Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
J Immunol ; 212(8): 1277-1286, 2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38381001

ABSTRACT

IL-33 is an inflammatory cytokine that promotes allergic disease by activating group 2 innate lymphoid cells, Th2 cells, and mast cells. IL-33 is increased in asthmatics, and its blockade suppresses asthma-like inflammation in mouse models. Homeostatic control of IL-33 signaling is poorly understood. Because the IL-33 receptor, ST2, acts via cascades used by the TLR family, similar feedback mechanisms may exist. MicroRNA (miR)-146a is induced by LPS-mediated TLR4 signaling and serves as a feedback inhibitor. Therefore, we explored whether miR-146a has a role in IL-33 signaling. IL-33 induced cellular and exosomal miR-146a expression in mouse bone marrow-derived mast cells (BMMCs). BMMCs transfected with a miR-146a antagonist or derived from miR-146a knockout mice showed enhanced cytokine expression in response to IL-33, suggesting that miR-146a is a negative regulator of IL-33-ST2 signaling. In vivo, miR-146a expression in plasma exosomes was elevated after i.p. injection of IL-33 in wild-type but not mast cell-deficient KitW-sh/W-sh mice. Finally, KitW-sh/W-sh mice acutely reconstituted with miR-146a knockout BMMCs prior to IL-33 challenge had elevated plasma IL-6 levels compared with littermates receiving wild-type BMMCs. These results support the hypothesis that miR-146a is a feedback regulator of IL-33-mediated mast cell functions associated with allergic disease.


Subject(s)
Asthma , MicroRNAs , Animals , Mice , Asthma/genetics , Cytokines/genetics , Feedback , Immunity, Innate , Interleukin-1 Receptor-Like 1 Protein/genetics , Interleukin-33 , Lymphocytes/metabolism , Mast Cells/metabolism , Mice, Knockout , MicroRNAs/genetics , MicroRNAs/metabolism
2.
Sci Immunol ; 9(91): eadg8691, 2024 Jan 19.
Article in English | MEDLINE | ID: mdl-38241399

ABSTRACT

Allergic diseases are common, affecting more than 20% of the population. Genetic variants in the TGFß pathway are strongly associated with atopy. To interrogate the mechanisms underlying this association, we examined patients and mice with Loeys-Dietz syndrome (LDS) who harbor missense mutations in the kinase domain of TGFΒR1/2. We demonstrate that LDS mutations lead to reduced TGFß signaling and elevated total and allergen-specific IgE, despite the presence of wild-type T regulatory cells in a chimera model. Germinal center activity was enhanced in LDS and characterized by a selective increase in type 2 follicular helper T cells (TFH2). Expression of Pik3cg was increased in LDS TFH cells and associated with reduced levels of the transcriptional repressor SnoN. PI3Kγ/mTOR signaling in LDS naïve CD4+ T cells was elevated after T cell receptor cross-linking, and pharmacologic inhibition of PI3Kγ or mTOR prevented exaggerated TFH2 and antigen-specific IgE responses after oral antigen exposure in an adoptive transfer model. Naïve CD4+ T cells from nonsyndromic allergic individuals also displayed decreased TGFß signaling, suggesting that our mechanistic discoveries may be broadly relevant to allergic patients in general. Thus, TGFß plays a conserved, T cell-intrinsic, and nonredundant role in restraining TFH2 development via the PI3Kγ/mTOR pathway and thereby protects against allergic disease.


Subject(s)
Hypersensitivity , Transforming Growth Factor beta , Animals , Humans , Mice , Hypersensitivity/metabolism , Immunoglobulin E , Th2 Cells , TOR Serine-Threonine Kinases
4.
Sci Signal ; 16(802): eabc9089, 2023 09 12.
Article in English | MEDLINE | ID: mdl-37699080

ABSTRACT

There is a clinical need for new treatment options addressing allergic disease. Selective serotonin reuptake inhibitors (SSRIs) are a class of antidepressants that have anti-inflammatory properties. We tested the effects of the SSRI fluoxetine on IgE-induced function of mast cells, which are critical effectors of allergic inflammation. We showed that fluoxetine treatment of murine or human mast cells reduced IgE-mediated degranulation, cytokine production, and inflammatory lipid secretion, as well as signaling mediated by the mast cell activator ATP. In a mouse model of systemic anaphylaxis, fluoxetine reduced hypothermia and cytokine production. Fluoxetine was also effective in a model of allergic airway inflammation, where it reduced bronchial responsiveness and inflammation. These data show that fluoxetine suppresses mast cell activation by impeding an FcɛRI-ATP positive feedback loop and support the potential repurposing of this SSRI for use in allergic disease.


Subject(s)
Fluoxetine , Mast Cells , Humans , Animals , Mice , Fluoxetine/pharmacology , Feedback , Inflammation/drug therapy , Cytokines , Adenosine Triphosphate , Immunoglobulin E
5.
Front Allergy ; 4: 1248954, 2023.
Article in English | MEDLINE | ID: mdl-37534194
6.
Nat Immunol ; 24(6): 1036-1048, 2023 06.
Article in English | MEDLINE | ID: mdl-37106040

ABSTRACT

Allergic diseases are a major global health issue. Interleukin (IL)-9-producing helper T (TH9) cells promote allergic inflammation, yet TH9 cell effector functions are incompletely understood because their lineage instability makes them challenging to study. Here we found that resting TH9 cells produced IL-9 independently of T cell receptor (TCR) restimulation, due to STAT5- and STAT6-dependent bystander activation. This mechanism was seen in circulating cells from allergic patients and was restricted to recently activated cells. STAT5-dependent Il9/IL9 regulatory elements underwent remodeling over time, inactivating the locus. A broader 'allergic TH9' transcriptomic and epigenomic program was also unstable. In vivo, TH9 cells induced airway inflammation via TCR-independent, STAT-dependent mechanisms. In allergic patients, TH9 cell expansion was associated with responsiveness to JAK inhibitors. These findings suggest that TH9 cell instability is a negative checkpoint on bystander activation that breaks down in allergy and that JAK inhibitors should be considered for allergic patients with TH9 cell expansion.


Subject(s)
Hypersensitivity , Janus Kinase Inhibitors , Humans , Interleukin-9/genetics , T-Lymphocytes, Helper-Inducer , STAT5 Transcription Factor/genetics , Chromatin/genetics , Inflammation , Hypersensitivity/genetics , Cell Differentiation , STAT6 Transcription Factor
7.
Int J Mol Sci ; 23(18)2022 Sep 17.
Article in English | MEDLINE | ID: mdl-36142776

ABSTRACT

Mast cells (MC) are a key effector cell in multiple types of immune responses, including atopic conditions. Allergic diseases have been steadily rising across the globe, creating a growing public health problem. IgE-mediated activation of MCs leads to the release of potent mediators that can have dire clinical consequences. Current therapeutic options to inhibit MC activation and degranulation are limited; thus, a better understanding of the mechanisms that regulate MC effector functions in allergic inflammation are necessary in order to develop effective treatment options with minimal side effects. Several cytokines have been identified that play multifaceted roles in regulating MC activation, including TGFß, IL-10, and IL-33, and others that appear to serve primarily anti-inflammatory functions, including IL-35 and IL-37. Here, we review the literature examining cytokines that regulate MC-mediated allergic immune responses.


Subject(s)
Cytokines , Mast Cells , Cell Degranulation , Cytokines/metabolism , Humans , Immunoglobulin E , Inflammation/metabolism , Interleukin-10/metabolism , Interleukin-33/metabolism , Mast Cells/metabolism , Transforming Growth Factor beta/metabolism
9.
Cell Immunol ; 371: 104470, 2022 01.
Article in English | MEDLINE | ID: mdl-34942481

ABSTRACT

Inflammatory responses are required to block pathogen infection but can also lead to hypersensitivity and chronic inflammation. Barrier tissues actively release IL-33, ATP, and other alarmins during cell stress, helping identify pathogenic stimuli. However, it is unclear how these signals are integrated. Mast cells are critical initiators of allergic inflammation and respond to IL-33 and ATP. We found that mouse mast cells had a 3-6-fold increase in ATP-induced cytokine production when pre-treated with IL-33. This effect was observed at ATP concentrations < 100 µM and required < 30-minute IL-33 exposure. ATP-induced degranulation was not enhanced by pretreatment nor was the response to several pathogen molecules. Mechanistic studies implicated the P2X7 receptor and calcineurin/NFAT pathway in the enhanced ATP response. Finally, we found that IL-33 + ATP co-stimulation enhanced peritoneal eosinophil and macrophage recruitment. These results support the hypothesis that alarmins collaborate to surpass a threshold necessary to initiate an inflammatory response.


Subject(s)
Adenosine Triphosphate/metabolism , Alarmins/immunology , Interleukin-33/metabolism , Mast Cells/metabolism , Peritonitis/pathology , Animals , Calcineurin/metabolism , Cell Degranulation/immunology , Cells, Cultured , Cytokines/biosynthesis , Eosinophils/immunology , Inflammation/pathology , Macrophages/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , NFATC Transcription Factors/metabolism , RNA Interference , RNA, Small Interfering/genetics , Receptors, Purinergic P2X7/genetics , Receptors, Purinergic P2X7/metabolism
10.
Cell Immunol ; 371: 104457, 2022 01.
Article in English | MEDLINE | ID: mdl-34883342

ABSTRACT

Statins are HMG-CoA reductase inhibitors prescribed for lowering cholesterol. They can also inhibit inflammatory responses by suppressing isoprenylation of small G proteins. Consistent with this, we previously found that fluvastatin suppresses IgE-mediated mast cell function. However, some studies have found that statins induced pro-inflammatory cytokines in macrophages and NK cells. In contrast to IgE signaling, we show that fluvastatin augments IL-33-induced TNF and IL-6 production by mast cells. This effect required the key mast cell growth factor, stem cell factor (SCF). Treatment of IL-33-activated mast cells with mevalonic acid or isoprenoids reduced fluvastatin effects, suggesting fluvastatin acts at least partly by reducing isoprenoid production. Fluvastatin also enhanced IL-33-induced NF-κB transcriptional activity and promoted neutrophilic peritonitis in vivo, a response requiring mast cell activation. Other statins tested did not enhance IL-33 responsiveness. Therefore, this work supports observations of unexpected pro-inflammatory effects of some statins and suggests mechanisms by which this may occur. Because statins are candidates for repurposing in inflammatory disorders, our work emphasizes the importance of understanding the pleiotropic and possible unexpected effects of these drugs.


Subject(s)
Fluvastatin/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Interleukin-33/metabolism , Interleukin-6/biosynthesis , Mast Cells/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , Animals , Cells, Cultured , Humans , Immunoglobulin E/immunology , Inflammation/immunology , Killer Cells, Natural/immunology , Macrophages/immunology , Mevalonic Acid/pharmacology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Peritonitis/chemically induced , Prenylation/drug effects , Stem Cell Factor/metabolism , Terpenes/pharmacology , Transcription Factor RelA/metabolism , Transcription, Genetic/drug effects
11.
Cell Immunol ; 364: 104344, 2021 06.
Article in English | MEDLINE | ID: mdl-33780747

ABSTRACT

Mast cells are found primarily at interfaces with the external environment, where they provide protection from pathogens but also elicit allergic inflammation. Mast cell activation by antigen-induced aggregation of IgE bound to the high affinity receptor, FcεRI, is a critical factor leading to inflammation and bronchoconstriction. We previously found that Stat5 is activated by FcεRI and that Stat5B suppression decreased IgE-induced cytokine production in vitro, but in vivo responses have not been assessed. We now show that Stat5B-deficient (KO) mice have reduced responses to IgE-mediated anaphylaxis, despite normal mast cell tissue distribution. Similarly, Stat5B KO mast cells have diminished IgE-induced degranulation and cytokine secretion in vitro. These mice have elevated IgE production that is not correlated with an intrinsic B cell defect. The current work demonstrates that the Stat5B isoform is required for normal mast cell function and suggests it limits IgE production in vivo.


Subject(s)
Anaphylaxis/immunology , B-Lymphocytes/immunology , Hypersensitivity/immunology , Immunoglobulin E/metabolism , Mast Cells/immunology , Receptors, IgE/metabolism , STAT5 Transcription Factor/metabolism , Animals , Cell Degranulation , Cells, Cultured , Cytokines/metabolism , Humans , Mice , Mice, Inbred BALB C , Mice, Knockout , STAT5 Transcription Factor/genetics
12.
Cell Immunol ; 356: 104134, 2020 10.
Article in English | MEDLINE | ID: mdl-32862025

ABSTRACT

Mast cells, well established effectors in allergic disease, can be activated by numerous stimuli. We previously found that the Fyn-Stat5B pathway is critical for FcεRI-stimulated mast cell function. Because IgG receptors employ similar signaling pathways, we investigated Fyn-Stat5B function downstream of FcγR. We report that FcγR elicits Fyn-dependent Stat5B tyrosine phosphorylation in mast cells. As we previously found for Fyn kinase, Stat5B is indispensable for IgG-mediated mast cell cytokine expression and secretion. However, Stat5B KO macrophages responded normally to FcγR signaling, indicating a lineage-restricted role for Stat5B. This was consistent in vivo, since passive FcγR activation induced anaphylaxis in a macrophage-dominated response even when Stat5B was deleted. We further investigated this lineage restriction using the K/BxN model of inflammatory arthritis. This model exhibits a rapid and transient mast cell-dependent joint inflammation followed days later by a macrophage- and neutrophil-dependent response. Consistent with our hypothesis, Fyn or Stat5B deficiency did not protect mice from late joint swelling, but greatly reduced the early mast cell-dependent response. This was associated with decreased joint and plasma histamine. We conclude that Fyn-Stat5B is a linage-restricted pathway critical for IgG-mediated mast cell responses.


Subject(s)
Mast Cells/physiology , Receptors, IgG/metabolism , STAT5 Transcription Factor/metabolism , Anaphylaxis/immunology , Animals , Cell Degranulation/physiology , Female , Humans , Male , Mast Cells/cytology , Mast Cells/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Phosphorylation , Proto-Oncogene Proteins c-fyn/metabolism , Receptors, IgE/metabolism , Receptors, IgG/immunology , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/immunology , Signal Transduction , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , src-Family Kinases/metabolism
13.
J Pharmacol Exp Ther ; 374(1): 104-112, 2020 07.
Article in English | MEDLINE | ID: mdl-32434944

ABSTRACT

Statin drugs are widely employed in the clinic to reduce serum cholesterol. Because of their hydroxymethylglutaryl coenzyme A reductase antagonism, statins also reduce isoprenyl lipids necessary for the membrane anchorage and signaling of small G-proteins in the Ras superfamily. We previously found that statins suppress immunoglobulin E (IgE)-mediated mast cell activation, suggesting these drugs might be useful in treating allergic disease. Although IgE-induced function is critical to allergic inflammation, mast cell proliferation and survival also impact atopic disease and mast cell neoplasia. In this study, we describe fluvastatin-mediated apoptosis in primary and transformed mast cells. An IC50 was achieved between 0.8 and 3.5 µM in both cell types, concentrations similar to the reported fluvastatin serum Cmax value. Apoptosis was correlated with reduced stem cell factor (SCF)-mediated signal transduction, mitochondrial dysfunction, and caspase activation. Complementing these data, we found that p53 deficiency or Bcl-2 overexpression reduced fluvastatin-induced apoptosis. We also noted evidence of cytoprotective autophagy in primary mast cells treated with fluvastatin. Finally, we found that intraperitoneal fluvastatin treatment reduced peritoneal mast cell numbers in vivo These findings offer insight into the mechanisms of mast cell survival and support the possible utility of statins in mast cell-associated allergic and neoplastic diseases. SIGNIFICANCE STATEMENT: Fluvastatin, a statin drug used to lower cholesterol, induces apoptosis in primary and transformed mast cells by antagonizing protein isoprenylation, effectively inhibiting stem cell factor (SCF)-induced survival signals. This drug may be an effective means of suppressing mast cell survival.


Subject(s)
Apoptosis/drug effects , Fluvastatin/pharmacology , Mast Cells/cytology , Mast Cells/drug effects , Animals , Bone Marrow Cells/cytology , Cell Line , Cell Survival/drug effects , Humans , Lipid Metabolism/drug effects , Mast Cells/metabolism , Mice
14.
J Immunol ; 203(2): 453-464, 2019 07 15.
Article in English | MEDLINE | ID: mdl-31160535

ABSTRACT

Sepsis has a well-studied inflammatory phase, with a less-understood secondary immunosuppressive phase. Elevated blood lactate and slow lactate clearance are associated with mortality; however, regulatory roles are unknown. We hypothesized that lactic acid (LA) contributes to the late phase and is not solely a consequence of bacterial infection. No studies have examined LA effects in sepsis models in vivo or a mechanism by which it suppresses LPS-induced activation in vitro. Because mast cells can be activated systemically and contribute to sepsis, we examined LA effects on the mast cell response to LPS. LA significantly suppressed LPS-induced cytokine production and NF-κB transcriptional activity in mouse bone marrow-derived mast cells and cytokine production in peritoneal mast cells. Suppression was MCT-1 dependent and reproducible with sodium lactate or formic acid. Further, LA significantly suppressed cytokine induction following LPS-induced endotoxemia in mice. Because glycolysis is linked to inflammation and LA is a byproduct of this process, we examined changes in glucose metabolism. LA treatment reduced glucose uptake and lactate export during LPS stimulation. LA effects were mimicked by glycolytic inhibitors and reversed by increasing ATP availability. These results indicate that glycolytic suppression and ATP production are necessary and sufficient for LA effects. Our work suggests that enhancing glycolysis and ATP production could improve immune function, counteracting LA suppressive effects in the immunosuppressive phase of sepsis.


Subject(s)
Adenosine Triphosphate/metabolism , Glycolysis/drug effects , Lactic Acid/pharmacology , Lipopolysaccharides/pharmacology , Mast Cells/drug effects , Animals , Cytokines/metabolism , Endotoxemia/drug therapy , Endotoxemia/metabolism , Inflammation/drug therapy , Inflammation/metabolism , Mast Cells/metabolism , Mice , Mice, Inbred C57BL , Sepsis/drug therapy , Sepsis/metabolism , Signal Transduction/drug effects
15.
Cell Immunol ; 341: 103918, 2019 07.
Article in English | MEDLINE | ID: mdl-31030957

ABSTRACT

Mast cells have functional plasticity affected by their tissue microenvironment, which greatly impacts their inflammatory responses. Because lactic acid (LA) is abundant in inflamed tissues and tumors, we investigated how it affects mast cell function. Using IgE-mediated activation as a model system, we found that LA suppressed inflammatory cytokine production and degranulation in mouse peritoneal mast cells, data that were confirmed with human skin mast cells. In mouse peritoneal mast cells, LA-mediated cytokine suppression was dependent on pH- and monocarboxylic transporter-1 expression. Additionally, LA reduced IgE-induced Syk, Btk, and ERK phosphorylation, key signals eliciting inflammation. In vivo, LA injection reduced IgE-mediated hypothermia in mice undergoing passive systemic anaphylaxis. Our data suggest that LA may serve as a feedback inhibitor that limits mast cell-mediated inflammation.


Subject(s)
Anaphylaxis/prevention & control , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Feedback, Physiological , Immunoglobulin E/genetics , Lactic Acid/pharmacology , Mast Cells/drug effects , Agammaglobulinaemia Tyrosine Kinase/genetics , Agammaglobulinaemia Tyrosine Kinase/immunology , Anaphylaxis/chemically induced , Anaphylaxis/immunology , Anaphylaxis/pathology , Animals , Dinitrophenols/administration & dosage , Dinitrophenols/antagonists & inhibitors , Female , Gene Expression Regulation , Ketoprofen/pharmacology , Lactic Acid/immunology , Lactic Acid/metabolism , Mast Cells/immunology , Mast Cells/pathology , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/immunology , Monocarboxylic Acid Transporters/genetics , Monocarboxylic Acid Transporters/immunology , Peritoneal Cavity/pathology , Phosphorylation/drug effects , Primary Cell Culture , Serum Albumin/administration & dosage , Serum Albumin/antagonists & inhibitors , Signal Transduction , Skin/drug effects , Skin/immunology , Skin/pathology , Syk Kinase/genetics , Syk Kinase/immunology , Symporters/genetics , Symporters/immunology
16.
J Biomed Mater Res A ; 107(4): 884-892, 2019 04.
Article in English | MEDLINE | ID: mdl-30615257

ABSTRACT

Implanted polymer scaffolds can induce inflammation leading to the foreign body response (FBR), fibrosis, and implant failure. Thus, it is important to understand how immune cells interact with scaffolds to mitigate inflammation and promote a regenerative response. We previously demonstrated that macrophage phenotype is modulated by fiber and pore diameters of an electrospun scaffold. However, it is unclear if this effect is consistent among other innate immune cells. Mast cells are inflammatory sentinels that play a vital role in the FBR of implanted biomaterials, as well as angiogenesis. We determined if altering electrospun scaffold architecture modulates mast cell responses, with the goal of promoting regenerative cell-scaffold interactions. Polydioxanone (PDO) scaffolds were made from 60 mg/mL or 140 mg/mL PDO solutions, yielding structures with divergent fiber and pore diameters. Mouse mast cells plated on these scaffolds were activated with IL-33 or lipopolysaccharide (LPS). Relative to the 60 mg/mL scaffold, 140 mg/mL scaffolds yielded less IL-6 and TNF, and greater VEGF secretion. Pores >4-6 µm elicited less IL-6 and TNF secretion. IL-33-induced VEGF regulation was more complex, showing effects of both pore size and fiber diameter. These data indicate parameters that can predict mast cell responses to scaffolds, informing biomaterial design to increase wound healing and diminish implant rejection. © 2019 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 107A: 884-892, 2019.


Subject(s)
Mast Cells/metabolism , Neovascularization, Physiologic , Polydioxanone/chemistry , Tissue Scaffolds/chemistry , Vascular Endothelial Growth Factor A/biosynthesis , Animals , Inflammation/metabolism , Inflammation/pathology , Mast Cells/pathology , Mice
17.
Methods Mol Biol ; 1799: 81-92, 2018.
Article in English | MEDLINE | ID: mdl-29956146

ABSTRACT

Mast cells and basophils are important innate immune cells involved in resistance to parasitic infection and are critical orchestrators of allergic disease. The relative ease with which they are cultured from mouse or human tissues allows one to work with primary cells that maintain a differentiated and functional phenotype. In this chapter, we describe the methods by which mouse mast cells and basophils can be cultured from bone marrow. We also provide methods for isolating and expanding mouse peritoneal mast cells and human skin mast cells.


Subject(s)
Basophils/immunology , Basophils/metabolism , Inflammation/immunology , Inflammation/metabolism , Mast Cells/immunology , Mast Cells/metabolism , Animals , Basophils/cytology , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Cell Differentiation , Cell Separation , Humans , Immunoglobulin E/immunology , Interleukin-3/metabolism , Mast Cells/cytology , Mice , Peritoneal Lavage , Skin/cytology , Skin/immunology , Skin/metabolism
18.
Front Immunol ; 9: 868, 2018.
Article in English | MEDLINE | ID: mdl-29755466

ABSTRACT

Mast cells are tissue resident, innate immune cells with heterogenous phenotypes tuned by cytokines and other microenvironmental stimuli. Playing a protective role in parasitic, bacterial, and viral infections, mast cells are also known for their role in the pathogenesis of allergy, asthma, and autoimmune diseases. Here, we review factors controlling mast cell activation, with a focus on receptor signaling and potential therapies for allergic disease. Specifically, we will discuss our work with FcεRI and FγR signaling, IL-4, IL-10, and TGF-ß1 treatment, and Stat5. We conclude with potential therapeutics for allergic disease. Much of these efforts have been influenced by the work of Bill Paul. With many mechanistic targets for mast cell activation and different classes of therapeutics being studied, there is reason to be hopeful for continued clinical progress in this area.


Subject(s)
Anti-Allergic Agents/therapeutic use , Homeostasis/immunology , Hypersensitivity/immunology , Mast Cells/immunology , Signal Transduction/immunology , Anti-Allergic Agents/pharmacology , Cytokines/immunology , Cytokines/metabolism , History, 20th Century , History, 21st Century , Homeostasis/drug effects , Humans , Hypersensitivity/drug therapy , Mast Cells/drug effects , Mast Cells/metabolism , Receptors, IgE/immunology , Receptors, IgE/metabolism , Receptors, IgG/immunology , Receptors, IgG/metabolism
19.
Front Immunol ; 9: 3026, 2018.
Article in English | MEDLINE | ID: mdl-30619366

ABSTRACT

Cellular metabolism and energy sensing pathways are closely linked to inflammation, but there is little understanding of how these pathways affect mast cell function. Mast cells are major effectors of allergy and asthma, and can be activated by the alarmin IL-33, which is linked to allergic disease. Therefore, we investigated the metabolic requirements for IL-33-induced mast cell function, to identify targets for controlling inflammation. We found that IL-33 increases glycolysis, glycolytic protein expression, and oxidative phosphorylation (OX PHOS). Inhibiting OX PHOS had little effect on cytokine production, but antagonizing glycolysis with 2-deoxyglucose or oxamate suppressed inflammatory cytokine production in vitro and in vivo. ATP reversed this suppression. Glycolytic blockade suppressed IL-33 signaling, including ERK phosphorylation, NFκB transcription, and ROS production in vitro, and suppressed IL-33-induced neutrophil recruitment in vivo. To test a clinically relevant way to modulate these pathways, we examined the effects of the FDA-approved drug metformin on IL-33 activation. Metformin activates AMPK, which suppresses glycolysis in immune cells. We found that metformin suppressed cytokine production in vitro and in vivo, effects that were reversed by ATP, mimicking the actions of the glycolytic inhibitors we tested. These data suggest that glycolytic ATP production is important for IL-33-induced mast cell activation, and that targeting this pathway may be useful in allergic disease.


Subject(s)
Hypersensitivity/drug therapy , Interleukin-33/immunology , Mast Cells/immunology , Metformin/therapeutic use , Peritonitis/drug therapy , Adenosine Triphosphate/biosynthesis , Animals , Antimetabolites/pharmacology , Cells, Cultured , Deoxyglucose/pharmacology , Disease Models, Animal , Female , Glycolysis/drug effects , Glycolysis/immunology , Humans , Hypersensitivity/immunology , Hypersensitivity/metabolism , Interleukin-33/metabolism , Male , Mast Cells/metabolism , Metformin/pharmacology , Mice , Mice, Inbred C57BL , Oxidative Phosphorylation/drug effects , Peritonitis/immunology , Peritonitis/metabolism , Primary Cell Culture , Treatment Outcome
20.
Cell Immunol ; 322: 41-48, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28964543

ABSTRACT

Mast cell activation via the high-affinity IgE receptor (FcεRI) elicits production of inflammatory mediators central to allergic disease. As a synthetic antioxidant and a potent ribonucleotide reductase (RNR) inhibitor, Didox (3,4-dihyroxybenzohydroxamic acid) has been tested in clinical trials for cancer and is an attractive therapeutic for inflammatory disease. We found that Didox treatment of mouse bone marrow-derived mast cells (BMMC) reduced IgE-stimulated degranulation and cytokine production, including IL-6, IL-13, TNF and MIP-1a (CCL3). These effects were consistent using BMMC of different genetic backgrounds and peritoneal mast cells. While the RNR inhibitor hydroxyurea had little or no effect on IgE-mediated function, high concentrations of the antioxidant N-acetylcysteine mimicked Didox-mediated suppression. Furthermore, Didox increased expression of the antioxidant genes superoxide dismutase and catalase, and suppressed DCFH-DA fluorescence, indicating reduced reactive oxygen species production. Didox effects were not due to changes in FcεRI expression or cell viability, suggesting it inhibits signaling required for inflammatory cytokine production. In support of this, we found that Didox reduced FcεRI-mediated AP-1 and NFκB transcriptional activity. Finally, Didox suppressed mast cell-dependent, IgE-mediated passive systemic anaphylaxis in vivo. These data demonstrate the potential use for Didox asa means of antagonizing mast cell responses in allergic disease.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Hydroxamic Acids/pharmacology , Hypersensitivity/drug therapy , Immunoglobulin E/immunology , Mast Cells/immunology , NF-kappa B/genetics , Transcription Factor AP-1/genetics , Acetylcysteine/pharmacology , Animals , Bone Marrow Cells/immunology , Catalase/biosynthesis , Cell Degranulation/drug effects , Cells, Cultured , Chemokine CCL3/biosynthesis , Hypersensitivity/immunology , Interleukin-13/biosynthesis , Interleukin-6/biosynthesis , Mast Cells/drug effects , Mice , Mice, Inbred C57BL , Oxidative Stress/drug effects , Oxidative Stress/immunology , Reactive Oxygen Species/metabolism , Superoxide Dismutase/biosynthesis , Transcription, Genetic/drug effects , Tumor Necrosis Factor-alpha/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...