Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
2.
PPAR Res ; 2008: 328172, 2008.
Article in English | MEDLINE | ID: mdl-18288284

ABSTRACT

Historically, platelets were viewed as simple anucleate cells responsible for initiating thrombosis and maintaining hemostasis, but clearly they are also key mediators of inflammation and immune cell activation. An emerging body of evidence links platelet function and thrombosis to vascular inflammation. peroxisome proliferator-activated receptors (PPARs) play a major role in modulating inflammation and, interestingly, PPARs (PPARbeta/delta and PPARgamma) were recently identified in platelets. Additionally, PPAR agonists attenuate platelet activation; an important discovery for two reasons. First, activated platelets are formidable antagonists that initiate and prolong a cascade of events that contribute to cardiovascular disease (CVD) progression. Dampening platelet release of proinflammatory mediators, including CD40 ligand (CD40L, CD154), is essential to hinder this cascade. Second, understanding the biologic importance of platelet PPARs and the mechanism(s) by which PPARs regulate platelet activation will be imperative in designing therapeutic strategies lacking the deleterious or unwanted side effects of current treatment options.

3.
J Thromb Haemost ; 5(5): 942-9, 2007 May.
Article in English | MEDLINE | ID: mdl-17461928

ABSTRACT

OBJECTIVES: To evaluate the effects of pioglitazone on insulin sensitivity and levels of biomarkers associated with thrombotic risk in overweight and obese, non-diabetic subjects with coronary artery disease. BACKGROUND: Little information is available regarding the effects of thiazolidinediones in the absence of diabetes. Further, although postprandial hyperlipemia is a risk factor for cardiovascular diseases, there is limited information about the postprandial effects. METHODS: Twenty overweight and obese, non-diabetic patients with coronary artery disease were enrolled in a randomized, placebo-controlled, double-blind study. Subjects were on atorvastatin for the duration of the study and received either placebo or pioglitazone (45 mg day(-1)) for 12 weeks and then crossed over to the alternative therapy for an additional 12 weeks. Insulin sensitivity, fasting and postprandial levels of lipid, hemostatic, and inflammatory variables were measured, and endothelial function was assessed. RESULTS: Insulin sensitivity improved from 0.03 micromol kg(-1) x min pM(-1) on placebo to 0.04 on pioglitazone (P = 0.0002), and there were decreases in fasting levels of factor (F) VII:C (102 +/- 17% to 92 +/- 18%, P = 0.001), FVII:Ag (68 +/- 12% to 60 +/- 14%, P = 0.01) and in von Willebrand factor (VWF) (174 +/- 94% to 142 +/- 69%, P = 0.01). Pioglitazone lowered postprandial levels of FVII:Ag, FVII:C, plasminogen activator inhibitor-1, VWF, and triglycerides, and increased high-density lipoproteins (+9%, P = 0.02). CONCLUSIONS: Pioglitazone improves insulin sensitivity and favorably modifies fasting and postprandial lipid, hemostatic and inflammatory markers of the metabolic syndrome in overweight and obese non-diabetic patients with coronary artery disease.


Subject(s)
Coronary Artery Disease/drug therapy , Fasting , Hemostasis/drug effects , Hyperlipidemias/drug therapy , Postprandial Period , Thiazolidinediones/therapeutic use , Adult , Aged , Coronary Artery Disease/complications , Cross-Over Studies , Double-Blind Method , Female , Humans , Hyperlipidemias/complications , Insulin/blood , Male , Middle Aged , Overweight , Pioglitazone , Thiazolidinediones/pharmacology
4.
J Thromb Haemost ; 4(1): 158-67, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16409465

ABSTRACT

Tissue factor (TF) is a transmembrane glycoprotein that initiates coagulation and plays a critical role in regulating hemostasis and thrombosis. We have recently reported a naturally occurring, soluble form of human tissue factor (asTF) generated by alternative splicing. This splice variant has a novel C-terminus with no homology to that of the full-length TF (flTF), lacks a transmembrane domain, and is active in the presence of phospholipids. Mouse models offer unique opportunities to examine the relative importance of flTF and asTF in mediating thrombosis, the response to arterial injury, and ischemic damage. To that end, we have identified and characterized murine asTF (masTF). Like the human splice variant, masTF lacks a transmembrane domain and has a unique C-terminus. We have generated antibodies specific to masTF and murine flTF (mflTF) to examine the expression of both forms of TF. masTF antigen is widely and abundantly expressed, with a pattern similar to that of mflTF, in adult tissues, in experimentally induced thrombi, and during development. These studies demonstrate that masTF contributes to the pool of total TF and may thus play an important role in mediating TF-dependent processes.


Subject(s)
Alternative Splicing , Thromboplastin/genetics , Amino Acid Sequence , Animals , Antibodies, Monoclonal , Cells, Cultured , Gene Expression Regulation, Developmental , Immunohistochemistry , Mice , Mice, Inbred C57BL , Myocytes, Smooth Muscle , RNA, Messenger/analysis , Solubility , Thromboplastin/analysis , Thromboplastin/chemistry , Thrombosis , Tissue Distribution
5.
Proc Natl Acad Sci U S A ; 98(18): 10142-7, 2001 Aug 28.
Article in English | MEDLINE | ID: mdl-11504923

ABSTRACT

There have been increasing reports of acute coronary thrombotic events in patients with HIV. Although these clinical events have been attributed primarily to dyslipidemia associated with protease inhibitor therapy, autopsy studies in children with HIV suggest the presence of an underlying arteriopathy. This study demonstrates that the HIV envelope protein, gp120, activates human arterial smooth muscle cells to express tissue factor, the initiator of the coagulation cascade. The induction of tissue factor by gp120 is mediated by two biologically relevant coreceptors for HIV infection, CXCR4 and CCR5, and is also dependent on the presence of functional CD4. Induction of tissue factor by gp120 requires activation of mitogen-activating protein kinases, activation of protein kinase C, and generation of reactive oxygen species, signaling pathways that have protean effects on smooth muscle cell physiology. The activation of smooth muscle cells by gp120 may play an important role in the vascular, thrombotic, and inflammatory responses to HIV infection.


Subject(s)
HIV Envelope Protein gp120/toxicity , Muscle, Smooth, Vascular/drug effects , CD4 Antigens/metabolism , Cells, Cultured , Chemokine CXCL12 , Chemokines, CXC/metabolism , Chemokines, CXC/pharmacology , Coronary Thrombosis/etiology , HIV Infections/complications , Humans , Ligands , Mitogen-Activated Protein Kinases/metabolism , Muscle, Smooth, Vascular/physiology , Muscle, Smooth, Vascular/virology , Protein Kinase C/metabolism , Reactive Oxygen Species/metabolism , Receptors, CCR5/metabolism , Receptors, CXCR4/metabolism , Recombinant Proteins/toxicity , Thromboplastin/biosynthesis
6.
Lab Invest ; 81(6): 895-903, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11406650

ABSTRACT

SUMMARY: Rapamycin, an immunosuppressant and antiproliferative agent, reduces intimal hyperplasia after arterial injury in animal models and in a preliminary study in humans. Rapamycin treatment reportedly increases expression of p27, a cyclin-dependent kinase inhibitor. This mechanism was tested using a p27-deficient (p27 -/-) murine model. Aortic smooth muscle cells from wild-type (WT) and p27 -/- mice were isolated and cultured. Cell proliferation, assessed by cell count and (3)H-thymidine incorporation, was inhibited significantly by rapamycin in WT and p27 -/- cells at concentrations of 1 ng/ml, 10 ng/ml, and 100 ng/ml (p < 0.05, versus control). The in vivo effect on intimal hyperplasia was studied in p27 -/- and WT mice after femoral artery transluminal injury. Rapamycin treatment was started 2 days before injury and maintained for 2 weeks (1 mg/kg per 48 hours, ip). No significant differences in intima-to-media ratio were found between WT (1.1 +/- 0.1) and p27 -/- mice (1.0 +/- 0.1) 4 weeks after injury. Rapamycin significantly (p < 0.05) reduced intima-to-media ratios in both WT (0.7 +/- 0.1) and p27 -/- mice (0.5 +/- 0.1), compared with untreated mice. p27 deficiency did not alter the arterial wall proliferative response to injury. The inhibitory effect of rapamycin on intimal hyperplasia occurred via a p27-independent mechanism. The in vitro data showed that this effect was mediated through decreased proliferation and enhanced apoptosis.


Subject(s)
Aorta/pathology , Cell Cycle Proteins , Immunosuppressive Agents/pharmacology , Microtubule-Associated Proteins/deficiency , Sirolimus/pharmacology , Tumor Suppressor Proteins , Tunica Intima/pathology , Animals , Aorta/drug effects , Aorta/injuries , Apoptosis/drug effects , Cell Division/drug effects , Cyclin-Dependent Kinase Inhibitor p27 , Female , Gene Deletion , Hyperplasia , Male , Mice , Mice, Inbred C57BL , Mice, Knockout/genetics , Microtubule-Associated Proteins/genetics , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Tunica Intima/drug effects , Tunica Intima/injuries , Wounds, Nonpenetrating/metabolism , Wounds, Nonpenetrating/pathology
7.
Atherosclerosis ; 155(2): 371-80, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11254907

ABSTRACT

Macrophages play a critical role in the development and progression of atherosclerosis. This study was designed to examine the effect of the glucocorticoid, dexamethasone, (Dex), on macrophage accumulation after acute arterial injury. Twenty New Zealand white rabbits were fed a 2% cholesterol, 6% peanut oil, rabbit chow diet for one month prior to bilateral balloon dilatation of the femoral arteries. Ten rabbits received Dex (1 mg/kg, im.) the day before and then daily for 7 days after arterial injury; control rabbits received vehicle only. Seven days after injury, Dex treatment resulted in a 96% and 77% reduction (P < 0.002) in the mean number of macrophages accumulating in the intima and media, respectively. This effect was apparently not due to a reduction in the number of circulating monocytes or to the ability of monocytes from Dex treated animals to adhere to endothelium or migrate in response to a chemotactic signal, determined in vitro under static conditions. It was associated with a 61% reduction in monocyte chemoattractant protein-1 (MCP-1) antigen (P < 0.004) in the injured arterial wall (media+intima). Glucocorticoids may be useful in attenuating the inflammatory response and subsequent foam-cell accumulation after arterial injury.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Arteriosclerosis/surgery , Catheterization/adverse effects , Chemotaxis/drug effects , Cholesterol, Dietary/toxicity , Dexamethasone/therapeutic use , Diet, Atherogenic , Femoral Artery/injuries , Graft Occlusion, Vascular/prevention & control , Macrophages/drug effects , Animals , Anti-Inflammatory Agents/pharmacology , Arteriosclerosis/chemically induced , Arteriosclerosis/pathology , Cell Adhesion/drug effects , Chemokine CCL2/metabolism , Dexamethasone/pharmacology , Drug Evaluation, Preclinical , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Femoral Artery/metabolism , Femoral Artery/pathology , Hyperplasia , Macrophages/pathology , Male , Monocytes/drug effects , Monocytes/pathology , Rabbits , Tunica Intima/drug effects , Tunica Intima/pathology , Wounds and Injuries/drug therapy
8.
Proc Natl Acad Sci U S A ; 98(3): 1288-93, 2001 Jan 30.
Article in English | MEDLINE | ID: mdl-11158632

ABSTRACT

Interaction between signaling pathways regulates many cellular functions, including proliferation. The Galpha(s)/cAMP pathway is known to inhibit signal flow from receptor tyrosine kinases to mitogen-activated protein kinase (MAPK)-1,2 and, thus, inhibit proliferation. Elevation of cAMP or adenovirus-directed expression of mutant (Q227L)-Galpha(s) (alpha(s)*) inhibits the proliferation of rat vascular smooth muscle cells (VSMCs) in culture. Platelet-derived growth factor (PDGF) stimulated MAPK activation and DNA synthesis was also blocked by expression of alpha(s)*. However, it is not known whether such mechanisms are operative in vivo. Proliferation of vascular smooth muscle cells in vivo was induced by balloon injury of carotid arteries in the rat. Recombinant adenovirus encoding beta-galactosidase (beta-gal) or alpha(s)* was applied to arterial segments injured by the balloon catheters. The alpha(s)*-treated vessels showed decreased phospho-MAPK staining in the intima as compared with beta-gal-treated vessels. Application of alpha(s)*, but not beta-gal containing adenovirus, inhibited formation of neointima by 50%. No change was observed in total vessel diameter or in the media or adventitia. These results suggest that the interaction between the Galpha(s) and MAPK pathways can regulate proliferation in vivo and that targeted expression of activated Galpha(s) may have therapeutic potential for the treatment of vascular pathophysiologies that arise from intimal hyperplasia.


Subject(s)
Angioplasty, Balloon/adverse effects , Aorta/metabolism , Heterotrimeric GTP-Binding Proteins/metabolism , Tunica Intima/metabolism , Tunica Intima/pathology , Adenoviridae , Amino Acid Substitution , Animals , Aorta/cytology , Aorta/pathology , Cells, Cultured , Cyclic AMP/metabolism , DNA Replication , Genetic Vectors , Heterotrimeric GTP-Binding Proteins/chemistry , Heterotrimeric GTP-Binding Proteins/genetics , Hyperplasia , Male , Mitogen-Activated Protein Kinases/metabolism , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Platelet-Derived Growth Factor/pharmacology , Rats , Rats, Sprague-Dawley , Recombinant Proteins/analysis , Recombinant Proteins/metabolism , Transfection , beta-Galactosidase/analysis , beta-Galactosidase/genetics
9.
Blood ; 97(1): 39-45, 2001 Jan 01.
Article in English | MEDLINE | ID: mdl-11133740

ABSTRACT

The CC chemokine receptor 8 (CCR8) is expressed on monocytes and type 2 T lymphocytes. CCR8 is the sole receptor for the human CC chemokine I-309, as well as for viral monocyte inflammatory protein-I (vMIP-I), a human chemokine homologue induced in human cells by the Kaposi sarcoma-related human herpesvirus-8. Recently it was found that I-309 messenger RNA and protein are expressed by human umbilical vein endothelial cells (HUVECs) and that the secretion of endothelial I-309 is stimulated by apolipoprotein(a). I-309, vMIP-I, and the conditioned medium from apolipoprotein(a)-stimulated HUVECs induce endothelial chemotaxis. A polyclonal anti-CCR8 antibody and a newly developed murine monoclonal antibody against CCR8 inhibited this activity. The G-protein inhibitor pertussis toxin also inhibited endothelial chemotaxis, providing further evidence for a chemokine receptor-mediated effect. Endothelial cells contain CCR8 mRNA as shown by RNA blot analysis as well by direct sequence analysis. Immunohistochemical studies identified CCR8 and I-309 on the endothelium of human atherosclerotic plaques and in endothelial-derived spindle cells of Kaposi sarcoma. These results indicate that CCR8 is an endothelial receptor that may modulate endothelial function.


Subject(s)
Chemotaxis/drug effects , Endothelium, Vascular/cytology , Receptors, Chemokine/physiology , Viral Proteins , Antibodies/pharmacology , Chemokine CCL1 , Chemokine CXCL12 , Chemokines, CC/metabolism , Chemokines, CC/pharmacology , Chemokines, CXC/pharmacology , Chemotactic Factors/pharmacology , Coronary Artery Disease/metabolism , Culture Media, Conditioned/pharmacology , Herpesvirus 8, Human/chemistry , Humans , Immunohistochemistry , Lipoprotein(a)/pharmacology , Macrophage Inflammatory Proteins/pharmacology , Pertussis Toxin , RNA, Messenger/biosynthesis , Receptors, CCR8 , Receptors, Chemokine/genetics , Receptors, Chemokine/immunology , Sarcoma, Kaposi/chemistry , Umbilical Veins/cytology , Virulence Factors, Bordetella/pharmacology
10.
J Am Coll Cardiol ; 36(7): 2303-10, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11127477

ABSTRACT

OBJECTIVES: We investigated the in vivo effects of tissue factor (TF) inhibition with recombinant tissue factor pathway inhibitor (rTFPI) on acute thrombus formation and intimal hyperplasia and the in vitro effects on smooth muscle cell migration and proliferation. BACKGROUND: Inhibition of TF with TFPI has been shown to reduce intimal hyperplasia in experimental models. However, its effects after coronary angioplasty and the cellular mechanisms involved have not been investigated. METHODS: Twenty-three swine underwent multivessel coronary angioplasty. Fifteen (n = 25 arteries) were euthanized at 72 h to assess thrombus formation and eight (n = 24 arteries) at 28 days to assess intimal hyperplasia. Animals in the 72-h time point received: 1) human rTFPI (0.5 mg bolus plus 25 microg/kg/min continuous infusion for 3 days) plus heparin (150 IU/kg intravenous bolus) plus acetyl salicylic acid (ASA) (325 mg/day); 2) rTFPI regimen plus ASA and 3) heparin (150 IU/kg intravenous bolus) plus ASA. RESULTS: On histology the control group had evidence of mural thrombus (area 0.8+/-0.4 mm2). Treatment with TFPI plus heparin abolished thrombus formation (mean area: 0.0+/-0.0 mm2, p < 0.05) but was associated with prolonged activated partial thromboplastin time and extravascular hemorrhage. Recombinant TFPI alone inhibited thrombosis without bleeding complications (mean area: 0.03+/-0.02 mm2, p < 0.05 vs. control). Animals in the 28-day time point received continuous intravenous infusion of rTFPI or control solution for 14 days. Tissue factor pathway inhibitor reduced neointimal formation with mean intimal area of 1.2+/-0.3 mm2 versus 3.2+/-0.4 mm2 in the control group; p < 0.01. Recombinant TFPI had no effect on human aortic smooth muscle cell growth but inhibited platelet-derived growth factor BB-induced migration. CONCLUSIONS: Inhibition of TF with rTFPI can prevent acute thrombosis and intimal hyperplasia after injury. Tissue factor plasma inhibitor may prove useful as an adjunct to intracoronary interventions.


Subject(s)
Angioplasty, Balloon, Coronary , Coronary Thrombosis/prevention & control , Fibrinolytic Agents/pharmacology , Lipoproteins/pharmacology , Peptide Fragments/pharmacology , Thromboplastin/antagonists & inhibitors , Tunica Intima/pathology , Angioplasty, Balloon, Coronary/adverse effects , Animals , Cells, Cultured , Coronary Thrombosis/etiology , Coronary Vessels/pathology , Drug Synergism , Heparin/pharmacology , Hyperplasia , Models, Animal , Swine
11.
Lab Invest ; 80(11): 1603-15, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11092521

ABSTRACT

Osteopontin (OPN) is an extracellular matrix protein that has been implicated in vascular smooth muscle cell (VSMC) adhesion. We have previously described the generation of OPN-deficient VSMC that displayed altered adhesion to collagen. We have examined further the causes and consequences of this altered adhesion. OPN-deficiency was associated with a significant reduction in surface expression of alpha1 and beta1 integrins (mean fluorescence intensity alpha1: OPN-deficient 0.135+/-0.04 vs. control 0.313+/-0.05, p < 0.0001; beta1: OPN-deficient 0.398+/-0.09 vs. control 0.570+/-0.05, p < 0.004). Treatment of normal VSMC with antibody to alpha1 recapitulated the adhesion defect. OPN-deficient cells without collagen exposure had an apoptotic fraction of 1.9%, which increased to 95.7% after 24 hours exposure to collagen. Exogenous OPN added to cultures within 15 minutes of plating restored normal cell adhesion, but did not prevent cells from undergoing apoptosis. Normal VSMC had no detectable apoptosis after 24 hours incubation in suspension, whereas OPN-deficient cells had an apoptotic fraction of 37.5% when incubated in suspension under the same conditions. The data suggest that OPN-deficient VSMC have two distinct abnormalities: an alpha1beta1-mediated inability to adhere normally to collagen and an increased propensity for apoptosis.


Subject(s)
Apoptosis , Collagen/metabolism , Muscle, Smooth, Vascular/metabolism , Sialoglycoproteins/metabolism , Animals , Antibodies, Monoclonal/immunology , Cell Adhesion , Collagen/genetics , Integrin alpha1beta1 , Integrins/immunology , Integrins/metabolism , Male , Muscle, Smooth, Vascular/cytology , Osteopontin , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley
12.
Circulation ; 102(7): 786-92, 2000 Aug 15.
Article in English | MEDLINE | ID: mdl-10942748

ABSTRACT

BACKGROUND: Lipoprotein(a) [Lp(a)] is a risk factor for atherosclerosis; however, the mechanisms are unclear. We previously reported that Lp(a) stimulated human vascular endothelial cells to produce monocyte chemotactic activity. The apolipoprotein(a) [apo(a)] portion of Lp(a) was the active moiety. METHODS AND RESULTS: We now describe the identification of the chemotactic activity as being due to the CC chemokine I-309. The carboxy-terminal domain of apo(a) containing 6 type-4 kringles (types 5 to 10), kringle V, and the protease domain was demonstrated to contain the I-309-inducing portion. Polyclonal and monoclonal anti-I-309 antibodies as well as an antibody against a portion of the extracellular domain of CCR8, the I-309 receptor, inhibited the increase in monocyte chemotactic activity induced by apo(a). I-309 antisense oligonucleotides also inhibited the induction of endothelial monocyte chemotactic activity by apo(a). I-309 mRNA was identified in human umbilical vein endothelial cells. Apo(a) induced an increase in I-309 protein in the endothelial cytoplasm and in the conditioned medium. Immunohistochemical studies have identified I-309 in endothelium, macrophages, and extracellular areas of human atherosclerotic plaques and have found that I-309 colocalized with apo(a). CONCLUSIONS: These data establish that I-309 is responsible for the monocyte chemotactic activity induced in human umbilical vein endothelial cells by Lp(a). The identification of the endothelial cell as a source for I-309 suggests that this chemokine may participate in vessel wall biology. Our data also suggest that I-309 may play a role in mediating the effects of Lp(a) in atherosclerosis.


Subject(s)
Apolipoproteins A/physiology , Chemokines, CC/metabolism , Chemotactic Factors/metabolism , Endothelium, Vascular/metabolism , Monocytes/metabolism , Antibodies/pharmacology , Apolipoproteins A/pharmacology , Blotting, Western , Cells, Cultured , Chemokine CCL1 , Chemokines, CC/genetics , Chemokines, CC/immunology , Chemotactic Factors/antagonists & inhibitors , Chemotactic Factors/genetics , Chemotactic Factors/immunology , Culture Media/metabolism , Cytoplasm/metabolism , Endothelium, Vascular/cytology , Humans , Oligonucleotides, Antisense/pharmacology , RNA, Messenger/metabolism , Receptors, CCR8 , Receptors, Chemokine/immunology , Recombinant Proteins/metabolism
13.
Circ Res ; 87(2): 126-32, 2000 Jul 21.
Article in English | MEDLINE | ID: mdl-10903996

ABSTRACT

Tissue factor (TF), the initiator of coagulation, is thought to function predominantly at the cell surface. Recent data have suggested that active TF is present extracellularly in atherosclerotic plaques, the arterial wall, and the blood. This study was conducted to determine whether smooth muscle cells (SMCs), a major source of arterial TF, could generate extracellular TF. Active TF accumulated in the medium of cultured human SMCs, representing approximately 10% of that measured in the underlying cells at 24 hours. Platelet-derived growth factor, phorbol ester, and tumor necrosis factor-alpha caused approximately 3-fold increases in TF activity in the medium. Release of TF into the medium was dependent on the presence of the TF transmembrane domain but not the cytoplasmic domain. Antibodies to TF precipitated most of the activity from the culture medium, whereas antibodies to the beta(1)-integrin subunit precipitated approximately 33% of the activity. Treatment with detergent or phosphatidylserine:phosphatidylcholine did not increase activity, suggesting that all TF released by SMCs was in the appropriate lipid milieu and not encrypted. Western blotting showed that the medium contained full-length TF protein. Fluorescent cytometry showed that extracellular TF was present largely in particles < or =200 nm, which had a density of 1.10 g/mL. We hypothesize that active extracellular TF found in the injured arterial wall and atherosclerotic plaques derives, in part, from SMC microparticles.


Subject(s)
Muscle, Smooth, Vascular/metabolism , Thromboplastin/metabolism , Aorta , Cells, Cultured , Coronary Vessels , Humans , Indomethacin/pharmacology , Interleukin-1/pharmacology , Kinetics , Melanoma , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Platelet-Derived Growth Factor/pharmacology , Recombinant Proteins/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Thromboplastin/genetics , Transfection , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/pharmacology
14.
Arterioscler Thromb Vasc Biol ; 20(5): 1293-300, 2000 May.
Article in English | MEDLINE | ID: mdl-10807745

ABSTRACT

The expression of monocyte-specific adhesion molecules and chemokines by cell types within the vessel wall plays an important role in foam cell accumulation during atherosclerotic plaque development. We previously identified IG9, a novel monocyte adhesion protein that is expressed on endothelial cells (ECs) overlying human and rabbit advanced atherosclerotic plaques. The present study was designed to determine the temporal and spatial expression of IG9 and the chemokine, monocyte chemoattractant protein-1 (MCP-1), after balloon injury with (double injury) or without (single injury) prior air desiccation EC injury in the femoral arteries of rabbits fed a high-cholesterol diet. By immunohistochemical analyses, intense reactivity with monoclonal antibodies to IG9 and MCP-1 was detected 24 hours after single injury in medial smooth muscle cells (SMCs) and in SMCs of adventitial microvessels. However, monocyte infiltration of the tunica media was minimal or not detected in these sections. IG9 and MCP-1 antibody reactivity in vessel sections 28 days after single injury and 24 hours, 7 days, and 28 days after double injury was localized to medial and neointimal SMCs, foam cells, and luminal ECs overlying the plaques. Uninjured rabbit (cholesterol or normal diet) vessel sections exhibited minimal IG9 and MCP-1 immunostaining. In vitro studies using human aortic SMCs demonstrated IG9 protein induction after 24 hours of treatment with platelet-derived growth factor-BB and interferon-gamma or epidermal growth factor. IG9 expression was further increased by pretreatment of SMCs with the proatherogenic lipid, minimally oxidized low density lipoprotein. After balloon injury (24 hours), IG9 is induced in vascular SMCs before the detectable accumulation of monocytes within the vessel wall. Thus, the expression of IG9 by SMCs as well as by ECs may be an important factor in the accumulation of foam cells in atherosclerotic plaque development after arterial injury.


Subject(s)
Carotid Artery Injuries/metabolism , Cell Adhesion Molecules/genetics , Cholesterol, Dietary/administration & dosage , Endothelium, Vascular/metabolism , Gene Expression Regulation , Muscle, Smooth, Vascular/metabolism , Animals , Aorta , Carotid Artery Injuries/etiology , Catheterization , Cell Adhesion , Cells, Cultured , Chemokine CCL2/genetics , Cytokines/pharmacology , Gene Expression Regulation/drug effects , Growth Substances/pharmacology , Humans , Lipoproteins, LDL/pharmacology , Monocytes , Rabbits
15.
Thromb Haemost ; 83(4): 622-8, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10780328

ABSTRACT

Tissue factor (TF), the initiator of coagulation, has been implicated as a critical mediator of arterial thrombosis. Previous studies have demonstrated that TF is rapidly induced in the normal rodent arterial wall by balloon injury, but is not associated with fibrin deposition. A second injury, however, performed 10-14 days after the first, is followed by small platelet-fibrin microthrombi. This study was undertaken to better localize active TF in balloon-injured rat arteries and to explore possible mechanisms underlying the apparent discrepancy between injury-induced TF expression and the lack of large platelet-fibrin thrombi. By immunohistochemistry, TF antigen was first detected in the media 24 h after injury to rat aortas, and subsequently accumulated in the neointima. Using an ex vivo flow chamber, no TF activity (Factor Xa generation) was found on the luminal surface of normal or injured aortas. Wiping the luminal surface with a cotton swab exposed TF activity in all vessels; levels were increased approximately 3-fold in arteries containing a neointima. The exposed TF activity was rapidly washed into the perfusate, rendering the luminal surface inactive. The loss of luminal TF into the circulation may attenuate thrombosis at sites of arterial injury.


Subject(s)
Angioplasty, Balloon, Coronary/adverse effects , Thromboplastin/metabolism , Tunica Intima/metabolism , Animals , Aorta, Abdominal/injuries , Aorta, Thoracic/injuries , Arterial Occlusive Diseases/etiology , Fibrin/metabolism , Male , Microscopy, Electron, Scanning , Rats , Rats, Sprague-Dawley , Recurrence , Thrombosis/etiology , Tunica Intima/injuries
16.
Arterioscler Thromb Vasc Biol ; 20(2): 335-42, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10669628

ABSTRACT

Techniques of arterial injury commonly used in animals to mimic endovascular procedures are not suitable for small mouse arteries. This has limited examination of the response to arterial injury in genetically modified mice. We therefore sought to develop a model of transluminal injury to the mouse femoral artery that would be reproducible and result in substantial levels of intimal hyperplasia. Mice of the C57BL/6 strain underwent bilateral femoral artery denudation by passage of an angioplasty guidewire. Intimal hyperplasia was observed in 10% of injured arteries at 1 week, in 88% at 2 weeks, and in 90% at 4 weeks. The mean intimal-to-medial area ratio reached 1.1+/-0.1 at 4 weeks. No intimal proliferation was found in control sham-operated arteries. One hour after injury, the denuded surface was covered with platelets and leukocytes, predominantly neutrophils. This was associated with the accumulation of P-selectin, intercellular adhesion molecule-1, and vascular cell adhesion molecule-1. Expression of these adhesion molecules was not seen in the underlying medial smooth muscle cells. At 24 hours, few neutrophils remained on the denuded surface. At 1 week, macrophages and platelets were present in the vessel wall, partially covered by regenerated endothelium. Transluminal wire injury to the mouse femoral artery induces abundant intimal hyperplasia formation by 2 and 4 weeks and elicits the rapid accumulation of leukocytes and adhesion molecules on the denuded luminal surface. This model will be a valuable tool to study arterial injury in genetically modified mouse models.


Subject(s)
Cell Adhesion Molecules/metabolism , Femoral Artery/injuries , Neutrophils/physiology , Tunica Intima/injuries , Wounds and Injuries/physiopathology , Animals , Mice , Mice, Inbred C57BL , Vasculitis/etiology , Vasculitis/pathology , Wounds and Injuries/complications , Wounds and Injuries/metabolism , Wounds and Injuries/pathology
17.
J Biol Chem ; 275(8): 5466-71, 2000 Feb 25.
Article in English | MEDLINE | ID: mdl-10681524

ABSTRACT

CC chemokine receptors are important modulators of inflammation. Although CC chemokine receptors have been found predominantly on leukocytes, recent studies have suggested that vascular smooth muscle cells respond to CC chemokines. We now report that human smooth muscle cells express CCR5, a co-receptor for human immunodeficiency virus. CCR5 mRNA was detectable by RNA blot hybridization in human aortic and coronary artery smooth muscle cells. The cDNA generated by reverse transcription-polymerase chain reaction from aortic smooth muscle cells had 100% identity throughout the entire coding region with the CCR5 cloned from THP-1 cells. By immunohistochemistry, CCR5 and the CCR5 ligand, macrophage inflammatory protein-1beta (MIP-1beta), were detected in smooth muscle cells and macrophages of the atherosclerotic plaque. In smooth muscle cell culture, MIP-1beta induced a significant increase in intracellular calcium concentrations, which was blocked by an antibody to CCR5. In addition, MIP-1beta caused a calcium-dependent increase in tissue factor activity. Tissue factor is the initiator of coagulation and is thought to play a key role in arterial thrombosis. These data suggest that human arterial smooth muscle cells express functional CCR5 receptors and MIP-1beta is an agonist for these cells.


Subject(s)
Muscle, Smooth, Vascular/metabolism , Receptors, CCR5/metabolism , Aorta/metabolism , Arteriosclerosis/metabolism , Calcium/metabolism , Chelating Agents/pharmacology , Chemokine CCL4 , Coronary Vessels/metabolism , Dose-Response Relationship, Drug , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Endothelium, Vascular/metabolism , Humans , Immunohistochemistry , Inflammation/metabolism , Macrophage Inflammatory Proteins/metabolism , RNA, Messenger/metabolism , Receptors, CCR5/genetics , Reverse Transcriptase Polymerase Chain Reaction , Thromboplastin/metabolism , Thrombosis/metabolism , Time Factors , Umbilical Cord/metabolism
18.
Thromb Haemost ; 82(2): 801-5, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10605785

ABSTRACT

The last few years have provided increasing evidence to support a major role for TF in the initiation and propagation of thrombosis after acute arterial injury. Although thrombotic occlusion occurs in a small minority of patients undergoing acute coronary interventions or bypass surgery, mural thrombi are likely to be present in almost all cases. These thrombi may stimulate SMC and promote the development of intimal hyperplasia and luminal narrowing. The use of inhibitors of TF and factor VIIa, therefore, may not only be valuable for inhibiting thrombus formation associated with acute arterial interventions, but may also have benefit in attenuating intimal hyperplasia. Although this paper focuses on the role of TF in establishing a procoagulant state after arterial injury, the fibrinolytic system undoubtedly plays a role in balancing the effects of increased TF production in the arterial wall. This is underscored by the success of activators of fibrinolysis (tissue plasminogen activator, streptokinase, urokinase) in revascularization in the setting of acute myocardial infarction and is reviewed elsewhere. Likewise, local regulation of TFPI in the atherosclerotic plaque and injured vessel wall may be important in attenuating the effects of increased TF synthesis and accumulation. It has been assumed that the primary source of active TF after arterial injury is either SMC or invading macrophages and that active TF is anchored to the surface of these cells. Recent data have suggested that the majority of cell-associated TF is either encrypted on the cell surface or present in an intracellular pool. Arterial injury may, therefore, involve the de-encryption of surface TF or the release of intracellular TF. In addition, active vascular TF may be present in microparticles that are not anchored to the arterial wall and may be washed into the circulation. The procoagulant state may be further accentuated by the accumulation of bloodborne TF at sites of arterial injury and in developing thrombi. This TF is likely to arise from circulating leukocytes, including neutrophils and monocytes. These studies suggest that the cellular processing of TF may be an important target for inhibiting thrombotic complications associated with arterial injury and acute coronary events.


Subject(s)
Arteries/physiology , Arteriosclerosis/physiopathology , Thromboplastin/physiology , Thrombosis/etiology , Animals , Arteries/injuries , Arteries/pathology , Endothelium, Vascular/pathology , Endothelium, Vascular/physiology , Fibrinolysis , Humans , Hyperplasia , Muscle, Smooth, Vascular/physiology , RNA, Messenger , Thromboplastin/biosynthesis
19.
Lab Invest ; 79(11): 1369-75, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10576207

ABSTRACT

Trapidil (triazolopyrimidine) is an antiplatelet agent that acts in part as a phosphodiesterase inhibitor and as a competitive inhibitor of the platelet-derived growth factor (PDGF) receptor. Trapidil has been shown to attenuate intimal hyperplasia in rat and hamster models of balloon arterial injury and to inhibit restenosis after percutaneous transluminal coronary angioplasty in several small clinical trials. Monocyte chemoattractant protein-1 (MCP-1) is a PDGF-inducible monocyte chemoattractant that is thought to play a particularly important role in recruiting monocyte/macrophages to sites of atherosclerosis and vessel injury. We hypothesized that, because of its ability to antagonize PDGF-mediated events, trapidil would inhibit the synthesis of MCP-1 and decrease macrophage accumulation in the injured arterial wall. Hypercholesterolemic rabbits were treated with trapidil (60 mg/kg/day subcutaneously) the day before and then daily for 6 days after balloon injury of the femoral artery; control rabbits received vehicle only. Trapidil resulted in a 75% reduction in MCP-1 expression and macrophage accumulation in the arterial wall 7 days after injury. This study suggests that trapidil has potent anti-inflammatory properties and that its activity in attenuating intimal hyperplasia may be in part attributed to its effects on macrophage accumulation.


Subject(s)
Catheterization/adverse effects , Chemokine CCL2/antagonists & inhibitors , Femoral Artery/injuries , Macrophages/pathology , Platelet Aggregation Inhibitors/pharmacology , Trapidil/pharmacology , Wounds and Injuries/metabolism , Wounds and Injuries/pathology , Animals , Cell Count/drug effects , Chemokine CCL2/metabolism , Femoral Artery/pathology , Macrophages/drug effects , Macrophages/physiology , Rabbits
20.
Gene Expr ; 8(1): 59-66, 1999.
Article in English | MEDLINE | ID: mdl-10543731

ABSTRACT

SM-20 is a novel, evolutionarily conserved "early response" gene originally cloned from a rat aortic smooth muscle cell (SMC) cDNA library. SM-20 encodes a cytoplasmic protein, which is induced by platelet-derived growth factor and angiotensin II in cultured SMC and is upregulated in intimal SMC of atherosclerotic plaques and injured arteries. We have now examined SM-20 expression during differentiation of cultured skeletal myoblasts and during skeletal myogenesis in vivo. Low levels of SM-20 mRNA and protein were expressed in proliferating mouse C2C12 myoblasts. Differentiation by serum withdrawal was associated with a marked induction of SM-20 mRNA and the expression of high levels of SM-20 antigen in myotubes. The induction was partially inhibited by blocking differentiation with bFGF or TGFbeta. Similar results were obtained with the nonfusing mouse C25 myoblast line, suggesting that SM-20 upregulation is a consequence of biochemical differentiation and is fusion independent. During mouse embryogenesis, SM-20 was first observed at 8.5E in the dermomyotomal cells of the rostral somites. SM-20 expression progressed in a rostral to caudal pattern, with highest levels seen in the muscle primordia and mature muscles. SM-20 thus represents a novel intracellular protein that is regulated during skeletal muscle differentiation and development.


Subject(s)
DNA-Binding Proteins , Gene Expression Regulation, Developmental , Immediate-Early Proteins/metabolism , Muscle, Skeletal/embryology , Muscle, Skeletal/metabolism , Animals , Cells, Cultured , Hypoxia-Inducible Factor-Proline Dioxygenases , Immediate-Early Proteins/genetics , Immunohistochemistry , Mice , Mice, Inbred Strains , Muscle, Skeletal/cytology , Procollagen-Proline Dioxygenase , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...