Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 215
Filter
1.
Mar Drugs ; 20(2)2022 Feb 10.
Article in English | MEDLINE | ID: mdl-35200662

ABSTRACT

In this study, we investigated the anti-allergic effects of 3,4-dihydroxybenzaldehyde (DHB) isolated from the marine red alga, Polysiphonia morrowii, in mouse bone-marrow-derived cultured mast cells (BMCMCs) and passive cutaneous anaphylaxis (PCA) in anti-dinitrophenyl (DNP) immunoglobulin E (IgE)-sensitized mice. DHB inhibited IgE/bovine serum albumin (BSA)-induced BMCMCs degranulation by reducing the release of ß-hexosaminidase without inducing cytotoxicity. Further, DHB dose-dependently decreased the IgE binding and high-affinity IgE receptor (FcεRI) expression and FcεRI-IgE binding on the surface of BMCMCs. Moreover, DHB suppressed the secretion and/or the expression of the allergic cytokines, interleukin (IL)-4, IL-5, IL-6, IL-13, and tumor necrosis factor (TNF)-α, and the chemokine, thymus activation-regulated chemokine (TARC), by regulating the phosphorylation of IκBα and the translocation of cytoplasmic NF-κB into the nucleus. Furthermore, DHB attenuated the passive cutaneous anaphylactic (PCA) reaction reducing the exuded Evans blue amount in the mouse ear stimulated by IgE/BSA. These results suggest that DHB is a potential therapeutic candidate for the prevention and treatment of type I allergic disorders.


Subject(s)
Anti-Allergic Agents/pharmacology , Benzaldehydes/pharmacology , Catechols/pharmacology , Mast Cells/drug effects , Rhodophyta/metabolism , Animals , Anti-Allergic Agents/administration & dosage , Anti-Allergic Agents/isolation & purification , Benzaldehydes/administration & dosage , Benzaldehydes/isolation & purification , Catechols/administration & dosage , Catechols/isolation & purification , Cells, Cultured , Cytokines/immunology , Disease Models, Animal , Dose-Response Relationship, Drug , Immunoglobulin E/immunology , Male , Mast Cells/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Passive Cutaneous Anaphylaxis/drug effects , Passive Cutaneous Anaphylaxis/immunology , Serum Albumin, Bovine/immunology
2.
J Allergy Clin Immunol ; 149(1): 200-211, 2022 01.
Article in English | MEDLINE | ID: mdl-34126155

ABSTRACT

BACKGROUND: Blocking the major cat allergen, Fel d 1, with mAbs was effective in preventing an acute cat allergic response. OBJECTIVES: This study sought to extend the allergen-specific antibody approach and demonstrate that a combination of mAbs targeting Bet v 1, the immunodominant and most abundant allergenic protein in birch pollen, can prevent the birch allergic response. METHODS: Bet v 1-specific mAbs, REGN5713, REGN5714, and REGN5715, were isolated using the VelocImmune platform. Surface plasmon resonance, x-ray crystallography, and cryo-electron microscopy determined binding kinetics and structural data. Inhibition of IgE-binding, basophil activation, and mast cell degranulation were assessed via blocking ELISA, flow cytometry, and the passive cutaneous anaphylaxis mouse model. RESULTS: REGN5713, REGN5714, and REGN5715 bind with high affinity and noncompetitively to Bet v 1. A cocktail of all 3 antibodies, REGN5713/14/15, blocks IgE binding to Bet v 1 and inhibits Bet v 1- and birch pollen extract-induced basophil activation ex vivo and mast cell degranulation in vivo. Crystal structures of the complex of Bet v 1 with immunoglobulin antigen-binding fragments of REGN5713 or REGN5715 show distinct interaction sites on Bet v 1. Cryo-electron microscopy reveals a planar and roughly symmetrical complex formed by REGN5713/14/15 bound to Bet v 1. CONCLUSIONS: These data confirm the immunodominance of Bet v 1 in birch allergy and demonstrate blockade of the birch allergic response with REGN5713/14/15. Structural analyses show simultaneous binding of REGN5713, REGN5714, and REGN5715 with substantial areas of Bet v 1 exposed, suggesting that targeting specific epitopes is sufficient to block the allergic response.


Subject(s)
Allergens/immunology , Antibodies, Monoclonal/pharmacology , Antigens, Plant/immunology , Immunodominant Epitopes/immunology , Immunoglobulin G/pharmacology , Passive Cutaneous Anaphylaxis/immunology , Animals , Basophils/drug effects , Basophils/immunology , Humans , Immunoglobulin E/immunology , Mast Cells/drug effects , Mast Cells/immunology , Mice, Inbred BALB C , Rhinitis, Allergic, Seasonal/blood , Rhinitis, Allergic, Seasonal/immunology
3.
J Dermatol Sci ; 102(1): 47-57, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33676788

ABSTRACT

BACKGROUND: Mast cells play an important role in allergic responses and persistently exposure to environmental fine particulate matter (PM2.5) exacerbates allergic diseases,but the details remained elucidative. OBJECTIVES: To investigate the effect of PM2.5 on IgE-mediated mast cell responses through an IgE-mediated mouse model and mast cell activation. METHODS: The ß-hexosaminidase release and a BALB/c model of passive cutaneous anaphylaxis (PCA) was used to test IgE-mediated mast cells activation in vitro and in vivo. RNA-Seq technique was conducted to study the gene expression profile. Reactive oxygen species (ROS) production was measured by flow-cytometry. RT-PCR,WB and ELISA were performed to examine targeting molecules expression. RESULTS: PM2.5 facilitated IgE-mediated degranulation and increased cytokines expression in mast cells. Meanwhile, the Evan's blue extravasation as well as serum cytokines in mice was increased after treatment with PM2.5. Furthermore, PM2.5 treatment dramatically increased the expression of Gadd45b which is an oxidative stress molecule that directly activates down-stream pathway, such as MEKK4/JNK. PM2.5 treatment activated MEKK4, JNK1/2 but not ERK1/2 and p38. Meanwhile, Knockdown of Gadd45b significantly attenuated PM2.5-mediated JNK1/2 activation and expression of cytokines. In addition, a JNK1/2-specific inhibitor SP600125 blocked IgE-mediated mast cell activation and cytokine release in PCA model mice. Moreover, PM2.5 treatment increased the ROS level and ROS inhibitor dramatically blocked the PM2.5-induced ROS production and reversed the PM2.5-mediated gene expression in the mitochondrial respiratory chain. CONCLUSIONS: PM2.5 regulates ROS production through Gadd45b/MEKK4/JNK pathway, facilitating IgE-mediated mast cell activation.


Subject(s)
Cell Degranulation/immunology , Dermatitis, Allergic Contact/immunology , Mast Cells/immunology , Particulate Matter/adverse effects , Skin/pathology , Animals , Anthracenes/administration & dosage , Antigens, Differentiation/metabolism , Cell Degranulation/drug effects , Cell Line , Cell Line, Tumor , Dermatitis, Allergic Contact/pathology , Disease Models, Animal , Electron Transport/drug effects , Electron Transport/immunology , Humans , Immunoglobulin E/administration & dosage , Immunoglobulin E/immunology , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Mast Cells/cytology , Mast Cells/metabolism , Mice , Mitochondria/metabolism , Particulate Matter/immunology , Passive Cutaneous Anaphylaxis/drug effects , Passive Cutaneous Anaphylaxis/immunology , RNA-Seq , Rats , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Signal Transduction/immunology , Skin/cytology , Skin/immunology
4.
J Immunol ; 206(3): 471-480, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33361207

ABSTRACT

Mast cells are tissue-resident immune cells that play pivotal roles in initiating and amplifying allergic/anaphylactic reactions in humans. Their activation occurs via multiple mechanisms, which include cross-linking of the IgE-bound, high-affinity IgE receptors (FcεRI) by allergens or Ags and the binding of anaphylatoxins such as C3a to its receptor, C3aR. We have previously demonstrated that the Na+/H+ exchanger regulatory factor 1 (NHERF1) promotes C3aR functions in human mast cells. In the current study, we show that NHERF1 regulates mast cell response following FcεRI stimulation. Specifically, intracellular Ca2+ mobilization, activation of the MAPKs (ERK1/2 and P38), and production of cytokines (IL-13 and IL-6) following exposure to IgE/Ag were significantly reduced in mast cells from NHERF1+/‒ mice. In agreement with our in vitro data, mast cell-mediated passive cutaneous anaphylaxis and passive systemic anaphylaxis were reduced in NHERF1+/‒ mice and mast cell-deficient KitW-sh/W-sh mice engrafted with NHERF1+/‒ mast cells. Mechanistically, the levels of microRNAs (miRNAs) that regulate mast cell responses, miRNA 155-3p and miRNA 155-5p, were altered in mast cells from NHERF1+/‒ mice. Moreover, NHERF1 rapidly localized to the nucleus of mast cells following FcεRI stimulation. In summary, our results suggest that the NHERF1 acts as an adapter molecule and promotes IgE/Ag-induced mast cell activation. Further elucidating the mechanisms through which NHERF1 modulates mast cell responses will lend insights into the development of new therapeutic strategies to target mast cells during anaphylaxis or other allergic diseases.


Subject(s)
Hypersensitivity/immunology , Mast Cells/immunology , Passive Cutaneous Anaphylaxis/immunology , Phosphoproteins/metabolism , Receptors, IgE/metabolism , Sodium-Hydrogen Exchangers/metabolism , Animals , Calcium Signaling , Cell Degranulation , Cells, Cultured , Immunoglobulin E/metabolism , Immunomodulation , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics , Phosphoproteins/genetics , Sodium-Hydrogen Exchangers/genetics
5.
J Pharm Pharmacol ; 72(9): 1221-1231, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32557699

ABSTRACT

OBJECTIVES: To investigate the inhibitory effects of Kaempferol, a natural flavonol active compound, on pseudo-allergic reactions (in vivo and in vitro), particularly on the mechanism underlying its effect in human mast cells. METHODS: Compound 48/80 (C48/80)-induced immunoglobulin E (IgE)-independent passive cutaneous anaphylaxis (PCA) model and systemic anaphylaxis were applied to investigate the anti-allergic activity of Kaempferol. The degranulation assay, calcium imaging and the secretion of cytokines and chemokines were used to evaluate the inhibitory effect on mast cell activation. Western blot analysis was performed to investigate intracellular calcium fluctuation-related signalling pathways. KEY FINDINGS: Kaempferol dose-dependently attenuated C48/80-induced mice hind paw swelling, dye extravasation and skin mast cell degranulation, and rehabilitated the hypothermia, as well as reduced the serum concentrations of histamine, tryptase, tumour necrosis factor-alpha (TNF-α), interleukin-8 (IL-8) and monocyte chemo-attractant protein-1 (MCP-1). Furthermore, Kaempferol suppressed C48/80-triggered human MC degranulation and calcium fluctuations by inhibiting phospholipase Cγ (PLCγ) phosphorylation and subsequent cytokines synthesis pathways. CONCLUSIONS: The inhibition of the process of PLCγ phosphorylation to Ca2+ mobilization represents a major strategy in Kaempferol-suppressed pseudo-allergic reactions. Thus, Kaempferol could be considered as a therapeutic drug candidate for non-IgE-mediated allergic reactions or inflammations.


Subject(s)
Anaphylaxis/drug therapy , Anti-Allergic Agents/pharmacology , Calcium/metabolism , Kaempferols/pharmacology , Anaphylaxis/immunology , Animals , Anti-Allergic Agents/administration & dosage , Cells, Cultured , Chemokines/metabolism , Cytokines/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Humans , Immunoglobulin E/immunology , Kaempferols/administration & dosage , Male , Mast Cells , Mice , Mice, Inbred C57BL , Passive Cutaneous Anaphylaxis/drug effects , Passive Cutaneous Anaphylaxis/immunology , Secretagogues/immunology , p-Methoxy-N-methylphenethylamine/immunology
6.
Nutrients ; 12(5)2020 05 09.
Article in English | MEDLINE | ID: mdl-32397556

ABSTRACT

Eckol, a precursor compound belonging to the dibenzo-1,4-dioxin class of phlorotannins, is a phloroglucinol derivative that exerts various activities. In the present study, we investigated the antiallergic effects of eckol isolated from the marine brown algae, Ecklonia cava using immunoglobulin E (IgE)/bovine serum albumin (BSA)-stimulated mouse bone marrow-derived cultured mast cells (BMCMC) and a mouse model of anaphylaxis. Eckol inhibited IgE/BSA-induced BMCMC degranulation by reducing ß-hexosaminidase release. A flow cytometric analysis revealed that eckol decreases FcεRI expression on cell surface and IgE binding to the FcεRI in BMCMC. Moreover, eckol suppressed the production of the cytokines, interleukin (IL)-4, IL-5, IL-6, and IL-13 and the chemokine, thymus activation-regulated chemokine (TARC) by downregulating, IκB-α degradation and NF-κB nuclear translocation. Furthermore, it attenuated the passive cutaneous anaphylactic reaction induced by IgE/BSA-stimulation in the ear of BALB/c mice. These results suggest that eckol is a potential therapeutic candidate for the prevention and treatment of allergic disorders.


Subject(s)
Anaphylaxis/drug therapy , Anti-Allergic Agents , Dioxins/pharmacology , Dioxins/therapeutic use , Immunoglobulin E/immunology , Mast Cells/immunology , Passive Cutaneous Anaphylaxis/drug effects , Passive Cutaneous Anaphylaxis/immunology , Phaeophyceae/chemistry , Phytotherapy , Anaphylaxis/immunology , Animals , Cells, Cultured , Dioxins/isolation & purification , Mice, Inbred BALB C , Mice, Inbred C57BL
7.
Int Immunopharmacol ; 84: 106500, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32311669

ABSTRACT

BACKGROUND: Mast cells (MCs) mediate a key role in allergic diseases. Detailed studies of how the neuroleptic drug pimozide affects MC activity are lacking. The aim of this study was to investigate pimozide inhibition of immunoglobulin E (IgE)-mediated MC activation and MC-mediated allergic responses. METHOD: MCs were stimulated with anti-dinitrophenyl (DNP) IgE antibodies and DNP-horse serum albumin (HSA) antigen (Ag), and anti-allergic pimozide effects were detected by measuring ß-hexosaminidase levels. Morphological changes were observed histologically. In vivo pimozide effects were assessed in passive cutaneous anaphylaxis (PCA) and ovalbumin (OVA)-sensitized active systemic anaphylaxis mouse (ASA) model experiments. Levels of phosphorylated (p-) SYK (spleen tyrosine kinase) and MAPKs (mitogen-activated protein kinases) were detected in western blots. RESULTS: We found that pimozide inhibited MC degranulation, reduced MC release of ß-hexosaminidase dose-dependently in activated RBL-2H3 (IC50: 13.52 µM) and bone marrow derived MC (BMMC) (IC50: 42.42 µM), and reduced MC morphological changes. The IgE/Ag-induced migration effect was suppressed by pimozide treatment dose-dependently. Pimozide down-regulated IgE/Ag-induced phosphorylation of SYK and MAPKs in activated MCs. Moreover, pimozide attenuated allergic reactions in PCA and ASA model mice, and decreased MC populations among splenic cells. CONCLUSIONS: The antipsychotic drug pimozide can suppress IgE-mediated MC activation in vitro and in vivo and should be considered for repurposing to suppress MC-mediated diseases.


Subject(s)
Anti-Allergic Agents/pharmacology , Immunoglobulin E/drug effects , Immunoglobulin E/metabolism , Mast Cells/drug effects , Mast Cells/metabolism , Pimozide/pharmacology , Anaphylaxis/drug therapy , Anaphylaxis/immunology , Animals , Anti-Allergic Agents/therapeutic use , Cell Degranulation/drug effects , Cell Line , Cell Movement/drug effects , Disease Models, Animal , Female , Mast Cells/cytology , Mice , Mice, Inbred BALB C , Mitogen-Activated Protein Kinases/metabolism , Passive Cutaneous Anaphylaxis/drug effects , Passive Cutaneous Anaphylaxis/immunology , Pimozide/therapeutic use , Rats , Syk Kinase/metabolism
8.
Front Immunol ; 11: 591975, 2020.
Article in English | MEDLINE | ID: mdl-33643282

ABSTRACT

The systemic anaphylactic reaction is a life-threatening allergic response initiated by activated mast cells. Sphingolipids are an essential player in the development and attenuation of this response. De novo synthesis of sphingolipids in mammalian cells is inhibited by the family of three ORMDL proteins (ORMDL1, 2, and 3). However, the cell and tissue-specific functions of ORMDL proteins in mast cell signaling are poorly understood. This study aimed to determine cross-talk of ORMDL2 and ORMDL3 proteins in IgE-mediated responses. To this end, we prepared mice with whole-body knockout (KO) of Ormdl2 and/or Ormdl3 genes and studied their role in mast cell-dependent activation events in vitro and in vivo. We found that the absence of ORMDL3 in bone marrow-derived mast cells (BMMCs) increased the levels of cellular sphingolipids. Such an increase was further raised by simultaneous ORMDL2 deficiency, which alone had no effect on sphingolipid levels. Cells with double ORMDL2 and ORMDL3 KO exhibited increased intracellular levels of sphingosine-1-phosphate (S1P). Furthermore, we found that concurrent ORMDL2 and ORMDL3 deficiency increased IκB-α phosphorylation, degranulation, and production of IL-4, IL-6, and TNF-α cytokines in antigen-activated mast cells. Interestingly, the chemotaxis towards antigen was increased in all mutant cell types analyzed. Experiments in vivo showed that passive cutaneous anaphylaxis (PCA), which is initiated by mast cell activation, was increased only in ORMDL2,3 double KO mice, supporting our in vitro observations with mast cells. On the other hand, ORMDL3 KO and ORMDL2,3 double KO mice showed faster recovery from passive systemic anaphylaxis, which could be mediated by increased levels of blood S1P presented in such mice. Our findings demonstrate that Ormdl2 deficiency potentiates the ORMDL3-dependent changes in mast cell signaling.


Subject(s)
Mast Cells/immunology , Mast Cells/metabolism , Membrane Proteins/deficiency , Signal Transduction , Amino Acid Sequence , Anaphylaxis/etiology , Anaphylaxis/metabolism , Animals , Biomarkers , Calcium/metabolism , Calcium Signaling , Chemotaxis/immunology , Cytokines/metabolism , Disease Susceptibility , Gene Expression , Lysophospholipids/blood , Lysophospholipids/metabolism , Membrane Proteins/chemistry , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Knockout , Multigene Family , Passive Cutaneous Anaphylaxis/genetics , Passive Cutaneous Anaphylaxis/immunology , Sphingolipids/blood , Sphingolipids/metabolism , Sphingosine/analogs & derivatives , Sphingosine/blood , Sphingosine/metabolism
10.
J Ethnopharmacol ; 250: 112484, 2020 Mar 25.
Article in English | MEDLINE | ID: mdl-31843576

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: A promising approach to treat a variety of diseases are considered as complementary and alternative herbal medicines. Prunus serrulata var. spontanea L. (Rosaceae) is used as herbal medicine to treat allergic diseases according to the Donguibogam, a tradition medical book of the Joseon Dynasty in Korea. AIM OF THE STUDY: We prepared the aqueous extract of the bark of P. serrulata (AEBPS) and aimed to investigate the effects in mouse anaphylaxis models and various types of mast cells, including RBL-2H3, primary cultured peritoneal and bone marrow-derived mast cells. MATERIALS AND METHODS: We used ovalbumin (OVA)-induced active systemic anaphylaxis (ASA) and immunoglobulin (Ig) E-mediated passive cutaneous anaphylaxis (PCA) models, in vivo. The control drug dexamethasone (10 mg/kg) was used to compare the effectiveness of AEBPS (1-100 mg/kg). In vitro, IgE-stimulated mast cells were used to confirm the role of AEBPS (1-100 µg/mL). For statistical analyses, p values less than 0.05 were considered to be significant. RESULTS: In ASA model, oral administration of AEBPS suppressed the hypothermia and increased level of serum histamine in a dose-dependent manner. AEBPS attenuated the serum IgE, OVA-specific IgE, and interleukin (IL)-4. Oral administration of AEBPS also blocked mast cell-dependent PCA. AEBPS suppressed degranulation of mast cells by reducing intracellular calcium level in mast cells. AEBPS inhibited tumor necrosis factor-α and IL-4 expression and secretion in a concentration-dependent manner through the reduction of nuclear factor-κB. CONCLUSIONS: On the basis of these findings, AEBPS could serve as a potential therapeutic target for the management of mast cell-mediated allergic inflammation and as a regulator of mast cell activation.


Subject(s)
Anaphylaxis/drug therapy , Mast Cells/immunology , Plant Extracts/pharmacology , Prunus/chemistry , Anaphylaxis/immunology , Animals , Dose-Response Relationship, Drug , Histamine/blood , Immunoglobulin E/immunology , Male , Medicine, Korean Traditional , Mice , Mice, Inbred ICR , Ovalbumin/immunology , Passive Cutaneous Anaphylaxis/drug effects , Passive Cutaneous Anaphylaxis/immunology , Plant Bark , Plant Extracts/administration & dosage , Rats , Rats, Sprague-Dawley
11.
Int Immunopharmacol ; 75: 105800, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31401386

ABSTRACT

Iopamidol is a radiographic contrast media which caused a very high incidence of anaphylactic reactions. Mast cells are sentinel cells in host defense reactions during immediate hypersensitivity responses and anaphylactic responses. Mas-related G protein-coupled receptor X2 (MRGPRX2) is a kind of mast cell specific receptor, which triggers mast cell degranulation in anaphylactic reactions. Mice MrgprB2 is a homologous gene of MRGPRX2. We sought to better understand the anaphylactic reactions induced by Iopamidol and the mechanisms involving MRGPRX2. The MRGPRX2-related anaphylactic reactions induced by Iopamidol were investigated using the hindpaw swelling and extravasation assay in vivo and a calcium imaging assay was used for mast cell intracellular calcium responses detection and mast cell release of anaphylactic mediators, such as ß-hexosaminidase, histamine and TNF-α, was also detected in vitro. The mast cell deficient KitW-sh/W-sh mice and MrgprB2 knockout mice exhibited a reduced Iopamidol-induced inflammation effect compared with wild type mice. Furthermore, human mast cells that express MRGPRX2 were activated by Iopamidol in a dose-dependent manner, meanwhile MRGPRX2 knockdown mast cells showed reduced intracellular calcium responses and anaphylactic mediators release effect. It could be concluded that Iopamidol-induced anaphylactoid reactions were MRGPRX2 mediated to provoke mast cells Ca2+ mobilization and degranulation.


Subject(s)
Drug Hypersensitivity/immunology , Iopamidol , Mast Cells/immunology , Passive Cutaneous Anaphylaxis/immunology , Receptors, G-Protein-Coupled/immunology , Animals , Calcium/immunology , Cell Line , Humans , Immunoglobulin E , Mice, Inbred C57BL , Mice, Knockout , Receptors, G-Protein-Coupled/genetics , Tumor Necrosis Factor-alpha/immunology
12.
Front Immunol ; 10: 3068, 2019.
Article in English | MEDLINE | ID: mdl-31993060

ABSTRACT

Protein 4.1R, a member of the 4.1 family, functions as a bridge between cytoskeletal and plasma membrane proteins. It is expressed in T cells, where it binds to a linker for activation of T cell (LAT) family member 1 and inhibits its phosphorylation and downstream signaling events after T cell receptor triggering. The role of the 4.1R protein in cell activation through other immunoreceptors is not known. In this study, we used 4.1R-deficient (4.1R-KO) and 4.1R wild-type (WT) mice and explored the role of the 4.1R protein in the high-affinity IgE receptor (FcεRI) signaling in mast cells. We found that bone marrow mast cells (BMMCs) derived from 4.1R-KO mice showed normal growth in vitro and expressed FcεRI and c-KIT at levels comparable to WT cells. However, 4.1R-KO cells exhibited reduced antigen-induced degranulation, calcium response, and secretion of tumor necrosis factor-α. Chemotaxis toward antigen and stem cell factor (SCF) and spreading on fibronectin were also reduced in 4.1R-KO BMMCs, whereas prostaglandin E2-mediated chemotaxis was not affected. Antibody-induced aggregation of tetraspanin CD9 inhibited chemotaxis toward antigen in WT but not 4.1R-KO BMMCs, implying a CD9-4.1R protein cross-talk. Further studies documented that in the absence of 4.1R, antigen-mediated phosphorylation of FcεRI ß and γ subunits was not affected, but phosphorylation of SYK and subsequent signaling events such as phosphorylation of LAT1, phospholipase Cγ1, phosphatases (SHP1 and SHIP), MAP family kinases (p38, ERK, JNK), STAT5, CBL, and mTOR were reduced. Immunoprecipitation studies showed the presence of both LAT1 and LAT2 (LAT, family member 2) in 4.1R immunocomplexes. The positive regulatory role of 4.1R protein in FcεRI-triggered activation was supported by in vivo experiments in which 4.1R-KO mice showed the normal presence of mast cells in the ears and peritoneum, but exhibited impaired passive cutaneous anaphylaxis. The combined data indicate that the 4.1R protein functions as a positive regulator in the early activation events after FcεRI triggering in mast cells.


Subject(s)
Chemotaxis/immunology , Mast Cells/immunology , Microfilament Proteins/immunology , Receptors, IgE/immunology , Animals , Cell Degranulation/immunology , Mast Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/metabolism , Passive Cutaneous Anaphylaxis/immunology , Receptors, IgE/metabolism
13.
Sci Rep ; 8(1): 14237, 2018 09 24.
Article in English | MEDLINE | ID: mdl-30250145

ABSTRACT

Immunoglobulin E (IgE) plays a central role in the pathogenesis of Type I hypersensitivity through interaction with a high-affinity receptor (FcεRIα). For therapeutic applications, substantial attention has been focused recently on the blockade of the IgE interaction with FcεRIα. While exploring better options for preventing allergic diseases, we found that the Fab fragment of the rat anti-murine IgE antibody (Fab-6HD5) strongly inhibited passive cutaneous anaphylaxis (PCA) in vivo, as well as spleen tyrosine kinase (Syk) activity and ß-hexosaminidase release from basophilic leukemia cells in vitro. The in vivo effects of Fab-6HD5 pre-administration were maintained over a long period of time for at least 10 days. Using flow cytometry analysis, we also found that Fab-6HD5 did not recognize the IgE Cε3 domain containing specific binding sites for FcεRIα. Furthermore, deletion-mapping studies revealed that Fab-6HD5 recognized conformational epitopes on the Cε2 domain of IgE. Given that the Cε2 domain plays a key role in stabilizing the interaction of IgE with FcRIα, our results suggest that the specific binding of Fab-6HD5 to the Cε2 domain prevents allergic reactions through destabilizing the preformed IgE-FcεRIα complex on rat mast cells. Although the present study was performed using animal models, these findings support the idea that a certain antibody directed against IgE CH domains may contribute to preventing allergic diseases through interacting with IgE-FcεRIα complex.


Subject(s)
Hypersensitivity, Immediate/immunology , Immunoglobulin Fab Fragments/immunology , Passive Cutaneous Anaphylaxis/immunology , Receptors, IgE/immunology , Animals , Antibodies, Anti-Idiotypic/immunology , Basophils/immunology , Binding Sites/immunology , Epitopes/immunology , Hexosaminidases/immunology , Humans , Hypersensitivity/genetics , Hypersensitivity/immunology , Immunoglobulin Domains/immunology , Immunoglobulin Fab Fragments/genetics , Mast Cells/immunology , Mice , Passive Cutaneous Anaphylaxis/genetics , Rats , Receptors, IgE/genetics , Syk Kinase/immunology
14.
Yakugaku Zasshi ; 137(5): 517-521, 2017.
Article in Japanese | MEDLINE | ID: mdl-28458281

ABSTRACT

Mast cells are effector cells in immunoglobulin E (IgE)-mediated immediate hypersensitivity and allergic diseases such as asthma and food allergy. Mast cells are activated by the aggregation of the IgE-bound high-affinity IgE receptor FcεRI with multivalent antigen. Activated mast cells secrete proinflammatory mediators such as histamine, serotonin, and proteases and produce cytokines and chemokines. However, it has been reported that mast cells are activated by crosslinking of FcεRI with monomeric IgE in the absence of antigen. We have recently demonstrated that histamine-releasing factor (HRF) is involved in IgE-mediated mast cell activation both in vitro and in vivo. HRF binds to a subset of IgE and IgG molecules [HRF-reactive antibodies (Abs)]. The Fab, but not Fc, portions of the IgE and IgG molecules are HRF-binding sites, and the N-terminal 19-residue (N19) and H3 portions of HRF are HRF-reactive Ab-binding sites. We observed that both N19 and H3 tagged with glutathione S transferase (GST) (GST-N19 and GST-H3) can inhibit the interaction between HRF and HRF-reactive Abs. Using acute- and late-phase passive cutaneous anaphylaxis mouse models, it was shown that HRF initiates mast cell activation through HRF-reactive, but not HRF-nonreactive, IgE in vivo. Antigen-induced passive cutaneous anaphylaxis was inhibited by pretreatment with GST-N19 and GST-H3. We demonstrated that pretreatment with GST-N19 before antigen challenge inhibited antigen-induced mast cell-dependent airway inflammation. In addition, GST-N19 partially inhibited Aspergillus fumigatus extract-induced IgE-dependent airway inflammation. However, GST-N19 did not inhibit T cell-dependent airway inflammation. These results suggest that mast cells are target cells for HRF to initiate IgE- and mast cell-dependent airway inflammation.


Subject(s)
Asthma/immunology , Biomarkers, Tumor/physiology , Mast Cells/immunology , Animals , Antibodies/immunology , Antibodies/metabolism , Binding Sites , Biomarkers, Tumor/immunology , Biomarkers, Tumor/metabolism , Glutathione Transferase/immunology , Glutathione Transferase/metabolism , Humans , Immunoglobulin E/immunology , Immunoglobulin E/metabolism , Mice , Passive Cutaneous Anaphylaxis/immunology , Protein Binding , Tumor Protein, Translationally-Controlled 1
15.
J Immunol ; 198(10): 3823-3834, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28396318

ABSTRACT

Allergic disorders have now become a major worldwide public health issue, but the effective treatment options remain limited. We report a novel approach to block allergic reactivity by targeting the surface-bound IgE of the allergic effector cells via low-affinity anti-human IgE Abs with dissociation constants in the 10-6 to 10-8 M range. We demonstrated that these low-affinity anti-IgE mAbs bind to the cell surface-bound IgE without triggering anaphylactic degranulation even at high concentration, albeit they would weakly upregulate CD203c expression on basophils. This is in contrast to the high-affinity anti-IgE mAbs that trigger anaphylactic degranulation at low concentration. Instead, the low-affinity anti-IgE mAbs profoundly block human peanut- and cat-allergic IgE-mediated basophil CD63 induction indicative of anaphylactic degranulation; suppress peanut-, cat-, and dansyl-specific IgE-mediated passive cutaneous anaphylaxis; and attenuate dansyl IgE-mediated systemic anaphylaxis in human FcεRIα transgenic mouse model. Mechanistic studies reveal that the ability of allergic reaction blockade by the low-affinity anti-IgE mAbs was correlated with their capacity to downregulate the surface IgE and FcεRI level on human basophils and the human FcεRIα transgenic mouse bone marrow-derived mast cells via driving internalization of the IgE/FcεRI complex. Our studies demonstrate that targeting surface-bound IgE with low-affinity anti-IgE Abs is capable of suppressing allergic reactivity while displaying an excellent safety profile, indicating that use of low-affinity anti-IgE mAbs holds promise as a novel therapeutic approach for IgE-mediated allergic diseases.


Subject(s)
Anaphylaxis/prevention & control , Antibodies, Anti-Idiotypic/immunology , Antibody Affinity , Hypersensitivity/prevention & control , Immunoglobulin E/immunology , Anaphylaxis/drug therapy , Anaphylaxis/immunology , Animals , Antibodies, Anti-Idiotypic/administration & dosage , Antibodies, Anti-Idiotypic/metabolism , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/metabolism , Basophils/immunology , Cell Degranulation/immunology , Cytokines/blood , Cytokines/immunology , Humans , Hypersensitivity/immunology , Immunoglobulin E/metabolism , Mice , Mice, Transgenic , Passive Cutaneous Anaphylaxis/immunology , Phosphoric Diester Hydrolases/immunology , Protein Binding , Pyrophosphatases/immunology , Tetraspanin 30/immunology
16.
Proc Natl Acad Sci U S A ; 114(8): E1490-E1499, 2017 02 21.
Article in English | MEDLINE | ID: mdl-28167789

ABSTRACT

DNA methylation and specifically the DNA methyltransferase enzyme DNMT3A are involved in the pathogenesis of a variety of hematological diseases and in regulating the function of immune cells. Although altered DNA methylation patterns and mutations in DNMT3A correlate with mast cell proliferative disorders in humans, the role of DNA methylation in mast cell biology is not understood. By using mast cells lacking Dnmt3a, we found that this enzyme is involved in restraining mast cell responses to acute and chronic stimuli, both in vitro and in vivo. The exacerbated mast cell responses observed in the absence of Dnmt3a were recapitulated or enhanced by treatment with the demethylating agent 5-aza-2'-deoxycytidine as well as by down-modulation of Dnmt1 expression, further supporting the role of DNA methylation in regulating mast cell activation. Mechanistically, these effects were in part mediated by the dysregulated expression of the scaffold protein IQGAP2, which is characterized by the ability to regulate a wide variety of biological processes. Altogether, our data demonstrate that DNMT3A and DNA methylation are key modulators of mast cell responsiveness to acute and chronic stimulation.


Subject(s)
Cell Proliferation/physiology , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation/physiology , Dermatitis, Contact/immunology , Epigenesis, Genetic/physiology , Mast Cells/physiology , Passive Cutaneous Anaphylaxis/immunology , ras GTPase-Activating Proteins/metabolism , Animals , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Cell Degranulation/genetics , Cell Proliferation/drug effects , Cells, Cultured , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methylation/drug effects , DNA Methyltransferase 3A , Decitabine , Dermatitis, Contact/etiology , Disease Models, Animal , Down-Regulation , Enzyme-Linked Immunosorbent Assay , Female , Fluorescent Antibody Technique , Immunoglobulin E/immunology , Interleukin-3/metabolism , Mast Cells/drug effects , Mastocytosis, Systemic/immunology , Mice , Mice, Knockout , Mutation , Oxazolone/toxicity , RNA Interference , RNA, Small Interfering/metabolism , ras GTPase-Activating Proteins/genetics
17.
Autoimmunity ; 50(2): 86-101, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28010135

ABSTRACT

Interleukin (IL) 17A in chronic inflammation is also produced by innate immune cells as neutrophils. Mice with chronic humoral response induced by venom of Thalassophryne nattereri (VTn) proved to be a good tool for evaluating the impact of IL-17A on the development of long-lived plasma cells in the inflamed peritoneal cavity. Here, we report that VTn induces IL-17A production by neutrophils accumulating in the peritoneal cavity and triggers the extrusion of IL-17A along with neutrophil extracellular traps (NETs). Neutrophil depletion reduced the number of IL17A-producing cells in VTn-immunized mice and blocked the differentiation of long-lived plasma cells. Specific antibody production and survival of long-lived plasma cells was ablated in VTn-immunized mice deficient in CD4, while CD28 signaling had the opposite effect on differentiation of long-lived plasma cells. Further, maturation of long-lived plasma cells in inflamed peritoneal cavity was IL-1R1 and COX-2 dependent. Finally, when both the Raf-MEK-ERK pathway and the IL-17A or IL-1R1 activities were blocked, neutrophils were unable to promote the differentiation of memory B cells into long-lived plasma cells, confirming the essential role of neutrophils and IL-17A along with NETs in an IL-1/IL-1R-dependent manner as the novel helping partner for plasma cell differentiation in chronically inflamed tissues.


Subject(s)
Cell Differentiation/immunology , Extracellular Traps/metabolism , Interleukin-17/metabolism , Neutrophils/immunology , Neutrophils/metabolism , Plasma Cells/immunology , Plasma Cells/metabolism , Receptors, Interleukin-1 Type I/metabolism , Animals , Cyclooxygenase 2/metabolism , Enzyme-Linked Immunosorbent Assay , Extracellular Traps/immunology , Fish Venoms/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Immunologic Memory , Lymphocyte Activation , Male , Mice , Mice, Knockout , Passive Cutaneous Anaphylaxis/immunology , Plasma Cells/cytology
18.
Nat Commun ; 7: 13198, 2016 10 27.
Article in English | MEDLINE | ID: mdl-27786273

ABSTRACT

Cross-linkage of the high-affinity immunoglobulin E (IgE) receptor (FcɛRI) on mast cells by antigen ligation has a critical role in the pathology of IgE-dependent allergic disorders, such as anaphylaxis and asthma. Restraint of intracellular signal transduction pathways that promote release of mast cell-derived pro-inflammatory mediators is necessary to dampen activation and restore homoeostasis. Here we show that the ligase Nedd4-2 and the adaptor Ndfip1 (Nedd4 family interacting protein 1) limit the intensity and duration of IgE-FcɛRI-induced positive signal transduction by ubiquitinating phosphorylated Syk, a tyrosine kinase that is indispensable for downstream FcɛRI signalosome activity. Importantly, loss of Nedd4-2 or Ndfip1 in mast cells results in exacerbated and prolonged IgE-mediated cutaneous anaphylaxis in vivo. Our findings reveal an important negative regulatory function for Nedd4-2 and Ndfip1 in IgE-dependent mast cell activity.


Subject(s)
Carrier Proteins/immunology , Immunoglobulin E/immunology , Mast Cells/immunology , Membrane Proteins/immunology , Nedd4 Ubiquitin Protein Ligases/immunology , Adoptive Transfer , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cells, Cultured , Cytokines/immunology , Cytokines/metabolism , Female , Immunoglobulin E/metabolism , Intercellular Signaling Peptides and Proteins , Male , Mast Cells/metabolism , Mast Cells/transplantation , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nedd4 Ubiquitin Protein Ligases/genetics , Nedd4 Ubiquitin Protein Ligases/metabolism , Passive Cutaneous Anaphylaxis/genetics , Passive Cutaneous Anaphylaxis/immunology , Receptors, IgE/immunology , Receptors, IgE/metabolism , Syk Kinase/immunology , Syk Kinase/metabolism
19.
Cell Immunol ; 306-307: 9-16, 2016.
Article in English | MEDLINE | ID: mdl-27311921

ABSTRACT

Mast cells play a critical role in allergic reactions. The cross-linking of FcεRI-bound IgE with multivalent antigen initiates a cascade of signaling events leading to mast cell activation. It has been well-recognized that cross linking of FcεRI mediates tyrosine phosphorylation. However, the mechanism involved in tyrosine dephosphorylation in mast cells is less clear. Here we demonstrated that protein tyrosine phosphatase 1B (PTP1B)-deficient mast cells showed increased IgE-mediated phosphorylation of the signal transducer and activator of transcription 5 (STAT5) and enhanced production of CCL9 (MIP-1γ) and IL-6 in IgE-mediated mast cells activation in vitro. However, IgE-mediated calcium mobilization, ß-hexaosaminidase release (degranulation), and phosphorylation of IκB and MAP kinases were not affected by PTP1B deficiency. Furthermore, PTP1B deficient mice showed normal IgE-dependent passive cutaneous anaphylaxis and late phase cutaneous reactions in vivo. Thus, PTP1B specifically regulates IgE-mediated STAT5 pathway, but is redundant in influencing mast cell function in vivo.


Subject(s)
Mast Cells/immunology , Passive Cutaneous Anaphylaxis/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Animals , Cells, Cultured , Chemokines, CC/metabolism , Humans , Immunoglobulin E/metabolism , Interleukin-4/metabolism , Interleukin-6/metabolism , Macrophage Inflammatory Proteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout , Phosphorylation/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , STAT5 Transcription Factor/metabolism
20.
J Allergy Clin Immunol ; 138(3): 769-779, 2016 09.
Article in English | MEDLINE | ID: mdl-27139822

ABSTRACT

BACKGROUND: Mast cells are a critical component of allergic responses in humans, and animal models that allow the in vivo investigation of their contribution to allergy and evaluation of new human-specific therapeutics are urgently needed. OBJECTIVE: To develop a new humanized mouse model that supports human mast cell engraftment and human IgE-dependent allergic responses. METHODS: This model is based on the NOD-scid IL2rg(null)SCF/GM-CSF/IL3 (NSG-SGM3) strain of mice engrafted with human thymus, liver, and hematopoietic stem cells (termed Bone marrow, Liver, Thymus [BLT]). RESULTS: Large numbers of human mast cells develop in NSG-SGM3 BLT mice and populate the immune system, peritoneal cavity, and peripheral tissues. The human mast cells in NSG-SGM3 BLT mice are phenotypically similar to primary human mast cells and express CD117, tryptase, and FcεRI. These mast cells undergo degranulation in an IgE-dependent and -independent manner, and can be readily cultured in vitro for additional studies. Intradermal priming of engrafted NSG-SGM3 mice with a chimeric IgE containing human constant regions resulted in the development of a robust passive cutaneous anaphylaxis response. Moreover, we describe the first report of a human mast cell antigen-dependent passive systemic anaphylaxis response in primed mice. CONCLUSIONS: NSG-SGM3 BLT mice provide a readily available source of human mast cells for investigation of mast cell biology and a preclinical model of passive cutaneous anaphylaxis and passive systemic anaphylaxis that can be used to investigate the pathogenesis of human allergic responses and to test new therapeutics before their advancement to the clinic.


Subject(s)
Anaphylaxis/immunology , Disease Models, Animal , Mast Cells/immunology , Passive Cutaneous Anaphylaxis/immunology , Animals , Hematopoietic Stem Cell Transplantation , Humans , Immunoglobulin E/immunology , Liver Transplantation , Mice , Thymus Gland/transplantation
SELECTION OF CITATIONS
SEARCH DETAIL
...