5
Review Mechanism of antiplatelet action of hypolipidemic, antidiabetic and antihypertensive drugs by PPAR activation PPAR agonists: New antiplatelet agents Eduardo Fuentes , Iván Palomo Department of Clinical Biochemistry and Immunohaematology, Faculty of Health Sciences, Interdisciplinary Excellence Research Program on Healthy Aging (PIEI-ES), Universidad de Talca, Talca, Chile Centro de Estudios en Alimentos Procesados (CEAP), CONICYT-Regional, Gore Maule, R09I2001, Chile abstract article info Article history: Received 20 February 2014 Received in revised form 8 May 2014 Accepted 15 May 2014 Available online 27 May 2014 Keywords: Antiplatelet Hypolipidemic Antidiabetic Antihypertensive peroxisome proliferator-activated receptors Given the prevalence of cardiovascular disease in patients with cardiovascular risk factors (i.e., hypertension, dia- betes, smoking and obesity) and that platelet activation plays an important pathogenic role in cardiovascular dis- eases, it is very important to identify the drugs that have multiple targets. In this sense, the present article describes the mechanism of antiplatelet action of hypolipidemic (statins and brates), antidiabetic (thiazolidinediones) and antihypertensive (nifedipine) drugs via peroxisome proliferator-activated receptor (PPAR) activation. The mecha- nism of antiplatelet action of the drugs is by direct activation of PPARs with the inhibition of cyclooxygenase-1, protein kinase C-alpha, calcium mobilization, thromboxane A2, sCD40L, platelet microparticles and cAMP- phosphodiesterase, and the stimulation of proteins kinase G and A. Thus, these observations highlight PPARs as a novel therapeutic target for the treatment and prevention of cardiovascular diseases. © 2014 Elsevier Inc. All rights reserved. Contents 1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 162 2. PPARs and atherothrombosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 163 3. Mechanism of antiplatelet action by PPAR activation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 163 4. Drugs and platelet inhibition: role of PPAR activation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 163 4.1. Statins and brates . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 163 4.2. Thiazolidinediones . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 164 4.3. Nifedipine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 164 5. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 165 Declaration of interest . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 165 Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 165 References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 165 1. Introduction Cardiovascular diseases (CVD) (i.e., acute myocardial infarction, cerebrovascular disease and peripheral arterial thrombosis) have signicantly increased in recent years [1,2]. The most frequent and independent risk factors for CVD are cigarette smoking, elevated blood pressure, elevated serum total cholesterol and diabetes, among others [3,4]. In most cases, CVD is the consequence of an atherosclerotic plaque rupture and thrombus formation. Accelerated atherosclerosis and an in- creased risk of thrombotic vascular events in diabetes may result from dyslipidemia, endothelial dysfunction and platelet hyperreactivity [5, 6]. Platelets from patients with type 1 and type 2 diabetes exhibit en- hanced platelet aggregation activity early in the disease course that may precede CVD development [7,8]. Thus, platelets adhere; secrete their granule contents, aggregate and initiate thrombus formation fol- lowing an atheromatous plaque rupture, [9,10]. Vascular Pharmacology 62 (2014) 162166 Corresponding authors at: Immunology and Haematology Laboratory, Faculty of Health Sciences, Universidad de Talca, Casilla, 747, Talca, Chile. Tel.: +56 71 200493; fax: +56 71 20048. E-mail addresses: [email protected] (E. Fuentes), [email protected] (I. Palomo). http://dx.doi.org/10.1016/j.vph.2014.05.008 1537-1891/© 2014 Elsevier Inc. All rights reserved. Contents lists available at ScienceDirect Vascular Pharmacology journal homepage: www.elsevier.com/locate/vph

jurnal

Embed Size (px)

DESCRIPTION

jurnal tersebut berisi tentang penelitian yang bertema kesehatan, penelitian tersebut tidak dilakukan di indonesia, namun di luar negeri berdasarkan evidance base yang ada

Citation preview

Vascular Pharmacology 62 (2014) 162–166

Contents lists available at ScienceDirect

Vascular Pharmacology

j ourna l homepage: www.e lsev ie r .com/ locate /vph

Review

Mechanism of antiplatelet action of hypolipidemic, antidiabeticand antihypertensive drugs by PPAR activation

PPAR agonists: New antiplatelet agents

Eduardo Fuentes ⁎, Iván Palomo ⁎Department of Clinical Biochemistry and Immunohaematology, Faculty ofHealth Sciences, Interdisciplinary Excellence Research ProgramonHealthy Aging (PIEI-ES), Universidad de Talca, Talca, ChileCentro de Estudios en Alimentos Procesados (CEAP), CONICYT-Regional, Gore Maule, R09I2001, Chile

⁎ Corresponding authors at: Immunology and HaemHealth Sciences, Universidad de Talca, Casilla, 747, Talcfax: +56 71 20048.

E-mail addresses: [email protected] (E. Fuentes), ipa

http://dx.doi.org/10.1016/j.vph.2014.05.0081537-1891/© 2014 Elsevier Inc. All rights reserved.

a b s t r a c t

a r t i c l e i n f o

Article history:Received 20 February 2014Received in revised form 8 May 2014Accepted 15 May 2014Available online 27 May 2014

Keywords:AntiplateletHypolipidemicAntidiabeticAntihypertensive peroxisomeproliferator-activated receptors

Given the prevalence of cardiovascular disease in patients with cardiovascular risk factors (i.e., hypertension, dia-betes, smoking and obesity) and that platelet activation plays an important pathogenic role in cardiovascular dis-eases, it is very important to identify the drugs that havemultiple targets. In this sense, the present article describesthe mechanism of antiplatelet action of hypolipidemic (statins and fibrates), antidiabetic (thiazolidinediones) andantihypertensive (nifedipine) drugs via peroxisome proliferator-activated receptor (PPAR) activation. Themecha-nism of antiplatelet action of the drugs is by direct activation of PPARs with the inhibition of cyclooxygenase-1,protein kinase C-alpha, calcium mobilization, thromboxane A2, sCD40L, platelet microparticles and cAMP-phosphodiesterase, and the stimulation of proteins kinase G and A. Thus, these observations highlight PPARs as anovel therapeutic target for the treatment and prevention of cardiovascular diseases.

© 2014 Elsevier Inc. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1622. PPARs and atherothrombosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1633. Mechanism of antiplatelet action by PPAR activation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1634. Drugs and platelet inhibition: role of PPAR activation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 163

4.1. Statins and fibrates . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1634.2. Thiazolidinediones . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1644.3. Nifedipine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 164

5. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 165Declaration of interest . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 165Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 165References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 165

1. Introduction

Cardiovascular diseases (CVD) (i.e., acute myocardial infarction,cerebrovascular disease and peripheral arterial thrombosis) havesignificantly increased in recent years [1,2]. The most frequent andindependent risk factors for CVD are cigarette smoking, elevated

atology Laboratory, Faculty ofa, Chile. Tel.: +56 71 200493;

[email protected] (I. Palomo).

blood pressure, elevated serum total cholesterol and diabetes,among others [3,4].

In most cases, CVD is the consequence of an atherosclerotic plaquerupture and thrombus formation. Accelerated atherosclerosis and an in-creased risk of thrombotic vascular events in diabetes may result fromdyslipidemia, endothelial dysfunction and platelet hyperreactivity [5,6]. Platelets from patients with type 1 and type 2 diabetes exhibit en-hanced platelet aggregation activity early in the disease course thatmay precede CVD development [7,8]. Thus, platelets adhere; secretetheir granule contents, aggregate and initiate thrombus formation fol-lowing an atheromatous plaque rupture, [9,10].

163E. Fuentes, I. Palomo / Vascular Pharmacology 62 (2014) 162–166

Given the central role of platelets in the atherosclerotic-related in-flammatory response and the subsequent thrombotic event, a varietyof antiplatelet agents have been developed [11,12]. Although antiplate-let drugs have been proven to be beneficial in patients with clinical ev-idence of CVD, outcomes still remain poor [13]. In this way, plateletsfrom diabetic patients release greater amounts of thromboxane A2(TXA2) than platelets from non-diabetic patients receiving treatmentwith low-dose aspirin [14]. This is because all the current available an-tiplatelet agents only target one signal pathway and moderately inhibitplatelet activation (e.g. aspirin and clopidogrel) [15]. Therefore, there ismuch room for further improvement of antiplatelet treatment and theneed to identify the mechanisms by which hypolipidemic (statins andfibrates), antidiabetic (thiazolidinediones) and antihypertensive (nifed-ipine) drugs inhibit platelet activation. It has also been observed thatthese drugs are ligands for peroxisome proliferator-activated receptors(PPARs) in cells which have a nucleus [16–18].

PPARs are involved inmany biological processes, including lipid andenergymetabolism inflammation responses, and atherosclerotic plaqueformation [19–21]. Selective agonists through their action on nuclearreceptors (e.g. PPARs) regulate platelet function despite the absence ofa nucleus in platelets. [22]. In this sense, the present article describesthe antiplatelet action mechanism of hypolipidemic, antidiabetic andantihypertensive drugs via PPAR activation.

2. PPARs and atherothrombosis

PPARs consist of a family of three nuclear receptor isoforms (γ, β/δ,andα) [23]. PPARs are the key regulators of metabolism and inflamma-tion, and play an important role in chronic inflammatory disease pro-cesses [24,25]. Accumulating evidence suggests that PPAR activation isa key mechanism to reduce atherosclerosis and improve cardiovascularfunction [26–28].

Recent studies suggest that PPAR-γ activation decreases atherogen-esis, not only by correcting metabolic disorders, but also through its di-rect effects on the vascular wall [29]. In addition, PPAR-α and PPAR-β/δhave also been suggested in the regulation of inflammation and slowatherosclerosis progression [20,30].

Ligands of PPAR-α are important regulators of lipid and lipoproteinmetabolism, thereby positively affecting plasma lipid risk factors relatedto atherosclerosis [31]. In this context, the lipid-lowering effect offenofibrate is achieved by activating PPAR-α andAMP-activated proteinkinase (AMPK) signaling pathway that results in increasing lipolysis andfatty acid β-oxidation [32].

PPARs appear to play a major role in the regulation of atherogenesisby countering the inflammation-provoking action of platelet adhesionand activation [33].

3. Mechanism of antiplatelet action by PPAR activation

Nuclear receptors are transcription factors that are activated by li-gands and subsequently bind to regulatory regions in target genes;allowing the organism to integrate signals coming from the environ-ment [34]. While steroid/nuclear receptors are recognized via for theirrole in gene regulation, increasing evidence supports nongenomic ac-tions of these receptors [35,36]. Thus, although platelets lack a nucleus,they express a number of transcription factors including steroid/nuclearreceptors such as PPARs [37,38], glucocorticoid receptor (GR) [39],retinoic X receptor (RXR) [41], estrogen receptor (ER) [40] and nuclearfactor kappa B (NF-κB) [42]. Thus, selective ligands for these receptorsregulate platelet aggregation and activation [22,43].

Whilemany reports focus on PPARexpression in the nucleus [44,45],in this article we focus on the role of PPAR-cytoplasm in platelet func-tion. Recent studies have provided the first evidence that human bonemarrow megakaryocytes and human platelets express PPAR-γ [37].PPAR-γ activation decreases platelet aggregation and delays intra-arterial thrombus formation in rats, at least partially, by an increase in

the expression of nitric oxide synthase (NOS) and thrombomodulin[46]. PPAR-γ activation also inhibits platelet function induced by colla-gen through the modulation of early glycoprotein (GP) VI signaling, atthe level of Syk and LAT [47]. In addition, platelet incubation with a nat-ural PPAR-γ agonist (15d-PGJ(2)) or with a potent synthetic PPAR-γ li-gand (rosiglitazone) not only attenuates platelet activation, but alsodecreases the release of platelet proinflammatory and procoagulantme-diators (sCD40L and TXA2) [33,37].

Agonists of PPAR-β, including GW0742 and L-165041, inhibit plate-let activation after 5min of incubation. Clearlywith such acute exposureand as platelets have no nucleus, PPAR-β is an active antithromboticpathway in platelets, whose effects are independent of the nucleus [38].

Moreover, PPAR-α activators may exert vasculo-protective actionthrough suppression of PDGF-BB production in amegakaryocyte/plateletpathway [48]. Prostacyclin formed by the consecutive actions ofcyclooxygenase-1 (COX-1) and prostacyclin synthase is arguably themost important endogenous antiplatelet hormone identified to date[49]. Prostacyclin inhibits platelet function via stimulation of a surfaceprostanoid receptor linked to the activation of adenylyl cyclase and in-creased intracellular levels of cyclic adenosine monophosphate (cAMP)[50]. Moreover, prostacyclin activates PPAR-β/δ in human platelets inthe low 10−9 M range. Therefore, an additional mechanism of antiplate-let action by prostacyclin occurs by activation of PPAR-β/δ and its effectson platelet aggregation is synergistic with nitric oxide (NO) [38,51].

4. Drugs and platelet inhibition: role of PPAR activation

Given the prevalence of CVD in patients with cardiovascular riskfactors (i.e., hypertension, diabetes, smoking and obesity) and thatplatelet activation plays an important pathogenic role in athero-thrombosis [3,52], it is very important to identify the drugs thathave multiple targets. In this sense, the present article describesthe mechanism of antiplatelet action of hypolipidemic (statins andfibrates), antidiabetic (thiazolidinediones) and antihypertensive(nifedipine) drugs by PPAR activation.

4.1. Statins and fibrates

The 3-hydroxy-3-methylglutaryl-coenzyme (HMG-CoA) reductaseinhibitors, also known as statins, are the most effective class of drugsfor lowering serum low-density lipoprotein cholesterol (LDL-C) concen-trations [53]. In this way, another class of drug called fibrates reducestriglycerides and increases high-density lipoprotein cholesterol (HDL-C) [54]. In this article, we discuss how statins and fibrates appear tohave more biological effects than those originally targeted. Statins andfibrates can exert beneficial anti-inflammatory and antithrombotic ef-fects in after patients with a high risk of coronary artery disease asearly as 3 days after therapy [55]. Thus, statins (simvastatin, atorvastat-in, or cerivastatin) decrease morbidity and mortality in patients withCVD by their effects on proinflammatory cytokines: interleukin 6 (IL-6), IL-8, and monocyte chemoattractant protein-1 (MCP-1) [56]. Mean-while, fibrates by PPAR-activation may inhibit atherosclerosis develop-ment in vivo [57].

Interestingly both statins and fibrates inhibit platelet function [54].Inhibition of platelet aggregation by simvastatin involves the activationof the cAMP-eNOS/NO-cGMP pathway, resulting in the inhibition of thePLC-γ2/PKC/p38 MAPK/TXA2 cascade [58,59]. Therefore, the direct in-hibitory effects of statins and fibrates on platelet activation are mediat-ed by PPAR activation and this new finding reveals some of thepleiotropic effects of these drugs [60]. In platelets, PPAR signaling path-way activation involves binding and repression of PKC, and increasing ofcAMP and cGMP levels [60]. The increase of intraplatelet levels of cAMPis due to the fact that the repression of PKC allows greater activity ofadenylyl cyclase, which converts ATP to cAMP [61,62]. In addition,cAMP induced inhibition of platelet P-selectin expression, platelet

Fig. 1. Mechanism of antiplatelet action of PPAR ligands. AA = arachidonic acid; AKT = known as protein kinase B; AMP = adenosine monophosphate; cAMP = cyclic adenosinemonophosphate; cGMP= cyclic guanosinemonophosphate; COX-1=cyclooxygenase-1;NO= nitric oxide;NOS= nitric oxide synthase; PDE3=phosphodiesterase-3; PGH2= prostaglan-dinH2; PGG2= prostaglandinG2; PKA= protein kinaseA; PKC= protein kinase C; PKG= protein kinaseG; PLA2= phospholipaseA2; PMP= plateletmicroparticles; PPARs= peroxisomeproliferator-activated receptors; TXA2 = thromboxane A2; VASP = vasodilator-stimulated phosphoprotein; VASP-P = vasodilator-stimulated phosphoprotein phosphorylation.

164 E. Fuentes, I. Palomo / Vascular Pharmacology 62 (2014) 162–166

aggregation and release of sCD40L, for the large part, is mediatedthrough the activation of protein kinase A (PKA) [63–66].

Platelet microparticles (PMP) have an average diameter of 0.1 μm[67], and are the most abundant type of microparticles (MP) in humanblood (representing 70–90%) and contribute to many biologically pro-cesses [68]. PMP levels are greatly elevated in patients (range from~3000–11,000/μL) with diseases including venous thromboembolism[69], hypertension [70], diabetes [71], cancer [72], immunologic plateletdisorders and arterial thrombosis [73,74]. In humans, plasma levels ofcirculating PMP are known to increase with the presence of cardiovas-cular risk factors and progression of atherosclerosis [75]. Even after ini-tiation of antithrombotic treatment, PMP are still elevated after sixmonths [76]. Possibly because a low dose of aspirinmight not be strongenough to suppress the shedding of PMP in the microcirculation [77].With respect to the role of statins, MP from hypercholesterolemia pa-tients with statin treatment had reduced markers of activated platelets(GPIIb-IIIa), activated inflammatory cells (Mac-1) and tissue factor [78].Meanwhile atorvastatin reduces thrombin generation and expression oftissue factor, GPIIIa and P-selectin on PMP in patients with peripheralvascular disease [79]. In this way statins may inhibit thrombotic eventgeneration partly through a PMP dependent mechanism.

4.2. Thiazolidinediones

The thiazolidinediones (TZDs) are a class of oral antidiabetic drugsthat exert effects through a mechanism that involves an activation ofPPAR-γ [80]. TZDs (rosiglitazone, pioglitazone and troglitazone) havebeen shown to protect LDL-C from oxidative modification in patientswith type 2 diabetes that were treated for 8 weeks, one of the key initialevents in atherogenesis [81]. In addition, TZD activation of PPAR-γ re-duced the progression of subclinical atherosclerosis by normalizing

proatherogenic metabolic abnormalities of the insulin resistance, andthrough an inhibition of vascular cell growth and movement [82].

Recent studies suggest that TZDs may also be clinically beneficial toinflammatory mediators [83]. Matrix metalloproteinase-9 (MMP-9) iscritically involved in the development of unstable plaques [84]. AlsoMMP-9 levels are increased in type 2 diabetes patients with coronaryartery disease, and the treatment of these patients with the antidiabeticPPARγ-activator rosiglitazone significantly reduces the serum levels ofMMP-9, tumor necrosis factor alpha (TNF-α) and serumamyloid A [85].

Troglitazonehas a potent inhibitory effect on platelet aggregation viathe suppression of the thrombin-induced activation of phosphoinositidesignaling in human platelets [86]. Based on the function of other cells(astrocyte cells), the TZDmode of action is likely related to the elevationof the intracellular cAMP level by PPAR-γ activation [87]. This could ex-plain why the treatment with TZDs reduces sCD40L serum levels in pa-tients with type 1 and 2 diabetes and coronary artery disease [88,89].

PPAR-γ containing PMP are quickly released fromα-granule into ex-tracellular parts when platelets are activated by collagen [90]. Then,PMP-PPAR-γ can be internalized by the monocytic cell line (THP-1). Inthis way, THP-1 cell activation can be attenuated in the presence of aPPAR-γ agonist (rosiglitazone) [91,92].

In addition, one possible mechanism explaining the anti-inflammatory activity of TZDs is its ability to activate GR [93]. Inthis way, classic anti-inflammatory glucocorticoids are due to thefact that human platelets contain GR [39].

4.3. Nifedipine

Nifedipine, a L-type calcium channel blocker, is widely used in thetreatment of hypertension and coronary heart disease [94,95]. Nifedi-pine increases the activity and intracellular expression of PPAR-β/γfromactivated platelets. Therefore, themechanism of antiplatelet action

165E. Fuentes, I. Palomo / Vascular Pharmacology 62 (2014) 162–166

of nifedipine is PAR-β/γ-dependent with upregulation of the PI3K/Akt/NO/cGMP/PKG pathway, and the inhibition of PKC activity and intracel-lular Ca2+ mobilization in platelets [96]. Taking into consideration allthese findings in vivo evidence of such in vitro-related nifedipine-antithrombotic effects is provided. Indeed, using a murine model ofthrombus formation, the administration of nifedipine substantiallyinhibitedfluorescein sodium-induced vessel thrombus formation. In ad-dition, its antithrombotic effect is considerably reduced in the presenceof PPAR-β/γ antagonists [96].

Finally, themechanismof antiplatelet action of statins, fibrates, TZDsand nifedipine by direct activation of PPARs is mediated by inhibition ofCOX-1 [37], PKC [62] and Ca2+mobilization [47], and indirect inhibitionof TXA2, sCD40L, PMP, P-selectin and cAMP-phosphodiesterase, andstimulation of PKG (increased levels of cGMP) and PKA (increased levelsof cAMP) [63,97] (Fig. 1).

5. Conclusions

The data presented in this article has demonstrated that the ability ofstatins, fibrates, TZDs and nifedipine to inhibit platelet activation is like-ly attributable to their influence on PPAR activation. Thus, these obser-vations highlight PPARs as a novel therapeutic target for the treatmentand prevention of cardiovascular risk factors and CVD.

Declaration of interest

The authors report no declarations of interest.

Acknowledgments

This work was funded by CONICYT REGIONAL/GORE MAULE/CEAP/R09I2001, Programa de Investigación de Excelencia Interdisciplinariaen Envejecimiento Saludable (PIEI-ES), and supported by grant no.1130216 (I.P., M.G., R.M., M.A., J.C.) from Fondecyt, Chile.

References

[1] Palomo GI, Icaza NG, Mujica EV, Nunez FL, Leiva ME, Vasquez RM. et al (Prevalenceof cardiovascular risk factors in adult from Talca, Chile). Rev Med Chil2007;135:904–12.

[2] Bautista LE, Orostegui M, Vera LM, Prada GE, Orozco LC, Herran OF. Prevalence andimpact of cardiovascular risk factors in Bucaramanga, Colombia: results from theCountrywide Integrated Noncommunicable Disease Intervention Programme(CINDI/CARMEN) baseline survey. Eur J Cardiovasc Prev Rehabil 2006;13:769–75.

[3] Grundy SM, Pasternak R, Greenland P, Smith Jr S, Fuster V. Assessment of cardiovas-cular risk by use of multiple-risk-factor assessment equations: a statement forhealthcare professionals from the American Heart Association and the AmericanCollege of Cardiology. Circulation 1999;100:1481–92.

[4] Mujica V, Urzua A, Leiva E, Diaz N, Moore-Carrasco R, Vasquez M, et al. Interventionwith education and exercise reverses the metabolic syndrome in adults. J Am SocHypertens 2010;4:148–53.

[5] Colwell JA, Nesto RW. The platelet in diabetes: focus on prevention of ischemicevents. Diabetes Care 2003;26:2181–8.

[6] Vinik AI, Erbas T, Park TS, Nolan R, Pittenger GL. Platelet dysfunction in type 2 diabe-tes. Diabetes Care 2001;24:1476–85.

[7] Sagel J, Colwell JA, Crook L, Laimins M. Increased platelet aggregation in earlydiabetus mellitus. Ann Intern Med 1975;82:733–8.

[8] Palomo I, Moore-Carrasco R, Alarcon M, Rojas A, Espana F, Andres V, et al. Patho-physiology of the proatherothrombotic state in the metabolic syndrome. FrontBiosci (Schol Ed) 2010;2:194–208.

[9] Fuentes QE, Fuentes QF, Andres V, Pello OM, de Mora JF, Palomo GI. Role of plateletsas mediators that link inflammation and thrombosis in atherosclerosis. Platelets2013;24:255–62.

[10] Ruggeri ZM. Mechanisms initiating platelet thrombus formation. Thromb Haemost1997;78:611–6.

[11] Collins B, Hollidge C. Antithrombotic drugmarket. Nat RevDrugDiscov 2003;2:11–2.[12] Palomo I, Toro C, Alarcon M. The role of platelets in the pathophysiology of athero-

sclerosis (Review). Mol Med Rep 2008;1:179–84.[13] Meadows TA, Bhatt DL. Clinical aspects of platelet inhibitors and thrombus forma-

tion. Circ Res 2007;100:1261–75.[14] DiMinno G, Silver MJ, Cerbone AM, Murphy S. Trial of repeated low-dose aspirin in

diabetic angiopathy. Blood 1986;68:886–91.[15] Reaume KT, Regal RE, DorschMP. Indications for dual antiplatelet therapywith aspi-

rin and clopidogrel: evidence-based recommendations for use. Ann Pharmacother2008;42:550–7.

[16] Qu S, Su D, Altomonte J, Kamagate A, He J, Perdomo G, et al. PPAR{alpha} mediatesthe hypolipidemic action of fibrates by antagonizing FoxO1. Am J Physiol EndocrinolMetab 2007;292:E421–34.

[17] Yano M, Matsumura T, Senokuchi T, Ishii N, Murata Y, Taketa K, et al. Statins activateperoxisome proliferator-activated receptor gamma through extracellular signal-reg-ulated kinase 1/2 and p38 mitogen-activated protein kinase-dependent cyclooxy-genase-2 expression in macrophages. Circ Res 2007;100:1442–51.

[18] Diaz-Delfin J, Morales M, Caelles C. Hypoglycemic action of thiazolidinediones/peroxisome proliferator-activated receptor gamma by inhibition of the c-JunNH2-terminal kinase pathway. Diabetes 2007;56:1865–71.

[19] Berger J, Moller DE. The mechanisms of action of PPARs. Annu Rev Med 2002;53:409–35.

[20] Lee CH, Chawla A, Urbiztondo N, Liao D, BoisvertWA, Evans RM, et al. Transcription-al repression of atherogenic inflammation: modulation by PPARdelta. Science2003;302:453–7.

[21] Fuentes E, Fuentes F, Vilahur G, Badimon L, Palomo I. Mechanisms of chronic state ofinflammation as mediators that link obese adipose tissue and metabolic syndrome.Mediators Inflamm 2013;2013:136584.

[22] Bishop-Bailey D. The platelet as a model system for the acute actions of nuclear re-ceptors. Steroids 2010;75:570–5.

[23] Moraes LA, Piqueras L, Bishop-Bailey D. Peroxisome proliferator-activated receptorsand inflammation. Pharmacol Ther 2006;110:371–85.

[24] Ahmed W, Ziouzenkova O, Brown J, Devchand P, Francis S, Kadakia M, et al. PPARsand their metabolic modulation: new mechanisms for transcriptional regulation?J Intern Med 2007;262:184–98.

[25] Fuentes E, Guzman-Jofre L, Moore-Carrasco R, Palomo I. Role of PPARs in inflammatoryprocesses associatedwithmetabolic syndrome (Review).MolMedRep2013;8:1611–6.

[26] Kersten S, Desvergne B, Wahli W. Roles of PPARs in health and disease. Nature2000;405:421–4.

[27] Inoue H, Jiang XF, Katayama T, Osada S, Umesono K, Namura S. Brain protection byresveratrol and fenofibrate against stroke requires peroxisome proliferator-activat-ed receptor alpha in mice. Neurosci Lett 2003;352:203–6.

[28] Verreth W, De Keyzer D, Pelat M, Verhamme P, Ganame J, Bielicki JK, et al. Weight-loss-associated induction of peroxisome proliferator-activated receptor-alpha andperoxisome proliferator-activated receptor-gamma correlate with reduced athero-sclerosis and improved cardiovascular function in obese insulin-resistant mice. Cir-culation 2004;110:3259–69.

[29] Winkler K, Konrad T, Fullert S, Friedrich I, Destani R, Baumstark MW, et al. Pioglita-zone reduces atherogenic dense LDL particles in nondiabetic patients with arterialhypertension: a double-blind, placebo-controlled study. Diabetes Care 2003;26:2588–94.

[30] Delerive P, Fruchart JC, Staels B. Peroxisome proliferator-activated receptors in in-flammation control. J Endocrinol 2001;169:453–9.

[31] Torra IP, Chinetti G, Duval C, Fruchart JC, Staels B. Peroxisome proliferator-activatedreceptors: from transcriptional control to clinical practice. Curr Opin Lipidol2001;12:245–54.

[32] Chen WL, Chen YL, Chiang YM, Wang SG, Lee HM. Fenofibrate lowers lipid accumu-lation in myotubes by modulating the PPARalpha/AMPK/FoxO1/ATGL pathway.Biochem Pharmacol 2012;84:522–31.

[33] Ray DM, Spinelli SL, O'Brien JJ, Blumberg N, Phipps RP. Platelets as a novel target forPPARgamma ligands: implications for inflammation, diabetes, and cardiovasculardisease. BioDrugs 2006;20:231–41.

[34] Chinetti G, Fruchart JC, Staels B. Peroxisome proliferator-activated receptors(PPARs): nuclear receptors at the crossroads between lipid metabolism and inflam-mation. Inflamm Res 2000;49:497–505.

[35] Croxtall JD, van Hal PT, Choudhury Q, Gilroy DW, Flower RJ. Different glucocorticoidsvary in their genomic and non-genomic mechanism of action in A549 cells. Br JPharmacol 2002;135:511–9.

[36] Losel RM, Falkenstein E, Feuring M, Schultz A, Tillmann HC, Rossol-Haseroth K, et al.Nongenomic steroid action: controversies, questions, and answers. Physiol Rev2003;83:965–1016.

[37] Akbiyik F, Ray DM, Gettings KF, Blumberg N, Francis CW, Phipps RP. Human bonemarrow megakaryocytes and platelets express PPARgamma, and PPARgamma ago-nists blunt platelet release of CD40 ligand and thromboxanes. Blood 2004;104:1361–8.

[38] Ali FY, Davidson SJ, Moraes LA, Traves SL, Paul-ClarkM, Bishop-Bailey D, et al. Role ofnuclear receptor signaling in platelets: antithrombotic effects of PPARbeta. FASEB J2006;20:326–8.

[39] Moraes LA, Paul-Clark MJ, Rickman A, Flower RJ, Goulding NJ, Perretti M. Ligand-specific glucocorticoid receptor activation in human platelets. Blood 2005;106:4167–75.

[40] Moraes LA, Swales KE, Wray JA, Damazo A, Gibbins JM, Warner TD, et al.Nongenomic signaling of the retinoid X receptor through binding and inhibitingGq in human platelets. Blood 2007;109:3741–4.

[41] Moro L, Reineri S, Piranda D, Pietrapiana D, Lova P, Bertoni A, et al. Nongenomiceffects of 17beta-estradiol in human platelets: potentiation of thrombin-inducedaggregation through estrogen receptor beta and Src kinase. Blood 2005;105:115–21.

[42] Malaver E, Romaniuk MA, D'Atri LP, Pozner RG, Negrotto S, Benzadon R, et al. NF-kappaB inhibitors impair platelet activation responses. J Thromb Haemost2009;7:1333–43.

[43] Fuentes E, Rojas A, Palomo I. Role of multiligand/RAGE axis in platelet activation.Thromb Res 2014;133:308–14.

[44] Padilla J, Leung E, Phipps RP. Human B lymphocytes and B lymphomas ex-press PPAR-gamma and are killed by PPAR-gamma agonists. Clin Immunol2002;103:22–33.

166 E. Fuentes, I. Palomo / Vascular Pharmacology 62 (2014) 162–166

[45] Chawla A. Control of macrophage activation and function by PPARs. Circ Res2010;106:1559–69.

[46] Li D, Chen K, Sinha N, Zhang X, Wang Y, Sinha AK, et al. The effects of PPAR-gammaligand pioglitazone on platelet aggregation and arterial thrombus formation.Cardiovasc Res 2005;65:907–12.

[47] Moraes LA, Spyridon M, Kaiser WJ, Jones CI, Sage T, Atherton RE, et al. Non-genomiceffects of PPARgamma ligands: inhibition of GPVI-stimulated platelet activation. JThromb Haemost 2010;8:577–87.

[48] Hashizume S, Akaike M, Azuma H, Ishikawa K, Yoshida S, Sumitomo-Ueda Y, et al.Activation of peroxisome proliferator-activated receptor alpha in megakaryocytesreduces platelet-derived growth factor-BB in platelets. J Atheroscler Thromb2011;18:138–47.

[49] Higgs EA, Higgs GA, Moncada S, Vane JR. Prostacyclin (PGI2) inhibits the formationof platelet thrombi in arterioles and venules of the hamster cheek pouch. Br JPharmacol 1978;63:535–9.

[50] Beck F, Geiger J, Gambaryan S, Veit J, Vaudel M, Nollau P, et al. Time-resolved char-acterization of cAMP/PKA-dependent signaling reveals that platelet inhibition is aconcerted process involving multiple signaling pathways. Blood.123:e1-e10.

[51] Fauti T, Muller-Brusselbach S, KreutzerM, RieckM, MeissnerW, Rapp U, et al. Induc-tion of PPARbeta and prostacyclin (PGI2) synthesis by Raf signaling: failure of PGI2to activate PPARbeta. FEBS J 2006;273:170–9.

[52] Davi G, Patrono C. Platelet activation and atherothrombosis. N Engl J Med2007;357:2482–94.

[53] Istvan ES, Deisenhofer J. Structural mechanism for statin inhibition of HMG-CoA re-ductase. Science 2001;292:1160–4.

[54] Staels B, Dallongeville J, Auwerx J, Schoonjans K, Leitersdorf E, Fruchart JC. Mecha-nism of action of fibrates on lipid and lipoprotein metabolism. Circulation1998;98:2088–93.

[55] Undas A, Celinska-Lowenhoff M, Domagala TB, Iwaniec T, Dropinski J, Lowenhoff T,et al. Early antithrombotic and anti-inflammatory effects of simvastatin versusfenofibrate in patients with hypercholesterolemia. Thromb Haemost 2005;94:193–9.

[56] Rezaie-Majd A, Maca T, Bucek RA, Valent P, Muller MR, Husslein P, et al. Simvastatinreduces expression of cytokines interleukin-6, interleukin-8, and monocytechemoattractant protein-1 in circulating monocytes from hypercholesterolemic pa-tients. Arterioscler Thromb Vasc Biol 2002;22:1194–9.

[57] Duez H, Chao YS, Hernandez M, Torpier G, Poulain P, Mundt S, et al. Reduction ofatherosclerosis by the peroxisome proliferator-activated receptor alpha agonistfenofibrate in mice. J Biol Chem 2002;277:48051–7.

[58] Lee YM, ChenWF, Chou DS, Jayakumar T, Hou SY, Lee JJ, et al. Cyclic nucleotides andmitogen-activated protein kinases: regulation of simvastatin in platelet activation. JBiomed Sci 2010;17:45.

[59] Chou TC, Lin YF, Wu WC, Chu KM. Enhanced nitric oxide and cyclic GMP formationplays a role in the anti-platelet activity of simvastatin. Br J Pharmacol 2008;153:1281–7.

[60] Ali FY, Armstrong PC, Dhanji AR, Tucker AT, Paul-Clark MJ, Mitchell JA, et al. Anti-platelet actions of statins and fibrates are mediated by PPARs. Arterioscler ThrombVasc Biol 2009;29:706–11.

[61] Levesque L, Crooke ST. Depletion of protein kinase C-alpha by antisense oligonucle-otides alters beta-adrenergic function and reverses the phorbol ester-induced reduc-tion of isoproterenol-induced adenosine 3′–5′-cyclic monophosphate accumulationin murine Swiss 3T3 fibroblasts. J Pharmacol Exp Ther 1998;287:425–34.

[62] Ali FY, Hall MG, Desvergne B, Warner TD, Mitchell JA. PPARbeta/delta agonistsmodulate platelet function via a mechanism involving PPAR receptors and spe-cific association/repression of PKCalpha—brief report. Arterioscler Thromb VascBiol 2009;29:1871–3.

[63] Libersan D, Rousseau G, Merhi Y. Differential regulation of P-selectin expression byprotein kinase A and protein kinase G in thrombin-stimulated human platelets.Thromb Haemost 2003;89:310–7.

[64] Yang D, Chen H, Koupenova M, Carroll SH, Eliades A, Freedman JE, et al. A new rolefor the A2b adenosine receptor in regulating platelet function. J Thromb Haemost2010;8:817–27.

[65] Loyau S, Dumont B, Ollivier V, Boulaftali Y, Feldman L, Ajzenberg N, et al. Platelet gly-coprotein VI dimerization, an active process inducing receptor competence, is an in-dicator of platelet reactivity. Arterioscler Thromb Vasc Biol 2012;32:778–85.

[66] Enomoto Y, Adachi S, Doi T, Natsume H, Kato K, Matsushima-Nishiwaki R, et al.cAMP regulates ADP-induced HSP27 phosphorylation in human platelets. Int J MolMed 2011;27:695–700.

[67] Wiedmer T, Shattil SJ, Cunningham M, Sims PJ. Role of calcium and calpain incomplement-induced vesiculation of the platelet plasma membrane and in the ex-posure of the platelet factor Va receptor. Biochemistry 1990;29:623–32.

[68] Berckmans RJ, Nieuwland R, Boing AN, Romijn FP, Hack CE, Sturk A. Cell-derived mi-croparticles circulate in healthy humans and support low grade thrombin genera-tion. Thromb Haemost 2001;85:639–46.

[69] Chirinos JA, Heresi GA, Velasquez H, JyW, Jimenez JJ, Ahn E, et al. Elevation of endo-thelial microparticles, platelets, and leukocyte activation in patients with venousthromboembolism. J Am Coll Cardiol 2005;45:1467–71.

[70] Preston RA, JyW, Jimenez JJ, Mauro LM, Horstman LL, Valle M, et al. Effects of severehypertension on endothelial and platelet microparticles. Hypertension 2003;41:211–7.

[71] Tripodi A, Branchi A, Chantarangkul V, Clerici M, Merati G, Artoni A, et al. Hyperco-agulability in patients with type 2 diabetes mellitus detected by a thrombin gener-ation assay. J Thromb Thrombolysis 2011;31:165–72.

[72] Hron G, Kollars M, Weber H, Sagaster V, Quehenberger P, Eichinger S, et al. Tissuefactor-positive microparticles: cellular origin and association with coagulation acti-vation in patients with colorectal cancer. Thromb Haemost 2007;97:119–23.

[73] Mallat Z, Benamer H, Hugel B, Benessiano J, Steg PG, Freyssinet JM, et al. Elevatedlevels of shed membrane microparticles with procoagulant potential in the periph-eral circulating blood of patients with acute coronary syndromes. Circulation2000;101:841–3.

[74] Galli M, Grassi A, Barbui T. Platelet-derived microvesicles in thrombotic throm-bocytopenic purpura and hemolytic uremic syndrome. Thromb Haemost1996;75:427–31.

[75] Nomura S, Suzuki M, Katsura K, Xie GL, Miyazaki Y, Miyake T, et al. Platelet-derivedmicroparticles may influence the development of atherosclerosis in diabetesmellitus. Atherosclerosis 1995;116:235–40.

[76] Skeppholm M, Mobarrez F, Malmqvist K, Wallen H. Platelet-derived microparticlesduring and after acute coronary syndrome. Thromb Haemost 2012;107:1122–9.

[77] Lubsczyk B, Kollars M, Hron G, Kyrle PA, Weltermann A, Gartner V. Low doseacetylsalicylic acid and shedding of microparticles in vivo in humans. Eur J Clin In-vest 2010;40:477–82.

[78] Suades R, Padro T, Alonso R, Mata P, Badimon L. Lipid-lowering therapy with statinsreduces microparticle shedding from endothelium, platelets and inflammatory cellsindicating protection against cell activation. Thromb Haemost 2013:110.

[79] Mobarrez F, He S, Broijersen A,Wiklund B, Antovic A, Antovic J, et al. Atorvastatin re-duces thrombin generation and expression of tissue factor, P-selectin and GPIIIa onplatelet-derivedmicroparticles in patients with peripheral arterial occlusive disease.Thromb Haemost 2011;106:344–52.

[80] Lehmann JM, Moore LB, Smith-Oliver TA, Wilkison WO, Willson TM, Kliewer SA. Anantidiabetic thiazolidinedione is a high affinity ligand for peroxisome proliferator-activated receptor gamma (PPAR gamma). J Biol Chem 1995;270:12953–6.

[81] Cominacini L, Garbin U, Fratta Pasini A, Campagnola M, Davoli A, Foot E, et al.Troglitazone reduces LDL oxidation and lowers plasma E-selectin concentration inNIDDM patients. Diabetes 1998;47:130–3.

[82] Xiang AH, Peters RK, Kjos SL, Ochoa C, Marroquin A, Goico J, et al. Effect ofthiazolidinedione treatment on progression of subclinical atherosclerosis in pre-menopausal women at high risk for type 2 diabetes. J Clin Endocrinol Metab2005;90:1986–91.

[83] ZhuM, Flynt L, Ghosh S, MellemaM, Banerjee A,Williams E, et al. Anti-inflammatoryeffects of thiazolidinediones in human airway smooth muscle cells. Am J Respir CellMol Biol 2011;45:111–9.

[84] Loftus IM, Naylor AR, Goodall S, Crowther M, Jones L, Bell PR, et al. Increased matrixmetalloproteinase-9 activity in unstable carotid plaques. A potential role in acuteplaque disruption. Stroke 2000;31:40–7.

[85] Marx N, Froehlich J, Siam L, Ittner J, Wierse G, Schmidt A, et al. Antidiabetic PPARgamma-activator rosiglitazone reduces MMP-9 serum levels in type 2 diabetic pa-tients with coronary artery disease. Arterioscler Thromb Vasc Biol 2003;23:283–8.

[86] Ishizuka T, Itaya S, Wada H, Ishizawa M, Kimura M, Kajita K, et al. Differential effectof the antidiabetic thiazolidinediones troglitazone and pioglitazone on human plate-let aggregation mechanism. Diabetes 1998;47:1494–500.

[87] Dello Russo C, Gavrilyuk V,Weinberg G, Almeida A, Bolanos JP, Palmer J, et al. Perox-isome proliferator-activated receptor gamma thiazolidinedione agonists increaseglucose metabolism in astrocytes. J Biol Chem 2003;278:5828–36.

[88] Varo N, Vicent D, Libby P, Nuzzo R, Calle-Pascual AL, Bernal MR, et al. Elevated plas-ma levels of the atherogenic mediator soluble CD40 ligand in diabetic patients: anovel target of thiazolidinediones. Circulation 2003;107:2664–9.

[89] Marx N, Imhof A, Froehlich J, Siam L, Ittner J, Wierse G, et al. Effect of rosiglitazonetreatment on soluble CD40L in patients with type 2 diabetes and coronary arterydisease. Circulation 2003;107:1954–7.

[90] Ray DM, Spinelli SL, Pollock SJ, Murant TI, O'Brien JJ, Blumberg N, et al. Peroxisomeproliferator-activated receptor gamma and retinoid X receptor transcription factorsare released from activated human platelets and shed in microparticles. ThrombHaemost 2008;99:86–95.

[91] Koppler B, Cohen C, Schlondorff D, Mack M. Differential mechanisms of microparti-cle transfer toB cells andmonocytes: anti-inflammatory properties of microparticles.Eur J Immunol 2006;36:648–60.

[92] Singh N, Webb R, Adams R, Evans SA, Al-Mosawi A, Evans M, et al. The PPAR-gammaactivator, Rosiglitazone, inhibits actin polymerisation in monocytes: involvement ofAkt and intracellular calcium. Biochem Biophys Res Commun 2005;333:455–62.

[93] Ialenti A, Grassia G, Di Meglio P, Maffia P, Di Rosa M, Ianaro A. Mechanism of theanti-inflammatory effect of thiazolidinediones: relationship with the glucocorticoidpathway. Mol Pharmacol 2005;67:1620–8.

[94] Eggertsen R, Hansson L. Effects of treatment with nifedipine and metoprolol in es-sential hypertension. Eur J Clin Pharmacol 1982;21:389–90.

[95] Kloner RA. Nifedipine in ischemic heart disease. Circulation 1995;92:1074–8.[96] Shih CY, Lin MH, Fan HC, Chen FC, Chou TC. Mechanisms of antiplatelet activity of

nifedipine: role of peroxisome proliferator-activated receptor-beta-gamma-depen-dent processes. J Hypertens 2014;32:181–92.

[97] Nishikawa M, Komada F, Morita K, Deguchi K, Shirakawa S. Inhibition of plateletaggregation by the cAMP-phosphodiesterase inhibitor, cilostamide, may not beassociated with activation of cAMP-dependent protein kinase. Cell Signal1992;4:453–63.