Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
Add more filters










Publication year range
1.
J Thromb Haemost ; 1(12): 2497-509, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14675084

ABSTRACT

Blood platelets maintain vascular integrity and promote primary and secondary hemostasis following interruption of vessel continuity. Biochemical or physical damage to the coronary, carotid or peripheral arteries is followed by excessive platelet activation and recruitment culminating in vascular occlusion and tissue ischemia. Currently inadequate therapeutic approaches to stroke and coronary artery disease are a public health issue. Following our demonstration of neutrophil leukotriene production from arachidonate released from activated aspirin-treated platelets, we studied interactions between platelets and other blood cells, leading to concepts of transcellular metabolism and thromboregulation. Thrombosis has a proinflammatory component whereby biologically active substances are synthesized by interactions between different cell types that could not individually synthesize the product(s). Endothelial cells control platelet reactivity via three biochemical systems-autacoids leading to production of prostacyclin and nitric oxide, and endothelial ecto-ADPase/CD39/NTPDase-1. The autacoids are fluid-phase reactants, not produced by tissues in the basal state. They are only synthesized intracellularly and released upon interactions of cells with an agonist. When released, autacoids exert fleeting actions in the immediate milieu, and are rapidly inactivated. CD39 is an integral component of the endothelial cell surface and is substrate-activated. It maintains vascular fluidity in the complete absence of prostacyclin and nitric oxide, indicating that they are ancillary components of hemostasis. Therapeutic implications for the autacoids have not been compelling because of their transient, local and fleeting action, and limited potency. Conversely, CD39, acting solely on the platelet releasate, is efficacious in three different animal models. It metabolically neutralizes a prothrombotic platelet releasate via deletion of ADP--the major recruiting agent responsible for formation of an occlusive thrombus. In addition, solCD39 reduced ATP- and ischemia-induced norepinephrine release in the heart. This reduction can prevent fatal arrhythmia. Moreover, solCD39 ameliorated the sequelae of stroke in CD39 null mice. CD39 represents the next generation of cardioprotective and cerebroprotective molecules.


Subject(s)
Adenosine Triphosphatases/physiology , Antigens, CD/physiology , Cell Communication/physiology , Adenosine Diphosphate/metabolism , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Apyrase , Aspirin/pharmacology , Aspirin/therapeutic use , Cell Communication/drug effects , Endothelial Cells/enzymology , Endothelial Cells/metabolism , Hemostasis , Humans , Thrombosis/blood , Thrombosis/drug therapy
2.
Ital Heart J ; 2(11): 824-30, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11770867

ABSTRACT

During their 7-9 day lifespan in the circulation platelets are mainly responsible for maintaining the integrity of the vasculature. In thrombocytopenic states, there is an increase in vascular permeability and fragility, presumably due to absence of this platelet function. In sharp contrast, biochemical or physical injury in the coronary, carotid or peripheral arteries induces platelet activation and platelet recruitment, which can culminate in thrombotic vascular occlusion. Since there is one death every 33 s from vascular occlusion in the United States, this situation constitutes a major public health issue. In the course of studying interactions between cells of the vascular wall and those in the circulation, we observed that platelets in close proximity to endothelial cells do not respond to agonists in vitro. Experiments initiated in the late 1980's cumulatively indicated that endothelial cell CD39--an ecto-ADPase--was mainly responsible for this phenomenon. CD39 rapidly and preferentially metabolizes ADP released from activated platelets. ADP is the final common pathway for platelet recruitment and thrombus formation, and platelet aggregation and recruitment are abolished by CD39. Our current hypothesis is that CD39 will be a novel antithrombotic agent for treating high risk patients who have activated platelets in their circulation--the identifying characteristic of coronary artery occlusion and thrombotic stroke. A recombinant, soluble form of human CD39 has been generated. This is solCD39, a glycosylated protein of 66 kDa whose enzymatic and biological properties are identical to the full-length form of the enzyme. In our in vitro experiments, solCD39 blocks ADP-induced human platelet aggregation, and inhibits collagen- and thrombin receptor agonist peptide-induced platelet reactivity. We studied solCD39 in vitro in a murine model of stroke, which was shown to be driven by excessive platelet recruitment. In studies with CD39 wild-type (CD39+/+) mice solCD39 completely abolished ADP-induced platelet aggregation, and strongly inhibited collagen- and arachidonate-induced platelet reactivity ex vivo. When solCD39 was administered prior to transient intraluminal middle cerebral artery occlusion, it reduced ipsilateral fibrin deposition, decreased (111)In-platelet deposition, and increased post-ischemic blood flow 2-fold at 24 hours. These results were superior to those we obtained with aspirin pre-treatment. CD39 null (CD39-/-) mice, which we generated by deletion of exons 4-6 (apyrase conserved regions 2-4), have a normal phenotype, normal hematologic profiles and bleeding times, but exhibit a decrease in post-ischemic perfusion and an increase in cerebral infarct volume when compared to genotypic CD39+/+ controls in our stroke model. "Reconstitution" of CD39 null mice with solCD39 reversed these pathologic changes. Thus, the CD39-/- mice were actually rescued from cerebral injury by solCD39, thereby fulfilling Koch's postulates. These experiments have led us to hypothesize that solCD39 has potential as a novel therapeutic agent for thrombotic stroke. In this review, we summarize our recent research results with CD39 and solCD39, and discuss our viewpoints on its present and future possibilities as a novel treatment for thrombosis.


Subject(s)
Apyrase/metabolism , Arterial Occlusive Diseases/metabolism , Blood Platelets/metabolism , Cell Communication/physiology , Animals , Antigens, CD , Apyrase/genetics , Apyrase/pharmacology , Blood Platelets/ultrastructure , Disease Models, Animal , Humans , Mice , Mutagenesis , Platelet Activation/drug effects , Platelet Aggregation/drug effects , Thrombosis/physiopathology
3.
Circulation ; 102(16): 1924-30, 2000 Oct 17.
Article in English | MEDLINE | ID: mdl-11034940

ABSTRACT

BACKGROUND: Fibrinogen binding to the active conformation of the alpha(IIb)beta(3) integrin receptor (glycoprotein IIb/IIIa) and cytoskeletal reorganization are important events in platelet function. Tyrosine phosphorylation of platelet proteins plays an essential role in platelet signal transduction pathways. We studied the participation of tyrosine kinases on these aspects of platelet reactivity and their importance in cyclooxygenase (COX)-1-independent mechanisms in thrombin-stimulated human platelets. METHODS AND RESULTS: Using washed platelets from normal donors and tyrphostin-A47 and aspirin as tyrosine kinase and COX-1 inhibitors, respectively, we found that tyrphostin-A47 downregulated (1) the thrombin-activated conformational change of alpha(IIb)beta(3), (2) actin polymerization and cytoskeletal reorganization, and (3) the quantity of tyrosine-phospho-rylated proteins associated with the reorganized cytoskeleton. The latter are important components of multimolecular signaling complexes. Concomitantly, platelet aggregation and secretion were significantly reduced. Aspirin did not affect receptor activation or tyrosine phosphorylation but did decrease the initial (30-second) burst of actin polymerization. Importantly, aspirin significantly amplified the inhibitory effect of tyrphostin-A47 on all aspects of platelet reactivity that we evaluated. CONCLUSIONS: Tyrosine protein phosphorylation is a regulatory control system of the inside-out mechanism of alpha(IIb)beta(3) activation and cytoskeletal assembly in thrombin-stimulated human platelets. Inhibition of these aspects of platelet function with tyrphostin-A47 is amplified when platelets are treated with aspirin. Therefore, tyrosine phosphorylation is a major component of early signaling events and of COX-1-independent mechanisms of thrombin-induced platelet reactivity. The study results may indicate a novel target for therapeutic intervention.


Subject(s)
Blood Platelets/metabolism , Cytoskeletal Proteins/metabolism , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Thrombin/metabolism , Aspirin/pharmacology , Blood Platelets/cytology , Blood Platelets/drug effects , Cyclooxygenase 1 , Down-Regulation , Enzyme Inhibitors/pharmacology , Humans , Isoenzymes/antagonists & inhibitors , Membrane Proteins , Phosphorylation/drug effects , Platelet Aggregation/drug effects , Prostaglandin-Endoperoxide Synthases , Protein-Tyrosine Kinases/antagonists & inhibitors , Signal Transduction/drug effects , Thrombin/pharmacology , Tyrosine/metabolism , Tyrphostins/pharmacology
4.
Biochemistry ; 39(23): 6936-43, 2000 Jun 13.
Article in English | MEDLINE | ID: mdl-10841775

ABSTRACT

Endothelial cell CD39/ecto-ADPase plays a major role in vascular homeostasis. It rapidly metabolizes ADP released from stimulated platelets, thereby preventing further platelet activation and recruitment. We recently developed a recombinant, soluble form of human CD39, solCD39, with enzymatic and biological properties identical to CD39. To identify amino acids essential for enzymatic/biological activity, we performed site-directed mutagenesis within the four highly conserved apyrase regions of solCD39. Mutation of glutamate 174 to alanine (E174A) and serine 218 to alanine (S218A) resulted in complete and approximately 90% loss of solCD39 enzymatic activity, respectively. Furthermore, compared to wild-type, S57A exhibited a 2-fold increase in ADPase activity without change in ATPase activity, while the tyrosine 127 to alanine (Y127A) mutant lost 50-60% of both ADPase and ATPase activity. The ADPase activity of wild-type solCD39 and each mutant, except for R135A, was greater with calcium as the required divalent cation than with magnesium, but for ATPase activity generally no such preference was observed. Y127A demonstrated the highest calcium/magnesium ADPase activity ratio, 2.8-fold higher than that of wild-type, even though its enzyme activity was greatly reduced. SolCD39 mutants were further characterized by correlating enzymatic with biological activity in an in vitro platelet aggregation system. Each solCD39 mutant was similar to wild-type in reversing platelet aggregation, except for E174A and S218A. E174A, completely devoid of enzymatic activity, failed to inhibit platelet responsiveness, as anticipated. S218A, with 91% loss of ADPase activity, could still reverse platelet aggregation, albeit much less effectively than wild-type solCD39. Thus, glutamate 174 and serine 218 are essential for both the enzymatic and biological activity of solCD39.


Subject(s)
Antigens, CD/genetics , Apyrase/genetics , Blood Platelets/metabolism , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Amino Acid Sequence , Antigens, CD/metabolism , Apyrase/metabolism , Cations, Divalent/metabolism , Conserved Sequence , Endopeptidases/metabolism , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , Nephelometry and Turbidimetry , Platelet Aggregation/genetics , Protein Conformation , Recombinant Proteins , Solubility
6.
J Clin Invest ; 101(9): 1851-9, 1998 May 01.
Article in English | MEDLINE | ID: mdl-9576748

ABSTRACT

Excessive platelet accumulation and recruitment, leading to vessel occlusion at sites of vascular injury, present major therapeutic challenges in cardiovascular medicine. Endothelial cell CD39, an ecto-enzyme with ADPase and ATPase activities, rapidly metabolizes ATP and ADP released from activated platelets, thereby abolishing recruitment. Therefore, a soluble form of CD39, retaining nucleotidase activities, would constitute a novel antithrombotic agent. We designed a recombinant, soluble form of human CD39, and isolated it from conditioned media from transiently transfected COS-1 cells and from stably transfected Chinese hamster ovary (CHO) cells. Conditioned medium from CHO cells grown under serum-free conditions was subjected to anti-CD39 immunoaffinity column chromatography, yielding a single approximately 66-kD protein with ATPase and ADPase activities. Purified soluble CD39 blocked ADP-induced platelet aggregation in vitro, and inhibited collagen-induced platelet reactivity. Kinetic analyses indicated that, while soluble CD39 had a Km for ADP of 5.9 microM and for ATP of 2.1 microM, the specificity constant kcat/Km was the same for both substrates. Intravenously administered soluble CD39 remained active in mice for an extended period of time, with an elimination phase half-life of almost 2 d. The data indicate that soluble CD39 is a potential therapeutic agent for inhibition of platelet-mediated thrombotic diatheses.


Subject(s)
Adenosine Triphosphatases , Antigens, CD/pharmacology , Apyrase/pharmacokinetics , Blood Platelets/drug effects , Fibrinolytic Agents/pharmacokinetics , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Animals , Antigens, CD/genetics , Apyrase/genetics , CHO Cells , COS Cells , Chromatography, Affinity , Cricetinae , Half-Life , Humans , Mice , Mice, Inbred BALB C , Recombinant Proteins/pharmacokinetics , Solubility , Thromboembolism/prevention & control
7.
Circulation ; 97(4): 350-5, 1998 Feb 03.
Article in English | MEDLINE | ID: mdl-9468208

ABSTRACT

BACKGROUND: Aspirin (acetylsalicylic acid, ASA) is widely used for secondary prevention of ischemic vascular events, although its protection only occurs in 25% of patients. We previously demonstrated that platelet reactivity is enhanced by a prothrombotic effect of erythrocytes in a thromboxane-independent manner. This diminishes the antithrombotic therapeutic potential of ASA. Recent data from our laboratory indicate that the prothrombotic effect of erythrocytes also contains an ASA-sensitive component. In accordance with this observation, intermittent treatment with high-dose ASA reduced the prothrombotic effects of erythrocytes ex vivo in healthy volunteers. In the present study, the effects of platelet-erythrocyte interactions were evaluated ex vivo in 82 patients with vascular disease: 62 patients with ischemic heart disease treated with 200 mg ASA/d and 20 patients with ischemic stroke treated with 300 mg ASA/d. METHODS AND RESULTS: Platelet activation (release reaction) and platelet recruitment (fluid-phase proaggregatory activity of cell-free releasates from activated platelets) were assessed after collagen stimulation (1 microg/mL) of platelets, platelet-erythrocyte mixtures, or whole blood. Platelet thromboxane A2 synthesis was inhibited by >94% by ASA administration in all patients. Importantly, platelet recruitment followed one of three distinct patterns. In group A (n=32; 39%), platelet recruitment was blocked by ASA both in the presence and absence of erythrocytes. In group B (n=37; 45%), recruitment was abolished when platelets were evaluated alone but continued in the presence of erythrocytes, indicating a suboptimal effect of ASA on erythrocytes of this patient group. In group C (n= 13; 16%), detectable recruitment in stimulated platelets alone persisted and was markedly enhanced by the presence of erythrocytes. CONCLUSIONS: In two thirds of a group of patients with vascular disease, 200 to 300 mg ASA was insufficient to block platelet reactivity in the presence of erythrocytes despite abolishing thromboxane A2 synthesis. Platelet activation in the presence of erythrocytes can induce the release reaction and generate biologically active products that recruit additional platelets into a developing thrombus. Insufficient blockade of this proaggregatory property of erythrocytes can lead to development of additional ischemic complications.


Subject(s)
Aspirin/therapeutic use , Blood Platelets/physiology , Erythrocytes/physiology , Fibrinolytic Agents/therapeutic use , Platelet Aggregation Inhibitors/therapeutic use , Vascular Diseases/drug therapy , Vascular Diseases/etiology , Adult , Aged , Aged, 80 and over , Aspirin/administration & dosage , Blood Platelets/metabolism , Brain Ischemia/drug therapy , Cerebrovascular Disorders/drug therapy , Dose-Response Relationship, Drug , Erythrocytes/drug effects , Female , Humans , Male , Middle Aged , Myocardial Ischemia/drug therapy , Platelet Aggregation Inhibitors/administration & dosage , Serotonin/metabolism , Vascular Diseases/blood
8.
J Clin Invest ; 99(6): 1351-60, 1997 Mar 15.
Article in English | MEDLINE | ID: mdl-9077545

ABSTRACT

We previously demonstrated that when platelets are in motion and in proximity to endothelial cells, they become unresponsive to agonists (Marcus, A.J., L.B. Safier, K.A. Hajjar, H.L. Ullman, N. Islam, M.J. Broekman, and A.M. Eiroa. 1991. J. Clin. Invest. 88:1690-1696). This inhibition is due to an ecto-ADPase on the surface of endothelial cells which metabolizes ADP released from activated platelets, resulting in blockade of the aggregation response. Human umbilical vein endothelial cells (HUVEC) ADPase was biochemically classified as an E-type ATP-diphosphohydrolase. The endothelial ecto-ADPase is herein identified as CD39, a molecule originally characterized as a lymphoid surface antigen. All HUVEC ecto-ADPase activity was immunoprecipitated by monoclonal antibodies to CD39. Surface localization of HUVEC CD39 was established by confocal microscopy and flow cytometric analyses. Transfection of COS cells with human CD39 resulted in both ecto-ADPase activity as well as surface expression of CD39. PCR analyses of cDNA obtained from HUVEC mRNA and recombinant human CD39 revealed products of the same size, and of identical sequence. Northern blot analyses demonstrated that HUVEC express the same sized transcripts for CD39 as MP-1 cells (from which CD39 was originally cloned). We established the role of CD39 as a prime endothelial thromboregulator by demonstrating that CD39-transfected COS cells acquired the ability to inhibit ADP-induced aggregation in platelet-rich plasma. The identification of HUVEC ADPase/CD39 as a constitutively expressed potent inhibitor of platelet reactivity offers new prospects for antithrombotic therapeusis.


Subject(s)
Adenosine Triphosphatases , Antigens, CD/pharmacology , Apyrase/pharmacology , Endothelium, Vascular/enzymology , Platelet Aggregation Inhibitors/pharmacology , Animals , Antibodies, Monoclonal/pharmacology , Antigens, CD/chemistry , Antigens, CD/genetics , Antigens, CD/immunology , Apyrase/chemistry , Apyrase/immunology , COS Cells , Cells, Cultured , DNA, Complementary/analysis , Endothelium, Vascular/cytology , Enzyme Activation/immunology , Humans , Intracellular Membranes/enzymology , Microsomes/enzymology , Platelet Aggregation Inhibitors/immunology , Precipitin Tests , RNA, Messenger/analysis , Recombinant Proteins/analysis , Transfection , Umbilical Veins
9.
Circulation ; 95(1): 63-8, 1997 Jan 07.
Article in English | MEDLINE | ID: mdl-8994418

ABSTRACT

BACKGROUND: Aspirin effectively reduces the incidence of secondary vascular occlusive events in only 25% of patients. Low-dose aspirin as currently used blocks platelet production of prothrombotic thromboxane A2 and allows endothelial synthesis of antithrombotic prostacyclin. This regimen minimizes gastrointestinal toxicity. We previously showed that intact erythrocytes markedly enhance platelet reactivity. Therefore we investigated whether supplementation of low-dose aspirin with a single high dose at 2-week intervals could more effectively block erythrocyte promotion of platelet reactivity. METHODS AND RESULTS: Effects of different aspirin regimens on erythrocyte enhancement of platelet reactivity in normal volunteers were measured with the use of an assay that evaluates both platelet activation and recruitment. After 15 days of daily ingestion of 50 mg aspirin, reactivity of platelets alone was inhibited. However, erythrocyte promotion of platelet activation and recruitment was only inhibited by approximately 50% and persisted in the total absence of thromboxane synthesis. In contrast, if 50 mg/d aspirin was preceded by a single loading dose of 500 mg aspirin, the erythrocyte prothrombotic effect was strongly inhibited (approximately 90%) for 2 to 3 weeks. However, over time, erythrocytes "escaped" from this inhibition, and once again became prothrombotic, even on a daily regimen of 50 mg aspirin. CONCLUSIONS: For clinical purposes, we recommend a loading dose of aspirin (500 mg), followed by daily administration of 50 mg. The loading dose should be repeated at 2-week intervals. This regimen blocks recovery of the erythrocyte capacity to promote platelet reactivity and may amplify the therapeutic potential of aspirin in cardiovascular disease.


Subject(s)
Aspirin/pharmacology , Blood Platelets/physiology , Erythrocytes/drug effects , Adult , Aspirin/administration & dosage , Blood Platelets/metabolism , Drug Administration Schedule , Erythrocytes/physiology , Female , Humans , Male , Middle Aged , Platelet Activation/drug effects , Serotonin/metabolism , Thromboxanes/biosynthesis , Time Factors
10.
Blood ; 87(1): 331-40, 1996 Jan 01.
Article in English | MEDLINE | ID: mdl-8547660

ABSTRACT

Lipoxygenases (LXs) catalyze formation of leukotrienes and hydroxy-eicosatetraenoic acids (HETEs), proinflammatory, and spasmogenic autacoids that are critical for host defense systems. We studied the expression and regulation of LX genes (12-LX, 5-LX, and 15-LX) and the 5-lipoxygenase activating protein (FLAP) in human mononuclear cells (MNC) and granulocytes using a quantitative reverse transcription polymerase chain reaction (RT-PCR) technique. We show that 12-LX mRNA is constitutively expressed in resting platelet-free MNC. 12-LX gene expression was upregulated by activation with lipopolysaccharide (LPS). The formation of 12-HETE was inducible with ionophore in MNC, as assessed by high-performance liquid chromatography (HPLC) and gas chromatography, and increased after LPS pretreatment. In addition to 12-LX, resting MNC expressed the genes for 5-LX and FLAP constitutively. Quantitative time course analyses of 12-LX, 5-LX, and FLAP gene expression suggested coregulation of 12-LX and FLAP mRNAs, and reciprocal regulation of 5-LX and FLAP mRNAs. During cell stimulation with LPS 5-LX mRNA levels remained unchanged, whereas FLAP gene expression increased. No 15-LX mRNA expression or 15-HETE formation was detectable in unstimulated and activated MNC. In contrast to MNC, quantitative RT-PCR mRNA analysis showed intermittent intraindividual expression of the 5-LX and FLAP genes in resting granulocytes. mRNAs for 12-LX and 15-LX were not expressed. On stimulation of granulocytes ex vivo, mRNA expression of 5-LX and FLAP was upregulated. Stimulation by LPS differed from that by ionophore A23187. Neither LPS nor ionophore induced gene expression of 12-LX or 15-LX in granulocytes. Our data indicate that resting human MNC and granulocytes express LX and FLAP genes in a cell-specific manner. Cell activation induces coordinated upregulation of 12-LX and FLAP genes in MNC, and 5-LX and FLAP genes in granulocytes, respectively. The constitutive expression of 12-LX mRNA, its upregulation on cell activation, and the formation of 12-HETE clearly indicate the presence of a functional 12-LX in human MNC.


Subject(s)
Arachidonate 12-Lipoxygenase/biosynthesis , Carrier Proteins/biosynthesis , Gene Expression Regulation , Granulocytes/enzymology , Leukocytes, Mononuclear/enzymology , Membrane Proteins/biosynthesis , RNA, Messenger/biosynthesis , 12-Hydroxy-5,8,10,14-eicosatetraenoic Acid , 5-Lipoxygenase-Activating Proteins , Arachidonate 12-Lipoxygenase/genetics , Base Sequence , Carrier Proteins/genetics , DNA, Complementary/genetics , Enzyme Induction , Humans , Hydroxyeicosatetraenoic Acids/biosynthesis , Lymphocyte Activation , Membrane Proteins/genetics , Molecular Sequence Data , Polymerase Chain Reaction , RNA, Messenger/genetics
12.
Thromb Haemost ; 74(1): 213-7, 1995 Jul.
Article in English | MEDLINE | ID: mdl-8578460

ABSTRACT

Platelet activation as a result of vascular injury provokes endothelial cells to respond in a manner which limits or reverses the occlusive consequences of platelet accumulation. If the agonistic forces are strong, platelet accumulation is irreversible. In vitro data from our laboratory have repeatedly demonstrated that platelets become unresponsive to all agonists when in proximity to endothelial cells. This unresponsiveness is due to at least three separate endothelial "thromboregulatory" systems: eicosanoids, endothelium-derived relaxing factor (EDRF/NO), and most importantly an endothelial cell ecto-nucleotidase which metabolizes released platelet adenosine diphosphate (ADP) with consequent restoration of platelets to the resting state. This nucleotidase is operative in the complete absence of EDRF/NO and eicosanoids, indicating that the latter two are dispensable thromboregulators. We have solubilized the human endothelial cell ectoADPase, as well as that from placental tissue. Candidate proteins from a purified ADPase fraction are now being studied in further detail. An understanding of the molecular biology of the ADPase gene may lead to development of therapeutic agents such as soluble forms of the enzyme as well as approaches toward up-regulation of ectoADPase activity. This could result in "early thromboregulation", i.e. prevention and/or reversal of platelet accumulation at sites of vascular damage via immediate metabolic removal of the prime platelet agonist-ADP.


Subject(s)
Inflammation/pathology , Thrombosis/pathology , Adenosine Diphosphate/physiology , Apyrase/physiology , Aspirin/pharmacology , Aspirin/therapeutic use , Blood Platelets/drug effects , Blood Platelets/pathology , Cyclooxygenase Inhibitors/pharmacology , Cyclooxygenase Inhibitors/therapeutic use , Eicosanoids/physiology , Endothelium, Vascular/injuries , Endothelium, Vascular/pathology , HSP70 Heat-Shock Proteins/physiology , Humans , Inflammation/drug therapy , Nitric Oxide/physiology , Platelet Activation/drug effects , Platelet Activation/physiology , Thrombosis/drug therapy
13.
J Biol Chem ; 270(14): 8179-87, 1995 Apr 07.
Article in English | MEDLINE | ID: mdl-7713923

ABSTRACT

Coagulation Factor IX/IXa has been shown to bind to cellular surfaces, and Factor IXa expresses its procoagulant activity by assembling into the intrinsic Factor X activating complex (Factors IXa/VIIIa/X), which also forms on membrane surfaces. This led us to identify cellular proteins which bind Factor IX/IXa; an approximately 55-kDa polypeptide was purified to homogeneity from bovine lung extracts based on its capacity to bind 125I-Factor IX in a dose-dependent and saturable manner. From protein sequence data of the amino terminus and internal peptides, the approximately 55-kDa polypeptide was identified as calreticulin, a previously identified intracellular calcium-binding protein. Recombinant calreticulin bound vitamin K-dependent coagulation factors, 125I-Factor IX, 125I-Factor X, and 125I-prothrombin (Kd values of approximately 2.7, 3.2, and 8.3 nM, respectively), via interaction with its C-domain, although it did not affect the coagulant properties of these proteins. 125I-Calreticulin also bound to endothelial cells in vitro (Kd approximately 7.4 nM), and mouse infusion studies showed an initial rapid phase of clearance in which calreticulin could be localized on the vascular endothelium. Exposure of endothelial cells to calreticulin led to dose-dependent, immediate, and sustained increase in the production of nitric oxide, as measured using a porphyrinic microsensor. In a canine electrically induced thrombosis model, intracoronary infusion of calreticulin (n = 7) prevented occlusion of the left circumflex coronary artery in a dose-dependent manner compared with vehicle-treated controls (n = 5). These results indicate that calreticulin interacts with the endothelium to stimulate release of nitric oxide and inhibit clot formation.


Subject(s)
Blood Coagulation Factors/metabolism , Calcium-Binding Proteins/pharmacology , Coronary Thrombosis/prevention & control , Endothelium, Vascular/drug effects , Nitric Oxide/biosynthesis , Ribonucleoproteins/pharmacology , Animals , Calcium-Binding Proteins/metabolism , Calreticulin , Cattle , Dogs , Endothelium, Vascular/metabolism , Lung/metabolism , Mice , Models, Biological , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Ribonucleoproteins/metabolism , Vitamin K/metabolism
15.
J Clin Invest ; 93(5): 2291-7, 1994 May.
Article in English | MEDLINE | ID: mdl-7514195

ABSTRACT

Nitric oxide (NO) is a novel biologic messenger with diverse effects but its role in organ transplantation remains poorly understood. Using a porphyrinic microsensor, the first direct measurements of coronary vascular and endocardial NO production were made. NO was measured directly in the effluent of preserved, heterotopically transplanted rat hearts stimulated with L-arginine and bradykinin; NO concentrations fell from 2.1 +/- 0.4 microM for freshly explanted hearts to 0.7 +/- 0.2 and 0.2 +/- 0.08 microM for hearts preserved for 19 and 38 h, respectively. NO levels were increased by SOD, suggesting a role for superoxide-mediated destruction of NO. Consistent with these data, addition of the NO donor nitroglycerin (NTG) to a balanced salt preservation solution enhanced graft survival in a time- and dose-dependent manner, with 92% of hearts supplemented with NTG surviving 12 h of preservation versus only 17% in its absence. NTG similarly enhanced preservation of hearts stored in University of Wisconsin solution, the clinical standard for preservation. Other stimulators of the NO pathway, including nitroprusside, L-arginine, or 8-bromoguanosine 3',5' monophosphate, also enhanced graft survival, whereas the competitive NO synthase antagonist NG-monomethyl-L-arginine was associated with poor preservation. Likely mechanisms whereby supplementation of the NO pathway enhanced preservation included increased blood flow to the reperfused graft and decreased graft leukostasis. NO was also measured in endothelial cells subjected to hypoxia/reoxygenation and detected based on its ability to inhibit thrombin-mediated platelet aggregation and serotonin release. NO became undetectable in endothelial cells exposed to hypoxia followed by reoxygenation and was restored to normoxic levels on addition of SOD. These studies suggest that the NO pathway fails during preservation/transplantation because of formation of oxygen free radicals during reperfusion, which quench available NO. Augmentation of NO/cGMP-dependent mechanisms enhances vascular function after ischemia and reperfusion and provides a new strategy for transplantation of vascular organs.


Subject(s)
Coronary Vessels/metabolism , Endocardium/metabolism , Heart Transplantation/physiology , Nitric Oxide/biosynthesis , Transplantation, Heterotopic/physiology , Amino Acid Oxidoreductases/analysis , Animals , Arginine/pharmacology , Biosensing Techniques , Bradykinin/pharmacology , Cyclic GMP/analogs & derivatives , Cyclic GMP/pharmacology , Graft Survival , Heart/drug effects , Male , Nitric Oxide Synthase , Nitroglycerin/pharmacology , Nitroprusside/pharmacology , Organ Preservation , Rats , Rats, Sprague-Dawley , Superoxide Dismutase/metabolism
16.
J Clin Invest ; 92(3): 1357-65, 1993 Sep.
Article in English | MEDLINE | ID: mdl-7690778

ABSTRACT

Unstimulated neutrophils inhibited activation and recruitment of thrombin- or collagen-stimulated platelets in an agonist-specific manner. This occurred under conditions of close physical cell-cell contact, although biochemical adhesion between the cells as mediated by P-selectin was not required. Moreover, in the presence of monoclonal P-selectin antibodies that blocked biochemical platelet-neutrophil adhesion, thrombin-stimulated platelets were more efficiently downregulated by neutrophils. This suggested a prothrombotic role for P-selectin under these circumstances. The neutrophil downregulatory effect on thrombin-stimulated platelets was amplified by lipoxygenase inhibition with 5,8,11,14-eicosatetraynoic acid. In contrast, the neutrophil inhibitory effect on platelets was markedly reduced by platelet-derived 12S-hydroxy-5,8-cis, 10-trans, 14-cis-eicosatetraenoic acid (12S-HETE), as well as by the platelet-neutrophil transcellular product, 12S,20-dihydroxy-5,8,10,14-eicosatetraenoic acid (12S,20-DiHETE), but not by another comparable metabolite, 5S,12S-dihydroxy-6-trans, 8-cis, 10-trans, 14-cis-eicosatetraenoic acid (5S,12S-DiHETE), or the neutrophil-derived hydroxy acid leukotriene B4. The neutrophil downregulatory effect on thrombin-induced platelet reactivity was enhanced by aspirin treatment. This may represent a novel action of aspirin as an inhibitor of platelet function. These results provide in vitro biochemical and functional evidence for the thromboregulatory role of neutrophils and emphasize the multicellular aspect of hemostasis and thrombosis.


Subject(s)
Blood Platelets/physiology , Cell Adhesion Molecules/physiology , Eicosanoids/physiology , Neutrophils/physiology , Adenosine Diphosphate/metabolism , Cell Adhesion , Collagen/pharmacology , Humans , In Vitro Techniques , Lipoxygenase/metabolism , Nitric Oxide/metabolism , P-Selectin , Platelet Activation , Platelet Membrane Glycoproteins/physiology , Serine Endopeptidases/metabolism , Serotonin/metabolism , Thrombin/pharmacology
17.
Biochim Biophys Acta ; 1166(1): 20-4, 1993 Feb 10.
Article in English | MEDLINE | ID: mdl-8381674

ABSTRACT

Addition of n-3 fatty acids to a human diet for more than 3 weeks lowers levels of the powerful proinflammatory compound, leukotriene (LT) B4. This can be shown ex vivo after stimulation of human granulocytes with ionophore A23187. In a controlled, randomized, observer-blind study in 14 human volunteers, we investigated the effect of adding 7 g/day of a 85% n-3 fatty acid concentrate to the diet of 7 volunteers (7 served as controls). Levels of LTB4, 20-OH-LTB4, 20-COOH-LTB4 as well as LTB5, 20-OH-LTB5 and 20-COOH-LTB5 were measured by high-pressure liquid chromatography (HPLC) after stimulation and extraction of a platelet-free granulocyte preparation (92% neutrophils). LTB5 and 20-COOH-LTB5 were only detected during n-3 fatty acids, when 20-OH-LTB5 increased from trace amounts to substantial quantities. Importantly, levels of catabolites of LTB4, i.e., 20-OH-LTB4 and 20-COOH-LTB4 were not significantly altered throughout the study. However, the level of LTB4 itself was reduced dramatically after 6 weeks (less so after 1 week) of dietary n-3 fatty acid administration. These data demonstrate that during dietary n-3 fatty acids levels of LTB4 are lowered by a combination of accelerated catabolism and diminished LTB4 generation. This newly observed mechanism of increased LT catabolism may be mediated via induction of peroxisomal enzymes catabolizing leukotrienes B.


Subject(s)
Dietary Fats, Unsaturated/pharmacology , Fatty Acids, Omega-3/pharmacology , Leukotriene B4/metabolism , Adult , Humans , Male , Time Factors
19.
J Clin Invest ; 88(5): 1690-6, 1991 Nov.
Article in English | MEDLINE | ID: mdl-1939654

ABSTRACT

We previously reported that platelets become unresponsive to agonists when stimulated in combined suspension with aspirin-treated human umbilical vein endothelial cells. Inhibition occurred concomitant with metabolism of platelet-derived endoperoxides to prostacyclin by endothelial cells. We now demonstrate that if aspirin-treated platelets which fully respond to appropriate doses of agonists are exposed to aspirin-treated endothelial cells, they remain unresponsive despite absence of prostacyclin. Platelet inhibition is due in large part to ecto-ADPase activity on the endothelial cells. This was established by incubating aspirin-treated endothelial cells with 14C-ADP. Radio-thin layer chromatography and aggregometry demonstrated that 14C-ADP and induction of platelet activation decreased rapidly and concurrently. AMP accumulated transiently, was further metabolized to adenosine, and deaminated to inosine. The apparent Km of the endothelial cell ADPase was 33-42 microM and the Vmax 17-43 nmol/min per 10(6) cells, values in the range of antithrombotic potential. Thus, at least three complementary systems in human endothelial cells control platelet responsiveness: a cell-associated, aspirin-insensitive ADPase which functions in parallel with fluid phase autacoids such as the aspirin-inhibitable eicosanoids, and the aspirin-insensitive endothelium-derived relaxing factor.


Subject(s)
Apyrase/physiology , Aspirin/pharmacology , Blood Platelets/drug effects , Endothelium, Vascular/physiology , Platelet Aggregation Inhibitors/pharmacology , Adenosine Diphosphate/physiology , Apyrase/analysis , Blood Platelets/physiology , Humans , In Vitro Techniques , Nitric Oxide/physiology , Platelet Aggregation/drug effects , Thrombin/pharmacology
20.
Blood ; 78(4): 1033-40, 1991 Aug 15.
Article in English | MEDLINE | ID: mdl-1868238

ABSTRACT

To determine a role for endothelium-derived relaxing factor/nitric oxide (EDRF/NO) in regulation of human platelet reactivity by human endothelial cells (EC), we studied combined suspensions of human umbilical vein endothelial cells (HU-VEC, passage 2 through 3) and washed human platelets. Confluent HUVEC monolayers were treated with aspirin (1 mmol/L) to prevent prostacyclin (PGI2) formation, washed, and harvested. Aspirin-treated platelets alone (58 x 10(6)) were fully aggregated by thrombin at 0.05 U/mL or more. In the presence of 10(6) HUVEC, however, platelet serotonin release and aggregation in response to thrombin at doses as high as 0.5 U/mL were blocked. We demonstrated for the first time that inhibition of aggregation and serotonin release, due to EDRF/NO, occurred in parallel. HUVEC-dependent inhibition of platelet responsiveness was enhanced by superoxide dismutase (SOD) and reversed by hemoglobin. The inhibitory effect was also reversed by preincubation of HUVEC with NG-monomethyl-L-arginine (NMA) or NG-nitro-L-arginine (NNA) through competitive blockade of arginine metabolism. Pretreatment of platelets with methylene blue indicated that EC-dependent inhibition of platelet reactivity occurred through activation of platelet soluble guanylate cyclase. When platelets and HUVEC were separated by a permeable membrane and both cells were stimulated by thrombin, platelets remained unresponsive. This indicated that inhibition was induced by a fluid-phase mediator, independent of direct cell-cell contact. These data demonstrate that EDRF/NO formation from L-arginine by human EC plays an important role as an aspirin-insensitive fluid-phase inhibitor of human platelet reactivity.


Subject(s)
Aspirin/pharmacology , Blood Platelets/physiology , Endothelium, Vascular/metabolism , Nitric Oxide/physiology , Platelet Aggregation , Serotonin/metabolism , Arginine/analogs & derivatives , Arginine/pharmacology , Blood Platelets/drug effects , Humans , Nitric Oxide/metabolism , Platelet Aggregation/drug effects , Superoxide Dismutase/pharmacology , Thrombin/pharmacology , Umbilical Veins , omega-N-Methylarginine
SELECTION OF CITATIONS
SEARCH DETAIL
...