Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
J Immunol ; 196(9): 3854-64, 2016 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-27001955

RESUMEN

The prevalence of allergies, including rhinitis, eczema, and anaphylaxis, is rising dramatically worldwide. This increase is especially problematic in children who bear the greatest burden of this rising trend. Increasing evidence identifies neutrophils as primary perpetrators of the more severe and difficult to manage forms of inflammation. A newly recognized mechanism by which neutrophils are recruited during the early phase of histamine-induced inflammation involves the sphingosine kinase (SK)/sphingosine-1-phosphate axis. This study examines whether topical application of fingolimod, an established SK/sphingosine-1-phosphate antagonist already in clinical use to treat multiple sclerosis, may be repurposed to treat cutaneous inflammation. Using two mouse models of ear skin inflammation (histamine- and IgE-mediated passive cutaneous anaphylaxis) we topically applied fingolimod prophylactically, as well as after establishment of the inflammatory response, and examined ear swelling, SK activity, vascular permeability, leukocyte recruitment, and production of proinflammatory mediators. The present study reveals that when applied topically, fingolimod attenuates both immediate and late-phase responses to histamine with reduced extravasation of fluid, SK-1 activity, proinflammatory cytokine and chemokine production, and neutrophil influx and prevents ear swelling. Intravital microscopy demonstrates that histamine-induced neutrophil rolling and adhesion to the postcapillary venules in the mouse ears is significantly attenuated even after 24 h. More importantly, these effects are achievable even once inflammation is established. Translation into humans was also accomplished with epicutaneous application of fingolimod resolving histamine-induced and allergen-induced inflammatory reactions in forearm skin. Overall, this study demonstrates, to our knowledge for the first time, that fingolimod may be repurposed to treat cutaneous inflammation.


Asunto(s)
Dermatitis/tratamiento farmacológico , Clorhidrato de Fingolimod/uso terapéutico , Esclerosis Múltiple/tratamiento farmacológico , Neutrófilos/efectos de los fármacos , Fosfotransferasas (Aceptor de Grupo Alcohol)/antagonistas & inhibidores , Piel/efectos de los fármacos , Administración Tópica , Animales , Permeabilidad Capilar/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Citocinas/metabolismo , Modelos Animales de Enfermedad , Clorhidrato de Fingolimod/farmacología , Histamina/metabolismo , Humanos , Inmunoglobulina E/sangre , Mediadores de Inflamación/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/inmunología , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Piel/inmunología
2.
FASEB J ; 29(9): 3638-53, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25985799

RESUMEN

Sphingosine 1-phosphate (S1P) is a bioactive lipid that can function both extracellularly and intracellularly to mediate a variety of cellular processes. Using lipid affinity matrices and a radiolabeled lipid binding assay, we reveal that S1P directly interacts with the transcription factor peroxisome proliferator-activated receptor (PPAR)γ. Herein, we show that S1P treatment of human endothelial cells (ECs) activated a luciferase-tagged PPARγ-specific gene reporter by ∼12-fold, independent of the S1P receptors. More specifically, in silico docking, gene reporter, and binding assays revealed that His323 of the PPARγ ligand binding domain is important for binding to S1P. PPARγ functions when associated with coregulatory proteins, and herein we identify that peroxisome proliferator-activated receptor-γ coactivator 1 (PGC1)ß binds to PPARγ in ECs and their progenitors (nonadherent endothelial forming cells) and that the formation of this PPARγ:PGC1ß complex is increased in response to S1P. ECs treated with S1P selectively regulated known PPARγ target genes with PGC1ß and plasminogen-activated inhibitor-1 being increased, no change to adipocyte fatty acid binding protein 2 and suppression of CD36. S1P-induced in vitro tube formation was significantly attenuated in the presence of the PPARγ antagonist GW9662, and in vivo application of GW9662 also reduced vascular development in Matrigel plugs. Interestingly, activation of PPARγ by the synthetic ligand troglitazone also reduced tube formation in vitro and in vivo. To support this, Sphk1(-/-)Sphk2(+/-) mice, with low circulating S1P levels, demonstrated a similar reduction in vascular development. Taken together, our data reveal that the transcription factor, PPARγ, is a bona fide intracellular target for S1P and thus suggest that the S1P:PPARγ:PGC1ß complex may be a useful target to manipulate neovascularization.


Asunto(s)
Células Endoteliales de la Vena Umbilical Humana/metabolismo , Lisofosfolípidos/metabolismo , Neovascularización Fisiológica/fisiología , PPAR gamma/metabolismo , Receptores de Lisoesfingolípidos/metabolismo , Esfingosina/análogos & derivados , Animales , Antígenos CD36/genética , Antígenos CD36/metabolismo , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana/citología , Humanos , Lisofosfolípidos/genética , Ratones , Ratones Noqueados , PPAR gamma/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Inhibidor 1 de Activador Plasminogénico/genética , Inhibidor 1 de Activador Plasminogénico/metabolismo , Proteínas de Unión al ARN , Receptores de Lisoesfingolípidos/genética , Serpina E2/genética , Serpina E2/metabolismo , Esfingosina/genética , Esfingosina/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Células U937
3.
Am J Pathol ; 180(4): 1740-50, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22322303

RESUMEN

Leukocyte recruitment to sites of inflammation is critical for the development of acute allergic responses. Rapid P-selectin up-regulation by endothelial cells is a key promoter of leukocyte infiltration in response to mediators such as histamine. However, the mechanisms underpinning this process are still incompletely understood. We examined the role of the sphingosine kinase/sphingosine-1-phosphate (SK/S1P) pathway and showed that in human umbilical vein endothelial cells, histamine rapidly activates SK in an extracellular signal-regulated kinase (ERK) 1/2-dependent manner, concurrent with the induction of P-selectin expression. Histamine activated both SK-1 and SK-2 isoforms; inhibition of SK-1, but not SK-2, attenuated histamine-induced P-selectin up-regulation and neutrophil rolling in vitro. S1P receptor antagonists failed to prevent histamine-induced P-selectin expression, and exogenous S1P did not increase P-selectin expression, suggesting that S1P cell surface receptors are not involved in this process. Finally, the role of both SK-1 and SK-2 in histamine-induced leukocyte rolling in vivo was assessed using pharmacological and genetic methods. Consistent with the in vitro findings, mice pretreated with either sphingosine kinase inhibitor or fingolimod (FTY720) significantly attenuated histamine-induced leukocyte rolling in the cremaster muscle. Similarly, Sphk1(-/-) but not Sphk2(-/-) mice exhibited reduced histamine-induced leukocyte rolling. These findings demonstrate a key role for SK-1 in histamine-induced rapid P-selectin up-regulation and associated leukocyte rolling, and suggest that endothelial SK-1 is an important contributor to allergic inflammation.


Asunto(s)
Histamina/farmacología , Infiltración Neutrófila/efectos de los fármacos , Fosfotransferasas (Aceptor de Grupo Alcohol)/fisiología , Animales , Células Cultivadas , Inhibidores Enzimáticos/farmacología , Clorhidrato de Fingolimod , Hemodinámica/fisiología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/enzimología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Inmunosupresores/farmacología , Recuento de Leucocitos , Rodamiento de Leucocito/efectos de los fármacos , Rodamiento de Leucocito/fisiología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/fisiología , Ratones , Ratones Noqueados , Infiltración Neutrófila/fisiología , Selectina-P/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/antagonistas & inhibidores , Fosfotransferasas (Aceptor de Grupo Alcohol)/deficiencia , Fosfotransferasas (Aceptor de Grupo Alcohol)/farmacología , Glicoles de Propileno/farmacología , Receptores de Lisoesfingolípidos/fisiología , Esfingosina/análogos & derivados , Esfingosina/farmacología , Regulación hacia Arriba/efectos de los fármacos
4.
Am J Pathol ; 177(1): 436-46, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20522645

RESUMEN

Leukocyte recruitment plays a major role in the immune response to infectious pathogens, as well as during inflammatory and autoimmune disorders. The process of leukocyte extravasation from the blood requires a complex cascade of adhesive events between the leukocytes and the endothelium, including initial leukocyte rolling, adhesion, and finally transendothelial migration. Current research in this area aims to identify the key leukocyte subsets that initiate a given disease and to identify the trafficking molecule(s) that will most specifically inhibit those cells. Herein we demonstrate that tumor necrosis factor (TNF)alpha activates the integrin alpha(5)beta(1) without altering total expression levels of beta(1) integrin on human umbilical vein endothelial cells. Moreover, our studies suggest that TNFalpha-induced beta(1) activation is dependent on sphingosine kinase-1, but independent of the sphingosine-1-phosphate family of G protein-coupled receptors. We also show, using a parallel plate flow chamber assay, that neutrophil adhesion to TNFalpha-activated endothelium can be attenuated by blocking alpha(5)beta(1) or its ligand angiopoietin-2. These observations add new complexities that broaden the accepted concept of cellular trafficking with neutrophil adhesion to TNFalpha activated endothelial cells being sphingosine kinase-1, alpha(5)beta(1), and angiopoietin-2 dependent. Moreover, this work supports the notion that sphingosine kinase-1 may be the single target required for an effective broad spectrum approach to combat inflammation and immune disorders.


Asunto(s)
Adhesión Celular/efectos de los fármacos , Integrina alfa5beta1/metabolismo , Neutrófilos/efectos de los fármacos , Neutrófilos/fisiología , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Angiopoyetina 2/metabolismo , Animales , Línea Celular , Humanos , Ratones , Neutrófilos/citología
5.
Blood ; 113(9): 2108-17, 2009 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-19109558

RESUMEN

Circulating endothelial progenitor cells (EPCs) are incorporated into foci of neovascularization where they undergo differentiation to mature endothelial cells (ECs). We show here that the enzyme sphingosine kinase-1 (SK-1) regulates the rate and direction of EPC differentiation without effect on the hematopoietic compartment. EPCs have high levels of SK-1 activity, which diminishes with differentiation and is, at least partially, responsible for maintaining their EPC phenotype. EPCs from SK-1 knockout mice form more adherent EC units and acquire a mature EC phenotype more rapidly. Conversely, EPCs from mice overexpressing SK-1 in the EC compartment are retarded in their differentiation. Exogenous regulation of SK-1 levels in normal EPCs, by genetic and pharmacologic means, including the immunomodulating drug FTY720, recapitulates these effects on EC differentiation. SK-1 knockout mice have higher levels of circulating EPCs, an exaggerated response to erythropoietin-induced EPC mobilization, and, in a mouse model of kidney ischemia reperfusion injury, exhibit a recovery similar to that of ischemic mice administered exogenous EPCs. Thus, SK-1 is a critical player in EPC differentiation into EC pointing to the potential utility of SK-1 modifying agents in the specific manipulation of endothelial development and repair.


Asunto(s)
Diferenciación Celular/genética , Células Endoteliales/fisiología , Fosfotransferasas (Aceptor de Grupo Alcohol)/fisiología , Células Madre/fisiología , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/fisiología , Proliferación Celular , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Células Endoteliales/metabolismo , Ratones , Ratones Noqueados , Fenotipo , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Proproteína Convertasas/fisiología , Serina Endopeptidasas/fisiología , Células Madre/metabolismo
6.
Am J Pathol ; 175(5): 2217-25, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19815712

RESUMEN

Sphingosine kinase (SK)-1 promotes endothelial cell (EC) survival through the cell junction molecule CD31 (platelet endothelial cell adhesion molecule-1). The integrin alpha(v)beta(3) is also essential for EC survival; inhibition of alpha(v)beta(3) ligation promotes apoptosis. Herein we demonstrate that under basal conditions, SK-1, alpha(v)beta(3), and CD31 exist as a heterotrimeric complex. Under conditions that affect EC survival such as loss of contact with the extracellular matrix or growth factor activation, more of this heterotrimeric complex forms. Overexpression studies demonstrate a requirement for SK-1 phosphorylation at serine 225 for increased heterotrimeric complex formation, activation of alpha(v)beta(3), and EC survival signals, including Bcl-X and nuclear factor-kappaB pathways. Moreover, beta(3) integrin depletion confirmed the requirement for this heterotrimeric complex in SK-1-mediated EC survival. Thus, with alpha(v)beta(3) integrin being identifiable primarily on angiogenic ECs and SK-1 being highly expressed in tumors, targeting SK-1 may affect multiple survival pathways, and its inhibition may be highly efficacious in controlling pathological EC survival.


Asunto(s)
Supervivencia Celular/fisiología , Células Endoteliales/fisiología , Integrina alfaVbeta3/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Apoptosis/fisiología , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteína 11 Similar a Bcl2 , Células Cultivadas , Células Endoteliales/citología , Proteína-Tirosina Quinasas de Adhesión Focal/genética , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Adhesiones Focales/metabolismo , Humanos , Integrina alfaVbeta3/genética , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo
7.
PLoS One ; 13(11): e0206802, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30395653

RESUMEN

BACKGROUND: The pericardial fluid may be representative of the interstitium of the heart. The aim of this study was to discriminate in cardiovascular disease patients between adipocytokines that are produced locally by the heart and those supplied by the circulation. METHODS: Enzyme-linked immunosorbent assays (ELISA) were used to determine levels of N-terminal pro-brain natriuretic peptide (NT-pBNP), fatty acid-binding protein 4 (FABP4), leptin, lipocalin-2, neutrophil elastase, proteinase-3, high sensitivity C-reactive protein (hsCRP) and adiponectin in venous plasma and pericardial fluid harvested during elective cardio-thoracic surgery (n = 132-152). RESULTS: In pericardial fluid compared to plasma, the levels were significantly smaller (p < 0.001) for leptin, lipocalin-2, neutrophil elastase, proteinase-3, hsCRP and adiponectin. For these biomarkers, the ratio of pericardial fluid-to-plasma level ([PF]/[P], median (interquartile range)) was 0.65 (0.47-1.01), 0.78 (0.56-1.09), 0.23 (0.11-0.60), 0.17 (0.09-0.36), 0.14 (0.08-0.35), and 0.25 (0.15-0.34), respectively. In contrast, pericardial fluid was significantly enriched (p < 0.001) in NT-pBNP ([PF]/[P]: 1.9 (1.06-2.73)) and even more so for FABP4 ([PF]/[P]: 3.90 (1.47-9.77)). Moreover, in pericardial fluid, the adipocytokines interrelated all significantly positive and correlated negative to hsCRP, whereas for NT-pBNP only a significantly positive correlation with adiponectin was found. These interrelations were distinct from those in the plasma, as were the correlations of the pericardial biomarkers with patient characteristics compared to plasma. CONCLUSIONS: In cardiovascular disease patients, the pericardial cavity is a distinct adipocytokine microenvironment in which especially FABP4 is mainly derived from the heart.


Asunto(s)
Enfermedades Cardiovasculares/metabolismo , Proteínas de Unión a Ácidos Grasos/metabolismo , Pericardio/metabolismo , Adipoquinas/sangre , Adipoquinas/metabolismo , Adiponectina/sangre , Adiponectina/metabolismo , Anciano , Biomarcadores/sangre , Biomarcadores/metabolismo , Proteína C-Reactiva/metabolismo , Enfermedades Cardiovasculares/sangre , Proteínas de Unión a Ácidos Grasos/sangre , Femenino , Humanos , Leptina/sangre , Leptina/metabolismo , Elastasa de Leucocito/sangre , Elastasa de Leucocito/metabolismo , Lipocalina 2/sangre , Lipocalina 2/metabolismo , Masculino , Persona de Mediana Edad , Mieloblastina/sangre , Mieloblastina/metabolismo , Péptido Natriurético Encefálico/sangre , Péptido Natriurético Encefálico/metabolismo , Fragmentos de Péptidos/sangre , Fragmentos de Péptidos/metabolismo
8.
Cell Transplant ; 24(1): 37-48, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-24069942

RESUMEN

The success of pancreatic islet transplantation is limited by delayed engraftment and suboptimal function in the longer term. Endothelial progenitor cells (EPCs) represent a potential cellular therapy that may improve the engraftment of transplanted pancreatic islets. In addition, EPCs may directly affect the function of pancreatic ß-cells. The objective of this study was to examine the ability of EPCs to enhance pancreatic islet transplantation in a murine syngeneic marginal mass transplant model and to examine the mechanisms through which this occurs. We found that cotransplanted EPCs improved the cure rate and initial glycemic control of transplanted islets. Gene expression data indicate that EPCs, or their soluble products, modulate the expression of the ß-cell surface molecule connexin 36 and affect glucose-stimulated insulin release in vitro. In conclusion, EPCs are a promising candidate for improving outcomes in islet transplantation, and their mechanisms of action warrant further study.


Asunto(s)
Conexinas/biosíntesis , Células Endoteliales/metabolismo , Regulación de la Expresión Génica , Células Secretoras de Insulina/metabolismo , Trasplante de Islotes Pancreáticos , Trasplante de Células Madre , Células Madre/metabolismo , Animales , Células Endoteliales/patología , Células Endoteliales/trasplante , Glucosa/farmacología , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/patología , Masculino , Ratones , Células Madre/patología , Edulcorantes/farmacología , Trasplante Isogénico , Proteína delta-6 de Union Comunicante
9.
Int Immunopharmacol ; 17(4): 1167-75, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23863858

RESUMEN

Neutrophils are a key mediator of the innate immune system and are pivotal in the inflammatory response to infection or tissue damage. Fundamental to the role that neutrophils play in host defence is their interaction with the vascular endothelium. From the initial mobilisation of neutrophils out of the bone marrow to their ultimate transmigration through the vasculature, endothelial cells are a vital component of the inflammatory process. This review focuses on the interactions that take place between neutrophils and the vascular endothelium during the various stages of the inflammatory response. The role of the vascular endothelium in detecting the initial episode of infection or tissue damage is explored and how this ultimately leads to mobilisation of the neutrophils from the bone marrow and into the bloodstream. The recruitment and adhesion of neutrophils to the vascular endothelium is also discussed, with particular emphasis on the various discrete stages of the adhesion cascade and what molecules mediate these steps. In addition, a novel role for the lipid kinase sphingosine kinase in neutrophil adhesion is examined. With the advent of improved imaging techniques and the development of new animal models, this is a dynamic area of research and this review aims to summarise some of the more recent findings.


Asunto(s)
Endotelio Vascular/fisiología , Neutrófilos/fisiología , Animales , Movimiento Celular , Humanos , Fosfotransferasas (Aceptor de Grupo Alcohol)/fisiología
10.
J Allergy (Cairo) ; 2012: 154174, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23316248

RESUMEN

Allergic inflammation is an immune response to foreign antigens, which begins within minutes of exposure to the allergen followed by a late phase leading to chronic inflammation. Prolonged allergic inflammation manifests in diseases such as urticaria and rhino-conjunctivitis, as well as chronic asthma and life-threatening anaphylaxis. The prevalence of allergic diseases is profound with 25% of the worldwide population affected and a rising trend across all ages, gender, and racial groups. The identification and avoidance of allergens can manage this disease, but this is not always possible with triggers being common foods, prevalent air-borne particles and only extremely low levels of allergen exposure required for sensitization. Patients who are sensitive to multiple allergens require prophylactic and symptomatic treatments. Current treatments are often suboptimal and associated with adverse effects, such as the interruption of cognition, sleep cycles, and endocrine homeostasis, all of which affect quality of life and are a financial burden to society. Clearly, a better therapeutic approach for allergic diseases is required. Herein, we review the current knowledge of allergic inflammation and discuss the role of sphingolipids as potential targets to regulate inflammatory development in vivo and in humans. We also discuss the benefits and risks of using sphingolipid inhibitors.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA