ArticlePDF AvailableLiterature Review

Shear Stress-Induced Activation of von Willebrand Factor and Cardiovascular Pathology

Authors:

Abstract and Figures

The von Willebrand factor (vWF) is a plasma protein that mediates platelet adhesion and leukocyte recruitment to vascular injury sites and carries coagulation factor VIII, a building block of the intrinsic pathway of coagulation. The presence of ultra-large multimers of vWF in the bloodstream is associated with spontaneous thrombosis, whereas its deficiency leads to bleeding. In cardiovascular pathology, the progression of the heart valve disease results in vWF deficiency and cryptogenic gastrointestinal bleeding. The association between higher plasma levels of vWF and thrombotic complications of coronary artery disease was described. Of note, it is not the plasma levels that are crucial for vWF hemostatic activity, but vWF activation, triggered by a rise in shear rates. vWF becomes highly reactive with platelets upon unfolding into a stretched conformation, at shear rates above the critical value (more than 5000 s−1), which might occur at sites of arterial stenosis and injury. The activation of vWF and its counterbalance by ADAMTS-13, the vWF-cleaving protease, might contribute to complications of cardiovascular diseases. In this review, we discuss vWF involvement in complications of cardiovascular diseases and possible diagnostic and treatment approaches.
Content may be subject to copyright.
International Journal of
Molecular Sciences
Review
Shear Stress-Induced Activation of von Willebrand
Factor and Cardiovascular Pathology
Sergey Okhota 1, Ivan Melnikov 1,2 , Yuliya Avtaeva 1, Sergey Kozlov 1
and Zufar Gabbasov 1, *
1National Medical Research Centre of Cardiology of the Ministry of Health of the Russian Federation,
15A, 3-rd Cherepkovskaya Street, 121552 Moscow, Russia; ae2007@mail.ru (S.O.); ivsgml@gmail.com (I.M.);
julia_94fs@mail.ru (Y.A.); bestofall@inbox.ru (S.K.)
2State Research Center of the Russian Federation—Institute of Biomedical Problems of Russian Academy
of Sciences, 76A, Khoroshevskoye Shosse, 123007 Moscow, Russia
*Correspondence: zufargabbasov@yandex.ru; Tel.: +7-(495)-414-62-79; Fax: +7-(495)-414-69-23
Received: 19 September 2020; Accepted: 20 October 2020; Published: 21 October 2020


Abstract:
The von Willebrand factor (vWF) is a plasma protein that mediates platelet adhesion
and leukocyte recruitment to vascular injury sites and carries coagulation factor VIII, a building
block of the intrinsic pathway of coagulation. The presence of ultra-large multimers of vWF in the
bloodstream is associated with spontaneous thrombosis, whereas its deficiency leads to bleeding.
In cardiovascular pathology, the progression of the heart valve disease results in vWF deficiency
and cryptogenic gastrointestinal bleeding. The association between higher plasma levels of vWF
and thrombotic complications of coronary artery disease was described. Of note, it is not the
plasma levels that are crucial for vWF hemostatic activity, but vWF activation, triggered by a
rise in shear rates. vWF becomes highly reactive with platelets upon unfolding into a stretched
conformation, at shear rates above the critical value (more than 5000 s
1
), which might occur at
sites of arterial stenosis and injury. The activation of vWF and its counterbalance by ADAMTS-13,
the vWF-cleaving protease, might contribute to complications of cardiovascular diseases. In this
review, we discuss vWF involvement in complications of cardiovascular diseases and possible
diagnostic and treatment approaches.
Keywords:
von Willebrand factor; ADAMTS-13; atherosclerosis; atherothrombosis; coronary artery
disease; Heyde’s syndrome
1. Introduction
The von Willebrand Factor (vWF) is a large multimeric glycoprotein, present in blood plasma,
endothelial cells, megakaryocytes, and platelets. It plays a major role in hemostasis, mediating platelet
adhesion to vascular injury sites. It also binds and protects coagulation factor VIII (FVIII) from
degradation [
1
]. Recent studies showed that vWF is also involved in inflammation, linking thrombosis
and inflammation. Inflammation can provoke thrombosis through the vWF-dependent pathway,
which includes endothelial activation, the secretion of vWF into the bloodstream, vWF activation and
interaction with platelets, and subsequent platelet adhesion to a vessel wall [
2
4
]. vWF multimers and
platelets, adhered to injured and activated endothelium, might serve as sites of leukocyte recruitment.
Altogether, this predisposes to the propagation of the inflammatory process. vWF is considered
a risk factor of arterial thrombosis and might contribute to the development of adverse events in
atherosclerosis and other cardiovascular diseases [58].
Int. J. Mol. Sci. 2020,21, 7804; doi:10.3390/ijms21207804 www.mdpi.com/journal/ijms
Int. J. Mol. Sci. 2020,21, 7804 2 of 18
2. Structure and Functions of VWF in Bloodstream
vWF is mainly produced in endothelial cells and stored in Weibel–Palade bodies—the storage
granules of endothelium, consisting of densely packed vWF multimers and P-selectin [
9
]. A small
amount of total vWF is produced in megakaryocytes and stored in the
α
-granules of platelets. The mature
vWF monomeric molecules form dimers through C-terminal disulphide bonds. Dimers compose the
basic vWF repeating structure, with a molecular mass of approximately 500 kDa [
10
]. Dimers are then
polymerized into ultra-large multimers through N-terminal disulphide bonds, with the size of stored
multimers ranging from 20 to 100 dimers [11].
vWF is constantly secreted from the Weibel–Palade bodies of endothelial cells into the bloodstream,
and from the
α
-granules of platelets upon activation. The pool of circulating vWF consists of
multimers of various sizes, ranging from a few dimers to high-molecular weight multimers (HMWM),
which contain 11–20 dimers. The former basically serve as FVIII carriers, while the latter present
the main hemostatically active force of vWF [
11
]. The more dimers that are in a vWF molecule,
the more hemostatically active it is. Upon secretion, large vWF multimers come into contact with
metalloproteinase ADAMTS-13, the vWF-cleaving protease. ADAMTS-13 regulates the size of
circulating vWF through proteolytic cleavage of its multimers [12].
vWF contains a variety of domains, which define the characteristics of the molecule: A1, A2, A3;
D assemblies: D1, D2, D’D3, D4; and the C-terminal cysteine knot (CTCK) (Figure 1) [
13
]. The A1
domain binds mainly glycoprotein (GP) Ib receptors of platelets (the only receptor on non-activated
platelets that has pronounced anity to vWF), and to a lesser extent, collagen types I, IV, VI, and heparin.
The A2 domain presents a binding cite for ADAMTS-13. The main collagen binding sites are located
on the A3 domain. The C1 domain can interact with activated platelets through the binding of
the IIb/IIIa receptors. The D’D3 assembly carries FVIII. The CTCK domain dimerizes vWF. All D
assemblies are involved in the assembly and disulfide linkage of vWF dimers into long tubules of
the Weibel–Palade bodies. [
9
,
13
]. vWF binds leukocytes through the interaction of the A1 domain
with P-selectin glycoprotein ligand-1 (PSGL-1), the D’D3, and the A1, A2, and A3 domains with
β2-integrins [14].
Int. J. Mol. Sci. 2020, 21, x FOR PEER REVIEW 2 of 18
2. Structure and Functions of VWF in Bloodstream
vWF is mainly produced in endothelial cells and stored in WeibelPalade bodiesthe storage
granules of endothelium, consisting of densely packed vWF multimers and P-selectin [9]. A small
amount of total vWF is produced in megakaryocytes and stored in the α-granules of platelets. The
mature vWF monomeric molecules form dimers through C-terminal disulphide bonds. Dimers
compose the basic vWF repeating structure, with a molecular mass of approximately 500 kDa [10].
Dimers are then polymerized into ultra-large multimers through N-terminal disulphide bonds, with
the size of stored multimers ranging from 20 to 100 dimers [11].
vWF is constantly secreted from the WeibelPalade bodies of endothelial cells into the
bloodstream, and from the α-granules of platelets upon activation. The pool of circulating vWF
consists of multimers of various sizes, ranging from a few dimers to high-molecular weight
multimers (HMWM), which contain 1120 dimers. The former basically serve as FVIII carriers, while
the latter present the main hemostatically active force of vWF [11]. The more dimers that are in a vWF
molecule, the more hemostatically active it is. Upon secretion, large vWF multimers come into contact
with metalloproteinase ADAMTS-13, the vWF-cleaving protease. ADAMTS-13 regulates the size of
circulating vWF through proteolytic cleavage of its multimers [12].
vWF contains a variety of domains, which define the characteristics of the molecule: A1, A2, A3;
D assemblies: D1, D2, D’D3, D4; and the C-terminal cysteine knot (CTCK) (Figure 1) [13]. The A1
domain binds mainly glycoprotein (GP) Ib receptors of platelets (the only receptor on non-activated
platelets that has pronounced affinity to vWF), and to a lesser extent, collagen types I, IV, VI, and
heparin. The A2 domain presents a binding cite for ADAMTS-13. The main collagen binding sites are
located on the A3 domain. The C1 domain can interact with activated platelets through the binding
of the IIb/IIIa receptors. The DD3 assembly carries FVIII. The CTCK domain dimerizes vWF. All D
assemblies are involved in the assembly and disulfide linkage of vWF dimers into long tubules of the
WeibelPalade bodies. [9,13]. vWF binds leukocytes through the interaction of the A1 domain with
P-selectin glycoprotein ligand-1 (PSGL-1), the DD3, and the A1, A2, and A3 domains with β2-
integrins [14].
Figure 1. (A) A schematic representation of a von Willebrand factor thread; and (B) a von Willebrand
factor dimer. The binding sites for ADAMTS-13, platelets, leukocytes, collagen, and heparin are
indicated.
vWF exists in the bloodstream in one of two conformationsglobular and unfolded [15]. vWF
conformation depends on the shear rate of blood flow in vessels. The shear rate is the rate of change
of velocity, at which one layer of fluid passes over another, and is measured in inverse, or reciprocal,
seconds (s−1). This parameter is used to estimate flow of liquids in tubes, which is important for the
Figure 1.
(
A
) A schematic representation of a von Willebrand factor thread; and (
B
) a von Willebrand factor
dimer. The binding sites for ADAMTS-13, platelets, leukocytes, collagen, and heparin are indicated.
vWF exists in the bloodstream in one of two conformations—globular and unfolded [
15
].
vWF conformation depends on the shear rate of blood flow in vessels. The shear rate is the rate
of change of velocity, at which one layer of fluid passes over another, and is measured in inverse,
Int. J. Mol. Sci. 2020,21, 7804 3 of 18
or reciprocal, seconds (s
1
). This parameter is used to estimate flow of liquids in tubes, which is
important for the understanding of vWF activation. Under conditions of low shear rates (e.g., in veins,
where shear rates are 15–200 s
1
, or in large arteries, where they are 300–800 s
1
), vWF remains in a
globular shape, which hides the binding sites, and does not interact with the circulating platelets [
16
].
In the case of vessel injury and under high shear rates (e.g., in intact small arteries and arterioles, shear
rates are 450–1600 s
1
, but at sites of advanced atherosclerosis, shear rates might reach up to 11,000 s
1
and even higher), the vWF shape unfolds, and opens binding sites, in particular for glycoprotein (GP)
Ib receptors of platelets.
A threshold value of shear rate, critical for unfolding and the activation of vWF, is estimated at
5000 s
1
[
15
]. Uniform shear rates occur in straight channels, but in pathological conditions in stenotic
or injured vessels, elongational flows occur, where the molecules are subjected to shear gradients.
The vasoconstriction of injured vessels and stenoses of arterial lumen by atherosclerotic plaques
create two zones of elongational flow. Flow and shear rate accelerate at the inflow and decelerate
at the outflow of a lesion. At the inflow, elongation occurs parallel to the flow direction. At the
outflow, elongation occurs perpendicular to the flow direction because the streamlines diverge. As the
adjacent shear layers have dierent velocities, large molecules, such as vWF multimers, are subjected
to the rotational and elongational components of the flow. In the straight sections of a blood vessel,
the rotational and elongational components are balanced. However, at the inflow and the outflow of a
lesion, the equilibrium is disturbed. At sites of vasoconstriction and stenoses, thrombus formation is
observed at the outflow zone of a lesion (Figure 2) [
9
]. Elongation flows are predicted to unfold vWF at
rates of two orders of magnitude below the corresponding pure shear rate values [
17
]. vWF-dependent
thrombus formation was observed at the outflow region of an
in vitro
model of stenosis at the inflow
shear rates of 600, 1000, and 2000 s1[18].
Int. J. Mol. Sci. 2020, 21, x FOR PEER REVIEW 3 of 18
understanding of vWF activation. Under conditions of low shear rates (e.g., in veins, where shear
rates are 15200 s−1, or in large arteries, where they are 300800 s−1), vWF remains in a globular shape,
which hides the binding sites, and does not interact with the circulating platelets [16]. In the case of
vessel injury and under high shear rates (e.g., in intact small arteries and arterioles, shear rates are
4501600 s−1, but at sites of advanced atherosclerosis, shear rates might reach up to 11,000 s−1 and even
higher), the vWF shape unfolds, and opens binding sites, in particular for glycoprotein (GP) Ib
receptors of platelets.
A threshold value of shear rate, critical for unfolding and the activation of vWF, is estimated at
5000 s−1 [15]. Uniform shear rates occur in straight channels, but in pathological conditions in stenotic
or injured vessels, elongational flows occur, where the molecules are subjected to shear gradients.
The vasoconstriction of injured vessels and stenoses of arterial lumen by atherosclerotic plaques
create two zones of elongational flow. Flow and shear rate accelerate at the inflow and decelerate at
the outflow of a lesion. At the inflow, elongation occurs parallel to the flow direction. At the outflow,
elongation occurs perpendicular to the flow direction because the streamlines diverge. As the
adjacent shear layers have different velocities, large molecules, such as vWF multimers, are subjected
to the rotational and elongational components of the flow. In the straight sections of a blood vessel,
the rotational and elongational components are balanced. However, at the inflow and the outflow of
a lesion, the equilibrium is disturbed. At sites of vasoconstriction and stenoses, thrombus formation
is observed at the outflow zone of a lesion (Figure 2) [9]. Elongation flows are predicted to unfold
vWF at rates of two orders of magnitude below the corresponding pure shear rate values [17]. vWF-
dependent thrombus formation was observed at the outflow region of an in vitro model of stenosis
at the inflow shear rates of 600, 1000, and 2000 s−1 [18].
Figure 2. Shear rate-induced activation of the von Willebrand factor at the site of atherosclerotic
narrowing of a vessel.
Unfolded by high shear rates, the vWF threads can self-associate and form long strands and
web-like structures, further stimulating platelet adhesion [19]. The ability of unfolded vWF to self-
associate under high-shear rates was studied in vitro in endothelialized microvessels. Smaller vessels,
Figure 2.
Shear rate-induced activation of the von Willebrand factor at the site of atherosclerotic
narrowing of a vessel.
Int. J. Mol. Sci. 2020,21, 7804 4 of 18
Unfolded by high shear rates, the vWF threads can self-associate and form long strands and web-like
structures, further stimulating platelet adhesion [
19
]. The ability of unfolded vWF to self-associate
under high-shear rates was studied
in vitro
in endothelialized microvessels. Smaller vessels, turns,
bifurcations, flow acceleration predisposed to thickening, and elongation of vWF strands. In regions
with complex flow, the vWF strands tended to form web-like structures. At the same time, at sites of
low shear rates, the vWF remained in a globular conformation and no vWF strands were visible [
19
].
Schneider et al. also reported spider web-like network formation by vWF, under high shear rates [
15
].
Zhang et al., demonstrated that vWF association was dependent on the A2 domain under shear rate of
9600 s
1
. Hence, the A2 domain might have an overlapping role, both in proteolysis and self-association
of vWF [20].
The role of platelet vWF in hemostasis is not clearly established, and the studies here are
quite sparse. A study on the vWF-knockout mice showed that replenishing of vWF in platelets after
bone marrow transplantation from wild-type mice, resulted in partial correction of bleeding time and
reduction of blood loss volume [
21
]. Recently, a study showed that chimeric mice with only platelet
vWF had a bleeding time comparable to that of the vWF-knockout mice [
22
]. Platelet vWF did not
contribute to thrombus formation in a model of carotid artery injury. On the other hand, after ischemic
brain injury, the mice with only platelet vWF and without plasma vWF, had a cerebral infarction size
similar to the wild-type mice. Platelet vWF mediated ischemic brain injury in a GPIb-dependent
mechanism and significantly contributed to intracerebral thrombosis formation [22].
Upon the unfolding of vWF multimers, FVIII is exposed on D’D3 domains [
1
]. FVIII participates
immediately in the intrinsic coagulation pathway, interacting with factor IXa as a cofactor to form the
intrinsic tenase (enzyme complex that cleaves inactive coagulation factor X into active Xa). FVIII is
essential for normal hemostasis—its deficiency leads to hemophilia A. vWF acts as a carrier of
FVIII in circulation. In the absence of vWF, FVIII is unstable and exposed to rapid degradation.
vWF significantly elongates its half-life and protects FVIII from proteolysis, eventually delivering it to
sites of vessel injury [1].
Unraveling, vWF faces its antagonist, ADAMTS-13 [
9
]. It is a member of the ADAMTS
(A Disintegrin And Metalloprotease with ThromboSpondin type 1 repeats) family of proteolytic
enzymes. ADAMTS-13 was first isolated in 2001, though the presence of Ca
2+
-dependent vWF-cleaving
protease was predicted earlier [
23
]. Human ADAMTS-13 mRNA and protein synthesis is localized
exclusively to hepatic stellate cells [
24
]. The plasma levels of ADAMTS-13 show negative correlation
with the plasma levels of vWF [
25
]. In the absence of vWF in the bloodstream, i.e., in patients with von
Willebrand disease (vWD) type 3, the plasma levels of ADAMTS-13 are 30% higher than in healthy
volunteers. On the contrary, after the administration of 1-deamino-8-D-arginine vasopressin (DDAVP),
which stimulates the release of vWF from endothelial Weibel–Palade bodies, the plasma levels of
ADAMTS-13 showed a 20% reduction in healthy volunteers [
25
]. The negative correlation is likely to
occur because of consumption in the cleavage of vWF. ADAMTS-13 circulates in the bloodstream as
an active enzyme, but vWF in the globular conformation remains resistant to proteolysis. Once vWF
unfolds, it exposes the binding sites for ADAMTS-13 on the A2 domain. The ensuing interaction
with ADAMTS-13 leads to the cleavage of vWF multimers between tyrosine in the 1605 position and
methionine in the 1606 position, which shortens the multimers and decreases the hemostatic activity of
vWF [16,26].
The growing evidence shows that vWF is an important mediator of vascular inflammation [
27
].
A murine model of acute peritonitis showed that vWF plays an important role in leukocyte
recruitment and extravasation into the site of inflammation. Notably, leukocyte extravasation
requires vWF-platelet binding, as inhibition of the platelet GPIb receptors prevented vWF-mediated
leukocyte extravasation [
28
]. A murine model of immune complex-mediated vasculitis and irritant
contact dermatitis showed that the interference with vWF activity with vWF-blocking antibody
led to a substantial decrease in vWF-mediated leukocyte recruitment and reduction of cutaneous
inflammatory response. In this study, the vWF-blocking antibody did not interfere with the GPIb
Int. J. Mol. Sci. 2020,21, 7804 5 of 18
domain [
29
]. Another murine model with the same pathology but with the use of the antibody targeting
the A1 domain showed comparable results [
30
]. A study comparing features of acute inflammation
after focal cerebral ischemia in vWF-deficient and ADAMTS-13-deficient mice, showed that the
vWF-deficient mice had a smaller injury size, reduced cytokine release and neutrophil infiltration of
the injury site, compared to the ADAMTS-13-deficient mice [
31
]. A myocardial ischemia/reperfusion
study in ADAMTS-13-knockout mice showed that infusion of recombinant ADAMTS-13 substantially
reduced myocardial apoptosis, troponin-I release, and resulted in a 9-fold reduction in the number of
neutrophils infiltrating the ischemic zone [32].
3. Diagnostic Tests for the von Willebrand Factor Deficiency and Dysfunction
Studies of acquired von Willebrand syndrome in aortic stenosis and obstructive hypertrophic
cardiomyopathy revealed that, despite the clinical manifestation of bleeding, plasma levels of vWF
and ristocetin cofactor assay remained at normal values [
33
]. The plasma levels of vWF show mass
concentration of all vWF multimers, from dimers to HMWM. The proteolyzed HMWM of vWF are
cleaved into smaller multimers and dimers. This significantly aects the hemostatic function of vWF,
but not its mass concentration. Thus, measuring the plasma levels of vWF shows the amount of protein
in the plasma, but not its functional status. The plasma levels of vWF depend on the blood group
of a person [
34
]. Variations of the plasma levels of vWF between blood groups might complicate
distinguishing healthy persons with low vWF from mild cases of vWD [
35
]. Unlike oligosaccharides of
other plasma glycoproteins, the vWF structure includes oligosaccharides of the ABO blood groups.
Persons in the O blood group have significantly lower levels of vWF than persons in other blood
groups [
36
]. A study, comprising 1117 blood donors showed that the plasma levels of vWF were the
lowest in the O blood group (74.8 IU/dL), higher in the A blood group (105.9 IU/dL), even higher in the
B blood group (116.9 IU/dL), and the highest in the AB blood group (123.3 IU/dL) [
37
]. In ristocetin
cofactor assay, ristocetin was used to bind the GPIb receptors of platelets to the A1 domain of
vWF [
38
]. The assay was performed under very low shear rate conditions, when the vWF multimers
remained folded. Thus, the assay reflected the presence of vWF in a sample per se, but not the
physiological hemostatic function of vWF. It also had a low sensitivity to the loss of HMWM of vWF.
Hence, it was useful in the diagnosis of vWD, especially in the case of severe vWF deficiency (
e.g., in type
3 vWD, in which vWF was almost or completely absent in the blood), but provided little diagnostic
information concerning the actual hemostatic function of vWF. Significant interlaboratory variations
in test results existed for both of these assays [
38
]. vWF multimer analysis by the electrophoresis
of vWF on agarose gels remained the main assay of qualitative deficiency of vWF [
10
]. The assay
had excellent sensitivity for the loss of HMWM of vWF. The inclusion of the luminescent methods
gave it the power to detect the presence of vWF in samples with a concentration of less than 1 IU/dL.
This allowed dierentiation between homozygous type 2 and type 3 vWD. vWF multimer analysis
is crucially important for subtyping of type 2 vWD [
10
]. It is also very useful in diagnostic of
HMWM of vWF deficiency in acquired von Willebrand syndrome, due to the heart valve disease [
39
].
However, the assay placed high demands on the technical competence of the laboratory personnel
and was unsuitable for standardization [
35
]. Therefore, its introduction to routine diagnostics in
cardiovascular disease is challenging. vWF collagen binding assay might be used as a substitution
for the detection of HMWM of vWF [
40
]. Thus, the broad panel of tests is needed to diagnose vWD
and its type. To simplify testing (e.g., in screening for bleeding disorders or in the emergency setting),
a screening tool that can quickly exclude platelet defects and vWD was introduced. The Platelet
Function Analyzer-100 (PFA-100) is an easy to use device, which utilizes whole blood and requires
only 5 min to complete a test [
41
]. Currently, this is the only commercially available system that
evaluates primary hemostasis under high shear rate conditions. PFA-100 pumps a whole blood sample
through a narrow orifice in a membrane, covered with collagen and platelet activator agents, such as
adenosine diphosphate (ADP) or epinephrine. The system measures time from the start of a sample
pumping to the closure of the orifice (the closure time). The shear rates in the orifice might reach
Int. J. Mol. Sci. 2020,21, 7804 6 of 18
5000–6000 s
1
, which is sucient to activate vWF. However, PFA-100 possesses a drawback concerning
vWF activation. The system is designed to assess primary hemostasis, not exclusively vWF hemostatic
function. The closure time is dependent on interactions between platelets and other blood cells,
not vWF activation alone. Its sensitivity varies, depending on the severity of vWF deficiency and
platelet defects, from moderate in mild cases to 100% sensitivity in the case of complete absence of
vWF in blood [
41
,
42
]. Thus, currently there is no widely available assay that exclusively measures
vWF shear rate-dependent hemostatic function. Experimental assays and devices, such as microfluidic
chambers, are created by some research laboratories to study dierent aspects of hemostasis, including
vWF functions. However, these devices are cumbersome and not standardized. Therefore, currently,
these devices fall short of satisfying clinical demands [43].
4. Diseases, Associated with von Willebrand Factor and ADAMTS-13 Dysfunction
vWF plays a major role in coagulation, and its deficiency or dysfunction predisposes one to
bleeding. A genetic disorder, caused by partial or total deficiency or structural aberrations of vWF
molecules, is called vWD [
35
]. This is one of the most widespread hemostasis disorders with an
incidence of around 1:100 persons. It is characterized by nasal and gingival bleeding, hemarthrosis,
muscular and subcutaneous hematomas, menorrhagia, and prolonged bleeding in traumas.
Desmopressin, which stimulates the release of vWF from endothelium, is the main medication
for the mild forms of vWD. In severe cases, the only choice is regular vWF-rich plasma transfusions [
11
].
Acquired von Willebrand syndrome is a rare disorder that occurs due to lymphoproliferative
(e.g., chronic lymphocytic leukemia), myeloproliferative (e.g., thrombocythemia), cardiovascular
(e.g., aortic stenosis) and immunological disorders (e.g., hypothyroidism), which provoke the qualitative
and quantitative deficiency of vWF [44].
On the contrary, increased concentrations of ultra-large multimers of vWF lead to thrombotic
thrombocytopenic purpura (TTP), which develops due to a deficiency of ADAMTS-13 [
45
].
ADAMTS-13 deficiency causes an unrestricted presence of ultra-large multimers of vWF in the
bloodstream, which might spontaneously interact with platelets and provoke thrombosis in multiple
small vessels. This results in thrombocytopenia with purpura, hemolytic anemia, and multiple organ
damage, due to microvascular thrombosis—acute renal, heart and neurological damage, mental
derangement, and fever. TTP can develop either due to genetic anomalies (congenital TTP) or
production of autoantibodies to ADAMTS-13 (acquired TTP), which interfere with its function or
enhance clearance. The determination of the ADAMTS-13 function is essential in TTP diagnosis.
TTP usually manifests when the ADAMTS-13 levels fall below 10%, which is below the detection
limit of many assays. As soon as ADAMTS-13 activity becomes detectable (i.e., higher than 10%),
TTP symptoms resolve. Dierentiation between congenital and acquired TTP is vitally important, as
treatment of these life-threatening conditions significantly dier. The diagnosis of congenital TTP
is built on the detection of pronounced fall in ADAMTS-13 activity, ruling out autoantibodies to
ADAMTS-13, and analyzing the ADAMTS-13 gene. The diagnosis of acquired TTP requires the lack of
ADAMTS-13 activity and the detection of autoantibodies to ADAMTS-13. Patients with congenital
TTP respond well to the plasma exchange therapy, which improves survival rates from 10% to 80–90%.
During the plasma exchange, ultra-large multimers of vWF, immune complexes, and autoantibodies are
removed. Infusions of donor plasma with normal content of ADAMTS-13 are also eective, and used
in combination with the plasma exchange therapy. Patients with acquired TTP respond to normal
plasma infusions poorly. Suppression of autoantibody production with corticosteroids, destruction of
CD20-posititve B-lymphocytes with rituximab, or the use of other immunosuppressive agents is
considered to be eective in acquired TTP. Splenectomy is eective, but reserved for refractory cases of
acquired TTP [
46
]. Recently, caplacizumab, an immunoglobulin fragment that targets the A1 domain
of vWF, and prevents its interaction with platelet GPIb receptors, was introduced in addition to plasma
exchange therapy [47].
Int. J. Mol. Sci. 2020,21, 7804 7 of 18
5. Acquired von Willebrand Syndrome in Heart Valve Disease and Hypertrophic Cardiomyopathy
The association between aortic stenosis and cryptogenic gastrointestinal bleeding was first
described in 1958 in a letter by E.C. Heyde [
48
]. In 1992, Warkentin et al. were the first to suggest
a link between the loss of HMWM of vWF that develops due to aortic stenosis or hypertrophic
cardiomyopathy and bleeding from gastrointestinal angiodysplasia [
49
]. In 2002, Warkentin et al.,
was also the first to provide a rationale for this association [
50
]. They reported two cases of severe
aortic stenosis with concomitant gastrointestinal bleeding that ceased after successful replacement of
the aortic valve. Notably, prior to the platelet count operation, activated partial-thromboplastin time,
the plasma level of FVIII, the plasma levels of vWF, and the ristocetin cofactor assay values were normal,
whereas the loss of HMWM of vWF was severe. After the valve replacement, the percentage of HMWM
recovered and remained normal during a 10-year follow up [
50
]. Acquired von Willebrand syndrome
in cardiovascular disease develops due to aortic stenosis, left ventricle outflow tract obstruction in
hypertrophic cardiomyopathy, and aortic and mitral regurgitation [
8
]. In severe aortic stenosis, a
steep increase in shear rates in the aortic orifice and left ventricular outflow tract occur. HMWM of
vWF, which pass through the narrowing aortic valve, undergo activation and subsequent proteolysis
by ADAMTS-13. Additionally, lower pulse pressure, which is observed in aortic stenosis, contributes to
the decreased secretion of HMWM of vWF from the endothelium [
8
]. In severe aortic stenosis,
HMWM of vWF might decline to approximately 1/2 of the normal levels [
51
]. Panzer et al., reported
a study of 47 patients with severe aortic stenosis, who were subjected to aortic valve replacement.
HMWM of vWF were depleted in all patients before valve replacement and became normal in most
patients. PFA-100 closure time in collagen-ADP cartridges were significantly prolonged at the baseline,
and normalized after the treatment. Using the cone and plate analyzer, the authors showed that
the loss of HMWM of vWF aects platelet adhesion and, to a larger extent, ADP-induced platelet
aggregation [
52
]. The quantitative deficiency of HMWM of vWF leads to the acquired von Willebrand
syndrome type 2A. Its clinical manifestation in patients with heart valve disease is called Heyde’s
syndrome [
53
]. It is characterized by cryptogenic gastrointestinal bleedings from submucous arterial
malformations, developing together with the progression of aortic stenosis and its cardiovascular
symptoms (such as fatigue and angina). Bleeding from arterial malformations is thought to be provoked
by increased shear rates, due to the complex structure of vessels, which cause further consumption of
vWF [
53
]. Patients with severe aortic stenosis also report skin and mucosal bleeding. In a study of
50 patients with aortic stenosis, skin or mucosal bleeding was reported by 21% of participants [
33
].
Primary hemostasis, measured by PFA-100 was significantly prolonged; the percentage of HMWM and
the collagen-binding activity of vWF were reduced in patients with severe aortic stenosis. The plasma
levels of vWF were normal in all patients. One day after surgical treatment, PFA-100 values and
the percentage of HMWM of vWF were completely corrected; however, after 6 months, these values
deteriorated, primarily in patients with prosthesis mismatch [
33
]. In two other studies of 183 and
21 patients with severe aortic stenosis, undergoing valve replacement, the same pattern was observed.
PFA-100 closure time was prolonged and the HMWM of vWF reduced at the baseline; they normalized
after successful valve replacement [
51
,
54
]. The volume of blood that is aected by high shear rates
is a crucial component in the development of Heyde’s syndrome, in which the whole volume of
circulating blood is aected. For example, in patients with severe coronary or peripheral atherosclerosis,
vWF deficiency does not occur because blood volume is aected only partially. The treatment of aortic
stenosis with valve replacement results in fast recovery and the cessation of bleeding. The normalization
of shear rates and pulse pressure leads to an increase in the plasma level of functional HMWM of vWF
within hours [
55
]. A prospective study with an 18-month follow-up of patients treated with aortic
valve replacement, and several other studies with follow-up from 2 weeks up to 6 months, showed that,
after recovery, the plasma levels of HMWM of vWF did not drop again [
51
]. However, if significant
aortic regurgitation and paravalvular leak develop after aortic valve replacement, the plasma levels
of HMWM of vWF might not recover [
54
]. A similar development pathway of the acquired von
Willebrand syndrome was also reported in patients with aortic or mitral valve regurgitation [56,57].
Int. J. Mol. Sci. 2020,21, 7804 8 of 18
In obstructive hypertrophic cardiomyopathy, the obstructed left ventricle outflow tract
predisposes the proteolysis of HMWM of vWF in a manner similar to aortic stenosis. A study by
Blackshear et al., included five patients with symptomatic obstructive hypertrophic cardiomyopathy [
58
].
Spontaneous gastrointestinal, mucosal, or excessive postsurgical bleeding was observed in all patients.
The plasma levels of vWF and ristocetin cofactor assay values were within normal limits, whereas
electrophoresis revealed loss of HMWM and excess of low-molecular weight multimers of vWF.
After surgical septal myectomy, bleeding resolved, and HMWM were restored to normal levels in all
patients [
58
]. In another study of 28 patients with obstructive hypertrophic cardiomyopathy, plasma
levels of vWF were normal in all patients [
59
]. PFA-100 closure time was significantly prolonged in all
but one patient. Additionally, all patients had a substantial reduction in HMWM of vWF. A strong
correlation of closure time in the PFA-100 tests and reduction in HMWM of vWF with peak gradients,
measured by ultrasonography, was observed. The mean peak gradient, sucient for the impairment
of vWF function and the reduction in HMWM, was estimated at 15 mm Hg [59].
The use of left ventricle assist devices (LVADs) might also lead to acquired von Willebrand
syndrome [
60
]. LVADs provide circulatory support for patients with end-stage heart failure, either
as a bridge or destination therapy. vWF might immobilize on the biomaterial surfaces of LVAD and
undergo cleavage at sites of high shear rates, which might occur in the LVAD system. This might
lead to the development of LVAD-associated complications, such as pump thrombosis and bleeding;
in particular, gastrointestinal bleeding from arteriovenous malformations [60].
6. von Willebrand Factor and Coronary Artery Disease
The relevance of vWF in platelet adhesion and thrombus formation under high shear rates
was shown in a study on pigs [
61
]. In this study, 8 healthy pigs and 6 pigs with vWD were
fed a high-cholesterol diet for 24 weeks with the assessment of coronary arteries for the presence
of atherosclerosis. The diet led to severe hypercholesterolemia (7–39 mMol/L). Coronary atherosclerosis
developed in both groups and was detected by histology in all but one healthy and all vWD pigs.
Left anterior descending coronary and carotid arteries were clamped to produce stenotic segments,
which were then injured. Occlusive thrombosis occurred in all stenotic segments in phenotypically
normal pigs, while it did not occur in the vWD pigs [61].
A study on mice showed the influence of ADAMTS-13 and vWF deficiency on a myocardial
infarction (MI), induced by the ligation of the left anterior descending coronary artery [
62
]. The infarct
size was significantly larger in homozygous ADAMTS-13-deficient mice (22.2
±
1.1%), compared to
heterozygous ADAMTS-13-deficient mice (17.3
±
0.8%) and wild-type mice (16.9
±
1.2%), which suggests
that a level of approximately 50% of ADAMTS-13 in plasma is sucient to prevent aggravated MI.
The infarct size was markedly reduced in the vWF-deficient mice (7.3
±
0.7%), compared to the
wild-type mice (18.6
±
1.3%). A group comprising ADAMTS-13 deficient and wild-type mice was
treated with a polyclonal antibody to vWF. In this group, the infarct size in the wild-type and in the
ADAMTS-13-deficient mice (8.5
±
0.7% and 8.2
±
1.3%, respectively) was significantly smaller than in
the control wild-type mice treated with a nonspecific antibody (17.48 ±0.7%) [62].
CAD in patients with vWD was observed less frequently. In a register study comprising 7556 cases
related to vWD and 19,918,970 cases unrelated to vWD, CAD was less common in patients with vWD
(15.0%) than in patients without vWD (26.0%). After multivariable logistic regression analysis with
adjustment for the main risk factors of CAD, the probability of CAD in patients with vWD remained
lower than in patients without it (OR 0.85; 95% CI 0.79–0.92) [63].
The plasma levels of vWF dier in healthy persons and patients with CAD. In 110 patients with a
mean age of 58
±
20 years with CAD, the plasma level of vWF was 141.78
±
20.53 IU/dL, whereas in a
control group of healthy volunteers it was 111.95
±
17.15 IU/dL [
64
]. A prospective multicenter study
comprising 3043 patients with stable angina or previous MI, showed that the higher plasma levels of
vWF correlated with an 8.5% increase in the rate of MI and sudden cardiac death [
65
]. A meta-analysis
of the prospective Reykjavik study, comprising 1925 persons who had primary nonfatal MI or died
Int. J. Mol. Sci. 2020,21, 7804 9 of 18
of CAD during a follow-up (median 19.4 years), and 3616 controls, showed that the baseline plasma
levels of vWF were higher in patients with CAD than in the control group [
66
]. The ENTIRE-TIMI 23
study, comprising 314 patients with ST-elevation myocardial infarction (STEMI) in whom the plasma
levels of vWF were measured before and 48–72 h after fibrinolysis, showed that an increase in vWF
levels after fibrinolysis was associated with the higher mortality and MI rate in 30 days (
11.2% vs. 4.1%
,
respectively) [
67
]. In 123 patients who had MI before the age of 70, plasma levels of vWF were measured
3 months after MI. A 4.9-year follow up showed that higher concentrations of vWF were independently
associated with recurrent MI and mortality [68].
A study, comprising 1026 patients with confirmed first STEMI and 652 healthy controls, showed
that the plasma levels of vWF were nearly 1.5-fold higher in patients with STEMI, than in healthy
controls (median 378.2 vs. 264.4 ng/mL, respectively). The plasma levels of ADAMTS-13 were lower in
patients with STEMI, than in the healthy controls (median 90% vs. 97%, respectively) [
69
]. In another
study, the plasma levels of vWF in 41 patient of mean age 68
±
23 years, hospitalized within 72 h
after the onset of MI, were significantly higher (2151
±
97 mU/mL) than in 33 patients with stable
angina and 90% narrowing of a major coronary artery (1445
±
93 mU/mL), who had angina attack
within 4 weeks before the study, or 30 patients with chest pain syndrome without hemodynamically
significant stenosis or coronary spasm (1425
±
76 mU/mL), in whom the vWF levels did not dier
significantly [
70
]. On the other hand, the plasma levels of ADAMTS-13 were significantly lower in
patients with acute MI (
799 ±29 mU/mL
) than in patients with stable angina (996
±
31 mU/mL) or
patients without hemodynamically significant stenosis or vasospasm (967
±
31 mU/mL). The enzymatic
activity of ADAMTS-13 was also lower in patients with acute MI (768
±
27 mU/mL) than in patients
with stable angina (893
±
27 mU/mL) or patients without hemodynamically significant stenosis or
vasospasm (936
±
29 mU/mL). [
70
]. In the GLAMIS study of 466 patients of mean age 55.1
±
7.45 years
with acute MI and 484 healthy controls, no correlation between ADAMTS-13 and vWF levels was found
in the healthy controls [
71
]. On the other hand, the plasma levels of vWF correlated positively and
the plasma levels of ADAMTS-13 correlated negatively with the risk of MI. After adjustment for the
main cardiovascular risk factors, each 60 IU/dL increase in plasma levels of vWF was expected to raise
the risk of MI by about 35%. Each 33% increase in the plasma levels of ADAMTS-13 was expected to
decrease the risk of MI by about 27% [
71
]. The SMILE study, which included 650 men, 18–70-years-old,
with stable CAD, who had an MI event at least 6 months before the study, and 646 healthy men,
revealed no clear association between ADAMTS-13 and the vWF plasma levels, measured more than
6 months after the onset of MI [
72
]. There was also no significant dierence between the ADAMTS-13
levels in men with stable CAD and the healthy controls (mean levels 101% and 100%, respectively).
Neither were the plasma levels of vWF dierent between the groups (mean 138% in the CAD group
and 135% in controls) [
72
]. The prospective PRIME study, comprising nearly 10,000 healthy men,
among whom 296 developed CAD during the 5 years of follow-up (158 persons with MI and 142 with
stable and unstable angina), showed that the baseline plasma levels of vWF were significantly higher in
men who developed MI (129.2
±
53.1 IU/dL) compared to the healthy controls (115.9
±
41.8 IU/dL) [
73
].
The relative risk of MI development was 3.34 in patients with plasma levels of vWF in the 4th quartile,
compared to 1.0 in persons with vWF plasma levels in the 1st quartile. Stable and unstable angina
incidence did not correlate with the plasma levels of vWF [73].
A study on rats attempted to determine time span for vWF plasma level recovery after STEMI [
74
].
The study comprised 57 male rats subjected to ligation of the left anterior descending artery, 2 mm
away from bifurcation, which provoked ECG-verified STEMI and myocardial fibrosis, on histological
examination. Rats were randomized into four groups—in the first, blood was taken from the coronary
sinus and the inferior vena cava, 1 h after the onset of MI; in the second, 24 h after MI; in the third,
7 days after MI; the fourth was used as a control. The plasma levels of vWF, obtained from the coronary
sinus, increased 1.31-fold after 1 h, and 0.88-fold, 24 h later. They decreased to normal levels on the
seventh day. In the inferior vena cava, the plasma levels of vWF were 0.37-fold higher 1 h after the
onset of MI, 0.18-fold higher 24 h later, and decreased to normal levels on the seventh day [74].
Int. J. Mol. Sci. 2020,21, 7804 10 of 18
7. Inflammatory and Stress Stimuli as a Possible Cause of Elevation in Plasma von Willebrand
Factor in Coronary Artery Disease
There are several reasons for the increase in plasma levels of vWF in CAD and, in particular, MI.
In a 5-year follow-up of 1592 persons 55–74 years old, smoking was associated with higher levels
of vWF and a higher risk of atherosclerosis development [
75
]. In a study of 2459 patients who had
nonfatal MI or died of CAD during the 12-year follow-up, higher plasma levels of vWF were associated
with smoking and older age [76].
In another study, 73 men of age 59
±
11 years, with stable CAD and in sinus rhythm, and 35 healthy
volunteers, underwent 24-h ambulatory blood pressure monitoring [
77
]. Patients were divided into
four groups: 1—with high pulse pressure; 2—with low pulse pressure; 3—dippers; and 4—non-dippers.
The results of the study showed that all patient groups had higher plasma levels of vWF compared
to controls (mean 197
±
58 vs. 120
±
18 IU/dL, respectively). The highest plasma levels of vWF were
detected in the high pulse pressure (219
±
58 IU/dL) and non-dipper (222
±
55 IU/dL) groups [
77
]. In a
study of 178 patients, 54
±
15 years-old with arterial hypertension, who had blood pressure higher than
160/90 mmHg on two visits to a physician, and 47 normotensive healthy controls, the plasma levels of
vWF were significantly higher in the hypertensive group, than in controls (mean 113 vs. 98 IU/dL,
respectively) [78].
Another prospective study of 631 patients 50–75 years-old showed that increased vWF
plasma levels correlated with cardiovascular and all-cause mortality, in patients with and without
diabetes mellitus. The increase in plasma levels of vWF significantly correlated with older age,
higher fasting glucose levels, glycated hemoglobin, body mass index, and systolic arterial pressure [
79
].
A study of 94 patients with non-insulin-dependent diabetes mellitus showed correlation between
albuminuria and increased plasma levels of vWF [
80
]. In the ASCET study, age, smoking, and diabetes
mellitus were associated with elevated vWF in plasma [
81
]. In the ENTIRE-TIMI 23 sub-study,
the reduced success of thrombolysis (estimated by the thrombolysis in myocardial infarction (TIMI)
flow grade and the corrected TIMI frame count) positively correlated with the plasma levels of VWF.
The plasma levels of vWF in the upper quartile were associated with a higher incidence of death within
30 days, compared with the lower quartile (11.2% vs. 4.1%, respectively) [67].
Elevated plasma vWF was observed in patients with systemic inflammatory diseases. In a
study of 113 patients with systemic inflammatory diseases (40 with rheumatoid arthritis, 38 with
systemic scleroderma, 35 with systemic vasculitis), the plasma levels of VWF were significantly
elevated, compared to 80 healthy controls [
82
]. Of note, in patients with chronic systemic inflammation,
experiencing exacerbation in the form of serositis, plasma vWF levels, and ristocetin cofactor assay
values were increased, compared to healthy controls, but were unrelated to clinical manifestations of
the disease, including thrombotic complications [83].
One of the reasons for elevated plasma vWF is the polymorphism of Thr789Ala allele in the vWF
gene, which is an independent predictor of CAD development [
84
]. Additionally, the drug-induced
elevation of plasma vWF might occur due to the intake of diuretics, digoxin, heparin, and oral
anticoagulants [
64
]. On the other hand, the administration of enoxaparin decreases plasma vWF levels,
compared to heparin [67].
Plasma vWF elevation might occur due to endothelial damage. Taking into consideration that
vWF is predominantly secreted by the endothelium, elevated plasma vWF might be considered to be a
marker of endothelial dysfunction. Endothelial dysfunction plays a major role in the pathogenesis
of atherosclerosis, increasing the risk of adverse cardiovascular events [
85
]. This is demonstrated
eectively in a study of 50 patients with CAD who underwent coronary artery stenting with single
or multiple bare-metal stents (BMS), and 8 controls with CAD who underwent only diagnostic
coronary angiography [
86
]. The single stent implantation group comprised 25 persons of mean
age
59.7 ±9.44 years
, and the multiple stent implantation group comprised 25 persons of mean age
57.5 ±8.1 years
. Blood for the measurement of vWF was taken from the coronary sinus, before and after
PCI. There was no significant dierence in vWF plasma levels before and after coronary angiography
Int. J. Mol. Sci. 2020,21, 7804 11 of 18
in the controls (123
±
10.8 IU/dL before, 125
±
11.6 IU/dL after). In patients with a single stent
implantation, the plasma levels of vWF rose slightly from 113.6
±
39.6 to 121.35
±
46.63 IU/dL after
stenting. In patients with multiple stent implantation, a steep rise in the plasma levels of vWF
was detected, from
112.7 ±25.16 IU/dL
to
152.78 ±41.03 IU/dL
. Thus, multiple stenting significantly
increased the plasma levels of vWF in coronary circulation [
86
]. Another study demonstrated the
dierent influence on vWF plasma levels of BMS and drug-eluting stents (DES) [
87
]. In total, 16 patients
with the proximal stenosis of left anterior descending artery received BMS or DES. The plasma levels
of vWF were measured in the aorta and the coronary sinus immediately before and 2 h after stenting.
The systemic plasma levels of vWF were measured at the baseline and 24 h after stenting. The baseline
plasma levels of vWF were similar in both groups. The change in plasma levels of vWF in the coronary
sinus, 2 h after stent implantation, was +20.1
±
26.9% in the BMS group,
and 5.7 ±23.02%
in the
DES group. The systemic baseline plasma levels rose from 132.8
±
58.8% to 169
±
40.7% in the BMS
group and slightly decreased from 140.6
±
84% to 136
±
39.5% in the DES group, 24 h after stent
implantation [87].
Therefore, the elevated plasma levels of vWF do not necessarily reflect a causal relationship
with the development of thrombotic complications of CAD. Rather, vWF levels rise in response to
acute inflammatory stimuli and stress. In acute inflammation, vWF levels rise and fall together with
C-reactive protein [
88
]. Dierent mediators of stress and inflammation influence vWF release from
endothelial cells. Thus, interleukin-6 (IL-6) in a complex with the soluble IL-6 receptor, IL-8, and tumor
necrosis factor-
α
(TNF-
α
), significantly stimulate the release of vWF from the endothelial Weibel–Palade
bodies. IL-6 interferes in vWF cleavage by ADAMTS-13 under flow, but not static, conditions [
89
].
Vasopressin, which rises in response to stress stimuli, induces the release of vWF from endothelial
Weibel–Palade bodies and substantially increases the plasma levels of vWF. Its analogue desmopressin
(DDAVP) is used in the treatment of vWD to sustain vWF plasma levels [
90
]. Thus, the rise in vWF
plasma levels in acute MI might reflect a reaction to ischemic injury and endothelial dysfunction,
rather than a causal role in MI development.
The physiology of vWF demands high shear rates for activation and proper functioning,
which future research into the role of vWF in the development of thrombotic complications of
CAD should take into consideration. Preliminary data show that the shear stress-induced activation of
vWF might play a role in the premature development of MI [91].
8. The Potential for New Treatment, Targeting von Willebrand Factor and ADAMTS-13
in Cardiovascular Diseases
Most medications, routinely used in CAD to prevent thrombotic complications, do not aect vWF
and ADAMTS-13. Only heparins were shown to bind to a site on vWF that overlapped the A1 domain,
thus impairing the GPIb-mediated platelet adhesion, measured by the ristocetin-cofactor activity [
92
].
In clinical studies, administration of enoxaparin, a low-molecular-weight heparin, was associated with
a decrease in the plasma levels of vWF, MI frequency, and death in MI [67,93].
At the turn of the century, antibodies against the A1 domain of vWF, such as AJvW-2 and
AJW200, were studied [
94
]. Despite promising preliminary results, no further studies and clinical
investigations were reported. Current vWF-specific therapeutics is under development focus on
aptamer antagonists of the A1 domain of vWF. The administration of the first-generation aptamer,
ARC1779, to healthy volunteers, resulted in the dose- and concentration-dependent inhibition of vWF
activity [
95
]. The ARC1779 antithrombotic eect was also studied on 36 patients who underwent carotid
endartherectomy. It was shown that ARC1779 inhibits vWF activity and reduces thromboembolism in
humans, but also provokes bleeding complications [
96
]. Recently, the development of novel aptamers
was reported [
97
,
98
]. The only drug targeting the A1 domain of vWF that is approved for clinical use is
ALX-0081 (caplacizumab) [
47
]. Caplacizumab is a humanized bivalent nanobody that specifically binds
to the GPIb binding site on the A1 domain of vWF. Following remarkable results of the HERCULES
trial, in which its administration resulted in a lower incidence of TTP-related death, thromboembolism
Int. J. Mol. Sci. 2020,21, 7804 12 of 18
and the recurrence of TTP [
47
], caplacizumab was approved in the European Union in 2018 and in the
USA in early 2019, to treat adults with TTP. Studies of caplacizumab safety and ecacy in patients with
CAD are sparse. The ecacy of caplacizumab was shown in ex vivo study of 9 patients with CAD
and 11 healthy controls [
99
]. Caplacizumab completely inhibited platelet adhesion and aggregation,
measured by ristocetin-cofactor assay, platelet function analyzer, and in flow chambers. The ecacy of
caplacizumab was not influenced by antithrombotic medications, which included aspirin, clopidogrel,
and heparin [
99
]. In a randomized, placebo-controlled phase Ib trial on 46 patients with stable CAD,
undergoing PCI, administration of caplacizumab was safe, and resulted in the complete inhibition of
platelet aggregation, measured by the ristocetin-cofactor assay [
100
]. In 2009, a phase II, randomized,
open-label clinical trial was initiated in 380 high-risk patients with ACS, undergoing PCI. The objective
was to compare the bleeding risk and eectiveness of caplacizumab and abciximab, a GPIIb/IIIa
inhibitor [101]. As of 2020, no results of the study are published.
Recently, in a study on mice, treatment with recombinant human ADAMTS-13 was shown to
decrease coronary vascular dysfunction and improve cardiac remodeling after left ventricular pressure
overload [
102
]. Another study on mice showed that the administration of recombinant human
ADAMTS-13 resulted in a reduction in infarct size, the neutrophil infiltration of ischemic myocardium,
and lower troponin-I release [
32
]. Currently, there are no therapeutics targeting vWF and ADAMTS-13
approved for the treatment of cardiovascular diseases.
9. Conclusions
vWF plays an important role in cardiovascular disease. The deficiency of HMWM of vWF in
valvular heart disease and obstructive hypertrophic cardiomyopathy manifests with gastrointestinal,
skin or mucosal bleeding. The bleeding might also complicate the surgical treatment of such patients.
In CAD, the involvement of vWF is more controversial. Though abundant data show that the plasma
levels of vWF are increased and ADAMTS-13 levels are decreased in CAD, especially in MI, this does
not necessarily reflect a causal relationship between elevated plasma vWF and MI. The existing data
show that vWF levels rise in response to stress and acute inflammatory stimuli, which occur in acute MI.
Rather than focusing on the measurement of plasma levels of vWF and the evaluation of vWF activity
in static or low shear rate conditions, future research should concentrate on physiologically relevant
studies of vWF functions under high shear rates. This might pave a way to new approaches to measure
vWF function, which eventually might turn vWF into a treatment target or an important tool for
diagnostics and risk assessment in cardiovascular disease.
Author Contributions:
Conceptualization, S.K.; Methodology, S.K., Z.G.; Data search and analysis, S.O., I.M., Y.A.;
Resources, Z.G.; Data Curation, S.K.; Writing—Original Draft Preparation, S.O., I.M., Y.A.; Writing—Review &
Editing, I.M., S.K., Z.G.; Visualization, Y.A.; Supervision, Z.G.; Project Administration, Z.G.; Funding Acquisition,
I.M. and Z.G. All authors have read and agreed to the published version of the manuscript.
Funding: This work was funded by Russian Science Foundation (project #16-15-10098).
Conflicts of Interest: The authors declare no conflict of interest.
References
1.
Terraube, V.; O’Donnell, J.S.; Jenkins, P.V. Factor VIII and von Willebrand factor interaction: Biological,
clinical and therapeutic importance. Haemophilia 2010,16, 3–13. [CrossRef] [PubMed]
2.
Chen, J.; Chung, D.W. Inflammation, von Willebrand factor, and ADAMTS13. Blood
2018
,132, 141–147.
[CrossRef] [PubMed]
3.
Kawecki, C.; Lenting, P.J.; Denis, C.V. von Willebrand factor and inflammation. J. Thromb. Haemost.
2017
,15,
1285–1294. [CrossRef] [PubMed]
4.
Pendu, R.; Terraube, V.; Christophe, O.D.; Gahmberg, C.G.; de Groot, P.G.; Lenting, P.J.; Denis, C.V. P-selectin
glycoprotein ligand 1 and
β
2-integrins cooperate in the adhesion of leukocytes to von Willebrand factor.
Blood 2006,108, 3746–3752. [CrossRef] [PubMed]
Int. J. Mol. Sci. 2020,21, 7804 13 of 18
5.
Fan, M.; Wang, X.; Peng, X.; Feng, S.; Zhao, J.; Liao, L.; Zhang, Y.; Hou, Y.; Liu, J. Prognostic value of plasma
von Willebrand factor levels in major adverse cardiovascular events: A systematic review and meta-analysis.
BMC Cardiovasc. Disord. 2020,20, 72. [CrossRef] [PubMed]
6.
Kovacevic, K.D.; Mayer, F.J.; Jilma, B.; Buchtele, N.; Obermayer, G.; Binder, C.J.; Blann, A.D.; Minar, E.;
Schillinger, M.; Hoke, M. Von Willebrand factor antigen levels predict major adverse cardiovascular events
in patients with carotid stenosis of the ICARAS study. Atherosclerosis
2019
,290, 31–36. [CrossRef] [PubMed]
7.
Lv, J.-X.; Kong, Q.; Ma, X. Current advances in circulating inflammatory biomarkers in atherosclerosis and
related cardio-cerebrovascular diseases. Chronic Dis. Transl. Med. 2017,3, 207–212. [CrossRef]
8.
Van Belle, E.; Vincent, F.; Rauch, A.; Casari, C.; Jeanpierre, E.; Loobuyck, V.; Rosa, M.; Delhaye, C.;
Spillemaeker, H.; Paris, C.; et al. von Willebrand Factor and Management of Heart Valve Disease. J. Am.
Coll. Cardiol. 2019,73, 1078–1088. [CrossRef]
9.
Springer, T.A. von Willebrand factor, Jedi knight of the bloodstream. Blood
2014
,124, 1412–1425. [CrossRef]
10.
Budde, U.; Pieconka, A.; Will, K.; Schneppenheim, R. Laboratory Testing for von Willebrand Disease:
Contribution of Multimer Analysis to Diagnosis and Classification. Semin. Thromb. Hemost.
2006
,32, 514–521.
[CrossRef]
11.
Stockschlaeder, M.; Schneppenheim, R.; Budde, U. Update on von Willebrand factor multimers: Focus on
high-molecular-weight multimers and their role in hemostasis. Blood Coagul. Fibrinolysis
2014
,25, 206–216.
[CrossRef]
12.
Zhang, Q.; Zhou, Y.-F.; Zhang, C.-Z.; Zhang, X.; Lu, C.; Springer, T.A. Structural specializations of A2,
a force-sensing domain in the ultralarge vascular protein von Willebrand factor. Proc. Natl. Acad. Sci. USA
2009,106, 9226–9231. [CrossRef] [PubMed]
13.
Sadler, J.E. Biochemistry and Genetics of von willebrand factor. Annu. Rev. Biochem.
1998
,67, 395–424.
[CrossRef]
14.
Denorme, F.; Vanhoorelbeke, K.; De Meyer, S.F. von Willebrand Factor and Platelet Glycoprotein Ib:
A Thromboinflammatory Axis in Stroke. Front. Immunol. 2019,10, 2884. [CrossRef]
15.
Schneider, S.W.; Nuschele, S.; Wixforth, A.; Gorzelanny, C.; Alexander-Katz, A.; Netz, R.R.; Schneider, M.F.
Shear-induced unfolding triggers adhesion of von Willebrand factor fibers. Proc. Natl. Acad. Sci. USA
2007
,
104, 7899–7903. [CrossRef]
16.
Crawley, J.T.B.; de Groot, R.; Xiang, Y.; Luken, B.M.; Lane, D.A. Unraveling the scissile bond: How
ADAMTS13 recognizes and cleaves von Willebrand factor. Blood
2011
,118, 3212–3221. [CrossRef] [PubMed]
17.
Sing, C.E.; Alexander-Katz, A. Elongational Flow Induces the Unfolding of von Willebrand Factor at
Physiological Flow Rates. Biophys. J. 2010,98, L35–L37. [CrossRef]
18.
Westein, E.; van der Meer, A.D.; Kuijpers, M.J.E.; Frimat, J.-P.; van den Berg, A.; Heemskerk, J.W.M.
Atherosclerotic geometries exacerbate pathological thrombus formation poststenosis in a von Willebrand
factor-dependent manner. Proc. Natl. Acad. Sci. USA 2013,110, 1357–1362. [CrossRef] [PubMed]
19.
Zheng, Y.; Chen, J.; L
ó
pez, J.A. Flow-driven assembly of VWF fibres and webs in
in vitro
microvessels.
Nat. Commun. 2015,6, 7858. [CrossRef]
20.
Zhang, C.; Kelkar, A.; Neelamegham, S. von Willebrand factor self-association is regulated by the
shear-dependent unfolding of the A2 domain. Blood Adv. 2019,3, 957–968. [CrossRef]
21.
Kanaji, S.; Fahs, S.A.; Shi, Q.; Haberichter, S.L.; Montgomery, R.R. Contribution of platelet vs. endothelial VWF
to platelet adhesion and hemostasis: Hemostatic eect of platelet VWF in murine VWD. J. Thromb. Haemost.
2012,10, 1646–1652. [CrossRef] [PubMed]
22.
Verhenne, S.; Denorme, F.; Libbrecht, S.; Vandenbulcke, A.; Pareyn, I.; Deckmyn, H.; Lambrecht, A.;
Nieswandt, B.; Kleinschnitz, C.; Vanhoorelbeke, K.; et al. Platelet-derived VWF is not essential for normal
thrombosis and hemostasis but fosters ischemic stroke injury in mice. Blood
2015
,126, 1715–1722. [CrossRef]
[PubMed]
23.
Fujikawa, K.; Suzuki, H.; McMullen, B.; Chung, D. Purification of human von Willebrand factor–cleaving
protease and its identification as a new member of the metalloproteinase family. Blood
2001
,98, 1662–1666.
[CrossRef]
24.
Uemura, M.; Tatsumi, K.; Matsumoto, M.; Fujimoto, M.; Matsuyama, T.; Ishikawa, M.; Iwamoto, T.; Mori, T.;
Wanaka, A.; Fukui, H.; et al. Localization of ADAMTS13 to the stellate cells of human liver. Blood
2005
,106,
922–924. [CrossRef]
Int. J. Mol. Sci. 2020,21, 7804 14 of 18
25.
Mannucci, P.M.; Capoferri, C.; Canciani, M.T. Plasma levels of von Willebrand factor regulate ADAMTS-13,
its major cleaving protease. Br. J. Haematol. 2004,126, 213–218. [CrossRef]
26.
Xiang, Y.; de Groot, R.; Crawley, J.T.B.; Lane, D.A. Mechanism of von Willebrand factor scissile bond cleavage
by a disintegrin and metalloproteinase with a thrombospondin type 1 motif, member 13 (ADAMTS13).
Proc. Natl. Acad. Sci. USA 2011,108, 11602–11607. [CrossRef]
27.
Gragnano, F.; Sperlongano, S.; Golia, E.; Natale, F.; Bianchi, R.; Crisci, M.; Fimiani, F.; Pariggiano, I.; Diana, V.;
Carbone, A.; et al. The Role of von Willebrand Factor in Vascular Inflammation: From Pathogenesis to
Targeted Therapy. Mediat. Inflamm. 2017,2017, 5620314. [CrossRef]
28.
Petri, B.; Broermann, A.; Li, H.; Khandoga, A.G.; Zarbock, A.; Krombach, F.; Goerge, T.; Schneider, S.W.;
Jones, C.; Nieswandt, B.; et al. von Willebrand factor promotes leukocyte extravasation. Blood
2010
,116,
4712–4719. [CrossRef] [PubMed]
29.
Hillgruber, C.; Steingräber, A.K.; Pöppelmann, B.; Denis, C.V.; Ware, J.; Vestweber, D.; Nieswandt, B.;
Schneider, S.W.; Goerge, T. Blocking von Willebrand Factor for Treatment of Cutaneous Inflammation.
J. Investig. Dermatol. 2014,134, 77–86. [CrossRef] [PubMed]
30.
Aym
é
, G.; Adam, F.; Legendre, P.; Bazaa, A.; Proulle, V.; Denis, C.V.; Christophe, O.D.; Lenting, P.J. A Novel
Single-Domain Antibody against von Willebrand Factor A1 Domain Resolves Leukocyte Recruitment and
Vascular Leakage during Inflammation—Brief Report. Arter. Thromb. Vasc. Biol.
2017
,37, 1736–1740.
[CrossRef]
31.
Khan, M.M.; Motto, D.G.; Lentz, S.R.; Chauhan, A.K. ADAMTS13 reduces VWF-mediated acute inflammation
following focal cerebral ischemia in mice: Role of ADAMTS13 and VWF in inflammatory brain injury.
J. Thromb. Haemost. 2012,10, 1665–1671. [CrossRef] [PubMed]
32.
De Meyer, S.F.; Savchenko, A.S.; Haas, M.S.; Schatzberg, D.; Carroll, M.C.; Schiviz, A.; Dietrich, B.;
Rottensteiner, H.; Scheiflinger, F.; Wagner, D.D. Protective anti-inflammatory eect of ADAMTS13 on
myocardial ischemia/reperfusion injury in mice. Blood 2012,120, 5217–5223. [CrossRef]
33.
Vincentelli, A.; Susen, S.; Le Tourneau, T.; Six, I.; Fabre, O.; Juthier, F.; Bauters, A.; Decoene, C.; Goudemand, J.;
Prat, A.; et al. Acquired von Willebrand Syndrome in Aortic Stenosis. N. Engl. J. Med.
2003
,349, 343–349.
[CrossRef] [PubMed]
34.
Sousa, N.C.; Anicchino-Bizzacchi, J.M.; Locatelli, M.F.; Castro, V.; Barjas-Castro, M.L. The relationship
between ABO groups and subgroups, factor VIII and von Willebrand factor. Haematologica
2007
,92, 236–239.
[CrossRef] [PubMed]
35. Budde, U.; Schneppenheim, R. von Willebrand Factor and von Willebrand disease. Rev. Clin. Exp. Hematol.
2001,5, 335–368. [CrossRef]
36.
Franchini, M.; Capra, F.; Targher, G.; Montagnana, M.; Lippi, G. Relationship between ABO blood group and
von Willebrand factor levels: From biology to clinical implications. Thromb. J. 2007,5, 14. [CrossRef]
37.
Gill, J.C.; Endres-Brooks, J.; Bauer, P.J.; Marks, W.J.; Montgomery, R.R. The eect of ABO blood group on the
diagnosis of von Willebrand disease. Blood 1987,69, 1691–1695. [CrossRef]
38. Smith, L.J. Laboratory Diagnosis of von Willebrand Disease. Clin. Lab. Sci. 2017,30, 65–74. [CrossRef]
39.
Budde, U.; Schneppenheim, R. Interactions of von Willebrand factor and ADAMTS13 in von Willebrand
disease and thrombotic thrombocytopenic purpura. Hamostaseologie 2014,34, 215–225. [CrossRef]
40.
Roberts, J.C.; Flood, V.H. Laboratory diagnosis of von Willebrand disease. Int. J. Lab. Hematol.
2015
,37,
11–17. [CrossRef]
41.
Favaloro, E.J. The Platelet Function Analyser (PFA)-100 and von Willebrand disease: A story well over
16 years in the making. Haemophilia 2015,21, 642–645. [CrossRef] [PubMed]
42.
Harrison, P. The role of PFA-100R testing in the investigation and management of haemostatic defects in
children and adults. Br. J. Haematol. 2005,130, 3–10. [CrossRef] [PubMed]
43.
Schoeman, R.M.; Lehmann, M.; Neeves, K.B. Flow chamber and microfluidic approaches for measuring
thrombus formation in genetic bleeding disorders. Platelets 2017,28, 463–471. [CrossRef]
44.
Federici, A.B. Acquired von Willebrand syndrome: Is it an extremely rare disorder or do we see only the tip
of the iceberg? J. Thromb. Haemost. 2008,6, 565–568. [CrossRef] [PubMed]
45.
Rizzo, C.; Rizzo, S. Thrombotic thrombocytopenic purpura: A review of the literature in the light of our
experience with plasma exchange. Blood Transfus. 2012. [CrossRef]
46.
Knöbl, P. Inherited and Acquired Thrombotic Thrombocytopenic Purpura (TTP) in Adults.
Semin. Thromb. Hemost. 2014,40, 493–502. [CrossRef] [PubMed]
Int. J. Mol. Sci. 2020,21, 7804 15 of 18
47.
Scully, M.; Cataland, S.R.; Peyvandi, F.; Coppo, P.; Knöbl, P.; Kremer Hovinga, J.A.; Metjian, A.;
de la Rubia, J.; Pavenski, K.; Callewaert, F.; et al. Caplacizumab Treatment for Acquired Thrombotic
Thrombocytopenic Purpura. N. Engl. J. Med. 2019,380, 335–346. [CrossRef]
48. Heyde, E.C. Gastrointestinal Bleeding in Aortic Stenosis. N. Engl. J. Med. 1958,259, 196. [CrossRef]
49.
Warkentin, T.; Morgan, D.G.; Moore, J.C. Aortic stenosis and bleeding gastrointestinal angiodysplasia:
Is acquired von Willebrand’s disease the link? Lancet 1992,340, 35–37. [CrossRef]
50.
Warkentin, T.E.; Moore, J.C.; Morgan, D.G. Gastrointestinal Angiodysplasia and Aortic Stenosis. N. Engl.
J. Med. 2002,347, 858–859. [CrossRef]
51.
Frank, R.D.; Lanzmich, R.; Haager, P.K.; Budde, U. Severe Aortic Valve Stenosis: Sustained Cure of Acquired
von Willebrand Syndrome after Surgical Valve Replacement. Clin. Appl. Thromb. Hemost.
2017
,23, 229–234.
[CrossRef] [PubMed]
52.
Panzer, S.; Eslam, R.B.; Schneller, A.; Kaider, A.; Koren, D.; Eichelberger, B.; Rosenhek, R.; Budde, U.;
Lang, I. Loss of high-molecular-weight von Willebrand factor multimers mainly aects platelet aggregation
in patients with aortic stenosis. Thromb. Haemost. 2010,103, 408–414. [CrossRef] [PubMed]
53.
Yasar, S.J.; Abdullah, O.; Fay, W.; Balla, S. Von Willebrand factor revisited. J. Interven. Cardiol.
2018
,31,
360–367. [CrossRef] [PubMed]
54.
Van Belle, E.; Rauch, A.; Vincent, F.; Robin, E.; Kibler, M.; Labreuche, J.; Jeanpierre, E.; Levade, M.; Hurt, C.;
Rousse, N.; et al. Von Willebrand Factor Multimers during Transcatheter Aortic-Valve Replacement. N. Engl.
J. Med. 2016,375, 335–344. [CrossRef] [PubMed]
55.
Solomon, C.; Budde, U.; Schneppenheim, S.; Czaja, E.; Hagl, C.; Schoechl, H.; von Depka, M.; Rahe-Meyer, N.
Acquired type 2A von Willebrand syndrome caused by aortic valve disease corrects during valve surgery.
Br. J. Anaesth. 2011,106, 494–500. [CrossRef] [PubMed]
56.
Blackshear, J.L.; Wysokinska, E.M.; Saord, R.E.; Thomas, C.S.; Shapiro, B.P.; Ung, S.; Stark, M.E.; Parikh, P.;
Johns, G.S.; Chen, D. Shear stress-associated acquired von Willebrand syndrome in patients with mitral
regurgitation. J. Thromb. Haemost. 2014,12, 1966–1974. [CrossRef]
57.
Susen, S.; Vincentelli, A.; Le Tourneau, T.; Caron, C.; Zawadzki, C.; Prat, A.; Goudemand, J.; Jude, B. Severe Aortic
and Mitral Valve Regurgitation Are Associated with von Willebrand Factor Defect. Blood
2005
,106, 1790. [CrossRef]
58.
Blackshear, J.L.; Scha, H.V.; Ommen, S.R.; Chen, D.; Nichols, W.L. Hypertrophic Obstructive
Cardiomyopathy, Bleeding History, and Acquired von Willebrand Syndrome: Response to Septal Myectomy.
Mayo Clin. Proc. 2011,86, 219–224. [CrossRef]
59.
Le Tourneau, T.; Susen, S.; Caron, C.; Millaire, A.; Mar
é
chaux, S.; Polge, A.-S.; Vincentelli, A.; Mouquet, F.;
Ennezat, P.-V.; Lamblin, N.; et al. Functional Impairment of von Willebrand Factor in Hypertrophic
Cardiomyopathy: Relation to Rest and Exercise Obstruction. Circulation 2008,118, 1550–1557. [CrossRef]
60.
Nascimbene, A.; Neelamegham, S.; Frazier, O.H.; Moake, J.L.; Dong, J. Acquired von Willebrand syndrome
associated with left ventricular assist device. Blood 2016,127, 3133–3141. [CrossRef]
61.
Nichols, T.C.; Bellinger, D.A.; Tate, D.A.; Reddick, R.L.; Read, M.S.; Koch, G.G.; Brinkhous, K.M.; Griggs, T.R.
von Willebrand factor and occlusive arterial thrombosis. A study in normal and von Willebrand’s disease pigs
with diet-induced hypercholesterolemia and atherosclerosis. Arteriosclerosis
1990
,10, 449–461. [CrossRef]
[PubMed]
62.
Gandhi, C.; Motto, D.G.; Jensen, M.; Lentz, S.R.; Chauhan, A.K. ADAMTS13 deficiency exacerbates
VWF-dependent acute myocardial ischemia/reperfusion injury in mice. Blood
2012
,120, 5224–5230. [CrossRef]
[PubMed]
63. Seaman, C.D.; Yabes, J.; Comer, D.M.; Ragni, M.V. Does deficiency of von Willebrand factor protect against
cardiovascular disease? Analysis of a national discharge register. J. Thromb. Haemost.
2015
,13, 1999–2003.
[CrossRef] [PubMed]
64.
Xu, A.-G.; Xu, R.-M.; Lu, C.-Q.; Yao, M.-Y.; Zhao, W.; Fu, X.; Guo, J.; Xu, Q.-F.; Li, D.-D. Correlation of von
Willebrand factor gene polymorphism and coronary heart disease. Mol. Med. Rep.
2012
,6, 1107–1110.
[CrossRef]
65.
Thompson, S.G.; Kienast, J.; Pyke, S.D.M.; Haverkate, F.; van de Loo, J.C.W. Hemostatic Factors and the
Risk of Myocardial Infarction or Sudden Death in Patients with Angina Pectoris. N. Engl. J. Med.
1995
,332,
635–641. [CrossRef]
Int. J. Mol. Sci. 2020,21, 7804 16 of 18
66.
Willeit, P.; Thompson, A.; Aspelund, T.; Rumley, A.; Eiriksdottir, G.; Lowe, G.; Gudnason, V.;
Di Angelantonio, E. Hemostatic Factors and Risk of Coronary Heart Disease in General Populations:
New Prospective Study and Updated Meta-Analyses. PLoS ONE 2013,8, e55175. [CrossRef]
67.
Ray, K.K.; Morrow, D.A.; Gibson, C.M.; Murphy, S.; Antman, E.M.; Braunwald, E. Predictors of the rise in
vWF after ST elevation myocardial infarction: Implications for treatment strategies and clinical outcome.
Eur. Heart J. 2005,26, 440–446. [CrossRef]
68.
Jansson, J.H.; Nilsson, T.K.; Johnson, O. von Willebrand factor in plasma: A novel risk factor for recurrent
myocardial infarction and death. Heart 1991,66, 351–355. [CrossRef]
69.
Rutten, B.; Maseri, A.; Cianflone, D.; Laricchia,A.; Cristell, N.; Durante, A.; Spartera, M.; Ancona, F.; Limite, L.;
Hu, D.; et al. Plasma levels of active von Willebrand factor are increased in patients with first ST-segment
elevation myocardial infarction: A multicenter and multiethnic study. Eur. Heart J. Acute Cardiovasc. Care
2015,4, 64–74. [CrossRef]
70. Kaikita, K.; Soejima, K.; Matsukawa, M.; Nakagaki, T.; Ogawa, H. Reduced von Willebrand factor-cleaving
protease (ADAMTS13) activity in acute myocardial infarction. J. Thromb. Haemost.
2006
,4, 2490–2493.
[CrossRef]
71.
Crawley, J.T.B.; Lane, D.A.; Woodward,M.; Rumley, A.; Lowe, G.D.O. Evidence that high von Willebrand factor
and low ADAMTS-13 levels independently increase the risk of a non-fatal heart attack. J. Thromb. Haemost.
2008,6, 583–588. [CrossRef] [PubMed]
72.
Chion, C.K.N.K.; Doggen, C.J.M.; Crawley, J.T.B.; Lane, D.A.; Rosendaal, F.R. ADAMTS13 and von Willebrand
factor and the risk of myocardial infarction in men. Blood 2007,109, 1998–2000. [CrossRef] [PubMed]
73.
Morange, P.E.; Simon, C.; Alessi, M.C.; Luc, G.; Arveiler, D.; Ferrieres, J.; Amouyel, P.; Evans, A.; Ducimetiere, P.;
Juhan-Vague, I. Endothelial Cell Markers and the Risk of Coronary Heart Disease: The Prospective Epidemiological
Study of Myocardial Infarction (PRIME) Study. Circulation 2004,109, 1343–1348. [CrossRef]
74.
Li, Y.; Li, L.; Dong, F.; Guo, L.; Hou, Y.; Hu, H.; Yan, S.; Zhou, X.; Liao, L.; Allen, T.D.; et al. Plasma von
Willebrand factor level is transiently elevated in a rat model of acute myocardial infarction. Exp. Ther. Med.
2015,10, 1743–1749. [CrossRef]
75.
Price, J. Relationship between smoking and cardiovascular risk factors in the development of peripheral
arterial disease and coronary artery disease; Edinburgh Artery Study Edinburgh Artery Study. Eur. Heart J.
1999,20, 344–353. [CrossRef] [PubMed]
76.
Danesh, J.; Wheeler, J.G.; Hirschfield, G.M.; Eda, S.; Eiriksdottir, G.; Rumley, A.; Lowe, G.D.O.; Pepys, M.B.;
Gudnason, V. C-Reactive Protein and Other Circulating Markers of Inflammation in the Prediction of
Coronary Heart Disease. N. Engl. J. Med. 2004,350, 1387–1397. [CrossRef]
77.
Lee, K.; Blann, A.; Lip, G. High pulse pressure and nondipping circadian blood pressure in patients
with coronary artery disease: Relationship to thrombogenesis and endothelial damage/dysfunction.
Am. J. Hypertens. 2005,18, 104–115. [CrossRef]
78.
Lip, G.Y.H.; Blann, A.D.; Jones, A.F.; Lip, P.L.; Beevers, D.G. Relation of Endothelium, Thrombogenesis,
and Hemorheology in Systemic Hypertension to Ethnicity and Left Ventricular Hypertrophy. Am. J. Cardiol.
1997,80, 1566–1571. [CrossRef]
79.
Jager, A.; van Hinsbergh, V.W.M.; Kostense, P.J.; Emeis, J.J.; Yudkin, J.S.; Nijpels, G.; Dekker, J.M.; Heine, R.J.;
Bouter, L.M.; Stehouwer, C.D.A. von Willebrand Factor, C-Reactive Protein, and 5-Year Mortality in Diabetic
and Nondiabetic Subjects: The Hoorn Study. Arter. Thromb. Vasc. Biol 1999,19, 3071–3078. [CrossRef]
80.
Stehouwer, C.A.; Zeldenrust, G.C.; den Ottolander, G.H.; Hackeng, W.H.L.; Donker, A.J.M.; Nauta, J.J.P.
Urinary albumin excretion, cardiovascular disease, and endothelial dysfunction in non-insulin-dependent
diabetes mellitus. Lancet 1992,340, 319–323. [CrossRef]
81.
Warlo, E.M.K.; Pettersen, A.-Å.R.; Arnesen, H.; Seljeflot, I. vWF/ADAMTS13 is associated with on-aspirin
residual platelet reactivity and clinical outcome in patients with stable coronary artery disease. Thromb. J.
2017,15, 28. [CrossRef] [PubMed]
82.
Blann, A.D.; Herrick, A.; Jayson, M.I.V. Altered levels of soluble adhesion molecules in rheumatoid arthritis,
vasculitis and systemic sclerosis. Rheumatology 1995,34, 814–819. [CrossRef] [PubMed]
83.
Nossent, J.C.; Raymond, W.D.; Eilertsen, G.Ø. Increased von Willebrand factor levels in patients with
systemic lupus erythematosus reflect inflammation rather than increased propensity for platelet activation.
Lupus Sci. Med. 2016,3, e000162. [CrossRef] [PubMed]
Int. J. Mol. Sci. 2020,21, 7804 17 of 18
84.
Lacquemant, C.; Gaucher, C.; Delorme, C.; Chatellier, G.; Gallois, Y.; Rodier, M.; Passa, P.; Balkau, B.;
Mazurier, C.; Marre, M.; et al. Association between high von Willebrand factor levels and the Thr789Ala
vWF gene polymorphism but not with nephropathy in type I diabetes. Kidney Int.
2000
,57, 1437–1443.
[CrossRef] [PubMed]
85.
Bonetti, P.O.; Lerman, L.O.; Lerman, A. Endothelial Dysfunction: A Marker of Atherosclerotic Risk.
ATVB 2003,23, 168–175. [CrossRef] [PubMed]
86.
Heper, G.; Murat, S.N.; Durmaz, T.; Kalkan, F. Prospective Evaluation of von Willebrand Factor Release after
Multiple and Single Stenting. Angiology 2004,55, 177–186. [CrossRef] [PubMed]
87.
Kefer, J.M.; Galanti, L.M.; Desmet, S.; Deneys, V.; Hanet, C.E. Time course of release of inflammatory markers
after coronary stenting: Comparison between bare metal stent and sirolimus-eluting stent. Coron. Artery Dis.
2005,16, 505–509. [CrossRef]
88.
Pottinger, B.E.; Read, R.C.; Paleolog, E.M.; Higgins, P.G.; Pearson, J.D. von Willebrand factor is an acute
phase reactant in man. Thromb. Res. 1989,53, 387–394. [CrossRef]
89.
Bernardo, A.; Ball, C.; Nolasco, L.; Choi, H.; Moake, J.L.; Dong, J.F. Platelets adhered to endothelial cell-bound
ultra-large von Willebrand factor strings support leukocyte tethering and rolling under high shear stress.
J. Thromb. Haemost. 2005,3, 562–570. [CrossRef]
90.
Kaufmann, J.E.; Oksche, A.; Wollheim, C.B.; Günther, G.; Rosenthal, W.; Vischer, U.M. Vasopressin-induced
von Willebrand factor secretion from endothelial cells involves V2 receptors and cAMP. J. Clin. Investig.
2000
,
106, 107–116. [CrossRef]
91.
Melnikov, I.; Avtaeva, Y.; Kozlov, S.; Nozadze, D.; Gabbasov, Z. P721Shear stress induced unfolding of von
willebrand factor may be involved in the premature development of myocardial infarction. Eur. Heart J.
2019,40, ehz747.0326. [CrossRef]
92.
Sobel, M.; McNeill, P.M.; Carlson, P.L.; Kermode, J.C.; Adelman, B.; Conroy, R.; Marques, D. Heparin
inhibition of von Willebrand factor-dependent platelet function
in vitro
and
in vivo
.J. Clin. Investig.
1991
,
87, 1787–1793. [CrossRef] [PubMed]
93.
Montalescot, G.; Philippe, F.; Ankri, A.; Vicaut, E.; Bearez, E.; Poulard, J.E.; Carrie, D.; Flammang, D.;
Dutoit, A.; Carayon, A.; et al. Early Increase of von Willebrand Factor Predicts Adverse Outcome in Unstable
Coronary Artery Disease: Beneficial Eects of Enoxaparin. Circulation 1998,98, 294–299. [CrossRef]
94.
Kageyama, S.; Matsushita, J.; Yamamoto, H. Eect of a humanized monoclonal antibody to von Willebrand
factor in a canine model of coronary arterial thrombosis. Eur. J. Pharmacol. 2002,443, 143–149. [CrossRef]
95.
Gilbert, J.C.; DeFeo-Fraulini, T.; Hutabarat, R.M.; Horvath, C.J.; Merlino, P.G.; Marsh, H.N.; Healy, J.M.;
BouFakhreddine, S.; Holohan, T.V.; Schaub, R.G. First-in-Human Evaluation of Anti–von Willebrand Factor
Therapeutic Aptamer ARC1779 in Healthy Volunteers. Circulation
2007
,116, 2678–2686. [CrossRef] [PubMed]
96.
Markus, H.S.; McCollum, C.; Imray, C.; Goulder, M.A.; Gilbert, J.; King, A. The von Willebrand Inhibitor
ARC1779 Reduces Cerebral Embolization after Carotid Endarterectomy: A Randomized Trial. Stroke
2011
,
42, 2149–2153. [CrossRef]
97.
Kovacevic, K.D.; Buchtele, N.; Schoergenhofer, C.; Derhaschnig, U.; Gelbenegger, G.; Brostjan, C.; Zhu, S.;
Gilbert, J.C.; Jilma, B. The aptamer BT200 eectively inhibits von Willebrand factor (VWF) dependent platelet
function after stimulated VWF release by desmopressin or endotoxin. Sci. Rep. 2020,10, 11180. [CrossRef]
98.
Sakai, K.; Someya, T.; Harada, K.; Yagi, H.; Matsui, T.; Matsumoto, M. Novel aptamer to von Willebrand
factor A1 domain (TAGX-0004) shows total inhibition of thrombus formation superior to ARC1779 and
comparable to caplacizumab. Haematologica 2019. [CrossRef]
99.
van Loon, J.E.; de Jaegere, P.P.T.; van Vliet, H.H.D.M.; de Maat, M.P.M.; de Groot, P.G.; Simoons, M.L.;
Leebeek, F.W.G. The
in vitro
eect of the new antithrombotic drug candidate ALX-0081 on blood samples of
patients undergoing percutaneous coronary intervention. Thromb. Haemost 2011,106, 165–171. [CrossRef]
100.
Bartunek, J.; Barbato, E.; Vercruysse, K.; Duby, C.; Wijns, W.; Heyndrickx, G.; Holz, J.-B. Abstract 15084: Safety
and Ecacy of Anti-von Willebrand Factor Nanobody
®
ALX-0081 in Stable Angina Patients Undergoing
Percutaneous Coronary Intervention. Circulation 2010,122, A15084.
Int. J. Mol. Sci. 2020,21, 7804 18 of 18
101.
Bartunek, J.; Barbato, E.; Heyndrickx, G.; Vanderheyden, M.; Wijns, W.; Holz, J.-B. Novel Antiplatelet Agents:
ALX-0081, a Nanobody Directed towards von Willebrand Factor. J. Cardiovasc. Trans. Res.
2013
,6, 355–363.
[CrossRef] [PubMed]
102.
Witsch, T.; Martinod, K.; Sorvillo, N.; Portier, I.; De Meyer, S.F.; Wagner, D.D. Recombinant Human
ADAMTS13 Treatment Improves Myocardial Remodeling and Functionality after Pressure Overload Injury
in Mice. JAHA 2018,7. [CrossRef] [PubMed]
Publisher’s Note:
MDPI stays neutral with regard to jurisdictional claims in published maps and institutional
aliations.
©
2020 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access
article distributed under the terms and conditions of the Creative Commons Attribution
(CC BY) license (http://creativecommons.org/licenses/by/4.0/).
... The data regarding MR influence of thromboembolic risk are contradictory [6][7][8][9][10]. Several studies have indicated that significant MR jets, likely attributed to the extensive blood flow around the left atrium (LA) and its appendage, may contribute to a reduction in thromboembolic events, including stroke [6,[11][12][13]. ...
... However, concerns arise about the turbulent nature of regurgitant flow, possibly contributing to coagulation activation [7,10]. Shear forces from turbulent blood flow may activate the coagulation cascade, enhancing the prothrombotic clot phenotype [7,10]. ...
... However, concerns arise about the turbulent nature of regurgitant flow, possibly contributing to coagulation activation [7,10]. Shear forces from turbulent blood flow may activate the coagulation cascade, enhancing the prothrombotic clot phenotype [7,10]. MR may, on one hand, be associated with a protective effect due to blood mobilization from the LA via the regurgitant jet. ...
Article
Full-text available
Introduction: The intricate management of heart failure (HF), especially in the context of reduced ejection fraction, is compounded by an elevated risk of thromboembolic events. Existing studies offer inconclusive insights into the interplay between MR and the coagulation system. Objectives: This study aimed to investigate the impact of transcatheter edge-to-edge repair (TEER) on specific coagulation parameters in HF patients. Patients and methods: A cohort of 31 HF patients with severe MR undergoing TEER underwent systematic evaluation at three time points (V1, V2, and V3). Coagulation parameters, including fibrinogen concentration, thrombin generation, fibrin clot permeability (Ks), and clot lysis time (CLT), were assessed (n = 27 [V2], and n = 25 [V3]). Results: TEER induced changes in fibrinogen levels (P = 0.01, V3 vs. V2) and improved fibrin clot properties over a 50-day follow-up (Ks, P = 0.01, V3 vs. V2). No significant differences were observed among time points in analyzed blood clot parameters. Correlation analysis showed that baseline CLT was significantly associated with delta NT-proBNP, (P = 0.049; r = 0.40). Multivariable analysis demonstrated that baseline CLT was an independent predictor of the early post-TEER NT-proBNP change (R2 = 0.55, P = 0.02). Conclusions: We found that fibrinogen levels decreased, and permeation coefficient increased over a median 50-day post-TEER follow-up, compared to early post-procedure assessments. Other blood coagulation parameters remained unchanged from baseline to both follow-up periods after TEER. Finally, CLT was an independent predictor of early NT-proBNP increase, emphasizing its role as an indicator of the hemodynamic response to TEER.
... Moreover, coagulation factor VIII is bound to vWF and thus protected [16]. ...
... The A2 domain of the molecule seems to function as a shear sensor within the inactive vWF polymer and serves as a region for further cleavage by ADAMTS13 [14]. In vivo studies confirmed decreased polymer length in pathophysiological settings with increased shear stress, such as severe aortic stenosis [17], hypertrophic cardiomyopathy [16], or non-endothelized extracorporeal circulation circuits [9] like intraoperative cardiopulmonary bypass, extracorporeal membrane oxygenation (ECMO) [18], or left ventricular assist devices [19]. This quantitative loss of HMW molecules is also known as acquired von Willebrand syndrome (AVWS). ...
Article
Full-text available
Background Von Willebrand factor (vWF) is an important part of blood coagulation since it binds platelets to each other and to endothelial cells. In traumatic and surgical haemorrhage, both blood cells and plasmatic factors are consumed, leading to consumption coagulopathy and fluid resuscitation. This often results in large amounts of crystalloids and blood products being infused. Additional administration of vWF complex and platelets might mitigate this problem. We hypothesize that administration of vWF concentrate additionally to platelet concentrates reduces blood loss and the amount of blood products (platelets, red blood cells [RBC], fresh frozen plasma [FFP]) administered. Methods We conducted a monocentric 6-year retrospective data analysis of cardiac surgery patients. Included were all patients receiving platelet concentrates within 48 h postoperatively. Patients who additionally received vWF concentrates were allocated to the intervention group and all others to the control group. Groups were compared in mixed regression models correcting for known confounders, based on nearest neighbour propensity score matching. Primary endpoints were loss of blood (day one and two) and amount of needed blood products on day one and two (platelets, RBC, FFP). Secondary endpoints were intensive care unit (ICU) and in-hospital length of stay, ICU and in-hospital mortality, and absolute difference of platelet counts before and after treatment. Results Of 497 patients analysed, 168 (34%) received vWF concentrates. 121 patients in both groups were considered for nearest neighbour matching. Patients receiving additional vWF were more likely to receive more blood products (RBC, FFP, platelets) in the first 24 h after surgery and had around 200 mL more blood loss at the same time. Conclusion In this retrospective analysis, no benefit in additional administration of vWF to platelet concentrates on perioperative blood loss, transfusion requirement (platelets, RBC, FFP), length of stay, and mortality could be found. These findings should be verified in a prospective randomized controlled clinical trial (www.clinicaltrials.gov identifier NCT04555785).
... It is synthesized in vascular endothelium and megakaryocytes, then stored in Weibel-Palade bodies of endotheliocytes and granules of platelets or released into the circulation. Upon vascular wall damage, plasma VWF binds to collagen in the exposed subendothelial matrix, and platelet glycoprotein Iba (GPIba) triggers platelet aggregation and thrombus formation [1]. ...
Article
Full-text available
Background Von Willebrand factor (VWF) is a large, multimeric glycoprotein that plays a role in thrombus formation; it is also an important mediator of inflammation. Our study aims to determine the association of VWF plasma level and acute ischemic stroke and determine plasma level of VWF in different subtypes of acute ischemic stroke. This case–control study was conducted on 90 subjects: 30 acute ischemic atherosclerotic stroke patients, 30 acute cardioembolic stroke patients and 30 healthy age and sex-matched control subjects. Stroke patients were recruited within the first week of stroke onset with an age range from 18 to 75 years. All subjects underwent complete neurological examination, duplex ultrasonography (U/S), CT brain, routine laboratory work-up and serum level of VWF. Results VWF serum levels were significantly elevated in patients of acute ischemic stroke, compared to control subjects. Higher plasma levels of VWF were observed in patients with acute ischemic atherosclerotic stroke. Conclusion Serum level of VWF can be used as a marker for acute ischemic stroke, especially the atherosclerotic subtype.
... von Willebrand Factor has different conformations depending on the magnitude of shear stress that the glycoprotein is exposed to (Okhota et al., 2020). The original form of von Willebrand Factor is very similar to a folded protein, existing in a globular state. ...
Article
Full-text available
This review examines the endothelial glycocalyx’s role in inflammation and explores its involvement in coagulation. The glycocalyx, composed of proteins and glycosaminoglycans, interacts with von Willebrand Factor and could play a crucial role in anchoring it to the endothelium. In inflammatory conditions, glycocalyx degradation may leave P-selectin as the only attachment point for von Willebrand Factor, potentially leading to uncontrolled release of ultralong von Willebrand Factor in the bulk flow in a shear stress-dependent manner. Identifying specific glycocalyx glycosaminoglycan interactions with von Willebrand Factor and P-selectin can offer insights into unexplored coagulation mechanisms.
... However, when shear stress is elevated, as in microvessels or stenotic arteries, platelets adhere largely to vWF, and vWF may also influence platelet aggregation. When shear stress is moderate, as in veins and bigger arteries, platelets attach primarily to collagen and fibronectin [40]. ...
Article
Full-text available
Cardiovascular disease is one of the chief factors that cause ischemic stroke, myocardial infarction, and venous thromboembolism. The elements that speed up thrombosis include nutritional consumption, physical activity, and oxidative stress. Even though the precise etiology and pathophysiology remain difficult topics that primarily rely on traditional medicine. The diagnosis and management of thrombosis are being developed using discrete non-invasive and non-surgical approaches. One of the emerging promising approach is ultrasound and photoacoustic imaging. The advancement of nanomedicines offers concentrated therapy and diagnosis, imparting efficacy and fewer side effects which is more significant than conventional medicine. This study addresses the potential of nanomedicines as theranostic agents for the treatment of thrombosis. In this article, we describe the factors that lead to thrombosis and its consequences, as well as summarize the findings of studies on thrombus formation in preclinical and clinical models and also provide insights on nanoparticles for thrombus imaging and therapy.
... This phenomenon is seen in various diseases, such as PE, disseminated intravascular coagulation (DIC), thrombotic thrombocytopenia purpura (TTP), scleroderma-associated pulmonary hypertension, and arterial hypertension. 6 Further, vWF is a massive heterogeneous, sticky glycoprotein which is constitutively produced by the endothelium and stored in Weibel-Palade bodies. The vWF is activated when it binds to sub-endothelial structures exposed after endothelial injury, or in small arterioles and atherosclerotic arteries with significant shear stresses. ...
Article
Full-text available
Objective: To determine and compare plasma thrombomodulin, von Willebrand factor and von Willebrand factor-cleaving protease levels between pre-eclamptic and healthy pregnant females. Method: The cross-sectional, comparative study was conducted at the Department of Haematology, University of Health Sciences, Lahore, Pakistan, from November 2019 to December 2020, and comprised pregnant females who were divided into healthy pregnant group A and pre-eclamptic group B. Plasma thrombomodulin and von Willebrand factor-cleaving protease levels were determined by using commercially available enzyme-linked immunosorbent assay kit, and von Willebrand factor level was determined by using immuno-turbidimetric assay kit. Data was analysed using SPSS 25. Results: Of the 88 participants, there were 44(50%) females with mean age 25.5±6 years in group A and 44(50%) in group B with mean age 26±5 years. Median thrombomodulin level in group B was significantly higher than group A (p=0.003). Median von Willebrand factor-cleaving protease levels were lower in group B compared to group A (p=0.838). A significant difference in von Willebrand factor level was observed between the groups (p=0.038). Conclusion: Females with pre-eclampsia had significantly higher plasma levels of von Willebrand factor and thrombomodulin than healthy pregnant subjects. Key Words: Thrombomodulin, vWF, ADAMTS13 protein, Pre-eclampsia.
Article
Degenerative aortic stenosis and coronary artery disease are considered to be the most prevalent cardiovascular diseases in industrialized countries. This study aims to determine the change over time in von Willebrand factor antigen, von Willebrand factor activity, and factor VIII and where there is a correlation with total post-operative drainage. The single-center retrospective study included 203 consecutive patients (64.5% male), undergoing coronary artery bypass surgery between March 1, 2019 and June 30, 2020 at the University Clinical Center of Serbia in the Clinic for Cardiac Surgery in Belgrade, Serbia. All patients 18 years or older who presented with isolated, hemodynamically significant aortic stenosis were included. The control group consisted of patients who presented with only coronary artery disease. Between patients with only coronary artery disease and patients with coronary artery diseases and aortic stenosis, there was a statistically significant difference between pre-op and 1-month post-op fibrinogen, factor VIII, von Willebrand factor antigen, and von Willebrand factor (p < 0.001), post-op drainage, with overall lower drainage in coronary artery disease patients, and consistent increase in von Willebrand factor antigen, von Willebrand factor activity, and Factor VIII post-operatively in patients with coronary artery diseases and aortic stenosis. This study has shown that there is a correlation between von Willebrand factor antigen, von Willebrand factor activity and total drainage to the level of statistical significance in aortic stenosis patients and in the overall study population.
Article
Full-text available
Immunomodulatory drugs (IMiDs) are key drugs for treating multiple myeloma and myelodysplastic syndrome with chromosome 5q deletion. IMiDs exert their pleiotropic effects through the interaction between cell-specific substrates and cereblon, a substrate receptor of the E3 ubiquitin ligase complex. Thus, identification of cell-specific substrates is important for understanding the effects of IMiDs. IMiDs increase the risk of thromboembolism, which sometimes results in fatal clinical outcomes. In this study, we sought to clarify the molecular mechanisms underlying IMiDs-induced thrombosis. We investigated cereblon substrates in human megakaryocytes using liquid chromatography-mass spectrometry and found that thrombospondin-1 (THBS-1), which is an inhibitor of a disintegrin-like and metalloproteinase with thrombospondin type 1 motifs 13 (ADAMTS13), functions as an endogenous substrate in human megakaryocytes. IMiDs inhibited the proteasomal degradation of THBS-1 by impairing the recruitment of cereblon to THBS-1, leading to aberrant accumulation of THBS-1. We observed a significant increase in THBS-1 in peripheral blood mononuclear cells (PBMNCs) as well as larger von Willebrand factor (vWF) multimers in the plasma of patients with myeloma, who were treated with IMiDs. These results collectively suggest that THBS-1 represents an endogenous substrate of cereblon. This pairing is disrupted by IMiDs, and the aberrant accumulation of THBS-1 plays an important role in the pathogenesis of IMiDs-induced thromboembolism.
Article
Full-text available
During the initial diagnosis of urgent medical conditions, which include acute infectious diseases, it is important to assess the severity of the patient’s clinical state as quickly as possible. Unlike individual biochemical or physiological indicators, derived indices make it possible to better characterize a complex syndrome as a set of symptoms, and therefore quickly take a set of adequate measures. Recently, we reported on novel diagnostic indices containing butyrylcholinesterase (BChE) activity, which is decreased in COVID-19 patients. Also, in these patients, the secretion of von Willebrand factor (vWF) increases, which leads to thrombosis in the microvascular bed. The objective of this study was the determination of the concentration and activity of vWF in patients with COVID-19, and the search for new diagnostic indices. One of the main objectives was to compare the prognostic values of some individual and newly derived indices. Patients with COVID-19 were retrospectively divided into two groups: survivors (n = 77) and deceased (n = 24). According to clinical symptoms and computed tomography (CT) results, the course of disease was predominantly moderate in severity. The first blood sample (first point) was taken upon admission to the hospital, the second sample (second point)—within 4–6 days after admission. Along with the standard spectrum of biochemical indicators, BChE activity (BChEa or BChEb for acetylthiocholin or butyrylthiocholin, respectively), malondialdehyde (MDA), and vWF analysis (its antigen level, AGFW, and its activity, ActWF) were determined and new diagnostic indices were derived. The pooled sensitivity, specificity, and area under the receiver operating curve (AUC), as well as Likelihood ratio (LR) and Odds ratio (OR) were calculated. The level of vWF antigen in the deceased group was 1.5-fold higher than the level in the group of survivors. Indices that include vWF antigen levels are superior to indices using vWF activity. It was found that the index [Urea] × [AGWF] × 1000/(BChEb × [ALB]) had the best discriminatory power to predict COVID-19 mortality (AUC = 0.91 [0.83, 1.00], p < 0.0001; OR = 72.0 [7.5, 689], p = 0.0002). In addition, [Urea] × 1000/(BChEb × [ALB]) was a good predictor of mortality (AUC = 0.95 [0.89, 1.00], p < 0.0001; OR = 31.5 [3.4, 293], p = 0.0024). The index [Urea] × [AGWF] × 1000/(BChEb × [ALB]) was the best predictor of mortality associated with COVID-19 infection, followed by [Urea] × 1000/(BChEb × [ALB]). After validation in a subsequent cohort, these two indices could be recommended for diagnostic laboratories.
Article
Purpose: To develop a mathematical model for predicting shear-induced von Willebrand factor (vWF) function modification which can be used to guide ventricular assist devices (VADs) design, and evaluate the damage of high molecular weight multimers (HMWM)-vWF in VAD patients for reducing clinical complications. Methods: Mathematical models were constructed based on three morphological variations (globular vWF, unfolded vWF and degraded vWF) of vWF under shear stress conditions, in which parameters were obtained from previous studies or fitted by experimental data. Different clinical support modes (pediatric vs. adult mode), different VAD operating states (pulsation vs. constant mode) and different clinical VADs (HeartMate II, HeartWare and CentriMag) were utilized to analyze shear-induced damage of HMWM-vWF based on our vWF model. The accuracy and feasibility of the models were evaluated using various experimental and clinical cases, and the biomechanical mechanisms of HMWM-vWF degradation induced by VADs were further explained. Results: The mathematical model developed in this study predicted VAD-induced HMWM-vWF degradation with high accuracy (correlation with experimental data r2 > 0.99). The numerical results showed that VAD in the pediatric mode resulted in more HMWM-vWF degradation per unit time and per unit flow rate than in the adult mode. However, the total degradation of HMWM-vWF is less in the pediatric mode than in the adult mode because the pediatric mode has fewer times of blood circulation than the adult mode in the same amount of time. The ratio of HMWM-vWF degradation was lower in the pulsation mode than in the constant mode. This is due to the increased flushing of VADs in the pulsation mode, which avoids prolonged stagnation of blood in high shear regions. This study also found that the design feature, rotor size and volume of the VADs, and the superimposed regions of high shear stress and long residence time inside VADs affect the degradation of HMWM-vWF. The axial flow VADs (HeartMate II) showed higher degradation of HMWM-vWF compared to centrifugal VADs (HeartWare and CentriMag). Compared to fully magnetically suspended VADs (CentriMag), hydrodynamic suspended VADs (HeartWare) produced extremely high degradation of HWMW-vWF in its narrow hydrodynamic clearance. Finally, the study used a mathematical model of HMWM-vWF degradation to interpret the clinical statistics from a biomechanical perspective and found that minimizing the rotating speed of VADs within reasonable limits helps to reduce HWMW-vWF degradation. All predicted conclusions are supported by the experimental and clinical data. Conclusion: This study provides a validated mathematical model to assess the shear-induced degradation of HMWM-vWF, which can help to evaluate the damage of HMWM-vWF in patients implanted with VADs for reducing clinical complications, and to guide the optimization of VADs for improving hemocompatibility.
Article
Full-text available
Von Willebrand factor (VWF) plays a major role in arterial thrombosis. Antiplatelet drugs induce only a moderate relative risk reduction after atherothrombosis, and their inhibitory effects are compromised under high shear rates when VWF levels are increased. Therefore, we investigated the ex vivo effects of a third-generation anti-VWF aptamer (BT200) before/after stimulated VWF release. We studied the concentration-effect curves BT200 had on VWF activity, platelet plug formation under high shear rates (PFA), and ristocetin-induced platelet aggregation (Multiplate) before and after desmopressin or endotoxin infusions in healthy volunteers. VWF levels increased > 2.5-fold after desmopressin or endotoxin infusion (p < 0.001) and both agents elevated circulating VWF activity. At baseline, 0.51 µg/ml BT200 reduced VWF activity to 20% of normal, but 2.5-fold higher BT200 levels were required after desmopressin administration (p < 0.001). Similarly, twofold higher BT200 concentrations were needed after endotoxin infusion compared to baseline (p < 0.011). BT200 levels of 0.49 µg/ml prolonged collagen-ADP closure times to > 300 s at baseline, whereas 1.35 µg/ml BT200 were needed 2 h after desmopressin infusion. Similarly, twofold higher BT200 concentrations were necessary to inhibit ristocetin induced aggregation after desmopressin infusion compared to baseline (p < 0.001). Both stimuli elevated plasma VWF levels in a manner representative of thrombotic or pro-inflammatory conditions such as arterial thrombosis. Even under these conditions, BT200 potently inhibited VWF activity and VWF-dependent platelet function, but higher BT200 concentrations were required for comparable effects relative to the unstimulated state.
Article
Full-text available
Background: Prediction of major adverse cardiovascular events (MACEs) may offer great benefits for patients with coronary artery disease (CAD). Von Willebrand factor (vWF) is stored in endothelial cells and released into blood plasma upon vascular dysfunction. This meta-analysis was performed to evaluate the prognostic value of plasma vWF levels in CAD patients with MACEs. Methods: A total of 15 studies were included in this meta-analysis through the search in PubMed, Embase and CNKI. Data were collected from 960 patients who had MACEs after CAD and 3224 controls nested without the adverse events. The standard mean difference (SMD) and 95% confidence intervals (95% CI) were calculated using random-effects model. Results: The plasma vWF levels examined at 24 h and 48 h after admission were significantly higher in CAD patients with MACEs than those without. The pooled SMD among the MACEs group and the non-MACEs group was 0.55 (95% CI = 0.30-0.80, P < 0.0001) and 0.70 (95% CI = 0.27-1.13, P = 0.001), respectively. However, no significant difference was found in plasma vWF levels on admission between the two groups. Conclusion: Plasma vWF level in CAD patients examined at 24 h and 48 h after admission might be an independent prognostic factor for MACE.
Article
Full-text available
von Willebrand factor (VWF) is a blood glycoprotein that plays an important role in platelet thrombus formation through interaction between its A1 domain and platelet glycoprotein Ib. ARC1779, an aptamer to the VWF A1 domain, was evaluated in a clinical trial for acquired thrombotic thrombocytopenic purpura (aTTP). Subsequently, caplacizumab, an anti-VWF A1 domain nanobody, was approved for aTTP in Europe and the United States. We recently developed a novel DNA aptamer, TAGX-0004, to the VWF A1 domain; it contains an artificial base and demonstrates high affinity for VWF. To compare the effects of these three agents on VWF A1, their ability to inhibit ristocetin- or botrocetin-induced platelet aggregation under static conditions was analyzed, and the inhibition of thrombus formation under high shear stress was investigated in a microchip flow chamber system. In both assays, TAGX-0004 showed stronger inhibition than ARC1779, and had comparable inhibitory effects to caplacizumab. The binding sites of TAGX-0004 and ARC1779 were analyzed with surface plasmon resonance performed using alanine scanning mutagenesis of the VWF A1 domain. An electrophoretic mobility shift assay showed that R1395 and R1399 in the A1 domain bound to both aptamers. R1287, K1362, and R1392 contributed to ARC1779 binding, and F1366 was essential for TAGX-0004 binding. Surface plasmon resonance analysis of the binding sites of caplacizumab identified five amino acids in the VWF A1 domain (K1362, R1392, R1395, R1399, and K1406). These results suggested that TAGX-0004 possessed better pharmacological properties than caplacizumab in vitro and might be similarly promising for aTTP treatment.
Article
Full-text available
von Willebrand factor (VWF) and platelets are key mediators of normal hemostasis. At sites of vascular injury, VWF recruits platelets via binding to the platelet receptor glycoprotein Ibα (GPIbα). Over the past decades, it has become clear that many hemostatic factors, including VWF and platelets, are also involved in inflammatory processes, forming intriguing links between hemostasis, thrombosis, and inflammation. The so-called “thrombo-inflammatory” nature of the VWF-platelet axis becomes increasingly recognized in different cardiovascular pathologies, making it a potential therapeutic target to interfere with both thrombosis and inflammation. In this review, we discuss the current evidence for the thrombo-inflammatory activity of VWF with a focus on the VWF-GPIbα axis and discuss its implications in the setting of ischemic stroke.
Article
Full-text available
Background and aims: Von Willebrand factor (VWF) plays an important role in thrombogenesis and mediates platelet adhesion particularly under high shear stress. Such conditions are generally found in stenotic arteries and can eventually cause myocardial infarction or stroke. We aimed to study whether levels of VWF antigen (VWF:Ag) predict future major adverse cardiovascular events (MACE) in patients suffering from carotid artery stenosis. Methods: Patients with atherosclerotic carotid artery disease defined by the presence of nonstenotic plaques or any degree of carotid stenosis were prospectively enrolled. Concentrations of VWF were measured by enzyme immunoassay. Results: VWF:Ag levels were more stable after 4 freeze-thaw cycles, when compared to VWF activity, and we showed similar concentrations of VWF in citrated plasma and serum (±4%). Levels of VWF:Ag predicted future cardiovascular events in 811 patients with carotid stenosis independent of known cardiovascular risk factors. Patients with VWF:Ag concentrations in the 4th quartile had a 44% event rate after an average 3-year follow up and a hazard ratio of 2.15 (95% confidence interval 1.46-3.16; p < 0.001). Conclusions: High concentrations of VWF:Ag predict major cardiovascular events in patients with carotid stenosis, and given their high event rate may be useful for risk stratification of such patients.
Article
Severe aortic stenosis can be associated with mucosal and gastrointestinal bleeding probably related to loss of high molecular weight (HMW) multimers of VWF. Possible mechanisms of this loss are enhanced proteolysis by shear-dependant metalloprotease (ADAMTS 13) of HMW multimers of VWF or increased platelet-VWF interactions leading to clearance or degradation of HMW multimers of VWF. The aim of our study was to investigate wether this high shear-stress induced loss of HMW multimers of VWF is observed in other cardiac defects associated with high pressure blood flow such as severe aortic or mitral valve regurgitation. Twenty four consecutive patients were operated on for aortic valve regurgitation (n=11) or mitral valve regurgitation (n=13). Before surgery recent mucosal bleeding was reported in 25% patients (n=5).VWF Closure Time of PFA 100® (CTADP) were prolonged in 87% of patients (n=20). The ratio of ristocetin cofactor activity to antigen and the ratio of collagen-binding activity to antigen were decreased in 28%(n=5) and 45%(n=10) of patients respectively. HMW multimers of VWF were decreased in 82 % (n=18) of patients. Six months after surgery, CTADP were normal in 71% (n=17) of patients, ratios were normal in all patients evaluable for this parameter (n=14) and HMW multimers of VWF increased in all patients as compared with preoperative value (8±2,5%vs10±2,3; p=0.04) but remain under value of control plasma in 63% (n=14) of patients. In this study we demonstrated that VWF defect can be associated with severe aortic or mitral valve regurgitation and that platelet function-analyzer closure time, abnormalities of von Willebrand factor, or both could be frequent in those valve disease and might cause mucosal bleeding.
Article
Background Normally, von Willebrand factor (vWF) becomes highly reactive with platelets upon unfolding into a fibrillar conformation at critical shear rate (more than 5000 s–1), that may occur in stenotic arteries. At shear rates below critical value (1200–1300 s–1), which occur in intact coronary arteries, normally there is no conformational rearrangement of vWF. Pathologic unfolding of vWF at shear rates below critical value may increase a risk of the development of coronary thrombosis. There is little information on the role of shear stress induced conformational rearrangement of vWF in the development of myocardial infarction in young individuals. Purpose To investigate vWF-dependent platelet adhesion of patients with premature myocardial infarction at shear rates below critical value (1200–1300 s–1). Methods Using a microfluidic system, we measured platelet adhesion to a fibrinogen-coated optical surface at shear rates of 1200–1300 s–1 during 10 minutes. We assessed platelet-rich plasma of 8 male persons 40–52 years old, who had previous myocardial infarction at the age of 34–39. The control group comprised 6 healthy male volunteers 30–55 years old. We compared the intensity of scattered laser light measured in volts (V) at 10th minute. To study vWF-dependent platelet adhesion, we blocked GPIb receptor with monoclonal antibody to inhibit platelet interaction with vWF. To compare the intensity of vWF-dependent platelet adhesion with normally occurring adhesion to fibrinogen, we blocked GPIIb/IIIa receptor with monoclonal antibody. Results The inhibition of GPIb vWF-receptor decreased platelet adhesion to fibrinogen surface at shear rates of 1200–1300 s–1 by 17.8±4.7% in healthy volunteers and by 92±2.8% in persons with premature myocardial infarction (p<0.05). Inhibition of GPIIb/IIIa receptor decreased platelet adhesion by 91.5±3.8% in healthy volunteers and by 97.3±3.2% in persons with premature myocardial infarction. Conclusion Pathologic unfolding of vWF at shear rates below critical value may be involved in the development of premature myocardial infarction. Acknowledgement/Funding This work was supported by the grant of the Russian Science Foundation (project #16-15-10098)
Article
In order to firmly establish a normal range for von Willebrand factor antigen (vWF:Ag), we determined plasma vWF:Ag concentrations in 1,117 volunteer blood donors by quantitative immunoelectrophoresis. The presence of the ABO blood group has a significant influence on vWF:Ag values; individuals with blood group O had the lowest mean vWF:Ag level (74.8 U/dL), followed by group A (105.9 U/dL), then group B (116.9 U/dL), and finally group AB (123.3 U/dL). Multiple regression analysis revealed that age significantly correlated with vWF:Ag levels in each blood group. We then performed reverse ABO typing on stored plasma from 142 patients with the diagnosis of von Willebrand disease (vWd). Of 114 patients with type I vWd, blood group O was found in 88 (77%), group A in 21 (18%), group B in 5 (4%), and group AB in none (0%), whereas the frequency of these blood groups in the normal population is significantly different (45%, 45%, 7% and 3%, respectively) (P less than .001). Patients with type II or III vWd had ABO blood group frequencies that were not different from the expected distribution. There may be a subset of symptomatic vWd patients with decreased concentrations of structurally normal vWf (vWd, type I) on the basis of blood group O. Some individuals of blood group AB with a genetic defect of vWF may have the diagnosis overlooked because vWF levels are elevated due to blood type.
Article
von Willebrand factor (VWF) self-association results in the homotypic binding of VWF upon exposure to fluid shear. The molecular mechanism of this process is not established. In this study, we demonstrate that the shear-dependent unfolding of the VWF A2 domain in the multimeric protein is a major regulator of protein self-association. This mechanism controls self-association on the platelet glycoprotein Ibα receptor, on collagen substrates, and during thrombus growth ex vivo. In support of this, A2-domain mutations that prevent domain unfolding due to disulfide bridging of N- and C-terminal residues ("Lock-VWF") reduce self-association and platelet activation under various experimental conditions. In contrast, reducing assay calcium concentrations, and 2 mutations that destabilize VWF-A2 conformation by preventing coordination with calcium (D1498A and R1597W VWD type 2A mutation), enhance self-association. Studies using a panel of recombinant proteins that lack the A1 domain ("ΔA1 proteins") suggest that besides pure homotypic A2 interactions, VWF-A2 may also engage other protein domains to control self-association. Addition of purified high-density lipoprotein and apolipoprotein-A1 partially blocked VWF self-association. Overall, similar conditions facilitate VWF self-association and ADAMTS13-mediated proteolysis, with low calcium and A2 disease mutations enhancing both processes, and locking-A2 blocking them simultaneously. Thus, VWF appears to have evolved 2 balancing molecular functions in a single A2 functional domain to dynamically regulate protein size in circulation: ADAMTS13-mediated proteolysis and VWF self-association. Modulating self-association rates by targeting VWF-A2 may provide novel methods to regulate the rates of thrombosis and hemostasis.
Article
For decades, numerous observations have shown an intimate relationship between von Willebrand factor (VWF) multimer profile and heart valve diseases (HVD). The current knowledge of the unique biophysical properties of VWF helps us to understand the longstanding observations concerning the bleeding complications in patients with severe HVD. Not only does the analysis of the VWF multimer profile provide an excellent evaluation of HVD severity, it is also a strong predictor of clinical events. Also of importance, VWF responds within minutes to any significant change in hemodynamic valve status, making it an accurate marker of the quality of surgical and transcatheter therapeutic interventions. The authors provide in this review a practical, comprehensive, and evidence-based framework of the concept of VWF as a biomarker in HVD, advocating for its implementation into the clinical decision-making process besides usual clinical and imaging evaluation. They also delineate critical knowledge gaps and research priorities to definitely validate this concept.