Overview of Hemostasis and Thrombosis

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

C H A P T E R 122 

OVERVIEW OF HEMOSTASIS AND THROMBOSIS


James C. Fredenburgh and Jeffrey I. Weitz

Hemostasis preserves vascular integrity by balancing the physiologic coagulation; and fibrinolytic agents that induce fibrin degradation
processes that maintain blood in a fluid state under normal circum- (see Chapter 149). With the predominance of platelets in arterial
stances and prevent excessive bleeding after vascular injury. Preserva- thrombi, strategies to inhibit or treat arterial thrombosis focus mainly
tion of blood fluidity depends on an intact vascular endothelium and on antiplatelet agents, although in the acute setting, strategies often
a complex series of regulatory pathways that maintain platelets in a include anticoagulants and fibrinolytic agents. When arterial thrombi
quiescent state and keep the coagulation system in check. In contrast, are occlusive and rapid restoration of blood flow is imperative,
arrest of bleeding requires rapid formation of hemostatic plugs at mechanical and pharmacologic methods enable thrombus extraction,
sites of vascular injury to prevent exsanguination. Perturbation of compression, or degradation. Although rarely used for this indication,
hemostasis can lead to bleeding or thrombosis. Bleeding will occur anticoagulants can also prevent recurrent ischemic events after acute
if there is failure to seal vascular leaks either because of defective myocardial infarction. Anticoagulants are the mainstay for prevention
hemostatic plug formation or because of premature breakdown of and treatment of venous thromboembolism because fibrin is the
the plugs. In contrast, thrombosis may occur if prothrombotic stimuli predominant component of venous thrombi (see Chapter 142).
are unregulated. Antiplatelet drugs are less effective than anticoagulants because of the
Thrombosis can occur in arteries or veins and is a major cause of limited platelet content of venous thrombi. Selected patients with
morbidity and mortality. Arterial thrombosis is the most common venous thromboembolism benefit from fibrinolytic therapy—for
cause of acute coronary syndromes, ischemic stroke, and limb gan- example, patients with massive or submassive pulmonary embolism
grene, whereas thrombosis in the deep veins of the leg leads to achieve more rapid restoration of pulmonary blood flow with systemic
postthrombotic syndrome and pulmonary embolism, which can be or catheter-directed fibrinolytic therapy than with anticoagulant
fatal. therapy alone. Certain patients with extensive deep vein thrombosis
Most arterial thrombi form on top of disrupted atherosclerotic in the iliac and/or femoral veins may also have a better outcome with
plaques, because plaque rupture exposes thrombogenic material in catheter-directed fibrinolytic therapy and/or mechanical thrombus
the plaque core to the blood.1 This material then triggers platelet extraction in addition to anticoagulants (see Chapter 143).
aggregation and fibrin formation, which results in the generation of This chapter provides an overview of hemostasis and thrombosis
a platelet-rich thrombus that temporarily or permanently occludes by highlighting the processes involved in platelet activation and
blood flow. Temporary occlusion of blood flow in coronary arteries aggregation, blood coagulation, and fibrinolysis.
may trigger unstable angina, whereas persistent obstruction causes
myocardial infarction. The same processes can occur in the cerebral
circulation, where temporary arterial occlusion may manifest as a HEMOSTATIC SYSTEM
transient ischemic attack, and persistent occlusion can lead to a
stroke. Likewise, critical limb ischemia can occur if there is superim- The major components of the hemostatic system are the vascular
posed thrombosis on ruptured atherosclerotic plaques in the major endothelium, platelets, and the coagulation and fibrinolytic systems.
arteries supplying blood to the lower extremities.
In contrast to arterial thrombi, venous thrombi rarely form at sites
of obvious vascular disruption. Although they can develop after surgi- Vascular Endothelium
cal trauma to veins, or secondary to indwelling venous catheters, they
usually originate in the valve cusps of the deep veins of the calf or in A monolayer of endothelial cells lines the intimal surface of the cir-
the muscular sinuses, where there is stasis.2 Sluggish blood flow in culatory tree and separates the blood from the prothrombotic suben-
these veins reduces the oxygen supply to the avascular valve cusps. dothelial components of the vessel wall (see Chapter 123). As such,
Hypoxemia induces endothelial cells lining the valve cusps to become the vascular endothelium encompasses about 1013 cells and covers a
activated and express adhesion molecules onto their surfaces. Tissue vast surface area. Rather than serving as a static barrier, the healthy
factor–bearing leukocytes and microparticles adhere to these activated vascular endothelium is a dynamic organ (Fig. 122.1) that actively
cells and induce coagulation. Impaired blood flow exacerbates local regulates hemostasis by inhibiting platelets, suppressing coagulation,
thrombus formation by reducing clearance of activated clotting promoting fibrinolysis, and modulating vascular tone and permeabil-
factors. These responses constitute the three axes of the Virchow triad ity.4 Defective vascular function can lead to bleeding if the endothe-
associated with development of thrombosis: stasis, hypercoagulability lium becomes more permeable to blood cells, if vasoconstriction does
of the blood, and activation or disruption of the endothelium. Calf not occur, or if premature degradation of hemostatic plugs reopens
vein thrombi that extend into the proximal veins of the leg can dis- repaired vasculature.
lodge and travel to the lungs to produce pulmonary embolism.3
Arterial and venous thrombi contain platelets and fibrin, but the
proportions differ. Arterial thrombi are rich in platelets because of Platelet Inhibition
the high shear on this side of the circulatory system. In contrast,
venous thrombi, which form under low shear conditions, contain Endothelial cells synthesize prostacyclin and nitric oxide and release
relatively few platelets and consist mostly of fibrin and trapped red them into the blood.4 These agents not only serve as potent vasodila-
blood cells. Because of the predominance of platelets, arterial thrombi tors but also inhibit platelet activation and subsequent aggregation
appear white, whereas venous thrombi appear red. by stimulating adenylate cyclase and increasing intracellular levels
The antithrombotic drugs used for prevention and treatment of of cyclic adenosine monophospahte (cAMP). In addition, endothe-
thrombosis target components of thrombi and include antiplatelet lial cells express CD39 on their surfaces, a membrane-associated
drugs, which inhibit platelets; anticoagulants, which attenuate ecto-adenosine diphosphatase (ADPase). By degrading adenosine

1831
1832 Part XII  Hemostasis and Thrombosis

Antiplatelet Anticoagulant Profibrinolytic

Heparan
NO CD39 Prostacyclin sulfate TM EPCR TFPI t-PA u-PA

Endothelium

Fig. 122.1  THE ANTITHROMBOTIC FUNCTIONS OF THE ENDOTHELIUM. The healthy endo-
thelium has (a) antiplatelet activity because of synthesis and release of prostacyclin and nitric oxide (NO) and
expression of CD39, a membrane-associated ectoADPase; (b) anticoagulant activity because of heparan sulfate
proteoglycan-mediated activation of antithrombin and expression of thrombomodulin (TM) and endothelial
protein C receptor (EPCR), which are involved in protein C activation, and surface-bound tissue factor
pathway inhibitor (TFPI); and (c) profibrinolytic activity because of release of tissue and urokinase-type
plasminogen activator (t-PA and u-PA, respectively).

diphosphate (ADP), which is a platelet agonist, CD39 attenuates Vascular Tone and Permeability
platelet activation.
In addition to synthesizing potent vasodilators, such as prostacyclin
and nitric oxide, endothelial cells also produce a group of counter-
Anticoagulant Activity regulatory peptides known as endothelins that induce vasoconstric-
tion. Endothelial cell permeability is influenced by the connections
Intact endothelial cells play an essential part in the regulation of that join endothelial cells to their neighbors. Macromolecules traverse
thrombin generation through a variety of mechanisms. Endothelial the endothelium via patent intercellular junctions, by endocytosis, or
cells produce heparan sulfate proteoglycans, which bind circulating through transendothelial pores. Vasodilatation, severe thrombocyto-
antithrombin and accelerate the rate at which it inhibits thrombin penia, and high doses of heparin can increase endothelial permeability,
and other coagulation enzymes. Tissue factor pathway inhibi- which may contribute to bleeding. Activated protein C may also
tor (TFPI), a naturally occurring inhibitor of coagulation, binds contribute to the barrier function of the endothelium.
heparan sulfate on the endothelial cell surface.5 Administration
of heparin or low-molecular-weight heparin (LMWH) displaces
glycosaminoglycan-bound TFPI from the vascular endothelium, and Platelets
released TFPI may contribute to the antithrombotic activity of these
drugs. Platelets are anucleate cellular particles released into the circulation
Endothelial cells regulate thrombin generation by expressing after programmed fragmentation of bone marrow megakaryocytes
thrombomodulin and endothelial cell protein C receptor (EPCR) on (see Chapter 124). Because they are anucleate, platelets have limited
their surfaces. Thrombomodulin binds thrombin and alters this capacity to synthesize proteins. Consequently, platelet protein
enzyme’s substrate specificity such that it no longer acts as a proco- composition is determined by the parent cell as well as by those
agulant but becomes a potent activator of protein C (see Chapter factors endocytosed from the circulation. Thrombopoietin, a glyco-
127). Activated protein C serves as an anticoagulant by degrading protein synthesized in the liver and kidneys, regulates megakaryo-
and inactivating activated factor V and factor VIII (factor Va and cytic proliferation and maturation as well as platelet production.
VIIIa, respectively), key cofactors involved in thrombin generation. Once they enter the circulation, platelets have a life span of 7 to
Protein S acts as a cofactor in this reaction, and EPCR enhances this 10 days.
pathway by binding protein C and presenting it to the thrombin– Damage to the intimal lining of the vessel exposes the underlying
thrombomodulin complex for activation. In addition to its role as an subendothelial matrix. Platelets home to sites of vascular disruption
anticoagulant, activated protein C also regulates inflammation and and adhere to the exposed matrix proteins (see Chapter 125). Adher-
preserves the barrier function of the endothelium.6 ent platelets undergo activation and not only release substances that
recruit additional platelets to the site of injury, but also promote
thrombin generation and subsequent fibrin formation (Fig. 122.2).
Fibrinolytic Activity A potent platelet agonist, thrombin amplifies platelet recruitment and
activation. Activated platelets then aggregate to form a plug that seals
The vascular endothelium promotes fibrinolysis by synthesizing and the leak in the vasculature.7 An understanding of the steps in these
releasing tissue-type and urokinase-type plasminogen activator (t-PA highly integrated processes helps pinpoint the sites of action of
and u-PA, respectively), which initiate fibrinolysis by converting antiplatelet drugs and rationalizes the utility of anticoagulants for the
plasminogen to plasmin (see Chapter 126). Endothelial cells in most treatment of arterial thrombosis and venous thrombosis.
vascular beds synthesize t-PA constitutively and release it in response
to stimuli such as thrombin or bradykinin. In contrast, perturbed
endothelial cells produce u-PA in the settings of inflammation and Adhesion
wound repair.
Endothelial cells also produce type 1 plasminogen activator Platelets adhere to exposed von Willebrand factor (vWF) and colla-
inhibitor (PAI-1), the major regulator of both t-PA and u-PA. There- gen, originating from endothelial cells and the subendothelium,
fore net fibrinolytic activity depends on the dynamic balance between respectively. The platelet monolayer promotes thrombin generation
the release of plasminogen activators and PAI-1. Fibrinolysis localizes and subsequent fibrin formation. These events depend on constitu-
to the endothelial cell surface because these cells express annexin II, tively expressed receptors on the platelet surface, α2β1 and glycopro-
a coreceptor for plasminogen and t-PA that promotes their interac- tein (GP) VI, which bind collagen, and GPIbα and GPIIb/IIIa
tion. Therefore healthy vessels actively resist thrombosis and help (αIIbβ3), which bind vWF. The platelet surface is crowded with recep-
maintain platelets in a quiescent state. tors, but those involved in adhesion are the most abundant: every
Chapter 122  Overview of Hemostasis and Thrombosis 1833

Fig. 122.2  SITES OF ACTION OF ANTIPLATELET DRUGS. Aspirin inhibits thromboxane A2 (TXA2)
synthesis by irreversibly acetylating cyclooxygenase-1 (COX-1). Reduced TXA2 release attenuates platelet
activation and recruitment to the site of vascular injury. Ticlopidine, clopidogrel, and prasugrel irreversibly
block P2Y12, a key ADP receptor on the platelet surface; whereas ticagrelor is a reversible inhibitor of P2Y12.
Abciximab, eptifibatide, and tirofiban inhibit the final common pathway of platelet aggregation by blocking
fibrinogen and von Willebrand factor (vWF) binding to activated glycoprotein (GP) IIb/IIIa. Vorapaxar
inhibits thrombin-mediated platelet activation by targeting protease activated receptor-1 (PAR-1), the major
thrombin receptor on platelets.
1834 Part XII  Hemostasis and Thrombosis

IIa
IIa

LDPR
IIa
RSFLLR
LDP
RLLFS

C C
C C

Response
Fig. 122.3  ACTIVATION OF PROTEASE-ACTIVATED RECEPTOR (PAR)-1 BY THROMBIN.
Thrombin (IIa) binds to the amino terminus of the extracellular domain of PAR-1 where it cleaves a specific
peptide bond. Cleavage of this bond generates a new amino-terminal sequence of Ser-Phe-Leu-Leu-Arg
(SFLLR) that acts as a tethered ligand and binds to the body of the receptor, thereby activating it. Thrombin
then dissociates from the receptor. Analogues of the first five or six amino acids of the tethered ligand sequences,
known as thrombin receptor agonist peptides, can independently activate PAR-1.

Although TP and the various ADP receptors signal through dif- IIIa exhibits minimal affinity for its ligands. Upon platelet activation,
ferent pathways, they all trigger an increase in the intracellular GPIIb/IIIa undergoes a conformational transformation, which
calcium concentration in platelets. This in turn induces shape change reflects transmission of inside-out signals from its cytoplasmic domain
via cytoskeletal rearrangement, granule mobilization and release, and to its extracellular domain.11 This transformation enhances the affin-
subsequent platelet aggregation. Activated platelets promote coagula- ity of GPIIb/IIIa for its ligands, fibrinogen, and, under high shear
tion by cycling phosphatidylserine from the inner membrane bilayer conditions, vWF (see Chapter 125). Cryptic Arg-Gly-Asp (RGD)
to the outer layer. Surface exposure of this anionic phospholipid is peptide sequences located in fibrinogen and vWF, as well as a platelet-
essential for assembly of coagulation factor complexes (see Chapter binding Lys-Gly-Asp (KGD) sequence in fibrinogen, mediate their
126). Once assembled, these clotting factor complexes trigger a burst interaction with GPIIb/IIIa. When subjected to high shear, circulat-
of thrombin generation and subsequent fibrin formation. In addition ing vWF elongates and exposes its platelet-binding domain, which
to converting fibrinogen to fibrin, thrombin amplifies platelet recruit- enables its interaction with the conformationally activated GPIIb/
ment and activation, thus promoting expansion of the platelet plug. IIIa. Similarly, initial binding to GPIIb/IIIa leads to a conformational
Thrombin binds to protease-activated receptors types 1 and 4 (PAR1 change in fibrinogen that unmasks additional binding sites. Divalent
and PAR4, respectively) on the platelet surface and cleaves their fibrinogen and multivalent vWF molecules serve as bridges and bind
extended amino-termini, thereby generating new amino-termini that adjacent platelets together. Once bound to GPIIb/IIIa, fibrinogen
serve as tethered ligands that bind internally and activate the receptors and vWF induce outside–inside signals that augment platelet activa-
(Fig. 122.3). Whereas low concentrations of thrombin cleave PAR1, tion and result in the activation of additional GPIIb/IIIa receptors,
PAR4 cleavage requires higher thrombin concentrations.10 Cleavage creating a positive feedback loop. Because GPIIb/IIIa acts as the final
of either receptor triggers platelet activation. effector in platelet aggregation, it is a logical target for potent anti-
In addition to providing a surface on which clotting factors platelet drugs (see Chapters 130 and 149). Fibrin, the ultimate
assemble, activated platelets also promote fibrin formation and sub- product of the coagulation system, tethers the platelet aggregates
sequent stabilization by releasing factor V, factor XI, fibrinogen, and together and anchors them to the site of injury.
factor XIII (see Chapter 125). Thus there is coordinated activation
of platelets and coagulation, and the fibrin network that results from
thrombin action helps anchor the platelet aggregates at the site of Coagulation
injury. Activated platelets also release adhesive proteins, such as vWF,
thrombospondin, and fibronectin, which augment platelet adhesion Coagulation results in the generation of thrombin, which converts
at sites of injury, as well as growth factors, such as platelet-derived soluble fibrinogen to fibrin. Coagulation occurs through a series of
growth factor (PDGF) and transforming growth factor-beta (TGF- concerted activation steps, wherein a nascent protease activates an
β), which promote wound healing. Platelet aggregation is the final inactive enzyme precursor (zymogen); a process that is repeated in a
step in the formation of the platelet plug. cascade-like fashion. The principal enzyme complexes are composed
of a vitamin K–dependent enzyme and a non-enzyme cofactor
assembled on anionic phospholipid membranes in a calcium-
Aggregation dependent fashion (see Chapter 126). Because each enzyme complex
activates a substrate that becomes the enzyme component of the
GPIIb/IIIa mediates platelet-to-platelet linkages that result in the subsequent complex, a small stimulus can produce a robust response.
formation of clumps of platelets. On nonactivated platelets, GPIIb/ Initially the small amount of thrombin generated activates
Chapter 122  Overview of Hemostasis and Thrombosis 1835

Vascular Contact
injury activation

VIIIaH
TF VIIa
Extrinsic IX X VIIIaL IXa X Intrinsic
IXa
tenase tenase

Prothrombinase

VaH
Xa Xa
VaL II
Xa

IIa

Fibrinogen Fibrin

Fig. 122.4  COAGULATION SYSTEM. Coagulation occurs through the action of discrete enzyme com-
plexes, which are composed of a vitamin K–dependent enzyme and a non-enzyme cofactor. These complexes
assemble on anionic phospholipid membranes in a calcium-dependent fashion. Vascular injury exposes tissue
factor (TF), which binds factor VIIa to form extrinsic tenase. Extrinsic tenase activates factors IX and X. Factor
IXa binds to factor VIIIa to form intrinsic tenase, which activates factor X. The contact system also leads to
activation of factor IX in response to exposure of anionic surfaces. Factor Xa binds to factor Va to form
prothrombinase, which converts prothrombin (II) to thrombin (IIa). Thrombin then converts soluble fibrino-
gen into insoluble fibrin.

non-enzyme cofactors and platelets. The activated platelets then in PAR4, the major thrombin receptor on mouse platelets, still
provide an anionic surface on which the coagulation complexes exhibit significant thrombus formation after laser injury. Red blood
assemble to promote rapid thrombin generation. The three predomi- cells also bind to damaged endothelium and express procoagulant
nant enzyme complexes involved in thrombin generation are extrinsic phospholipids on their surface, thereby providing additional sites for
tenase, intrinsic tenase, and prothrombinase, named for their sub- assembly of coagulation complexes. In addition, activated neutrophils
strates, factor X and prothrombin (Fig. 122.4). extrude web-like structures known as neutrophil extracellular traps
Coagulation can be divided into three phases: initiation, propaga- (NETs). Composed of nuclear DNA, histones, and metalloproteases,
tion, and termination.12,13 These phases reflect the exquisite control NETs promote coagulation by binding and activating platelets, trap-
over thrombin activity necessary to effect hemostasis. The initiation ping red blood cells, and activating the contact pathway. Thus cells
phase, which is predominantly mediated by extrinsic tenase, is contribute to coagulation at numerous sites in the cascade.
responsible for generating the initial burst of thrombin required for
platelet and coagulation factor activation. The propagation phase is
largely mediated by intrinsic tenase and is responsible for the explosive Extrinsic Tenase
thrombin generation necessary to sustain the coagulant response. The
termination phase, which is mediated by numerous protease inhibi- This complex forms upon exposure of tissue factor–expressing cells
tors, ensures that thrombin generation is localized and finite. Each to the blood.14 Tissue factor exposure occurs after atherosclerotic
of the activation complexes exhibits similar composition, assembly, plaque rupture because the core of the plaque is rich in macrophages
and regulation. and other cells that express tissue factor. Denuding injury to the vessel
Because initiation and propagation reactions require a membrane wall also exposes tissue factor constitutively expressed by subendothe-
surface for assembly of factors, cells play a key role in coagulation. lial fibroblasts and smooth muscle cells. In addition to cells in the
Tissue factor exposure on monocytes and extravascular cells and vessel wall, circulating monocytes and monocyte-derived micropar-
platelet membrane capacitation are recognized key steps. However, ticles (small membrane fragments) also provide a source of tissue
other cells also make important contributions. For example, damaged factor. When tissue factor–bearing monocytes or microparticles bind
endothelium appears to support coagulation because mice deficient to platelets or other leukocytes and their plasma membranes fuse,
1836 Part XII  Hemostasis and Thrombosis

tissue factor transfer occurs. By binding to adhesion molecules Emerging Role of the Contact Pathway
expressed on activated endothelial cells or to P-selectin on activated
platelets, these tissue factor–bearing cells or microparticles can initiate The contact pathway has emerged as an important player in thrombo-
or augment coagulation. This phenomenon likely explains how sis. It has long been recognized that the contact pathway is dispensable
venous thrombi develop in the absence of obvious vessel wall injury. for hemostasis because of the lack of a bleeding diathesis in patients
Tissue factor is an integral membrane protein that serves as a with hereditary deficiency of factor XII, prekallikrein, or high-molecular-
receptor for factor VIIa. Blood contains trace amounts of factor VIIa, weight kininogen and the mild bleeding diathesis with factor XI defi-
which has negligible activity in the absence of tissue factor.14 Although ciency relative to deficiencies of factor VIII or factor IX. However, until
recently, the role of the contact pathway remained elusive. The pathway
tissue factor is present on cell surfaces, it is proposed to exist in an was ignored for several decades because its activators were mainly
encrypted, inactive form. The decryption step is thought to occur by nonphysiologic substances such as kaolin or ellagic acid, compounded
a disulfide bond rearrangement catalyzed by protein-disulfide isom- by the observation that thrombin could activate factor XI, providing a
erase and exposure of phosphatidylserine on the outer membrane potential physiologic bypass for the contact system as the activator of
surface. Factor VIIa binds tissue factor in a calcium-dependent intrinsic tenase.
fashion to form the extrinsic tenase complex, which is a potent activa- The renaissance in our understanding of the important contribution
tor of factors IX and X. Once activated, factor IXa and factor Xa of the contact pathway to thrombus stabilization and propagation
serve as the enzyme components of intrinsic tenase and prothrombi- occurred as a result of the identification of new physiologic activators,
nase, respectively. Because sufficient levels of factor Xa and thrombin the attenuated thrombosis after venous or arterial injury observed in
mice deficient in factor XII or XI, and the development of contact
are formed in response to exposure of tissue factor, the extrinsic tenase pathway–specific inhibitors. Reports that factor XI-deficient mice were
complex is considered the essential mediator of the initiation phase. protected from arterial injury–induced thrombosis to the same extent
as factor IX-deficient mice, but did not experience bleeding, provide a
clear demarcation between hemostasis and thrombosis. The role of the
Intrinsic Tenase contact pathway in thrombosis gained credibility with the observation
that mice lacking factor XII were also resistant to clot formation at sites
Factor IXa binds to factor VIIIa on anionic platelet or cell surfaces of vascular injury. However, the physiologic mechanism for activation
to form the intrinsic tenase complex.15 Factor VIII circulates in blood of the pathway remained unclear until studies showed that nuclear
in complex with vWF. Thrombin cleaves factor VIII and releases it material released from neutrophils in the form of neutrophil extra-
cellular traps, nucleic acids, and inorganic polyphosphates released
from vWF, converting it to its activated form. Activated platelets from activated platelets or microorganisms, activated coagulation in a
express binding sites for factor VIIIa. Once bound, factor VIIIa binds contact pathway–dependent fashion. With a valid association between
factor IXa in a calcium-dependent fashion to form the intrinsic tenase the contact pathway and physiologic activators, investigation into the
complex, which then activates factor X. The loss in catalytic efficiency role of the contact pathway in thrombosis exploded.
of intrinsic tenase that occurs with a deficiency of factors VIII or IX Renewed interest in the contact pathway has spurred investigation of
in hemophilia A and B, respectively, highlights their importance. factors XI and XII as targets for new antithrombotic drugs. Agents tar-
Absence of the membrane or factor VIIIa almost completely abolishes geting factors XI and XII include biological inhibitors, antibodies, small
enzymatic activity, and the catalytic efficiency of the complete molecule inhibitors, and aptamers. Another approach is to reduce factor
complex is 106-fold greater than that of factor IXa alone.15 Because levels by inhibiting protein expression through the use of antisense
oligonucleotides (ASO). This approach has been successfully applied
intrinsic tenase activates factor X at a rate 50- to 100-fold faster than to factor XI, where administration of an ASO reduced postoperative
extrinsic tenase, it plays a critical role in the amplification of factor thrombosis in patients undergoing total knee arthroplasty, without an
Xa and subsequent thrombin generation. Thus intrinsic tenase is increase in bleeding. Similar interest in targeting the contact pathway is
crucial to the propagation phase of coagulation. directed at device-related thrombosis. Thrombosis is a major cause of
failure of blood-contacting medical devices, a problem that can lead to
life-threatening complications including pulmonary embolism, coronary
Contact Pathway occlusion, and stroke. and activation of the contact pathway by the
artificial surface is thought to be the primary cause. Therefore the
The contact pathway is so named because initial identification of its contact pathway has emerged as an attractive target for development
of agents that reduce thrombosis with little impact on hemostasis.
constituents (factors XII, XI, IX, and kallikrein), required contact
with artificial agents such as ellagic acid or silica for activation. For
this reason, the contact pathway lost prominence when the physiologic
tissue factor pathway was identified. Current thinking is that tissue
factor exposure represents the sole pathway for activation of coagula- In addition to its role in device-related thrombosis, the contact
tion and thus the contact system is unimportant for hemostasis pathway may also contribute to the stability of arterial and venous
because patients deficient in factor XII, prekallikrein, and high- thrombi.16 NETs, and DNA and RNA released from damaged cells
molecular-weight kininogen do not have bleeding problems (see in atherosclerotic plaques, activate factor XII, and mice given DNA-
Chapter 126). Although patients with severe deficiency of factor XI or RNA-degrading enzymes exhibit attenuated thrombosis at sites of
can bleed after trauma or surgery, spontaneous bleeding is uncom- arterial injury. Polyphosphates released from activated platelets also
mon, and the plasma level of factor XI does not reliably predict the activate factor XII, and may provide another stimulus for contact
propensity for bleeding (see Chapter 137). The capacity of thrombin pathway activation. Mice deficient in factor XII or factor XI form
to feedback and activate platelet-bound factor XI may help to explain small unstable thrombi at sites of arterial or venous damage, suggest-
this phenomenon. It also is possible that platelet-derived factor XI ing that factor XII and factor XI contribute to thrombogenesis (see
may be more important for hemostasis than circulating factor XI. Chapter 137). There is mounting evidence that the same is true in
We cannot ignore the contact pathway, however, because catheters humans. Thus patients with unstable angina have increased plasma
and other blood-contacting medical devices, such as stents or levels of factor XIa which could reflect activation by factor XIIa,
mechanical valves, likely trigger clotting through this mechanism. although activation by thrombin remains a possibility. The best evi-
Factor XII bound to charged or artificial surfaces undergoes a con- dence for the importance of the contact system comes from the results
formational change that results in its activation. Factor XIIa converts of a phase II study that demonstrated that knock down of factor XI
prekallikrein to kallikrein in a reaction accelerated by high-molecular- with an antisense oligonucleotide in patients undergoing elective
weight kininogen, and factor XIIa and kallikrein both feed back knee arthroplasty reduced the risk of postoperative venous thrombo-
to activate additional factor XII. Factor XIIa propagates coagulation embolism to a greater extent than enoxaparin. Furthermore, the
by activating factor XI and generating factor XIa, the predominant thrombi that did form in patients with low levels of factor XI were
activator of factor IX (Fig. 122.5). (See box on Emerging Role of the very small in size, consistent with the role of factor XI in thrombus
Contact Pathway) growth. Therefore the contact system is not only an important driver
Chapter 122  Overview of Hemostasis and Thrombosis 1837

inhibitors modulate coagulation: TFPI and antithrombin. TFPI,


K PK which is located on platelets and microvascular endothelial cells,
inhibits factor VIIa in a factor Xa-dependent manner. TFPI effectively
HK HK halts tissue factor-mediated initiation of coagulation, but not before
sufficient factor Xa is generated to propagate clotting. The high levels
of thrombin produced during the amplification phase are controlled
XII XIIa
Surface by antithrombin. This serine protease inhibitor (serpin) also inacti-
vates other coagulation proteases, including factors VIIa, IXa, Xa, and
Xia. Although antithrombin is abundant, it exhibits only moderate
inhibitory activity, except in the presence of cell-associated glycosami-
noglycans, such as heparan sulfate. This is the biochemical basis for
XI XIa use of heparin as an anticoagulant (see Chapter 149). Further regula-
tion of thrombin generation is mediated by the protein C anticoagu-
lant pathway which is catalyzed by thrombin. These processes ensure
IX IXa that thrombin generation is localized and limited. Sufficient thrombin
is produced, however, to ensure that coagulation occurs.

Surface
Common Fibrin Formation
Pathway
Thrombin converts soluble fibrinogen into insoluble fibrin. Fibrino-
gen is a dimeric molecule, each half of which is composed of three
polypeptide chains, the Aα, Bβ, and γ chains. Numerous disulfide
Thrombin
bonds covalently link the chains together and join the two halves of
Fig. 122.5  CONTACT SYSTEM. Factor XII (XII) is activated by contact the fibrinogen molecule (Fig. 122.6). Electron micrographic studies
with negatively charged surfaces. XIIa converts prekallikrein (PK) to kallikrein of fibrinogen reveal a trinodular structure with a central E domain
(K), which can feed back to activate more XII. Likewise, XIIa also can feed flanked by two D domains. Crystal structures show symmetry of
back to amplify its own generation. About 75% of circulating PK is bound design with the central E domain, which contains the amino termini
to high-molecular-weight kininogen (HK), which localizes it to anionic of the fibrinogen chains, joined to the lateral D domains by coiled-
surfaces and promotes PK activation. XIIa propagates clotting by activating coil regions.
XI, which then activates IX. The resultant IXa assembles into the intrinsic Fibrinogen circulates in a soluble form. Thrombin binds to the
tenase complex, which activates X to initiate the common pathway of coagu- amino termini of the Aα and Bβ chains of fibrinogen, where it cleaves
lation. Thrombin can feedback activate factor XI to further propagate specific peptide bonds to release fibrinopeptide A and fibrinopeptide
coagulation. B and generates fibrin monomer (Fig. 122.6). Because they are
products of thrombin action on fibrinogen, plasma levels of these
fibrinopeptides provide an index of thrombin activity. Fibrinopeptide
of thrombosis in blood-contacting medical devices but may also release creates new amino termini that extend as knobs from the E
contribute to thrombus propagation in situations where tissue factor domain of one fibrin monomer and insert into preformed holes in
initiates coagulation, such as after major surgery. the D domains of other fibrin monomers. This creates long strands
known as protofibrils, consisting of fibrin monomers noncovalently
linked together in a half-staggered, overlapping fashion.18
Prothrombinase Noncovalently linked fibrin protofibrils lack tensile strength.18
The stability of the fibrin network is enhanced by platelets and
Being the only physiologic producer of thrombin, the prothrombinase procoagulant cells.19 Platelets not only bind fibrin via GPIIb/IIIa
complex is essential for hemostasis. Factor Xa binds to factor Va, its and promote formation of a dense fibrin network, but they also
activated cofactor, on anionic phospholipid membrane surfaces to release factor XIII. By covalently cross-linking α and γ chains
form the prothrombinase complex. Activated platelets release factor of adjacent fibrin monomers, factor XIIIa stabilizes the fibrin in
V from their α-granules, and this platelet-derived factor V may play a calcium-dependent fashion and renders it relatively resistant to
a more important role in hemostasis than its plasma counterpart.17 physical strain and degradation. Factor XIII circulates in blood as a
Whereas plasma factor V requires thrombin activation to exert its heterodimer consisting of pairs of A and B subunits. The active and
cofactor activity, the partially activated factor V released from platelets calcium binding sites on factor XIII are localized to the A subunit.
already exhibits substantial cofactor activity. Activated platelets Platelets contain large amounts of factor XIII in their cytoplasm,
express specific factor Va binding sites on their surface, and bound but platelet-derived factor XIII consists only of the A subunits (see
factor Va serves as a receptor for factor Xa. The catalytic efficiency of Chapter 125). Both plasma and platelet factor XIII are activated by
factor Xa activation of prothrombin increases by 105-fold when factor thrombin.
Xa incorporates into the prothrombinase complex.13 Prothrombin Hemostasis depends on the dynamic balance between the forma-
binds to the prothrombinase complex, where it undergoes conversion tion of fibrin and its degradation. The fibrinolytic system mediates
to thrombin in a reaction that releases prothrombin fragment 1.2 fibrin breakdown.
(F1.2). Plasma levels of F1.2, therefore, provide a marker of pro-
thrombin activation. Prothrombin is the most abundant coagulation
factor, and the efficiency of activation generates high local levels of Fibrinolytic System
thrombin.
Fibrinolysis initiates when plasminogen activators convert plasmino-
gen to plasmin, which then degrades fibrin into soluble fragments.
Termination Blood contains two immunologically and functionally distinct plas-
minogen activators, t-PA and u-PA. t-PA mediates intravascular fibrin
Because thrombin clots fibrinogen, activates cells and platelets, and degradation, whereas u-PA binds to a specific u-PA receptor (u-PAR)
mediates anticoagulant and antifibrinolytic processes, it is imperative on the surface of cells, where it activates cell-bound plasminogen.
to regulate its activity and location. Thus the termination phase plays Consequently, pericellular proteolysis during cell migration and tissue
a critical role in balancing the procoagulant forces. Two principal remodeling and repair are the major functions of u-PA.20
1838 Part XII  Hemostasis and Thrombosis

COOH NH2 NH2 COOH


Aα FPA FPA Aα

Bβ FPB FPB Bβ Fibrinogen

γ γ

D domain Colied E domain Colied D domain


coil coil

D E D Fibrinogen

Thrombin FPA, FPB

D E D Fibrin monomer

D E D D E D
Fibrin polymer
E D D E D D E

Factor XIIIa

D E D D E D Cross-linked
fibrin polymer
E D D E D D E

Fig. 122.6  FIBRINOGEN STRUCTURE AND CONVERSION OF FIBRINOGEN TO FIBRIN. A


dimeric molecule, each half of fibrinogen is composed of three polypeptide chains, Aα, Bβ, and γ. Numerous
disulfide bonds (lines) covalently link the chains together and join the two halves of the fibrinogen molecule
to yield a trinodular structure with a central E domain linked via the coiled-coil regions to two lateral D
domains. To convert fibrinogen to fibrin, thrombin cleaves specific peptide bonds at the amino (NH2) termini
of the Aα and Bβ chains of fibrinogen to release fibrinopeptide A (FPA) and fibrinopeptide B (FPB), thereby
generating fibrin monomer. Fibrin monomers polymerize to generate protofibrils arranged in a half-staggered
overlapping fashion. By covalently cross-linking α and γ chains of adjacent fibrin monomers, factor XIIIa
stabilizes the fibrin network and renders it resistant to degradation.

Regulation of fibrinolysis occurs on a number of levels (see Single- and two-chain forms of t-PA convert plasminogen to plasmin.
Chapter 127). The substrate of the fibrinolytic system, fibrin, serves Native Glu-plasminogen is a single-chain polypeptide with a Glu
a transient but essential stimulatory role that subsides as it degrades. residue at its amino-terminus. Plasmin cleavage near the amino-
The serpins, PAI-1, and to a lesser extent, PAI-2, inhibit the plas- terminus generates Lys-plasminogen, a truncated form with a Lys
minogen activators, whereas α2-antiplasmin inhibits plasmin. residue at its new amino terminus.20 t-PA cleaves a single peptide
Endothelial cells synthesize PAI-1, which inhibits both t-PA and bond to convert single-chain Glu- or Lys-plasminogen into two-chain
u-PA, whereas monocytes and the placenta synthesize PAI-2, which plasmin, composed of a heavy chain containing five kringle domains
specifically inhibits u-PA. Thrombin-activatable fibrinolysis inhibitor and a light chain containing the catalytic domain. Because its open
(TAFI) also modulates fibrinolysis and provides a link between conformation exposes the t-PA cleavage site, Lys-plasminogen is a
fibrinolysis and coagulation.21 Thrombosis can occur if there is better substrate than Glu-plasminogen, which assumes a circular
impaired activation of the fibrinolytic system, whereas excessive closed conformation that renders this bond less accessible.
activation leads to bleeding. Therefore a review of the mechanisms t-PA has little enzymatic activity in the absence of fibrin, but its
of action of t-PA, u-PA, and TAFI is worthwhile. activity increases by at least three orders of magnitude when fibrin is
present. This increase in activity reflects the capacity of fibrin to serve
as a template that binds t-PA and plasminogen and promotes their
Mechanism of Action of Tissue-Type interaction. t-PA binds to fibrin via its finger and second kringle
Plasminogen Activator domains, whereas plasminogen binds fibrin via its kringle domains.
Kringle domains are triple loop-like structures that bind Lys residues
t-PA, a serine protease, contains five discrete domains: a fibronectin- on fibrin and other proteins. As fibrin undergoes degradation, more
like finger domain, an epidermal growth factor (EGF) domain, two Lys residues are exposed, which provide additional binding sites for
kringle domains, and a protease domain. Synthesized as a single-chain t-PA and plasminogen. Consequently, degraded fibrin stimulates t-PA
polypeptide, t-PA is converted into a two-chain form by plasmin. activation of plasminogen more than intact fibrin.
Chapter 122  Overview of Hemostasis and Thrombosis 1839

α2-Antiplasmin, another serpin, rapidly inhibits circulating TABLE Comparison of the Features of Disorders of Primary,
plasmin by docking to its first kringle domain and then inhibiting 122.1 Secondary, or Tertiary Hemostasis
the active site.20 Because plasmin binds to fibrin via its kringle
domains, plasmin generated on the fibrin surface resists inhibition by Features Primary Secondary Tertiary
α2-antiplasmin. This phenomenon endows fibrin-bound plasmin Components Platelets, vWF, Coagulation Fibrinolysis
with the capacity to degrade fibrin. Factor XIIIa cross-links small involved and vessel wall factors
amounts of α2-antiplasmin onto fibrin, which prevents premature
Site of Skin and Muscles, joints, Wounds and
fibrinolysis.19
bleeding mucocutaneous and deep genitourinary
Like fibrin, endothelial cells bind t-PA and plasminogen and
and soft tissues tissues tract
markedly promote activation by colocalization of enzyme and sub-
strate. Cell-surface binding is mediated by receptors such as annexin Physical Petechiae and Hematomas and Hematuria and
II, gangliosides, and α-enolase, as well as an orphan transmembrane findings ecchymoses hemarthroses menorrhagia
protein expressed with a carboxy-terminal lysine residue. Plasminogen Timing of Immediate Delayed Delayed
binds to exposed lysine residues on these receptors via its kringle bleeding
domains. Lipoprotein a, which also possesses kringle domains, Inheritance Autosomal Autosomal or Autosomal
impairs cell-based fibrinolysis by competing with plasminogen for dominant X-linked recessive
cell-surface binding. This phenomenon may explain the association recessive
between elevated lipoprotein a levels and atherosclerosis.
vWF, von Willebrand factor.

Mechanism of Action of Urokinase-Type


Plasminogen Activator TABLE
Disorders of Primary Hemostasis
122.2
Synthesized as a single-chain polypeptide, single-chain u-PA (scu-PA)
has minimal enzymatic activity. Plasmin converts scu-PA into a two- Components Affected Causes
chain form that is enzymatically active and capable of binding u-PAR, Platelets Quantitative or qualitative platelet disorders
the u-PA receptor on cell surfaces. Further cleavage at the amino-
vWF Inherited or acquired deficiency or
terminus of two-chain u-PA yields a truncated, lower-molecular-
dysfunction of vWF
weight form that lacks the u-PAR binding domain.
Two-chain forms of u-PA readily convert plasminogen to plasmin Vessel wall Vasculitis or abnormalities of connective
in the absence or presence of fibrin.20 In contrast, scu-PA does not tissue supporting the vasculature
activate plasminogen in the absence of fibrin, but it can activate vWF, von Willebrand factor.
fibrin-bound plasminogen, because plasminogen adopts the readily
activatable open conformation. Like the higher-molecular-weight
form of two-chain u-PA, scu-PA binds to cell surface u-PAR, where
plasmin can activate it. Many tumor cells elaborate u-PA and express Hemostatic Disorders
u-PAR on their surface. As with fibrin and plasminogen receptors,
colocalization of the reactants greatly promotes activation. Plasmin Bleeding can occur if there is abnormal platelet plug formation and/
generated on these cancer cells endows them with the capacity for or reduced thrombin generation and subsequent fibrin clot formation
metastasis because plasmin readily degrades components of the at the site of vascular injury; disorders of primary and secondary
extracellular matrix and activates growth factors and other degrada- hemostasis, respectively. Bleeding also can occur if the platelet/fibrin
tive proteases. clot is prematurely degraded because of excessive fibrinolysis; a dis-
order of tertiary hemostasis. The features distinguishing disorders of
primary, secondary, and tertiary hemostasis are outlined in Table
Mechanism of Action of TAFI 122.1. Hemorrhagic disorders can be inherited or acquired, and the
clinical and laboratory evaluation of such disorders is detailed in
TAFI, a procarboxypeptidase B–like molecule synthesized in the liver, Chapters 128 and 129, respectively.
circulates in blood in a latent form where thrombin bound to throm-
bomodulin can activate it to TAFIa (see Chapters 126 and 127).
Unless bound to thrombomodulin, thrombin activates TAFI ineffi- Disorders of Primary Hemostasis
ciently.21 TAFIa attenuates fibrinolysis by cleaving Lys residues from
the carboxy termini of chains of degrading fibrin, thereby removing Platelet plug formation, the first step in the arrest of bleeding at sites
binding sites for plasminogen, plasmin, and t-PA, attenuating activa- of injury, requires three key components (a) an adequate number of
tion, and promoting inhibition. TAFI links fibrinolysis to coagulation functional platelets, (b) vWF, the molecular glue that mediates
because the thrombin–thrombomodulin complex not only activates platelet adhesion to the damaged vessel wall even in the face of high
TAFI, which attenuates fibrinolysis, but also activates protein C, shear, and (c) a normal blood vessel that constricts in response to
which mutes thrombin generation. injury (Table 122.2). Because the platelet plug provides the first line
TAFIa has a short half-life in plasma because the enzyme is of defense against bleeding, patients with disorders of primary
unstable.21 Genetic polymorphisms can result in synthesis of more hemostasis often present with immediate bleeding after injury,
stable forms of TAFIa. Persistent attenuation of fibrinolysis by these and petechiae (pinpoint hemorrhages) may be noted. In addition to
variant forms of TAFIa may render patients susceptible to thrombosis. skin bleeding, mucocutaneous bleeding, which may manifest as
epistaxis, bleeding gums, or hematochezia, is common as is excessive
menstrual bleeding in women (see Chapter 128).
DISORDERS OF HEMOSTASIS OR THROMBOSIS Disorders of primary hemostasis may be inherited or acquired.22
Thrombocytopenia or congenital or acquired disorders of platelet
A physiologic host defense mechanism, hemostasis focuses on arrest function are common causes of bleeding. Thrombocytopenia can be
of bleeding by forming hemostatic plugs composed of platelets and the result of decreased production, which can occur because of failure,
fibrin at sites of vessel injury. In contrast, thrombosis reflects a infiltration, or fibrosis of the bone marrow (see Chapters 29 and 30),
pathologic process associated with intravascular thrombi that fill the increased platelet destruction, or abnormal distribution because
lumens of arteries or veins. of platelet pooling in the spleen (see Chapter 132). Increased
1840 Part XII  Hemostasis and Thrombosis

destruction of platelets can occur via immune mechanisms, such as bleeding with minimal trauma. Those with factor levels between 1%
immune thrombocytopenic purpura (ITP), alloimmune thrombocy- and 5% have an intermediate phenotype, whereas patients with factor
topenia, posttransfusion purpura, and drug-induced thrombocytope- VIII or IX levels above 5% usually have mild disease and bleed only
nia (see Chapter 131), or nonimmune mechanisms, which include with trauma or surgery. The frequency of bleeding episodes in patients
microangiopathic disorders, such as thrombotic thrombocytopenic with severe hemophilia can be reduced with prophylactic administra-
purpura and hemolytic uremic syndrome (see Chapter 134), as well tion of the appropriate factor concentrate; such treatment is also
as consumption because of activation of coagulation, such as occurs administered to hemophiliacs with overt bleeding, or in preparation
with disseminated intravascular coagulation (see Chapter 139). for surgery or other major interventions. Long-lasting factor VIII and
Platelet function disorders include disorders of platelet (a) adhesion, IX molecules have been developed to reduce the frequency of factor
such as von Willebrand disease (see Chapter 138) and Bernard-Soulier replacement. The half-lives of these recombinant full-length or
syndrome (see Chapter 125); (b) thromboxane synthesis; (c) secretion, truncated proteins have been prolonged by conjugating them to
such as alpha or dense granule deficiency or aspirin-like secretion hydrophilic polymers such as polyethylene glycol or by fusing them
defects; (d) aggregation, such as Glanzmann thrombasthenia (see with albumin or the Fc fragment of IgG1. Conjugation to polyethyl-
Chapter 125); or (e) procoagulant activity (Scott syndrome) where the ene glycol protects the proteins from proteolytic degradation, whereas
platelets fail to support clotting factor complex assembly (see Chapter fusion technology creates new recycling pathways that diminish
126). Acquired disorders of platelet function can occur in patients natural protein breakdown (see Chapter 136). Management of
taking drugs that impair platelet function, such as aspirin or nonste- hemophilia becomes more complicated if patients develop inhibitory
roidal antiinflammatory drugs, or in patients with uremia, paraproteins antibodies that attenuate or abolish the activity of the infused factor.
or myelodysplastic or myeloproliferative disorders (see Chapter 130). Congenital deficiencies of prothrombin (factor II), factors V, VII,
Bleeding can also occur with inflammation or malformations of X, or XI (hemophilia C), or fibrinogen are less common causes of
the blood vessels or abnormalities of the connective tissue supporting bleeding (see Chapter 137). In contrast, deficiencies of components of
the blood vessels. Inflammatory disorders include Henoch-Schonlein the contact pathway—factor XII, high-molecular-weight kininogen,
purpura (see Chapter 152) and the vasculitis that occurs with para- and prekallikrein—are not associated with bleeding. The clinical and
proteins or cryoglobulins or in patients with systemic lupus erythe- laboratory evaluation of such patients is detailed in Chapters 128 and
matosis or other immune disorders.23 Hereditary hemorrhagic 129, respectively, whereas their treatment is outlined in Chapter 115.
telangiectasia is an inherited disorder associated with malformations Acquired deficiencies of coagulation factors can result from
of the capillaries. Telangiectatic vessels can often be seen in the oral decreased synthesis due to severe liver disease, vitamin K deficiency
and nasal cavities of patients with this disorder and bleeding episodes, or intake of drugs that interfere with vitamin K metabolism, con-
primarily from the nose and gastrointestinal tract, are common. sumption because of excessive activation of coagulation (e.g., dis-
Abnormalities of the connective tissue matrix supporting the blood seminated intravascular coagulation; see Chapter 139), or accelerated
vessels include Marfan syndrome, Ehlers-Danlos syndrome, and clearance due to adsorption by paraproteins or amyloid (see Chapters
pseudoxanthoma elasticum.24 Patients with these disorders frequently 86 and 87) or to autoantibodies that shorten the half-life or attenuate
report easy bruising. or abolish clotting factor activity.
Congenital disorders of fibrinogen include absence or low levels
of fibrinogen (afibrinogenemia and hypofibrinogenemia, respectively)
Disorders of Secondary Hemostasis or synthesis of a dysfunctional protein (dysfibrinogenemia). Acquired
disorders of fibrinogen include decreased synthesis or production of
Secondary hemostasis depends on rapid generation of sufficient an abnormal fibrinogen, increased fibrinogen consumption or the
amounts of thrombin to generate a fibrin mesh that not only con- presence of inhibitors that interfere with fibrin polymerization, such
solidates the platelet aggregates that form at sites of vascular injury, as paraproteins, autoantibodies, particularly in patients with systemic
but also is stable enough to provide a barrier that prevents leakage of lupus erythematosis or other immune disorders or elevated levels of
blood from the damaged blood vessel.19 Secondary hemostasis can be fibrin(ogen) degradation products.
compromised by (a) impaired thrombin generation because of con- Stabilization of fibrin requires cross-linking of the α and γ chains
genital or acquired deficiencies of coagulation factors or cofactors or of adjacent fibrin monomers to yield a polymer that is resistant to
intake of drugs that inhibit one or more steps in the coagulation premature breakdown. Factor XIIIa, a transglutaminase, performs
pathways, (b) congenital or acquired fibrinogen deficiency or dys- this function by catalyzing the condensation of lysine residues on one
function, and/or (c) impaired cross-linking of fibrinogen because of chain with glutamic acid residues on another chain.25 Congenital or
congenital or acquired deficiency of factor XIII (Table 122.3). acquired deficiency of factor XIII can impair cross-linking, resulting
Examples of inherited deficiencies of coagulation factors include in bleeding. The hallmarks of severe factor XIII deficiency include
hemophilia A and B, deficiencies of factor VIII and factor IX, respec- umbilical stump bleeding in the neonatal period (see Chapter 150),
tively (see Chapter 135). Because of redundancy in the coagulation intracranial hemorrhage with little or no trauma, recurrent soft tissue
system, only patients with a factor VIII or factor IX level less than hemorrhages, and, in females, recurrent spontaneous miscarriages.
1% have severe disease characterized by spontaneous bleeding or

Disorders of Tertiary Hemostasis


TABLE
Disorders of Secondary Hemostasis
122.3 Tertiary hemostasis depends on the generation of plasmin, which
Components Affected Causes degrades fibrin and restores blood flow in damaged vessels. Premature
Coagulation factors Congenital deficiency, autoantibodies,
lysis of fibrin in hemostatic plugs can lead to bleeding; this can occur
increased consumption, or drugs that
systemically or can be localized (Table 122.4). Systemic fibrinolysis
attenuate thrombin generation or thrombin
that occurs in the absence of activation of coagulation, so-called
activity
primary hyperfibrinolysis, is rare but can occur with inherited defi-
ciency of PAI-1 or α2-antiplasmin, the inhibitors of the plasminogen
Fibrinogen Decreased production; increased consumption activators and plasmin, respectively, advanced liver disease, and some
or synthesis of an abnormal protein snake bites. More commonly, systemic hyperfibrinolysis is secondary
Impaired fibrin polymerization because of to activation of coagulation by procoagulants such as tissue factor
fibrin(ogen) degradation products or (e.g., in patients with metastatic cancer) or artificial surfaces (e.g.,
paraproteins in cardiopulmonary bypass surgery or with cardiac assist devices).
Fibrin cross-linking Congenital or acquired factor XIII deficiency Examples of localized hyperfibrinolysis include menorrhagia or hema-
turia after prostatectomy triggered by excessive plasmin generation
Chapter 122  Overview of Hemostasis and Thrombosis 1841

TABLE infarction and abnormal blood flow are the major triggers for left
Disorders of Tertiary Hemostasis ventricular thrombus formation. With rapid atrial fibrillation, there
122.4
also is stasis and turbulent blood flow in the left atrial appendage,
Component Affected Causes which is a long, blind-ended trabeculated pouch.27 This may lead to
Plasminogen activators Increased t-PA or u-PA release in the GU localized activation of endothelial cells and subsequent loss of their
tract or other tissues anticoagulant phenotype, a process amplified by adhesion of leuko-
cytes and subsequent elaboration of proinflammatory cytokines. The
Plasmin Deficiency of PAI-1 or α2-antiplasmin,
generation of thrombin creates a local hypercoagulable state that
resulting in an increased plasmin
likely promotes thrombus formation on the abnormal endothelium.
concentration
Embolization of these thrombi to the brain is a common cause of
Plasminogen activation Enhanced plasminogen activation ischemic stroke and the major cause of mortality and morbidity in
secondary to activation of coagulation patients with atrial fibrillation.
by procoagulants, such as cancer cells,
artificial surfaces, or snake venoms
GU, Genitourinary; PAI-1, plasminogen activator inhibitor 1; t-PA, tissue Venous Thrombosis
plasminogen activator; u-PA, urokinase-type plasminogen activator.
The causes of venous thrombosis include those associated with hyper-
coagulability, which can be genetic or acquired, and the mainly acquired
induced by the high concentrations of t-PA and u-PA in the uterus risk factors, such as advanced age, obesity, or cancer, which are associ-
and genitourinary tract, respectively. ated with immobility (see Chapters 140 and 142). Inherited hyperco-
agulable states and these acquired risk factors combine to establish the
intrinsic risk of thrombosis for each individual.2 Superimposed trigger-
Thrombotic Disorders ing factors, such as surgery, pregnancy, or hormonal therapy, modify
this risk, and thrombosis occurs when the combination of genetic,
Thrombosis may occur in arteries, in the chambers of the heart, or acquired, and triggering forces exceed a critical threshold.28
in the veins. Factors contributing to thrombosis in these sites include Some acquired or triggering factors entail a higher risk than
endothelial injury or activation, reduced blood flow, and hypercoagu- others. For example, major orthopedic surgery, neurosurgery, multiple
lability of the blood, the so-called Virchow triad. trauma, and metastatic cancer (particularly adenocarcinoma) are
associated with the highest risk; prolonged bed rest, antiphospholipid
antibodies (see Chapter 141), and the puerperium are associated with
Arterial Thrombosis an intermediate risk; whereas pregnancy, obesity, long-distance travel,
or the use of oral contraceptives or hormonal replacement therapy
Most arterial thrombi occur on top of disrupted atherosclerotic are mild risk factors. Up to half of patients who present with venous
plaques. Plaques with a thin fibrous cap and a lipid-rich core are most thromboembolism before the age of 45 have inherited hypercoagu-
prone to disruption. Erosion or rupture of the fibrous cap exposes lable disorders—so-called thrombophilia (see Chapter 140)—particu-
thrombogenic material in the lipid-rich core to the blood and triggers larly those whose event occurred in the absence of risk factors or with
platelet activation and thrombin generation. The extent of plaque minimal provocation, such as after minor trauma or a long-haul flight
disruption and the content of thrombogenic material in the plaque or with estrogen use.29
determine the consequences of the event, regardless of whether it
occurs in the cerebral circulation (see Chapter 145), the coronary
circulation (see Chapter 146), or the major arteries of the legs (see TREATMENT OF DISORDERS OF HEMOSTASIS
Chapter 148), but host factors also contribute. Breakdown of regula- AND THROMBOSIS
tory mechanisms that limit platelet activation and inhibit coagulation
can augment thrombosis at sites of plaque disruption. By the midpoint of the 20th century, two major anticoagulant drugs
Decreased production of nitric oxide and prostacyclin by diseased had been discovered, characterized, and given to humans for preven-
endothelial cells can trigger vasoconstriction and platelet activation. tion or treatment of thrombotic disorders. Heparin and vitamin K
Proinflammatory cytokines lower thrombomodulin expression by antagonists, such as warfarin, dominated treatment regimens for
endothelial cells, which promotes thrombin generation, and they decades. In the same era, determination of their mechanisms of action
stimulate PAI-1 expression which inhibits fibrinolysis. and dosing was aided by the discovery of the main players of the
Products of blood coagulation contribute to atherogenesis, as well coagulation system, resulting from the development of sensitive
as to its complications (see Chapter 144). Microscopic erosions in functional assays to monitor their activity. Heparin and warfarin still
the vessel wall trigger the formation of tiny platelet-rich thrombi.26 represent effective members of the anticoagulant armamentarium;
Activated platelets release PDGF and TGF-β, which promote a however, detailed understanding of the biochemistry and cell biology
fibrotic response. Thrombin generated at the site of injury not only of hemostasis has directly contributed to the development of new
activates platelets and converts fibrinogen to fibrin, but also activates therapies. Heparin derivatives with more predictable pharmacokinet-
PAR-1 on smooth muscle cells and induces their proliferation, migra- ics have improved therapy and reduced complications (see Chapter
tion, and elaboration of extracellular matrix. Incorporation of thrombi 149). Small molecule, direct inhibitors of thrombin and factor Xa have
into plaques promotes plaque growth, and decreased endothelial cell been developed as non–vitamin K antagonist (or direct) oral antico-
production of heparan sulfate—which normally limits smooth muscle agulants, such as dabigatran, rivaroxaban, apixaban, and edoxaban,
proliferation—contributes to plaque expansion.26 The multiple links that are replacing conventional therapies (see Chapter 149). The
between atherosclerosis and thrombosis have prompted the term resurgence of interest in the contact system as a potential mediator of
atherothrombosis (see Chapter 144). thrombosis has led to the investigation of factors IX, XI, and XII as
new targets for therapy. Likewise new antiplatelet agents that antago-
nize activation or aggregation steps are being used in conjunction with
Intracardiac Thrombosis aspirin to prevent and treat arterial thrombosis (see Chapter 146). On
the hemostasis side, regimens to treat bleeding disorders include
Thrombi can form in the left ventricle after transmural myocardial administration of factors VIIa, VIII, or IX, and prothrombin complex
infarction or with an aneurysm or dyskinetic ventricle, or in the concentrates (see Chapters 135, 136, and 137). The new agents and
left atrial appendage, particularly in patients with atrial fibrillation treatment regimens highlight the therapeutic benefit that has resulted
(see Chapter 147). Damage to the endothelium after myocardial from our detailed understanding of hemostasis and thrombosis.
1842 Part XII  Hemostasis and Thrombosis

REFERENCES 16. Woodruff RS, Sullenger B, Becker RC: The many faces of the contact
pathway and their role in thrombosis. J Thromb Thrombolysis 32(1):9–20,
1. Lippi G, Franchini M, Targher G: Arterial thrombus formation in 2011.
cardiovascular disease. Nat Rev Cardiol 8:502, 2011. 17. Fager AM, Wood JP, Bouchard BA, et al: Properties of procoagulant
2. Reitsma PH, Versteeg HH, Middledorp S: Mechanistic view of risk platelets: defining and characterizing the subpopulation binding a func-
factors for venous thromboembolism. Arterioscler Thromb Vasc Biol tional prothrombinase. Arterioscler Thromb Vasc Biol 30(12):2400–2407,
32(3):563–568, 2012. 2010.
3. Morris TA: Natural history of venous thromboembolism. Crit Care Clin 18. Lord ST: Molecular mechanisms affecting fibrin structure and stability.
27(4):869–884, 2011. Arterioscler Thromb Vasc Biol 31(3):494–499, 2011.
4. van Hinsbergh VW: Endothelium – role in regulation and coagulation 19. Wolberg AS: Plasma and cellular contributions to fibrin network forma-
and inflammation. Semin Immunopathol 34(1):93–106, 2012. tion, structure and stability. Haemophilia 16(Suppl 3):7–12, 2010.
5. Wood JP, Ellery PE, Maroney SA, et al: Biology of tissue factor pathway 20. Schaller J, Gerber SS: The plasmin-antiplasmin system: structural and
inhibitor. Blood 123(19):2934–2943, 2014. functional aspects. Cell Mol Life Sci 68(5):785–801, 2011.
6. Esmon CT: Protein C anticoagulant system–anti-inflammatory effects. 21. Heylen E, Willemse J, Hendriks D: An update on the role of car-
Semin Immunopathol 34(1):127–132, 2012. boxypeptidase U (TAFIa) in fibrinolysis. Front Biosci 17:2427–2450,
7. Nieswandt B, Pleines I, Bender M: Platelet adhesion and activation 2011.
mechanisms in arterial thrombosis and ischaemic stroke. J Thromb 22. Broos K, Feys HB, DeMeyer SF, et al: Platelets at work in primary
Haemost 9(Suppl 1):92–104, 2011. hemostasis. Blood Rev 25(4):155–167, 2011.
8. Di Stasio E, De Cristofaro R: The effect of shear stress on protein 23. Eby C: Pathogenesis and management of bleeding and thrombosis in
conformation: physical forces operating on biochemical systems: the case plasma cell dyscrasias. Br J Haematol 145(2):151–163, 2009.
of von Willebrand factor. Biophys Chem 153(1):1–8, 2010. 24. Malfait F, DePaepe A: Bleeding in the heritable connective tissue disor-
9. Hechler B, Gachet C: P2 receptors and platelet function. Purinergic ders: mechanisms, diagnosis and treatment. Blood Rev 23(5):191–197,
Signal 7(3):293–303, 2011. 2009.
10. Alberelli MA, De Candia E: Functional role of protease activated recep- 25. Komaromi I, Bagoly Z, Muszbek L: Factor XIII: novel structural and
tors in vascular biology. Vascul Pharmacol 62(2):72–81, 2014. functional aspects. J Thromb Haemost 9(1):9–20, 2011.
11. Bennett JS, Moore DT: Regulation of platelet beta 3 integrins. Haema- 26. Borissoff JI, Spronk HM, ten Cate H: The hemostatic system as a
tologica 95(7):1049–1051, 2010. modulator of atherosclerosis. N Engl J Med 364(18):1746–1760,
12. Versteeg HH, Heemskerk JW, Levi M, et al: New fundamentals in 2011.
hemostasis. Physiol Rev 93(1):327–358, 2013. 27. Watson T, Shantsila E, Lip GY: Mechanisms of thrombogenesis in atrial
13. Mann KG: Thrombin generation in hemorrhage control and vascular fibrillation: Virchow’s triad revisited. Lancet 373(9658):155–166, 2009.
occlusion. Circulation 124(2):225–235, 2011. 28. Tchaikovski SN, Rosing J: Mechanisms of estrogen-induced venous
14. Rao LV, Pendurthi UR: Regulation of tissue factor coagulant activity on thromboembolism. Thromb Res 126(1):5–11, 2010.
cell surfaces. J Thromb Haemost 10(11):2242–2253, 2012. 29. Anderson JA, Weitz JI: Hypercoagulable states. Crit Care Clin
15. Ahmad SS, London FS, Walsh PN: The assembly of the factor X-activating 27(4):933–952, 2011.
complex on activated human platelets. J Thromb Haemost 1(1):48–59,
2003.

You might also like