platelets and the vessel wall

Upload: racquel-joseph

Post on 03-Jun-2018

227 views

Category:

Documents


0 download

TRANSCRIPT

  • 8/12/2019 Platelets and the Vessel Wall

    1/20

  • 8/12/2019 Platelets and the Vessel Wall

    2/20

    1380 HEMOSTASIS

    In a 1881 communication to the Turin Royal Academy

    of Medicine, the Italian physician Giulio Bizzozero dis-

    closed the presence in circulating human blood of dis-

    crete elements that he termed piastrine (blutplttchen

    in a 1882 publication in a German journal and petites

    plaques in a communication in French).1 Previously

    speculated to be merely nonphysiologic granular aggre-

    gates, blood platelets have since become central to ourunderstanding of thrombosis and hemostasis, and detailed

    understanding of their participation in cardiovascular

    disease, stroke, and even cancer has led to remarkable

    progress in the rational treatment of these disorders.

    Although platelets are most often studied in the

    context of their ability to form a hemostatically effective

    plug, it is now widely recognized that their influence

    extends far beyond this process to all aspects of hemo-

    stasis, as well as to wound healing and vascular remodel-

    ing. For example, platelets generate or secrete biologically

    active mediators such as thromboxane A2 (TXA2) and

    serotonin, which not only amplify platelet activation

    responses but also modulate vascular tone. In addition,platelets secrete a broad array of granule constituents

    that stimulate vessel repair, induce megakaryocytopoie-

    sis, promote coagulation, and limit fibrinolysis.

    The same pathways that lead to platelet plug forma-

    tion can also produce pathologic thrombosis, a process

    that has been described as hemostasis occurring at the

    wrong time or in the wrong place. Platelets are particu-

    larly important for hemostasis on the arterial side of the

    circulation, where blood flows under higher pressure and

    experiences greater shear force. As a result, platelet func-

    tion is generally considered to be critical to the patho-

    genesis of arterial thrombosis and less so for venous

    thrombosis, and antiplatelet drugs are most widely usedin the former setting. However, this distinction between

    the mechanisms underlying arterial and venous throm-

    bosis is not absolute, and the spectrum of thrombotic

    disorders should be considered a continuum.

    Arterial thrombosis is a particularly common problem

    in middle-aged and older adults and is a major cause of

    morbidity and mortality in developed countries. The

    thrombi that arise in atherosclerotic vessels are predomi-

    nantly platelet in origin and are the proximate cause of

    myocardial infarction and most cerebrovascular acci-

    dents. Although arterial thrombosis is considerably less

    common in children than adults, it may contribute to

    major morbidity in patients with sickle cell disease, aswell as complications of some childhood infections,

    Kawasakis syndrome, and various forms of arteritis,

    autoimmune disorders, hemolytic-uremic syndrome,

    and thrombotic thrombocytopenic purpura (see Chapter

    33).

    In this chapter we review platelet structure and func-

    tion, with special emphasis on the cell surface glycopro-

    teins that function as sentries for areas of vascular damage

    and the signal transduction events that both amplify and

    limit platelet responsiveness. The information provided

    here should be helpful in understanding subsequent

    chapters that describe inherited and acquired platelet

    disorders (see Chapters 29 and 33) and the role of the

    adhesive protein von Willebrand factor (VWF) (see

    Chapter 30) in hemostasis. Finally, there is growing

    appreciation of the role that platelets play in inflamma-

    tion and the pathogenesis of atherothrombosis, which is

    briefly discussed at the end of the chapter.

    PLATELET MORPHOLOGY ANDSUBCELLULAR ORGANIZATION

    Platelets are adhesion and signaling machines that circu-

    late as small, disc-shaped cellular fragments in the whole

    blood of healthy individuals at a concentration of approx-

    imately 150,000 to 300,000/L. Early studies suggestedthat platelets might be produced via cytoplasmic frag-

    mentation along a network of internal demarcation

    membranes that were observed in large, polyploid mega-

    karyocytes.2,3 More recent studies,4-6 however, support

    the notion that proplatelets are assembled and packagedwith their various constituents at the ends of long cyto-

    plasmic extensions of differentiated megakaryocytes that

    have migrated from the proliferative osteoblastic niche to

    the capillary-rich vascular niche of the bone marrow

    microenvironment,7 with the invaginated demarcation

    membrane system serving simply as a reservoir of inter-

    nal membrane used for proplatelet extension.8,9 Once

    adjacent to the adluminal face of the endothelium,

    proplatelets are released into the bloodstream, where

    they circulate as mature platelets for approximately 7 to

    10 days before being cleared by the liver and spleen 10

    their life span being controlled, at least in part, by

    an antagonistic balance between the apoptotic proteinsBcl-xL and Bak.11

    The size of resting platelets is somewhat variable,

    averaging approximately 1.5 m in diameter and 0.5 to1 m in thickness. Platelet size is undoubtedly regulatedby numerous factors during their biogenesis, but both the

    224-kd nonmuscle myosin heavy chain IIA (MYHIIA)

    and the cell surface glycoprotein Ib (GPIb) complex

    appear to play critical roles. Thus, mutations in the

    MYH9 gene predominantly interfere with contractile

    events important for platelet formation,12whereas failure

    to express GPIbthe molecular basis for the platelet

    disorder known as Bernard-Soulier syndrome13,14dis-

    rupts critical associations with the cytoskeletal proteinfilamin15,16 that play an essential role in both platelet

    formation and platelet compliance.17In both these inher-

    ited platelet disorders, platelets can appear as large as

    lymphocytes (see Chapter 29). Correction of GPIb

    expression in GPIb-deficient (Bernard-Soulier) mice has

    been shown to restore platelets to their normal size.18

    The volume of a platelet (mean platelet volume)

    normally ranges from 6 to 10 fL (1 fL =1015L). Plateletdensity is also variable,19and the issue of whether young

    platelets are more20,21or less22dense as they gain versus

    lose content during their circulating lifetime has never

  • 8/12/2019 Platelets and the Vessel Wall

    3/20

    Chapter 25 Platelets and the Vessel Wall 1381

    been satisfactorily resolved. Because platelets retain most

    species of messenger RNA (mRNA) for a short period

    after their release from bone marrow megakaryocytes,23

    young platelets can be distinguished from older ones by

    their RNA content.24

    As shown in Figure 25-1A and C, resting platelets are

    discoid in shape, largely because of the presence of a cir-

    cumferential coil of microtubules,25,26

    and they are packedwith numerous electron-opaque alpha granules, a few

    dense granules (granule contents and their functions are

    discussed later), several mitochondria, and lysosomes.27

    Platelets also retain a few Golgi remnants, as well as occa-

    sional vestiges of rough endoplasmic reticulumthe

    exception being platelets from patients with rapid platelet

    turnover, in whom very young platelets containing more

    abundant protein synthesis machinery are readily observed

    in the circulation. Platelets also contain two highly spe-

    cialized membrane systems not found in other cells of the

    body: the surface-connected open canalicular system

    (OCS) (see Fig. 25-1B and C) and the dense tubular

    system (DTS). The OCS is a series of tortuous invagina-tions of the plasma membrane that appear to tunnel

    throughout the cytoplasm of the cell28 and serve as an

    internal reservoir of plasma membrane that is called upon

    when platelets round up, extend lamellipods and filopods

    (see Fig. 25-1B and D), and spread during platelet activa-

    tiona process that can increase the surface area of

    exposed plasma membrane by more than 400%.29Because

    OCS channels are proximal to internal granules, they also

    probably function as a conduit for the rapid expulsion of

    alpha and dense granule contents during platelet activa-

    tion.30The DTS, on the other hand, is a remnant of the

    smooth endoplasmic reticulum31and is found randomly

    dispersed throughout the cytoplasm. The DTS appears to

    be one of several organelles within the platelet known to

    harbor high concentrations of calcium,32,33

    and it isthought to contain a 100-kd calcium adenosine triphos-

    phatase (ATPase) known as SERCA2b34that functions to

    sequester and store cytosolic calcium in resting cells.

    Recent evidence suggests that adenosine diphosphate

    (ADP) is able to induce selective release of calcium from

    the DTS35whereas activation of the GPIb/V/IX receptor

    for VWF releases calcium primarily from a poorly defined

    acidic compartment36 within the cell.35 Thrombin, a

    strong platelet agonist, appears to elicit release of calcium

    from both stores on binding to the platelet thrombin

    receptor PAR1.35

    The platelet cytoskeleton is composed of a single

    rigid, but dynamic microtubule approximately 100 m inlength that is coiled about 8 to 12 times around theequatorial plane of the cell.37-39This marginal band of

    microtubules is largely responsible for maintaining the

    discoid shape of the resting cell, as illustrated by the

    observations that (1) incubation of platelets with colchi-

    cinean agent that dissolves microtubulesresults

    in their rounding,40 (2) platelets from mice lacking

    A

    B

    C D

    FIGURE 25-1.Platelet morphology. Resting platelets (shown in thin section in Aand from a scanning electron micrograph of a flash-frozen,freeze-dried platelet in C) are shaped like a disc and contain numerous electron-opaque alpha granules, a few dense granules, and several mito-

    chondria and lysosomes. A circumferential coil of microtubules (mchighlighted with an oval) is responsible for maintaining their discoid shape

    Platelets also contain a number of cytoplasmic membrane systems that subserve specialized functions, including vestiges of the smooth endoplasmic

    reticulum that sequester calcium and tortuous invaginations of the plasma membrane that form a surface-connected open canalicular system

    (OCS). When platelets become activated (Band D), they rapidly round up, extend lamellipodia (lam) and filopodia (fil), and release the contents

    of their granules, often into the nearby OCS. (Photographs generously provided by John H. Hartwig and used with permission.)

  • 8/12/2019 Platelets and the Vessel Wall

    4/20

    1382 HEMOSTASIS

    1-tubulin remain largely spherical,41 and (3) plateletspherocytosis in humans results when tubulin fails to

    polymerize normally into microtubules.42Directly under-

    neath the plasma membrane lies an intricate, two-dimen-

    sional, tightly woven membrane skeleton43composed of

    nonerythroid spectrin,44,45a network of actin filaments,43,45

    vinculin,46and the actin-binding protein filamin,47which

    itself is tethered to the inner face of the plasma mem-brane via linkages with the cytoplasmic domain of

    GPIb.15,47The membrane skeleton, because of its loca-

    tion, serves as a scaffold that links elements of the plasma

    membrane with contractile elements of the cytoskeleton

    and cytosolic signaling proteins and thereby regulates

    such diverse functions as receptor mobility,48,49receptor

    clustering,50-52 and signal transduction.53 Finally, the

    platelet is filled with an extensive cytoplasmic network of

    actin filaments45,54 organized by the actin-binding pro-

    teins filamin55,56 and -actinin57 that constitute itscytoskeleton.

    When platelets become exposed to components of

    the extracellular matrix58

    or to soluble agonists such asADP59or thrombin,60,61they undergo dramatic changes

    in their morphology.62,63The marginal band of microtu-

    bules disappears,54which allows the platelet to transform

    from a disc to an irregular sphere. At nearly the same

    time, the actin filamentcapping protein -adducinbecomes phosphorylated and dissociates from existing

    F-actin filaments,64thereby exposing the barbed end of

    the filament to cytosolic actin monomers and driving

    rapid polymerization of actin into microfilaments.62This

    has the dual effect of driving the extension of lamellipo-

    dia and filopodia and forcing granules toward the center

    of the platelet, where they can fuse with membranes of

    the OCS and release their contents to the exterior of thecell. Phosphorylation of myosin additionally induces

    contractile events that facilitate centralization of the

    granules.65

    PLATELET GENOMICS AND PROTEOMICS

    Though anucleate, platelets contain measurable and

    manipulable levels of megakaryocyte-derived mRNA,23

    at least some of which is capable of being synthesized

    into small, but detectable amounts of protein.66,67Both

    serial analysis of gene expression (SAGE) and gene

    microarray analysis have been used to estimate the sizeand composition of the platelet transcriptome.68-70A con-

    sistent finding of all genomic analyses performed to date

    is that mitochondrially derived transcripts dominate the

    platelet transcriptomepresumably because of persis-

    tent transcription of the mitochondrial genome after

    platelet release from the bone marrow. This problem has

    recently been addressed by analyzing the transcriptome

    of cultured megakaryocytes derived from cord blood

    stem cells.71Of the 20,488 genes present in the human

    genome, 2000 to 3000 distinct transcripts have been

    identified in unstimulated plateletsconsiderably fewer

    than normally found in a nucleated cell, but perhaps

    more than one might have expected from an anucleate

    circulating cellular fragment. One of the more surprising

    findings in recent years has been the identification of

    heterogeneous nuclear RNA (hnRNA) in the platelet

    cytosol, as well as all of the spliceosome components

    necessary to splice the hnRNA into mature message that

    can thereafter be translated into protein.72

    Enlisted duringthe activation process, signal-dependent protein transla-

    tion has thus far been demonstrated for mRNA mole-

    cules encoding interleukin-1 (IL-1),72 tissue factor,73and Bcl-3,74 the protein products of which have the

    potential to influence inflammation, thrombosis, and

    wound repair.

    The platelet proteome appears to be equally complex

    and diverse and, unlike the transcriptome, reports both

    the breadth and relative amounts of protein products

    actually present in the cell. Obtained by refined two-

    dimensional gel electrophoretic techniques that were

    originally developed in the 1970s75,76or by liquid chro-

    matographic separation, proteins are fragmented andseparated via a combination of proteolytic and ionization

    techniques and then analyzed by mass spectrometry.

    Such analysis has allowed the identification of dozens of

    proteins present in complex cellular lysates or subcellular

    fractions (see elsewhere77,78 for recent reviews of this

    topic). In addition to yielding the expected menu of

    major plasma membrane glycoprotein receptors, one of

    the more complete global profiling analyses to date79

    identified a core platelet proteome composed of 641 pro-

    teins, including an abundance of molecules involved in

    signal transduction, cytoskeletal change, and metabo-

    lismunderstandable given the importance of cellular

    activation and its control in platelet function. By combin-ing prefractionation methods with suitable separation

    techniques, proteomic analysis has also been used to

    compile an inventory of proteins that are either (1) post-

    translationally modified (normally by phosphorylation)

    during the platelet activation process80-82or (2) present

    at low abundance in the total platelet proteome but

    enriched within various subcellular compartments,

    including the platelet cytoskeleton,83alpha granules,81,84,85

    membrane fraction,86 membrane rafts,87 and

    microparticles.88

    ANTITHROMBOTIC COMPONENTSOF THE VESSEL WALL

    Although hundreds of thousands of platelets per micro-

    liter circulate in blood, under normal conditions very few,

    if any, interact with the intact vessel wall because the

    endothelial lining of the blood vessel presents an excel-

    lent nonthrombogenic surface. In fact, this property of

    the vessel wall has not yet been duplicated in any pros-

    thetic or extracorporeal device. Healthy endothelium not

    only provides an effective barrier between blood compo-

    nents and the highly thrombogenic components of the

  • 8/12/2019 Platelets and the Vessel Wall

    5/20

    Chapter 25 Platelets and the Vessel Wall 1383

    subendothelium (see later) but also actively produces

    both membrane-bound and secretory products that limit

    fibrin generation and promote clot dissolution. For

    example, heparin-like glycosaminoglycans present on the

    luminal side of the endothelial cell surface recruit plasma

    antithrombin, which effects a conformational change that

    promotes binding and neutralization of thrombin and

    other serine proteases.89

    Thrombin, when bound to theendothelial cell surface receptor thrombomodulin, takes

    on anticoagulant properties via its cleavage and activation

    of protein C, which in turn cleaves coagulation factors V

    and VIII, thereby further suppressing thrombin genera-

    tion.90Endothelial cells also express a specific receptor

    for activated protein C that serves to concentrate the

    protein on the endothelial surface (see Chapter 26).91

    Finally, endothelial cells synthesize, secrete, and rebind

    tissue plasminogen activator,92,93 which activates plas-

    minogen to facilitate fibrin dissolution (see Chapter 27).

    These activities are summarized in schematic form in

    Figure 25-2.

    The endothelial cell also produces two importantinhibitors of platelet activation: prostacyclin (PGI2)

    94-96

    and nitric oxide (NO).97-99A labile oxygenated metabolite

    of arachidonic acid generated by endothelial cell cyclo-

    oxygenase-2 (COX-2), PGI2diffuses out of the cell and

    binds to a platelet Gsproteincoupled receptor (GPCR)

    known as the isoprostenoid (IP) receptor.100,101 Such

    binding stimulates adenylate cyclase to increase cytosolic

    cyclic adenosine monophosphate (cAMP) levels, which

    (1) activates a pump in the DTS that decreases cytosolic

    Ca2+, thereby helping keep platelets quiescent, and (2)

    activates protein kinase A (PKA), the actions of which

    will be discussed later. PGI2also has potent vasodilatory

    effects by binding to IP on arterial smooth muscles cellsto effect vessel relaxation.94 The PGI2 produced by

    vascular endothelium thus serves to counterbalance

    the proaggregatory and vasoconstrictor activities of the

    platelet-derived prostanoid TXA2, the biology of which is

    discussed later. In fact, upsetting the delicate balance

    between COX-1derived TXA2 and COX-2derived

    PGI2 has been shown to increase the risk for adverse

    cardiovascular events.102

    Whereas PGI2stimulates adenylate cyclase to produce

    cAMP, NO, a product of -arginine generated by endo-

    thelial nitric oxide synthase (eNOS),103directly activates

    platelet guanylate cyclase, which results in increased cyto-

    solic levels of cyclic guanosine monophosphate (cGMP).Although platelet responses to low levels of this cyclic

    nucleotide can at first be mildly stimulatory,104 cGMP,

    largely via its activation of protein kinase G (PKG), has

    the overall effect of dampening platelet responses, inhibit-

    ing platelet adhesion105,106 and aggregation,107-110 and

    cAMP

    CD39

    EPCR

    TM

    COX-2eNOS

    NO PGI2

    GAGs

    APC

    ATIII Plasminogen

    AMP

    Plasmin

    FVon, FVIIIon

    ADP Fvoff, FVIIIoff

    thrombin

    PC

    TPA

    Adenylatecyclase

    Guanylatecyclase

    Gs

    Gs

    Dense tubularsystem

    PKG

    Platelet

    Endothelium

    Multiple inhibitory signaling pathways

    PKA

    SERCA2b Ca2+Ca2+

    Ca2+

    Ca2+ Ca2+Ca2+Ca

    2+Ca2+

    Ca2+Ca2+

    cGMP

    FIGURE 25-2. Anticoagulant and antithromboticcomponents of the vascular endothelium. Endothelial

    cells produce a number of substances, including nitric

    oxide (NO) and prostacyclin (PGI2), that act on plate-

    let surface receptors to dampen platelet responsiveness.

    They also scavenge the platelet agonist adenosine

    diphosphate (ADP), inactivate thrombin, and activate

    the fibrinolytic enzyme plasmin. APC, activated protein

    C; AMP, adenosine monophosphate; ATIII, antithrom-

    bin III; cAMP, cyclic adenosine monophosphate;

    cGMP, cyclic guanosine monophosphate; COX-2,

    cyclooxygenase-2; eNOS, endothelial nitric oxide syn-

    thase; EPCR, endothelial cell protein C receptor; FV,factor V; GAGs, glycosaminoglycans; TM, thrombo-

    modulin; IP, isoprostenoid; PC, protein C; PKA,

    protein kinase A; PKG, protein kinase G; TPA, tissue

    plasminogen activator.

  • 8/12/2019 Platelets and the Vessel Wall

    6/20

    1384 HEMOSTASIS

    impeding platelet-mediated recruitment of leukocytes

    during the inflammatory response.111 Its mechanism of

    action is discussed in more detail later.

    In addition to the soluble metabolites PGI2and NO,

    endothelial cells also express on their surface a potent

    adenosine diphosphatase (ADPase) known as CD39 that

    scavenges plasma ADP to prevent platelet aggrega-

    tion.112,113

    Finally, it is important to note that inflamma-tory cytokines, oxidized lipids, and immune complexes

    can, under pathologic conditions, inhibit these protective

    biochemical pathways and impair the antithrombotic

    state of the endothelial cell. The latter changes permit

    unrestrained formation of platelet- and fibrin-containing

    thrombi, as well as thrombus formation beyond sites of

    vascular injury, and can thus contribute to atherothrom-

    bosisa topic that is discussed more extensively at the

    end of this chapter.

    REACTING TO THE BREACHCELL SURFACE

    RECEPTORS THAT MEDIATE TETHERINGAND ADHESION AND TRANSMIT EARLYACTIVATION SIGNALS

    As antithrombotic as the endothelial lining is, the under-

    lying extracellular matrix consists of a rich mixture of

    glycosaminoglycans into which are embedded an abun-

    dance of highly concentrated prothrombotic proteins,

    including structural components such as collagen and

    elastin (which constitute 30% of body weight) andadhesive proteins such as laminin, fibronectin, and VWF.

    Not surprisingly, platelets have evolved receptors for

    most of these proteins and initiate a series of rapid bio-

    chemical events both on the surface and inside the cellwhen exposed to them. As a result, adhesion is an activat-

    ing event!

    The large number of circulating red blood cells serve

    to marginate platelets, and when the vessel wall is

    breached, either by mechanical injury or after rupture

    of atherosclerotic plaque, the first layer of platelets to

    encounter exposed matrix undergoes a series of sequen-

    tial events similar to what leukocytes experience during

    the inflammatory responsenamely, tethering, initial

    signaling to the cell interior, integrin-mediated adhesion,

    and cytoskeletally directed cell spreading. Whereas leu-

    kocyte tethering is mediated by members of the selectin

    family, the first layer of platelets become tethered onVWF,114which is sprinkled on exposed collagen fibers.

    VWF interacts with a high-affinity, platelet-specific mul-

    tisubunit receptor known as the GPIb/V/IX complex.115

    This latter complex, which is expressed at approximately

    25,000 copies per cell,116 binds to the A1 domain of

    VWF117with high-enough affinity to tether platelets even

    under conditions of arterial shear.118Loss of the GPIB/

    V/IX receptor in both humans and mice results in a clini-

    cal condition known as Bernard-Soulier syndrome,13,14

    which is characterized not only by an increase in platelet

    size but also by prolonged bleeding caused, in large part,

    by the inability of platelets to adhere to the vessel wall.

    After engagement with its ligand, GPIb acts through

    membrane-proximal Src family kinases,119 through

    adapter molecules,120and to a lesser extent, via its asso-

    ciation with immunoreceptor tyrosinebased activation

    motif (ITAM)-bearing subunits121,122 to transmit early

    activation signals123,124that together result in the recruit-

    ment and activation by tyrosine phosphorylation of phos-pholipase C2 (PLC2),125,126a key enzymatic componentof platelet amplification that is required to achieve throm-

    bus growth and stability (Fig. 25-3).127

    Once tethered, two different platelet integrinseach

    of which exists in a low-affinity state on the platelet

    surfacebegin to engage specific extracellular matrix

    components and, together with the small calcium tran-

    sients and kinase-generated signals emanating from the

    GPIb complex and from the mechanical shear force gen-

    erated by the flowing blood,128initiate the reciprocal pro-

    cesses of platelet adhesion and activation. Thus, the 21integrin binds to exposed collagen fibrils,129whereas the

    integrin receptor 61 engages laminin.130

    Both theseintegrins hand off to a member of the immunoglobulin

    superfamily, GPVI,131,132which via its noncovalent asso-

    ciation in the plane of the plasma membrane with the

    ITAM-bearing Fc receptor chain dimer131,133 elicitsstrong PLC2-dependent events that (1) begin theprocess of cytoskeletally directed shape change and

    cell spreading (discussed earlier); (2) initiate signal trans-

    duction pathways (illustrated in Fig. 25-3) that cause

    dramatic structural changes in platelet integrins and

    thereby result in their adopting a high-affinity, ligand-

    bindingcompetent conformation134a process known

    as inside-out signal transduction (to be described in

    more detail later); and (3) facilitate fusion of alpha anddense granules with the OCS and underlying plasma

    membrane.

    PLATELET GRANULES AND THEIRROLE IN HEMOSTASIS

    Platelet-specific granules are synthesized, assembled, and

    packaged during megakaryocyte biogenesis, and at later

    stages of maturation they appear to come into contact

    with microtubules, which then transport them, via the

    microtubule motor protein kinesin, along the shafts of

    proplatelets until they reach the proplatelet tips.135

    Onceinside a mature platelet, platelet granules remain rela-

    tively evenly dispersed throughout the cytoplasm, their

    contents awaiting threshold signals for cellular activation,

    at which time their membranes fuse with the plasma

    membrane or, more likely, the invaginated subdomains

    of the plasma membrane known as the OCS136 (see

    earlier). The regulated secretion of granule contents

    ensures that hemostasis remains highly localizedan

    event that has recently been exploited to deliver non

    platelet-derived procoagulant proteins such as factor VIII

    to sites of vascular injury.137,138

  • 8/12/2019 Platelets and the Vessel Wall

    7/20

    Chapter 25 Platelets and the Vessel Wall 1385

    Collagen

    Laminin

    VWF

    GPVI

    IT AM

    Fyn/Lyn

    Syk

    PLC2

    IT AM

    SH2

    SH2

    SH2

    SH2

    Granule secretion

    Rap1 Integrin activation

    Cytoskeletal rearrangementsShape change

    Integrin clustering

    GPIb-V-IX

    FcR

    CRP

    PI3KSrc

    Ca2+

    release from DTSIP3+

    DAG

    PKC

    CalDAG-GEF

    Syk

    Akt2GSK

    3

    RIAM

    Talin

    21

    LAT,S

    LP-76

    61

    Kinase

    Kinase

    PI3K,

    Ca2+

    S S

    Gads,B

    tk

    Gads,B

    TKLAT,SL

    P-76

    FIGURE 25-3. Platelet adhesion receptors that signal through phospholipase C2 (PLC2). Each of the cell surface receptors shown recognizesdifferent component of the extracellular matrix and thus works in coordinated fashion to send early activation signals into the cell. Binding of

    platelets to von Willebrand factor (VWF) slows platelets down so that integrins can associate productively with matrix collagen and laminin. Signals

    emanating from each of these events are transmitted into the cell interior, in part via the action of receptor-associated Src family kinases (Src, Fyn,

    Lyn), which phosphorylate tyrosine residues within nearby immunoreceptor tyrosinebased activation motifs (ITAMs), thus forming a nucleation

    point for the assembly of miniature organelles sometime referred to as signalosomes. Signalosomes are themselves composed of the adaptor proteins

    LAT, SLP-76, and Gads and the receptor tyrosine kinase Btk and function to localize, phosphorylate, and then activate PLC2, which coordinatesall these responses by generating the classic signaling molecules 1,4,5-inositol triphosphate (IP 3) and diacylglycerol (DAG). Details regarding the

    molecular events that take place after the generation of IP3and DAG are shown in Figures 25-4 and 25-6. Btk, Brutons tyrosine kinase; CRP, C-

    reactive protein; DTS, dense tubular system; GP1b, glycoprotein Ib; GSK3, glycogen synthase kinase 3; PI3K, posphatidylinositol-3-kinasePKC, protein kinase C; RIAM, Rap1GTP-interacting adaptor molecule; SH2, Src homology domain 2.

    Platelets harbor three distinct types of granules (Box

    25-1). Twoalpha and dense granulesare found only in

    platelets, whereas lysosomes are present in nearly all cell

    types. Alpha granules are by far the most numerous, with

    as many as 40 to 80 per cell, and they contain a wide array

    of proteins and bioactive peptides. For ease of discussion,

    Box 25-1 classifies alpha granule proteins as those that

    reside within the alpha granule membrane (P-selectin

    being the most diagnostic), those pinocytosed from

    plasma and packaged (IgG, fibrinogen, albumin),139and

    those synthesized by megakaryocytes and stored (VWF,

    platelet factor 4, thrombospondin). mRNA molecules

    encoding the latter group have all been identified in the

    platelet cytoplasm. Upon platelet activation, granules

    become redistributed toward the center of the cell,136at

    which time SNARE (solubleN-ethylmaleimidesensitive

    attachment protein receptor) proteins within the alpha

    granule membrane facilitate fusion,140with members of

    the Rab family of low-molecular-weight guanosine tri-

    phosphatases (GTPases) playing a prominent role in

    vesicle docking and exocytosis.141,142 After membrane

    fusion, P-selectin143-145 and alpha granule membrane

  • 8/12/2019 Platelets and the Vessel Wall

    8/20

    1386 HEMOSTASIS

    that contain the vasoconstrictive substance seroto-

    nin,160-162adenine and guanine nucleotides such as ADP

    and ATP, inorganic pyrophosphates163 and polyphos-

    phates,164and the divalent cation calcium.165Complexes

    of the latter two are probably responsible for the dark

    appearance of these bodies on thin-section electron

    microscopy.166Dense granule membranes contain a few

    components in common with lysosomal membranes, suchas granulophysin (CD63, lysosome-associated membrane

    protein-3 [LAMP-3])167and LAMP-2,168as well as mem-

    brane proteins also present in alpha granule membranes,

    such as P-selectin,167 thus suggesting a common origin

    during biogenesis. Like their alpha granule counterparts,

    these dense granule membrane proteins become expressed

    on the platelet cell surface after granule fusion and secre-

    tion and can be used as platelet activation markers. Curi-

    ously, a number of plasma membrane glycoproteins,

    including GPIb and GPIIb-IIIa, have also been reported

    in dense granule membranes.169Dense granule contents,

    especially ADP,170play a physiologically important role in

    hemostasis, as evidenced by characteristic platelet func-tion defects in patients whose platelets lack dense gran-

    ules or their contents,171 collectively known as storage

    pool disorders.172-174 Chdiak-Higashi and Hermansky-

    Pudlak175syndromes are two such examples of autosomal

    recessive dense granule defects that lead to platelet dys-

    function and bleeding, the former being associated with

    immunodeficiency and the latter with albinism (see

    Chapter 29).

    Primary lysosomes are the third organelle whose

    contents are secreted upon platelet activation, but only

    three or fewer per cell are normally identifiable.176

    Although a clear role for lysosomes in platelet function

    has not been identified, they do contain more than adozen different acid hydrolases, cathepsins D and E, and

    other degradative enzymes that can be secreted if plate-

    lets are subjected to strong agonist stimulation. Their

    contents have been shown to be mildly reduced in the

    platelets of individuals with GPS,177in keeping with the

    notion that the latter disorder is caused by a defect in

    packaging. The membrane proteins on platelet lysosomes

    are typical of lysosomes in other cells and include LAMP-

    1,178LAMP-2,168and LAMP-3.167

    FEED-FORWARD AMPLIFICATION

    PATHWAYS INVOLVED IN PLATELETRECRUITMENT AND THROMBUS STABILITY

    Although platelet adhesion, early activation signals, and

    granule release are prerequisites for thrombus formation,

    efficientrecruitment of additional platelets to the site of

    the vascular lesion to yield a stable platelet plug requires

    a host of additional receptor/ligand interactionseach of

    which results in signal transmission and subsequent bio-

    chemical and cell biologic changes that help sustain

    platelet activation. Among the most important of these is

    the binding of released ADP to one of its two platelet G

    Box 25-1 Platelet Granules and Their Contents

    ALPHA GRANULES

    Membrane proteins enriched in the granule membrane: P-

    selectin, TLT-1, CD40 ligand (which is cleaved after

    exposure on the platelet surface to release soluble

    CD40L), and tissue factor.

    Membrane proteins present at similar concentrations as they arein the plasma membrane: GPIIb-IIIa, GPIb, PECAM-1,

    and perhaps many others

    Granule contents:

    Synthesized by megakaryocytes: Thrombospondin, VWF,

    platelet factor 4, -thromboglobulin, PDGF Endocytosed from plasma or origin not determined:

    Albumin, fibrinogen, fibronectin, IgG, Gas6,

    coagulation factor V, and many chemokines and growth

    factors, including RANTES, bFGF, EGF, TGF-, andVEGF

    DENSE GRANULES

    ADP, ATP, 5-HT, Ca2+, polyphosphate

    LYSOSOMES

    Acid hydrolases, elastase, cathepsins, and other

    degradative enzymes

    ADP, adenosine diphosphate; ATP, adenosine triphosphate;

    bFGF, basic fibroblast growth factor; EGF, epidermal growth

    factor; Gas6, growth arrestspecific gene 6; GP, glycoprotein;

    5-HT, 5-hydroxytryptamine; PDGF, platelet-derived growthfactor; PECAM-1, platelet endothelial cell adhesion molecule-

    1; RANTES, regulated on activation, T cell expressed and

    secreted; TGF-, transforming growth factor ; TLT-1, TREM(triggering receptor expressed on myeloid cells)-like transcript-

    1; VEGF, vascular endothelial growth factor; VWF, von

    Willebrand factor.

    specific proteins such as TLT-1146become expressed on

    the platelet surface, and the contents of the granule are

    released into the plasma milieu. Exposed P-selectin, diag-

    nostic of an activated platelet,147,148serves to recruit leu-

    kocytes to the site of injury,149one of a number of impor-

    tant links between thrombosis and inflammation150

    (discussed at the end of this chapter). Proteins secreted

    from platelets include the adhesive ligands VWF and

    fibrinogen, which serve to support platelet-platelet inter-

    actions; growth factors and cytokines, which promote cell

    migration151and wound healing152and maintain vascular

    integrity153

    ; and autocrine factors such as growth arrestspecific gene 6 (Gas6)154and CD40L,155which are released

    and rebind platelet receptors to help amplify platelet

    responsiveness. Finally, alpha granules and their contents

    are a source of procoagulant proteins, with release of

    factor V156and exposure of tissue factor73,157promoting

    localized fibrin deposition at sites of vascular injury. Plate-

    let alpha granules, or at least their contents,158are severely

    reduced in an inherited bleeding disorder known as gray

    platelet syndrome (GPS) (see Chapter 29).159

    Dense granules (four to eight per platelet) are mor-

    phologically distinct, electron-opaque storage organelles

  • 8/12/2019 Platelets and the Vessel Wall

    9/20

    Chapter 25 Platelets and the Vessel Wall 1387

    as a result of the action of phospholipase-generated 1,4,5-

    inositol triphosphate (IP3, discussed in more detail

    later)to remain cytosolic. Bioavailable calcium ions,

    in turn, support a host of additional cellular events,

    including more robust granule secretion, activation of

    metal iondependent proteases, and activation of cell

    surface integrins. The importance of ADP in amplifying

    platelet responses is illustrated by the clinical effective-ness of ticlopidine and clopidogrelwidely used phar-

    macologic agents that antagonize the activity of P2Y12

    in pacifying platelet reactivity and inhibiting platelet

    aggregation.181

    In addition to ADP-induced, P2Y12-mediated signal-

    ing, nearly a dozen other soluble ligands are either gener-

    ated or released at sites of vascular injury and function

    in signal amplification and platelet activation. These

    ligands can, for the sake of simplicity, be broken into

    three classes according to the type of platelet receptor to

    which they bind (Fig. 25-5). The first class of ligands is

    composed of ADP, thrombin, TXA2, and serotonin (5-

    hydroxytryptamine [5-HT]), each of which binds to aspecific GPCR that is coupled to the heterotrimeric subunit, Gq. Thus, ADP binds to P2Y1,

    182-185thrombin to

    the protease-activated receptors PAR1 and PAR4,186,187

    TXA2to the thromboxane receptor,188,189and serotonin

    to 5-HT2A.190When released as a consequence of ligand

    binding to any of these GPCRs, the Gqsubunit binds to

    the isoform of phospholipase C (PLC). PLCs are lipidhydrolases that act on membrane-associated phosphati-

    dylinositol 4,5-diphosphate (PIP2) to produce the second

    messengers IP3and diacylglycerol (DAG). IP3binds and

    opens calcium channels, whereas DAG activates the most

    abundant forms of protein kinase C (PKC), thereby

    epi

    2A P2Y12Adenylatecyclase

    cAMP Many other cellbiologic effects

    Dense tubularsystem

    SERCA2b

    Granulesecretion

    Ca2+ Ca2+

    Ca2+

    Ca2+

    Ca2+

    Ca2+Ca2+

    Ca2+Ca2+

    Gi2

    ADP

    Gz Gi2Gz

    FIGURE 25-4. Gi proteincoupled receptors on platelets inhibitadenylate cyclase and lower cyclic adenosine monophosphate (cAMP)

    levels. Shown are the two major receptors responsible for dampening

    the activity of adenylate cyclase. As cAMP levels drop, the ability of the

    SERCA2b calcium pump to sequester cytosolic calcium ions is

    impaired, thereby allowing calcium-mediated activation events to occur

    more readily.

    Ca2+

    releasefrom DTS

    IP3+

    DAGPKC

    Granule secretion

    Cytoskeletal rearrangementsShape change

    Integrin clustering

    Rap1 RIAM

    Talin

    CalDAG-GEF

    Rap

    1

    Integrinactivation

    PLCPLC

    Gq

    Gq

    Integrins

    Src

    Kinase

    SH2

    Syk

    ITAMSH2

    ADPThrombin

    TXA25'HT

    Fg, VWF, Collagen,sCD40L, LM, FN

    Gas6Ephrins

    GPCR RTK

    LAT,

    SLP-7

    6

    Gads,

    Btk

    FIGURE 25-5.Agonist receptors that initiate or amplifyplatelet activation responses (or both). Three different

    families of receptors are involved in signal amplification

    pathways: Gq-coupled GPCRs, integrins, and receptor

    tyrosine kinases (RTKs). Note how each activates either

    the or isoform of phospholipase C (PLC). The sumtotal of PLC-generated products serves to determine the

    activation state of the platelet, its ability to respond to

    vascular injury, and its participation in thrombus growth.

    ADP, adenosine triphosphate; Btk, Brutons tyrosine

    kinase; DAG, diacylglycerol; DTS, dense tubular system;

    GPCR, Gs proteincoupled receptor; 5-HT, 5-hydroxy-tryptamine; IP3, 1,4,5-inositol triphosphate; ITAM, immu-

    noreceptor tyrosinebased activation motif; PKC, protein

    kinase C; RIAM, Rap1GTP-interacting adaptor molecule;

    SH2, Src homology domain 2; TXA2, thromboxane A2;

    VWF, von Willebrand factor.

    proteincoupled receptors, P2Y12.179,180Like the 2recep-

    tor for epinephrine, P2Y12is coupled to an inhibitory G

    protein that slows down the activity of adenylate cyclase,

    thus lowering cytosolic levels of cAMP (Fig. 25-4). This

    greatly potentiates platelet responses by other agonists

    because it allows calcium ionsreleased from the DTS

  • 8/12/2019 Platelets and the Vessel Wall

    10/20

    1388 HEMOSTASIS

    initiating additional signaling cascades downstream of

    this serine/threonine kinase. As shown in Figure 25-5, it

    is the sum of these productsgenerated by the 2 isoformof PLC in response to adhesion and by the isoform ofPLC in response to ligand-GPCR interactionsthat the

    platelet integrates when deciding whether to become

    fully activated. This concept is important in the context

    of designing pharmacologic strategies to inhibit plateletfunction because blocking adhesion and its consequent

    activation of PLC2 leaves PLC-mediated platelet acti-vation largely intact, and vice versa.

    The second class of signal amplifiers consists of the

    cell surface integrins themselves.191As illustrated in the

    middle section of Figure 25-5, when ligands bind to

    integrins, the Src family kinases associated with integrin

    cytoplasmic tails192trigger a series of incompletely under-

    stood amplification events193that have been collectively

    termed outside-in signaling.194-196Although this has best

    been demonstrated after interaction of the major platelet

    integrin IIb3 with its ligand fibrinogen, signals also

    probably emanate from 21134,197,198

    and 61132

    uponengaging collagen and laminin, respectively. Activation

    signals from the latter two may be relatively weak by

    comparison because of the fact that only a few thousand

    of each are expressed on each platelet as compared with

    50,000 to 80,000 IIb3 receptors per cell.199,200 Theprotein kinases Syk and FAK have been shown to become

    activated downstream of IIb3engagement, as has activa-tion of PLC2.201 However, the details of these eventsremain to be worked out. Finally, there is at least one

    autocrine loop that uses integrin-mediated outside-in

    signal amplificationthat being cleavage and rebinding

    of soluble CD40L after alpha granule fusion and

    secretion.155

    The third class of feed-forward amplification reac-

    tions is mediated by ligand-activated plasma membrane

    receptor tyrosine kinases. The first of these to be described

    were receptors for Gas6, a vitamin Kdependent protein

    related to the anticoagulant protein S. Gas6 is thought

    to reside in platelet alpha granules202,203and, like other

    alpha granule proteins, becomes secreted upon platelet

    activation. Interestingly, platelets have three different

    receptors for Gas6Axl, Sky, and Merall of which

    have active cytoplasmic tyrosine kinase activity (see Fig.

    25-5). Upon engagement, Gas6 receptors appear to be

    able to trigger tyrosine phosphorylation of the 3integrin

    cytoplasmic domain and thereby support outside-in inte-grin signaling, as well as activate phosphatidylinositol-

    3-kinase (PI3K) to further sustain granule secretion.154Platelets also express two members of the Eph receptor

    tyrosine kinase family, EphA4 and EphB1, which when

    in contact with their membrane-bound counter-receptor

    Ephrin B1, stimulate tyrosine phosphorylation of the

    integrin 3tail and activate the integrin activator Rap1b.204Like Gas6 signaling,154,205,206 genetic loss or pharmaco-

    logic blockade of Ephrin/Eph kinase interactions results

    in decreased ability to form a stable thrombus or retract

    a fibrin clot.207

    ACTIVATION OF THE MAJOR PLATELETINTEGRIN aIIBb3(GPIIB-IIIA COMPLEX)THEFINAL COMMON END POINT OF PLATELETACTIVATION

    Human platelets express at least five different members

    of the 24-member integrin family,196,208 including three

    1 integrins (21, 51, and 61specific for collagen,fibronectin, and laminin, respectively) and two 3integ-rinsv3and its close relative IIb3(also known as theGPIIb-IIIa complex). IIb3is by far the most abundantand well studied. This section focuses on our current

    understanding of how IIb3becomes transformed froma resting to an active ligand-bindingcompetent confor-

    mation, with the understanding that the biochemical and

    cell biologic principles described for this integrin may

    well apply to the others.

    As shown in schematic form on the left side of Figure

    25-6, IIb3exists on the platelet surface in a bent-overconformation that is unable to associate effectively with its

    major soluble ligands fibrinogen, VWF, and fibronec-tin.209,210Though relatively short, the cytoplasmic domains

    of IIband 3are thought to play a key role in maintainingthe off state of this integrin complex as a result of weak

    charge interactions between them211,212 that allow the

    hydrophobic transmembrane domain helices of each

    subunit to interact and maintain the integrin in a low-

    affinity state.213,214 When platelets become activated

    either by adhesion- or soluble agonist-mediated

    eventscalcium and DAG, generated as a result of the

    actions of PLC2 and PLC(see Figs. 25-3 and 25-5),bind to and activate PKC and the guanine exchange factor

    CalDAG-GEF1. As illustrated in Figure 25-6, each of

    these can independently activate Rap1215-219

    a low-molecular-weight GTPase that has been implicated in

    integrin activation.220-222Rap1 appears to activate integrins

    via an effector molecule known as RIAM (Rap1GTP-inter-

    acting adaptor molecule), which recruits the highly abun-

    dant cytosolic protein talin to the inner face of the plasma

    membrane to form an integrin activation complex. Binding

    of RIAM-associated talin to the 3integrin subunit repre-sents the final common step in integrin activation223-225

    because it disrupts the weak ionic clasp between the IIb3tails and thereby allows tail separation and a dramatic,

    rapid unfolding of the extracellular domain.210Simultane-

    ous conformational changes in the integrin head226,227

    result in the formation of an integrin receptor with highaffinity for its soluble adhesive ligands. Finally, clustering

    of integrins occurs228and ensures that bound ligands effec-

    tively broker with high avidity the platelet-platelet interac-

    tions that permit thrombus growth and stabilization.

    CELL SURFACE AND CYTOSOLIC PROTEINSTHAT LIMIT PLATELET RESPONSES

    As anyone who has suffered a myocardial infarction or

    thrombotic stroke can attest to, unrestrained thrombus

  • 8/12/2019 Platelets and the Vessel Wall

    11/20

    Chapter 25 Platelets and the Vessel Wall 1389

    growth at inappropriate sites can be as harmful as exces-

    sive bleeding because it can result in vessel occlusion,

    ischemia, and tissue damage. Numerous active processes

    are therefore in place to limit platelet responsiveness in

    healthy vessels so that thrombus growth is kept localizedto specific sites of vascular injury and dissolution of the

    platelet plug during recovery is facilitated.

    As discussed earlier, healthy endothelium contrib-

    utes to platelet passivation via rather continuous genera-

    tion of PGI2and NO, which act on platelets by activating

    adenylate and guanylate cyclases to increase intracellular

    levels of cAMP and cGMP. These messengers activate

    PKA and PKG, respectively (illustrated in Fig. 25-2).

    PKG controls the threshold for platelet activation pri-

    marily by phosphorylating the IP3 receptorassociated

    cGMP kinase substrate IRAG,229a protein that associates

    with PKG and IP3receptor type I to inhibit IP3-induced

    calcium release from intracellular stores.230,231Both PKA

    and PKG interfere with platelet activation by phosphory-

    lating and inactivating VASP (vasodilator-stimulated

    phosphoprotein),232a molecule with anticapping activity

    that is important for the processes of actin polymeriza-

    tion and filopod formation.233-237 PKC can also bind

    VASP and interfere with its ability to promote filopodiaformation, although this pathway is unique to collagen-

    stimulated platelets and does not involve regulation of

    PKA- or PKG-mediated VASP phosphorylation.238

    One of the better characterized inhibitory receptors

    in platelets is platelet endothelial cell adhesion molecule-

    1 (PECAM-1)a cell surface molecule composed of

    six extracellular immunoglobulin domains, the most

    amino-terminal of which engages in homophilic interac-

    tions with PECAM-1 molecules on other cells, and two

    cytoplasmic immunoreceptor tyrosinebased inhibitory

    motifs (ITIMs) that upon phosphorylation, recruit and

    activate the cytosolic SH2 domaincontaining protein

    tyrosine phosphatase-2 (SHP-2).239,240

    PECAM-1 hasbeen shown to negatively regulate both GPVI- and GPIb/

    V/IX-mediated platelet activation241-243perhaps by con-

    trolling the phosphorylation state of these two ITAM-

    bearing signaling receptorsand appears to be one of

    several inhibitory receptors that control the rate and

    extent of platelet thrombus formation in vivo.244Platelets

    have also recently been found to express two other immu-

    noglobulin/ITIM-containing molecules: triggering recep-

    tor expressed on myeloid (TREM) cellslike transcript-1

    (TLT-1) and products of the G6b gene. TLT-1 is con-

    tained within platelet alpha granules and is expressed

    on the platelet surface in an activation-dependent

    manner.146,245

    Although the two cytoplasmic ITIMs ofTLT-1 are capable of becoming phosphorylated and

    recruiting SHP-2,146 the extent to which TLT-1/SHP-2

    complexes regulate platelet function is not yet known.

    The G6bgene, which is located within the class III region

    of the human major histocompatibility complex,246gives

    rise to multiple alternatively spliced transcripts (G6b-A

    through G6b-G).247Platelets contain at least two (G6b-A

    and G6b-B)71,86,248 and possibly four (G6b-A, G6b-B,

    G6b-D, and G6b-E)249of these transcripts, and the G6b-

    B isoform contains cytoplasmic ITIMs that are capable

    of becoming tyrosine-phosphorylated and recruiting

    SHP-1 and SHP-2.247In platelets, the G6b-B isoform has

    been shown to be tyrosine-phosphorylated in resting andactivated platelets, but to associate with SHP-1 only

    upon platelet activation.248 Cross-linking of antibodies

    specific for G6bgene products has been shown to inhibit

    platelet aggregation in response to multiple stimuli249;

    however, whether these effects are due to the inhibitory

    function of G6b-B remains to be determined.

    Several inhibitory pathways have been identified in

    platelets that either regulate or are regulated by PI3Ka

    lipid kinase that phosphorylates the 3position of PIP2to generate phosphatidylinositol 3,4,5-triphoshate (PIP3),

    thereby creating docking sites on the inner face of the

    Ca2+

    +DAG

    CalDAG-GEF1

    PKCPKD

    1 Rap1RIAM/Talin

    Integrinactivation

    complex (IAC)

    PLCPLC

    Bent stalk

    +

    +

    +++++

    EGF4

    EGF3

    EGF2

    EGF1PSI

    Hybrid

    Fully

    exp

    osed

    ligan

    dbi

    ndin

    g

    domain

    -TD

    Calf2

    Calf1

    Thigh

    Clasped tails

    IIb

    3

    Crypticligandbinding

    domain

    FIGURE 25-6. Integrin activation. As shown in the schematic at thebottom, calcium and diacylglycerol (DAG), generated as a result of the

    combined actions of phospholipase (PLC) and PLC, activate twoproteins: (1) protein kinase C (PKC) and (2) the Rap guanine exchange

    factor CalDAG-GEF1. Each of these is able to independently activate

    the small guanosine triphosphates (GTPase) Rap1. In its GTP-bound

    form, Rap1 binds to and activates one of its effector molecules, Rap1GTP-

    interacting adaptor molecule (RIAM), which then binds talin to form

    an integrin activation complex (IAC). When the IAC binds specific sites

    within the 3cytoplasmic domain, the clasp breaks, thereby destabiliz-ing transmembrane domain helix associations that are thought to main-

    tain the integrin in its low-affinity state (shown on theleft

    ). Breaking

    the hinge causes extensive conformational changes in the extracellular

    domain and produces a high-affinity, ligand-bindingcompetent integ-

    rin (shown on the right). EGF, epidermal growth factor; PSI, plexin-

    semiphorin-integrin; -TD, beta terminal domain. (Portions adaptedfrom Wegener KL, Partridge AW, Han J, et al: Structural basis of integrin

    activation by talin. Cell 2007;128:171, with permission.)

  • 8/12/2019 Platelets and the Vessel Wall

    12/20

    1390 HEMOSTASIS

    plasma membrane for Pleckstrin homology (PH)

    domaincontaining molecules.250The actions of PI3K are

    opposed by the lipid phosphatase SHIP1 (SH2 domain

    containing inositol 5-phosphatase 1, which hydrolyzesthe 5-phosphate of PIP3).251,252In platelets, SHIP1 hasbeen shown to downregulate PIP3generation after IIb3-mediated outside-in signaling253and thus may interfere

    with the feed-forward amplification pathways thatincrease the efficiency with which platelets are recruited

    to growing thrombi. Interestingly, the 5-inositol phos-phatase activity of SHIP1 appears to be enhanced, at

    least in part, by the actions of the Src family tyrosine

    kinase Lyn,254-256 which itself has been shown to limit

    platelet aggregation in response to GPVI-specific

    stimuli257 and after platelet spreading on immobilized

    fibrinogen.253

    Akt (also known as protein kinase B) is a PH domain

    containing serine/threonine kinase that is a well-charac-

    terized effector of PI3K.258,259Akt contributes positively

    to platelet activation in multiple ways, one of which

    appears to be by inactivating the serine/threonine kinaseglycogen synthase kinase 3 (GSK3). The GSK3 family is

    composed of three isoforms (, , 2) that are constitu-tively active in resting cells but become inactivated in

    activated cells by Akt-mediated phosphorylation.260Plate-

    lets express two isoforms of GSK3 ( and ), both ofwhich become phosphorylated and inactivated after

    exposure of the platelet to multiple agonists that activate

    PI3K and Akt.261 Whereas initial studies reported that

    specific inhibitors of GSK3 activity block rather than

    enhance platelet responses to agonist stimulation,261 a

    recent report suggest that as in other cells, the isoformof GSK3 acts as a negative regulator of platelet function

    both in vitro and in vivo.262

    ADDITIONAL ROLES FOR PLATELETS INVASCULAR PHYSIOLOGY: VESSEL REPAIR(ANGIOGENESIS), INFLAMMATION, ANDATHEROTHROMBOSIS

    In addition to being essential for primary hemostasis,

    activated platelets and their secreted products have the

    ability to influence a broad array of pathophysiologic

    processes, including leukocyte trafficking and inflamma-

    tion, tissue regeneration and angiogenesis, and both the

    beginning and end stages of atherosclerosis.Activated platelets that become spread on compo-

    nents of the extracellular matrix, or on each other, display

    an altered surface phenotypethe most prominent of

    which is exposure of several thousand copies of the alpha

    granulederived membrane protein P-selectin. P-selectin

    is also expressed on cytokine-activated endothelial cells.

    Thus, after either a thrombotic or inflammatory event,

    P-selectin appears on the luminal face of the vessel wall,

    where it serves to recruit monocytes and neutrophils into

    the underlying tissue by binding PSGL-1a constitu-

    tively expressed counter-receptor for P-selectin that is

    present on most leukocytes. In vivo, mice lacking P-selec-

    tin exhibit greatly diminished leukocyte rolling, delayed

    recruitment into sites of inflammation, and increased

    susceptibility to infection.263,264Although endothelial P-

    selectin no doubt has a major role in leukocyte capture,

    platelet P-selectin probably plays a prominent role in

    secondary capture.265,266As in platelets, tethering also

    initiates activation of leukocyte integrins, which are thenable to mediate cell spreading and transendothelial migra-

    tion. P-selectin/PSGL-1 interactions therefore constitute

    an important link between thrombosis and inflamma-

    tion.150,267 Other platelet/leukocyte receptor/counter-

    receptor pairs have also been shown to facilitate the

    inflammatory response, including binding of platelet-

    associated fibrinogen to the leukocyte integrin MAC-1268

    and platelet JAM-3 binding to MAC-1 on monocytes269

    and dendritic cells.270

    In addition to forming a platform for leukocyte

    recruitment during acute inflammation, platelets also

    deliver to the vessel wall proinflammatory chemokines

    that are thought to play a role in the development ofatherosclerosis by promoting further chemoattraction of

    leukocytes and stimulating proliferation of vessel wall

    smooth muscle cells and fibroblasts. Such secreted factors

    include the C-X-C chemokine platelet factor 4, macro-

    phage inflammatory protein 1a (MIP-1a), the C-C

    chemokine RANTES (regulated on activation, T cell

    expressed and secreted), CD40 ligand, platelet-derived

    growth factor (PDGF), and transforming growth factor

    (TGF-).150,267,271,272Activated platelets also synthesizede novo IL-1,273,274 a potent stimulator of endothelialcells and monocytes that upregulates adhesion molecule

    expression. Thus, platelets appear to contribute in a

    number of ways to the development and progression ofatherosclerotic lesions.

    Finally, so that one is not left with the impression

    that platelets only exacerbate chronic human disease, it

    should be noted that platelets and their secreted products

    were shown as early as 1969153 to be able to nurture

    the vascular endothelium, and they have recently been

    proposed as a source of biologic response modifiers for

    a plethora of uses, including organ preservation, gum

    restoration after dental procedures, and tissue repair after

    surgery.152 Their ability to adhere at sites of vascular

    injury and secrete both degradative enzymes and at the

    same time growth-promoting factors such as vascular

    endothelial growth factor (VEGF), PDGF, fibroblastgrowth factor (FGF), epidermal growth factor (EGF),

    and angiopoietin 1 allows them to play a uniquely sup-

    portive role in endothelial cell migration and survival

    during the process of wound healing and

    angiogenesis.151

    Acknowledgment

    The authors thank Robert I. Handin for valuable insights

    gleaned from versions of this chapter that appeared in

    earlier editions of this book. Research in the authors

  • 8/12/2019 Platelets and the Vessel Wall

    13/20

    Chapter 25 Platelets and the Vessel Wall 1391

    laboratories is supported by grants from the American

    Heart Association and the National Heart, Lung, and

    Blood Institute of the National Institutes of Health.

    REFERENCES

    1. de Gaetano G. A new blood corpuscle: an impossible

    interview with Giulio Bizzozero. Thromb Haemost. 2001;86:973-979.

    2. Yamada E. The fine structure of the megakaryocyte in the

    mouse spleen. Acta Anat (Basel). 1957;29:267-290.

    3. Shaklai M, Tavassoli M. Demarcation membrane system

    in rat megakaryocyte and the mechanism of platelet for-

    mation: a membrane reorganization process. J Ultrastruct

    Res. 1978;62:270-285.

    4. Italiano JE, Lecine P, Shivdasani RA, Hartwig JH. Blood

    platelets are assembled principally at the ends of proplate-

    let processes produced by differentiated megakaryocytes.

    J Cell Biol. 1999;147:1299-1312.

    5. Italiano JE Jr, Shivdasani RA. Megakaryocytes and

    beyond: the birth of platelets. J Thromb Haemost. 2003;1:

    1174-1182. 6. Patel SR, Hartwig JH, Italiano JE Jr. The biogenesis of

    platelets from megakaryocyte proplatelets. J Clin Invest.

    2005;115:3348-3354.

    7. Avecilla ST, Hattori K, Heissig B, et al. Chemokine-medi-

    ated interaction of hematopoietic progenitors with the

    bone marrow vascular niche is required for thrombopoi-

    esis. Nat Med. 2004;10:64-71.

    8. Radley JM, Haller CJ. The demarcation membrane system

    of the megakaryocyte: a misnomer? Blood. 1982;60:

    213-219.

    9. Schulze H, Korpal M, Hurov J, et al. Characterization of

    the megakaryocyte demarcation membrane system and its

    role in thrombopoiesis. Blood. 2006;107:3868-3875.

    10. Aster RH. Platelet sequestration studies in man. Br JHaematol. 1972;22:259-263.

    11. Mason KD, Carpinelli MR, Fletcher JI, et al. Programmed

    anuclear cell death delimits platelet life span. Cell. 2007;

    128:1173-1186.

    12. Franke JD, Dong F, Rickoll WL, et al. Rod mutations

    associated with MYH9-related disorders disrupt nonmus-

    cle myosin-IIA assembly. Blood. 2005;105:161-169.

    13. Caen JP, Nurden AT, Jeanneau C, et al. Bernard-Soulier

    syndrome: a new platelet glycoprotein abnormality. Its

    relationship with platelet adhesion to the subendothelium

    and with the factor VIII von Willebrand protein. J Lab

    Clin Med. 1976;87:586-596.

    14. Jenkins CS, Phillips DR, Clemetson KJ, et al. Platelet

    membrane glycoproteins implicated in ristocetin-inducedaggregation. Studies of the proteins on platelets from

    patients with Bernard-Soulier syndrome and von Wille-

    brands disease. J Clin Invest. 1976;57:112-124.

    15. Okita JR, Pidard D, Newman PJ, et al. On the association

    of glycoprotein Ib and actin-binding protein in human

    platelets. J Cell Biol. 1985;100:317-321.

    16. Fox JE. Linkage of a membrane skeleton to integral mem-

    brane glycoproteins in human platelets. Identification of

    one of the glycoproteins as glycoprotein Ib. J Clin Invest.

    1985;76:1673-1683.

    17. White JG, Burris SM, Hasegawa D, Johnson M. Micro-

    pipette aspiration of human blood platelets: a defect in

    Bernard-Souliers syndrome. Blood. 1984;63:1249-

    1252.

    18. Ware J, Russell S, Ruggeri ZM. Generation and rescue of

    a murine model of platelet dysfunction: the Bernard-

    Soulier syndrome. Proc Natl Acad Sci U S A. 2000;97:

    2803-2808.

    19. Corash L, Tan H, Gralnick HR. Heterogeneity of human

    whole blood platelet subpopulations. I. Relationship

    between buoyant density, cell volume, and ultrastructure.Blood. 1977;49:71-87.

    20. Karpatkin S. Heterogeneity of rabbit platelets. VI. Further

    resolution of changes in platelet density, volume, and

    radioactivity following cohort labelling with 75Se-seleno-

    methione. Br J Haematol. 1978;39:459-469.

    21. Corash L, Costa JL, Shafer B, et al. Heterogeneity of

    human whole blood platelet subpopulations. III. Density-

    dependent differences in subcellular constituents. Blood.

    1984;64:185-193.

    22. Mezzano D, Hwang K, Catalano P, Aster RH. Evidence

    that platelet buoyant density, but not size, correlates with

    platelet age in man. Am J Hematol. 1981;11:61-76.

    23. Newman PJ, Gorski J, White GC, et al. Enzymatic ampli-

    fication of platelet-specific messenger RNA using thepolymerase chain reaction. J Clin Invest. 1988;82:739-

    743.

    24. Kienast J, Schmitz G. Flow cytometric analysis of thiazole

    orange uptake by platelets: a diagnostic aid in the evalua-

    tion of thrombocytopenic disorders. Blood. 1990;75:

    116-121.

    25. White JG, Krivit W. An ultrastructural basis for the shape

    changes induced in platelets by chilling. Blood. 1967;30:

    625-635.

    26. Kenney DM, Linck RW. The cystoskeleton of unstimu-

    lated blood platelets: structure and composition of the

    isolated marginal microtubular band. J Cell Sci. 1985;

    78:1-22.

    27. Bentfeld ME, Bainton DF. Cytochemical localization oflysosomal enzymes in rat megakaryocytes and platelets. J

    Clin Invest. 1975;56:1635-1649.

    28. Behnke O. Electron microscopic observations on the

    membrane systems of the rat blood platelet. Anat Rec.

    1967;158:121-137.

    29. Escolar G, Leistikow E, White JG. The fate of the open

    canalicular system in surface and suspension-activated

    platelets. Blood. 1989;74:1983-1988.

    30. White JG, Escolar G. The blood platelet open canalicular

    system: a two-way street. Eur J Cell Biol. 1991;56:

    233-242.

    31. Gerrard JM, White JG, Peterson DA. The platelet dense

    tubular system: its relationship to prostaglandin synthesis

    and calcium flux. Thromb Haemost. 1978;40:224-231. 32. Papp B, Enyedi A, Pszty K, et al. Simultaneous presence

    of two distinct endoplasmic-reticulumtype calcium-

    pump isoforms in human cells. Characterization by

    radio-immunoblotting and inhibition by 2,5-di-(t-butyl)-

    1,4-benzohydroquinone. Biochem J. 1992;288:297-

    302.

    33. Wuytack F, Papp B, Verboomen H, et al. A sarco/endo-

    plasmic reticulum Ca2+-ATPase 3type Ca2+ pump is

    expressed in platelets, in lymphoid cells, and in mast

    cells [published erratum appears in J Biol Chem.

    1994;269(17):13056]. J Biol Chem. 1994;269:1410-

    1416.

  • 8/12/2019 Platelets and the Vessel Wall

    14/20

    1392 HEMOSTASIS

    34. Enouf J, Bredoux R, Papp B, et al. Human platelets

    express the SERCA2-b isoform of Ca2+-transport ATPase.

    Biochem J. 1992;286:135-140.

    35. Jardin I, Ben-amor N, Bartegi A, et al. Differential involve-

    ment of thrombin receptors in Ca2+release from two dif-

    ferent intracellular stores in human platelets. Biochem J.

    2007;401:167-174.

    36. Lopez JJ, Camello-Almaraz C, Pariente JA, et al. Ca2+

    accumulation into acidic organelles mediated by Ca2+-and vacuolar H+-ATPases in human platelets. Biochem J.

    2005;390:243-252.

    37. Haydon GB, Taylor DA. Microtubules in hamster plate-

    lets. J Cell Biol. 1965;26:673-676.

    38. Behnke O. Further studies on microtubules. A marginal

    bundle in human and rat thrombocytes. J Ultrastruct Res.

    1965;13:469-477.

    39. White JG, Krumwiede M. Isolation of microtubule coils

    from normal human platelets. Blood. 1985;65:1028-

    1032.

    40. White JG. Effects of colchicine and vinca alkaloids on

    human platelets. I. Influence on platelet microtubules and

    contractile function. Am J Pathol. 1968;53:281-291.

    41. Schwer HD, Lecine P, Tiwari S, et al. A lineage-restrictedand divergent beta-tubulin isoform is essential for the

    biogenesis, structure and function of blood platelets. Curr

    Biol. 2001;11:579-586.

    42. White JG, de Alarcon PA. Platelet spherocytosis: a new

    bleeding disorder. Am J Hematol. 2002;70:158-166.

    43. Fox JE, Boyles JK, Berndt MC, et al. Identification of a

    membrane skeleton in platelets. J Cell Biol. 1988;106:

    1525-1538.

    44. Fox JE, Reynolds CC, Morrow JS, Phillips DR. Spectrin

    is associated with membrane-bound actin filaments in

    platelets and is hydrolyzed by the Ca2+-dependent prote-

    ase during platelet activation. Blood. 1987;69:537-545.

    45. Hartwig JH, DeSisto M. The cytoskeleton of the resting

    human blood platelet: structure of the membrane skeletonand its attachment to actin filaments. J Cell Biol. 1991;

    112:407-425.

    46. Asijee GM, Sturk A, Bruin T, et al. Vinculin is a perma-

    nent component of the membrane skeleton and is incor-

    porated into the (re)organising cytoskeleton upon platelet

    activation. Eur J Biochem. 1990;189:131-136.

    47. Fox JEB. Identification of actin-binding protein as the

    protein linking the membrane skeleton to glycoproteins

    on platelet plasma membranes. J Biol Chem. 1985;260:

    11970-11977.

    48. Loftus JC, Choate J, Albrecht RM. Platelet activation and

    cytoskeletal reorganization: high voltage electron micro-

    scopic examination of intact and Triton-extracted whole

    mounts. J Cell Biol. 1984;98:2019-2025. 49. Newman PJ, Hillery CA, Albrecht R, et al. Activation-

    dependent changes in human platelet PECAM-1: phos-

    phorylation, cytoskeletal association, and surface

    membrane redistribution. J Cell Biol. 1992;119:

    239-246.

    50. Bennett JS, Zigmond S, Vilaire G, et al. The platelet cyto-

    skeleton regulates the affinity of the integrin IIb3 forfibrinogen. J Biol Chem. 1999;274:25301-25307.

    51. Hato T, Pampori N, Shattil SJ. Complementary roles for

    receptor clustering and conformational change in the

    adhesive and signaling functions of integrin IIb3. J CellBiol. 1998;141:1685-1695.

    52. Fox JEB, Shattil SJ, Kinlough-Rathbone RL, et al. The

    platelet cytoskeleton stabilizes the interaction between

    IIb3 and its ligand and induces selective movementof ligand-occupied integrin. J Biol Chem. 1996;271:

    7004-7011.

    53. Fox JE, Lipfert L, Clark EA, et al. On the role of the

    platelet membrane skeleton in mediating signal transduc-

    tion. Association of GP IIb-IIIa, pp60c-src, pp62c-yes,

    and the p21ras GTPase-activating protein with themembrane skeleton. J Biol Chem. 1993;268:25973-

    25984.

    54. Nachmias VT. Cytoskeleton of human platelets at rest and

    after spreading. J Cell Biol. 1980;86:795-802.

    55. Phillips DR, Jennings LK, Edwards HH. Identification of

    membrane proteins mediating the interaction of human

    platelets. J Cell Biol. 1980;86:77-86.

    56. Rosenberg S, Stracher A, Lucas RC. Isolation and char-

    acterization of actin and actin-binding protein from

    human platelets. J Cell Biol. 1981;91:201-211.

    57. Rosenberg S, Stracher A, Burridge K. Isolation and char-

    acterization of a calcium-sensitive alpha-actininlike pro-

    tein from human platelet cytoskeletons. J Biol Chem.

    1981;256:12986-12991. 58. Kroll MH, Harris TS, Moake JL, et al. von Willebrand

    factor binding to platelet GPIb initiates signals for platelet

    activation. J Clin Invest. 1991;88:1568-1573.

    59. White JG. Fine structural alterations induced in platelets

    by adenosine diphosphate. Blood. 1968;31:604-622.

    60. Carlsson L, Markey F, Blikstad I, et al. Reorganization of

    actin in platelets stimulated by thrombin as measured by

    the DNase I inhibition assay. Proc Natl Acad Sci U S A.

    1979;76:6376-6380.

    61. Jennings LK, Fox JE, Edwards HH, Phillips DR. Changes

    in the cytoskeletal structure of human platelets following

    thrombin activation. J Biol Chem. 1981;256:6927-6932.

    62. Fox JE, Phillips DR. Polymerization and organization

    of actin filaments within platelets. Semin Hematol.1983;20:243-260.

    63. Hartwig JH. Mechanisms of actin rearrangements medi-

    ating platelet activation. J Cell Biol. 1992;118:1421-

    1442.

    64. Barkalow KL, Italiano JE Jr, Chou DE, et al. -Adducindissociates from F-actin and spectrin during platelet acti-

    vation. J Cell Biol. 2003;161:557-570.

    65. Fox JE, Phillips DR. Role of phosphorylation in mediating

    the association of myosin with the cytoskeletal structures

    of human platelets. J Biol Chem. 1982;257:4120-4126.

    66. Booyse F, Rafelson ME Jr. In vitro incorporation of

    amino-acids into the contractile protein of human blood

    platelets. Nature. 1967;215:283-284.

    67. Kieffer N, Guichard J, Farcet JP, et al. Biosynthesis ofmajor platelet proteins in human blood platelets. Eur J

    Biochem. 1987;164:189-195.

    68. Gnatenko DV, Dunn JJ, McCorkle SR, et al. Transcript

    profiling of human platelets using microarray and serial

    analysis of gene expression. Blood. 2003;101:2285-

    2293.

    69. McRedmond JP, Park SD, Reilly DF, et al. Integration of

    proteomics and genomics in platelets: a profile of platelet

    proteins and platelet-specific genes. Mol Cell Proteomics.

    2004;3:133-144.

    70. Dittrich M, Birschmann I, Pfrang J, et al. Analysis of

    SAGE data in human platelets: features of the transcrip-

  • 8/12/2019 Platelets and the Vessel Wall

    15/20

    Chapter 25 Platelets and the Vessel Wall 1393

    tome in an anucleate cell. Thromb Haemost. 2006;95:

    643-651.

    71. Macaulay IC, Tijssen MR, Thijssen-Timmer DC, et al.

    Comparative gene expression profiling of in vitro differ-

    entiated megakaryocytes and erythroblasts identifies novel

    activatory and inhibitory platelet membrane proteins.

    Blood. 2007;109:3260-3269.

    72. Denis MM, Tolley ND, Bunting M, et al. Escaping the

    nuclear confines: signal-dependent pre-mRNA splicing inanucleate platelets. Cell. 2005;122:379-391.

    73. Schwertz H, Tolley ND, Foulks JM, et al. Signal-depen-

    dent splicing of tissue factor pre-mRNA modulates the

    thrombogenicity of human platelets. J Exp Med. 2006;203:

    2433-2440.

    74. Weyrich AS, Denis MM, Schwertz H, et al. mTOR-

    dependent synthesis of Bcl-3 controls the retraction of

    fibrin clots by activated human platelets. Blood. 2007;109:

    1975-1983.

    75. OFarrell PH. High resolution two-dimensional electro-

    phoresis of proteins. J Biol Chem. 1975;250:4007-4021.

    76. Ames GF, Nikaido K. Two-dimensional gel electro-

    phoresis of membrane proteins. Biochemistry. 1976;15:

    616-623. 77. Macaulay IC, Carr P, Gusnanto A, et al. Platelet genomics

    and proteomics in human health and disease. J Clin

    Invest. 2005;115:3370-3377.

    78. Dittrich M, Birschmann I, Stuhlfelder C, et al. Under-

    standing platelets. Lessons from proteomics, genomics

    and promises from network analysis. Thromb Haemost.

    2005;94:916-925.

    79. Martens L, Van DP, Van DJ, et al. The human platelet

    proteome mapped by peptide-centric proteomics: a func-

    tional protein profile. Proteomics. 2005;5:3193-3204.

    80. Marcus K, Moebius J, Meyer HE. Differential analysis of

    phosphorylated proteins in resting and thrombin-stimu-

    lated human platelets. Anal Bioanal Chem. 2003;376:

    973-993. 81. Maguire PB, Fitzgerald DJ. Platelet proteomics. J Thromb

    Haemost. 2003;1:1593-1601.

    82. Garcia A, Senis YA, Antrobus R, et al. A global proteomics

    approach identifies novel phosphorylated signaling pro-

    teins in GPVI-activated platelets: involvement of G6f, a

    novel platelet Grb2-binding membrane adapter. Pro-

    teomics. 2006;6:5332-5343.

    83. Gevaert K, Eggermont L, Demol H, Vandekerckhove J. A

    fast and convenient MALDI-MS based proteomic

    approach: identification of components scaffolded by the

    actin cytoskeleton of activated human thrombocytes. J

    Biotechnol. 2000;78:259-269.

    84. Maguire PB, Moran N, Cagney G, Fitzgerald DJ. Applica-

    tion of proteomics to the study of platelet regulatorymechanisms. Trends Cardiovasc Med. 2004;14:207-220.

    85. Coppinger JA, Cagney G, Toomey S, et al. Characteriza-

    tion of the proteins released from activated platelets leads

    to localization of novel platelet proteins in human athero-

    sclerotic lesions. Blood. 2004;103:2096-2104.

    86. Moebius J, Zahedi RP, Lewandrowski U, et al. The human

    platelet membrane proteome reveals several new potential

    membrane proteins. Mol Cell Proteomics. 2005;4:1754-

    1761.

    87. Maguire PB, Foy M, Fitzgerald DJ. Using proteomics to

    identify potential therapeutic targets in platelets. Biochem

    Soc Trans. 2005;33:409-412.

    88. Garcia BA, Smalley DM, Cho H, et al. The platelet mic-

    roparticle proteome. J Proteome Res. 2005;4:1516-

    1521.

    89. Damus PS, Hicks M, Rosenberg RD. Anticoagulant

    action of heparin. Nature. 1973;246:355-357.

    90. Esmon CT. The roles of protein C and thrombomodulin

    in the regulation of blood coagulation. J Biol Chem.

    1989;264:4743-4746.

    91. Fukudome K, Esmon CT. Identification, cloning, and reg-ulation of a novel endothelial cell protein C/activated

    protein C receptor. J Biol Chem. 1994;269:26486-26491.

    92. Levin EG. Latent tissue plasminogen activator produced

    by human endothelial cells in culture: evidence for an

    enzyme-inhibitor complex. Proc Natl Acad Sci U S A.

    1983;80:6804-6808.

    93. Hajjar KA, Hamel NM, Harpel PC, Nachman RL.

    Binding of tissue plasminogen activator to cultured human

    endothelial cells. J Clin Invest. 1987;80:1712-1719.

    94. Bunting S, Gryglewski R, Moncada S, Vane JR. Arterial

    walls generate from prostaglandin endoperoxides a sub-

    stance (prostaglandin X) which relaxes strips of mesen-

    teric and coeliac arteries and inhibits platelet aggregation

    Prostaglandins. 1976;12:897-913. 95. Gryglewski RJ, Bunting S, Moncada S, et al. Arterial walls

    are protected against deposition of platelet thrombi by a

    substance (prostaglandin X) which they make from pros-

    taglandin endoperoxides. Prostaglandins. 1976;12:685-

    713.

    96. Moncada S, Gryglewski R, Bunting S, Vane JR. An enzyme

    isolated from arteries transforms prostaglandin endoper-

    oxides to an unstable substance that inhibits platelet

    aggregation. Nature. 1976;263:663-665.

    97. Furchgott RF, Zawadzki JV. The obligatory role of endo-

    thelial cells in the relaxation of arterial smooth muscle by

    acetylcholine. Nature. 1980;288:373-376.

    98. Ignarro LJ, Buga GM, Wood KS, et al. Endothelium-

    derived relaxing factor produced and released from arteryand vein is nitric oxide. Proc Natl Acad Sci U S A.

    1987;84:9265-9269.

    99. Palmer RM, Ferrige AG, Moncada S. Nitric oxide release

    accounts for the biological activity of endothelium-derived

    relaxing factor. Nature. 1987;327:524-526.

    100. Boie Y, Rushmore TH, Darmon-Goodwin A, et al. Cloning

    and expression of a cDNA for the human prostanoid IP

    receptor. J Biol Chem. 1994;269:12173-12178.

    101. Namba T, Oida H, Sugimoto Y, et al. cDNA cloning of a

    mouse prostacyclin receptor. Multiple signaling pathways

    and expression in thymic medulla. J Biol Chem. 1994;

    269:9986-9992.

    102. Cheng Y, Austin SC, Rocca B, et al. Role of prostacyclin

    in the cardiovascular response to thromboxane A2. Science.2002;296:539-541.

    103. Pollock JS, Forstermann U, Mitchell JA, et al. Purification

    and characterization of particulate endothelium-derived

    relaxing factor synthase from cultured and native bovine

    aortic endothelial cells. Proc Natl Acad Sci U S A. 1991;

    88:10480-10484.

    104. Li Z, Xi X, Gu M, et al. A stimulatory role for cGMP-

    dependent protein kinase in platelet activation. Cell.

    2003;112:77-86.

    105. Radomski MW, Palmer RM, Moncada S. Endogenous

    nitric oxide inhibits human platelet adhesion to vascular

    endothelium. Lancet. 1987;2:1057-1058.

  • 8/12/2019 Platelets and the Vessel Wall

    16/20

    1394 HEMOSTASIS

    106. Sneddon JM, Vane JR. Endothelium-derived relaxing

    factor reduces platelet adhesion to bovine endothelial

    cells. Proc Natl Acad Sci U S A. 1988;85:2800-2804.

    107. Azuma H, Ishikawa M, Sekizaki S. Endothelium-depen-

    dent inhibition of platelet aggregation. Br J Pharmacol.

    1986;88:411-415.

    108. Radomski MW, Palmer RM, Moncada S. The anti-aggre-

    gating properties of vascular endothelium: interactions

    between prostacyclin and nitric oxide. Br J Pharmacol.1987;92:639-646.

    109. Macdonald PS, Read MA, Dusting GJ. Synergistic inhibi-

    tion of platelet aggregation by endothelium-derived relax-

    ing factor and prostacyclin. Thromb Res. 1988;49:

    437-449.

    110. Stamler J, Mendelsohn ME, Amarante P, et al.N-acetyl-

    cysteine potentiates platelet inhibition by endothelium-

    derived relaxing factor. Circ Res. 1989;65:789-795.

    111. Michelson AD, Benoit SE, Furman MI, et al. Effects of

    nitric oxide/EDRF on platelet surface glycoproteins. Am

    J Physiol. 1996;270:H1640-H1648.

    112. Kaczmarek E, Koziak K, Sevigny J, et al. Identification

    and characterization of CD39/vascular ATP diphospho-

    hydrolase. J Biol Chem. 1996;271:33116-33122.113. Marcus AJ, Broekman MJ, Drosopoulos JH, et al. The

    endothelial cell ecto-ADPase responsible for inhibition of

    platelet function is CD39. J Clin Invest. 1997;99:

    1351-1360.

    114. Dopheide SM, Maxwell MJ, Jackson SP. Shear-dependent

    tether formation during platelet translocation on von Wil-

    lebrand factor. Blood. 2002;99:159-167.

    115. Lopez JA, Berndt MC. The GPIb-IX-V Complex.

    Platelets. San Diego, CA, Academic Press, 2002, pp

    85-97.

    116. Coller BS, Peerschke EL, Scudder IE, Sullivan CA.

    Studies with a murine monoclonal antibody that abolishes

    ristocetin-induced binding of von Willebrand factor to

    platelets: additional evidence in support of GPIb as aplatelet receptor for von Willebrand factor. Blood. 1983;

    61:99-110.

    117. Lankhof H, Wu YP, Vink T, et al. Role of the glycoprotein

    Ib-binding A1 repeat and the RGD sequence in platelet

    adhesion to human recombinant von Willebrand factor.

    Blood. 1995;86:1035-1042.

    118. Savage B, Saldivar E, Ruggeri ZM. Initiation of platelet

    adhesion by arrest onto fibrinogen or translocation on von

    Willebrand factor. Cell. 1996;84:289-297.

    119. Kasirer-Friede A, Cozzi MR, Mazzucato M, et al.

    Signaling through GP Ib-IX-V activates alpha IIb beta

    3 independently of other receptors. Blood. 2004;103:

    3403-3411.

    120. Kasirer-Friede A, Moran B, Nagrampa-Orje J, et al.ADAP is required for normal lIb3activation by VWF/GPIb-IX-V and other agonists. Blood. 2007;109:

    1018-1025.

    121. Sullam PM, Hyun WC, Szollosi J, et al. Physical proximity

    and functional interplay of the glycoprotein Ib-IX-V

    complex and the Fc receptor FcRIIA on the plateletplasma membrane. J Biol Chem. 1998;273:5331-

    5336.

    122. Falati S, Edmead CE, Poole AW. Glycoprotein Ib-V-IX, a

    receptor for von Willebrand factor, couples physically and

    functionally to the Fc receptor -chain, fyn, and lyn toactivate human platelets. Blood. 1999;94:1648-1656.

    123. Nesbitt WS, Kulkarni S, Giuliano S, et al. Distinct glyco-

    protein Ib/V/IX and integrin lIb3-dependent calciumsignals cooperatively regulate platelet adhesion under

    flow. J Biol Chem. 2002;277:2965-2972.

    124. Mazzucato M, Pradella P, Cozzi MR, et al. Sequential

    cytoplasmic calcium signals in a 2-stage platelet activation

    process induced by the glycoprotein Ibmechanorecep-tor. Blood. 2002;100:2793-2800.

    125. Jackson SP, Nesbitt WS, Kulkarni S. Signaling eventsunderlying thrombus formation. J Thromb Haemost.

    2003;1:1602-1612.

    126. Mangin P, Yuan Y, Goncalves I, et al. Signaling role for

    PLC2 in platelet glycoprotein Ibcalcium flux and cyto-skeletal reorganization involvement of a pathway distinct

    from FcR-chain and FcRIIA. J Biol Chem. 2003;278:32880-32891.

    127. Rathore V, Wang D, Newman DK, Newman PJ. Phospho-

    lipase C2 contributes to stable thrombus formation onVWF. Febs Lett. 2004;573:26-30.

    128. Goncalves I, Nesbitt WS, Yuan Y, Jackson SP. Importance

    of temporal flow gradients and integrin IIb3 mechano-transduction for shear activation of platelets. J Biol Chem.

    2005;280:15430-15437.129. Staatz WD, Rajpara SM, Wayner EA, et al. The membrane

    glycoprotein Ia-IIa (VLA-2) complex mediates the Mg2+-

    dependent adhesion of platelets to collagen. J Cell Biol.

    1989;108:1917-1924.

    130. Sonnenberg A, Modderman PW, Hogervorst F. Laminin

    receptor on platelets is the integrin VLA-6. Nature.

    1988;336:487-489.

    131. Gibbins JM, Okuma M, Farndale R, et al. Glycoprotein

    VI is the collagen receptor in platelets which underlies

    tyrosine phosphorylation of the Fc receptor -chain. FebsLett. 1997;413:255-259.

    132. Inoue O, Suzuki-Inoue K, McCarty OJ, et al. Laminin

    stimulates spreading of platelets through integrin 61-

    dependent activation of GPVI. Blood. 2006;107:1405-1412.

    133. Tsuji M, Ezumi Y, Arai M, Takayama H. A novel

    association of Fc receptor -chain with glycoproteinVI and their co-expression as a collagen receptor

    in human platelets. J Biol Chem. 1997;272:23528-

    23531.

    134. Chen H, Kahn ML. Reciprocal signaling by integrin and

    nonintegrin receptors during collagen activation of plate-

    lets. Mol Cell Biol. 2003;23:4764-4777.

    135. Richardson JL, Shivdasani RA, Boers C, et al. Mecha-

    nisms of organelle transport and capture along proplate-

    lets during platelet production. Blood. 2005;106:

    4066-4075.

    136. Stenberg PE, Shuman MA, Levine SP, Bainton DF.Redistribution of alpha-granules and their contents in

    thrombin-stimulated platelets. J Cell Biol. 1984;98:

    748-760.

    137. Yarovoi HV, Kufrin D, Eslin DE, et al. Factor VIII ectopi-

    cally expressed in platelets: efficacy in hemophilia A treat-

    ment. Blood. 2003;102:4006-4013.

    138. Shi Q, Wilcox DA, Fahs SA, et al. Factor VIII ectopically

    targeted to platelets is therapeutic in hemophilia A with

    high-titer inhibitory antibodies. J Clin Invest. 2006;116:

    1974-1982.

    139. Handagama PJ, George JN, Shuman MA, et al. Incorpo