Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Allergy ; 77(3): 856-869, 2022 03.
Article in English | MEDLINE | ID: mdl-34460953

ABSTRACT

BACKGROUND: Children with asthma have impaired production of interleukin (IL) 37; in mice, IL-37 reduces hallmarks of experimental allergic asthma (EAA). However, it remains unclear how IL-37 exerts its inhibitory properties in asthma. This study aimed to identify the mechanism(s) by which IL-37 controls allergic inflammation. METHODS: IL-37 target cells were identified by single-cell RNA-seq of IL-1R5 and IL-1R8. Airway tissues were isolated by laser-capture microdissection and examined by microarray-based gene expression analysis. Mononuclear cells (MNC) and airway epithelial cells (AECs) were isolated and stimulated with allergen, IL-1ß, or IL-33 together with recombinant human (rh) IL-37. Wild-type, IL-1R1- and IL-33-deficient mice with EAA were treated with rhIL-37. IL-1ß, IL-33, and IL-37 levels were determined in sputum and nasal secretions from adult asthma patients without glucocorticoid therapy. RESULTS: IL-37 target cells included AECs, T cells, and dendritic cells. In mice with EAA, rhIL-37 led to differential expression of >90 genes induced by IL-1ß and IL-33. rhIL-37 reduced production of Th2 cytokines in allergen-activated MNCs from wild-type but not from IL-1R1-deficient mice and inhibited IL-33-induced Th2 cytokine release. Furthermore, rhIL-37 attenuated IL-1ß- and IL-33-induced pro-inflammatory mediator expression in murine AEC cultures. In contrast to wild-type mice, hIL-37 had no effect on EAA in IL-1R1- or IL-33-deficient mice. We also observed that expression/production ratios of both IL-1ß and IL-33 to IL-37 were dramatically increased in asthma patients compared to healthy controls. CONCLUSION: IL-37 downregulates allergic airway inflammation by counterbalancing the disease-amplifying effects of IL-1ß and IL-33.


Subject(s)
Asthma , Interleukin-33 , Allergens , Animals , Asthma/metabolism , Cytokines , Disease Models, Animal , Humans , Inflammation , Lung/metabolism , Mice , Th2 Cells
2.
Med Microbiol Immunol ; 209(4): 531-543, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32507938

ABSTRACT

Mast cells (MCs) are long-living immune cells highly specialized in the storage and release of different biologically active compounds and are involved in the regulation of innate and adaptive immunity. MC degranulation and replacement of MC granules are accompanied by active membrane remodelling. Tetraspanins represent an evolutionary conserved family of transmembrane proteins. By interacting with lipids and other membrane and intracellular proteins, they are involved in organisation of membrane protein complexes and act as "molecular facilitators" connecting extracellular and cytoplasmic signaling elements. MCs express different tetraspanins and MC degranulation is accompanied by changes in membrane organisation. Therefore, tetraspanins are very likely involved in the regulation of MC exocytosis and membrane reorganisation after degranulation. Antiviral response and production of exosomes are further aspects of MC function characterized by dynamic changes of membrane organization. In this review, we pay a particular attention to tetraspanin gene expression in different human and murine MC populations, discuss tetraspanin involvement in regulation of key MC signaling complexes, and analyze the potential contribution of tetraspanins to MC antiviral response and exosome production. In-depth knowledge of tetraspanin-mediated molecular mechanisms involved in different aspects of the regulation of MC response will be beneficial for patients with allergies, characterized by overwhelming MC reactions.


Subject(s)
Mast Cells/immunology , Mast Cells/metabolism , Tetraspanins/immunology , Tetraspanins/metabolism , Animals , Cell Degranulation , Exosomes/metabolism , Humans , Mice , Signal Transduction , Tetraspanins/genetics , Virus Diseases/immunology
3.
J Allergy Clin Immunol ; 144(4S): S4-S18, 2019 10.
Article in English | MEDLINE | ID: mdl-30468774

ABSTRACT

Mast cells (MCs), which are well known for their effector functions in TH2-skewed allergic and also autoimmune inflammation, have become increasingly acknowledged for their role in protection of health. It is now clear that they are also key modulators of immune responses at interface organs, such as the skin or gut. MCs can prime tissues for adequate inflammatory responses and cooperate with dendritic cells in T-cell activation. They also regulate harmful immune responses in trauma and help to successfully orchestrate pregnancy. This review focuses on the beneficial effects of MCs on tissue homeostasis and elimination of toxins or venoms. MCs can enhance pathogen clearance in many bacterial, viral, and parasitic infections, such as through Toll-like receptor 2-triggered degranulation, secretion of antimicrobial cathelicidins, neutrophil recruitment, or provision of extracellular DNA traps. The role of MCs in tumors is more ambiguous; however, encouraging new findings show they can change the tumor microenvironment toward antitumor immunity when adequately triggered. Uterine tissue remodeling by α-chymase (mast cell protease [MCP] 5) is crucial for successful embryo implantation. MCP-4 and the tryptase MCP-6 emerge to be protective in central nervous system trauma by reducing inflammatory damage and excessive scar formation, thereby protecting axon growth. Last but not least, proteases, such as carboxypeptidase A, released by FcεRI-activated MCs detoxify an increasing number of venoms and endogenous toxins. A better understanding of the plasticity of MCs will help improve these advantageous effects and hint at ways to cut down detrimental MC actions.


Subject(s)
Immunity, Innate , Infections/immunology , Mast Cells/immunology , Animals , Cathelicidins/metabolism , Cell Degranulation , Embryo Implantation , Female , Homeostasis , Humans , Pregnancy , Toll-Like Receptor 2/metabolism
4.
Allergy ; 74(4): 767-779, 2019 04.
Article in English | MEDLINE | ID: mdl-30341777

ABSTRACT

BACKGROUND: Food-induced anaphylaxis is a serious allergic reaction caused by Fcε-receptor activation on mast cells (MCs). The exact mechanisms breaking oral tolerance and the effector pathways driving food allergy remain elusive. As complement is activated in food-induced anaphylaxis, we aimed to assess the role of C5a in disease pathogenesis. METHODS: Oral antigen-induced food-induced anaphylaxis was induced in BALB/c wild-type (wt) and C5ar1-/- mice. Readouts included diarrhea development, changes in rectal temperature, hematocrit, antigen-specific serum IgE, MCPT-1, and intestinal MC numbers, as well as FcεR1-mediated MC functions including C5a receptor 1 (C5aR1) regulation. Further, histamine-mediated hypothermia and regulation of endothelial tight junctions were determined. RESULTS: Repeated oral OVA challenge resulted in diarrhea, hypothermia, increased hematocrit, high OVA-specific serum IgE, and MCPT-1 levels in wt mice. Male C5ar1-/- mice were completely whereas female C5ar1-/- mice were partially protected from anaphylaxis development. Serum MCPT-1 levels were reduced gender-independent, whereas IgE levels were reduced in male but not in female C5ar1-/- mice. Mechanistically, IgE-mediated degranulation and IL-6 production from C5ar1-/- BMMCs of both sexes were significantly reduced. Importantly, FcεR1 cross-linking strongly upregulated C5aR1 MC expression in vitro and in vivo. Finally, C5ar1-/- male mice were largely protected from histamine-induced hypovolemic shock, which was associated with protection from histamine-induced barrier dysfunction in vitro following C5aR targeting. CONCLUSIONS: Our findings identify C5aR1 activation as an important driver of IgE-mediated food allergy through regulation of allergen-specific IgE production, FcεR1-mediated MC degranulation, and histamine-driven effector functions preferentially in male mice.


Subject(s)
Food Hypersensitivity/etiology , Immunoglobulin E/blood , Receptor, Anaphylatoxin C5a/genetics , Sex Factors , Anaphylaxis , Animals , Cell Degranulation , Chymases/blood , Female , Male , Mast Cells/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout , Receptor, Anaphylatoxin C5a/deficiency , Receptors, IgE/immunology
5.
Immunity ; 31(4): 665-76, 2009 Oct 16.
Article in English | MEDLINE | ID: mdl-19818652

ABSTRACT

The characteristics, importance, and molecular requirements for interactions between mast cells (MCs) and CD8(+) T cells have not been elucidated. Here, we demonstrated that MCs induced antigen-specific CD8(+) T cell activation and proliferation. This process required direct cell contact and MHC class I-dependent antigen cross-presentation by MCs and induced the secretion of interleukin-2, interferon-gamma, and macrophage inflammatory protein-1alpha by CD8(+) T cells. MCs regulated antigen-specific CD8(+) T cell cytotoxicity by increasing granzyme B expression and by promoting CD8(+) T cell degranulation. Because MCs also upregulated their expression of costimulatory molecules (4-1BB) and released osteopontin upon direct T cell contact, MC-T cell interactions probably are bidirectional. In vivo, adoptive transfer of antigen-pulsed MCs induced MHC class I-dependent, antigen-specific CD8(+) T cell proliferation, and MCs regulated CD8(+) T cell-specific priming in experimental autoimmune encephalomyelitis. Thus, MCs are important players in antigen-specific regulation of CD8(+) T cells.


Subject(s)
Antigen Presentation/immunology , CD8-Positive T-Lymphocytes/immunology , Cross-Priming/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Mast Cells/immunology , Adoptive Transfer , Animals , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/microbiology , Cell Degranulation/immunology , Chemokine CCL3/biosynthesis , Chemokine CCL3/immunology , Coculture Techniques , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Glycoproteins/immunology , Granzymes/immunology , Granzymes/metabolism , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Interleukin-2/biosynthesis , Interleukin-2/immunology , Listeria monocytogenes/immunology , Listeriosis/immunology , Listeriosis/microbiology , Mast Cells/metabolism , Mast Cells/microbiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myelin-Oligodendrocyte Glycoprotein , Osteopontin/immunology , Osteopontin/metabolism , Ovalbumin/immunology , Peptide Fragments/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism
6.
Eur J Immunol ; 46(2): 432-9, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26564814

ABSTRACT

The interaction between tissue-resident mast cells (MCs) and recruited immune cells contributes to tissue immunosurveillance. However, the cells, mechanisms, and receptors involved in this crosstalk remain ill defined. Invariant natural killer T (iNKT) cells are CD1d-restricted innate lymphocytes that recognize glycolipid antigens and have emerged as critical players in immunity. Here, we show that primary mouse peritoneal MCs express surface CD1d, which is upregulated in vivo following administration of alpha-galactosylceramide. In contrast, in BM-derived MCs CD1d was found to be stored intracellularly and to relocate at the cell surface upon IgE-mediated degranulation. Activated BM-derived MCs expressing surface CD1d and loaded with alpha-galactosylceramide were found to induce iNKT-cell proliferation and the release of IFN-γ, IL-13, and IL-4 in a CD1d-restricted manner. Moreover, the costimulatory molecules CD48, CD137L, CD252, CD274, and CD275 affected MC-induced IFN-γ release and iNKT-cell proliferation. Interestingly, among the costimulatory molecules, CD48 and CD252 exhibited a distinctly regulatory activity on iNKT-cell release of both IFN-γ and IL-13. In conclusion, we demonstrate that the crosstalk between MCs and iNKT cells may regulate inflammatory immune responses.


Subject(s)
Antigens, CD1d/metabolism , Lymphocyte Activation , Mast Cells/immunology , Natural Killer T-Cells/immunology , OX40 Ligand/metabolism , Animals , Antigens, CD1d/genetics , Cell Communication , Cell Differentiation , Cells, Cultured , Cytokines/metabolism , Galactosylceramides/administration & dosage , Gene Expression Regulation , Immunity, Innate/drug effects , Immunomodulation , Lymphocyte Activation/drug effects , Mast Cells/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Monitoring, Immunologic , OX40 Ligand/genetics
7.
Eur J Immunol ; 43(4): 979-88, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23404570

ABSTRACT

Mast cells are important cellular constituents of epithelial-mesenchymal interactions, densely located at sites of microbial entry into the host where they are continuously exposed to products from commensals. In order to avoid excessive activation and the associated pathology, mast cell responses to TLR agonists must be tightly regulated. Here, we show that exposure in vitro to subactivating levels of the epithelial cell product, IL-33, renders mast cells insensitive to bacterial cell wall products. Mast cell responsiveness to Ag, cytoplasmic dsDNA, and TLR7/8 agonists is unaffected or enhanced by IL-33. The IL-33-induced mast cell selective tolerance requires the IL-33 receptor ST2 and peritoneal mast cells from St2(-/-) mice display a constitutively activated phenotype, demonstrated by increased expression of activation markers including CD11b and CD28. IL-33 exposure neither affects the levels of TLR4, MyD88, TIRAP, IL-1R associated kinase 2 (IRAK2), or IRAK4, nor induces persistent A20 or Tollip expression, but potently causes ST2-dependent IRAK1 degradation. We show that while IRAK2 is redundant for TLR4 signaling, IRAK1 is essential for TLR4 signaling in mast cells. We suggest that IL-33 produced during homeostasis retains mast cells in an unresponsive state to bacterial cell wall products via IRAK1 degradation, thus preventing chronic inflammation and tissue destruction.


Subject(s)
Immune Tolerance/drug effects , Interleukin-1 Receptor-Associated Kinases/metabolism , Interleukins/pharmacology , Lipopolysaccharides/immunology , Mast Cells/immunology , Mast Cells/metabolism , Animals , Cells, Cultured , Endotoxins/immunology , Immune Tolerance/genetics , Interleukin-1 Receptor-Associated Kinases/genetics , Interleukin-1 Receptor-Like 1 Protein , Interleukin-33 , Mast Cells/drug effects , Mice , Mice, Knockout , Models, Biological , Proteolysis , Receptors, Interleukin/genetics , Receptors, Interleukin/metabolism , Signal Transduction
8.
Eur J Immunol ; 43(1): 194-208, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23042080

ABSTRACT

Syntaxin 11 (STX11) controls vesicular trafficking and is a key player in exocytosis. Since Stx11 mutations are causally associated with a familial hemophagocytic lymphohistio-cytosis, we wanted to clarify whether STX11 is functionally important for key immune cell populations. This was studied in primary cells obtained from newly generated Stx11(-/-) mice. Our data revealed that STX11 is not only widely expressed in different immune cells, but also induced upon LPS or IFN-γ treatment. However, Stx11 deficiency does not affect macrophage phagocytic function and cytokine secretion, mast cell activation, or antigen presentation by DCs. Instead, STX11 selectively controls lymphocyte cytotoxicity in NK and activated CD8(+) T cells and degranulation in neutrophils. Stx11(-/-) NK cells and CTLs show impaired degranulation, despite a comparable activation, maturation and expression of the complex-forming partners MUNC18-2 and VTI1B. In addition, Stx11(-/-) CTLs and NK cells produce abnormal levels of IFN-γ. Since functional reconstitution rescues the defective phenotype of Stx11(-/-) CTLs, we suggest a direct, specific and key role of STX11 in controlling lymphocyte cytotoxicity, cytokine production and secretion. Finally, we show that these mice are a very useful tool for dissecting the role of STX11 in vesicular trafficking and secretion.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Killer Cells, Natural/immunology , Neutrophils/immunology , Qa-SNARE Proteins/immunology , Animals , Cell Degranulation/genetics , Cell Line , Cytotoxicity, Immunologic/genetics , Humans , Interferon-gamma/immunology , Lipopolysaccharides/immunology , Lymphohistiocytosis, Hemophagocytic/genetics , Lymphohistiocytosis, Hemophagocytic/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Munc18 Proteins/immunology , Mutation/genetics , Qa-SNARE Proteins/genetics , Qb-SNARE Proteins/immunology
9.
Nat Med ; 13(8): 927-34, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17643110

ABSTRACT

Sepsis remains a global clinical problem. By using the mouse cecal ligation and puncture model of sepsis, here we identify an important aspect of mast cell (MC)-dependent, innate immune defenses against Gram-negative bacteria by demonstrating that MC protease activity is regulated by interleukin-15 (IL-15). Mouse MCs express both constitutive and lipopolysaccharide-inducible IL-15 and store it intracellularly. Deletion of Il15 in mice markedly increases chymase activities, leading to greater MC bactericidal responses, increased processing and activation of neutrophil-recruiting chemokines, and significantly higher survival rates of mice after septic peritonitis. By showing that intracellular IL-15 acts as a specific negative transcriptional regulator of a mouse MC chymase (mast cell protease-2), we provide evidence that defined MC protease activity is transcriptionally regulated by an intracellularly retained cytokine. Our results identify an unexpected breach in MC-dependent innate immune defenses against sepsis and suggest that inhibiting intracellular IL-15 in MCs may improve survival from sepsis.


Subject(s)
Chymases/metabolism , Interleukin-15/metabolism , Mast Cells/metabolism , Sepsis/metabolism , Sepsis/microbiology , Animals , Chemokine CCL8 , Chemotaxis , Down-Regulation , Escherichia coli/physiology , Gene Deletion , Interleukin-15/deficiency , Interleukin-15/genetics , Mast Cells/cytology , Mice , Mice, Knockout , Monocyte Chemoattractant Proteins/genetics , Sepsis/genetics , Sepsis/pathology , Signal Transduction , Survival Rate , Transcription, Genetic/genetics , Up-Regulation
10.
Blood ; 116(15): 2665-75, 2010 Oct 14.
Article in English | MEDLINE | ID: mdl-20595514

ABSTRACT

Mast cell (MC) differentiation, survival, and activation are controlled by the membrane tyrosine kinase c-Kit upon interaction with stem cell factor (SCF). Here we describe a single point mutation induced by N-ethyl-N-nitrosurea (ENU) mutagenesis in C57BL/6J mice-an A to T transversion at position 2388 (exon 17) of the c-Kit gene, resulting in the isoleucine 787 substitution by phenylalanine (787F), and analyze the consequences of this mutation for ligand binding, signaling, and MC development. The Kit(787F/787F) mice carrying the single amino acid exchange of c-Kit lacks both mucosal and connective tissue-type MCs. In bone marrow-derived mast cells (BMMCs), the 787F mutation does not affect SCF binding and c-Kit receptor shedding, but strongly impairs SCF-induced cytokine production, degranulation enhancement, and apoptosis rescue. Interestingly, c-Kit downstream signaling in 787F BMMCs is normally initiated (Erk1/2 and p38 activation as well as c-Kit autophosphorylation) but fails to be sustained thereafter. In addition, 787F c-Kit does not efficiently mediate Cbl activation, leading to the absence of subsequent receptor ubiquitination and impaired c-Kit internalization. Thus, I787 provides nonredundant signals for c-Kit internalization and functionality.


Subject(s)
Cell Differentiation/physiology , Mast Cells/cytology , Mast Cells/metabolism , Proto-Oncogene Proteins c-kit/chemistry , Proto-Oncogene Proteins c-kit/metabolism , Amino Acid Sequence , Amino Acid Substitution , Animals , Base Sequence , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line , Cell Survival/genetics , Cell Survival/physiology , DNA Primers/genetics , In Vitro Techniques , Interleukin-3/pharmacology , Isoleucine/chemistry , Mast Cells/drug effects , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Mutant Strains , Mutagenesis, Site-Directed , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Point Mutation , Proto-Oncogene Proteins c-cbl/metabolism , Proto-Oncogene Proteins c-kit/genetics , Signal Transduction , Stem Cell Factor/metabolism
11.
Int Arch Allergy Immunol ; 158(1): 43-53, 2012.
Article in English | MEDLINE | ID: mdl-22212634

ABSTRACT

BACKGROUND: Allergen-specific immunoglobulin (Ig) E initiates the effector cascade of allergic asthma and has been identified as a valuable target for therapeutic treatment of this disease. The proteasome inhibitor bortezomib was previously shown to deplete Ig-secreting plasma cells and to efficiently suppress Ig serum titers. The present study aimed at evaluating the therapeutic potential of the proteasome inhibitor bortezomib in allergic bronchial asthma. METHODS: To address this question, a chronic experimental asthma mouse model was used in a therapeutic setting. Mice were sensitized to ovalbumin (OVA) and challenged with OVA aerosol for 12 weeks. After 6 weeks of challenge, bortezomib treatment was started and continued for 1 week (short-term) or 6 weeks (long-term) with a dosage of 0.75 mg/kg body weight twice a week. Lung function, lung histology, Ig serum titers and plasma cell numbers were assessed. RESULTS: Whereas short-term treatment lowered bronchoalveolar lavage eosinophils, long-term treatment considerably reduced serum titers of anti-OVA IgE in mice with chronic experimental asthma. However, neither short-term nor long-term treatment significantly reduced plasma cell numbers, anti-OVA IgG1 serum titers or allergic airway inflammation or ablated airway hyperresponsiveness. CONCLUSION: Our results suggest that bortezomib treatment has only limited value as plasma cell-depleting therapy against allergic bronchial asthma.


Subject(s)
Allergens/immunology , Anti-Asthmatic Agents/therapeutic use , Asthma/drug therapy , Boronic Acids/therapeutic use , Immunoglobulin E/immunology , Pyrazines/therapeutic use , Animals , Asthma/immunology , Asthma/physiopathology , Bortezomib , Chronic Disease , Disease Models, Animal , Eosinophils/drug effects , Eosinophils/immunology , Female , Immunoglobulin E/blood , Immunoglobulin G/blood , Immunoglobulin G/immunology , Lung/immunology , Lung/pathology , Lung/physiopathology , Mice , Mice, Inbred BALB C , Plasma Cells/drug effects , Plasma Cells/immunology , Treatment Failure
12.
J Immunol ; 183(5): 3004-13, 2009 Sep 01.
Article in English | MEDLINE | ID: mdl-19657094

ABSTRACT

Axl, a prototypic member of the transmembrane tyrosine kinase receptor family, is known to regulate innate immunity. In this study, we show that Axl expression is induced by IFN-alpha during human dendritic cell (DC) differentiation from monocytes (IFN/DC) and that constitutively Axl-negative, IL-4-differentiated DC (IL-4/DC) can be induced to up-regulate Axl by IFN-alpha. This effect is inhibited by TLR-dependent maturation stimuli such as LPS, poly(I:C), TLR7/8 ligand, and CD40L. LPS-induced Axl down-regulation on the surface of human IFN-alpha-treated DC correlates with an increased proteolytic cleavage of Axl and with elevated levels of its soluble form. GM6001 and TAPI-1, general inhibitors of MMP and ADAM family proteases, restored Axl expression on the DC surface and diminished Axl shedding. Furthermore, stimulation of Axl by its ligand, Gas6, induced chemotaxis of human DC and rescued them from growth factor deprivation-induced apoptosis. Our study provides the first evidence that Gas6/Axl-mediated signaling regulates human DC activities, and identifies Gas6/Axl as a new DC chemotaxis pathway. This encourages one to explore whether dysregulation of this novel pathway in human DC biology is involved in autoimmunity characterized by high levels of IFN-alpha.


Subject(s)
Cell Movement/immunology , Dendritic Cells/immunology , Intercellular Signaling Peptides and Proteins/physiology , Interferon-alpha/physiology , Oncogene Proteins/physiology , Receptor Protein-Tyrosine Kinases/physiology , Signal Transduction/immunology , Cell Differentiation/immunology , Cell Survival/immunology , Cells, Cultured , Chemotaxis, Leukocyte/immunology , Dendritic Cells/cytology , Dendritic Cells/metabolism , Humans , Intercellular Signaling Peptides and Proteins/biosynthesis , Interleukin-4/physiology , Lipopolysaccharides/physiology , Oncogene Proteins/antagonists & inhibitors , Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/biosynthesis , Up-Regulation/immunology , Axl Receptor Tyrosine Kinase
13.
J Biol Chem ; 284(47): 32635-41, 2009 Nov 20.
Article in English | MEDLINE | ID: mdl-19801677

ABSTRACT

Mast cells (MCs) play a critical role in innate and adaptive immunity through the release of cytokines, chemokines, lipid mediators, biogenic amines, and proteases. We recently showed that the activities of MC proteases are transcriptionally regulated by intracellularly retained interleukin-15 (IL-15), and we provided evidence that this cytokine acts as a specific regulator of mouse mast cell protease-2 (mMCP-2). Here, we show that in wild-type bone marrow-derived mast cells (BMMCs) IL-15 inhibits mMCP-2 transcription indirectly by inducing differential expression and mMCP-2 promoter binding of the bifunctional transcription factors C/EBPbeta and YY1. In wild-type BMMCs, C/EBPbeta expression predominates over YY1 expression, and thus C/EBPbeta preferentially binds to the mMCP-2 promoter. In IL-15-deficient BMMCs, the opposite is found: YY1 expression predominates and binds to the mMCP-2 promoter at the expense of C/EBPbeta. Hypertranscription of the mMCP-2 gene in IL-15-deficient BMMCs is associated with histone acetylation and, intriguingly, with methylation of non-CpG dinucleotides within the MCP-2 promoter. This suggests a novel model of cytokine-controlled protease transcription: non-CpG methylation maintains a chromosomal domain in an "open" configuration that is permissive for gene expression.


Subject(s)
Chymases/biosynthesis , Interleukin-15/physiology , Transcription, Genetic , Animals , Base Sequence , Bone Marrow Cells/cytology , CCAAT-Enhancer-Binding Protein-beta/metabolism , COS Cells , Chlorocebus aethiops , Epigenesis, Genetic , Histones/chemistry , Humans , Mast Cells/cytology , Mice , Models, Biological , Molecular Sequence Data
14.
Exp Cell Res ; 315(17): 3064-75, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19632221

ABSTRACT

The regulation of mast cell activities and survival is a central issue in inflammatory immune responses. Here, we have investigated the role of mouse interleukin-15, a pro-inflammatory and pleiotropic cytokine, in the control of mast cell survival and homeostasis. We report that aged IL-15-/- mice show a reduced number of peritoneal mast cells compared to WT mice. Furthermore, IL-15 deficiency in bone marrow derived mouse mast cells (BMMCs) results in increased susceptibility to apoptosis mediated by growth factor deprivation and A-SMase-treatment. IL-15-/- BMMCs show a constitutive stronger mRNA and protein expression as well as enzymatic activity of the members of the mitochondrial apoptotic pathways including acidic lysosomal aspartate protease cathepsin D (CTSD), endogenous acid sphingomyelinase (A-SMase), caspase-3 and -7 compared to wild type (WT) BMMCs. Furthermore, IL-15-/- BMMCs constitutively generate more A-SMase-derived ceramide than WT controls and display a decreased expression of pro-survival sphingosin-1-phosphate (SPP) both in cytosol and membrane cell fractions. Furthermore, pre-treatment of mast cells with imipramine or pepstatin A, inhibitors of the intracellular acid sphingomyelinase and cathepsin D pathways respectively, increases survival in IL-15-/- BMMCs. These findings suggest that intracellular IL-15 is a key regulator of pathways controlling primary mouse mast cell homeostasis.


Subject(s)
Cell Survival/physiology , Interleukin-15/physiology , Mast Cells/cytology , Mast Cells/physiology , Animals , Apoptosis/drug effects , Apoptosis/physiology , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/physiology , Ceramides/metabolism , Growth Substances/pharmacology , Growth Substances/physiology , Homeostasis/physiology , Interleukin-15/deficiency , Interleukin-15/genetics , Mast Cells/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , RNA, Messenger/genetics
15.
Front Immunol ; 10: 1174, 2019.
Article in English | MEDLINE | ID: mdl-31191542

ABSTRACT

Mast cells (MCs) have long been mainly regarded as effector cells in IgE-associated allergic disorders with potential immunoregulatory roles. Located close to the allergen entry sites in the skin and mucosa, MCs can capture foreign substances such as allergens, toxins, or noxious substances and are exposed to the danger signals produced by epithelial cells. MC reactivity shaped by tissue-specific factors is crucial for allergic responses ranging from local skin reactions to anaphylactic shock. Development of Th2 response leading to allergen-specific IgE production is a prerequisite for MC sensitization and induction of FcεRI-mediated MC degranulation. Up to now, IgE production has been mainly associated with proteins, whereas lipids present in plant pollen grains, mite fecal particles, insect venoms, or food have been largely overlooked regarding their immunostimulatory and immunomodulatory properties. Recent studies, however, have now demonstrated that lipids affect the sensitization process by modulating innate immune responses of epithelial cells, dendritic cells, and NK-T cells and thus crucially contribute to the outcome of sensitization. Whether and how lipids affect also MC effector functions in allergic reactions has not yet been fully clarified. Here, we discuss how lipids can affect MC responses in the context of allergic inflammation. Direct effects of immunomodulatory lipids on MC degranulation, changes in local lipid composition induced by allergens themselves and changes in lipid transport affecting MC reactivity are possible mechanisms by which the function of MC might be modulated.


Subject(s)
Immunomodulation , Lipids/immunology , Mast Cells/immunology , Mast Cells/metabolism , Allergens/chemistry , Allergens/immunology , Animals , Cell Degranulation/immunology , Disease Susceptibility , Humans , Hypersensitivity/etiology , Hypersensitivity/metabolism , Lipid Metabolism , Lipids/chemistry , Membrane Lipids/immunology , Membrane Lipids/metabolism , Structure-Activity Relationship
16.
Front Immunol ; 10: 974, 2019.
Article in English | MEDLINE | ID: mdl-31134071

ABSTRACT

Plant pollen are an important source of antigens that evoke allergic responses. Protein antigens have been the focus of studies aiming to elucidate the mechanisms responsible for allergic reactions to pollen. However, proteins are not the sole active agent present in pollen. It is known that pollen grains contain lipids essential for its reproduction and bioactive lipid mediators. These small molecular compounds are co-delivered with the allergens and hence have the potential to modulate the immune response of subjects by activating their innate immune cells. Previous reports showed that pollen associated lipid mediators exhibited neutrophil- and eosinophil-chemotactic activity and induced polarization of dendritic cells (DCs) toward a Th2-inducing phenotype. In our study we performed chemical analyses of the pollen associated lipids, that are rapidly released upon hydration. As main components we have identified different types of phytoprostanes (PhytoPs), and for the first time phytofurans (PhytoFs), with predominating 16-F1t-PhytoPs (PPF1-I), 9-F1t-PhytoPs (PPF1-II), 16-E1t-PhytoPs (PPE1-I) and 9-D1t-PhytoPs (PPE1-II), and 16(RS)-9-epi-ST-Δ14-10-PhytoFs. Interestingly 16-E1t-PhytoP and 9-D1t-PhytoPs were found to be bound to glycerol. Lipid-containing samples (aqueous pollen extract, APE) induced murine mast cell chemotaxis and IL-6 release, and enhanced their IgE-dependent degranulation, demonstrating a role for these lipids in the immediate effector phase of allergic inflammation. Noteworthy, mast cell degranulation seems to be dependent on glycerol-bound, but not free phytoprostanes. On murine dendritic cells, APE selectively induced the upregulation of CD1d, likely preparing lipid-antigen presentation to iNKT cells. Our report contributes to the understanding of the activity of lipid mediators in the immediate effector phase of allergic reactions but identifies a yet undescribed pathway for the recognition of pollen-derived glycolipids by iNKT cells.


Subject(s)
Allergens/immunology , Dendritic Cells/immunology , Glycolipids/immunology , Hypersensitivity/immunology , Lipids/immunology , Phleum/immunology , Allergens/analysis , Allergens/isolation & purification , Animals , Antigens, CD1d/genetics , Antigens, CD1d/metabolism , Cell Degranulation/immunology , Chemotaxis, Leukocyte/immunology , Dendritic Cells/metabolism , Fatty Acids, Unsaturated/immunology , Fatty Acids, Unsaturated/isolation & purification , Furans/immunology , Furans/isolation & purification , Glycolipids/metabolism , Humans , Lipids/analysis , Lipids/isolation & purification , Mast Cells/immunology , Mast Cells/physiology , Mice, Inbred C57BL , Mice, Knockout , Phleum/chemistry , Pollen/chemistry , Pollen/immunology
17.
Mol Cell Biol ; 25(21): 9324-39, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16227584

ABSTRACT

Axl receptor tyrosine kinase exists as a transmembrane protein and as a soluble molecule. We show that constitutive and phorbol 12-myristate 13-acetate-induced generation of soluble Axl (sAxl) involves the activity of disintegrin-like metalloproteinase 10 (ADAM10). Spontaneous and inducible Axl cleavage was inhibited by the broad-spectrum metalloproteinase inhibitor GM6001 and by hydroxamate GW280264X, which is capable of blocking ADAM10 and ADAM17. Furthermore, murine fibroblasts deficient in ADAM10 expression exhibited a significant reduction in constitutive and inducible Axl shedding, whereas reconstitution of ADAM10 restored sAxl production, suggesting that ADAM10-mediated proteolysis constitutes a major mechanism for sAxl generation in mice. Partially overlapping 14-amino-acid stretch deletions in the membrane-proximal region of Axl dramatically affected sAxl generation, indicating that these regions are involved in regulating the access of the protease to the cleavage site. Importantly, relatively high circulating levels of sAxl are present in mouse sera in a heterocomplex with Axl ligand Gas6. Conversely, two other family members, Tyro3 and Mer, were not detected in mouse sera and conditioned medium. sAxl is constitutively released by murine primary cells such as dendritic and transformed cell lines. Upon immobilization, sAxl promoted cell migration and induced the phosphorylation of Axl and phosphatidylinositol 3-kinase. Thus, ADAM10-mediated generation of sAxl might play an important role in diverse biological processes.


Subject(s)
ADAM Proteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , ADAM Proteins/antagonists & inhibitors , ADAM Proteins/genetics , ADAM10 Protein , Amyloid Precursor Protein Secretases , Animals , Cell Line , Cell Movement , Dendritic Cells/enzymology , Dipeptides/pharmacology , Female , Fibroblasts/metabolism , Hydroxamic Acids/pharmacology , Intercellular Signaling Peptides and Proteins/blood , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Inbred Strains , Oncogene Proteins/blood , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins , Receptor Protein-Tyrosine Kinases/blood , Axl Receptor Tyrosine Kinase
20.
Sci Rep ; 8(1): 8636, 2018 06 05.
Article in English | MEDLINE | ID: mdl-29872093

ABSTRACT

IL-9 is a cytokine with pleiotropic function that mediates allergic inflammation and immunity to intestinal helminth parasites. Accumulating evidence suggests that IL-9 acts via both, initiation and regulation of adaptive immune responses and direct activation of intestinal effector pathways. Here we use IL-9 receptor deficient mice on BALB/c and C57BL/6 genetic background to dissect effector and regulatory functions of IL-9 during infection with the parasitic nematode Strongyloides ratti. IL-9 receptor-deficient mice displayed increased intestinal parasite burden and prolonged infection irrespective of the genetic background of the mice. Increased parasite burden was correlated to a reciprocally reduced early degranulation of mucosal mast cells, reduced intestinal IL-13 expression and caused by IL-9 receptor deficiency on hematopoietic cells. We observed additional significant changes in the adaptive immune response to S. ratti infection in the absence of the IL-9 receptor that depended on the mouse strain. However, the generation of protective memory to a second infection was intact in IL-9 receptor-deficient mice, irrespective of the genetic background. In summary, our results support a central role for IL-9 as an early mast cell activating effector cytokine during intestinal helminth infection while non-redundant functions in the initiation and amplification of adaptive immune responses were not apparent.


Subject(s)
Interleukin-9/metabolism , Mast Cells/immunology , Strongyloides ratti/immunology , Strongyloidiasis/immunology , Animals , Disease Models, Animal , Immunologic Factors/metabolism , Immunologic Memory , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Receptors, Interleukin-9/deficiency
SELECTION OF CITATIONS
SEARCH DETAIL