You are on page 1of 11

ANTIPLATELET THERAPY

The Role of Platelets in Atherothrombosis


Zane S. Kaplan1 and Shaun P. Jackson1

1Australian
Centre for Blood Diseases, Alfred Medical Research and Education Precinct, Monash University,
Melbourne, Australia

Platelets have evolved highly specialized adhesion mechanisms that enable cell-matrix and cell-cell interactions
throughout the entire vasculature irrespective of the prevailing hemodynamic conditions. This unique property of
platelets is critical for their ability to arrest bleeding and promote vessel repair. Platelet adhesion under conditions of
high shear stress, as occurs in stenotic atherosclerotic arteries, is central to the development of arterial thrombosis;
therefore, precise control of platelet adhesion must occur to maintain blood fluidity and to prevent thrombotic or
hemorrhagic complications. Whereas the central role of platelets in hemostasis and thrombosis has long been
recognized and well defined, there is now a major body of evidence supporting an important proinflammatory function
for platelets that is linked to host defense and a variety of autoimmune and inflammatory diseases. In the context of the
vasculature, experimental evidence indicates that the proinflammatory function of platelets can regulate various
aspects of the atherosclerotic process, including its initiation and propagation. The mechanisms underlying the
proatherogenic function of platelets are increasingly well defined and involve specific adhesive interactions between
platelets and endothelial cells at atherosclerotic-prone sites, leading to the enhanced recruitment and activation of
leukocytes. Through the release of chemokines, proinflammatory molecules, and other biological response modula-
tors, the interaction among platelets, endothelial cells, and leukocytes establishes a localized inflammatory response
that accelerates atherosclerosis. These inflammatory processes typically occur in regions of the vasculature
experiencing low shear and perturbed blood flow, a permissive environment for leukocyte-platelet and leukocyte-
endothelial interactions. Therefore, the concept has emerged that platelets are a central element of the atherothrom-
botic process and that future therapeutic strategies to combat this disease need to take into consideration both the
prothrombotic and proinflammatory function of platelets.

Introduction leukocyte interaction. The importance of local blood flow distur-


Platelets are anuclear cell fragments derived from the cytoplasm of bances in regulating the atherothrombotic process is briefly dis-
BM megakaryocytes1 that circulate in the bloodstream of humans cussed. A brief overview of the well-defined platelet adhesion
for 7-10 days. The primary hemostatic function of platelets has been mechanisms regulating platelet-vessel wall and platelet-platelet
recognized for more than a century, and the molecular processes interactions and a discussion of recent advances in the understand-
regulating the adhesive function of platelets have been elucidated in ing of the platelet procoagulant response are presented first.
great detail.2-4 In addition to preventing excess bleeding, platelets
also play an important role in surveying and maintaining the Hemostatic and prothrombotic function of platelets
integrity of the endothelium, in part through the release of proangio- Under normal physiological conditions, platelets circulate in close
genic cytokines and growth factors. Dysregulated platelet- proximity to the endothelium without forming stable adhesion
endothelial interactions have increasingly been recognized as an contacts due to the anti-adhesive properties of quiescent endothelial
important pathogenic mechanism in the development of atheroscle- cells. However, after vascular injury, platelets rapidly adhere to sites
rosis. In response to specific proinflammatory signals, endothelial of endothelial disruption to establish a hemostatic plug that prevents
cells become more adhesive toward platelets, stimulating the excess blood loss. This process is critically dependent on the
production of various platelet-derived inflammatory molecules that efficiency of platelet adhesion to the subendothelial matrix, as well
provide a positive feedback loop for further endothelial cell as on the ability of the platelets to undergo rapid biochemical and
activation. Endothelial-bound platelets are highly effective at recruit- morphological changes that support aggregation and the localized
ing leukocytes from flowing blood and also enhance leukocyte activation of the coagulation cascade.
adhesion and transmigration to the site of the proinflammatory
stimulus. Therefore, pathological derangement of these key interac- Platelet recruitment (Figure 1)
tions among platelets, endothelial cells, and leukocytes facilitates The mechanisms supporting platelet adhesion to the damaged vessel
the inflammatory process that underlies the development of wall have been thoroughly investigated and well defined, so only a
atherosclerosis. brief description will be provided here. After damage to the vessel
wall, subendothelial matrix proteins that are highly reactive to
This review outlines the key pathways used by platelets to facilitate platelets, including collagen, VWF, fibronectin, and laminin, be-
the development of vascular inflammatory lesions linked to the come exposed to the blood and immediately engage specific
development of atherosclerosis, with a focus on the mechanisms receptors on the platelet surface. The contribution of specific ligands
leading to dysregulation of the platelet-endothelium and platelet- and receptors mediating platelet adhesion is critically dependent on

Hematology 2011 51
Figure 1. Hemostatic and prothrombotic function of platelets under flow. (A) Platelet adhesion mechanisms operating under arterial flow. After
disruption of the endothelium, platelets are rapidly recruited from flowing blood via a tethering mechanism dependent on the interaction of platelet
GPIb-V-IX and VWF. This adhesive bond has a rapid dissociation rate, resulting in platelet translocation on the vessel wall. (B) Platelet firm adhesion
and aggregation. Translocating platelets engage collagen in the vessel wall through their adhesion receptors GPVI and 21. GPVI is the major
collagen receptor inducing intracellular calcium flux necessary for stable platelet adhesion, cytoskeletal reorganization, IIb3 activation, and the release
of the soluble agonists (ADP and TxA2). These agonists act through specific G-protein coupled receptors to amplify the platelet activation response.
TxA2 is derived from the metabolism of arachidonic acid via the COX-1 pathway, whereas ADP is released from platelet-dense granules. Locally
generated thrombin also enhances platelet activation through proteolytic cleavage and activation of PARs. Activated platelets form stable aggregates
through IIb3 engagement of VWF and fibrinogen. These molecular pathways of amplification of platelet activation are the target of both clinically
available and experimental therapeutic interventions in the treatment of atherothrombotic disease. (C) Stabilization of the platelet thrombus. The primary
hemostatic plug is consolidated by fibrin generation at the site of injury and throughout the developing thrombus. -Thrombin generation at the injured
vessel wall is critically dependent on TF, whereas thrombin generation on the surface of activated platelets is likely to evolve both TF and contact
factor dependent activation of blood coagulation. UHF indicates unfractionated heparin; LMWH, low-molecular-weight heparin; VKA, vitamin K
antagonist; PGH2, prostaglandin H2; AA, arachidonic acid, PLA2, phospholipase A2; IP3R, inositol trisphosphate receptor; ITAM, immunoreceptor
tyrosine-based activation motif; FcR, Fc receptor gamma.

the prevailing blood-flow conditions. Under conditions of high component of the platelet GPIb-V-IX complex.6 Circulating plasma
shear stress, as encountered in arterioles and stenotic arteries, VWF VWF has limited binding potential for GPIb, however, once
plays a critical role in recruiting platelets from blood flow.5 This immobilized onto subendothelial collagen (type I, III, and VI), the
tethering function of subendothelial VWF is dependent on the unfolded VWF macromolecule provides a linear array of A1
interaction of its A1 domain with the glycoprotein Ib (GPIb) domains that facilitate binding to multiple GPIb receptors.7,8 The

52 American Society of Hematology


bond between GPIb and VWF has a rapid dissociation rate that is cascade, thus affecting thrombin generation at the vessel wall as
unable to support stable adhesion, requiring the contribution of well as within the body of thrombus. However, recent progress in
other ligand-receptor interactions to promote stable adhesion.9 The understanding the mechanisms by which platelets support blood
fundamental importance of both VWF and GPIb for the adhesive coagulation have raised the possibility that selectively inhibiting
function of platelets is underscored by the severe bleeding disorder platelet procoagulant function may provide a targeted approach to
experienced by patients with qualitative or quantitative defects in specifically reduce thrombin generation within the thrombus.
these molecules.10
Platelet regulation of the contact phase of blood
Firm adhesion and activation (Figure 1) coagulation (Figure 2)
Once tethered to the site of vascular injury, platelets form stable The generation of thrombin at the injured vessel wall is contingent
adhesion contacts with collagen and other matrix macromolecules. upon the expression of tissue factor (TF) on the surface of
Platelet binding to collagen is mediated by GPVI11,12 and integrin fibroblasts, smooth muscle cells, and potentially endothelial cells
21,13 whereas fibronectin and laminin engage integrin 51 and and leukocytes. TF expression and activation helps to stabilize the
61, respectively.4 Isolated deficiency of any of these receptors active conformation of FVIIa,30 triggering the extrinsic pathway of
does not cause a severe bleeding disorder, suggesting considerable blood coagulation. The mechanisms underlying the generation of
redundancy in the mechanisms regulating platelet-vessel wall thrombin within the body of a developing thrombus remains
interactions. Once adherent, platelets undergo a remarkably com- controversial. Whereas TF is expressed on the surface of activated
plex series of morphological and biochemical changes that lead to leukocytes and microparticles and has been demonstrated to be
the release of platelet granule contents and up-regulation of the incorporated within the thrombus, recent experimental evidence has
adhesive function of integrin IIb3. Activated IIb3 binds multiple suggested a potentially important role for the contact phase of blood
ligands, including VWF,14,15 fibrinogen,16 fibrin and fibronectin,17 coagulation in promoting arterial thrombus growth. This rekindled
and is indispensable for the formation of stable platelet aggregates.18 interest in the role of contact factors (eg, FXII, FXI, and high-
Therefore, similar to GPIb, quantitative or qualitative defects in molecular-weight kininogen) in promoting blood coagulation in
IIb3 leads to a major defect in the hemostatic function of platelets vivo stemmed from the observation that FXII-deficient mice are
and a severe bleeding disorder (Glanzmann thrombasthenia).19 protected from vasoocclusive thrombi in arterial thrombosis mod-
Integrin IIb3 deficiency also provides protection against arterial els.31 Similarly, FXI deficiency32,33 and pharmacological antago-
thrombosis, thus pharmacological IIb3 antagonism represents a nism34 of the contact phase of coagulation leads to a similar defect in
highly effective antithrombotic approach.20,21 arterial thrombosis. Given that humans with complete FXII defi-
ciency do not have a significant hemostatic defect, this has raised the
Soluble agonists amplifying platelet activation (Figure 1) possibility that therapeutic targeting of this process may lead to the
Central to platelet activation is the generation and release of soluble development of safer antithrombotics. How platelets activate the
agonists at sites of vascular injury. These include thromboxane A2 contact phase of coagulation during thrombus development has
(TxA2) which is synthesized from arachidonic acid via cyclooxygen- remained a vexing issue.35 Recently, Muller et al identified inor-
ase (COX) and thromboxane synthase, and ADP released from ganic polyphosphate released from activated platelets as a major
platelet dense granules. These endogenous agonists can act in an regulator of FXII activation.36 Pharmacologic exploitation of this
autocrine or paracrine manner to enhance platelet activation by pathway may enable targeting of platelet polyphosphate by phospha-
engaging specific G protein coupled receptors; P2Y122 and P2Y12 23 tases to reduce thrombotic risk while leaving hemostatic responses
(ADP) and the thromboxane receptors TP and TP.24 ADP and intact.
TxA2 act in a cooperative manner to enhance platelet activation and
thrombus formation, thus pharmacological targeting of TxA225 Identification of the elusive phospholipid scramblase
generation and/or the P2Y1226 receptor are effective strategies to The contribution of platelet-derived phospholipids to thrombin
reduce thrombus propagation at sites of vascular injury.27,28 generation has long been recognized; however, the biological
mechanisms regulating this procoagulant platelet function have
remained poorly defined. In resting platelets, membrane phospholip-
Thrombin (Figure 1) ids are asymmetrically distributed: phosphatidylcholine and sphin-
In addition to the synthesis and release of soluble agonists, platelets gomyelin are primarily localized in the outer leaflet of the plasma
provide a catalytic surface for the assembly of coagulation com- membrane, whereas phosphatidylethanolamine and phosphatidylser-
plexes necessary for -thrombin generation. Thrombin is among the ine (PS) are largely confined to the inner leaflet. This asymmetry is
most potent stimulators of platelets through proteolytic cleavage maintained through the action of two membrane lipid transporters,
and activation of platelet protease-activated receptors (PARs), flippase and floppase.37,38 After platelet stimulation by potent
specifically PAR1 and PAR4, on human platelets.29 Also central to agonists, this asymmetry is lost due to activation of Ca2-dependent
the hemostatic function of thrombin is its ability to generate fibrin phospholipid scramblases that bidirectionally transport phospholip-
polymers, a key step stabilizing formed thrombi. Therefore, pharma- ids.39 Scramblase activity therefore results in the expression of
cological inhibitors of thrombin (eg, direct thrombin inhibitors), negatively charged phospholipids, especially PS, on the outer leaflet
upstream coagulation proteases (vitamin K antagonists and factor of the platelet membrane, thus providing the requisite surface for
Xa [FXa] inhibitors), and, potentially, thrombin receptors (PAR1 assembly of coagulation factors required for thrombin generation
antagonists), represent effective strategies to limit arterial thrombus (Figure 2). Defects in platelet PS expression are rare; however, in
growth.27 affected individuals, this leads to a major hemostatic disturbance, as
described in Scott syndrome.40 Until recently, the identity of the
Procoagulant function of platelets: emerging phospholipid scramblase had remained elusive, but recent studies
concepts have identified a plasma membrane protein transmembrane protein
Current therapeutic strategies aimed at reducing thrombins effects 16F (TMEM16F) that confers Ca2-dependent scrambling of phos-
in vivo primarily target various components of the coagulation pholipids. A mutation in the TMEM16F gene resulting in premature

Hematology 2011 53
Figure 2. Procoagulant function of platelets. (A) Plasma membrane phospholipids are asymmetrically distributed in resting platelets, with PS
confined to the inner leaflet. After platelet activation by potent agonists, high levels of sustained intracellular calcium triggers scramblase activity.
Scramblase activation results in the loss of phospholipid asymmetry and PS exposure on the outer leaflet of the platelet membrane, providing the
requisite surface for rapid thrombin generation. (B) PS exposure can be induced through two distinct cell-death pathways; apoptosis and necrosis.
Apoptosis results in assembly of the Bak/Bax mitochondrial membrane pore, causing release of cytochrome C (CytC), caspase activation, and
substrate proteolysis. Necrotic cell death can be initiated by elevated cytosolic Ca2, causing a loss in mitochondrial membrane potential and
subsequent formation of a mitochondrial permeability transition pore (mPTP). mPTP formation results in bioenergetic failure through ATP depletion with
consequent reactive oxygen species (ROS) generation, leading to loss of membrane integrity. (C) Platelets can also contribute to thrombin generation
via activation of FXII through the release of polyphosphate from dense granules. (D) Whether there are additional cell death pathways regulating PS
exposure and the procoagulant function of platelets remains unclear. PC indicates phosphatidylcholine.

termination of the protein has been demonstrated in a patient with instability and the release of intracellular contents into the extracel-
Scott syndrome.41 lular environment. Recently, evidence has emerged that platelet
stimulation by potent agonists initiates a tightly regulated cell death
Cell death and the procoagulant function of platelets pathway mediated by sustained high levels of cytosolic Ca2,
(Figure 2) culminating in distinct morphological and biochemical changes that
Procoagulant platelets expressing PS exhibit distinct morphologic are consistent with a necrotic cell death pathway.43 Direct induction
and biochemical features relative to fully activated platelets. Many of apoptosis can also induce PS expression and platelet procoagu-
of these changes are indicative of dead or dying cells, and include lant function,43 raising the possibility that multiple cell death
membrane blebbing, microvesiculation, activation of intracellular pathways may regulate thrombin generation on the surface of
proteases, and the cleavage of cytoskeletal proteins, PS surface platelets. In addition to promoting thrombin generation, procoagu-
exposure, and alterations in mitochondrial membrane potential.42 lant platelets may also contribute to the inflammatory response by
These results suggest that the pathways regulating cell death in generating high levels of the proinflammatory lipid platelet-
platelets may play an important role in regulating the platelet activating factor (PAF) and by enhancing leukocyte adhesion.44
procoagulant phenotype. Programmed cell death is orchestrated by
several pathways, including apoptosis, necrosis, and autophagy. Proinflammatory role of platelets
Apoptosis is an energy-dependent process contingent upon caspase Promiscuous platelets
activation and is critical for the removal of senescent cells. Necrosis The growing list of pathophysiological processes in which platelets
is typified by bioenergetic failure of the cell resulting in membrane have a proposed role is in part a reflection of the large number of

54 American Society of Hematology


Figure 3. Platelet-endothelial cell interactions. (A) The healthy endothelium. The anti-adhesive phenotype of endothelial cells is maintained through
3 intrinsic pathways: the ecto-ADPase/CD39/NTPDase pathway, which metabolizes ADP, and the PGI2 and NO pathways, which inhibit platelet
activation through the stimulation of cAMP and cGMP production, respectively. (B) Endothelial activation in hyperlipemia. In a hyperlipidemic milieu, the
endothelium becomes inflamed as a result of modified lipoprotein particles and reactive oxygen species (ROS) accumulating in the intima. This leads to
the expression of adhesive ligands (VWF and P- and E-selectins) on the endothelium. (C) Platelet adhesion to the inflamed endothelium and platelet
endothelial cross-talk. The expression of VWF and P-selectin on the endothelial surface supports platelet tethering and rolling. Subsequently, stable
platelet adhesion occurs through fibrinogen-IIb3 complexes binding to v3 or ICAM-1 on endothelial cells. Adherent platelets secrete numerous
bioactive substances that alter the chemotactic and adhesive properties of the endothelial cells. Platelet-derived IL-1 induces endothelial secretion of
IL-6 and IL-8 and surface expression of ICAM-1, v3, and MCP-1. Platelet CD40 ligand binds CD40 on endothelial cells, resulting in up-regulation of
adhesive molecules (ICAM-1, VCAM-1, and E- and P-selectin), cytokine and TF release, and reduction in NO synthesis. Ecto ADPase indicates
ecto-adenosine diphosphatase; NTPDase, nucleoside triphosphate diphosphohydrolase.

different cell types with which platelets can interact. These include for P-selectin include GPIb and P-selectin glycoprotein-1 (PSGL-
endothelial cells, neutrophils, monocytes, dendritic cells, cytotoxic 1),51,52 which support tethering and rolling, but not firm adhesion on
T-lymphocytes, malaria-infected red blood cells, and various tumor inflamed endothelium. Whereas there is some uncertainty as to the
cells. In the context of atherosclerosis, the interaction of platelets exact mechanism supporting stable platelet adhesion to activated
with endothelial cells and leukocytes appears to be important for the endothelium, in vitro and in vivo models suggest that binding of
initiation and propagation of inflammatory processes in the arterial IIb3 to fibrinogen bound to endothelial v3 or ICAM-1 facilitates
wall.45 firm platelet adhesion and activation.53,54

Platelet-endothelial cell adhesive interactions (Figure 3) Chemical signals


While platelets continuously circulate in close proximity to the Platelets activated through their interactions with inflamed endothe-
endothelium, under physiological conditions, they typically do not lium release a vast array of inflammatory and mitogenic mediators
form sustained adhesive interactions. The anti-adhesive phenotype (Table 1) into the local microenvironment.55 These platelet-derived
of endothelial cells is controlled by the three intrinsic pathways; the bioactive substances alter the chemotactic and adhesive properties
nitric oxide (NO) pathway, the ecto-ADPase/CD39/NTPDase path- of the endothelial cells, enabling monocytes and other leukocytes to
way, and the ecosanoid-arachidonic acid-prostacyclin (PGI2) path- adhere and transmigrate through the endothelium to sites of
way.46 It is increasingly recognized that inflammatory stimuli inflammation.48,56 In vivo models of atherogenesis suggest that
facilitate sustained endothelial-platelet adhesion by perturbing these these interactions between endothelial cells and activated platelets
anti-adhesive mechanisms and increasing surface expression of are likely to be critical in the initiation of atherosclerosis.57,58
endothelial adhesion molecules.47 The adhesion of platelets to
inflamed endothelium involves a similar coordinated multistep IL-I (Figure 3). IL-I is a platelet derived cytokine that has been
process as occurs in hemostasis and thrombosis, including platelet postulated to be a principal mediator of platelet-induced endothelial
tethering, surface translocation, and firm adhesion.48 Initiation of the activation.59,60 It is not stored in platelet granules, but is believed to
inflammatory insult to the endothelium in atherosclerosis is partly be synthesized by platelets from constitutional prepackaged mRNA
due to the accumulation of modified oxidized lipoprotein particles several hours after thrombin stimulation or integrin-mediated adhe-
in the setting of hyperlipidemia.49 This leads to surface expression sion.61 IL-I stimulation of endothelial cells induces secretion of
of endothelial selectins, including P- and E-selectin, as well as IL-6 and IL-8 and significantly increases the endothelial surface
membrane expression of VWF.48,50 The platelet counterreceptors expression of ICAM-1, v3, and monocyte chemotactic protein-1

Hematology 2011 55
Table 1. Bioactive platelet mediators underlying atherothrombosis
Localization within the
platelet Released molecule Target cell Function
Dense granules ADP Platelet P2Y1 and P2Y12 Platelet activation
ATP Platelet P2X1 Platelet activation and sensitization to other
agonists
5HT Platelet 5HT2A Weak platelet agonist
Endothelium 5HT2B NO synthesis: vasodilation, platelet inhibitor
Vascular smooth muscle Vasoconstriction
Monocytes T-cell activation: proinflammatory
granules
Adhesion receptors IIb3 Platelet Platelet aggregation, platelet-endothelial, and
platelet-leukocyte adhesion
Coagulation factors FV Procoagulant
Adhesive ligands VWF Platelet Platelet-endothelium and platelet-vessel wall
tethering and platelet aggregation
Fibrinogen Platelet Platelet aggregation, fibrin clot formation, and
platelet-leukocyte adhesion
Fibronectin Platelet Platelet-endothelial and platelet-leukocyte
adhesion
P-selectin Platelets, endothelial cell and Adhesion of leukocytes to platelets and
leukocytesPSGL-1 endothelial cells
Chemokines CXCL4 (PF-4) Leukocytes Synergizes with other cytokines to promote
leukocyte adhesion and monocyte
differentiation
CXCL7 (PBP, -TG, CTAP-III, Leukocytes Enhances neutrophil and monocyte adhesion
and NAP-2) and neutrophil transendothelial migration
CCL5 (RANTES) Endothelium/leukocytes Promotes monocytes adhesion to the
endothelium and chemokine synthesis
Growth factors PDGF Vascular smooth muscle Stimulates vascular smooth muscle cell
cells migration and proliferation associated with
intimal hyperplasia
Synthesized cytokines IL-1 Endothelial cells Up-regulation of leukocyte adhesion molecules
and stimulation of endothelial release of
proinflammatory cytokines
granule and platelet
membrane
CD40 Ligand Endothelial cell CD40 Upregulation of leukocyte adhesion molecules
and stimulation of endothelial release of
proinflammatory cytokines
Proinflammatory lipids TxA2 Platelets Platelet activation
Leukotriene B4 Leukocytes (BLT-1 and Chemo-attractant and promotion of monocyte
BLT-2) differentiation into foam cells and neutrophil
activation
PAF Platelet/leukocytes Platelet agonist and promotes neutrophil
adhesion

(MCP-1).48,60,62 ICAM-1 and MCP-1 up-regulation by endothelial with its cognate receptor CD40 results in up-regulation of ICAM-1,
cells in response to IL-I is dependent on the activation of NF-B.63 VCAM-1, and E- and P-,selectin as well as the release of IL-6 and
Through this potential mechanism, platelets are able to significantly TF.69 CD40L inhibits endothelial NO production by destabilizing
enhance monocyte and neutrophil adhesion to the endothelium. NO synthase mRNA64,70; therefore, CD40L binding to endothelial
cells results in a proatherogenic and proinflammatory phenotype.64
CD40 ligand (CD40L-CD154; Figure 3). CD40 ligand is an
important platelet-derived bioactive mediator of endothelial cell Platelet-leukocyte cross-talk in atherogenesis
activation. CD40L is a transmembrane protein with structural The central role of leukocytes, especially monocytes and T lympho-
homology to TNF-.64 CD40L was initially identified on the surface cytes, in the development of atherosclerosis is well established.71
of activated CD4 T cells and is central to the function of the There is increasing evidence that activated platelets adherent to the
humoral immune system by supporting B-cell differentiation and inflamed endothelium may enhance leukocyte recruitment, activa-
immunoglobulin class switching.65 It was subsequently demon- tion, and transmigration, thereby enhancing the inflammatory
strated that platelets are a rich source of CD40L,66 with 95% of processes underlying atherosclerosis.
circulating CD40L being of platelet origin.67 After release from
platelets, CD40L interacts which CD40 expressed on numerous cell Leukocyte recruitment to endothelial-bound platelets is a coordi-
types, including endothelial cells, smooth muscle cells, leukocytes, nated, multistep process involving selectin-mediated tethering and
fibroblasts, and platelets.64,68 In endothelial cells, CD40L binding rolling, followed by 2 integrinmediated stable adhesion (Figure

56 American Society of Hematology


Figure 4. Platelet-leukocyte interactions promoting atherogenesis. (A) Platelet-mediated leukocyte recruitment and adhesion. Leukocyte
recruitment to endothelial-bound platelets occurs in a multistep coordinated process. Initial tethering of leukocytes is mediated by the interaction of
P-selectin expressed on the platelet surface with its cognate receptor leukocyte PSGL-1. Ligation of PSGL-1 promotes activation of leukocyte 2
integrins (Mac-1 and LFA-1), necessary for stable leukocyte adhesion. Note that Mac-1 can engage several platelet ligands, including GPIb, ICAM-2,
and JAM-3, as well as fibrinogen bound to IIb3. Release of the chemokine RANTES at sites of atherogenesis leads to enhanced monocyte adhesion.
Monocytes subsequently migrate through the endothelium and differentiate into macrophages, which engulf lipids and transform into foam cells.
(B) Platelet-leukocyte cross-talk. Bioactive mediators derived from -granules of activated platelets, including PF4, synergize with RANTES to enhance
leukocyte adhesion and monocyte differentiation. RANTES, acting in concert with P-selectin, results in MCP-1 and IL-8 secretion by monocytes.
CTAP-III is another chemokine released from the platelet -granules, which is subsequently converted into active NAP-2 by the neutrophil membrane
associated serine-protease cathepsin G. NAP-2 enhances leukocyte adhesion and neutrophil transendothelial migration. Up-regulation of COX-2 in
monocytes via pathways involving IL-I and P-selectin results in increased production of proinflammatory mediators, including PAF, leukotrienes, and
TxA2 via transcellular eicosanoid metabolism. P-sel indicates P-selectin; AA, arachidonic acid.

4). Platelet P-selectin has a central role in mediating the initial displayed on the platelet surface, where they exert numerous
capture and rolling of leukocytes through interactions with its key biological activities instrumental in atherosclerosis.77,78
counterreceptor P-selectin glycoprotein 1 (PSGL-1).51,72 Ligation of
PSGL-1 induces signaling events culminating in the activation of
the leukocyte 2 integrins, including macrophage antigen-1 (Mac-1, CXC chemokines NAP-2 and PF4 (Figure 4)
M2) and lymphocyte function-associated antigen-1 (LFA-1, L2), The two most abundant CXC chemokines stored in platelet
which are required to mediate stable leukocyte adhesion. Mac-1 is granules are CXCL7 and platelet factor 4 (PF4). CXCL7 has
the principal 2 integrin that facilitates firm adhesion onto the several molecular variants, including the inactive precursor, connec-
surface of platelets via its binding to multiple ligands, including tive tissue-activating peptide III (CTAP-III), which is enzymatically
GPIb,73 ICAM-2,74 and junctional adhesion molecule-3 (JAM- converted into active neutrophil activating protein-2 (NAP-2)
3),75 as well as through fibrinogen bound to IIb3.48,76 through proteolysis by the neutrophil membrane-associated serine-
protease cathepsin G.79 NAP-2 has been demonstrated to induce
Platelet activation with consequent degranulation results in the neutrophil and monocyte adhesion in vitro and neutrophil transendo-
liberation of numerous chemokines stored in platelet -granules. thelial migration.78,80 The chemotactic properties of PF4 on its own
These chemokines can either be released into the circulation or are controversial because it does not exert classical chemokine

Hematology 2011 57
functions at submicromolar concentrations.78,81 However, PF4 act- Shear effects on the endothelium
ing in concert with other chemokines such as the platelet-derived Endothelial cells in arteries are constantly exposed to dynamic
CC chemokine regulated upon activation normal T-cell expressed alterations in shear due to the pulsatile nature of blood flow.90
and secreted (RANTES) may enhance leukocyte adhesion and However, the rate of fluctuation in blood flow appears to be the
promote monocyte differentiation and atherosclerosis. These PF4- major parameter altering endothelial cell function. Endothelial cells
RANTES heterodimers have been proposed to represent potential respond to flow changes through a variety of mechanotransduction
therapeutic targets in the treatment of atherosclerosis.82 mechanisms, including mechanically gated ion channels and G pro-
teins, force-dependent alterations in membrane fluidity, nuclear
CC chemokine RANTES (Figure 4) deformation, cytoskeletal-dependent junctional signaling, and via
The proinflammatory CC chemokine RANTES is released from primary cilia and the glycocalyx.91 These mechanosensory signaling
activated platelets and deposited on activated endothelium, where it mechanisms are exquisitely sensitive to changes in wall shear stress,
is able to promote adhesion of monocytes.83 A prerequisite for the in leading to alterations in cell morphology, gene-expression profiles,
vivo activity of RANTES function is for it to bind to glycosamino- and increased adhesiveness.92
glycans and to form higher-order oligomers.84 Further, through its
interactions with other modulators, including MCP-4, PF4, and Atheromatous lesions in permissive flow environments for
P-selectin, RANTES has been implicated in inflammation, athero-
leukocyte adhesion
genesis, and vascular remodeling after injury. Interestingly, P-
The mechanisms by which disturbed blood flow influences leuko-
selectin engagement with PSGL-1 on monocytes is insufficient for
cyte adhesion mechanisms remain ill defined. In contrast to
the induction of MCP-1, requiring the cooperation of platelet-
platelets, leukocytes adhere preferentially at low shear regions in the
derived RANTES for chemokine synthesis by monocytes.85 Further,
vasculature, typically at wall shear rates 1000/s. Whereas a
monocytes costimulated by RANTES and P-selectin secrete a
minimum threshold shear is required for leukocyte adhesion,93 there
different cytokine profile, including MCP-1 and IL-8.85 Platelet
is typically an inverse correlation between wall shear and the
activation results in the release of microparticles that are able to
efficiency of leukocyte recruitment, with optimal adhesion around
facilitate the deposition of RANTES on the inflamed and atheroscle-
150/s.93 The site density of P- and E-selectin molecules also plays a
rotic endothelium, thus promoting monocyte adhesion.86
major role in regulating the efficiency of leukocyte adhesion;
therefore, the prevailing flow conditions and the degree of P-selectin
Proinflammatory lipids (Figure 4) expression on the surface of activated endothelial cells and endothe-
In addition to being storage houses for cytokines and chemokines, lial-bound platelets are likely to be major determinants regulating
platelets also generate lipid-derived inflammatory mediators. The leukocyte recruitment to atherogenesis-prone sites.94 Experimental
adhesion of platelets to leukocytes and the endothelium results in models of disturbed blood flow lead to enhanced leukocyte accumu-
transcellular metabolism of eicosanoids, leading to the local synthe- lation in low shear recirculation zones,95 supporting the concept that
sis of proinflammatory lipids such as PAF, leukotrienes, and flow disturbances may facilitate leukocyte recruitment to atheroscle-
TxA2.87 The imbalance of inflammatory mediators is further exacer- rotic sites.
bated by monocyte up-regulation of COX-2 expression via interac-
tions involving P-selectin derived from activated platelets and
IL-I. This up-regulation of COX-2 enables further synthesis of Impact of high shear and disturbed blood flow on platelet
proinflammatory eicosanoids.88 PAF is a potent platelet agonist and adhesive function
inflammatory mediator and has also been shown to regulate firm The importance of shear forces in regulating platelet adhesive
neutrophil adhesion on the surface of immobilized spread platelets.44 function has long been recognized. At normal wall shear rates
(20-2000/s), shear does not stimulate the activation of platelets in
Atherothrombosis and disturbed blood flow suspension. However at high stress rates ( 5000/s) as occurs in
An important determinant influencing platelet-endothelial and plate- stenosed atherosclerotic arteries, shear can directly induce platelet
let-leukocyte interactions at atherosclerotic-prone sites is the local activation.5 Based on in vitro findings, shear-induced platelet
blood flow conditions. It is well established that atherosclerotic activation is critically dependent upon the sequential binding of
lesions develop at arterial branch points, which are typically areas of VWF to GPIb and integrin IIb3, as well as on the release of
low shear and disturbed flow. Altered flow has a significant effect on ADP.96 Shear-induced platelet activation is insensitive to COX
the adhesive properties of platelets and leukocytes, as well on the inhibitors, a finding that may partly explain the relative weak
morphology and function of endothelial cells. Rheological factors antithrombotic effect of aspirin.97 In a complex shear environment
also contribute to the deposition of lipids and other modulators of characterized by rapid phases of shear acceleration and deceleration,
atherosclerosis. platelet aggregation can occur independently of ADP and TxA2,98
raising the possibility that shear gradients can promote platelet
Throughout much of the vasculature, blood flow is laminar, with deposition and initial thrombus growth even in the presence of
adjacent fluid layers traveling parallel to each other but at different aspirin and a P2Y12 inhibitor.
velocities because of the fluid drag exerted by the vessel wall. This
phenomenon leads to the generation of shear forces between Conclusion
adjacent fluid planes. These flow profiles apply to straight, healthy It has long been recognized that arterial thrombosis primarily
vesselsat arterial branch points, curvatures, and areas of stenosis, represents an exaggerated physiological hemostatic response at sites
flow profiles become significantly more complex. These alterations of atherosclerotic plaque disruption. A similar concept can be
in flow produce shear gradients, turbulence, flow separation, and developed for the role of platelets in the initiation and propagation
eddy formation, each of which can affect the atherogenic process. of atherosclerosis. Platelets normally interact with endothelial cells
Progression of the atherosclerotic lesion exacerbates flow distur- to preserve their physiological function and to enhance leukocyte
bances, thus inducing a potential hazardous cycle of shear- recruitment to sites of inflammation. An exaggeration of these
dependent acceleration of atherosclerosis.89 physiological responses at atherosclerosis-prone sites appears to

58 American Society of Hematology


play an important role in the pathogenesis of this disease. This nia: deficient binding of von Willebrand factor to thrombin-
improved understanding of the role of platelets in the development stimulated platelets. Proc Natl Acad Sci U S A. 1982;79(19):
of atherothrombosis and elucidation of the key pathways involved 6038-6041.
should stimulate further investigation into the possibility of target- 15. Hantgan RR. Fibrin protofibril and fibrinogen binding to
ing both the proinflammatory and prothrombotic function of plate- ADP-stimulated platelets: evidence for a common mechanism.
lets to reduce the clinical complications of this major disease. Biochim Biophys Acta. 1988;968(1):24-35.
16. Bennett JS, Vilaire G. Exposure of platelet fibrinogen receptors
Acknowledgments by ADP and epinephrine. J Clin Invest. 1979;64(5):1393-1401.
We thank Dr Simone Schoenwaelder for assistance with the 17. Ni H, Denis CV, Subbarao S, et al. Persistence of platelet
preparation of the figures. Z.S.K. is an National Health and Medical thrombus formation in arterioles of mice lacking both von
Research Council (NHMRC) and National Heart Foundation post- Willebrand factor and fibrinogen. J Clin Invest. 2000;106(3):385-
graduate scholarship holder. S.P.J. is a NHMRC Australia Fellow. 392.
18. Jackson SP. The growing complexity of platelet aggregation.
Disclosures Blood. 2007;109(12):5087-5095.
Conflict-of-interest disclosure: The authors declare no competing 19. George JN, Caen JP, Nurden AT. Glanzmanns thrombasthe-
financial interests. Off-label drug use: None disclosed. nia: the spectrum of clinical disease. Blood. 1990;75(7):1383-
1395.
20. Quinn MJ, Plow EF, Topol EJ. Platelet glycoprotein IIb/IIIa
Correspondence inhibitors: recognition of a two-edged sword? Circulation.
Shaun P. Jackson, Australian Centre for Blood Diseases, 6th Level
2002;106(3):379-385.
Burnet Bldg, AMREP, 89 Commercial Rd, Melbourne, Victoria,
21. Boersma E, Harrington RA, Moliterno DJ, et al. Platelet
Australia 3004; Phone: 61-3-9903-0131; Fax: 61-3-9903-0228;
glycoprotein IIb/IIIa inhibitors in acute coronary syndromes: a
e-mail: shaun.jackson@monash.edu.
meta-analysis of all major randomised clinical trials. Lancet.
2002;359(9302):189-198.
References 22. Jin J, Daniel JL, Kunapuli SP. Molecular basis for ADP-
1. Junt T, Schulze H, Chen Z, et al. Dynamic visualization of induced platelet activation. II. The P2Y1 receptor mediates
thrombopoiesis within bone marrow. Science. 2007;317(5845): ADP-induced intracellular calcium mobilization and shape
1767-1770. change in platelets. J Biol Chem. 1998;273(4):2030-2034.
2. Andrews RK, Lopez JA, Berndt MC. Molecular mechanisms of 23. Hollopeter G, Jantzen HM, Vincent D, et al. Identification of
platelet adhesion and activation. Int J Biochem Cell Biol.
the platelet ADP receptor targeted by antithrombotic drugs.
1997;29(1):91-105.
Nature. 2001;409(6817):202-207.
3. Ruggeri ZM, Mendolicchio GL. Adhesion mechanisms in
24. Huang JS, Ramamurthy SK, Lin X, Le Breton GC. Cell
platelet function. Circ Res. 2007;100(12):1673-1685.
signalling through thromboxane A2 receptors. Cell Signal.
4. Ruggeri ZM. Platelet adhesion under flow. Microcirculation.
2004;16(5):521-533.
2009;16(1):58-83.
25. Collaborative meta-analysis of randomised trials of antiplatelet
5. Savage B, Saldivar E, Ruggeri ZM. Initiation of platelet
therapy for prevention of death, myocardial infarction, and
adhesion by arrest onto fibrinogen or translocation on von
stroke in high risk patients. BMJ. 2002;324(7329):71-86.
Willebrand factor. Cell. 1996;84(2):289-297.
26. A randomised, blinded, trial of clopidogrel versus aspirin in
6. Ruggeri ZM. Structure and function of von Willebrand factor.
Thromb Haemost. 1999;82(2):576-584. patients at risk of ischaemic events (CAPRIE). CAPRIE
7. Siedlecki CA, Lestini BJ, Kottke-Marchant KK, Eppell SJ, Steering Committee. Lancet. 1996;348(9038):1329-1339.
Wilson DL, Marchant RE. Shear-dependent changes in the 27. Michelson AD. Antiplatelet therapies for the treatment of
three-dimensional structure of human von Willebrand factor. cardiovascular disease. Nat Rev Drug Discov. 2010;9(2):154-
Blood. 1996;88(8):2939-2950. 169.
8. Barg A, Ossig R, Goerge T, et al. Soluble plasma-derived von 28. Patrono C, Baigent C, Hirsh J, Roth G. Antiplatelet drugs:
Willebrand factor assembles to a haemostatically active filamen- American College of Chest Physicians Evidence-Based Clini-
tous network. Thromb Haemost. 2007;97(4):514-526. cal Practice Guidelines (8th Ed). Chest. 2008;133(6 suppl):199S-
9. Savage B, Almus-Jacobs F, Ruggeri ZM. Specific synergy of 233S.
multiple substrate-receptor interactions in platelet thrombus 29. Coughlin SR. How the protease thrombin talks to cells. Proc
formation under flow. Cell. 1998;94(5):657-666. Natl Acad Sci U S A. 1999;96(20):11023-11027.
10. Lopez JA, Andrews RK, Afshar-Kharghan V, Berndt MC. 30. Persson E, Olsen OH. Current status on tissue factor activation
Bernard-Soulier syndrome. Blood. 1998;91(12):4397-4418. of factor VIIa. Thromb Res. 2010;125 Suppl 1:S11-12.
11. Moroi M, Jung SM, Shinmyozu K, Tomiyama Y, Ordinas A, 31. Renne T, Pozgajova M, Gruner S, et al. Defective thrombus
Diaz-Ricart M. Analysis of platelet adhesion to a collagen- formation in mice lacking coagulation factor XII. J Exp Med.
coated surface under flow conditions: the involvement of 2005;202(2):271-281.
glycoprotein VI in the platelet adhesion. Blood. 1996;88(6): 32. Salomon O, Steinberg DM, Koren-Morag N, Tanne D, Selig-
2081-2092. sohn U. Reduced incidence of ischemic stroke in patients with
12. Nieswandt B, Watson SP. Platelet-collagen interaction: is severe factor XI deficiency. Blood. 2008;111(8):4113-4117.
GPVI the central receptor? Blood. 2003;102(2):449-461. 33. Rosen ED, Gailani D, Castellino FJ. FXI is essential for
13. Santoro SA. Identification of a 160,000 dalton platelet mem- thrombus formation following FeCl3-induced injury of the
brane protein that mediates the initial divalent cation-dependent carotid artery in the mouse. Thromb Haemost. 2002;87(4):774-
adhesion of platelets to collagen. Cell. 1986;46(6):913-920. 776.
14. Ruggeri ZM, Bader R, de Marco L. Glanzmann thrombasthe- 34. Zhang H, Lowenberg EC, Crosby JR, et al. Inhibition of the

Hematology 2011 59
intrinsic coagulation pathway factor XI by antisense oligonucle- 56. Dav G, Patrono C. Platelet activation and atherothrombosis.
otides: a novel antithrombotic strategy with lowered bleeding N Engl J Med. 2007;357(24):2482-2494.
risk. Blood. 2010;116(22):4684-4692. 57. Massberg S, Brand K, Gruner S, et al. A critical role of platelet
35. Caen J, Wu Q. Hageman factor, platelets and polyphosphates: adhesion in the initiation of atherosclerotic lesion formation. J
early history and recent connection. J Thromb Haemost. Exp Med. 2002;196(7):887-896.
2010;8(8):1670-1674. 58. Belton OA, Duffy A, Toomey S, Fitzgerald DJ. Cyclooxygen-
36. Muller F, Mutch NJ, Schenk WA, et al. Platelet polyphosphates ase isoforms and platelet vessel wall interactions in the
are proinflammatory and procoagulant mediators in vivo. Cell. apolipoprotein E knockout mouse model of atherosclerosis.
2009;139(6):1143-1156. Circulation. 2003;108(24):3017-3023.
37. Daleke DL. Regulation of transbilayer plasma membrane 59. Hawrylowicz CM, Howells GL, Feldmann M. Platelet-derived
phospholipid asymmetry. J Lipid Res. 2003;44(2):233-242. interleukin 1 induces human endothelial adhesion molecule
38. Leventis PA, Grinstein S. The distribution and function of expression and cytokine production. J Exp Med. 1991;174(4):
phosphatidylserine in cellular membranes. Annu Rev Biophys. 785-790.
2010;39:407-427. 60. Kaplanski G, Farnarier C, Kaplanski S, et al. Interleukin-1
39. Bevers EM, Williamson PL. Phospholipid scramblase: an induces interleukin-8 secretion from endothelial cells by a
update. FEBS Lett. 2010;584(13):2724-2730. juxtacrine mechanism. Blood. 1994;84(12):4242-4248.
40. Weiss HJ, Vicic WJ, Lages BA, Rogers J. Isolated deficiency of 61. Lindemann S, Tolley ND, Dixon DA, et al. Activated platelets
platelet procoagulant activity. Am J Med. 1979;67(2):206-213. mediate inflammatory signaling by regulated interleukin 1beta
41. Suzuki J, Umeda M, Sims PJ, Nagata S. Calcium-dependent synthesis. J Cell Biol. 2001;154(3):485-490.
phospholipid scrambling by TMEM16F. Nature. 2010; 62. Gawaz M, Brand K, Dickfeld T, et al. Platelets induce
468(7325):834-838. alterations of chemotactic and adhesive properties of endothe-
42. Jackson SP, Schoenwaelder SM. Procoagulant platelets: are lial cells mediated through an interleukin-1-dependent mecha-
they necrotic? Blood. 2010;116(12):2011-2018. nism. Implications for atherogenesis. Atherosclerosis. 2000;
43. Schoenwaelder SM, Yuan Y, Josefsson EC, et al. Two distinct 148(1):75-85.
pathways regulate platelet phosphatidylserine exposure and 63. Gawaz M, Neumann FJ, Dickfeld T, et al. Activated platelets
procoagulant function. Blood. 2009;114(3):663-666. induce monocyte chemotactic protein-1 secretion and surface
44. Kulkarni S, Woollard KJ, Thomas S, Oxley D, Jackson SP. expression of intercellular adhesion molecule-1 on endothelial
Conversion of platelets from a proaggregatory to a proinflamma- cells. Circulation. 1998;98(12):1164-1171.
tory adhesive phenotype: role of PAF in spatially regulating 64. Antoniades C, Bakogiannis C, Tousoulis D, Antonopoulos AS,
neutrophil adhesion and spreading. Blood. 2007;110(6):1879- Stefanadis C. The CD40/CD40 ligand system: linking inflam-
1886. mation with atherothrombosis. J Am Coll Cardiol. 2009;54(8):
45. Borissoff JI, Spronk HM, ten Cate H. The hemostatic system as 669-677.
a modulator of atherosclerosis. N Engl J Med. 2011;364(18): 65. Kroczek RA, Graf D, Brugnoni D, et al. Defective expression
1746-1760. of CD40 ligand on T cells causes X-linked immunodeficiency
46. Jin RC, Voetsch B, Loscalzo J. Endogenous mechanisms of with hyper-IgM (HIGM1). Immunol Rev. 1994;138:39-59.
inhibition of platelet function. Microcirculation. 2005;12(3):247- 66. Henn V, Slupsky JR, Grafe M, et al. CD40 ligand on activated
258. platelets triggers an inflammatory reaction of endothelial cells.
47. Gawaz M, Neumann FJ, Ott I, Schiessler A, Schomig A. Nature. 1998;391(6667):591-594.
Platelet function in acute myocardial infarction treated with 67. Heeschen C, Dimmeler S, Hamm CW, et al. Soluble CD40
direct angioplasty. Circulation. 1996;93(2):229-237. ligand in acute coronary syndromes. N Engl J Med. 2003;
48. Gawaz M, Langer H, May AE. Platelets in inflammation and 348(12):1104-1111.
atherogenesis. J Clin Invest. 2005;115(12):3378-3384. 68. Schonbeck U, Libby P. The CD40/CD154 receptor/ligand
49. Libby P, Ridker PM, Maseri A. Inflammation and atherosclero- dyad. Cell Mol Life Sci. 2001;58(1):4-43.
sis. Circulation. 2002;105(9):1135-1143. 69. Semple JW, Italiano JE, Freedman J. Platelets and the immune
50. Wagner DD, Frenette PS. The vessel wall and its interactions. continuum. Nat Rev Immunol. 2011;11(4):264-274.
Blood. 2008;111(11):5271-5281. 70. Schonbeck U, Libby P. CD40 signaling and plaque instability.
51. Frenette PS, Denis CV, Weiss L, et al. P-Selectin glycoprotein Circulation Research. 2001;89(12):1092-1103.
ligand 1 (PSGL-1) is expressed on platelets and can mediate 71. Ross R. Atherosclerosisan inflammatory disease. N Engl
platelet-endothelial interactions in vivo. J Exp Med. 2000;191(8): J Med. 1999;340(2):115-126.
1413-1422. 72. McEver RP, Cummings RD. Perspectives series: cell adhesion
52. Romo GM, Dong JF, Schade AJ, et al. The glycoprotein in vascular biology. Role of PSGL-1 binding to selectins in
Ib-IX-V complex is a platelet counterreceptor for P-selectin. leukocyte recruitment. J Clin Invest. 1997;100(3):485-491.
J Exp Med. 1999;190(6):803-814. 73. Simon DI, Chen Z, Xu H, et al. Platelet glycoprotein ibalpha is
53. Gawaz M, Neumann FJ, Dickfeld T, et al. Vitronectin receptor a counterreceptor for the leukocyte integrin Mac-1 (CD11b/
(alpha(v)beta3) mediates platelet adhesion to the luminal aspect CD18). J Exp Med. 2000;192(2):193-204.
of endothelial cells: implications for reperfusion in acute 74. Diacovo TG, deFougerolles AR, Bainton DF, Springer TA. A
myocardial infarction. Circulation. 1997;96(6):1809-1818. functional integrin ligand on the surface of platelets: intercellu-
54. Massberg S, Enders G, Matos FC, et al. Fibrinogen deposition lar adhesion molecule-2. J Clin Invest. 1994;94(3):1243-1251.
at the postischemic vessel wall promotes platelet adhesion 75. Santoso S, Sachs UJ, Kroll H, et al. The junctional adhesion
during ischemia-reperfusion in vivo. Blood. 1999;94(11):3829- molecule 3 (JAM-3) on human platelets is a counterreceptor for
3838. the leukocyte integrin Mac-1. J Exp Med. 2002;196(5):679-
55. Langer HF, Gawaz M. Platelet-vessel wall interactions in 691.
atherosclerotic disease. Thromb Haemost. 2008;99(3):480-486. 76. Weber C, Springer TA. Neutrophil accumulation on activated,

60 American Society of Hematology


surface-adherent platelets in flow is mediated by interaction of 87. Smyth SS, McEver RP, Weyrich AS, et al. Platelet functions
Mac-1 with fibrinogen bound to alphaIIbbeta3 and stimulated beyond hemostasis. J Thromb Haemost. 2009;7(11):1759-
by platelet-activating factor. J Clin Invest. 1997;100(8):2085- 1766.
2093. 88. Dixon DA, Tolley ND, Bemis-Standoli K, et al. Expression of
77. Koenen RR, Weber C. Platelet-derived chemokines in vascular COX-2 in platelet-monocyte interactions occurs via combinato-
remodeling and atherosclerosis. Semin Thromb Hemost. 2010; rial regulation involving adhesion and cytokine signaling.
36(2):163-169. J Clin Invest. 2006;116(10):2727-2738.
78. Gleissner CA, von Hundelshausen P, Ley K. Platelet chemo- 89. Nesbitt WS, Mangin P, Salem HH, Jackson SP. The impact of
kines in vascular disease. Arterioscler Thromb Vasc Biol. blood rheology on the molecular and cellular events underlying
2008;28(11):1920-1927. arterial thrombosis. J Mol Med. 2006;84(12):989-995.
79. Brandt E, Van Damme J, Flad HD. Neutrophils can generate 90. Lehoux S, Tedgui A. Cellular mechanics and gene expression
their activator neutrophil-activating peptide 2 by proteolytic in blood vessels. J Biomech. 2003;36(5):631-643.
cleavage of platelet-derived connective tissue-activating pep- 91. Davies PF. Hemodynamic shear stress and the endothelium in
tide III. Cytokine. 1991;3(4):311-321. cardiovascular pathophysiology. Nat Clin Pract Cardiovasc
80. von Hundelshausen P, Koenen RR, Weber C. Platelet-mediated Med. 2009;6(1):16-26.
enhancement of leukocyte adhesion. Microcirculation. 2009; 92. Davies PF. Flow-mediated endothelial mechanotransduction.
16(1):84-96. Physiol Rev. 1995;75(3):519-560.
81. Brandt E, Ludwig A, Petersen F, Flad HD. Platelet-derived 93. Lawrence MB, Kansas GS, Kunkel EJ, Ley K. Threshold levels
CXC chemokines: old players in new games. Immunol Rev. of fluid shear promote leukocyte adhesion through selectins
2000;177:204-216. (CD62L,P,E). J Cell Biol. 1997;136(3):717-727.
82. Koenen RR, von Hundelshausen P, Nesmelova IV, et al. 94. Woollard KJ, Kling D, Kulkarni S, Dart AM, Jackson S,
Disrupting functional interactions between platelet chemokines Chin-Dusting J. Raised plasma soluble P-selectin in peripheral
inhibits atherosclerosis in hyperlipidemic mice. Nature medi- arterial occlusive disease enhances leukocyte adhesion. Circ
cine. 2009;15(1):97-103. Res. 2006;98(1):149-156.
83. von Hundelshausen P, Weber KS, Huo Y, et al. RANTES 95. Skilbeck CA, Walker PG, David T, Nash GB. Disturbed flow
deposition by platelets triggers monocyte arrest on inflamed promotes deposition of leucocytes from flowing whole blood in
and atherosclerotic endothelium. Circulation. 2001;103(13): a model of a damaged vessel wall. Br J Haematol. 2004;126(3):
1772-1777. 418-427.
84. Baltus T, Weber KS, Johnson Z, Proudfoot AE, Weber C. 96. Goto S, Tamura N, Handa S. Effects of adenosine 5-
Oligomerization of RANTES is required for CCR1-mediated diphosphate (ADP) receptor blockade on platelet aggregation
arrest but not CCR5-mediated transmigration of leukocytes on under flow. Blood. 2002;99(12):4644-4645; author reply 4645-
inflamed endothelium. Blood. 2003;102(6):1985-1988. 4646.
85. Weyrich AS, Elstad MR, McEver RP, et al. Activated platelets 97. Barstad RM, Orvim U, Hamers MJ, Tjonnfjord GE, Brosstad
signal chemokine synthesis by human monocytes. J Clin Invest. FR, Sakariassen KS. Reduced effect of aspirin on thrombus
1996;97(6):1525-1534. formation at high shear and disturbed laminar blood flow.
86. Mause SF, von Hundelshausen P, Zernecke A, Koenen RR, Thromb Haemost. 1996;75(5):827-832.
Weber C. Platelet microparticles: a transcellular delivery sys- 98. Nesbitt WS, Westein E, Tovar-Lopez FJ, et al. A shear
tem for RANTES promoting monocyte recruitment on endothe- gradient-dependent platelet aggregation mechanism drives
lium. Arterioscler Thromb Vasc Biol. 2005;25(7):1512-1518. thrombus formation. Nat Med. 2009;15(6):665-673.

Hematology 2011 61

You might also like