Inhibition of Platelet Function by the Endothelium

Inhibition of Platelet Function by the Endothelium

C H A P T E R 17 Inhibition of Platelet Function by the Endothelium Lea M. Beaulieu and Jane E. Freedman Department of Medicine, University of Massac...

853KB Sizes 33 Downloads 119 Views

C H A P T E R

17 Inhibition of Platelet Function by the Endothelium Lea M. Beaulieu and Jane E. Freedman Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts

I. INTRODUCTION Cellcell interactions, environmental factors, and cytokine/hormone concentrations play a major role in modulating the process of hemostasis and thrombus formation after vessel injury. A fine balance between prothrombotic and antithrombotic processes must be achieved to prevent either hemorrhage or thrombosis. During the activation, adhesion, and aggregation of platelets at the site of injury, the endothelium responds by limiting the size and growth of the hemostatic plug or thrombus or even reversing platelet reactivity. These responses are referred to as endothelial thromboregulation13 and require several forms of communication between endothelial cells and platelets. There are three primary (and functionally independent) pathways during the early stages of thromboregulation (Fig. 17-1 and Table 17-1):14 (1) nitric oxide (NO); (2) the eicosanoid prostacyclin; and (3) the ectonucleotidase CD39. In this chapter, these three thromboregulatory mechanisms are discussed in detail. In particular, this chapter highlights the enzymes, receptors, and signaling pathways involved and discusses animal models, clinical investigations, and the role of these pathways in disease development, including relevant genetic variants.

II. NITRIC OXIDE A. Biosynthesis of NO and Characteristics of Endothelial NO Synthase NO, a well-characterized platelet inhibitor and vasodilator, was first discovered in endothelial cells by Furchgott and Zawadzki in 1980.5 NO can exist in

Platelets, 3rd edition

three interrelated redox forms that expand the biological spectrum of NO: the free radical NO, the nitrosonium anion (NO1 ), and the nitrosyl anion (NO).6,7 NO is inactivated by superoxide8 but stabilized by superoxide dismutase.9,10 It can also bind to heme-containing proteins, such as hemoglobin, to activate or inhibit their enzymatic activity.7 The biosynthesis of NO is carried out by a family of enzymes called nitric oxide synthases (NOS). The family consists of three members: endothelial NOS (eNOS), inducible NOS (iNOS), and neuronal NOS (nNOS). Several recent reports also describe the existence of a distinct form of mitochondrial NOS (mtNOS).1114 eNOS is expressed in endothelial cells,15,16 cardiomyocytes,17 bronchiolar epithelial cells,18,19 megakaryocytes, and platelets.20,21 The basal level of NO released from platelets has been reported to be similar to that from endothelial cells.22 eNOS is composed of an N-terminal oxygenase domain and a C-terminal reductase domain. The oxygenase domain—which contains binding sites for heme, tetrahydrobiopterin (BH4), and the substrate L-arginine—is linked by a calmodulin-recognition site to the reductase domain, which has binding sites for flavin adenine dinucleotide, flavin mononucleotide, and NADPH.23 The dimerization interface includes binding sites for BH4 and the Zn21-binding heme,2426 which is mandatory for dimerization. An “N-terminal hook” domain,26 as well as the presence of L-arginine, further stabilize the dimerization process.23 eNOS catalyzes the multi-electron oxidation reaction of L-arginine with oxygen in a Ca21-dependent manner, forming L-citrulline and releasing NO.27 Intracellular localization, posttranslational modifications, and enzymatic activity of eNOS are tightly

313

© 2013 Elsevier Inc. All rights reserved.

314

17. INHIBITION OF PLATELET FUNCTION BY THE ENDOTHELIUM

Nitric Oxide

Prostacyclin

cGMP sGC cAMP

CD39

cAMP IP receptor

NO

PGI2 eNOS

cAMP A2 Receptor ADP

Adenosine AMP

CD39

eNOS

CD73

PGIS PGIS

ER

FIGURE 17-1 Overview of the 3 mechanisms utilized by endothelial cells to inhibit platelets. Nitric Oxide: Endothelial nitric oxide synthase (eNOS), located in the caveolae, releases nitric oxide (NO). Upon diffusion across the platelet plasma membrane, NO stimulates soluble guanylyl cyclase (sGC), which leads to the upregulation of cyclic guanosine monophosphate (cGMP) and cyclic adenosine monophosphate (cAMP). Activated platelets will also produce NO through eNOS expressed on their cell surface. Prostacyclin: Prostacyclin synthase (PGIS), located on the endoplasmic reticulum (ER) and in the caveolae of the endothelial cells, releases prostacyclin (PGI2). On the platelet cell surface, prostacyclin receptor (IP receptor) binds PGI2, which leads to the increase in cellular cAMP. CD39: Adenosine diphosphate (ADP) that is released from activated platelets is hydrolyzed into adenosine monophosphate (AMP) on the endothelial cell surface by CD39. AMP is then converted into adenosine by CD73, also present on the endothelial cell surface. Adenosine will bind the platelet adenosine receptor (A2 receptor) and increase cAMP levels. All of these processes will lead to the inhibition of platelet aggregation and adhesion, controlling the size of the clot to allow for proper blood flow and healing of the endothelium.

regulated. In the endoplasmic reticulum, eNOS becomes irreversibly myristoylated at the N-terminal residue Gly2, whereas reversible palmitoylation at residues Cys15 and Cys26 takes place in the Golgi apparatus. Both acylation processes are crucial for the optimal targeting of eNOS to the plasma membrane.2830 In the plasma membrane, eNOS is found to be highly enriched within caveolae,31 which are specialized membrane invaginations rich in signaling molecules and coated with the protein caveolin.32,33 The enzymatic activity is sevenfold higher in membrane-associated eNOS than cytosolic.33 In the plasma membrane, its activity is about tenfold greater in the caveolae fraction as compared to the whole plasma membrane.33 It has been reported that eNOS binds to the scaffolding domain of caveolin, which has an inhibitory effect on eNOS activity in vivo.34 The activity of eNOS is regulated by several other mechanisms, such as changes in intracellular calcium concentration, phosphorylation, and dephosphorylation at various tyrosine, serine and

threonine residues, as well as the association or dissociation of eNOS-interacting proteins (reviewed in Refs. 23, 30, and 35). Mechanical stimuli, such as shear flow in the blood vessel, can activate eNOS36 by Akt-mediated phosphorylation at Ser1179,37,38 leading to NO production and vasodilation. Estrogen also affects eNOS function. 17β-estradiol increases eNOS expression and activity (reviewed in Ref. 39), which can be inhibited by progestins40 and medroxyprogesterone acetate.41 Estrogen replacement therapy should therefore be beneficial by increasing eNOS levels and NO production. However, clinical studies have shown that there is an increased risk for DVT in postmenopausal women under hormone replacement therapy. A new aminoestrogen with anticoagulant properties, prolame, has been tested in vitro and in mouse models and has shown to induce NO production and reduced thrombi production in a thrombosis model.42 Conversely, under conditions such as the absence of L-arginine or BH4, or in the presence of the eNOS

I. PLATELET BIOLOGY

315

II. NITRIC OXIDE

TABLE 17-1

Endothelial-derived Mediators of Platelet Activation

Class of Thromboregulator

Nitrovasodilator

Eicosanoid

Ecto-nucleotidase

Type of Thromboregulator

NO

Prostacyclin

CD39 and CD73

Site of Action

Released into blood stream

Released into blood stream

ADP/ATP removed from blood stream; adenosine released into blood stream

Levels

Basal levels, increased upon stimulation

Induced upon stimulation

Basal levels

Endothelial Thromboregulating Enzyme

eNOS

Prostacyclin synthase (PGIS)

CD39, CD73

Platelet Receptor for Endothelial Thromboregulator

NO diffusion across membrane, sGC

Prostacyclin receptor (IP receptor)

Adenosine receptor (A2)

Thromboregulatory Mechanism to Inhibit Platelet Reactivity

Upregulation of platelet cGMP production

Upregulation of platelet cAMP production

Enzymatic removal of ADP secreted from activated platelets; adenosine-induced upregulation of platelet cAMP

Platelet “Auto-regulation”

Platelet eNOS (NO production), superoxide, ROS

TXA2 synthesis

ADP/ATP purinergic receptors; CD39 (low expression)

Sensitivity to Aspirin

No

Yes

No

Abbreviations: ADP, adenosine diphosphate; ATP, adenosine triphosphate; cAMP, cyclic adenosine monophosphate; cGMP, cyclic guanosine 5’-monophosphate; eNOS, endothelial nitric oxide synthase; NO, nitric oxide; PGI2, prostaglandin I2; ROS, reactive oxygen species; sGC, soluble guanylyl cyclase; TXA2, thromboxane A2.

inhibitor NG-monomethyl-L-arginine (L-NMMA), eNOS can undergo a process called eNOS uncoupling whereby eNOS catalyzes an uncoupled NADPH oxidation leading to the formation of superoxide, instead of NO.4345 Superoxide can react very rapidly with NO, forming the powerful oxidant peroxynitrite,46 which can then readily modify protein residues and lipid moieties.8,47

B. The Effect of Endothelial and Platelet NO on Platelet Reactivity NO has both antithrombotic and vasodilatory effects. In the nondiseased blood vessel, the intact endothelium releases NO to inhibit platelet adhesion to the endothelium48,49 and platelet aggregation.5053 In addition, the vasodilatory effect of NO leads to smooth muscle relaxation. NO inhibits platelet activity primarily by binding to the heme-containing enzyme soluble guanylyl cyclase (sGC), which triggers a conformational change that increases its catalytic activity.54 Intracellular cyclic guanosine 50 -monophosphate (cGMP) levels are rapidly increased by sGC,55,56 affecting multiple signaling pathways,57,58 including cGMP-dependent receptor proteins, cGMP-regulated phosphodiesterases (PDE), and cGMP-dependent protein kinases.59 The increase in cGMP levels is accompanied by a decrease in intracellular Ca21 flux60,61 caused by activation of cGMP-dependent protein kinase G.7,62 Ca21 levels are

furthered lowered by a decreased rate of Ca21 entry from the extracellular environment and inhibition of receptor-mediated Ca21 release from the dense tubular system.63 The decrease in Ca21 levels inhibits the conformational change of GPIIb-IIIa into its active form64 and thus decreases platelet association with fibrinogen.65 The binding affinity of GPIIb-IIIa for fibrinogen is also decreased by cGMP-dependent inhibition of phosphoinositide 3-kinase activation (PI3K)66 and by cGMP-dependent phosphorylation of the focal adhesion vasodilator-stimulated phosphoprotein (VASP) at Ser157.67 In addition, stimulation of cGMP-dependent protein kinases leads to the inhibition of phospholipase A2- and C-mediated responses,68 such as the inhibition of arachidonic acid release and inhibition of phospholipase C/G protein/receptor coupling.62 Thromboxane (TXA2) receptor becomes phosphorylated by cGMPdependent protein kinase and is unable to mediate platelet activation.69 Another cGMP-dependent mechanism preventing platelet adhesion to the endothelium is the downregulation of P-selectin expression by platelet NOS via an inhibitory effect on protein kinase C (PKC).70 NO-mediated platelet inhibition can also occur through cGMP-independent pathways. Such pathways include the modification of cellular or plasma proteins by S-nitrosylation of cysteine residues forming S-nitrosothiols.58,71,72 Transnitrosation reactions between protein thiol-bound NO and low-molecular-weight thiolbound NO occur in vivo.73,74 The bioavailability of albumin thiols, or low-molecular-weight thiols such as

I. PLATELET BIOLOGY

316

17. INHIBITION OF PLATELET FUNCTION BY THE ENDOTHELIUM

glutathione, was found to prolong the antiplatelet action of endothelial NO.75,76 Extracellular NO in the nanomolar range is required for cGMP-independent inhibition of platelet activation. Plasma components may play an important role in the activation of cGMP-independent signaling by S-nitrosothiols or peroxynitrite generators.77 NO is important not only for inhibiting platelet aggregation and adhesion but also for modulating platelet function itself. Platelet-derived NO is released both at rest22 and during aggregation7880 and has been linked to important autoregulatory functions in platelet reactivity. The NO released from platelets in the resting state is in the nanomolar range.22 Shortly after activation, platelets release a large amount of NO (in the micromolar range) to prevent further platelet aggregation and adhesion in the growing thrombus.80,81 SHP-1 protein tyrosine phosphatase associates with eNOS in resting platelets. Upon activation, SHP-1 dephosphorylates eNOS, which leads to increased NO synthesis.82 Resveratrol, a main polyphenol in wine, has been shown to increase NO in platelets, in vitro, through protein kinase B.83 Estrogen has also been shown to regulate platelet NO release through the increase in cAMP and cGMP levels.84 The outcome of NO-mediated effects are concentration dependent, such that NO synthesized in a nanomolar range is cytoprotective to maintain vascular hemostasis, whereas NO released in the micromolar range is considered to produce cytotoxic effects promoting vascular pathology.7 One study showed that excessive NO production can inhibit Ca21 flux, prostacyclin production, and eNOS expression in endothelial cells.85 Thus, the bioavailability of NO is regulated in at least two ways: reaction with various reactive oxygen species (ROS)86,87 and desensitization of the NO/cGMP system.88 As discussed previously, under certain conditions eNOS can release the prothrombotic oxidant superoxide, which can react rapidly with NO to form peroxynitrite.46 NADPH oxidase has been identified as one primary source of superoxide in platelets87,89,90 and was found to play a role in platelet activation.90 Superoxide production by NADPH oxidase is decreased by NO-mediated PI3K inhibition, resulting in a reversal of platelet aggregation.91 This modulation of reactive oxygen and nitrogen species is a potential mechanism of regulating the bioavailability of NO at the site of platelet aggregation.87 Upon activation, platelets generate superoxide via the glutathione cycle and lipoxygenase pathway,92 leading to a reduction in NO levels. Hydrogen peroxide and lipid hydroperoxides are additional forms of ROS, which affect the bioavailability of NO. The plasma enzyme gluthathione peroxidase protects both endothelial cells and platelets93 from

oxidative damage by reduction of ROS.63 More recent work in vitro has shown that sphingosine-1-phosphate (S1P) released from platelets will increase eNOS activity and reverse the effects of hydrogen peroxide on the endothelium.94

C. NO-Mediated EndothelialPlatelet Interactions and Thrombotic Disease NO plays an important protective role in vascular hemostasis by suppressing thrombosis, atherosclerosis, and proliferation of vascular smooth muscle cells.95 Diverse animal and human in vivo studies examining eNOS activity and NO production have reported the level of NO bioavailability and eNOS function during thrombotic disease states, as well as studied the effect of NO-donating or NO-enhancing substances in various pathological settings. In atherosclerosis and hyperlipidemia, eNOS is dysfunctional and produces superoxide, which is implicated in endothelial dysfunction and impaired endothelium-dependent relaxations.96 Endothelial dysfunction or diminished NO bioavailability result in increased neutrophil adhesion to the endothelium and the initiation of atherosclerosis and thrombosis.9799 During sepsis, there is increased interaction among immune cells, including neutrophils, the endothelium, and platelets. In vitro data has shown that increases in eNOS activity can decrease neutrophil activation and adhesion onto endothelial cells and platelet activation, aggregation, and adhesion to endothelial cells and neutrophils.100 Animal studies have helped elucidate the importance of NO in plateletendothelial mediated homeostasis and vascular patency. Mice deficient in eNOS have decreased vascular reactivity and are hypertensive.101 Using platelets deficient in eNOS, it was shown that platelet-derived NO is important in regulating hemostasis, specifically by regulating platelet recruiment.81 eNOS deficiency is also associated with enhanced fibrinolysis due to lack of NO-dependent inhibition of the release of endothelial WeibelPalade bodies,102 which contain tissue plasminogen activator.5 More recently, it was demonstrated that in eNOS/ mice collateral circulation and angiogenesis was reduced due to impaired proliferation of vascular cells.103 Mice overexpressing the human eNOS gene were studied in a murine model of infarct-induced congestive heart failure.104 Both cardiac and pulmonary dysfunctions were attenuated and survival was drastically improved in the mice overexpressing human eNOS.104 The genetic overexpression of human eNOS in mice also attenuated myocardial infarction after ischemia/reperfusion injury but failed to significantly protect against post-ischemic myocardial contractile dysfunction.105

I. PLATELET BIOLOGY

II. NITRIC OXIDE

Overexpression of bovine eNOS was found to inhibit lesion formation in a mouse model of vascular remodeling,106 but when crossed with ApoE2/2 mice, atherosclerotic lesion formation was accelerated.107 In the human diseased blood vessel and during unstable coronary syndromes, release of both endothelial and platelet NO is impaired and thus contributes to thrombus formation. NO availability in the vascular system has been associated with various disease states and genetic variants. Risk factors for atherosclerosis— such as high cholesterol, male gender, family history, and age—have also been linked to this impaired endothelium-dependent vasodilation in coronary arteries.108 NO deficiency has also been linked to an increasing number of cardiac and noncardiac thrombotic disorders in humans. There is more platelet activation in unstable coronary disease,109 and these platelets produce significantly less NO compared to patients with stable coronary artery disease.110,111 Furthermore, in human atherosclerotic coronary arteries, endothelial NO-dependent vessel dilation is impaired.112 Endothelial dysfunction has also been associated with pulmonary hypertension, leading to eventual pulmonary vascular hypertrophy and thrombosis.113 Measurement of eNOS expression in the pulmonary vascular tissue of patients with pulmonary hypertension is decreased compared to healthy controls,114 and inhibition of eNOS leads to an increase in platelet deposition.115 Impaired bioavailability of NO due to a deficiency in plasma glutathione peroxidase was found to be the cause of thrombotic childhood stroke.116 Pharmacological agents may have direct beneficial effects by improving the function of endothelial cells and platelets.95,117 In a canine model of coronary artery stenosis, intravenous nitroglycerin infusion has been shown to inhibit platelet thrombus formation.118 In a rat study, inhaled NO resulted in a lower incidence of collagen-induced platelet aggregation in small pulmonary vessels compared to controls.119 The antithrombotic activity of NO was similar to the platelet GPIIb-IIIa antagonistic peptide G4120, suggesting that NO had a direct effect on inhibiting platelet activation.119 Chronic treatment with L-arginine, a substrate for eNOS, inhibits formation of atherosclerotic lesions in animal models of atherosclerosis.120,121 Human in vivo studies have shown that modulating NO clinically affects platelet function. Oral supplementation of L-arginine in healthy adults inhibits ADP-dependent platelet aggregation when compared to placebo,122 and inhibition of platelet eNOS enhances platelet aggregation and reduces bleeding times.123 Furthermore, infusion of L-arginine reversed the vasoconstrictive effect of eNOS inhibitor L-NMMA in coronary arteries.124 Some statins have also been used to upregulated eNOS. However, the

317

results have been inconsistent. Clinical studies have shown that the presence of asymmetric dimethylarginine (ADMA) affects how statins work. Individuals with high levels of ADMA did not show any enhancement of NO synthesis upon treatment with simvastatin; those with low ADMA levels benefited from the treatment. Adding L-arginine to simvastatin treatment in patients with elevated ADMA levels improves endothelial function.125 Clinical studies have also looked at how dietary supplements can affect eNOS and NO levels. A peroxynitrite scavenger found in walnuts, γ-tocopherol, has shown in enhance platelet eNOS activity and endothelial-dependent vasodilation, potentially through upregulation of expression and phosphorylation of eNOS, as shown in rat models.126 In vitro, fish oil treatment of human endothelial cells increases eNOS gene expression and decreases NADPH oxidase expression.127 EGb 761, an extract of ginkgo biloba, increases eNOS expression and activation through phosphorylation at Ser478, resulting in the reduction of blood pressure and vasodilation in rat models.128 Folic acid has been shown to promote eNOS dimerization, which has been suggested as one way folic acid may improve endothelial function in patients with coronary artery disease.129 Finally, acetate has been shown to increase phosphorylation of eNOS by PKA and AMPK in HUVEC, which is suggested to explain the increase in forearm vasodilation in postmenopausal women given vinegar (acetic acid).130 The use of GPIIb-IIIa antagonists is known to reduce myocardial infarction and death in patients undergoing percutaneous coronary intervention and in patients with acute coronary syndromes (Chapter 55).131,132 Inhibition of GPIIb-IIIa in patients with known cardiovascular disease increases NO bioactivity.133 NO donors have also been found to directly reverse GPIIb-IIIa activation of platelets,134 although the therapeutic use of GPIIb-IIIa antagonists remains in flux.135,136 Angiotensin-converting enzyme (ACE) inhibitors have been largely used to treat hypertension. Experimental models and clinical studies have shown how some of these inhibitors can affect eNOS levels and activity. Some ACE inhibitors were shown to increase eNOS levels and activity in rat aortas; however, some were better than others.137 In patients with coronary artery disease or heart failure, ACE inhibitor treatment increased eNOS levels and activity in endothelial cells and cardiac tissue.138 S1P, which has been shown to augment eNOS levels in endothelial cells, has been studied as a potent immunosuppressor in kidney transplantation. Clinical trials, using its analogue FTY720, have shown that treatment traps lymphocytes in lymphoid organs, preventing migration to inflammatory sites. ApoE/

I. PLATELET BIOLOGY

318

17. INHIBITION OF PLATELET FUNCTION BY THE ENDOTHELIUM

mice given FTY720 had a reduction in atherosclerotic plaque formation, suggested to occur through its immunosuppressive abilities and activation of eNOS.139 A PPARγ agonist, fenofibrate, has been studied for its role in reducing cardiovascular events, particularly in diabetic patients who have undergone limb amputation. Fenofibrate increases adiponectin as a way to protect the vasculature and increase revascularization. As previously mentioned, work in mice looking at collateral circulation has indicated a role of eNOS in angiogenesis and collateral formation.103 Fenofibrate treatment in mice was shown to stimulate the phosphorylation of AMPK and eNOS in ischemic tissue. Blockage of both pathways, individually, led to a reversal of fenofibrate effects on blood flow.140

D. Genetic Variants and Polymorphisms of Endothelial and Platelet eNOS Numerous investigations have elucidated eNOS polymorphisms and linked them to various thrombotic diseases in association studies, although many of these studies are now considered to be underpowered. The most common and thus extensively studied polymorphisms are the Glu298Asp polymorphism in exon 7 (also known as G894T), the T786C polymorphism in the promoter region of the eNOS gene, and the intron 4b/a. Table 17-2 provides an overview of these and other polymorphisms with regard to their association with thrombotic diseases. Genetic variability due to heritable factors has a major influence on platelet TABLE 17-2 Disease

aggregation,141 and the availability of plasma NO has been linked to several of these polymorphisms.142,143 In a recent meta-analysis, the effects of three of the most common eNOS genetic variations on the susceptibility to ischemic heart disease were examined:144 Glu298Asp, T786C, and intron 4. In this study,144 only homozygosity for Glu298Asp and intron 4a alleles were associated with respective 31% and 34% increased risks of ischemic heart disease. Studies attempting to demonstrate an association of clinical coronary artery disease or acute myocardial infarction with eNOS polymorphisms are inconsistent and vary between ethnic groups (Table 17-2). Japanese patients homozygous for the intron 4 polymorphism were associated with an increased risk of myocardial infarction.145 However, in a similar German population, there was no increased risk of cardiovascular disease,146 whereas a decreased risk of myocardial infarction was reported in Finnish men.147 In Koreans, both T768C and 4b/a polymorphisms are associated with coronary artery disease.148 In an study conducted in the United States, the Glu298Asp polymorphism is associated with the first myocardial infarction occurring in individuals younger than 50 years old.149 Part of the apparent variations in studies may lie in the various presentations of acute and chronic coronary artery disease, including coronary vasospasm, intraarterial thrombosis, and chronic plaque formation or plaque rupture. There are also numerous difficulties associated with reporting genetic associations linked to complex outcomes;150 for instance, many of the current trials involving eNOS polymorphisms are relatively

Polymorphisms of the eNOS Gene and Associated Disease States Cardiovascular Disease: MI, CAD

Hypertension

Stroke and Aneurysm

Diabetes

Other

T786C161,162,451

T786C152155,158,157

T786C164,165

Retinopathy of PREMATURITY  T786C; G894T174

Intron 4b/a144148,444,452,453,455

Intron 4b/a161,162,454

Intron 4b/a152,156,163

Intron 4b/a163,164,167

Lupus  T786C172,173

G894T 144,149,180,444,445,457461

G894T161,162,453,461465

G894T152154,156,466

G894T164,165

Breast Cancer  T786C178

A922G444

Intron 13 CA VNTR468

A922G157

786/4b/ 894G163

Renal Disease  T786C; Intron 4b/a; G894T164,165,179,180

Intron 13 CA VNTR467,469

Intron 23 G10T463,465

298Asp/4b/-786T156

C Asp b166

ED  G894T175,176

C Glu b159

298Asp/4b/-786C156

C Glu a160

-786C/4b/894G163

Polymorphism/ T786C144,148,180,444450 VNTR/SNP/ Haplotype

C Asp b166 Abbreviations: CAD, coronary artery disease; ED, erectile dysfunction; MI, myocardial infarction; SNP, single nucleotide polymorphisms; VNTR, variable number of tandem repeat polymorphisms.

I. PLATELET BIOLOGY

319

III. PROSTACYCLIN

small and likely underpowered, in addition to the possible different distribution of eNOS polymorphisms among ethnic groups.151 eNOS polymorphisms are also associated with ischemic stroke and aneurysms. eNOS polymorphisms including G894T, T786C, and 4b/a, and haplotypes were associated with silent brain infarctions in Koreans.152 However, in a separate study, there was no link between these eNOS polymorphisms and intracranial aneurysms in Koreans153 and Indians.154 In a study conducted in the United States, there was an association between the T786C polymorphism and the increased risk for angiographic vasospasm after an aneurysm.155 Both eNOS polymorphisms and haplotypes have been associated with a susceptibility of ischemic stroke in Tunisians156 and Chinese.157 Conversely, intron 4aa polymorphism was shown to be protective of lacunar infarction, or silent strokes; T786C increased the chances in a Turkish population.158 In hypertension, various studies have found correlations between eNOS polymorphisms and the development or progression of the disease. In obese children and adolescents, eNOS haplotypes, but not polymorphisms, were associated with the development of hypertension.159 Haplotypes were also associated with gestation hypertension and preeclampsia.160 Interestingly, as seen with coronary artery disease and myocardial infarction, studies in Italians have shown no associations between eNOS variants and hypertension.161,162 In relationship to diabetes, eNOS haplotypes have been associated with both type 2 diabetes and the risk of ischemic stroke in individuals with diabetes in Koreans.163 There is no association of eNOS polymorphisms in Caucasian Brazilians with diabetes and the development of renal disease.164 However, in Asian Indians, there is an association of eNOS polymorphisms and haplotypes with the development of renal disease in diabetic patients, related to lower serum NO levels.165Additionally, another study done in Brazil, showed a relationship between eNOS haplotypes and NO levels in individuals with hypertension and type 2 diabetes.166 Polymorphisms in intron 4b/a in Germans are associated with lower eNOS-dependent vasodilation in individuals at an increased risk for type 2 diabetes.167 eNOS polymorphisms have been shown to alter treatment efficacy. T786C polymorphism has been shown to affect atorvastatin’s ability to modulate NO bioavailability and inflammation.168,169 The effect of exercise on lowering blood pressure in postmenopausal women has also shown to be more effective in individuals with the T786C polymorphism.170 Finally, eNOS haplotypes have been shown to determine the effectiveness of antihypertension therapy in women

with preeclampsia but not in gestational hypertension.171 eNOS polymorphisms have also been associated with various other diseases. eNOS haplotypes in combination with the T786C polymorphism is associated with susceptibility to systemic lupus erythematous in subjects from Kuwait172 but not in subjects from China.173 Retinopathy of prematurity in infants is associated with both T768C and G894T polymorphisms.174 G894T is associated with an increased risk of erectile dysfunction.175,176 eNOS polymorphisms are also associated with a reduced risk of radiation therapy toxicity used to treatment breast cancer.177 However, another polymorphism, T786C, is associated with sporadic breast cancer in women younger than 55 years old.178 Finally, in renal disease, eNOS polymorphisms are associated with functional deterioration179 and cardiovascular events.180

III. PROSTACYCLIN A. Prostacyclin Biosynthesis and Characteristics of Endothelial Prostacyclin Synthase Prostaglandin I2 (PGI2) or prostacyclin is a derivative of the C-20 unsaturated fatty acid arachidonic acid (5,8,11,14-eicosatetraenoic acid),181 which was discovered in 1976 as a substance inhibiting platelet secretion, platelet aggregation, and vasoconstriction.182,183 With a half-life of about 3 minutes,184,185 prostacyclin is extremely labile under physiological conditions, but this half-life is increased by binding to serum albumin to stabilize PGI2 activity and enhance its receptor binding.186 The biosynthesis of prostacyclin is part of the arachidonic acid metabolic pathway. Arachidonic acid is a member of the ω-6 series of essential fatty acids and is covalently bound at the sn-2-position in membrane phospholipids. It is released by phospholipase A2 upon activation of the enzyme by an increase in intracellular Ca21 concentration. Arachidonic acid is further metabolized by at least two major enzyme complexes: (1) prostaglandin endoperoxide H (PGH) synthase, which is commonly known as cyclooxygenase (COX), and (2) 5-lipoxygenase187 (see Chapter 53). COX is a bifunctional enzyme with two related catalytic functions, a bis-oxygenase (cyclooxygenase) that catalyzes first the formation of prostaglandin G2 (PGG2),188 and a hydroperoxidase that subsequently catalyzes the two-electron reduction of PGG2 to the unstable PGH2.189 PGH2 is further converted by several specific synthases into prostanoids such as PGE2, PGD2, PGF2α, prostacyclin, or TXA2, depending on the tissue in which this reaction occurs.187,190 Prostacyclin

I. PLATELET BIOLOGY

17. INHIBITION OF PLATELET FUNCTION BY THE ENDOTHELIUM

synthase (PGIS), in the endothelium, catalyzes the formation of prostacyclin from PGH2. PGIS is widely distributed, predominantly in endothelial and smooth muscle cells,191196 but it is not expressed in platelets. PGIS, a membrane-bound enzyme of approximately 57 kDa, has one membrane anchor domain at the N-terminus197 and a large cytoplasmic domain containing a substrate channel, hemebinding sites, and two Couet-motifs.198,199 The enzyme is mainly localized in the membrane of the endoplasmic reticulum (ER)200 whereas the heme-containing domain faces the cytoplasmic site of the ER.198,201 The two isoforms of COX are also located in the ER membrane, though their active site is positioned toward the ER luminal side.202 In addition, recent studies demonstrated the presence of PGIS in caveolae of human endothelial cell membranes. It binds to caveolin-1, the primary coat protein of caveolae, which does not affect the enzymatic activity.203 Previous studies suggested that PGIS is subject to multilevel regulation in vivo. One proposed mechanism is “suicide” inactivation, by which PGIS undergoes a catalytic inactivation by its substrate PGH2.204 Another control mechanism has been described as selective inactivation of PGIS by tyrosine nitration at the active site at Tyrosine 430 due to exposure to micromolar peroxynitrite concentrations.205,206 At the gene expression level, it has been previously established that variations in the PGIS gene sequence can affect the enzymatic activity (see Section III.E).

B. The Platelet Prostacyclin Receptor: Biochemistry, Structure, and Function Prostacyclin is a potent vasodilator, antithrombotic, and antiplatelet agent that mediates its effects through a specific membrane-bound receptor, the prostacyclin receptor (IP receptor). The receptor belongs to the prostanoid family of G-protein-coupled receptors (GPCRs)190 and is expressed on platelets207,208 and smooth muscle cells.209 IP receptor expression within the cardiovascular system is most abundant in the aorta.210 It is also expressed in the atrium and ventricle of the heart, indicating possible roles for prostacyclin in cardiac tissue.211 There is no IP receptor expression found in veins.190 The human prostacyclin receptor has a molecular weight of 41 to 83 kDa,212,213 is located in the plasma membrane, seven transmembrane domains with a short extracellular N-terminal region and an extended intracellular C-terminal region.211 Prostaglandins have two structural features, a cyclopentane ring and side chains, that are recognized by their receptor to stabilize ligand binding. In the IP receptor, transmembrane

domains VI and VII bind with PGI2 side chains. Transmembrane domains I and II confer broader binding functions, including recognition and interaction with the cyclopentane ring of prostacyclin.181,214 The transmembrane domain I also contains a dimerization motif.211 The prostaglandins are not necessarily specific for an individual receptor. The binding pocket of the IP receptor can accommodate the cyclopentane rings of PGI2, PGE1, and PGE2.211 Prostacyclin analogs such as iloprost and cicaprost bind the receptor with the same affinity as prostacyclin. The rank order of agonist affinity for the cloned human prostacyclin receptor is: iloprost 5 cicaprost . PGE1 . carbacyclin PGE2 . PGD2, PGF2α.190,210,215,216 The cross-reactivity of IP ligands with several receptors for PGE2 (EP) has been reported.190,211 Further elucidation of substrate specificity of the IP receptor as well as the evaluation of the role of endogenous prostacyclin have been hampered by the lack of a potent, selective IP receptor antagonist and highly selective agonists.211,217,218 The human prostacyclin receptor contains two potential N-glycosylation sites, one in the extracellular N-terminal domain at Asn7 and one in the first extracellular loop at Asn78.211 A functional study of the glycosylation sites in HEK and COS-7 cells revealed a greater degree of glycosylation at Asn78 than at Asn7, which might play a role in membrane localization, ligand binding, and signal transduction of the IP receptor.219 The C-terminus of the IP receptor contains a consensus sequence for isoprenylation and is modified at Cys383 by a C-15 farnesyl isoprenoid chain.220 This modification has no effect on the ligand binding characteristics but is required for efficient coupling of the prostacyclin receptor to adenylyl cyclase as well as phospholipase C, both via the G proteins Gαs and Gαq, respectively.220 In addition, it was established that the isoprenyl modification is necessary for efficient agonist-mediated receptor internalization221 following receptor desensitization222 and G-protein uncoupling.223 Palmitoylation of the intracellular C-terminal domain at residues Cys308 and Cys311224 is not required for ligand binding but plays a crucial role in Gαs/adenylyl cyclase coupling. Cys308 is necessary for Gαq coupling and phospholipase C activation.224 Several sites in the intracellular loops and the C-terminal domain of the IP receptor represent consensus sequences for protein kinase A (PKA) and PKC phosphorylation.211,223 In particular, Ser328 was found to be a primary site for PKC phosphorylation.223 In addition to protein regulation via posttranslational modifications, the composition of the platelet membrane also plays a role in the expression of prostacyclin binding sites whereby an increase in cholesterol

I. PLATELET BIOLOGY



320

III. PROSTACYCLIN

content lowers and an increase in phospholipids enhances the amount of bound prostacyclin.225 Furthermore, the expression of the prostacyclin receptor was shown to be upregulated by proinflammatory and megakaryocytopoietic cytokines.226

C. Prostacyclin: Signaling Mechanisms and Platelet Inhibition Prostacyclin is a very potent endogenous inhibitor of platelet aggregation, as well as a strong vasodilator that also inhibits the growth of vascular smooth muscle cells.182,191,193,215,227231 It inhibits platelet activation induced by various stimulants such as thrombin, collagen, ADP, TXA2, or the calcium ionophore A23187.218 However, it shows selectivity towards TXA2-initiated platelet activation and its inhibitory potential decreases in the following order: TXA2 A23187 . thrombin . ADP.232 Besides inhibiting platelet activation and limiting thrombus size,191 prostacyclin can also prevent platelet56 and leukocyte adhesion to endothelial cells.233 After release from the vessel wall, prostacyclin mediates its inhibitory effects via the high-affinity prostacyclin receptors on the platelet surface, causing an increase in cAMP levels.215,234,235 The binding of prostacyclin to its receptor induces a signaling cascade through G-protein Gαs.181,236 This stimulates adenylate cyclase, assumed to be located in the dense tubular system,237 and leads to an increase in cAMP levels.234,235 Increased cAMP production activates PKA,238 which causes the phosphorylation of several key proteins239,240 such as myosin light chain kinase (MLCK),241 the platelet inositol 1,4,5-triphosphate receptor,242 and VASP.243 Activation of these proteins leads to the inhibition of several pathways: (1) Rhokinase and MLCK-activation and subsequent granule secretion, (2) GPIIb-IIIa activation, and (3) PKC activation and increase in intracellular calcium levels.218,244 Phosphorylated MLCK is inactive and has a reduced affinity for calmodulin, which then reduces the amount of phosphorylated myosin.245 The effect of this is a decreased platelet contractile activity, including secretion, and a decreased association of myosin with the platelet cytoskeleton.246 Through these mechanisms, prostacyclin is able to inhibit platelet activation and to cause disaggregation of existing platelet aggregates.247 Prostacyclin action is a dynamic process that is regulated on many levels. The binding of prostacyclin to its receptor and subsequent activation of adenylyl cyclase via Gαs is a concentration-dependent process. Similar to other GPCRs, the IP receptor may activate more than one G protein.211 At higher ligand

321

concentrations, the IP receptor activates phospholipase C, most likely through Gαq coupling,248 and causes calcium mobilization.249 Furthermore, it has been proposed that the net effect on cAMP levels by prostacyclin depends on its ability to stimulate as well as inactivate adenylyl cyclase.250 Prostacyclin receptor desensitization is another regulatory mechanism of PGI2 responsiveness. It is known that elevated prostacyclin levels are accompanied by reduced binding to the IP receptor and decreased responsiveness in human vascular disease, such as myocardial infarction251,252 and preeclampsia,253 as well as during administration of prostacyclin and its analogs.211,254 The desensitization process involves uncoupling of the receptor from G-proteins, inhibition of adenylyl cyclase activity, and subsequent receptor internalization. The result is a diminished response due to extended exposure to agonists or a high concentrations of agonist.222,223 Several studies have shown that desensitization and internalization of the human IP receptor takes place in platelets,222,255,256 in culture,249,257 as well as in vivo.258,259 PKC-mediated phosphorylation of the human IP receptor is a critical determinant of agonist-induced desensitization,223 whereas sequestration is independent of the PKC pathway and proceeds partially via an endocytotic pathway involving clathrin-coated vesicles.249 In platelets, the response to desensitization is augmented plateletendothelial cell adhesion in the presence of thrombin, resulting in an enhanced tendency toward thrombus formation.251,256 The short-term desensitization of the platelet IP receptor is a reversible phenomenon whereby the receptor is not degraded but is rapidly recycled as a functionally active form after prostacyclin withdrawal.260 IP receptor responsiveness is also regulated by receptor co-expression and co-localization, receptor cross-regulation through shared G-proteins, as well as proposed heterodimerization with other prostanoid receptors.211,215,218,261,262 In platelets the IP receptor (a stimulatory receptor) co-localizes with the EP3 receptor (an inhibitory receptor) to regulate cAMP production and to act as a buffering mechanism against a local increase in prostacyclin levels.263,264 This observation emphasizes the functional advantage of nonspecific agonists. Maintenance of the balance between prostacyclin and TXA2 appears to be a critical regulator for the maintenance of vessel integrity,265 the interaction between platelets, and the endothelium193,266 and cardiovascular processes.218,267 Synthesized by platelets268 and released after platelet activation,269,270 TXA2 is a platelet agonist and vasoconstrictor participating in a positive feedback loop that mediates additional platelet activation and aggregation. The actions of PGI2

I. PLATELET BIOLOGY



322

17. INHIBITION OF PLATELET FUNCTION BY THE ENDOTHELIUM

counteract the actions of TXA2. Platelet activation and vasoconstriction induced by TXA2 can be inhibited by prostacyclin.215 Through increased platelet cAMP levels, prostacyclin blocks arachidonic acid formation271 and thus prevents synthesis of prothrombotic prostaglandins and thromboxanes by the human platelet,272,273 which further inhibits platelet activation.274 The formation of heterodimers between the prostacyclin receptor and the TXA2 receptor α (TPα) has been shown to regulate the prothrombotic role of TXA2.262 IP and TPα heterodimerization results in cAMP generation through TPα, changing the effect of this receptor to be antithrombotic.262 In addition, platelet-derived endoperoxides (like TXA2) can act as substrate and thus upregulate prostacyclin synthesis in endothelial cells.218,275,276 In general, there appear to be two mechanisms for prostacyclin synthesis by cultured endothelial cells: the first one involves prostacyclin synthesis from endogenous precursors, and the second one involves endoperoxides derived from stimulated platelets.275 As discussed previously, platelets release other antithrombotic substances, such as NO, that provide a negative feedback for thrombus formation. Indeed, low doses of NO and PGI2 synergize in their anti-aggregatory effect,277,278 although not at a level to inhibit platelet adhesion to the endothelium.56,279 Platelets also release factors that upregulate prostacyclin production. Bile salt-dependent lipase is released from platelets upon activation and has been shown to bind to HUVEC and increase prostacyclin synthesis.280

D. The Relevance of Prostacyclin In Vivo Prostacyclin as an antiplatelet and antithrombotic mediator plays an important role in the development of cardiovascular disease.281 Dysfunctional prostacyclin activity has been implicated in the development of various cardiovascular diseases including thrombosis, myocardial infarction, stroke, atherosclerosis, and hypertension.190 A reduction in the binding affinity of platelets for prostacyclin and cAMP synthesis has been observed in patients with acute myocardial infarction compared to healthy subjectss.252 Because PGIS is upregulated by various cytokines, prostacyclin biosynthesis has been reported to be increased in the presence of atherosclerosis and platelet activation.282 Mice deficient in the PGIS gene are hypertensive and develop vascular disorders with vascular wall thickening and interstitial fibrosis, particularly in the kidneys.283 Inhibition or deficiency of PGIS, in vivo, causes a marked enhancement of white platelet thrombus formation.284286 Several investigators have reported that abnormal

prostacyclin synthesis or metabolism may be a risk factor for myocardial and cerebral infarction.287,288 Delivery of PGIS in vivo can prevent proliferation and migration of smooth muscle cells, key steps in the development of restenosis and atherosclerosis.289,290 Although prostacyclin reduces cerebral infarction in animal models,291,292 many therapeutic trials of prostacyclin have failed to show significant clinical improvement.293295 A decrease in prostacyclin production has been implicated in the pathogenesis of severe pulmonary hypertension296 but not in human essential hypertension. However, in pregnancy-induced hypertension, a decrease in prostacyclin has been reported to precede the clinical manifestation.297 Mice deficient in the IP receptor have been studied in various disease settings.217,298302 Murata et al. found that IP receptor knockout mice display an increased thrombotic tendency but have a reduction in inflammatory swelling and pain responses and are not susceptible to hypertension.298 Injury of the carotid artery leads to obstruction in the mice lacking the IP receptor, and this response was abolished in mice lacking TXA2.300 In the IP null mice, the production of TXA2 by platelets and components of the injured vessel wall was increased.300 Ovariectomized LDLR/ female mice also lacking the IP receptor are no longer atheroprotective when estrogen is reintroduced. Estrogen, when bound to estrogen receptor alpha, upregulates prostacyclin through COX-2 activation.302 The opposing actions of prostacyclin and TXA2 on platelets and the vessel wall and their concentration levels at the site of injury are considered to be critical for thrombus formation183 and in various occlusive vascular diseases including coronary heart disease.262,303 During cardiac ischemia/reperfusion, the synthesis of PGI2 and TXA2 are significantly increased.304,305 It has been reported that PGI2 and its analogs attenuate cardiac ischemia/reperfusion injury when administered exogenously in animal models,306309 possibly due to their inhibitory effect on platelets and neutrophils. Clinical studies suggested that loss of the IP receptor may contribute to atherogenesis in patients with chronic spinal cord injury.310 During spontaneous angina pectoris,311 severe atherosclerosis,312 and acute myocardial infarction,313 the prostacyclin binding capacity and the number of IP receptors has been reported to decrease, but not in other patients with angiographically proven coronary artery disease and stable angina.252,314 It has been shown that in myocardial infarction and unstable angina, biosynthesis of prostacyclin is considerably increased,109,313 which could lead to agonist-induced receptor changes such as desensitization of the PGI2 binding sites.314

I. PLATELET BIOLOGY

III. PROSTACYCLIN

Prostacyclin’s short half-life has led to the development and experimental application of several prostacyclin mimetics.215 The ones most commonly used are cicaprost,315 iloprost,316 and carbacyclin,215,317,318 which are all analogs of PGI2.319 Cicaprost is often the analog of choice as iloprost is less selective and partially acts as an agonist for prostaglandin E1 receptors.215 Administration of prostacyclin mimetics, such as epoprostenol, in the clinical setting has been used to limit platelet aggregation, but a major drawback is the profound vascular dilatation they induce.215 In pulmonary arterial hypertension (PAH), treatment with prostacyclin analogs, such as epoprostenol, treprostinil, iloprost and beraprost, continues to play an important role despite the complications induced by their generally short half-lives and complicated drug delivery systems.320 More recently, epoprostenol was shown to have no effect in patients with pulmonary embolism.321 This same drug has been shown to induce headaches in healthy patients322 and migraine-like events in migraine suffers323 because of the sensitization of sensory afferents around extracranial arteries. However, epoprostenol has been shown to increase the survival of patients with PAH324 and improve pulmonary function,325 even during thoracic transplantation.326 To improve the effects of beraprost on PAH, this drug has been used in conjuncture with other drugs, such as an angiotensin blocker,327 or has been reformulated to last longer to improve pulmonary pressure and quality of life.328 Iloprost is also being studied for its effectiveness in PAH, even during heart transplantations,329 and shows, alone or in conjuncture with other drugs,330 effectiveness in improving pulmonary arterial pressure and hemodynamics.331 Treprostinil has been extensively studied more recently, particularly as a replacement for epoprostenol because it is longer lasting.332,333 All studies report improved pulmonary vasodilation, exercise capacity, hemodynamics, and survival.334338 Iloprost has been used for the treatment of systemic sclerosis, a connective tissue disease in which the immune system attacks body tissues, leading to a buildup of collagen in these tissues. Studies have looked at its effects on oxidative stress that occurs due to frequent ischemia/reperfusion events during the pathogenesis of this disease, but there has been no agreement if this drug has a role.339,340 However, iloprost has been shown to be effective in preventing pulmonary hypertension in patients with systemic sclerosis.341 Two prostacyclin analogues have been studied for the treatment of peripheral artery disease (PAD; arteriosclerosis obliterans)—iloprost and beraprost. Iloprost has been shown to be effective in reducing inflammation and oxidative stress in conjuncture with

323

aspirin treatment;342 alone, it has not been effective to treat the disease.343 Beraprost has shown to be effective in the treatment of PAD by reducing renal anemia,344 decreasing endothelin-1 levels, and improving quality of life.345 Prostacyclin analogs are also being tested in renal diseases. Individuals with acute renal failure requiring continuous venovenous hemodiafiltration have an increased risk of thrombosis. Treatments to prevent this cause an increase in bleeding. A prostacyclin analog was shown to reduce platelet aggregation induced by ADP and collagen these patients.346,347 In another study, patients with renal failure undergoing coronary intervention were given iloprost prophylactically protected against contrast-induced nephropathy.348 Finally, individuals on chronic peritoneal dialysis experience endothelial injury and thrombosis. Patients treated with beraprost had a reduction in both Ddimer and von Willebrand factor markers, indicative of thrombosis and endothelial damage, respectively.349 Aspirin (acetylsalicylic acid) has an anti-thrombotic effect350 based on its preferential action on blocking COX in platelets and the endothelium by acetylating COX’s active center (Chapter 53).351 Aspirin has also been reported to increase NO production in neutrophils352,353 and in the arterial wall.354 There are two different cyclooxygenases, COX-1 and COX-2.187,188,355 COX-1 is involved in TXA2 synthesis in platelets while COX-2 is involved in the synthesis of prostacyclin in endothelial cells.187,356 Aspirin at low doses acetylates COX-1 in platelets and thus irreversibly blocks TXA2 synthesis for their lifetime in the circulation. At the same low doses, aspirin has little effect on the synthesis of PGI2. Thus, the overall effect of low dose aspirin is a reduced risk of thrombosis.187 The combined inhibition of both COX isoforms, but not the selective inhibition of COX-2, attenuates atherogenesis in LDLR/ mice.357

E. Polymorphisms of the PGIS and Prostacyclin Receptor Genes The genomic organization of the PGIS gene has been investigated for several polymorphisms in association studies performed, primarily, in the Japanese population.358369 Correlations between some of these polymorphisms, including single nucleotide polymorphisms (SNP), variable number of tandem repeat polymorphism (VNTR), and splicing mutations, and have found an increased risk for essential hypertension, myocardial infarction, and cerebral infarction.360,364,366368 Two additional studies found polymorphisms in the French Caucasian population and two in African ethnic groups, but with no

I. PLATELET BIOLOGY

324 TABLE 17-3

17. INHIBITION OF PLATELET FUNCTION BY THE ENDOTHELIUM

Polymorphisms of the Prostacyclin Synthase (PGIS) Gene and Associated Disease States

Disease

Cardiovascular disease: MI, CAD, Thrombosis

Hypertension

Stroke and Aneurysm Other

Polymorphism/ VNTR/SNP/ Haplotype

SNP A1117C in Exon 8368,374

Nonsense Mutation in Exon 2, Codon 26365

Nonsense Mutation in Exon 2, Codon 26365

Renal Disease - Splicing Site Mutation in Intron 9359,360

M113T, L104R. R279C382

VNTR in Promoter  3-7 repeats of 9 bp362,363,369,372

VNTR in Promoter  3-7 repeats of 9 bp367

Respiratory Syncytial Virus Infection  VNTR378

V53V/S328S381

Splicing Site Mutation in Intron 9359,360

Lung Cancer  CpG Methylation in VNTRs in Promoter and Exon 1377

R212C381

SNP T192G366

Adenomatous Polyps  VNTR in Promoter376

SNP375

T192G/VNTR/A117C361 SNP373

Abbreviations: CAD, coronary artery disease; MI, myocardial infarction; SNP, single nucleotide polymorphisms; VNTR, variable number of tandem repeat polymorphism.

associated functional changes or disease-associations (Table 17-3).370,371 Other mutations have been identified, including a nonsense mutation in exon 2 at codon 26 (CGA/TGA) of the PGIS gene, which may be associated with hypertension. This polymorphism causes a large part of the PGIS mRNA to not be translated and is assumed to decrease the enzymatic activity of PGIS.365 A VNTR polymorphism in the 5’-upstream promoter region has also been found in Japanese subjects and is associated with cerebral infarction.367 This region contains binding sites for the transcription factors Sp1 and AP-2. The alleles vary in size from three to seven repeats of nine base pairs. In a study with almost 5000 Japanese subjects, the 3 and 4 repeats of the VNTR was found to be associated with increased systolic hypertension and higher pulse pressure.363,372 This repeat polymorphism was found to influence the promoter activity of the PGIS gene in human endothelial cells.363 Another mutation includes a splicing variant in intron 9 that results in a truncated protein lacking the heme-binding region, which is associated with hypertension.359,360 In the Croatian population, a SNP near PGIS and the potassium voltage gate channel genes is associated with a high prevalence of hypertension, particularly in inhabitants of the island of Vis, where 75% of the population has hypertension.373 The SNP, C1117A, which is a silent mutation, was identified in exon 8 and found to be associated with a risk for myocardial infarction, in a Japanese population368 and Chinese population.374 More recently, a study done in the United States showed that common SNPs in PGIS were associated with myocardial infarctions. One haplotype was associated with a higher risk of myocardial infarction and stroke. One SNP was associated with a lower risk for both diseases.375

Polymorphisms in PGIS that affect the promoter region and expression level have also been associated with cancer. Less than 6 repeats in the promoter region of PGIS was associated with an increased risk of adenomatous polyps, while more than 6 repeats was associated with a reduced risk. The presence of these repeats also determined the benefit of NSAID treatment for colorectal polps.376 Finally, CpG methylation within VNTRs in the promoter and in exon 1 and the first intron control PGIS expression level. Overexpression of PGIS was found to be protective in lung cancer. Human lung cancer cell lines were found to have lower levels of PGIS due to a high number of these CpG methylation sites.377 Finally, the number of VNTRs in the promoter of PGIS are inversely correlated with the severity of respiratory syncytial virus in Japanese infants. A low number of VNTRs, resulting in a lower level of urinary PGI2 metabolite levels correlated with the most severe respiratory syncytial virus infection in the lower respiratory tract. The higher number of VNTRs correlated with a less severe infection.378 A genetic analysis revealed the first two naturally occurring polymorphisms within the coding sequence of the human prostacyclin receptor, Val25Met in the transmembrane domain I and Arg212His in the third intracellular loop.379 Structure-function characterizations of these polymorphisms were performed at physiological pH (7.4) and an acidic pH (6.8) that would be encountered during stress such as renal, respiratory or heart failure. In particular, the Arg212His polymorphism showed significant reduction in receptor binding affinity at low pH and in signal transduction activity at both pH values, whereas the Val25Met polymorphism had no significant effects.379 A nuclear prostacyclin receptor has been identified which belongs

I. PLATELET BIOLOGY

IV. CD39 (NTPDase-1)

to the peroxisome proliferator-activated receptor (PPAR) family (PPARδ)380 and was suggested to be involved in angiogenesis.199 More recently, polymorphisms in the human prostacyclin receptor have been associated with thrombosis and cardiovascular disease. Two synonymous polymorphisms (V53V/ S328S) were associated with an increase in sP-selectin and 11-dehydro-thromboxane B2 levels in DVT patients compared to control.381 Another polymorphism, R212C, results in a dysfunctional receptor and is associated with intimal hyperplasia.381 Rare receptor polymorphisms that lead to impaired binding and activation of the prostacyclin receptor, including M113T, L104R, and R279C, were associated with coronary artery obstruction.382

IV. CD39 (NTPDase-1) A. Endothelial CD39—Biochemistry, Structure and Function Ecto-nucleoside triphosphate diphosphohydrolases (ENTPDases) is a family of membrane proteins that are ubiquitously expressed in eukaryotic cells and play a pivotal role in mediating platelet-endothelial interactions. These enzymes hydrolyze nucleoside 5’-di- and triphosphates in a Ca21 or Mg21-dependent manner.383,384 Endothelial CD39 (NTPDase-1), the major vascular ENTPDase, is a membrane-anchored glycoprotein with ecto-apyrase activity.385,386 As an integral component of the endothelial cell surface, it is activated by its extracellular substrates in the blood stream, ATP and ADP, which are rapidly hydrolyzed into AMP.383,387 An ecto-5’-nucleotidase (CD73) further converts AMP into adenosine.388,389 CD39 enzyme expression is found in a wide variety of vascular cells. The enzyme is highly expressed on human umbilical vein endothelial cells (HUVECs),390,391 which also has the highest enzymatic activity. It is also expressed on natural killer cells,392 a subset of T cells,390 activated B cells,393 EpsteinBarr virus-transformed B cells,385 megakaryocytes, and platelets.394 Structural analysis of CD39 showed that the protein is membrane-anchored at its N- and C- terminus, whereas each terminus is composed of one transmembrane domain and a short cytoplasmic tail.393 The middle of the protein forms a large extracellular loop containing a more central hydrophobic region.383,395 This large extracellular domain has four apyrase conserved regions (ACR) which were suggested to contain the sites of catalytic activities.396,397 The four ACR are highly conserved throughout the plant and animal kingdoms suggesting their importance in the biological function of CD39.396,398 A fifth ACR (termed ACR-5)

325

has been described in the C-terminal region of the extracellular domain.399,400 The ACR-4 in CD39 was suggested to contain the putative γ-phosphate binding motif, to be highly homologous with the actin-HSP70hexokinase superfamily,396 and to be involved in ATP hydrolysis.397 ACR-1 has been proposed to be the β-phosphate binding domain by analogy with the same superfamily396 and to play a role in ADP hydrolysis.397 The enzyme’s ADPase (but not ATPase) activity depends on the presence of divalent cations, with Ca21 preferred over Mg21.398 Heterologous interactions between both transmembrane domains of CD39 cause the tetramerization of the enzyme in the plasma membrane, which increases its activity over the monomeric form.401 Human CD39 has six potential N-linked glycosylation sites.383,393 The extent of glycosylation is different in endothelial cells, platelets, and leukocytes.394 The enzymatic activity of CD39 was reported to remain essentially unaltered by deglycosylation395 after the protein is properly folded and targeted to the membrane surface.402 CD39 also has several sites that may be modified by ectoprotein kinases,403,404 a few potential phosphorylation sites for intracellular PKC,403 as well as one N-terminal palmitoylation site.405 Oxidative modifications and proteolytic cleavages may modulate and regulate the enzymatic activity of CD39.406,407 CD39 is preferentially localized in caveolae of HUVECs and COS-7 cells,408 a process which is mediated by S-palmitoylation at the residue Cys13 of the intracytoplasmic N-terminal region.405 The activity of CD39 is cholesterol-dependent, whereby depletion or sequestering of membrane cholesterol results in inhibition of the enzymatic activity.409 The absence of caveolin-1 and the subsequent loss of caveolae formation does not affect the enzymatic activity or the targeting of CD39 to the membrane, as large aggregates of endogenous CD39 were found to colocalize with membrane-anchored CD73 and lipid rafts.409

B. The Effect of Endothelial CD39 on Platelet Reactivity Nucleotides serve as intracellular energy sources as well as extracellular signaling molecules, and they can be released upon cellular activation or injury and induce biological responses through specific receptors.384 Some of them can have pathological consequences, for example, the excessive ADP released from platelets and injured tissue have prothrombotic effects.2 Endothelial CD39 does not act on platelets per se; instead it reacts with the ADP released from activated

I. PLATELET BIOLOGY

326

17. INHIBITION OF PLATELET FUNCTION BY THE ENDOTHELIUM

platelets.391 Upon activation, platelet release ADP from their dense granules leads to further activation and aggregation of platelets. By hydrolyzing ADP to AMP, CD39 prevents further prothrombotic platelet activation387 with consequent restoration of platelets to the resting state.410 Activation of platelets with collagen or low levels of thrombin is inhibited by CD39.386 solCD39 strongly inhibits aggregation of human platelets induced by collagen, ADP, arachidonate, and thrombin receptor agonist peptide (TRAP).411,412 Platelet-released ADP is transiently hydrolyzed to AMP, which is further broken down to adenosine by endothelial CD73 (Figure 17-1).386,388 Adenosine, an antithrombotic and anti-inflammatory mediator,3 blocks ADP-induced platelet activation and aggregation413 by elevating intracellular cyclic AMP levels and blocking calcium mobilization,414 through the adenosine A2 receptor on platelets.415,416 A study of transgenic CD73/ mice supports these findings by demonstrating a significant reduction in the occlusion time of the carotid artery, as well as a reduction in the tail bleeding time compared to wild-type mice.417 Platelet cAMP levels, but not cGMP levels, were also found to be significantly lower in these mice.417 Thus, endothelial CD39 and CD73 act in concert to prevent platelet activation and aggregation, while neither NO nor prostacyclin are involved in their inhibitory activity.387,413 ADP and ATP are also substrates for the purinergic type 2 receptors (P2) (see Chapter 14). Several P2 receptors have been localized on megakaryocytes, platelets,418420 as well as on endothelial cells421,422 (see Chapter 14). ATP is a competitive antagonist of ADP for P2Y receptors on human platelets.423 The situation is further complicated by purinergic receptor desensitization where adenine nucleotides could also limit ADP-induced platelet activation423 and cause platelet dysfunction.424 CD39 appears to modulate the functional expression of P2Y receptors. The coexpression of CD39 and various P2 receptors in endothelial cells as well as platelets has been confirmed.394 A more recent study observed that P2Y1 and CD39 colocalize in the caveolae of endothelial cells.425 ADP and ATP released from activated platelets also affect the endothelium of the vessel wall. Through various G-protein-coupled P2 receptors both molecules stimulate the release of NO and prostacyclin from endothelial cells422,426 via a phospholipase C-mediated pathway that leads to increased intracellular Ca21 and subsequent activation of eNOS and phospholipase A2. This negative feedback mechanism can further limit the extent of platelet aggregation.422 The strong thromboregulatory potential of CD39 has been confirmed by experiments in which prostacyclin and NO production in the endothelium was

blocked by treatment with aspirin and hemoglobin, respectively. The inhibition of platelet activation and aggregation was maintained due to the presence of CD39 in the endothelial cell membrane.387,391,414 CD39 is therefore able to act independently of NO and prostacyclin; however, the presence of these thromboregulatory molecules enhances its activity.389 Interestingly, activation of endothelial cells by proinflammatory cytokines like tumor necrosis factor α (TNFα) or exposure to oxidative stress leads to loss of the enzymatic activity of CD39,386,406,407 but not the cell surface expression.406 Cholesterol depletion of CD39-containing membranes causes a significant delay in the inhibition of platelet aggregation and a substantial decrease in platelet disaggregation.409 Although the function of endothelial CD39 has been related to the inhibition of platelet activation and aggregation by ADP/ATP hydrolysis,386387 the function of CD39 expressed on platelets and megakaryocytes has not yet been established. Interesting, vigorous exercise in men involves a reduction in CD39 expression in platelets but an increase in expression on Blymphocytes.427

C. CD39  In Vivo Studies The clinical relevance of CD39 has been primarily established by animal studies. Recombinant solCD39 is able to inhibit platelet aggregation,428 remaining active over an extended period of time. Following reperfusion injury429 or after exposure to oxidative stress during vascular inflammation,406 the enzymatic and antithrombotic properties of endothelial CD39 are rapidly lost. Locally administered soluble apyrase (a soluble enzyme with identical function to CD39) was found to be beneficial in inhibiting platelet reactivity within the vasculature of transplanted organs.430 Thus, targeted expression of a stable and active CD39 could be an effective therapeutic means to intervene in vascular inflammation395 and transplantation-associated diseases.431 Administration of solCD39 either before or 3 hours after arterial occlusion improves the neurological score and reduces the infarct size in a rat stroke model.432 SolCD39 may, therefore, have potential for the treatment of focal ischemic stroke in humans.432 In sympathetic nerve endings from guinea pig hearts, neuronal ATP enhances norepinephrine exocytosis. Treatment with solCD39 significantly attenuates norepinephrine release,433 suggesting that CD39 could provide cardioprotection by reducing ATP-mediated norepinephrine release. In a murine stroke model driven by an excessive platelet recruitment, solCD39 reduced the extent of stroke without increasing intracerebral hemorrhage.433

I. PLATELET BIOLOGY

327

IV. CD39 (NTPDase-1)

The transgenic expression of human CD39 (hCD39) in mice has been investigated in different disease settings. Mice that express hCD39, at base line, have no spontaneous bleeding tendencies; however, platelet aggregation is impaired, bleeding times are prolonged, and the mice are resistance to systemic thromboembolism.389 In denudated rat aortas adenovirally transfected with hCD39, neointimal formation was reduced compared to controls, which was associated with a decrease in vascular smooth muscle cell proliferation.434 In mice with dysfunctional CD39, bleeding times were normal, but they had increased cerebral infarct volumes and reduced postischemic perfusion. Treatment with solCD39 restored postischemic cerebral perfusion and rescued the mice from cerebral injury, suggesting a protective function of CD39 in stroke.435 WT mice that have undergone wire-induced femoral artery injury have a reduced platelet adhesion, leukocyte accumulation, and neointimal hyperplasia with treatment of human solCD39 compared to saline controls.436 Additionally, treating mice with solCD39 that is incorporated into lipid bilayer increases enzymatic activity and decreased mortality in mice that have undergone thromboplastin-induce thromboembolism.437 Studies with CD39 knockout mice have investigated the lack of this gene on cardiovascular properties and disease development.389,424,435,438440 Although CD39deficient (CD39/) mice and wild-type mice displayed no differences in their phenotype, the CD39/ mice had substantially prolonged bleeding times and prolonged platelet plug formation times with minimally perturbed coagulation parameters.424 This was due to a reduced response of the CD39/ platelets to ADP, collagen, and low doses of thrombin. The platelet count was found to be 20% lower in CD39/mice compared to wild-type mice, but the CD39/platelets had no morphological abnormalities. The interaction between platelet and injured vasculature in vivo was considerably reduced.424 Platelet dysfunction was reversible and correlated with purinergic receptor P2Y1 desensitization, suggesting a dual role for CD39 in hemostasis and thrombotic reactions.424 The CD39/mice were also very sensitive to ischemia-reperfusion injuries as they were incapable of producing adenosine at the local endothelial vessel wall layer. Treatment with apyrase reversed the effect and protected the mice.424 Using endothelial cell cultures from the CD39/mice, it was shown both ADPase and ATPase activities were diminished.424 This suggests that other nucleotidases associated with endothelial cells, contribute only minimally to the hydrolysis of extracellular adenine nucleotides. Other disease models have been studied using CD39/ mice, including intestinal ischemia and reperfusion injuries.438 In CD39-null or -deficient mice, 80% died due to intestinal injury as compared to wild-type

mice. Apyrase supplementation protected all wild-type mice from intestinal ischemia-related death, but did not fully protect CD39/ mice. Adenosine treatment failed to improve the survival rate. Platelet adherence to postcapillary venules in wild-type mice was significantly decreased and vascular integrity was well preserved following apyrase treatment. The potential of CD39 to maintain vascular integrity suggests potential pharmacological benefit in mesenteric ischemic injury.438 Balloon injury in rabbit arteries decreases native CD39 activity, which was reversed by adenovirus-mediated gene transfer of CD39; however, platelet deposition was not altered.439 Studies of ApoE/ mice showed that deletion of CD39 accelerates the development of atherosclerotic lesions, a process reversed by solCD39 administration.440 Furthermore, supplementation of solCD39 slows atherosclerosis in CD39 wild-type/ ApoE/ mice.440 In conclusion, the deletion of CD39 renders the mice sensitive to vascular injury. Supplementation of CD39 by somatic gene transfer or administration of soluble CD39 has been shown to be advantageous in models of transplantation, inflammation, and atherosclerosis.

D. Polymorphisms of Endothelial CD39 No polymorphisms of CD39 have been associated with cardiovascular diseases. However, a few have been associated with diabetes and colitis (Table 17-4). A two-SNP haplotype is associated with an increased risk for type 2 diabetes in African Americans. This SNP results in a higher expression level of CD39. A four-SNP haplotype was found to be protective against diabetes. This SNP results in a lower expression level of CD39 compared to control subjects.441 Another CD39 polymorphism has been associated with an increased risk for Crohn’s disease. This SNP results in a lower expression level of CD39. Mouse studies using a CD39/ model, showed that dextran sodium sulfate-induced colitis was more severe in the knockout mice compared to WT.442 However, in another study, CD39/ mice given trinitrobenzene sulfonic acid-induced colitis had improved survival and a TABLE 17-4 Polymorphisms of the Ecto-nucleotidase CD39 Gene and Associated Disease States Disease Polymorphism/ VNTR/SNP/ Haplotype

Diabetes AG Haplotype

Other 441

Crohn’s Disease  Loss of Function Mutation442

GAGA Haplotype441 Abbreviations: SNP, single nucleotide polymorphisms; VNTR, variable number of tandem repeat polymorphisms.

I. PLATELET BIOLOGY

328

17. INHIBITION OF PLATELET FUNCTION BY THE ENDOTHELIUM

reduced inflammatory response compared to WT mice.443 Thus, although CD39 polymorphisms have been associated with colitis, the exact role of CD39 in the progression of this disease remains controversial.

V. SUMMARY Endothelial thromboregulation requires several forms of communication between endothelial cells and platelets in order to regulate clot formation, but still allow proper blood vessel repair. The three primary and functionally independent pathways involve NO, prostacyclin, and the ecto-nucleotidase CD39. Taken together, the endothelium utilizes these pathways to regulate and prevent uncontrolled platelet activation, aggregation, and adhesion to the vessel wall. In addition, the platelet itself possesses several protective and counter-regulatory mechanisms to balance its own prothrombotic actions and to stimulate endothelial antithrombotic factors and processes. As each of these pathways is defined, it is becoming more apparent that the underlying thromboregulatory mechanisms and enzymatic systems involved are interconnected. Disturbances of these balanced systems contribute to not only the pathophysiology of vascular thrombotic diseases, but other diseases including hemorrhage. Understanding how we can target these pathways to control platelet function without causing hemorrhage or thrombosis will be key in developing effective therapeutics for vascular diseases in the future.

Acknowledgements The authors thank Sybille Rex for her contribution to the writing of this chapter.

References [1] Marcus AJ, Safier LB, Kaminski WE, Islam N, Hajjar KA, Jendraschak E, et al. Principles of thromboregulation: control of platelet reactivity in vascular disease. Adv Prostaglandin Thromboxane Leukot Res 1995;23:4138. [2] Marcus AJ, Safier LB. Thromboregulation: multicellular modulation of platelet reactivity in hemostasis and thrombosis. Faseb J 1993;7(6):51622. [3] Marcus AJ, Broekman MJ, Drosopoulos JH, Islam N, Pinsky DJ, Sesti C, et al. Heterologous cell-cell interactions: thromboregulation, cerebroprotection and cardioprotection by CD39 (NTPDase-1). J Thromb Haemost 2003;1(12):2497509. [4] Marcus AJ, Broekman MJ, Drosopoulos JH, Olson KE, Islam N, Pinsky DJ, et al. Role of CD39 (NTPDase-1) in thromboregulation, cerebroprotection, and cardioprotection. Semin Thromb Hemost 2005;31(2):23446. [5] Furchgott RF, Zawadzki JV. The obligatory role of endothelial cells in the relaxation of arterial smooth muscle by acetylcholine. Nature 1980;288(5789):3736.

[6] Stamler JS, Singel DJ, Loscalzo J. Biochemistry of nitric oxide and its redox-activated forms. Science 1992;258(5090):1898902. [7] Walford G, Loscalzo J. Nitric oxide in vascular biology. J Thromb Haemost 2003;1(10):21128. [8] Radi R, Beckman JS, Bush KM, Freeman BA. Peroxynitrite oxidation of sulfhydryls. The cytotoxic potential of superoxide and nitric oxide. J Biol Chem 1991;266(7):424450. [9] Rubanyi G, Vanhoutte P. Superoxide anions and hyperoxia inactivate endothlium-derived relaxing factor. Am J Physiol 1986;250(5, Pt. 2):H8827. [10] Rubanyi GM, Vanhoutte PM. Oxygen-derived free radicals, endothelium, and responsiveness of vascular smooth muscle. Am J Physiol 1986;250(5, Pt. 2):H81521. [11] Lacza Z, Snipes JA, Zhang J, Horvath EM, Figueroa JP, Szabo C, et al. Mitochondrial nitric oxide synthase is not eNOS, nNOS or iNOS. Free Radic Biol Med 2003;35(10):121728. [12] Carreras MC, Franco MC, Peralta JG, Poderoso JJ. Nitric oxide, complex I, and the modulation of mitochondrial reactive species in biology and disease. Mol Aspects Med 2004;25 (12):12539. [13] Haynes V, Elfering S, Traaseth N, Giulivi C. Mitochondrial nitric-oxide synthase: enzyme expression, characterization, and regulation. J Bioenerg Biomembr 2004;36(4):3416. [14] Ghafourifar P, Cadenas E. Mitochondrial nitric oxide synthase. Trends Pharmacol Sci 2005;26(4):1905. [15] Pollock JS, Forstermann U, Mitchell JA, Warner TD, Schmidt HH, Nakane M, et al. Purification and characterization of particulate endothelium-derived relaxing factor synthase from cultured and native bovine aortic endothelial cells. Proc Natl Acad Sci USA 1991;88(23):104804. [16] Nathan C. Nitric oxide as a secretory product of mammalian cells. Faseb J 1992;6(12):305164. [17] Michel T, Feron O. Nitric oxide synthases: Which, where, how, and why? J Clin Invest 1997;100(9):214652. [18] Shaul PW, North AJ, Wu LC, Wells LB, Brannon TS, Lau KS, et al. Endothelial nitric oxide synthase is expressed in cultured human bronchiolar epithelium. J Clin Invest 1994;94 (6):22316. [19] German Z, Chambliss KL, Pace MC, Arnet UA, Lowenstein CJ, Shaul PW. Molecular basis of cell-specific endothelial nitricoxide synthase expression in airway epithelium. J Biol Chem 2000;275(11):81839. [20] Mehta JL, Chen LY, Kone BC, Mehta P, Turner P. Identification of constitutive and inducible forms of nitric oxide synthase in human platelets. J Lab Clin Med 1995;125:3707. [21] Sase K, Michel T. Expression of constitutive endothelial nitric oxide synthase in human blood platelets. Life Sci 1995;57:204955. [22] Zhou Q, Hellermann G, Solomonson L. Nitric oxide release from resting human platelets. Thromb Res 1995;77:8796. [23] Alderton WK, Cooper CE, Knowles RG. Nitric oxide synthases: structure, function and inhibition. Biochem J 2001;357(Pt. 3):593615. [24] Raman CS, Li H, Martasek P, Kral V, Masters BS, Poulos TL. Crystal structure of constitutive endothelial nitric oxide synthase: a paradigm for pterin function involving a novel metal center. Cell 1998;95(7):93950. [25] Fischmann TO, Hruza A, Niu XD, Fossetta JD, Lunn CA, Dolphin E, et al. Structural characterization of nitric oxide synthase isoforms reveals striking active-site conservation. Nat Struct Biol 1999;6(3):23342. [26] Crane BR, Rosenfeld RJ, Arvai AS, Ghosh DK, Ghosh S, Tainer JA, et al. N-terminal domain swapping and metal ion binding in nitric oxide synthase dimerization. Embo J 1999;18(22):627181.

I. PLATELET BIOLOGY

REFERENCES

[27] Marletta MA, Yoon PS, Iyengar R, Leaf CD, Wishnok JS. Macrophage oxidation of L-arginine to nitrite and nitrate: nitric oxide is an intermediate. Biochemistry 1988;27(24):870611. [28] Lamas S, Marsden PA, Li GK, Tempst P, Michel T. Endothelial nitric oxide synthase: molecular cloning and characterization of a distinct constitutive enzyme isoform. Proc Natl Acad Sci USA 1992;89(14):634852. [29] Robinson LJ, Michel T. Mutagenesis of palmitoylation sites in endothelial nitric oxide synthase identifies a novel motif for dual acylation and subcellular targeting. Proc Natl Acad Sci USA 1995;92(25):1177680. [30] Shaul PW. Regulation of endothelial nitric oxide synthase: Location, location, location. Annu Rev Physiol 2002;64:74974. [31] Feron O, Belhassen L, Kobzik L, Smith TW, Kelly RA, Michel T. Endothelial nitric oxide synthase targeting to caveolae. Specific interactions with caveolin isoforms in cardiac myocytes and endothelial cells. J Biol Chem 1996;271(37):228104. [32] Conrad PA, Smart EJ, Ying YS, Anderson RG, Bloom GS. Caveolin cycles between plasma membrane caveolae and the Golgi complex by microtubule-dependent and microtubuleindependent steps. J Cell Biol 1995;131(6, Pt. 1):142133. [33] Shaul PW, Smart EJ, Robinson LJ, German Z, Yuhanna IS, Ying Y, et al. Acylation targets emdothelial nitric-oxide synthase to plasmalemmal caveolae. J Biol Chem 1996;271 (11):651822. [34] Bucci M, Gratton JP, Rudic RD, Acevedo L, Roviezzo F, Cirino G, et al. In vivo delivery of the caveolin-1 scaffolding domain inhibits nitric oxide synthesis and reduces inflammation. Nat Med 2000;6(12):13627. [35] Fleming I, Busse R. Molecular mechanisms involved in the regulation of the endothelial nitric oxide synthase. Am J Physiol Regul Integr Comp Physiol 2003;284(1):R112. [36] Rubanyi GM, Romero JC, Vanhoutte PM. Flow-induced release of endothelium-derived relaxing factor. Am J Physiol 1986;250(6, Pt. 2):H11459. [37] Fulton D, Gratton JP, McCabe TJ, Fontana J, Fujio Y, Walsh K, et al. Regulation of endothelium-derived nitric oxide production by the protein kinase Akt. Nature 1999;399(6736):597601. [38] Dimmeler S, Fleming I, Fisslthaler B, Hermann C, Busse R, Zeiher AM. Activation of nitric oxide synthase in endothelial cells by Akt-dependent phosphorylation. Nature 1999;399 (6736):6015. [39] Ross RL, Serock MR, Khalil RA. Experimental benefits of sex hormones on vascular function and the outcome of hormone therapy in cardiovascular disease. Curr Cardiol Rev 2008;4 (4):30922. [40] Zerr-Fouineau M, Jourdain M, Boesch C, Hecker M, Bronner C, Schini-Kerth VB. Certain progestins prevent the enhancing effect of 17beta-estradiol on NO-mediated inhibition of platelet aggregation by endothelial cells. Arterioscler Thromb Vasc Biol 2009;29(4):58693. [41] Simoncini T, Caruso A, Garibaldi S, Fu XD, Giretti MS, Baldacci C, et al. Activation of nitric oxide synthesis in human endothelial cells using nomegestrol acetate. Obstet Gynecol 2006;108(4):96978. [42] Gonzalez G, Alvarado-Vasquez N, Fernandez GJ, Cruz-Robles D, Del Valle L, Pinzon E, et al. The antithrombotic effect of the aminoestrogen prolame (N-(3-hydroxy-1,3,5(10)-estratrien-17BYL)-3-hydroxypropylamine) is linked to an increase in nitric oxide production by platelets and endothelial cells. Atherosclerosis 2010;208(1):628. [43] Papapetropoulos A, Rudic RD, Sessa WC. Molecular control of nitric oxide synthases in the cardiovascular system. Cardiovasc Res 1999;43(3):50920.

329

[44] Andrew PJ, Mayer B. Enzymatic function of nitric oxide synthases. Cardiovasc Res 1999;43(3):52131. [45] Govers R, Rabelink TJ. Cellular regulation of endothelial nitric oxide synthase. Am J Physiol Renal Physiol 2001;280(2): F193206. [46] Beckman J, Beckman T, Chen J, Marshall P, Freeman B. Apparent hydroxyl radical production by peroxynitrite: Implications for endothelial injury from nitric oxide and superoxide. Proc Natl Acad Sci USA 1990;87:16204. [47] Radi R, Beckman JS, Bush KM, Freeman BA. Peroxynitriteinduced membrane lipid peroxidation: the cytotoxic potential of superoxide and nitric oxide. Arch Biochem Biophys 1991;288 (2):4817. [48] Ignarro LJ. Biological actions and properties of endotheliumderived nitric oxide formed and released from artery and vein. Circ Res 1989;65(1):121. [49] de Graaf JC, Banga JD, Moncada S, Palmer RM, de Groot PG, Sixma JJ. Nitric oxide functions as an inhibitor of platelet adhesion under flow conditions. Circulation 1992;85: 228490. [50] Azuma H, Ishikawa M, Sekizaki S. Endothelium-dependent inhibition of platelet aggregation. Br J Pharmacol 1986;88 (2):4115. [51] Broekman MJ, Eiroa AM, Marcus AJ. Inhibition of human platelet reactivity by endothelium-derived relaxing factor from human umbilical vein endothelial cells in suspension: blockade of aggregation and secretion by an aspirin-insensitive mechanism. Blood 1991;78(4):103340. [52] Stamler J, Mendelsohn ME, Amarante P, Smick D, Andon N, Davies PF, et al. N-acetylcysteine potentiates platelet inhibition by endothelium-derived relaxing factor. Circ Res 1989;65:78995. [53] Cooke JP, Stamler J, Andon N, Davies PF, McKinley G, Loscalzo J. Flow stimulates endothelial cells to release a nitrovasodilator that is potentiated by reduced thiol. Am J Physiol 1990;259(3, Pt. 2):H80412. [54] Bellamy TC, Garthwaite J. The receptor-like properties of nitric oxide-activated soluble guanylyl cyclase in intact cells. Mol Cell Biochem 2002;230(12):16576. [55] Murad F, Mittal CK, Arnold WP, Katsuki S, Kimura H. Guanylate cyclase: activation by azide, nitro compounds, nitric oxide, and hydroxyl radical and inhibition by hemoglobin and myoglobin. Adv Cyclic Nucleotide Res 1978;9:14558. [56] Warner TD. Influence of endothelial mediators on the vascular smooth muscle and circulating platelets and blood cells. Int Angiol 1996;15(2):939. [57] Beavo JA, Brunton LL. Cyclic nucleotide research—still expanding after half a century. Nat Rev Mol Cell Biol 2002;3 (9):7108. [58] Hanafy KA, Krumenacker JS, Murad F. NO, nitrotyrosine, and cyclic GMP in signal transduction. Med Sci Monit 2001;7 (4):80119. [59] Feil R, Lohmann SM, de Jonge H, Walter U, Hofmann F. Cyclic GMP-dependent protein kinases and the cardiovascular system: insights from genetically modified mice. Circ Res 2003;93 (10):90716. [60] Moro M, Russel R, Cellek S, Lizasoain I, Su Y, Darley-Usman V, et al. cGMP mediates the vascular and platelet action of nitric oxide: confirmation using an inhibitor of the soluble guanylyl cylase. Proc Natl Acad Sci USA 1996;93:14805. [61] Rao GH, Krishnamurthi S, Raij L, White JG. Influence of nitric oxide on agonist-mediated calcium mobilization in platelets. Biochem Med Metab Biol 1990;43(3):2715.

I. PLATELET BIOLOGY

330

17. INHIBITION OF PLATELET FUNCTION BY THE ENDOTHELIUM

[62] Walter U, Geiger J, Haffner C, Markert T, Nehls C, Silber RE, et al. Platelet-vessel wall interactions, focal adhesions, and the mechanism of action of endothelial factors. Agents Actions Suppl 1995;45:25568. [63] Jin RC, Voetsch B, Loscalzo J. Endogenous mechanisms of inhibition of platelet function. Microcirculation 2005;12(3): 24758. [64] Michelson AD, Benoit SE, Furman MI, Breckwoldt WL, Rohrer MJ, Barnard MR, et al. Effects of nitric oxide/endotheliumderived relaxing factor on platelet surface glycoproteins. Am J Physiol 1996;270(5, Pt. 2):H16408. [65] Mendelsohn ME, O’Neill S, George D, Loscalzo J. Inhibition of fibrinogen binding to human platelets by S-nitroso-N-acetylcysteine. J Biol Chem 1990;265(31):1902834. [66] Pigazzi A, Heydrick S, Folli F, Benoit S, Michelson A, Loscalzo J. Nitric oxide inhibits thrombin receptor-activating peptide-induced phosphoinositide 3-kinase activity in human platelets. J Biol Chem 1999;274(20):1436875. [67] Horstrup K, Jablonka B, Honig-Liedl P, Just M, Kochsiek K, Walter U. Phosphorylation of focal adhesion vasodilator-stimulated phosphoprotein at Ser157 in intact human platelets correlates with fibrinogen receptor inhibition. Eur J Biochem 1994;225(1):217. [68] Radomski M, Moncada S. Regulation of vascular homeostasis by nitric oxide. Thrombosis Haemostasis 1993;70:3641. [69] Wang GR, Zhu Y, Halushka PV, Lincoln TM, Mendelsohn ME. Mechanism of platelet inhibition by nitric oxide: in vivo phosphorylation of thromboxane receptor by cyclic GMPdependent protein kinase. Proc Natl Acad Sci USA 1998;95 (9):488893. [70] Murohara T, Parkinson S, Waldman S, Lefer A. Inhibition of nitric oxide biosynthesis promotes P-selectin expression in platelets. Arterioscler Thromb Vasc Biol 1995;15:206875. [71] Stamler JS, Simon DI, Osborne JA, Mullins ME, Jaraki O, Michel T, et al. S-nitrosylation of proteins with nitric oxide: synthesis and characterization of biologically active compounds. Proc Natl Acad Sci USA 1992;89(1):4448. [72] Stamler JS, Simon DI, Jaraki O, Osborne JA, Francis S, Mullins M, et al. S-nitrosylation of tissue-type plasminogen activator confers vasodilatory and antiplatelet properties on the enzyme. Proc Natl Acad Sci USA 1992;89(17):808791. [73] Scharfstein JS, Keaney Jr JF, Slivka A, Welch GN, Vita JA, Stamler JS, et al. In vivo transfer of nitric oxide between a plasma protein-bound reservoir and low molecular weight thiols. J Clin Invest 1994;94(4):14329. [74] Liu Z, Rudd MA, Freedman JE, Loscalzo J. S-Transnitrosation reactions are involved in the metabolic fate and biological actions of nitric oxide. J Pharmacol Exp Ther 1998;284 (2):52634. [75] de Belder AJ, MacAllister R, Radomski MW, Moncada S, Vallance PJ. Effects of S-nitroso-glutathione in the human forearm circulation: evidence for selective inhibition of platelet activation. Cardiovasc Res 1994;28(5):6914. [76] Crane MS, Ollosson R, Moore KP, Rossi AG, Megson IL. Novel role for low molecular weight plasma thiols in nitric oxidemediated control of platelet function. J Biol Chem 2002;277 (49):4685863. [77] Crane MS, Rossi AG, Megson IL. A potential role for extracellular nitric oxide generation in cGMP-independent inhibition of human platelet aggregation: biochemical and pharmacological considerations. Br J Pharmacol 2005;144(6):84959. [78] Malinski T, Radomski MW, Taha Z, Moncada S. Direct electrochemical measurement of nitric oxide released from human platelets. Biochem Biophys Res Commun 1993;194:9605.

[79] Radomski MW, Palmer RMJ, Moncada S. An L-arginine/nitric oxide pathway present in human platelets regulates aggregation. Proc Natl Acad Sci USA 1990;87:51937. [80] Freedman JE, Loscalzo J, Barnard MR, Alpert C, Keaney Jr JF, Michelson A. Nitric oxide released from activated platelets inhibits platelet recruitment. J Clin Invest 1997;100:3506. [81] Freedman J, Sauter R, Battinelli B, Ault A, Knowles C, Huang P, et al. Deficient platelet-derived nitric oxide and enhanced hemostasis in mice lacking the NOS3 gene. Circ Res 1999;84:141621. [82] Patel B, Sharifi M, Milward AD, Oberprieler NG, Gibbins JM, Parkin S, et al. Platelet nitric oxide synthase is activated by tyrosine dephosphorylation: possible role for SHP-1 phosphatase. J Thromb Haemost 2006;4(11):242332. [83] Gresele P, Pignatelli P, Guglielmini G, Carnevale R, Mezzasoma AM, Ghiselli A, et al. Resveratrol, at concentrations attainable with moderate wine consumption, stimulates human platelet nitric oxide production. J Nutr 2008;138 (9):16028. [84] Wu GJ, Lee JJ, Chou DS, Jayakumar T, Hsiao G, Chen WF, et al. Inhibitory signaling of 17beta-estradiol in platelet activation: the pivotal role of cyclic AMP-mediated nitric oxide synthase activation. Eur J Pharmacol 2010;649 (13):1409. [85] Takeuchi K, Watanabe H, Tran QK, Ozeki M, Sumi D, Hayashi T, et al. Nitric oxide: inhibitory effects on endothelial cell calcium signaling, prostaglandin I2 production and nitric oxide synthase expression. Cardiovasc Res 2004;62 (1):194201. [86] Loscalzo J. Nitric oxide insufficiency, platelet activation, and arterial thrombosis. Circ Res 2001;88(8):75662. [87] Krotz F, Sohn HY, Pohl U. Reactive oxygen species: players in the platelet game. Arterioscler Thromb Vasc Biol 2004;24 (11):198896. [88] Friebe A, Koesling D. Regulation of nitric oxide-sensitive guanylyl cyclase. Circ Res 2003;93(2):96105. [89] Salvemini D, Radziszewski W, Mollace V, Moore A, Willoughby D, Vane J. Diphenylene iodonium, an inhibitor of free radical formation, inhibits platelet aggregation. Eur J Pharmacol 1991;199(1):158. [90] Chlopicki S, Olszanecki R, Janiszewski M, Laurindo FR, Panz T, Miedzobrodzki J. Functional role of NADPH oxidase in activation of platelets. Antioxid Redox Signal 2004;6(4):6918. [91] Clutton P, Miermont A, Freedman JE. Regulation of endogenous reactive oxygen species in platelets can reverse aggregation. Arterioscler Thromb Vasc Biol 2004;24 (1):18792. [92] Jahn B, Hansch GM. Oxygen radical generation in human platelets: dependence on 12-lipoxygenase activity and on the glutathione cycle. Int Arch Allergy Appl Immunol 1990;93 (1):739. [93] Freedman JE, Frei B, Welch GN, Loscalzo J. Glutathione peroxidase potentiates the inhibition of platelet function by S-nitrosothiols. J Clin Invest 1995;96(1):394400. [94] Igarashi J, Miyoshi M, Hashimoto T, Kubota Y, Kosaka H. Hydrogen peroxide induces S1P1 receptors and sensitizes vascular endothelial cells to sphingosine 1-phosphate, a plateletderived lipid mediator. Am J Physiol Cell Physiol 2007;292(2): C7408. [95] Ignarro LJ, Napoli C. Novel features of nitric oxide, endothelial nitric oxide synthase, and atherosclerosis. Curr Atheroscler Rep 2004;6(4):2817. [96] Kawashima S, Yokoyama M. Dysfunction of endothelial nitric oxide synthase and atherosclerosis. Arterioscler Thromb Vasc Biol 2004;24(6):9981005.

I. PLATELET BIOLOGY

331

REFERENCES

[97] Cohen RA. The role of nitric oxide and other endotheliumderived vasoactive substances in vascular disease. Prog Cardiovasc Dis 1995;38(2):10528. [98] Harrison DG. Endothelial dysfunction in atherosclerosis. Basic Res Cardiol 1994;89(Suppl. 1):87102. [99] Flavahan NA. Atherosclerosis or lipoprotein-induced endothelial dysfunction. Potential mechanisms underlying reduction in EDRF/nitric oxide activity. Circulation 1992;85 (5):192738. [100] Khan R, Kirschenbaum LA, LaRow C, Berna G, Griffin K, Astiz ME. Augmentation of platelet and endothelial cell eNOS activity decreases sepsis-related neutrophil-endothelial cell interactions. Shock 2010;33(3):2426. [101] Huang PL, Huang Z, Mashimo H, Bloch KD, Moskowitz MA, Bevan JA, et al. Hypertension in mice lacking the gene for endothelial nitric oxide synthase. Nature 1995;377:23942. [102] Iafrati MD, Vitseva O, Tanriverdi K, Blair P, Rex S, Chakrabarti S, et al. Compensatory mechanisms influence hemostasis in setting of eNOS deficiency. Am J Physiol Heart Circ Physiol 2005;288(4):H162732. [103] Dai X, Faber JE. Endothelial nitric oxide synthase deficiency causes collateral vessel rarefaction and impairs activation of a cell cycle gene network during arteriogenesis. Circ Res 2010;106(12):187081. [104] Jones SP, Greer JJ, van Haperen R, Duncker DJ, de Crom R, Lefer DJ. Endothelial nitric oxide synthase overexpression attenuates congestive heart failure in mice. Proc Natl Acad Sci USA 2003;100(8):48916. [105] Jones SP, Greer JJ, Kakkar AK, Ware PD, Turnage RH, Hicks M, et al. Endothelial nitric oxide synthase overexpression attenuates myocardial reperfusion injury. Am J Physiol Heart Circ Physiol 2004;286(1):H27682. [106] Kawashima S, Yamashita T, Ozaki M, Ohashi Y, Azumi H, Inoue N, et al. Endothelial NO synthase overexpression inhibits lesion formation in mouse model of vascular remodeling. Arterioscler Thromb Vasc Biol 2001;21 (2):2017. [107] Ozaki M, Kawashima S, Yamashita T, Hirase T, Namiki M, Inoue N, et al. Overexpression of endothelial nitric oxide synthase accelerates atherosclerotic lesion formation in apoEdeficient mice. J Clin Invest 2002;110(3):33140. [108] Vita JA, Treasure CB, Nabel EG, McLenachan JM, Fish R, Yeung AC, et al. Coronary vasomotor response to acetylcholine relates to risk factors for coronary artery disease. Circulation 1990;81:4917. [109] Fitzgerald DJ, Roy L, Catella F, FitzGerald A. Platelet activation in unstable coronary disease. N Engl J Med 1986;315:9839. [110] Freedman JE, Ting B, Hankin B, Loscalzo J, Keaney JFJ, Vita JA. Impaired platelet production of nitric oxide in patients with unstable angina. Circulation 1998;98:14816. [111] Freedman JE, Ting B, Hankin B, Loscalzo J, Keaney Jr JF, Vita JA. Impaired platelet production of nitric oxide predicts presence of acute coronary syndromes. Circulation 1998;98 (15):14816. [112] Bossaller C, Habib GB, Yamamoto H, Williams C, Wells S, Henry PD. Impaired muscarinic endothelium-dependent relaxation and cyclic guanosine 5’-monophosphate formation in atherosclerotic human coronary artery and rabbit aorta. J Clin Invest 1986;79(79):1704. [113] Cool CD, Stewart JS, Werahera P, Miller GJ, Williams RL, Voelkel NF, et al. Three-dimensional reconstruction of pulmonary arteries in plexiform pulmonary hypertension using cellspecific markers. Evidence for a dynamic and heterogeneous

[114]

[115]

[116]

[117]

[118]

[119]

[120]

[121]

[122]

[123]

[124]

[125]

[126]

[127]

[128]

[129]

process of pulmonary endothelial cell growth. Am J Pathol 1999;155(2):4119. Giaid A, Saleh D. Reduced expression of endothelial nitric oxide synthase in the lungs of patients with pulmonary hypertension. N Engl J Med 1995;333(4):21421. Stagliano N, Zhao W, Prado R, Dewanjee M, Ginsberg M, Dietrich W. The effect of nitric oxide synthase inhibition on acute platelet accumulation and hemodynamic depression in a rat model of thromboembolic stroke. J Cereb Blood Flow Metab 1997;17:118290. Freedman JE, Loscalzo J, Benoit SE, Valeri CR, Barnard MR, Michelson AD. Decreased platelet inhibition by nitric oxide in two brothers with a history of arterial thrombosis. J Clin Invest 1996;97:97987. Goumas G, Tentolouris C, Tousoulis D, Stefanadis C, Toutouzas P. Therapeutic modification of the L-arginineeNOS pathway in cardiovascular diseases. Atherosclerosis 2001;154(2):25567. Folts JD, Stamler J, Loscalzo J. Intravenous nitroglycerin infusion inhibits cyclic blood flow responses caused by periodic platelet thrombus formation in stenosed canine coronary arteries. Circulation 1991;83(6):21227. Nong Z, Hoylaerts M, Van Pelt N, Collen D, Janssens S. Nitric oxide inhalation inhibits platelet aggregation and plateletmediated pulmonary thrombosis in rats. Circ Res 1997;81 (5):8659. Candipan RC, Wang BY, Buitrago R, Tsao PS, Cooke JP. Regression or progression. Dependency on vascular nitric oxide. Arterioscler Thromb Vasc Biol 1996;16(1):4450. Aji W, Ravalli S, Szabolcs M, Jiang XC, Sciacca RR, Michler RE, et al. L-arginine prevents xanthoma development and inhibits atherosclerosis in LDL receptor knockout mice. Circulation 1997;95(2):4307. Adams M, Forsyth C, Jessup W, Robinson J, Celermajer D. Oral L-arginine inhibits platelet aggregation but does not enhance endothelium-dependent dilation in healthy young men. J Am Coll Cardiol 1995;26:105461. Simon DI, Stamler JS, Loh E, Loscalzo J, Frances SA, Creager MA. Effect of nitric oxide synthase inhibition on bleeding time in humans. J Cardiovasc Pharm 1995;26:33942. Quyyumi AA, Dakak N, Andrews NP, Husain S, Arora S, Gilligan DM, et al. Nitric oxide activity in the human coronary circulation. Impact of risk factors for coronary atherosclerosis. J Clin Invest 1995;95(4):174755. Boger GI, Rudolph TK, Maas R, Schwedhelm E, Dumbadze E, Bierend A, et al. Asymmetric dimethylarginine determines the improvement of endothelium-dependent vasodilation by simvastatin: effect of combination with oral L-arginine. J Am Coll Cardiol 2007;49(23):227482. McCarty MF. Gamma-tocopherol may promote effective no synthase function by protecting tetrahydrobiopterin from peroxynitrite. Med Hypotheses 2007;69(6):136770. Armah CK, Jackson KG, Doman I, James L, Cheghani F, Minihane AM. Fish oil fatty acids improve postprandial vascular reactivity in healthy men. Clin Sci (Lond) 2008;114 (11):67986. Koltermann A, Hartkorn A, Koch E, Furst R, Vollmar AM, Zahler S. Ginkgo biloba extract EGb 761 increases endothelial nitric oxide production in vitro and in vivo. Cell Mol Life Sci 2007;64(13):171522. Moat SJ, Madhavan A, Taylor SY, Payne N, Allen RH, Stabler SP, et al. High- but not low-dose folic acid improves endothelial function in coronary artery disease. Eur J Clin Invest 2006;36(12):8509.

I. PLATELET BIOLOGY

332

17. INHIBITION OF PLATELET FUNCTION BY THE ENDOTHELIUM

[130] Sakakibara S, Murakami R, Takahashi M, Fushimi T, Murohara T, Kishi M, et al. Vinegar intake enhances flowmediated vasodilatation via upregulation of endothelial nitric oxide synthase activity. Biosci Biotechnol Biochem 2010;74(5):105561. [131] Randomised placebo-controlled trial of abciximab before and during coronary intervention in refractory unstable angina: the CAPTURE Study. Lancet. 1997;349(9063):14291435. [132] Kleiman NS, Lincoff AM, Ohman EM, Harrington RA. Glycoprotein IIb/IIIa inhibitors in acute coronary syndromes: pathophysiologic foundation and clinical findings. Am Heart J 1998;136(4, Pt. 2 Su.):S3242. [133] Heitzer T, Ollmann I, Koke K, Meinertz T, Munzel T. Platelet glycoprotein IIb/IIIa receptor blockade improves vascular nitric oxide bioavailability in patients with coronary artery disease. Circulation 2003;108(5):53641. [134] Keh D, Gerlach M, Kurer I, Seiler S, Kerner T, Falke KJ, et al. The effects of nitric oxide (NO) on platelet membrane receptor expression during activation with human alpha-thrombin. Blood Coagul Fibrinolysis 1996;7(6):61524. [135] Quinn MJ, Byzova TV, Qin J, Topol EJ, Plow EF. Integrin alphaIIbbeta3 and its antagonism. Arterioscler Thromb Vasc Biol 2003;23(6):94552. [136] Quinn MJ, Plow EF, Topol EJ. Platelet glycoprotein IIb/IIIa inhibitors: recognition of a two-edged sword? Circulation 2002;106(3):37985. [137] Comini L, Bachetti T, Cargnoni A, Bastianon D, Gitti GL, Ceconi C, et al. Therapeutic modulation of the nitric oxide: all ACE inhibitors are not equivalent. Pharmacol Res 2007;56 (1):428. [138] Morawietz H, Rohrbach S, Rueckschloss U, Schellenberger E, Hakim K, Zerkowski HR, et al. Increased cardiac endothelial nitric oxide synthase expression in patients taking angiotensin-converting enzyme inhibitor therapy. Eur J Clin Invest 2006;36(10):70512. [139] Keul P, Tolle M, Lucke S, von Wnuck Lipinski K, Heusch G, Schuchardt M, et al. The sphingosine-1-phosphate analogue FTY720 reduces atherosclerosis in apolipoprotein E-deficient mice. Arterioscler Thromb Vasc Biol 2007;27(3):60713. [140] Li P, Shibata R, Maruyama S, Kondo M, Ohashi K, Ouchi N, et al. Fenofibrate promotes ischemia-induced revascularization through the adiponectin-dependent pathway. Am J Physiol Endocrinol Metab 2010;299(4):E5606. [141] O’Donnell CJ, Larson MG, Feng D, Sutherland PA, Lindpaintner K, Myers RH, et al. Genetic and environmental contributions to platelet aggregation: the Framingham heart study. Circulation 2001;103(25):30516. [142] Tsukada T, Yokoyama K, Arai T, Takemoto F, Hara S, Yamada A, et al. Evidence of association of the ecNOS gene polymorphism with plasma NO metabolite levels in humans. Biochem Biophys Res Commun 1998;245(1):1903. [143] Yoon Y, Song J, Hong SH, Kim JQ. Plasma nitric oxide concentrations and nitric oxide synthase gene polymorphisms in coronary artery disease. Clin Chem 2000;46(10):162630. [144] Casas JP, Bautista LE, Humphries SE, Hingorani AD. Endothelial nitric oxide synthase genotype and ischemic heart disease: Meta-analysis of 26 studies involving 23028 subjects. Circulation 2004;109(11):135965. [145] Ichihara S, Yamada Y, Fujimura T, Nakashima N, Yokota M. Association of a polymorphism of the endothelial constitutive nitric oxide synthase gene with myocardial infarction in the Japanese population. Am J Cardiol 1998;81(1):836. [146] Sigusch HH, Surber R, Lehmann MH, Surber S, Weber J, Henke A, et al. Lack of association between 27-bp repeat

[147]

[148]

[149]

[150]

[151]

[152]

[153]

[154]

[155]

[156]

[157]

[158]

[159]

[160]

[161]

polymorphism in intron 4 of the endothelial nitric oxide synthase gene and the risk of coronary artery disease. Scand J Clin Lab Invest 2000;60(3):22935. Kunnas TA, Ilveskoski E, Niskakangas T, Laippala P, Kajander OA, Mikkelsson J, et al. Association of the endothelial nitric oxide synthase gene polymorphism with risk of coronary artery disease and myocardial infarction in middleaged men. J Mol Med 2002;80(9):6059. Kim IJ, Bae J, Lim SW, Cha DH, Cho HJ, Kim S, et al. Influence of endothelial nitric oxide synthase gene polymorphisms (-786T.C, 4a4b, 894G.T) in Korean patients with coronary artery disease. Thromb Res 2007;119(5):57985. Morray B, Goldenberg I, Moss AJ, Zareba W, Ryan D, McNitt S, et al. Polymorphisms in the paraoxonase and endothelial nitric oxide synthase genes and the risk of earlyonset myocardial infarction. Am J Cardiol 2007;99(8):11005. Colhoun HM, McKeigue PM, Davey Smith G. Problems of reporting genetic associations with complex outcomes. Lancet 2003;361(9360):86572. Tanus-Santos JE, Desai M, Flockhart DA. Effects of ethnicity on the distribution of clinically relevant endothelial nitric oxide variants. Pharmacogenetics 2001;11(8):71925. Song J, Kim OJ, Kim HS, Bae SJ, Hong SP, Oh D, et al. Endothelial nitric oxide synthase gene polymorphisms and the risk of silent brain infarction. Int J Mol Med 2010;25(5): 81923. Kim TG, Kim NK, Baek MJ, Huh R, Chung SS, Choi JU, et al. The relationships between endothelial nitric oxide synthase polymorphisms and the formation of intracranial aneurysms in the Korean population. Neurosurg Focus 2011;30(6):E23. Koshy L, Easwer HV, Neetha NV, Natarajan C, Bhattacharya RN, Banerjee M. Role of endothelial nitric oxide synthase gene polymorphisms in predicting aneurysmal subarachnoid hemorrhage in South Indian patients. Dis Markers 2008;24 (6):3339. Ko NU, Rajendran P, Kim H, Rutkowski M, Pawlikowska L, Kwok PY, et al. Endothelial nitric oxide synthase polymorphism (-786T-.C) and increased risk of angiographic vasospasm after aneurysmal subarachnoid hemorrhage. Stroke 2008;39(4):11038. Saidi S, Mallat SG, Almawi WY, Mahjoub T. Endothelial nitric oxide synthase Glu298Asp, 4b/a, and -786T.C gene polymorphisms and the risk of ischemic stroke. Acta Neurol Scand 2010;121(2):1149. Cheng J, Liu J, Li X, Yu L, Peng J, Zhang R, et al. Effect of polymorphisms of endothelial nitric oxide synthase on ischemic stroke: a case-control study in a Chinese population. Clin Chim Acta 2008;392(12):4651. Yemisci M, Sinici I, Ozkara HA, Hayran M, Ay H, Celtikci B, et al. Protective role of 27bp repeat polymorphism in intron 4 of eNOS gene in lacunar infarction. Free Radic Res 2009;43 (3):2729. Souza-Costa DC, Belo VA, Silva PS, Sertorio JT, Metzger IF, Lanna CM, et al. eNOS haplotype associated with hypertension in obese children and adolescents. Int J Obes (Lond) 2011;35(3):38792. Sandrim VC, Palei AC, Cavalli RC, Araujo FM, Ramos ES, Duarte G, et al. eNOS haplotypes associated with gestational hypertension or preeclampsia. Pharmacogenomics 2008;9 (10):146773. Colomba D, Duro G, Corrao S, Argano C, Di Chiara T, Nuzzo D, et al. Endothelial nitric oxide synthase gene polymorphisms and cardiovascular damage in hypertensive subjects: an Italian case-control study. Immun Ageing 2008;5:4.

I. PLATELET BIOLOGY

REFERENCES

[162] Dell’Omo G, Penno G, Pucci L, Fotino C, Lucchesi D, Del Prato S, et al. Lack of association between endothelial nitric oxide synthase gene polymorphisms, microalbuminuria and endothelial dysfunction in hypertensive men. J Hypertens 2007;25(7):138995. [163] Kim OJ, Kim UK, Oh SH, Cho YW, Oh KI, Oh D, et al. Association of endothelial nitric oxide synthase polymorphisms and haplotypes with ischemic stroke in Korean individuals with or without diabetes mellitus. Mol Med Report 2010;3(3):50913. [164] Santos KG, Crispim D, Canani LH, Ferrugem PT, Gross JL, Roisenberg I. Association of eNOS gene polymorphisms with renal disease in caucasians with type 2 diabetes. Diabetes Res Clin Pract 2011;91(3):35362. [165] Ahluwalia TS, Ahuja M, Rai TS, Kohli HS, Sud K, Bhansali A, et al. Endothelial nitric oxide synthase gene haplotypes and diabetic nephropathy among Asian Indians. Mol Cell Biochem 2008;314(12):917. [166] Sandrim VC, de Syllos RW, Lisboa HR, Tres GS, Tanus-Santos JE. Influence of eNOS haplotypes on the plasma nitric oxide products concentrations in hypertensive and type 2 diabetes mellitus patients. Nitric Oxide 2007;16(3):34855. [167] Rittig K, Holder K, Stock J, Tschritter O, Peter A, Stefan N, et al. Endothelial NO-synthase intron 4 polymorphism is associated with disturbed in vivo nitric oxide production in individuals prone to type 2 diabetes. Horm Metab Res 2008;40(1):137. [168] Nagassaki S, Sertorio JT, Metzger IF, Bem AF, Rocha JB, Tanus-Santos JE. eNOS gene T-786C polymorphism modulates atorvastatin-induced increase in blood nitrite. Free Radic Biol Med 2006;41(7):10449. [169] Souza-Costa DC, Sandrim VC, Lopes LF, Gerlach RF, Rego EM, Tanus-Santos JE. Anti-inflammatory effects of atorvastatin: modulation by the T-786C polymorphism in the endothelial nitric oxide synthase gene. Atherosclerosis 2007;193(2):43844. [170] Sponton CH, Rezende TM, Mallagrino PA, Franco-Penteado CF, Bezerra MA, Zanesco A. Women with TT genotype for eNOS gene are more responsive in lowering blood pressure in response to exercise. Eur J Cardiovasc Prev Rehabil 2010;17 (6):67681. [171] Sandrim VC, Palei AC, Luizon MR, Izidoro-Toledo TC, Cavalli RC, Tanus-Santos JE. eNOS haplotypes affect the responsiveness to antihypertensive therapy in preeclampsia but not in gestational hypertension. Pharmacogenomics J 2010;10(1):405. [172] Alfadhli S, AlTamimy B, AlSaeid K, Haider M. Endothelial nitric oxide synthase gene haplotype association with systemic lupus erythematosus. Lupus 2011;20(7):7008. [173] Tang FY, Xie XW, Ling GH, Liu FY. Endothelial nitric oxide synthase and nicotinamide adenosine dinucleotide phosphate oxidase p22phox gene (C242T) polymorphisms and systemic lupus erythematosus in a Chinese population. Lupus 2010;19 (2):1926. [174] Yanamandra K, Napper D, Pramanik A, Bocchini Jr JA, Dhanireddy R. Endothelial nitric oxide synthase genotypes in the etiology of retinopathy of prematurity in premature infants. Ophthalmic Genet 2010;31(4):1737. [175] Wang JL, Wang HG, Gao HQ, Zhai GX, Chang P, Chen YG. Endothelial nitric oxide synthase polymorphisms and erectile dysfunction: a meta-analysis. J Sex Med 2010;7(12):388998. [176] Erol B, Bozdogan G, Akduman B, Dursun A, Bozdogan S, Onem K, et al. eNOS gene intron 4 VNTR and exon 7-G894T polymorphisms in Turkish men with erectile dysfunction: a case control study. J Sex Med 2009;6(5):14239.

333

[177] Kuptsova N, Chang-Claude J, Kropp S, Helmbold I, Schmezer P, von Fournier D, et al. Genetic predictors of long-term toxicities after radiation therapy for breast cancer. Int J Cancer 2008;122(6):13339. [178] Lu J, Wei Q, Bondy ML, Yu TK, Li D, Brewster A, et al. Promoter polymorphism (-786t.C) in the endothelial nitric oxide synthase gene is associated with risk of sporadic breast cancer in non-Hispanic white women age younger than 55 years. Cancer 2006;107(9):224553. [179] Rodriguez-Perez JC, Macias-Reyes A, Jimenez-Sosa A, Companioni O, Rodriguez-Esparragon FJ, Cobo MA, et al. A synergistic association of ACE I/D and eNOS G894T gene variants with the progression of immunoglobulin a nephropathy—a pilot study. Am J Nephrol 2009;30(3):3039. [180] Spoto B, Benedetto FA, Testa A, Tripepi G, Mallamaci F, Maas R, et al. An additive effect of endothelial nitric oxide synthase gene polymorphisms contributes to the severity of atherosclerosis in patients on dialysis. Am J Hypertens 2007;20(7):75863. [181] Kobayashi T, Ushikubi F, Narumiya S. Amino acid residues conferring ligand binding properties of prostaglandin I and prostaglandin D receptors. Identification by site-directed mutagenesis. J Biol Chem 2000;275(32):24294303. [182] Moncada S, Gryglewski R, Bunting S, Vane JR. An enzyme isolated from arteries transforms prostaglandin endoperoxides to an unstable substance that inhibits platelet aggregation. Nature 1976;263(5579):6635. [183] Gryglewski RJ, Bunting S, Moncada S, Flower RJ, Vane JR. Arterial walls are protected against deposition of platelet thrombi by a substance (prostaglandin X) which they make from prostaglandin endoperoxides. Prostaglandins 1976;12 (5):685713. [184] Cho MJ, Allen MA. Chemical stability of prostacyclin (PGI2) in aqueous solutions. Prostaglandins 1978;15(6):94354. [185] Dusting GJ, Moncada S, Vane JR. Recirculation of prostacyclin (PGI2) in the dog. Br J Pharmacol 1978;64(2):31520. [186] Tsai AL, Hsu MJ, Patsch W, Wu KK. Regulation of PGI2 activity by serum proteins: serum albumin but not high density lipoprotein is the PGI2 binding and stabilizing protein in human blood. Biochim Biophys Acta 1991;1115 (2):13140. [187] Parente L, Perretti M. Advances in the pathophysiology of constitutive and inducible cyclooxygenases: two enzymes in the spotlight. Biochem Pharmacol 2003;65(2):1539. [188] Marnett LJ, Rowlinson SW, Goodwin DC, Kalgutkar AS, Lanzo CA. Arachidonic acid oxygenation by COX-1 and COX-2. Mechanisms of catalysis and inhibition. J Biol Chem 1999;274(33):229036. [189] Smith WL, DeWitt DL, Garavito RM. Cyclooxygenases: structural, cellular, and molecular biology. Annu Rev Biochem 2000;69:14582. [190] Narumiya S, Sugimoto Y, Ushikubi F. Prostanoid receptors: structures, properties, and functions. Physiol Rev 1999;79 (4):1193226. [191] Weksler BB, Marcus AJ, Jaffe EA. Synthesis of prostaglandin I2 (prostacyclin) by cultured human and bovine endothelial cells. Proc Natl Acad Sci USA 1977;74(9):39226. [192] Ingerman-Wojenski C, Silver MJ, Smith JB, Macarak E. Bovine endothelial cells in culture produce thromboxane as well as prostacyclin. J Clin Invest 1981;67(5):12926. [193] Moncada S. Biological importance of prostacyclin. Br J Pharmacol 1982;76(1):331 Eighth Gaddum Memorial Lecture. University of London Institute of Education, December 1980.

I. PLATELET BIOLOGY

334

17. INHIBITION OF PLATELET FUNCTION BY THE ENDOTHELIUM

[194] Bunting S, Moncada S, Vane JR. The prostacyclinthromboxane A2 balance: pathophysiological and therapeutic implications. Br Med Bull 1983;39(3):2716. [195] Smith WL, DeWitt DL, Allen ML. Bimodal distribution of the prostaglandin I2 synthase antigen in smooth muscle cells. J Biol Chem 1983;258(9):59226. [196] Oates JA, FitzGerald GA, Branch RA, Jackson EK, Knapp HR, Roberts II LJ. Clinical implications of prostaglandin and thromboxane A2 formation (1). N Engl J Med 1988;319(11): 68998. [197] Lin Y, Wu KK, Ruan KH. Characterization of the secondary structure and membrane interaction of the putative membrane anchor domains of prostaglandin I2 synthase and cytochrome P450 2C1. Arch Biochem Biophys 1998;352(1):7884. [198] Ruan KH, So SP, Zheng W, Wu J, Li D, Kung J. Solution structure and topology of the N-terminal membrane anchor domain of a microsomal cytochrome P450: prostaglandin I2 synthase. Biochem J 2002;368(Pt. 3):7218. [199] Massimino ML, Griffoni C, Spisni E, Toni M, Tomasi V. Involvement of caveolae and caveolae-like domains in signalling, cell survival and angiogenesis. Cell Signal 2002;14(2): 938. [200] Smith WL. Prostaglandin biosynthesis and its compartmentation in vascular smooth muscle and endothelial cells. Annu Rev Physiol 1986;48:25162. [201] Deng H, Huang A, So SP, Lin YZ, Ruan KH. Substrate access channel topology in membrane-bound prostacyclin synthase. Biochem J 2002;362(Pt. 3):54551. [202] Ruan KH, Deng H, Wu J, So SP. The N-terminal membrane anchor domain of the membrane-bound prostacyclin synthase involved in the substrate presentation of the coupling reaction with cyclooxygenase. Arch Biochem Biophys 2005;435(2): 37281. [203] Spisni E, Griffoni C, Santi S, Riccio M, Marulli R, Bartolini G, et al. Colocalization prostacyclin (PGI2) synthasecaveolin-1 in endothelial cells and new roles for PGI2 in angiogenesis. Exp Cell Res 2001;266(1):3143. [204] Wade ML, Voelkel NF, Fitzpatrick FA. “Suicide” inactivation of prostaglandin I2 synthase: Characterization of mechanismbased inactivation with isolated enzyme and endothelial cells. Arch Biochem Biophys 1995;321(2):4538. [205] Zou M, Martin C, Ullrich V. Tyrosine nitration as a mechanism of selective inactivation of prostacyclin synthase by peroxynitrite. Biol Chem 1997;378(7):70713. [206] Schmidt P, Youhnovski N, Daiber A, Balan A, Arsic M, Bachschmid M, et al. Specific nitration at tyrosine 430 revealed by high resolution mass spectrometry as basis for redox regulation of bovine prostacyclin synthase. J Biol Chem 2003;278(15):128139. [207] Dutta-Roy AK, Sinha AK. Purification and properties of prostaglandin E1/prostacyclin receptor of human blood platelets. J Biol Chem 1987;262(26):1268591. [208] Tsai AL, Hsu MJ, Vijjeswarapu H, Wu KK. Solubilization of prostacyclin membrane receptors from human platelets. J Biol Chem 1989;264(1):617. [209] Jones RL, Qian Y, Wong HN, Chan H, Yim AP. Prostanoid action on the human pulmonary vascular system. Clin Exp Pharmacol Physiol 1997;24(12):96972. [210] Nakagawa O, Tanaka I, Usui T, Harada M, Sasaki Y, Itoh H, et al. Molecular cloning of human prostacyclin receptor cDNA and its gene expression in the cardiovascular system. Circulation 1994;90(4):16437. [211] Smyth EM, FitzGerald GA. Human prostacyclin receptor. Vitam Horm 2002;65:14965.

[212] Ogawa Y, Tanaka I, Inoue M, Yoshitake Y, Isse N, Nakagawa O, et al. Structural organization and chromosomal assignment of the human prostacyclin receptor gene. Genomics 1995;27(1):1428. [213] Leigh PJ, Cramp WA, MacDermot J. Identification of the prostacyclin receptor by radiation inactivation. J Biol Chem 1984;259(20):124316. [214] Kobayashi T, Kiriyama M, Hirata T, Hirata M, Ushikubi F, Narumiya S. Identification of domains conferring ligand binding specificity to the prostanoid receptor. Studies on chimeric prostacyclin/prostaglandin D receptors. J Biol Chem 1997;272 (24):1515460. [215] Armstrong RA. Platelet prostanoid receptors. Pharmacol Ther 1996;72(3):17191. [216] Boie Y, Rushmore TH, Darmon-Goodwin A, Grygorczyk R, Slipetz DM, Metters KM, et al. Cloning and expression of a cDNA for the human prostanoid IP receptor. J Biol Chem 1994;269(16):121738. [217] Xiao CY, Hara A, Yuhki K, Fujino T, Ma H, Okada Y, et al. Roles of prostaglandin I(2) and thromboxane A(2) in cardiac ischemia-reperfusion injury: a study using mice lacking their respective receptors. Circulation 2001;104 (18):22105. [218] Wise H, Wong YH, Jones RL. Prostanoid signal integration and cross talk. Neurosignals 2002;11(1):208. [219] Zhang Z, Austin SC, Smyth EM. Glycosylation of the human prostacyclin receptor: role in ligand binding and signal transduction. Mol Pharmacol 2001;60(3):4807. [220] Hayes JS, Lawler OA, Walsh MT, Kinsella BT. The prostacyclin receptor is isoprenylated. Isoprenylation is required for efficient receptor-effector coupling. J Biol Chem 1999;274 (34):2370718. [221] Miggin SM, Lawler OA, Kinsella BT. Investigation of a functional requirement for isoprenylation by the human prostacyclin receptor. Eur J Biochem 2002;269(6):171425. [222] Giovanazzi S, Accomazzo MR, Letari O, Oliva D, Nicosia S. Internalization and down-regulation of the prostacyclin receptor in human platelets. Biochem J 1997;325(Pt. 1): 717. [223] Smyth EM, Li WH, FitzGerald GA. Phosphorylation of the prostacyclin receptor during homologous desensitization. A critical role for protein kinase C. J Biol Chem 1998;273 (36):2325866. [224] Miggin SM, Lawler OA, Kinsella BT. Palmitoylation of the human prostacyclin receptor. Functional implications of palmitoylation and isoprenylation. J Biol Chem 2003;278 (9):694758. [225] Modesti PA, Abbate R, Prisco D, Gensini GF. Human prostacyclin platelet receptors and platelet lipid composition. J Lipid Mediat 1990;2(6):30915. [226] Sasaki Y, Takahashi T, Tanaka I, Nakamura K, Okuno Y, Nakagawa O, et al. Expression of prostacyclin receptor in human megakaryocytes. Blood 1997;90(3):103946. [227] Svensson J, Hamberg M, Samuelsson B. On the formation and effects of thromboxane A2 in human platelets. Acta Physiol Scand 1976;98(3):28594. [228] Moncada S, Vane JR. Pharmacology and endogenous roles of prostaglandin endoperoxides, thromboxane A2, and prostacyclin. Pharmacol Rev 1978;30(3):293331. [229] Moncada S, Vane JR. Prostacyclin in the cardiovascular system. Adv Prostaglandin Thromboxane Res 1980;6:4360. [230] Needleman P, Turk J, Jakschik BA, Morrison AR, Lefkowith JB. Arachidonic acid metabolism. Annu Rev Biochem 1986;55: 69102.

I. PLATELET BIOLOGY

REFERENCES

[231] Pober JS, Cotran RS. Cytokines and endothelial cell biology. Physiol Rev 1990;70(2):42751. [232] Manganello JM, Djellas Y, Borg C, Antonakis K, Le Breton GC. Cyclic AMP-dependent phosphorylation of thromboxane A(2) receptor-associated Galpha(13). J Biol Chem 1999;274 (39):2800310. [233] Panes J, Perry M, Granger DN. Leukocyte-endothelial cell adhesion: avenues for therapeutic intervention. Br J Pharmacol 1999;126(3):53750. [234] Gorman RR, Bunting S, Miller OV. Modulation of human platelet adenylate cyclase by prostacyclin (PGX). Prostaglandins 1977;13(3):37788. [235] Tateson JE, Moncada S, Vane JR. Effects of prostacyclin (PGX) on cyclic AMP concentrations in human platelets. Prostaglandins 1977;13(3):38997. [236] Wise H, Jones RL. Prostacyclin and its receptors. New York: Kluwer Academic/Plenum Publishers; 2000. [237] Gonzalez-Utor AL, Sanchez-Aguayo I, Hidalgo J. Cytochemical localization of K(1)-dependent p-nitrophenyl phosphatase and adenylate cyclase by using one-step method in human washed platelets. Histochemistry 1992;97(6):5037. [238] Seiss W. Molecular mechanisms of platelet activation. Physiol Rev 1989;69:58178. [239] Fox JE, Say AK, Haslam RJ. Subcellular distribution of the different platelet proteins phosphorylated on exposure of intact platelets to ionophore A23187 or to prostaglandin E1. Possible role of a membrane phosphopolypeptide in the regulation of calcium-ion transport. Biochem J 1979;184(3):65161. [240] Kaser-Glanzmann R, Gerber E, Luscher EF. Regulation of the intracellular calcium level in human blood platelets: cyclic adenosine 3’,5’-monophosphate dependent phosphorylation of a 22,000 dalton component in isolated Ca21 -accumulating vesicles. Biochim Biophys Acta 1979;558(3):3447. [241] Hathaway DR, Eaton CR, Adelstein RS. Regulation of human platelet myosin light chain kinase by the catalytic subunit of cyclic AMP-dependent protein kinase. Nature 1981;291(5812):2526. [242] Cavallini L, Coassin M, Borean A, Alexandre A. Prostacyclin and sodium nitroprusside inhibit the activity of the platelet inositol 1,4,5-trisphosphate receptor and promote its phosphorylation. J Biol Chem 1996;271(10):554551. [243] Aszodi A, Pfeifer A, Ahmad M, Glauner M, Zhou XH, Ny L, et al. The vasodilator-stimulated phosphoprotein (VASP) is involved in cGMP- and cAMP-mediated inhibition of agonistinduced platelet aggregation, but is dispensable for smooth muscle function. Embo J 1999;18(1):3748. [244] Siess W, Lapetina EG. Prostacyclin inhibits platelet aggregation induced by phorbol ester or Ca21 ionophore at steps distal to activation of protein kinase C and Ca21 -dependent protein kinases. Biochem J 1989;258(1):5765. [245] Conti MA, Adelstein RS. The relationship between calmodulin binding and phosphorylation of smooth muscle myosin kinase by the catalytic subunit of 3’:5’ cAMP-dependent protein kinase. J Biol Chem 1981;256(7):317881. [246] Fox JE, Phillips DR. Role of phosphorylation in mediating the association of myosin with the cytoskeletal structures of human platelets. J Biol Chem 1982;257(8):41206. [247] Body SC. Platelet activation and interactions with the microvasculature. J Cardiovasc Pharmacol 1996;27(Suppl. 1):S1325. [248] Narumiya S, FitzGerald GA. Genetic and pharmacological analysis of prostanoid receptor function. J Clin Invest 2001;108(1):2530. [249] Smyth EM, Austin SC, Reilly MP, FitzGerald GA. Internalization and sequestration of the human prostacyclin receptor. J Biol Chem 2000;275(41):3203745.

335

[250] Ashby B. Comparison of Iloprost, cicaprost and prostacyclin effects on cyclic AMP metabolism in intact platelets. Prostaglandins 1992;43(3):25561. [251] Darius H, Veit K, Binz C, Fisch A, Meyer J. Diminished inhibition of adhesion molecule expression in prostacyclin receptor desensitized human platelets. Agents Actions Suppl 1995;45:7783. [252] Jaschonek K, Karsch KR, Weisenberger H, Tidow S, Faul C, Renn W. Platelet prostacyclin binding in coronary artery disease. J Am Coll Cardiol 1986;8(2):25966. [253] Klockenbusch W, Hohlfeld T, Wilhelm M, Somville T, Schror K. Platelet PGI2 receptor affinity is reduced in pre-eclampsia. Br J Clin Pharmacol 1996;41(6):6168. [254] Modesti PA, Fortini A, Poggesi L, Boddi M, Abbate R, Gensini GF. Acute reversible reduction of PGI2 platelet receptors after iloprost infusion in man. Thromb Res 1987;48 (6):6639. [255] Alt U, Leigh PJ, Wilkins AJ, Morris PK, MacDermot J. Desensitization of iloprost responsiveness in human platelets follows prolonged exposure to iloprost in vitro. Br J Clin Pharmacol 1986;22(1):1189. [256] Darius H, Binz C, Veit K, Fisch A, Meyer J. Platelet receptor desensitization induced by elevated prostacyclin levels causes plateletendothelial cell adhesion. J Am Coll Cardiol 1995;26 (3):8006. [257] Krane A, MacDermot J, Keen M. Desensitization of adenylate cyclase responses following exposure to IP prostanoid receptor agonists. Homologous and heterologous desensitization exhibit the same time course. Biochem Pharmacol 1994;47 (6):9539. [258] Sinzinger H, Silberbauer K, Horsch AK, Gall A. Decreased sensitivity of human platelets to PGI2 during long-term intraarterial prostacyclin infusion in patients with peripheral vascular disease—a rebound phenomenon? Prostaglandins 1981;21(1):4951. [259] MacDermot J. Desensitization of prostacyclin responsiveness in platelets. Apparent differences in the mechanism in vitro or in vivo. Biochem Pharmacol 1986;35(16):26459. [260] Fisch A, Tobusch K, Veit K, Meyer J, Darius H. Prostacyclin receptor desensitization is a reversible phenomenon in human platelets. Circulation 1997;96(3):75660. [261] Ashby B. Interactions among prostaglandin receptors. Receptor 1994;4(1):3142. [262] Wilson SJ, Roche AM, Kostetskaia E, Smyth EM. Dimerization of the human receptors for prostacyclin and thromboxane facilitates thromboxane receptor-mediated cAMP generation. J Biol Chem 2004;279(51):5303647. [263] Ortiz-Vega S, Ashby B. Human prostacyclin receptor: cloning and co-expression with EP3 prostaglandin receptor. Adv Exp Med Biol 1997;433:2358. [264] Paul BZ, Ashby B, Sheth SB. Distribution of prostaglandin IP and EP receptor subtypes and isoforms in platelets and human umbilical artery smooth muscle cells. Br J Haematol 1998;102(5):120411. [265] Moncada S, Vane JR. Arachidonic acid metabolites and the interactions between platelets and blood-vessel walls. N Engl J Med 1979;300(20):11427. [266] Jorgensen KA. Studies on the biological balance between thromboxanes and prostacyclins in relation to the platelet-vessel wall interaction. Dan Med Bull 1982;29(4): 16997. [267] Marcus AJ, Broekman MJ, Pinsky DJ. COX inhibitors and thromboregulation. N Engl J Med 2002;347(13):10256. [268] Coleman R, Kennedy I, Humphrey P, et al. Prostanoids and their receptors. In: Emmett J, editor. Comprehensive medicinal

I. PLATELET BIOLOGY

336

[269]

[270] [271]

[272]

[273]

[274]

[275]

[276]

[277]

[278]

[279]

[280]

[281] [282]

[283]

[284]

[285]

[286]

17. INHIBITION OF PLATELET FUNCTION BY THE ENDOTHELIUM

chemistry: membranes and receptors. Oxford: Pergamon Press; 1990. p. 643714. Smith WL, DeWitt DL, Shimokawa T, Kraemer SA, Meade EA. Molecular basis for the inhibition of prostanoid biosynthesis by nonsteroidal anti-inflammatory agents. Stroke 1990;21(12 Suppl.):IV248. Smith WL. Prostanoid biosynthesis and mechanisms of action. Am J Physiol 1992;263(2, Pt. 2):F18191. Minkes M, Stanford N, Chi MM, Roth GJ, Raz A, Needleman P, et al. Cyclic adenosine 3’,5’-monophosphate inhibits the availability of arachidonate to prostaglandin synthetase in human platelet suspensions. J Clin Invest 1977;59(3):44954. Malmsten C, Granstrom E, Samuelsson B. Cyclic AMP inhibits synthesis of prostaglandin endoperoxide (PGG2) in human platelets. Biochem Biophys Res Commun 1976;68 (2):56976. Gerrard JM, Peller JD, Krick TP, White JG. Cyclic AMP and platelet prostaglandin synthesis. Prostaglandins 1977;14 (1):3950. Nony P, Ffrench P, Girard P, Delair S, Azoulay S, Girre JP, et al. Platelet-aggregation inhibition and hemodynamic effects of beraprost sodium, a new oral prostacyclin derivative: a study in healthy male subjects. Can J Physiol Pharmacol 1996;74(8):88793. Marcus AJ, Weksler BB, Jaffe EA, Broekman MJ. Synthesis of prostacyclin from platelet-derived endoperoxides by cultured human endothelial cells. J Clin Invest 1980;66(5):97986. Caughey GE, Cleland LG, Gamble JR, James MJ. Up-regulation of endothelial cyclooxygenase-2 and prostanoid synthesis by platelets. Role of thromboxane A2. J Biol Chem 2001;276 (41):3783945. Radomski MW, Palmer RM, Moncada S. The anti-aggregating properties of vascular endothelium: interactions between prostacyclin and nitric oxide. Br J Pharmacol 1987;92 (3):63946. Lidbury PS, Antunes E, de Nucci G, Vane JR. Interactions of iloprost and sodium nitroprusside on vascular smooth muscle and platelet aggregation. Br J Pharmacol 1989;98(4):127580. Radomski MW, Palmer MJ, Moncada S. The role of nitric oxide and cGMP in platelet adhesion to vascular endothelium. Biochem Biophys Res Commun 1987;148:14829. Panicot-Dubois L, Thomas GM, Furie BC, Furie B, Lombardo D, Dubois C. Bile salt-dependent lipase interacts with platelet CXCR4 and modulates thrombus formation in mice and humans. J Clin Invest 2007;117(12):370819. Vane JR, Botting RM. Pharmacodynamic profile of prostacyclin. Am J Cardiol 1995;75(3):3A10A. FitzGerald GA, Smith B, Pedersen AK, Brash AR. Increased prostacyclin biosynthesis in patients with severe atherosclerosis and platelet activation. N Engl J Med 1984;310 (17):10658. Yokoyama C, Yabuki T, Shimonishi M, Wada M, Hatae T, Ohkawara S, et al. Prostacyclin-deficient mice develop ischemic renal disorders, including nephrosclerosis and renal infarction. Circulation 2002;106(18):2397403. Bourgain RH. Inhibition of PGI2 (prostacyclin) synthesis in the arterial wall enhances the formation of white platelet thrombi in vivo. Haemostasis 1978;7(4):2525. Pettigrew LC, Wu KK. Platelet function and antiaggregant therapy in ischemic stroke. Semin Neurol 1986;6 (3):2938. Vane J. Prostacyclin in the cardiovascular system in health and disease. In: Schror K, editor. Prostaglandins and other eicosanoids in the cardiovascular system. Basel: Karger; 1985. p. 728.

[287] Stein RW, Papp AC, Weiner WJ, Wu KK. Reduction of serum prostacyclin stability in ischemic stroke. Stroke 1985;16(1):168. [288] Akopov S, Darbinian V, Grigorian G, Kosyan A, Gabrielian E. Elevated velocity of prostacyclin degradation in blood as a possible risk factor in patients with cerebrovascular disorders. Eur Neurol 1993;33(3):2525. [289] Numaguchi Y, Naruse K, Harada M, Osanai H, Mokuno S, Murase K, et al. Prostacyclin synthase gene transfer accelerates reendothelialization and inhibits neointimal formation in rat carotid arteries after balloon injury. Arterioscler Thromb Vasc Biol 1999;19(3):72733. [290] Harada M, Toki Y, Numaguchi Y, Osanai H, Ito T, Okumura K, et al. Prostacyclin synthase gene transfer inhibits neointimal formation in rat balloon-injured arteries without bleeding complications. Cardiovasc Res 1999;43 (2):48191. [291] Uchiyama-Tsuyuki Y, Kawashima K, Araki H, Otomo S. Prostacyclin analogue TTC-909 reduces memory impairment in rats with cerebral embolism. Pharmacol Biochem Behav 1995;52(3):5559. [292] Stier Jr CT, Chander PN, Belmonte A, Inamdar RS, Mistry M. Beneficial action of beraprost sodium, a prostacyclin analog, in stroke-prone rats. J Cardiovasc Pharmacol 1997;30 (3):28593. [293] Huczynski J, Kostka-Trabka E, Sotowska W, Bieron K, Grodzinska L, Dembinska-Kiec A, et al. Double-blind controlled trial of the therapeutic effects of prostacyclin in patients with completed ischaemic stroke. Stroke 1985;16 (5):8104. [294] Martin JF, Hamdy N, Nicholl J, Lewtas N, Bergvall U, Owen P, et al. Prostacyclin in cerebral infarction. N Engl J Med 1985;312(25):1642. [295] Hsu CY, Faught Jr RE, Furlan AJ, Coull BM, Huang DC, Hogan EL, et al. Intravenous prostacyclin in acute nonhemorrhagic stroke: a placebo-controlled double-blind trial. Stroke 1987;18(2):3528. [296] Rubin LJ. Pathology and pathophysiology of primary pulmonary hypertension. Am J Cardiol 1995;75(3):51A4A. [297] Fitzgerald DJ, Entman SS, Mulloy K, FitzGerald GA. Decreased prostacyclin biosynthesis preceding the clinical manifestation of pregnancy-induced hypertension. Circulation 1987;75(5):95663. [298] Murata T, Ushikubi F, Matsuoka T, Hirata M, Yamasaki A, Sugimoto Y, et al. Altered pain perception and inflammatory response in mice lacking prostacyclin receptor. Nature 1997;388(6643):67882. [299] Ueno A, Naraba H, Ikeda Y, Ushikubi F, Murata T, Narumiya S, et al. Intrinsic prostacyclin contributes to exudation induced by bradykinin or carrageenin: a study on the paw edema induced in IP-receptor-deficient mice. Life Sci 2000;66(12):PL15560. [300] Cheng Y, Austin SC, Rocca B, Koller BH, Coffman TM, Grosser T, et al. Role of prostacyclin in the cardiovascular response to thromboxane A2. Science 2002;296(5567): 53941. [301] Yang J, Wu J, Jiang H, Mortensen R, Austin S, Manning DR, et al. Signaling through Gi family members in platelets. Redundancy and specificity in the regulation of adenylyl cyclase and other effectors. J Biol Chem 2002;277(48): 4603542. [302] Egan KM, Lawson JA, Fries S, Koller B, Rader DJ, Smyth EM, et al. COX-2-derived prostacyclin confers atheroprotection on female mice. Science 2004;306(5703):19547. [303] Majerus PW. Arachidonate metabolism in vascular disorders. J Clin Invest 1983;72(5):15215.

I. PLATELET BIOLOGY

REFERENCES

[304] de Deckere EA, Nugteren DH, Ten Hoor F. Prostacyclin is the major prostaglandin released from the isolated perfused rabbit and rat heart. Nature 1977;268(5616):1603. [305] Coker SJ, Parratt JR, Ledingham IM, Zeitlin IJ. Thromboxane and prostacyclin release from ischaemic myocardium in relation to arrhythmias. Nature 1981;291(5813):3234. [306] Johnson III G, Furlan LE, Aoki N, Lefer AM. Endothelium and myocardial protecting actions of taprostene, a stable prostacyclin analogue, after acute myocardial ischemia and reperfusion in cats. Circ Res 1990;66(5):136270. [307] Simpson PJ, Mickelson J, Fantone JC, Gallagher KP, Lucchesi BR. Iloprost inhibits neutrophil function in vitro and in vivo and limits experimental infarct size in canine heart. Circ Res 1987;60(5):66673. [308] Simpson PJ, Mitsos SE, Ventura A, Gallagher KP, Fantone JC, Abrams GD, et al. Prostacyclin protects ischemic reperfused myocardium in the dog by inhibition of neutrophil activation. Am Heart J 1987;113(1):12937. [309] Aherne T, Price DC, Yee ES, Hsieh WR, Ebert PA. Prevention of ischemia-induced myocardial platelet deposition by exogenous prostacyclin. J Thorac Cardiovasc Surg 1986;92 (1):99104. [310] Kahn NN, Bauman WA, Sinha AK. Loss of high-affinity prostacyclin receptors in platelets and the lack of prostaglandininduced inhibition of platelet-stimulated thrombin generation in subjects with spinal cord injury. Proc Natl Acad Sci USA 1996;93(1):2459. [311] Neri Serneri GG, Modesti PA, Fortini A, Abbate R, Lombardi A, Gensini GF. Reduction in prostacyclin platelet receptors in active spontaneous angina. Lancet 1984;2 (8407):83841. [312] Betteridge DJ, El Tahir KE, Reckless JP, Williams KI. Platelets from diabetic subjects show diminished sensitivity to prostacyclin. Eur J Clin Invest 1982;12(5):3958. [313] Jaschonek K, Weisenberger H, Karsch KR, Renn W, Daiss W, Schenzle D, et al. Impaired platelet prostacyclin binding in acute myocardial infarction. Lancet 1984;2(8415):13412. [314] Jaschonek K, Muller CP. Platelet and vessel associated prostacyclin and thromboxane A2/prostaglandin endoperoxide receptors. Eur J Clin Invest 1988;18(1):18. [315] Sturzebecher Sr C, Haberey M, Muller B, Schillinger E, Schroder G, Skuballa W, et al. Pharmacological profile of ZK 96480, a new chemically and metabolically stable prostacyclin analogue with oral availability and high PGI2 intrinsic activity. In: Schror K, editor. Prostaglandins and other eicosanoids in the cardiovascular system. Basel: Karger; 1985. p. 48591. [316] Schror K, Darius H, Matzky R, Ohlendorf R. The antiplatelet and cardiovascular actions of a new carbacyclin derivative (ZK 36 374)—equipotent to PGI2 in vitro. Naunyn Schmiedebergs Arch Pharmacol 1981;316(3):2525. [317] Whittle BJ, Moncada S, Whiting F, Vane JR. Carbacyclin—a potent stable prostacyclin analogue for the inhibition of platelet aggregation. Prostaglandins 1980;19(4):60527. [318] Whittle BJ, Moncada S. Platelet actions of stable carbocyclic analogues of prostacyclin. Circulation 1985;72(6):121925. [319] Nickolson RC, Town MH, Vorbruggen H. Prostacyclinanalogs. Med Res Rev 1985;5(1):153. [320] Badesch DB, McLaughlin VV, Delcroix M, Vizza CD, Olschewski H, Sitbon O, et al. Prostanoid therapy for pulmonary arterial hypertension. J Am Coll Cardiol 2004;43(12, Suppl. S):56S61S. [321] Kooter AJ, Ijzerman RG, Kamp O, Boonstra AB, Smulders YM. No effect of epoprostenol on right ventricular diameter in patients with acute pulmonary embolism: a randomized controlled trial. BMC Pulm Med 2010;10:18.

337

[322] Wienecke T, Olesen J, Oturai PS, Ashina M. Prostacyclin (epoprostenol) induces headache in healthy subjects. Pain 2008;139 (1):10616. [323] Wienecke T, Olesen J, Ashina M. Prostaglandin I2 (epoprostenol) triggers migraine-like attacks in migraineurs. Cephalalgia 2010;30(2):17990. [324] Badesch DB, McGoon MD, Barst RJ, Tapson VF, Rubin LJ, Wigley FM, et al. Longterm survival among patients with scleroderma-associated pulmonary arterial hypertension treated with intravenous epoprostenol. J Rheumatol 2009;36 (10):22449. [325] Akagi S, Nakamura K, Miyaji K, Ogawa A, Kusano KF, Ito H, et al. Marked hemodynamic improvements by high-dose epoprostenol therapy in patients with idiopathic pulmonary arterial hypertension. Circ J 2010;74(10):22005. [326] Khan TA, Schnickel G, Ross D, Bastani S, Laks H, Esmailian F, et al. A prospective, randomized, crossover pilot study of inhaled nitric oxide versus inhaled prostacyclin in heart transplant and lung transplant recipients. J Thorac Cardiovasc Surg 2009;138(6):141724. [327] Nakayama T, Masubuchi Y, Kawauchi K, Masaki R, Hironaga T, Ishima H, et al. Beneficial effect of beraprost sodium plus telmisartan in the prevention of arterial stiffness development in elderly patients with hypertension and cerebral infarction. Prostaglandins Leukot Essent Fatty Acids 2007;76(6):30914. [328] Kunieda T, Nakanishi N, Matsubara H, Ohe T, Okano Y, Kondo H, et al. Effects of long-acting beraprost sodium (TRK100STP) in Japanese patients with pulmonary arterial hypertension. Int Heart J 2009;50(4):51329. [329] Theodoraki K, Tsiapras D, Tsourelis L, Zarkalis D, Sfirakis P, Kapetanakis E, et al. Inhaled iloprost in eight heart transplant recipients presenting with post-bypass acute right ventricular dysfunction. Acta Anaesthesiol Scand 2006;50 (10):12137. [330] McLaughlin VV, Oudiz RJ, Frost A, Tapson VF, Murali S, Channick RN, et al. Randomized study of adding inhaled iloprost to existing bosentan in pulmonary arterial hypertension. Am J Respir Crit Care Med 2006;174 (11):125763. [331] Baughman RP, Judson MA, Lower EE, Highland K, Kwon S, Craft N, et al. Inhaled iloprost for sarcoidosis associated pulmonary hypertension. Sarcoidosis Vasc Diffuse Lung Dis 2009;26(2):11020. [332] Sitbon O, Manes A, Jais X, Pallazini M, Humbert M, Presotto L, et al. Rapid switch from intravenous epoprostenol to intravenous treprostinil in patients with pulmonary arterial hypertension. J Cardiovasc Pharmacol 2007;49(1):15. [333] Rubenfire M, McLaughlin VV, Allen RP, Elliott G, Park MH, Wade M, et al. Transition from IV epoprostenol to subcutaneous treprostinil in pulmonary arterial hypertension: a controlled trial. Chest 2007;132(3):75763. [334] Voswinckel R, Enke B, Reichenberger F, Kohstall M, Kreckel A, Krick S, et al. Favorable effects of inhaled treprostinil in severe pulmonary hypertension: results from randomized controlled pilot studies. J Am Coll Cardiol 2006;48(8): 167281. [335] Channick RN, Olschewski H, Seeger W, Staub T, Voswinckel R, Rubin LJ. Safety and efficacy of inhaled treprostinil as add-on therapy to bosentan in pulmonary arterial hypertension. J Am Coll Cardiol 2006;48(7):14337. [336] Skoro-Sajer N, Bonderman D, Wiesbauer F, Harja E, Jakowitsch J, Klepetko W, et al. Treprostinil for severe inoperable chronic thromboembolic pulmonary hypertension. J Thromb Haemost 2007;5(3):4839.

I. PLATELET BIOLOGY

338

17. INHIBITION OF PLATELET FUNCTION BY THE ENDOTHELIUM

[337] McLaughlin VV, Benza RL, Rubin LJ, Channick RN, Voswinckel R, Tapson VF, et al. Addition of inhaled treprostinil to oral therapy for pulmonary arterial hypertension: a randomized controlled clinical trial. J Am Coll Cardiol 2010;55 (18):191522. [338] Hiremath J, Thanikachalam S, Parikh K, Shanmugasundaram S, Bangera S, Shapiro L, et al. Exercise improvement and plasma biomarker changes with intravenous treprostinil therapy for pulmonary arterial hypertension: a placebo-controlled trial. J Heart Lung Transplant 2010;29(2):13749. [339] Balbir-Gurman A, Braun-Moscovici Y, Livshitz V, Schapira D, Markovits D, Rozin A, et al. Antioxidant status after iloprost treatment in patients with Raynaud’s phenomenon secondary to systemic sclerosis. Clin Rheumatol 2007;26(9):151721. [340] Volpe A, Biasi D, Caramaschi P, Bambara LM, Carletto A, Degan M, et al. Iloprost infusion does not reduce oxidative stress in systemic sclerosis. Rheumatol Int 2008;28(4):3357. [341] Caramaschi P, Volpe A, Tinazzi I, Bambara LM, Carletto A, Biasi D. Does cyclically iloprost infusion prevent severe isolated pulmonary hypertension in systemic sclerosis? Preliminary results. Rheumatol Int 2006;27(2):2035. [342] Lessiani G, Vazzana N, Cuccurullo C, Di Michele D, Laurora G, Sgro G, et al. Inflammation, oxidative stress and platelet activation in aspirin-treated critical limb ischaemia: beneficial effects of iloprost. Thromb Haemost 2011;105(2):3218. [343] Creager MA, Pande RL, Hiatt WR. A. randomized trial of iloprost in patients with intermittent claudication. Vasc Med 2008;13(1):513. [344] Moriya H, Ishioka K, Honda K, Oka M, Maesato K, Ikee R, et al. Beraprost sodium, an orally active prostaglandin I(2) analog, improves renal anemia in hemodialysis patients with peripheral arterial disease. Ther Apher Dial 2010;14(5):4726. [345] Matsumoto T, Iwasa K, Kyuragi R, Honma K, Guntani A, Ohmine T, et al. The efficacy of oral beraprost sodium, a prostaglandin I2 analogue, for treating intermittent claudication in patients with arteriosclerosis obliterans. Int Angiol 2010;29(2 Suppl.):4954. [346] Arcangeli A, Rocca B, Salvatori G, Ciancia M, De Cristofaro R, Antonelli M. Heparin versus prostacyclin in continuous hemodiafiltration for acute renal failure: effects on platelet function in the systemic circulation and across the filter. Thromb Res 2010;126(1):2431. [347] Fabbri LP, Nucera M, Al Malyan M, Becchi C. Regional anticoagulation and antiaggregation for CVVH in critically ill patients: a prospective, randomized, controlled pilot study. Acta Anaesthesiol Scand 2010;54(1):927. [348] Spargias K, Adreanides E, Demerouti E, Gkouziouta A, Manginas A, Pavlides G, et al. Iloprost prevents contrastinduced nephropathy in patients with renal dysfunction undergoing coronary angiography or intervention. Circulation 2009;120(18):17939. [349] Kim KM, Kim HW, Lee JH, Chang JW, Park JS, Kim SB. Effects of beraprost sodium, an oral prostaglandin I2 analog, on hemostatic factors and inflammation in chronic peritoneal dialysis patients. Perit Dial Int 2009;29(2):17881. [350] Moncada S, Amezcua JL. Prostacyclin, thromboxane A2 interactions in haemostasis and thrombosis. Haemostasis 1979;8 (35):25265. [351] Vane JR. Inhibition of prostaglandin synthesis as a mechanism of action for aspirin-like drugs. Nat New Biol 1971;231 (25):2325. [352] Lopez-Farre A, Caramelo C, Esteban A, Alberola ML, Millas I, Monton M, et al. Effects of aspirin on platelet-neutrophil interactions. Role of nitric oxide and endothelin-1. Circulation 1995;91(7):20808.

[353] De La Cruz JP, Blanco E, Sanchez de la Cuesta F. Effect of dipyridamole and aspirin on the platelet-neutrophil interaction via the nitric oxide pathway. Eur J Pharmacol 2000;397 (1):3541. [354] De La Cruz JP, Arrebola MM, Guerrero A, Sanchez de la Cuesta F. Influence of nitric oxide on the in vivo antiaggregant effect of ticlopidine. Vascul Pharmacol 2002;38(3):1836. [355] Smith WL, Garavito RM, DeWitt DL. Prostaglandin endoperoxide H synthases (cyclooxygenases)-1 and -2. J Biol Chem 1996;271(52):3315760. [356] McAdam BF, Catella-Lawson F, Mardini IA, Kapoor S, Lawson JA, FitzGerald GA. Systemic biosynthesis of prostacyclin by cyclooxygenase (COX)-2: the human pharmacology of a selective inhibitor of COX-2. Proc Natl Acad Sci USA 1999;96(1):2727. [357] Pratico D, Tillmann C, Zhang ZB, Li H, FitzGerald GA. Acceleration of atherogenesis by COX-1-dependent prostanoid formation in low density lipoprotein receptor knockout mice. Proc Natl Acad Sci USA 2001;98(6):335863. [358] Nakayama T, Soma M, Takahashi Y, Uwabo J, Izumi Y, Kanmatsuse K. Novel polymorphic CA/TG repeat identified in the human prostacyclin synthase gene. Hum Hered 1997;47 (3):1767. [359] Nakayama T, Soma M, Watanabe Y, Hasimu B, Kanmatsuse K, Kokubun S, et al. Splicing mutation of the prostacyclin synthase gene in a family associated with hypertension. Adv Exp Med Biol 2003;525:1658. [360] Nakayama T, Soma M, Watanabe Y, Hasimu B, Sato M, Aoi N, et al. Splicing mutation of the prostacyclin synthase gene in a family associated with hypertension. Biochem Biophys Res Commun 2002;297(5):11359. [361] Nakayama T, Soma M, Haketa A, Aoi N, Kosuge K, Sato M, et al. Haplotype analysis of the prostacyclin synthase gene and essential hypertension. Hypertens Res 2003;26(7):5537. [362] Amano S, Tatsumi K, Tanabe N, Kasahara Y, Kurosu K, Takiguchi Y, et al. Polymorphism of the promoter region of prostacyclin synthase gene in chronic thromboembolic pulmonary hypertension. Respirology 2004;9(2):1849. [363] Iwai N, Katsuya T, Ishikawa K, Mannami T, Ogata J, Higaki J, et al. Human prostacyclin synthase gene and hypertension: The Suita Study. Circulation 1999;100(22):22316. [364] Nakayama T, Soma M, Kanmatsuse K. Organization of the human prostacyclin synthase gene and association analysis of a novel CA repeat in essential hypertension. Adv Exp Med Biol 1997;433:12730. [365] Nakayama T, Soma M, Rahmutula D, Izumi Y, Kanmatsuse K. Nonsense mutation of prostacyclin synthase gene in a family. Lancet 1997;349(9069):18878. [366] Nakayama T, Soma M, Rahmutula D, Tobe H, Sato M, Uwabo J, et al. Association study between a novel single nucleotide polymorphism of the promoter region of the prostacyclin synthase gene and essential hypertension. Hypertens Res 2002;25(1):658. [367] Nakayama T, Soma M, Rehemudula D, Takahashi Y, Tobe H, Satoh M, et al. Association of 5’ upstream promoter region of prostacyclin synthase gene variant with cerebral infarction. Am J Hypertens 2000;13(12):12637. [368] Nakayama T, Soma M, Saito S, Honye J, Yajima J, Rahmutula D, et al. Association of a novel single nucleotide polymorphism of the prostacyclin synthase gene with myocardial infarction. Am Heart J 2002;143(5):797801. [369] Nakayama T, Soma M, Takahashi Y, Rehemudula D, Tobe H, Sato M, et al. Polymorphism of the promoter region of prostacyclin synthase gene is not related to essential hypertension. Am J Hypertens 2001;14(5 Pt 1):40911.

I. PLATELET BIOLOGY

REFERENCES

[370] Chevalier D, Cauffiez C, Bernard C, Lo-Guidice JM, Allorge D, Fazio F, et al. Characterization of new mutations in the coding sequence and 5’-untranslated region of the human prostacylcin synthase gene (CYP8A1). Hum Genet 2001;108(2):14855. [371] Chevalier D, Allorge D, Lo-Guidice JM, Cauffiez C, Lepetit C, Migot-Nabias F, et al. Sequence analysis, frequency and ethnic distribution of VNTR polymorphism in the 5’-untranslated region of the human prostacyclin synthase gene (CYP8A1). Prostaglandins Other Lipid Mediat 2002;70(12):317. [372] Luft FC. Molecular genetics of human hypertension. Curr Opin Nephrol Hypertens 2000;9(3):25966. [373] Barbalic M, Narancic NS, Skaric-Juric T, Salihovic MP, Klaric IM, Lauc LB, et al. A quantitative trait locus for SBP maps near KCNB1 and PTGIS in a population isolate. Am J Hypertens 2009;22(6):6638. [374] Xie X, Ma YT, Fu ZY, Yang YN, Ma X, Chen BD, et al. Association of polymorphisms of PTGS2 and CYP8A1 with myocardial infarction. Clin Chem Lab Med 2009;47(3):34752. [375] Lemaitre RN, Rice K, Marciante K, Bis JC, Lumley TS, Wiggins KL, et al. Variation in eicosanoid genes, non-fatal myocardial infarction and ischemic stroke. Atherosclerosis 2009;204(2):e5863. [376] Poole EM, Bigler J, Whitton J, Sibert JG, Potter JD, Ulrich CM. Prostacyclin synthase and arachidonate 5-lipoxygenase polymorphisms and risk of colorectal polyps. Cancer Epidemiol Biomarkers Prev 2006;15(3):5028. [377] Stearman RS, Grady MC, Nana-Sinkam P, Varella-Garcia M, Geraci MW. Genetic and epigenetic regulation of the human prostacyclin synthase promoter in lung cancer cell lines. Mol Cancer Res 2007;5(3):295308. [378] Hashimoto K, Ishibashi K, Gebretsadik T, Hartert TV, Yamamoto A, Nakayama T, et al. Functional polymorphism of the promoter region of the prostacyclin synthase gene and severity of RSV infection in hospitalized children. J Med Virol 2008;80(11):201522. [379] Stitham J, Stojanovic A, Hwa J. Impaired receptor binding and activation associated with a human prostacyclin receptor polymorphism. J Biol Chem 2002;277(18): 1543944. [380] Forman BM, Chen J, Evans RM. Hypolipidemic drugs, polyunsaturated fatty acids, and eicosanoids are ligands for peroxisome proliferator-activated receptors alpha and delta. Proc Natl Acad Sci USA 1997;94(9):43127. [381] Patrignani P, Di Febbo C, Tacconelli S, Douville K, Guglielmi MD, Horvath RJ, et al. Differential association between human prostacyclin receptor polymorphisms and the development of venous thrombosis and intimal hyperplasia: a clinical biomarker study. Pharmacogenet Genomics 2008;18(7):61120. [382] Stitham J, Arehart E, Elderon L, Gleim SR, Douville K, Kasza Z, et al. Comprehensive biochemical analysis of rare prostacyclin receptor variants: study of association of signaling with coronary artery obstruction. J Biol Chem 2011;286(9):70609. [383] Plesner L. Ecto-ATPases: identities and functions. Int Rev Cytol 1995;158:141214. [384] Zimmermann H. Two novel families of ectonucleotidases: molecular structures, catalytic properties and a search for function. Trends Pharmacol Sci 1999;20(6):2316. [385] Wang TF, Guidotti G. CD39 is an ecto-(Ca21 ,Mg21 )-apyrase. J Biol Chem 1996;271(17):9898901. [386] Kaczmarek E, Koziak K, Sevigny J, Siegel JB, Anrather J, Beaudoin AR, et al. Identification and characterization of CD39/vascular ATP diphosphohydrolase. J Biol Chem 1996;271(51):3311622.

339

[387] Marcus AJ, Safier LB, Hajjar KA, Ullman HL, Islam N, Broekman MJ, et al. Inhibition of platelet function by an aspirininsensitive endothelial cell ADPase. Thromboregulation by endothelial cells. J Clin Invest 1991;88(5):16906. [388] Zimmermann H. 5’-Nucleotidase: molecular structure and functional aspects. Biochem J 1992;285(Pt. 2):34565. [389] Dwyer KM, Robson SC, Nandurkar HH, Campbell DJ, Gock H, Murray-Segal LJ, et al. Thromboregulatory manifestations in human CD39 transgenic mice and the implications for thrombotic disease and transplantation. J Clin Invest 2004;113 (10):14406. [390] Kansas GS, Wood GS, Tedder TF. Expression, distribution, and biochemistry of human CD39. Role in activationassociated homotypic adhesion of lymphocytes. J Immunol 1991;146(7):223544. [391] Marcus AJ, Broekman MJ, Drosopoulos JH, Islam N, Alyonycheva TN, Safier LB, et al. The endothelial cell ectoADPase responsible for inhibition of platelet function is CD39. J Clin Invest 1997;99(6):135160. [392] Dombrowski KE, Trevillyan JM, Cone JC, Lu Y, Phillips CA. Identification and partial characterization of an ectoATPase expressed by human natural killer cells. Biochemistry 1993;32 (26):651522. [393] Maliszewski CR, Delespesse GJ, Schoenborn MA, Armitage RJ, Fanslow WC, Nakajima T, et al. The CD39 lymphoid cell activation antigen. Molecular cloning and structural characterization. J Immunol 1994;153(8):357483. [394] Koziak K, Sevigny J, Robson SC, Siegel JB, Kaczmarek E. Analysis of CD39/ATP diphosphohydrolase (ATPDase) expression in endothelial cells, platelets and leukocytes. Thromb Haemost 1999;82(5):153844. [395] Schulte am Esch II J, Sevigny J, Kaczmarek E, Siegel JB, Imai M, Koziak K, et al. Structural elements and limited proteolysis of CD39 influence ATP diphosphohydrolase activity. Biochemistry 1999;38(8):224858. [396] Handa M, Guidotti G. Purification and cloning of a soluble ATP-diphosphohydrolase (apyrase) from potato tubers (Solanum tuberosum). Biochem Biophys Res Commun 1996;218 (3):91623. [397] Grinthal A, Guidotti G. Substitution of His59 converts CD39 apyrase into an ADPase in a quaternary structure dependent manner. Biochemistry 2000;39(1):916. [398] Drosopoulos JH, Broekman MJ, Islam N, Maliszewski CR, Gayle III RB, Marcus AJ. Site-directed mutagenesis of human endothelial cell ecto-ADPase/soluble CD39: requirement of glutamate 174 and serine 218 for enzyme activity and inhibition of platelet recruitment. Biochemistry 2000;39(23):693643. [399] Vasconcelos EG, Nascimento PS, Meirelles MN, VerjovskiAlmeida S, Ferreira ST. Characterization and localization of an ATP-diphosphohydrolase on the external surface of the tegument of Schistosoma mansoni. Mol Biochem Parasitol 1993;58(2):20514. [400] Vasconcelos EG, Ferreira ST, Carvalho TM, Souza W, Kettlun AM, Mancilla M, et al. Partial purification and immunohistochemical localization of ATP diphosphohydrolase from Schistosoma mansoni. Immunological cross-reactivities with potato apyrase and Toxoplasma gondii nucleoside triphosphate hydrolase. J Biol Chem 1996;271(36):2213945. [401] Wang TF, Ou Y, Guidotti G. The transmembrane domains of ectoapyrase (CD39) affect its enzymatic activity and quaternary structure. J Biol Chem 1998;273(38):2481421. [402] Zhong X., Kriz R., Kumar R., Guidotti G. Distinctive roles of endoplasmic reticulum and golgi glycosylation in functional surface expression of mammalian E-NTPDase1, CD39. Biochim Biophys Acta. 2005.

I. PLATELET BIOLOGY

340

17. INHIBITION OF PLATELET FUNCTION BY THE ENDOTHELIUM

[403] Zimmermann H, Braun N, Kegel B, Heine P. New insights into molecular structure and function of ectonucleotidases in the nervous system. Neurochem Int 1998;32(56):4215. [404] Kegel B, Braun N, Heine P, Maliszewski CR, Zimmermann H. An ecto-ATPase and an ecto-ATP diphosphohydrolase are expressed in rat brain. Neuropharmacology 1997;36(9): 1189200. [405] Koziak K, Kaczmarek E, Kittel A, Sevigny J, Blusztajn JK, Schulte Am Esch II J, et al. Palmitoylation targets CD39/endothelial ATP diphosphohydrolase to caveolae. J Biol Chem 2000;275(3):205762. [406] Robson SC, Kaczmarek E, Siegel JB, Candinas D, Koziak K, Millan M, et al. Loss of ATP diphosphohydrolase activity with endothelial cell activation. J Exp Med 1997;185(1): 15363. [407] Krotz F, Sohn HY, Keller M, Gloe T, Bolz SS, Becker BF, et al. Depolarization of endothelial cells enhances platelet aggregation through oxidative inactivation of endothelial NTPDase. Arterioscler Thromb Vasc Biol 2002;22(12): 20039. [408] Kittel A, Kaczmarek E, Sevigny J, Lengyel K, Csizmadia E, Robson SC. CD39 as a caveolar-associated ectonucleotidase. Biochem Biophys Res Commun 1999;262(3):5969. [409] Papanikolaou A., Papafotika A., Murphy C., Papamarcaki T., Tsolas O., Drab M., et al. Cholesterol-dependent lipid assemblies regulate the activity of the ecto-nucleotidase CD39. J Biol Chem. 2005. [410] Marcus AJ, Safier LB, Broekman MJ, Islam N, Fliessbach JH, Hajjar KA, et al. Thrombosis and inflammation as multicellular processes: significance of cellcell interactions. Thromb Haemost 1995;74(1):2137. [411] Marcus AJ, Broekman MJ, Drosopoulos JH, Pinsky DJ, Islam N, Maliszewsk CR. Inhibition of platelet recruitment by endothelial cell CD39/ecto-ADPase: significance for occlusive vascular diseases. Ital Heart J 2001;2(11):82430. [412] Marcus A, Broekman M, Drosopoulos J, Pinsky D, Islam N, Gayle III R, et al. Thromboregulation by endothelial cells: significance for occlusive vascular diseases. Arterioscler Thromb Vasc Biol 2001;21(2):17882. [413] Kawashima Y, Nagasawa T, Ninomiya H. Contribution of ecto-5’-nucleotidase to the inhibition of platelet aggregation by human endothelial cells. Blood 2000;96(6):215762. [414] Cote YP, Filep JG, Battistini B, Gauvreau J, Sirois P, Beaudoin AR. Characterization of ATP-diphosphohydrolase activities in the intima and media of the bovine aorta: evidence for a regulatory role in platelet activation in vitro. Biochim Biophys Acta 1992;1139(12):13342. [415] Linden J. Molecular approach to adenosine receptors: receptor-mediated mechanisms of tissue protection. Annu Rev Pharmacol Toxicol 2001;41:77587. [416] Yang D, Chen H, Koupenova M, Carroll SH, Eliades A, Freedman JE, et al. A new role for the A2b adenosine receptor in regulating platelet function. J Thromb Haemost 2010;8 (4):81727. [417] Koszalka P, Ozuyaman B, Huo Y, Zernecke A, Flogel U, Braun N, et al. Targeted disruption of cd73/ecto-5’-nucleotidase alters thromboregulation and augments vascular inflammatory response. Circ Res 2004;95(8):81421. [418] Vial C, Hechler B, Leon C, Cazenave JP, Gachet C. Presence of P2X1 purinoceptors in human platelets and megakaryoblastic cell lines. Thromb Haemost 1997;78(6):15004. [419] Leon C, Hechler B, Vial C, Leray C, Cazenave JP, Gachet C. The P2Y1 receptor is an ADP receptor antagonized by ATP and expressed in platelets and megakaryoblastic cells. FEBS Lett 1997;403(1):2630.

[420] Daniel JL, Dangelmaier C, Jin J, Ashby B, Smith JB, Kunapuli SP. Molecular basis for ADP-induced platelet activation. I. Evidence for three distinct ADP receptors on human platelets. J Biol Chem 1998;273(4):20249. [421] Leon C, Vial C, Cazenave JP, Gachet C. Cloning and sequencing of a human cDNA encoding endothelial P2Y1 purinoceptor. Gene 1996;171(2):2957. [422] Motte S, Communi D, Pirotton S, Boeynaems JM. Involvement of multiple receptors in the actions of extracellular ATP: the example of vascular endothelial cells. Int J Biochem Cell Biol 1995;27(1):17. [423] Fijnheer R, Boomgaard MN, van den Eertwegh AJ, Homburg CH, Gouwerok CW, Veldman HA, et al. Stored platelets release nucleotides as inhibitors of platelet function. Thromb Haemost 1992;68(5):5959. [424] Enjyoji K, Sevigny J, Lin Y, Frenette PS, Christie PD, Esch II JS, et al. Targeted disruption of CD39/ATP diphosphohydrolase results in disordered hemostasis and thromboregulation. Nat Med 1999;5(9):10107. [425] Kittel A, Csapo ZS, Csizmadia E, Jackson SW, Robson SC. Colocalization of P2Y1 receptor and NTPDase1/CD39 within caveolae in human placenta. Eur J Histochem 2004;48 (3):2539. [426] Boeynaems JM, Pearson JD. P2 purinoceptors on vascular endothelial cells: physiological significance and transduction mechanisms. Trends Pharmacol Sci 1990;11(1):347. [427] Coppola A, Coppola L, dalla Mora L, Limongelli FM, Grassia A, Mastrolorenzo L, et al. Vigorous exercise acutely changes platelet and B-lymphocyte CD39 expression. J Appl Physiol 2005;98(4):14149. [428] Gayle III RB, Maliszewski CR, Gimpel SD, Schoenborn MA, Caspary RG, Richards C, et al. Inhibition of platelet function by recombinant soluble ecto-ADPase/CD39. J Clin Invest 1998;101(9):18519. [429] Candinas D, Koyamada N, Miyatake T, Siegel J, Hancock WW, Bach FH, et al. Loss of rat glomerular ATP diphosphohydrolase activity during reperfusion injury is associated with oxidative stress reactions. Thromb Haemost 1996;76 (5):80712. [430] Koyamada N, Miyatake T, Candinas D, Hechenleitner P, Siegel J, Hancock WW, et al. Apyrase administration prolongs discordant xenograft survival. Transplantation 1996;62 (12):173943. [431] Robson SC, Wu Y, Sun X, Knosalla C, Dwyer K, Enjyoji K. Ectonucleotidases of CD39 family modulate vascular inflammation and thrombosis in transplantation. Semin Thromb Hemost 2005;31(2):21733. [432] Belayev L, Khoutorova L, Deisher TA, Belayev A, Busto R, Zhang Y, et al. Neuroprotective effect of SolCD39, a novel platelet aggregation inhibitor, on transient middle cerebral artery occlusion in rats. Stroke 2003;34(3):75863. [433] Marcus AJ, Broekman MJ, Drosopoulos JH, Islam N, Pinsky DJ, Sesti C, et al. Metabolic control of excessive extracellular nucleotide accumulation by CD39/ecto-nucleotidase-1: implications for ischemic vascular diseases. J Pharmacol Exp Ther 2003;305(1):916. [434] Koziak K, Bojakowska M, Robson SC, Bojakowski K, Soin J, Csizmadia E, et al. Overexpression of CD39/nucleoside triphosphate diphosphohydrolase-1 decreases smooth muscle cell proliferation and prevents neointima formation after angioplasty. J Thromb Haemost 2008;6(7): 11917. [435] Pinsky DJ, Broekman MJ, Peschon JJ, Stocking KL, Fujita T, Ramasamy R, et al. Elucidation of the thromboregulatory role

I. PLATELET BIOLOGY

REFERENCES

[436]

[437]

[438]

[439]

[440]

[441]

[442]

[443]

[444]

[445]

[446]

[447]

[448]

[449]

of CD39/ectoapyrase in the ischemic brain. J Clin Invest 2002;109(8):103140. Drosopoulos JH, Kraemer R, Shen H, Upmacis RK, Marcus AJ, Musi E. Human solCD39 inhibits injury-induced development of neointimal hyperplasia. Thromb Haemost 2010;103 (2):42634. Haller CA, Cui W, Wen J, Robson SC, Chaikof EL. Reconstitution of CD39 in liposomes amplifies nucleoside triphosphate diphosphohydrolase activity and restores thromboregulatory properties. J Vasc Surg 2006;43(4): 81623. Guckelberger O, Sun XF, Sevigny J, Imai M, Kaczmarek E, Enjyoji K, et al. Beneficial effects of CD39/ecto-nucleoside triphosphate diphosphohydrolase-1 in murine intestinal ischemia-reperfusion injury. Thromb Haemost 2004;91(3): 57686. Gangadharan SP, Imai M, Rhynhart KK, Sevigny J, Robson SC, Conte MS. Targeting platelet aggregation: CD39 gene transfer augments nucleoside triphosphate diphosphohydrolase activity in injured rabbit arteries. Surgery 2001;130(2): 296303. Mazer S, Fedarau M, Liu Y-L, Huang DW, Towe CW, Liu CF, et al. Deletion of endothelial ectoapyrase (CD39) promotes atherogenesis in hyperlipidemic mice. American Heart Scientific Sessions, November 2004. Friedman DJ, Talbert ME, Bowden DW, Freedman BI, Mukanya Y, Enjyoji K, et al. Functional ENTPD1 polymorphisms in African Americans with diabetes and end-stage renal disease. Diabetes 2009;58(4):9991006. Friedman DJ, Kunzli BM, A-Rahim YI, Sevigny J, Berberat PO, Enjyoji K, et al. CD39 deletion exacerbates experimental murine colitis and human polymorphisms increase susceptibility to inflammatory bowel disease. Proc Natl Acad Sci USA 2009;106(39):1678893. Kunzli BM, Berberat PO, Dwyer K, Deaglio S, Csizmadia E, Cowan P, et al. Variable impact of CD39 in experimental murine colitis. Dig Dis Sci 2011;56(5):1393403. Poirier O, Mao C, Mallet C, Nicaud V, Herrmann SM, Evans A, et al. Polymorphisms of the endothelial nitric oxide synthase gene—no consistent association with myocardial infarction in the ECTIM study. Eur J Clin Invest 1999;29(4):28490. Jeerooburkhan N, Jones LC, Bujac S, Cooper JA, Miller GJ, Vallance P, et al. Genetic and environmental determinants of plasma nitrogen oxides and risk of ischemic heart disease. Hypertension 2001;38(5):105461. Granath B, Taylor RR, van Bockxmeer FM, Mamotte CD. Lack of evidence for association between endothelial nitric oxide synthase gene polymorphisms and coronary artery disease in the Australian Caucasian population. J Cardiovasc Risk 2001;8 (4):23541. Alvarez R, Gonzalez P, Batalla A, Reguero JR, IglesiasCubero G, Hevia S, et al. Association between the NOS3 (-786 T/C) and the ACE (I/D) DNA genotypes and early coronary artery disease. Nitric Oxide 2001;5(4):3438. Rossi GP, Cesari M, Zanchetta M, Colonna S, Maiolino G, Pedon L, et al. The T-786C endothelial nitric oxide synthase genotype is a novel risk factor for coronary artery disease in Caucasian patients of the GENICA study. J Am Coll Cardiol 2003;41(6):9307. Ghilardi G, Biondi ML, DeMonti M, Bernini M, Turri O, Massaro F, et al. Independent risk factor for moderate to severe internal carotid artery stenosis: T786C mutation of the endothelial nitric oxide synthase gene. Clin Chem 2002;48(7): 98993.

341

[450] Sharan K, Surrey S, Ballas S, Borowski M, Devoto M, Wang KF, et al. Association of T-786C eNOS gene polymorphism with increased susceptibility to acute chest syndrome in females with sickle cell disease. Br J Haematol 2004;124(2): 2403. [451] Hyndman ME, Parsons HG, Verma S, Bridge PJ, Edworthy S, Jones C, et al. The T-786–.C mutation in endothelial nitric oxide synthase is associated with hypertension. Hypertension 2002;39(4):91922. [452] Hwang JJ, Tsai CT, Yeh HM, Chiang FT, Hsu KL, Tseng CD, et al. The 27-bp tandem repeat polymorphism in intron 4 of the endothelial nitric oxide synthase gene is not associated with coronary artery disease in a hospital-based Taiwanese population. Cardiology 2002;97(2):6772. [453] Cine N, Hatemi AC, Erginel-Unaltuna N. Association of a polymorphism of the ecNOS gene with myocardial infarction in a subgroup of Turkish MI patients. Clin Genet 2002;61 (1):6670. [454] Benjafield AV, Morris BJ. Association analyses of endothelial nitric oxide synthase gene polymorphisms in essential hypertension. Am J Hypertens 2000;13(9):9948. [455] Wang XL, Sim AS, Badenhop RF, McCredie RM, Wilcken DE. A smoking-dependent risk of coronary artery disease associated with a polymorphism of the endothelial nitric oxide synthase gene. Nat Med 1996;2(1):415. [456] Spence MS, McGlinchey PG, Patterson CC, Allen AR, Murphy G, Bayraktutan U, et al. Endothelial nitric oxide synthase gene polymorphism and ischemic heart disease. Am Heart J 2004;148(5):84751. [457] Jia CQ, Ning Y, Liu TT, Liu ZL. Association between G894T mutation in endothelial nitric oxide synthase gene and premature coronary heart disease. Zhonghua Liu Xing Bing Xue Za Zhi 2005;26(1):513. [458] Shimasaki Y, Yasue H, Yoshimura M, Nakayama M, Kugiyama K, Ogawa H, et al. Association of the missense Glu298Asp variant of the endothelial nitric oxide synthase gene with myocardial infarction. J Am Coll Cardiol 1998;31 (7):150610. [459] Hingorani AD, Liang CF, Fatibene J, Lyon A, Monteith S, Parsons A, et al. A common variant of the endothelial nitric oxide synthase (Glu298–.Asp) is a major risk factor for coronary artery disease in the UK. Circulation 1999;100 (14):151520. [460] Lembo G, De Luca N, Battagli C, Iovino G, Aretini A, Musicco M, et al. A common variant of endothelial nitric oxide synthase (Glu298Asp) is an independent risk factor for carotid atherosclerosis. Stroke 2001;32(3):73540. [461] Chang K, Baek SH, Seung KB, Kim PJ, Ihm SH, Chae JS, et al. The Glu298Asp polymorphism in the endothelial nitric oxide synthase gene is strongly associated with coronary spasm. Coron Artery Dis 2003;14(4):2939. [462] Wolff B, Grabe HJ, Schluter C, Popowski K, Volzke H, Ludemann J, et al. Endothelial nitric oxide synthase Glu298Asp gene polymorphism, blood pressure and hypertension in a general population sample. J Hypertens 2005;23 (7):13616. [463] Miyamoto Y, Saito Y, Kajiyama N, Yoshimura M, Shimasaki Y, Nakayama M, et al. Endothelial nitric oxide synthase gene is positively associated with essential hypertension. Hypertension 1998;32(1):38. [464] Kishimoto T, Misawa Y, Kaetu A, Nagai M, Osaki Y, Okamoto M, et al. eNOS Glu298Asp polymorphism and hypertension in a cohort study in Japanese. Prev Med 2004;39(5):92731.

I. PLATELET BIOLOGY

342

17. INHIBITION OF PLATELET FUNCTION BY THE ENDOTHELIUM

[465] Lacolley P, Gautier S, Poirier O, Pannier B, Cambien F, Benetos A. Nitric oxide synthase gene polymorphisms, blood pressure and aortic stiffness in normotensive and hypertensive subjects. J Hypertens 1998;16(1):315. [466] Elbaz A, Poirier O, Moulin T, Chedru F, Cambien F, Amarenco P. Association between the Glu298Asp polymorphism in the endothelial constitutive nitric oxide synthase gene and brain infarction. The GENIC Investigators. Stroke 2000;31(7):16349. [467] Stangl K, Cascorbi I, Laule M, Klein T, Stangl V, Rost S, et al. High CA repeat numbers in intron 13 of the endothelial

nitric oxide synthase gene and increased risk of coronary artery disease. Pharmacogenetics 2000;10(2):13340. [468] Bonnardeaux A, Nadaud S, Charru A, Jeunemaitre X, Corvol P, Soubrier F. Lack of evidence for linkage of the endothelial cell nitric oxide synthase gene to essential hypertension. Circulation 1995;91(1):96102. [469] Nakayama T, Soma M, Takahashi Y, Izumi Y, Kanmatsuse K, Esumi M. Association analysis of CA repeat polymorphism of the endothelial nitric oxide synthase gene with essential hypertension in Japanese. Clin Genet 1997;51(1): 2630.

I. PLATELET BIOLOGY