Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters










Publication year range
1.
Thromb Res ; 227: 17-24, 2023 07.
Article in English | MEDLINE | ID: mdl-37207560

ABSTRACT

BACKGROUND: Antithrombotic therapy is inevitably associated with a risk for bleeding and these bleeding complications can be life-threatening. Recently, specific reversal agents were developed for the direct factor Xa and thrombin inhibitors (DOACs). However, next to the fact that these agents are relatively expensive, the use of selective reversal agents complicates treatment of bleeding patients in practice. In a series of screening experiments, we discovered a class of cyclodextrins with procoagulant properties. In this study we characterize a lead compound, OKL-1111, and demonstrate its potential use as a universal reversal agent. OBJECTIVES: To assess the anticoagulant reversal properties of OKL-1111, in vitro and in vivo. METHODS: The effect of OKL-1111 on coagulation in the absence and presence of DOACs was investigated in a thrombin generation assay. Its reversal effect on a variety of anticoagulants in vivo was investigated in a rat tail cut bleeding model. A possible prothrombotic action of OKL-1111 was assessed in a Wessler model in rabbits. RESULTS: OKL-1111 concentration-dependently reversed the in vitro anticoagulant effects of dabigatran, rivaroxaban, apixaban and edoxaban in the thrombin generation assay. Also in the absence of a DOAC, OKL-1111 concentration-dependently accelerated coagulation in this assay, but did not initiate coagulation. The reversal effect was also seen for all DOACs in the rat tail cut bleeding model. In addition, when tested with other anticoagulants, OKL-1111 also reversed the anticoagulant effect of the vitamin K antagonist warfarin, the low molecular weight heparin enoxaparin, the pentasaccharide fondaparinux and the platelet inhibitor clopidogrel in vivo. OKL-1111 did not have prothrombotic effects in the Wessler model. CONCLUSION: OKL-1111 is a procoagulant cyclodextrin with a currently unknown working mechanism that has potential to become a universal reversal agent for anticoagulants and platelet inhibitors.


Subject(s)
Anticoagulants , Thrombin , Animals , Rabbits , Rats , Anticoagulants/adverse effects , Thrombin/therapeutic use , Factor Xa Inhibitors/pharmacology , Factor Xa Inhibitors/therapeutic use , Dabigatran/therapeutic use , Rivaroxaban/therapeutic use , Hemorrhage/chemically induced , Administration, Oral
2.
Pregnancy Hypertens ; 19: 100-105, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31927322

ABSTRACT

OBJECTIVES: Ceramide is a sphingolipid with anti-angiogenic and pro-apoptotic properties that has shown to be increased in plasma of women with pre-eclampsia. We aimed to compare plasma and placental sphingolipid content among normotensive pregnant women and pre-eclamptic women with and without HELLP syndrome and we aimed to assess whether ceramide is related to hypertension and proteinuria in pre-eclampsia. STUDY DESIGN: Case-control study. Participants were recruited from the Department of Obstetrics at the Academic Medical Center in Amsterdam, The Netherlands. In total 48 pregnant women were included: 24 with pre-eclampsia and 24 normotensive controls. Of the 24 pre-eclamptic women, 11 had HELLP syndrome. MAIN OUTCOME MEASURES: Plasma and placental ceramide content and correlation with blood pressure and protein excretion in pre-eclampsia. RESULTS: Total plasma, but not placental, ceramide was higher in pre-eclamptic women with HELLP syndrome (11200 95% CI 9531-12870 nmol/ml, n = 11) compared to pre-eclamptic women without HELLP (7413 95% CI 5928-8898 nmol/ml, n = 13, p < 0.001) and normotensive pregnant women (7404 95% CI 6695-8112 nmol/ml, n = 24, p < 0.001). Maternal circulating ceramide levels were strongly associated with proteinuria (r = 0.621, n = 24, p = 0.001) in pre-eclamptic women and inversely correlated with gestational age at delivery (r = 0.771, p < 0.01) in pre-eclamptic women with HELLP syndrome. Plasma ceramide was not correlated with blood pressure. CONCLUSION: Plasma but not placental ceramide content is increased in women with pre-eclampsia and HELLP syndrome. The strong positive correlation with proteinuria and the inverse correlation with gestational age at delivery indicate that excess plasma ceramide may contribute to the pathophysiology of pre-eclampsia and HELLP.


Subject(s)
Ceramides/metabolism , HELLP Syndrome/blood , Pre-Eclampsia/blood , Proteinuria/blood , Adult , Case-Control Studies , Female , Gestational Age , Humans , Placenta/metabolism , Platelet Count , Pregnancy
3.
J Am Heart Assoc ; 5(5)2016 05 20.
Article in English | MEDLINE | ID: mdl-27207969

ABSTRACT

BACKGROUND: Sphingosine-1-phosphate plays vital roles in cardiomyocyte physiology, myocardial ischemia-reperfusion injury, and ischemic preconditioning. The function of the cardiomyocyte sphingosine-1-phosphate receptor 1 (S1P1) in vivo is unknown. METHODS AND RESULTS: Cardiomyocyte-restricted deletion of S1P1 in mice (S1P1 (α) (MHCC) (re)) resulted in progressive cardiomyopathy, compromised response to dobutamine, and premature death. Isolated cardiomyocytes from S1P1 (α) (MHCC) (re) mice revealed reduced diastolic and systolic Ca(2+) concentrations that were secondary to reduced intracellular Na(+) and caused by suppressed activity of the sarcolemmal Na(+)/H(+) exchanger NHE-1 in the absence of S1P1. This scenario was successfully reproduced in wild-type cardiomyocytes by pharmacological inhibition of S1P1 or sphingosine kinases. Furthermore, Sarcomere shortening of S1P1 (α) (MHCC) (re) cardiomyocytes was intact, but sarcomere relaxation was attenuated and Ca(2+) sensitivity increased, respectively. This went along with reduced phosphorylation of regulatory myofilament proteins such as myosin light chain 2, myosin-binding protein C, and troponin I. In addition, S1P1 mediated the inhibitory effect of exogenous sphingosine-1-phosphate on ß-adrenergic-induced cardiomyocyte contractility by inhibiting the adenylate cyclase. Furthermore, ischemic precondtioning was abolished in S1P1 (α) (MHCC) (re) mice and was accompanied by defective Akt activation during preconditioning. CONCLUSIONS: Tonic S1P1 signaling by endogenous sphingosine-1-phosphate contributes to intracellular Ca(2+) homeostasis by maintaining basal NHE-1 activity and controls simultaneously myofibril Ca(2+) sensitivity through its inhibitory effect on adenylate cyclase. Cardioprotection by ischemic precondtioning depends on intact S1P1 signaling. These key findings on S1P1 functions in cardiac physiology may offer novel therapeutic approaches to cardiac diseases.


Subject(s)
Calcium/metabolism , Cardiomyopathies/genetics , Ischemic Preconditioning, Myocardial , Myocardial Reperfusion Injury/genetics , Myocytes, Cardiac/metabolism , Receptors, Lysosphingolipid/genetics , Sodium-Hydrogen Exchangers/metabolism , Action Potentials , Adenylyl Cyclases/metabolism , Animals , Blotting, Western , Cardiac Myosins/metabolism , Cardiomyopathies/diagnostic imaging , Cardiomyopathies/metabolism , Carrier Proteins/metabolism , Echocardiography , Magnetic Resonance Imaging , Mice , Mice, Knockout , Myocardial Reperfusion Injury/metabolism , Myocytes, Cardiac/drug effects , Myosin Light Chains/metabolism , Phosphorylation , Positron-Emission Tomography , Real-Time Polymerase Chain Reaction , Receptors, Lysosphingolipid/antagonists & inhibitors , Sarcomeres/metabolism , Sphingosine-1-Phosphate Receptors , Troponin I/metabolism
4.
PLoS One ; 9(3): e91346, 2014.
Article in English | MEDLINE | ID: mdl-24632890

ABSTRACT

Sphingosine-1-phosphate (S1P) is an agonist for five distinct G-protein coupled receptors, that is released by platelets, mast cells, erythrocytes and endothelial cells. S1P promotes endothelial cell barrier function and induces release of endothelial cell-specific storage-organelles designated Weibel-Palade bodies (WPBs). S1P-mediated enhancement of endothelial cell barrier function is dependent on S1P receptor 1 (S1PR1) mediated signaling events that result in the activation of the small GTPase Rac1. Recently, we have reported that Rac1 regulates epinephrine-induced WPB exocytosis following its activation by phosphatidylinositol-3,4,5-triphosphate-dependent Rac exchange factor 1 (PREX1). S1P has also been described to induce WPB exocytosis. Here, we confirm that S1P induces release of WPBs using von Willebrand factor (VWF) as a marker. Using siRNA mediated knockdown of gene expression we show that S1PR1 is not involved in S1P-mediated release of WPBs. In contrast depletion of the S1PR3 greatly reduced S1P-induced release of VWF. S1P-mediated enhancement of endothelial barrier function was not affected by S1PR3-depletion whereas it was greatly impaired in cells lacking S1PR1. The Rho kinase inhibitor Y27632 completely abrogated S1P-mediated release of VWF. Also, the calcium chelator BAPTA-AM significantly reduced S1P-induced release of VWF. Our findings indicate that S1P-induced release of haemostatic, inflammatory and angiogenic components stored within WPBs depends on the S1PR3.


Subject(s)
Endothelial Cells/metabolism , Receptors, Lysosphingolipid/metabolism , Weibel-Palade Bodies/metabolism , Amides/pharmacology , Cell Line , Down-Regulation , Endothelial Cells/drug effects , Enzyme Inhibitors/pharmacology , Gene Expression , Humans , Protein Binding , Pyridines/pharmacology , Receptors, Lysosphingolipid/genetics , Sphingosine-1-Phosphate Receptors
5.
J Hypertens ; 32(5): 1050-8; discussion 1058, 2014 May.
Article in English | MEDLINE | ID: mdl-24569415

ABSTRACT

OBJECTIVE: Long-chain n-3 polyunsaturated fatty acids from oily fish reduce blood pressure (BP) in hypertension. Previously, we demonstrated that hypertension is associated with marked alterations in sphingolipid biology and elevated ceramide-induced vasoconstriction. Here we investigated in spontaneously hypertensive rats (SHRs) whether fish oil improves endothelial function including reduced vascular contraction induced via the sphingolipid cascade, resulting in reduced BP. METHODS: Twelve-week-old SHRs were fed a control or fish oil-enriched diet during 12 weeks, and BP was recorded. Plasma sphingolipid levels were quantified by mass spectrometry and the response of isolated carotid arteries towards different stimuli was measured. Furthermore, erythrocyte membrane fatty acid composition, thromboxane A2 formation and cytokine secretion in ex-vivo lipopolysaccharide-stimulated thoracic aorta segments were determined. RESULTS: The fish oil diet reduced the mean arterial BP (P < 0.001) and improved endothelial function, as indicated by a substantially increased relaxation potential towards ex-vivo methacholine exposure of the carotid arteries (P < 0.001). The long-chain n-3 polyunsaturated fatty acid diet resulted in altered levels of specific (glucosyl)ceramide subspecies (P < 0.05), reduced membrane arachidonic acid content (P < 0.001) and decreased thromboxane concentrations in plasma (P < 0.01). Concomitantly, the fish oil diet largely reduced ceramide-induced contractions (P < 0.01), which are predominantly mediated by thromboxane. Furthermore, thromboxane A2 and interleukin-10 were reduced in supernatants of lipopolysaccharide-stimulated thoracic aorta of SHRs fed the fish oil diet while RANTES (regulated on activation, normal T-cell expressed and secreted) was enhanced. This may contribute to reduced vasoconstriction in vivo. CONCLUSIONS: Dietary fish oil lowers BP in SHRs and improves endothelial function in association with suppression of sphingolipid-dependent vascular contraction.


Subject(s)
Blood Pressure/drug effects , Dietary Fats, Unsaturated/pharmacology , Endothelium, Vascular/drug effects , Fish Oils/pharmacology , Sphingolipids/physiology , Animals , Chromatography, Liquid , Mass Spectrometry , Muscle Contraction/drug effects , Rats , Rats, Inbred SHR , Sphingomyelin Phosphodiesterase/metabolism , Thromboxane B2/blood
6.
Br J Pharmacol ; 166(4): 1411-8, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22251137

ABSTRACT

BACKGROUND AND PURPOSE: FTY720 (Fingolimod) is a recently approved orally administered drug for the treatment of multiple sclerosis. Phase II and III clinical trials have demonstrated that this drug modestly increases BP. We previously showed that inhibition of sphingosine kinase increases vascular tone and BP in hypertensive, but not normotensive rats. Since FTY720 is reported to have inhibitory effects on sphingosine kinase, we investigated whether FTY720 increases vascular tone and BP only in hypertensive rats via this mechanism. EXPERIMENTAL APPROACH: The contractile and BP modulating effects of FTY720 were studied in vivo and ex vivo (wire myography) in age-matched normotensive Wistar Kyoto (WKY) rats and spontaneously hypertensive rats (SHRs). KEY RESULTS: Oral administration of FTY720 induced an increase in mean arterial pressure in SHR, whereas a decrease in BP was observed in WKY rats, as measured 24 h after administration. Similar to the sphingosine kinase inhibitor dimethylsphingosine (DMS), FTY720 induced large contractions in isolated carotid arteries from SHR, but not in those from WKY. In contrast, the phosphorylated form of FTY720 did not induce contractions in isolated carotid arteries from SHR. FTY720-induced contractions were inhibited by endothelium denudation, COX and thromboxane synthase inhibitors, and by thromboxane receptor antagonism, indicating that (like DMS-induced contractions) they were endothelium-dependent and mediated by thromboxane A2. CONCLUSIONS AND IMPLICATIONS: These data demonstrate that FTY720 increases vascular tone and BP only in hypertensive rats, most likely as a result of its inhibitory effect on sphingosine kinase.


Subject(s)
Carotid Arteries/metabolism , Endothelium, Vascular/metabolism , Hypertension/metabolism , Muscle, Smooth, Vascular/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Vascular Resistance , Animals , Blood Pressure/drug effects , Carotid Arteries/cytology , Carotid Arteries/drug effects , Carotid Arteries/pathology , Cyclooxygenase Inhibitors/pharmacology , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Enzyme Inhibitors/adverse effects , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Fingolimod Hydrochloride , Hypertension/chemically induced , Hypertension/pathology , Hypertension/physiopathology , Immunosuppressive Agents/adverse effects , Immunosuppressive Agents/chemistry , Immunosuppressive Agents/pharmacology , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Male , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/pathology , Phosphorylation , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Propylene Glycols/adverse effects , Propylene Glycols/chemistry , Propylene Glycols/pharmacology , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Sphingosine/adverse effects , Sphingosine/analogs & derivatives , Sphingosine/chemistry , Sphingosine/pharmacology , Thromboxane A2/antagonists & inhibitors , Thromboxane A2/metabolism , Vascular Resistance/drug effects , Vasoconstrictor Agents/adverse effects , Vasoconstrictor Agents/chemistry , Vasoconstrictor Agents/pharmacology
7.
PLoS One ; 6(12): e29222, 2011.
Article in English | MEDLINE | ID: mdl-22195025

ABSTRACT

BACKGROUND: We have previously shown that essential hypertension in humans and spontaneously hypertensive rats (SHR), is associated with increased levels of ceramide and marked alterations in sphingolipid biology. Pharmacological elevation of ceramide in isolated carotid arteries of SHR leads to vasoconstriction via a calcium-independent phospholipase A(2), cyclooxygenase-1 and thromboxane synthase-dependent release of thromboxane A(2). This phenomenon is almost absent in vessels from normotensive Wistar Kyoto (WKY) rats. Here we investigated whether lowering of blood pressure can reverse elevated ceramide levels and reduce ceramide-mediated contractions in SHR. METHODS AND FINDINGS: For this purpose SHR were treated for 4 weeks with the angiotensin II type 1 receptor antagonist losartan or the vasodilator hydralazine. Both drugs decreased blood pressure equally (SBP untreated SHR: 191±7 mmHg, losartan: 125±5 mmHg and hydralazine: 113±14 mmHg). The blood pressure lowering was associated with a 20-25% reduction in vascular ceramide levels and improved endothelial function of isolated carotid arteries in both groups. Interestingly, losartan, but not hydralazine treatment, markedly reduced sphingomyelinase-induced contractions. While both drugs lowered cyclooxygenase-1 expression, only losartan and not hydralazine, reduced the endothelial expression of calcium-independent phospholipase A(2). The latter finding may explain the effect of losartan treatment on sphingomyelinase-induced vascular contraction. CONCLUSION: In summary, this study corroborates the importance of sphingolipid biology in blood pressure control and specifically shows that blood pressure lowering reduces vascular ceramide levels in SHR and that losartan treatment, but not blood pressure lowering per se, reduces ceramide-mediated arterial contractions.


Subject(s)
Antihypertensive Agents/pharmacology , Blood Vessels/drug effects , Blood Vessels/metabolism , Hydralazine/pharmacology , Losartan/pharmacology , Sphingolipids/metabolism , Animals , Blood Pressure/drug effects , Blood Vessels/physiopathology , Carotid Arteries/drug effects , Carotid Arteries/pathology , Carotid Arteries/physiopathology , Ceramides/blood , Endothelium, Vascular/drug effects , Endothelium, Vascular/enzymology , Endothelium, Vascular/physiopathology , Group VI Phospholipases A2/metabolism , In Vitro Techniques , Male , Rats , Rats, Inbred SHR , Sphingomyelin Phosphodiesterase/metabolism , Vasoconstriction/drug effects
8.
PLoS One ; 6(7): e21817, 2011.
Article in English | MEDLINE | ID: mdl-21818267

ABSTRACT

BACKGROUND: Hypertension is, amongst others, characterized by endothelial dysfunction and vascular remodeling. As sphingolipids have been implicated in both the regulation of vascular contractility and growth, we investigated whether sphingolipid biology is altered in hypertension and whether this is reflected in altered vascular function. METHODS AND FINDINGS: In isolated carotid arteries from spontaneously hypertensive rats (SHR) and normotensive Wistar-Kyoto (WKY) rats, shifting the ceramide/S1P ratio towards ceramide dominance by administration of a sphingosine kinase inhibitor (dimethylsphingosine) or exogenous application of sphingomyelinase, induced marked endothelium-dependent contractions in SHR vessels (DMS: 1.4±0.4 and SMase: 2.1±0.1 mN/mm; n = 10), that were virtually absent in WKY vessels (DMS: 0.0±0.0 and SMase: 0.6±0.1 mN/mm; n = 9, p<0.05). Imaging mass spectrometry and immunohistochemistry indicated that these contractions were most likely mediated by ceramide and dependent on iPLA(2), cyclooxygenase-1 and thromboxane synthase. Expression levels of these enzymes were higher in SHR vessels. In concurrence, infusion of dimethylsphingosine caused a marked rise in blood pressure in anesthetized SHR (42±4%; n = 7), but not in WKY (-12±10%; n = 6). Lipidomics analysis by mass spectrometry, revealed elevated levels of ceramide in arterial tissue of SHR compared to WKY (691±42 vs. 419±27 pmol, n = 3-5 respectively, p<0.05). These pronounced alterations in SHR sphingolipid biology are also reflected in increased plasma ceramide levels (513±19 pmol WKY vs. 645±25 pmol SHR, n = 6-12, p<0.05). Interestingly, we observed similar increases in ceramide levels (correlating with hypertension grade) in plasma from humans with essential hypertension (185±8 pmol vs. 252±23 pmol; n = 18 normotensive vs. n = 19 hypertensive patients, p<0.05). CONCLUSIONS: Hypertension is associated with marked alterations in vascular sphingolipid biology such as elevated ceramide levels and signaling, that contribute to increased vascular tone.


Subject(s)
Ceramides/metabolism , Hypertension/metabolism , Adult , Anesthesia , Animals , Arachidonic Acid/metabolism , Blood Pressure/drug effects , Carotid Arteries/drug effects , Carotid Arteries/physiopathology , Ceramides/blood , Chromatography, Liquid , Coronary Vessels/drug effects , Coronary Vessels/physiopathology , Cyclooxygenase 1/metabolism , Female , Humans , Hypertension/blood , Hypertension/physiopathology , Immunohistochemistry , In Vitro Techniques , Male , Mass Spectrometry , Middle Aged , Phospholipases A2, Calcium-Independent/metabolism , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Sphingomyelin Phosphodiesterase/pharmacology , Sphingosine/administration & dosage , Sphingosine/pharmacology , Thromboxane A2/biosynthesis , Vasoconstriction/drug effects
9.
Eur J Pharmacol ; 667(1-3): 105-12, 2011 Sep 30.
Article in English | MEDLINE | ID: mdl-21663738

ABSTRACT

The sphingosine-1-phosphate type 1 (S1P(1)) receptor is a new target in the treatment of auto-immune diseases as evidenced by the recent approval of FTY720 (Fingolimod). The ligand-binding pocket of the S1P(1) receptor has been generally characterised but detailed insight into ligand-specific differences is still lacking. The aim of the current study is to determine differences in ligand-induced S1P(1) receptor activation using an in silico guided site-directed mutagenesis strategy. S1P(1) mutant receptors (modifications of residues Y98(2.57), R120(3.28), F125(3.33)) were probed with a chemically diverse set of S1P(1) agonists (S1P, dihydro-S1P (dhS1P), R-, S- and racemic FTY720-P, VPC24191, SEW2871). Mutation of the R(3.28) residue generally results in a reduction of the potency of all ligands although the synthetic ligands including FTY720-P are less sensitive to these mutations. The Y(2.57)F mutation does not affect the potency of any of the ligands tested, but for all ligands except FTY720-P a significant decrease in potency is observed at the Y(2.57)A mutant. The F(3.33)A mutation significantly decreased the potency of FTY720-P and is detrimental for SEW2871 and VPC24191. The non-aromatic endogenous ligands S1P and dhS1P are less affected by this mutation. Our in silico guided mutagenesis studies identified new molecular determinants of ligand-induced S1P(1) receptor activation: 1) the flexibility of the polar head of the agonist to maintain a tight H-bond network with R(3.28) and 2) the ability of the agonist to make aromatic π-stacking interactions with F(3.33). Interestingly, FTY720-P has both chemical properties and is the only ligand that can efficiently activate the Y(2.57)A mutant.


Subject(s)
Mutation , Receptors, Lysosphingolipid/agonists , Receptors, Lysosphingolipid/genetics , Animals , Computational Biology , Drug Design , Humans , Ligands , Models, Molecular , Mutagenesis, Site-Directed , Protein Binding , Protein Conformation , Protein Stability , Receptors, Lysosphingolipid/chemistry , Receptors, Lysosphingolipid/metabolism , Sphingosine-1-Phosphate Receptors , Stereoisomerism
10.
J Cereb Blood Flow Metab ; 31(4): 1085-92, 2011 Apr.
Article in English | MEDLINE | ID: mdl-20978518

ABSTRACT

The adipocytokine leptin has distinct functions regulating vascular tone, inflammation, and collateral artery growth. Arteriogenesis is an inflammatory process and provides a mechanism to overcome the effects of vascular obstruction. We, therefore, tested the effects of leptin in hypoperfused rat brain (three-vessel occlusion). Systemic leptin administration for 1 week after occlusion surgery increased cerebral hemodynamic reserve similar to granulocyte-macrophage colony-stimulating factor (GM-CSF), as indicated by improved CO(2) reactivity (vehicle 0.53%±0.26% versus leptin 1.05%±0.6% per mm Hg arterial pCO(2), P<0.05). Infusion of microspheres under maximal vasodilation failed to show a positive effect of leptin on cerebral perfusion (vehicle 64.9%±4.5% versus leptin 66.3%±7.0%, occluded/nonoccluded hemisphere). Acute treatment with GM-CSF led to a significant increased CO(2) reactivity and cerebral perfusion (79.2%±8.1% versus 64.9%±4.5%, P<0.05). Vasoconstrictive response of isolated rat carotid artery rings, after phenylephrine was attenuated at 24 hours following preincubation with leptin, was unaffected by removal of endothelium but abrogated by coculture with N-(omega)-nitro-L-arginine methylester, pointing toward an inducible nitric oxide synthase-mediated mechanism. In chronic cerebral hypoperfusion, acute leptin treatment restored the hemodynamic reserve of the cerebral vasculature through its effects on vascular tone, while leaving vascular outward remodeling unaffected. Our results, for the first time, reveal a protective role of leptin on vascular function in hemodynamically compromised brain tissue.


Subject(s)
Cerebrovascular Circulation/drug effects , Cerebrovascular Disorders/drug therapy , Cerebrovascular Disorders/physiopathology , Leptin/pharmacology , Muscle Tonus/drug effects , Animals , Anterior Cerebral Artery/physiology , Body Weight/physiology , Carbon Dioxide/metabolism , Carotid Arteries/drug effects , Carotid Arteries/pathology , Cell Proliferation/drug effects , Cerebrovascular Disorders/pathology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Hemodynamics/drug effects , Hemodynamics/physiology , Male , Monocytes/drug effects , Monocytes/physiology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiology , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/physiology , Nitric Oxide Synthase Type II/antagonists & inhibitors , Oxygen Consumption/drug effects , Phenylephrine/pharmacology , Posterior Cerebral Artery/physiology , Rats , Rats, Sprague-Dawley , Vasoconstrictor Agents/pharmacology
11.
Eur J Pharmacol ; 654(1): 86-91, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21172342

ABSTRACT

Nebivolol is a selective ß1-adrenoceptor antagonist which, in addition, displays endothelium-dependent vasodilating properties in humans and other species. ß3-adrenoceptors have been proposed to be a molecular target of nebivolol-induced vasodilatation. Therefore, we have investigated possible ß3-adrenoceptor agonism by nebivolol for relaxation of the human and rat urinary bladder (prototypical ß3-adrenoceptor-mediated responses) as well as for cAMP accumulation in Chinese hamster ovary cells stably transfected with the human ß-adrenoceptor subtypes. Nebivolol concentration-dependently relaxed both human and rat isolated urinary bladder strips but with low potency, similar to that reported for vasodilatation. However, nebivolol-induced bladder relaxation in either species was not inhibited by the ß3-adrenoceptor antagonist SR 59,230A (10µM), although this compound inhibited the isoprenaline-induced relaxation with the expected potency. In radioligand binding studies nebivolol had lower affinity for human ß3-adrenoceptors than the other two ß-adrenoceptor subtypes, but this low affinity was in line with its potency to relax the bladder or isolated blood vessels. In functional studies nebivolol even in high concentrations did not stimulate cAMP formation via any of the three cloned human ß-adrenoceptors or in rat bladder smooth muscle cells. Taken together these data demonstrate that nebivolol can relax not only vascular but also urinary bladder smooth muscle. However, they do not support the hypothesis that nebivolol is an agonist at cloned human ß3-adrenoceptors or in rat or human urinary bladder.


Subject(s)
Adrenergic beta-1 Receptor Antagonists/pharmacology , Benzopyrans/pharmacology , Ethanolamines/pharmacology , Receptors, Adrenergic, beta-3/drug effects , Urinary Bladder/drug effects , Adrenergic beta-1 Receptor Antagonists/administration & dosage , Animals , Benzopyrans/administration & dosage , CHO Cells , Cricetinae , Cricetulus , Cyclic AMP/metabolism , Dose-Response Relationship, Drug , Ethanolamines/administration & dosage , Humans , Male , Nebivolol , Propanolamines/pharmacology , Rats , Rats, Wistar , Receptors, Adrenergic, beta-3/metabolism , Urinary Bladder/metabolism
12.
Glia ; 58(12): 1465-76, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20648639

ABSTRACT

Sphingolipids are a class of biologically active lipids that have a role in multiple biological processes including inflammation. Sphingolipids exert their functions by direct signaling or through signaling by their specific receptors. Phosphorylated FTY720 (FTY720P) is a sphingosine 1-phosphate (S1P) analogue that is currently in trial for treatment of multiple sclerosis (MS), which targets all S1P receptors but S1P(2). To date, however, it remains unknown whether FTY720P may exert direct anti-inflammatory effects within the central nervous system (CNS), because data concerning S1P receptor expression and regulation under pathological conditions in the human brain are lacking. To investigate potential regulation of S1P receptors in the human brain during MS, we performed immunohistochemical analysis of S1P receptor 1 and 3 expression in well-characterized MS lesions. A strong increase in S1P receptor 1 and 3 expression on reactive astrocytes was detected in active and chronic inactive MS lesions. In addition, we treated primary cultures of human astrocytes with the proinflammatory cytokine tumor necrosis factor-alpha to identify the regulation of S1P(1/3) on astrocytes under pathological conditions. Importantly, we demonstrate that FTY720P exerts an anti-inflammatory action on human astrocytes by limiting secretion of proinflammatory cytokines. Our data demonstrate that reactive astrocytes in MS lesions and cultured under proinflammatory conditions strongly enhance expression of S1P receptors 1 and 3. Results from this study indicate that astrocytes may act as a yet-unknown target within the CNS for the anti-inflammatory effects observed after FTY720P administration in the treatment of MS.


Subject(s)
Multiple Sclerosis/physiopathology , Receptors, Lysosphingolipid/metabolism , Up-Regulation/physiology , Adult , Aged , Aged, 80 and over , Astrocytes/metabolism , Brain/cytology , Cells, Cultured , Chemokines/metabolism , Enzyme-Linked Immunosorbent Assay/methods , Female , Fingolimod Hydrochloride , Humans , Immunosuppressive Agents/pharmacology , Male , Middle Aged , Propylene Glycols/pharmacology , Receptors, Lysosphingolipid/genetics , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , Sphingosine-1-Phosphate Receptors , T-Lymphocytes/metabolism , Up-Regulation/drug effects
13.
Cardiovasc Res ; 87(3): 561-8, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20189954

ABSTRACT

AIMS: Structural adaptation of the vessel wall in response to sustained alterations in haemodynamic forces is known as vascular remodelling. Detailed knowledge on the mechanism underlying this vascular response is limited, and we aimed to study the function of Nur77 in smooth muscle cells (SMCs) in arterial remodelling. METHODS AND RESULTS: Carotid artery ligation in mice results in flow-induced, outward remodelling of the contralateral carotid artery, and we observed enhanced Nur77 expression during this process. Transgenic mice that express Nur77 or its dominant-negative variant, denoted as 'DeltaTA' in arterial SMCs, were exposed to carotid artery ligation, and after 4 weeks pressure-diameter relationships were measured. Structural outward remodelling is inhibited in Nur77-transgenic mice when compared with wild-type and DeltaTA-transgenic mice. The key determinants of remodelling vascular tone and macrophage accumulation were studied. No difference in contractile and relaxant responses was detected in isolated aorta, carotid, and mesenteric artery segments between transgenic and wild-type mice. SMC-specific overexpression of Nur77 in transgenic mice reduced macrophage accumulation and repressed matrix metalloproteinase (MMP)1 and MMP9 expression at early time points. MMP2 protein expression was reduced in Nur77-transgenic mice, whereas in DeltaTA-transgenic mice MMP2 expression was increased. CONCLUSION: Nur77 is induced during outward remodelling and inhibits this vascular adaptation in mice. Nur77-mediated inhibition of arterial remodelling involves a reduction in both macrophage accumulation and MMP expression levels.


Subject(s)
Carotid Arteries/enzymology , Carotid Artery Diseases/enzymology , Hemodynamics , Macrophages/pathology , Matrix Metalloproteinases/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Adaptation, Physiological , Animals , Blood Pressure , Carotid Arteries/drug effects , Carotid Arteries/pathology , Carotid Arteries/physiopathology , Carotid Artery Diseases/genetics , Carotid Artery Diseases/pathology , Carotid Artery Diseases/physiopathology , Disease Models, Animal , Dose-Response Relationship, Drug , Down-Regulation , Gene Expression Regulation, Enzymologic , Hemodynamics/drug effects , Matrix Metalloproteinase 13/metabolism , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinases/genetics , Mice , Mice, Transgenic , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , RNA, Messenger/metabolism , Regional Blood Flow , Time Factors , Vasoconstriction , Vasoconstrictor Agents/pharmacology , Vasodilation , Vasodilator Agents/pharmacology
14.
Mol Cells ; 29(2): 105-11, 2010 Feb 28.
Article in English | MEDLINE | ID: mdl-20127284

ABSTRACT

Vasomotor tone is regulated by a complex interplay of a variety of extrinsic neurohumoral and intrinsic factors. It is the endothelium that has a major influence on smooth muscle cell tone via the release of intrinsic vasoactive factors and is therefore an important regulator of vasomotor tone. Sphingolipids are an emerging class of lipid mediators with important physiological properties. In the last two decades it has not only become increasingly clear that sphingolipid signaling plays a pivotal role in immune function, but also its role in the vascular system is now becoming more recognized. In this mini-review we will highlight the possible cross-talk between sphingolipids and intrinsic vasoactive factors released by the endothelium. Via this cross-talk sphingolipids can orchestrate vasomotor tone and may therefore also be involved in the pathophysiology of disease states associated with endothelial dysfunction.


Subject(s)
Biological Factors/metabolism , Endothelium, Vascular/physiology , Sphingolipids/metabolism , Vasomotor System/metabolism , Animals , Humans , Models, Biological
15.
Mol Cells ; 29(2): 99-104, 2010 Feb 28.
Article in English | MEDLINE | ID: mdl-20127285

ABSTRACT

Regulatory processes including receptor phosphorylation and intracellular trafficking, also referred to as receptor internalization, are important processes to terminate G protein-coupled receptor (GPCR) signaling. Compelling evidence now indicates that internalization of a receptor is not necessarily the endpoint of signaling, but can also be the beginning of the activation of intracellular signaling pathways. Sphingosine-1-phosphate (S1P) receptors, which are activated by the endogenous phospholipid S1P, belong to the family of GPCRs. Interestingly, there is evidence indicating differential intracellular trafficking of one of the S1P receptor subtypes, the S1P1 receptor, upon agonist activation by either S1P or the synthetic agonist FTY720-P. Moreover, the differential effect of FTY720-P on S1P1 receptor regulation has been suggested to be the mechanism of action of this drug, which is now in Phase III clinical trials for the treatment of multiple sclerosis. It is thus of importance to get a good insight into the regulation of S1P receptors. This review therefore gives a detailed overview about the current state of knowledge on S1P receptor internalization and its functional implications, including some data on nuclear signaling of S1P receptors.


Subject(s)
Intracellular Space/metabolism , Receptors, Lysosphingolipid/metabolism , Amino Acid Sequence , Animals , Endocytosis , Humans , Ligands , Molecular Sequence Data , Protein Processing, Post-Translational , Protein Transport , Receptors, Lysosphingolipid/chemistry
16.
Basic Res Cardiol ; 105(1): 73-87, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19639379

ABSTRACT

We investigated the contribution of the intracellular calcium (Ca (i) (2+) ) transient to acetylcholine (ACh)-mediated reduction of pacemaker frequency and cAMP content in rabbit sinoatrial nodal (SAN) cells. Action potentials (whole cell perforated patch clamp) and Ca (i) (2+) transients (Indo-1 fluorescence) were recorded from single isolated rabbit SAN cells, whereas intracellular cAMP content was measured in SAN cell suspensions using a cAMP assay (LANCE((R))). Our data show that the Ca (i) (2+) transient, like the hyperpolarization-activated "funny current" (I (f)) and the ACh-sensitive potassium current (I (K,ACh)), is an important determinant of ACh-mediated pacemaker slowing. When I (f) and I (K,ACh) were both inhibited, by cesium (2 mM) and tertiapin (100 nM), respectively, 1 micro M ACh was still able to reduce pacemaker frequency by 72%. In these I (f) and I (K,ACh)-inhibited SAN cells, good correlations were found between the ACh-mediated change in interbeat interval and the ACh-mediated change in Ca (i) (2+) transient decay (r (2) = 0.98) and slow diastolic Ca (i) (2+) rise (r (2) = 0.73). Inhibition of the Ca (i) (2+) transient by ryanodine (3 microM) or BAPTA-AM (5 microM) facilitated ACh-mediated pacemaker slowing. Furthermore, ACh depressed the Ca (i) (2+) transient and reduced the sarcoplasmic reticulum (SR) Ca(2+) content, all in a concentration-dependent fashion. At 1 microM ACh, the spontaneous activity and Ca (i) (2+) transient were abolished, but completely recovered when cAMP production was stimulated by forskolin (10 microM) and I (K,ACh) was inhibited by tertiapin (100 nM). Also, inhibition of the Ca (i) (2+) transient by ryanodine (3 microM) or BAPTA-AM (25 microM) exaggerated the ACh-mediated inhibition of cAMP content, indicating that Ca (i) (2+) affects cAMP production in SAN cells. In conclusion, muscarinic receptor stimulation inhibits the Ca (i) (2+) transient via a cAMP-dependent signaling pathway. Inhibition of the Ca (i) (2+) transient contributes to pacemaker slowing and inhibits Ca (i) (2+) -stimulated cAMP production. Thus, we provide functional evidence for the contribution of the Ca (i) (2+) transient to ACh-induced inhibition of pacemaker activity and cAMP content in rabbit SAN cells.


Subject(s)
Acetylcholine/metabolism , Calcium/metabolism , Cyclic AMP/metabolism , Receptors, Muscarinic/metabolism , Sinoatrial Node/metabolism , Animals , Egtazic Acid/analogs & derivatives , Muscarinic Agonists , Patch-Clamp Techniques , Rabbits , Ryanodine , Sarcoplasmic Reticulum/metabolism , Sinoatrial Node/cytology
17.
Eur J Pharmacol ; 606(1-3): 25-31, 2009 Mar 15.
Article in English | MEDLINE | ID: mdl-19374869

ABSTRACT

Regulator of G protein signalling (RGS) protein expression is altered under growth promoting conditions in vascular smooth muscle cells (VSMCs). Since sphingosine-1-phosphate (S1P) is an important growth stimulatory factor, we investigated whether stimulation of VSMCs with S1P results in alterations in mRNA expression levels of several RGS proteins and which signalling components are involved. VSMCs were stimulated with S1P and mRNA expression levels of RGS2, RGS3, RGS4, RGS5 and RGS16 were measured by real-time polymerase chain reaction. S1P caused a time-dependent up-regulation of RGS2 and RGS16 mRNA expression. FTY720-P, a S1P(1)/S1P(3-5) agonist, did not regulate RGS2 mRNA levels although it did up-regulate RGS16 mRNA expression. Pertussis toxin treatment revealed that the S1P-induced RGS16 expression was G(i/o)-dependent whereas up-regulation of RGS2 mRNA was not. Phosphatidylinositol 3-kinase, protein kinase C and mitogen-activated protein kinase kinase apparently were not involved in the S1P-induced up-regulation of both RGS proteins. The present study demonstrates that S1P induces RGS2 and RGS16 mRNA expression but uses distinct S1P receptor subtypes and signalling pathways to regulate expression of these RGS proteins.


Subject(s)
Gene Expression Regulation/drug effects , Lysophospholipids/pharmacology , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , RGS Proteins/genetics , Sphingosine/analogs & derivatives , Animals , Lysophospholipids/metabolism , Male , Muscle, Smooth, Vascular/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptors, Lysosphingolipid/metabolism , Signal Transduction/drug effects , Sphingosine/metabolism , Sphingosine/pharmacology , Up-Regulation/drug effects
18.
Neurourol Urodyn ; 28(4): 349-55, 2009.
Article in English | MEDLINE | ID: mdl-19034955

ABSTRACT

AIMS: To explore possible changes in expression and/or function of alpha(1)- and beta-adrenoceptor subtypes as a cause for bladder dysfunction in a rat model of bladder outlet obstruction (BOO). METHODS: BOO was induced in rats by partial urethral ligature. Contraction and relaxation experiments were performed with isolated bladder strips from BOO, sham-operated and non-operated (control) rats 7 days after BOO induction. mRNA expression of alpha(1)- and beta-adrenoceptor subtypes was assessed by quantitative real-time PCR. RESULTS: Receptor-independent contraction or relaxation did not differ between BOO and sham rats. The alpha(1)-agonists methoxamine and A-61,603 caused only weak contraction without major differences between groups. Against KCl-induced tone, the beta-adrenoceptor agonists noradrenaline and isoprenaline caused similar relaxation in BOO and sham rats, whereas relaxation in response to the beta(3)-selective BRL 37,344 was attenuated. Against passive tension, noradrenaline induced relaxation in sham and control rats; in contrast, noradrenaline induced contraction at low concentrations and relaxation at high concentrations in BOO rats. The contraction component was abolished by the alpha(1)-antagonist prazosin. The mRNA expression of alpha(1D)-adrenoceptors was increased in BOO, whereas none of the other receptor mRNAs were up-regulated. CONCLUSIONS: In a rat BOO model, weak contraction responses to alpha(1)-agonists and relaxation responses to beta-agonists are not altered to a major extent. Nevertheless, relaxation responses to the endogenous agonist noradrenaline are turned into alpha(1)-adrenoceptor-mediated contraction responses in BOO, possibly due to an up-regulation of alpha(1D)-adrenoceptors.


Subject(s)
Receptors, Adrenergic, alpha-1/biosynthesis , Receptors, Adrenergic, alpha-1/physiology , Receptors, Adrenergic, beta/biosynthesis , Receptors, Adrenergic, beta/physiology , Urinary Bladder Neck Obstruction/metabolism , Adrenergic alpha-Agonists/pharmacology , Animals , Ligation , Male , Molecular Sequence Data , Muscle Contraction/drug effects , Muscle Contraction/physiology , Muscle Relaxation/drug effects , Muscle Relaxation/physiology , Norepinephrine/pharmacology , Potassium Chloride/pharmacology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Wistar , Regression Analysis , Reverse Transcriptase Polymerase Chain Reaction
19.
Basic Res Cardiol ; 104(1): 50-9, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18777003

ABSTRACT

Local formation of the sphingomyelin metabolite sphingosine-1-phosphate (S1P) within the vascular wall has been shown to modulate vascular reactivity. In this study we investigated whether sphingosine kinase, the enzyme responsible for S1P synthesis, plays a role in muscarinic receptor-mediated NO production and vascular relaxation in different blood vessel types. For this purpose, sphingosine kinase translocation and sphingolipid-dependent NO-production after muscarinic receptor stimulation were assessed in an endothelial cell line. Furthermore, we used the sphingosine kinase inhibitor N,N-dimethylsphingosine (DMS) to investigate the role of sphingosine kinase in the relaxant responses to the muscarinic agonist methacholine (MCh) in isolated rat aorta and mesenteric arteries. Activation of M(3)-receptors in an endothelial cell line induced a fast translocation of YFP-tagged sphingosine kinase-1 from the cytosol to the plasma membrane. Concomitant NO-production in this cell line was partially inhibited by DMS. Accordingly, in rat aorta the relaxant responses to MCh were attenuated in the presence of DMS, while the responses to the NO-donor sodium nitroprusside were unaltered. In contrast, DMS enhanced the relaxant responses to MCh in mesenteric artery preparations. This effect could also be observed in the presence of NO synthase and cyclooxygenase inhibitors, indicating that sphingosine kinase inhibition specifically enhanced endothelium-derived hyperpolarizing factor-mediated (i.e. non-NO and non-prostacyclin-dependent) relaxation. We conclude that sphingosine kinase differentially regulates vascular tone in different vessel types, enhancing NO-dependent vasorelaxation but counteracting EDHF-dependent vasorelaxation. This observation enhances our understanding of the complex mechanisms by which sphingolipids regulate vascular homeostasis. Moreover, a disturbed regulation of sphingolipid metabolism in the vascular wall may therefore play a role in the aetiology/pathology of disease states characterized by endothelial dysfunction.


Subject(s)
Biological Factors/physiology , Endothelium, Vascular/physiology , Enzyme Activation/physiology , Nitric Oxide/physiology , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Receptors, Muscarinic/physiology , Vasodilation/physiology , Animals , Cerebrovascular Circulation/physiology , DNA Primers , Endothelium, Vascular/cytology , Genetic Markers , Luminescent Proteins/genetics , Mice , Phosphotransferases (Alcohol Group Acceptor)/genetics , Polymerase Chain Reaction , Protein Transport
20.
Eur J Pharmacol ; 600(1-3): 1-9, 2008 Dec 14.
Article in English | MEDLINE | ID: mdl-18854184

ABSTRACT

Sphingosine-1-phosphate (S1P) signalling via G protein-coupled receptors is important for the regulation of cell function and differentiation. Specific Regulators of G protein Signalling (RGS) proteins modulate the function of these receptors in many cell types including vascular smooth muscle cells (VSMCs). Therefore, we investigated the role of altered expression levels of RGS proteins in S1P receptor function in VSMCs and transfected CHO cells. The mRNA expression of the S1P(1) receptor, RGS4 and RGS16 were down-regulated in VSMCs during phenotypic modulation induced by culturing, whereas mRNA levels of RGS2, RGS3, S1P(2) and S1P(3) receptors were unchanged. Interestingly, the expression level of RGS5 was transiently up-regulated. Despite major alterations in RGS levels, S1P-induced calcium elevation in VSMCs was not altered. Co-transfection of RGS2, RGS3, RGS4, RGS5 and RGS16 into CHO-Flp-In cells stably expressing the S1P(1) or S1P(3) receptor did not modify S1P-induced inhibition of cAMP accumulation to a major extent. Similar results were obtained with SEW2871, a selective S1P(1) receptor agonist. However, the inhibition of cAMP accumulation by the agonist FTY720-P via the S1P(1) receptor was significantly decreased by co-transfection with RGS5. These results indicate that mRNA of the S1P(1) receptor, RGS4, RGS5 and RGS16 is differentially regulated during phenotypic modulation. However, major alterations in RGS protein expression have only limited effect on S1P receptor function.


Subject(s)
Gene Expression Regulation , RGS Proteins/metabolism , Receptors, Lysosphingolipid/metabolism , Signal Transduction , Animals , CHO Cells , Calcium/metabolism , Cells, Cultured , Cricetinae , Cricetulus , Cyclic AMP/metabolism , Male , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , RGS Proteins/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...