Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Mol Med ; 22: 115-123, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27031787

ABSTRACT

Endothelial cell (EC) dysfunction is a critical mediator of the acute respiratory distress syndrome (ARDS). Recent studies have demonstrated that stromal cell-derived factor 1α (SDF-1α) promotes EC barrier integrity. Our previous studies used a SDF-1α analogue CTCE-0214 (CTCE) in experimental sepsis and demonstrated that it attenuated vascular leak and modulated microRNA (miR) levels. We examined the hypothesis that CTCE improves EC function in lipopolysaccharide (LPS)-induced ARDS through increasing miR-126 expression. Human microvascular endothelial cells (HMVECs) were treated with thrombin to disrupt the EC integrity followed by incubation with CTCE or SDF-1α. Barrier function was determined by trans-endothelial electrical resistance assay. CTCE-induced alterations in miRNA expression and signaling pathways involved in barrier function were determined. Thrombin-induced vascular leak was abrogated by both CTCE and SDF-1α. CTCE also prevented thrombin-induced decreases of vascular endothelial (VE)-cadherin cell surface expression and expansion of the intercellular space. CTCE increased miR-126 levels and induced activation of AKT/Rac 1 signaling. Cotreatment with a miR-126 inhibitor blocked the protective effects of CTCE on AKT activation and endothelial permeability. In subsequent in vivo studies, ARDS was induced by intratracheal instillation of LPS. Intravenous injection of CTCE diminished the injury severity as evidenced by significant reductions in protein, immune cells, inflammatory cytokines and chemokines in the bronchoalveolar lavage fluid, increased miR-126 expression and decreased pulmonary vascular leak and alveolar edema. Taken together, our data show that CTCE improves endothelial barrier integrity through increased expression of miR-126 and activation of Rac 1 signaling and represents an important potential therapeutic strategy in ARDS.

2.
Lipids Health Dis ; 14: 66, 2015 Jul 04.
Article in English | MEDLINE | ID: mdl-26138336

ABSTRACT

BACKGROUND: Periodontitis is a bacteria-driven inflammatory bone loss disease. Previous studies showed that the oral pathogen Aggregatibacter actinomycetemcomitans (A. actinomycetemcomitans) stimulated the generation of sphingosine 1 phosphate (S1P). In addition, S1P signaling regulated the migration of osteoclast precursors and affected osteoclastogenesis. Furthermore, treatment with FTY720 (also called fingolimod, a modulator of multiple S1P receptors) alleviated osteoporosis and suppressed arthritis in animals. This study determined the effect of FTY720 on proinflammatory cytokine production and osteoclastogenesis in murine bone marrow cells with or without A. actinomycetemcomitans stimulation. METHODS: Murine bone marrow-derived monocytes and macrophages (BMMs) were treated with vehicle ethanol or FTY720, and were either unstimulated or stimulated for 0.5 to 6 h with A. actinomycetemcomitans. The protein levels of interleukin (IL)-1ß, IL-6, and tumor necrosis factor (TNF)-α in the media of BMMs were quantified by enzyme-linked immunosorbent assay (ELISA). Protein expressions, including phosphorylated phosphoinositide 3-kinase (p-PI3K), p-Akt, p-extracellular signal-regulated kinase (p-ERK), PI3K, Akt, and ERK were evaluated by Western blot. In addition, murine bone marrow-derived pre-osteoclasts were treated with macrophage colony-stimulating factor (M-CSF) and receptor activator of nuclear factor kappa-B ligand (RANKL) for three days. Then the cells were treated with either vehicle or FTY720 and were either unstimulated or stimulated with A. actinomycetemcomitans for 4 to 24 h. Control cells were treated with M-CSF alone with or without bacterial stimulation. Osteoclasts were stained by tartrate-resistant acid phosphatase (TRAP) staining. The mRNA levels of osteoclastogenic factors, including nuclear factor of activated T-cells cytoplasmic calcineurin-dependent 1 (Nfatc1), cathepsin K (Ctsk), acid phosphatase 5 (Acp5), osteoclast-associated receptor (Oscar), and RANKL were quantified by quantitative real-time polymerase chain reaction (PCR). RESULTS: FTY720 dose-dependently inhibited IL-1ß, IL-6, and TNF-α protein levels induced by A. actinomycetemcomitans in BMMs compared with controls. Additionally, FTY720 attenuated p-PI3K, p-Akt, and p-ERK expressions induced by A. actinomycetemcomitans. Furthermore, FTY720 suppressed osteoclastogenesis in bone marrow-derived pre-osteoclasts with or without bacterial stimulation and reduced the mRNA levels of Nfatc1, Ctsk, Acp5, and Oscar, but not RANKL in bone marrow-derived pre-osteoclasts. CONCLUSION: FTY720 inhibited proinflammatory cytokine production and suppressed osteoclastogenesis, supporting FTY720 as a potential therapy for inflammatory bone loss diseases.


Subject(s)
Aggregatibacter actinomycetemcomitans/physiology , Cytokines/metabolism , Fingolimod Hydrochloride/pharmacology , Inflammation Mediators/metabolism , Osteoclasts/metabolism , Osteoclasts/microbiology , Osteogenesis/drug effects , Animals , Bone Marrow Cells/cytology , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation/drug effects , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Macrophages/drug effects , Macrophages/metabolism , Macrophages/microbiology , Male , Mice, Inbred C57BL , Osteoclasts/drug effects , Osteoclasts/pathology , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
3.
Mech Dev ; 136: 123-32, 2015 May.
Article in English | MEDLINE | ID: mdl-25575930

ABSTRACT

Endothelial to mesenchymal transition (EMT) that occurs during cardiac outflow tract (OFT) development is critical for formation of the semilunar valves. Fibulin-1 (Fbln1) is an extracellular matrix protein that is present at several sites of EMT, including the OFT (i.e., E9.5-10.5). The aim of this study was to determine the role of Fbln1 in EMT during the earliest events of OFT development. Examination of proximal OFT cushions in Fbln1 null embryos detected hypercellularity at both E9.5 (93% increase; p = 0.002) and E10.5 (43% increase; p = 0.01) as compared to wild type, suggesting that Fbln1 normally suppresses OFT endocardial cushion EMT. This was supported by studies of proximal OFT cushion explants, which showed that explants from Fbln1 null embryos displayed a 58% increase in cells migrating from the explants as compared to wild type (p = 0.005). We next evaluated the effects of Fbln1 deficiency on the expression of factors that regulate proximal OFT EMT. At E9.5, Fbln1 null proximal OFT endocardium and EMT-derived mesenchyme showed increased TGFß2 (58% increase; p = 0.01) and increased Snail1-positive nuclei (27% increase; p = 0.0003). Histological examination of OFT cushions in Fbln1 null embryos (E9.5) also detected cells present in the cushion that were determined to be erythrocytes based on round morphology, autofluorescence, and positive staining for hemoglobin. Erythrocytes were also detected in Fbln1 null OFT cushions at E10.5. Together, the findings indicate that Fbln1 normally suppresses proximal OFT EMT preventing proximal cushion hypercellularity and blood cell accumulation.


Subject(s)
Calcium-Binding Proteins/metabolism , Endocardial Cushions/metabolism , Endocardium/metabolism , Extracellular Matrix Proteins/metabolism , Myocardium/metabolism , Animals , Apoptosis , Calcium-Binding Proteins/genetics , Cell Proliferation , Endocardial Cushions/cytology , Endocardium/cytology , Extracellular Matrix Proteins/genetics , Mice , Mice, Knockout , Myocardium/cytology
4.
Biochim Biophys Acta ; 1841(10): 1403-1412, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25009123

ABSTRACT

Loss of endothelial barrier function is implicated in the etiology of metastasis, atherosclerosis, sepsis and many other diseases. Studies suggest that sphingosine-1-phosphate (S1P), particularly HDL-bound S1P (HDL-S1P) is essential for endothelial barrier homeostasis and that HDL-S1P may be protective against the loss of endothelial barrier function in disease. This review summarizes evidence providing mechanistic insights into how S1P maintains endothelial barrier function, highlighting the recent findings that implicate the major S1P carrier, HDL, in the maintenance of the persistent S1P-signaling needed to maintain endothelial barrier function. We review the mechanisms proposed for HDL maintenance of persistent S1P-signaling, the evidence supporting these mechanisms and the remaining fundamental questions.

5.
J Am Soc Nephrol ; 25(5): 1028-36, 2014 May.
Article in English | MEDLINE | ID: mdl-24357674

ABSTRACT

Cubilin is an endocytic receptor highly expressed in renal proximal tubules, where it mediates uptake of albumin and filtered forms of apoA-I/HDL. Cubilin deficiency leads to urinary loss of albumin and apoA-I; however, the consequences of cubilin loss on the homeostasis of blood albumin and apoA-I/HDL have not been studied. Using mice heterozygous for cubilin gene deletion (cubilin HT mice), we show that cubilin haploinsufficiency leads to reduced renal proximal tubular uptake of albumin and apoA-I and significantly increased urinary loss of albumin and apoA-I. Moreover, cubilin HT mice displayed significantly decreased blood levels of albumin, apoA-I, and HDL. The levels of albumin and apoA-I protein or mRNA expressed in the liver, kidney, or intestine of cubilin HT mice did not change significantly. The clearance rate of small HDL3 particles (density>1.13 g/ml) from the blood increased significantly in cubilin HT mice. In contrast, the rate of clearance of larger HDL2 particles from the blood did not change significantly, indicating a decreased half-life for HDL particles capable of filtering through the glomerulus. On the basis of these findings, we conclude that cubilin deficiency reduces renal salvage and delivery back to the blood of albumin and apoA-I, which decreases blood levels of albumin and apoA-I/HDL. These findings raise the possibility that therapeutic increase of renal cubilin expression might reduce proteinuria and increase blood levels of albumin and HDL.


Subject(s)
Albuminuria/etiology , Albuminuria/genetics , Apolipoprotein A-I/urine , Lipoproteins, HDL/blood , Receptors, Cell Surface/physiology , Albumins/antagonists & inhibitors , Albumins/metabolism , Albuminuria/metabolism , Animals , Apolipoprotein A-I/antagonists & inhibitors , Apolipoprotein A-I/blood , Gene Deletion , Genetic Carrier Screening , Humans , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Lipoproteins, HDL/antagonists & inhibitors , Lipoproteins, HDL/biosynthesis , Lipoproteins, HDL3/antagonists & inhibitors , Lipoproteins, HDL3/blood , Lipoproteins, HDL3/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/genetics
6.
J Biol Chem ; 287(53): 44645-53, 2012 Dec 28.
Article in English | MEDLINE | ID: mdl-23135269

ABSTRACT

Sphingosine 1-phosphate (S1P) is a blood-borne lysosphingolipid that acts to promote endothelial cell (EC) barrier function. In plasma, S1P is associated with both high density lipoproteins (HDL) and albumin, but it is not known whether the carriers impart different effects on S1P signaling. Here we establish that HDL-S1P sustains EC barrier longer than albumin-S1P. We showed that the sustained barrier effects of HDL-S1P are dependent on signaling by the S1P receptor, S1P1, and involve persistent activation of Akt and endothelial NOS (eNOS), as well as activity of the downstream NO target, soluble guanylate cyclase (sGC). Total S1P1 protein levels were found to be higher in response to HDL-S1P treatment as compared with albumin-S1P, and this effect was not associated with increased S1P1 mRNA or dependent on de novo protein synthesis. Several pieces of evidence indicate that long term EC barrier enhancement activity of HDL-S1P is due to specific effects on S1P1 trafficking. First, the rate of S1P1 degradation, which is proteasome-mediated, was slower in HDL-S1P-treated cells as compared with cells treated with albumin-S1P. Second, the long term barrier-promoting effects of HDL-S1P were abrogated by treatment with the recycling blocker, monensin. Finally, cell surface levels of S1P1 and levels of S1P1 in caveolin-enriched microdomains were higher after treatment with HDL-S1P as compared with albumin-S1P. Together, the findings reveal S1P carrier-specific effects on S1P1 and point to HDL as the physiological mediator of sustained S1P1-PI3K-Akt-eNOS-sGC-dependent EC barrier function.


Subject(s)
Human Umbilical Vein Endothelial Cells/metabolism , Lipoproteins, HDL/metabolism , Lysophospholipids/metabolism , Receptors, Lysosphingolipid/metabolism , Serum Albumin/metabolism , Sphingosine/analogs & derivatives , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Humans , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Protein Transport , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Lysosphingolipid/genetics , Signal Transduction , Sphingosine/metabolism
7.
Pediatr Cardiol ; 33(7): 1154-9, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22411716

ABSTRACT

Despite the emerging relevance of high-density lipoprotein (HDL) in the inflammatory cascade and vascular barrier integrity, HDL levels in children undergoing cardiac surgery are unexplored. As a measure of HDL levels, the HDL-cholesterol (HDL-C) in single-ventricle patients was quantified before and after the Fontan operation, and it was determined whether relationships existed between the duration and the type of postoperative pleural effusions. The study prospectively enrolled 12 children undergoing the Fontan operation. Plasma HDL-C levels were measured before and after cardiopulmonary bypass. The outcome variables of interest were the duration and type of chest tube drainage (chylous vs. nonchylous). The Kendall rank correlation coefficient and the Wilcoxon rank sum test were used. There were 11 complete observations. The median preoperative HDL-C level for all the subjects was 30 mg/dl (range, 24-53 mg/dl), and the median postcardiopulmonary bypass level was 21 mg/dl (range, 14-46 mg/dl) (p = 0.004). There was a tendency toward a moderate inverse correlation (-0.42) between the postcardiopulmonary bypass HDL-C level and the duration of chest tube drainage, but the result was not statistically significant (p = 0.07). In the chylous effusion group, the median postcardiopulmonary bypass HDL-C tended to be lower (16 vs. 23 mg/dl; p = 0.09). After the Fontan operation, the plasma HDL-C levels in children are significantly reduced. It is reasonable to conclude that the reduction in HDL-C reflects reduced plasma levels of HDL particles, which may have pertinent implications in postoperative pleural effusions given the antiinflammatory and endothelial barrier functions of HDL.


Subject(s)
Cholesterol, HDL/blood , Fontan Procedure , Heart Defects, Congenital/blood , Heart Defects, Congenital/surgery , Cardiopulmonary Bypass , Chest Tubes , Child , Child, Preschool , Female , Humans , Infant , Male , Pleural Effusion/blood , Postoperative Complications/blood , Prospective Studies , Statistics, Nonparametric , Treatment Outcome
8.
BMC Cancer ; 11: 520, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-22172030

ABSTRACT

BACKGROUND: Perturbing Hsp90 chaperone function targets hypoxia inducible factor (HIF) function in a von Hippel-Lindau (VHL) independent manner, and represents an approach to combat the contribution of HIF to cell renal carcinoma (CCRCC) progression. However, clinical trials with the prototypic Hsp90 inhibitor 17-AAG have been unsuccessful in halting the progression of advanced CCRCC. METHODS: Here we evaluated a novel next generation small molecule Hsp90 inhibitor, EC154, against HIF isoforms and HIF-driven molecular and functional endpoints. The effects of EC154 were compared to those of the prototypic Hsp90 inhibitor 17-AAG and the histone deacetylase (HDAC) inhibitor LBH589. RESULTS: The findings indicate that EC154 is a potent inhibitor of HIF, effective at doses 10-fold lower than 17-AAG. While EC154, 17-AAG and the histone deacetylase (HDAC) inhibitor LBH589 impaired HIF transcriptional activity, CCRCC cell motility, and angiogenesis; these effects did not correlate with their ability to diminish HIF protein expression. Further, our results illustrate the complexity of HIF targeting, in that although these agents suppressed HIF transcripts with differential dynamics, these effects were not predictive of drug efficacy in other relevant assays. CONCLUSIONS: We provide evidence for EC154 targeting of HIF in CCRCC and for LBH589 acting as a suppressor of both HIF-1 and HIF-2 activity. We also demonstrate that 17-AAG and EC154, but not LBH589, can restore endothelial barrier function, highlighting a potentially new clinical application for Hsp90 inhibitors. Finally, given the discordance between HIF activity and protein expression, we conclude that HIF expression is not a reliable surrogate for HIF activity. Taken together, our findings emphasize the need to incorporate an integrated approach in evaluating Hsp90 inhibitors within the context of HIF suppression.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Renal Cell/drug therapy , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Hypoxia-Inducible Factor 1/antagonists & inhibitors , Kidney Neoplasms/drug therapy , Neoplasm Proteins/antagonists & inhibitors , Blotting, Western , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Disease Progression , Electric Impedance , HSP90 Heat-Shock Proteins/metabolism , Humans , Hydroxamic Acids/pharmacology , Hypoxia-Inducible Factor 1/metabolism , Indoles , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Luciferases/metabolism , Neoplasm Proteins/metabolism , Panobinostat , Real-Time Polymerase Chain Reaction , Transcription, Genetic/drug effects , Vascular Endothelial Growth Factor A/drug effects , Vascular Endothelial Growth Factor A/metabolism
9.
Lipids Health Dis ; 10: 70, 2011 May 09.
Article in English | MEDLINE | ID: mdl-21554699

ABSTRACT

BACKGROUND: The lysosphingolipid sphingosine 1-phosphate (S1P) is carried in the blood in association with lipoproteins, predominantly high density lipoproteins (HDL). Emerging evidence suggests that many of the effects of HDL on cardiovascular function may be attributable to its S1P cargo. METHODS: Here we have evaluated how levels of S1P and related sphingolipids in an HDL-containing fraction of human serum correlate with occurrence of ischemic heart disease (IHD). To accomplish this we used liquid chromatography-mass spectrometry to measure S1P levels in the HDL-containing fraction of serum (depleted of LDL and VLDL) from 204 subjects in the Copenhagen City Heart Study (CCHS). The study group consisted of individuals having high serum HDL cholesterol (HDL-C) (females:≥ 73.5 mg/dL; males:≥ 61.9 mg/dL) and verified IHD; subjects with high HDL-C and no IHD; individuals with low HDL-C (females:≤ 38.7 mg/dL; males:≤ 34.1 mg/dL) and IHD, and subjects with low HDL-C and no IHD. RESULTS: The results show a highly significant inverse relationship between the level of S1P in the HDL-containing fraction of serum and the occurrence of IHD. Furthermore, an inverse relationship with IHD was also observed for two other sphingolipids, dihydro-S1P and C24:1-ceramide, in the HDL-containing fraction of serum. Additionally, we demonstrated that the amount of S1P on HDL correlates with the magnitude of HDL-induced endothelial cell barrier signaling. CONCLUSIONS: These findings indicate that compositional differences of sphingolipids in the HDL-containing fraction of human serum are related to the occurrence of IHD, and may contribute to the putative protective role of HDL in IHD.


Subject(s)
Lipoproteins, HDL/blood , Lysophospholipids/blood , Myocardial Ischemia/blood , Myocardial Ischemia/epidemiology , Sphingosine/analogs & derivatives , Cell Movement/drug effects , Ceramides/blood , Chemical Fractionation , Chromatography, Liquid , Denmark/epidemiology , Electric Impedance , Endothelial Cells/drug effects , Endothelial Cells/pathology , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Female , Humans , Lysophospholipids/pharmacology , Male , Mass Spectrometry , Middle Aged , ROC Curve , Sphingolipids/blood , Sphingosine/blood , Sphingosine/pharmacology
10.
World J Biol Chem ; 1(10): 291-7, 2010 Oct 26.
Article in English | MEDLINE | ID: mdl-21537462

ABSTRACT

Blood vessels either form de novo through the process of vasculogenesis or through angiogenesis that involves the sprouting and proliferation of endothelial cells in pre-existing blood vessels. A complex interactive network of signaling cascades downstream from at least three of the nine known G-protein-coupled sphingosine-1-phosphate (S1P) receptors act as a prime effector of neovascularization that occurs in embryonic development and in association with various pathologies. This review focuses on the current knowledge of the roles of S1P signaling in vasculogenesis and angiogenesis, with particular emphasis on vascular cell adhesion and motility responses.

11.
Histochem Cell Biol ; 132(5): 559-65, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19693531

ABSTRACT

Fibulin-1 is a fibrinogen-binding blood protein and a component of many extracellular matrices (ECM) including those of blood vessels. In this study, the deposition patterns of fibulin-1 and fibrinogen were examined in human coronary artery atherosclerotic lesions excised by atherectomy from 20 patients. Fibulin-1 deposition was found to be closely overlapping with fibrinogen located within the atherosclerotic lesions and in regions containing fresh thrombi. Pronounced intracellular fibulin-1 immunostaining was apparent in lesion areas rich in macrophages and foam cells, although THP-1 macrophages and foam cells were found not to express fibulin-1. Strong ECM deposition of fibulin-1 was observed in acellular atheromatous and myxomatous regions. By contrast, fibulin-1 was present at relatively low levels in the ECM associated with smooth muscle cells within and outside of lesions and was not detected in sclerotic regions. These results reveal the pattern of fibulin-1 within human atherosclerotic lesions and highlight the potential for fibulin-1, perhaps derived from the blood and acting in conjunction with fibrinogen, to play a role in the etiology and cardiovascular disease progression, particularly with respect to thrombotic aspects of atherosclerosis.


Subject(s)
Atherosclerosis/metabolism , Atherosclerosis/pathology , Calcium-Binding Proteins/metabolism , Fibrinogen/metabolism , Adult , Aged , Antibody Specificity , Calcium-Binding Proteins/immunology , Cells, Cultured , Coronary Artery Disease/metabolism , Coronary Artery Disease/pathology , Fibrinogen/immunology , Humans , Immunohistochemistry , Middle Aged
12.
Dev Dyn ; 237(10): 2918-25, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18816835

ABSTRACT

Embryonic mouse allantoic tissue (E8.5) was cultured in hanging drops to generate a three-dimensional vascular micro-tissue. The resulting tissue spheroids had an inner network of small diameter vessels expressing platelet endothelial cell adhesion molecule-1 (PECAM-1) and an outer layer of cells expressing SMalphaA, SM22-alpha, and SM-MHC. In a subsequent phase of culture, the fusion-promoting activity of vascular endothelial growth factor (VEGF) was used to transform the inner network of small diameter endothelial tubes into a contiguous layer of cells expressing PECAM-1, CD34, and VE-cadherin that circumscribed a central lumen-like cavity. The blood vessel-like character of the VEGF-treated spheroids was further demonstrated by their physiologically relevant vasodilatory and contractile responses, including contraction induced by KCl and relaxation stimulated by high-density lipoproteins and acetylcholine-induced nitric oxide production.


Subject(s)
Neovascularization, Physiologic/drug effects , Vascular Endothelial Growth Factor A/pharmacology , Actins/metabolism , Allantois/blood supply , Allantois/drug effects , Allantois/embryology , Allantois/metabolism , Animals , Endothelial Cells/metabolism , Histocompatibility Antigens/metabolism , Mice , Mice, Inbred ICR
13.
J Biol Chem ; 283(36): 25074-81, 2008 Sep 05.
Article in English | MEDLINE | ID: mdl-18606817

ABSTRACT

High density lipoproteins (HDL) are major plasma carriers of sphingosine 1-phosphate (S1P). Here we show that HDL increases endothelial barrier integrity as measured by electric cell substrate impedance sensing. S1P was implicated as the mediator in this process through findings showing that pertussis toxin, an inhibitor of Gi-coupled S1P receptors, as well as antagonists of the S1P receptor, S1P1, inhibited barrier enhancement by HDL. Additional findings show that HDL stimulates endothelial cell activation of Erk1/2 and Akt, signaling pathway intermediates that have been implicated in S1P-dependent endothelial barrier activity. HDL was also found to promote endothelial cell motility, a process that may also relate to endothelial barrier function in the context of a vascular injury response. The effects of HDL on endothelial cell Erk1/2 and Akt activation and motility were suppressed by pertussis toxin and S1P1 antagonists. However, both HDL-induced barrier enhancement and HDL-induced motility showed a greater dependence on Akt activation as compared with Erk1/2 activation. Together, the findings indicate that HDL has endothelial barrier promoting activities, which are attributable to its S1P component and signaling through the S1P1/Akt pathway.


Subject(s)
Cell Movement/physiology , Endothelial Cells/metabolism , Lipoproteins, HDL/metabolism , Lysophospholipids/metabolism , MAP Kinase Signaling System/physiology , Receptors, Lysosphingolipid/metabolism , Sphingosine/analogs & derivatives , Cell Movement/drug effects , Cells, Cultured , Endothelial Cells/cytology , Enzyme Activation/drug effects , Enzyme Activation/physiology , Humans , Lipoproteins, HDL/pharmacology , Lysophospholipids/pharmacology , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Pertussis Toxin/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Sphingosine/metabolism , Sphingosine/pharmacology
14.
J Lipid Res ; 48(11): 2325-33, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17698855

ABSTRACT

The lysosphingolipid sphingosine 1-phosphate (S1P) is a component of HDL. Findings from a growing number of studies indicate that S1P is a mediator of many of the cardiovascular effects of HDL, including the ability to promote vasodilation, vasoconstriction, and angiogenesis, protect against ischemia/reperfusion injury, and inhibit/reverse atherosclerosis. These latter cardioprotective effects are being shown to involve the S1P-mediated suppression of inflammatory processes, including reduction of the endothelial expression of monocyte and lymphocyte adhesion molecules, decreased recruitment of polymorphonuclear cells to sites of infarction, and blocking of cardiomyocyte apoptosis after myocardial infarction. This review article summarizes the evidence that S1P as a component of HDL serves to regulate vascular cell and lymphocyte behaviors associated with cardiovascular (patho)physiology.


Subject(s)
Cardiovascular System/drug effects , Lipoproteins, HDL/physiology , Lysophospholipids/physiology , Signal Transduction/physiology , Sphingosine/analogs & derivatives , ATP-Binding Cassette Transporters/physiology , Animals , Atherosclerosis/etiology , Cell Movement/drug effects , Cystic Fibrosis Transmembrane Conductance Regulator/physiology , Endothelial Cells/drug effects , Endothelial Cells/physiology , Humans , Immunologic Factors/physiology , Muscle, Smooth, Vascular/physiology , Phosphotransferases (Alcohol Group Acceptor)/blood , Reperfusion Injury/prevention & control , Sphingosine/physiology , Vasoconstriction/physiology , Vasodilation/physiology
15.
J Biol Chem ; 279(48): 50580-90, 2004 Nov 26.
Article in English | MEDLINE | ID: mdl-15377653

ABSTRACT

Here we have investigated the role of sphingosine-1-phosphate (S1P) signaling in the process of vasculogenesis in the mouse embryo. At stages preceding the formation of blood vessels (7.5-8 dpc) in the embryo proper, yolk sac, and allantois, the S1P receptor S1P(2) is expressed in conjunction with S1P(1) and/or S1P(3). Additionally, sphingosine kinase-2 (SK2), an enzyme that catalyzes the formation of S1P, is expressed in these tissues throughout periods of vasculogenesis. Using the cultured mouse allantois explant model of blood vessel formation, we found that vasculogenesis was dependent on S1P signaling. We showed that S1P could replace the ability of serum to promote vasculogenesis in cultured allantois explants. Instead of small poorly reticulated clusters of rounded endothelial cells that formed under serum-free conditions, S1P promoted the formation of elongated endothelial cells that arranged into expansive branched networks of capillary-like vessels. These effects could not be reproduced by vascular endothelial growth factor or basic fibroblast growth factor administration. The ability of S1P to promote blood vessel formation was not due to effects on cell survival or on changes in numbers of endothelial cells (Flk1(+)/PECAM(+)), angioblasts (Flk1(+)/PECAM(-)), or undifferentiated mesodermal cells (Flk1(-)/PECAM(-)). The S1P effect on blood vessel formation was attributed to it promoting migratory activities of angioblasts and early endothelial cells required for the expansion of vascular networks. Together, our findings suggest that migratory events critical to the de novo formation of blood vessels are under the influence of S1P, possibly synthesized via the action of SK2, with signaling mediated by S1P receptors that include S1P(1), S1P(2), and S1P(3).


Subject(s)
Cell Movement/physiology , Lysophospholipids/metabolism , Neovascularization, Physiologic/physiology , Signal Transduction/physiology , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Allantois/blood supply , Animals , Mice , Phosphotransferases (Alcohol Group Acceptor)/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...