Вы находитесь на странице: 1из 9

Vascular Pharmacology 59 (2013) 6775

Contents lists available at ScienceDirect

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

Review

Antiplatelet properties of natural products


Gemma Vilahur a,, Lina Badimon a,b
a
b

Centro de Investigacin Cardiovascular, CSIC-ICCC, Hospital de la Santa Creu i Sant Pau and IIB-Santpau, Spain
Ctedra de Investigacin Cardiovascular, UAB-HSCSP-Fundacin Jess Serra, Barcelona, Spain

a r t i c l e

i n f o

Article history:
Received 16 August 2013
Received in revised form 19 August 2013
Accepted 20 August 2013
Chemical compounds studied in this article:
Arachidonic acid (PubMed CID: 444899)
Thromboxane (PubChem CID: 114873)
Adenosine diphosphate (PubChem CID: 6022)
Omega-3 fatty acids (PubChem CID: 56842239)
Onion (PubChem CID: 53472027)
Garlic (PubChem CID: 6850761)
Lycopene (PubChem CID: 446925)
Resveratrol (PubChem CID: 445154)
Flavonol (PubChem CID: 11349)
Ethanol (PubChem CID: 702)

a b s t r a c t
Cardiovascular diseases (CVD) and its main underlying cause, atherothrombosis, are the major culprits of morbidity
and mortality worldwide. Apart from the treatment of cardiovascular risk factors and the use of antithrombotic
agents there is considerable interest in the role of natural food products and their bioactive components in the prevention and treatment of cardiovascular disorders. The consumption of healthy diets rich in functional foods, such as
the Mediterranean diet, has shown to exert profound cardioprotective effects in the primary and secondary prevention of CVD. Moreover, accumulating data have attributed these benecial effects, at least in part, to the modulation
of key players in the pathogenesis of atherosclerosis, including amelioration in the lipid prole and vascular function
and a decrease in oxidative stress and inammation. Although with a much less clear picture, natural dietary compounds have also demonstrated to exert antiplatelet activities, further contributing to reduce the thrombotic risk.
This article provides a brief overview of the atherothrombotic process to further provide an up-to-date review of
the antiplatelet properties exerted by natural products and/or food-derived bioactive constituents including -3
PUFA, olive oil, garlic and onions, tomatoes, mushrooms, polyphenol-rich beverages, and avonol-rich cocoa as
well as to describe the mechanisms underlying these antiplatelet activities.
2013 Elsevier Inc. All rights reserved.

Keywords:
Atherothrombosis
Healthy diet
Natural foods
Antiplatelet activity

Contents
1.
2.

Introduction . . . . . . . . . . . . . . . . . .
Atherothrombosis . . . . . . . . . . . . . . .
2.1.
Atherosclerosis . . . . . . . . . . . . .
2.2.
Role of platelets in thrombus formation . .
3.
Antiplatelet properties of natural products . . . .
3.1.
-3 polyunsaturated fatty acids (PUFA) . .
3.2.
Olive oil . . . . . . . . . . . . . . . .
3.3.
Onion and garlic (ajoene) . . . . . . . . .
3.4.
Tomatoes . . . . . . . . . . . . . . . .
3.5.
Mushrooms . . . . . . . . . . . . . . .
3.6.
Polyphenol-rich beverages (wine and beer)
3.7.
Flavonol-rich cocoa . . . . . . . . . . .
4.
Summary and conclusions . . . . . . . . . . . .
Acknowledgments . . . . . . . . . . . . . . . . . .
References . . . . . . . . . . . . . . . . . . . . .

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

Corresponding author at: Cardiovascular Research Center, C/Sant Antoni M Claret 167, 08025 Barcelona, Spain. Tel.: +34 93 5537100; fax: +34 93 556 55 59.
E-mail address: gvilahur@csic-iccc.org (G. Vilahur).
1537-1891/$ see front matter 2013 Elsevier Inc. All rights reserved.
http://dx.doi.org/10.1016/j.vph.2013.08.002

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

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

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

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

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

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

68
68
68
68
71
71
71
71
72
72
72
73
73
73
73

68

G. Vilahur, L. Badimon / Vascular Pharmacology 59 (2013) 6775

1. Introduction
According to a World Health Organization fact sheet (EURO/03/06)
CVD is the number one killer in Europe, with heart disease and stroke
being the major cause of death in all 56 member states. Atherosclerosis
and its thrombotic complications (i.e., atherothrombosis) is the major
underlying cause of CVD [1]. Despite all the improved knowledge
on the etiopathogenesis and treatment of atherothrombosis, it is estimated that the socio-economic impact of this disease will increase,
rather than decrease, in the near future. It is predicted that coronary
heart disease will be the dominant cause of mortality worldwide by
2020.
The pioneering contributions from the Framingham Heart Study
launched in 1948 brought into mind the connection between cardiovascular risk factors (especially hypercholesterolemia) and CVD [2].
However, it was not until the 1960s when the Seven Countries study
established the rst link between dietary patterns and CVD [3]. In this
seminal study, Ancel Keys and colleagues evidenced that regular dietary
saturated fat intake was signicantly associated with serum cholesterol
levels and the risk of coronary heart disease [3]. Moreover, the outcome
of the study brought up the concept of the cardioprotective properties
of the dietary habits within the Mediterranean regions (low in saturated fat, rich in monounsaturated and -3 polyunsaturated fatty
acids and abundant in fruits and vegetables as well as a moderate
wine consumption) in comparison with other regions of the Western
world (high in saturated fats and simple sugars and low in -3 polyunsaturated fatty acids, bers and antioxidants) [4]. This initial observation was further supported by many other epidemiological studies
and several intervention trials. Among these, the recent PREDIMED
(Prevencin con Dieta Mediterranea) trial [5] and the Lyon Diet Heart
Study [6], which have robustly demonstrated the protective effects associated to the adherence of a Mediterranean type of diet in primary
and secondary prevention of CVD, respectively, deserve special attention. Recent analyses suggest that, together with regular physical activity and smoking cessation, over 80% of coronary heart disease and 70%
of stroke could be avoided by healthy food choices that are consistent
with the traditional Mediterranean Diet [7]. These studies have also
raised an enormous interest in identifying the food components and
the mechanisms involved in the protection against CVD. So far, compiling data from experimental, epidemiological and clinical studies have
supported that dietary Mediterranean components provide cardiovascular health benets, at least in part, by modulating key players in the
pathogenesis of atherosclerosis (e.g., amelioration in the lipid prole,
improvement of vascular function, oxidative stress diminishment
and anti-inammatory properties) [4,8]. In addition, although a much
less clear picture exists as to their antithrombotic potential, several
Mediterranean-related foods have also demonstrated to exert antiplatelet
activities, leading to a lesser pro-atherothrombotic prole.
In this mini-review we will rst provide a brief overview of the
atherothrombotic process in order to further outline some relevant
functional foods (whole foods and their bioactive ingredients) which
have been shown to modulate platelet function, and then describe the
mechanisms by which this is thought to occur.
2. Atherothrombosis
2.1. Atherosclerosis
Atherosclerosis is an inammatory disease that involves the arterial
wall, and is characterized by the progressive accumulation of lipids and
inammatory cells within the intima of large arteries [9,10]. The rst
step is the internalization of lipids (low density lipoproteins or LDLs)
in the intima, with the concomitant endothelial activation/dysfunction
(Fig. 1). The vascular endothelium is a semi-permeable barrier that
controls the diffusion of plasma molecules and regulates vascular
tone, inammation and prevents thrombus formation [11,12].

However, dysfunctional/activated endothelium is characterized by


the loss of such properties (Fig. 1). Consequently, LDL particles further inltrate and accumulate in the extracellular matrix (ECM); circulating monocytes are then recruited and attach through the exposure of
endothelial adhesion molecules. Once attached, they transmigrate into
the subendothelial space where they transform into macrophages.
Moreover, the injury in the endothelial-related antithrombotic properties facilitates platelet adhesion and activation in the dysfunctional area.
Adhered platelets, in concert with dysfunctional endothelial cells, secrete chemotactic and growth factors which stimulate migration, accumulation and proliferation of vascular smooth muscle cells (VSMC) and
leukocytes in the intimal layer, promoting plaque progression (Fig. 1)
[13,14]. LDLs retained in the ECM mainly by proteoglycans become
targets for oxidative and enzymatic modications. Oxidized LDLs enhance a series of pro-inammatory reactions via different mediators
(TNF-alpha, IL-1, MCSF, etc), perpetuating the activation, recruitment
and transmigration of monocytes and other inammatory cells across
the endothelial layer into the intima. On the other hand, the attracted
macrophages scavenge modied/oxidized LDLs, become lipid-laden,
and convert into foam cells (Fig. 1) [9]. In the early stages of atherosclerosis, the accumulation of foam cells evolves into fatty streak. Lesion
complication occurs when foam cells release growth factors and cytokines, which further stimulate VSMC migration from the media
into the intima where they divide and produce ECM components
such as collagen, and contribute to the formation of a brous cap
(Fig. 1) [15]. If the pathological triggering process persists and macrophages fail to remove accumulated cholesterol from the vessel,
they become apoptotic, releasing cholesterol to the vessel wall and,
more importantly, prothrombotic molecules (e.g., tissue factor) and
metalloproteinases (enzymes able to digest the ECM scaffold) [16].
Progression and complication of atherosclerotic plaques are also
characterized by a decreased number of VSMCs as well as the formation of immature and leaky new vessels, making atherosclerotic lesions more prone to rupture (the so-called vulnerable plaques)
[8]. Indeed, human atherectomy specimens and necropsy studies
have revealed that the main intrinsic features that characterize
plaques as vulnerable are large necrotic lipid core, increased inammatory content, reduced collagen and VSMC amount, and angiogenesis [8,17,18]. Plaque disruption and the subsequent exposure of
thrombogenic substrates initiate both platelet adhesion/activation
and aggregation on the exposed vascular surface and the activation
of the coagulation cascade, leading to thrombus formation and the
clinical manifestations of the atherosclerotic disease, acute myocardial infarction or sudden death (Fig. 1) [17].
2.2. Role of platelets in thrombus formation
Platelets, cytoplasmic fragments (25 m) originated from bonemarrow megakaryocytes, circulate within the vascular tree without
signicant interaction with the vessel wall. The endothelium inhibits
platelet reactivity by producing several local active substances, including nitric oxide (NO) and prostacyclin (prostaglandin I2; PGI2)
as well as by the presence on its surface of an ecto-ADPase (CD39)
and thrombomodulin. The endothelium also blocks the activation
of the coagulation cascade by expressing tissue factor pathway inhibitor (TFPi) and enhances brinolysis (clot dissolution) via activation of tissue plasminogen activator (tPA) (Fig. 1). However, the
clustering of cardiovascular risk factors, among which high-LDL
levels play a critical role, results in the aforementioned endothelial
dysfunction, characterized by a decrease in NO bioavailability [19].
NO is produced through the conversion of L-arginine to L-citrulline
by the enzyme endothelial NO synthase (eNOS). NO then diffuses
to the underlying vascular smooth muscle cells or circulating platelets to stimulate the production of cyclic guanosine monophosphate
(cGMP) through the activation of the enzyme guanylate cyclase
(GC)(Fig. 2). GC activation ultimately induces the activation of

G. Vilahur, L. Badimon / Vascular Pharmacology 59 (2013) 6775

69

Dysfunctional endothelium
Blood
coagulation

Platelet activation

Thrombin

Fibrinolysis

TF-FVIIa
PGI2
TFPi

TPa

Heparin sulphate
proteoglycan

NO

Thrombomodulin

Cardiovascular Risk Factors


(e.g., hypercholesterolemia)

Platelet
activation/aggregation
CXCL-2

LDL

Monocytes

MCP-1

Platelets
CXCL-2

Coagulation
cascade

vWF
MCP-1
GPIb
I-CAM

TF

V-CAM

LDL

Intima

ECM

MCP-1

Scavenger
receptors

Foam Cell

Dying
macrophage

Colony
Stimulating
factor

oxLDL
Macrophage

Media

ROS

Lipid-ladden VSMC

VSMC

Fig. 1. The following gure illustrates the complexity of events that drive the atherosclerotic process and further thrombotic complications. The rst step in atherosclerosis is the internalization of lipids in the intima that induces endothelial secretion of chemotactic substances and the expression of adhesion receptors, in turn favoring monocyte recruitment, adhesion, and
transmigration into the arterial wall. Once there, macrophages accumulate lipids, leading to foam cell formation. Foam cells aggravate the atherogenic process by releasing growth factors,
cytokines, metalloproteinases and reactive oxygen species, all of which perpetuate and amplify the vascular remodeling process and activate VSMC migration. On the other hand, the disruption of endothelial-related antithrombotic properties in concurrence with vascular damage induces platelet activation. MCP-1: monocyte chemoattractant protein-1; LDL: low density
lipoproteins; CXCL-2: Chemokine (C-X-C motif) ligand 2; vWF: Von Willebrand factor; V-CAM: Vascular cell adhesion molecule; E-CAM: endothelial cell adhesion molecule; mLDL:
modied LDL; TF: tissue factor; ROS: reactive oxygen species; TPa: tissue plasminogen activator; TFPi: tissue factor pathway inhibitor; PGI2: prostacyclin; NO: nitric oxide.

cGMP and/or cAMP-related kinases and the subsequent vasodilation


and/or inhibition of platelet aggregation, respectively [20]. In fact,
enhanced cAMP has shown to induce VASP-phosphorylation and
subsequent platelet inactivation (GPIIb/IIIa inactivation, Fig. 2).
Hence, the reduction in endothelial-related antithrombotic properties in concurrence with high reactive oxygen species generated by
atherosclerotic risk factors and the local increase in pro-thrombotic
and pro-inammatory mediators seem to contribute to platelet activation in the onset of atherosclerosis (Fig. 1) [21,22]. In advanced
disrupted plaques, platelet adhesion varies according to the shear
rate. Under low shear rate conditions platelet attachment mainly occurs through the collagen receptor that directly binds to the platelet
receptor GPIa/IIa (integrin II1) (Fig. 2). To a lesser extent, bronectin, laminin, vitronectin and thrombospondin also contribute to
platelet adhesion by binding to GPIc-IIa, GP Ic-IIa, vitronectin receptors, and to GPIV, respectively. Thereafter, the binding of platelet
GPVI receptor to collagen promotes rm platelet adhesion and mediates platelet activation and aggregation [23]. At high shear rates
platelet adhesion is mainly driven by the interaction of circulating
vWF (via its A3 domain) with exposed collagen. The binding of vWF
to collagen allows vWF A1 domain interaction with the platelet GPIb
component of the GPIb/IX/V platelet receptor complex (Fig. 2) [24].

The multimeric nature of vWF increases the local amount of active


A1 domain sites, thus reinforcing platelet-vessel wall interaction.
However, the vWF-GPIb/V/IX is known to be characterized by a fast
dissociation rate, and thus, formed bonds cannot provide stable arrest of platelets on the subendothelial matrix [25]. Again, platelet
GPVI receptor, by binding directly to collagen, induces platelet rm
adhesion and further activation and aggregation. As such, it promotes the activation of GPIIb/IIIa (integrin IIb3) and GPIa/IIa.
Both GPIIb/IIIa and GPIa/IIa act in concert to promote subsequent
rm, irreversible and stable platelet arrest on the endothelial surface, by direct binding both to collagen (GPIa/IIa) and to the vWF
C1 domain (GPIIb/IIIa; Fig. 2) [2527].
P-selectin has also been shown to mediate platelet adhesion to the
damaged vessel. P-selectin is localized in the -granules of platelets
and the WeibelPalade bodies of endothelial cells, and upon cell activation, it is translocated to the cell surface [28]. There are many putative Pselectin glycoprotein ligands that are either expressed on platelets
(sulfatides, GPIb, P-selectin glycoprotein ligand-1/PSGL-1), endothelial
cells (glycolysated cell adhesion molecule-1/GlyCAM-1, CD34, mucosal
cell adhesion molecule-1/MAdCAM-1) or leukocytes (PSGL-1) [29].
Besides platelet-collagen interaction, circulating agents such as epinephrine, serotonin, adenosine diphosphate (ADP; released from lysed

70

G. Vilahur, L. Badimon / Vascular Pharmacology 59 (2013) 6775

cGMP
PLC

AC

DAG I3P

cAMP

NITRIC
OXIDE

AMP

GC

PKA

GMP

inhibition

PKG

activation
5-HT2

SEROTONIN
Ca2+

Ca2+

VASP

-3 PUFAs
TOMATOES?
RED WINE
COCOA
TOMATOES
MUSHROOMS

GPIIb/IIIa

GARLIC
ONIONS
MUSHROOMS
RED WINE

GPIIb/IIIa

vWF

Ca2+

VASP-P

A1 A3

GARLIC (ajoene)

RGD

P2X1
PAR-1/PAR-4
DAG

EPINEPHRIN

COCOA

ATP

PKC

Fibrinogen

GARLIC
(ajoene)

PIP2

GPIIb/IIIa

PLC

OLIVE OIL
TOMATOES
MUSHROOMS
RED WINE
COCOA

VASP-P

P2Y12

PLATELET
GRANULES

AC

PLC

IP3

VASP

GPIIb/IIIa

VITRONECTIN

GPIIb/IIIa

RED WINE

PKA

Ca2+
Ca2+

cAMP

Ca2+

PI3K
MAPK

ALA?

ADP
vWF
Fgibrinogen

PGH2
PGG2

Ca2+

MAPK

TP

GARLIC
ONIONS
COCOA

TXA2
PLA2

ATP
ADP

GPIb-IX-V
vWF

PLATETL
DEGRANULATION

GPIa/IIa

AA

GPVI

-3 PUFAs
OLIVE OIL
GARLIC

COLLAGEN
A1

OLIVE OIL?

A3

RGD

RGD

vWF

COLLAGEN

OLIVE OIL
TOMATOES
MUSHROOMS
RED WINE

Fig. 2. Key agonists, their receptors and triggering signaling pathways involved in platelet activation and subsequent aggregation. In addition, the mechanisms by which natural food compounds exert antiplatelet effects are detailed. VWF: Von Willebrand factor; GP: glycoprotein; Fg: brinogen; AA: arachidonic acid; TXA2: thromboxane-A2; TP: thromboxane receptor;
ADP: adenosine diphosphate; PAR: proteinase activated receptor; TF: tissue factor, ALA: -linoleic acid.

erythrocytes at the site of injury) and thrombin (generated upon


atherosclerotic plaque exposure of tissue factor) may also activate
platelets via specic platelet surface receptors (Fig. 2). Once activated,
platelets undergo shape change, ensuing calcium translocation within
the platelet and the subsequent release of the platelet granule components (i.e., platelet degranulation). Platelet granule secretion leads
to the local release of ADP/ATP, serotonin, Ca2 +, adhesion proteins
(e.g., brinogen, bronectin, vWF, thrombospondin, vitronectin, Pselectin, GPIIb/IIIa) and coagulation factors (e.g., factor V, factor XI,
plasminogen activator inhibitor type 1, plasminogen, protein S), all of
which contribute to perpetuate and amplify the thrombotic response.
ADP released from platelet granules may exert both an autocrine effect,
promoting stable platelet aggregation by interacting with specic ADP
receptors in the membrane (P2Y1 and P2Y12; Fig. 2), and a paracrine
effect, by binding to ADP-receptors of neighboring platelets, thus
amplifying the activation process. Activated platelets are also

characterized by the externalization of phosphatidylserine in the


cell membrane, becoming a substrate for coagulation cofactor/
enzyme complexes (VIIa/IXa and Va/Xa), and thereby driving
procoagulant reactions and the eventual thrombin generation [30].
However, platelets are not only stores of several bioactive molecules,
but also generate lipid mediators, such as thromboxane A2 (TXA2).
Platelet activation induces phospholipase-A2 (PLA2) activation that
triggers arachidonic acid metabolism. Platelet cyclooxygenase 1 (COX1) catalyzes the conversion of arachidonic acid to prostaglandin G2/H2,
and the latter is converted to TXA2 (Fig. 2). TXA2 is released into the
bloodstream, where it binds to TX receptors (TP) present on the surface
of adjacent platelets were it activates phospholipase C (PLC),
resulting in the production of diacylglycerol (DAG) and inositol trisphosphate (IP3). DAG and IP3 activate protein kinase C and mobilize
cytoplasmic Ca2 +, respectively (Fig. 2) [31]. Platelets also possess
mitogen activated protein (MAP) kinases [extracellular signal-

G. Vilahur, L. Badimon / Vascular Pharmacology 59 (2013) 6775

regulated kinase 2 (ERK2), p38MAPK and c-Jun NH2-terminal kinase


1 (JNK1)], which are activated by different agonists [32]. Reports indicated that ERK2 plays a role in ADP-induced TXA2 generation [33],
whereas JNK1 and p38 MAPK are involved in collagen-inducedplatelet aggregation (Fig. 2) [34].
Regardless of the trigger, platelet aggregation is regulated in the nal
part of the pathway by activation of the platelet heterodimer GPIIb/IIIa
receptor, the most abundant protein on the platelet surface. The activation of this receptor is calcium-dependent and requires a conformational change in the 2 subunits, such that a new binding domain is
exposed. Fibrinogen (of plasma or platelet origin) is the main ligand
for the GPIIb/IIIa receptor (Fig. 2). The dimeric structure of brinogen allows its interaction with 2 platelets at the same time, thereby favoring
platelet aggregation. Hence, thrombus grows as active GPIIb/IIIa captures
new platelets by means of brinogen (Fig. 2). Other ligands for GPIIb/IIIa
that participate in platelet aggregation (although to a lesser extent)
include vWF, bronectin, and vitronectin (Fig. 2). Fibrinogen binding
to GPIIb/IIIa also triggers an in-side out signaling, causing amplication of the initial signal and further platelet activation. This leads to
further aggregation of platelets and accumulation at the site of vessel
injury; thrombus formation. In the nal phase of thrombus formation,
brinogen is converted to brin by thrombin, leading to the stabilization of the platelet aggregates, with more platelets and blood cells (leukocytes and red blood cells) thus getting trapped and contributing to
thrombus growth.
3. Antiplatelet properties of natural products
Within the following section we will provide an overview of the
antiplatelet properties exerted by natural products and/or foodderived bioactive constituents, and will describe the mechanisms
by which their antiplatelet activity may contribute to reduce the
thrombotic risk (Fig. 2).
3.1. -3 polyunsaturated fatty acids (PUFA)
Ever since a very low incidence of coronary heart disease in
Greenland Eskimos was reported, which was believed to be related to
the high intake of seafood containing -3 PUFAs [35], many large
scale studies have extensively characterized the cardioprotective effects
of sh-derived dietary -3 fatty polyunsaturated fatty acids (PUFA)
eicosapentaenoic acid (EPA; C20:5 n-3) and docosahexaenoic acid
(DHA; C22:6 n-3) [4,36]. Indeed each 20 g per day increase in sh intake has been associated with a 7% lower risk of coronary heart disease
mortality [37]. The exact mechanisms by which sh-derived -3 PUFAs
perform such protective functions are still into debate, but the main
mechanisms proposed include amelioration in the lipid prole (mainly
lowering plasma triglyceride levels), anti-inammatory responses
(decrease NF-B activation and cytokine/chemokine synthesis and
release), plaque stabilization (lower monocyte inltration) and decreased platelet aggregation [4,3841]. The antithrombotic properties of sh-derived -3 PUFAs have been well-characterized and
have been mainly attributed to the incorporation of -3 PUFAs into
the phospholipid membrane, altering the arachidonic acid metabolism
and subsequent reduction in TXA2 release (Fig. 2). As such, -3 PUFA
compete with arachidonic acid (-6 PUFA) as a substrate for COX enzyme promoting the synthesis of platelet biologically inactive eicosanoids (TXA3) rather than -6-derived platelet activator eicocainoids
(TXA2) [41]. Hence, a dominant incorporation of -3 PUFA in the cell
membrane phospholipids ultimately shifts eicosanoid production equilibrium toward an anti-thrombotic prole. Alternatively, -3 PUFA
have also demonstrated to ameliorate vascular function and enhance
NO release, thereby modulating platelet function (Fig. 2) [42]. Linolenic acid (ALA), another -3 PUFA found in vegetable oils, has
also shown to exert antiplatelet effects. Following oral intake, ALA is
partially converted (10%) into EPA and DHA, questioning whether ALA

71

conversion to EPA and DHA accounts for the detected benecial effects
or whether ALA directly exerts its own biological effects [43]. Nevertheless, an elegant recent study by Holy and colleagues has demonstrated
that ALA by itself is capable of impairing in vivo arterial thrombus formation, tissue factor expression, and platelet activation via p38MAPK
blockade (Fig. 2) [44].
3.2. Olive oil
Olive oil is the principal component of Mediterranean diet, both
by its predominance as the main energy source and its abundance
in health-promoting components [8,33]. The protective role of olive
oil against CVD can be seen in multiple benecial effects, including
an improvement in glucose metabolism and lipid prole, blood pressure
control, amelioration in endothelial function, a reduction of LDL
oxidizability, and decreased systemic inammation [33]. In addition,
olive oil has shown to ameliorate the prothrombotic environment by
favorably inuencing coagulation factors (tissue factor, TFPI, FVII,
and FXII), brinolysis (PAI-1) and platelet function [33]. We have recently reported, in asymptomatic subjects with high cardiovascular
risk, that intake (for 3 months) of traditional Mediterranean diet
supplemented with virgin olive oil actively modulates the expression
of key genes involved in vascular inammation, foam cell formation,
and thrombosis towards an anti-atherothrombotic prole [45].
With regard to platelet function, olive oil has been shown to partially
inhibit ADP-, collagen- and arachidonic acid- induced platelet aggregation, resulting in lower TXA2 release (Fig. 2) [46,47,33]. The protective properties of olive oil have been often attributed to its high
content in -9 monounsaturated fatty acids (MUFA; 7085% content), mainly in the form of oleic acid. However, olive oil, unlike
other vegetable oils, also contains high amounts of several micronutrient
constituents (e.g., phenolic compounds, such as hydroxytyrosol and
oleuropein) that have demonstrated important bioactive functions. In
fact, Karantonis and colleagues found, when comparing different sources
of oleic acid (olive oil, soya, maize, sunower and sesame oils), a
higher antiaggregating effect in the olive oil sample, supporting the
capacity of this minor components to exert antiplatelet effects [48].
In effect, olive oil phenolic compounds are thought to be responsible
for the detected reduction in TXA2 release. Finally, although controversial, some reports have been attributed to olive oil about the potential to reduce circulating vWF levels, likely impairing platelet
adhesion and aggregation (Fig. 2). Yet, more studies are needed to
support this hypothesis [49,50].
3.3. Onion and garlic (ajoene)
Plants of the genus Allium such as onion (Allium cepa) and garlic
(Allium sativum) have shown to exert multiple benecial cardiovascular effects, including lowering of blood pressure, decreasing cholesterol and triglyceride levels, and enhancement of brinolysis
and antiplatelet activities [5153]. Both raw garlic and onion have
been reported to inhibit platelet aggregation, however boiled preparations have shown little or no antiplatelet effect [54,55]. So far, several trials have demonstrated a profound effect of garlic to reduce the
ability of platelets to aggregate. Concretely, six trials have assessed
the effectiveness of garlic on platelet aggregation; of these, ve reported positive observations (decrease in platelet aggregation)
[5660] and one reported no effect [61]. No effects were observed
in brinogen levels, and inconclusive data were obtained for the brinolytic activity. Garlic and onion compounds have also shown to
exert potent inhibitory effects over TX production, likely as a result
of direct non-competitive inhibition with the COX enzyme (Fig. 2)
[54]. A study examining the effect of consuming a clove of fresh garlic
on platelet TX production reported a marked 80% reduction in serum
TXA2 levels after 26 weeks [54]. Although onions also decrease TXA2
synthesis [62] their antiplatelet activity is 13 times lower than that of

72

G. Vilahur, L. Badimon / Vascular Pharmacology 59 (2013) 6775

garlic. As such, a study in rabbits reported that, while raw garlic extract
reduced serum TXB2 in rabbits, onion extract at an equivalent dose did
not [54].
Several compounds isolated from Allium species have been identied as inhibitors of platelet aggregation. These include allicin, parafnic
sulphide, and adenosine. Most of these compounds have in common a
high sulfur content. In fact, the antiplatelet activity of onions is sulfurdependent (lower sulfur content results in lower antiplatelet activity)
[63]. Allicin, and more specically, ajoene, a self-condensation product
of allicin, is thought to be the primary substance responsible for the inhibitory effect of garlic on platelet aggregation. We have reported that
ajoene inhibits the binding of brinogen and vWF to the GPIIb/IIIa receptor without affecting the binding of vWF to GPIb (Fig. 2) [64]. Ajoene
seems to diminish the number of functional GPIIb/IIIa receptor sites on
the platelet membrane [65] by reducing the viscosity in the inner part of
the lipid bilayer rather than affecting brinogen afnity to GPIIb/IIIa
receptor or GPIIb/IIIa receptor conformation [66]. The inhibitory effect of ajoene on the binding to GPIIb/IIIa supports the ndings that
ajoene inhibits the aggregation of platelets induced by all known agonists [52]. In addition, ajoene has also shown to reduce TXA2 production by altering arachidonic acid metabolism (Fig. 2) [61,66].
A parafnic sulphide component of Allium, dyallyl trisulde, has
also shown to exhibit antiplatelet functions [61]. Studies have indicated that this polysulde inhibits platelet aggregation and Ca2 +
mobilization induced by thrombin in a concentration-dependent
manner, as well as TXA2 synthesis without affecting inositol-1,4,5triphosphate formation [67].

family of mushrooms to exert anti-platelet activities by interfering


with different platelet receptors, as well as diverse signaling pathways [77]. In 2004, a methanol extract of Pleurotus orida Eger (an
edible and commercially grown mushroom) was demonstrated to almost abolish ADP-induced platelet aggregation (95% reduction) after
a 5 min incubation in washed-human platelets [77]. On the other
hand, a dry extract of Stereum hirsutum demonstrated to partly diminish platelet activation by blocking thrombin active site (Fig. 2)
[78] and an extract from Hericium erinaceus (i.e., hericenone B) to selectively impede collagen signaling from GPIIb/Ia [79]. Additionally,
recent studies by Kamruzzaman and colleagues have provided detailed insights as to the mechanisms by which certain medicinal
mushrooms may provide antiplatelet protection. They have reported
the capability of Phellinus baummii (a mushroom under the family of
Hymenochaetaceae basidiomycetes) [80] and P-terphenyl curtisian
e (an antioxidant component of the wild mushroom Paxillus curtissi)
to inhibit collagen-, thrombin- and ADPinduced platelet aggregation, suppress ATP secretion, cytoplasmic Ca2 + mobilization and brinogen binding. Interestingly, the Authors have also provide
evidence that P-terphenyl curtisian E suppresses collagen-induced
platelet GIIb/IIIa activation by interfering with the three known
platelet MAPKs (JNK, ERK2, and p38) and Akt signaling, and by
upregulating cAMP and subsequent PKA activation and VASPphosphorylation (Fig. 2) [81]. These later observations, published in
the current issue of Vascular Pharmacology, support the notion of the
power of mushrooms in interfering with platelet activation via multiple
potential targets.

3.4. Tomatoes
3.6. Polyphenol-rich beverages (wine and beer)
Both fresh and processed tomatoes (Solanum lycopersicum) have
been associated with a decrease in the prevalence of CVD through a
reduction in lipid levels, antioxidant and antiplatelet activities [68].
Tomato has shown antiaggregating activity by inhibiting ADP- and
collagen-, but not to arachidonic acid-induced platelet aggregation
in healthy subjects 3 h after ingestion [69]. The mechanism of action
by which tomatoes inhibits platelet aggregation has yet to be elucidated.
It has been suggested that adenosine and other nucleosides may be responsible for this inhibition by interacting with three platelet receptors GPVI, P2Y1 and P2Y12 (ADP-receptors; Fig. 2) [70]. As such,
bioactive tomato compounds have been suggested to interact with
intraplatelet signaling pathways downstream the activation of
these receptors, likely blocking PLC, cAMP and Akt/VASP phosphorylation. However, it remains to be identied both the tomato components possessing the antiplatelet activity (skin, pulp, seeds, etc) and
their bioactivity. Interestingly, it has been recently reported that
both green and red tomato pulp exert similar antiplatelet activity
triggered by ADP despite green tomato pulp being devoid of lycopene [71], the main natural carotenoid antioxidant present in tomatoes. Other two studies further agreed that lycopene does not seem
to contribute to modulate platelet function [72,73]. However, in contrast to these observations, Hsiao and colleagues [74] demonstrated,
in vitro and in vivo, that lycopene does exert antiplatelet activity
which may involve PLC inhibition followed by inhibition of intracellular Ca+ 2 mobilization. Moreover, lycopene also showed to activate
the formation of NO-derived cyclic GMP, resulting in the inhibition of
platelet aggregation (Fig. 2) [74].
3.5. Mushrooms
Mushrooms are an important natural source of foods and biologically
active compounds. A wide range of activities, including antitumor, antimicrobial, neuroprotective, and cardiovascular benecial (antioxidant, antihypertensive, antidiabetic, and anti-inammatory) activities have been
reported in mushrooms and fungi [75,76]. In addition, in the last
ten years several reports have revealed the capability of certain

A large number of studies have consistently found that moderate


alcohol consumption (1 to 3 drinks/day) is associated with a decreased risk of CVD [8287]. Some of the protective effects appeared
to be linked to ethanol per se; however, several studies have indicated, when comparing different beverage types, that red wine seems to
exert more protective effects than other forms of alcohol, supporting
a protective role for polyphenolic compounds [83,8589]. The benecial effects derived from polyphenols appear to be mediated via a
plethora of biochemical pathways and signaling mechanisms, acting
either independently or synergistically. Polyphenols have shown to
modulate inammation, lipid metabolism, improve antioxidant status and endothelial function, increase NO release, and protect
against platelet aggregation [33,90]. Collectively, there is emerging
evidence that resveratrol (the main polyphenol of wine found in
grape skin and seeds) has antithrombotic effects that appear to be
the result of reduced susceptibility to platelet activation and aggregation, reduced synthesis of prothrombotic mediators (eicosainoid
synthesis) and decrease gene expression of tissue factor [91,92]. Indeed,
resveratrol has shown to inhibit, in a concentration-dependent manner,
platelet aggregation induced by collagen, ADP, and thrombin (Fig. 2)
[93]. It has also demonstrated to attenuate TP-induced platelet activation by reducing PLC activity [94] and inhibit cAMP and cGMP phosphodiesterases [95], thereby increasing platelet levels of cAMP and
cGMP and the subsequent decrease in intracellular Ca2+ levels reducing
platelet activation (Fig. 2). Moreover, resveratrol, by activating eNOS
and/or inhibiting the generation of reactive oxygen species, has demonstrated to modulate the production of endothelial-related NO further
reducing platelet reactivity [96,97]. In addition, we have evidenced,
in an ex vivo perfusion experimental porcine model (the Badimon
perfusion chamber), that moderate daily intake of red wine inhibited
mural thrombosis. Such antithrombotic effects were associated with
inhibition of RhoA translocation to the platelet membrane (active
site) supporting platelet shape change regulation via calciumindependent pathways and a marked reduction in monocyte tissue
factor expression [98].

G. Vilahur, L. Badimon / Vascular Pharmacology 59 (2013) 6775

3.7. Flavonol-rich cocoa


Short-term studies have evidenced a cardioprotective effect of cocoa
and chocolate [99]. Cocoa and dark-chocolate are suggested to have
antioxidant, anti-inammatory, and antihypertensive effects, as
well as to improve vascular function and diminish platelet reactivity
[100,101]. Many benecial effects of chocolate consumption are mediated, at least in part, through naturally occurring phenolic compounds [88]. Indeed, chocolate is a rich source of polyphenolic
compounds (e.g., 41 g of chocolate contain nearly as much phenol
as 140 mL of red wine). Major phenolic compounds include quercetin, epicatechin, procyanindin, and cocoa-red (the color component of
cocoa). Human interventional studies have reported a consistent inhibition of platelet activation and aggregation upon polyphenol-rich cocoa
or dark chocolate consumption in moderate amounts, either on a
acute or a chronic basis [102]. However, the mechanism accounting
for the inhibitory effect of this food on platelet function has not yet
been claried as well as whether their high phenolic content is mainly
responsible for such antiplatelet effects. So far, cocoa and dark chocolate have shown to prevent platelet aggregation by reducing ADP-,
adrenaline- and, epinephrine- induced GPIIb/IIIa membrane activation, ADP-induced P-selectin membrane expression and PLA2 and
COX activity (Fig. 2) [93,103]. Moreover, they have also demonstrated an increase in endothelial-related NO production [104,105].
4. Summary and conclusions
CVD is the leading cause of death in the Western world and it is
foreseen to be the major cause of death worldwide in 2020. The
major reasons for the increase include the aging of the population,
the increase of certain risk factors (obesity/diabetes) especially
among the youth, and the implementation of unhealthy life-styles.
As such, unhealthy diet is considered as a main cause of increased
atherothrombotic disease in industrialized countries. Conversely,
epidemiological evidence has supported the cardinal importance of
a well-balanced diet rich in MUFAs and PUFAs, vegetables, fruits
and polyphenol-rich beverages (i.e., the Mediterranean diet) against
the development and progression of CVD. The biological actions by
which this diet inuences CVD remain to be properly demonstrated.
Yet, multiple data have evidenced that Mediterranean functional
foods display critical anti-atherosclerotic and anti-thrombotic effects.
As such, certain foods and/or their bioactive components have proven
effective in decreasing platelet activation through a variety of mechanisms, suggesting benets in attenuating the future risk of thrombosis.
Nevertheless, most data are based on in vitro experiments and only few
are based on animal studies; for this, mechanistic effects active in
humans are sometimes conjectured or extrapolated. With a few exceptions, the number of studies in humans is limited, so that, although nutritional approaches offer interesting possibilities to assist the ght
against thrombosis, further long-term randomized controlled trials are
indicated or, in the case of natural/whole foods, cohort or good quality
casecontrol studies are much awaited. These will allow to draw denitive conclusions for providing a useful preventive approach or an adjunct to current pharmacological treatments for thrombotic diseases.
Moreover, advances in the knowledge of both platelet biology and of
the biological functions of dietary foods will provide new avenues to develop pharmaceutical and/or dietary strategies aimed at promoting cardiovascular health.
Acknowledgments
This work was supported by SAF 2012-40208 (to GV), SAF 201016549 (to LB) and RD12/0019/0026 - TerCel and RD12/0042/0027Red de Investigacin Cardiovascular from Institute Carlos III (to
LB). We thank the support received by l'Oreal-UNESCO 2012 "For
women in science", Madrid (to GV) and Fundacion de Investigacin

73

Cardiovascular-Fundacin Jesus Serra, Barcelona, GV is a recipient


of a contract from the Innovation and Science Spanish Ministry
(RyC-2009-5495, MICINN, Spain).
References
[1] www.oms.org.
[2] Dawber TR, Meadors GF, Moore Jr FE. Epidemiological approaches to heart disease:
the Framingham Study. Am J Public Health Nations Health 1951;41:27981.
[3] Keys A, Menotti A, Karvonen MJ, Aravanis C, Blackburn H, Buzina R, et al. The diet and
15-year death rate in the seven countries study. Am J Epidemiol 1986;124:90315.
[4] Massaro M, Scoditti E, Carluccio MA, De Caterina R. Nutraceuticals and prevention
of atherosclerosis: focus on omega-3 polyunsaturated fatty acids and Mediterranean diet polyphenols. Cardiovasc Ther 2010;28:e139.
[5] Estruch R, Ros E, Salas-Salvado J, Covas MI, Pharm D, Corella D, et al. Primary
prevention of cardiovascular disease with a Mediterranean diet. N Engl J Med
2013;368:127990.
[6] de Lorgeril M, Renaud S, Mamelle N, Salen P, Martin JL, Monjaud I, et al. Mediterranean alpha-linolenic acid-rich diet in secondary prevention of coronary heart disease. Lancet 1994;343:14549.
[7] Willett WC. The Mediterranean diet: science and practice. Public Health Nutr
2006;9:10510.
[8] Badimon L, Vilahur G, Padro T. Nutraceuticals and atherosclerosis: human trials.
Cardiovasc Ther 2010;28:20215.
[9] Badimon L, Storey RF, Vilahur G. Update on lipids, inammation and
atherothrombosis. Thromb Haemost 2012;105(Suppl. 1):S3442.
[10] Badimon L, Vilahur G, Padro T. Lipoproteins, platelets and atherothrombosis. Rev
Esp Cardiol 2009;62:116178.
[11] Mehta D, Malik AB. Signaling mechanisms regulating endothelial permeability.
Physiol Rev 2006;86:279367.
[12] Otsuka F, Finn AV, Yazdani SK, Nakano M, Kolodgie FD, Virmani R. The importance
of the endothelium in atherothrombosis and coronary stenting. Nat Rev Cardiol
2012;9:43953.
[13] Andrews RK, Berndt MC. Platelet physiology and thrombosis. Thromb Res
2004;114:44753.
[14] Jennings LK. Mechanisms of platelet activation: need for new strategies to protect
against platelet-mediated atherothrombosis. Thromb Haemost 2009;102:24857.
[15] Koga J, Aikawa M. Crosstalk between macrophages and smooth muscle cells in atherosclerotic vascular diseases. Vascul Pharmacol 2013;57:248.
[16] Ghosh S, Zhao B, Bie J, Song J. Macrophage cholesteryl ester mobilization and atherosclerosis. Vascul Pharmacol 2013;52:110.
[17] Fuster V, Moreno PR, Fayad ZA, Corti R, Badimon JJ. Atherothrombosis and high-risk
plaque: part I: evolving concepts. J Am Coll Cardiol 2005;46:93754.
[18] Stary H, Blankenhorn D, Chandler A. A denition of the intima of human arteries
and of its atherosclerosis-prone regions. A report from the Committee on Vascular
Lesions of the Council on Arteriosclerosis, American Heart Association. Circulation
1992;85:391405.
[19] Russo G, Leopold JA, Loscalzo J. Vasoactive substances: nitric oxide and endothelial
dysfunction in atherosclerosis. Vascul Pharmacol 2002;38:25969.
[20] Lubos E, Handy DE, Loscalzo J. Role of oxidative stress and nitric oxide in
atherothrombosis. Front Biosci 2008;13:532344.
[21] Nowak J, Murray JJ, Oates JA, FitzGerald GA. Biochemical evidence of a chronic abnormality in platelet and vascular function in healthy individuals who smoke cigarettes. Circulation 1987;76:614.
[22] Massberg S, Brand K, Gruner S, Page S, Muller E, Muller I, et al. A critical role of
platelet adhesion in the initiation of atherosclerotic lesion formation. J Exp Med
2002;196:88796.
[23] Penz S, Reininger AJ, Brandl R, Goyal P, Rabie T, Bernlochner I, et al. Human atheromatous plaques stimulate thrombus formation by activating platelet glycoprotein
VI. FASEB J 2005;19:898909.
[24] Savage B, Saldivar E, Ruggeri ZM. Initiation of platelet adhesion by arrest onto brinogen or translocation on von Willebrand factor. Cell 1996;84:65766.
[25] Massberg S, Shulz C, Gawaz M. Role of platelets in the pathophysiology of acute
coronary syndrome. Semin Vasc Med 2003;3:14761.
[26] Gibbins J, Okuma M, Farndale R, Barnes M, Watson S. Glicoprotein VI is the collagen
receptor in platelets which underlies tyrosine phosphorylation of the Fc receptor
gamma-chain. FEBS Lett 1997;413:2559.
[27] Nieswandt B, Brakebush C, Bergmeier W. Glycoprotein VI but not
alpha2beta1 integrin is essential for platelet interaction with collagen.
EMBO J 2001;20:212030.
[28] Stenberg P, McEver R, Shuman M, Jacques Y, Bainton DF. A platelet alpha granule
membrane protein (GMP-140) is expressed on the plasma membrane after activation. J Cell Biol 1985;101:8806.
[29] Merten M, Thiagarajan P. P-selectin in arterial thrombosis. Z Kardiol
2004;93:85563.
[30] Hemker HC, van Rijn JL, Rosing J, van Dieijen G, Bevers EM, Zwaal RF. Platelet membrane involvement in blood coagulation. Blood Cells 1983;9:30317.
[31] Andrews RK, Gardiner EE, Shen Y, Berndt MC. Platelet interactions in thrombosis.
IUBMB Life 2004;56:138.
[32] Adam F, Kauskot A, Rosa JP, Bryckaert M. Mitogen-activated protein kinases in
hemostasis and thrombosis. J Thromb Haemost 2008;6:200716.
[33] Delgado-Lista J, Garcia-Rios A, Perez-Martinez P, Lopez-Miranda J, Perez-Jimenez F.
Olive oil and haemostasis: platelet function, thrombogenesis and brinolysis. Curr
Pharm Des 2011;17:77885.

74

G. Vilahur, L. Badimon / Vascular Pharmacology 59 (2013) 6775

[34] Kauskot A, Adam F, Mazharian A, Ajzenberg N, Berrou E, Bonnefoy A, et al. Involvement of the mitogen-activated protein kinase c-Jun NH2-terminal kinase 1 in
thrombus formation. J Biol Chem 2007;282:319909.
[35] Bang HO, Dyerberg J, Nielsen AB. Plasma lipid and lipoprotein pattern in Greenlandic West-coast Eskimos. Lancet 1971;1:11435.
[36] Kromhout D, Giltay EJ, Geleijnse JM. n-3 fatty acids and cardiovascular events after
myocardial infarction. N Engl J Med 2010;363:201526.
[37] He K, Song Y, Daviglus ML, Liu K, Van Horn L, Dyer AR, et al. Accumulated evidence
on sh consumption and coronary heart disease mortality: a meta-analysis of cohort studies. Circulation 2004;109:270511.
[38] Serhan CN, Chiang N, Van Dyke TE. Resolving inammation: dual anti-inammatory
and pro-resolution lipid mediators. Nat Rev Immunol 2008;8:34961.
[39] Breslow JL. n-3 fatty acids and cardiovascular disease. Am J Clin Nutr
2006;83:1477S82S.
[40] De Caterina R, Madonna R, Massaro M. Effects of omega-3 fatty acids on cytokines
and adhesion molecules. Curr Atheroscler Rep 2004;6:48591.
[41] von Schacky C, Fischer S, Weber PC. Long-term effects of dietary marine omega-3
fatty acids upon plasma and cellular lipids, platelet function, and eicosanoid formation in humans. J Clin Invest 1985;76:162631.
[42] Abeywardena MY, Head RJ. Longchain n-3 polyunsaturated fatty acids and blood
vessel function. Cardiovasc Res 2001;52:36171.
[43] Harris WS. Alpha-linolenic acid: a gift from the land? Circulation 2005;111:28724.
[44] Holy EW, Forestier M, Richter EK, Akhmedov A, Leiber F, Camici GG, et al. Dietary
alpha-linolenic acid inhibits arterial thrombus formation, tissue factor expression,
and platelet activation. Arterioscler Thromb Vasc Biol 31:177280.
[45] Llorente-Cortes V, Estruch R, Mena MP, Ros E, Gonzalez MA, Fito M, et al. Effect of
Mediterranean diet on the expression of pro-atherogenic genes in a population at
high cardiovascular risk. Atherosclerosis 2010;208:44250.
[46] Sirtori CR, Tremoli E, Gatti E, Montanari G, Sirtori M, Colli S, et al. Controlled
evaluation of fat intake in the Mediterranean diet: comparative activities of
olive oil and corn oil on plasma lipids and platelets in high-risk patients. Am J
Clin Nutr 1986;44:63542.
[47] Karantonis HC, Fragopoulou E, Antonopoulou S, Rementzis J, Phenekos C,
Demopoulos CA. Effect of fast-food Mediterranean-type diet on type 2 diabetics and healthy human subjects' platelet aggregation. Diabetes Res Clin
Pract 2006;72:3341.
[48] Karantonis HC, Antonopoulou S, Demopoulos CA. Antithrombotic lipid minor constituents from vegetable oils. Comparison between olive oils and others. J Agric
Food Chem 2002;50:115060.
[49] Turpeinen AM, Mutanen M. Similar effects of diets high in oleic or linoleic acids on
coagulation and brinolytic factors in healthy humans. Nutr Metab Cardiovasc Dis
1999;9:6572.
[50] Mezzano D, Leighton F. Haemostatic cardiovascular risk factors: differential
effects of red wine and diet on healthy young. Pathophysiol Haemost Thromb
2003;33:4728.
[51] Grifths G, Trueman L, Crowther T, Thomas B, Smith B. Onions a global benet to
health. Phytother Res 2002;16:60315.
[52] Makheja AN, Bailey JM. Antiplatelet constituents of garlic and onion. Agents Actions
1990;29:3603.
[53] Rahman K, Lowe GM. Garlic and cardiovascular disease: a critical review. J Nutr
2006;136:736S40S.
[54] Ali M, Thomson M, Afzal M. Garlic and onions: their effect on eicosanoid metabolism and its clinical relevance. Prostaglandins Leukot Essent Fatty Acids
2000;62:5573.
[55] Chen JH, Chen HI, Tsai SJ, Jen CJ. Chronic consumption of raw but not boiled Welsh
onion juice inhibits rat platelet function. J Nutr 2000;130:347.
[56] Kiesewetter H, Jung F, Pindur G, Jung EM, Mrowietz C, Wenzel E. Effect of garlic on
thrombocyte aggregation, microcirculation, and other risk factors. Int J Clin
Pharmacol Ther Toxicol 1991;29:1515.
[57] Kiesewetter H, Jung F, Jung EM, Blume J, Mrowietz C, Birk A, et al. Effects of
garlic coated tablets in peripheral arterial occlusive disease. Clin Investig
1993;71:3836.
[58] Steiner M, Lin RS. Changes in platelet function and susceptibility of lipoproteins to
oxidation associated with administration of aged garlic extract. J Cardiovasc
Pharmacol 1998;31:9048.
[59] Czerny B, Samochowiec J. Klinische Untersuchunger mit einem KnoblauchLezithin-Praparat. Arztezeitschr Naturheilverf 1996;37:1269.
[60] Barrie SA, Wright JV, JE P. Effects of garlic oil on platelet aggregation, serum
lipids and blood pressure in humans. J Orthomolecular Med 1987;2:1521.
[61] Bordia A, Verma SK, Srivastava KC. Effect of garlic (Allium sativum) on blood lipids,
blood sugar, brinogen and brinolytic activity in patients with coronary artery
disease. Prostaglandins Leukot Essent Fatty Acids 1998;58:25763.
[62] Moon CH, Jung YS, Kim MH, Lee SH, Baik EJ, Park SW. Mechanism for antiplatelet
effect of onion: AA release inhibition, thromboxane A(2)synthase inhibition and
TXA(2)/PGH(2)receptor blockade. Prostaglandins Leukot Essent Fatty Acids
2000;62:27783.
[63] Goldman IL, Kopelberg M, Debaene JE, Schwartz BS. Antiplatelet activity in onion
(Allium cepa) is sulfur dependent. Thromb Haemost 1996;76:4502.
[64] Apitz-Castro R, Badimon JJ, Badimon L. Effect of ajoene, the major
antiplatelet compound from garlic, on platelet thrombus formation. Thromb
Res 1992;68:14555.
[65] Apitz-Castro R, Ledezma E, Escalante J, Jain MK. The molecular basis of the
antiplatelet action of ajoene: direct interaction with the brinogen receptor.
Biochem Biophys Res Commun 1986;141:14550.
[66] Rendu F, Daveloose D, Debouzy JC, Bourdeau N, Levy-Toledano S, Jain MK, et al.
Ajoene, the antiplatelet compound derived from garlic, specically inhibits platelet

[67]

[68]

[69]

[70]

[71]

[72]
[73]
[74]

[75]
[76]

[77]

[78]

[79]

[80]

[81]

[82]
[83]

[84]
[85]

[86]
[87]

[88]
[89]
[90]

[91]

[92]

[93]

[94]

[95]

release reaction by affecting the plasma membrane internal microviscosity.


Biochem Pharmacol 1989;38:13218.
Qi R, Liao F, Inoue K, Yatomi Y, Sato K, Ozaki Y. Inhibition by diallyl trisulde,
a garlic component, of intracellular Ca(2 +) mobilization without affecting
inositol-1,4, 5-trisphosphate (IP(3)) formation in activated platelets. Biochem
Pharmacol 2000;60:147583.
Palomo I, Fuentes E, Padro T, Badimon L. Platelets and atherogenesis: platelet antiaggregation activity and endothelial protection from tomatoes (Solanum
lycopersicum L.). Exp Ther Med 2012;23:10917.
O'Kennedy N, Crosbie L, Whelan S, Luther V, Horgan G, Broom JI, et al. Effects of tomato extract on platelet function: a double-blinded crossover study in healthy
humans. Am J Clin Nutr 2006;84:5619.
Fuentes EJ, Astudillo LA, Gutierrez MI, Contreras SO, Bustamante LO, Rubio PI, et al.
Fractions of aqueous and methanolic extracts from tomato (Solanum lycopersicum L.)
present platelet antiaggregant activity. Blood Coagul Fibrinolysis 2013;2013:867578.
Fuentes E, Carle R, Astudillo L, Guzman L, Gutierrez M, Carrasco G, et al. Antioxidant
and antiplatelet activities in extracts from green and fully ripe tomato fruits
(Solanum lycopersicum) and pomace from industrial tomato processing. Evid
Based Complement Altern Med 2013;867578.
Phang M, Lazarus S, Wood LG, Garg M. Diet and thrombosis risk: nutrients for prevention of thrombotic disease. Semin Thromb Hemost 2011;37:199208.
Yamamoto J, Taka T, Yamada K, Ijiri Y, Murakami M, Hirata Y, et al. Tomatoes have
natural anti-thrombotic effects. Br J Nutr 2003;90:10318.
Hsiao G, Wang Y, Tzu NH, Fong TH, Shen MY, Lin KH, et al. Inhibitory effects of lycopene on in vitro platelet activation and in vivo prevention of thrombus formation. J Lab Clin Med 2005;146:21626.
Wasser SP. Medicinal mushrooms as a source of antitumor and immunomodulating
polysaccharides. Appl Microbiol Biotechnol 2002;60:25874.
Mahajna J, Dotan N, Zaidman BZ, Petrova RD, Wasser SP. Pharmacological values of
medicinal mushrooms for prostate cancer therapy: the case of Ganoderma lucidum.
Nutr Cancer 2009;61:1626.
Jose N, Ajith TA, Janardhanan KK. Methanol extract of the oyster mushroom,
Pleurotus orida, inhibits inammation and platelet aggregation. Phytother Res
2004;18:436.
Doljak B, Cateni F, Anderluh M, Procida G, Zilic J, Zacchigna M. Glycerolipids as selective thrombin inhibitors from the fungus Stereum hirsutum. Drug Dev Ind Pharm
2006;32:63543.
Mori K, Kikuchi H, Obara Y, Iwashita M, Azumi Y, Kinugasa S, et al. Inhibitory effect
of hericenone B from Hericium erinaceus on collagen-induced platelet aggregation.
Phytomedicine 2010;17:10825.
Kamruzzaman SM, Endale M, Oh WJ, Park SC, Kim TH, Lee IK, et al. Antiplatelet
activity of Phellinus baummii methanol extract is mediated by cyclic AMP elevation
and inhibition of collagen-activated integrin-alpha(IIb) beta(3) and MAP kinase.
Phytother Res 2011;25:1596603.
Kamruzzaman SM, Yayeh T, Ji HD, Park JY, Kwon YS, Lee IK, et al. p-Terphenyl
curtisian E inhibits in vitro platelet aggregation via cAMP elevation and VASP phosphorylation. Vascul Pharmacol 2013;59:839.
Flesch M, Rosenkranz S, Erdmann E, Bohm M. Alcohol and the risk of myocardial
infarction. Basic Res Cardiol 2001;96:12835.
Gronbaek M, Becker U, Johansen D, Gottschau A, Schnohr P, Hein HO, et al. Type of
alcohol consumed and mortality from all causes, coronary heart disease, and cancer. Ann Intern Med 2000;133:4119.
Kloner RA, Rezkalla SH. To drink or not to drink? That is the question. Circulation
2007;116:130617.
Levantesi G, Marsi R, Mozaffarian D, Franzosi MG, Maggioni A, Nicolosi GL,
et al. Wine consumption and risk of cardiovascular events after myocardial
infarction: results from the GISSI-Prevenzione trial. Int J Cardiol
2013;163:2827.
Renaud S, de Lorgeril M. Wine, alcohol, platelets, and the French paradox for coronary heart disease. Lancet 1992;339:15236.
St Leger AS, Cochrane AL, Moore F. Factors associated with cardiac mortality in developed countries with particular reference to the consumption of wine. Lancet
1979;1:101720.
Carnevale R, Nocella C. Alcohol and cardiovascular disease: still unresolved underlying mechanisms. Vascul Pharmacol 2012;57:6971.
Massaro M, Scoditti E, Carluccio MA, De Caterina R. Alcohol and atherosclerosis: a
double edged sword. Vascul Pharmacol 2012;57:658.
Hozumi T, Sugioka K, Shimada K, Kim SH, Kuo MY, Miyake Y, et al. Benecial effect
of short term intake of red wine polyphenols on coronary microcirculation in patients with coronary artery disease. Heart 2006;92:6812.
Pace-Asciak CR, Hahn S, Diamandis EP, Soleas G, Goldberg DM. The red wine phenolics trans-resveratrol and quercetin block human platelet aggregation and eicosanoid synthesis: implications for protection against coronary heart disease. Clin
Chim Acta 1995;235:20719.
Pendurthi UR, Meng F, Mackman N, Rao LV. Mechanism of resveratrolmediated suppression of tissue factor gene expression. Thromb Haemost
2002;87:15562.
Pearson DA, Paglieroni TG, Rein D, Wun T, Schramm DD, Wang JF, et al. The effects
of avanol-rich cocoa and aspirin on ex vivo platelet function. Thromb Res
2002;106:1917.
Yang YM, Chen JZ, Wang XX, Wang SJ, Hu H, Wang HQ. Resveratrol attenuates
thromboxane A2 receptor agonist-induced platelet activation by reducing phospholipase C activity. Eur J Pharmacol 2008;583:14855.
Dell'Agli M, Galli GV, Vrhovsek U, Mattivi F, Bosisio E. In vitro inhibition of human
cGMP-specic phosphodiesterase-5 by polyphenols from red grapes. J Agric Food
Chem 2005;53:19605.

G. Vilahur, L. Badimon / Vascular Pharmacology 59 (2013) 6775


[96] Penumathsa SV, Maulik N. Resveratrol: a promising agent in promoting
cardioprotection against coronary heart disease. Can J Physiol Pharmacol
2009;87:27586.
[97] Schmitt CA, Dirsch VM. Modulation of endothelial nitric oxide by plant-derived
products. Nitric Oxide 2009;21:7791.
[98] Casani L, Segales E, Vilahur G, Bayes de Luna A, Badimon L. Moderate daily intake of
red wine inhibits mural thrombosis and monocyte tissue factor expression in an
experimental porcine model. Circulation 2004;110:4605.
[99] Buijsse B, Weikert C, Drogan D, Bergmann M, Boeing H. Chocolate consumption in
relation to blood pressure and risk of cardiovascular disease in German adults. Eur
Heart J 2010;31:161623.
[100] Cohen DL, Townsend RR. Cocoa ingestion and hypertension another cup please?
J Clin Hypertens (Greenwich) 2007;9:6478.

75

[101] Flammer AJ, Hermann F, Sudano I, Spieker L, Hermann M, Cooper KA, et al. Dark
chocolate improves coronary vasomotion and reduces platelet reactivity. Circulation 2007;116:237682.
[102] Ostertag LM, O'Kennedy N, Kroon PA, Duthie GG, de Roos B. Impact of dietary polyphenols on human platelet function a critical review of controlled dietary intervention studies. Mol Nutr Food Res 2010;54:6081.
[103] Rein D, Paglieroni TG, Wun T, Pearson DA, Schmitz HH, Gosselin R, et al. Cocoa inhibits platelet activation and function. Am J Clin Nutr 2000;72:305.
[104] Rimbach G, Melchin M, Moehring J, Wagner AE. Polyphenols from cocoa and
vascular health a critical review. Int J Mol Sci 2009;10:4290309.
[105] Heiss C, Schroeter H, Balzer J, Kleinbongard P, Matern S, Sies H, et al. Endothelial
function, nitric oxide, and cocoa avanols. J Cardiovasc Pharmacol 2006;47(Suppl.
2):S12835 [discussion S72-6].

Вам также может понравиться