Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Pharmacol ; 80(2): 337-44, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21596928

ABSTRACT

Thrombin promotes vascular smooth muscle cell (SMC) proliferation and inflammation via protease-activated receptor (PAR)-1. A further thrombin receptor, PAR-3, acts as a PAR-1 cofactor in some cell-types. Unlike PAR-1, PAR-3 is dynamically regulated at the mRNA level in thrombin-stimulated SMC. This study investigated the mechanisms controlling PAR-3 expression. In human vascular SMC, PAR-3 siRNA attenuated thrombin-stimulated interleukin-6 expression and extracellular signal-regulated kinases 1/2 phosphorylation, indicating PAR-3 contributes to net thrombin responses in these cells. Thrombin slowed the decay of PAR-3 but not PAR-1 mRNA in the presence of actinomycin D and induced cytosolic shuttling and PAR-3 mRNA binding of the mRNA-stabilizing protein human antigen R (HuR). HuR siRNA prevented thrombin-induced PAR-3 expression. By contrast, forskolin inhibited HuR shuttling and destabilized PAR-3 mRNA, thus reducing PAR-3 mRNA and protein expression. Other cAMP-elevating agents, including the prostacyclin-mimetic iloprost, also down-regulated PAR-3, accompanied by decreased HuR/PAR-3 mRNA binding. Iloprost-induced suppression of PAR-3 was reversed with a myristoylated inhibitor of protein kinase A and mimicked by phorbol ester, an inducer of cyclooxygenase-2. In separate studies, iloprost attenuated PAR-3 promoter activity and prevented binding of nuclear factor of activated T cells (NFAT2) to the human PAR-3 promoter in a chromatin immunoprecipitation assay. Accordingly, PAR-3 expression was suppressed by the NFAT inhibitor cyclosporine A or NFAT2 siRNA. Thus human PAR-3, unlike PAR-1, is regulated post-transcriptionally via the mRNA-stabilizing factor HuR, whereas transcriptional control involves NFAT2. Through modulation of PAR-3 expression, prostacyclin and NFAT inhibitors may limit proliferative and inflammatory responses to thrombin after vessel injury.


Subject(s)
Muscle, Smooth, Vascular/physiology , NFATC Transcription Factors/physiology , RNA Stability/genetics , RNA, Messenger/genetics , Receptors, Thrombin/physiology , Antigens, Surface/genetics , Antigens, Surface/physiology , Cyclic AMP/metabolism , ELAV Proteins , ELAV-Like Protein 1 , Gene Knockdown Techniques/methods , Humans , NFATC Transcription Factors/genetics , Oligopeptides/genetics , Oligopeptides/physiology , RNA Processing, Post-Transcriptional/genetics , RNA Processing, Post-Transcriptional/physiology , RNA-Binding Proteins/genetics , RNA-Binding Proteins/physiology , Receptors, Thrombin/genetics
2.
Arterioscler Thromb Vasc Biol ; 31(3): 624-33, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21164077

ABSTRACT

OBJECTIVE: Diabetes is associated with vascular remodeling and increased thrombin generation. Thrombin promotes vascular smooth muscle cell (SMC) mitogenesis and migration via protease-activated receptors (PAR)-1, PAR-3, and PAR-4. We investigated the effect of high glucose on expression and function of vascular thrombin receptors. METHODS AND RESULTS: In human vascular SMCs, high glucose (25 versus 5.5 mmol/L) induced a rapid and sustained increase in PAR-4 mRNA, protein, and cell surface expression. PAR-1 and PAR-3 expression were not changed. High glucose pretreatment (48 hours) enhanced thrombin or PAR-4-activating peptide but not PAR-1-activating peptide evoked intracellular calcium mobilization, migration, and tumor necrosis factor α gene expression. This enhancement of thrombin-stimulated migration and gene expression by high glucose was abolished by endogenous PAR-4 knockdown. PAR-4 regulation was prevented by inhibition of protein kinase (PK)C-ß and -δ isoforms or nuclear factor (NF)κB. Nuclear translocation of NFκB in high glucose-stimulated SMCs led to PKC-dependent NFκB binding to the PAR-4 promoter in a chromatin immunoprecipitation assay. Furthermore, in situ hybridization and immunohistochemistry confirmed high abundance of PAR-4 in human diabetic vessels as compared with nondiabetic vessels. CONCLUSIONS: High glucose enhances SMC responsiveness to thrombin through transcriptional upregulation of PAR-4, mediated via PKC-ß, -δ, and NFκB. This may play an important role in the vascular complications of diabetes.


Subject(s)
Calcium Signaling , Diabetic Angiopathies/metabolism , Glucose/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Receptors, Thrombin/metabolism , Thrombin/metabolism , Binding Sites , Calcium/metabolism , Cell Movement , Cells, Cultured , Chromatin Immunoprecipitation , Diabetic Angiopathies/genetics , Humans , Immunohistochemistry , In Situ Hybridization , NF-kappa B/metabolism , Promoter Regions, Genetic , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Protein Kinase C beta , Protein Kinase C-delta/genetics , Protein Kinase C-delta/metabolism , Protein Kinase Inhibitors/pharmacology , RNA Interference , RNA, Messenger/metabolism , Receptor, PAR-1/metabolism , Receptors, Thrombin/genetics , Time Factors , Transcriptional Activation , Tumor Necrosis Factor-alpha/genetics , Up-Regulation
3.
Thromb Haemost ; 103(5): 884-90, 2010 May.
Article in English | MEDLINE | ID: mdl-20143010

ABSTRACT

The vast majority of thrombin (>95%) is generated after clotting is completed, suggesting that thrombin formation serves purposes beyond coagulation, such as tissue repair after vessel injury. Two types of vascular thrombin binding sites exist: protease-activated receptors (PARs) and thrombomodulin (TM). Their expression is low in contractile vascular smooth muscle cells (SMC), the dominating subendothelial cell population, but becomes markedly up-regulated upon injury. In human SMC, PAR-1, PAR-3, and PAR-4 mediate thrombin-induced proliferation, migration and matrix biosynthesis as well as generation of inflammatory and growth-promoting mediators. Thrombin-responsive PARs are transcriptionally down-regulated in human vascular SMC by vasodilatory prostaglandins (PGI2/PGE2). For PAR-1 and PAR-3 this mechanism involves cAMP-dependent inactivation of the transcription factor NFAT. The human PAR-4 promoter does not possess NFAT recognition motifs suggesting involvement of other cAMP-regulated effectors. Unlike PARs, TM is induced in SMC exposed to vasodilatory prostaglandins. Enhanced thrombin binding to TM might ameliorate PAR-mediated SMC stimulation. Also expressed in human SMC is the endothelial protein C receptor (EPCR), which serves as an anchor to facilitate generation of activated protein C (aPC) by TM-bound thrombin. Whether prostaglandins affect aPC-generation is not known. In SMC, thrombin and aPC act synergistically via PAR-1 to modify tissue remodelling, in contrast to their antagonistic interaction in the coagulation pathways. Overall, this will contribute to plaque stability and wound healing. The processes outlined here are likely to become clinically relevant after up-regulation of vascular cyclooxygenase2, the rate limiting step in vascular PGE2/PGI2 biosynthesis, such as in advanced atherosclerosis and acute coronary syndromes.


Subject(s)
Myocytes, Smooth Muscle/metabolism , Prostaglandins/metabolism , Receptors, Thrombin/metabolism , Thrombosis/metabolism , Vasodilation , Acute Coronary Syndrome/metabolism , Acute Coronary Syndrome/pathology , Animals , Atherosclerosis/metabolism , Atherosclerosis/pathology , Blood Coagulation , Gene Expression Regulation , Humans , Myocytes, Smooth Muscle/pathology , Receptors, Thrombin/genetics , Thrombosis/pathology , Thrombosis/physiopathology , Wound Healing
4.
Circ Res ; 100(2): 255-62, 2007 Feb 02.
Article in English | MEDLINE | ID: mdl-17170365

ABSTRACT

The endothelial cell protein C receptor (EPCR) is expressed on endothelial cells and regulates the protein C anticoagulant pathway via the thrombin-thrombomodulin complex. Independent of its anticoagulant activity, activated protein C (APC) can directly signal to endothelial cells and upregulate antiapoptotic and antiinflammatory genes. Here we show that vascular smooth muscle cells (SMCs) also express EPCR. EPCR protein on SMCs was detected by flow cytometry and Western blotting. EPCR mRNA was identified by quantitative RT-PCR. To examine the functionality of EPCR, intracellular signaling in APC-stimulated SMCs was analyzed by determination of intracellular free calcium transients using confocal laser scanning microscopy. Phosphorylation of extracellular signal-regulated kinases 1 and 2 (ERK-1/2) was detected by immunoblotting. APC-induced ERK-1/2 phosphorylation was inhibited by an anti-EPCR antibody and by a cleavage site blocking anti-PAR-1 antibody, indicating that binding of APC to EPCR and cleavage of protease-activated receptor-1 (PAR-1) were involved. APC elicited an increase in [(3)H]-thymidine incorporation. The mitogenic effect of APC was significantly enhanced in the presence of thrombin. EPCR expression was also detected in SMCs in the fibrous cap of human carotid artery plaques. The present data demonstrate functionally active EPCR in SMCs and suggest that EPCR-bound APC might modulate PAR-1-mediated responses of SMCs to vascular injury.


Subject(s)
Antigens, CD/biosynthesis , Antigens, CD/genetics , Endothelium, Vascular/metabolism , Gene Expression Regulation/physiology , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Protein C/metabolism , Receptors, Cell Surface/biosynthesis , Receptors, Cell Surface/genetics , Antigens, CD/physiology , Carotid Stenosis/enzymology , Carotid Stenosis/metabolism , Carotid Stenosis/pathology , Cells, Cultured , Coculture Techniques , Endothelial Protein C Receptor , Endothelium, Vascular/enzymology , Endothelium, Vascular/pathology , Humans , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/enzymology , Myocytes, Smooth Muscle/pathology , Protein Binding/physiology , Receptors, Cell Surface/physiology
5.
Proc Natl Acad Sci U S A ; 103(16): 6326-31, 2006 Apr 18.
Article in English | MEDLINE | ID: mdl-16606835

ABSTRACT

Cysteinyl leukotrienes (cysLT), i.e., LTC4, LTD4, and LTE4, are lipid mediators derived from the 5-lipoxygenase pathway, and the cysLT receptors cysLT1-R/cysLT2-R mediate inflammatory tissue reactions. Although endothelial cells (ECs) predominantly express cysLT2-Rs, their role in vascular biology remains to be fully understood. To delineate cysLT2-R actions, we stimulated human umbilical vein EC with LTD4 and determined early induced genes. We also compared LTD4 effects with those induced by thrombin that binds to protease-activated receptor (PAR)-1. Stringent filters yielded 37 cysLT2-R- and 34 PAR-1-up-regulated genes (>2.5-fold stimulation). Most LTD4-regulated genes were also induced by thrombin. Moreover, LTD4 plus thrombin augmented gene expression when compared with each agonist alone. Strongly induced genes were studied in detail: Early growth response (EGR) and nuclear receptor subfamily 4 group A transcription factors; E-selectin; CXC ligand 2; IL-8; a disintegrin-like and metalloprotease (reprolysin type) with thrombospondin type 1 motif 1 (ADAMTS1); Down syndrome critical region gene 1 (DSCR1); tissue factor (TF); and cyclooxygenase 2. Transcripts peaked at approximately 60 min, were unaffected by a cysLT1-R antagonist, and were superinduced by cycloheximide. The EC phenotype was markedly altered: LTD4 induced de novo synthesis of EGR1 protein and EGR1 localized in the nucleus; LTD4 up-regulated IL-8 formation and secretion; and LTD4 raised TF protein and TF-dependent EC procoagulant activity. These data show that cysLT2-R activation results in a proinflammatory EC phenotype. Because LTD4 and thrombin are likely to be formed concomitantly in vivo, cysLT2-R and PAR-1 may cooperate to augment vascular injury.


Subject(s)
Endothelium, Vascular/metabolism , Gene Expression Regulation , Leukotriene C4/metabolism , Membrane Proteins/physiology , Receptor, PAR-1/physiology , Receptors, Leukotriene/physiology , Thrombin/metabolism , Blood Coagulation/drug effects , Blood Coagulation/physiology , Cells, Cultured , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelium, Vascular/drug effects , Humans , Leukotriene C4/pharmacology , Membrane Proteins/agonists , Receptor, PAR-1/agonists , Receptors, Cytoplasmic and Nuclear , Receptors, Leukotriene/agonists , Thrombin/pharmacology , Transcription, Genetic/drug effects , Umbilical Veins/cytology
6.
Circ Res ; 96(1): e1-6, 2005 Jan 07.
Article in English | MEDLINE | ID: mdl-15591227

ABSTRACT

There is concern that cyclooxygenase (COX)-2 inhibitors may promote atherothrombosis by inhibiting vascular formation of prostacyclin (PGI2) and an increased thrombotic risk of COX-2 inhibitors has been reported. It is widely accepted that the prothrombotic effects of COX-2 inhibitors can be explained by the removal of platelet-inhibitory PGI2. Using microarray chip technology, we have previously demonstrated that thrombomodulin (TM) mRNA is upregulated in cultured human coronary artery smooth muscle cells by the stable prostacyclin mimetic iloprost. This study is the first to demonstrate a stimulation of the expression of functionally active thrombomodulin in human smooth muscle cells by prostaglandins, endogenously formed via the COX-2 pathway. Because TM is an important inhibitor of blood coagulation, these findings provide a novel platelet-independent mechanism to explain the prothrombotic effects of COX-2 inhibitors. The full text of this article is available online at http://circres.ahajournals.org.


Subject(s)
Alprostadil/analogs & derivatives , Cyclooxygenase Inhibitors/toxicity , Epoprostenol/analogs & derivatives , Gene Expression Regulation/physiology , Myocytes, Smooth Muscle/drug effects , Prostaglandin-Endoperoxide Synthases/physiology , Pyridines/toxicity , Sulfones/toxicity , Thrombomodulin/biosynthesis , Thrombophilia/chemically induced , Alprostadil/pharmacology , Blood Coagulation/physiology , Bucladesine/pharmacology , Carotid Artery Diseases/enzymology , Carotid Artery Diseases/pathology , Carotid Artery, Internal/chemistry , Carotid Artery, Internal/enzymology , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Colforsin/pharmacology , Coronary Vessels/cytology , Culture Media, Serum-Free , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Diclofenac/pharmacology , Dinoprostone/pharmacology , Epoprostenol/pharmacology , Etoricoxib , Gene Expression Profiling , Gene Expression Regulation/drug effects , Humans , Iloprost/pharmacology , Isoquinolines/pharmacology , Mammary Arteries/cytology , Membrane Proteins , Models, Biological , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/enzymology , Myocytes, Smooth Muscle/metabolism , Oligonucleotide Array Sequence Analysis , Prostaglandins/deficiency , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Prostaglandin/antagonists & inhibitors , Receptors, Prostaglandin E/antagonists & inhibitors , Receptors, Prostaglandin E, EP3 Subtype , Saphenous Vein/cytology , Second Messenger Systems/drug effects , Sulfonamides/pharmacology , Tetradecanoylphorbol Acetate/pharmacology , Thrombomodulin/genetics , Thrombophilia/blood , Thrombophilia/physiopathology , Vasodilator Agents/pharmacology
7.
Thromb Haemost ; 90(4): 704-9, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14515192

ABSTRACT

The present study investigates whether vascular smooth muscle cells of the human saphenous vein (SMC) express a functionally active protease-activated receptor-3 (PAR-3). PAR-3 mRNA was detected by RT-PCR. In the presence of thrombin, a rapid and transient increase in PAR-3 mRNA was observed. Stimulation of SMC with thrombin or the synthetic PAR-3-activating peptide, TFRGAP, resulted in transient mobilization of intracellular calcium. After a preceding challenge with thrombin, the calcium signal to TFRGAP was abolished, suggesting cleavage and subsequent desensitization of PAR-3 by thrombin. Activation of PAR-3 by TFRGAP elicited a time-dependent activation of the extracellular-signal-regulated kinase (ERK)-1/2 with a maximum response 10-20 min after stimulation. At 200 microM, TFRGAP increased [3H]-thymidine incorporation into cellular DNA about two-fold. These data indicate that PAR-3 is expressed in human SMC and triggers intracellular signaling. Thus, in the SMC PAR-3 might contribute to thrombin-induced responses.


Subject(s)
Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/chemistry , Receptors, Thrombin/analysis , Calcium Signaling , DNA Replication/drug effects , Humans , Kinetics , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Muscle, Smooth, Vascular/chemistry , RNA, Messenger/analysis , RNA, Messenger/drug effects , Receptors, Thrombin/genetics , Receptors, Thrombin/physiology , Saphenous Vein , Thrombin/pharmacology
8.
Arterioscler Thromb Vasc Biol ; 23(8): e32-6, 2003 Aug 01.
Article in English | MEDLINE | ID: mdl-12816882

ABSTRACT

OBJECTIVE: Inflammatory infiltrates and atherosclerotic lesions emerge when monocytes adhere to endothelial cells (ECs), migrate into the subendothelial space, and become macrophages (MPhi(s)). Leukotrienes (LTs), products of 5-lipoxygenase, are powerful inflammatory mediators. 5-lipoxygenase+ MPhi(s) have been shown to increase during atherogenesis, and LT receptor (LT-R) transcripts were identified in diseased arteries. To investigate LT-Rs in cells involved in inflammation and atherogenesis, we used the in vitro models of human umbilical vein ECs (HUVECs) and monocyte-derived MPhi(s). METHODS AND RESULTS: HUVECs primarily expressed transcripts of the cysteinyl (cys) LT2-R, which was strongly upregulated by interleukin-4. By contrast, MPhi(s) predominantly expressed transcripts of the cysLT1-R. Calcium responses toward LTs revealed differential cysLT-R utilization by both cell types: HUVECs responded to both cysLTs, whereas MPhi(s) preferentially responded to LTD4; HUVECs, but not MPhi(s), were resistant toward a cysLT1-R antagonist, montelukast; cysLTs generated regular calcium oscillations in HUVECs that lasted >60 minutes, resulting in >500 oscillations per cell. By contrast, calcium elevations in MPhi(s) returned to baseline within seconds and were nonoscillatory. CONCLUSIONS: Our data raise the possibility that MPhi-derived LTs differentially activate cysLT2-Rs via paracrine stimulation and cysLT1-Rs via autocrine and paracrine stimulation during inflammation and atherogenesis.


Subject(s)
Arachidonate 5-Lipoxygenase/metabolism , Arteriosclerosis/metabolism , Calcium/metabolism , Endothelium, Vascular/metabolism , Macrophages/metabolism , Receptors, Leukotriene/genetics , Receptors, Leukotriene/metabolism , Arteriosclerosis/etiology , Cells, Cultured , Gene Expression Profiling , Humans , Inflammation/complications , Inflammation/physiopathology , Up-Regulation
9.
Circ Res ; 90(10): 1122-7, 2002 May 31.
Article in English | MEDLINE | ID: mdl-12039803

ABSTRACT

Pro-matrix metalloproteinase-2 (pro-MMP-2) is expressed in vascular smooth muscle cells (SMCs). We report that activated coagulation factor X (FXa) induces the release of MMP-2 (65 kDa) from human SMCs. In addition, FXa cleaves pro-MMP-2 (72 kDa) into MMP-2. Pro-MMP-2 and MMP-2 were determined by gelatin zymography. MMP-2 was generated in conditioned medium containing pro-MMP-2 in a concentration-dependent fashion by FXa (3 to 100 nmol/L). FX at concentrations up to 300 nmol/L was ineffective. The conversion of pro-MMP-2 to MMP-2 was inhibited by a selective FXa inhibitor (DX-9065a) at 3 to 10 micromol/L. There was a concentration-dependent induction of an intermediate MMP-2 form (68 kDa) in lysates of FXa-treated cells. This indicates that cellular mechanisms are involved in FXa-induced conversion of pro-MMP-2. As a possible biological consequence of MMP-2 activation by FXa, DNA synthesis and matrix invasion of SMCs were determined. Both were stimulated by FXa and inhibited by the selective FXa inhibitor DX-9065a and the MMP inhibitor GM 6001 but not by hirudin or aprotinin. It is concluded that stimulation of SMCs by FXa increases the levels of MMP-2 in the extracellular space and that two different mechanisms are involved: release of active MMP-2 and cleavage of secreted pro-MMP-2. Both might contribute to the mitogenic potency of FXa and FXa-stimulated matrix invasion of SMCs.


Subject(s)
Enzyme Precursors/metabolism , Factor Xa/pharmacology , Gelatinases/metabolism , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 2/physiology , Metalloendopeptidases/metabolism , Muscle, Smooth, Vascular/enzymology , Arteries/cytology , Arteries/enzymology , Cell Movement , Cells, Cultured , Collagenases/metabolism , Culture Media, Conditioned , DNA/biosynthesis , Dose-Response Relationship, Drug , Extracellular Matrix/metabolism , Factor Xa Inhibitors , Humans , Matrix Metalloproteinase 9 , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiology , Naphthalenes/pharmacology , Propionates/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...