Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros

Banco de datos
Tipo del documento
Asunto de la revista
Intervalo de año de publicación
1.
J Immunol ; 206(6): 1127-1139, 2021 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-33558372

RESUMEN

T effector cells promote inflammation in asthmatic patients, and both Th2 and Th17 CD4 T cells have been implicated in severe forms of the disease. The metabolic phenotypes and dependencies of these cells, however, remain poorly understood in the regulation of airway inflammation. In this study, we show the bronchoalveolar lavage fluid of asthmatic patients had markers of elevated glucose and glutamine metabolism. Further, peripheral blood T cells of asthmatics had broadly elevated expression of metabolic proteins when analyzed by mass cytometry compared with healthy controls. Therefore, we hypothesized that glucose and glutamine metabolism promote allergic airway inflammation. We tested this hypothesis in two murine models of airway inflammation. T cells from lungs of mice sensitized with Alternaria alternata extract displayed genetic signatures for elevated oxidative and glucose metabolism by single-cell RNA sequencing. This result was most pronounced when protein levels were measured in IL-17-producing cells and was recapitulated when airway inflammation was induced with house dust mite plus LPS, a model that led to abundant IL-4- and IL-17-producing T cells. Importantly, inhibitors of the glucose transporter 1 or glutaminase in vivo attenuated house dust mite + LPS eosinophilia, T cell cytokine production, and airway hyperresponsiveness as well as augmented the immunosuppressive properties of dexamethasone. These data show that T cells induce markers to support metabolism in vivo in airway inflammation and that this correlates with inflammatory cytokine production. Targeting metabolic pathways may provide a new direction to protect from disease and enhance the effectiveness of steroid therapy.


Asunto(s)
Asma/tratamiento farmacológico , Dexametasona/farmacología , Transportador de Glucosa de Tipo 1/antagonistas & inhibidores , Glutaminasa/antagonistas & inhibidores , Inmunosupresores/farmacología , Adulto , Alternaria/inmunología , Animales , Asma/sangre , Asma/inmunología , Biomarcadores/análisis , Biomarcadores/metabolismo , Glucemia/metabolismo , Líquido del Lavado Bronquioalveolar/inmunología , Estudios de Casos y Controles , Células Cultivadas , Dexametasona/uso terapéutico , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Femenino , Transportador de Glucosa de Tipo 1/metabolismo , Glutaminasa/metabolismo , Glutamina/metabolismo , Voluntarios Sanos , Humanos , Inmunosupresores/uso terapéutico , Pulmón/citología , Pulmón/efectos de los fármacos , Pulmón/inmunología , Masculino , Ratones , Persona de Mediana Edad , Cultivo Primario de Células , Pyroglyphidae/inmunología , Células Th17/efectos de los fármacos , Células Th17/inmunología , Células Th17/metabolismo , Células Th2/efectos de los fármacos , Células Th2/inmunología , Células Th2/metabolismo , Adulto Joven
2.
J Immunol ; 199(2): 510-519, 2017 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-28576981

RESUMEN

The appropriate orchestration of different arms of the immune response is critical during viral infection to promote efficient viral clearance while limiting immunopathology. However, the signals and mechanisms that guide this coordination are not fully understood. IFNs are produced at high levels during viral infection and have convergent signaling through STAT1. We hypothesized that STAT1 signaling during viral infection regulates the balance of innate lymphoid cells (ILC), a diverse class of lymphocytes that are poised to respond to environmental insults including viral infections with the potential for both antiviral or immunopathologic functions. During infection with respiratory syncytial virus (RSV), STAT1-deficient mice had reduced numbers of antiviral IFN-γ+ ILC1 and increased numbers of immunopathologic IL-5+ and IL-13+ ILC2 and IL-17A+ ILC3 compared with RSV-infected wild-type mice. Using bone marrow chimeric mice, we found that both ILC-intrinsic and ILC-extrinsic factors were responsible for this ILC dysregulation during viral infection in STAT1-deficient mice. Regarding ILC-extrinsic mechanisms, we found that STAT1-deficient mice had significantly increased expression of IL-33 and IL-23, cytokines that promote ILC2 and ILC3, respectively, compared with wild-type mice during RSV infection. Moreover, disruption of IL-33 or IL-23 signaling attenuated cytokine-producing ILC2 and ILC3 responses in STAT1-deficient mice during RSV infection. Collectively, these data demonstrate that STAT1 is a key orchestrator of cytokine-producing ILC responses during viral infection via ILC-extrinsic regulation of IL-33 and IL-23.


Asunto(s)
Inmunidad Innata , Linfocitos/inmunología , Infecciones por Virus Sincitial Respiratorio/inmunología , Factor de Transcripción STAT1/metabolismo , Animales , Citocinas/biosíntesis , Regulación de la Expresión Génica , Interferón gamma/biosíntesis , Interferón gamma/genética , Interferón gamma/inmunología , Interleucina-13/genética , Interleucina-13/inmunología , Interleucina-17/genética , Interleucina-17/inmunología , Interleucina-23/genética , Interleucina-23/inmunología , Interleucina-33/genética , Interleucina-33/inmunología , Interleucina-5/genética , Interleucina-5/inmunología , Linfocitos/clasificación , Ratones , Infecciones por Virus Sincitial Respiratorio/virología , Factor de Transcripción STAT1/deficiencia , Factor de Transcripción STAT1/genética , Transducción de Señal
3.
J Immunol ; 197(5): 1577-86, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27456482

RESUMEN

Allergic airway diseases are immune disorders associated with heightened type 2 immune responses and IL-5 and IL-13 production at the site of inflammation. We have previously reported that cyclooxygenase (COX) inhibition by indomethacin augmented allergic airway inflammation in a STAT6-independent manner. However, the key COX product(s) responsible for restraining indomethacin-mediated STAT6-independent allergic inflammation is unknown. In this study, using the mouse model of OVA-induced allergic airway inflammation, we identified that PGI2 receptor (IP) signaling was critical for indomethacin-induced, STAT6-independent proallergic effects. We demonstrated that IP deficiency increased inflammatory cell infiltration, eosinophilia, and IL-5 and IL-13 expression in the lung in a STAT6-independent manner. The augmented STAT6-independent allergic inflammation correlated with enhanced primary immune responses to allergic sensitization and elevated production of multiple inflammatory chemokines (CCL11, CCL17, CCL22, and CXCL12) in the lung after allergen challenge. We also showed that the PGI2 analogue cicaprost inhibited CD4 T cell proliferation and IL-5 and IL-13 expression in vitro, and IP deficiency diminished the stimulatory effect of indomethacin on STAT6-independent IL-5 and IL-13 responses in vivo. The inhibitory effects of PGI2 and the IP signaling pathway on CD4 T cell activation, inflammatory chemokine production, and allergic sensitization and airway inflammation suggest that PGI2 and its analogue iloprost, both Food and Drug Administration-approved drugs, may be useful in treating allergic diseases and asthma. In addition, inhibiting PGI2 signaling by drugs that either block PGI2 production or restrain IP signaling may augment STAT6-independent pathways of allergic inflammation.


Asunto(s)
Alérgenos/inmunología , Pulmón/inmunología , Activación de Linfocitos/efectos de los fármacos , Receptores de Epoprostenol/metabolismo , Factor de Transcripción STAT6/metabolismo , Alérgenos/administración & dosificación , Animales , Antihipertensivos/farmacología , Asma/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/fisiología , Proliferación Celular , Quimiocinas/biosíntesis , Quimiocinas/inmunología , Epoprostenol/administración & dosificación , Epoprostenol/análogos & derivados , Epoprostenol/farmacología , Hipersensibilidad , Indometacina , Inflamación , Interleucina-13/genética , Interleucina-13/inmunología , Interleucina-5/genética , Interleucina-5/inmunología , Pulmón/fisiopatología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Ovalbúmina/inmunología , Receptores de Epoprostenol/deficiencia , Receptores de Epoprostenol/genética , Factor de Transcripción STAT6/deficiencia , Factor de Transcripción STAT6/genética , Factor de Transcripción STAT6/inmunología , Transducción de Señal , Células Th2/inmunología
4.
Am J Respir Crit Care Med ; 193(1): 31-42, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26378386

RESUMEN

RATIONALE: Group 2 innate lymphoid cells (ILC2s) robustly produce IL-5 and IL-13, cytokines central to the asthma phenotype; however, the effect of prostaglandin (PG) I2 on ILC2 function is unknown. OBJECTIVES: To determine the effect of PGI2 on mouse and human ILC2 cytokine expression in vitro and the effect of endogenous PGI2 and the PGI2 analog cicaprost on lung ILC2s in vivo. METHODS: Flow-sorted bone marrow ILC2s of wild-type (WT) and PGI2 receptor-deficient (IP(-/-)) mice were cultured with IL-33 and treated with the PGI2 analog cicaprost. WT and IP(-/-) mice were challenged intranasally with Alternaria alternata extract for 4 consecutive days to induce ILC2 responses, and these were quantified. Prior to A. alternata extract, challenged WT mice were treated with cicaprost. Human flow-sorted peripheral blood ILC2s were cultured with IL-33 and IL-2 and treated with the PGI2 analog cicaprost. MEASUREMENT AND MAIN RESULTS: We demonstrate that PGI2 inhibits IL-5 and IL-13 protein expression by IL-33-stimulated ILC2s purified from mouse bone marrow in a manner that was dependent on signaling through the PGI2 receptor IP. In a mouse model of 4 consecutive days of airway challenge with an extract of A. alternata, a fungal aeroallergen associated with severe asthma exacerbations, endogenous PGI2 signaling significantly inhibited lung IL-5 and IL-13 protein expression, and reduced the number of lung IL-5- and IL-13-expressing ILC2s, as well as the mean fluorescence intensity of IL-5 and IL-13 staining. In addition, exogenous administration of a PGI2 analog inhibited Alternaria extract-induced lung IL-5 and IL-13 protein expression, and reduced the number of lung IL-5- and IL-13-expressing ILC2s and the mean fluorescence intensity of IL-5 and IL-13 staining. Finally, a PGI2 analog inhibited IL-5 and IL-13 expression by human ILC2s that were stimulated with IL-2 and IL-33. CONCLUSIONS: These results suggest that PGI2 may be a potential therapy to reduce the ILC2 response to protease-containing aeroallergens, such as Alternaria.


Asunto(s)
Epoprostenol/fisiología , Linfocitos/fisiología , Transducción de Señal/fisiología , Alternaria/inmunología , Animales , Epoprostenol/análogos & derivados , Epoprostenol/farmacología , Humanos , Técnicas In Vitro , Interleucina-13/fisiología , Interleucina-33/farmacología , Interleucina-5/fisiología , Pulmón/citología , Pulmón/inmunología , Linfocitos/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Transducción de Señal/efectos de los fármacos
5.
Infect Immun ; 84(5): 1548-1555, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26953325

RESUMEN

γδ T cells are prevalent at mucosal and epithelial surfaces and are a critical first line of defense against bacterial and fungal pathogens. γδ17 cells are a subset of γδ T cells which, in the presence of IL-23 and IL-1ß, produce large quantities of interleukin-17A (IL-17A), a cytokine crucial to these cells' antibacterial and antifungal function. STAT6, an important transcription factor in Th2 differentiation and inhibition of Th1 differentiation, is expressed at high levels in the T cells of people with parasitic infections and asthma. Our group and others have shown that STAT6 attenuates IL-17A protein expression by CD4(+) T cells. By extension, we hypothesized that STAT6 activation also inhibits innate γδ17 cell cytokine secretion. We show here that γδ17 cells expressed the type I IL-4 receptor (IL-4R), and IL-4 increased STAT6 phosphorylation in γδ T cells. IL-4 inhibited γδ17 cell production of IL-17A. IL-4 also decreased γδ17 cell expression of IL-23R as well as Sgk1. To determine whether STAT6 signaling regulates γδ17 cell numbers in vivo, we used a model of Klebsiella pneumoniae in mice deficient in STAT6. We chose K. pneumoniae for our in vivo model, since K. pneumoniae increases IL-17A expression and γδ17 numbers. K. pneumoniae infection of STAT6 knockout mice resulted in a statistically significant increase in the number of γδ17 cells compared to that of wild-type mice. These studies are the first to demonstrate that γδ17 cells express the type I IL-4R and that STAT6 signaling negatively regulates γδ17 cells, a cell population that plays a front-line role in mucosal immunity.


Asunto(s)
Interleucina-17/metabolismo , Infecciones por Klebsiella/patología , Klebsiella pneumoniae/inmunología , Factor de Transcripción STAT6/metabolismo , Transducción de Señal , Subgrupos de Linfocitos T/inmunología , Células Th17/inmunología , Animales , Modelos Animales de Enfermedad , Femenino , Inmunidad Mucosa , Interleucina-4/metabolismo , Ratones Endogámicos BALB C , Ratones Noqueados , Receptores de Superficie Celular/metabolismo
6.
J Exp Med ; 215(1): 263-281, 2018 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-29222107

RESUMEN

Group 2 innate lymphoid cells (ILC2s) are effector cells within the mucosa and key participants in type 2 immune responses in the context of allergic inflammation and infection. ILC2s develop in the bone marrow from common lymphoid progenitor cells, but little is known about how ILC2s egress from the bone marrow for hematogenous trafficking. In this study, we identified a critical role for IL-33, a hallmark peripheral ILC2-activating cytokine, in promoting the egress of ILC2 lineage cells from the bone marrow. Mice lacking IL-33 signaling had normal development of ILC2s but retained significantly more ILC2 progenitors in the bone marrow via augmented expression of CXCR4. Intravenous injection of IL-33 or pulmonary fungal allergen challenge mobilized ILC2 progenitors to exit the bone marrow. Finally, IL-33 enhanced ILC2 trafficking to the lungs in a parabiosis mouse model of tissue disruption and repopulation. Collectively, these data demonstrate that IL-33 plays a critical role in promoting ILC2 egress from the bone marrow.


Asunto(s)
Células de la Médula Ósea/metabolismo , Inmunidad Innata , Interleucina-33/metabolismo , Subgrupos Linfocitarios/metabolismo , Alérgenos/inmunología , Animales , Antígenos Fúngicos/inmunología , Médula Ósea , Células de la Médula Ósea/citología , Células de la Médula Ósea/inmunología , Recuento de Células , Diferenciación Celular , Proteína 1 Similar al Receptor de Interleucina-1/deficiencia , Interleucina-33/genética , Subgrupos Linfocitarios/citología , Subgrupos Linfocitarios/inmunología , Ratones , Ratones Noqueados , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA