You are on page 1of 11

J. of Cardiovasc. Trans. Res.

(2013) 6:299309
DOI 10.1007/s12265-012-9445-9

Platelet Biology and Receptor Pathways


Giovanni Cimmino & Paolo Golino

Received: 19 November 2012 / Accepted: 27 December 2012 / Published online: 10 January 2013
# Springer Science+Business Media New York 2013

Abstract The main function of platelets is to participate in Keywords Platelet . Aggregation . Membrane receptors .
primary hemostasis through four distinct steps: adhesion, Coagulation
activation, secretion, and aggregation. Unraveling the mo-
lecular mechanisms underlying these steps has led to a better
understanding of the pathophysiology of bleeding disorders, Introduction
on one hand, and of thrombotic diseases, such as acute
coronary syndromes, on the other. Platelets are cytoplasmic Under normal conditions, platelets circulate freely in the
fragments of megakaryocytes formed in the bone marrow. blood without interacting with each other or with the vas-
They lack nuclei but contain organelles and structures, such cular endothelium. In the presence of endothelial damage,
as mitochondria, microtubules, and granules. Platelet gran- whether from a vascular injury or rupture of an atheroscle-
ules contain different bioactive chemical mediators, many of rotic plaque, a chain of events is triggered, ultimately lead-
which have a fundamental role in hemostasis and/or tissue ing to platelet-rich clot formation. Depending on the
healing. The platelet cytoplasm contains an open canalicular initiating event, this may represent normal hemostasis or
system that increases the effective surface area for the intake pathologic intravascular thrombosis [1].
of stimulatory agonists and the release of effector substan-
ces. The submembrane region contains microfilaments of
actin and myosin that mediate morphologic alterations char-
Platelet Morphology
acteristic of shape change. Resting platelets remain in the
circulation for an average of approximately 10 days before
Platelets are anucleated cytoplasmic fragments originating
being removed by macrophages of the reticuloendothelial
from megakaryocytes in the bone marrow; they form as a
system. A wide variety of transmembrane receptors cover
result of cytoplasmic divisions not associated with concom-
the platelet membrane, including many integrins, leucine-
itant nuclear division (a process called endomitosis) of
rich repeat receptors, G protein-coupled receptors, proteins
megakaryocytes in response to thrombopoietin [2]. After
belonging to the immunoglobulin superfamily, C-type lectin
leaving the bone marrow, platelets circulate in the blood
receptors, tyrosine kinase receptors, and a variety of other
for about 10 days. Their volume is about 7 m3 with a
types. In this article, we will review platelet biology under
discoid shape and a major diameter of about 300 nm. A
physiological and pathological conditions, with particular
complex internal system of microtubules and actin cytoskel-
emphasis on the function of their membrane receptors.
eton is responsible for shape change modifications after
activation.
G. Cimmino : P. Golino
The resting platelet can be divided into three zones: (a)
Section of Cardiology, Department of Cardio-Thoracic and
Respiratory Sciences, Second University of Naples, Naples, Italy peripheral zone: responsible for adhesion and aggregation,
consists of a fluffy glycocalyx coat, platelet membrane, and
P. Golino (*) cytoskeleton; it may contain absorbed coagulation factors I,
Division of Cardiology, Second University of Naples, c/o
V, VIII, XI, and XII, receptors for adenosine diphosphate
Ospedale S. Sebastiano Via Tescione, 1,
81100 Caserta, Italy (ADP), thrombin, von Willebrand factor (vWF), collagen,
e-mail: paolo.golino@unina2.it fibrinogen, fibrin, fibronectin, epinephrine, platelet-
300 J. of Cardiovasc. Trans. Res. (2013) 6:299309

activating factor, thrombospondin (TSP), thromboxane A2 The initial tethering of platelets at sites of vascular injury
(TXA2), prostacyclin, epinephrine, serotonin, and glycosyl- is mediated by glycoprotein (GP) Ib/V/IX, a structurally
transferase [3]; (b) solgel zone: responsible for contraction unique receptor complex expressed in megakaryocytes and
and support of the microtubule system; it contains the con- platelets. vWF is the major ligand for one component of this
necting system called the open canalicular system and the complex, GPIb [1, 10, 11], and its absence causes important
dense tubular system; and (c) organelle zone: contains the defects in primary hemostasis and coagulation. Besides
dense body system, nonmetabolic ADP, serotonin, catechol- GPIb, several collagen receptors with a tethering function
amines, calcium, -granules, platelet factor 4 (PF4), platelet are found on the platelet surface, notably GPVI and GPIa/IIa
mitogenic factors (such as platelet-derived growth factor (or 21 integrin), members of the immunoglobulin super-
[PDGF]), fibrinogen, -thromboglobulin, lysosomal gran- family [1, 10, 11]. GPIV, a non-integrin protein, may also
ules, mitochondria, and glycogen granules [46]. play a role in plateletcollagen interactions, as well as acting
Three specific granule populations (dense granules, lyso- as a receptor for TSP [10, 11]. Other integrins that contribute
somes, and -granules) store different types of constituents, to platelet adhesion include GPIc/IIa (a51), a fibronectin
some of which are at high concentrations. Each granule receptor, a laminin receptor, and a vitronectin receptor that
population has specific properties concerning both the struc- also recognizes many of the ligands that bind to the GPIIb/
ture and the role played by the released constituents. Dense IIIa receptor [11].
granules contain small nonprotein molecules that are secret- Shear stress is an important factor that modulates platelet
ed upon platelet activation. The -granules are the most function. Shear stress is related to flow rate, diameter of the
abundant, comprising roughly 10 % of the platelet volume, vessel, and fluid viscosity [12]. Blood circulating in vessels is
10-fold more than dense granules. They contain large adhe- exposed to shear stress caused by the force necessary to
sive and healing proteins, such as hemostatic factors (e.g., produce flow; the difference in velocity between layers situ-
factor V, vWF, fibrinogen), angiogenic factors (e.g., angio- ated at varying distances from the vessel wall determines the
genin, vascular endothelial growth factor [VEGF]), antian- shear rate, which is directly proportional to the shear force and
giogenic factors (e.g., angiostatin, PF4), growth factors inversely proportional to the viscosity of blood. Studies per-
(e.g., PDGF, bFGF, SDF1), proteases (e.g., MMP2, formed under controlled conditions of high shear stress, to
MMP9), necrotic factors (e.g., TNF, TNF), and other mimic the rheological situation existing in certain districts of
cytokines. Among the platelet-specific proteins are several the arterial circulation, have suggested that shear stress may
peptides that modulate cell growth. Of these, PDGF is one influence platelet aggregation through different adhesive
of the most extensively studied [7]. Lysosomes contain ligands, including vWF, GPIb/IX, and GPIIb/IIIa [13, 14].
hydrolases, the function of which is to eliminate circulating Platelets can directly or indirectly interact with collagen
platelet aggregates. through their surface receptors: GPIb/V/IX complex, GPVI,
and GPIa/IIa (or integrin a2b1). Under high shear flow
conditions (i.e., arterial blood flow), the initial interaction
Platelet Function is indirect and mediated by the interposition of vWF, a
circulating protein which is able to bind exposed collagen.
The main physiological role of platelets is to contribute to As a consequence of this binding, vWF switches from its
primary hemostasis, a defense mechanism aimed at prevent- inactive to its activated conformation [15]. The GPIb/V/IX
ing blood loss when the continuity of the vasculature is complex interacts with activated vWF immobilized on ex-
interrupted. In addition to their critical role in hemostasis, posed collagen, and the fast on and off rates of this interac-
new evidence suggests that platelets may exert other func- tion causes rolling of platelets [16]. This process does not
tions in regulating the immune response, promoting inflam- mediate firm adhesion of the cells, but reducing platelets
mation, and cancer metastasis [9]. velocity, allows platelets to bind directly to collagen through
Classically, the complex series of biochemical and cellu- its two chief collagen receptors, GPVI and GPIa/IIa, stabi-
lar processes responsible for the biological functions of lizing the growing thrombus [17]. GPIa/IIa (or integrin
platelets can be divided into four different steps: adhesion, 21) is the second most important platelet integrin (next
activation, secretion, and aggregation (Fig. 1). to IIb3). It is expressed on resting platelets surface in
low-affinity state for collagen, but platelets activation shifts
Adhesion the receptor to a high-affinity state [18].

Adhesion and aggregation should be considered as distinct Activation


processes through which platelets establish individual con-
tacts with extracellular surfaces or stick to one another Platelet activation follows adhesion and can be initiated by
forming aggregates, respectively. several mechanical and chemical stimuli. Adhesion of
J. of Cardiovasc. Trans. Res. (2013) 6:299309 301

Fig. 1 Schematic view of


platelets function (see text for
details)

platelets to collagen and other components of the subendo- fibrinogen molecules, which form bridges between adjacent
thelial matrix (collagen, vWF) are among the strongest platelets and facilitate platelet aggregation [22]. Inhibitors of
stimulators of platelet activation. The activation of platelets GPIIb/IIIa receptors also bind to these receptors, blocking
is associated with stimulation of several metabolic path- the binding of fibrinogen and thus preventing platelet ag-
ways, changes in the shape of platelets, and secretion [1]. gregation [2124]. Other adhesive GPs, including fibronec-
Binding of collagen to GPVI, a type I transmembrane tin, vWF, and vitronectin, also bind to these receptors. As a
protein belonging to the Ig superfamily, plays a central role result of multiple reactions of this type, platelets become
in platelet activation through multiple intracytoplasmic path- aggregated into a hemostatic plug.
ways (i.e., elevation of intracellular Ca2+, phosphoinositide
metabolism, phosphorylation of cytoplasmic and nuclear pro-
teins); this results in the conversion of platelet shape from
discoid to spherical and degranulation [19]. During platelet Platelet Membrane Receptors
shape change, the discoid cells undergo cytoskeletal modifi-
cations, including the disassembly of a microtubule ring that Two ADP receptors are expressed in platelets: P2Y1, which
results in an intermediate spherical shape. This is followed by is coupled to Gq and contributes to initial aggregation, and
actin polymerization and the slower extension of filopodia [1]. P2Y12, which is coupled to G12 and decreases intracellu-
lar levels of cyclic adenosine monophosphate (cAMP). The
P2Y1 is a purinoceptor widely distributed in many tissues
Secretion including the heart, blood vessels and blood cells, neural
tissue, testis, prostate, and ovary [25, 26]. About 150 P2Y1
Following activation, platelets release the substances stored receptor-binding sites are expressed per platelet [27]. The
in their specific granules. This release reaction is an P2Y1 receptor triggers the mobilization of calcium from
important step of primary hemostasis. Energy required for internal stores, which results in platelet shape change and
platelet reactivity is provided by the mitochondria [3]. Once a weak, transient aggregation in response to ADP. As a
activated, platelets readily release the constituents present in consequence, at the intracellular level, the calcium signal
their cytoplasmic granule, such as ADP, ATP, serotonin, is abolished, while the ability of ADP to inhibit cAMP
TXA2 [20]. formation is preserved [28, 29]. The P2Y1 receptor also
participates in the aggregation response to collagen and
Aggregation plays a key role in collagen-induced shape change when
TXA2 formation is prevented [30].
Irrespective of the agonist, the final step after activation is The P2Y12 receptor is coupled to the inhibition of
the formation of platelet aggregates. This event is mediated adenylyl cyclase activity through the activation of a Gi2
by the binding of adhesive proteins to the now ligand- G protein subtype, which is a critical component of the
receptive form of the GPIIb/IIIa receptor. It is well-known signaling pathway for integrin IIb3 activation [31].
that activation causes changes in the shape of platelets and However, adenylyl cyclase inhibition resulting in lowering
conformational changes in GPIIb/IIIa receptors, transform- cAMP levels is not sufficient to cause platelet aggregation
ing the receptors from a ligand-unreceptive to a ligand- per se [32]; thus, other signaling events are required for full
receptive state [21]. The receptive form of GPIIb/IIIa binds activation of the IIb3 integrin and subsequent aggregation.
302 J. of Cardiovasc. Trans. Res. (2013) 6:299309

One important intracellular pathway, which regulates Gi- in each case. The cleavage process is irreversible, thus PAR-
dependent integrin IIb3 activation, is represented by phos- 1 pathway needs several inactivating mechanisms to switch
phoinositide 3-kinase (PI3K). The PI3K isoform p110 reg- off signal transduction: receptor desensitization (via phos-
ulates integrin activation through a classical lipid kinase- phorylation for PAR-1, but not for PAR-4) [8, 44], receptor
dependent mechanism, involving the small GTPase Rap1 internalization, and lysosomes degradation [41] are the main
and/or the serinethreonine protein kinase B/Akt (PKB/Akt) known mechanisms.
[33], whereas p110 appears to regulate integrins principally Thrombin-mediated platelet activation in humans occurs
through a noncatalytic signaling mechanism [34]. through the activation of PAR-1 and PAR-4. PAR-1 is con-
Coactivation of the P2Y1 and P2Y12 receptors is necessary sidered the principal proteaseplatelet receptor and mediates
for normal ADP-induced platelet aggregation, since separate the activation of human platelets at low (<1 nM) thrombin
inhibition of either of them with selective antagonists results concentrations; PAR-4 necessitates higher (>30 nM) concen-
in a dramatic decrease in aggregation [35]. tration of thrombin [42]. PAR-1 signaling in platelets results in
P2Y12 receptor signaling also stimulates surface expres- heterotrimeric G proteins activation (G12/13, Gq, and
sion of P-selectin and secretion of TXA2 [36]. TXA2 is Gi/z) with consequent TXA2 production, ADP release, mo-
produced de novo and, like ADP, is released by activated bilization of P-selectin and CD40L, integrin activation, and
platelets. It is enzymatically generated from arachidonic platelet aggregation. PAR-1, in addition, enhances thrombin
acid (AA) by cyclooxygenase-1 (COX-1) and TX synthase. formation and fibrin generation inducing platelets procoagu-
TXA2 binds platelet receptors TP and TP which differ in lant activity [43]. Recently, several studies also showed a
their cytoplasmic tail; however, its effects in platelets are cross-reaction of P2Y12 and PAR-1 pathways in mediating
mediated primarily through TP [37]. TP and TP couple platelets activation. Despite the lower affinity of PAR-4 for
to the proteins Gq and G12 or G13, all of which activate thrombin, both PAR-1 and PAR-4 contributes to platelet acti-
phospholipase C. This enzyme degrades the membrane vation. PAR-4 is activated, shut off, and internalized more
phosphoinositides (such as phosphatidylinositol 4,5- slowly than PAR-1 and produces the majority of the integrated
bisphosphate [PIP2]), releasing the second messenger ino- thrombin-triggered calcium signals of the two receptors. In
sitol triphosphate (IP3) and diacylglycerol (DAG). DAG addition, PAR-1 and PAR-4 counter-regulate the release of
activates intracellular protein kinase C, which causes protein endostatin and VEGF from human platelets [44]. The impor-
phosphorylation. The release of IP3 increases cytosolic lev- tance of these differences in PARs signaling in human platelet
els of Ca2+, which is released from the endoplasmic reticu- is still not clear. The current hypothesis is that PAR-4 acts as
lum. Both ADP and TXA2 are secreted from adherent an auxiliary receptor mediating thrombin signaling to distinct
platelets, contribute to the recruitment of circulating plate- effectors (i.e., release of distinct subsets of platelets granules)
lets, and promote alterations in platelet shape and granule or with different kinetics compared with PAR-1. A schematic
secretion. view of the major platelet receptors with its own function is
Thrombin is the ultimate effector of the coagulation reported in Table 1.
cascade and, in addition to cleaving fibrinogen and other
circulating substrates, it acts as a circulating mediator able to
induce different cellular responses in platelets, endothelial Regulation of Platelet Function: The Novel Role
cells, and SMCs. Thrombin binds a G protein-coupled class of microRNAs (miRNA)
of receptor named protease-activated receptors (PARs) [38,
39]. Four subtypes of PARs have been identified in humans, Platelets activation is a highly regulated process. Despite the
which are widely distributed among different cell types and fact that they are anucleate cell fragments, it has been shown
tissues. PAR-1, PAR-3, and PAR-4 are activated by throm- that platelets contain a number of megakaryocyte-derived
bin, whereas PAR-2 can be activated by trypsin, tryptase, mRNAs [45] encoding for different proteins involved in
and coagulation factors VIIa and Xa [8, 9], but not by metabolism, signaling, inflammation, and immunity [46].
thrombin. PAR-1 can be considered as the prototype of this Platelets also contain rough endoplasmic reticulum and pol-
receptor family. It is converted in the active form by the yribosomes, enabling them to synthesize different proteins
cleavage of its N-terminal exodomains at a specific site [47]. These mechanisms allow platelets to modify their
(thrombin cleavage site) that unmasks a new N terminus protein expression and, as a consequence, their functions
domain (SFLLRN) able to bind intramolecularly to the body [48]. For example, specific mRNA translation may vary in
of the receptor, resulting in a transmembrane signaling via G clinical conditions, such as acute myocardial infarction, as
protein activation [40]. Thus, PAR-1 carries its own silent recently reported [49]. Identification of disease-associated
agonist peptide and achieves a sort of self-activation pro- platelet-specific transcripts is of particular relevance in
cess after thrombin cleavage. Each of the four PARs is platelet pathophysiology, since it may lead to the discovery
activated via this mechanism, with some minor variations of novel therapeutic targets.
J. of Cardiovasc. Trans. Res. (2013) 6:299309 303

Table 1 Major Platelet Mem-


brane Receptors Name Type Agonists Role

glycoprotein Ib (GPIb) Integrin vWF Adhesion


glycoprotein VI (GPVI) non-integrin collagen Adhesion
glycoprotein Ia/IIa (GPIa/IIa) integrin collagen Adhesion
glicoprotein Ib-V-IX complex (GPIb-V-IX) integrin collagen Adhesion
glycoprotein VI (GPVI) non-integrin collagen Activation
P2Y1 G protein-coupled receptor ADP Aggregation
P2Y12 G protein-coupled receptor ADP Aggregation
Thromboxane Receptor (TP) G protein-coupled receptor TXA2 Aggregation
Protease Activated Receptor (PAR) G protein-coupled receptor Thrombin Aggregation
glycoprotein IIb/IIIa (GPIIb/IIIa) integrin Fibrinogen Aggregation

The evidence that platelets contain mRNAs and are ca- function could be modulated by targeting the previously
pable of protein synthesis has raised the issue of how these discussed ligands and/or receptors; this approach has
mRNAs are regulated. Among the various mechanisms in- resulted in the development of several pharmacological
volved, mRNA splicing [50, 51] and miRNAs are of great interventions.
relevance. miRNAs are a class of small noncoding RNAs
(approximately 21 to 23 nucleotides) which function pre- ADP Receptors Antagonism
dominantly as sequence-targeted modifiers of gene expres-
sion acting on posttranscriptional processes, such as mRNA P2Y12 receptor antagonists include ticlopidine, clopidogrel,
degradation and translational repression. They originate prasugrel, ticagrelor, and other compounds under late-stage
from longer precursor RNAs (100 to 1,000 nucleotides development (cangrelor, elinogrel) [56]. Ticlopidine, clopi-
primary miRNAs) that are processed in the nucleus into a dogrel, and prasugrel are thienopyridines that selectively
70- to 100-nucleotide hairpin-shaped precursor miRNA. and irreversibly inhibit the P2Y12 receptor. Other new re-
Pre-miRNAs are then transported into the cytoplasm by a versible P2Y12 inhibitors are currently at different stages of
nuclear export factor and processed into a 19- to 25- clinical development, including those which reversibly in-
nucleotide double-stranded RNA. This duplex miRNA is hibit the P2Y12 receptor available for oral (such as ticagrelor
then incorporated into the RNA-induced silencing complex. that has been recently approved for routine use in patients
One strand remains in the complex and becomes the ma- with acute coronary syndrome [ACS]), intravenous
ture miRNA, whereas the other strand is often rapidly (cangrelor), or both oral and intravenous (elinogrel) routes
degraded. The mature miRNA interacts with the target of administration.
mRNAs as a negative regulator; downregulating protein
expression [52] miRNAs has been implicated in the patho- TXA2 Pathway Antagonism
genesis of various diseases and has become an intriguing
target for therapeutic interventions [53]. Antagonists of TP receptors may have different advantages
Recently, the existence and functionality of miRNA path- over aspirin as they not only block the effect of TXA2 on
way components in human platelets has been reported [54]. platelets but also inhibit other ligands such as prostaglandin
Moving from these observations, in preliminary experi- (PG) endoperoxides and isoprostanes. Given the distribution
ments, our group has analyzed miRNA modulation in an of TP receptors in platelets, in circulating inflammatory
ex vivo model of ADP-induced platelet activation, founding cells, in the vascular wall, and in atherosclerotic plaques,
that platelets activation resulted in the expression of 435 they also inhibit the effects of TXA2 over TP receptors on
miRNAs, 308 of which were modulated up to 2 h after vascular cells or in the plaque. A large trial is currently
activation. These findings suggest, for the first time, a phys- comparing the efficacy and safety of terutroban versus as-
iological role for miRNAs in the regulation of platelet pirin in secondary prevention of cardiovascular events in
function [55]. patients who have suffered a stroke or transient ischemic
attack (PERFORM Study) [57].

Therapeutic Modulation of Platelet Function Collagen Receptors Antagonism

In the last two decades, better understanding of platelets Collagen receptors antagonism is considered as a new prom-
biology induced researchers to investigate how platelet ising strategy for antithrombotic therapy. GPVI has emerged
304 J. of Cardiovasc. Trans. Res. (2013) 6:299309

as a powerful and safe pharmacological target preventing low-molecular-weight nonpeptide, competitive antagonist
thrombus formation without causing major bleeding com- of PAR-1. Despite preclinical and early clinical studies that
plications in experimental animal model. Injection of JAQ1, reported no significant increase in bleeding, the Thrombin
a monoclonal antibody selective for GPVI, in mice results in Receptor Antagonist for Clinical Event Reduction in ACS
an acquired deficiency of the receptor that mimics a GPVI (TRACER) trial, a phase 3 study designed to test the antith-
knockout phenotype, causing a long-term antithrombotic rombotic effect of vorapaxar administered in combination
protection, despite a mild increase in bleeding times [58]. with aspirin and/or clopidogrel, was stopped because of
An alternative way of antagonism is the use of F(ab) frag- increased intracranial bleeding in the vorapaxar group
ments of anti-GPVI that block the collagen-binding site of [65]. These data have been also confirmed by the recently
the receptor. Also, this type of approach has resulted in published TRA2P trial, in which vorapaxar reduced the risk
limited bleeding complications [59]. Other modalities of of cardiovascular death or ischemic events in patients with
antagonism are represented by competitive inhibition of stable atherosclerosis but it was associated with an increas-
GPVI-binding sites on its substrate collagen through ing risk of moderate or severe bleeding, including intracra-
GPVI-Fc fusion protein [60] or by the interference with nial hemorrhage [66]. Atopaxar is a low-molecular-weight,
the GPVI/FcR gamma-chain intracellular signaling pathway competitive antagonist of PAR-1 trialed for the treatment
[61]. The favorable profile of GPVI antagonism derives and prevention of arterial thrombosis. In a small phase 2 trial
from a relatively minor role of GPVI in the physiological (Japanese-Lesson from Antagonizing the Cellular Effect of
hemostatic process, as opposed to a prime role in the for- Thrombin [J-LANCELOT]), atopaxar has been proved to be
mation of in vivo (experimental) arterial thrombi following generally well-tolerated, with no increase in major bleeding
injury of the arterial wall. [67]. These results presented atopaxar as a potential effec-
The collagenvWFGPIb complex, for its dominant role tive treatment of thrombosis and led to phase 3 trial for the
in platelets adhesion, represents an alternative interesting evaluation of therapeutic efficacy in the treatment and pre-
target for new antithrombotic drugs. Anti-vWF and anti- vention of cardiovascular disease [67]. Thus, in the near
GPIb antibody showed a potent antithrombotic effect in future, PAR antagonism might represent a valid alternative
vivo, without significant increase of bleeding risk [62], to the current antithrombotic therapy. Further studies are
while despite its unquestionable involvement in adhesion needed to clarify its efficacy and safety profile.
process, contradictory results have been obtained in experi-
ments with integrin 21 antagonist [62]. More recently a GPIIb/IIIa Antagonism
bivalent nanobody (ALX-0081) that binds with high affinity
to the A1 domain of vWF has been developed. It consists of Since the GPIIb/IIIa receptor is the final common pathway
two identical building blocks bound to each other via a short that leads to platelet aggregation, direct inhibition of this
linker that stops the interactions between platelets and vas- receptor is one of the therapeutic strategies widely used in
cular collagen. It selectively prevents thrombus formation clinical practice [22]. This inhibition has been obtained by
under high shear stress conditions [63]. In the proof of using monoclonal antibody or blocking peptides. Presently,
concept phase 2 trial comparing ALX-0081 versus GPIIb/ abciximab, eptifibatide, and tirofiban are the GPIIb/IIIa
IIIa inhibitor ReoPro in ACS patients, the nanobody showed inhibitors most extensively studied in several randomized
that 36 (19.9 %) patients reported bleeding events in the trials involving thousands of patients with ACS or undergo-
ALX-0081 remedy group and 28 (15.3 %) patients reported ing PCI [23]. Abciximab is a large monoclonal antibody that
bleeding events in the ReoPro remedy group. Only three binds to the GPIIb/IIIa receptor as well as to the v3
ALX-0081-treated patients (1.7 %) and two ReoPro-treated integrin receptor of endothelial and smooth muscle cells
patients (1.1 %) showed a key bleeding event during the 30- and to the m2 integrin found in leucocytes. Tirofiban
day period following the percutaneous coronary interven- and eptifibatide are nonpeptide molecules with marked
tion (PCI) process. However, ALX-0081 failed to meet the specificity for the GPIIb/IIIa receptor. Thus, only abciximab
primary endpoint of a 40 % reduction in total bleeding has the potential for direct inhibition of adhesion of platelets
events compared to the GPIIb/IIIa inhibitor ReoPro [64]. and endothelial cells and of platelets and white cells, and as
a consequence, to inhibit or limit the inflammatory process
PAR Antagonism invariably present in ACS and after PCI. In randomized
placebo-controlled trials, the risk of recurrent ischemic com-
Blocking exclusively thrombin-mediated platelet activation, plications within 30 days after PCI was reduced by 4060 %
PAR modulation might act selectively on pathological with abciximab and by 1535 % with eptifibatide or tirofi-
thrombosis. Two distinct PAR-1 antagonists, vorapaxar ban [23]. Moreover, abciximab has been associated with a
and atopaxar, were investigated in phase 2 and phase 3 long-term decrease in mortality, an effect that cannot be
clinical studies. Vorapaxar is a high-affinity, orally active, entirely attributed to the suppression of acute periprocedural
J. of Cardiovasc. Trans. Res. (2013) 6:299309 305

ischemic events, whereas mortality reduction has not been COX-1 and COX-2 are differently regulated: (a) COX-2
observed to date with eptifibatide or tirofiban [24]. requires considerably lower levels of hydroperoxides to
initiate COX catalysis than those required by COX-1; (b)
COX-2 activity occurs at lower levels of free AA than COX-
COX Pathway: Thrombosis, Inflammation, and Beyond 1 activity [11]; (c) COX-1 has the structural features of a
housekeeping gene, while COX-2 has the structural fea-
COX is the central enzyme of cascade reactions leading to tures of an immediate early gene; and (d) both mRNA and
the conversion of AA to prostanoids, a cluster of bioactive protein of COX-1 are more stable than those of COX-2 [74].
lipids known to affect multiple signaling pathways that Pharmacological modulation of COX-1 activity can be
modulate physiological processes such as inflammation, achieved by the use of aspirin, the oldest and most widely
blood clotting, wound healing, blood vessel tone, and im- prescribed antithrombotic drug. The antiplatelet effect has
mune responses. The first step of this pathway is the liber- been attributed to the irreversible acetylation of the serine
ation of AA from membrane phospholipid by phospholipase residue (Ser529) in platelet-expressed COX-1 that inhibits
A2. The free AA is converted via two independent enzymat- the synthesis of PGH2 and, subsequently, the generation of
ic reactions catalyzed by COX, first in PGG2 via a COX TXA2. The effect of aspirin on platelet TXA2 production
function and then in PGH2 via a peroxidase function. PGH2 continues for the all lifetime of platelets, as consequence of
is then converted to other PGs, such as prostacyclins (PGI2) the aspirin ability to acetylate irreversibly COX-1 and the
and TXA2, by tissue-specific isomerases [68]. inability of anucleated platelets to synthesize new COX-1
There are two different COX isoenzymes, known as COX- protein. A high level of inhibition of platelet COX-1 is
1 and COX-2, that differ for regulatory mechanisms of ex- required in vivo to obtain antiplatelet efficacy (more than
pression, tissue distribution, substrate specificity, and prefer- 95 % inhibition of TXA2 synthesis is required to observe
ential coupling to upstream and downstream enzymes [69]. any clinical efficacy of aspirin). The inhibition of COX-2-
Recently, an alternative splice variant of COX-1 has been related pathologic manifestations (hyperalgesia and inflam-
identified (COX-3). This variant is highly unlikely to be a mation) requires large doses of aspirin and a shorter dosing
functional human protein because it has no COX activity [70]. interval (nucleated cells rapidly resynthesize the enzyme).
COX-1 is a membrane-bound hemoglycoprotein of
71 kDa found in the endoplasmic reticulum of PG-
producing cells, constitutively expressed in most of tissue. Novel Platelets Function: Link Between Inflammation
The enzyme structure is characterized by an EGF-like do- and Immunity
main, a membrane-binding domain, and an enzymatic do-
main. The COX active site is a long hydrophobic channel that The central role of platelets in hemostasis and thrombosis is
is blocked by nonsteroidal anti-inflammatory drugs to prevent well-known; however, platelets probably contribute to several
the access of AA. COX-1-derived PGs are thought to mediate other processes, such as inflammation and immunity. This
physiological processes that require constant or rapid modula- concept took a long time to get consensus in the scientific
tion (or both), the so-called housekeeping functions, including community. In addition to their traditional contribution in clot
vascular homeostasis, gastrointestinal protection, and sodium formation, platelets are also specialized in further activities
reabsorption in the kidney [71]. Furthermore, COX-1 is one of and intercellular interactions that make them key effectors in
the main controllers of platelet function, providing intermedi- inflammation and in the continuum of innate and adaptive
ate precursors for TXA2 synthesis. immunity [75]. For example, they can recruit leukocytes and
Originally discovered as a protein upregulated during progenitor cells to sites of vascular injury and inflammation
inflammation and strongly decreased by glucocorticoids, [75]; they release proinflammatory, anti-inflammatory, and
COX-2 is an inducible enzyme normally absent from tissues angiogenic factors and microparticles into the circulation
(with the exception of kidney and parts of the brain, where [76], and they spur thrombin generation [74].
COX-2 is constitutive) that is mainly expressed by cells Platelets also express immunoreceptors on their surfa-
involved in inflammation (e.g., macrophages, monocytes, ces, including Fc receptors that recognize immunoglobulins
synoviocytes) in response to a wide range of extracellular of the IgG, IgE, and IgA classes. Like platelet integrins,
and intracellular stimuli (such as growth factors, tumor these immunoreceptors seem to influence adhesion and,
promoters, or cytokines) [72]. Thus, it has emerged as the more importantly, inflammatory and immune responses
isoform primarily responsible for the synthesis of prosta- [77]. Via these receptors, platelets may contribute to innate
noids involved in acute and chronic inflammatory states and adaptive immunity. As part of innate immunity, they
(i.e., PGE2). COX-2 structure closely resembles that of possess, in fact, rudimentary antibacterial and phagocytic
COX-1; however, its active site has a slightly larger volume activity by which platelets interact with bacteria, viruses,
and can accommodate specifically larger drugs [73]. and parasites. This interaction induces platelet activation
306 J. of Cardiovasc. Trans. Res. (2013) 6:299309

and secretion of antimicrobial peptides. Furthermore, in with peripheral lymph nodes addressin on high endothelial
platelet granules are stored many proinflammatory cyto- venules (HEV) and P-selectin glycoprotein ligand-1 (PSGL-
kines (e.g., IL-1), which modulate the inflammatory/im- 1) on lymphocytes simultaneously, thus mediating the roll-
mune responses [77]. More interestingly, platelets express ing and recruitment of lymphocytes to HEV of peripheral
several functional Toll-like receptors (TLRs), such as TLR- lymph nodes. Interestingly, platelets also contain multiple
2, TLR-4, and TLR-9. It has been shown that the interaction anti-inflammatory cytokines, such as TGF- [81], and it has
between platelets TLR-4 and lipopolysaccharide from the been reported that metastasizing tumor cells induce platelets
gram-negative bacteria activates platelets and induces plate- to secrete TGF-, which inhibits the antitumor activity of
letneutrophil cross-talk, leading to neutrophil degranula- natural killer cells. Furthermore, platelets contain a large
tion and release of extracellular traps that can kill the amount of TSP-1, which comprises around 25 % of the total
bacteria [77]. protein content of platelet -granules [82]. It has been
Platelets are also actively involved in adaptive immunity. demonstrated that TSP-1 not only activates the anti-
They contain functional transforming growth factor beta inflammatory cytokine TGF-1 [83] but also inhibits the
(TGF-), which is essential for the development of naturally phagocytic capacity of macrophages. In summary, platelets
occurring regulatory T cells (nTreg) or T helper 17 cells participate not only in proinflammatory responses but also
(Th17), depending on the local cytokine environment [78]. they might modulate that balance between inflammation and
This involvement seems to be mediated by P-selectin immune responses (Fig. 2).
(CD62P), which plays an important role in the development
of the Th1 immune response [79], and CD40L, which plays an
important role in supporting immunoglobulin class switch and Conclusions
augmenting CD8+ T cell function during viral infection [80].
Platelet activation induces P-selectin translocation from During the last decade, important advances in our under-
the -granule to the cell surface. P-selectin can then interact standing of the pathophysiology of platelets have been

Fig. 2 Platelets as bridge between hemostasis and immunity. Throm- activation. In a similar capacity, 5HT can activate T cells and induce
bin activation of PARs, in concert with platelet-derived CD154, sero- autocrine production of 5HT to amplify this process. PF4 directly
tonin (5HT), platelet factor 4 (PF4), and regulated and normal T cell interacts with CXCR3 to increase T cell recruitment or forms dimers
expressed and secreted (RANTES), has now been discovered to opti- with RANTES that recruits monocytes and T cells. The activation of
mize the process of antigen presentation to initiate and steer the PAR on dendritic cells enhanced the production of proinflammatory
phenotype of subsequent adaptive immune responses. Platelets express and prothrombotic mediators, a potential feedback system to amplify
CD154, which can be secreted by TLR induction or remotely delivered antigen presentation
by platelet-derived microvesicles to augment both dendritic and B cell
J. of Cardiovasc. Trans. Res. (2013) 6:299309 307

made. These small cell fragments play a critical role in all 13. Ballerman, B. J., Dardik, A., Eng, E., & Liu, A. (1998). Shear
stress and the endothelium. Kidney International. Supplement, 67,
stages of the complex, dynamic process of hemostasis and
S100S108.
thrombus formation. Their function is the result of the 14. Ikeda, Y., Handa, M., Kawano, K., Kamata, T., Murata, M., Araki,
interaction between different agonists and specific receptors Y., et al. (1991). The role of von Willebrand factor and fibrinogen
at platelet surface, triggering intracellular signaling that in platelet aggregation under varying shear stress. The Journal of
Clinical Investigation, 87(4), 12341240.
leads to platelet activation; this is accompanied by the
15. Lisman, T., Raynal, N., Groeneveld, D., Maddox, B., Peachey, A.
release of different factors and inflammatory substances. R., Huizinga, E. G., et al. (2006). A single high-affinity binding
These mediators act in an orchestrated manner with the site for von Willebrand Factor in collagen III, identified using
vascular endothelium and other blood cells and coagulation synthetic triple-helical peptides. Blood, 108, 37533756.
16. Kumar, R. A., Dong, J. F., Thaggard, J. A., Cruz, M. A., Lopez, J.
factors to promote not only the growth of the platelet plug A., & McIntire, L. V. (2003). Kinetics of GPIb alpha-vWF-A1
by recruiting new platelets but also to consolidate the throm- tether bond under flow: Effect of GPIb alpha mutations on the
bus by means of post-aggregation signaling events, clot association and dissociation rates. Biophysical Journal, 85, 4099
retraction, and controlled formation of a fibrin network. 4109.
17. Savage, B., Almus-Jacobs, F., & Ruggeri, Z. M. (1998). Specific
On the other hand, compensatory molecules and signaling
synergy of multiple substratereceptor interactions in platelet
events also starts to limit undesirable platelet accumulation thrombus formation under flow. Cell, 94, 657666.
and pathological thrombosis. Based on new evidence, pla- 18. Van de Walle, G. R., Vanhoorelbeke, K., Majer, Z., Illyes, E.,
telets might be a bridge between different extracellular Baert, J., Pareyn, I., et al. (2005). Two functional active conforma-
tions of the integrin alpha 2 beta 1, depending on activation
functions, beyond the coagulative cascade, thus the im-
condition and cell type. Journal of Biological Chemistry, 280,
provement of our knowledge on these extracoagulative 3687336882.
actions may also result in the development of novel thera- 19. Nieswandt, B., & Watson, S. P. (2003). Platelet-collagen interac-
peutic approaches. tion: Is GPVI the central receptor? Blood, 102, 449461.
20. Dale, G. L., Friese, P., Batar, P., et al. (2002). Stimulated platelets
use serotonin to enhance their retention of procoagulant proteins
on the cell surface. Nature, 415, 175179.
References 21. Coller, B. S., Scudder, L. E., Beer, J., Gold, H. K., Folts, J. D.,
Cavagnaro, J., et al. (1991). Monoclonal antibodies to platelet
glycoprotein IIb/IIIa as antithrombotic agents. Annals of the New
1. Jurk, K., & Kehrel, B. E. (2005). Platelets: Physiology and bio- York Academy of Sciences, 614, 193213.
chemistry. Seminars in Thrombosis and Hemostasis, 31(4), 381 22. Madan, M., Berkowitz, S. D., & Tcheng, J. E. (1998).
392. Glycoprotein IIb/IIIa integrin blockade. Circulation, 98, 2629
2. Schulze, H., & Shivdasani, R. A. (2005). Mechanisms of throm- 2635.
bopoiesis. Journal of Thrombosis and Haemostasis, 3(8), 1717 23. Linkoff, A. M., Califf, R. M., & Topol, E. J. (2000). Platelet
1724. glycoprotein blockade in coronary artery disease. Journal of the
3. Rendu, F., & Brohard-Bohn, B. (2001). The platelet release reac- American College of Cardiology, 35, 11031115.
tion: Granules constituents, secretion and functions. Platelets, 12, 24. Anderson, K. M., Califf, R. M., Stone, G. W., Nemann, F.-J.,
261273. Montalescot, G., Miller, D. P., et al. (2001). Long-term mortality
4. Escolar, G., & White, J. G. (1991). The platelet open canalicular benefit with abciximab in patients undergoing percutaneous coro-
system: A final common pathway. Blood Cells, 17, 467485. nary intervention. Journal of the American College of Cardiology,
5. Fox, J. E. (1993). The platelet cytoskeleton. Thrombosis and 37, 20592065.
Haemostasis, 70, 884893. 25. Offermanns, S. (2006). Activation of platelet function through G
6. McNicol, A., & Israels, S. J. (1999). Platelet dense granules: protein coupled receptors. Circulation Research, 99, 12931304.
Structure, function and implications for haemostasis. Thrombosis 26. Abbracchio, M. P., Burnstock, G., Boeynaems, J. M., Barnard, E.
Research, 95, 118. A., Boyer, J. L., Kennedy, C., et al. (2006). International Union of
7. Heijnen, H. F., Debili, N., Vainchencker, W., Breton-Gorius, J., Pharmacology LVIII: Update on the P2Y G protein-coupled nu-
Geuze, H. J., & Sixma, J. J. (1998). Multivesicular bodies are an cleotide receptors: From molecular mechanisms and pathophysiol-
intermediate stage in the formation of platelet alpha-granules. ogy to therapy. Pharmacological Reviews, 58, 281341.
Blood, 91(7), 23132325. 27. Ohlmann, P., Castro, S., Brown, G. G., Jr., Gachet, C., Jacobson,
8. Harrison, P., & Cramer, E. M. (1993). Platelet alpha-granules. K. A., & Harden, T. K. (2010). Quantification of recombinant and
Blood Reviews, 7, 5262. platelet P2Y(1) receptors utilizing a [(125)I]-labeled high-affinity
9. Semple, J. W., Italiano, J. E., Jr., & Freedman, J. (2011). Platelets antagonist 2-iodo-N(6)-methyl-(N)-methanocarba-2-deoxyadeno-
and the immune continuum. Nature Reviews Immunology, 11(4), sine-3,5-bisphosphate ([(125)I]MRS2500). Pharmacological
264274. Research, 62(4), 344351.
10. Angiolillo, D. J., Ueno, M., & Goto, S. (2010). Basic principles of 28. Jin, J., Daniel, J. L., & Kunapuli, S. P. (1998). Molecular basis for
platelet biology and clinical implications. Circulation Journal, 74, ADP-induced platelet activation. II. The P2Y1 receptor mediates
597607. ADP-induced intracellular calcium mobilization and shape change
11. Varga-Szabo, D., Pleines, I., & Nieswandt, B. (2008). Cell adhe- in platelets. Journal of Biological Chemistry, 273(4), 20302034.
sion mechanisms in platelets. Arteriosclerosis, Thrombosis, and 29. Savi, P., Beauverger, P., Labouret, C., Delfaud, M., Salel, V.,
Vascular Biology, 28, 403412. Kaghad, M., et al. (1998). Role of P2Y1 purinoceptor in ADP-
12. Kroll, M. H., Hellums, J. D., McIntire, L. V., Schafer, A. I., & induced platelet activation. FEBS Letters, 422(3), 291295.
Moake, J. L. (1996). Platelets and shear stress. Blood, 88, 1525 30. Mangin, P., Ohlmann, P., Eckly, A., Cazenave, J. P., Lanza, F., &
1541. Gachet, C. (2004). The P2Y receptor plays an essential role in the
308 J. of Cardiovasc. Trans. Res. (2013) 6:299309

platelet shape change induced by collagen when TxA2 formation is 48. Weyrich, A. S., Lindemann, S., & Tolley, N. D. (2004). Change in
prevented. Journal of Thrombosis and Haemostasis, 2(6), 969 protein phenotype without a nucleus: Translational control in pla-
977. telets. Seminars in Thrombosis and Hemostasis, 30, 491498.
31. Hardy, A. R., Jones, M. L., Mundell, S. J., & Poole, A. W. (2004). 49. Healy, A. M., Pickard, M. D., & Pradhan, A. D. (2006). Platelet
Reciprocal cross-talk between P2Y1 and P2Y12 receptors at the level expression profiling and clinical validation of myeloid-related
of calcium signaling in human platelets. Blood, 104(6), 17451752. protein-14 as a novel determinant of cardiovascular events.
32. Yang, J., Wu, J., Jiang, H., Mortensen, R., Austin, S., Manning, D. Circulation, 113, 22782284.
R., et al. (2002). Signaling through Gi family members in platelets. 50. Denis, M. M., Tolley, N. D., & Bunting, M. (2005). Escaping the
Redundancy and specificity in the regulation of adenylyl cyclase nuclear confines: Signal-dependent pre-mRNA splicing in anu-
and other effectors. Journal of Biological Chemistry, 277(48), cleate platelets. Cell, 122, 379391.
4603546042. 51. Schwertz, H., Tolley, N. D., & Foulks, J. M. (2006). Signal-
33. Martin, V., Guillermet-Guibert, J., Chicanne, G., Cabou, C., dependent splicing of tissue factor pre-mRNA modulates the
Jandrot-Perrus, M., Plantavid, M., et al. (2010). Deletion of the thrombogenicity of human platelets. The Journal of Experimental
p110beta isoform of phosphoinositide 3-kinase in platelets reveals Medicine, 203, 24332440.
its central role in Akt activation and thrombus formation in vitro 52. Baek, D., Villen, J., & Shin, C. (2008). Nature, 455, 6471.
and in vivo. Blood, 115(10), 20082013. 53. Small, E.M., Frost, R.J., Olson, E.N. (2010). MicroRNAs add a
34. Hirsch, E., Bosco, O., Tropel, P., Laffargue, M., Calvez, R., Altruda, new dimension to cardiovascular disease. Circulation, 121, 1022
F., et al. (2001). Resistance to thromboembolism in PI3Kgamma- 1032.
deficient mice. The FASEB Journal, 15(11), 20192021. 54. Landry, P., Plante, I., & Ouellet, D. L. (2009). Existence of a
35. Jin, J., & Kunapuli, S. P. (1998). Coactivation of two different G microRNA pathway in anucleate platelets. Nature Structural and
protein-coupled receptors is essential for ADP-induced platelet Molecular Biology, 16(9), 961966.
aggregation. Proceedings National Academy Science USA, 95 55. Cimmino, G., Tarallo, R., Nassa, G., De Filippo, M.R., Conte, S.,
(14), 80708074. Pellegrino, G., Morello, A., Cirillo, P., Weisz, A., Golino, P.
36. Storey, R. F., Judge, H. M., Wilcox, R. G., & Heptinstall, S. (2012). Activation modulates miRNA expression profile in plate-
(2002). Inhibition of ADP-induced P-selectin expression and lets: New insights into the pathophysiology of platelet activation.
platelet-leukocyte conjugate formation by clopidogrel and the AHA Scientific Session, Abstract 12998
P2Y 12 receptor antagonist AR-C69931MX but not aspirin. 56. Damman, P., Woudstra, P., Kuijt, W. J., de Winter, R. J., & James,
Thrombosis and Haemostasis, 88, 488494. S. K. (2012). P2Y12 platelet inhibition in clinical practice. Journal
37. FitzGerald, G. A. (1991). Mechanisms of platelet activation: of Thrombosis and Thrombolysis, 33(2), 143153.
Thromboxane A2 as an amplifying signal for other agonists. The 57. Chamorro, A. (2009). TP receptor antagonism: A new concept in
American Journal of Cardiology, 68, 11B15B. atherothrombosis and stroke prevention. Cerebrovascular
38. Coughlin, S. R. (1999). How the protease thrombin talks to cells. Diseases, 27(Suppl 3), 2027. Epub 2009 May 14.
Proceedings of the National Academy of Sciences of the United 58. Nieswandt, B., Schulte, V., Bergmeier, W., Mokhtari-Nejad, R.,
States of America, 96, 110237. Rackebrandt, K., Cazenave, J. P., et al. (2001). Long-term antith-
39. Ossovskaya, V. S., & Bunnett, N. W. (2004). Protease-activated rombotic protection by in vivo depletion of platelet glycoprotein
receptors: Contribution to physiology and disease. Physiological VI in mice. The Journal of Experimental Medicine, 193, 459469.
Reviews, 84, 579621. 59. Li, H., Lockyer, S., Concepcion, A., Gong, X., Takizawa, H.,
40. Vu, T. K., Hung, D. T., Wheaton, VI, & Coughlin, S. R. (1991). Guertin, M., et al. (2007). The Fab fragment of a novel anti-
Molecular cloning of a functional thrombin receptor reveals a GPVI monoclonal antibody, OM4, reduces in vivo thrombosis
novel proteolytic mechanism of receptor activation. Cell, 64, without bleeding risk in rats. Arteriosclerosis, Thrombosis, and
10571068. Vascular Biology, 27, 11991205.
41. OBrien, P. J., Molino, M., Kahn, M., & Brass, L. F. (2001). 60. Massberg, S., Konrad, I., Bltmann, A., Schulz, C., Mnch, G.,
Protease activated receptors: Theme and variations. Oncogene, Peluso, M., et al. (2004). Soluble glycoprotein VI dimer inhibits
20, 15701581. platelet adhesion and aggregation to the injured vessel wall in vivo.
42. Kahn, M. L., Nakanishi-Matsui, M., Shapiro, M. J., Ishihara, H., & The FASEB Journal, 18, 397399.
Coughlin, S. R. (1999). Protease-activated receptors 1 and 4 me- 61. Ungerer, M., Rosport, K., Bltmann, A., Piechatzek, R.,
diate activation of human platelets by thrombin. The Journal of Uhland, K., Schlieper, P., et al. (2011). Novel antiplatelet
Clinical Investigation, 103, 879887. drug revacept (dimeric glycoprotein VI-Fc) specifically and
43. Steinhoff, M., Buddenkotte, J., Shpacovitch, V., Rattenholl, A., efficiently inhibited collagen-induced platelet aggregation
Moormann, C., Vergnolle, N., et al. (2005). Proteinase-activated without affecting general hemostasis in humans. Circulation,
receptors: Transducers of proteinase-mediated signaling in inflam- 123, 18911899.
mation and immune response. Endocrine Reviews, 26, 143. 62. Vanhoorelbeke, K., Ulrichts, H., Schoolmeester, A., & Deckmyn,
44. Shapiro, M. J., Weiss, E. J., Faruqi, T. R., & Coughlin, S. R. H. (2003). Inhibition of platelet adhesion to collagen as a new
(2000). Protease-activated receptors 1 and 4 are shut off with target for antithrombotic drugs. Current Drug Targets.
distinct kinetics after activation by thrombin. Journal of Cardiovascular & Haematological Disorders, 3, 125140.
Biological Chemistry, 275, 2521625221. 63. Van Bockstaele, F., Holz, J. B., & Revets, H. (2009). The devel-
45. Newman, P. J., Gorski, J., & White, G. C., 2nd. (1988). Enzymatic opment of nanobodies for therapeutic applications. Current
amplification of platelet-specific messenger RNA using the poly- Opinion in Investigational Drugs, 10(11), 12121224.
merase chain reaction. The Journal of Clinical Investigation, 82, 64. ClinicalTrials.gov. (2009). Comparative study of ALX-0081 ver-
739743. sus GPIIb/IIIa inhibitor in high risk percutaneous coronary inter-
46. Gnatenko, D. V., Dunn, J. J., & McCorkle, S. R. (2003). Transcript vention (PCI) patients. Retrieved from http://clinicaltrials.gov/ct2/
profiling of human platelets using microarray and serial analysis of show/record/NCT01020383.
gene expression. Blood, 101, 22852293. 65. Tricoci, P., Huang, Z., Held, C., Moliterno, D. J., Armstrong, P. W.,
47. Kieffer, N., Guichard, J., & Farcet, J. P. (1987). Biosynthesis of Van de Werf, F., et al. (2012). Thrombin-receptor antagonist vor-
major platelet proteins in human blood platelets. European Journal apaxar in acute coronary syndromes. The New England Journal of
of Biochemistry, 164, 189195. Medicine, 366(1), 2033. Epub 2011 Nov 13.
J. of Cardiovasc. Trans. Res. (2013) 6:299309 309

66. Morrow, D. A., Braunwald, E., Bonaca, M. P., Ameriso, S. F., 74. Kulmacz, R. J., & Wang, L. H. (1995). Comparison of hydroper-
Dalby, A. J., Fish, M. P., et al. (2012). Vorapaxar in the secondary oxide initiator requirements for the cyclooxygenase activities of
prevention of atherothrombotic events. The New England Journal prostaglandin H synthase-1 and -2. Journal of Biological
of Medicine, 366(15), 14041413. Chemistry, 270, 24019.
67. Morrow, D. A., Scirica, B. M., Fox, K. A., Berman, G., Strony, J., 75. Smyth, S. S., Mcever, R. P., Weyrich, A. S., Morrell, C. N.,
Veltri, E., et al. (2009). Evaluation of a novel antiplatelet agent for Hoffman, M. R., Arepally, G. M., et al. (2009). Platelet functions
secondary prevention in patients with a history of atherosclerotic beyond hemostasis. Journal of Thrombosis and Haemostasis, 7,
disease: Design and rationale for the Thrombin-Receptor 17591766.
Antagonist in Secondary Prevention of Atherothrombotic 76. Li, C., Li, J., Li, Y., Lang, S., Yougbare, I., Zhu, G., et al. (2012).
Ischemic Events (TRA 2 degrees P)-TIMI 50 trial. American Crosstalk between platelets and the immune system: Old systems
Heart Journal, 158(335341), e333. with new discoveries. Advances in Hematology, 2012, 384685.
68. Warner, T. D., & Mitchell, J. A. (2004). Cyclooxygenases: New Epub 2012 Sep 12.
forms, new inhibitors, and lessons from the clinic. The FASEB 77. Aslam, R., Speck, E. R., Kim, M., et al. (2006). Platelet Toll-like
Journal, 18, 790804. receptor expression modulates lipopolysaccharide-induced throm-
69. Smith, W. L., & Langenbach, R. (2001). Why there are two cyclo- bocytopenia and tumor necrosis factor- production in vivo.
oxygenase isozymes. The Journal of Clinical Investigation, 107 Blood, 107(2), 637641.
(12), 14911495. 78. Littman, D. R., & Rudensky, A. Y. (2010). Th17 and regulatory T cells
70. Chandrasekharan, N. V., Dai, H., Roos, L. T., Evanson, N. K., in mediating and restraining inflammation. Cell, 140(6), 845858.
Tomsik, J., Elton, T. S., et al. (2002). COX-3, a cyclooxygenase-1 79. Austrup, F., Vestweber, D., Borges, E., et al. (1997). P- and E-
variant inhibited by acetaminophen and other analgesic/antipyretic selectin mediate recruitment of T-helper-1 but not T-helper-2 cells
drugs: Cloning, structure, and expression. Proceedings of the into inflamed tissues. Nature, 385(6611), 8183.
National Academy of Sciences of the United States of America, 80. Elzey, B. D., Tian, J., Jensen, R. J., et al. (2003). Platelet-mediated
99(21), 1392613931. modulation of adaptive immunity: A communication link between
71. Dubois, R. N., Abramson, S. B., Crofford, L., Gupta, R. A., innate and adaptive immune compartments. Immunity, 19(1), 919.
Simon, L. S., Van De Putte, L. B., et al. (1998). Cyclooxygenase 81. Kopp, H. G., Placke, T., & Salih, H. R. (2009). Platelet-derived
in biology and disease. The FASEB Journal, 12(12), 10631073. transforming growth factor- down-regulates NKG2D thereby
72. Xie, W., Chipman, J. G., Roberston, D. L., Erikson, R. L., & inhibiting natural killer cell antitumor reactivity. Cancer
Simmons, D. L. (1991). Expression of a mitogen-responsive gene Research, 69(19), 77757783.
encoding prostaglandin synthase is regulated by mRNA splicing. 82. Baenziger, N. L., Brodie, G. N., & Majerus, P. W. (1971). A
Proceedings of the National Academy of Sciences of the United thrombin-sensitive protein of human platelet membranes.
States of America, 88(7), 26822686. Proceedings of the National Academy of Sciences of the United
73. Smith, W. L., & DeWitt, D. L. (1995). Biochemistry of prosta- States of America, 68(1), 240243.
glandin endoperoxide H synthase and their differential susceptibil- 83. Crawford, S. E., Stellmach, V., Murphy-Ullrich, J. E., et al. (1998).
ity to nonsteroidal anti-inflammatory drugs. Seminars in Thrombospondin-1 is a major activator of TGF-1 in vivo. Cell,
Nephrology, 15, 179194. 93(7), 11591170.

You might also like