to be confirmed on a larger and independent cohort.


Subject(s)
Eosinophilic Esophagitis , Humans , Child , Multiomics , Cytokines/metabolism , Adaptive Immunity , Biomarkers
2.
Int J Mol Sci ; 22(19)2021 Sep 30.
Article in English | MEDLINE | ID: mdl-34638950

ABSTRACT

Mucosal-associated invariant T (MAIT) cells represent a distinct T cell population restricted by the MHC-class-I-related molecule, MR1, which recognizes microbial-derived vitamin B2 (riboflavin) metabolites. Their abundance in humans, together with their ability to promptly produce distinct cytokines including interferon γ (IFNγ) and tumor necrosis factor α (TNFα), are consistent with regulatory functions in innate as well as adaptive immunity. Here, we tested whether the alarmin interleukin 33 (IL-33), which is secreted following inflammation or cell damage, could activate human MAIT cells. We found that MAIT cells stimulated with IL-33 produced high levels of IFNγ, TNFα and Granzyme B (GrzB). The action of IL-33 required IL-12 but was independent of T cell receptor (TCR) cross-linking. MAIT cells expressed the IL-33 receptor ST2 (suppression of tumorigenicity 2) and upregulated Tbet (T-box expressed in T cells) in response to IL-12 or IL-33. Electronically sorted MAIT cells also upregulated the expression of CCL3 (Chemokine C-C motif ligand 3), CD40L (CD40 Ligand), CSF-1 (Colony Stimulating Factor 1), LTA (Lymphotoxin-alpha) and IL-2RA (IL-2 receptor alpha chain) mRNAs in response to IL-33 plus IL-12. In conclusion, IL-33 combined with IL-12 can directly target MAIT cells to induce their activation and cytokine production. This novel mechanism of IL-33 activation provides insight into the mode of action by which human MAIT cells can promote inflammatory responses in a TCR-independent manner.


Subject(s)
Interferon-gamma/biosynthesis , Interleukin-33/pharmacology , Lymphocyte Activation/drug effects , Mucosal-Associated Invariant T Cells/drug effects , Mucosal-Associated Invariant T Cells/immunology , Signal Transduction/drug effects , Th1 Cells/immunology , Tumor Necrosis Factor-alpha/biosynthesis , Adult , Blood Donors , Cells, Cultured , Granzymes/biosynthesis , Healthy Volunteers , Humans , Interleukin-1 Receptor-Like 1 Protein/metabolism , Interleukin-12/biosynthesis , Interleukin-12/pharmacology , Interleukin-33/biosynthesis , Receptors, Antigen, T-Cell/immunology , Signal Transduction/immunology , T-Box Domain Proteins/metabolism
3.
Front Immunol ; 12: 700521, 2021.
Article in English | MEDLINE | ID: mdl-34349761

ABSTRACT

Background: Targeted approaches may not account for the complexity of inflammation involved in children with severe asthma (SA), highlighting the need to consider more global analyses. We aimed to identify sets of immune constituents that distinguish children with SA from disease-control subjects through a comprehensive analysis of cells and immune constituents measured in bronchoalveolar lavage (BAL) and blood. Methods: Twenty children with SA and 10 age-matched control subjects with chronic respiratory disorders other than asthma were included. Paired blood and BAL samples were collected and analyzed for a large set of cellular (eosinophils, neutrophils, and subsets of lymphocytes and innate lymphoid cells) and soluble (chemokines, cytokines, and total antibodies) immune constituents. First, correlations of all immune constituents between BAL and blood and with demographic and clinical data were assessed (Spearman correlations). Then, all data were modelled using supervised multivariate analyses (partial least squares discriminant analysis, PLS-DA) to identify immune constituents that significantly discriminate between SA and control subjects. Univariate analyses were performed (Mann-Whitney tests) and then PLS-DA and univariate analyses were combined to identify the most discriminative and significant constituents. Results: Concentrations of soluble immune constituents poorly correlated between BAL and blood. Certain constituents correlated with age or body mass index and, in asthmatics, with clinical symptoms, such as the number of exacerbations in the previous year, asthma control test score, or forced expiratory volume. Multivariate supervised analysis allowed construction of a model capable of distinguishing children with SA from control subjects with 80% specificity and 100% sensitivity. All immune constituents contributed to the model but some, identified by variable-important-in-projection values > 1 and p < 0.1, contributed more strongly, including BAL Th1 and Th2 cells and eosinophilia, CCL26 (Eotaxin 3), IgA and IL-19 concentrations in blood. Blood concentrations of IL-26, CCL13, APRIL, and Pentraxin-3 may also help in the characterization of SA. Conclusions: The analysis of a large set of immune constituents may allow the identification of a biological immune signature of SA. Such an approach may provide new leads for delineating the pathogenesis of SA in children and identifying new targets for its diagnosis, prediction, and personalized treatment.


Subject(s)
Asthma/blood , Asthma/immunology , Biomarkers/analysis , Bronchoalveolar Lavage Fluid/immunology , Child , Female , Humans , Male
5.
Front Immunol ; 11: 595003, 2020.
Article in English | MEDLINE | ID: mdl-33643284

ABSTRACT

Allergic asthma is characterized by airway inflammation with a Th2-type cytokine profile, hyper-IgE production, mucus hypersecretion, and airway hyperreactivity (AHR). It is increasingly recognized that asthma is a heterogeneous disease implicating complex immune mechanisms resulting in distinct endotypes observed in patients. In this study, we showed that non-obese diabetic (NOD) mice, which spontaneously develop autoimmune diabetes, undergo more severe allergic asthma airway inflammation and AHR than pro-Th2 BALB/c mice upon house dust mite (HDM) sensitization and challenge. The use of IL-4-deficient NOD mice and the in vivo neutralization of IL-17 demonstrated that both IL-4 and IL-17 are responsible by the exacerbated airway inflammation and AHR observed in NOD mice. Overall, our findings indicate that autoimmune diabetes-prone NOD mice might become useful as a new HDM-induced asthma model to elucidate allergic dysimmune mechanisms involving Th2 and Th17 responses that could better mimic some asthmatic endoytpes.


Subject(s)
Asthma/immunology , Interleukin-17/immunology , Interleukin-4/immunology , Pyroglyphidae/immunology , Th17 Cells/immunology , Th2 Cells/immunology , Animals , Asthma/pathology , Disease Models, Animal , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Th17 Cells/pathology , Th2 Cells/pathology
7.
PLoS One ; 14(6): e0219047, 2019.
Article in English | MEDLINE | ID: mdl-31251783

ABSTRACT

BACKGROUND: The implication of lymphocytes in sickle cell disease pathogenesis is supported by a number of recent reports. These studies provided evidence for the activation of invariant natural killer T (iNKT) cells in adult patients, but did not investigate the involvement of other innate-like T cell subsets so far. METHODS: Here we present a monocentric prospective observational study evaluating the number and functional properties of both circulating conventional and innate-like T cells, namely iNKT, Mucosal-Associated Invariant T (MAIT) and gammadelta (γδ) T cells in a cohort of 39 children with sickle cell disease. RESULTS: Relative to age-matched healthy controls, we found that patients had a higher frequency of IL-13- and IL-17-producing CD4+ T cells, as well as higher MAIT cell counts with an increased frequency of IL-17-producing MAIT cells. Patients also presented increased Vδ2 γδ T cell counts, especially during vaso-occlusive crisis, and a lower frequency of IFNγ-producing Vδ2 γδ T cells, except during crisis. iNKT cell counts and the frequency of IFNγ-producing iNKT cells were unchanged compared to controls. Our study revealed positive correlations between 1) the frequency of IFNγ-producing CD4+, CD8+ and Vδ2 γδ T cells and the number of hospitalizations for vaso-occlusive crisis in the previous year; 2) the frequency of IFNγ-producing iNKT cells and patients' age and 3) the frequency of IL-17-producing Vδ2 γδ T cells and hemoglobin S level. CONCLUSION: These results strongly suggest a role of innate-like T cells in sickle cell disease pathophysiology, especially that of IL-17-producing MAIT and γδ T cells.


Subject(s)
Anemia, Sickle Cell/immunology , Immunity, Innate/immunology , Mucosal-Associated Invariant T Cells/immunology , Natural Killer T-Cells/immunology , T-Lymphocyte Subsets/immunology , Adolescent , Anemia, Sickle Cell/metabolism , Child , Female , Humans , Male , Mucosal-Associated Invariant T Cells/metabolism , Natural Killer T-Cells/metabolism , Prospective Studies , Receptors, Antigen, T-Cell, gamma-delta/metabolism , T-Lymphocyte Subsets/metabolism
9.
J Diabetes Res ; 2015: 965056, 2015.
Article in English | MEDLINE | ID: mdl-26090474

ABSTRACT

Recent evidence has highlighted the role of histamine in inflammation. Since this monoamine has also been strongly implicated in the pathogenesis of type-1 diabetes, we assessed its effect in the nonobese diabetic (NOD) mouse model. To this end, we used mice (inactivated) knocked out for the gene encoding histidine decarboxylase, the unique histamine-forming enzyme, backcrossed on a NOD genetic background. We found that the lack of endogenous histamine in NOD HDC(-/-) mice decreased the incidence of diabetes in relation to their wild-type counterpart. Whereas the proportion of regulatory T and myeloid-derived suppressive cells was similar in both strains, histamine deficiency was associated with increased levels of immature macrophages, as compared with wild-type NOD mice. Concerning the cytokine pattern, we found a decrease in circulating IL-12 and IFN-γ in HDC(-/-) mice, while IL-6 or leptin remained unchanged, suggesting that histamine primarily modulates the inflammatory environment. Paradoxically, exogenous histamine given to NOD HDC(-/-) mice provided also protection against T1D. Our study supports the notion that histamine is involved in the pathogenesis of diabetes, thus providing additional evidence for its role in the regulation of the immune response.


Subject(s)
Cytokines/immunology , Diabetes Mellitus, Type 1/genetics , Histamine/immunology , Histidine Decarboxylase/genetics , Mice, Inbred NOD/genetics , Animals , Diabetes Mellitus, Type 1/immunology , Interferon-gamma/immunology , Interleukin-12/immunology , Interleukin-6/immunology , Leptin/immunology , Mice , Mice, Inbred NOD/immunology , Mice, Knockout , T-Lymphocytes, Regulatory/immunology
10.
PLoS One ; 8(9): e70292, 2013.
Article in English | MEDLINE | ID: mdl-24086246

ABSTRACT

Basophils co-express FcεRIα and CD49b, the α-2 chain of integrin-type receptor VLA-2 (α2ß1), which recognizes type-1 collagen as a major natural ligand. The physiological relevance of this integrin for interactions with extracellular bone marrow matrix remains unknown. Herein, we examined the expression of several receptors of this family by bone marrow-derived basophils sorted either ex-vivo or after culture with IL-3. Having established that both populations display CD49d, CD49e and CD49f (α-4, α-5 and α-6 integrins subunits, respectively), we addressed receptor functions by measuring migration, adhesion, proliferation and survival after interacting with matched natural ligands. Type I collagen, laminin and fibronectin promoted basophil migration/adhesion, the former being the most effective. None of these ligands affected basophil viability and expansion. Interactions between basophils and extracellular matrix are likely to play a role in situ, as supported by confocal 3D cell imaging of femoral bone marrow sections, which revealed basophils exclusively in type-1 collagen-enriched niches that contained likewise laminin and fibronectin. This is the first evidence for a structure/function relationship between basophils and extracellular matrix proteins inside the mouse bone marrow.


Subject(s)
Basophils/cytology , Bone Marrow Cells/cytology , Cell Movement , Extracellular Matrix , Animals , Basophils/metabolism , Bone Marrow Cells/metabolism , Cell Adhesion , Cells, Cultured , Female , Fluorescent Antibody Technique , Gene Expression Profiling , Male , Mice , Mice, Inbred C57BL , Real-Time Polymerase Chain Reaction
11.
J Allergy Clin Immunol ; 128(4): 864-871.e2, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21636115

ABSTRACT

BACKGROUND: Murine basophils can contribute to the T(H)2 polarization of the immune response by providing rapidly large amounts of IL-4, which suggests that pharmacologic downregulation of this cytokine might provide a strategy to attenuate pathologies associated with excessive production. OBJECTIVE: We examined a number of physiological and pharmacologic ligands of the organic cation transporter 3 (OCT3), a membrane carrier of biogenic amines, for their inhibitory effect on IL-4 production by basophils, selecting the most efficient compounds for in vivo evaluation in basophil-dependent experimental models. METHODS: IL-4 production by basophils isolated ex vivo or from bone marrow cultures was assessed in response to various stimuli with or without biogenic monoamines or pharmacologic analogs. Selected compounds were administered in vivo to examine their effect on levels of circulating IgE generated during a basophil-dependent T(H)2 response and on basophil activation in mice receiving IL-33. RESULTS: We found a drastic decrease in IL-4 production by stimulated basophils on exposure to serotonin (5-hydroxytryptamine [5-HT]) that is taken up by basophils through the specific high-affinity transporters serotonin transporter and the polyspecific, high-capacity organic cation transporter 3 (OCT3; or Slc22a3) but inhibits their function exclusively through the latter. This downregulation is likewise observed in vivo in response to 5-HT and other OCT3 ligands, as well as in human basophils sorted from PMBCs of nonatopic donors. CONCLUSIONS: We provide evidence for a new means of downregulating IL-4 production by basophils, both in vitro and in vivo, through OCT3 targeted by 5-HT and pharmacologic ligands.


Subject(s)
Basophils/immunology , Immunoglobulin E/immunology , Interleukin-4/immunology , Organic Cation Transport Proteins/agonists , Serotonin/immunology , Th2 Cells/immunology , Animals , Basophils/metabolism , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Cells, Cultured , Down-Regulation/drug effects , Down-Regulation/genetics , Down-Regulation/immunology , Female , Humans , Immunoglobulin E/metabolism , Interleukin-33 , Interleukin-4/biosynthesis , Interleukin-4/genetics , Interleukins/immunology , Interleukins/pharmacology , Ligands , Male , Mice , Mice, Knockout , Organic Cation Transport Proteins/genetics , Organic Cation Transport Proteins/immunology , Organic Cation Transport Proteins/metabolism , Serotonin/genetics , Serotonin/metabolism , Serotonin/pharmacology , Serotonin Receptor Agonists/immunology , Serotonin Receptor Agonists/metabolism , Serotonin Receptor Agonists/pharmacology , Th2 Cells/metabolism
12.
PLoS One ; 4(8): e6504, 2009 Aug 07.
Article in English | MEDLINE | ID: mdl-19662098

ABSTRACT

The most recently characterized H4 histamine receptor (H4R) is expressed preferentially in the bone marrow, raising the question of its role during hematopoiesis. Here we show that both murine and human progenitor cell populations express this receptor subtype on transcriptional and protein levels and respond to its agonists by reduced growth factor-induced cell cycle progression that leads to decreased myeloid, erythroid and lymphoid colony formation. H4R activation prevents the induction of cell cycle genes through a cAMP/PKA-dependent pathway that is not associated with apoptosis. It is mediated specifically through H4R signaling since gene silencing or treatment with selective antagonists restores normal cell cycle progression. The arrest of growth factor-induced G1/S transition protects murine and human progenitor cells from the toxicity of the cell cycle-dependent anticancer drug Ara-C in vitro and reduces aplasia in a murine model of chemotherapy. This first evidence for functional H4R expression in hematopoietic progenitors opens new therapeutic perspectives for alleviating hematotoxic side effects of antineoplastic drugs.


Subject(s)
Cell Cycle/physiology , Hematopoietic Stem Cells/drug effects , Intercellular Signaling Peptides and Proteins/pharmacology , Receptors, G-Protein-Coupled/physiology , Receptors, Histamine/physiology , Animals , Cell Proliferation , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Gene Silencing , Hematopoietic Stem Cells/cytology , Humans , Mice , Receptors, Histamine H4 , Signal Transduction
13.
Blood ; 108(3): 1065-72, 2006 Aug 01.
Article in English | MEDLINE | ID: mdl-16597595

ABSTRACT

Two classes of oncogenic mutations of the c-kit tyrosine kinase have been described: the juxtamembrane domain V560G mutation, which is preferentially found in gastrointestinal stromal tumors (GISTs), and the kinase domain D816V mutation, which is highly representative of systemic mastocytosis (SM). Here we show that both mutations constitutively activate the mammalian target of rapamycin (mTOR) signaling pathway. Surprisingly, the mTOR inhibitor rapamycin induces only apoptosis in HMC-1 cells bearing the D816V but not the V560G mutation. In support of this unexpected selectivity, rapamycin inhibits the phosphorylation of 4E-BP1, a downstream substrate of the mTOR pathway, but only in D816V HMC-1 cells. Importantly, D816V mast cells isolated from SM patients or from transgenic mice are sensitive to rapamycin whereas normal human or mouse mast cells are not. Thus, rapamycin inhibition appears specific to the D816V mutation. At present there is no effective cure for SM patients with the D816V mutation. The data presented here provide a rationale to test whether rapamycin could be a possible treatment for SM and other hematologic malignancies with the D816V mutation.


Subject(s)
Mastocytosis, Systemic/drug therapy , Mastocytosis, Systemic/genetics , Mutation, Missense , Pharmacogenetics , Proto-Oncogene Proteins c-kit/genetics , Sirolimus/pharmacology , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Humans , Mast Cells/drug effects , Mast Cells/pathology , Mastocytosis, Systemic/pathology , Mice , Mice, Transgenic , Protein Kinases/metabolism , Signal Transduction , TOR Serine-Threonine Kinases , Tumor Cells, Cultured
14.
J Exp Med ; 202(3): 387-93, 2005 Aug 01.
Article in English | MEDLINE | ID: mdl-16061728

ABSTRACT

In this study, we identify the bidirectional organic cation transporter 3 (OCT3/Slc22a3) as the molecule responsible for histamine uptake by murine basophils. We demonstrate that OCT3 participates in the control of basophil functions because exogenous histamine can inhibit its own synthesis--and that of interleukin (IL)-4, IL-6, and IL-13--through this means of transport. Furthermore, ligands of H3/H4 histamine receptors or OCT3 inhibit histamine uptake, and outward transport of newly synthesized histamine. By doing so, they increase the histamine content of basophils, which explains why they mimic the effect of exogenous histamine. These drugs were no longer effective in histamine-free histidine decarboxylase (HDC)-deficient mice, in contrast with histamine itself. Histamine was not taken up and lost its inhibitory effect in mice deficient for OCT3, which proved its specific involvement. Intracellular histamine levels were increased strongly in IL-3-induced OCT3-/- bone marrow basophils, and explained why they generated fewer cytokines than their wild-type counterpart. Their production was enhanced when histamine synthesis was blocked by the specific HDC inhibitor alpha-fluoro-methyl histidine, and underscored the determinant role of histamine in the inhibitory effect. We postulate that pharmacologic modulation of histamine transport might become instrumental in the control of basophil functions during allergic diseases.


Subject(s)
Basophils/metabolism , Histamine Release/physiology , Histamine/metabolism , Histidine Decarboxylase/metabolism , Organic Cation Transport Proteins/metabolism , Animals , Basophils/cytology , Biological Transport, Active/genetics , Biological Transport, Active/physiology , Cytokines/metabolism , Histamine Release/genetics , Histidine Decarboxylase/genetics , Hypersensitivity/genetics , Hypersensitivity/metabolism , Mice , Mice, Knockout , Organic Cation Transport Proteins/genetics , Receptors, Histamine H3/metabolism
15.
J Biol Chem ; 278(21): 18859-67, 2003 May 23.
Article in English | MEDLINE | ID: mdl-12618431

ABSTRACT

4E-BP1 plays a major role in translation by inhibiting cap-dependent translation initiation. Several reports have investigated the regulation of 4E-BP1 phosphorylation, which varies along with cell differentiation and upon various stimulations, but very little is known about the regulation of its expression. In a first part, we show that the expression of 4E-BP1 protein and transcript decreases in hematopoietic cell lines cultivated in the presence of phorbol 12-myristate 13-acetate (PMA). This decrease depends on the activation of the ERK/mitogen-activated protein kinases. 4E-BP1 expression also decreases when the p38/mitogen-activated protein kinase pathway is activated by granulocyte/macrophage colony-stimulating factor but to a lesser extent than with PMA. In a second part, we examine how 4e-bp1 promoter activity is regulated. PMA and granulocyte/macrophage colony-stimulating factor induce Egr-1 expression through ERK and p38 activation, respectively. Using a dominant negative mutant of Egr, ZnEgr, we show that this transcription factor is responsible for the inhibition of 4e-bp1 promoter activity. In a third part we show that histidine decarboxylase, whose activity and expression are inversely correlated with 4E-BP1 expression, is a potential target for the translational machinery. These data (i) are the first evidence of a new role of ERK and p38 on the translational machinery and (ii) demonstrate that 4E-BP1 is a new target for Egr-1.


Subject(s)
Carrier Proteins/genetics , DNA-Binding Proteins/physiology , Gene Expression , Immediate-Early Proteins , JNK Mitogen-Activated Protein Kinases , Mitogen-Activated Protein Kinases/physiology , Phosphoproteins/genetics , Promoter Regions, Genetic/genetics , Repressor Proteins/genetics , Transcription Factors/physiology , Adaptor Proteins, Signal Transducing , Base Sequence , Blotting, Northern , Blotting, Western , Carrier Proteins/metabolism , Cell Cycle Proteins , Cell Line , Chromones/pharmacology , Cycloheximide/pharmacology , DNA-Binding Proteins/genetics , Early Growth Response Protein 1 , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Gene Expression/drug effects , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Histamine/metabolism , Histidine Decarboxylase/genetics , Histidine Decarboxylase/metabolism , Humans , Hypotonic Solutions , Kinetics , MAP Kinase Kinase 4 , Mitogen-Activated Protein Kinase 11 , Mitogen-Activated Protein Kinase 8 , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Molecular Sequence Data , Morpholines/pharmacology , Mutation , Phosphoinositide-3 Kinase Inhibitors , Phosphoproteins/metabolism , Phosphorylation , Phosphoserine/metabolism , RNA, Messenger/analysis , Response Elements , Sirolimus/pharmacology , Tetradecanoylphorbol Acetate/pharmacology , Transcription Factors/genetics , p38 Mitogen-Activated Protein Kinases
16.
Eur J Immunol ; 32(7): 1897-904, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12115609

ABSTRACT

Natural killer (NK) T cells are prominent for their prompt IL-4 and IFN-gamma production upon TCR ligation that enables them to influence acquired immune responses. In the present study we provide evidence that the regulatory functions of this particular T cell subset extend to the myeloid compartment of bone marrow and spleen through its production of hematopoietic growth factors. Bone marrow and spleen NKT cells responded to a single injection of their specific ligand alpha-galactosylceramide (alpha-GalCer) by producing both IL-3 and granulocyte-macrophage colony stimulating factor (GM-CSF), whose colony-stimulating activity became detectable in the serum as early as 1 h post treatment. These cytokines were not produced in mice lacking NKT cells (CD1d-/-), whose exclusive involvement in this biological activity was further confirmed by intracellular immuno-staining. Growth factor production was accompanied by significant changes in the myeloid compartment of treated mice, namely mobilization of myeloid progenitors (colony-forming unit cells, CFU-C) and neutrophils from the bone marrow to the periphery. Taken together, our data support the notion that activated NKT cells influence innate immune responses by recruiting myeloid progenitors and granulocytes to the periphery through their production of hematopoietic growth factors.


Subject(s)
Galactosylceramides/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Interleukin-3/biosynthesis , Killer Cells, Natural/immunology , Myeloid Cells/immunology , T-Lymphocytes/immunology , Animals , Galactosylceramides/administration & dosage , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocytes/cytology , Granulocytes/immunology , Hematopoiesis, Extramedullary , Injections , Interferon-gamma/genetics , Interleukin-3/genetics , Interleukin-4/genetics , Killer Cells, Natural/drug effects , Ligands , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/cytology , T-Lymphocytes/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...