Associate Editor: K. E. Suckling
Cyclooxygenase and prostaglandin synthases in atherosclerosis: Recent insights and future perspectives

https://doi.org/10.1016/j.pharmthera.2008.01.002Get rights and content

Abstract

Cyclooxygenase (COX) is the key enzyme in the conversion of arachidonic acid to prostanoids, lipid mediators involved in several physiological and pathological processes. Two COX isoenzymes have been characterized, COX-1 and COX-2, that differ in terms of regulatory mechanisms of expression, tissue distribution, substrate specificity, and preferential coupling to upstream and downstream enzymes. Both isoforms play fundamental roles in atherothrombosis; however, whereas the function of COX-1 in this setting is well established, the role of COX-2 remains unclear. Indeed, the intracellular pathways regulating COX-2 induction appear numerous and complicated, varying between cell types and cellular stimulus. In recent years a long series of studies has been performed with the aim of clarifying the role of COX-2 in atherothrombosis, with the major finding that the COX-2 expression pattern in arterial vessels may be associated with either protective or plaque-destabilyzing phenotypes according to the downstream synthase that couples with COX-2. In this review we summarize the role of COX-2 as well as the different downstream synthases in atherosclerosis and atherothrombosis. Finally, we briefly review the controversial vascular effects on prostanoid inhibition by COX-2 inhibitors.

Introduction

Atherosclerosis is a complex disease and the precise molecular mechanisms contributing to its pathogenesis are not completely defined (Libby et al., 2002). While atherosclerosis was traditionally considered a mere lipid storage disorder, solid evidence in the last decades has led to the concept of atherosclerosis as a chronic inflammatory disease. In fact, inflammation characterizes all the phases of atherosclerosis progression, from the initial formation of the early lesions to the rupture of advanced plaques, both at systemic and local level. Indeed, because the systemic status of a patient has been associated with the incidence of plaque rupture, there has been much interest in potential circulating markers able to identify subjects at increased risk of plaque rupture. In this context, it has been shown that upregulation of inflammatory mediators such as C reactive protein (CRP), adhesion molecules [soluble intercellular adhesion molecule (sICAM)-1, soluble vascular cell adhesion molecule (sVCAM)-1, P-selectin], interleukin (IL)-6, IL-18, tumor necrosis factor (TNF)-α, and soluble CD40L predict future cardiovascular risk in a variety of clinical settings. From a local perspective, atherosclerotic plaques prone to rupture have histological characteristics that are different from stable plaques. Indeed, it has been widely shown that an inflammatory infiltrate, mainly composed of activated macrophages and T-lymphocytes, is present in all atherosclerotic plaques, but it is markedly more abundant in unstable than in stable plaques, and this strongly supports the crucial role of inflammation in plaque instability (Shah, 2003).

Several mechanisms linking inflammation to plaque instability have been proposed so far. In this context, it is recognized that important inflammatory enzymes and lipid mediators playing a crucial role in the pathophysiology of atherosclerosis progression derive from the cascade of arachidonic acid, a polyunsaturated fatty acid present in the phospholipids of cell membranes. This acid can be metabolized by different pathways originating a variety of biologically important compounds collectively known as prostanoids.

In recent years, we investigated some potential molecular mechanisms of atherosclerotic plaque instability (reviewed in Cipollone et al., 2005a, Cipollone et al., 2005b), all related to the expression of the inducible isoform of cyclooxygenase (COX), COX-2, the key enzyme in the conversion of arachidonic acid to prostanoids. The role of this protein in atherothrombosis appears to be quite complex, as it is an intermediate enzyme in the cascade of arachidonic acid and may generate not only proinflammatory but also anti-inflammatory compounds, according to the downstream synthase that is co-expressed with it.

In this article we review the role of cyclooxygenase-2 and downstream synthases in the vascular system and in atherothrombosis, summarizing our newest findings linking COX-2 to different plaque phenotypes.

The first steps of atherogenesis occur as a result of injury to endothelium by several factors, including elevated and modified low density lipoproteins (LDL), excessive blood pressure, free radicals, the products of glycoxidation associated with hyperglycemia, infectious agents, shear stress, or proinflammatory cytokines derived from excess adipose tissue. Following these stimuli, the endothelium undergoes rapid phenotypic changes, leading to activation of endothelial cells and impairment of their physiological functions, i.e. maintenance of blood circulation and fluidity, regulation of vascular tone, and inflammatory responses (Libby et al., 2002). Thus, homeostatic imbalance, impairment in vasorelaxation, oxidative stress and increased adhesiveness of the endothelium lining for circulating inflammatory cells become predominant in the injured vascular wall. The enhanced adhesion of blood leukocytes to the inner surface of the arterial wall is a key event in atherogenesis and is strongly promoted by the upregulation of the endothelial expression of cellular adhesion molecules, including ICAM-1, VCAM-1 and selectins. Once adherent to the endothelium, the blood leukocytes—mainly mononuclear phagocytes and T-lymphocytes—transmigrate into the tunica intima, a process promoted by chemoattractant mediators including monocyte chemotactic protein-1 (MCP-1). In the arterial wall, the blood-derived inflammatory cells promote and amplify a local inflammatory response that leads to generation of cytokines and foam cells, the lipid laden macrophages that characterize early atherosclerotic lesions. As this inflammatory process continues, the activated leukocytes and vascular cells release a variety of mediators, including growth factors that stimulate replication of smooth muscle cells (SMCs). These cells also are important players in atherosclerosis progression because of their consistent proliferation, migration from the tunica media into the intima, and synthesis of components of extracellular matrix, all of which contribute to plaque progression toward advanced lesions (Aikawa & Libby, 2004). Advanced plaques are mainly composed of connective tissue extracellular matrix (including collagen, proteoglycans, fibronectin elastic fibers), lipids (crystalline cholesterol, cholesteryl esters, phospholipids), calcium deposits, as well as inflammatory cells and SMCs (Davies, 1997). However, varying proportions of these components occur in different plaques, thus leading to a spectrum of lesion compositions. The extracellular lipids accumulated in the intima form the classic “lipid core”, a necrotic region containing oxidized lipids and prothrombotic factors, making the lipid highly thrombogenic when exposed to circulating blood. Under ill-defined circumstances, some plaques in some patients undergo physical disruption, and the subsequent formation of a superimposed platelet thrombus lead to the onset of the clinical manifestations of acute ischemic syndromes (Fuster et al., 2005).

The analysis of angiograms reveal that in about 75% of patients with acute ischemic syndromes, the culprit lesion site produced a < 70% diameter narrowing (Hackett et al., 1988). Thus plaques producing non-flow limiting stenoses account for more cases of plaque rupture and thrombosis than plaques producing a more severe stenosis. These observations support the hypothesis that plaque composition rather than plaque size is the real determinant of the plaque evolution toward rupture. Indeed, histological comparison of stable versus unstable plaques indicates that the latter have histomorphologic features quite different from intact plaques. Plaques prone to rupture are characterized by a large lipid core, often occupying > 40% plaque volume, as well as a thin fibrous cap, a structure that protects the deeper components of the plaque from contact with circulating blood. The biomechanical strength of the plaque's fibrous cap has considerable importance as a determinant of lesion stability, and thinning of the fibrous cap is generally considered to be a sign of vulnerability and a prelude to rupture. In this context, it is worth noting that fibrous caps from ruptured plaques typically contain less extracellular matrix (collagen and proteoglycans), a greater area of inflammatory cell infiltration and fewer SMCs than caps from intact plaques. Depletion of matrix components from the fibrous cap leading to cap thinning is caused, at least in part, by an imbalance between matrix synthesis and breakdown, leading to enhanced degradation and reduced synthesis of extracellular matrix. While depletion of matrix may be due to a reduction of the number or function of SMCs, enhanced matrix breakdown has been attributed to a family of matrix-degrading enzymes known as metalloproteinases (MMPs, in particular MMP-1, MMP-2, MMP-3, MMP-9) which are generated in atherosclerotic plaques mainly by inflammatory cells (macrophages, foam cells) and to a lesser extent by SMCs and endothelial cells (Jones et al., 2003). This family of enzymes plays a major role in both physiological and pathological vascular remodelling, entailing degradation and reorganization of the extracellular matrix (ECM) scaffold of the vessel wall. The activity of MMPs is tightly regulated at the level of gene transcription and is also regulated by their secretion in an inactive zymogen form that requires extracellular activation. Latent MMPs can be activated by plasmin, trypsin, and chymase (derived from degranulating mast cells), oxidized lipids, reactive oxygen species, chlamydial heat shock protein, CD40 ligation, inflammatory cytokines, and hemodynamic stress, all existing in atherosclerotic plaques. Finally, MMP generation and activity are also modulated by co-secretion of the tissue inhibitors of metalloproteinases (TIMPs). Thus, increased gene transcription, enhanced activation, and reduced activity of TIMPs can individually or together create a milieu for increased matrix proteolysis.

In the last few years, we focused our interest on two metalloproteinases, 92 kDa (MMP-9) and 72 kDa (MMP-2) gelatinases, specialized in the digestion of collagen fragments, that have been critically associated with acute ischemic events in humans (Cipollone et al., 2005a, Cipollone et al., 2005b). Firstly we characterized their expression pattern in carotid plaques from patients enlisted to undergo carotid endarterectomy for high-grade internal carotid artery stenosis. Patients were subdivided in symptomatic and asymptomatic according to NASCET classification (1991). The first group (symptomatic patients) included subjects who presented with clinical symptoms of atherothrombotic ischemic stroke or transient ischemic attack (TIA). Ischemic strokes were all well-defined atherothrombotic strokes clearly related to ulceration of a culprit plaque of the internal carotid artery and endarterectomy was performed within 120 days after the onset of symptoms in these patients. The second group (asymptomatic patients) included patients who had an asymptomatic carotid stenosis. We found that both of MMPs were expressed in the two groups of patients, but their expression and activity were markedly higher in plaques from symptomatic patients as compared to specimens from asymptomatic individuals, suggesting that MMPs play a crucial role in plaque instability. Thus the subsequent step of our research was to identify potential molecular mechanisms leading to MMP overexpression in this setting.

The starting point of our investigations was the observation that secretion of MMP-2 as well as MMP-9 by macrophages in human atherosclerotic plaques occurs through a mechanism dependent on prostaglandin (PG)E2, one of the compounds generated by the cyclooxygenase-mediated pathway (Corcoran et al., 1994). Thus, in the last years, we identified some of the mechanisms contributing to MMP overexpression in unstable plaques, all related to PGE2 biosynthetic pathway along the arachidonic acid cascade.

Section snippets

The cyclooxygenase-mediated pathway

Arachidonic acid metabolism plays an important role in the pathophysiology of acute ischemic syndromes, as compounds generated by this cascade, known as eicosanoids, are critical regulators of several pathophysiological responses, such as inflammation, blood clotting, wound healing, blood vessel tone, and immune responses. The first step in the formation of eicosanoids is the liberation of arachidonic acid from membrane-bound phospholipids, usually by the action of phospholipases, primarily

Prostaglandin E synthase

Prostaglandin E synthase catalyzes the conversion of the COX-product PGH2 to PGE2. This enzyme was characterized in 1999 by Jakobsson et al. (1999), who demonstrated for the first time that recombinant human microsomal glutathione-S-transferase (GST)-1-like 1 (MGST1-L1), a member of the MAPEG superfamily [for membrane-associated proteins involved in eicosanoid and glutathione (GSH) metabolism] was responsible for PGE2 biosynthesis. In the subsequent years, several isoforms of PGES have been

Prostacyclin synthase-mediated pathway in the vascular system

PGI2 is generated by the sequential action of COX and PGI2 synthase (PGIS), a member of the cytochrome P450 superfamily that specifically converts PGH2 to PGI2. PGIS was previously reported to be localized to plasma and nuclear membranes. In the last few years it was shown that PGIS, like its related cytochrome P450 proteins, is localized to endoplasmic reticulum but is also found at the perinuclear region including nuclear envelope (Helliwell et al., 2004). PGIS is constitutively expressed in

Prostaglandin F synthase

Prostaglandin F2 is synthesized via three pathways from PGE2, PGD2, or PGH2 by PGE2 9-ketoreductase, PGD2 11-ketoreductase, or PGH2 9,11-endoperoxide reductase, respectively (Watanabe, 2002). PGE2 9-ketoreductase has been demonstrated to be the key enzyme in the regulation of specific PGs in the endometrium during the perimplantation period. PGD2 11-ketoreductase exists as two isoforms: lung-type PGFS (or PGFS I) and liver-type PGFS (or PGFS II). PGFS-1 is expressed in lung and peripheral blood

Aspirin

Antithrombotic therapy is currently the cornerstone of the treatment of acute coronary syndromes. Numerous studies indicate the importance of inflammation in atherothrombosis and support therapeutic use of anti-inflammatory treatment. The use of aspirin has increased since it was shown to reduce the risk of myocardial infarction and stroke: because platelet aggregation is known to play a crucial role in thrombosis the administration of aspirin translates to clinical benefit in CAD, as

Coxibs and the message from the clinical trials

Selective inhibitors of COX-2 were developed on the basis of the assumption that COX-1 mediates the biosynthesis of physiological prostanoids that regulate vascular tone and mucosal integrity, whereas COX-2 is the inducible isoform responsible for the PG involved in inflammation. As predicted from biological experiments and animal models, specific blockade of the COX-2 system has a therapeutic anti-inflammatory effect associated with a lower risk of gastrointestinal (GI) complications,

Conclusions and future perspectives

The presence of different downstream synthases, the multiple and opposite biological effects of prostanoids, as well as the existence of different isoforms of prostanoid receptors make it difficult to define the role of COX-2 expression in atherosclerosis progression and in plaque rupture. The complexity surrounding the effects of COX-2 should be taken into consideration in the assessment of the potential benefits and risks of COX-2 inhibition. From a pharmacological standpoint, development of

References (144)

  • DegraeveF. et al.

    Modulation of COX-2 expression by statins in human aortic smooth muscle cells: Involvement of geranylgeranylated proteins

    J Biol Chem

    (2001)
  • FahmiH. et al.

    15d-PGJ(2) is acting as a ‘dual agent’ on the regulation of COX-2 expression in human osteoarthritic chondrocytes

    Osteoarthr Cartil

    (2002)
  • FaourW.H. et al.

    Prostaglandin E2 regulates the level and stability of cyclooxygenase-2 mRNA through activation of p38 mitogen-activated protein kinase in interleukin-1 beta-treated human synovial fibroblasts

    J Biol Chem

    (2001)
  • FarkouhM.E. et al.

    Comparison of lumiracoxib with naproxen and ibuprofen in the Therapeutic Arthritis Research and Gastrointestinal Event Trial (TARGET), cardiovascular outcomes, randomised controlled trial

    Lancet

    (2004)
  • FlavahanN.A.

    Balancing prostanoid activity in the human vascular system

    Trends Pharmacol Sci

    (2007)
  • FusterV. et al.

    Atherothrombosis and high-risk plaque: Part I: Evolving concepts

    J Am Coll Cardiol

    (2005)
  • HataA.N. et al.

    Pharmacology and signaling of prostaglandin receptors: Multiple roles in inflammation and immune modulation

    Pharmacol Ther

    (2004)
  • HelliwellR.J. et al.

    Prostaglandin synthases: Recent developments and a novel hypothesis

    Prostaglandins Leukot Essent Fat Acids

    (2004)
  • Hernandez-PresaM.A. et al.

    Atorvastatin reduces the expression of cyclooxygenase-2 in a rabbit model of atherosclerosis and in cultured vascular smooth muscle cells

    Atherosclerosis

    (2002)
  • HolmesD.R. et al.

    Prostaglandin E2 synthesis, and cyclooxygenase expression in abdominal aortic aneurysms

    J Vasc Surg

    (1997)
  • HirataY. et al.

    Occurrence of 9-deoxy-delta 9,delta 12-13,14-dihydroprostaglandin D2 in human urine

    J Biol Chem

    (1988)
  • KameiD. et al.

    Reduced pain hypersensitivity and inflammation in mice lacking microsomal prostaglandin E synthase-1

    J Biol Chem

    (2004)
  • KanaokaY. et al.

    Hematopoietic prostaglandin D synthase

    Prostaglandins Leukot Essent Fat Acids

    (2003)
  • LangenbachR. et al.

    Cyclooxygenase knockout mice: Models for elucidating isoform-specific functions

    Biochem Pharmacol

    (1999)
  • LehrkeM. et al.

    The many faces of PPARgamma

    Cell

    (2005)
  • LiouJ.Y. et al.

    Colocalization of prostacyclin synthase with prostaglandin H synthase-1 (PGHS-1) but not phorbol ester-induced PGHS-2 in cultured endothelial cells

    J Biol Chem

    (2000)
  • MathurS.N. et al.

    Decreased prostaglandin production by cholesterol-rich macrophages

    J Lipid Res

    (1989)
  • MatsumotoH. et al.

    Concordant induction of prostaglandin E2 synthase with cyclooxygenase-2 leads to preferred production of prostaglandin E2 over thromboxane and prostaglandin D2 in lipopolysaccharide-stimulated rat peritoneal macrophages

    Biochem Biophys Res Commun

    (1997)
  • MiwaY. et al.

    Identification of gene polymorphism in lipocalin-type prostaglandin D synthase and its association with carotid atherosclerosis in Japanese hypertensive patients

    Biochem Biophys Res Commun

    (2004)
  • MonneretG. et al.

    Prostaglandin D2 is a potent chemoattractant for human eosinophils that acts via a novel DP receptor

    Blood

    (2001)
  • ArberN. et al.

    Celecoxib for the prevention of colorectal adenomatous polyps

    N Engl J Med

    (2006)
  • ArdansJ.A. et al.

    Oxidized low-density and high-density lipoproteins regulate the production of matrix metalloproteinase-1 and-9 by activated monocytes

    J Leukoc Biol

    (2002)
  • AudolyL.P. et al.

    Cardiovascular responses to the isoprostanes iPF(2alpha)-III and iPE(2)-III are mediated via the thromboxane A(2) receptor in vivo

    Circulation

    (2000)
  • BeltonO. et al.

    Cyclooxygenase-1 and-2-dependent prostacyclin formation in patients with atherosclerosis

    Circulation

    (2000)
  • BertagnolliM.M. et al.

    Celecoxib for the prevention of sporadic colorectal adenomas

    N Engl J Med

    (2006)
  • BombardierC. et al.

    VIGOR Study Group. Comparison of upper gastrointestinal toxicity of rofecoxib and naproxen in patients with rheumatoid arthritis. VIGOR Study Group

    N Engl J Med

    (2000)
  • BrashA.R.

    Arachidonic acid as a bioactive molecule

    J Clin Invest

    (2001)
  • BresalierR.S. et al.

    Cardiovascular events associated with rofecoxib in a colorectal adenoma chemoprevention trial

    N Engl J Med

    (2005)
  • BrunR.P. et al.

    Differential activation of adipogenesis by multiple PPAR isoforms

    Genes Dev

    (1996)
  • BuerkleM.A. et al.

    Selective inhibition of cyclooxygenase-2 enhances platelet adhesion in hamster arterioles in vivo

    Circulation

    (2004)
  • BurleighM.E. et al.

    Cyclooxygenase-2 promotes early atherosclerotic lesion formation in LDL receptor-deficient mice

    Circulation

    (2002)
  • CaponeM.L. et al.

    Clinical pharmacology of platelet, monocyte, and vascular cyclooxygenase inhibition by naproxen and low-dose aspirin in healthy subjects

    Circulation

    (2004)
  • Catella-LawsonF. et al.

    Effects of specific inhibition of cyclooxygenase-2 on sodium balance, hemodynamics, and vasoactive eicosanoids

    J Pharmacol Exp Ther

    (1999)
  • Catella-LawsonF. et al.

    Systemic biosynthesis of prostacyclin by cyclooxygenase (COX)-2: The human pharmacology of a selective inhibitor of COX-2

    Proc Natl Acad Sci U S A

    (1999)
  • CayatteA.J. et al.

    The thromboxane receptor antagonist S18886 but not aspirin inhibits atherogenesis in apo E-deficient mice: Evidence that eicosanoids other than thromboxane contribute to atherosclerosis

    Arterioscler Thromb Vasc Biol

    (2000)
  • CensarekP. et al.

    Cyclooxygenase COX-2a, a novel COX-2 mRNA variant, in platelets from patients after coronary artery bypass grafting

    Thromb Haemost

    (2004)
  • ChandrasekharanN.V. et al.

    COX-3, a cyclooxygenase-1 variant inhibited by acetaminophen and other analgesic/antipyretic drugs: Cloning, structure, and expression

    Proc Natl Acad Sci U S A

    (2002)
  • ChawlaA. et al.

    PPAR-gamma dependent and independent effects on macrophage-gene expression in lipid metabolism and inflammation

    Nat Med

    (2001)
  • ChengY. et al.

    Role of prostacyclin in the cardiovascular response to thromboxane A2

    Science

    (2002)
  • ChinettiG. et al.

    CLA-1/SR-BI is expressed in atherosclerotic lesion macrophages and regulated by activators of peroxisome proliferator-activated receptors

    Circulation

    (2000)
  • Cited by (94)

    • Discovery of novel 1,3-diaryl pyrazolyl ester derivatives as COX-2 inhibitory candidates with anti-tumor effect

      2023, Journal of Molecular Structure
      Citation Excerpt :

      During the investigations on COX, at least two isoenzymes have been paid close attention to. One is the structural protein COX-1 with the functions including vasoconstriction in blood vessels and renal regulation in kidney [4,5]; while the other is the inducible enzyme COX-2 on the nuclear membrane with the high-level expression in various disorders [6,7]. Despite the known correlation with inflammation, COX-2 has been found overexpressed in several tumor types including breast cancer and pancreatic cancer, while COX-1 commonly keeps unchanged in these tumor cases [8–11].

    • Molecular feature of arterial remodeling in the brain arteriovenous malformation revealed by arteriovenous shunt rat model and RNA sequencing

      2022, International Immunopharmacology
      Citation Excerpt :

      Last, we predict 9 small molecular drugs targeting the key genes. Nitrendipine, prostaglandin, fostamatinib, isotretinoin have shown their efficacy in vascular diseases [36–39]. Among these, nitrendipine could decrease the accumulation of macrophages and synthetic VSMCs in hypercholesterolemic rabbits [36].

    View all citing articles on Scopus
    View full text