Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
Add more filters








Publication year range
1.
J Allergy Clin Immunol ; 142(2): 381-393, 2018 08.
Article in English | MEDLINE | ID: mdl-29454835

ABSTRACT

FcεRI is the primary receptor in mast cells that mediates allergic reactions by inducing rapid release of mediators, an adaptive immune response that might have evolved as a host defense against parasites and venoms. Yet it is apparent that mast cells are also activated through non-IgE receptors, the significance of which is just beginning to be understood. This includes the Mas-related G protein-coupled receptor X2, which might contribute to reactions to diverse antimicrobials and polybasic compounds, and the adhesion G protein-coupled receptor E2, variants of which are associated with familial vibratory urticaria and are activated by mechanical vibration. Similarly, mast cells have long been recognized as the main repository for histamine, heparin, and proteases. Recent evidence also points to new functions, modes of delivery, and mechanisms of action of mast cell proteases that add new dimensions to the roles of mast cells in human biology. In addition, exposure of mast cells to environmental cues can quantitatively and qualitatively modulate their responses and thus their effect on allergic inflammation. Illustrating this paradigm, we summarize a number of recent studies implicating the injury/tissue damage cytokine IL-33 as a modulator of allergen-induced mast cell responses. We also discuss the discovery of markers associated with transformed mast cells and new potential directions in suppressing mast cell activity.


Subject(s)
Hypersensitivity/immunology , Inflammation/immunology , Interleukin-33/metabolism , Mast Cells/immunology , Mastocytosis/immunology , Receptors, IgE/metabolism , Urticaria/immunology , Animals , Carboxypeptidases A/metabolism , Cell Degranulation , Histamine/metabolism , Humans , Nerve Tissue Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, Neuropeptide/metabolism , Signal Transduction , Vibration
2.
J Allergy Clin Immunol ; 142(1): 195-206.e8, 2018 07.
Article in English | MEDLINE | ID: mdl-29031599

ABSTRACT

BACKGROUND: DJ-1 is a redox-sensitive protein with multiple roles in cell homeostasis, levels of which are altered in patients with mast cell (MC)-related disorders. However, whether DJ-1 can regulate human MC function is unknown. OBJECTIVE: We sought to investigate the potential role of DJ-1 in the responses of human MCs to antigen stimulation. METHODS: DJ-1 was silenced in human CD34+-derived MCs and in the LAD2 MC line by using lentiviral short hairpin RNA constructs. Release of ß-hexosaminidase, prostaglandin D2, and GM-CSF and changes in reactive oxygen species levels were measured after FcεRI engagement. Enzymatic assays, sucrose density gradient centrifugation, immunoprecipitation, dot and Western blotting, and confocal imaging were performed for signaling, cellular localization, and coassociation studies. RESULTS: DJ-1 knockdown substantially reduced mediator release, as well as Lyn kinase and spleen tyrosine kinase activation and signaling through mechanisms that appeared largely unrelated to DJ-1 antioxidant activity. Following FcεRI activation, nonoxidized rather than oxidized DJ-1 translocated to lipid rafts, where it associated with Lyn, an interaction that appeared critical for maximal Lyn activation and initiation of signaling. Using purified recombinant proteins, we demonstrated that DJ-1 directly bound to Lyn but not to other Src kinases, and this interaction was specific for human but not mouse proteins. In addition, DJ-1 reduced Src homology 2 domain-containing phosphatase 2 phosphatase activity by scavenging reactive oxygen species, thus preventing spleen tyrosine kinase dephosphorylation and perpetuating MC signaling. CONCLUSION: We demonstrate a novel role for DJ-1 in the early activation of Lyn by FcεRI, which is essential for human MC responses and provides the basis for an alternative target in allergic disease therapy.


Subject(s)
Immunoglobulin E/immunology , Mast Cells/immunology , Protein Deglycase DJ-1/immunology , src-Family Kinases/immunology , Cell Degranulation/immunology , Cells, Cultured , Humans , Receptors, IgE
3.
Nat Commun ; 8(1): 1519, 2017 11 15.
Article in English | MEDLINE | ID: mdl-29142196

ABSTRACT

Receptor activator of NF-kB ligand (RANKL) generates intracellular reactive oxygen species (ROS), which increase RANKL-mediated signaling in osteoclast (OC) precursor bone marrow macrophages (BMMs). Here we show that a ROS scavenging protein DJ-1 negatively regulates RANKL-driven OC differentiation, also called osteoclastogenesis. DJ-1 ablation in mice leads to a decreased bone volume and an increase in OC numbers. In vitro, the activation of RANK-dependent signals is enhanced in DJ-1-deficient BMMs as compared to wild-type BMMs. DJ-1 suppresses the activation of both RANK-TRAF6 and RANK-FcRγ/Syk signaling pathways because of activation of Src homology region 2 domain-containing phosphatase-1, which is inhibited by ROS. Ablation of DJ-1 in mouse models of arthritis and RANKL-induced bone disease leads to an increase in the number of OCs, and exacerbation of bone damage. Overall, our results suggest that DJ-1 plays a role in bone homeostasis in normal physiology and in bone-associated pathology by negatively regulating osteoclastogenesis.


Subject(s)
Bone and Bones/metabolism , Cell Differentiation , Homeostasis , Osteoclasts/metabolism , Protein Deglycase DJ-1/metabolism , Animals , Female , Macrophages/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Osteogenesis , Protein Deglycase DJ-1/genetics , RANK Ligand/metabolism , Signal Transduction , TNF Receptor-Associated Factor 6/metabolism
4.
Proc Natl Acad Sci U S A ; 113(49): 14115-14120, 2016 12 06.
Article in English | MEDLINE | ID: mdl-27872312

ABSTRACT

Allergic diseases are driven by activation of mast cells and release of mediators in response to IgE-directed antigens. However, there are no drugs currently available that can specifically down-regulate mast cell function in vivo when chronically administered. Here, we describe an innovative approach for targeting mast cells in vitro and in vivo using antisense oligonucleotide-mediated exon skipping of the ß-subunit of the high-affinity IgE receptor (FcεRIß) to eliminate surface high-affinity IgE receptor (FcεRI) expression and function, rendering mast cells unresponsive to IgE-mediated activation. As FcεRIß expression is restricted to mast cells and basophils, this approach would selectively target these cell types. Given the success of exon skipping in clinical trials to treat genetic diseases such as Duchenne muscular dystrophy, we propose that exon skipping of FcεRIß is a potential approach for mast cell-specific treatment of allergic diseases.


Subject(s)
Cell Degranulation/drug effects , Dermatitis, Allergic Contact/therapy , Mast Cells/drug effects , Oligonucleotides, Antisense/therapeutic use , RNA Splicing/drug effects , Receptors, IgE/metabolism , Animals , Calcium/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Cytokines/biosynthesis , Disease Models, Animal , Female , Humans , Mast Cells/metabolism , Mice, Inbred BALB C , Mice, Inbred C57BL , Oligonucleotides, Antisense/pharmacology , Passive Cutaneous Anaphylaxis/genetics , Receptors, IgE/genetics
5.
PLoS One ; 11(9): e0162831, 2016.
Article in English | MEDLINE | ID: mdl-27611333

ABSTRACT

Neoplastic accumulation of mast cells in systemic mastocytosis (SM) associates with activating mutations in the receptor tyrosine kinase KIT. Constitutive activation of tyrosine kinase oncogenes has been linked to imbalances in oxidant/antioxidant mechanisms in other myeloproliferative disorders. However, the impact of KIT mutations on the redox status in SM and the potential therapeutic implications are not well understood. Here, we examined the regulation of reactive oxygen species (ROS) and of the antioxidant protein DJ-1 (PARK-7), which increases with cancer progression and acts to lessen oxidative damage to malignant cells, in relationship with SM severity. ROS levels were increased in both indolent (ISM) and aggressive variants of the disease (ASM). However, while DJ-1 levels were reduced in ISM with lower mast cell burden, they rose in ISM with higher mast cell burden and were significantly elevated in patients with ASM. Studies on mast cell lines revealed that activating KIT mutations induced constant ROS production and consequent DJ-1 oxidation and degradation that could explain the reduced levels of DJ-1 in the ISM population, while IL-6, a cytokine that increases with disease severity, caused a counteracting transcriptional induction of DJ-1 which would protect malignant mast cells from oxidative damage. A mouse model of mastocytosis recapitulated the biphasic changes in DJ-1 and the escalating IL-6, ROS and DJ-1 levels as mast cells accumulate, findings which were reversed with anti-IL-6 receptor blocking antibody. Our findings provide evidence of increased ROS and a biphasic regulation of the antioxidant DJ-1 in variants of SM and implicate IL-6 in DJ-1 induction and expansion of mast cells with KIT mutations. We propose consideration of IL-6 blockade as a potential adjunctive therapy in the treatment of patients with advanced mastocytosis, as it would reduce DJ-1 levels making mutation-positive mast cells vulnerable to oxidative damage.


Subject(s)
Antioxidants/metabolism , Mastocytosis/metabolism , Protein Deglycase DJ-1/metabolism , Reactive Oxygen Species/metabolism , Adoptive Transfer , Adult , Animals , Cell Line , Extracellular Space/metabolism , Homeostasis , Humans , Mast Cells/metabolism , Mastocytoma/pathology , Mastocytosis/blood , Mice , Middle Aged , Mutation/genetics , Protein Deglycase DJ-1/blood , Protein Deglycase DJ-1/genetics , Proteolysis , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/metabolism , Reactive Oxygen Species/blood , Receptors, Interleukin-6/metabolism , Transcription, Genetic
6.
J Allergy Clin Immunol ; 137(6): 1863-1871.e6, 2016 06.
Article in English | MEDLINE | ID: mdl-26774658

ABSTRACT

BACKGROUND: IL-6, levels of which are reported to be increased in association with mastocytosis, asthma, and urticaria, is used in conjunction with stem cell factor to generate CD34(+) cell-derived primary human mast cell (HuMC) cultures. Despite these associations, the effects on and mechanisms by which prolonged exposure to IL-6 alters HuMC numbers and function are not well understood. OBJECTIVES: We sought to study the effect of IL-6 on HuMC function, the mechanisms by which IL-6 exerts its effects, and the relationship of these findings to mastocytosis. METHODS: HuMCs were cultured in stem cell factor with or without IL-6. Responses to FcεRI aggregation and expression of proteases and receptors, including the soluble IL-6 receptor (sIL-6R), were then quantitated. Epigenetic changes in suppressor of cytokine signaling 3 (SOCS3) were determined by using methylation-specific PCR. Serum samples from healthy control subjects and patients with mastocytosis were assayed for IL-6, tryptase, and sIL-6R. RESULTS: IL-6 enhanced mast cell (MC) proliferation, maturation, and reactivity after FcεRI aggregation. IL-6 reduced expression of SOCS3, which correlated with methylation of the SOCS3 promoter and increased expression and activation of signal transducer and activator of transcription 3. IL-6 also suppressed constitutive production of sIL-6R, and serum levels of sIL-6R were similarly reduced in patients with mastocytosis. CONCLUSION: IL-6 increases MC proliferation and formation of a more reactive phenotype enabled by suppressing proteolytic cleavage of sIL-6R from IL-6R and downregulation of the SOCS3 autoinhibitory pathway. We suggest IL-6 blockade might ameliorate MC-related symptoms and pathology in patients with MC-related diseases associated with increased IL-6 levels, including mastocytosis.


Subject(s)
Interleukin-6/metabolism , Mast Cells/immunology , Mast Cells/metabolism , Suppressor of Cytokine Signaling 3 Protein/metabolism , Cell Count , Cell Proliferation/drug effects , Chymases/metabolism , DNA Methylation , Humans , Interleukin-6/pharmacology , Mast Cells/drug effects , Phosphorylation , Promoter Regions, Genetic , Protein Binding , Proto-Oncogene Proteins c-kit/metabolism , Receptors, IgE/metabolism , Receptors, Interleukin-6/metabolism , Signal Transduction/drug effects
7.
Sci Rep ; 6: 19685, 2016 01 20.
Article in English | MEDLINE | ID: mdl-26785945

ABSTRACT

Food allergy is a hypersensitive immune reaction to food proteins. We have previously demonstrated the presence of IL-10-producing CD5(+) B cells and suggested their potential role in regulating cow's milk casein allergy in humans and IgE-mediated anaphylaxis in mice. In this study, we determined whether IL-10-producing CD5(+) regulatory B cells control casein-induced food allergic responses in mice and, if so, the underlying mechanisms. The induction of oral tolerance (OT) by casein suppressed casein-induced allergic responses including the decrease of body temperature, symptom score, diarrhea, recruitment of mast cells and eosinophils into jejunum, and other biological parameters in mice. Notably, the population of IL-10-producing CD5(+) B cells was increased in mesenteric lymph node (MLN), but not in spleen or peritoneal cavity (PeC) in OT mice. The adoptive transfer of CD5(+) B cells from MLN, but not those from spleen and PeC, suppressed the casein-induced allergic responses in an allergen-specific and IL-10-dependent manner. The inhibitory effect of IL-10-producing CD5(+) B cells on casein-induced allergic response was dependent on Foxp3(+) regulatory T cells. Taken together, mesenteric IL-10-producing regulatory B cells control food allergy via Foxp3(+) regulatory T cells and could potentially act as a therapeutic regulator for food allergy.


Subject(s)
Allergens/immunology , B-Lymphocytes, Regulatory/immunology , B-Lymphocytes, Regulatory/metabolism , Caseins/immunology , Interleukin-10/biosynthesis , Milk Hypersensitivity/immunology , Milk Hypersensitivity/metabolism , Milk/immunology , Adoptive Transfer , Animals , B-Lymphocyte Subsets , CD5 Antigens , Cattle , Cell Communication , Disease Models, Animal , Female , Immune Tolerance , Immunoglobulin E/immunology , Immunomodulation , Lymph Nodes/immunology , Lymph Nodes/metabolism , Mesentery , Mice , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
8.
Eur J Immunol ; 45(11): 3034-44, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26256265

ABSTRACT

IL-33 released from damaged cells plays a central role in allergic inflammation by acting through its membrane-bound receptor, ST2 receptor (ST2L). IL-33 activity can be neutralized by the soluble spliced variant of ST2 (sST2) that has been associated with allergic inflammation but its source is not well defined. We investigated whether mast cells (MCs) are a significant source of sST2 following activation through FcεRI or ST2. We find that antigen and IL-33 induce substantial production and release of sST2 from human and mouse MCs in culture and do so synergistically when added together or in combination with stem cell factor. Moreover, increases in circulating sST2 during anaphylaxis in mice were dependent on the presence of MCs. Human MCs activated via FcεRI failed to generate IL-33 and IL-33 produced by mouse bone marrow-derived MCs was retained within the cells. Therefore, FcεRI-mediated sST2 production is independent of MC-derived IL-33 acting in an autocrine manner. These results are consistent with the conclusion that both mouse and human MCs when activated are a significant inducible source of sST2 but not IL-33 and thus have the ability to modulate the biologic impact of IL-33 produced locally by other cell types during allergic inflammation.


Subject(s)
Interleukin-33/immunology , Mast Cells/immunology , Receptors, Cell Surface/immunology , Receptors, Cell Surface/metabolism , Animals , Cells, Cultured , Flow Cytometry , Humans , Immunoblotting , Inflammation/immunology , Interleukin-1 Receptor-Like 1 Protein , Mice , Mice, Inbred C57BL , Real-Time Polymerase Chain Reaction , Receptors, Interleukin/immunology , Receptors, Interleukin/metabolism , Signal Transduction/immunology
9.
Sci Signal ; 8(368): ra28, 2015 Mar 17.
Article in English | MEDLINE | ID: mdl-25783157

ABSTRACT

Subsets of B cells inhibit various immune responses through their production of the cytokine interleukin-10 (IL-10). We found that IL-10-producing CD5(+) B cells suppressed the immunoglobulin E (IgE)- and antigen-mediated activation of mast cells in vitro as well as allergic responses in mice in an IL-10-dependent manner. Furthermore, the suppressive effect of these B cells on mast cells in vitro and in vivo depended on direct cell-to-cell contact through the costimulatory receptor CD40 on CD5(+) B cells and the CD40 ligand on mast cells. This contact enhanced the production of IL-10 by the CD5(+) B cells. Through activation of the Janus-activated kinase-signal transducer and activator of transcription 3 pathway, IL-10 decreased the abundance of the kinases Fyn and Fgr and inhibited the activation of the downstream kinase Syk in mast cells. Together, these findings suggest that an important function of IL-10-producing CD5(+) B cells is inhibiting mast cells and IgE-mediated allergic responses.


Subject(s)
B-Lymphocytes/immunology , CD5 Antigens/immunology , Hypersensitivity/immunology , Immunoglobulin E/immunology , Interleukin-10/immunology , Mast Cells/immunology , Animals , B-Lymphocytes/pathology , CD40 Antigens/genetics , CD40 Antigens/immunology , CD5 Antigens/genetics , Hypersensitivity/genetics , Hypersensitivity/pathology , Immunoglobulin E/genetics , Interleukin-10/genetics , Mast Cells/pathology , Mice , Mice, Knockout , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/immunology , Proto-Oncogene Proteins c-fyn/genetics , Proto-Oncogene Proteins c-fyn/immunology , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/immunology , Signal Transduction/genetics , Signal Transduction/immunology , src-Family Kinases/genetics , src-Family Kinases/immunology
10.
Mol Biol Cell ; 26(9): 1711-27, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25717186

ABSTRACT

MS4A family members differentially regulate the cell cycle, and aberrant, or loss of, expression of MS4A family proteins has been observed in colon and lung cancer. However, the precise functions of MS4A family proteins and their mechanistic interactions remain unsolved. Here we report that MS4A4 facilitates trafficking of the receptor tyrosine kinase KIT through endocytic recycling rather than degradation pathways by a mechanism that involves recruitment of KIT to caveolin-1-enriched microdomains. Silencing of MS4A4 in human mast cells altered ligand-induced KIT endocytosis pathways and reduced receptor recycling to the cell surface, thus promoting KIT signaling in the endosomes while reducing that in the plasma membrane, as exemplified by Akt and PLCγ1 phosphorylation, respectively. The altered endocytic trafficking of KIT also resulted in an increase in SCF-induced mast cell proliferation and migration, which may reflect altered signaling in these cells. Our data reveal a novel function for MS4A family proteins in regulating trafficking and signaling, which could have implications in both proliferative and immunological diseases.


Subject(s)
Clathrin/metabolism , Mast Cells/metabolism , Membrane Proteins/physiology , Proto-Oncogene Proteins c-kit/metabolism , Antigens, CD20/physiology , Cell Line , Cell Membrane/metabolism , Endocytosis , Endosomes/metabolism , Gene Expression , Humans , Phospholipase C gamma/metabolism , Phosphorylation , Protein Processing, Post-Translational , Protein Transport , Signal Transduction , rab5 GTP-Binding Proteins/metabolism
11.
J Immunol ; 193(12): 5924-32, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25378594

ABSTRACT

Rictor is a regulatory component of the mammalian target of rapamycin (mTOR) complex 2 (mTORC2). We have previously demonstrated that rictor expression is substantially downregulated in terminally differentiated mast cells as compared with their immature or transformed counterparts. However, it is not known whether rictor and mTORC2 regulate mast cell activation. In this article, we show that mast cell degranulation induced by aggregation of high-affinity receptors for IgE (FcεRI) is negatively regulated by rictor independently of mTOR. We found that inhibition of mTORC2 by the dual mTORC1/mTORC2 inhibitor Torin1 or by downregulation of mTOR by short hairpin RNA had no impact on FcεRI-induced degranulation, whereas downregulation of rictor itself resulted in an increased sensitivity (∼50-fold) of cells to FcεRI aggregation with enhancement of degranulation. This was linked to a similar enhancement in calcium mobilization and cytoskeletal rearrangement attributable to increased phosphorylation of LAT and PLCγ1. In contrast, degranulation and calcium responses elicited by the G protein-coupled receptor ligand, C3a, or by thapsigargin, which induces a receptor-independent calcium signal, was unaffected by rictor knockdown. Overexpression of rictor, in contrast with knockdown, suppressed FcεRI-mediated degranulation. Taken together, these data provide evidence that rictor is a multifunctional signaling regulator that can regulate FcεRI-mediated degranulation independently of mTORC2.


Subject(s)
Carrier Proteins/metabolism , Cell Degranulation/immunology , Mast Cells/immunology , Mast Cells/metabolism , Receptors, IgE/metabolism , Actins/metabolism , Calcium Signaling , Carrier Proteins/genetics , Cell Degranulation/genetics , Cell Line , Enzyme Activation , Gene Knockdown Techniques , Humans , Protein Aggregates , Protein Transport , RNA Interference , Rapamycin-Insensitive Companion of mTOR Protein , Signal Transduction , TOR Serine-Threonine Kinases/metabolism
12.
Sci Signal ; 6(292): ra80, 2013 Sep 10.
Article in English | MEDLINE | ID: mdl-24023254

ABSTRACT

The protein prohibitin (PHB) is implicated in diverse cellular processes, including cell signaling, transcriptional control, and mitochondrial function. We found that PHB was abundant in the intracellular granules of mast cells, which are critical for allergic responses to antigens. Thus, we investigated whether PHB played a role in signaling mediated by the high-affinity receptor for antigen-bound immunoglobulin E (IgE), FcεRI. PHB-specific small interfering RNAs (siRNAs) inhibited antigen-mediated signaling, degranulation, and cytokine secretion by mast cells in vitro. Knockdown of PHB inhibited the antigen-dependent association of the tyrosine kinase Syk with FcεRI and inhibited the activation of Syk. Fractionation studies revealed that PHB translocated from intracellular granules to plasma membrane lipid rafts in response to antigen, and knockdown of PHB suppressed the movement of FcεRIγ and Syk into lipid rafts. Tyrosine phosphorylation of PHB by Lyn was observed early after exposure to antigen, and point mutations in PHB indicated that Tyr(114) and Tyr(259) were required for the recruitment of Syk to FcεRIγ and mast cell activation. In mice, PHB-specific siRNAs inhibited antigen-initiated mast cell degranulation, passive cutaneous anaphylaxis, and passive systemic anaphylaxis. Together, these results suggest that PHB is essential for FcεRI-mediated mast cell activation and allergic responses in vivo, raising the possibility that PHB might serve as a therapeutic target for the treatment of allergic diseases.


Subject(s)
Antigens/immunology , Mast Cells/immunology , Repressor Proteins/immunology , Signal Transduction/immunology , Animals , Blotting, Western , Cell Degranulation/immunology , Cells, Cultured , Cytokines/immunology , Cytokines/metabolism , Immunoglobulin E/immunology , Immunoglobulin E/metabolism , Intracellular Signaling Peptides and Proteins/immunology , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mast Cells/metabolism , Mast Cells/physiology , Mice , Mice, Inbred BALB C , Microscopy, Confocal , Models, Immunological , Mutation , Passive Cutaneous Anaphylaxis/genetics , Passive Cutaneous Anaphylaxis/immunology , Phosphorylation/immunology , Prohibitins , Protein Binding/immunology , Protein-Tyrosine Kinases/immunology , Protein-Tyrosine Kinases/metabolism , RNA Interference , Receptors, IgE/immunology , Receptors, IgE/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Signal Transduction/genetics , Syk Kinase , Tyrosine/immunology , Tyrosine/metabolism
13.
Immunity ; 38(5): 906-17, 2013 May 23.
Article in English | MEDLINE | ID: mdl-23643722

ABSTRACT

Human linkage analyses have implicated the MS4A2-containing gene locus (encoding FcεRIß) as a candidate for allergy susceptibility. We have identified a truncation of FcεRIß (t-FcεRIß) in humans that contains a putative calmodulin-binding domain and thus, we sought to identify the role of this variant in mast cell function. We determined that t-FcεRIß is critical for microtubule formation and degranulation and that it may perform this function by trafficking adaptor molecules and kinases to the pericentrosomal and Golgi region in response to Ca2+ signals. Mutagenesis studies suggest that calmodulin binding to t-FcεRIß in the presence of Ca2+ could be critical for t-FcεRIß function. In addition, gene targeting of t-FcεRIß attenuated microtubule formation, degranulation, and IL-8 production downstream of Ca2+ signals. Therefore, t-FcεRIß mediates Ca2+ -dependent microtubule formation, which promotes degranulation and cytokine release. Because t-FcεRIß has this critical function, it represents a therapeutic target for the downregulation of allergic inflammation.


Subject(s)
Calcium Signaling/immunology , Cell Degranulation/immunology , Mast Cells/immunology , Microtubules/metabolism , Receptors, IgE/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Calcium/metabolism , Calmodulin-Binding Proteins/genetics , Calmodulin-Binding Proteins/immunology , Golgi Apparatus/metabolism , Humans , Hypersensitivity/immunology , Immunoglobulin E/immunology , Interleukin-8/metabolism , Mast Cells/metabolism , Prostaglandin D2/biosynthesis , Prostaglandin D2/immunology , Protein Isoforms/immunology , RNA Interference , RNA Splicing , RNA, Messenger , RNA, Small Interfering , Receptors, IgE/genetics
14.
Eur J Immunol ; 43(7): 1873-82, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23616175

ABSTRACT

Following antigen/IgE-mediated aggregation of high affinity IgE-receptors (FcεRI), mast cells (MCs) degranulate and release inflammatory mediators leading to the induction of allergic reactions including anaphylaxis. Migration of MCs to resident tissues and sites of inflammation is regulated by tissue chemotactic factors such as stem cell factor (SCF (KIT ligand)). Despite inducing similar early signaling events to antigen, chemotactic factors, including SCF, produce minimal degranulation in the absence of other stimuli. We therefore investigated whether processes regulating MC chemotaxis are rate limiting for MC mediator release. To investigate this issue, we disrupted actin polymerization, a requirement for MC chemotaxis, with latrunculin B and cytochalasin B, then examined chemotaxis and mediator release in human (hu)MCs induced by antigen or SCF. As expected, such disruption minimally affected early signaling pathways, but attenuated SCF-induced human mast cell chemotaxis. In contrast, SCF, in the absence of other stimuli, induced substantial degranulation in a concentration-dependent manner following actin disassembly. It also moderately enhanced antigen-mediated human mast cell degranulation which was further enhanced in the presence of SCF. These observations suggest that processes regulating cell migration limit MC degranulation as a consequence of cytoskeletal reorganization.


Subject(s)
Actins/metabolism , Cell Degranulation/immunology , Chemotaxis, Leukocyte/immunology , Mast Cells/metabolism , Stem Cell Factor/immunology , Actins/immunology , Cell Degranulation/drug effects , Cells, Cultured , Chemotaxis, Leukocyte/drug effects , Flow Cytometry , Humans , Immunoblotting , Mast Cells/immunology , Microscopy, Confocal , Stem Cell Factor/pharmacology
15.
Cell Calcium ; 53(5-6): 315-26, 2013.
Article in English | MEDLINE | ID: mdl-23489970

ABSTRACT

Antigen-mediated mast cell (MC) degranulation is the critical early event in the induction of allergic reactions. Transient receptor potential channels (TRPC), particularly TRPC1, are thought to contribute to such MC activation. To explore the contribution of TRPC1 in MC-driven allergic reactions, we examined antigen-mediated anaphylaxis in Trpc1⁻/⁻ and WT mice, and TRPC1 involvement in the activation of MCs derived from the bone marrow (BMMCs) of these mice. In vivo, we observed a similar induction of passive systemic anaphylaxis in the Trpc1⁻/⁻ mice compared to WT controls. Nevertheless, there was delayed recovery from this response in Trpc1⁻/⁻ mice. Furthermore, contrary to expectations, Trpc1⁻/⁻ BMMCs responded to antigen with enhanced calcium signaling but with little defect in degranulation or associated signaling. In contrast, antigen-mediated production of TNF-α, and other cytokines, was enhanced in the Trpc1⁻/⁻ BMMCs, as were calcium-dependent events required for these responses. Additionally, circulating levels of TNF-α in response to antigen were preferentially elevated in the Trpc1⁻/⁻ mice, and administration of an anti-TNF-α antibody blocked the delay in recovery from anaphylaxis in these mice. These data thus provide evidence that, in this model, TRPC1 promotes recovery from the anaphylactic response by repressing antigen-mediated TNF-α release from MCs.


Subject(s)
Anaphylaxis/immunology , Mast Cells/immunology , TRPC Cation Channels/metabolism , Tumor Necrosis Factor-alpha/metabolism , Allergens/immunology , Animals , Calcium Signaling/genetics , Cell Degranulation/genetics , Cells, Cultured , Humans , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , TRPC Cation Channels/genetics , TRPC6 Cation Channel
16.
J Immunol Methods ; 390(1-2): 52-62, 2013 Apr 30.
Article in English | MEDLINE | ID: mdl-23357051

ABSTRACT

Activation of KIT, by its ligand, stem cell factor (SCF), results in the initiation of signal transduction pathways that influence mast cell survival and proliferation. Activating mutations in KIT have thus been linked to clonal MC proliferation associated with systemic mastocytosis. SCF also modulates MC function by inducing MC chemotaxis and by potentiating antigen (Ag)/IgE-mediated MC degranulation. Thus, mutations in KIT also have the potential to affect these processes in allergic and other mast cell-related diseases. Studies to determine how native and mutated KIT may modulate MC chemotaxis and activation have, however, been limited due to the lack of availability of a suitable functional MC line lacking native KIT which would allow transduction of KIT constructs. Here we describe a novel mouse MC line which allows the study of normal and mutated KIT constructs. These cells originated from a bone marrow-derived mouse MC culture out of which a rapidly dividing mast cell sub-population spontaneously arose. Over time, these cells lost KIT expression while continuing to express functional high affinity receptors for IgE (FcεRI). As a consequence, these cells degranulated in response to Ag/IgE but did not migrate nor show any evidence of potentiation of Ag/IgE degranulation in response to SCF. Retroviral transduction of the cells with a human (hu)KIT construct resulted in surface expression of huKIT which responded to huSCF by potentiation of Ag/IgE-induced degranulation and chemotaxis. This cell line thus presents a novel system to delineate how MC function is modulated by native and mutated KIT and for the identification of novel inhibitors of these processes.


Subject(s)
Antigens/immunology , Cell Degranulation/immunology , Mast Cells/immunology , Proto-Oncogene Proteins c-kit/immunology , Animals , Benzamides/immunology , Benzamides/pharmacology , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Calcium/immunology , Calcium/metabolism , Cell Degranulation/drug effects , Cell Survival/drug effects , Cell Survival/immunology , Cells, Cultured , Chemotaxis/drug effects , Chemotaxis/immunology , Cytokines/immunology , Cytokines/metabolism , Flow Cytometry , Humans , Imatinib Mesylate , Immunoblotting , Mast Cells/metabolism , Mast Cells/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Confocal , Mutation/immunology , Phosphorylation/drug effects , Phosphorylation/immunology , Piperazines/immunology , Piperazines/pharmacology , Protein Kinase Inhibitors/immunology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/metabolism , Pyrimidines/immunology , Pyrimidines/pharmacology , Receptors, IgE/genetics , Receptors, IgE/immunology , Receptors, IgE/metabolism , Stem Cell Factor/immunology , Stem Cell Factor/pharmacology , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/immunology , TOR Serine-Threonine Kinases/metabolism
17.
J Immunol ; 190(2): 531-8, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23248261

ABSTRACT

IL-33 is elevated in afflicted tissues of patients with mast cell (MC)-dependent chronic allergic diseases. Based on its acute effects on mouse MCs, IL-33 is thought to play a role in the pathogenesis of allergic disease through MC activation. However, the manifestations of prolonged IL-33 exposure on human MC function, which best reflect the conditions associated with chronic allergic disease, are unknown. In this study, we found that long-term exposure of human and mouse MCs to IL-33 results in a substantial reduction of MC activation in response to Ag. This reduction required >72 h exposure to IL-33 for onset and 1-2 wk for reversion following IL-33 removal. This hyporesponsive phenotype was determined to be a consequence of MyD88-dependent attenuation of signaling processes necessary for MC activation, including Ag-mediated calcium mobilization and cytoskeletal reorganization, potentially as a consequence of downregulation of the expression of phospholipase Cγ(1) and Hck. These findings suggest that IL-33 may play a protective, rather than a causative, role in MC activation under chronic conditions and, furthermore, reveal regulated plasticity in the MC activation phenotype. The ability to downregulate MC activation in this manner may provide alternative approaches for treatment of MC-driven disease.


Subject(s)
Immunosuppression Therapy , Interleukins/immunology , Mast Cells/immunology , Mast Cells/metabolism , Phenotype , Actins/metabolism , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Humans , Interleukin-1 Receptor-Like 1 Protein , Interleukin-33 , Interleukins/pharmacology , Mast Cells/drug effects , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/immunology , Phospholipase C gamma/genetics , Phospholipase C gamma/immunology , Phospholipase C gamma/metabolism , Protein Multimerization/drug effects , Proto-Oncogene Proteins c-hck/genetics , Proto-Oncogene Proteins c-hck/immunology , Receptors, Interleukin/genetics , Receptors, Interleukin/immunology
18.
J Allergy Clin Immunol ; 131(6): 1653-62, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23182168

ABSTRACT

BACKGROUND: DJ-1 is an antioxidant protein known to reduce levels of reactive oxygen species (ROS), but its presence or function in mast cells and allergic diseases is unknown. OBJECTIVES: We sought to determine the role and mechanism of DJ-1 in allergic responses in vitro and in vivo. METHODS: ROS and DJ-1 levels in serum or culture medium were measured with ELISA kits. The role of DJ-1 was evaluated in mast cell cultures and passive cutaneous anaphylaxis in normal or DJ-1 knockout (KO) mice. The mechanism of DJ-1 action was examined by using immunoblotting, immunoprecipitation, RT-PCR, and other molecular biological approaches. RESULTS: Patients with atopic dermatitis had increased levels of ROS and diminished levels of DJ-1. DJ-1 KO mice exhibited enhanced passive cutaneous anaphylaxis and augmented ROS levels in sera and bone marrow-derived mast cells (BMMCs). Furthermore, antigen-induced degranulation and production of TNF-α and IL-4 were significantly amplified in DJ-1 KO and anti-DJ-1 small interfering RNA-transfected BMMCs compared with that seen in wild-type (WT) BMMCs. Studies with these cells and BMMCs transfected with small interfering RNAs against the phosphatases Src homology domain 2-containing protein tyrosine phosphatase (SHP) 1 and SHP-2 revealed that the DJ-1 KO phenotype could be attributed to suppression of SHP-1 activity and enhancement of SHP-2 activity, leading to strengthened signaling through linker for activation of T cells, phospholipase Cγ, and mitogen-activated protein kinases. CONCLUSIONS: A deficiency or constitutive activation of DJ-1 can have implications in mast cell-driven allergic diseases, such as asthma and anaphylaxis.


Subject(s)
Hypersensitivity, Immediate/immunology , Hypersensitivity, Immediate/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Mast Cells/immunology , Mast Cells/metabolism , Oncogene Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Adolescent , Adult , Animals , Antigens/immunology , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Cell Degranulation/immunology , Child , Child, Preschool , Cytokines/biosynthesis , Dermatitis, Atopic/immunology , Dermatitis, Atopic/metabolism , Disease Models, Animal , Female , Humans , Hypersensitivity, Immediate/genetics , Interleukin-4/metabolism , Intracellular Signaling Peptides and Proteins/blood , Intracellular Signaling Peptides and Proteins/genetics , Male , Membrane Proteins/metabolism , Mice , Mice, Knockout , Middle Aged , Oncogene Proteins/blood , Oncogene Proteins/genetics , Passive Cutaneous Anaphylaxis , Phosphoproteins/metabolism , Phosphorylation , Protein Deglycase DJ-1 , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins c-fyn/metabolism , RNA Interference , Reactive Oxygen Species/metabolism , Receptors, IgE/metabolism , Signal Transduction , Syk Kinase , Tumor Necrosis Factor-alpha/metabolism , Young Adult
19.
J Immunol ; 188(11): 5428-37, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22529299

ABSTRACT

Mast cells, activated by Ag via FcεRI, release an array of proinflammatory mediators that contribute to allergic disorders, such as asthma and anaphylaxis. The KIT ligand, stem cell factor (SCF), is critical for mast cell expansion, differentiation, and survival, and under acute conditions, it enhances mast cell activation. However, extended SCF exposure in vivo conversely protects against fatal Ag-mediated anaphylaxis. In investigating this dichotomy, we identified a novel mode of regulation of the mast cell activation phenotype through SCF-mediated programming. We found that mouse bone marrow-derived mast cells chronically exposed to SCF displayed a marked attenuation of FcεRI-mediated degranulation and cytokine production. The hyporesponsive phenotype was not a consequence of altered signals regulating calcium flux or protein kinase C, but of ineffective cytoskeletal reorganization with evidence implicating a downregulation of expression of the Src kinase Hck. Collectively, these findings demonstrate a major role for SCF in the homeostatic control of mast cell activation with potential relevance to mast cell-driven disease and the development of novel approaches for the treatment of allergic disorders.


Subject(s)
Mast Cells/immunology , Mast Cells/metabolism , Stem Cell Factor/physiology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Cell Degranulation/immunology , Cell Proliferation , Cells, Cultured , Coculture Techniques , Homeostasis/immunology , Hypersensitivity/immunology , Hypersensitivity/metabolism , Hypersensitivity/pathology , Immunophenotyping , Mast Cells/cytology , Mice , Mice, Inbred C57BL , NIH 3T3 Cells
20.
Cell ; 148(3): 421-33, 2012 Feb 03.
Article in English | MEDLINE | ID: mdl-22304913

ABSTRACT

Resveratrol, a polyphenol in red wine, has been reported as a calorie restriction mimetic with potential antiaging and antidiabetogenic properties. It is widely consumed as a nutritional supplement, but its mechanism of action remains a mystery. Here, we report that the metabolic effects of resveratrol result from competitive inhibition of cAMP-degrading phosphodiesterases, leading to elevated cAMP levels. The resulting activation of Epac1, a cAMP effector protein, increases intracellular Ca(2+) levels and activates the CamKKß-AMPK pathway via phospholipase C and the ryanodine receptor Ca(2+)-release channel. As a consequence, resveratrol increases NAD(+) and the activity of Sirt1. Inhibiting PDE4 with rolipram reproduces all of the metabolic benefits of resveratrol, including prevention of diet-induced obesity and an increase in mitochondrial function, physical stamina, and glucose tolerance in mice. Therefore, administration of PDE4 inhibitors may also protect against and ameliorate the symptoms of metabolic diseases associated with aging.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/antagonists & inhibitors , Aging/metabolism , Caloric Restriction , Signal Transduction , Stilbenes/administration & dosage , 3',5'-Cyclic-AMP Phosphodiesterases/chemistry , 3',5'-Cyclic-AMP Phosphodiesterases/metabolism , AMP-Activated Protein Kinase Kinases , Adipose Tissue, White/drug effects , Animals , Cyclic Nucleotide Phosphodiesterases, Type 4/chemistry , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Diet , Glucose Intolerance/prevention & control , Guanine Nucleotide Exchange Factors/metabolism , Mice , Models, Molecular , Muscle, Skeletal/drug effects , NAD/metabolism , Obesity/prevention & control , Protein Kinases/metabolism , Resveratrol , Rolipram/administration & dosage , Ryanodine Receptor Calcium Release Channel/metabolism , Sirtuin 1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL