Zum Hauptinhalt springen

Prostanoid function and cardiovascular disease

INIGUEZ, Miguel A ; CACHEIRO-LLAGUNO, Cristina ; et al.
In: Archives of physiology and biochemistry, Jg. 114 (2008), Heft 3, S. 201-209
Online academicJournal - print, 2 p.1/4

Prostanoid function and cardiovascular disease. 

Prostanoids, including prostaglandins (PGs) and thromboxanes (TXs) are synthesized from arachidonic acid by the combined action of cyclooxygenases (COXs) and PG and TX synthases. Finally after their synthesis, prostanoids are quickly released to the extracellular medium exerting their effects upon interaction with prostanoid receptors present in the neighbouring cells. These agents exert important actions in the cardiovascular system, modulating vascular homeostasis and participating in the pathogenesis of vascular diseases as thrombosis and atherosclerosis. Among prostanoids, Tromboxane (TX)A2, a potent platelet activator and vasoconstrictor and prostacyclin (PGI2), a platelet inhibitor and vasodilator, are the most important in controlling vascular homeostasis. Although multiple studies using pharmacological inhibitors and genetically deficient mice have demonstrated the importance of prostanoid-mediated actions on cardiovascular physiology, further analysis on the prostanoid mediated actions in the vascular system are required to better understand the benefits and risks for the use of COX inhibitors in cardiovascular diseases.

Keywords: Prostaglandins; thromboxanes; cyclooxygenases; prostanoid receptors; NSAIDs; cardiovascular physiopathology

Abbreviations COX Cyclooxygenase DP PGD receptor EP PGE receptor FP PGF receptor IL Interleukin IP PGI receptor NSAIDs non-steroidal anti-inflammatory drugs PG prostaglandin PGES PGE synthase PGDS PGD synthase PGFS PGF synthase PGIS PGI synthase TP Thromboxane receptor TX Thromboxane TXS Thromboxane synthase

Introduction

Arachidonic acid, liberated from membrane phospholipids by several phospholipases, is metabolized by the sequential action of cyclooxygenases (COX) and prostaglandin or thromboxane synthases to produce the diverse classes of prostanoids (Simmons et al., [64]; Smith et al., [65]). These agents are a group of bioactive lipids including prostaglandins (PGs) and thromboxanes (TXs) that play a very important role in many physiological and pathological processes, including inflammation (Williams et al., [84]), cancer (Subbaramaiah & Dannenberg, [68]), angiogenesis (Iñiguez et al., 2003) and cardiovascular diseases (FitzGerald et al., [24]; Davidge, [20]; Alfranca et al., [1]; Belton & Fitzgerald, [4]). COX enzymes catalyse the formation of an unstable endoperoxide intermediate, PGH2, which in turn can be metabolized by cell-specific isomerases and synthases to a range of eicosanoids with potent and diverse biological effects, as PGD2, PGE2, PGF, PGI2, and TXA2. Prostanoids released to the extracellular medium exert their biological effects in an autocrine or paracrine fashion upon interaction with prostanoid receptors present in target cells (Figure 1).

Graph: Figure 1. Biosynthesis of prostanoids. Arachidonic acid is liberated from the membrane phospholipids by phospholipase A2 (PLA2) and converted to PGH2 and then to PGG2 by cyclooxygenases (COX-1 or COX-2). These enzymes are the target of NSAIDs that inhibit PG and TX synthesis. Glucocorticoids diminish prostanoid production through the inhibition of COX-2 transcriptional induction by inflammatory stimuli. Subsequent conversion of PGH2 to different prostanoids (PGs, prostaglandins and TXs, thromboxanes) is catalyzed by the respective synthases and isomerases. Prostanoids signal through G protein-coupled receptors with seven trans-membrane domains. PGE2 signals via four different receptors (EP1, EP2, EP3 and EP4). PGD2, PGF2α, PGI and TXA2 bind to the DP and CRTH2, FP, IP and TP receptors respectively.

In the cardiovascular system, prostanoids have been shown to modulate the pathogenesis of vascular diseases as thrombosis and atherosclerosis through a variety of processes, including platelet aggregation, vasorelaxation and vasoconstriction, local inflammatory response and leukocyte-endothelial cell adhesion (Vila, [78]; Kobayashi et al., [43]). Multiple studies using NSAIDs as pharmacological inhibitors of COXs and genetically deficient mice have revealed the importance of prostanoid-mediated actions on cardiovascular physiology (Belton & Fitzgerald, [4]; Burleigh et al., [10]; FitzGerald & Patrono, [25]; Fosslien, [27]; Ray et al., [56]; Martinez-Gonzalez & Badimon, [46]). However, recent withdrawal of COX-2 selective inhibitors from the clinic because of their adverse effects in patients with potential cardiovascular risk has opened a debate about the role of COX–derived prostanoids in vascular pathologies and the benefits and risks for the use of COX inhibitors in cardiovascular diseases.

Biosynthesis of prostanoids

Cyclooxygenases

Two main COX isoenzymes have been identified, named COX-1 and COX-2, encoded by two separate genes (Smith et al., [65]; Simmons et al., [64]; Warner & Mitchell, [82]). A COX-3 enzyme derived from alternative splicing of the COX-1 gene has been also described, although its role is still unclear (Chandrasekharan et al., [16]). COX-1 is constitutively expressed in most cell types and tissues being responsible for vascular, renal and gastric homeostasis (Morita, [48]). In contrast, COX-2 expression is generally induced at sites of inflammation by many stimuli that includes among others, pro-inflammatory cytokines as interleukin (IL)-1β, tumour necrosis factor (TNF)-α, growth factors, mechanical stress, oxidized lipids, free radicals and bacterial products (Smith et al., [65]; Tanabe & Tohnai, [69]). Those stimuli activate many transcription factors such as nuclear factor (NF)-κB, NF-IL6 (C/EBP), cAMP response element-binding protein (CREBP), Activator protein (AP)-1, interferon regulatory factors (IRFs) and nuclear factor of activated T cells (NFAT) (Tanabe & Tohnai, [69]; Blanco et al., [7]; Iniguez et al., [36]) that induce COX-2 transcription in different cell types. COX-2 transcripcional induction can be inhibited by anti-inflammatory and immunosuppressive drugs as glucocorticoids and Cyclosporin A (Iniguez et al., [37]; Kujubu & Herschman, [44]) as well as by anti-inflammatory cytokines such as IL-4, IL-10 and IL-13 (Niiro et al., [53]; Diaz-Cazorla et al., [21]). Increasing research on the physiological roles of COX-2 has revealed that, in addition to inflammation, this isoform also plays an important role in renal homeostasis, mucosal defence, reproduction and vascular homeostasis (Morita, [48]; Simmons et al., [64]; Warner & Mitchell, [82]).

Cyclooxygenases are the target of non-steroidal anti-inflammatory drugs (NSAIDs). According to their selectivity in the inhibition of both COX isoforms, these drugs can be divided into classic NSAIDs, such as aspirin and ibuprofen that inhibit both isoforms at standard doses, and highly selective COX-2 NSAIDs as those belonging to the family of "coxibs". The fact that COX-1 was mainly involved in house-keeping functions and COX-2 was associated to inflammation, led to the theory that inhibition of COX-1 may account for some of the unwanted side effects of these drugs such as gastrointestinal and renal toxicity whereas the ability of NSAIDs to inhibit COX-2 activity may explain their therapeutic effects as anti-inflammatory drugs. Therefore, most of the new research on anti-inflammatory drugs was aimed at targeting the COX-2 inducible production of PGs (Dannhardt & Kiefer, [19]; Simmons et al., [64]). As a consequence, new drugs with high selectivity against COX-2 such as Celecoxib and Rofecoxib and the newest coxibs Etoricoxib, Valdecoxib, and Lumiracoxib, have been developed with the idea of providing safer anti-inflammatory compounds with reduced gastrointestinal and renal toxicity. However, although these drugs have been proved to be potent anti-inflammatory compounds with less gastric damage than conventional, non-selective COX inhibitors, their use is still associated with significant gastrointestinal injury and renal side effects. (Warner et al., [81]; Rostom et al., [60]; Coruzzi et al., [15]). Particular concerns have emerged by the fact that some coxibs increase the risk of serious cardiovascular events, including myocardial infarction and stroke (Fosslien, [27]; Caldwell et al., [12]; Ray et al., [56]). This has led to the withdrawal of rofecoxib from the market, and has raised serious questions about the cardiovascular safety of NSAIDs (Kearney et al., [42]; Martinez-Gonzalez & Badimon, [46]; McGettigan & Henry, [47]). It is also important to note that, although it is generally accepted that these drugs exert their anti-inflammatory and analgesic actions through inhibition of COX enzymatic activity, increasing evidence has shown that certain actions of NSAIDs could be mediated through mechanisms independent of cyclooxygenase activity and prostaglandin production that may be relevant to their effects in vivo (Tegeder et al., [71]).

Prostaglandin and Thromboxane synthases

Despite the importance of COX isoenzymes in prostanoid generation, the actual prostanoid profile synthesized by a particular cell type is determined by the presence of different downstream PG and TX synthases and isomerases (Helliwell et al., [33]) (Figure 1). A particular cell type may predominantly express a particular isomerase, which will largely determine which prostanoid is generated. Moreover, some cell types can express more than one isomerase and a particular COX isoenzyme can couple to different isomerases in the same cell (Ueno et al., [75]). Thus, COX-1 preferentially couples with Thromboxane synthase (TXS) and PGF synthase (PGFS), while COX-2 is associated to Prostacyclin synthase (PGIS) and microsomal PGE2 synthase-1 (mPGES-1) (Murakami et al., [49]; Ueno et al., [75]). Evenmore, PGH2 and other endoperoxides generated in one cell type may be secreted and metabolized by isomerases present in surrounding cells by the so-called "transcellular metabolism" (Karim et al., [41]).

PGE 2 and PGD 2 synthases

Three types of PGE synthases (PGES) participating in the synthesis of PGE2 have been described: one cytosolic (cPGES) and two membrane-associated PGE synthases (mPGES)-1 and -2. The (cPGES) belongs to the glutathione transferase family, is ubiquitously expressed and is preferentially functionally coupled to COX-1, thus, involved in housekeeping production of PGE2 (Murakami et al., [51]; Tanioka et al., [70]). In contrast, mPGES-1, which belongs to the membrane-associated proteins involved in Eicosanoid and Glutathione metabolism (MAPEG) superfamily, is inducible by similar stimuli that induce COX-2, being its induction also suppressed by glucocorticoids. Moreover, mPGES-1 appears functionally coupled with COX-2 and its induction is usually co-ordinated with COX-2 (Samuelsson et al., [62]). A second type of membrane-bound microsomal PGES is the mPGES-2, able to couple with both COX isoenzymes, although its functional significance is still unclear (Murakami et al., [50]).

PGD2 is synthesized from PGH2 by the action of PGD synthases (PGDS) of which there are two isoforms. The haematopoietic PGDS is present in mast cells, basophils, and in a subset of T helper cells (Th2) and may play a role in allergy (Kanaoka & Urade, [40]). The lipocalin-type PGD synthase is mostly expressed in brain and it is especially abundant in the cerebrospinal fluid (Urade & Hayaishi, [76]). Interestingly, PGD2 can be further metabolized by dehydration to PGJ2, delta12-PGJ2, and 15-deoxy-delta(12,14)-PGJ2, which is a ligand for the nuclear transcription factor peroxisomal proliferator activated receptor (PPAR)-gamma having anti-inflammatory activity (Scher & Pillinger, [63]).

Synthesis of PGF 2α , PGI 2 and TXA 2

PGF can be synthesized from PGH2 by PGH 9-11-endoperoxide reductase or PGF synthase (PGFS). In addition, it can be generated from PGE2 by PGE 9-ketoreductase or from PGD2 by PGD-11-ketoreductase (Watanabe, [83]).

Prostacyclin, PGI2, is produced by the action of PGI synthase (PGIS) which can be induced by mechanical stress, via activation of AP-1 (Wu & Liou, [85]).

TXA2 is synthesized from PGH2 by thromboxane synthase (TXS), which is constitutively expressed in platelets as well as other blood cells and various tissues as kidney, lung, liver and placenta. Its regulation takes place mainly at the transcriptional level (Wang & Kulmacz, [79]).

Prostanoid receptors

Prostanoids actions, with the possible exception of cyclopentenone PGs, are mediated through cell surface receptors. These are seven membrane spanning G-protein coupled receptors that are classified into five basic types, named according to their ligands: DP for PGD2, EP for PGE2, FP for PGF2, IP for PGI2, and TP for TXA2 (Alfranca et al., [1]; Hata & Breyer, [32]).

DP (also named DP1) is weakly expressed in brain despite the important action of PGD2 in this organ. It is moderately expressed in the ileum with very weak expression in the lung. DP2, also called CRTH2 (chemoattractant receptor-homologous molecule expressed on Th2 cells), is expressed on Th2 immune cells being a chemoattractant receptor for Th2 lymphocyte subset in allergic inflammation (Kabashima & Narumiya, [39]).

FP is most abundantly expressed in the corpus luteum according to the role played by PGF2α in menstrual cycle. It is also expressed in the heart, lung, kidney, and stomach, although its expression in these tissues does not vary during the estrous cycle as does in the corpus luteum (Tsuboi et al., [74]). There are several splice variants of FP (FPA and FPB) but its particular role is unclear.

IP is expressed in larger quantities by dorsal root ganglion neurons but also by platelet precursors (megakaryocytes) as well as in the smooth muscle cells of arteries consistent with the important action of PGI2 in the cardiovascular system (Smyth & FitzGerald, [66]).

PGE2 has four receptors, EP1 to 4. In addition, there are several splice variants of EP3. More importantly, each one is linked to a different transduction pathway that may even give rise to opposite effects (activation or inhibition) on cellular responses (Alfranca et al., [1]). Thus, EP1 induces an inhibition of adenylate cyclase leading to a decrease in cAMP whereas EP2 and EP4 receptors activate this enzyme. On the other hand, EP3 is coupled to Gαq and its activation results in intracellular calcium increase. EP3 and EP4 receptors have a wide distribution throughout the body, being expressed in almost all tissues examined. In contrast, of EP1 is restricted to the lung, kidney and stomach, and EP2 is scarcely expressed.

In the cardiovascular system, IP, DP, EP4 and EP2 receptors that mediate cAMP increases are termed "relaxant" receptors, whereas TP, FP, and EP1 receptors, which induce calcium mobilization, represent the "contractile" receptor group. EP3, which reduces cAMP levels, has been named the "inhibitory" receptor (Alfranca et al., [1]).

Nuclear actions of prostanoids have also been reported. Thus, PGJ2 derivatives can bind and activate peroxisome proliferator-activated receptors (PPARs) nuclear transcription factors (Straus & Glass, [67]). There are four types of PPARs (alpha, delta, gamma-1, and gamma-2), which may bind various PGs with different sensitivity (Berger & Moller, [6]). In this way, PGs may also act as intracellular signalling molecules and regulate gene expression (Ide et al., [35]). Generally PPARs induces transcription of anti-inflammatory genes as well as inhibits activation of pro-inflammatory ones (Straus & Glass, [67]). Besides, cyclopentenone PGs (that includes PGA1) have a reactive ring which may lead to have receptor-independent actions through redox alterations (Grau et al., [30]).

Prostanoids in cardiovascular homeostasis

TXA2 and PGI2 are thought to be the most important prostanoids in controlling the homeostasis of the cardiovascular system, as proposed more than 20 years ago by Bunting, Moncada and Vane (Bunting et al., [9]; Davidge, [20]; Cheng et al., [17]). They are synthesized by blood platelets and vascular endothelium, respectively, and have opposed biological activities. TXA2 is potent vasoconstrictor as well as a potent inducer of platelet adhesion and regulates renal hemodynamics and sodium handling by the kidney (FitzGerald et al., [24]). On the other hand, PGI2 is the predominant prostanoid produced by cells of the vasculature, having an important vasodilator effect by promoting renal sodium excretion and regulating the growth of vascular smooth muscle cells. Moreover, it opposes TXA2 effects on platelets inhibiting their aggregation and action (Helliwell et al., [33]; Yokoyama et al., [87]).

Due to this, many aspects of cardiovascular disease have traditionally been explained by alterations in the balance between PGI2 and TXA2 during the interactions between platelets and vessel wall (Vane, [77]) although recent studies argue about this theory (Flavahan, [26]). Since platelets express COX-1 but not COX-2, it is inferred that TXA2 production by TXAS uses PGH2 derived from COX-1. In this regard, platelet TXA2 is blunted in COX-1 deficient mice, which have decreased platelet aggregation and thrombosis (Yu et al., [88]). Mice deficient in the TP (Cheng et al., [17]; Thomas et al., [72]) or human patients with a genetic disorder in the TP receptor (Fuse et al., [29]; Hirata et al., [34]) exhibit an increase in bleeding tendency and resistance to platelet aggregation, confirming the role of TXA2 in those activities and consistent with the effects of TP antagonists in humans. Conversely, transgenic mice specifically over-expressing TP in the vasculature results in placental ischemia during pregnancy and suppression of TXA2 formation rescue the phenotype (Rocca et al., [58]). Since TXA2 is a mitogen of vascular smooth muscle cells, TP KO mice have decreased vascular proliferation and platelet response after artery injury to the carotid although they are normotensive (Cheng et al., [17]).

Whether PGI2 is synthesized through COX-1 or COX-2 has been of paramount importance in classical cardiovascular research. Initial reports indicated that vascular endothelial cells and smooth muscle cells have COX-1, and that PGI2 can be formed through COX-1 which was supported by the fact that in endothelial cells, PGIS co-localizes with COX-1 but not with COX-2 (Flavahan, [26]; Warner & Mitchell, [82]). However, PGIS expression can be induced together with COX-2 in endothelial cells by haemodynamic shear stress or by oxidized low density lipoproteins (oxLDLs) leading to PGI2 production (Norata et al., [54]; Topper et al., [73]). Moreover, in healthy individuals COX-2 specific inhibitors reduce PGI2 metabolites in urine without affecting TXA2 metabolites (FitzGerald & Patrono, [25]). This suggests that COX-2 is a major source of PGI2 in the human cardiovascular system and plays a role under physiological conditions. The predominant coupling of PGI2 to COX-2 suggests that COX-2 specific inhibitor could be prothrombotic, since anti-aggregatory PGI2 circulating levels within would be reduced. This has been proposed to explain the putative associations between COX-2 inhibitors and the development of cardiovascular complications (Belton & Fitzgerald, [4]; FitzGerald, [23]). In contrast to TP deficient mice, IP knockout mice have normal blood pressure and develop normally without suffering from spontaneous thrombosis. However, after endothelial damage they show an enhanced thrombotic response compared to control littermates (Murata et al., [52]), indicating that PGI2 does play a major role only in response to stress and not in the basal systemic circulation. Mice lacking IP have enhanced vascular smooth cell proliferation and platelet activation in response to vascular injury likely caused by enhanced TXA2/PGI2 ratio (Cheng et al., [17]). Simultaneous deletion of the TP and the IP abrogated both increased responses (Cheng et al., [17]). Selective inhibition, knockout, mutation or deletion of COX-2 or IP has been shown to accelerate thrombogenesis. These responses were attenuated by COX-1 knock down (Cheng et al., [18]). IP KO mice also show vascular hyperplasia and increased vascular remodeling (Rudic et al., [61]) as well as cardiac hypertrophy and fibrosis (Francois et al., [28]). Taken together those results indicate that PGI2 regulates the cardiovascular activity of TXA2 and further support the hypothesis that cardiovascular homeostasis is resulting from the balance between these two eicosanoids.

Prostanoids and atherosclerosis

Atherosclerosis is one of the most important diseases of the developed countries and can be considered as a multifactorial inflammatory disease triggered by high levels of cholesterol in serum leading to an inflammation in the intima of large arteries and involving several cell types including immune cells as T lymphocytes and monocytes/macrophages as well as endothelial and smooth muscle cells and platelets (Ross, [59]; Hansson & Libby, [31]). Among those, monocytes/macrophages play an important key role in many phases of atherogenic process. Thus, after an atherogenic stimulus, monocyte-macrophages reversibly adhere to the endothelium and migrate across it, leading to a prolonged retention of those cells in the intima which is central in atherogenesis. A variety of substances including prostanoids have been implicated in the pathogenesis of atherosclerosis.

Prostanoids may be involved in atherosclerosis through its ability to regulate a variety of mechanisms potentially involved in the pathogenesis such as inflammation, vasodilatation, vasoconstriction, platelet aggregation and leukocyte–endothelial cell adhesion, and leukocyte migration among others (Vila, [78]; Kobayashi et al., [43]). In this regard, COX-2 expression has been observed in symptomatic atherosclerotic lesions (Cipollone et al., [14]). This enzyme may play a dual role in the pathogenesis of the atherosclerosis. Initially, COX-2 expression is induced in monocytes by pro-inflammatory cytokines and several growth factors. Then, COX-2-mediated PG production by activated macrophages may promote atherosclerosis in the artery wall through several mechanisms, as induction of other pro-inflammatory mediators or by favouring migration of macrophages and other immune cells or by induction of adhesion molecules. Later on, COX-2-derived PGI2, likely from the endothelial cells, may have a protective role in atherogenesis by favouring vasodilatation. Moreover, in atherosclerotic patients, PGI2 can be formed through the action of both COX-1 and COX-2 (Flavahan, [26]). COX-1 but not COX-2 is expressed in normal arteries, whereas both isoforms are expressed in atherosclerotic lesions. In those lesions COX-2 is expressed not only by monocyte/macrophages, but also by endothelial and proliferating smooth muscle cells (Belton & Fitzgerald, [5]). Atherogenic lipoproteins, such as oxidized low density lipoproteins (oxLDL) may promote atherosclerosis first by contributing to inflammation by activating monocytes/macrophages and later by stimulating lipid uptake by macrophages, leading to foam cell formation. Interestingly, oxLDL has apparently contradictory effect on macrophages, since they can activate the expression of some pro-inflammatory genes whereas reduce COX-2 expression (Banfi et al., [3]). In agreement with this is the fact that macrophage-derived foam cells from the atherosclerotic lesions in mice, did not express COX-2. Thus induction COX-2 expression in macrophages takes place before their transformation into foam cells in the plaques.

In spite of the clear involvement of COX-2 derived prostanoids in vascular atherosclerosis, results on the effect of COX-2 selective inhibitors on the formation and progression of atherosclerotic plaques are controversial. Results from different studies have shown increased, reduced as well as unaltered atherogenesis (Belton & Fitzgerald, [5]; Burleigh et al., [10]; FitzGerald & Patrono, [25]; Fosslien, [27]; Ray et al., [56]; Martinez-Gonzalez & Badimon, [46]). Those discrepant effects of COX-2 may reflect the dual role of COX-2 in promoting early, but protecting late atherosclerotic lesions, mentioned above. Results about the influence of COXs in atherosclerosis in animal models, apart from those using pharmacological inhibitors, have been obtained by means of cell transplantation from fetal liver or bone marrow from COX-1 or COX- 2 deficient mice into ApoE or LDLR KO mice (Babaev et al., [2]; Burleigh et al., [10]; Burleigh et al., [11]). The size of the atherosclerotic lesions was significantly reduced when cells deficient in COX-2 were transplanted compared to the mice transplanted with wild type fetal liver cells (Burleigh et al., [10]; Burleigh et al., [11]). Those results implicate COX-2 expression in the macrophage and not in other cells as endothelial cells, smooth muscle cells, or T-cells in promoting atherosclerotic lesion formation. Efforts to obtain information from mice deficient in both COX-2 and ApoE or LDLR genes have been unsuccessful due to the severe renal defects of COX-2 mice. Regarding to COX-1, both proatherosclerotic and anti-atherosclerotic roles also have been reported (Babaev et al., [2]; Pratico et al., [55]).

Various prostanoids may play a significant role in the atherogenic process (Reiss & Edelman, [57]; Vila, [78]). Patients with extensive disease as well as murine models of atherosclerosis have enhanced formation of TXA2. Even more, TP antagonists decrease atherogenesis in mice. TP deficiency in atherosclerotic mice models induced a significant delay in atherogenesis, compared with mice deficient in apoE alone (Kobayashi et al., [43]).

In contrast, PGI2 may theoretically have a beneficial effect in the atherogenic process by limiting platelet adhesion to the endothelium and activation in the plaques. In accordance with this, local delivery of PIGS gene through an adenoviral vector reduces the platelet deposition seen following vascular injury (Yamada et al., [86]). Genetic deletion of the IP receptor, both in the LDRL and in the ApoE KO mice models, aggravated atherogenesis (Egan et al., [22]; Kobayashi et al., [43]). As platelets are though to contribute to the development and progression of atherosclerosis in the late phase, PGI2 may likely suppress lesion formation by limiting platelet deposition. Those mice exhibited a significant acceleration in atherogenesis with enhanced platelet activation and increased rolling of leukocytes on the vessel walls. Those results indicate that TXA2 promotes whereas PGI2 prevents the initiation and progression of atherogenesis by modulating platelet activation and leukocyte-endothelial cell interaction.

In addition to PGI2 and TXA2, other PGs, such as PGE2 and PGD2, could also play an important role in the pathophysiology of atherosclerosis. In this regard, vasoconstrictor responses to PGE2 are greatly increased in atherosclerosis (Lopez et al., [45]) and PGE2 may also promote angiogenesis (Iniguez et al., [38]). Moreover, inducible mPGES-1 has been detected in activated macrophages in atherosclerotic lesions where it colocalizes with COX-2 both in mice and humans (Cipollone et al., [14]). Disruption of this enzyme in mice reduced foam cell formation and atherosclerosis in fat-fed LDLR−/− mice. mPGES-1 deletion augmented both PGIS and TXS expression in endothelial cells (Wang, M. et al., [80]). On the other hand, COX-2 derived PGs as PGD2 may possess anti-inflammatory and anti-atherosclerotic properties in such a way that the balance between PGDS and PGES has been shown to be a major determinant of atherosclerotic plaque instability (Cipollone et al., [13]).

Future directions

Based on the hypothesis that atherosclerosis is an inflammatory disease, it was proposed that COX inhibition by NSAIDs, and in particular selective inhibition of COX-2 by the Coxibs family of NSAIDs, might have protective and even anti-atherogenic effects. However, clinical studies have indicated that there is an increased risk of atherothrombosis in individuals taking these drugs in such a way that some of them as Rofecoxib have been recently withdrawn from the market (Bombardier et al., [8]; McGettigan & Henry, [47]; Caldwell et al., [12]; Kearney et al., [42]). These undesirable effects of COX-2 selective inhibitors have been explained by the TXA2/PGI2 balance theory by which selective inhibition of COX-2 lead to a reduction in the production of the vasodilator PGI2 whereas production of the vasoconstrictor and pro-aggregatory TXA2, mostly COX-1–dependent, remains unaffected. Thus, disruption of the physiological balance between TXA2 and PGI2 accelerates atherosclerosis and increases the risk of thrombosis and other cardiovascular complications (Figure 2).

Graph: Figure 2. The balance hypothesis for the cardiovascular effects of NSAIDs. Cardiovascular homeostasis is controlled by the balance between COX-1 – dependent production of TXA2 by platelets, and COX-2 – mediated PGI2 (prostacyclin) production by endothelial cells. Whereas TXA2 function as a potent platelet activator and vasoconstrictor, PGI2 inhibits platelet aggregation and thrombosis. Classical non-selective NSAIDs reduce the production of TXA2 by platelets through their ability to inhibit COX-1 activity thus displaying anti-thrombotic properties. In contrast, COX-2 selective NSAIDs reduce PGI2 formation by endothelial cells consequently disturbing the equilibrium between TXA2 and PGI2 and potentially favouring pro-thrombotic cardiovascular events.

Nevertheless, the mechanisms underlying the pathogenesis of cardiovascular complications upon NSAIDs administration remain to be clarified as the TXA2/PGI2 balance theory is somewhat simple and it does not consider some important clinical and experimental evidences as the increased risk of cardiovascular events by some non-selective NSAIDs, the contribution of other prostanoids to the overall effect in cardiovascular physiopathology, and the COX-2 independent effects of NSAIDs. In this sense, a more profound knowledge of the complex relations between prostanoids-mediated actions and function of cells in the cardiovascular system is required to clearly understand the benefits and risks of NSAIDs on cardiovascular diseases.

Acknowledgements

The authors thank the financial support of Comunidad Autónoma de Madrid (S-SAL-0159-2006), Ministerio de Educación y Ciencia-FEDER (SAF 2004-05109, BFU2004-04157 and BFU2007-62659), European Commission (EICOSANOX integrated project LSH-CT-2004-005033; and MAIN network of excellence) and Laboratorios del Dr. ESTEVE S.A. We apologize that many valuable studies, especially original contributions, have not been cited due to space limitation.

Declaration of interest: The authors report no conflicts of interest. The authors alone are responsible for the content and writing of the paper.

Footnotes 1 Centro de Biología Molecular Severo Ochoa, Departamento de Biología Molecular, Universidad Autónoma de Madrid, Consejo Superior de Investigaciones Científicas, Cantoblanco, 28049, Madrid, Spain, + 34 911904565, + 34 911964420 mfresno@cbm.uam.es References Alfranca A, Iniguez M A, Fresno M, Redondo J M. Prostanoid signal transduction and gene expression in the endothelium: role in cardiovascular diseases. Cardiovasc Res 2006; 70(3)446–456 2 Babaev V R, Ding L, Reese J, Morrow J D, Breyer M D, Dey S K, Fazio S, Linton M F. Cyclooxygenase-1 deficiency in bone marrow cells increases early atherosclerosis in apolipoprotein E- and low-density lipoprotein receptor-null mice. Circulation 2006; 113(1)108–117 3 Banfi C, Colli S, Eligini S, Mussoni L, Tremoli E. Oxidized LDLs influence thrombotic response and cyclooxygenase 2. Prostaglandins Leukot Essent Fatty Acids 2002; 67(2–3)169–173 4 Belton O, Fitzgerald D. Cyclooxygenase-2 inhibitors and atherosclerosis. J Am Coll Cardiol 2003; 41(10)1820–1822 5 Belton O, Fitzgerald D J. Cyclooxygenase isoforms and atherosclerosis. Expert Rev Mol Med 2003; 2003: 1–18 6 Berger J, Moller D E. The mechanisms of action of PPARs. Annu Rev Med 2002; 53: 409–435 7 Blanco J C, Contursi C, Salkowski C A, DeWitt D L, Ozato K, Vogel S N. Interferon regulatory factor (IRF)- 1 and IRF-2 regulate interferon gamma-dependent cyclooxygenase 2 expression. J Exp Med 2000; 191(12)2131–2144 8 Bombardier C, Laine L, Reicin A, Shapiro D, Burgos-Vargas R, Davis B, Day R, Ferraz M B, Hawkey C J, Hochberg M C, Kvien T K, Schnitzer T J. Comparison of upper gastrointestinal toxicity of rofecoxib and naproxen in patients with rheumatoid arthritis. VIGOR Study Group. N Engl J Med 2000; 343(21)1520–1528, (2 pages following 1528) 9 Bunting S, Moncada S, Vane J R. The prostacyclin-thromboxane A2 balance: pathophysiological and therapeutic implications. Br Med Bull 1983; 39(3)271–276 Burleigh M E, Babaev V R, Oates J A, Harris R C, Gautam S, Riendeau D, Marnett L J, Morrow J D, Fazio S, Linton M F. Cyclooxygenase-2 promotes early atherosclerotic lesion formation in LDL receptor-deficient mice. Circulation 2002; 105(15)1816–1823 Burleigh M E, Babaev V R, Yancey P G, Major A S, McCaleb J L, Oates J A, Morrow J D, Fazio S, Linton M F. Cyclooxygenase-2 promotes early atherosclerotic lesion formation in ApoE-deficient and C57BL/6 mice. J Mol Cell Cardiol 2005; 39(3)443–452 Caldwell B, Aldington S, Weatherall M, Shirtcliffe P, Beasley R. Risk of cardiovascular events and celecoxib: a systematic review and meta-analysis. J R Soc Med 2006; 99(3)132–140 Cipollone F, Fazia M, Iezzi A, Ciabattoni G, Pini B, Cuccurullo C, Ucchino S, Spigonardo F, De Luca M, Prontera C, Chiarelli F, Cuccurullo F, Mezzetti A. Balance between PGD synthase and PGE synthase is a major determinant of atherosclerotic plaque instability in humans. Arterioscler Thromb Vasc Biol 2004; 24(7)1259–1265 Cipollone F, Prontera C, Pini B, Marini M, Fazia M, De Cesare D, Iezzi A, Ucchino S, Boccoli G, Saba V, Chiarelli F, Cuccurullo F, Mezzetti A. Overexpression of functionally coupled cyclooxygenase-2 and prostaglandin E synthase in symptomatic atherosclerotic plaques as a basis of prostaglandin E(2)-dependent plaque instability. Circulation 2001; 104(8)921–927 Coruzzi G, Venturi N, Spaggiari S. Gastrointestinal safety of novel nonsteroidal antiinflammatory drugs: selective COX-2 inhibitors and beyond. Acta Biomed 2007; 78(2)96–110 Chandrasekharan N V, Dai H, Roos K L, Evanson N K, Tomsik J, Elton T S, Simmons D L. COX-3, a cyclooxygenase-1 variant inhibited by acetaminophen and other analgesic/antipyretic drugs: cloning, structure, and expression. Proc Natl Acad Sci USA 2002; 99(21)13926–13931 Cheng Y, Austin S C, Rocca B, Koller B H, Coffman T M, Grosser T, Lawson J A, FitzGerald G A. Role of prostacyclin in the cardiovascular response to thromboxane A2. Science 2002; 296(5567)539–541 Cheng Y, Wang M, Yu Y, Lawson J, Funk C D, Fitzgerald G A. Cyclooxygenases, microsomal prostaglandin E synthase-1, and cardiovascular function. J Clin Invest 2006; 116(5)1391–1399 Dannhardt G, Kiefer W. Cyclooxygenase inhibitors – current status and future prospects. Eur J Med Chem 2001; 36(2)109–126 Davidge S T. Prostaglandin H synthase and vascular function. Circ Res 2001; 89(8)650–660 Diaz-Cazorla M, Perez-Sala D, Ros J, Jimenez W, Fresno M, Lamas S. Regulation of cyclooxygenase-2 expression in human mesangial cells – transcriptional inhibition by IL-13. Eur J Biochem 1999; 260(1)268–274 Egan K M, Lawson J A, Fries S, Koller B, Rader D J, Smyth E M, Fitzgerald G A. COX-2-derived prostacyclin confers atheroprotection on female mice. Science 2004; 306(5703)1954–1957 FitzGerald G A. COX-2 and beyond: Approaches to prostaglandin inhibition in human disease. Nat Rev Drug Discov 2003; 2(11)879–890 FitzGerald G A, Austin S, Egan K, Cheng Y, Pratico D. Cyclo-oxygenase products and atherothrombosis. Ann Med 2000; 32(Suppl 1)21–26 FitzGerald G A, Patrono C. The coxibs, selective inhibitors of cyclooxygenase-2. N Engl J Med 2001; 345(6)433–442 Flavahan N A. Balancing prostanoid activity in the human vascular system. Trends Pharmacol Sci 2007; 28(3)106–110 Fosslien E. Cardiovascular complications of non-steroidal anti-inflammatory drugs. Ann Clin Lab Sci 2005; 35(4)347–385 Francois H, Athirakul K, Howell D, Dash R, Mao L, Kim H S, Rockman H A, FitzGerald G A, Koller B H, Coffman T M. Prostacyclin protects against elevated blood pressure and cardiac fibrosis. Cell Metab 2005; 2(3)201–207 Fuse I, Higuchi W, Aizawa Y. Pathogenesis of a bleeding disorder characterized by platelet unresponsiveness to thromboxane A2. Semin Thromb Hemost 2000; 26(1)43–45 Grau R, Iniguez M A, Fresno M. Inhibition of activator protein 1 activation, vascular endothelial growth factor, and cyclooxygenase-2 expression by 15-deoxy-Delta12,14-prostaglandin J2 in colon carcinoma cells: evidence for a redox-sensitive peroxisome proliferator-activated receptor-gamma-independent mechanism. Cancer Res 2004; 64(15)5162–5171 Hansson G K, Libby P. The immune response in atherosclerosis: a double-edged sword. Nat Rev Immunol 2006; 6(7)508–519 Hata A N, Breyer R M. Pharmacology and signaling of prostaglandin receptors: multiple roles in inflammation and immune modulation. Pharmacol Ther 2004; 103(2)147–166 Helliwell R J, Adams L F, Mitchell M D. Prostaglandin synthases: recent developments and a novel hypothesis. Prostaglandins Leukot Essent Fatty Acids 2004; 70(2)101–113 Hirata T, Kakizuka A, Ushikubi F, Fuse I, Okuma M, Narumiya S. Arg60 to Leu mutation of the human thromboxane A2 receptor in a dominantly inherited bleeding disorder. J Clin Invest 1994; 94(4)1662–1667 Ide T, Egan K, Bell-Parikh L C, FitzGerald G A. Activation of nuclear receptors by prostaglandins. Thromb Res 2003; 110(5–6)311–315 Iniguez M A, Martinez-Martinez S, Punzon C, Redondo J M, Fresno M. An essential role of the nuclear factor of activated T cells in the regulation of the expression of the cyclooxygenase-2 gene in human T lymphocytes. J Biol Chem 2000; 275(31)23627–23635 Iniguez M A, Punzon C, Fresno M. Induction of cyclooxygenase-2 on activated T lymphocytes: regulation of T cell activation by cyclooxygenase-2 inhibitors. J Immunol 1999; 163(1)111–119 Iniguez M A, Rodriguez A, Volpert O V, Fresno M, Redondo J M. Cyclooxygenase-2: a therapeutic target in angiogenesis. Trends Mol Med 2003; 9(2)73–78 Kabashima K, Narumiya S. The DP receptor, allergic inflammation and asthma. Prostaglandins Leukot Essent Fatty Acids 2003; 69(2–3)187–194 Kanaoka Y, Urade Y. Hematopoietic prostaglandin D synthase. Prostaglandins Leukot Essent Fatty Acids 2003; 69(2–3)163–167 Karim S, Habib A, Levy-Toledano S, Maclouf J. Cyclooxygenase-1 and -2 of endothelial cells utilize exogenous or endogenous arachidonic acid for transcellular production of thromboxane. J Biol Chem 1996; 271(20)12042–12048 Kearney P M, Baigent C, Godwin J, Halls H, Emberson J R, Patrono C. Do selective cyclooxygenase-2 inhibitors and traditional non-steroidal anti-inflammatory drugs increase the risk of atherothrombosis? Meta-analysis of randomised trials. BMJ 2006; 332(7553)1302–1308 Kobayashi T, Tahara Y, Matsumoto M, Iguchi M, Sano H, Murayama T, Arai H, Oida H, Yurugi-Kobayashi T, Yamashita J K, Katagiri H, Majima M, Yokode M, Kita T, Narumiya S. Roles of thromboxane A(2) and prostacyclin in the development of atherosclerosis in apoE-deficient mice. J Clin Invest 2004; 114(6)784–794 Kujubu D A, Herschman H R. Dexamethasone inhibits mitogen induction of the TIS10 prostaglandin synthase/cyclooxygenase gene. J Biol Chem 1992; 267(12)7991–7994 Lopez J A, Armstrong M L, Harrison D G, Piegors D J, Heistad D D. Vascular responses to leukocyte products in atherosclerotic primates. Circ Res 1989; 65(4)1078–1086 Martinez-Gonzalez J, Badimon L. Mechanisms underlying the cardiovascular effects of COX-inhibition: benefits and risks. Curr Pharm Des 2007; 13(22)2215–2227 McGettigan P, Henry D. Cardiovascular risk and inhibition of cyclooxygenase: a systematic review of the observational studies of selective and nonselective inhibitors of cyclooxygenase 2. Jama 2006; 296(13)1633–1644 Morita I. Distinct functions of COX-1 and COX-2. Prostaglandins Other Lipid Mediat 2002; 68–69: 165–175 Murakami M, Kambe T, Shimbara S, Kudo I. Functional coupling between various phospholipase A2s and cyclooxygenases in immediate and delayed prostanoid biosynthetic pathways. J Biol Chem 1999; 274(5)3103–3115 Murakami M, Nakashima K, Kamei D, Masuda S, Ishikawa Y, Ishii T, Ohmiya Y, Watanabe K, Kudo I. Cellular prostaglandin E2 production by membrane-bound prostaglandin E synthase-2 via both cyclooxygenases-1 and -2. J Biol Chem 2003; 278(39)37937–37947 Murakami M, Nakatani Y, Tanioka T, Kudo I. Prostaglandin E synthase. Prostaglandins Other Lipid Mediat 2002; 68/69: 383–399 Murata T, Ushikubi F, Matsuoka T, Hirata M, Yamasaki A, Sugimoto Y, Ichikawa A, Aze Y, Tanaka T, Yoshida N, Ueno A, Oh-ishi S, Narumiya S. Altered pain perception and inflammatory response in mice lacking prostacyclin receptor. Nature 1997; 388(6643)678–682 Niiro H, Otsuka T, Izuhara K, Yamaoka K, Ohshima K, Tanabe T, Hara S, Nemoto Y, Tanaka Y, Nakashima H, Niho Y. Regulation by interleukin-10 and interleukin-4 of cyclooxygenase-2 expression in human neutrophils. Blood 1997; 89(5)1621–1628 Norata G D, Callegari E, Inoue H, Catapano A L. HDL3 induces cyclooxygenase-2 expression and prostacyclin release in human endothelial cells via a p38 MAPK/CRE-dependent pathway: effects on COX-2/PGI-synthase coupling. Arterioscler Thromb Vasc Biol 2004; 24(5)871–877 Pratico D, Tillmann C, Zhang Z B, Li H, FitzGerald G A. Acceleration of atherogenesis by COX-1-dependent prostanoid formation in low density lipoprotein receptor knockout mice. Proc Natl Acad Sci USA 2001; 98(6)3358–3363 Ray W A, Stein C M, Daugherty J R, Hall K, Arbogast P G, Griffin M R. COX-2 selective non-steroidal anti-inflammatory drugs and risk of serious coronary heart disease. Lancet 2002; 360(9339)1071–1073 Reiss A B, Edelman S D. Recent insights into the role of prostanoids in atherosclerotic vascular disease. Curr Vasc Pharmacol 2006; 4(4)395–408 Rocca B, Loeb A L, Strauss J F, 3rd, Vezza R, Habib A, Li H, FitzGerald G A. Directed vascular expression of the thromboxane A2 receptor results in intrauterine growth retardation. Nat Med 2000; 6(2)219–221 Ross R. Atherosclerosis – an inflammatory disease. N Engl J Med 1999; 340(2)115–126 Rostom A, Muir K, Dube C, Jolicoeur E, Boucher M, Joyce J, Tugwell P, Wells G W. Gastrointestinal safety of cyclooxygenase-2 inhibitors: a Cochrane Collaboration systematic review. Clin Gastroenterol Hepatol 2007; 5(7)818–828, 828 e1–5; quiz 768 Rudic R D, Brinster D, Cheng Y, Fries S, Song W L, Austin S, Coffman T M, FitzGerald G A. COX-2-derived prostacyclin modulates vascular remodeling. Circ Res 2005; 96(12)1240–1247 Samuelsson B, Morgenstern R, Jakobsson P J. Membrane prostaglandin E synthase-1: a novel therapeutic target. Pharmacol Rev 2007; 59(3)207–224 Scher J U, Pillinger M H. 15d-PGJ2: the anti-inflammatory prostaglandin. Clin Immunol 2005; 114(2)100–109 Simmons D L, Botting R M, Hla T. Cyclooxygenase isozymes: the biology of prostaglandin synthesis and inhibition. Pharmacol Rev 2004; 56(3)387–437 Smith W L, DeWitt D L, Garavito R M. Cyclooxygenases: structural, cellular, and molecular biology. Annu Rev Biochem 2000; 69: 145–182 Smyth E M, FitzGerald G A. Human prostacyclin receptor. Vitam Horm 2002; 65: 149–165 Straus D S, Glass C K. Cyclopentenone prostaglandins: new insights on biological activities and cellular targets. Med Res Rev 2001; 21(3)185–210 Subbaramaiah K, Dannenberg A J. Cyclooxygenase 2: a molecular target for cancer prevention and treatment. Trends Pharmacol Sci 2003; 24(2)96–102 Tanabe T, Tohnai N. Cyclooxygenase isozymes and their gene structures and expression. Prostaglandins Other Lipid Mediat 2002; 68/69: 95–114 Tanioka T, Nakatani Y, Semmyo N, Murakami M, Kudo I. Molecular identification of cytosolic prostaglandin E2 synthase that is functionally coupled with cyclooxygenase-1 in immediate prostaglandin E2 biosynthesis. J Biol Chem 2000; 275(42)32775–32782 Tegeder I, Pfeilschifter J, Geisslinger G. Cyclooxygenase-independent actions of cyclooxygenase inhibitors. Faseb J 2001; 15(12)2057–2072 Thomas D W, Mannon R B, Mannon P J, Latour A, Oliver J A, Hoffman M, Smithies O, Koller B H, Coffman T M. Coagulation defects and altered hemodynamic responses in mice lacking receptors for thromboxane A2. J Clin Invest 1998; 102(11)1994–2001 Topper J N, Cai J, Falb D, Gimbrone M A, Jr. Identification of vascular endothelial genes differentially responsive to fluid mechanical stimuli: cyclooxygenase-2, manganese superoxide dismutase, and endothelial cell nitric oxide synthase are selectively up-regulated by steady laminar shear stress. Proc Natl Acad Sci USA 1996; 93(19)10417–10422 Tsuboi K, Sugimoto Y, Ichikawa A. Prostanoid receptor subtypes. Prostaglandins. Other Lipid Mediat 2002; 68/69: 535–556 Ueno N, Takegoshi Y, Kamei D, Kudo I, Murakami M. Coupling between cyclooxygenases and terminal prostanoid synthases. Biochem Biophys Res Commun 2005; 338(1)70–76 Urade Y, Hayaishi O. Biochemical, structural, genetic, physiological, and pathophysiological features of lipocalin-type prostaglandin D synthase. Biochim Biophys Acta 2000; 1482(1–2)259–271 Vane J R. Biomedicine. Back to an aspirin a day? Science 2002; 296(5567)474–475 Vila L. Cyclooxygenase and 5-lipoxygenase pathways in the vessel wall: role in atherosclerosis. Med Res Rev 2004; 24(4)399–424 Wang L H, Kulmacz R J. Thromboxane synthase: structure and function of protein and gene. Prostaglandins Other Lipid Mediat 2002; 68/69: 409–422 Wang M, Zukas A M, Hui Y, Ricciotti E, Pure E, FitzGerald G A. Deletion of microsomal prostaglandin E synthase-1 augments prostacyclin and retards atherogenesis. Proc Natl Acad Sci USA 2006; 103(39)14507–14512 Warner T D, Giuliano F, Vojnovic I, Bukasa A, Mitchell J A, Vane J R. Nonsteroid drug selectivities for cyclo-oxygenase-1 rather than cyclo-oxygenase-2 are associated with human gastrointestinal toxicity: a full in vitro analysis. Proc Natl Acad Sci USA 1999; 96(13)7563–7568 Warner T D, Mitchell J A. Cyclooxygenases: new forms, new inhibitors, and lessons from the clinic. Faseb J 2004; 18(7)790–804 Watanabe K. Prostaglandin F synthase. Prostaglandins Other Lipid Mediat 2002; 68/69: 401–407 Williams C S, Mann M, DuBois R N. The role of cyclooxygenases in inflammation, cancer, and development. Oncogene 1999; 18(55)7908–7916 Wu K K, Liou J Y. Cellular and molecular biology of prostacyclin synthase. Biochem Biophys Res Commun. 2005 Yamada M, Numaguchi Y, Okumura K, Harada M, Naruse K, Matsui H, Ito T, Hayakawa T. Prostacyclin synthase gene transfer modulates cyclooxygenase-2-derived prostanoid synthesis and inhibits neointimal formation in rat balloon-injured arteries. Arterioscler Thromb Vasc Biol 2002; 22(2)256–262 Yokoyama C, Yabuki T, Shimonishi M, Wada M, Hatae T, Ohkawara S, Takeda J, Kinoshita T, Okabe M, Tanabe T. Prostacyclin-deficient mice develop ischemic renal disorders, including nephrosclerosis and renal infarction. Circulation 2002; 106(18)2397–2403 Yu Y, Cheng Y, Fan J, Chen X S, Klein-Szanto A, Fitzgerald G A, Funk C D. Differential impact of prostaglandin H synthase 1 knockdown on platelets and parturition. J Clin Invest 2005; 115(4)986–995

By Miguel A. Iñiguez; Cristina Cacheiro-Llaguno; Natalia Cuesta; Manuel D. Díaz-Muñoz and Manuel Fresno

Reported by Author; Author; Author; Author; Author

Titel:
Prostanoid function and cardiovascular disease
Autor/in / Beteiligte Person: INIGUEZ, Miguel A ; CACHEIRO-LLAGUNO, Cristina ; CUESTA, Natalia ; DIAZ-MUNOZ, Manuel D ; FRESNO, Manuel
Link:
Zeitschrift: Archives of physiology and biochemistry, Jg. 114 (2008), Heft 3, S. 201-209
Veröffentlichung: Basingstoke: Taylor & Francis, 2008
Medientyp: academicJournal
Umfang: print, 2 p.1/4
ISSN: 1381-3455 (print)
Schlagwort:
  • Biochemistry, molecular biology, biophysics
  • Biochimie, biologie moléculaire, biophysique
  • Physiology, morphology
  • Physiologie, morphologie
  • Sciences biologiques et medicales
  • Biological and medical sciences
  • Sciences biologiques fondamentales et appliquees. Psychologie
  • Fundamental and applied biological sciences. Psychology
  • Vertebres: anatomie et physiologie, organisme dans son ensemble ou etude de plusieurs organes ou systemes
  • Vertebrates: anatomy and physiology, studies on body, several organs or systems
  • Dérivé de l'acide arachidonique
  • Arachidonic acid derivatives
  • Araquidónico ácido derivado
  • Eicosanoïde
  • Eicosanoid
  • Eicosanoide
  • Enzyme
  • Enzima
  • Oxidoreductases
  • Vertebrata
  • Mammalia
  • Pathologie de l'appareil circulatoire
  • Cardiovascular disease
  • Aparato circulatorio patología
  • Prostaglandin-endoperoxide synthase
  • Prostaglandine
  • Prostaglandin
  • Prostaglandina
  • Prostanoïde
  • Prostanoid
  • Prostanoide
  • Récepteur biologique
  • Biological receptor
  • Receptor biológico
  • Thromboxane
  • Tromboxano
  • NSAIDs
  • Prostaglandins
  • cardiovascular physiopathology
  • cyclooxygenases
  • prostanoid receptors
  • thromboxanes
Sonstiges:
  • Nachgewiesen in: PASCAL Archive
  • Sprachen: English
  • Original Material: INIST-CNRS
  • Document Type: Article
  • File Description: text
  • Language: English
  • Author Affiliations: Centro de Biologia Molecular Severo Ochoa, Departamento de Biologia Molecular, Universidad Autónoma de Madrid, Consejo Superior de Investigaciones Cientificas, Cantoblanco 28049, Madrid, Spain
  • Rights: Copyright 2008 INIST-CNRS ; CC BY 4.0 ; Sauf mention contraire ci-dessus, le contenu de cette notice bibliographique peut être utilisé dans le cadre d’une licence CC BY 4.0 Inist-CNRS / Unless otherwise stated above, the content of this bibliographic record may be used under a CC BY 4.0 licence by Inist-CNRS / A menos que se haya señalado antes, el contenido de este registro bibliográfico puede ser utilizado al amparo de una licencia CC BY 4.0 Inist-CNRS
  • Notes: Vertebrates : anatomy and physiology, studies on body, several organs or systems

Klicken Sie ein Format an und speichern Sie dann die Daten oder geben Sie eine Empfänger-Adresse ein und lassen Sie sich per Email zusenden.

oder
oder

Wählen Sie das für Sie passende Zitationsformat und kopieren Sie es dann in die Zwischenablage, lassen es sich per Mail zusenden oder speichern es als PDF-Datei.

oder
oder

Bitte prüfen Sie, ob die Zitation formal korrekt ist, bevor Sie sie in einer Arbeit verwenden. Benutzen Sie gegebenenfalls den "Exportieren"-Dialog, wenn Sie ein Literaturverwaltungsprogramm verwenden und die Zitat-Angaben selbst formatieren wollen.

xs 0 - 576
sm 576 - 768
md 768 - 992
lg 992 - 1200
xl 1200 - 1366
xxl 1366 -