Vascular Pharmacology: Eduardo Fuentes, Iván Palomo

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 5

Vascular Pharmacology 62 (2014) 162166

Contents lists available at ScienceDirect

Vascular Pharmacology
journal homepage: www.elsevier.com/locate/vph

Review

Mechanism of antiplatelet action of hypolipidemic, antidiabetic


and antihypertensive drugs by PPAR activation
PPAR agonists: New antiplatelet agents
Eduardo Fuentes , Ivn 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

a r t i c l e

i n f o

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

a b s t r a c t
Given the prevalence of cardiovascular disease in patients with cardiovascular risk factors (i.e., hypertension, diabetes, smoking and obesity) and that platelet activation plays an important pathogenic role in cardiovascular diseases, 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 mechanism 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 cAMPphosphodiesterase, 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.
2.
3.
4.

Introduction . . . . . . . . . . . . . . . . . .
PPARs and atherothrombosis . . . . . . . . . .
Mechanism of antiplatelet action by PPAR activation
Drugs and platelet inhibition: role of PPAR activation
4.1.
Statins and brates . . . . . . . . . . .
4.2.
Thiazolidinediones . . . . . . . . . . .
4.3.
Nifedipine . . . . . . . . . . . . . . .
5.
Conclusions . . . . . . . . . . . . . . . . . .
Declaration of interest . . . . . . . . . . . . . . . .
Acknowledgments . . . . . . . . . . . . . . . . .
References . . . . . . . . . . . . . . . . . . . . .

. . . .
. . . .
. . . .
. . .
. . . .
. . . .
. . . .
. . . .
. . . .
. . . .
. . . .

.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.

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
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: edfuentes@utalca.cl (E. Fuentes), ipalomo@utalca.cl (I. Palomo).

http://dx.doi.org/10.1016/j.vph.2014.05.008
1537-1891/ 2014 Elsevier Inc. All rights reserved.

.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.

162
163
163
163
163
164
164
165
165
165
165

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 increased 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 enhanced 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 following an atheromatous plaque rupture, [9,10].

E. Fuentes, I. Palomo / Vascular Pharmacology 62 (2014) 162166

Given the central role of platelets in the atherosclerotic-related inammatory response and the subsequent thrombotic event, a variety
of antiplatelet agents have been developed [11,12]. Although antiplatelet drugs have been proven to be benecial in patients with clinical evidence of CVD, outcomes still remain poor [13]. In this way, platelets
from diabetic patients release greater amounts of thromboxane A2
(TXA2) than platelets from non-diabetic patients receiving treatment
with low-dose aspirin [14]. This is because all the current available antiplatelet agents only target one signal pathway and moderately inhibit
platelet activation (e.g. aspirin and clopidogrel) [15]. Therefore, there is
much room for further improvement of antiplatelet treatment and the
need to identify the mechanisms by which hypolipidemic (statins and
brates), antidiabetic (thiazolidinediones) and antihypertensive (nifedipine) drugs inhibit platelet activation. It has also been observed that
these drugs are ligands for peroxisome proliferator-activated receptors
(PPARs) in cells which have a nucleus [1618].
PPARs are involved in many biological processes, including lipid and
energy metabolism inammation responses, and atherosclerotic plaque
formation [1921]. Selective agonists through their action on nuclear
receptors (e.g. PPARs) regulate platelet function despite the absence of
a nucleus in platelets. [22]. In this sense, the present article describes
the antiplatelet action mechanism of hypolipidemic, antidiabetic and
antihypertensive drugs via PPAR activation.

163

the expression of nitric oxide synthase (NOS) and thrombomodulin


[46]. PPAR- activation also inhibits platelet function induced by collagen through the modulation of early glycoprotein (GP) VI signaling, at
the level of Syk and LAT [47]. In addition, platelet incubation with a natural PPAR- agonist (15d-PGJ(2)) or with a potent synthetic PPAR- ligand (rosiglitazone) not only attenuates platelet activation, but also
decreases the release of platelet proinammatory and procoagulant mediators (sCD40L and TXA2) [33,37].
Agonists of PPAR-, including GW0742 and L-165041, inhibit platelet activation after 5 min of incubation. Clearly with such acute exposure
and as platelets have no nucleus, PPAR- is an active antithrombotic
pathway in platelets, whose effects are independent of the nucleus [38].
Moreover, PPAR- activators may exert vasculo-protective action
through suppression of PDGF-BB production in a megakaryocyte/platelet
pathway [48]. Prostacyclin formed by the consecutive actions of
cyclooxygenase-1 (COX-1) and prostacyclin synthase is arguably the
most important endogenous antiplatelet hormone identied to date
[49]. Prostacyclin inhibits platelet function via stimulation of a surface
prostanoid receptor linked to the activation of adenylyl cyclase and increased intracellular levels of cyclic adenosine monophosphate (cAMP)
[50]. Moreover, prostacyclin activates PPAR-/ in human platelets in
the low 109 M range. Therefore, an additional mechanism of antiplatelet action by prostacyclin occurs by activation of PPAR-/ and its effects
on platelet aggregation is synergistic with nitric oxide (NO) [38,51].

2. PPARs and atherothrombosis


PPARs consist of a family of three nuclear receptor isoforms (, /,
and ) [23]. PPARs are the key regulators of metabolism and inammation, and play an important role in chronic inammatory disease processes [24,25]. Accumulating evidence suggests that PPAR activation is
a key mechanism to reduce atherosclerosis and improve cardiovascular
function [2628].
Recent studies suggest that PPAR- activation decreases atherogenesis, not only by correcting metabolic disorders, but also through its direct effects on the vascular wall [29]. In addition, PPAR- and PPAR-/
have also been suggested in the regulation of inammation and slow
atherosclerosis progression [20,30].
Ligands of PPAR- are important regulators of lipid and lipoprotein
metabolism, thereby positively affecting plasma lipid risk factors related
to atherosclerosis [31]. In this context, the lipid-lowering effect of
fenobrate is achieved by activating PPAR- and AMP-activated protein
kinase (AMPK) signaling pathway that results in increasing lipolysis and
fatty acid -oxidation [32].
PPARs appear to play a major role in the regulation of atherogenesis
by countering the inammation-provoking action of platelet adhesion
and activation [33].
3. Mechanism of antiplatelet action by PPAR activation
Nuclear receptors are transcription factors that are activated by ligands and subsequently bind to regulatory regions in target genes;
allowing the organism to integrate signals coming from the environment [34]. While steroid/nuclear receptors are recognized via for their
role in gene regulation, increasing evidence supports nongenomic actions of these receptors [35,36]. Thus, although platelets lack a nucleus,
they express a number of transcription factors including steroid/nuclear
receptors such as PPARs [37,38], glucocorticoid receptor (GR) [39],
retinoic X receptor (RXR) [41], estrogen receptor (ER) [40] and nuclear
factor kappa B (NF-B) [42]. Thus, selective ligands for these receptors
regulate platelet aggregation and activation [22,43].
While many reports focus on PPAR expression in the nucleus [44,45],
in this article we focus on the role of PPAR-cytoplasm in platelet function. Recent studies have provided the rst evidence that human bone
marrow megakaryocytes and human platelets express PPAR- [37].
PPAR- activation decreases platelet aggregation and delays intraarterial thrombus formation in rats, at least partially, by an increase in

4. Drugs and platelet inhibition: role of PPAR activation


Given the prevalence of CVD in patients with cardiovascular risk
factors (i.e., hypertension, diabetes, smoking and obesity) and that
platelet activation plays an important pathogenic role in atherothrombosis [3,52], 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 by PPAR activation.
4.1. Statins and brates
The 3-hydroxy-3-methylglutaryl-coenzyme (HMG-CoA) reductase
inhibitors, also known as statins, are the most effective class of drugs
for lowering serum low-density lipoprotein cholesterol (LDL-C) concentrations [53]. In this way, another class of drug called brates reduces
triglycerides and increases high-density lipoprotein cholesterol (HDLC) [54]. In this article, we discuss how statins and brates appear to
have more biological effects than those originally targeted. Statins and
brates can exert benecial anti-inammatory and antithrombotic effects in after patients with a high risk of coronary artery disease as
early as 3 days after therapy [55]. Thus, statins (simvastatin, atorvastatin, or cerivastatin) decrease morbidity and mortality in patients with
CVD by their effects on proinammatory cytokines: interleukin 6 (IL6), IL-8, and monocyte chemoattractant protein-1 (MCP-1) [56]. Meanwhile, brates by PPAR-activation may inhibit atherosclerosis development in vivo [57].
Interestingly both statins and brates inhibit platelet function [54].
Inhibition of platelet aggregation by simvastatin involves the activation
of the cAMP-eNOS/NO-cGMP pathway, resulting in the inhibition of the
PLC-2/PKC/p38 MAPK/TXA2 cascade [58,59]. Therefore, the direct inhibitory effects of statins and brates on platelet activation are mediated by PPAR activation and this new nding reveals some of the
pleiotropic effects of these drugs [60]. In platelets, PPAR signaling pathway activation involves binding and repression of PKC, and increasing of
cAMP and cGMP levels [60]. The increase of intraplatelet levels of cAMP
is due to the fact that the repression of PKC allows greater activity of
adenylyl cyclase, which converts ATP to cAMP [61,62]. In addition,
cAMP induced inhibition of platelet P-selectin expression, platelet

164

E. Fuentes, I. Palomo / Vascular Pharmacology 62 (2014) 162166

Fig. 1. Mechanism of antiplatelet action of PPAR ligands. AA = arachidonic acid; AKT = known as protein kinase B; AMP = adenosine monophosphate; cAMP = cyclic adenosine
monophosphate; cGMP = cyclic guanosine monophosphate; COX-1 = cyclooxygenase-1; NO = nitric oxide; NOS = nitric oxide synthase; PDE3 = phosphodiesterase-3; PGH2 = prostaglandin H2; PGG2 = prostaglandin G2; PKA = protein kinase A; PKC = protein kinase C; PKG = protein kinase G; PLA2 = phospholipase A2; PMP = platelet microparticles; PPARs = peroxisome
proliferator-activated receptors; TXA2 = thromboxane A2; VASP = vasodilator-stimulated phosphoprotein; VASP-P = vasodilator-stimulated phosphoprotein phosphorylation.

aggregation and release of sCD40L, for the large part, is mediated


through the activation of protein kinase A (PKA) [6366].
Platelet microparticles (PMP) have an average diameter of 0.1 m
[67], and are the most abundant type of microparticles (MP) in human
blood (representing 7090%) and contribute to many biologically processes [68]. PMP levels are greatly elevated in patients (range from
~ 300011,000/L) with diseases including venous thromboembolism
[69], hypertension [70], diabetes [71], cancer [72], immunologic platelet
disorders and arterial thrombosis [73,74]. In humans, plasma levels of
circulating PMP are known to increase with the presence of cardiovascular risk factors and progression of atherosclerosis [75]. Even after initiation of antithrombotic treatment, PMP are still elevated after six
months [76]. Possibly because a low dose of aspirin might not be strong
enough to suppress the shedding of PMP in the microcirculation [77].
With respect to the role of statins, MP from hypercholesterolemia patients with statin treatment had reduced markers of activated platelets
(GPIIb-IIIa), activated inammatory cells (Mac-1) and tissue factor [78].
Meanwhile atorvastatin reduces thrombin generation and expression of
tissue factor, GPIIIa and P-selectin on PMP in patients with peripheral
vascular disease [79]. In this way statins may inhibit thrombotic event
generation partly through a PMP dependent mechanism.

4.2. Thiazolidinediones
The thiazolidinediones (TZDs) are a class of oral antidiabetic drugs
that exert effects through a mechanism that involves an activation of
PPAR- [80]. TZDs (rosiglitazone, pioglitazone and troglitazone) have
been shown to protect LDL-C from oxidative modication in patients
with type 2 diabetes that were treated for 8 weeks, one of the key initial
events in atherogenesis [81]. In addition, TZD activation of PPAR- reduced the progression of subclinical atherosclerosis by normalizing

proatherogenic metabolic abnormalities of the insulin resistance, and


through an inhibition of vascular cell growth and movement [82].
Recent studies suggest that TZDs may also be clinically benecial to
inammatory mediators [83]. Matrix metalloproteinase-9 (MMP-9) is
critically involved in the development of unstable plaques [84]. Also
MMP-9 levels are increased in type 2 diabetes patients with coronary
artery disease, and the treatment of these patients with the antidiabetic
PPAR-activator rosiglitazone signicantly reduces the serum levels of
MMP-9, tumor necrosis factor alpha (TNF-) and serum amyloid A [85].
Troglitazone has a potent inhibitory effect on platelet aggregation via
the suppression of the thrombin-induced activation of phosphoinositide
signaling in human platelets [86]. Based on the function of other cells
(astrocyte cells), the TZD mode of action is likely related to the elevation
of the intracellular cAMP level by PPAR- activation [87]. This could explain why the treatment with TZDs reduces sCD40L serum levels in patients with type 1 and 2 diabetes and coronary artery disease [88,89].
PPAR- containing PMP are quickly released from -granule into extracellular parts when platelets are activated by collagen [90]. Then,
PMP-PPAR- can be internalized by the monocytic cell line (THP-1). In
this way, THP-1 cell activation can be attenuated in the presence of a
PPAR- agonist (rosiglitazone) [91,92].
In addition, one possible mechanism explaining the antiinammatory activity of TZDs is its ability to activate GR [93]. In
this way, classic anti-inammatory glucocorticoids are due to the
fact that human platelets contain GR [39].
4.3. Nifedipine
Nifedipine, a L-type calcium channel blocker, is widely used in the
treatment of hypertension and coronary heart disease [94,95]. Nifedipine increases the activity and intracellular expression of PPAR-/
from activated platelets. Therefore, the mechanism of antiplatelet action

E. Fuentes, I. Palomo / Vascular Pharmacology 62 (2014) 162166

of nifedipine is PAR-/-dependent with upregulation of the PI3K/Akt/


NO/cGMP/PKG pathway, and the inhibition of PKC activity and intracellular Ca2+ mobilization in platelets [96]. Taking into consideration all
these ndings in vivo evidence of such in vitro-related nifedipineantithrombotic effects is provided. Indeed, using a murine model of
thrombus formation, the administration of nifedipine substantially
inhibited uorescein sodium-induced vessel thrombus formation. In addition, its antithrombotic effect is considerably reduced in the presence
of PPAR-/ antagonists [96].
Finally, the mechanism of antiplatelet action of statins, brates, TZDs
and nifedipine by direct activation of PPARs is mediated by inhibition of
COX-1 [37], PKC [62] and Ca2+ mobilization [47], and indirect inhibition
of TXA2, sCD40L, PMP, P-selectin and cAMP-phosphodiesterase, and
stimulation of PKG (increased levels of cGMP) and PKA (increased levels
of cAMP) [63,97] (Fig. 1).
5. Conclusions
The data presented in this article has demonstrated that the ability of
statins, brates, TZDs and nifedipine to inhibit platelet activation is likely attributable to their inuence on PPAR activation. Thus, these observations highlight PPARs as a novel therapeutic target for the treatment
and 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 Investigacin de Excelencia Interdisciplinaria
en 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 (Prevalence
of cardiovascular risk factors in adult from Talca, Chile). Rev Med Chil
2007;135:90412.
[2] Bautista LE, Orostegui M, Vera LM, Prada GE, Orozco LC, Herran OF. Prevalence and
impact of cardiovascular risk factors in Bucaramanga, Colombia: results from the
Countrywide Integrated Noncommunicable Disease Intervention Programme
(CINDI/CARMEN) baseline survey. Eur J Cardiovasc Prev Rehabil 2006;13:76975.
[3] Grundy SM, Pasternak R, Greenland P, Smith Jr S, Fuster V. Assessment of cardiovascular risk by use of multiple-risk-factor assessment equations: a statement for
healthcare professionals from the American Heart Association and the American
College of Cardiology. Circulation 1999;100:148192.
[4] Mujica V, Urzua A, Leiva E, Diaz N, Moore-Carrasco R, Vasquez M, et al. Intervention
with education and exercise reverses the metabolic syndrome in adults. J Am Soc
Hypertens 2010;4:14853.
[5] Colwell JA, Nesto RW. The platelet in diabetes: focus on prevention of ischemic
events. Diabetes Care 2003;26:21818.
[6] Vinik AI, Erbas T, Park TS, Nolan R, Pittenger GL. Platelet dysfunction in type 2 diabetes. Diabetes Care 2001;24:147685.
[7] Sagel J, Colwell JA, Crook L, Laimins M. Increased platelet aggregation in early
diabetus mellitus. Ann Intern Med 1975;82:7338.
[8] Palomo I, Moore-Carrasco R, Alarcon M, Rojas A, Espana F, Andres V, et al. Pathophysiology of the proatherothrombotic state in the metabolic syndrome. Front
Biosci (Schol Ed) 2010;2:194208.
[9] Fuentes QE, Fuentes QF, Andres V, Pello OM, de Mora JF, Palomo GI. Role of platelets
as mediators that link inammation and thrombosis in atherosclerosis. Platelets
2013;24:25562.
[10] Ruggeri ZM. Mechanisms initiating platelet thrombus formation. Thromb Haemost
1997;78:6116.
[11] Collins B, Hollidge C. Antithrombotic drug market. Nat Rev Drug Discov 2003;2:112.
[12] Palomo I, Toro C, Alarcon M. The role of platelets in the pathophysiology of atherosclerosis (Review). Mol Med Rep 2008;1:17984.
[13] Meadows TA, Bhatt DL. Clinical aspects of platelet inhibitors and thrombus formation. Circ Res 2007;100:126175.
[14] DiMinno G, Silver MJ, Cerbone AM, Murphy S. Trial of repeated low-dose aspirin in
diabetic angiopathy. Blood 1986;68:88691.
[15] Reaume KT, Regal RE, Dorsch MP. Indications for dual antiplatelet therapy with aspirin and clopidogrel: evidence-based recommendations for use. Ann Pharmacother
2008;42:5507.

165

[16] Qu S, Su D, Altomonte J, Kamagate A, He J, Perdomo G, et al. PPAR{alpha} mediates


the hypolipidemic action of brates by antagonizing FoxO1. Am J Physiol Endocrinol
Metab 2007;292:E42134.
[17] Yano M, Matsumura T, Senokuchi T, Ishii N, Murata Y, Taketa K, et al. Statins activate
peroxisome proliferator-activated receptor gamma through extracellular signal-regulated kinase 1/2 and p38 mitogen-activated protein kinase-dependent cyclooxygenase-2 expression in macrophages. Circ Res 2007;100:144251.
[18] Diaz-Deln J, Morales M, Caelles C. Hypoglycemic action of thiazolidinediones/
peroxisome proliferator-activated receptor gamma by inhibition of the c-Jun
NH2-terminal kinase pathway. Diabetes 2007;56:186571.
[19] Berger J, Moller DE. The mechanisms of action of PPARs. Annu Rev Med 2002;53:
40935.
[20] Lee CH, Chawla A, Urbiztondo N, Liao D, Boisvert WA, Evans RM, et al. Transcriptional repression of atherogenic inammation: modulation by PPARdelta. Science
2003;302:4537.
[21] Fuentes E, Fuentes F, Vilahur G, Badimon L, Palomo I. Mechanisms of chronic state of
inammation as mediators that link obese adipose tissue and metabolic syndrome.
Mediators Inamm 2013;2013:136584.
[22] Bishop-Bailey D. The platelet as a model system for the acute actions of nuclear receptors. Steroids 2010;75:5705.
[23] Moraes LA, Piqueras L, Bishop-Bailey D. Peroxisome proliferator-activated receptors
and inammation. Pharmacol Ther 2006;110:37185.
[24] Ahmed W, Ziouzenkova O, Brown J, Devchand P, Francis S, Kadakia M, et al. PPARs
and their metabolic modulation: new mechanisms for transcriptional regulation?
J Intern Med 2007;262:18498.
[25] Fuentes E, Guzman-Jofre L, Moore-Carrasco R, Palomo I. Role of PPARs in inammatory
processes associated with metabolic syndrome (Review). Mol Med Rep 2013;8:16116.
[26] Kersten S, Desvergne B, Wahli W. Roles of PPARs in health and disease. Nature
2000;405:4214.
[27] Inoue H, Jiang XF, Katayama T, Osada S, Umesono K, Namura S. Brain protection by
resveratrol and fenobrate against stroke requires peroxisome proliferator-activated receptor alpha in mice. Neurosci Lett 2003;352:2036.
[28] Verreth W, De Keyzer D, Pelat M, Verhamme P, Ganame J, Bielicki JK, et al. Weightloss-associated induction of peroxisome proliferator-activated receptor-alpha and
peroxisome proliferator-activated receptor-gamma correlate with reduced atherosclerosis and improved cardiovascular function in obese insulin-resistant mice. Circulation 2004;110:325969.
[29] Winkler K, Konrad T, Fullert S, Friedrich I, Destani R, Baumstark MW, et al. Pioglitazone reduces atherogenic dense LDL particles in nondiabetic patients with arterial
hypertension: a double-blind, placebo-controlled study. Diabetes Care 2003;26:
258894.
[30] Delerive P, Fruchart JC, Staels B. Peroxisome proliferator-activated receptors in inammation control. J Endocrinol 2001;169:4539.
[31] Torra IP, Chinetti G, Duval C, Fruchart JC, Staels B. Peroxisome proliferator-activated
receptors: from transcriptional control to clinical practice. Curr Opin Lipidol
2001;12:24554.
[32] Chen WL, Chen YL, Chiang YM, Wang SG, Lee HM. Fenobrate lowers lipid accumulation in myotubes by modulating the PPARalpha/AMPK/FoxO1/ATGL pathway.
Biochem Pharmacol 2012;84:52231.
[33] Ray DM, Spinelli SL, O'Brien JJ, Blumberg N, Phipps RP. Platelets as a novel target for
PPARgamma ligands: implications for inammation, diabetes, and cardiovascular
disease. BioDrugs 2006;20:23141.
[34] Chinetti G, Fruchart JC, Staels B. Peroxisome proliferator-activated receptors
(PPARs): nuclear receptors at the crossroads between lipid metabolism and inammation. Inamm Res 2000;49:497505.
[35] Croxtall JD, van Hal PT, Choudhury Q, Gilroy DW, Flower RJ. Different glucocorticoids
vary in their genomic and non-genomic mechanism of action in A549 cells. Br J
Pharmacol 2002;135:5119.
[36] Losel RM, Falkenstein E, Feuring M, Schultz A, Tillmann HC, Rossol-Haseroth K, et al.
Nongenomic steroid action: controversies, questions, and answers. Physiol Rev
2003;83:9651016.
[37] Akbiyik F, Ray DM, Gettings KF, Blumberg N, Francis CW, Phipps RP. Human bone
marrow megakaryocytes and platelets express PPARgamma, and PPARgamma agonists blunt platelet release of CD40 ligand and thromboxanes. Blood 2004;104:
13618.
[38] Ali FY, Davidson SJ, Moraes LA, Traves SL, Paul-Clark M, Bishop-Bailey D, et al. Role of
nuclear receptor signaling in platelets: antithrombotic effects of PPARbeta. FASEB J
2006;20:3268.
[39] Moraes LA, Paul-Clark MJ, Rickman A, Flower RJ, Goulding NJ, Perretti M. Ligandspecic glucocorticoid receptor activation in human platelets. Blood 2005;106:
416775.
[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 inhibiting
Gq in human platelets. Blood 2007;109:37414.
[41] Moro L, Reineri S, Piranda D, Pietrapiana D, Lova P, Bertoni A, et al. Nongenomic
effects of 17beta-estradiol in human platelets: potentiation of thrombin-induced
aggregation through estrogen receptor beta and Src kinase. Blood 2005;105:
11521.
[42] Malaver E, Romaniuk MA, D'Atri LP, Pozner RG, Negrotto S, Benzadon R, et al. NFkappaB inhibitors impair platelet activation responses. J Thromb Haemost
2009;7:133343.
[43] Fuentes E, Rojas A, Palomo I. Role of multiligand/RAGE axis in platelet activation.
Thromb Res 2014;133:30814.
[44] Padilla J, Leung E, Phipps RP. Human B lymphocytes and B lymphomas express PPAR-gamma and are killed by PPAR-gamma agonists. Clin Immunol
2002;103:2233.

166

E. Fuentes, I. Palomo / Vascular Pharmacology 62 (2014) 162166

[45] Chawla A. Control of macrophage activation and function by PPARs. Circ Res
2010;106:155969.
[46] Li D, Chen K, Sinha N, Zhang X, Wang Y, Sinha AK, et al. The effects of PPAR-gamma
ligand pioglitazone on platelet aggregation and arterial thrombus formation.
Cardiovasc Res 2005;65:90712.
[47] Moraes LA, Spyridon M, Kaiser WJ, Jones CI, Sage T, Atherton RE, et al. Non-genomic
effects of PPARgamma ligands: inhibition of GPVI-stimulated platelet activation. J
Thromb Haemost 2010;8:57787.
[48] Hashizume S, Akaike M, Azuma H, Ishikawa K, Yoshida S, Sumitomo-Ueda Y, et al.
Activation of peroxisome proliferator-activated receptor alpha in megakaryocytes
reduces platelet-derived growth factor-BB in platelets. J Atheroscler Thromb
2011;18:13847.
[49] Higgs EA, Higgs GA, Moncada S, Vane JR. Prostacyclin (PGI2) inhibits the formation
of platelet thrombi in arterioles and venules of the hamster cheek pouch. Br J
Pharmacol 1978;63:5359.
[50] Beck F, Geiger J, Gambaryan S, Veit J, Vaudel M, Nollau P, et al. Time-resolved characterization of cAMP/PKA-dependent signaling reveals that platelet inhibition is a
concerted process involving multiple signaling pathways. Blood.123:e1-e10.
[51] Fauti T, Muller-Brusselbach S, Kreutzer M, Rieck M, Meissner W, Rapp U, et al. Induction of PPARbeta and prostacyclin (PGI2) synthesis by Raf signaling: failure of PGI2
to activate PPARbeta. FEBS J 2006;273:1709.
[52] Davi G, Patrono C. Platelet activation and atherothrombosis. N Engl J Med
2007;357:248294.
[53] Istvan ES, Deisenhofer J. Structural mechanism for statin inhibition of HMG-CoA reductase. Science 2001;292:11604.
[54] Staels B, Dallongeville J, Auwerx J, Schoonjans K, Leitersdorf E, Fruchart JC. Mechanism of action of brates on lipid and lipoprotein metabolism. Circulation
1998;98:208893.
[55] Undas A, Celinska-Lowenhoff M, Domagala TB, Iwaniec T, Dropinski J, Lowenhoff T,
et al. Early antithrombotic and anti-inammatory effects of simvastatin versus
fenobrate in patients with hypercholesterolemia. Thromb Haemost 2005;94:
1939.
[56] Rezaie-Majd A, Maca T, Bucek RA, Valent P, Muller MR, Husslein P, et al. Simvastatin
reduces expression of cytokines interleukin-6, interleukin-8, and monocyte
chemoattractant protein-1 in circulating monocytes from hypercholesterolemic patients. Arterioscler Thromb Vasc Biol 2002;22:11949.
[57] Duez H, Chao YS, Hernandez M, Torpier G, Poulain P, Mundt S, et al. Reduction of
atherosclerosis by the peroxisome proliferator-activated receptor alpha agonist
fenobrate in mice. J Biol Chem 2002;277:480517.
[58] Lee YM, Chen WF, Chou DS, Jayakumar T, Hou SY, Lee JJ, et al. Cyclic nucleotides and
mitogen-activated protein kinases: regulation of simvastatin in platelet activation. J
Biomed Sci 2010;17:45.
[59] Chou TC, Lin YF, Wu WC, Chu KM. Enhanced nitric oxide and cyclic GMP formation
plays a role in the anti-platelet activity of simvastatin. Br J Pharmacol 2008;153:12817.
[60] Ali FY, Armstrong PC, Dhanji AR, Tucker AT, Paul-Clark MJ, Mitchell JA, et al. Antiplatelet actions of statins and brates are mediated by PPARs. Arterioscler Thromb
Vasc Biol 2009;29:70611.
[61] Levesque L, Crooke ST. Depletion of protein kinase C-alpha by antisense oligonucleotides alters beta-adrenergic function and reverses the phorbol ester-induced reduction of isoproterenol-induced adenosine 35-cyclic monophosphate accumulation
in murine Swiss 3T3 broblasts. J Pharmacol Exp Ther 1998;287:42534.
[62] Ali FY, Hall MG, Desvergne B, Warner TD, Mitchell JA. PPARbeta/delta agonists
modulate platelet function via a mechanism involving PPAR receptors and specic association/repression of PKCalphabrief report. Arterioscler Thromb Vasc
Biol 2009;29:18713.
[63] Libersan D, Rousseau G, Merhi Y. Differential regulation of P-selectin expression by
protein kinase A and protein kinase G in thrombin-stimulated human platelets.
Thromb Haemost 2003;89:3107.
[64] Yang D, Chen H, Koupenova M, Carroll SH, Eliades A, Freedman JE, et al. A new role
for the A2b adenosine receptor in regulating platelet function. J Thromb Haemost
2010;8:81727.
[65] Loyau S, Dumont B, Ollivier V, Boulaftali Y, Feldman L, Ajzenberg N, et al. Platelet glycoprotein VI dimerization, an active process inducing receptor competence, is an indicator of platelet reactivity. Arterioscler Thromb Vasc Biol 2012;32:77885.
[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 Mol
Med 2011;27:695700.
[67] Wiedmer T, Shattil SJ, Cunningham M, Sims PJ. Role of calcium and calpain in
complement-induced vesiculation of the platelet plasma membrane and in the exposure of the platelet factor Va receptor. Biochemistry 1990;29:62332.
[68] Berckmans RJ, Nieuwland R, Boing AN, Romijn FP, Hack CE, Sturk A. Cell-derived microparticles circulate in healthy humans and support low grade thrombin generation. Thromb Haemost 2001;85:63946.
[69] Chirinos JA, Heresi GA, Velasquez H, Jy W, Jimenez JJ, Ahn E, et al. Elevation of endothelial microparticles, platelets, and leukocyte activation in patients with venous
thromboembolism. J Am Coll Cardiol 2005;45:146771.
[70] Preston RA, Jy W, Jimenez JJ, Mauro LM, Horstman LL, Valle M, et al. Effects of severe
hypertension on endothelial and platelet microparticles. Hypertension 2003;41:
2117.
[71] Tripodi A, Branchi A, Chantarangkul V, Clerici M, Merati G, Artoni A, et al. Hypercoagulability in patients with type 2 diabetes mellitus detected by a thrombin generation assay. J Thromb Thrombolysis 2011;31:16572.

[72] Hron G, Kollars M, Weber H, Sagaster V, Quehenberger P, Eichinger S, et al. Tissue


factor-positive microparticles: cellular origin and association with coagulation activation in patients with colorectal cancer. Thromb Haemost 2007;97:11923.
[73] Mallat Z, Benamer H, Hugel B, Benessiano J, Steg PG, Freyssinet JM, et al. Elevated
levels of shed membrane microparticles with procoagulant potential in the peripheral circulating blood of patients with acute coronary syndromes. Circulation
2000;101:8413.
[74] Galli M, Grassi A, Barbui T. Platelet-derived microvesicles in thrombotic thrombocytopenic purpura and hemolytic uremic syndrome. Thromb Haemost
1996;75:42731.
[75] Nomura S, Suzuki M, Katsura K, Xie GL, Miyazaki Y, Miyake T, et al. Platelet-derived
microparticles may inuence the development of atherosclerosis in diabetes
mellitus. Atherosclerosis 1995;116:23540.
[76] Skeppholm M, Mobarrez F, Malmqvist K, Wallen H. Platelet-derived microparticles
during and after acute coronary syndrome. Thromb Haemost 2012;107:11229.
[77] Lubsczyk B, Kollars M, Hron G, Kyrle PA, Weltermann A, Gartner V. Low dose
acetylsalicylic acid and shedding of microparticles in vivo in humans. Eur J Clin Invest 2010;40:47782.
[78] Suades R, Padro T, Alonso R, Mata P, Badimon L. Lipid-lowering therapy with statins
reduces microparticle shedding from endothelium, platelets and inammatory cells
indicating protection against cell activation. Thromb Haemost 2013:110.
[79] Mobarrez F, He S, Broijersen A, Wiklund B, Antovic A, Antovic J, et al. Atorvastatin reduces thrombin generation and expression of tissue factor, P-selectin and GPIIIa on
platelet-derived microparticles in patients with peripheral arterial occlusive disease.
Thromb Haemost 2011;106:34452.
[80] Lehmann JM, Moore LB, Smith-Oliver TA, Wilkison WO, Willson TM, Kliewer SA. An
antidiabetic thiazolidinedione is a high afnity ligand for peroxisome proliferatoractivated receptor gamma (PPAR gamma). J Biol Chem 1995;270:129536.
[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 in
NIDDM patients. Diabetes 1998;47:1303.
[82] Xiang AH, Peters RK, Kjos SL, Ochoa C, Marroquin A, Goico J, et al. Effect of
thiazolidinedione treatment on progression of subclinical atherosclerosis in premenopausal women at high risk for type 2 diabetes. J Clin Endocrinol Metab
2005;90:198691.
[83] Zhu M, Flynt L, Ghosh S, Mellema M, Banerjee A, Williams E, et al. Anti-inammatory
effects of thiazolidinediones in human airway smooth muscle cells. Am J Respir Cell
Mol Biol 2011;45:1119.
[84] Loftus IM, Naylor AR, Goodall S, Crowther M, Jones L, Bell PR, et al. Increased matrix
metalloproteinase-9 activity in unstable carotid plaques. A potential role in acute
plaque disruption. Stroke 2000;31:407.
[85] Marx N, Froehlich J, Siam L, Ittner J, Wierse G, Schmidt A, et al. Antidiabetic PPAR
gamma-activator rosiglitazone reduces MMP-9 serum levels in type 2 diabetic patients with coronary artery disease. Arterioscler Thromb Vasc Biol 2003;23:2838.
[86] Ishizuka T, Itaya S, Wada H, Ishizawa M, Kimura M, Kajita K, et al. Differential effect
of the antidiabetic thiazolidinediones troglitazone and pioglitazone on human platelet aggregation mechanism. Diabetes 1998;47:1494500.
[87] Dello Russo C, Gavrilyuk V, Weinberg G, Almeida A, Bolanos JP, Palmer J, et al. Peroxisome proliferator-activated receptor gamma thiazolidinedione agonists increase
glucose metabolism in astrocytes. J Biol Chem 2003;278:582836.
[88] Varo N, Vicent D, Libby P, Nuzzo R, Calle-Pascual AL, Bernal MR, et al. Elevated plasma levels of the atherogenic mediator soluble CD40 ligand in diabetic patients: a
novel target of thiazolidinediones. Circulation 2003;107:26649.
[89] Marx N, Imhof A, Froehlich J, Siam L, Ittner J, Wierse G, et al. Effect of rosiglitazone
treatment on soluble CD40L in patients with type 2 diabetes and coronary artery
disease. Circulation 2003;107:19547.
[90] Ray DM, Spinelli SL, Pollock SJ, Murant TI, O'Brien JJ, Blumberg N, et al. Peroxisome
proliferator-activated receptor gamma and retinoid X receptor transcription factors
are released from activated human platelets and shed in microparticles. Thromb
Haemost 2008;99:8695.
[91] Koppler B, Cohen C, Schlondorff D, Mack M. Differential mechanisms of microparticle transfer toB cells and monocytes: anti-inammatory properties of microparticles.
Eur J Immunol 2006;36:64860.
[92] Singh N, Webb R, Adams R, Evans SA, Al-Mosawi A, Evans M, et al. The PPAR-gamma
activator, Rosiglitazone, inhibits actin polymerisation in monocytes: involvement of
Akt and intracellular calcium. Biochem Biophys Res Commun 2005;333:45562.
[93] Ialenti A, Grassia G, Di Meglio P, Mafa P, Di Rosa M, Ianaro A. Mechanism of the
anti-inammatory effect of thiazolidinediones: relationship with the glucocorticoid
pathway. Mol Pharmacol 2005;67:16208.
[94] Eggertsen R, Hansson L. Effects of treatment with nifedipine and metoprolol in essential hypertension. Eur J Clin Pharmacol 1982;21:38990.
[95] Kloner RA. Nifedipine in ischemic heart disease. Circulation 1995;92:10748.
[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-dependent processes. J Hypertens 2014;32:18192.
[97] Nishikawa M, Komada F, Morita K, Deguchi K, Shirakawa S. Inhibition of platelet
aggregation by the cAMP-phosphodiesterase inhibitor, cilostamide, may not be
associated with activation of cAMP-dependent protein kinase. Cell Signal
1992;4:45363.

You might also like