Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Allergy Clin Immunol ; 145(3): 818-833.e11, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31812575

RESUMEN

BACKGROUND: Idiopathic pulmonary fibrosis (IPF) is a disease with high 5-year mortality and few therapeutic options. Prostaglandin (PG) E2 exhibits antifibrotic properties and is reduced in bronchoalveolar lavage from patients with IPF. 15-Prostaglandin dehydrogenase (15-PGDH) is the key enzyme in PGE2 metabolism under the control of TGF-ß and microRNA 218. OBJECTIVE: We sought to investigate the expression of 15-PGDH in IPF and the therapeutic potential of a specific inhibitor of this enzyme in a mouse model and human tissue. METHODS: In vitro studies, including fibrocyte differentiation, regulation of 15-PGDH, RT-PCR, and Western blot, were performed using peripheral blood from healthy donors and patients with IPF and A549 cells. Immunohistochemistry, immunofluorescence, 15-PGDH activity assays, and in situ hybridization as well as ex vivo IPF tissue culture experiments were done using healthy donor and IPF lungs. Therapeutic effects of 15-PGDH inhibition were studied in the bleomycin mouse model of pulmonary fibrosis. RESULTS: We demonstrate that 15-PGDH shows areas of increased expression in patients with IPF. Inhibition of this enzyme increases PGE2 levels and reduces collagen production in IPF precision cut lung slices and in the bleomycin model. Inhibitor-treated mice show amelioration of lung function, decreased alveolar epithelial cell apoptosis, and fibroblast proliferation. Pulmonary fibrocyte accumulation is also decreased by inhibitor treatment in mice, similar to PGE2 that inhibits fibrocyte differentiation from blood of healthy donors and patients with IPF. Finally, microRNA 218-5p, which is downregulated in patients with IPF, suppressed 15-PGDH expression in vivo and in vitro. CONCLUSIONS: These findings highlight the role of 15-PGDH in IPF and suggest 15-PGDH inhibition as a promising therapeutic approach.


Asunto(s)
Hidroxiprostaglandina Deshidrogenasas/metabolismo , Fibrosis Pulmonar Idiopática/enzimología , MicroARNs/metabolismo , Animales , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Dinoprostona/metabolismo , Eicosanoides/metabolismo , Inhibidores Enzimáticos/farmacología , Regulación de la Expresión Génica , Humanos , Fibrosis Pulmonar Idiopática/patología , Ratones , Piridinas/farmacología , Tiofenos/farmacología
2.
J Allergy Clin Immunol ; 144(3): 764-776, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31082458

RESUMEN

BACKGROUND: Lung eosinophilia is a hallmark of asthma, and eosinophils are believed to play a crucial role in the pathogenesis of allergic inflammatory diseases. Short-chain fatty acids (SCFAs), such as acetate, propionate, and butyrate, are produced in high amounts in the gastrointestinal tract by commensal bacteria and can be absorbed into the bloodstream. Although there is recent evidence that SCFAs are beneficial in allergic asthma models, the effect on eosinophils has remained elusive. OBJECTIVE: The role of SCFAs was investigated in human eosinophil function and a mouse model of allergic asthma. METHODS: Eosinophils were purified from self-reported allergic or healthy donors. Migration, adhesion to the endothelium, and eosinophil survival were studied in vitro. Ca2+ flux, apoptosis, mitochondrial membrane potential, and expression of surface markers were determined by using flow cytometry and in part by using real-time PCR. Allergic airway inflammation was assessed in vivo in an ovalbumin-induced asthma model by using invasive spirometry. RESULTS: For the first time, we observed that SCFAs were able to attenuate human eosinophils at several functional levels, including (1) adhesion to the endothelium, (2) migration, and (3) survival. These effects were independent from GPR41 and GPR43 but were accompanied by histone acetylation and mimicked by trichostatin A, a pan-histone deacetylase inhibitor. In vivo butyrate ameliorated allergen-induced airway and lung eosinophilia, reduced type 2 cytokine levels in bronchial fluid, and improved airway hyperresponsiveness in mice. CONCLUSION: These in vitro and in vivo findings highlight the importance of SCFAs, especially butyrate as a promising therapeutic agent in allergic inflammatory diseases.


Asunto(s)
Antiinflamatorios/uso terapéutico , Asma/tratamiento farmacológico , Butiratos/farmacología , Butiratos/uso terapéutico , Eosinófilos/efectos de los fármacos , Eosinofilia Pulmonar/tratamiento farmacológico , Animales , Antiinflamatorios/farmacología , Apoptosis/efectos de los fármacos , Asma/genética , Asma/inmunología , Movimiento Celular/efectos de los fármacos , Eosinófilos/inmunología , Eosinófilos/fisiología , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Ratones Endogámicos BALB C , Eosinofilia Pulmonar/genética , Eosinofilia Pulmonar/inmunología
3.
Int J Cancer ; 142(1): 121-132, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-28875496

RESUMEN

The putative cannabinoid receptor GPR55 has been shown to play a tumor-promoting role in various cancers, and is involved in many physiological and pathological processes of the gastrointestinal (GI) tract. While the cannabinoid receptor 1 (CB1 ) has been reported to suppress intestinal tumor growth, the role of GPR55 in the development of GI cancers is unclear. We, therefore, aimed at elucidating the role of GPR55 in colorectal cancer (CRC), the third most common cancer worldwide. Using azoxymethane (AOM)- and dextran sulfate sodium (DSS)-driven CRC mouse models, we found that GPR55 plays a tumor-promoting role that involves alterations of leukocyte populations, i.e. myeloid-derived suppressor cells and T lymphocytes, within the tumor tissues. Concomitantly, expression levels of COX-2 and STAT3 were reduced in tumor tissue of GPR55 knockout mice, indicating reduced presence of tumor-promoting factors. By employing the experimental CRC models to CB1 knockout and CB1 /GPR55 double knockout mice, we can further show that GPR55 plays an opposing role to CB1 . We report that GPR55 and CB1 mRNA expression are differentially regulated in the experimental models and in a cohort of 86 CRC patients. Epigenetic methylation of CNR1 and GPR55 was also differentially regulated in human CRC tissue compared to control samples. Collectively, our data suggest that GPR55 and CB1 play differential roles in colon carcinogenesis where the former seems to act as oncogene and the latter as tumor suppressor.


Asunto(s)
Carcinogénesis/metabolismo , Neoplasias Colorrectales/patología , Receptores Acoplados a Proteínas G/metabolismo , Animales , Neoplasias Colorrectales/metabolismo , Humanos , Ratones , Ratones Noqueados , Receptor Cannabinoide CB1/metabolismo , Receptores de Cannabinoides/metabolismo
4.
J Allergy Clin Immunol ; 137(3): 833-43, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26792210

RESUMEN

BACKGROUND: Prostaglandin (PG) D2 is an early-phase mediator in inflammation, but its action and the roles of the 2 D-type prostanoid receptors (DPs) DP1 and DP2 (also called chemoattractant receptor-homologous molecule expressed on T(H)2 cells) in regulating macrophages have not been elucidated to date. OBJECTIVE: We investigated the role of PGD2 receptors on primary human macrophages, as well as primary murine lung macrophages, and their ability to influence neutrophil action in vitro and in vivo. METHODS: In vitro studies, including migration, Ca(2+) flux, and cytokine secretion, were conducted with primary human monocyte-derived macrophages and neutrophils and freshly isolated murine alveolar and pulmonary interstitial macrophages. In vivo pulmonary inflammation was assessed in male BALB/c mice. RESULTS: Activation of DP1, DP2, or both receptors on human macrophages induced strong intracellular Ca(2+) flux, cytokine release, and migration of macrophages. In a murine model of LPS-induced pulmonary inflammation, activation of each PGD2 receptor resulted in aggravated airway neutrophilia, tissue myeloperoxidase activity, cytokine contents, and decreased lung compliance. Selective depletion of alveolar macrophages abolished the PGD2-enhanced inflammatory response. Activation of PGD2 receptors on human macrophages enhanced the migratory capacity and prolonged the survival of neutrophils in vitro. In human lung tissue specimens both DP1 and DP2 receptors were located on alveolar macrophages along with hematopoietic PGD synthase, the rate-limiting enzyme of PGD2 synthesis. CONCLUSION: For the first time, our results show that PGD2 markedly augments disease activity through its ability to enhance the proinflammatory actions of macrophages and subsequent neutrophil activation.


Asunto(s)
Pulmón/inmunología , Pulmón/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Infiltración Neutrófila/inmunología , Receptores Inmunológicos/metabolismo , Receptores de Prostaglandina/metabolismo , Animales , Señalización del Calcio , Quimiotaxis de Leucocito/inmunología , Citocinas/biosíntesis , Endotoxinas/efectos adversos , Endotoxinas/inmunología , Expresión Génica , Humanos , Mediadores de Inflamación/metabolismo , Pulmón/patología , Lesión Pulmonar/etiología , Lesión Pulmonar/metabolismo , Lesión Pulmonar/patología , Macrófagos/efectos de los fármacos , Ratones , Neutrófilos/efectos de los fármacos , Neutrófilos/inmunología , Neutrófilos/metabolismo , Prostaglandina D2/farmacología , Receptores Inmunológicos/genética , Receptores de Prostaglandina/genética , Síndrome de Dificultad Respiratoria/etiología , Síndrome de Dificultad Respiratoria/metabolismo , Síndrome de Dificultad Respiratoria/patología , Factor de Necrosis Tumoral alfa/biosíntesis
5.
Eur J Immunol ; 45(5): 1548-59, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25645675

RESUMEN

Prostaglandin E2 (PGE2 ) protects against allergic responses via binding to prostanoid receptor EP4, which inhibits eosinophil migration in a PI3K/PKC-dependent fashion. The phosphoinositide-dependent protein kinase 1 (PDK1) is known to act as a downstream effector in PI3K signaling and has been implicated in the regulation of neutrophil migration. Thus, here we elucidate whether PDK1 mediates inhibitory effects of E-type prostanoid receptor 4 (EP4) receptors on eosinophil function. Therefore, eosinophils were isolated from human peripheral blood or differentiated from mouse BM. PDK1 signaling was investigated in shape change, chemotaxis, CD11b, respiratory burst, and Ca(2+) mobilization assays. The specific PDK1 inhibitors BX-912 and GSK2334470 prevented the inhibition by prostaglandin E2 and the EP4 agonist ONO-AE1-329. Depending on the cellular function, PDK1 seemed to act through PI3K-dependent or PI3K-independent mechanisms. Stimulation of EP4 receptors caused PDK1 phosphorylation at Ser396 and induced PI3K-dependent nuclear translocation of PDK1. EP4-induced inhibition of shape change and chemotaxis was effectively reversed by the Akt inhibitor triciribine. In support of this finding, ONO-AE1-329 induced a PI3K/PDK1-dependent increase in Akt phosphorylation. In conclusion, our data illustrate a critical role for PDK1 in transducing inhibitory signals on eosinophil effector function. Thus, our results suggest that PDK1 might serve as a novel therapeutic target in diseases involving eosinophilic inflammation.


Asunto(s)
Proteínas Quinasas Dependientes de 3-Fosfoinosítido/metabolismo , Eosinófilos/metabolismo , Proteínas Quinasas Dependientes de 3-Fosfoinosítido/antagonistas & inhibidores , Transporte Activo de Núcleo Celular , Animales , Antígeno CD11b/metabolismo , Señalización del Calcio , Forma de la Célula , Dinoprostona/metabolismo , Eosinófilos/efectos de los fármacos , Eosinófilos/inmunología , Humanos , Indazoles/farmacología , Éteres Metílicos/farmacología , Ratones , Ratones Endogámicos BALB C , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/metabolismo , Pirimidinas/farmacología , Subtipo EP4 de Receptores de Prostaglandina E/agonistas , Subtipo EP4 de Receptores de Prostaglandina E/metabolismo , Estallido Respiratorio , Ribonucleósidos/farmacología , Transducción de Señal/efectos de los fármacos
6.
J Immunol ; 193(2): 827-39, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-24929001

RESUMEN

Proresolution functions were reported for PGD2 in colitis, but the role of its two receptors, D-type prostanoid (DP) and, in particular, chemoattractant receptor homologous molecule expressed on Th2 cells (CRTH2), is less well defined. We investigated DP and CRTH2 expression and function during human and murine ulcerative colitis (UC). Expression of receptors was measured by flow cytometry on peripheral blood leukocytes and by immunohistochemistry and immunoblotting in colon biopsies of patients with active UC and healthy individuals. Receptor involvement in UC was evaluated in a mouse model of dextran sulfate sodium colitis. DP and CRTH2 expression changed in leukocytes of patients with active UC in a differential manner. In UC patients, DP showed higher expression in neutrophils but lower in monocytes as compared with control subjects. In contrast, CRTH2 was decreased in eosinophils, NK, and CD3(+) T cells but not in monocytes and CD3(+)/CD4(+) T cells. The decrease of CRTH2 on blood eosinophils clearly correlated with disease activity. DP correlated positively with disease activity in eosinophils but inversely in neutrophils. CRTH2 internalized upon treatment with PGD2 and 11-dehydro TXB2 in eosinophils of controls. Biopsies of UC patients revealed an increase of CRTH2-positive cells in the colonic mucosa and high CRTH2 protein content. The CRTH2 antagonist CAY10595 improved, whereas the DP antagonist MK0524 worsened inflammation in murine colitis. DP and CRTH2 play differential roles in UC. Although expression of CRTH2 on blood leukocytes is downregulated in UC, CRTH2 is present in colon tissue, where it may contribute to inflammation, whereas DP most likely promotes anti-inflammatory actions.


Asunto(s)
Colitis Ulcerosa/metabolismo , Colon/metabolismo , Receptores Inmunológicos/metabolismo , Receptores de Prostaglandina/metabolismo , Adolescente , Adulto , Animales , Western Blotting , Complejo CD3/metabolismo , Colitis/inducido químicamente , Colitis/metabolismo , Colon/efectos de los fármacos , Colon/patología , Sulfato de Dextran , Femenino , Citometría de Flujo , Humanos , Inmunohistoquímica , Indoles/farmacología , Células Asesinas Naturales/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Neutrófilos/metabolismo , Prostaglandina D2/metabolismo , Receptores Inmunológicos/antagonistas & inhibidores , Receptores de Prostaglandina/antagonistas & inhibidores , Linfocitos T/metabolismo , Células Th2/metabolismo , Adulto Joven
7.
Pharmacology ; 96(3-4): 137-43, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26228833

RESUMEN

Platelets express the EP2, EP3 and EP4 receptors. Prostaglandin (PG) E2 has a biphasic effect on platelets. Low concentrations of PGE2 enhance platelet aggregation through the activation of the EP3 receptors, while at high concentrations it attenuates aggregation via the EP4 receptor. Consequently, EP3 receptor inhibition was shown to inhibit artherothrombosis, but had no influence on bleeding time in vivo. In this study, we investigated the role of the EP3 receptor in adhesion and thrombus formation under flow conditions in vitro. The EP3 agonist sulprostone caused an increase in the adhesion of washed platelets to fibrinogen as well as to collagen under low shear stress, an effect that was blocked by the EP3 antagonist L-798106. In contrast, when whole blood was perfused over collagen-coated surfaces, sulprostone did not enhance binding and thrombus formation of platelets on collagen; at high concentrations it even attenuated this response. We conclude that in more physiological models of thrombus formation, the role for EP3 receptors is limited, indirectly suggesting that the primary action of PGE2 in haemostasis might be an inhibitory one.


Asunto(s)
Plaquetas/metabolismo , Dinoprostona/análogos & derivados , Adhesividad Plaquetaria/efectos de los fármacos , Subtipo EP3 de Receptores de Prostaglandina E/agonistas , Trombosis/sangre , Plaquetas/efectos de los fármacos , Colágeno/farmacología , Dinoprostona/farmacología , Fibrinógeno/farmacología , Humanos , Técnicas In Vitro , Agregación Plaquetaria/efectos de los fármacos , Sulfonamidas/farmacología , Trombosis/inducido químicamente
9.
Kidney Int ; 86(5): 923-31, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24940796

RESUMEN

The dramatic cardiovascular mortality of patients with chronic kidney disease is attributable in a significant proportion to endothelial dysfunction. Cyanate, a reactive species in equilibrium with urea, is formed in excess in chronic kidney disease. Cyanate is thought to have a causal role in promoting cardiovascular disease, but the underlying mechanisms remain unclear. Immunohistochemical analysis performed in the present study revealed that carbamylated epitopes associate mainly with endothelial cells in human atherosclerotic lesions. Cyanate treatment of human coronary artery endothelial cells reduced expression of endothelial nitric oxide synthase, and increased tissue factor and plasminogen activator inhibitor-1 expression. In mice, administration of cyanate, promoting protein carbamylation at levels observed in uremic patients, attenuated arterial vasorelaxation of aortic rings in response to acetylcholine without affecting the sodium nitroprusside-induced relaxation. Total endothelial nitric oxide synthase and nitric oxide production were significantly reduced in aortic tissue of cyanate-treated mice. This coincided with a marked increase of tissue factor and plasminogen activator inhibitor-1 protein levels in aortas of cyanate-treated mice. Thus, cyanate compromises endothelial functionality in vitro and in vivo. This may contribute to the dramatic cardiovascular risk of patients suffering from chronic kidney disease.


Asunto(s)
Aorta/efectos de los fármacos , Cianatos/farmacología , Células Endoteliales/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Vasodilatación/efectos de los fármacos , Administración por Inhalación , Animales , Aorta/metabolismo , Aorta/fisiopatología , Células Cultivadas , Citrulina/análogos & derivados , Citrulina/metabolismo , Cianatos/administración & dosificación , Relación Dosis-Respuesta a Droga , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Endotelio Vascular/fisiopatología , Humanos , Masculino , Ratones Endogámicos C57BL , Óxido Nítrico Sintasa de Tipo III/genética , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fenotipo , Inhibidor 1 de Activador Plasminogénico/genética , Inhibidor 1 de Activador Plasminogénico/metabolismo , Procesamiento Proteico-Postraduccional , ARN Mensajero/metabolismo , Tromboplastina/metabolismo , Factores de Tiempo , Vasodilatadores/farmacología
10.
Biochem Biophys Res Commun ; 450(4): 1643-9, 2014 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-25044109

RESUMEN

The serum amyloid A (SAA) family of proteins is encoded by multiple genes, which display allelic variation and a high degree of homology in mammals. The SAA1/2 genes code for non-glycosylated acute-phase SAA1/2 proteins, that may increase up to 1000-fold during inflammation. The SAA4 gene, well characterized in humans (hSAA4) and mice (mSaa4) codes for a SAA4 protein that is glycosylated only in humans. We here report on a previously uncharacterized SAA4 gene (rSAA4) and its product in Rattus norvegicus, the only mammalian species known not to express acute-phase SAA. The exon/intron organization of rSAA4 is similar to that reported for hSAA4 and mSaa4. By performing 5'- and 3'RACE, we identified a 1830-bases containing rSAA4 mRNA (including a GA-dinucleotide tandem repeat). Highest rSAA4 mRNA expression was detected in rat liver. In McA-RH7777 rat hepatoma cells, rSAA4 transcription was significantly upregulated in response to LPS and IL-6 while IL-1α/ß and TNFα were without effect. Luciferase assays with promoter-truncation constructs identified three proximal C/EBP-elements that mediate expression of rSAA4 in McA-RH7777 cells. In line with sequence prediction a 14-kDa non-glycosylated SAA4 protein is abundantly expressed in rat liver. Fluorescence microscopy revealed predominant localization of rSAA4-GFP-tagged fusion protein in the ER.


Asunto(s)
Proteína Amiloide A Sérica/metabolismo , Animales , Línea Celular Tumoral , Hígado/metabolismo , Reacción en Cadena de la Polimerasa , Regiones Promotoras Genéticas , Ratas , Ratas Sprague-Dawley , Proteína Amiloide A Sérica/química , Proteína Amiloide A Sérica/genética
12.
Pharmacology ; 94(5-6): 280-6, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25531811

RESUMEN

Prostaglandin (PG) E2 has been implicated in the pathogenesis of aspirin-exacerbated respiratory disease (AERD). E-type prostanoid (EP) receptor 4 is known to confer inhibitory signals to eosinophils and monocytes, amongst others. In this study, we investigated whether the responsiveness of eosinophils and monocytes to PGE2 and EP4 receptor activation is altered in AERD patients. While the expression of the EP4 receptor in eosinophils was unaltered in AERD patients, inhibition of eosinophil chemotaxis by PGE2 or the EP4 agonist CAY10598 was less pronounced in AERD patients as compared to healthy control subjects. In monocytes, we found no changes in basal or lipopolysaccharide (LPS)-stimulated PGE2 synthesis, but the response to EP4 receptor activation with respect to inhibition of LPS-induced tumor necrosis factor-α release was reduced in AERD patients, especially in the presence of aspirin (acetylsalicylic acid). Our data point towards a decreased sensitivity of inhibitory EP4 receptor that may play a role in AERD.


Asunto(s)
Aspirina/efectos adversos , Asma/inducido químicamente , Asma/metabolismo , Eosinófilos/metabolismo , Monocitos/metabolismo , Subtipo EP4 de Receptores de Prostaglandina E/metabolismo , Adulto , Anciano , Aspirina/farmacología , Calcio/metabolismo , Movimiento Celular , Dinoprostona/metabolismo , Dinoprostona/farmacología , Eosinófilos/fisiología , Femenino , Humanos , Lipopolisacáridos/farmacología , Masculino , Persona de Mediana Edad , Pirrolidinonas/farmacología , Subtipo EP4 de Receptores de Prostaglandina E/agonistas , Tetrazoles/farmacología , Factor de Necrosis Tumoral alfa/metabolismo
13.
J Allergy Clin Immunol ; 131(2): 532-40.e1-2, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22704539

RESUMEN

BACKGROUND: Increased vascular permeability is a fundamental characteristic of inflammation. Substances that are released during inflammation, such as prostaglandin (PG) E(2), can counteract vascular leakage, thereby hampering tissue damage. OBJECTIVE: In this study we investigated the role of PGE(2) and its receptors in the barrier function of human pulmonary microvascular endothelial cells and in neutrophil trafficking. METHODS: Endothelial barrier function was determined based on electrical impedance measurements. Neutrophil recruitment was assessed based on adhesion and transendothelial migration. Morphologic alterations are shown by using immunofluorescence microscopy. RESULTS: We observed that activation of E-type prostanoid (EP) 4 receptor by PGE(2) or an EP4-selective agonist (ONO AE1-329) enhanced the barrier function of human microvascular lung endothelial cells. EP4 receptor activation prompted similar responses in pulmonary artery and coronary artery endothelial cells. These effects were reversed by an EP4 antagonist (ONO AE3-208), as well as by blocking actin polymerization with cytochalasin B. The EP4 receptor-induced increase in barrier function was independent of the classical cyclic AMP/protein kinase A signaling machinery, endothelial nitric oxide synthase, and Rac1. Most importantly, EP4 receptor stimulation showed potent anti-inflammatory activities by (1) facilitating wound healing of pulmonary microvascular endothelial monolayers, (2) preventing junctional and cytoskeletal reorganization of activated endothelial cells, and (3) impairing neutrophil adhesion to endothelial cells and transendothelial migration. The latter effects could be partially attributed to reduced E-selectin expression after EP4 receptor stimulation. CONCLUSION: These data indicate that EP4 agonists as anti-inflammatory agents represent a potential therapy for diseases with increased vascular permeability and neutrophil extravasation.


Asunto(s)
Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Neutrófilos/metabolismo , Subtipo EP4 de Receptores de Prostaglandina E/metabolismo , Actinas/metabolismo , Permeabilidad Capilar/efectos de los fármacos , Permeabilidad Capilar/fisiología , Adhesión Celular/efectos de los fármacos , Adhesión Celular/fisiología , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Células Cultivadas , Vasos Coronarios/efectos de los fármacos , Vasos Coronarios/metabolismo , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Citocalasina B/farmacología , Dinoprostona/metabolismo , Células Endoteliales/efectos de los fármacos , Células Endoteliales/inmunología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/inmunología , Humanos , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Éteres Metílicos/farmacología , Microvasos/efectos de los fármacos , Microvasos/inmunología , Microvasos/metabolismo , Naftalenos/farmacología , Neutrófilos/efectos de los fármacos , Neutrófilos/inmunología , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fenilbutiratos/farmacología , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/metabolismo , Cicatrización de Heridas/efectos de los fármacos , Cicatrización de Heridas/fisiología , Proteína de Unión al GTP rac1/metabolismo
14.
Eur J Immunol ; 41(8): 2379-89, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21681739

RESUMEN

Eosinophil extravasation across the endothelium is a key feature of allergic inflammation. Here, we investigated the role of PGE(2) and its receptor, E-type prostanoid receptor (EP)-4, in the regulation of eosinophil interaction with human pulmonary microvascular endothelial cells. PGE(2) and the EP4 receptor agonist ONO AE1-329 significantly reduced eotaxin-induced eosinophil adhesion to fibronectin, and formation of filamentous actin and gelsolin-rich adhesive structures. These inhibitory effects were reversed by a selective EP4 receptor antagonist, ONO AE3-208. PGE(2) and the EP4 agonist prevented the activation and cell-surface clustering of ß2 integrins, and L-selectin shedding of eosinophils. Under physiological flow conditions, eosinophils that were treated with the EP4 agonist showed reduced adhesion to endothelial monolayers upon stimulation with eotaxin, as well as after TNF-α-induced activation of the endothelial cells. Selective activation of EP1, EP2, and EP3 receptors did not alter eosinophil adhesion to endothelial cells, whereas the EP4 antagonist prevented PGE(2) from decreasing eosinophil adhesion. Finally, eosinophil transmigration across thrombin- and TNF-α-activated endothelial cells was effectively reduced by the EP4 agonist. These data suggest that PGE(2) -EP4 signaling might be protective against allergic responses by inhibiting the interaction of eosinophils with the endothelium and might hence be a useful therapeutic option for controlling inappropriate eosinophil infiltration.


Asunto(s)
Comunicación Celular , Células Endoteliales/metabolismo , Eosinófilos/metabolismo , Subtipo EP4 de Receptores de Prostaglandina E/metabolismo , Antígenos CD18/metabolismo , Adhesión Celular/efectos de los fármacos , Línea Celular , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Quimiocina CCL11/farmacología , Ciclopentanos/farmacología , Dinoprostona/farmacología , Células Endoteliales/citología , Eosinófilos/citología , Fibronectinas/metabolismo , Citometría de Flujo , Gelsolina/metabolismo , Humanos , Selectina L/metabolismo , Éteres Metílicos , Microscopía Confocal , Naftalenos/farmacología , Fenilbutiratos/farmacología , Subtipo EP1 de Receptores de Prostaglandina E/metabolismo , Subtipo EP2 de Receptores de Prostaglandina E/metabolismo , Subtipo EP3 de Receptores de Prostaglandina E/metabolismo , Subtipo EP4 de Receptores de Prostaglandina E/agonistas , Subtipo EP4 de Receptores de Prostaglandina E/antagonistas & inhibidores , Tioglicolatos/farmacología , Trombina/farmacología , Factor de Necrosis Tumoral alfa/farmacología
15.
Cell Mol Life Sci ; 68(21): 3573-87, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21365278

RESUMEN

Accumulation of eosinophils in tissue is a hallmark of allergic inflammation. Here we observed that a selective agonist of the PGE(2) receptor EP4, ONO AE1-329, potently attenuated the chemotaxis of human peripheral blood eosinophils, upregulation of the adhesion molecule CD11b and the production of reactive oxygen species. These effects were accompanied by the inhibition of cytoskeletal rearrangement and Ca(2+) mobilization. The involvement of the EP4 receptor was substantiated by a selective EP4 antagonist, which reversed the inhibitory effects of PGE(2) and the EP4 agonist. Selective kinase inhibitors revealed that the inhibitory effect of EP4 stimulation on eosinophil migration depended upon activation of PI 3-kinase and PKC, but not cAMP. Finally, we found that EP4 receptors are expressed by human eosinophils, and are also present on infiltrating leukocytes in inflamed human nasal mucosa. These data indicate that EP4 agonists might be a novel therapeutic option in eosinophilic diseases.


Asunto(s)
Regulación hacia Abajo , Eosinófilos/metabolismo , Subtipo EP4 de Receptores de Prostaglandina E/metabolismo , Antígeno CD11b/metabolismo , Calcio/metabolismo , Canales de Calcio/química , Canales de Calcio/metabolismo , Movimiento Celular , Ciclopentanos/farmacología , Dinoprostona/farmacología , Eosinófilos/efectos de los fármacos , Eosinófilos/inmunología , Humanos , Inflamación/metabolismo , Inflamación/patología , Isoindoles/farmacología , Éteres Metílicos , Mucosa Nasal/metabolismo , Mucosa Nasal/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteína Quinasa C/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Especies Reactivas de Oxígeno/metabolismo , Subtipo EP4 de Receptores de Prostaglandina E/agonistas , Subtipo EP4 de Receptores de Prostaglandina E/antagonistas & inhibidores , Sulfonamidas/farmacología , Tioglicolatos/farmacología
16.
J Neurochem ; 119(5): 1016-28, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21951135

RESUMEN

Currently, little is known about the role of intracellular triacylglycerol (TAG) lipases in the brain. Adipose triglyceride lipase (ATGL) is encoded by the PNPLA2 gene and catalyzes the rate-limiting step of lipolysis. In this study, we investigated the effects of ATGL deficiency on brain lipid metabolism in vivo using an established knock-out mouse model (ATGL-ko). A moderate decrease in TAG hydrolase activity detected in ATGL-ko versus wild-type brain tissue was accompanied by a 14-fold increase in TAG levels and an altered composition of TAG-associated fatty acids in ATGL-ko brains. Oil Red O staining revealed a severe accumulation of neutral lipids associated to cerebrovascular cells and in distinct brain regions namely the ependymal cell layer and the choroid plexus along the ventricular system. In situ hybridization histochemistry identified ATGL mRNA expression in ependymal cells, the choroid plexus, pyramidal cells of the hippocampus, and the dentate gyrus. Our findings imply that ATGL is involved in brain fatty acid metabolism, particularly in regions mediating transport and exchange processes: the brain-CSF interface, the blood-CSF barrier, and the blood-brain barrier.


Asunto(s)
Barrera Hematoencefálica/enzimología , Encéfalo/enzimología , Lipasa/fisiología , Metabolismo de los Lípidos , Animales , Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Femenino , Lipasa/deficiencia , Lipasa/genética , Metabolismo de los Lípidos/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Triglicéridos/metabolismo
17.
Arterioscler Thromb Vasc Biol ; 30(12): 2416-23, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21071691

RESUMEN

OBJECTIVE: Low concentrations of prostaglandin (PG) E(2) enhance platelet aggregation, whereas high concentrations inhibit it. The effects of PGE(2) are mediated through 4 G protein-coupled receptors, termed E-type prostaglindin (EP) receptor EP1, EP2, EP3, and EP4. The platelet-stimulating effect of PGE(2) has been suggested to involve EP3 receptors. Here we analyzed the receptor usage relating to the inhibitory effect of PGE(2). METHODS AND RESULTS: Using flow cytometry, we found that human platelets expressed EP4 receptor protein. A selective EP4 agonist (ONO AE1-329) potently inhibited the platelet aggregation as induced by ADP or collagen. This effect could be completely reversed by an EP4 antagonist, but not by PGI(2), PGD(2), and thromboxane receptor antagonists or cyclooxygenase inhibition. Moreover, an EP4 antagonist enhanced the PGE(2)-induced stimulation of platelet aggregation, indicating a physiological antiaggregatory activity of EP4 receptors. The inhibitory effect of the EP4 agonist was accompanied by attenuated Ca(2+) flux, inhibition of glycoprotein IIb/IIIa, and downregulation of P-selectin. Most importantly, adhesion of platelets to fibrinogen under flow and in vitro thrombus formation were effectively prevented by the EP4 agonist. In this respect, the EP4 agonist synergized with acetylsalicylic acid. CONCLUSIONS: These results are suggestive of EP4 receptor activation as a novel antithrombotic strategy.


Asunto(s)
Coagulación Sanguínea , Plaquetas/metabolismo , Dinoprostona/metabolismo , Agregación Plaquetaria , Subtipo EP4 de Receptores de Prostaglandina E/metabolismo , Trombosis/prevención & control , Aspirina/farmacología , Coagulación Sanguínea/efectos de los fármacos , Plaquetas/efectos de los fármacos , Calcio/metabolismo , Relación Dosis-Respuesta a Droga , Fibrinógeno/metabolismo , Fibrinolíticos/farmacología , Citometría de Flujo , Humanos , Éteres Metílicos/farmacología , Naftalenos/farmacología , Selectina-P/metabolismo , Fenilbutiratos/farmacología , Adhesividad Plaquetaria , Agregación Plaquetaria/efectos de los fármacos , Inhibidores de Agregación Plaquetaria/farmacología , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Subtipo EP4 de Receptores de Prostaglandina E/efectos de los fármacos , Trombosis/sangre
18.
J Allergy Clin Immunol ; 125(5): 1105-13, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20153037

RESUMEN

BACKGROUND: Enhanced eosinophil migration from the blood into the tissue is a hallmark of allergic diseases. Prostaglandin (PG) I(2) is the major prostanoid released by endothelial cells. Mice deficient in PGI(2) receptors (IPs) show exaggerated eosinophilic inflammation in response to allergen. OBJECTIVE: We set out to determine the role of PGI(2) in eosinophil trafficking. METHODS: Human lung microvascular endothelial cells and purified human eosinophils were used to study adhesion and transendothelial migration. Morphologic studies were performed with fluorescence microscopy. RESULTS: PGI(2) markedly attenuated the migration of eosinophils through cell-free filters but had no effect on neutrophil migration. The inhibitory effect of PGI(2) on eosinophils was prevented by the IP antagonist Cay10441 and the adenylyl cyclase inhibitor SQ22536. Similarly, PGI(2) prevented the adhesion of eosinophils to fibronectin and the rapid upregulation and activation of the adhesion molecule CD11b. IP expression on eosinophils was confirmed by means of flow cytometry and Western blotting. Furthermore, when endothelial cells were treated with the COX inhibitor diclofenac to abolish PGI(2) production, adhesion of eosinophils to endothelial monolayers and subsequent transendothelial migration were markedly enhanced. Similarly, the IP antagonist enhanced eosinophil adhesion to endothelial cells. Inhibition of PGI(2) biosynthesis decreased the electrical resistance of endothelial monolayers and compromised the texture of adherent junctions, as visualized by means of VE-cadherin and F-actin staining. CONCLUSION: We propose that endothelium-derived PGI(2) might be fundamental for the maintenance of the endothelial barrier function against infiltrating cells. These results suggest that selective IP agonists might have beneficial effects in allergic inflammation.


Asunto(s)
Quimiotaxis de Leucocito/efectos de los fármacos , Células Endoteliales/metabolismo , Endotelio Vascular/citología , Eosinófilos/fisiología , Epoprostenol/farmacología , Adhesión Celular/efectos de los fármacos , Células Cultivadas , Células Endoteliales/citología , Endotelio Vascular/metabolismo , Eosinófilos/efectos de los fármacos , Epoprostenol/metabolismo , Humanos , Pulmón/irrigación sanguínea , Pulmón/citología , Pulmón/inmunología
19.
J Immunol ; 181(10): 7273-83, 2008 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-18981149

RESUMEN

The accumulation of eosinophils in lung tissue is a hallmark of asthma, and it is believed that eosinophils play a crucial pathogenic role in allergic inflammation. Prostaglandin (PG) E(2) exerts anti-inflammatory and bronchoprotective mechanisms in asthma, but the underlying mechanisms have remained unclear. In this study we show that PGE(2) potently inhibits the chemotaxis of purified human eosinophils toward eotaxin, PGD(2), and C5a. Activated monocytes similarly attenuated eosinophil migration, and this was reversed after pretreatment of the monocytes with a cyclooxygenase inhibitor. The selective E-prostanoid (EP) 2 receptor agonist butaprost mimicked the inhibitory effect of PGE(2) on eosinophil migration, whereas an EP2 antagonist completely prevented this effect. Butaprost, and also PGE(2), inhibited the C5a-induced degranulation of eosinophils. Moreover, selective kinase inhibitors revealed that the inhibitory effect of PGE(2) on eosinophil migration depended upon activation of PI3K and protein kinase C, but not cAMP. In animal models, the EP2 agonist butaprost inhibited the rapid mobilization of eosinophils from bone marrow of the in situ perfused guinea pig hind limb and prevented the allergen-induced bronchial accumulation of eosinophils in OVA-sensitized mice. Immunostaining showed that human eosinophils express EP2 receptors and that EP2 receptor expression in the murine lungs is prominent in airway epithelium and, after allergen challenge, in peribronchial infiltrating leukocytes. In summary, these data show that EP2 receptor agonists potently inhibit eosinophil trafficking and activation and might hence be a useful therapeutic option in eosinophilic diseases.


Asunto(s)
Quimiotaxis de Leucocito/inmunología , Dinoprostona/inmunología , Eosinófilos/inmunología , Receptores de Prostaglandina E/inmunología , Alprostadil/análogos & derivados , Alprostadil/farmacología , Animales , Células Cultivadas , Quimiotaxis de Leucocito/efectos de los fármacos , Dinoprostona/metabolismo , Inhibidores Enzimáticos/farmacología , Eosinófilos/efectos de los fármacos , Eosinófilos/metabolismo , Cobayas , Humanos , Inmunohistoquímica , Ratones , Fosfatidilinositol 3-Quinasas/inmunología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteína Quinasa C/inmunología , Proteína Quinasa C/metabolismo , Receptores de Prostaglandina E/metabolismo , Subtipo EP2 de Receptores de Prostaglandina E
20.
J Leukoc Biol ; 85(1): 136-45, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18835884

RESUMEN

The major mast cell product PGD2 is released during the allergic response and stimulates the chemotaxis of eosinophils, basophils, and Th2-type T lymphocytes. The chemoattractant receptor homologous molecule of Th2 cells (CRTH2) has been shown to mediate the chemotactic effect of PGD2. PGH2 is the common precursor of all PGs and is produced by several cells that express cyclooxygenases. In this study, we show that PGH2 selectively stimulates human peripheral blood eosinophils and basophils but not neutrophils, and this effect is prevented by the CRTH2 receptor antagonist (+)-3-[[(4-fluorophenyl)sulfonyl] methyl amino]-1,2,3,4-tetrahydro-9H-carbazole-9-acetic acid (Cay10471) but not by the hematopoietic PGD synthase inhibitor 4-benzhydryloxy-1-[3-(1H-tetrazol-5-yl)-propyl]piperidine (HQL79). In chemotaxis assays, eosinophils showed a pronounced migratory response toward PGH2, but eosinophil degranulation was inhibited by PGH2. Moreover, collagen-induced platelet aggregation was inhibited by PGH2 in platelet-rich plasma, which was abrogated in the presence of the D-type prostanoid (DP) receptor antagonist 3-[(2-cyclohexyl-2-hydroxyethyl)amino]-2,5-dioxo-1-(phenylmethyl)-4-imidazolidine-heptanoic acid (BWA868c). Each of these effects of PGH2 was enhanced in the presence of plasma and/or albumin. In eosinophils, PGH2-induced calcium ion (Ca2+) flux was subject to homologous desensitization with PGD2. Human embryo kidney (HEK)293 cells transfected with human CRTH2 or DP likewise responded with Ca2+ flux, and untransfected HEK293 cells showed no response. These data indicate that PGH2 causes activation of the PGD2 receptors CRTH2 and DP via a dual mechanism: by interacting directly with the receptors and/or by giving rise to PGD2 after catalytic conversion by plasma proteins.


Asunto(s)
Quimiotaxis , Eosinófilos/fisiología , Prostaglandina H2/fisiología , Receptores Inmunológicos/fisiología , Receptores de Prostaglandina/fisiología , Basófilos/efectos de los fármacos , Basófilos/fisiología , Proteínas Sanguíneas/farmacología , Proteínas Sanguíneas/fisiología , Calcio/fisiología , Carbazoles/farmacología , Cationes Bivalentes , Degranulación de la Célula , Línea Celular , Colágeno/metabolismo , Eosinófilos/efectos de los fármacos , Humanos , Hidantoínas/farmacología , Oxidorreductasas Intramoleculares/antagonistas & inhibidores , Oxidorreductasas Intramoleculares/metabolismo , Lipocalinas/antagonistas & inhibidores , Lipocalinas/metabolismo , Neutrófilos/efectos de los fármacos , Neutrófilos/fisiología , Piperidinas/farmacología , Prostaglandina H2/farmacología , Receptores Inmunológicos/agonistas , Receptores Inmunológicos/antagonistas & inhibidores , Receptores de Prostaglandina/agonistas , Receptores de Prostaglandina/antagonistas & inhibidores , Sulfonamidas/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA