Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Cell Immunol ; 371: 104470, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34942481

RESUMEN

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.


Asunto(s)
Adenosina Trifosfato/metabolismo , Alarminas/inmunología , Interleucina-33/metabolismo , Mastocitos/metabolismo , Peritonitis/patología , Animales , Calcineurina/metabolismo , Degranulación de la Célula/inmunología , Células Cultivadas , Citocinas/biosíntesis , Eosinófilos/inmunología , Inflamación/patología , Macrófagos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Factores de Transcripción NFATC/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/genética , Receptores Purinérgicos P2X7/genética , Receptores Purinérgicos P2X7/metabolismo
2.
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
3.
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
4.
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
5.
Cell Immunol ; 356: 104134, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32862025

RESUMEN

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.


Asunto(s)
Mastocitos/fisiología , Receptores de IgG/metabolismo , Factor de Transcripción STAT5/metabolismo , Anafilaxia/inmunología , Animales , Degranulación de la Célula/fisiología , Femenino , Humanos , Masculino , Mastocitos/citología , Mastocitos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Fosforilación , Proteínas Proto-Oncogénicas c-fyn/metabolismo , Receptores de IgE/metabolismo , Receptores de IgG/inmunología , Factor de Transcripción STAT5/genética , Factor de Transcripción STAT5/inmunología , Transducción de Señal , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Familia-src Quinasas/metabolismo
6.
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
7.
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
8.
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
9.
J Immunol ; 191(9): 4505-13, 2013 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-24068671

RESUMEN

We previously demonstrated that TGF-ß1 suppresses IgE-mediated signaling in human and mouse mast cells in vitro, an effect that correlated with decreased expression of the high-affinity IgE receptor, FcεRI. The in vivo effects of TGF-ß1 and the means by which it suppresses mast cells have been less clear. This study shows that TGF-ß1 suppresses FcεRI and c-Kit expression in vivo. By examining changes in cytokine production concurrent with FcεRI expression, we found that TGF-ß1 suppresses TNF production independent of FcεRI levels. Rather, IgE-mediated signaling was altered. TGF-ß1 significantly reduced expression of Fyn and Stat5, proteins critical for cytokine induction. These changes may partly explain the effects of TGF-ß1, because Stat5B overexpression blocked TGF-mediated suppression of IgE-induced cytokine production. We also found that Stat5B is required for mast cell migration toward stem cell factor, and that TGF-ß1 reduced this migration. We found evidence that genetic background may alter TGF responses. TGF-ß1 greatly reduced mast cell numbers in Th1-prone C57BL/6, but not Th2-prone 129/Sv mice. Furthermore, TGF-ß1 did not suppress IgE-induced cytokine release and did increase c-Kit-mediated migration in 129/Sv mast cells. These data correlated with high basal Fyn and Stat5 expression in 129/Sv cells, which was not reduced by TGF-ß1 treatment. Finally, primary human mast cell populations also showed variable sensitivity to TGF-ß1-mediated changes in Stat5 and IgE-mediated IL-6 secretion. We propose that TGF-ß1 regulates mast cell homeostasis, and that this feedback suppression may be dependent on genetic context, predisposing some individuals to atopic disease.


Asunto(s)
Inmunoglobulina E/inmunología , Mastocitos/metabolismo , Receptores de IgE/inmunología , Factor de Transcripción STAT5/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Movimiento Celular/inmunología , Células Cultivadas , Citocinas/metabolismo , Humanos , Inmunoglobulina E/metabolismo , Mastocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas c-fyn/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Interferencia de ARN , ARN Interferente Pequeño , Receptores de IgE/biosíntesis , Receptores de IgE/metabolismo , Factor de Transcripción STAT5/genética , Factor de Transcripción STAT5/inmunología , Transducción de Señal/inmunología , Factor de Crecimiento Transformador beta1/inmunología , Factores de Necrosis Tumoral/biosíntesis
10.
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
11.
J Biol Chem ; 287(3): 2045-54, 2012 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-22130676

RESUMEN

Previous studies indicate that STAT5 expression is required for mast cell development, survival, and IgE-mediated function. STAT5 tyrosine phosphorylation is swiftly and transiently induced by activation of the high affinity IgE receptor, FcεRI. However, the mechanism for this mode of activation remains unknown. In this study we observed that STAT5 co-localizes with FcεRI in antigen-stimulated mast cells. This localization was supported by cholesterol depletion of membranes, which ablated STAT5 tyrosine phosphorylation. Through the use of various pharmacological inhibitors and murine knock-out models, we found that IgE-mediated STAT5 activation is dependent upon Fyn kinase, independent of Syk, PI3K, Akt, Bruton's tyrosine kinase, and JAK2, and enhanced in the context of Lyn kinase deficiency. STAT5 immunoprecipitation revealed that unphosphorylated protein preassociates with Fyn and that this association diminishes significantly during mast cell activation. SHP-1 tyrosine phosphatase deficiency modestly enhanced STAT5 phosphorylation. This effect was more apparent in the absence of Gab2, a scaffolding protein that docks with multiple negative regulators, including SHP-1, SHP-2, and Lyn. Targeting of STAT5A or B with specific siRNA pools revealed that IgE-mediated mast cell cytokine production is selectively dependent upon the STAT5B isoform. Altogether, these data implicate Fyn as the major positive mediator of STAT5 after FcεRI engagement and demonstrate importantly distinct roles for STAT5A and STAT5B in mast cell function.


Asunto(s)
Citocinas/biosíntesis , Mastocitos/metabolismo , Receptores de IgE/metabolismo , Factor de Transcripción STAT5/metabolismo , Transducción de Señal/fisiología , Proteínas Adaptadoras Transductoras de Señales , Agammaglobulinemia Tirosina Quinasa , Animales , Células Cultivadas , Colesterol/genética , Colesterol/metabolismo , Citocinas/genética , Janus Quinasa 2/genética , Janus Quinasa 2/metabolismo , Mastocitos/citología , Ratones , Ratones Noqueados , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Fosforilación/fisiología , Proteína Tirosina Fosfatasa no Receptora Tipo 6/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-fyn/genética , Proteínas Proto-Oncogénicas c-fyn/metabolismo , Receptores de IgE/genética , Factor de Transcripción STAT5/genética , Familia-src Quinasas/genética , Familia-src Quinasas/metabolismo
12.
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
13.
Exp Hematol ; 34(5): 579-87, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16647563

RESUMEN

Mast cells are potent effectors of the inflammatory response, playing an important role in atopy, bacterial immunity, and animal models of arthritis, multiple sclerosis, and heart disease. Hence controlling mast cell numbers and responsiveness is essential for preventing inflammatory disease. We demonstrate that the cytokine transforming growth factor (TGF) beta1 is a potent inducer of mast cell apoptosis, a finding that was consistent in cultured mouse bone marrow-derived mast cells, peritoneal mast cells, and human mast cells. Cell death appeared to be caused by TGF-mediated repression of interleukin-3 (IL-3) receptor expression and function, leading to mitochondrial damage and activation of an apoptotic cascade acting via p53 and caspases. Although IL-3 receptor expression was reduced within 1 day of TGFbeta1 stimulation, apoptosis required at least 3 days to occur. This delay in onset is postulated to allow protective mast cell effector functions, protecting the host from infection while preventing the establishment of chronic inflammation. Our data support the theory that TGFbeta1 is an inhibitor of mast cell survival. The widespread expression of TGFbeta1 offers this cytokine as an ideal candidate for control of mast cell homeostasis.


Asunto(s)
Apoptosis/fisiología , Mastocitos/citología , Factor de Crecimiento Transformador beta/fisiología , Animales , Células Cultivadas , Humanos , Ratones , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas c-kit/metabolismo , Receptores de Interleucina-3/antagonistas & inhibidores , Receptores de Interleucina-3/fisiología , Factor de Transcripción STAT5/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
14.
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
15.
J Leukoc Biol ; 95(4): 643-50, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24338630

RESUMEN

Mast cells and MDSCs are increased by parasitic infection and tumor growth. We previously demonstrated that enhanced MDSC development in ADAM10 transgenic mice yielded resistance to Nb infection and that coculturing MDSCs and mast cells enhanced cytokine production. In the current work, we show that MDSC-mast cell coculture selectively enhances IgE-mediated cytokine secretion among mast cells, without increasing MDSC cytokine production. This effect was independent of cell contact and elicited by Ly6C(+) and Ly6C/G+ MDSC subsets. These interactions were functionally important. MDSC depletion with the FDA-approved drug gemcitabine exacerbated Nb or Trichinella spiralis infection and reduced mast cell-dependent AHR and lung inflammation. Adoptive transfer of MDSC worsened AHR in WT but not mast cell-deficient Wsh/Wsh mice. These data support the hypothesis that MDSCs enhance mast cell inflammatory responses and demonstrate that this interaction can be altered by an existing chemotherapeutic.


Asunto(s)
Inmunoglobulina E/inmunología , Mastocitos/inmunología , Células Mieloides/fisiología , Animales , Asma/inmunología , Células Cultivadas , Citocinas/biosíntesis , Femenino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Nippostrongylus/inmunología , Trichinella spiralis/inmunología
16.
J Immunol ; 177(5): 3421-6, 2006 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-16920984

RESUMEN

The mast cell (MC) inflammatory response is now linked not only to atopy, but also to arthritis, multiple sclerosis, heart disease, and resistance to bacterial infection. In the current study, we demonstrate that the signal transducer and activator of transcription 5 (Stat5) is rapidly activated by IgE cross-linkage, and that its expression is critical to the MC response. Stat5-deficient (Stat5KO) MC demonstrated a significant decrease in IgE-mediated degranulation, leukotriene B4 production, cytokine secretion, and survival signals. The defect in cytokine production may be caused by decreased cytokine mRNA stability. Stat5KO MC-induced cytokine mRNAs normally following IgE cross-linkage, but these mRNAs were not sustained over time and were degraded at twice the rate observed in WT cells. Interestingly, the RNA destabilizing protein tristetraprolin was induced following IgE cross-linkage in Stat5KO but not wild-type cells. Moreover, reducing tristetraprolin expression via short hairpin RNA transfection significantly increased IL-13 production in Stat5KO MC. Our work demonstrates that Stat5 is a critical factor in IgE-induced MC activation, acting in part via posttranscriptional control of cytokine mRNA stability. These data have a direct impact on MC-associated inflammatory and autoimmune diseases.


Asunto(s)
Inmunoglobulina E/inmunología , Mastocitos/inmunología , Mastocitos/metabolismo , Factor de Transcripción STAT5/metabolismo , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Diferenciación Celular , Células Cultivadas , Citocinas/biosíntesis , Citocinas/genética , Activación Enzimática , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Leucotrienos/biosíntesis , Mastocitos/citología , Ratones , Ratones Noqueados , Estabilidad del ARN/genética , Receptores de IgE/inmunología , Factor de Transcripción STAT5/deficiencia , Factor de Transcripción STAT5/genética , Transducción de Señal , Tristetraprolina/metabolismo
17.
J Immunol ; 175(5): 3000-5, 2005 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-16116187

RESUMEN

Mast cells are critical effectors of allergic disease, and are now implicated in immune responses observed in arthritis, multiple sclerosis, and heart disease. Because of their role in inflammation, understanding how mast cells develop is of clinical importance. In this study we determined the effects of IFN-gamma on mast cell survival. Using in vitro culture of bone marrow cells in IL-3 plus stem cell factor, we found that the addition of IFN-gamma induced apoptosis, as exhibited by the presence of subdiploid DNA and caspase activation. IFN-gamma-mediated apoptosis was Stat1-dependent, and was accompanied by loss of mitochondrial membrane potential. Apoptosis was reduced in cultures of bone marrow cells derived from p53- or Bax-deficient mice, as well as H2K-Bcl-2 transgenic mice. IFN-gamma hyperresponsiveness has been shown to result in inflammatory disease and death in mice lacking the regulatory protein suppressor of cytokine signaling (SOCS)-1. Bone marrow cells from SOCS-1 knockout (KO) mice failed to give rise to viable mast cells after culture in IL-3 plus stem cell factor, with profound apoptosis occurring as the cultures matured. However, bone marrow cells lacking both SOCS-1 and IFN-gamma survived normally. This in vitro defect in mast cell development was recapitulated in vivo. SOCS-1 KO mice demonstrated a 67% decrease in peritoneal mast cell numbers relative to wild-type mice, a deficiency that was reversed in SOCS-1/IFN-gamma KO mice. These data demonstrate the potent regulatory effects of IFN-gamma on mast cell survival and show that this cytokine can elicit mast cell death in vitro and in vivo.


Asunto(s)
Apoptosis/efectos de los fármacos , Interferón gamma/farmacología , Mastocitos/efectos de los fármacos , Animales , Proteínas Portadoras/fisiología , Recuento de Células , Proteínas de Unión al ADN/fisiología , Mastocitos/citología , Ratones , Ratones Noqueados , Mitocondrias/fisiología , Proteínas Proto-Oncogénicas c-kit/análisis , Proteínas Represoras/fisiología , Factor de Transcripción STAT1 , Proteína 1 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas , Transactivadores/fisiología
18.
J Immunol ; 174(10): 5987-93, 2005 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-15879091

RESUMEN

Mast cell activation through the high affinity IgE receptor (FcepsilonRI) is a critical component of atopic inflammation. The cytokine TGF-beta1 has been shown to inhibit IgE-dependent mast cell activation, possibly serving to dampen mast cell-mediated inflammatory responses. We present proof that TGF-beta1 inhibits mast cell FcepsilonRI expression through a reversible pathway that diminishes protein, but not mRNA, expression of the FcepsilonRI subunit proteins alpha, beta, and gamma. The stability of the expressed proteins and the assembled cell surface complex was unaltered by TGF-beta1 treatment. However, TGF-beta1 decreased the rate of FcepsilonRI beta-chain synthesis, arguing that this inhibitory cytokine exerts its effects at the level of mRNA translation. TGF-beta1 consistently diminished FcepsilonRI expression on cultured human or mouse mast cells as well as freshly isolated peritoneal mast cells. The related cytokines, TGF-beta2 and TGF-beta3, had similar effects. We propose that TGF-beta1 acts as a negative regulator of mast cell function, in part by decreasing FcepsilonRI expression.


Asunto(s)
Mastocitos/inmunología , Mastocitos/metabolismo , Receptores de IgE/antagonistas & inhibidores , Receptores de IgE/biosíntesis , Factor de Crecimiento Transformador beta/fisiología , Animales , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Células Cultivadas , Ratones , Ratones Endogámicos C57BL , Subunidades de Proteína/antagonistas & inhibidores , Subunidades de Proteína/biosíntesis , Subunidades de Proteína/genética , ARN Mensajero/biosíntesis , Receptores de IgE/genética , Factor de Crecimiento Transformador beta1
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA