Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Circ Res ; 119(8): e110-26, 2016 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-27582371

RESUMO

RATIONALE: Sphingosine-1-phosphate (S1P) signaling is essential for vascular development and postnatal vascular homeostasis. The relative importance of S1P sources sustaining these processes remains unclear. OBJECTIVE: To address the level of redundancy in bioactive S1P provision to the developing and mature vasculature. METHODS AND RESULTS: S1P production was selectively impaired in mouse platelets, erythrocytes, endothelium, or smooth muscle cells by targeted deletion of genes encoding sphingosine kinases -1 and -2. S1P deficiency impaired aggregation and spreading of washed platelets and profoundly reduced their capacity to promote endothelial barrier function ex vivo. However, and in contrast to recent reports, neither platelets nor any other source of S1P was essential for vascular development, vascular integrity, or hemostasis/thrombosis. Yet rapid and profound depletion of plasma S1P during systemic anaphylaxis rendered both platelet- and erythrocyte-derived S1P essential for survival, with a contribution from blood endothelium observed only in the absence of circulating sources. Recovery was sensitive to aspirin in mice with but not without platelet S1P, suggesting that platelet activation and stimulus-response coupling is needed. S1P deficiency aggravated vasoplegia in this model, arguing a vital role for S1P in maintaining vascular resistance during recovery from circulatory shock. Accordingly, the S1P2 receptor mediated most of the survival benefit of S1P, whereas the endothelial S1P1 receptor was dispensable for survival despite its importance for maintaining vascular integrity. CONCLUSIONS: Although source redundancy normally secures essential S1P signaling in developing and mature blood vessels, profound depletion of plasma S1P renders both erythrocyte and platelet S1P pools necessary for recovery and high basal plasma S1P levels protective during anaphylactic shock.


Assuntos
Anafilaxia/metabolismo , Plaquetas/metabolismo , Endotélio Vascular/metabolismo , Eritrócitos/metabolismo , Homeostase/fisiologia , Lisofosfolipídeos/deficiência , Esfingosina/análogos & derivados , Anafilaxia/patologia , Animais , Vasos Sanguíneos/crescimento & desenvolvimento , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/patologia , Endotélio Vascular/crescimento & desenvolvimento , Endotélio Vascular/patologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Esfingosina/deficiência
2.
Crit Care ; 19: 372, 2015 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-26498205

RESUMO

INTRODUCTION: Sphingosine-1-phosphate (S1P) is a signaling lipid that regulates pathophysiological processes involved in sepsis progression, including endothelial permeability, cytokine release, and vascular tone. The aim of this study was to investigate whether serum-S1P concentrations are associated with disease severity in patients with sepsis. METHODS: This single-center prospective-observational study includes 100 patients with systemic inflammatory response syndrome (SIRS) plus infection (n = 40), severe sepsis (n = 30), or septic shock (n = 30) and 214 healthy blood donors as controls. Serum-S1P was measured by mass spectrometry. Blood parameters, including C-reactive protein (CRP), procalcitonin (PCT), interleukin-6 (IL-6), lactate, and white blood cells (WBCs), were determined by routine assays. The Sequential Organ Failure Assessment (SOFA) score was generated and used to evaluate disease severity. RESULTS: Serum-S1P concentrations were lower in patients than in controls (P < 0.01), and the greatest difference was between the control and the septic shock groups (P < 0.01). Serum-S1P levels were inversely correlated with disease severity as determined by the SOFA score (P < 0.01) as well as with IL-6, PCT, CRP, creatinine, lactate, and fluid balance. A receiver operating characteristic analysis for the presence or absence of septic shock revealed equally high sensitivity and specificity for S1P compared with the SOFA score. In a multivariate logistic regression model calculated for prediction of septic shock, S1P emerged as the strongest predictor (P < 0.001). CONCLUSIONS: In patients with sepsis, serum-S1P levels are dramatically decreased and are inversely associated with disease severity. Since S1P is a potent regulator of endothelial integrity, low S1P levels may contribute to capillary leakage, impaired tissue perfusion, and organ failure in sepsis.


Assuntos
Lisofosfolipídeos/sangue , Insuficiência de Múltiplos Órgãos/mortalidade , Sepse/mortalidade , Esfingosina/análogos & derivados , Adulto , Feminino , Alemanha , Humanos , Lisofosfolipídeos/deficiência , Masculino , Pessoa de Meia-Idade , Insuficiência de Múltiplos Órgãos/sangue , Estudos Prospectivos , Sepse/sangue , Sepse/terapia , Índice de Gravidade de Doença , Esfingosina/sangue , Esfingosina/deficiência
3.
Stroke ; 46(8): 2260-70, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26138121

RESUMO

BACKGROUND AND PURPOSE: Subarachnoid hemorrhage (SAH) is a complex stroke subtype characterized by an initial brain injury, followed by delayed cerebrovascular constriction and ischemia. Current therapeutic strategies nonselectively curtail exacerbated cerebrovascular constriction, which necessarily disrupts the essential and protective process of cerebral blood flow autoregulation. This study identifies a smooth muscle cell autocrine/paracrine signaling network that augments myogenic tone in a murine model of experimental SAH: it links tumor necrosis factor-α (TNFα), the cystic fibrosis transmembrane conductance regulator, and sphingosine-1-phosphate signaling. METHODS: Mouse olfactory cerebral resistance arteries were isolated, cannulated, and pressurized for in vitro vascular reactivity assessments. Cerebral blood flow was measured by speckle flowmetry and magnetic resonance imaging. Standard Western blot, immunohistochemical techniques, and neurobehavioral assessments were also used. RESULTS: We demonstrate that targeting TNFα and sphingosine-1-phosphate signaling in vivo has potential therapeutic application in SAH. Both interventions (1) eliminate the SAH-induced myogenic tone enhancement, but otherwise leave vascular reactivity intact; (2) ameliorate SAH-induced neuronal degeneration and apoptosis; and (3) improve neurobehavioral performance in mice with SAH. Furthermore, TNFα sequestration with etanercept normalizes cerebral perfusion in SAH. CONCLUSIONS: Vascular smooth muscle cell TNFα and sphingosine-1-phosphate signaling significantly enhance cerebral artery tone in SAH; anti-TNFα and anti-sphingosine-1-phosphate treatment may significantly improve clinical outcome.


Assuntos
Lisofosfolipídeos/biossíntese , Esfingosina/análogos & derivados , Hemorragia Subaracnóidea/metabolismo , Hemorragia Subaracnóidea/fisiopatologia , Fator de Necrose Tumoral alfa/biossíntese , Vasoconstrição/fisiologia , Animais , Artérias Cerebrais/efeitos dos fármacos , Artérias Cerebrais/fisiologia , Marcação de Genes/métodos , Lisofosfolipídeos/deficiência , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/fisiologia , Técnicas de Cultura de Órgãos , Fenilefrina/administração & dosagem , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Esfingosina/biossíntese , Esfingosina/deficiência , Hemorragia Subaracnóidea/terapia , Fator de Necrose Tumoral alfa/deficiência , Vasoconstrição/efeitos dos fármacos , Sistema Vasomotor/efeitos dos fármacos , Sistema Vasomotor/fisiologia
4.
FASEB J ; 27(5): 1830-46, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23322166

RESUMO

There has been much recent interest in lysophosphatidic acid (LPA) signaling through one of its receptors, LPA1, in fibrotic diseases, but the mechanisms by which LPA-LPA1 signaling promotes pathological fibrosis remain to be fully elucidated. Using a mouse peritoneal fibrosis model, we demonstrate central roles for LPA and LPA1 in fibroblast proliferation. Genetic deletion or pharmacological antagonism of LPA1 protected mice from peritoneal fibrosis, blunting the increases in peritoneal collagen by 65.4 and 52.9%, respectively, compared to control animals and demonstrated that peritoneal fibroblast proliferation was highly LPA1 dependent. Activation of LPA1 on mesothelial cells induced these cells to express connective tissue growth factor (CTGF), driving fibroblast proliferation in a paracrine fashion. Activation of mesothelial cell LPA1 induced CTGF expression by inducing cytoskeleton reorganization in these cells, causing nuclear translocation of myocardin-related transcription factor (MRTF)-A and MRTF-B. Pharmacological inhibition of MRTF-induced transcription also diminished CTGF expression and fibrosis in the peritoneal fibrosis model, mitigating the increase in peritoneal collagen content by 57.9% compared to controls. LPA1-induced cytoskeleton reorganization therefore makes a previously unrecognized but critically important contribution to the profibrotic activities of LPA by driving MRTF-dependent CTGF expression, which, in turn, drives fibroblast proliferation.


Assuntos
Lisofosfolipídeos/metabolismo , Fibrose Peritoneal/prevenção & controle , Receptores de Ácidos Lisofosfatídicos/fisiologia , Anilidas/farmacologia , Animais , Benzamidas/farmacologia , Compostos de Bifenilo/farmacologia , Proliferação de Células , Clorexidina/análogos & derivados , Fator de Crescimento do Tecido Conjuntivo/biossíntese , Fator de Crescimento do Tecido Conjuntivo/fisiologia , Citoesqueleto/fisiologia , Modelos Animais de Doenças , Células Epiteliais/metabolismo , Epitélio/fisiologia , Isoxazóis/farmacologia , Lisofosfolipídeos/deficiência , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miofibroblastos/citologia , Fibrose Peritoneal/induzido quimicamente , Fibrose Peritoneal/patologia , Receptores de Ácidos Lisofosfatídicos/antagonistas & inibidores , Transativadores/antagonistas & inibidores , Transativadores/metabolismo , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/metabolismo
5.
PLoS One ; 7(5): e37218, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22606352

RESUMO

Sphingosine-1-phosphate (S1P) activates a widely expressed family of G protein-coupled receptors, serves as a muscle trophic factor and activates muscle stem cells called satellite cells (SCs) through unknown mechanisms. Here we show that muscle injury induces dynamic changes in S1P signaling and metabolism in vivo. These changes include early and profound induction of the gene encoding the S1P biosynthetic enzyme SphK1, followed by induction of the catabolic enzyme sphingosine phosphate lyase (SPL) 3 days later. These changes correlate with a transient increase in circulating S1P levels after muscle injury. We show a specific requirement for SphK1 to support efficient muscle regeneration and SC proliferation and differentiation. Mdx mice, which serve as a model for muscular dystrophy (MD), were found to be S1P-deficient and exhibited muscle SPL upregulation, suggesting that S1P catabolism is enhanced in dystrophic muscle. Pharmacological SPL inhibition increased muscle S1P levels, improved mdx muscle regeneration and enhanced SC proliferation via S1P receptor 2 (S1PR2)-dependent inhibition of Rac1, thereby activating Signal Transducer and Activator of Transcription 3 (STAT3), a central player in inflammatory signaling. STAT3 activation resulted in p21 and p27 downregulation in a S1PR2-dependent fashion in myoblasts. Our findings suggest that S1P promotes SC progression through the cell cycle by repression of cell cycle inhibitors via S1PR2/STAT3-dependent signaling and that SPL inhibition may provide a therapeutic strategy for MD.


Assuntos
Lisofosfolipídeos/farmacologia , Distrofia Muscular de Duchenne/tratamento farmacológico , Distrofia Muscular de Duchenne/metabolismo , Receptores de Lisoesfingolipídeo/metabolismo , Fator de Transcrição STAT3/metabolismo , Células Satélites de Músculo Esquelético/efeitos dos fármacos , Células Satélites de Músculo Esquelético/metabolismo , Esfingosina/análogos & derivados , Animais , Proliferação de Células , Feminino , Lisofosfolipídeos/deficiência , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Camundongos Knockout , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/lesões , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Distrofia Muscular de Duchenne/patologia , Fosfotransferases (Aceptor do Grupo Álcool)/deficiência , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Regeneração/efeitos dos fármacos , Regeneração/fisiologia , Células Satélites de Músculo Esquelético/patologia , Transdução de Sinais/efeitos dos fármacos , Esfingosina/deficiência , Esfingosina/farmacologia , Receptores de Esfingosina-1-Fosfato
6.
Methods Mol Biol ; 874: 129-39, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22528444

RESUMO

We describe a method to visualize the migration of osteoclast precursors within intact murine bone -marrow in real time using intravital multiphoton microscopy. Conventionally, cell migration has been evaluated using in vitro systems, such as transmigration assays. Although these methods are convenient for quantification and are highly reproducible, these in vitro assay systems may not accurately reflect in vivo cellular behavior. In addition to in vitro analyses, recent technological progress in two-photon excitation-based laser microscopy has enabled the visualization of dynamic cell behavior deep inside intact living organs. Combining this imaging method with in vitro chemoattraction analyses, we have revealed that sphingosine-1-phosphate (S1P), a lipid mediator enriched in blood, bidirectionally controls the trafficking of osteoclast precursors between the circulation and bone marrow cavities via G protein-coupled receptors (GPCRs).


Assuntos
Osso e Ossos/citologia , Osso e Ossos/metabolismo , Ensaios de Migração Celular/métodos , Quimiotaxia , Homeostase , Lisofosfolipídeos/metabolismo , Microscopia de Fluorescência por Excitação Multifotônica , Esfingosina/análogos & derivados , Animais , Células da Medula Óssea/citologia , Movimento Celular , Feminino , Lisofosfolipídeos/deficiência , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Osteoclastos/citologia , Osteoclastos/metabolismo , Esfingosina/deficiência , Esfingosina/metabolismo
7.
Exp Cell Res ; 316(2): 232-44, 2010 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-19796636

RESUMO

Three-dimensional (3D) cell-matrix cultures provide a useful model to analyze and dissect the structural, functional, and mechanical aspects of cell-matrix interactions and motile behavior important for cell and tissue morphogenesis. In the current studies we tested the effects of serum and physiological growth factors on the morphogenetic behavior of human fibroblasts cultured on the surfaces of 3D collagen matrices. Fibroblasts in medium containing serum contracted into clusters, whereas cells in medium containing platelet-derived growth factor (PDGF) were observed to migrate as individuals. The clustering activity of serum appeared to depend on lysophosphatidic acid, required cell contraction based on inhibition by blocking Rho kinase or myosin II, and was reversed upon switching to PDGF. Oncogenic Ras transformed human fibroblasts did not exhibit serum-stimulated cell clustering. Our findings emphasize the importance of cell-specific promigratory and procontractile growth factor environments in the differential regulation of cell motile function and cell morphogenesis.


Assuntos
Agregação Celular/fisiologia , Movimento Celular/fisiologia , Forma Celular/fisiologia , Fibroblastos/citologia , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Actinas/metabolismo , Amidas/farmacologia , Agregação Celular/efeitos dos fármacos , Técnicas de Cultura de Células , Linhagem Celular Transformada , Movimento Celular/efeitos dos fármacos , Forma Celular/efeitos dos fármacos , Colágeno/fisiologia , Inibidores Enzimáticos/farmacologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Lisofosfolipídeos/deficiência , Lisofosfolipídeos/farmacologia , Masculino , Miosina Tipo II/antagonistas & inibidores , Miosina Tipo II/metabolismo , Proteína Oncogênica p21(ras)/genética , Fator de Crescimento Derivado de Plaquetas/farmacologia , Piridinas/farmacologia , Soro/fisiologia , Esfingosina/análogos & derivados , Esfingosina/farmacologia , Vinculina/metabolismo , Quinases Associadas a rho/antagonistas & inibidores , Quinases Associadas a rho/metabolismo
8.
J Exp Med ; 206(11): 2469-81, 2009 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-19808259

RESUMO

During a screen for ethylnitrosourea-induced mutations in mice affecting blood natural killer (NK) cells, we identified a strain, designated Duane, in which NK cells were reduced in blood and spleen but increased in lymph nodes (LNs) and bone marrow (BM). The accumulation of NK cells in LNs reflected a decreased ability to exit into lymph. This strain carries a point mutation within Tbx21 (T-bet), which generates a defective protein. Duane NK cells have a 30-fold deficiency in sphingosine-1-phosphate receptor 5 (S1P5) transcript levels, and S1P5-deficient mice exhibit an egress defect similar to Duane. Chromatin immunoprecipitation confirms binding of T-bet to the S1pr5 locus. S1P-deficient mice exhibit a more severe NK cell egress block, and the FTY720-sensitive S1P1 also plays a role in NK cell egress from LNs. S1P5 is not inhibited by CD69, a property that may facilitate trafficking of activated NK cells to effector sites. Finally, the accumulation of NK cells within BM of S1P-deficient mice was associated with reduced numbers in BM sinusoids, suggesting a role for S1P in BM egress. In summary, these findings identify S1P5 as a T-bet-induced gene that is required for NK cell egress from LNs and BM.


Assuntos
Células da Medula Óssea/citologia , Movimento Celular , Células Matadoras Naturais/citologia , Células Matadoras Naturais/metabolismo , Linfonodos/citologia , Receptores de Lisoesfingolipídeo/metabolismo , Proteínas com Domínio T/metabolismo , Animais , Células da Medula Óssea/efeitos dos fármacos , Contagem de Células , Movimento Celular/efeitos dos fármacos , Etilnitrosoureia , Cloridrato de Fingolimode , Células Matadoras Naturais/efeitos dos fármacos , Linfonodos/efeitos dos fármacos , Ativação Linfocitária/efeitos dos fármacos , Lisofosfolipídeos/deficiência , Lisofosfolipídeos/farmacologia , Camundongos , Camundongos Mutantes , Mutação/genética , Propilenoglicóis/farmacologia , Esfingosina/análogos & derivados , Esfingosina/deficiência , Esfingosina/farmacologia
9.
Circ Res ; 102(8): 950-8, 2008 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-18323526

RESUMO

Activated macrophages acquire a proinflammatory (classic) or antiinflammatory (alternative) phenotype that influences atherosclerosis. The present study investigated whether sphingosine-1-phosphate (S1P), with its known antiinflammatory effects, could regulate the inflammatory phenotype of lipopolysaccharide (LPS)-stimulated mouse macrophages. Activation of macrophages by LPS significantly increases proinflammatory cytokine secretion. Pretreatment of macrophages with 500 nmol/L S1P markedly reduced LPS-mediated secretion of tumor necrosis factor-alpha, monocyte chemoattractant protein-1, and interleukin-12. Such antiinflammatory actions were also evident in LPS-stimulated macrophages treated with the S1P1 receptor-specific agonist SEW2871. Pharmacological antagonism of the S1P1 receptor on macrophages using the S1P1-specific antagonist VPC44116 also blocked proinflammatory cytokine secretion in response to LPS. Studies using bone marrow-derived macrophages from S1P2-deficient mice revealed that the S1P2 receptor did not play a pivotal role in this process. Thus, activation of the S1P1 receptor in mouse macrophages limits the expression of proinflammatory cytokines. Furthermore, we demonstrated that S1P increased arginase I activity and inhibited LPS-induced inducible NO synthase activity in LPS-treated macrophages, again through S1P1 receptor activation on macrophages. Analysis of a 1.7-kb region of the murine inducible NO synthase promoter revealed the presence of putative nuclear factor kappaB, activator protein-1, and STAT-1 response elements. Using inducible NO synthase promoter-reporter constructs, we found that S1P significantly reduced the nuclear factor kappaB-mediated induction of inducible NO synthase. These findings demonstrate an important role for S1P in the regulation of macrophage phenotypic switching. Therefore, we conclude that S1P promotes the production of an alternative antiinflammatory macrophage phenotype through activation of the macrophage S1P1 receptor.


Assuntos
Inflamação , Lisofosfolipídeos/fisiologia , Macrófagos/imunologia , Receptores de Lisoesfingolipídeo/metabolismo , Esfingosina/análogos & derivados , Animais , Células da Medula Óssea , Células Cultivadas , Citocinas/biossíntese , Lipopolissacarídeos/farmacologia , Lisofosfolipídeos/deficiência , Lisofosfolipídeos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Óxido Nítrico Sintase Tipo II , Fenótipo , Receptores de Lisoesfingolipídeo/imunologia , Esfingosina/deficiência , Esfingosina/imunologia , Esfingosina/fisiologia
10.
Science ; 316(5822): 295-8, 2007 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-17363629

RESUMO

Lymphocytes require sphingosine-1-phosphate (S1P) receptor-1 to exit lymphoid organs, but the source(s) of extracellular S1P and whether S1P directly promotes egress are unknown. By using mice in which the two kinases that generate S1P were conditionally ablated, we find that plasma S1P is mainly hematopoietic in origin, with erythrocytes a major contributor, whereas lymph S1P is from a distinct radiation-resistant source. Lymphocyte egress from thymus and secondary lymphoid organs was markedly reduced in kinase-deficient mice. Restoration of S1P to plasma rescued egress to blood but not lymph, and the rescue required lymphocyte expression of S1P-receptor-1. Thus, separate sources provide S1P to plasma and lymph to help lymphocytes exit the low-S1P environment of lymphoid organs. Disruption of compartmentalized S1P signaling is a plausible mechanism by which S1P-receptor-1 agonists function as immunosuppressives.


Assuntos
Medula Óssea/metabolismo , Linfócitos/fisiologia , Lisofosfolipídeos/biossíntese , Lisofosfolipídeos/fisiologia , Esfingosina/análogos & derivados , Animais , Quimiotaxia de Leucócito/fisiologia , Cromatografia Líquida , Endotélio Vascular , Feminino , Células-Tronco Hematopoéticas/metabolismo , Linfócitos/metabolismo , Lisofosfolipídeos/sangue , Lisofosfolipídeos/deficiência , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Receptores de Lisoesfingolipídeo/fisiologia , Esfingosina/biossíntese , Esfingosina/sangue , Esfingosina/deficiência , Esfingosina/fisiologia , Espectrometria de Massas em Tandem
11.
Curr Pharm Des ; 12(5): 575-87, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16472149

RESUMO

Blood platelets are very unique in that they store sphingosine 1-phosphate (Sph-1-P) abundantly (possibly due to the existence of highly active sphingosine kinase and a lack of Sph-1-P lyase) and release this bioactive lipid extracellularly upon stimulation. Vascular endothelial cells (ECs) and smooth muscle cells (SMCs) respond dramatically to this platelet-derived bioactive lipid mainly through a family of G protein-coupled Sph-1-P receptors named S1P1, 2, 3, 4, and 5, originally referred to as EDG-1, 5, 3, 6, and 8, respectively. In fact, the importance of Sph-1-P in platelet-vascular cell interactions has been revealed in a number of recent reports. Through interaction with ECs, Sph-1-P can mediate physiological wound healing processes such as vascular repair, although this important bioactive lipid can become atherogenic and thrombogenic, and cause or aggravate cardiovascular diseases especially under certain pathological conditions. On the other hand, Sph-1-P induces vasoconstriction through interaction with SMCs. It is likely that regulation of Sph-1-P biological activities is important for the therapeutical purpose to control vascular disorders. Particularly, the development of specific S1P receptor agonists or antagonists seems a reasonable strategy to selectively regulate the bioactivity of Sph-1-P, considering that a great diversity of Sph-1-P actions has been reported and that this diversity depends mainly on the S1P receptor subtype involved. In this review, I will summarize recent findings on possible roles of Sph-1-P in vascular biology and its therapeutical implications.


Assuntos
Vasos Sanguíneos/fisiologia , Lisofosfolipídeos/fisiologia , Lisofosfolipídeos/uso terapêutico , Esfingosina/análogos & derivados , Doenças Vasculares/tratamento farmacológico , Animais , Plaquetas/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/fisiologia , Humanos , Lisofosfolipídeos/sangue , Lisofosfolipídeos/deficiência , Músculo Liso Vascular/citologia , Músculo Liso Vascular/efeitos dos fármacos , Neovascularização Patológica/tratamento farmacológico , Esfingosina/sangue , Esfingosina/deficiência , Esfingosina/fisiologia , Esfingosina/uso terapêutico
12.
Blood ; 106(4): 1314-22, 2005 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-15870184

RESUMO

Sphingosine-1-phosphate (S1P) and its receptor S1P1 control T-cell egress from thymus and secondary lymphoid organs (SLOs). To further define the role of S1P1 in lymphocyte trafficking, we performed adoptive transfer experiments and intravital microscopy (IVM) using both S1P1-/- lymphocytes and recipient wild-type (WT) mice treated with FTY720, an immunosuppressant that downmodulates S1P receptors. S1P1 deficiency and FTY720 caused rapid disappearance of T cells from blood, prolonged retention in SLOs, and accumulation in bone marrow, but did not alter interstitial T-cell motility in peripheral lymph nodes (PLNs) as assessed by multiphoton IVM. However, S1P1-/- lymphocytes displayed reduced short-term homing to PLNs due to attenuated integrin-mediated firm arrest in high endothelial venules (HEVs). By contrast, S1P1-/- T cells homed normally to Peyer patches (PPs), whereas S1P1-/- B cells had a marked defect in homing to PPs and arrested poorly in PP HEVs. Therefore, S1P1 not only controls lymphocyte egress from SLOs, but also facilitates in a tissue- and subset-specific fashion integrin activation during homing. Interestingly, FTY720 treatment enhanced accumulation of both S1P1 sufficient and S1P1-/- T cells in PPs by enhancing integrin-mediated arrest in HEVs. Thus, FTY720 exerts unique effects on T-cell traffic in PPs that are independent of T-cell-expressed S1P1.


Assuntos
Quimiotaxia de Leucócito/efeitos dos fármacos , Imunossupressores/farmacologia , Lisofosfolipídeos/fisiologia , Propilenoglicóis/farmacologia , Esfingosina/análogos & derivados , Linfócitos T/fisiologia , Transferência Adotiva , Animais , Quimiotaxia de Leucócito/fisiologia , Cloridrato de Fingolimode , Imunossupressores/imunologia , Integrinas/metabolismo , Linfonodos , Vasos Linfáticos , Contagem de Linfócitos , Linfócitos , Lisofosfolipídeos/deficiência , Lisofosfolipídeos/imunologia , Camundongos , Camundongos Knockout , Microscopia de Vídeo , Nódulos Linfáticos Agregados , Propilenoglicóis/imunologia , Esfingosina/deficiência , Esfingosina/imunologia , Esfingosina/fisiologia , Linfócitos T/efeitos dos fármacos
13.
Mol Cell Neurosci ; 20(2): 271-82, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12093159

RESUMO

Lysophosphatidic acid (LPA) is a bioactive lysophospholipid that produces process retraction and cell rounding through its cognate receptors in neuroblastoma cell lines. Although the expression profile of LPA receptors in developing brains suggests a role for LPA in central nervous system (CNS) development, how LPA influences the morphology of postmitotic CNS neurons remains to be determined. Here we have investigated the effects of exogenous LPA on the morphology of young, postmitotic neurons in primary culture. When treated with LPA, these neurons responded by not only retracting processes but also producing retraction fiber "caps" characterized by fine actin filaments emanating from a dense core. Retraction fiber caps gradually vanished due to the outward spread of regrowing membranes along the fibers, suggesting a role for caps as scaffolds for regrowth of retracted processes. Furthermore, LPA also affects neuronal migration in vitro and in vivo. Taken together, these results implicate LPA as an extracellular lipid signal affecting process outgrowth and migration of early postmitotic neurons during development.


Assuntos
Citoesqueleto de Actina/metabolismo , Diferenciação Celular/fisiologia , Movimento Celular/fisiologia , Córtex Cerebral/embriologia , Lisofosfolipídeos/deficiência , Neuritos/metabolismo , Citoesqueleto de Actina/efeitos dos fármacos , Animais , Diferenciação Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Tamanho Celular/efeitos dos fármacos , Tamanho Celular/fisiologia , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/metabolismo , Citocalasina D/farmacologia , Relação Dose-Resposta a Droga , Feminino , Feto , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Membranas Intracelulares/efeitos dos fármacos , Membranas Intracelulares/metabolismo , Membranas Intracelulares/ultraestrutura , Lisofosfolipídeos/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Proteínas Associadas aos Microtúbulos/efeitos dos fármacos , Proteínas Associadas aos Microtúbulos/metabolismo , Neuritos/efeitos dos fármacos , Neuritos/ultraestrutura , Polímeros/metabolismo , Gravidez , Pseudópodes/efeitos dos fármacos , Pseudópodes/metabolismo , Pseudópodes/ultraestrutura , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Tubulina (Proteína)/efeitos dos fármacos , Tubulina (Proteína)/metabolismo
14.
Kardiologiia ; 31(6): 82-4, 1991 Jun.
Artigo em Russo | MEDLINE | ID: mdl-1833581

RESUMO

In addition to hypertrophy of the myocardium, there are changes in its lipid composition as evidenced by activated lipid peroxidation, lower quantities of unsaturated fatty acids, and higher amounts of saturated phospholipids and lysophospholipids in overload heart failure induced by aortic coarctation. The derangement of the lipid environment may contribute to a decrease in the powerful capacity of membraneous enzyme systems, disturbances in ion transport and cardiac diastolic relaxation, which is one of the causes of heart failure.


Assuntos
Baixo Débito Cardíaco/etiologia , Modelos Animais de Doenças , Metabolismo dos Lipídeos , Peroxidação de Lipídeos/fisiologia , Miocárdio/metabolismo , Animais , Cardiomegalia/complicações , Ácidos Graxos Insaturados/metabolismo , Lisofosfolipídeos/deficiência , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...