Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Immunol ; 212(8): 1277-1286, 2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38381001

RESUMEN

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.


Asunto(s)
Asma , MicroARNs , Animales , Ratones , Asma/genética , Citocinas/genética , Retroalimentación , Inmunidad Innata , Proteína 1 Similar al Receptor de Interleucina-1/genética , Interleucina-33 , Linfocitos/metabolismo , Mastocitos/metabolismo , Ratones Noqueados , MicroARNs/genética , MicroARNs/metabolismo
2.
Sci Signal ; 16(802): eabc9089, 2023 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-37699080

RESUMEN

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.


Asunto(s)
Fluoxetina , Mastocitos , Humanos , Animales , Ratones , Fluoxetina/farmacología , Retroalimentación , Inflamación/tratamiento farmacológico , Citocinas , Adenosina Trifosfato , Inmunoglobulina E
3.
bioRxiv ; 2023 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-37163060

RESUMEN

Group 2 innate lymphoid cells (ILC2s) cooperate with adaptive Th2 cells as key organizers of tissue type 2 immune responses, while a spectrum of innate and adaptive lymphocytes coordinate early type 3/17 immunity. Both type 2 and type 3/17 lymphocyte associated cytokines are linked to tissue fibrosis, but how their dynamic and spatial topographies may direct beneficial or pathologic organ remodelling is unclear. Here we used volumetric imaging in models of liver fibrosis, finding accumulation of periportal and fibrotic tract IL-5 + lymphocytes, predominantly ILC2s, in close proximity to expanded type 3/17 lymphocytes and IL-33 high niche fibroblasts. Ablation of IL-5 + lymphocytes worsened carbon tetrachloride-and bile duct ligation-induced liver fibrosis with increased niche IL-17A + type 3/17 lymphocytes, predominantly γδ T cells. In contrast, concurrent ablation of IL-5 + and IL-17A + lymphocytes reduced this progressive liver fibrosis, suggesting a cross-regulation of type 2 and type 3 lymphocytes at specialized fibroblast niches that tunes hepatic fibrosis.

4.
Immunity ; 55(2): 254-271.e7, 2022 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-35139352

RESUMEN

Allergic immunity is orchestrated by group 2 innate lymphoid cells (ILC2s) and type 2 helper T (Th2) cells prominently arrayed at epithelial- and microbial-rich barriers. However, ILC2s and Th2 cells are also present in fibroblast-rich niches within the adventitial layer of larger vessels and similar boundary structures in sterile deep tissues, and it remains unclear whether they undergo dynamic repositioning during immune perturbations. Here, we used thick-section quantitative imaging to show that allergic inflammation drives invasion of lung and liver non-adventitial parenchyma by ILC2s and Th2 cells. However, during concurrent type 1 and type 2 mixed inflammation, IFNγ from broadly distributed type 1 lymphocytes directly blocked both ILC2 parenchymal trafficking and subsequent cell survival. ILC2 and Th2 cell confinement to adventitia limited mortality by the type 1 pathogen Listeria monocytogenes. Our results suggest that the topography of tissue lymphocyte subsets is tightly regulated to promote appropriately timed and balanced immunity.


Asunto(s)
Inflamación/inmunología , Interferón gamma/inmunología , Subgrupos Linfocitarios/inmunología , Células Th2/inmunología , Animales , Muerte Celular/inmunología , Movimiento Celular/inmunología , Hipersensibilidad/inmunología , Inmunidad Innata , Interleucina-33/inmunología , Interleucina-5/metabolismo , Listeria monocytogenes , Listeriosis/inmunología , Listeriosis/mortalidad , Hígado/inmunología , Pulmón/inmunología , Subgrupos Linfocitarios/metabolismo , Lisofosfolípidos/inmunología , Ratones , Tejido Parenquimatoso/inmunología , Esfingosina/análogos & derivados , Esfingosina/inmunología , Células TH1/inmunología , Células Th2/metabolismo
5.
Cell Immunol ; 371: 104457, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34883342

RESUMEN

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.


Asunto(s)
Fluvastatina/farmacología , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Interleucina-33/metabolismo , Interleucina-6/biosíntesis , Mastocitos/metabolismo , Factor de Necrosis Tumoral alfa/biosíntesis , Animales , Células Cultivadas , Humanos , Inmunoglobulina E/inmunología , Inflamación/inmunología , Células Asesinas Naturales/inmunología , Macrófagos/inmunología , Ácido Mevalónico/farmacología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Peritonitis/inducido químicamente , Prenilación/efectos de los fármacos , Factor de Células Madre/metabolismo , Terpenos/farmacología , Factor de Transcripción ReIA/metabolismo , Transcripción Genética/efectos de los fármacos
6.
J Pharmacol Exp Ther ; 374(1): 104-112, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32434944

RESUMEN

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.


Asunto(s)
Apoptosis/efectos de los fármacos , Fluvastatina/farmacología , Mastocitos/citología , Mastocitos/efectos de los fármacos , Animales , Células de la Médula Ósea/citología , Línea Celular , Supervivencia Celular/efectos de los fármacos , Humanos , Metabolismo de los Lípidos/efectos de los fármacos , Mastocitos/metabolismo , Ratones
7.
J Immunol ; 203(2): 453-464, 2019 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-31160535

RESUMEN

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.


Asunto(s)
Adenosina Trifosfato/metabolismo , Glucólisis/efectos de los fármacos , Ácido Láctico/farmacología , Lipopolisacáridos/farmacología , Mastocitos/efectos de los fármacos , Animales , Citocinas/metabolismo , Endotoxemia/tratamiento farmacológico , Endotoxemia/metabolismo , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Mastocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Sepsis/tratamiento farmacológico , Sepsis/metabolismo , Transducción de Señal/efectos de los fármacos
8.
Cell Immunol ; 341: 103918, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31030957

RESUMEN

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.


Asunto(s)
Anafilaxia/prevención & control , Antiinflamatorios no Esteroideos/farmacología , Retroalimentación Fisiológica , Inmunoglobulina E/genética , Ácido Láctico/farmacología , Mastocitos/efectos de los fármacos , Agammaglobulinemia Tirosina Quinasa/genética , Agammaglobulinemia Tirosina Quinasa/inmunología , Anafilaxia/inducido químicamente , Anafilaxia/inmunología , Anafilaxia/patología , Animales , Dinitrofenoles/administración & dosificación , Dinitrofenoles/antagonistas & inhibidores , Femenino , Regulación de la Expresión Génica , Cetoprofeno/farmacología , Ácido Láctico/inmunología , Ácido Láctico/metabolismo , Mastocitos/inmunología , Mastocitos/patología , Ratones , Ratones Endogámicos C57BL , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/inmunología , Transportadores de Ácidos Monocarboxílicos/genética , Transportadores de Ácidos Monocarboxílicos/inmunología , Cavidad Peritoneal/patología , Fosforilación/efectos de los fármacos , Cultivo Primario de Células , Albúmina Sérica/administración & dosificación , Albúmina Sérica/antagonistas & inhibidores , Transducción de Señal , Piel/efectos de los fármacos , Piel/inmunología , Piel/patología , Quinasa Syk/genética , Quinasa Syk/inmunología , Simportadores/genética , Simportadores/inmunología
9.
Methods Mol Biol ; 1799: 81-92, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29956146

RESUMEN

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.


Asunto(s)
Basófilos/inmunología , Basófilos/metabolismo , Inflamación/inmunología , Inflamación/metabolismo , Mastocitos/inmunología , Mastocitos/metabolismo , Animales , Basófilos/citología , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Diferenciación Celular , Separación Celular , Humanos , Inmunoglobulina E/inmunología , Interleucina-3/metabolismo , Mastocitos/citología , Ratones , Lavado Peritoneal , Piel/citología , Piel/inmunología , Piel/metabolismo
10.
Front Immunol ; 9: 868, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29755466

RESUMEN

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.


Asunto(s)
Antialérgicos/uso terapéutico , Homeostasis/inmunología , Hipersensibilidad/inmunología , Mastocitos/inmunología , Transducción de Señal/inmunología , Antialérgicos/farmacología , Citocinas/inmunología , Citocinas/metabolismo , Historia del Siglo XX , Historia del Siglo XXI , Homeostasis/efectos de los fármacos , Humanos , Hipersensibilidad/tratamiento farmacológico , Mastocitos/efectos de los fármacos , Mastocitos/metabolismo , Receptores de IgE/inmunología , Receptores de IgE/metabolismo , Receptores de IgG/inmunología , Receptores de IgG/metabolismo
11.
Front Immunol ; 9: 3026, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30619366

RESUMEN

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.


Asunto(s)
Hipersensibilidad/tratamiento farmacológico , Interleucina-33/inmunología , Mastocitos/inmunología , Metformina/uso terapéutico , Peritonitis/tratamiento farmacológico , Adenosina Trifosfato/biosíntesis , Animales , Antimetabolitos/farmacología , Células Cultivadas , Desoxiglucosa/farmacología , Modelos Animales de Enfermedad , Femenino , Glucólisis/efectos de los fármacos , Glucólisis/inmunología , Humanos , Hipersensibilidad/inmunología , Hipersensibilidad/metabolismo , Interleucina-33/metabolismo , Masculino , Mastocitos/metabolismo , Metformina/farmacología , Ratones , Ratones Endogámicos C57BL , Fosforilación Oxidativa/efectos de los fármacos , Peritonitis/inmunología , Peritonitis/metabolismo , Cultivo Primario de Células , Resultado del Tratamiento
12.
Cell Immunol ; 322: 41-48, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28964543

RESUMEN

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.


Asunto(s)
Antiinflamatorios/farmacología , Antioxidantes/farmacología , Ácidos Hidroxámicos/farmacología , Hipersensibilidad/tratamiento farmacológico , Inmunoglobulina E/inmunología , Mastocitos/inmunología , FN-kappa B/genética , Factor de Transcripción AP-1/genética , Acetilcisteína/farmacología , Animales , Células de la Médula Ósea/inmunología , Catalasa/biosíntesis , Degranulación de la Célula/efectos de los fármacos , Células Cultivadas , Quimiocina CCL3/biosíntesis , Hipersensibilidad/inmunología , Interleucina-13/biosíntesis , Interleucina-6/biosíntesis , Mastocitos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/inmunología , Especies Reactivas de Oxígeno/metabolismo , Superóxido Dismutasa/biosíntesis , Transcripción Genética/efectos de los fármacos , Factor de Necrosis Tumoral alfa/biosíntesis
13.
Cell Immunol ; 319: 10-16, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28750923

RESUMEN

While IgE is considered the primary mediator of mast cell activation, IL-33 contributes substantially in asthma, allergic rhinitis, and atopic dermatitis. To develop effective treatments for allergic disease, it is important to understand the role of therapeutic agents on IL-33 activation. We examined the effect of Didox (3,4-dihydroxybenzohydroxamic acid), an antioxidant and ribonucleotide reductase (RNR) inhibitor, on IL-33-mediated mast cell activation. Didox suppressed IL-6, IL-13, TNF, and MIP-1α (CCL3) production in bone marrow derived mast cells following IL-33 activation. This suppression was observed in different genetic backgrounds and extended to peritoneal mast cells. The antioxidant N-acetylcysteine mimicked the suppression of Didox, albeit at a much higher dose, while the RNR inhibitor hydroxyurea had no effect. Didox substantially suppressed IL-33-mediated NFκB and AP-1 transcriptional activities. These results suggest that Didox attenuates IL-33-induced mast cell activation and should be further studied as a potential therapeutic agent for inflammatory diseases involving IL-33.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Ácidos Hidroxámicos/farmacología , Inmunosupresores/farmacología , Interleucina-33/farmacología , Mastocitos/efectos de los fármacos , Acetilcisteína/farmacología , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/inmunología , Quimiocina CCL3/antagonistas & inhibidores , Quimiocina CCL3/genética , Quimiocina CCL3/inmunología , Femenino , Regulación de la Expresión Génica/inmunología , Genes Reporteros , Hidroxiurea/farmacología , Interleucina-13/antagonistas & inhibidores , Interleucina-13/genética , Interleucina-13/inmunología , Interleucina-33/antagonistas & inhibidores , Lipopolisacáridos/farmacología , Luciferasas/genética , Luciferasas/inmunología , Masculino , Mastocitos/citología , Mastocitos/inmunología , Ratones , Ratones Endogámicos C57BL , FN-kappa B/antagonistas & inhibidores , FN-kappa B/genética , FN-kappa B/inmunología , Cultivo Primario de Células , Transducción de Señal , Factor de Transcripción AP-1/antagonistas & inhibidores , Factor de Transcripción AP-1/genética , Factor de Transcripción AP-1/inmunología , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología
14.
J Immunol ; 199(3): 866-873, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28637902

RESUMEN

TGF-ß1 is involved in many pathological conditions, including autoimmune disorders, cancer, and cardiovascular and allergic diseases. We have previously found that TGF-ß1 can suppress IgE-mediated mast cell activation of human and mouse mast cells. IL-33 is a member of the IL-1 family capable of inducing mast cell responses and enhancing IgE-mediated activation. In this study, we investigated the effects of TGF-ß on IL-33-mediated mast cell activation. Bone marrow-derived mast cells cultured in TGF-ß1, ß2, or ß3 showed reduced IL-33-mediated production of TNF, IL-6, IL-13, and MCP-1 in a concentration-dependent manner. TGF-ß1 inhibited IL-33-mediated Akt and ERK phosphorylation as well as NF-κB- and AP-1-mediated transcription. These effects were functionally important, as TGF-ß1 injection suppressed IL-33-induced systemic cytokines in vivo and inhibited IL-33-mediated cytokine release from human mast cells. TGF-ß1 also suppressed the combined effects of IL-33 and IgE-mediated activation on mouse and human mast cells. The role of IL-33 in the pathogenesis of allergic diseases is incompletely understood. These findings, consistent with our previously reported effects of TGF-ß1 on IgE-mediated activation, demonstrate that TGF-ß1 can provide broad inhibitory signals to activated mast cells.


Asunto(s)
Interleucina-33/inmunología , Mastocitos/inmunología , Factor de Crecimiento Transformador beta1/fisiología , Animales , Células Cultivadas , Citocinas/antagonistas & inhibidores , Citocinas/biosíntesis , Citocinas/inmunología , Humanos , Inmunoglobulina E/inmunología , Interleucina-6/biosíntesis , Interleucina-6/inmunología , Activación de Linfocitos/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Mastocitos/efectos de los fármacos , Mastocitos/metabolismo , Ratones , FN-kappa B/genética , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de IgE/inmunología , Factor de Transcripción AP-1/genética , Factor de Crecimiento Transformador beta1/farmacología , Factor de Crecimiento Transformador beta3/farmacología
15.
J Immunol ; 197(7): 2909-17, 2016 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-27559047

RESUMEN

Lactic acid (LA) is present in tumors, asthma, and wound healing, environments with elevated IL-33 and mast cell infiltration. Although IL-33 is a potent mast cell activator, how LA affects IL-33-mediated mast cell function is unknown. To investigate this, mouse bone marrow-derived mast cells were cultured with or without LA and activated with IL-33. LA reduced IL-33-mediated cytokine and chemokine production. Using inhibitors for monocarboxylate transporters (MCT) or replacing LA with sodium lactate revealed that LA effects are MCT-1- and pH-dependent. LA selectively altered IL-33 signaling, suppressing TGF-ß-activated kinase-1, JNK, ERK, and NF-κB phosphorylation, but not p38 phosphorylation. LA effects in other contexts have been linked to hypoxia-inducible factor (HIF)-1α, which was enhanced in bone marrow-derived mast cells treated with LA. Because HIF-1α has been shown to regulate the microRNA miR-155 in other systems, LA effects on miR-155-5p and miR-155-3p species were measured. In fact, LA selectively suppressed miR-155-5p in an HIF-1α-dependent manner. Moreover, overexpressing miR-155-5p, but not miR-155-3p, abolished LA effects on IL-33-induced cytokine production. These in vitro effects of reducing cytokines were consistent in vivo, because LA injected i.p. into C57BL/6 mice suppressed IL-33-induced plasma cytokine levels. Lastly, IL-33 effects on primary human mast cells were suppressed by LA in an MCT-dependent manner. Our data demonstrate that LA, present in inflammatory and malignant microenvironments, can alter mast cell behavior to suppress inflammation.


Asunto(s)
Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Inflamación/prevención & control , Interleucina-33/inmunología , Ácido Láctico/farmacología , Mastocitos/efectos de los fármacos , Mastocitos/inmunología , MicroARNs/genética , Animales , Células Cultivadas , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Inflamación/inmunología , Masculino , Mastocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , MicroARNs/biosíntesis , Relación Estructura-Actividad
16.
J Leukoc Biol ; 100(6): 1395-1404, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27443878

RESUMEN

Mast cells are critical effectors of allergic disease and can be activated by IL-33, a proinflammatory member of the IL-1 cytokine family. IL-33 worsens the pathology of mast cell-mediated diseases, but therapies to antagonize IL-33 are still forthcoming. Because steroids are the mainstay of allergic disease treatment and are well known to suppress mast cell activation by other stimuli, we examined the effects of the steroid dexamethasone on IL-33-mediated mast cell function. We found that dexamethasone potently and rapidly suppressed cytokine production elicited by IL-33 from murine bone marrow-derived and peritoneal mast cells. IL-33 enhances IgE-mediated mast cell cytokine production, an activity that was also antagonized by dexamethasone. These effects were consistent in human mast cells. We additionally observed that IL-33 augmented migration of IgE-sensitized mast cells toward antigen. This enhancing effect was similarly reversed by dexamethasone. Simultaneous addition of dexamethasone with IL-33 had no effect on the phosphorylation of MAP kinases or NFκB p65 subunit; however, dexamethasone antagonized AP-1- and NFκB-mediated transcriptional activity. Intraperitoneal administration of dexamethasone completely abrogated IL-33-mediated peritoneal neutrophil recruitment and prevented plasma IL-6 elevation. These data demonstrate that steroid therapy may be an effective means of antagonizing the effects of IL-33 on mast cells in vitro and in vivo, acting partly by suppressing IL-33-induced NFκB and AP-1 activity.


Asunto(s)
Dexametasona/farmacología , Interleucina-33/antagonistas & inhibidores , Mastocitos/efectos de los fármacos , Factores de Transcripción/antagonistas & inhibidores , Transcripción Genética/efectos de los fármacos , Animales , Movimiento Celular/efectos de los fármacos , Quimiotaxis de Leucocito/efectos de los fármacos , Citocinas/biosíntesis , Citocinas/genética , Humanos , Inmunoglobulina E/inmunología , Proteína 1 Similar al Receptor de Interleucina-1/biosíntesis , Proteína 1 Similar al Receptor de Interleucina-1/genética , Interleucina-33/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Mifepristona/farmacología , Neutrófilos/efectos de los fármacos , Receptores de Glucocorticoides/efectos de los fármacos , Receptores de Glucocorticoides/fisiología , Proteínas Recombinantes/farmacología , Piel/patología , Factores de Transcripción/genética
17.
J Immunol ; 196(11): 4457-67, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27183599

RESUMEN

IL-10 is an important regulatory cytokine that modulates a wide range of immune cells. Whereas it is best known for its ability to suppress immune responses, IL-10 has been found to be pathogenic in several human and animal studies of immune-mediated diseases. There is a considerable gap in our understanding of the molecular mechanisms behind the stimulatory effects of IL-10 during allergic inflammation. IL-10 treatment has been shown to suppress mast cell TNF production. In this study, we report that whereas TNF secretion was reduced, IL-10 surprisingly enhanced IgE-mediated protease and cytokine production both in vitro and in vivo. This stimulatory effect was consistent in mouse and human skin mast cells. IL-10 enhanced activation of the key FcεRI signaling proteins Stat5, JNK, and ERK. We demonstrate that IL-10 effects are dependent on Stat3 activation, eliciting miR-155 expression, with a resulting loss of suppressor of cytokine signaling-1. The importance of miR-155 was demonstrated by the inability of IL-10 to enhance anaphylaxis in miR-155-deficient mice. Taken together, our results reveal an IL-10-induced, Stat3-miR-155 signaling pathway that can promote mast cell responses.


Asunto(s)
Inmunoglobulina E/inmunología , Interleucina-10/inmunología , Mastocitos/inmunología , MicroARNs/genética , Proteína 1 Supresora de la Señalización de Citocinas/genética , Animales , Células Cultivadas , Mastocitos/citología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , MicroARNs/inmunología
18.
J Immunol ; 196(4): 1461-70, 2016 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-26773154

RESUMEN

Mast cell (MC)- and basophil-associated inflammatory diseases are a considerable burden to society. A significant portion of patients have symptoms despite standard-of-care therapy. Statins, used to lower serum cholesterol, have immune-modulating activities. We tested the in vitro and in vivo effects of statins on IgE-mediated MC and basophil activation. Fluvastatin showed the most significant inhibitory effects of the six statins tested, suppressing IgE-induced cytokine secretion among mouse MCs and basophils. The effects of fluvastatin were reversed by mevalonic acid or geranylgeranyl pyrophosphatase, and mimicked by geranylgeranyl transferase inhibition. Fluvastatin selectively suppressed key FcεRI signaling pathways, including Akt and ERK. Although MCs and basophils from the C57BL/6J mouse strain were responsive to fluvastatin, those from 129/SvImJ mice were completely resistant. Resistance correlated with fluvastatin-induced upregulation of the statin target HMG-CoA reductase. Human MC cultures from eight donors showed a wide range of fluvastatin responsiveness. These data demonstrate that fluvastatin is a potent suppressor of IgE-mediated MC activation, acting at least partly via blockade of geranyl lipid production downstream of HMG-CoA reductase. Importantly, consideration of statin use for treating MC-associated disease needs to incorporate genetic background effects, which can yield drug resistance.


Asunto(s)
Basófilos/efectos de los fármacos , Ácidos Grasos Monoinsaturados/farmacología , Inmunoglobulina E/biosíntesis , Indoles/farmacología , Mastocitos/efectos de los fármacos , Acilcoenzima A/genética , Acilcoenzima A/inmunología , Animales , Apoptosis , Basófilos/inmunología , Células Cultivadas , Citocinas/biosíntesis , Farnesiltransferasa/metabolismo , Femenino , Fluvastatina , Genotipo , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Inmunoglobulina E/inmunología , Mastocitos/inmunología , Ácido Mevalónico/farmacología , Ratones , Ratones Endogámicos C57BL , Transducción de Señal/efectos de los fármacos , Células Th2/inmunología
19.
Cell Immunol ; 290(1): 80-8, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24950026

RESUMEN

A Disintegrin and Metalloproteinase (ADAM)-10 plays critical roles in neuronal migration and distribution. Recently, ADAM10 deletion was shown to disrupt myelopoiesis. We found that inducible deletion of ADAM10 using Mx1-driven Cre recombinase for a period of three weeks resulted in mast cell hyperplasia in the skin, intestine and spleen. Mast cells express surface ADAM10 in vitro and in vivo, at high levels compared to other immune cells tested. ADAM10 is important for mast cell migration, since ADAM10-deficiency reduced c-Kit-mediated migration. As with some mast cell proteases, ADAM10 expression could be altered by the cytokine microenvironment, being inhibited by IL-10 or TGFß1, but not by several other T cell-derived cytokines. Collectively these data show that the ADAM10 protease is an important factor in mast cell migration and tissue distribution, and can be manipulated by environmental cues.


Asunto(s)
Proteínas ADAM/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Movimiento Celular/genética , Mastocitos/fisiología , Proteínas de la Membrana/metabolismo , Factor de Células Madre/metabolismo , Proteínas ADAM/biosíntesis , Proteínas ADAM/genética , Proteína ADAM10 , Secretasas de la Proteína Precursora del Amiloide/biosíntesis , Secretasas de la Proteína Precursora del Amiloide/genética , Animales , Proliferación Celular , Células Cultivadas , Hiperplasia/genética , Interleucina-10/farmacología , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Peritoneo/citología , Interferencia de ARN , ARN Interferente Pequeño , Linfocitos T/inmunología , Factor de Crecimiento Transformador beta/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...