Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 142
Filter
1.
Acta Neurol Scand ; 131(4): 253-7, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25346212

ABSTRACT

OBJECTIVES: To investigate long-term outcome in patients with spontaneous spinal cord infarctions and secondly to compare outcome with that of patients with cerebral infarction. MATERIAL AND METHODS: The study includes 30 patients with spinal cord infarction discharged between 1995 and 2010. Surviving patients were contacted by telephone and sent a questionnaire. Data on employment, function, depression, fatigue, pain, and quality of life were obtained and compared to similar data obtained from a group of patients with cerebral infarction. RESULTS: Seven patients with spinal cord infarction had died after a mean follow-up of 7.1 years. Mortality was associated with poor functioning in the acute phase. Thirteen of 20 responding patients were able to walk. Compared to patients with cerebral infarction, patients with spinal cord infarction had significantly lower mortality, poorer functioning, higher re-employment rate, and more pain. CONCLUSION: Many patients with spinal cord infarction experience significant improvement. Even though functional outcome is worse, the mortality rate is lower and the frequency of re-employment higher among patients with spinal cord infarction compared to patients with cerebral infarction.


Subject(s)
Infarction/complications , Quality of Life , Recovery of Function , Spinal Cord Ischemia/complications , Spinal Cord/blood supply , Aged , Female , Humans , Infarction/mortality , Kaplan-Meier Estimate , Male , Middle Aged , Spinal Cord Ischemia/mortality , Surveys and Questionnaires , Young Adult
2.
Thromb Haemost ; 107(4): 690-8, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22370911

ABSTRACT

Protein S (ProS) is an essential plasma protein that enhances the anticoagulant activity of activated protein C (APC). In vitro , purified native human Zn2+-containing ProS also exerts direct anticoagulant activity by inhibiting prothrombinase and extrinsic FXase activities independently of APC. We investigated antithrombotic effects of ProS infused without APC in a baboon shunt model of thrombogenesis that employs a device consisting of arterial and venous shear flow segments. In in vitro experiments, the Zn2+-containing human ProS used for the studies displayed >10-fold higher prothrombinase inhibitory activity and anticoagulant activity in tissue factor-stimulated plasma, and four-fold higher inhibition of the intrinsic pathway than the Zn2+-deficient ProS used. In the thrombosis model, ProS (33 µg/minute for 1 hour) or saline was infused locally; platelet and fibrin deposition in the shunt were measured over 2 hours. During experiments performed at 50 ml/minute blood flow, Zn2+-containing ProS inhibited platelet deposition 73-96% in arterial-type flow segments and 90-99% in venous-type flow segments; Zn2+-deficient ProS inhibited platelet deposition 52% in arterial-type flow segments and 65-73% in venous-type flow segments. At 100 ml/min blood flow rate, Zn2+-containing ProS inhibited platelet deposition by 39% and 73% in the respective segments; Zn2+-deficient ProS inhibited platelet deposition by 5% and 0% in the respective segments. Zn2+-containing ProS suppressed fibrin deposition by 67-90%. Systemic APC-independent ProS activity was significantly increased and thrombin-antithrombin complex levels were significantly decreased after infusion of ProS. Thus, infused human Zn2+-containing ProS is antithrombotic in primates, and may have therapeutic potential even in protein C-deficient human patients.


Subject(s)
Protein C/metabolism , Protein S/metabolism , Thrombosis/metabolism , Animals , Anticoagulants/metabolism , Blood Platelets/metabolism , Cysteine Endopeptidases/metabolism , Disease Models, Animal , Fibrin/metabolism , Fibrinolytic Agents/pharmacology , Hemostasis , Humans , Male , Neoplasm Proteins/metabolism , Papio , Protein C Deficiency/metabolism , Thrombosis/pathology , Time Factors , Zinc/chemistry
3.
J Thromb Haemost ; 4(2): 392-7, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16420571

ABSTRACT

Anticoagulation with activated protein C (APC) reduces the mortality of severe sepsis. We investigated whether the circulating protein C (PC) pool could be utilized for sustained anticoagulation by endogenous APC. To generate APC without procoagulant effects, we administered the anticoagulant thrombin mutant W215A;E217A (WE) to baboons. In preliminary studies, administration of high dose WE (110 microg kg(-1) i.v. bolus every 120 min; n = 2) depleted PC levels by 50% and elicited transient APC bursts and anticoagulation. The response to WE became smaller with each successive injection. Low dose WE infusion (5 microg kg(-1) loading + 5 microg kg(-1) h(-1) infusion; n = 5) decreased plasma PC activity by 15%, from 105% to 90%, to a new equilibrium within 60 min. APC levels increased from 7.5 ng mL(-1) to 86 ng mL(-1) by 40 min, then declined, but remained elevated at 34 ng mL(-1) at 240 min. A 22-fold higher dose WE (n = 5) decreased PC levels to 60% by 60 min without significant further depletion in 5 h. The APC level was 201 ng mL(-1) at 40 min and decreased to 20 ng mL(-1) within 120 min despite continued activator infusion. Co-infusion of WE and equimolar soluble thrombomodulin (n = 5) rapidly consumed about 80% of the PC pool with significant temporal increase in APC generation. In conclusion, low-grade PC activation by WE produced sustained, clinically relevant levels of circulating APC. Limited PC consumption in WE excess was consistent with the rapid depletion of cofactor activity before depletion of the PC zymogen. Reduced utilization of circulating PC might have similar mechanism in some patients.


Subject(s)
Protein C/metabolism , Thrombin/pharmacology , Amino Acid Substitution , Animals , Anticoagulants/administration & dosage , Anticoagulants/pharmacology , Hemostasis/drug effects , Humans , Infusions, Intravenous , Injections, Intravenous , Mutagenesis, Site-Directed , Papio , Recombinant Proteins/administration & dosage , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Thrombin/administration & dosage , Thrombin/genetics
4.
Curr Pharm Des ; 9(28): 2367-74, 2003.
Article in English | MEDLINE | ID: mdl-14529397

ABSTRACT

Thrombosis is the collective term for diseases caused by the localized accumulation of circulating blood elements within the vasculature that result in vessel occlusion. Conventional antithrombotic drugs can inhibit thrombus growth by targeting coagulation pathways (e.g., heparin, warfarin) or platelet-dependent mechanisms (e.g., aspirin, clopidogrel). Thrombolytic agents (e.g., streptokinase) are used to degrade thrombi in situ, thereby restoring the blood flow. Despite advances, the search for new strategies continues because existing treatments impair hemostasis, and must be administered at dose levels that do not achieve maximum efficacy. Only a few drugs are used at markedly efficacious doses, for short periods of time in closely watched clinical situations, such as interventional cardiology and surgery. Ideally, new targets for therapy would lead to the development of agents that are specific for thrombus-forming mechanisms without affecting hemostasis. In the absence of such agents, new products should preferentially inhibit the thrombotic process at doses that are relatively safe. The symptomatology of naturally occurring or experimentally-induced alterations of relevant hemostatic pathways can serve as basis for target selection. Hemostatic disorders that are compatible with life, do not pose a significantly increased risk of bleeding, but potentially protect against thrombosis provide guidance for rational design strategies. Theoretical considerations and recent experimental data suggest that: 1) inhibition of intrinsic coagulation pathway activity, 2) reduction of circulating platelet count, or 3) activation or enhancement of endogenous protein C or thrombolytic pathways could improve antithrombotic therapy.


Subject(s)
Fibrinolytic Agents/pharmacology , Thrombosis/drug therapy , Animals , Blood Coagulation/drug effects , Fibrinolytic Agents/therapeutic use , Humans , Lipoproteins/metabolism , Plasminogen Activator Inhibitor 1/metabolism , Platelet Aggregation/drug effects , Protein C/metabolism , Selectins/metabolism , Thrombin/antagonists & inhibitors
6.
J Med Chem ; 44(26): 4497-500, 2001 Dec 20.
Article in English | MEDLINE | ID: mdl-11741468

ABSTRACT

Myoseverin, a trisubstituted purine, inhibits microtubule assembly in vitro, interferes with normal mitotic spindle assembly, and arrests the cell cycle in mitosis in U937 cells. We synthesized a variety of myoseverin derivatives and screened them for inhibition of spindle assembly in Xenopus egg extracts and for microtubule disassembly in vitro. Selected compounds were tested against 60 cancer cell lines at the National Cancer Institute as possible anticancer drug candidates.


Subject(s)
Antineoplastic Agents/chemical synthesis , Purines/chemical synthesis , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Biopolymers , Cell Division/drug effects , Depression, Chemical , Flow Cytometry , Humans , In Vitro Techniques , Oocytes , Purines/chemistry , Purines/pharmacology , Spindle Apparatus/drug effects , Structure-Activity Relationship , Tissue Extracts , Tubulin/chemistry , U937 Cells , Xenopus laevis
7.
Thromb Res ; 104(3): 149-74, 2001 Nov 01.
Article in English | MEDLINE | ID: mdl-11672758

ABSTRACT

The purpose of the present communication is to evaluate the importance of blood flow and surface reactivity for measurement of antithrombotic drug activity or efficacy in selected model systems of thrombus formation. Such information is essential for proper evaluation of antithrombotic drug profiles. The continuous development of flow-dependent thrombosis models for in vitro (anticoagulated blood) and ex vivo (native blood) studies and their application in in vivo animal models from the early 1970s and onwards are briefly considered. Central to this process was the development of various types of perfusion chambers in which a thrombogenic surface is exposed to flowing blood. Such perfusion chambers have been inserted into arteriovenous (AV) shunts in baboon, pig, dog, and rabbit. These approaches have allowed reproducible testing of traditional and novel experimental antithrombotic drugs, and studies on novel drug strategies under well-defined shear conditions and surface reactivity. Shear-dependent antithrombotic efficacy in these models is observed with anticoagulants such as unfractionated heparin, low-molecular weight heparins, or selective inhibitors of thrombin, Factor Xa, or Factor VIIa. However, the degree of shear dependency depends on the nature of the thrombogenic surface, e.g., the inhibition is more pronounced on a tissue factor (TF)-rich surface than on a collagen-rich surface, particularly at venous or low arterial shear. Platelet antagonists such as the COX-1 inhibitor aspirin, inhibitors of thromboxane A2 (TxA2) synthetase, the TxA2 platelet receptor, and of von Willebrand factor (vWf) are shear dependent also, being more efficient at high arterial shear. In contrast, the platelet ADP antagonist clopidogrel, or antagonists to the active platelet membrane glycoprotein IIb-IIIa complex (GPIIb-IIIa) are shear independent. At extremely high arterial shear, which activates platelets and elicit aggregates of circulating platelets, aspirin looses its antithrombotic effect, whereas ADP and GPIIb-IIIa antagonists still interrupt thrombus formation. In general, results obtained with these models mimic and predict antithrombotic efficacy in man when comparison is possible. Information on antithrombotic efficacy in flow devices with various thrombogenic surfaces is now sufficiently available to suggest recommendations for experimental conditions, particularly with regard to blood flow and reactive surfaces.


Subject(s)
Drug Evaluation, Preclinical/methods , Fibrinolytic Agents/pharmacology , Hemorheology/drug effects , Animals , Disease Models, Animal , Drug Evaluation, Preclinical/instrumentation , Hemorheology/instrumentation , Hemorheology/methods , Humans , Models, Cardiovascular , Platelet Activation/drug effects , Platelet Aggregation Inhibitors/pharmacology , Stress, Mechanical , Therapeutic Equivalency , Thrombosis/blood , Thrombosis/drug therapy , Thrombosis/etiology
8.
Ann Biomed Eng ; 29(8): 657-64, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11556722

ABSTRACT

The clinical histories of the Medtronic Parallel (MP) and St. Jude Medical (SJM) Standard valves suggest pivot geometry influences the thrombogenic characteristics of bileaflet prostheses. This work studied the effects of various pivot geometries on markers of platelet damage in a controlled, in vitro apparatus. The Medtronic Parallel valve, two St. Jude Medical valves, and two demonstration prostheses were used to study the effects of bileaflet pivot design, gap width, and size on platelet secretion and anionic phospholipid expression during leakage flow. A centrifugal pump was used to drive blood through a circuit containing a bileaflet prosthesis. Samples were taken at set time intervals after the start of the pump. These samples were analyzed by cell counting, flow cytometry, and enzyme-linked immunosorbant assay. No significant differences were observed in platelet secretion or anionic phospholipid expression between experiments with the SJM 27 Standard regular leaker, the SJM 20 regular leaker, and the MP 27 valves. Significant differences in platelet secretion and anionic phospholipid expression were observed between a SJM 27 Standard regular leaker and a SJM 27 high leaker valve. These studies suggest that leakage gap width within bileaflet valve pivots has a significant effect on platelet damage initiated by leakage flow.


Subject(s)
Aortic Valve , Blood Platelets/physiology , Heart Valve Prosthesis , Phospholipids/blood , Prosthesis Design , Anions , Biomedical Engineering , Heart Valve Prosthesis/adverse effects , Hemodynamics , Humans , In Vitro Techniques , Phospholipids/chemistry , Platelet Factor 4/metabolism , Thrombosis/etiology
9.
Ann Biomed Eng ; 29(4): 321-9, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11339329

ABSTRACT

Thrombosis on an atherosclerotic lesion can cause heart attack or stroke. Thrombosis may be triggered by plaque rupture or erosion, creating a thrombogenic stenosis. To measure and model this situation, collagen-coated stenoses have been exposed to nonanticoagulated blood in a baboon ex vivo shunt. The maximum rate of platelet accumulation, measured using a gamma camera, was highest in the throat region of moderate and severe stenoses, and increased with increasing stenosis severity. A species transport model of platelet accumulation was developed, which included mechanisms of convection, shear-enhanced diffusion, near-wall platelet concentration, and a kinetic model of platelet activation and aggregation. The model accurately reproduced the average spatial pattern and time rate of platelet accumulation in the upstream and throat regions of the stenosis, where shear-enhanced diffusivity increased platelet transport in the stenosis throat. Downstream of the throat where flow is complicated by recirculation, the model computed a transport-limited region with lower than measured platelet accumulation, suggesting that fluid-phase platelet activation may significantly affect both transport and adhesion rates in the poststenotic region. This model may provide an initial quantitative estimate of the likelihood of occlusive thrombus in individual patients due to plaque erosion, artery spasm, incomplete angioplasty, or plaque rupture.


Subject(s)
Arteriosclerosis/blood , Models, Cardiovascular , Platelet Adhesiveness/physiology , Thrombosis/blood , Animals , Biomedical Engineering , Constriction, Pathologic , Hemodynamics , Humans , In Vitro Techniques , Papio , Platelet Activation/physiology
10.
Thromb Haemost ; 85(3): 488-93, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11307820

ABSTRACT

Thromboembolic complications have been attributed to the use of radiographic contrast media (CM) during interventional procedures for arterial revascularization. However, due to the low frequency of adverse events, comparisons between different CM have been difficult to perform, although it has been suggested that ionic (vs. non-ionic) CM may be associated with fewer thrombotic events. The present study was undertaken using well-characterized baboon thrombosis models in order to compare different CM under physiologically relevant and controlled conditions of blood flow, exposure time, and CM concentration. Three CM were studied: ioxaglate, iohexol, and iodixanol. CM were locally infused into the proximal segment of femoral arteriovenous shunts. Palmaz-Schatz stents (4 mm i.d.) and expanded tubular segments (9 mm i.d.), which exhibited venous-type flow recirculation and stasis, were deployed into the shunts distally. Saline was infused in identical control studies. Blood flow was maintained at 100 ml/min. Thrombosis was measured over a blood exposure period of 2 hours by gamma camera imaging of 111In-platelets and by gamma counting of deposited 125I-fibrin. CM concentrations within the flowfield were predicted using computational fluid dynamics. At infusion rates of 0.1 and 0.3 ml/min, the low-osmolar ionic CM ioxaglate reduced both platelet and fibrin deposition on the stents by 75-80% (p <0.005), while both iohexol and iodixanol reduced platelet deposition by 30-50% (p <0.05). In the regions of low shear flow, ioxaglate (0.3 ml/min) also reduced platelet deposition significantly (by 52% vs. control results; p <0.05). Thus the three agents evaluated--ioxaglate, iohexol, and iodixanol--all produced anticoagulant and antiplatelet effects and were inherently antithrombotic in vivo. The most striking effects were seen with the low osmolarity, ionic contrast agent ioxaglate.


Subject(s)
Contrast Media/therapeutic use , Fibrinolytic Agents/pharmacology , Thrombosis/prevention & control , Animals , Arteriovenous Shunt, Surgical , Blood Flow Velocity , Contrast Media/pharmacology , Disease Models, Animal , Hemodynamics/drug effects , Iohexol/pharmacology , Iohexol/therapeutic use , Ions/pharmacology , Ioxaglic Acid/pharmacology , Ioxaglic Acid/therapeutic use , Models, Cardiovascular , Papio , Thrombosis/drug therapy , Triiodobenzoic Acids/pharmacology , Triiodobenzoic Acids/therapeutic use
11.
J Heart Valve Dis ; 10(2): 228-38, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11297211

ABSTRACT

BACKGROUND AND AIM OF THE STUDY: The recent clinical history and experimental studies of the Medtronic Parallel (MP) valve suggest that bileaflet valve leakage flow is a primary initiator of thrombosis. These studies investigated the effects of physiologic leakage flow through a MP valve on various markers of blood damage. METHODS: A centrifugal pump was used to drive whole, human blood anticoagulated with PPACK through a circuit containing a MP 27 mm valve in the closed position (experimental runs) or a MP 27 mm valve in the open position (control runs). Samples were taken at set time intervals after the start of the pump. These samples were analyzed by cell counting, flow cytometry, and ELISA. RESULTS: Cell counts remained relatively constant in both the experimental and control runs. Increases in plasma hemoglobin concentration and the percentage of glycophorin A-positive fragments in the cell population were not significant in either the experimental or the control runs. Plasma platelet factor 4 activity and the percentage of the CD41-positive population which was positive for annexin V increased significantly (p <0.05) in the experimental runs compared with the control runs. CONCLUSION: The results indicate that bileaflet valve leakage flow causes significant platelet disruption, that erythrocytes are more resistant to disruption by leakage flow than platelets and granulocytes, and that annexin V binding to platelets and plasma platelet factor 4 activity are more sensitive markers of leakage induced blood damage than plasma hemoglobin concentration.


Subject(s)
Biomarkers/blood , Coronary Thrombosis/blood , Heart Valve Diseases/blood , Heart Valve Prosthesis/adverse effects , Prosthesis Failure , Annexin A5/blood , Cell Count , Glycophorins/analysis , Heart Valve Diseases/surgery , Hemoglobins/analysis , Humans , P-Selectin/blood , Platelet Factor 4/analysis , Sensitivity and Specificity
12.
J Trace Elem Med Biol ; 15(4): 243-53, 2001.
Article in English | MEDLINE | ID: mdl-11846014

ABSTRACT

Virgin and lactating Sprague Dawley rats were used to determine whether the pathways of silver transport to tissues and milk resemble those for copper. Rats were injected i.p.with small amounts of 110AgNO3. Blood and tissues were examined at various times thereafter for total radioactivity and for incorporation into copper binding proteins in plasma and milk. As with 67Cu, much of the 110Ag was rapidly incorporated into the liver. Skeletal muscle, spleen, mammary gland, ovaries, uterus and adrenals also were significant initial accumulation sites, with or without lactation. Lactation enhanced uptake by the mammary gland, and radioactivity rapidly entered the milk and milk ceruloplasmin. In the plasma, most of the 110Ag bound to a single component of apparent molecular weight 800 k throughout the 52 h period examined. A small proportion was also incorporated into plasma ceruloplasmin, as determined by immunoprecipitation and native gel electrophoresis. There was little or no association of 110Ag with albumin or transcuprein. The binding of 110Ag to the 800 kDa protein was tight. Off rates during pH 7 dialysis were <2.5%/day even in the presence of 100 microM histidine or Cu(II), but were accelarated by mercaptoethanol. Subunits of 145 and 45 kDa in virtually pure peak fractions were those of alpha1-macroglobulin. We conclude that silver resembles copper in aspects of its tissue distribution, response to lactation, and incorporation into ceruloplasmin. However its main plasma carrier appears to be alpha1-macroglobulin, a different macroglobulin than that involved in copper transport.


Subject(s)
Copper/pharmacokinetics , Silver/pharmacokinetics , Animals , Biological Transport , Ceruloplasmin/metabolism , Chromatography , Dose-Response Relationship, Drug , Electrophoresis, Polyacrylamide Gel , Female , Lactation , Milk/metabolism , Precipitin Tests , Rats , Rats, Sprague-Dawley , Time Factors , Tissue Distribution , alpha-Macroglobulins/metabolism
13.
Arch Gen Psychiatry ; 57(9): 875-82, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10986551

ABSTRACT

BACKGROUND: Alterations in platelet reactivity have been previously posited to underlie the increased vulnerability of patients with depression to ischemic heart disease (IHD). The present study sought to determine whether the increased platelet reactivity associated with major depression is reduced after antidepressant treatment. METHODS: Patients diagnosed as having DSM-IV major depression (n = 15) (mean age, 37 +/- 7 years; range, 23-48 years) and 12 normal comparison subjects (mean age, 36 +/- 7; range, 23-48 years) were recruited. None of the controls or depressed group had evidence of IHD; 10 of 15 patients who were depressed had 1 or more traditional IHD risk factors. In vivo platelet activation, secretion, and dose-response aggregation of the controls and patients was measured after overnight bedrest under basal conditions, and after a mild exercise challenge. After 6 weeks of open-label treatment with the selective serotonin reuptake inhibitor paroxetine (20 mg/d), the patients with depression were readmitted and procedures of the first General Clinical Research Center admission repeated. RESULTS: In comparison with the control group, the depressed group exhibited greater procoagulant activity as detected by increased platelet binding of the monoclonal antibodies anti-ligand-induced binding site and GA6, and increased plasma concentrations of platelet factor 4 under basal conditions. After paroxetine treatment, the patients with depression exhibited significant reductions in all 3 parameters. CONCLUSIONS: Normalization of platelet activation is associated with paroxetine treatment of patients with depression. Because this study design did not allow for the determination of whether this effect of paroxetine on platelet function is caused by a direct effect of the drug or placebo or, alternatively, because of recovery from depression, studies containing a placebo and/or psychotherapy treatment arm may resolve this issue.


Subject(s)
Blood Platelets/metabolism , Depressive Disorder/blood , Depressive Disorder/drug therapy , Paroxetine/therapeutic use , Platelet Activation , Selective Serotonin Reuptake Inhibitors/therapeutic use , Adenosine Triphosphate/metabolism , Blood Platelets/immunology , Depressive Disorder/metabolism , Dose-Response Relationship, Drug , Flow Cytometry , Humans , Paroxetine/pharmacology , Peptide Fragments/metabolism , Physical Exertion/physiology , Platelet Activation/drug effects , Platelet Aggregation/drug effects , Platelet Count , Platelet Factor 4/metabolism , Receptors, Thrombin/antagonists & inhibitors , Regression Analysis , Risk Factors , Selective Serotonin Reuptake Inhibitors/pharmacology , beta-Thromboglobulin/metabolism
14.
Exp Eye Res ; 71(2): 195-207, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10930324

ABSTRACT

This investigation of the water-insoluble crystallins from human lenses has used multiple chromatographic separations to obtain proteins of sufficient purity for mass spectrometric analysis. Each fraction was analysed to determine the molecular masses of the constituent proteins as well as peptides in tryptic digests of these proteins. The major components of the water-insoluble crystallins were identified as alphaA- and alphaB-crystallins. In addition, gammaS-, betaB1-, gammaD-, betaA3/A1- and betaB2-crystallins were found, in order of decreasing abundance. Although there was evidence of some backbone cleavage, the predominant forms of alphaA-, alphaB, betaB2-, gammaS- and gammaD-crystallins were the intact polypeptide chains. The major modifications distinguishing the water-soluble crystallins were increased disulfide bonding, oxidation of Met, deamidation of Gln and Asn and backbone cleavage. Of the many reactions hypothesized to lead to crystallin insolubility and cataract, these results most strongly support metal-catalysed oxidation, deamidation and truncation as initiators of conformational changes that favor aggregation.


Subject(s)
Crystallins/chemistry , Disulfides/chemistry , Lens, Crystalline/chemistry , Methionine/chemistry , Aged , Chromatography, High Pressure Liquid , Deamination , Humans , Lens, Crystalline/metabolism , Mass Spectrometry , Methionine/metabolism , Middle Aged , Oxidation-Reduction
15.
Arterioscler Thromb Vasc Biol ; 20(4): 1168-72, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10764689

ABSTRACT

Biocompatible stent coatings may alleviate problems of increased (sub)acute thrombosis after stent implantation. Hyaluronic acid (HA), a ubiquitous, nonsulfated glycosaminoglycan, inhibits platelet adhesion and aggregation and prolongs bleeding when administered systemically. However, the effects of immobilized HA for reducing stent platelet deposition in vivo are unknown. We therefore quantified the antithrombotic effects of coating stainless steel stents and tubes with HA using an established baboon thrombosis model under physiologically relevant blood flow conditions. HA-coated and uncoated (control) stents (3.5 mm in diameter, n=32) and stainless steel tubes (4.0 mm in diameter, n=18) were deployed into exteriorized arteriovenous shunts of conscious, nonanticoagulated baboons. Accumulation of (111)In-radiolabeled platelets was quantified by continuous gamma-camera imaging during a 2-hour blood exposure period. HA coating resulted in a significant reduction in platelet deposition in long (4 cm) tubes (0.24+/-0.15 x 10(9) versus 6.12+/-0.49 x 10(9) platelets; P<0.03), short (2 cm) stainless steel tubes (0.18+/-0.06 x 10(9) versus 3.03+/-0.56 x 10(9) platelets; P<0.008), and stents (0.82+/-0.20 x 10(9) versus 1.83+/-0. 23 x 10(9) platelets; P<0.02) compared with uncoated control devices. Thus, HA coating reduces platelet thrombus formation on stainless steel stents and tubes in primate thrombosis models. These results indicate that immobilized HA may represent an attractive strategy for improving the thromboresistance of endovascular devices.


Subject(s)
Biocompatible Materials , Hyaluronic Acid , Stents/adverse effects , Thrombosis/prevention & control , Animals , Blood Platelets/physiology , Kinetics , Male , Microscopy, Electron, Scanning , Papio , Platelet Adhesiveness , Thrombosis/etiology , Thrombosis/pathology
16.
Thromb Res ; 98(2): 195-201, 2000 Apr 15.
Article in English | MEDLINE | ID: mdl-10713321

ABSTRACT

Two cDNAs encoding the thrombin receptor of baboon vascular smooth muscle cells have 5'-untranslated regions that begin upstream of multiple putative transcription initiation sites reported for the closely related human receptor gene. The extent of these baboon 5'-untranslated cDNA regions and their close similarity to the corresponding human sequences suggest that there is only one transcription initiation site of the primate thrombin receptor gene, which might be linked to a typical TATA-box previously identified in the upstream region of the human gene. It is possible that all primates have a unique thrombin receptor gene transcription start site. Inferences drawn from the baboon system may be usefully extrapolated to the human, in view of the extensive similarities seen between the nucleotide sequences of baboon and human thrombin receptor gene sequences in the 5'- untranslated and coding regions. The extents of the 5'-untranslated region of the baboon cDNAs argue that the "multiple" transcription start sites identified for the human gene are artifactual. The striking differences in the reported baboon and human transcription start sites warrant further investigation in view of the significant role played by the thrombin receptor in numerous vascular and cellular growth responses.


Subject(s)
DNA, Complementary/genetics , Papio/genetics , Receptors, Thrombin/genetics , Amino Acid Sequence , Animals , Base Sequence , Cells, Cultured , Humans , Molecular Sequence Data , Muscle, Smooth, Vascular/metabolism , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Species Specificity , Transcription, Genetic
17.
J Vasc Surg ; 31(2): 354-63, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10664503

ABSTRACT

PURPOSE: Recently, we designed and characterized a novel expanded polytetrafluoroethylene (ePTFE)-based local drug delivery approach that selectively concentrates infused pharmacologic agents specifically within those blood layers adjacent to the graft wall and at downstream anastomotic sites. In this study, we locally administrated standard heparin therapy and evaluated its effects on neointimal hyperplasia formation in a baboon model of aortoiliac bypass graft placement. METHODS: Six adult male baboons underwent bilateral aortoiliac bypass grafting with ringed ePTFE (4 mm internal diameter x 5 cm length). In each animal, the distal anastomosis of one graft was continuously infused with heparin (50 U/h) and the distal anastomosis of the contralateral graft was infused with saline solution at the same rate (2.5 microL/h), with osmotic pumps implanted for 4 weeks. Platelet counts and activated partial thromboplastin time measurements were performed weekly. The specimens were harvested at 4 weeks and were subjected to morphometric analysis. Cell proliferation was assessed with bromodeoxyuridine immunostaining. RESULTS: All the harvested grafts were patent except for one control graft. There were no significant differences in platelet counts or activated partial thromboplastin time measurements taken before and during heparin infusion. As expected, there were no significant differences in graft neointimal hyperplasia and cell proliferation at the proximal anastomoses between the heparin-infused and control grafts. In contrast, at the treated distal anastomoses, heparin infusion significantly reduced the graft neointimal area by 65% and the cell proliferation index by 47% as compared with the untreated control distal anastomoses. CONCLUSION: These results show that local infusion of heparin significantly reduces distal anastomotic neointimal hyperplasia and cell proliferation without measurable systemic anticoagulation or other side effects. Thus, this approach may represent an attractive strategy for prolonging ePTFE bypass graft patency.


Subject(s)
Aorta, Abdominal/drug effects , Blood Vessel Prosthesis , Fibrinolytic Agents/administration & dosage , Heparin/administration & dosage , Iliac Artery/drug effects , Infusion Pumps, Implantable , Polytetrafluoroethylene , Postoperative Complications/prevention & control , Anastomosis, Surgical , Animals , Aorta, Abdominal/metabolism , Aorta, Abdominal/pathology , Aorta, Abdominal/surgery , Drug Evaluation, Preclinical , Hyperplasia/metabolism , Hyperplasia/pathology , Hyperplasia/prevention & control , Iliac Artery/metabolism , Iliac Artery/pathology , Iliac Artery/surgery , Immunohistochemistry , Male , Papio , Postoperative Complications/metabolism , Postoperative Complications/pathology , Time Factors
18.
Circulation ; 100(19): 2018-24, 1999 Nov 09.
Article in English | MEDLINE | ID: mdl-10556229

ABSTRACT

BACKGROUND: The hypothesis that thrombin mediates the formation of neointimal vascular lesions at sites of mechanical vascular injury has been tested in baboons by measurement of the effects of hirudin delivered by retrovirus-transduced hirudin-secreting vascular endothelial cells (ECs) lining surgically implanted arterial vascular grafts (AVGs). METHODS AND RESULTS: The antithrombotic efficacy of baboon ECs transduced with cDNA encoding hirudin was assessed in vitro and in vivo on thrombogenic segments in chronically exteriorized femoral arteriovenous (AV) shunts. Bilateral brachial AVGs lined with hirudin-transduced versus nonhirudin control ECs at confluent density were surgically implanted, and vascular lesion formations at distal graft-vessel anastomoses were compared after 30 days. Hirudin-transduced ECs secreted 20+/-6 ng x 10(6) cells(-1) x 24 h(-1) (range, 14 to 24 ng x 10(6) cells(-1) x 24 h(-1)) hirudin in supernatants of static cultures. Hirudin-secreting ECs on segments of collagen-coated graft interposed in chronic AV shunts decreased the accumulation of (111)In-labeled platelets to 0.52+/-0.34 x 10(9) platelets, compared with 0.82+/-0.49 x 10(9) platelets in controls (P = 0.03) and reduced platelet deposition in propagated thrombotic tails extending downstream from segments of vascular graft from 1.38+/-0.41 x 10(9) platelets in controls to 0.59+/-0.22 x 10(9) platelets (P = 0.04). ECs recovered from 30-day AVG implants generated 17+/-9 ng x 10(6) cells(-1) x 24 h(-1) (range, 9 to 25 ng x 10(6) cells(-1) x 24 h(-1)) hirudin. Hirudin-secreting ECs reduced neointimal lesion formation at distal graft-vessel anastomoses, ie, 1.02 mm(2) (range, 0.88 to 1.95 mm(2)) versus 1.82 mm(2) (range, 0.88 to 2.56 mm(2)) in contralateral AVGs bearing nonhirudin control ECs (P<0.01). CONCLUSIONS: Viral vector-directed secretion of hirudin from ECs lining implanted AVGs significantly reduces the formation of thrombus and neointimal vascular lesions.


Subject(s)
Antithrombins/therapeutic use , Blood Vessel Prosthesis , Endothelium, Vascular/metabolism , Hirudin Therapy , Muscle, Smooth, Vascular/pathology , Retroviridae/genetics , Thrombosis/prevention & control , Animals , Hirudins/genetics , Male , Papio , Transfection
19.
Ultrasound Med Biol ; 25(4): 561-6, 1999 May.
Article in English | MEDLINE | ID: mdl-10386731

ABSTRACT

We tested the ability of ultrasound radiofrequency (RF) signal analysis to characterize thrombus accumulation in a Dacron graft incorporated into the exteriorized arteriovenous shunt in 3 baboons with constant blood flow for 60 min. Thrombus formation was quantified by sequential measurements of 111Indium-labeled platelet deposition. RF signals were acquired every 15 min at 2 sites in the graft, using a 2.9 Fr intravascular ultrasound catheter-based transducer (30 MHz) and digitized at 250 MHz in 8-bit resolution. Regions of interest were placed within a 0.5-mm perimeter adjacent to the graft wall. Integrated backscatter increased significantly (p < 0.001) with increasing platelet deposition. However, mean-to-standard deviation ratio of the RF envelope showed no significant change and the distribution pattern of the RF probability function remained constant and consistent with a Rayleigh scattering process. These results provide a basis for using RF analysis to monitor the time-course of thrombus formation.


Subject(s)
Disease Models, Animal , Thrombosis/diagnostic imaging , Ultrasonography, Interventional/methods , Analysis of Variance , Animals , Arteriovenous Shunt, Surgical , Blood Vessel Prosthesis , Disease Progression , Femoral Artery , Femoral Vein , Graft Occlusion, Vascular/diagnostic imaging , Papio , Polyethylene Terephthalates , Time Factors , Ultrasonography, Interventional/instrumentation , Ultrasonography, Interventional/statistics & numerical data
20.
Arterioscler Thromb Vasc Biol ; 19(4): 900-9, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10195916

ABSTRACT

Restenosis remains a significant clinical problem associated with mechanical interventional procedures for arterial revascularization or repair, including coronary angioplasty and stenting. Studies with rodents have established that platelet-derived growth factor (PDGF), a potent chemotactic and mitogenic agent for vascular smooth muscle cells, is a key mediator of lesion formation after vascular injury. To further explore this hypothesis in a more clinically relevant model, neutralizing monoclonal antibodies (mAbs) were used to examine the effect of selective inhibition of alpha or beta PDGF receptor (PDGFR) on neointima formation in nonhuman primates. Carotid arteries were injured by surgical endarterectomy and femoral arteries by balloon catheter dilatation. Immunostaining revealed that both injuries induced cell proliferation and the upregulation of beta PDGFR but not alpha PDGFR. By 7 days after injury, beta PDGFR staining was limited to the luminal region of the media, the small areas of neointima, and the adventitia. Nearly all bromodeoxyuridine-positive cells were found in these regions as well. After 30 days, a concentric neointima that stained strongly for beta PDGFR had formed in the carotid and femoral arteries. Treatment of baboons with anti-beta PDGFR mAb 2A1E2 for 6 days after injury reduced the carotid artery and femoral artery lesion sizes by 37% (P<0.05) and 48% (P<0.005), respectively, when measured at 30 days. Under the same conditions, treatment with anti-alpha PDGFR mAb 2H7C5 had no effect. These findings suggest that PDGF mediates neointima formation through the beta PDGFR, and that antagonism of this pathway may be a promising therapeutic strategy for reducing clinical restenosis.


Subject(s)
Carotid Artery Injuries , Femoral Artery/injuries , Receptors, Platelet-Derived Growth Factor/physiology , Animals , Antibodies, Blocking/pharmacology , Antibodies, Monoclonal/pharmacology , Carotid Arteries/pathology , Catheterization , Cell Division/immunology , Endarterectomy , Femoral Artery/pathology , Male , Papio , Phosphorylation , Receptor, Platelet-Derived Growth Factor alpha , Receptor, Platelet-Derived Growth Factor beta , Receptors, Platelet-Derived Growth Factor/antagonists & inhibitors , Receptors, Platelet-Derived Growth Factor/biosynthesis , Receptors, Platelet-Derived Growth Factor/immunology , Tunica Intima/immunology , Tunica Intima/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...