Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Catheter. cardiovasc. interv ; 97(suppl. 1): S55-S56, Apr., 2021.
Article in English | CONASS, Sec. Est. Saúde SP, SESSP-IDPCPROD, Sec. Est. Saúde SP | ID: biblio-1349087

ABSTRACT

BACKGROUND: While dual antiplatelet therapy (DAPT) constitutes the cornerstone of post-PCI pharmacotherapy, duration of DAPT in high bleeding risk (HBR) patients has not been fully defined especially with regard to sex. The results from the Onyx ONE Clear trial demonstrated favorable safety and efficacy after PCI with 1-month dual antiplatelet therapy (DAPT) in HBR patients treated with Resolute Onyx drug-eluting stents (DES). We sought to evaluate impact of sex on clinical outcomes in this trial. METHODS: In this prespecified subgroup analysis from Onyx ONE Clear, patients were divided into 2 groups according to sex. Primary endpoint was cardiac death or myocardial infarction (MI) from 1 month to 1 year. RESULTS: A total of 487 female patients (32%) and 1019 males (68%) were free from major ischemic events 1-month after PCI and were transitioned to single antiplatelet therapy.Women were older (p<0.001), had more HBR criteria (p=0.02), and higher rates of moderate/severe calcific lesions (p=0.03) compared to men. Men had higher rates of previous MI (p=0.003), atrial fibrillation (p=0.001), and multivessel coronary artery disease (p<0.001). Clinical outcomes between 1 and 12 months are shown in (Figure) and were similar for males and females except for target vessel revascularization which was greater for males (p=0.04). CONCLUSIONS: In HBR patients treated with Resolute Onyx DES and an abbreviated DAPT course of one month, rates of the primary endpoint of cardiac death or MI between 1 and 12 months were low and did not show any sex-based differences. These data support the use of an abbreviated DAPT regimen in men and women with HBR after PCI with Resolute Onyx DES.


Subject(s)
Sex , Coronary Artery Disease , Drug-Eluting Stents
2.
N. Engl. j. med ; 382(13): 1208-1218, Mar., 2020. tab., graf.
Article in English | Sec. Est. Saúde SP, SESSP-IDPCPROD, Sec. Est. Saúde SP | ID: biblio-1053448

ABSTRACT

BACKGROUND Polymer-free drug-coated stents provide superior clinical outcomes to bare-metal stents in patients at high bleeding risk who undergo percutaneous coronary intervention (PCI) and are treated with 1 month of dual antiplatelet therapy. Data on the use of polymer-based drug-eluting stents, as compared with polymer-free drug-coated stents, in such patients are limited. METHODS In an international, randomized, single-blind trial, we compared polymer-based zotarolimus-eluting stents with polymer-free umirolimus­coated stents in patients at high bleeding risk. After PCI, patients were treated with 1 month of dual antiplatelet therapy, followed by single antiplatelet therapy. The primary outcome was a safety composite of death from cardiac causes, myocardial infarction, or stent thrombosis at 1 year. The principal secondary outcome was target-lesion failure, an effectiveness composite of death from cardiac causes, target-vessel myocardial infarction, or clinically indicated target-lesion revascularization. Both outcomes were powered for noninferiority. RESULTS A total of 1996 patients at high bleeding risk were randomly assigned in a 1:1 ratio to receive zotarolimus-eluting stents (1003 patients) or polymer-free drugcoated stents (993 patients). At 1 year, the primary outcome was observed in 169 of 988 patients (17.1%) in the zotarolimus-eluting stent group and in 164 of 969 (16.9%) in the polymer-free drug-coated stent group (risk difference, 0.2 percentage points; upper boundary of the one-sided 97.5% confidence interval [CI], 3.5; noninferiority margin, 4.1; P=0.01 for noninferiority). The principal secondary outcome was observed in 174 patients (17.6%) in the zotarolimus-eluting stent group and in 169 (17.4%) in the polymer-free drug-coated stent group (risk difference, 0.2 percentage points; upper boundary of the one-sided 97.5% CI, 3.5; noninferiority margin, 4.4; P=0.007 for noninferiority). CONCLUSIONS Among patients at high bleeding risk who received 1 month of dual antiplatelet therapy after PCI, use of polymer-based zotarolimus-eluting stents was noninferior to use of polymer-free drug-coated stents with regard to safety and effectiveness composite outcomes. (Funded by Medtronic; ONYX ONE ClinicalTrials.gov number, NCT03344653.). (AU)


Subject(s)
Coronary Artery Disease/drug therapy , Combined Modality Therapy , Sirolimus , Drug-Eluting Stents , Polymers , Double-Blind Method
3.
Circ Res ; 89(10): 859-65, 2001 Nov 09.
Article in English | MEDLINE | ID: mdl-11701612

ABSTRACT

The leukocyte integrin Mac-1 (alphaMbeta2, CD11b/CD18) regulates important cell functions in inflammation, including adhesion, phagocytosis, and oxidative burst. Deficiency of Mac-1 reduces vessel wall inflammation and neointimal thickening after murine carotid artery injury. Although Mac-1 has been implicated in modulating AP-1 and NF-kappaB activity, the signal transduction pathways involved are undefined. cDNA array analysis of Mac-1-clustered compared with -nonclustered monocytic THP-1 cells showed increased expression of the signal transducer TRAF6 (TNF receptor-associated factor 6), leading us to consider the possibility that Mac-1 used a Toll/IL-1 receptor family-like signaling pathway. Mac-1-dependent activation of NF-kappaB was potentiated by wild-type, and attenuated by dominant negative, TRAF6- and TGF-beta-activated kinase (TAK1) constructs. IRAK1 (IL-1 receptor associated kinase), a kinase immediately upstream of TRAF6, coimmunoprecipitated with Mac-1. Taken together, these observations indicate that Mac-1 recruits a Toll/IL-1 receptor family-like cascade to modulate NF-kappaB activity. This represents a new pathway for integrin-dependent modulation of gene expression.


Subject(s)
Macrophage-1 Antigen/metabolism , Membrane Glycoproteins , Membrane Proteins/metabolism , NF-kappa B/metabolism , Receptors, Cell Surface , Receptors, Immunologic/metabolism , Receptors, Interleukin-1/metabolism , Adaptor Proteins, Signal Transducing , Antigens, Differentiation/genetics , Antigens, Differentiation/metabolism , Cell Adhesion/immunology , Cell Line , Gene Expression Regulation/immunology , Genes, Reporter , Humans , Interleukin-1 Receptor-Associated Kinases , MAP Kinase Kinase Kinases/metabolism , Macrophage-1 Antigen/genetics , Monocytes/cytology , Monocytes/immunology , Monocytes/metabolism , Myeloid Differentiation Factor 88 , NF-kappa B/genetics , Oligonucleotide Array Sequence Analysis , Protein Kinases/metabolism , Proteins/metabolism , Receptor Aggregation/immunology , Signal Transduction/immunology , TNF Receptor-Associated Factor 6 , Toll-Like Receptors , Transfection
4.
Am J Cardiol ; 88(1): 5-9, 2001 Jul 01.
Article in English | MEDLINE | ID: mdl-11423050

ABSTRACT

The objectives of this analysis were to develop and validate simplified risk score models for predicting the risk of major in-hospital complications after percutaneous coronary intervention (PCI) in the era of widespread stenting and use of glycoprotein IIb/IIIa antagonists. We then sought to compare the performance of these simplified models with those of full logistic regression and neural network models. From January 1, 1997 to December 31, 1999, data were collected on 4,264 consecutive interventional procedures at a single center. Risk score models were derived from multiple logistic regression models using the first 2,804 cases and then validated on the final 1,460 cases. The area under the receiver operating characteristic (ROC) curve for the risk score model that predicted death was 0.86 compared with 0.85 for the multiple logistic model and 0.83 for the neural network model (validation set). For the combined end points of death, myocardial infarction, or bypass surgery, the corresponding areas under the ROC curves were 0.74, 0.78, and 0.81, respectively. Previously identified risk factors were confirmed in this analysis. The use of stents was associated with a decreased risk of in-hospital complications. Thus, risk score models can accurately predict the risk of major in-hospital complications after PCI. Their discriminatory power is comparable to those of logistic models and neural network models. Accurate bedside risk stratification may be achieved with these simple models.


Subject(s)
Angioplasty, Balloon, Coronary/adverse effects , Coronary Disease/mortality , Hospital Mortality , Postoperative Complications/mortality , Risk Assessment/methods , Cardiopulmonary Bypass , Coronary Disease/therapy , Female , Humans , Logistic Models , Male , Middle Aged , Myocardial Infarction/mortality , Neural Networks, Computer , Postoperative Complications/epidemiology , Predictive Value of Tests , Prognosis , Prospective Studies , ROC Curve , Risk Factors
5.
Arterioscler Thromb Vasc Biol ; 21(5): 759-64, 2001 May.
Article in English | MEDLINE | ID: mdl-11348871

ABSTRACT

Motexafin lutetium is a photosensitizer that accumulates in atherosclerotic plaque and, after activation by far-red light, produces cytotoxic singlet oxygen. The combination of photosensitizer and illumination, known as photodynamic therapy (PDT), has been shown to reduce atheroma formation in animal models and is under clinical investigation. However, the effects of PDT with motexafin lutetium on isolated vascular cells are unknown. This study was designed to characterize the effects of PDT on vascular cell viability and to define the cell-death pathway for this agent. Fluorescence microscopy of RAW macrophages and human vascular smooth muscle cells revealed time-dependent uptake of motexafin lutetium. Illumination of motexafin lutetium-loaded cells with 732-nm light (2 J/cm(2)) impaired cellular viability and growth (IC(50) 5 to 20 micromol/L). Depletion of intracellular glutathione potentiated (P=0.035) and the addition of antioxidant N-acetylcysteine attenuated (P=0.002) cell death, suggesting that the intracellular redox state influences motexafin lutetium action. PDT was associated with the loss of mitochondrial membrane potential, mitochondrial release of cytochrome c, and caspase activation. PDT promoted phosphatidylserine externalization and induced apoptotic DNA fragmentation, with the number of apoptotic cells increasing from 7+/-2% to 34+/-3% of total cells. Reducing plaque cellularity by the induction of apoptosis may be one mechanism by which PDT reduces plaque burden, possibly modulates plaque vulnerability, and inhibits restenosis in vivo.


Subject(s)
Apoptosis/drug effects , Metalloporphyrins/pharmacology , Muscle, Smooth, Vascular/drug effects , Photochemotherapy , Photosensitizing Agents/pharmacology , Animals , Arteriosclerosis/drug therapy , Cell Division/drug effects , Cell Line , Cell Survival/drug effects , Cells, Cultured , Cytochrome c Group/metabolism , Humans , Macrophages/cytology , Macrophages/drug effects , Membrane Potentials , Metalloporphyrins/pharmacokinetics , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Muscle, Smooth, Vascular/cytology , Oxidation-Reduction , Photosensitizing Agents/pharmacokinetics
6.
Catheter Cardiovasc Interv ; 53(1): 56-63, 2001 May.
Article in English | MEDLINE | ID: mdl-11329219
7.
Catheter Cardiovasc Interv ; 52(4): 425-32, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11285593

ABSTRACT

Platelet inhibition is central to the efficacy of glycoprotein (GP) IIb-IIIa antagonist therapy, but is not routinely measured during percutaneous coronary intervention (PCI). Data directly comparing the antiplatelet effects of these agents are also limited. Therefore, we compared ex vivo platelet function by standard light transmission aggregometry (LTA) and two automated bedside platelet function assays in 36 patients undergoing PCI with GP IIb-IIIa inhibitors. At baseline and 10 min following clinically recommended bolus and infusion of abciximab (0.25 mg/kg, 0.125 microg/kg/min), eptifibatide (180 microg/kg, 2 microg/kg/min), or tirofiban (10 microg/kg, 0.1 microg/kg/min), we measured 20 microM ADP- and 1.9 mg/mL collagen-induced platelet aggregation using LTA. Platelet function was also assessed using the bedside Accumetrics Ultegra-Rapid Platelet Function Assay (RPFA) and the Xylum Clot Signature Analyzer (CSA). The degree of platelet inhibition, as assessed by LTA, varied significantly between the clinically recommended doses of these GP IIb-IIIa antagonists. RPFA measurements agreed closely with LTA for abciximab, but tended to overestimate the degree of platelet inhibition for small molecules. CSA demonstrated profoundly inhibited shear-induced platelet function, but lacked sensitivity to discriminate between agents. These findings may have implications for the results of trials comparing the efficacy of these agents in patients undergoing PCI.


Subject(s)
Angioplasty, Balloon , Platelet Aggregation Inhibitors/administration & dosage , Platelet Aggregation , Tyrosine/analogs & derivatives , Abciximab , Aged , Antibodies, Monoclonal/administration & dosage , Blood Platelets/drug effects , Eptifibatide , Female , Humans , Immunoglobulin Fab Fragments/administration & dosage , Light , Male , Middle Aged , Peptides/administration & dosage , Physiology/methods , Platelet Function Tests/methods , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , Point-of-Care Systems , Tirofiban , Tyrosine/administration & dosage
9.
Arterioscler Thromb Vasc Biol ; 20(12): 2553-8, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11116052

ABSTRACT

Macrophages are abundant after stent-induced arterial injury. Inhibition of macrophage recruitment blocks neointimal growth in this model. In contrast, after superficial injury from balloon endothelial denudation, macrophages are sparse. However, many anti-inflammatory therapies remain effective against neointimal growth after balloon injury. To investigate further the role of leukocytes after injury, 41 New Zealand White rabbits underwent iliac artery balloon denudation. In 18, subcutaneous pumps were placed to deliver intravenous heparin (0.3 mg/kg per hour). Arteries were harvested at 6 hours and at 3, 7, and 14 days. In 8 animals, either M1/70 (a monoclonal antibody [mAb] against adhesion molecule Mac-1) or a nonspecific IgG was given (5 mg/kg IV bolus and then 1 mg/kg SC QOD), and arteries were harvested at 6 hours and 3 days. Computer-aided morphometry was performed as was immunohistochemistry to assess smooth muscle cell (SMC) proliferation (bromodeoxyuridine-positive cells), neutrophil content (RPN357, mAb against rabbit neutrophil/thymocyte), and macrophage content (RAM-11, mAb against rabbit macrophage). Heparin inhibited neointimal growth at 7 and 14 days (64% and 32.5% reduction, respectively; P:<0.05). Neutrophils were observed in the media early after balloon injury, and heparin and M1/70 inhibited this infiltration (82% and 83% reduction, respectively; P:<0.05 each) with a coincident inhibition of medial SMC proliferation at 3 days (49% and 84% reduction, respectively; P:<0.05 each). Macrophages were absent at all time points. Neutrophil, but not macrophage, infiltration occurs early after endothelial denudation. Inhibition of this process is associated with a reduction in medial SMC proliferation. These data suggest a central role for neutrophils in restenosis and help to explain prior reports of an inhibitory effect of anti-inflammatory therapies on neointimal growth after balloon injury.


Subject(s)
Angioplasty, Balloon , Iliac Artery/pathology , Neutrophils/immunology , Animals , Antibodies, Monoclonal/administration & dosage , Anticoagulants/administration & dosage , Anticoagulants/pharmacology , Cell Adhesion/drug effects , Cell Division/drug effects , Coloring Agents , Heparin/administration & dosage , Heparin/pharmacology , Hyperplasia/prevention & control , Iliac Artery/immunology , Immunoglobulin G/administration & dosage , Immunohistochemistry , Leukocyte Count , Macrophages/immunology , Models, Animal , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/pathology , Neutrophil Infiltration/drug effects , Rabbits , Time Factors , Tunica Intima/drug effects , Tunica Intima/pathology , Wound Healing/drug effects
10.
J Exp Med ; 192(2): 193-204, 2000 Jul 17.
Article in English | MEDLINE | ID: mdl-10899906

ABSTRACT

The firm adhesion and transplatelet migration of leukocytes on vascular thrombus are both dependent on the interaction of the leukocyte integrin, Mac-1, and a heretofore unknown platelet counterreceptor. Here, we identify the platelet counterreceptor as glycoprotein (GP) Ibalpha, a component of the GP Ib-IX-V complex, the platelet von Willebrand factor (vWf) receptor. THP-1 monocytic cells and transfected cells that express Mac-1 adhered to GP Ibalpha-coated wells. Inhibition studies with monoclonal antibodies or receptor ligands showed that the interaction involves the Mac-1 I domain (homologous to the vWf A1 domain), and the GP Ibalpha leucine-rich repeat and COOH-terminal flanking regions. The specificity of the interaction was confirmed by the finding that neutrophils from wild-type mice, but not from Mac-1-deficient mice, bound to purified GP Ibalpha and to adherent platelets, the latter adhesion being inhibited by pretreatment of the platelets with mocarhagin, a protease that specifically cleaves GP Ibalpha. Finally, immobilized GP Ibalpha supported the rolling and firm adhesion of THP-1 cells under conditions of flow. These observations provide a molecular target for disrupting leukocyte-platelet complexes that promote vascular inflammation in thrombosis, atherosclerosis, and angioplasty-related restenosis.


Subject(s)
Macrophage-1 Antigen/physiology , Platelet Glycoprotein GPIb-IX Complex/physiology , Animals , Binding Sites , Blood Platelets/physiology , Cell Adhesion , Cell Line , Humans , Lymphocyte Function-Associated Antigen-1/physiology , Mice , Mice, Inbred C57BL , Neutrophils/physiology
11.
Circ Res ; 87(2): 133-9, 2000 Jul 21.
Article in English | MEDLINE | ID: mdl-10903997

ABSTRACT

Extracellular proteolysis is likely to be a feature of vascular remodeling associated with atherosclerotic and restenotic arteries. To investigate the role of plasminogen-mediated proteolysis in remodeling, polyethylene cuffs were placed around the femoral arteries of mice with single and combined deficiencies in plasminogen and fibrinogen. Neointimal development occurred in all mice and was unaffected by genotype. Significant compensatory medial remodeling occurred in the cuffed arteries of control mice but not in plasminogen-deficient mice. Furthermore, focal areas of medial atrophy were frequently observed in plasminogen-deficient mice but not in control animals. A simultaneous deficit of fibrinogen restored the potential of the arteries of plasminogen-deficient mice to enlarge in association with neointimal development but did not eliminate the focal medial atrophy. An intense inflammatory infiltrate occurred in the adventitia of cuffed arteries, which was associated with enhanced matrix deposition. Adventitial collagen deposition was apparent after 28 days in control and fibrinogen-deficient arteries but not in plasminogen-deficient arteries, which contained persistent fibrin. These studies demonstrate that plasmin(ogen) contributes to favorable arterial remodeling and adventitial collagen deposition via a mechanism that is related to fibrinogen, presumably fibrinolysis. In addition, these studies reveal a fibrin-independent role of plasminogen in preventing medial atrophy in challenged vessels.


Subject(s)
Femoral Artery/physiopathology , Fibrinogen/physiology , Plasminogen/physiology , Tunica Intima/physiopathology , Animals , Collagen/metabolism , Crosses, Genetic , Femoral Artery/pathology , Femoral Artery/physiology , Inflammation , Macrophages/physiology , Mice , Mice, Knockout , Plasminogen/deficiency , Tunica Intima/pathology , Tunica Intima/physiology
12.
J Biol Chem ; 275(14): 10228-34, 2000 Apr 07.
Article in English | MEDLINE | ID: mdl-10744708

ABSTRACT

Adhesion and signaling by integrins require their dynamic association with nonintegrin membrane proteins. One such protein, the glycolipid-anchored urokinase receptor (uPAR), associates with and modifies the function of the beta(2)-integrin Mac-1 (CD11b/CD18). In this study, a critical non-I-domain binding site for uPAR on CD11b (M25; residues 424-440) is identified by homology with a phage display peptide known to bind uPAR. Recombinant soluble uPAR and cells expressing uPAR bound to immobilized M25, binding being promoted by urokinase and blocked by soluble M25, but not a scrambled control or homologous peptides from other beta(2)-associated alpha-chains. Mac-1, but not a mutated Mac-1 in which M25 was replaced with the homologous sequence of CD11c, co-precipitated with uPAR. In the beta-propeller model of alpha-chain folding, M25 spans an exposed loop on the ligand-binding, upper surface of alphaM, identifying uPAR as an atypical alphaM ligand. Although not blocking ligand binding to Mac-1, M25 (25-100 microM) inhibited leukocyte adhesion to fibrinogen, vitronectin, and cytokine-stimulated endothelial cells. M25 also blocked the association of uPAR with beta(1)-integrins and impaired beta(1)-integrin-dependent spreading and migration of human vascular smooth muscle cells on fibronectin and collagen. These observations indicate that uPAR associates with integrins directly and that disruption of this association broadly impairs integrin function, suggesting a novel strategy for regulation of integrins in the settings of inflammation and tumor progression.


Subject(s)
CD18 Antigens/chemistry , CD18 Antigens/metabolism , Macrophage-1 Antigen/metabolism , Muscle, Smooth, Vascular/physiology , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/metabolism , Amino Acid Sequence , Antigens, CD/chemistry , Antigens, CD/metabolism , Binding Sites , Cell Line , Chemotaxis , Humans , Macromolecular Substances , Macrophage-1 Antigen/chemistry , Molecular Sequence Data , Muscle, Smooth, Vascular/cytology , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Plasminogen/metabolism , Plasminogen Activators/metabolism , Receptors, Urokinase Plasminogen Activator , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Saphenous Vein/cytology , Saphenous Vein/physiology , Transfection
13.
J Clin Invest ; 105(3): 293-300, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10675355

ABSTRACT

Inflammation plays an essential role in the initiation and progression of atherosclerosis, but its role in vascular repair after mechanical arterial injury (i.e., percutaneous transluminal coronary angioplasty, PTCA) is unknown. In animal models of vascular injury, leukocytes are recruited as a precursor to intimal thickening. Furthermore, markers of leukocyte activation - in particular, increased expression of the beta2-integrin Mac-1 (alphaMbeta2, or CD11b/CD18), which is responsible for firm leukocyte adhesion to platelets and fibrinogen on denuded vessels - predict restenosis after PTCA. To determine whether Mac-1-mediated leukocyte recruitment is causally related to neointimal formation, we subjected mice lacking Mac-1 to a novel form of mechanical carotid artery dilation and complete endothelial denudation. We now report that the selective absence of Mac-1 impairs transplatelet leukocyte migration into the vessel wall, reducing leukocyte accumulation over time. Diminished medial leukocyte accumulation was accompanied by markedly reduced neointimal thickening after vascular injury. These data establish a role for inflammation in neointimal thickening and suggest that leukocyte recruitment to mechanically injured arteries may prevent restenosis.


Subject(s)
Carotid Arteries/pathology , Carotid Arteries/physiopathology , Inflammation , Macrophage-1 Antigen/physiology , Angioplasty, Balloon , Animals , Gene Deletion , Inflammation/genetics , Male , Mice , Tunica Intima/pathology , Tunica Intima/physiopathology
14.
Thromb Haemost ; 82(2): 291-7, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10605716

ABSTRACT

Emerging evidence indicates a prominent role for non-integrin membrane adaptors in the dynamic regulation of integrin signaling. Two such integrin-associated proteins are the glycosylphosphatidyl-inositol (GPI)-linked urokinase receptor (u-PAR) and the cholesterol-binding protein, caveolin-1. Recent studies indicate that caveolin is required for the association of Src-family kinases with beta 1 integrins. Loss of caveolin/beta 1 integrin association results in loss of ligand-induced focal adhesion kinase (FAK) phosphorylation and impaired development of focal adhesion sites. Similarly, fibronectin-dependent fyn signaling through alpha 5/beta 1 leading to mitogen-activated protein (MAP) kinase activation requires the presence of caveolin-1. Caveolin binds Src-family kinases and such binding maintains these kinases in an inactive state. Current evidence favors a model in which ligand-induced integrin clustering, a central event in integrin activation, promotes caveolin oligomerization leading to release and/or activation of Src-family kinases and initiation of integrin signaling. The presence of u-PAR promotes these events because the extracellular domain(s) of u-PAR binds to beta 1 and beta 2 integrins and the GPI anchor of u-PAR, like that of other GPI-anchored proteins, interacts with cholesterol-rich membrane domains enriched in caveolin and tyrosine kinases. Integrins, caveolin, and u-PAR form interdependent functional complexes, promoting the association of integrins with caveolin-rich signaling domains. During states of accelerated cellular migration, such as during inflammation and tumorigenesis, expression of u-PAR may be a key facilitator of integrin signaling. Interruption of u-PAR/integrin interactions may be a strategy to regulate cellular migration in these settings.


Subject(s)
Caveolins , Integrins/metabolism , Membrane Proteins/metabolism , Receptors, Cell Surface/metabolism , Signal Transduction , Animals , Caveolin 1 , Cell Adhesion , Cell Movement , Humans , Integrin beta1/metabolism , Ligands , Receptors, Urokinase Plasminogen Activator
16.
FASEB J ; 13(9): 965-75, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10336880

ABSTRACT

Considerable epidemiologic data suggest that dietary consumption of vitamin E reduces the incidence of cardiovascular disease. The precise mechanisms are not clear, but emerging data indicate that vitamin E has numerous activities that may, in part, explain its effect on vascular disease. In particular, vitamin E enhances the bioactivity of nitric oxide, inhibits smooth muscle proliferation, and limits platelet aggregation. One common mechanism to account for these effects of vitamin E is the inhibition of protein kinase C stimulation. In the setting of atherosclerosis, inhibition of protein kinase C by vitamin E would be expected to maintain normal vascular homeostasis and thus reduce the clinical incidence of cardiovascular disease.


Subject(s)
Antioxidants/metabolism , Arteriosclerosis/etiology , Blood Vessels/physiology , Models, Biological , Vitamin E/metabolism , Antioxidants/pharmacology , Antioxidants/therapeutic use , Arteriosclerosis/drug therapy , Blood Cells/drug effects , Endothelium, Vascular/drug effects , Homeostasis , Humans , Muscle, Smooth, Vascular/drug effects , Nitric Oxide/metabolism , Protein Kinase C/antagonists & inhibitors , Vitamin E/pharmacology , Vitamin E/therapeutic use
17.
Appl Opt ; 38(8): 1388-92, 1999 Mar 10.
Article in English | MEDLINE | ID: mdl-18305758

ABSTRACT

We discuss the design and performance of diffractive ring-toric lenses for focus-error sensing in optical data storage. A ring-toric lens images a point source of light to a ring-shaped image. Focus-error sensing is accomplished by means of monitoring the change in ring radius: The ring expands in response to a diverging wave front, and the ring contracts in response to a converging wave front. We describe the use of a segmented phi detector to generate a focus-error signal (FES). We found that the FES slope, a measure of sensitivity to disk defocus, is higher for the ring-toric lenses described in this paper than for other techniques such as the astigmatic and the obscuration methods. We measured an FES slope of 0.7 per micrometer of disk defocus (microm(-1)). The corresponding theoretical FES slope is 0.96 microm(-1).

18.
Proc Natl Acad Sci U S A ; 95(17): 10134-9, 1998 Aug 18.
Article in English | MEDLINE | ID: mdl-9707613

ABSTRACT

Leukocytes are recruited early and abundantly to experimentally injured vessels, in direct proportion to cell proliferation and intimal growth. Activated circulating leukocytes and Mac-1 (CD11b/CD18, alphaMbeta2) expression are markers of restenosis risk in patients undergoing angioplasty. As angioplastied vessels lack endothelium but have extensive fibrin(ogen) and platelet deposition, we hypothesized that Mac-1-dependent adhesion to fibrin(ogen) is an important determinant of leukocyte recruitment and function, which may in turn promote intimal growth. To study this hypothesis we administered M1/70, an anti-CD11b blocking mAb, to rabbits (1 mg/kg i.v.) immediately before, and every 48 hr for 3, 6, or 14 days after, iliac artery balloon denudation or deeper stent-induced injury. M1/70, which bound to isolated rabbit monocytes and dose-dependently inhibited Mac-1-mediated fibrinogen binding in vitro, reduced leukocyte recruitment more than 2-fold 3, 6, and 14 days after injury. Neointimal growth 14 days after injury was markedly attenuated by treatment with M1/70 (intimal area after balloon injury, 0.12 +/- 0.09 mm2, compared with 0.32 +/- 0.08 mm2 in vehicle-treated controls, P < 0.01, and 0.38 +/- 0.08 mm2 in IgG-treated controls, P < 0.005; intimal area after stent injury, 0. 56 +/- 0.16 mm2, compared with 0.84 +/- 0.13 mm2 in vehicle-treated controls, P < 0.05, and 0.90 +/- 0.15 mm2 in IgG-treated controls, P < 0.02). Mac-1 blockade reduces experimental neointimal thickening, suggesting that leukocyte recruitment to and infiltration of injured arteries may be a valid target for preventing intimal hyperplasia.


Subject(s)
Angioplasty, Balloon/adverse effects , Antibodies, Monoclonal/pharmacology , CD18 Antigens/physiology , Iliac Artery/injuries , Stents/adverse effects , Animals , CD18 Antigens/immunology , Cell Adhesion , Cell Division , Cell Movement , Humans , Hyperplasia , Iliac Artery/immunology , Iliac Artery/pathology , Inflammation/pathology , Inflammation/prevention & control , Leukocytes/immunology , Leukocytes/pathology , Leukocytes/physiology , Male , Rabbits
19.
J Clin Invest ; 102(3): 576-83, 1998 Aug 01.
Article in English | MEDLINE | ID: mdl-9691094

ABSTRACT

Formation of the atherosclerotic intima must involve altered metabolism of the elastin-rich arterial extracellular matrix. Proteases potentially involved in these processes remain unclear. This study examined the expression of the potent elastases cathepsins S and K in human atheroma. Normal arteries contained little or no cathepsin K or S. In contrast, macrophages in atheroma contained abundant immunoreactive cathepsins K and S. Intimal smooth muscle cells (SMC), especially cells appearing to traverse the internal elastic laminae, also contained these enzymes. Extracts of atheromatous tissues had approximately twofold greater elastase-specific activity than extracts of uninvolved arteries, mostly due to cysteine proteases. Cultured human SMC displayed no immunoreactive cathepsins K and S and exhibited little or no elastolytic activity when incubated with insoluble elastin. SMC stimulated with the atheroma-associated cytokines IL-1beta or IFN-gamma secreted active cathepsin S and degraded substantial insoluble elastin (15-20 microg/10(6) cells/24 h). A selective inhibitor of cathepsin S blocked > 80% of this elastolytic activity. The presence of cathepsins K and S at sites of vascular matrix remodeling and the ability of SMC and macrophages to use these enzymes to degrade elastin supports a role for elastolytic cathepsins in vessel wall remodeling and identifies novel therapeutic targets in regulating plaque stability.


Subject(s)
Arteriosclerosis/enzymology , Cathepsins/biosynthesis , Elastin/metabolism , Muscle, Smooth, Vascular/enzymology , Arteriosclerosis/genetics , Carotid Stenosis/enzymology , Carotid Stenosis/genetics , Cathepsin K , Cathepsins/genetics , Cells, Cultured , Coronary Disease/enzymology , Coronary Disease/genetics , Enzyme Induction , Humans , Interferon-gamma/pharmacology , Muscle, Smooth, Vascular/drug effects , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Tunica Intima/cytology , Tunica Intima/metabolism
20.
Opt Lett ; 23(1): 13-5, 1998 Jan 01.
Article in English | MEDLINE | ID: mdl-18084396

ABSTRACT

The microtag concept is an anticounterfeiting and security measure. Microtags are computer-generated holograms (CGH's) consisting of 150-nm lines arranged to form 300-nm-period gratings. The microtags that we describe were designed for readout at 442nm . The smallest microtag measures 56micromx80 microm when viewed at normal incidence. The CGH design process uses a modified iterative Fourier-transform algorithm to create either phase-only or phase-and-amplitude microtags. We also report on a simple and compact readout system for recording the diffraction pattern formed by a microtag. The measured diffraction patterns agree very well with predictions.

SELECTION OF CITATIONS
SEARCH DETAIL
...