Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Circ Res ; 117(4): 376-87, 2015 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-26129975

RESUMEN

RATIONALE: Platelets are known to play a crucial role in hemostasis. Sphingosine kinases (Sphk) 1 and 2 catalyze the conversion of sphingosine to the bioactive metabolite sphingosine 1-phosphate (S1P). Although platelets are able to secrete S1P on activation, little is known about a potential intrinsic effect of S1P on platelet function. OBJECTIVE: To investigate the role of Sphk1- and Sphk2-derived S1P in the regulation of platelet function. METHODS AND RESULTS: We found a 100-fold reduction in intracellular S1P levels in platelets derived from Sphk2(-/-) mutants compared with Sphk1(-/-) or wild-type mice, as analyzed by mass spectrometry. Sphk2(-/-) platelets also failed to secrete S1P on stimulation. Blood from Sphk2-deficient mice showed decreased aggregation after protease-activated receptor 4-peptide and adenosine diphosphate stimulation in vitro, as assessed by whole blood impedance aggregometry. We revealed that S1P controls platelet aggregation via the sphingosine 1-phosphate receptor 1 through modulation of protease-activated receptor 4-peptide and adenosine diphosphate-induced platelet activation. Finally, we show by intravital microscopy that defective platelet aggregation in Sphk2-deficient mice translates into reduced arterial thrombus stability in vivo. CONCLUSIONS: We demonstrate that Sphk2 is the major Sphk isoform responsible for the generation of S1P in platelets and plays a pivotal intrinsic role in the control of platelet activation. Correspondingly, Sphk2-deficient mice are protected from arterial thrombosis after vascular injury, but have normal bleeding times. Targeting this pathway could therefore present a new therapeutic strategy to prevent thrombosis.


Asunto(s)
Plaquetas/enzimología , Lisofosfolípidos/sangre , Fosfotransferasas (Aceptor de Grupo Alcohol)/sangre , Agregación Plaquetaria , Esfingosina/análogos & derivados , Animales , Ácido Araquidónico/sangre , Coagulación Sanguínea , Pruebas de Coagulación Sanguínea , Traumatismos de las Arterias Carótidas/sangre , Traumatismos de las Arterias Carótidas/enzimología , Modelos Animales de Enfermedad , Eritrocitos/enzimología , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Fosfotransferasas (Aceptor de Grupo Alcohol)/deficiencia , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Adhesividad Plaquetaria , Pruebas de Función Plaquetaria , Receptores de Lisoesfingolípidos/sangre , Transducción de Señal , Esfingosina/sangre , Receptores de Esfingosina-1-Fosfato , Trombosis/sangre , Trombosis/enzimología , Trombosis/prevención & control , Tromboxano A2/sangre , Lesiones del Sistema Vascular/sangre , Lesiones del Sistema Vascular/enzimología
2.
Nephrol Dial Transplant ; 32(8): 1313-1322, 2017 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-28206609

RESUMEN

BACKGROUND: C5a plays a crucial role in anti-neutrophil cytoplasmic antibody (ANCA)-mediated neutrophil recruitment and activation. Our previous studies found that the interaction between sphingosine-1-phosphate (S1P) and C5a plays an important role in the ANCA-mediated activation of neutrophils. In the current study, the expression levels of S1P in plasma and its receptors (S1PR1-5) in kidneys were analysed in patients with ANCA-associated vasculitis (AAV). METHODS: Plasma samples from 32 AAV patients in active stage and 20 AAV patients in remission were collected. The plasma levels of S1P were determined by an enzyme-linked immunosorbent assay (ELISA). The expression of S1PR1-5 in the renal specimens from 24 AAV patients was detected by immunohistochemistry. The associations of the plasma levels of S1P and renal expression of S1PRs with clinical and pathological parameters were analysed. RESULTS: The level of plasma S1P was significantly higher in AAV patients in active stage than it was in both patients in remission and in normal controls. Correlation analysis showed that the plasma levels of S1P correlated with the initial serum creatinine levels (r = 0.502, P = 0.003) and inversely correlated with the estimated glomerular filtration rate (eGFR; r = -0.358, P = 0.044) in AAV patients. Double-labelling immunofluorescence assay suggested that S1PR1-5 were expressed on endothelial cells in the glomeruli and that S1PR1, 4 and 5 were expressed on neutrophils. CONCLUSIONS: In AAV patients, the circulating S1P levels were elevated and the renal expression of S1PR2-5 was upregulated. The levels of circulating S1P and the renal expression of S1PR were associated with the renal involvement and disease activity of AAV.


Asunto(s)
Vasculitis Asociada a Anticuerpos Citoplasmáticos Antineutrófilos/sangre , Biomarcadores/sangre , Lisofosfolípidos/metabolismo , Receptores de Lisoesfingolípidos/sangre , Esfingosina/análogos & derivados , Vasculitis Asociada a Anticuerpos Citoplasmáticos Antineutrófilos/diagnóstico , Vasculitis Asociada a Anticuerpos Citoplasmáticos Antineutrófilos/inmunología , Ensayo de Inmunoadsorción Enzimática , Femenino , Tasa de Filtración Glomerular , Humanos , Glomérulos Renales/patología , Masculino , Persona de Mediana Edad , Neutrófilos/inmunología , Esfingosina/metabolismo , Receptores de Esfingosina-1-Fosfato
3.
Handb Exp Pharmacol ; 224: 455-82, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25522998

RESUMEN

The cholesterol of high-density lipoproteins (HDLs) and its major proteic component, apoA-I, have been widely investigated as potential predictors of acute cardiovascular (CV) events. In particular, HDL cholesterol levels were shown to be inversely and independently associated with the risk of acute CV diseases in different patient populations, including autoimmune and chronic inflammatory disorders. Some relevant and direct anti-inflammatory activities of HDL have been also recently identified targeting both immune and vascular cell subsets. These studies recently highlighted the improvement of HDL function (instead of circulating levels) as a promising treatment strategy to reduce inflammation and associated CV risk in several diseases, such as systemic lupus erythematosus and rheumatoid arthritis. In these diseases, anti-inflammatory treatments targeting HDL function might improve both disease activity and CV risk. In this narrative review, we will focus on the pathophysiological relevance of HDL and apoA-I levels/functions in different acute and chronic inflammatory pathophysiological conditions.


Asunto(s)
Apolipoproteína A-I/sangre , Enfermedades Autoinmunes/sangre , Inflamación/sangre , Lipoproteínas HDL/sangre , Animales , Apolipoproteína A-I/química , Enfermedades Autoinmunes/inmunología , Biomarcadores/sangre , Humanos , Inmunidad Innata , Inflamación/inmunología , Lipoproteínas HDL/química , Linfocitos/inmunología , Linfocitos/metabolismo , Lisofosfolípidos/sangre , Microdominios de Membrana/inmunología , Microdominios de Membrana/metabolismo , Conformación Proteica , Receptores de Lisoesfingolípidos/sangre , Esfingosina/análogos & derivados , Esfingosina/sangre , Relación Estructura-Actividad
4.
Mediators Inflamm ; 2015: 831059, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26604433

RESUMEN

Sphingosine-1-phosphate (S1P) is a versatile lipid signaling molecule and key regulator in vascular inflammation. S1P is secreted by platelets, monocytes, and vascular endothelial and smooth muscle cells. It binds specifically to a family of G-protein-coupled receptors, S1P receptors 1 to 5, resulting in downstream signaling and numerous cellular effects. S1P modulates cell proliferation and migration, and mediates proinflammatory responses and apoptosis. In the vascular barrier, S1P regulates permeability and endothelial reactions and recruitment of monocytes and may modulate atherosclerosis. Only recently has S1P emerged as a critical mediator which directly links the coagulation factor system to vascular inflammation. The multifunctional proteases thrombin and FXa regulate local S1P availability and interact with S1P signaling at multiple levels in various vascular cell types. Differential expression patterns and intracellular signaling pathways of each receptor enable S1P to exert its widespread functions. Although a vast amount of information is available about the functions of S1P and its receptors in the regulation of physiological and pathophysiological conditions, S1P-mediated mechanisms in the vasculature remain to be elucidated. This review summarizes recent findings regarding the role of S1P and its receptors in vascular wall and blood cells, which link the coagulation system to inflammatory responses in the vasculature.


Asunto(s)
Coagulación Sanguínea/fisiología , Inflamación/sangre , Inflamación/inmunología , Lisofosfolípidos/sangre , Lisofosfolípidos/inmunología , Receptores de Lisoesfingolípidos/sangre , Receptores de Lisoesfingolípidos/inmunología , Esfingosina/análogos & derivados , Coagulación Sanguínea/inmunología , Vasos Sanguíneos/fisiología , Endotelio Vascular/fisiología , Humanos , Modelos Cardiovasculares , Modelos Inmunológicos , Activación Plaquetaria , Receptores de Trombina/metabolismo , Transducción de Señal , Esfingosina/sangre , Esfingosina/inmunología
5.
Am J Physiol Lung Cell Mol Physiol ; 305(7): L467-77, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-23911438

RESUMEN

The genetic mechanisms underlying the susceptibility to acute respiratory distress syndrome (ARDS) are poorly understood. We previously demonstrated that sphingosine 1-phosphate (S1P) and the S1P receptor S1PR3 are intimately involved in lung inflammatory responses and vascular barrier regulation. Furthermore, plasma S1PR3 protein levels were shown to serve as a biomarker of severity in critically ill ARDS patients. This study explores the contribution of single nucleotide polymorphisms (SNPs) of the S1PR3 gene to sepsis-associated ARDS. S1PR3 SNPs were identified by sequencing the entire gene and tagging SNPs selected for case-control association analysis in African- and ED samples from Chicago, with independent replication in a European case-control study of Spanish individuals. Electrophoretic mobility shift assays, luciferase activity assays, and protein immunoassays were utilized to assess the functionality of associated SNPs. A total of 80 variants, including 29 novel SNPs, were identified. Because of limited sample size, conclusive findings could not be drawn in African-descent ARDS subjects; however, significant associations were found for two promoter SNPs (rs7022797 -1899T/G; rs11137480 -1785G/C), across two ED samples supporting the association of alleles -1899G and -1785C with decreased risk for sepsis-associated ARDS. In addition, these alleles significantly reduced transcription factor binding to the S1PR3 promoter; reduced S1PR3 promoter activity, a response particularly striking after TNF-α challenge; and were associated with lower plasma S1PR3 protein levels in ARDS patients. These highly functional studies support S1PR3 as a novel ARDS candidate gene and a potential target for individualized therapy.


Asunto(s)
Regiones Promotoras Genéticas , Receptores de Lisoesfingolípidos/genética , Síndrome de Dificultad Respiratoria/genética , Sepsis/complicaciones , Secuencia de Bases , Biomarcadores/sangre , Estudios de Casos y Controles , Ensayo de Cambio de Movilidad Electroforética , Femenino , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Genotipo , Humanos , Lisofosfolípidos/metabolismo , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Polimorfismo de Nucleótido Simple , Receptores de Lisoesfingolípidos/sangre , Síndrome de Dificultad Respiratoria/sangre , Síndrome de Dificultad Respiratoria/etiología , Análisis de Secuencia de ADN , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Receptores de Esfingosina-1-Fosfato
6.
Am J Respir Cell Mol Biol ; 47(5): 628-36, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22771388

RESUMEN

The inflamed lung exhibits oxidative and nitrative modifications of multiple target proteins, potentially reflecting disease severity and progression. We identified sphingosine-1-phosphate receptor-3 (S1PR3), a critical signaling molecule mediating cell proliferation and vascular permeability, as a nitrated plasma protein in mice with acute lung injury (ALI). We explored S1PR3 as a potential biomarker in murine and human ALI. In vivo nitrated and total S1PR3 concentrations were determined by immunoprecipitation and microarray studies in mice, and by ELISA in human plasma. In vitro nitrated S1PR3 concentrations were evaluated in human lung vascular endothelial cells (ECs) or within microparticles shed from ECs after exposure to barrier-disrupting agonists (LPS, low-molecular-weight hyaluronan, and thrombin). The effects of S1PR3-containing microparticles on EC barrier function were assessed by transendothelial electrical resistance (TER). Nitrated S1PR3 was identified in the plasma of murine ALI and in humans with severe sepsis-induced ALI. Elevated total S1PR3 plasma concentrations (> 251 pg/ml) were linked to sepsis and ALI mortality. In vitro EC exposure to barrier-disrupting agents induced S1PR3 nitration and the shedding of S1PR3-containing microparticles, which significantly reduced TER, consistent with increased permeability. These changes were attenuated by reduced S1PR3 expression (small interfering RNAs). These results suggest that microparticles containing nitrated S1PR3 shed into the circulation during inflammatory lung states, and represent a novel ALI biomarker linked to disease severity and outcome.


Asunto(s)
Lesión Pulmonar Aguda/sangre , Receptores de Lisoesfingolípidos/sangre , Lesión Pulmonar Aguda/inmunología , Lesión Pulmonar Aguda/mortalidad , Adulto , Anciano , Animales , Biomarcadores/sangre , Permeabilidad Capilar , Estudios de Casos y Controles , Micropartículas Derivadas de Células/metabolismo , Células Cultivadas , Impedancia Eléctrica , Células Endoteliales/inmunología , Células Endoteliales/metabolismo , Células Endoteliales/patología , Endotelio Vascular/patología , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Estimación de Kaplan-Meier , Lipopolisacáridos/farmacología , Pulmón/patología , Masculino , Ratones , Persona de Mediana Edad , Arteria Pulmonar/patología , Interferencia de ARN , Receptores de Lisoesfingolípidos/genética , Receptores de Esfingosina-1-Fosfato , Tirosina/análogos & derivados , Tirosina/sangre , Lesión Pulmonar Inducida por Ventilación Mecánica/metabolismo
7.
Am J Physiol Cell Physiol ; 302(10): C1460-8, 2012 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-22357735

RESUMEN

Lymph nodes are highly organized structures specialized for efficient regulation of adaptive immunity. The blood and lymphatic systems within a lymph node play essential roles by providing functionally distinct environments for lymphocyte entry and egress, respectively. Direct imaging and measurement of vascular microenvironments by intravital multiphoton microscopy provide anatomical and mechanistic insights into the essential events of lymphocyte trafficking. Lymphocytes, blood endothelial cells, and lymphatic endothelial cells express sphingosine 1-phosphate receptor 1, a key G protein-coupled receptor regulating cellular egress and a modulator of endothelial permeability. Here we report the development of a differential vascular labeling (DVL) technique in which a single intravenous injection of a fluorescent dextran, in combination with fluorescent semiconductor quantum dot particles, differentially labels multiple blood and lymphatic compartments in a manner dependent on the size of the fluorescent particle used. Thus DVL allows measurement of endothelial integrity in multiple vascular compartments and the affects or pharmacological manipulation in vascular integrity. In addition, this technique allows for real-time observation of lymphocyte trafficking across physiological barriers differentiated by DVL. Last, single-field fluid movement dynamics can be derived, allowing for the simultaneous determination of fluid flow rates in diverse blood and lymphatic compartments.


Asunto(s)
Sistemas de Computación , Células Endoteliales/fisiología , Líquido Extracelular/química , Vasos Linfáticos/química , Flujo Sanguíneo Regional , Coloración y Etiquetado/métodos , Animales , Células Endoteliales/química , Líquido Extracelular/fisiología , Ganglios Linfáticos/irrigación sanguínea , Ganglios Linfáticos/química , Ganglios Linfáticos/fisiología , Vasos Linfáticos/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores de Lisoesfingolípidos/biosíntesis , Receptores de Lisoesfingolípidos/sangre , Flujo Sanguíneo Regional/fisiología
8.
Sci Rep ; 8(1): 15371, 2018 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-30337577

RESUMEN

There are no data on the effects of fingolimod, an immunomodulatory drug used in treatment of multiple sclerosis (MS), on circulating tight-junction (TJ) protein levels as well as on peripheral blood mononuclear cells (PBMC) migration. Serum TJ protein [occludin (OCLN), claudin-5 (CLN-5) and zonula occludens-1 (ZO-1)] levels, sphingosine-1 phosphate 1 (S1P1) receptor expression on circulating leukocyte populations as well as in vitro PBMC migration were longitudinally assessed in 20 MS patients under 12-months fingolimod treatment and correlated with clinical and magnetic resonance imaging (MRI) parameters. After 12 months of treatment, a significant reduction of mean relapse rate as well as number of active lesions at MRI was found. TJ protein levels significantly decreased and were associated with reduction of S1P1 expression as well as of PBMC in vitro migratory activity. A significant correlation of CLN-5/OCLN ratio with new T2 MRI lesions and a significant inverse correlation of CLN-5/ZO-1 ratio with disability scores were found. These findings support possible in vivo effects of fingolimod on the blood-brain barrier (BBB) functional activity as well as on peripheral cell trafficking that could result in avoiding passage of circulating autoreactive cells into brain parenchyma. Circulating TJ protein levels and respective ratios could be further studied as a novel candidate biomarker of BBB functional status to be monitored in course of fingolimod as well as of other immunomodulatory treatments in MS.


Asunto(s)
Biomarcadores/sangre , Movimiento Celular , Clorhidrato de Fingolimod/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Leucocitos Mononucleares/efectos de los fármacos , Esclerosis Múltiple/patología , Proteínas de Uniones Estrechas/sangre , Adulto , Quimiotaxis , Femenino , Humanos , Inmunosupresores/farmacología , Técnicas In Vitro , Estudios Longitudinales , Masculino , Esclerosis Múltiple/sangre , Esclerosis Múltiple/tratamiento farmacológico , Estudios Prospectivos , Receptores de Lisoesfingolípidos/sangre
9.
J Atheroscler Thromb ; 24(9): 954-969, 2017 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-28321011

RESUMEN

AIM: Sphingosine 1-phosphate (S1P) has been suggested to be a positive regulator of plasminogen activator inhibitor 1 (PAI-1) in adipocytes, while some studies are not consistent with this prothrombotic property of S1P. Since S1P is bound to apolipoprotein M (apoM) on HDL or to albumin in plasma, we compared the properties of these two forms on the PAI-1 induction. METHODS: We investigated the associations of S1P, apoM, and PAI-1 concentrations in the plasma of normal coronary artery (NCA), stable angina pectoris (SAP), and acute coronary syndrome (ACS) subjects (n=32, 71, and 38, respectively). Then, we compared the effects of S1P with various vehicles on the PAI-1 expression in 3T3L1 adipocytes. We also investigated the modulation of the PAI-1 levels in mice infected with adenovirus coding apoM. RESULTS: Among ACS subjects, the PAI-1 level was positively correlated with the S1P level, but not the apoM level. In adipocytes, S1P bound to an apoM-rich vehicle induced PAI-1 expression to a lesser extent than the control vehicle, while S1P bound to an apoM-depleted vehicle induced PAI-1 expression to a greater extent than the control vehicle in 3T3L1 adipocytes. Additionally, apoM overexpression in mice failed to modulate the plasma PAI-1 level and the adipose PAI-1 expression level. S1P bound to albumin increased PAI-1 expression through the S1P receptor 2-Rho/ROCK-NFκB pathway. CONCLUSION: S1P bound to albumin, but not to apoM, induces PAI-1 expression in adipocytes, indicating that S1P can exert different properties on the pathogenesis of vascular diseases, depending on its vehicle.


Asunto(s)
Lisofosfolípidos/administración & dosificación , Lisofosfolípidos/sangre , Inhibidor 1 de Activador Plasminogénico/sangre , Esfingosina/análogos & derivados , Células 3T3-L1 , Síndrome Coronario Agudo/sangre , Adipocitos/metabolismo , Angina Estable/sangre , Animales , Apolipoproteínas M/sangre , Células Endoteliales de la Vena Umbilical Humana , Humanos , Lipoproteínas HDL/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , FN-kappa B/sangre , Activación Plaquetaria , Unión Proteica , Receptores de Lisoesfingolípidos/sangre , Proteínas Recombinantes/sangre , Serpina E2/sangre , Albúmina Sérica Humana/metabolismo , Transducción de Señal , Esfingosina/administración & dosificación , Esfingosina/sangre , Receptores de Esfingosina-1-Fosfato
10.
Stem Cells Dev ; 22(11): 1645-56, 2013 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-23282236

RESUMEN

Acute myocardial infarction (AMI) triggers mobilization of stem cells from bone marrow (BM) into peripheral blood (PB). Based on our observation that the bioactive sphingophospholipids, sphingosine-1 phosphate (S1P), and ceramide-1 phosphate (C1P) regulate trafficking of hematopoietic stem cells (HSCs), we explored whether they also direct trafficking of non-hematopoietic stem cells (non-HSCs). We detected a 3-6-fold increase in circulating CD34+, CD133+, and CXCR4+ lineage-negative (Lin-)/CD45- cells that are enriched in non-HSCs [including endothelial progenitors (EPCs) and very small embryonic-like stem cells (VSELs)] in PB from AMI patients (P<0.05 vs. controls). Concurrently, we measured a ∼3-fold increase in S1P and C1P levels in plasma from AMI patients. At the same time, plasma obtained at hospital admission and 6 h after AMI strongly chemoattracted human BM-derived CD34+/Lin- and CXCR4+/Lin- cells in Transwell chemotaxis assays. This effect of plasma was blunted after depletion of S1P level by charcoal stripping and was further inhibited by the specific S1P1 receptor antagonist such as W146 and VPC23019. We also noted that the expression of S1P receptor 1 (S1P1), which is dominant in naïve BM, is reduced after the exposure to S1P at concentrations similar to the plasma S1P levels in patients with AMI, thus influencing the role of S1P in homing to the injured myocardium. Therefore, we examined mechanisms, other than bioactive lipids, that may contribute to the homing of BM non-HSCs to the infarcted myocardium. Hypoxic cardiac tissue increases the expression of cathelicidin and ß-2 defensin, which could explain why PB cells isolated from patients with AMI migrated more efficiently to a low, yet physiological, gradient of stromal-derived factor-1 in Transwell migration assays. Together, these observations suggest that while elevated S1P and C1P levels early in the course of AMI may trigger mobilization of non-HSCs into PB, cathelicidin and ß-2 defensin could play an important role in their homing to damaged myocardium.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/metabolismo , Ceramidas/metabolismo , Trasplante de Células Madre Hematopoyéticas , Lisofosfolípidos/metabolismo , Infarto del Miocardio/terapia , Esfingosina/análogos & derivados , Antígeno AC133 , Animales , Antígenos CD/sangre , Antígenos CD34/sangre , Péptidos Catiónicos Antimicrobianos/biosíntesis , Células de la Médula Ósea/metabolismo , Hipoxia de la Célula , Movimiento Celular , Quimiocina CXCL12/metabolismo , Glicoproteínas/sangre , Movilización de Célula Madre Hematopoyética , Células Madre Hematopoyéticas/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Miocardio/citología , Miocardio/metabolismo , Péptidos/sangre , Receptores CXCR4/sangre , Receptores de Lisoesfingolípidos/sangre , Esfingosina/metabolismo , beta-Defensinas/biosíntesis , Catelicidinas
11.
Dev Cell ; 23(3): 600-10, 2012 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-22975328

RESUMEN

During angiogenesis, nascent vascular sprouts fuse to form vascular networks, enabling efficient circulation. Mechanisms that stabilize the vascular plexus are not well understood. Sphingosine 1-phosphate (S1P) is a blood-borne lipid mediator implicated in the regulation of vascular and immune systems. Here we describe a mechanism by which the G protein-coupled S1P receptor-1 (S1P1) stabilizes the primary vascular network. A gradient of S1P1 expression from the mature regions of the vascular network to the growing vascular front was observed. In the absence of endothelial S1P1, adherens junctions are destabilized, barrier function is breached, and flow is perturbed, resulting in abnormal vascular hypersprouting. Interestingly, S1P1 responds to S1P as well as laminar shear stress to transduce flow-mediated signaling in endothelial cells both in vitro and in vivo. These data demonstrate that blood flow and circulating S1P activate endothelial S1P1 to stabilize blood vessels in development and homeostasis.


Asunto(s)
Vasos Sanguíneos/crecimiento & desarrollo , Vasos Sanguíneos/metabolismo , Células Endoteliales/citología , Células Endoteliales/metabolismo , Receptores de Lisoesfingolípidos/metabolismo , Transducción de Señal , Animales , Hemorreología , Homeostasis , Ratones , Receptores de Lisoesfingolípidos/sangre
12.
Shock ; 33(1): 31-42, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19752779

RESUMEN

Hepatic ischemia/reperfusion (I/R) injury is a major complication after liver transplantation, major hepatic resection, or prolonged portal vein occlusion. Furthermore, acute kidney injury is frequent after hepatic I/R and greatly increases postoperative complications. Sphinganine-1-phosphate is a sphingolipid with uncharacterized physiological effects. We serendipitously determined that plasma levels of sphinganine-1-phosphate fell significantly after liver I/R in mice. In this study, we hypothesized that repletion of plasma sphinganine-1-phosphate would protect against liver and kidney injuries after liver I/R. C57BL/6 mice were subjected to 60 min of partial hepatic I/R and treated with either vehicle or with sphinganine-1-phosphate (given immediately before and 2 h after reperfusion). Vehicle-treated mice subjected to liver I/R developed acute liver and kidney injuries with elevated plasma alanine aminotransferase and creatinine 5 and 24 h after liver I/R. However, liver and kidney injuries were significantly attenuated with sphinganine-1-phosphate treatment. Sphinganine-1-phosphate markedly inhibited liver and kidney necrosis and apoptosis 24 h after liver I/R. Moreover, sphinganine-1-phosphate attenuated neutrophil infiltration, reduced plasma IL-6 and TNF-alpha upregulation, and preserved liver and kidney vascular integrity while reducing liver and kidney F-actin degradation after liver I/R. Finally, sphinganine-1-phosphate-mediated hepatic and renal protection was blocked by VPC23019, an antagonist for sphingosine-1-phosphate type 1 receptor. Therefore, sphinganine-1-phosphate improves acute liver and kidney injuries after hepatic I/R via sphingosine-1-phosphate type 1 receptor-mediated inhibition of necrosis and apoptosis and by improving vascular integrity. Harnessing the mechanisms of cytoprotection with sphinganine-1-phosphate activation may lead to new therapies for perioperative hepatic I/R injury and subsequent remote organ injury.


Asunto(s)
Riñón/efectos de los fármacos , Riñón/patología , Hígado/efectos de los fármacos , Hígado/patología , Lisofosfolípidos/uso terapéutico , Daño por Reperfusión/tratamiento farmacológico , Esfingosina/análogos & derivados , Actinas/metabolismo , Animales , Apoptosis/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayo de Inmunoadsorción Enzimática , Interleucina-6/sangre , Riñón/metabolismo , Hígado/metabolismo , Lisofosfolípidos/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Infiltración Neutrófila/efectos de los fármacos , Receptores de Lisoesfingolípidos/agonistas , Receptores de Lisoesfingolípidos/sangre , Daño por Reperfusión/sangre , Daño por Reperfusión/patología , Esfingosina/sangre , Esfingosina/uso terapéutico , Factor de Necrosis Tumoral alfa/sangre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA