Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Am J Pathol ; 191(5): 829-837, 2021 05.
Article in English | MEDLINE | ID: mdl-33617784

ABSTRACT

The endothelial glycoprotein thrombomodulin regulates coagulation, inflammation, and apoptosis. In diabetic mice, reduced thrombomodulin function results in diabetic nephropathy (DN). Furthermore, thrombomodulin treatment reduces renal inflammation and fibrosis. Herein, thrombomodulin expression was examined in human kidney samples to investigate the possibility of targeting thrombomodulin in patients with DN. Glomerular thrombomodulin was analyzed together with the number of glomerular macrophages in 90 autopsied diabetic cases with DN, 55 autopsied diabetic cases without DN, and 37 autopsied cases without diabetes or kidney disease. Thrombomodulin mRNA was measured in glomeruli microdissected from renal biopsies from patients with DN and nondiabetic controls. Finally, glomerular thrombomodulin was measured in diabetic mice following treatment with the selective endothelin A receptor (ETAR) blocker, atrasentan. In diabetic patients, glomerular thrombomodulin expression was increased at the mRNA level, but decreased at the protein level, compared with nondiabetic controls. Reduced glomerular thrombomodulin was associated with an increased glomerular influx of macrophages. Blocking the ETAR with atrasentan restored glomerular thrombomodulin protein levels in diabetic mice to normal levels. The reduction in glomerular thrombomodulin in diabetes likely serves as an early proinflammatory step in the pathogenesis of DN. Thrombomodulin protein may be cleaved under diabetic conditions, leading to a compensatory increase in transcription. The nephroprotective effects of ETAR antagonists in diabetic patients may be attributed to the restoration of glomerular thrombomodulin.


Subject(s)
Atrasentan/pharmacology , Diabetes Mellitus, Experimental/pathology , Diabetic Nephropathies/pathology , Endothelin A Receptor Antagonists/pharmacology , Fibrosis/pathology , Thrombomodulin/metabolism , Animals , Endothelium/pathology , Humans , Inflammation/pathology , Kidney/pathology , Kidney Glomerulus/pathology , Macrophages/pathology , Male , Mice , Mice, Knockout , Thrombomodulin/drug effects , Thrombomodulin/genetics
2.
Blood Coagul Fibrinolysis ; 30(7): 324-330, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31490208

ABSTRACT

: The gut microbial metabolite, trimethylamine N-oxide (TMAO), was previously reported to induce platelet hypersensitivity, which leads to thrombotic risk. However, the molecular mechanism underlying the effects of TMAO on endothelial cells (EC), which is the primary vessel wall contact with the lumen, remains unclear. Here, we investigated the impact of TMAO on procoagulant activity (PCA) in EC and mice, for a possible link between microbiota and coagulation. To test the PCA of TMAO in EC, we performed one-stage clotting assays and converted into PCA. Antitissue factor (TF) antibody was used to test the TF role in PCA. Quantitative PCR was performed to measure the TF, thrombomodulin, IL-6, TF pathway inhibitor and IL-1b expressions at mRNA levels. To test the PCA and thrombotic risk by TMAO in mice, we challenged the mice with TMAO (8 mg/kg; 3 h) and measured the thrombin-anti-thrombin complex (TAT) and D-dimer levels as well as ferric chloride (FeCl3)-induced carotid artery thrombosis model. TMAO-induced TF expression in EC at mRNA and protein levels, dose-dependently. TF blocking experiment confirmed that the increased PCA by TMAO is TF-dependent. Also, mitogen-activated protein kinase pathway inhibitors abolished TMAO-induced TF expression. However, TMAO challenged mice failed to develop systemic activation of coagulation (TAT and D-dimer), as well as a FeCl3-induced carotid arterial thrombosis model. Our results indicated that TMAO triggered TF-dependent PCA via activation of nuclear factor-κB and downregulated thrombomodulin expression in human EC, but failed to develop systemic activation of coagulation in mice.


Subject(s)
Blood Coagulation/drug effects , Endothelial Cells/drug effects , Hemostasis/drug effects , Methylamines/pharmacology , Animals , Cells, Cultured , Humans , Mice , NF-kappa B/metabolism , Oxidants/pharmacology , Thrombomodulin/drug effects , Thrombomodulin/metabolism
3.
Heart Vessels ; 34(2): 368-374, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30143885

ABSTRACT

Interventions targeting the serum eicosapentaenoic acid (EPA)/arachidonic acid (AA) ratio could be useful for the prevention of coronary artery disease (CAD). Few data exist regarding the effects of administration of EPA on the serum levels of soluble thrombomodulin (sTM) as a marker of endothelial damage, or on the relationship between the sTM and EPA/AA ratio in patients with CAD receiving statin treatment. We assigned stable CAD patients already receiving statin therapy to an EPA group (1800 mg/day: n = 50) or control group (n = 50). A significant increase of the sTM level was observed in the EPA group as compared to that in the control group 0.40 (0.10/0.70) FU/mL vs. 0.20 (0/0.40) FU/mL, p = 0.004 at the 6-month follow-up examination. Multivariate regression analysis after adjustments for coronary risk factors and changes of the serum lipid levels identified an increased EPA/AA ratio as an independent predictor of increased serum sTM level (ß = 0.244, p = 0.02). The results suggest that an increased sTM level caused by additional administration of EPA to statin might be associated with an increased EPA/AA ratio. The increase of the serum sTM after administration of EPA might reflect an increase of the TM expression on the endothelial surface rather than endothelial damage in CAD patients under statin treatment.Clinical Trial Registration Information UMIN ( http://www.umin.ac.jp/ ), Study ID: UMIN000010452.


Subject(s)
Coronary Artery Disease/drug therapy , Eicosapentaenoic Acid/administration & dosage , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Thrombomodulin/blood , Aged , Biomarkers/blood , Coronary Angiography , Coronary Artery Disease/blood , Coronary Artery Disease/diagnosis , Dose-Response Relationship, Drug , Female , Follow-Up Studies , Humans , Male , Prognosis , Prospective Studies , Thrombomodulin/drug effects , Time Factors
4.
Life Sci ; 87(3-4): 92-9, 2010 Jul 17.
Article in English | MEDLINE | ID: mdl-20493886

ABSTRACT

AIMS: Statins (3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors) and fluid wall shear stress have been reported to modulate the expression of genes related to inflammation, blood coagulation, thrombosis, and vascular constriction in cultured endothelial cells. In this study, we investigated the combined effect of laminar shear stress (LSS) and statins on endothelial cell gene expression. MAIN METHODS: Kruppel-like factor 2 (KLF2), endothelial nitric oxide synthase (eNOS), and thrombomodulin (TM) mRNA and protein expression were evaluated in human abdominal aortic endothelial cells (HAAEC) treated with simvastatin (0.1, 1 or 10 microM) at various levels of LSS (0, 1.25, 12.5 or 25 dynes/cm(2)). KEY FINDINGS: As expected, simvastatin and LSS separately enhanced KLF2, eNOS, and TM mRNA expressions. The combination of simvastatin and LSS resulted in significantly higher mRNA levels of all three genes compared to cells treated with LSS only. The highest KLF2, eNOS, and TM mRNA levels were detected at 10 microM simvastatin and 25 dynes/cm(2). Under these conditions, eNOS and TM protein levels were also elevated. Combining LSS and simvastatin produced an overall additive increase in KLF2, eNOS, and TM mRNA. Treatment of the endothelial cells with 10 microM simvastatin and 200 microM mevalonate completely eliminated the effect of simvastatin. SIGNIFICANCE: Our results suggest an additive increase in KLF2, eNOS, and TM expressions when simvastatin and LSS are combined. These results may help to explain the proposed non-lipid lowering benefits of statins observed in the clinic.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Kruppel-Like Transcription Factors/drug effects , Nitric Oxide Synthase Type III/drug effects , Simvastatin/pharmacology , Thrombomodulin/drug effects , Aorta, Abdominal/cytology , Aorta, Abdominal/drug effects , Aorta, Abdominal/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Gene Expression Regulation/drug effects , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , RNA, Messenger/metabolism , Simvastatin/administration & dosage , Stress, Mechanical , Thrombomodulin/genetics
5.
J Cardiovasc Pharmacol ; 55(3): 276-85, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20075745

ABSTRACT

To overcome the limitations of balloon expandible metal stent-induced neointimal smooth muscle cell proliferation, drug-coated stent devices have been developed. Drug eluting stents release high concentrations of antiproliferative agents, such as paclitaxel, to reduce neointimal hyperplasia. The proinflammatory cytokine, tumor necrosis factor-alpha (TNF-alpha), is known to cause severe endothelial dysfunction and accelerate atherosclerotic lesion progression. The interaction of TNF-alpha and paclitaxel on the release of prothrombotic molecules was examined in endothelial cells. Treatment of endothelial cells with paclitaxel had no direct effect on tissue factor (TF) expression, but TNF-alpha increased TF. Cotreatment of paclitaxel with TNF-alpha markedly augmented the release of TF. TNF-alpha induced release of plasminogen activator inhibitor but no synergism occurred with paclitaxel. Treatment of endothelial cells with paclitaxel and TNF-alpha reduced expression of thrombomodulin and protein C receptor. Tissue factor pathway inhibitor expression was reduced by prolonged treatment with either paclitaxel or TNF-alpha. The adhesion molecule, CD62 E, was induced by TNF-alpha; however, CD31, CD62 P, and CD106 were not affected by paclitaxel and TNF-alpha. Apoptosis was not observed with cotreatment of endothelial cells with paclitaxel and TNF-alpha. CD59-positive microparticles were released in response to TNF-alpha, but the release was not augmented by paclitaxel. Paclitaxel and TNF-alpha increased the nitrotyrosination of proteins. These findings indicate that paclitaxel enhances TNF-alpha-induced release of TF, and downregulated thrombomodulin, increased protein nitration, which may subsequently favor prothrombotic intimal surface.


Subject(s)
Endothelium, Vascular/drug effects , Paclitaxel/toxicity , Tubulin Modulators/toxicity , Tumor Necrosis Factor-alpha/toxicity , Cells, Cultured , Cytokines/drug effects , Cytokines/metabolism , Down-Regulation/drug effects , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Gene Expression Regulation/drug effects , Humans , Inflammation/chemically induced , Inflammation/physiopathology , Nitrates/metabolism , Paclitaxel/pharmacology , Plasminogen Inactivators/metabolism , Proteins/metabolism , Thrombomodulin/drug effects , Thrombomodulin/genetics , Thromboplastin/drug effects , Thromboplastin/genetics , Tubulin Modulators/pharmacology , Tumor Necrosis Factor-alpha/metabolism
6.
J Periodontol ; 80(9): 1511-7, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19722803

ABSTRACT

BACKGROUND: The endothelial cell surface glycoprotein thrombomodulin (TM) inhibits vascular coagulation and inflammation via regulation of thrombin-mediated activation of protein C. Porphyromonas gingivalis is the major periodontopathic bacterium and has been found in vessel walls and atherosclerotic lesions in humans. P. gingivalis-derived cysteine proteases (gingipains) are known to enhance inflammatory and coagulant responses of vascular endothelial cells. However, it has not been elucidated whether gingipains affect vascular endothelial TM. METHODS: Purified arginine-specific gingipains (Rgps) and lysine-specific gingipain (Kgp) from P. gingivalis were used to investigate the effects of gingipains on recombinant human TM by immunoblot analyses. Flow cytometry and activated protein C assay were carried out to examine the effects of gingipains on vascular endothelial cell surface TM. Immunohistochemistry was performed to investigate TM expression in microvascular endothelia in gingival tissues taken from patients with periodontitis. RESULTS: Rgps and Kgp cleaved TM in vitro. Endothelial cell surface TM was also degraded by Rgps. Thrombin-mediated activation of protein C was reduced by Rgps through TM inactivation. Gingival microvascular endothelial TM was reduced in patients with periodontitis. CONCLUSIONS: P. gingivalis gingipains induced the degradation and inactivation of endothelial TM, which may promote vascular coagulation and inflammation. In addition, in vivo relevance was demonstrated by reduced expression of TM in gingival microvascular endothelia in patients with periodontitis, which may be involved in the pathogenesis of periodontitis.


Subject(s)
Adhesins, Bacterial/pharmacology , Cysteine Endopeptidases/pharmacology , Endothelial Cells/drug effects , Endothelium, Vascular/drug effects , Hemagglutinins/pharmacology , Porphyromonas gingivalis/enzymology , Thrombomodulin/drug effects , Cells, Cultured , Electrophoresis, Polyacrylamide Gel , Endothelial Cells/metabolism , Endothelium, Vascular/pathology , Female , Flow Cytometry , Gingipain Cysteine Endopeptidases , Gingiva/blood supply , Gingivitis/pathology , Humans , Immunoblotting , Immunohistochemistry , Male , Microvessels/pathology , Middle Aged , Periodontal Attachment Loss/pathology , Periodontal Pocket/pathology , Periodontitis/pathology , Protein C/analysis , Thrombomodulin/analysis
7.
Blood ; 113(3): 714-22, 2009 Jan 15.
Article in English | MEDLINE | ID: mdl-18971420

ABSTRACT

Various virus infections cause dysfunctional hemostasis and in some instances lead to the development of viral hemorrhagic fever syndrome. How do diverse viruses induce the expression of tissue factor on vascular cells? We hypothesize that a direct stimulation of pattern recognition receptors (PRR) by viral nucleic acids may be the key. Double-stranded RNA (dsRNA) is produced by many viruses and is recognized by various PRR, including Toll-like receptor-3 (TLR3). We have investigated whether poly I:C, a model for viral dsRNA, can influence cellular hemostasis. Poly I:C could up-regulate tissue factor and down-regulate thrombomodulin expression on endothelial cells but not on monocytes. The response to poly I:C was diminished upon small interfering RNA (siRNA)-mediated inhibition of TLR3, but not other PRR. In vivo, application of poly I:C induced similar changes in the aortic endothelium of mice as determined by enface microscopy. D-dimer, a circulating marker for enhanced coagulation and fibrinolysis, and tissue fibrin deposition was elevated. All the hemostasis-related responses to poly I:C, but not cytokine secretion, were blunted in TLR3(-/-) mice. Hence, the activation of TLR3 can induce the procoagulant state in the endothelium, and this could be relevant for understanding the mechanisms of viral stimulation of hemostasis.


Subject(s)
Endothelial Cells/metabolism , Hemostasis/physiology , Thromboplastin/metabolism , Toll-Like Receptor 3/metabolism , Animals , Blood Coagulation/drug effects , Blood Coagulation/physiology , Blotting, Western , Cells, Cultured , Cytokines/biosynthesis , Endothelial Cells/drug effects , Female , Fluorescent Antibody Technique , Gene Expression/drug effects , Humans , Interferon Inducers/pharmacology , Male , Mice , Mice, Mutant Strains , Microscopy, Fluorescence , Poly I-C/pharmacology , RNA, Small Interfering , Receptors, Pattern Recognition/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Thrombomodulin/drug effects , Thrombomodulin/metabolism , Thromboplastin/drug effects , Toll-Like Receptor 3/drug effects , Toll-Like Receptor 3/genetics
8.
Przegl Lek ; 65(2): 68-72, 2008.
Article in Polish | MEDLINE | ID: mdl-18663903

ABSTRACT

Inflammation and disturbances of the hemostatic system may play a role in pathogenesis and complications of ischemic heart disease. More and more reports indicate that apart from their cholesterol-lowering effect statins also exert other beneficial effects in cardiovascular diseases. Taking this into consideration, the aim of the study was to assess the influence of simvastatin (20 mg per day) on a marker of inflammation - CRP and some parameters of coagulation and fibrinolysis in 22 patients with ischaemic heart disease. Serum lipids, levels of hsCRP, thrombomodulin (TM), vWF, prothrombin fragment 1+2 (F1+2), thrombin-antithrombin complex (TAT), thrombin activatable fibrinolysis inhibitor (TAFI), t-PA, plasmin-antiplasmin complex (PAP) and TAFI activity were assessed before and after one, three and six months simvastatin treatment. After one month therapy of simvastatin, there have been significant reduction of levels of total cholesterol, LDL-cholesterol and triglycerides and these values have remained until the end of the study. No influence on the level of HDL-cholesterol has been observed. After 6 months of treatment significant decrease in the level of hsCRP and increase of the levels TM and vWF with reference to baselines results have been observed. After a 1-and 6-month therapy, the level of TAFI have been significantly increased. Other hemostatic parameters, i.e. levels of F1+2, TAT, t-PA, PAP and TAFI activity have not changed significantly. This prospective study has confirmed high efficacy of lipid-lowering effect and anti-inflammatory properties of simvastatin. Simvastatin influenced some hemo-static parameters, however, these effects were not, in majority, significant.


Subject(s)
C-Reactive Protein/drug effects , Hemostasis/drug effects , Myocardial Ischemia/prevention & control , Simvastatin/pharmacology , Aged , Carboxypeptidase B2/metabolism , Carboxypeptidase B2/pharmacology , Cholesterol/blood , Cholesterol, LDL/blood , Cholesterol, LDL/drug effects , Female , Humans , Male , Myocardial Ischemia/etiology , Myocarditis/complications , Myocarditis/metabolism , Myocarditis/prevention & control , Thrombomodulin/drug effects , Triglycerides/blood
9.
Int J Cardiol ; 123(2): 180-2, 2008 Jan 11.
Article in English | MEDLINE | ID: mdl-17292497

ABSTRACT

INTRODUCTION: Thrombomodulin is an integral endothelial cell membrane protein, exists not only on the surface of endothelial cells but also as soluble fragments circulating in plasma. Probucol has anti-oxidant properties as well as cholesterol-lowering effects and may affect soluble thrombomodulin (sTM). METHODS: Sixteen rabbits fed with high-cholesterol diet for 8 weeks were randomly divided into two groups: (1) high-cholesterol group (n=8): maintained high-cholesterol diet; (2) probucol group (n=8): the same diet plus probucol for 6 weeks. Control group (n=8) was fed with normal diet for 14 weeks. The levels of sTM and oxidized low-density lipoprotein (OX-LDL) were measured using ELISA. RESULTS: There were atherosclerotic lesions in aortas and intimal thickness significantly increased in high-cholesterol group. Probucol significantly reduced the lesion area (56.4%+/-9.8% vs 82.5%+/-10.5%) and decreased the intimal thickness (44.65+/-7.25 mum vs 72.21+/-8.32) as compared with high-cholesterol group, all P<0.01. Probucol decreased the level of OX-LDL and sTM as compared with high-cholesterol group, all P<0.05. CONCLUSIONS: Probucol retarded the plaques formation may relate to decrease plasma OX-LDL and sTM concentration, which may improve endothelial function in hypercholesterolemic rabbit.


Subject(s)
Anticholesteremic Agents/pharmacology , Antioxidants/pharmacology , Hypercholesterolemia/blood , Probucol/pharmacology , Thrombomodulin/drug effects , Animals , Anticholesteremic Agents/therapeutic use , Antioxidants/therapeutic use , Hypercholesterolemia/drug therapy , Male , Probucol/therapeutic use , Rabbits
10.
Thromb Res ; 120(4): 559-66, 2007.
Article in English | MEDLINE | ID: mdl-17239427

ABSTRACT

Stromal cell-derived factor-1 (SDF-1) is a CXC chemokine that activates and directs the migration of leukocytes that have CXCR4, which is the unique receptor for SDF-1. Although SDF-1/CXCR4 interaction has been implicated in various inflammatory conditions, its role in modulating coagulation has not been determined. We studied the plasma SDF-1 levels in 90 patients with suspected disseminated intravascular coagulation (DIC) and we found that circulating SDF-1 was significantly increased in the overt DIC patients and was also increased in overt DIC patients who have a poor outcome. We then tested in vitro whether SDF-1 can affect the expression of monocyte tissue factor (TF) and endothelial thrombomodulin (TM), and both of these play important roles in coagulopathy. SDF-1 did not affect the expression of surface TF protein and its function and the TF mRNA level in both monocytes and the monocytic leukemia cell line THP-1. SDF-1 also did not change the surface TM expression of endothelial cells. SDF-1 could enhance low-dose ADP induced platelet aggregation, although it failed by itself to induce aggregation. These findings suggest that plasma SDF-1 might be closely associated with hypercoagulability though its action as a platelet activator.


Subject(s)
Chemokines, CXC/blood , Disseminated Intravascular Coagulation/blood , Thrombophilia/etiology , Cells, Cultured , Chemokine CXCL12 , Chemokines, CXC/pharmacology , Endothelial Cells/metabolism , Humans , Monocytes/metabolism , Platelet Function Tests , Prognosis , Thrombomodulin/drug effects , Thromboplastin/drug effects
11.
Thromb Res ; 119(6): 769-75, 2007.
Article in English | MEDLINE | ID: mdl-16844201

ABSTRACT

Salviae miltiorrhizae (SM), a clinical, commonly used herb, can activate blood circulation and resolve stasis. We have investigated the effects of salvianolic acid B (Sal B), a pure compound extracted from the dried SM roots, on fibrinolytic (tissue-type plasminogen activator and plasminogen activator inhibitor, t-PA and PAI) and anticoagulant (thrombomodulin,TM) properties of cultured human umbilical vein endothelial cells (HUVECs). When HUVECs were treated with Sal B, a dose- (0.0125-0.5 mg/ml) and a time-dependent decrease in PAI activity were observed. PAI type 1 (PAI-1) antigen and PAI-1 mRNA expression significantly decreased compared to control values in the conditioned media of HUVECs pretreated with Sal B for 12 h. Moreover, TM activity reached a maximum stimulation of 1.25-fold over control levels in the pretreatment of Sal B for 12 h and t-PA and TM specific mRNA expression also increased (1.7- and 1.8-fold, respectively). In conclusion, Sal B increased the fibrinolytic and anticoagulant potential of cultured HUVECs by up-regulating the expression of t-PA and TM and by down-regulating the expression of PAI-1. These data suggest that Sal B is clinically effective because of its ability to change the gene expression profile of endothelial cells thereby preventing vascular events.


Subject(s)
Benzofurans/pharmacology , Drugs, Chinese Herbal/pharmacology , Endothelial Cells/physiology , Hemostasis/drug effects , Umbilical Veins/physiology , Cell Survival/drug effects , Cells, Cultured , Endothelial Cells/metabolism , Fibrinolytic Agents/metabolism , Humans , Plasminogen Activator Inhibitor 1/genetics , Plasminogen Activator Inhibitor 1/metabolism , Plasminogen Inactivators/metabolism , RNA, Messenger/metabolism , Thrombomodulin/drug effects , Thrombomodulin/genetics , Thrombomodulin/physiology , Tissue Plasminogen Activator/genetics , Umbilical Veins/cytology , Umbilical Veins/metabolism
12.
Crit Care Med ; 35(1): 57-63, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17080003

ABSTRACT

OBJECTIVE: Various lung diseases are associated with local activation of coagulation and concurrent inhibition of fibrinolysis. Although salmeterol, a beta2-adrenoceptor agonist with profound bronchodilatory properties, has been studied extensively, the effects of this compound on the pulmonary hemostatic balance are not elucidated. DESIGN: A single-blinded, placebo-controlled study. SETTING: University hospital and laboratory. SUBJECTS: A total of 32 human volunteers. INTERVENTIONS: Subjects inhaled 100 microg of salmeterol or placebo (t = -30 mins) followed by 100 microg of lipopolysaccharide (LPS) or normal saline (t = 0 mins; n = 8 per group). MEASUREMENTS AND MAIN RESULTS: Measurements were performed in bronchoalveolar lavage fluid obtained 6 hrs postchallenge. Inhalation of LPS enhanced pulmonary coagulation as determined by an increase in the concentrations of thrombin-antithrombin complexes, factor VIIa, and soluble tissue factor in bronchoalveolar lavage fluid (all p < .05 vs. saline). LPS concurrently inhibited pulmonary fibrinolysis, as reflected by a decrease in bronchoalveolar lavage fluid plasminogen activator activity together with an increase in plasminogen activator inhibitor type 1 (both p < .05 vs. saline). Moreover, LPS inhalation was associated with a suppression of the anticoagulant protein C pathway, as indicated by an increase in soluble thrombomodulin and decreases in protein C and activated protein C levels in bronchoalveolar lavage fluid (all p < .05 vs. saline). Salmeterol, either with or without LPS inhalation, enhanced fibrinolysis (plasminogen activator activity and tissue-type and urokinase-type plasminogen activator levels) but did not influence LPS-induced changes in coagulation or the protein C pathway. CONCLUSIONS: Salmeterol has profibrinolytic properties in the normal lung and when applied in a model of sterile pulmonary inflammation.


Subject(s)
Adrenergic beta-Agonists/therapeutic use , Albuterol/analogs & derivatives , Fibrinolysis/drug effects , Lung/drug effects , Administration, Inhalation , Adrenergic beta-Agonists/pharmacology , Adult , Albuterol/pharmacology , Albuterol/therapeutic use , Analysis of Variance , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Bronchodilator Agents/pharmacology , Bronchodilator Agents/therapeutic use , Factor VIIa/analysis , Factor VIIa/drug effects , Humans , Inflammation , Lipopolysaccharides/adverse effects , Lung Diseases/drug therapy , Lung Diseases/immunology , Lung Diseases/microbiology , Macrophages, Alveolar/drug effects , Male , Plasminogen Activator Inhibitor 1/analysis , Plasminogen Activators/analysis , Plasminogen Activators/drug effects , Protein C/analysis , Protein C/drug effects , Salmeterol Xinafoate , Single-Blind Method , Thrombomodulin/analysis , Thrombomodulin/drug effects , Thromboplastin/analysis , Thromboplastin/drug effects , Tissue Plasminogen Activator/analysis , Tissue Plasminogen Activator/drug effects , Urokinase-Type Plasminogen Activator/analysis , Urokinase-Type Plasminogen Activator/drug effects
13.
J Gastroenterol Hepatol ; 20(6): 882-9, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15946135

ABSTRACT

BACKGROUND: Recombinant human factor VIIa (rhFVIIa) is used to treat hemophilia and occasionally individuals with liver disease. The aim of the present study was to investigate the consequences of rhFVIIa in individuals with advanced liver disease in an attempt to understand the mechanism of action of rhFVIIa in this unique population. METHODS: Levels of plasma tissue factor, plasminogen activator inhibitor-1, tissue factor pathway inhibitor, fibrin split products, D-dimers and free thrombomodulin were measured following the administration of rhFVIIa in 17 subjects. The results were compared to normal controls. RESULTS: The prothrombin time declined from 20.2 +/- 2.8 s to 14.3 +/- 3.9 s (P < 0.01). No change in the activated partial thromboplastin time occurred. A 15.6% reduction in thrombomodulin was observed (P < 0.05). A mean 75.2% reduction in plasma tissue factor occurred (P < 0.01). Tissue factor pathway inhibitor levels declined to less than the control value (P < 0.05). No changes in plasminogen activator inhibitor-1, fibrin split products or D-dimer levels occurred. CONCLUSIONS: These data demonstrate that rhFVIIa administration to individuals with liver disease results in (i) a transient improvement in the prothrombin time; (ii) no change in the activated partial thromboplastin time; and (iii) a marked reduction in the levels of thrombomodulin and tissue factor. These data suggest that rhFVIIa binds tissue factor and enhances tissue factor and thrombomodulin clearance from the circulation.


Subject(s)
Factor VII/therapeutic use , Liver Cirrhosis/drug therapy , Thrombomodulin/blood , Thromboplastin/metabolism , Adult , Aged , Biomarkers/blood , Enzyme-Linked Immunosorbent Assay , Factor VIIa , Female , Humans , Liver Cirrhosis/blood , Male , Middle Aged , Recombinant Proteins/therapeutic use , Thrombomodulin/drug effects , Thromboplastin/drug effects , Treatment Outcome
14.
Eur J Surg Oncol ; 31(3): 217-20, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15780553

ABSTRACT

BACKGROUND: Thrombomodulin (TM) is an endothelial receptor that exerts anti-coagulant, anti-fibrinolytic, and anti-inflammatory activity by inhibiting thrombin and cellular adhesion. There is growing evidence that TM plays a role in tumour behaviour. METHODS: The electronic literature (1966-2004) was reviewed with a specific focus on tumour biology. RESULTS: TM is expressed on both the endothelium and tumour cells in several cancers. Loss of expression denotes a more malignant profile with poorer prognosis. Loss of TM is mediated by hypoxia, endotoxin, and various cytokines, while up-regulation can be achieved by pharmacological manipulation (e.g. pentoxyfylline and statins). CONCLUSION: Originally described as an endothelial anticoagulant, TM plays a key role in tumour biology and prognostics, and provides a potential therapeutic target in impeding cancer spread.


Subject(s)
Gene Expression Regulation, Neoplastic , Neoplasms/metabolism , Thrombomodulin/metabolism , Animals , Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/metabolism , Down-Regulation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Neoplasms/drug therapy , Pentoxifylline/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Predictive Value of Tests , Prognosis , Thrombomodulin/drug effects , Thrombomodulin/genetics
15.
Am J Clin Nutr ; 81(3): 583-9, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15755826

ABSTRACT

BACKGROUND: Dietary factors and very-long-chain n-3 polyunsaturated fatty acids (n-3 PUFAs) may influence the atherothrombotic process. Elevated concentrations of circulating cell adhesion molecules, thrombomodulin (TM), von Willebrand factor (vWF), and tissue-type plasminogen activator antigen (tPAag) are related to atherothrombotic cardiovascular disease. OBJECTIVE: The randomized Diet and Omega-3 Intervention Trial (DOIT) targeted a comparison of the effect of 3-y dietary counseling, n-3 PUFA supplementation (2.4 g/d), or both on circulating markers of endothelial activation. DESIGN: The study included 563 elderly men with long-standing hyperlipidemia. The men were randomly assigned by factorial design into 4 groups: control (no dietary counseling and placebo capsules), dietary counseling (and placebo capsules), n-3 PUFA supplementation (no dietary counseling), and dietary counseling and n-3 PUFA supplementation. RESULTS: Serum concentrations of fatty acids reflected good compliance. Dietary counseling was followed by significantly reduced concentrations of soluble intercellular adhesion molecule 1 (sICAM-1; P < 0.001), sTM (P = 0.004), and tPAag (P < 0.001) than in subjects without dietary counseling. After n-3 PUFA supplementation, significantly reduced concentrations of sICAM-1 (P < 0.001) and sTM (P = 0.006) were observed when compared with subjects receiving placebo capsules. An increase in tPAag was not significantly different from that observed in subjects receiving placebo capsules. For sICAM-1, a significant effect was observed for both interventions combined. CONCLUSIONS: Each intervention (dietary counseling or n-3 PUFA supplements) reduced sTM and sICAM-1 concentrations, indicating decreased endothelial activation. The tPAag increase in the groups not receiving dietary counseling (pooled), which indicates progression of atherosclerosis, was significantly counteracted by dietary counseling.


Subject(s)
Arteriosclerosis/blood , Fatty Acids, Omega-3/therapeutic use , Hyperlipidemias/diet therapy , Intercellular Adhesion Molecule-1/blood , Thrombomodulin/blood , Tissue Plasminogen Activator/blood , Aged , Arteriosclerosis/prevention & control , Biomarkers/blood , Counseling , Diet , Dietary Supplements , Disease Progression , Double-Blind Method , Fatty Acids, Omega-3/administration & dosage , Fatty Acids, Omega-3/blood , Humans , Intercellular Adhesion Molecule-1/drug effects , Male , Middle Aged , Thrombomodulin/drug effects , Tissue Plasminogen Activator/drug effects , von Willebrand Factor/analysis
16.
Thromb Res ; 115(5): 417-26, 2005.
Article in English | MEDLINE | ID: mdl-15733976

ABSTRACT

The objective of this study was to elucidate the effects of tumor necrosis factor-alpha (TNF-alpha) on the expression of thrombomodulin (TM) and endothelial protein C receptor (EPCR) in human endothelial cells as well as the effect of curcumin, a spice and coloring food compound, as a potential therapeutic agent. Human umbilical vein endothelial cells (HUVECs) treated with TNF-alpha (2.0 ng/ml) showed reduced TM mRNA levels by 80%, 97%, 94%, and 97% at 3, 6, 12, and 24 h, respectively (P<0.05), by real-time PCR analysis. Dose-dependent study showed that TM mRNA levels of HUVECs were decreased by 86%, 89%, 91%, and 94% after treatment of TNF-alpha (0, 0.25, 0.5, 1, and 2 ng/ml) for 6 h, respectively (P<0.05). TM protein levels in HUVECs were significantly reduced by 69% in TNF-alpha-treated cells as compared to controls (P<0.05) by Western blot analysis. Secreted protein and activity of TM of HUVEC cultures were also significantly reduced in TNF-alpha-treated cells. In addition, EPCR mRNA levels of HUVECs were significantly reduced in TNF-alpha-treated group as compared to controls (P<0.05). Furthermore, these effects were observed in other types of endothelial cells from human coronary arteries, lung, and skin. Curcumin effectively blocked these effects of TNF-alpha on downregulation of TM and EPCR. These data demonstrate that TNF-alpha significantly decreases expression of TM and EPCR at both mRNA and protein levels in several human endothelial cells. Curcumin can effectively block TNF-alpha-induced endothelial dysfunction. This study suggests a new molecular mechanism of inflammation-induced thrombosis and a new therapeutic strategy to prevent this clinical problem.


Subject(s)
Curcumin/pharmacology , Endothelial Cells/drug effects , Gene Expression Regulation/drug effects , Glycoproteins/genetics , Thrombomodulin/genetics , Tumor Necrosis Factor-alpha/pharmacology , Antigens, CD , Dose-Response Relationship, Drug , Down-Regulation , Endothelial Cells/cytology , Endothelial Cells/metabolism , Endothelial Protein C Receptor , Glycoproteins/drug effects , Glycoproteins/metabolism , Humans , RNA, Messenger/genetics , Receptors, Cell Surface , Thrombomodulin/drug effects , Thrombomodulin/metabolism
17.
Drugs Exp Clin Res ; 30(3): 125-32, 2004.
Article in English | MEDLINE | ID: mdl-15366789

ABSTRACT

Essential hypertension is often accompanied by abnormalities of the coagulation/fibrinolytic system predisposing to a procoagulant state. The aim of the present study was to examine the comparative efficacy of the angiotensin II type 1 receptor antagonists eprosartan and losartan on plasma levels of hemostatic/fibrinolytic and endothelial function markers in a cohort of previously untreated hypertensive patients. A total of 86 patients whose hypertension was controlled by monotherapy with eprosartan 600 mg (45 patients) or losartan 100 mg (41 patients) were studied. The plasma levels of plasminogen activator inhibitor-1 (PAI-1) antigen, tissue plasminogen activator inhibitor (tPA) antigen, thrombomodulin (TM), tissue factor pathway inhibitor (TFPI) antigen, and fibrinogen were determined before and after 6 months of therapy. Age, sex distribution, body mass index, lipid profile, systolic and diastolic blood pressure levels, and baseline values of the measured markers were similar in both groups. After 6 months of therapy, systolic blood pressure was significantly lower in patients treated with eprosartan, while no differences were observed with respect to diastolic blood pressure. Treatment with both drugs was associated with a significant decrease in PAI-1 antigen, TM, fibrinogen plasma levels and an increase in tPA antigen. The favorable modification of all the above parameters was significantly greater in the eprosartan than in the losartan group, while TFPI plasma levels were decreased to a similar extent with both drugs. In conclusion, the results of our study indicate that 6-month monotherapy with a new angiotensin II type 1 receptor blocker, eprosartan, is associated with a more favorable modification of hemostatic/fibrinolytic status than with losartan.


Subject(s)
Acrylates/therapeutic use , Fibrinolysis/drug effects , Hemostasis/drug effects , Hypertension/drug therapy , Imidazoles/therapeutic use , Losartan/therapeutic use , Thiophenes/therapeutic use , Acrylates/pharmacology , Aged , Angiotensin II Type 1 Receptor Blockers , Drug Administration Schedule , Female , Fibrinogen/chemistry , Fibrinogen/drug effects , Fibrinolysis/physiology , Follow-Up Studies , Hemostasis/physiology , Humans , Hypertension/physiopathology , Imidazoles/pharmacology , Lipoproteins/blood , Losartan/pharmacology , Male , Middle Aged , Plasminogen Activator Inhibitor 1/blood , Receptor, Angiotensin, Type 1/therapeutic use , Thiophenes/pharmacology , Thrombomodulin/blood , Thrombomodulin/drug effects , Time Factors , Tissue Plasminogen Activator/antagonists & inhibitors , Tissue Plasminogen Activator/blood , Tissue Plasminogen Activator/drug effects
18.
Blood Coagul Fibrinolysis ; 14(6): 575-85, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12960612

ABSTRACT

Expression of functionally active thrombomodulin (TM) on the luminal surface of endothelial cells is critical for vascular thromboresistance. TM maintains thrombohemorrhagic homeostasis by forming a complex with thrombin, which subsequently loses its procoagulant properties and instead activates protein C. Acquired deficiency of endothelial TM is of particular pathophysiological significance in sepsis and related disorders. We show here that two different 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors (statins), atorvastatin and simvastatin, strongly increase the expression and functional activity of TM in human umbilical vein endothelial cells, human coronary artery endothelial cells, and EA.hy926 endothelial cells. The increase in endothelial TM conferred by statin was prevented by the addition of mevalonic acid, geranylgeranyl-pyrophosphate, and nitric oxide scavenger, and was mimicked by the addition of a specific inhibitor of geranylgeranyl transferase, as well as by nitric oxide donors. Moreover, statin counteracted tumor necrosis factor alpha-induced downregulation of endothelial cell TM. The increase in endothelial cell TM activity in response to statin constitutes a novel pleiotropic (non-lipid-related) effect of these commonly used compounds, and may be of clinical significance in disorders where deficient endothelial TM and protein C activation play a pathophysiological role.


Subject(s)
Endothelium, Vascular/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Nitric Oxide/metabolism , Thrombomodulin/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Anticholesteremic Agents/pharmacology , Atorvastatin , Cells, Cultured , Dose-Response Relationship, Drug , Endothelium, Vascular/cytology , Gene Expression Regulation/drug effects , Heptanoic Acids/pharmacology , Humans , Kinetics , Polyisoprenyl Phosphates/metabolism , Pyrroles/pharmacology , Simvastatin/pharmacology , Thrombomodulin/biosynthesis
20.
Liver Int ; 23(2): 110-5, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12654133

ABSTRACT

BACKGROUND: Damage to endothelial cells may be an important factor in the complications of acute liver failure, resulting in multi-organ failure. The aim of this study was to assess endothelial cell function in patients with severe hepatotoxicity due to paracetamol ingestion. PATIENTS AND METHODS: Fifty-eight patients with paracetamol-induced hepatotoxicity were studied for up to 7 days. Serum hyaluronic acid (HA), as a marker of hepatic sinusoidal endothelial cell function, was determined using an enzyme-linked binding assay. Plasma von Willebrand Factor, thrombomodulin and interleukin-8 were also determined using ELISA. RESULTS: Serum HA on admission was significantly increased (median 6777 ng/ml, range 24-50 967 ng/ml) as compared to normal controls (n = 10, median 21 ng/ml, range 0-50 ng/ml; P < 0.001). In non-survivors (n = 21) HA levels peaked on day 2 after admission (P = 0.044), and then decreased. In the survivors (n = 37) the levels of HA did not increase further. Plasma von Willebrand Factor, plasma thrombomodulin and serum interleukin-8 were significantly increased in the patients as compared to the normal controls (P < 0.001). Serum interleukin-8 was significantly higher in non-survivors in the first 2 days. CONCLUSIONS: Endothelial function is abnormal in paracetamol-induced hepatotoxicity. Damage to hepatic sinusoidal endothelial cells assessed by serum HA was greater in non-survivors than survivors.


Subject(s)
Acetaminophen/adverse effects , Analgesics, Non-Narcotic/adverse effects , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Hyaluronic Acid/blood , Liver Failure, Acute/chemically induced , Adolescent , Adult , Aged , Biomarkers/blood , Endothelium, Vascular/metabolism , Female , Humans , Interleukin-8/blood , Liver Failure, Acute/mortality , Liver Failure, Acute/therapy , Liver Transplantation , London , Male , Middle Aged , Patient Admission , Statistics as Topic , Survival Analysis , Thrombomodulin/blood , Thrombomodulin/drug effects , Time Factors , Treatment Outcome , Ultrafiltration , von Willebrand Factor/drug effects , von Willebrand Factor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...