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










Base de datos
Intervalo de año de publicación
2.
Am J Respir Crit Care Med ; 191(8): 914-23, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25632992

RESUMEN

RATIONALE: Patients with cystic fibrosis (CF) lung disease have chronic airway inflammation driven by disrupted balance of T-cell (Th17 and Th2) responses. Regulatory T cells (Tregs) dampen T-cell activation, but their role in CF is incompletely understood. OBJECTIVES: To characterize numbers, function, and clinical impact of Tregs in CF lung disease. METHODS: Tregs were quantified in peripheral blood and airway samples from patients with CF and from lung disease control patients without CF and healthy control subjects. The role of Pseudomonas aeruginosa and CF transmembrane conductance regulator (CFTR) in Treg regulation was analyzed by using in vitro and murine in vivo models. MEASUREMENTS AND MAIN RESULTS: Tregs were decreased in peripheral blood and airways of patients with CF compared with healthy controls or lung disease patients without CF and correlated positively with lung function parameters. Patients with CF with chronic P. aeruginosa infection had lower Tregs compared with patients with CF without P. aeruginosa infection. Genetic knockout, pharmacological inhibition, and P. aeruginosa infection studies showed that both P. aeruginosa and CFTR contributed to Treg dysregulation in CF. Functionally, Tregs from patients with CF or from Cftr(-/-) mice were impaired in suppressing conventional T cells, an effect that was enhanced by P. aeruginosa infection. The loss of Tregs in CF affected memory, but not naive Tregs, and manifested gradually with disease progression. CONCLUSIONS: Patients with CF who have chronic P. aeruginosa infection show an age-dependent, quantitative, and qualitative impairment of Tregs. Modulation of Tregs represents a novel strategy to rebalance T-cell responses, dampen inflammation, and ultimately improve outcomes for patients with infective CF lung disease.


Asunto(s)
Fibrosis Quística/complicaciones , Fibrosis Quística/inmunología , Infecciones por Pseudomonas/complicaciones , Linfocitos T Reguladores/inmunología , Adolescente , Adulto , Factores de Edad , Niño , Preescolar , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Adulto Joven
3.
Eur Respir J ; 44(6): 1608-15, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25142483

RESUMEN

Cystic fibrosis (CF) lung disease is characterised by chronic Pseudomonas aeruginosa infection and leukocyte infiltration. Chemokines recruit leukocytes to sites of infection. Gene expression analysis identified the chemokine CCL18 as upregulated in CF leukocytes. We hypothesised that CCL18 characterises infection and inflammation in patients with CF lung disease. Therefore, we quantified CCL18 protein levels in the serum and airway fluids of CF patients and healthy controls, and studied CCL18 protein production by airway cells ex vivo. These studies demonstrated that CCL18 levels were increased in the serum and airway fluids from CF patients compared with healthy controls. Within CF patients, CCL18 levels were increased in P. aeruginosa-infected CF patients. CCL18 levels in the airways, but not in serum, correlated with severity of pulmonary obstruction in CF. Airway cells isolated from P. aeruginosa-infected CF patients produced significantly higher amounts of CCL18 protein compared with airway cells from CF patients without P. aeruginosa infection or healthy controls. Collectively, these studies show that CCL18 levels characterise chronic P. aeruginosa infection and pulmonary obstruction in patients with CF. CCL18 may, thus, serve as a potential biomarker and therapeutic target in CF lung disease.


Asunto(s)
Quimiocinas CC/metabolismo , Fibrosis Quística/metabolismo , Leucocitos/metabolismo , Infecciones por Pseudomonas/metabolismo , Adolescente , Adulto , Estudios de Casos y Controles , Quimiocina CXCL1/inmunología , Quimiocina CXCL1/metabolismo , Quimiocina CXCL2/inmunología , Quimiocina CXCL2/metabolismo , Quimiocinas CC/inmunología , Niño , Fibrosis Quística/complicaciones , Fibrosis Quística/inmunología , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Interleucina-8/inmunología , Interleucina-8/metabolismo , Leucocitos/inmunología , Masculino , Infecciones por Pseudomonas/complicaciones , Infecciones por Pseudomonas/inmunología , Esputo/metabolismo , Adulto Joven
4.
J Biol Chem ; 289(30): 20516-25, 2014 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-24914212

RESUMEN

CXCL8 (IL-8) recruits and activates neutrophils through the G protein-coupled chemokine receptor CXCR1. We showed previously that elastase cleaves CXCR1 and thereby impairs antibacterial host defense. However, the molecular intracellular machinery involved in this process remained undefined. Here we demonstrate by using flow cytometry, confocal microscopy, subcellular fractionation, co-immunoprecipitation, and bioluminescence resonance energy transfer that combined α- and γ-secretase activities are functionally involved in elastase-mediated regulation of CXCR1 surface expression on human neutrophils, whereas matrix metalloproteases are dispensable. We further demonstrate that PAR-2 is stored in mobilizable compartments in neutrophils. Bioluminescence resonance energy transfer and co-immunoprecipitation studies showed that secretases, PAR-2, and CXCR1 colocalize and physically interact in a novel protease/secretase-chemokine receptor network. PAR-2 blocking experiments provided evidence that elastase increased intracellular presenilin-1 expression through PAR-2 signaling. When viewed in combination, these studies establish a novel functional network of elastase, secretases, and PAR-2 that regulate CXCR1 expression on neutrophils. Interfering with this network could lead to novel therapeutic approaches in neutrophilic diseases, such as cystic fibrosis or rheumatoid arthritis.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/metabolismo , Regulación de la Expresión Génica/fisiología , Neutrófilos/metabolismo , Elastasa Pancreática/metabolismo , Receptor PAR-2/metabolismo , Receptores de Interleucina-8A/biosíntesis , Artritis Reumatoide/metabolismo , Artritis Reumatoide/patología , Fibrosis Quística/metabolismo , Fibrosis Quística/patología , Femenino , Humanos , Masculino , Presenilina-1/metabolismo
5.
J Biol Chem ; 289(8): 5320-9, 2014 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-24398679

RESUMEN

Neutrophils represent the major fraction of circulating immune cells and are rapidly recruited to sites of infection and inflammation. The inflammasome is a multiprotein complex that regulates the generation of IL-1 family proteins. The precise subcellular localization and functionality of the inflammasome in human neutrophils are poorly defined. Here we demonstrate that highly purified human neutrophils express key components of the NOD-like receptor family, pyrin domain containing 3 (NLRP3), and absent in melanoma 2 (AIM2) inflammasomes, particularly apoptosis-associated speck-like protein containing a CARD (ASC), AIM2, and caspase-1. Subcellular fractionation and microscopic analyses further showed that inflammasome components were localized in the cytoplasm and also noncanonically in secretory vesicle and tertiary granule compartments. Whereas IL-1ß and IL-18 were expressed at the mRNA level and released as protein, highly purified neutrophils neither expressed nor released IL-1α at baseline or upon stimulation. Upon inflammasome activation, highly purified neutrophils released substantially lower levels of IL-1ß protein compared with partially purified neutrophils. Serine proteases and caspases were differentially involved in IL-1ß release, depending on the stimulus. Spontaneous activation of the NLRP3 inflammasome in neutrophils in vivo affected IL-1ß, but not IL-18 release. In summary, these studies show that human neutrophils express key components of the inflammasome machinery in distinct intracellular compartments and release IL-1ß and IL-18, but not IL-1α or IL-33 protein. Targeting the neutrophil inflammasome may represent a future therapeutic strategy to modulate neutrophilic inflammatory diseases, such as cystic fibrosis, rheumatoid arthritis, or sepsis.


Asunto(s)
Inflamasomas/metabolismo , Neutrófilos/metabolismo , Adulto , Proteínas Portadoras/metabolismo , Caspasas/metabolismo , Compartimento Celular , Perfilación de la Expresión Génica , Humanos , Inflamasomas/genética , Interleucinas/metabolismo , Espacio Intracelular/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR , Neutrófilos/ultraestructura , Transporte de Proteínas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Serina Proteasas/metabolismo , Fracciones Subcelulares/metabolismo
6.
J Biol Chem ; 287(23): 19409-17, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-22532562

RESUMEN

RNAs are capable of modulating immune responses by binding to specific receptors. Neutrophils represent the major fraction of circulating immune cells, but receptors and mechanisms by which neutrophils sense RNA are poorly defined. Here, we analyzed the mRNA and protein expression patterns and the subcellular localization of the RNA receptors RIG-I, MDA-5, TLR3, TLR7, and TLR8 in primary neutrophils and immortalized neutrophil-like differentiated HL-60 cells. Our results demonstrate that both neutrophils and differentiated HL-60 cells express RIG-I, MDA-5, and TLR8 at the mRNA and protein levels, whereas TLR3 and TLR7 are not expressed at the protein level. Subcellular fractionation, flow cytometry, confocal laser scanning microscopy, and immuno-transmission electron microscopy provided evidence that, besides the cytoplasm, RIG-I and MDA-5 are stored in secretory vesicles of neutrophils and showed that RIG-I and its ligand, 3p-RNA, co-localize at the cell surface without triggering neutrophil activation. In summary, this study demonstrates that neutrophils express a distinct pattern of RNA recognition receptors in a non-canonical way, which could have essential implications for future RNA-based therapeutics.


Asunto(s)
ARN Helicasas DEAD-box/metabolismo , Regulación de la Expresión Génica/fisiología , Activación Neutrófila/fisiología , ARN , Receptores de Ácido Retinoico/metabolismo , Receptores Toll-Like/metabolismo , ARN Helicasas DEAD-box/genética , Femenino , Células HL-60 , Humanos , Helicasa Inducida por Interferón IFIH1 , Masculino , Neutrófilos , Receptores de Ácido Retinoico/genética , Vesículas Secretoras/genética , Vesículas Secretoras/metabolismo , Receptores Toll-Like/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...