ArticlePDF AvailableLiterature Review

The endothelial cell protein C receptor: Its role in thrombosis

Authors:

Abstract and Figures

The protein C anticoagulant pathway plays a crucial role as a regulator of the blood clotting cascade. Protein C is activated on the vascular endothelial cell membrane by the thrombin-thrombomodulin complex. The endothelial protein C receptor binds protein C and further enhances protein C activation. Once formed, activated protein C down-regulates thrombin formation by inactivating factors Va and VIIIa and exerts cytoprotective effects through endothelial protein C receptor binding. An adequate generation of activated protein C depends on the precise assembly, on the surface of the endothelial cells, of thrombin, thrombomodulin, protein C, and endothelial protein C receptor. Therefore, any change in the efficiency of this assembly may cause a reduction or increase in activated protein C generation and modulate the risk of thrombosis. This review highlights the role of the endothelial protein C receptor in disease and discusses the association of its mutations with the risk of thrombosis.
Content may be subject to copyright.
Review Article
The endothelial cell protein C receptor: Its role in thrombosis
Silvia Navarro
a
, Elena Bonet
a,b
, Amparo Estellés
a
, Ramón Montes
c
, José Hermida
c
, Laura Martos
a
,
Francisco España
a
, Pilar Medina
a,
a
Hemostasis and Thrombosis Unit, Research Center, La Fe University Hospital, Valencia, Spain
b
Clinical Pathology Service, La Fe University Hospital, Valencia, Spain
c
Division of Cardiovascular Sciences, Laboratory of Thrombosis and Haemostasis, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
abstractarticle info
Article history:
Received 13 May 2011
Received in revised form 14 July 2011
Accepted 1 August 2011
Available online 7 September 2011
Keywords:
activated protein C
endothelial protein C receptor
factor VII
venous thrombosis
myocardial infarction
The protein C anticoagulant pathway plays a crucial role as a regulator of the blood clotting cascade. Protein C
is activated on the vascular endothelial cell membrane by the thrombin-thrombomodulin complex. The
endothelial protein C receptor binds protein C and further enhances protein C activation. Once formed,
activated protein C down-regulates thrombin formation by inactivating factors Va and VIIIa and exerts
cytoprotective effects through endothelial protein C receptor binding. An adequate generation of activated
protein C depends on the precise assembly, on the surface of the endothelial cells, of thrombin,
thrombomodulin, protein C, and endothelial protein C receptor. Therefore, any change in the efciency of
this assembly may cause a reduction or increase in activated protein C generation and modulate the risk of
thrombosis. This review highlights the role of the endothelial protein C receptor in disease and discusses the
association of its mutations with the risk of thrombosis.
© 2011 Elsevier Ltd. All rights reserved.
Contents
Introduction ................................................................ 410
EPCR structure ............................................................... 411
Soluble EPCR and disease .......................................................... 411
EPCR and FVII/FVIIa ............................................................. 412
Anti-EPCR autoantibodies .......................................................... 412
EPCR polymorphisms and thrombosis .................................................... 412
EPCR and venous thromboembolism ................................................... 412
EPCR and arterial thrombosis ...................................................... 413
Summary .................................................................. 414
Conict of interest statement ........................................................ 414
Acknowledgment .............................................................. 414
References ................................................................. 414
Introduction
The protein C (PC) anticoagulant pathway plays a crucial role in
the regulation of brin formation via proteolytic inactivation of the
procoagulant cofactors Factor (F) Va and FVIIIa [13]. PC is a vitamin
K-dependent plasma glycoprotein that circulates in plasma as an
inactive zymogen, which is activated to activated PC (APC) [4] on the
surface of endothelial cells by the thrombin-thrombomodulin (TM)
complex. Another endothelial cell-specic protein, which is involved
in the PC anticoagulant pathway, is the endothelial cell PC receptor
(EPCR). EPCR binds PC to on the endothelial cell surface. This binding
Thrombosis Research 128 (2011) 410416
Abbreviations: PC, protein C; F, factor; APC, activated protein C; TM, thrombomo-
dulin; EPCR, endothelial protein C receptor; TNF-α, tumor necrosis factor-α;PROCR,
endothelial protein C receptor gene; aa, amino acid; UTR, untranslated region; sEPCR,
soluble EPCR; TF, tissue factor; PAR-1, protease-activated receptor-1; MAPK, mitogen
activated protein kinase; VTE, venous thromboembolism; SNPs, single nucleotide
polymorphisms; T, thrombin; PS, protein S.
Corresponding author at: Cen tro de Investigación, Hospita l Universitario y
Politécnico La Fe. Av. Campanar 21, 46009 Valencia, Spain. Tel.: + 34 963862797; fax:
+34 961973018.
E-mail address: medina_pil@gva.es (P. Medina).
0049-3848/$ see front matter © 2011 Elsevier Ltd. All rights reserved.
doi:10.1016/j.thromres.2011.08.001
Contents lists available at ScienceDirect
Thrombosis Research
journal homepage: www.elsevier.com/locate/thromres
enhances the rate of PC activation by the thrombin-TM complex by 5-
to 20-fold [5] by decreasing the K
m
of PC for its activation by the
thrombin-TM complex. The relevance of the anticoagulant role of
EPCR in vivo has been well documented: EPCR blockade in baboons
notably reduced the amount of APC generated upon thrombin
administration [6], and an anti-EPCR monoclonal antibody accelerat-
ed thrombus development in a murine model of thrombosis [7].
TM is uniformly distributed on all endothelial cells, which results
in relatively low TM concentrations in large vessels. This would result
in inefcient activation of PC in these vessels [8]. This effect is
counterbalanced by EPCR, which is highly expressed on the
endothelium of large vessels and is present in trace levels in most
capillary beds [9]. This effective location may ensure efcacious PC
activation on the surface of large vessels where the ratio of the blood
volume to the surface area is high.
Once activated, APC may either dissociate from EPCR, bind to its
cofactor protein S [1,3], and exhibit its anticoagulant functions, or it
may remain bound to EPCR and display cell-signaling cytoprotective
activities [1016] (Fig. 1a).
The clinical relevance of the PC pathway is evident from reports
showing a clear association between deciencies of PC [2,17,18] and
protein S [1921] or reduced APC levels [2224] with thrombosis. In
fact, the deciencies of PC and protein S or the FV Leiden mutation are
present in more than 50% of patients with inherited thrombophilia
[25].
EPCR is widely expressed on the surface of the endothelium of
large vessels. Inammatory stimuli-like tumor necrosis factor-α
(TNF-α) or an atherosclerotic setting have been shown to reduce its
expression [26,27]. The presence of EPCR on the surface of other cells
has also been reported: trophoblasts, monocytes, neutrophils and
eosinophils [28,29], brain endothelial cells [30], lymphocytes [31],
osteoblasts [32], gastric epithelial cells [33], chondrocytes [34],
tenocytes [35], epidermal keratinocytes [36], and human vascular
smooth muscle cells [37]. More recently, EPCR was reported to be
expressed in murine CD8
+
dendritic cells [38], hematopoietic stem
cells [39], and cardiomyocytes [40].
EPCR structure
The human EPCR gene (PROCR) spans approximately 8 kb, is
located on chromosome 20 at position q11.2, and consists of 4 exons
[26,4143]. Exon 1 [138 bp; 1 to 24 amino acids (aa)] encodes the 5-
untranslated region (UTR), the signal peptide, and 7 additional
residues. Exons 2 (252 bp; 24 to 108 aa) and 3 (279 bp; 108 to 201
aa) encode most of the extracellular region of EPCR. Exon 4 (659 bp;
201 to 238 aa) encodes an additional 10 residues of the extracellular
region of EPCR, the transmembrane domain, the cytoplasmic tail, and
the 3UTR [42].
EPCR is a 46-kD type I transmembrane protein, constitutively
expressed on the luminal surface of endothelium by endothelial cells,
and is structurally similar to the major histocompatibility class 1/CD1
family proteins involved in the immune and inammation responses
[26]. The crystal structure of EPCR showed that a tightly bound
phospholipid resides in the groove typically involved in antigen
presentation, and its extraction results in loss of PC binding, which can
be restored by lipid reconstitution [44]. The crystal structure, solved
alone and in complex with the phospholipid-binding domain of
protein C, revealed that most of the residues contacting the lipid in
EPCR are identical to or highly conserved in CD1d, which may help to
further understand the role of EPCR in immune regulation. Finally, it
showed that the PC binding site is outside this conserved groove and
is distal from the membrane-spanning domain [44].
The EPCR protein is comprised of 238 aa with a signal sequence at
the amino-terminal end (17 aa); an extracellular domain composed of
2 alpha domains (α1 and α2), 4 potential N-glycosylation sites, and 3
Cys residues; a transmembrane domain near the carboxy-terminal
(25 aa); and a short cytosolic domain (3 aa) [41,45,46].
Soluble EPCR and disease
A soluble form of EPCR (sEPCR), which lacks the transmembrane
and cytoplasmic tail domain, is present in normal human plasma [47].
sEPCR is generated in vitro through proteolytic cleavage by metallo-
protease activity inducible by thrombin and other inammatory
mediators, a process called shedding. This metalloprotease has been
identied as the TNF-αconverting enzyme or ADAM17 [48] and
cleaves EPCR between aa's 192 and 200. Moreover, ADAM17 promotes
the release of pro-inammatory and adhesion molecules [48,49], and
TNF-αsignicantly decreases the expression of EPCR and TM in
several human endothelial cells [50]. A variety of mediators, including
interleukin-1, hydrogen peroxide, phorbol esters, and thrombin
dramatically increase EPCR shedding from the endothelium [51,52].
Like the membrane-bound form, sEPCR binds PC and APC with similar
afnity. However, its binding to APC inhibits its anticoagulant and
anti-inammatory properties, and its binding to PC prevents PC
activation by the thrombin-TM complex [53]. This would suggest that
sEPCR may display procoagulant activity. sEPCR may also bind to
activated neutrophils in a process that involves proteinase-3 and Mac-
1. There is a controversy whether or not this interaction exerts an
anti-inammatory effect. On the one hand, it was proposed that
binding of sEPCR to proteinase-3/Mac-1 may reduce leukocyte-
Fig. 1. Endothelial protein C receptor (EPCR) functions. 1a. Protein C (PC) is activated by
the thrombin (T)-thrombomodulin (TM)-EPCR ternary complex on the endothelial cell
surface. Upon activation, activated PC (APC) may dissociate from the complex, bind to
protein S (PS) and display its anticoagulant functions, or may remain bound to EPCR
and display its cytoprotective activities through cell signaling mechanisms, most of
them via protease-activated receptor-1 (PAR-1). 1b. Factor (F) VII and FVIIa also bind to
EPCR. Binding of FVII prevents the FXa-dependent generation of FVIIa, which may
represent a new anticoagulant role for EPCR. On the other hand, FVIIa, upon binding to
EPCR on endothelial cells, activates PAR1-mediated cell signaling and provides a
barrier-protective effect.
411S. Navarro et al. / Thrombosis Research 128 (2011) 410416
endothelial cells interactions, thus modulating inammation and
preventing endothelium damage [54,55]. However, it has also been
reported that proteinase-3 is able to proteolyze EPCR, thus triggering
an additional mechanism by which anticoagulant and cell protective
pathways may be down-regulated during inammation [56]. In sum,
the physiological signicance of sEPCR in vivo is not well known.
sEPCR levels showed a bimodal distribution in a healthy
population. Approximately 15% to 20% of the general population has
plasma sEPCR levels between 200 and 800 ng/mL, whereas the
remainder have levels below 200 ng/mL [57]. Plasma sEPCR levels
responded to anticoagulant treatment, suggesting that sEPCR may be
a marker for a hypercoagulable state [58].
sEPCR levels increase in a wide variety of pathophysiological
conditions, and may reect endothelial dysfunction, and contribute to
a procoagulant phenotype and increased risk of thrombosis. Accordingly,
sEPCR levels increase in patients with sepsis, a disorder that results from
a complex dysregulation of hemostatic mechanisms, with activation of
procoagulant pathways and impairment of the brinolytic system and
natural anticoagulant pathways, especially the PC pathway. In fact, as
previously explained, the anti-inammatory properties of APC seem to
be benecial in the treatment of sepsis [59,60]. sEPCR level also increases
in systemic lupus erythematosus [61,62], a potentially fatal autoimmune
disease affecting multiple organ systems, and it is associated with
thrombotic manifestations, inammation, and widespread activation of
the vascular endothelium.
It has been shown that 5080% of plasma sEPCR variations are
under genetic control [6365] and that most subjects with elevated
sEPCR levels carry the H3 haplotype, one of the EPCR haplotypes
described, but this will be discussed in detail later.
Recently, a new mechanism to generate sEPCR has been reported,
consisting of the alternative mRNA splicing that generates a truncated
EPCR mRNA lacking the sequence encoding the transmembrane and
intra-cytoplasmic domains [66]. The resulting protein is not able to
anchor to the membrane and, as a consequence, it is secreted and can
be detected in plasma as sEPCR. The truncated sEPCR can bind PC and
APC, and its generation is particularly efcient in H3-carrying subjects
[66,67], and their endothelial cells might therefore be able to produce
a soluble receptor that might compete with the membrane-bound
EPCR for APC binding.
EPCR and FVII/FVIIa
It has been reported that EPCR may exhibit PC-independent
anticoagulant activities. FVIIa is a serine protease that binds to tissue
factor (TF) and initiates the coagulation cascade. The Gla domain of
FVIIa exhibits an important degree of homology with the Gla domain
of PC, and all the residues directly involved in the binding of PC to
EPCR are conserved in FVII [6870]. A binding analysis has shown that
FVII, FVIIa, PC, and APC bind to EPCR with similar afnity[7072],
suggesting that the interaction of FVII/FVIIa with EPCR on endothelial
cells may inuence the activation of PC and APC-mediated cell
signaling. In addition, EPCR mediates the internalization of FVIIa
bound to it on the cell surface, indicating that it may play a role in
FVIIa clearance [71,73,74].
More recently, it has been shown that endothelial cells may down-
regulate the FXa-dependent generation of FVIIa through EPCR binding
[75]. This regulation is probably made by moving FVII from
phosphatidylserine-rich regions, suggesting a new anticoagulant
role for EPCR. Moreover, FVIIa, upon binding to the EPCR on the
endothelial cell surface, activates the endogenous protease-activated
receptor-1 (PAR-1) and induces PAR-1-mediated p44/42 mitogen-
activated protein kinase (MAPK) activation, thus providing a barrier-
protective effect [76] (Fig. 1b).
It must be noted that, very recently, FXa has been shown to bind to
EPCR, which opens new venues in serine-protease signaling [77].
Anti-EPCR autoantibodies
The presence of high titers of anti-EPCR autoantibodies has been
described in patients with antiphospholipid syndrome, fetal death
[78], deep vein thrombosis in the general population [79], and women
with acute myocardial infarction [80]. Also, a case report described a
patient with stroke and massive cutaneous necrosis who had high
titers of anti-EPCR autoantibodies [81]. Two of these autoantibodies
blocked the binding of PC to EPCR, and thus inhibited the generation
of APC on the endothelium. [78]. For this reason, anti-EPCR
autoantibodies may play a causative role in thrombosis, since low
APC levels have been associated with an increased risk of venous and
arterial thrombosis [24,82].
There is an association between elevated levels of the anti-EPCR
autoantibodies, high levels of coagulation activity estimated by D-
dimer levels, and levels of sEPCR [79], which could be related with
endothelial injury induced by these autoantibodies. Anyhow, the
mechanisms by which anti-EPCR autoantibodies confer a risk for
thrombotic events are not fully understood.
EPCR polymorphisms and thrombosis
As mentioned before, normal APC generation depends on the
precise assembly of thrombin and PC to their respective receptors, TM
and EPCR, on the surface of endothelial cells. Any change in the
efciency of this coupling may cause altered APC generation and a
modication in the risk of thrombosis. In fact, several mutations and
polymorphisms have been reported in the EPCR gene, some of them
associated with the risk of venous or arterial thrombosis.
EPCR and venous thromboembolism
Merati et al. [83] described a 23-bp insertion in exon 3. This
mutation duplicates the preceding 23 bases and results in a STOP
codon downstream from the insertion point [84]. Although statistical
analysis did not reveal a signicant association between the mutation
and the risk of thrombosis, expression studies in mammalian cells
showed that the truncated protein is not localized on the cell surface,
cannot be secreted in the culture medium, and does not bind APC,
suggesting that the insertion is a risk factor for arterial and venous
thrombosis. However, given its low population frequency (b1%)
[85,86], it is difcult to assess the effect of this mutation on the risk of
venous and arterial thrombosis. In fact, after combining data from 7
studies that genotyped a total of 2,508 venous thromboembolism
(VTE) patients and 2,617 controls [8389], the prevalence of the
mutation was 0.48% and 0.38%, respectively.
Hermida et al. described the point mutation C2769T, which causes
a substitution of Arg to Cys at position 96 in the mature protein [90].In
vitro expression and characterization studies of the EPCR R96C variant
revealed no biochemical differences with the wild-type counterpart,
supporting no role of this mutation in VTE [90].
Up to 4 haplotypes of EPCR have been reported [6365]: H1, H2,
H3, and H4; 3 of which contain 1 or more single-nucleotide
polymorphisms (SNPs) that are haplotype-specic(Fig. 2), while H2
contains the common allele of each SNP.
The H1 haplotype, tagged by the rare allele of 4678G/C (rs9574),
has been associated with increased circulating APC levels [64] and a
reduced risk of VTE in 2 independent studies [64,91]. H1 also reduced
the risk of thrombosis in carriers of FV Leiden [92]. In patients with the
FV Leiden mutation, the mean age at the rst thrombosis was
signicantly higher in H1H1 propositi than in non-carriers of the H1
haplotype. In contrast, 2 other groups found no association of the H1
haplotype with the risk of thrombosis [63,65]. A possible explanation
for the protective effect of EPCR H1 against VTE would be its
association with increased APC levels. In fact, it has been described
412 S. Navarro et al. / Thrombosis Research 128 (2011) 410416
that a low level of APC in plasma is a strong, prevalent, independent
risk factor for VTE [93,94].
EPCR is essential for normal embryonic development and plays a
key role in preventing thrombosis at the maternal-embryonic
interface [95]. Recently, it has been shown that the EPCR H1 seems
to protect against recurrent pregnancy loss, particularly in carriers of
FV Leiden [96], but not in the absence of this thrombophilic defect
[97]. Presently it is unknown which SNP in EPCR H1 is responsible for
the reported protective effect. The H1 haplotype contains 10 specic
alleles, the 1451T (rs2069943), 1541A (rs2069944), 1880C
(rs2069945), 2532C (rs2069948), 2897A (rs945960), 3424C
(rs871480), 3997C (rs2069952), 4678C (rs9574), 5632G
(rs1415773), and 5663A (rs1415774) (Fig. 2). Therefore, any of
these nucleotides may be responsible for the observed association of
H1 with increased levels of plasma APC and reduced risk of VTE
[64,92]. Further studies are required to identify which polymorphism/
s is responsible for the observed associations, although some efforts
are being made [98].
The H3 haplotype, tagged bythe rare allele of 4600A/G (rs867186),is
associated with increased plasma levels of sEPCR, but its association
with the risk of VTE is controversial [6365,99].Onestudy[63] reported
that carriers of the H3 haplotype have an increased risk of VTE in men
but not in women, whereas others [64,65,91,99] did not nd a
signicant association between EPCR H3 and the risk of thrombosis.
The presence of EPCR H3 and concomitant elevated sEPCR plasma levels
in carriers of the 2 dysfunctional PC variants, Arg-1Cys and Arg-1Leu, is
associated with severe thrombotic manifestations [100]. In addition, it
has been observed that EPCR H3 increases the risk of VTE in carriers of
the prothrombin 20210A mutation, probably due to its association with
increased sEPCR levels [101]. Furthermore, H3 carriers experienced the
rst VTE episode at a young age [101]. Recently, the maternal EPCR H3
allele has been found to be a mild risk factor for iliac VTE during
pregnancy and puerperium [102]. Overall, the thrombogenicity of the
EPCR H3, evenif weak, does not seemanecdotal. First,the high incidence
of this polymorphism in the Caucasian Mediterranean population
(21.4%) and the fact that it may potentiate the prothrombotic effect of
other thrombophilias, like the prothrombin 20210A allele [101],
suggeststhat its contributiontowards a VTE event maynot be negligible.
Second, the H3 haplotype may be a risk factor not only for VTE but also
for pregnancy loss [103,104].
It has been suggested that high sEPCR levels associated with the H3
haplotype could be the mechanism that increases the thrombotic risk,
even if sEPCR levels have not been deeply studied in this regard and
some contradictory results exist [65,101]. Regarding the SNPs that
comprise the H3 haplotype, the 4600G (219Gly) allele arises as the
more obvious candidate responsible for the association of the H3
haplotype with increased sEPCR levels, in view of the fact that the
cleavage of the EPCR anchored in the endothelial cell membrane to
generate sEPCR occurs within the region of the protein encoded by
exon 4, near the 4,600 position. The 4600G variant predicts a
conformational change in the protein due to the Ser 219 to Gly
substitution, which may render an EPCR more susceptible to cleavage
by metalloproteases such as ADAM17 [48,63,64]. This hypothesis has
been supported by 2 independent studies [105,106]. And other
mechanism that could link the H3 haplotype and high levels of sEPCR
is the alternative splicing previously explained [66]. The mean
physiological sEPCR concentration is about 3 nM, well below the
concentration of circulating PC, which is about 70 nM, and the Kd of
the EPCR-PC/APC interactions is about 30 nM. It has been hypothe-
sized that in individuals with markedly increased sEPCR levels due to
the H3 haplotype (between 6 and 20 nM), the local concentration at
the endothelial cell surface may approach or exceed the Kd value of
the PC interaction, resulting in a decreased APC generation and
inhibition of the APC generated [63].
Recently, a study that looked for genetic determinants for PC levels
has shown that the EPCR H3 is associated with higher levels of plasma
PC [107,108]. Additionally, the H3 haplotype has also been associated
with higher levels of FVII [109,110], which could hypothetically confer
its risk of thrombosis. An alternative explanation for the thrombo-
genicity that the H3 haplotype may induce is that the increased
shedding of EPCR could reduce the amount of EPCR at the endothelial
surface. In favor of this argument is the fact that inducing EPCR
shedding in cells bearing the H3 haplotype notably reduced their
ability to sustain PC activation as compared with non-H3 cells [106].
Finally, the EPCR H4 was reported to be associated with a slight
increase in the risk of VTE [65], although no further studies have
conrmed these results.
EPCR and arterial thrombosis
In accordance to the risk of VTE, the role of the EPCR 23-bp insertion
[83] in the risk of arterial disease is hard to ascertain mainly due to the
low prevalence of this mutation in the population. Actually, the
combination of the data from the 3 reported studies that genotyped a
total of 669 patients with myocardial infarction and 372 controls
[83,84,111] revealed prevalences of 1.20% and 0.27%, respectively.
With regard to EPCR haplotypes and arterial disease, the results are
also controversial. In the widest study performed so far, no association
between the H3 haplotype and risk of coronary heart disease, stroke,
or mortality was found [107]. In contrast, H1 was associated with
lymphoid EPCR mRNA expression and with increased risk of incident
stroke, all-cause mortality, and decreased healthy survival during
follow-up [107]. These results are in discrepancy with the other wide
study available, which showed that, surprisingly, both H1 and H3
haplotypes were associated with a reduction in the risk of premature
myocardial infarction, and that these effects were additive [112]. The
protective mechanism by which the H1 haplotype would reduce the
risk of premature myocardial infarction may be related to an increase
Fig. 2. The 4 haplotypes of the EPCR gene. Numbering according to Simmonds RE and Lane DA [42]. Circled letters correspond to specic alleles for each haplotype. Bold numbers
indicate supposed numbering for these single-nucleotide polymorphisms according to this numbering since the sequence described by Simmonds RE and Lane DA do not reach these
positions.
413S. Navarro et al. / Thrombosis Research 128 (2011) 410416
in the circulating APC levels, which are characteristic of the carriers of
the H1 haplotype [64,92]. The protection conferred by the H3
haplotype would be more difcult to explain as increased sEPCR
levels would promote neither the activation of protein C nor the APC-
dependent anticoagulant and cytoprotective actions. The rest of the
studies available are not wide enough to withdraw powerful
conclusions. One study [105] suggested that individuals carrying
one EPCR H3 allele were protected from myocardial infarction,
provided that they were neither diabetic nor showing symptoms of
metabolic syndrome, since in patients with these pathologies, the H3
allele would increase the risk. Finally, there is another study which
suggested that elevated sEPCR levels, linked to the H3 haplotype,
might increase the risk of stroke at pediatric age [113].
Summary
EPCR plays an important role in the regulation of coagulation and
in the APC-evoked cell signaling. Whether or not functional poly-
morphisms in the EPCR gene increase or decrease the risk of
thrombosis, alone or in combination with other risk factors, has not
been undoubtedly demonstrated and requires further investigation.
Conict of interest statement
The authors state that they have no conict of interest.
Acknowledgment
This work was supported by the PN de I+ D + I 200820011 of
Instituto de Salud Carlos III-Fondo de Investigación Sanitaria and FEDER
(PS09/00610, PI10/01432 and Red Temática de Investigación RECAVA
RD06/0014/0004 and RD06/0014/0008), Conselleria de Educación-
Generalitat Valenciana (Prometeo/2011/027), and Fundación para la
Investigación del Hospital Universitario La Fe (20070185), Spain. Pilar
Medina is a Miguel Servet Researcher (ISCIII CP09/00065).
References
[1] Walker FJ. Regulation of activated protein C by a new protein. A possible function
for bovine protein S. J Biol Chem 1980;255:55214.
[2] Grifn JH, Evatt B, Zimmerman TS, Kleiss AJ, Wideman C. Deciency of protein C
in congenital thrombotic disease. J Clin Invest 1981;68:13703.
[3] Fulcher CA, Gardiner JE, Grifn JH, Zimmerman TS. Proteolytic inactivation of
human factor VIII procoagulant protein by activated human protein C and its
analogy with factor V. Blood 1984;63:4869.
[4] Grifn JH, Fernandez JA, Gale AJ, Mosnier LO. Activated protein C. J Thromb
Haemost 2007;5(Suppl 1):7380.
[5] Stearns-Kuros awa DJ, Kurosawa S, Mollica JS, Ferrell GL, Esmo n CT. The
endothelial cell protein C receptor augments protein C activation by the
thrombin-thrombomodulin complex. Proc Natl Acad Sci USA 1996;93:102126.
[6] Taylor Jr FB, Peer GT, Lockhart MS, Ferrell G, Esmon CT. Endothelial cell protein C
receptor plays an important role in protein C activation in vivo. Blood 2001;97:
16858.
[7] Centelles MN, Puy C, López-Sagaseta J, Fukudome K, Montes R, Hermida J.
Blocking endothelial protein C receptor (EPCR) accelerates thrombus develop-
ment in vivo. Thromb Haemost 2010;103:123944.
[8] Esmon CT. The roles of protein C and thrombomodulin in the regulation of blood
coagulation. J Biol Chem 1989;264:47436.
[9] Laszik Z, Mitro A, Taylor FB, Ferrell G, Esmon CT. Human protein C receptor is
present primarily on endothelium of large blood vessels: implications for the
control of the protein C pathway. Circulation 1997;96:363340.
[10] España F, Medina P, Navarro S, Zorio E, Estellés A, Aznar J. The multifunctional
protein C system. Curr Med Chem Cardiovasc Hematol Agents 2005;3:11931.
[11] Riewald M, Petrovan RJ, Donner A, Mueller BM, Ruf W. Activation of endothelial
cell protease activated receptor 1 by the protein C pathway. Science 2002;296:
18802.
[12] Coughlin SR. Protease-activated receptors in hemosta sis, thrombosis and
vascular biology. J Thromb Haemost 2005;3:180014.
[13] Bae JS, Yang L, Rezaie AR. Receptors of the protein C activation and activated
protein C signaling pathways are colocalized in lipid rafts of endothelial cells.
Proc Natl Acad Sci USA 2007;104:286772.
[14] Niessen F, Furlan-Freguia C, Fernandez JA, Mosnier LO, Castellino FJ, Weiler H,
et al. Endogenous EPCR/aPC-PAR1 signaling prevents inammation-induced
vascular leakage and lethality. Blood 2009;113:285966.
[15] Bezuhly M, Cullen R, Esmon CT, Morris SF, West KA, Johnston B, et al. Role of
activated protein C and its receptor in inhibition of tumor metastasis. Blood
2009;113:33714.
[16] Cheng T, Liu D, Grifn JH, Fernández JA, Castellino F, Rosen ED, et al. Activated
protein C blocks p53-mediated apoptosis in ischemic human brain endothelium
and is neuroprotective. Nat Med 2003;9:33842.
[17] Bertina RM, Broekmans AW, van der Linden IK, Mertens K. Protein C deciency in
a Dutch family with thrombotic disease. Thromb Haemost 1982;48:15.
[18] Estellés A, García-Plaza I, Dasí A, Aznar J, Duart M, Sanz G, et al. Severe inherited
"homozygous" protein C deciency in a newborn infant. Thromb Haemost
1984;52:536.
[19] Schwarz HP, Fischer M, Hopmeier P, Batard MA, Grifn JH. Plasma protein S
deciency in familial thrombotic disease. Blood 1984;64:1297300.
[20] Comp PC, Nixon RR, Cooper MR, Esmon CT. Familial protein S deciency is
associated with recurrent thrombosis. J Clin Invest 1984;74:20828.
[21] Engesser L, Broekmans AW, Briët E, Brommer EJ, Bertina RM. Hereditary protein S
deciency: clinical manifestations. Ann Intern Med 1987;106:67782.
[22] España F, Zorio E, Medina P, Osa A, Estellés A, Palencia M, et al. Low levels of
circulating activated protein C are a risk factor for myocardial infarction.
Pathophysiol Haemost Thromb 2002;32(Suppl 2):61.
[23] España F, Medina P, Navarro S, Estellés A, Aznar J. Inherited abnormalities in the
protein C activation pathway. Pathophysiol Haemost Thromb 2002;32:2414.
[24] Zorio E, Navarro S, Medina P, Estellés A, Osa A, Rueda J, et al. Circulating activated
protein C is reduced in young survivors of myocardial infarction and inversely
correlates with the severity of coronary lesions. J Thromb Haemost 2006;4:
15306.
[25] Bertina RM. Molecular risk factors for thrombosis. Thromb Haemost 1999;82:
6019.
[26] Fukudome K, Esmon CT. Identication, cloning, and regulation of a novel
endothelial cell protein C/activated protein C receptor. J Biol Chem 1994;269:
2648691.
[27] Laszik ZG, Zhou XJ, Ferrell GL, Silva FG, Esmon CT. Down-regulation of endothelial
expression of endothelial cell protein C receptor and thrombomodulin in
coronary atherosclerosis. Am J Pathol 2001;159:797802.
[28] Galligan L, Livingstone W, Volkov Y, Hokamp K, Murphy C, Lawler M, et al.
Characterization of protein C receptor expression in monocytes. Br J Haematol
2001;115:40814.
[29] Sturn DH, Kaneider NC, Feistritzer C, Djanani A, Fukudome K, Wiedermann CJ.
Expression and function of the endo thelial protein C receptor in human
neutrophils. Blood 2003;102:1499505.
[30] Mosnier LO, Grifn JH. Protein C anticoagulant activity in relation to anti-
inammatory and anti-apoptotic activities. Front Biosci 2006;11:238199.
[31] Feistritzer C, Mosheimer BA, Sturn DH, Riewald M, Patsch JR, Wiedermann CJ.
Endothelial protein C receptor-dependent inhibition of migration of human
lymphocytes by protein C involves epidermal growth factor receptor. J Immunol
2006;176:101925.
[32] Kurata T, Hayashi T, Yoshikawa T, Okamoto T, Yoshida K, Iino T, et al. Activated
protein C stimulates osteoblast proliferation via endothelial protein C receptor.
Thromb Res 2010;125:18491.
[33] Nakamura M, Gabazza EC, Imoto I, Yano Y, Taguchi O, Horiki N, et al. Anti-
inammatory effect of activated protein C in gastric epithelial cells. J Thromb
Haemost 2005;3:27219.
[34] Jackson MT, Smith MM, Smith SM, Jackson CJ, Xue M, Little CB. Activation of
cartilage matrix metalloproteinases by activated protein C. Arthritis Rheum
2009;60:78091.
[35] Xue M, Smith MM, Little CB, Sambrook P, March L, Jackson CJ. Activated protein C
mediates a healing phenotype in cultured tenocytes. J Cell Mol Med 2009;13:
74957.
[36] Xue M, Campbell D, Jackson CJ. Protein C is an autocrine growth factor for human
skin keratinocytes. J Biol Chem 2007;282:136106.
[37] Bretschneider E, Uzonyi B, Weber AA, Fischer JW, Pape R, Lotzer K, et al. Human
vascular smooth muscle cells express functionally active endothelial cell protein
C receptor. Circ Res 2007;100:25562.
[38] Kerschen E, Hernández I, Zogg M, Jia S, Hessner MJ, Fernández JA, et al. Activated
protein C targets CD8+ dendritic cells to reduce the mortality of endotoxemia in
mice. J Clin Invest 2010;120:316778.
[39] Balazs AB, Fabian AJ, Esmon CT, Mulligan RC. Endothelial protein C receptor
(CD201) explicitly identies hematopoietic stem cells in murine bone marrow.
Blood 2006;107:231721.
[40] Wang J, Yang L, Rezaie AR, Li J. Activated protein C protects against myocardial
ischemic/reperfusion injury through AMP-activated protein kinase signaling. J
Thromb Haemost 2011;9:130817.
[41] Fukudome K, Esmon CT. Molecular cloning and expression of murine and bovine
endothelial cell protein C/activated protein C receptor (EPCR). The structural and
functional conservation in human, bovine, and murine EPCR. J Biol Chem
1995;270:55717.
[42] Simmonds RE, Lane DA. Structural and functional implications of the intron/exon
organization of the human endothelial cell protein C/activated protein C receptor
(EPCR) gene: comparison with the structure of CD1/major histocompatibility
complex alpha1 and alpha2 domains. Blood 1999;94:63241.
[43] Hayashi T, Nakamura H, Okada A, Takebayashi S, Wakita T, Yuasa H, et al.
Organization and chromosomal localization of the human endothelial protein C
receptor gene. Gene 1999;238:36773.
[44] Oganesyan V, Oganesyan N, Terzyan S, Qu D, Dauter Z, Esmon NL, et al. The crystal
structure of the endothelial protein C receptor and a bound phospholipid. J Biol
Chem 2002;277:248514.
414 S. Navarro et al. / Thrombosis Research 128 (2011) 410416
[45] Esmon CT, Gu JM, Xu J, Qu D, Stearns-Kurosawa DJ, Kurosawa S. Regulation and
functions of the protein C anticoagulant pathway. Haematologica 1999;84:3638.
[46] Esmon CT. The endothelial cell protein C receptor. Thromb Haemost 2000;83:
63943.
[47] Kurosawa S, Stearns-Kurosawa DJ, Hidari N, Esmon CT. Identication of functional
endothelialprotein C receptor in human plasma. J Clin Invest 1997;100:4118.
[48] Qu D, Wang Y, Esmon NL, Esmon CT. Regulated endothelial protein C receptor
shedding is mediated by tumor necrosis factor-alpha converting enzyme/
ADAM17. J Thromb Haemost 2007;5:395402.
[49] Peiretti F, Canault M, Morange P, Alessi MC, Nalbone G. The two sides of ADAM17
in inammation: implications in atherosclerosis and obesity. Med Sci (Paris)
2009;25:4550.
[50] Nan B, Lin P, Lumsden AB, Yao Q, Chen C. Effects of TNF-alpha and curcumin on
the expression of thrombomodulin and endothelial protein C receptor in human
endothelial cells. Thromb Res 2005;115:41726.
[51] Menschikowski M, Hagelgans A, Eisenhofer G, Siegert G. Regulation of
endothelial protein C receptor shedding by cytokines is mediated through
differential activation of MAP kinase signaling pathways. Exp Cell Res 2009;315:
267382.
[52] Menschikowski M, Hagelgans A, Tiebel O, Klinsmann L, Eisenhofer G, Siegert G.
Expression and shedding of endothelial protein C receptor in prostate cancer
cells. Cancer Cell Int 2011;11:4.
[53] Liaw PC, Neuenschwander PF, Smirnov MD, Esmon CT. Mechanisms by which
soluble endothelial cell protein C receptor modulates protein C and activated
protein C function. J Biol Chem 2000;275:544752.
[54] Kurosawa S, Esmon C, Stearns-Kurosawa D. The soluble endothelial protein C
receptor binds to activated neutro phils: involvement of proteinase-3 and
CD11b/CD18. J Immunol 2000;165:4697703.
[55] Esmon CT. The protein C pathway. Chest 2003;124:26S32S.
[56] Villegas-Méndez A, Montes R, Ambrose LR, Warrens AN, Laffan M, Lane DA.
Proteolysis of the endothelial cell protein C receptor by neutrophil proteinase 3. J
Thromb Haemost 2007;5:9808.
[57] Stearns-Kurosawa DJ, Burgin C, Parker D, Comp P, Ku rosawa S. Bimodal
distribution of soluble endothelial protein C receptor levels in healthy
populations. J Thromb Haemost 2003;1:8556.
[58] Stearns-Kurosawa DJ, Swindle K, D'Angelo A. Della Valle P, Fattorini A, Caron N,
Grimaux M, Woodhams B and Kurosawa S. Plasma levels of endothelial protein C
receptor respond to anticoagulant treatment. Blood 2002;99:52630.
[59] Menschikowski M, Hagelgans A, Hempel U, Lattke P, Ismailov I, Siegert G. On
interaction of activated protein C with human aortic smooth muscle cells
attenuating the secretory group IIA phospholipase A2 expression. Thromb Res
2008;122:6976.
[60] Bae JS, Rezaie AR. Thrombin and activated protein C inhibit the expression of
secretory group IIA phospholipase A(2) in the TNF-alpha-activated endothelial
cells by EPCR and PAR-1 dependent mechanisms. Thromb Res 2010;125:e9e15.
[61] Kurosawa S, Stearns-Kurosawa DJ, Carson CW, D'Angelo A, Della Valle P, Esmon CT.
Plasma levels of endothelial cell protein C receptor are elevated in patients with
sepsis andsystemic lupus erythematosus: lackof correlationwith thrombomodulin
suggests involvement of different pathological processes. Blood 1998;91:7257.
[62] Sesin CA, Yin X, Esmon CT, Buyon JP, Clancy RM. Shedding of endothelial protein
C receptor contributes to vasculopathy and renal injury in lupus: in vivo and in
vitro evidence. Kidney Int 2005;68:11020.
[63] Saposnik B, Reny JL, Gaussem P, Emmerich J, Aiach M, Gandrille S. A haplotype of
the EPCR gene is associated with increased plasma levels of sEPCR and is a
candidate risk factor for thrombosis. Blood 2004;103:13118.
[64] Medina P, Navarro S, Estellés A, Vayá A, Woodhams B, Mira Y, et al. Contribution
of polymorphisms in th e endothelial protein C receptor gene to solu ble
endothelial protein C receptor and circulating activated protein C levels, and
thrombotic risk. Thromb Haemost 2004;91:90511.
[65] Uitte de Willige S, Van Marion V, Rosendaal FR, Vos HL, de Visser MC, Bertina RM.
Haplotypes of the EPCR gene, plasma sEPCR levels and the risk of deep venous
thrombosis. J Thromb Haemost 2004;2:130510.
[66] Saposnik B, Lesteven E, Lokajczyk A, Esmon CT, Aiach M, Gandrille S. Alternative
mRNA is favored by the A3 haplotype of the EPCR gene PROCR and generates a
novel soluble form of EPCR in plasma. Blood 2008;111:344251.
[67] Molina E, Hermida J, López-Sagaseta J, Puy C, Montes R. The functional properties
of a truncated form of endothelial cell protein C receptor generated by alternative
splicing. Haematologica 2008;93:87884.
[68] Regan LM, Mollica JS, Rezaie AR, Esmon CT. The interaction between the
endothelial cell protein C receptor and protein C is dictated by the gamma-
carboxyglutamic acid domain of protein C. J Biol Chem 1997;272:2627984.
[69] Preston RJ, Villegas-Méndez A, Sun YH, Hermida J, Simioni P, Philippou H, et al.
Selective modulation of protein C afnity for EPCR and phospholipids by Gla
domain mutation. FEBS J 2005;272:97108.
[70] Preston RJ, Ajzner E, Razzari C, Karageorgi S, Dua S, Dahlbäck B, et al.
Multifunctional specicity of the protein C/activated protein C Gla domain. J
Biol Chem 2006;281:288507.
[71] Ghosh S, Pendurthi UR, Steinoe A, Esmon CT, Rao LV. Endothelial cell protein C
receptor acts as a cellular receptor for factor VIIa on endothelium. J Biol Chem
2007;282:1184957.
[72] López-Sagaseta J, Montes R, Puy C, Díez N, Fukudome K, Hermida J. Binding of
factor VIIa to the endothelial cell protein C receptor reduces its coagulant activity.
J Thromb Haemost 2007;5:181724.
[73] Nayak RC, Sen P, Ghosh S, Gopalakrishnan R, Esmon CT, Pendurthi UR, et al.
Endothelial cell protein C receptor cellular localization and trafcking: potential
functional implications. Blood 2009;114:197486.
[74] Pendurthi UR, Rao LV. Factor VIIa interaction with endothelial cells and
endothelial cell protein C receptor. Thromb Res 2010;125(Suppl 1):S1922.
[75] Puy C, López-Sagaseta J, Hermida J, Montes R. The endothelial cells downregulate
the generation of factor VIIa through EPCR binding. Br J Haematol 2010;149:
1117.
[76] Sen P, Gopalakrishnan R, Kothari H, Keshava S, Clark CA, Esmon CT, et al. Factor
VIIa bound to endothelial cell protein C receptor activates protease activated
receptor-1 and mediates cell signaling and barrier protection. Blood 2011;117:
3199208.
[77] Disse J, Petersen HH, Larsen KS, Persson E, Esmon N, Esmon CT, et al. The
endothelial protein C receptor supports tissue factor ternary coagulation
initiation complex signaling through protease-activated receptors. J Biol Chem
2011;286:575667.
[78] Hurtado V, Montes R, Gris JC, Bertolaccini ML, Alonso A, Martínez-González MA,
et al. Autoantibodies against EPCR are found in antiphospholipid syndrome and
are a risk factor for fetal death. Blood 2004;104:136974.
[79] van Hylckama Vlieg A, Montes R, Rosendaal FR, Hermida J. Autoantibodies
against endothelial protein C receptor and the risk of a rst deep vein thrombosis.
J Thromb Haemost 2007;5:144954.
[80] Montes R, Hurtado V, Alonso A, Foco L, Zonzin P, Mannucci PM, et al.
Autoantibodies against the endothelial receptor of protein C are associated
with acute myocardial infarction in young women. J Thromb Haemost 2005;3:
14548.
[81] Lavigne-Lissalde G, Cochery-Nouvellon E, Granier G, Quere I, Gris JC. Diffuse skin
necrosis in a patient with an anti-endothelial cell protein C receptor
autoantibody which blocks protein C activation. J Thromb Haemost 2005;3:
4135.
[82] Espana F, Vaya A, Mira Y, Medina P, Estelles A, Villa P, et al. Low level of
circulating activated protein C is a risk factor for venous thromboembolism.
Thromb Haemost 2001;86:136873.
[83] Merati G, Biguzzi E, Oganesyan N, Fetiveau R, Qu D, Bucciarelli P, et al. A 23bp
insertion in the endothelial protein C receptor (EPCR) gene in patients with
myocardial infarction and deep vein thrombosis. Supplement to the journal
Thromb Haemost 1999;507.
[84] Biguzzi E, Merati G, Liaw PC, Bucciarelli P, Oganesyan N, Qu D, et al. A 23 bp
insertion in the endothelial protein C receptor (EPCR) gene impairs EPCR
function. Thromb Haemost 2001;86:9458.
[85] von Depka M, Czwalinna A, Eisert R, Wermes C, Scharrer I, Ganser A, et al.
Prevalence of a 23 bp insertion in exon 3 of the endothelial cell protein C receptor
gene in venous thrombophilia. Thromb Haemost 2001;86:13602.
[86] Poort SR, Vos HL, Rosendaal FR, Bertina RM. The endothelial protein C receptor
(EPCR) 23 bp insert mutation and the risk of venous thrombosis. Thromb
Haemost 2002;88:1602.
[87] Akar N, Gokdemir R, Ozel D, Akar E. Endothelial cell protein C receptor (EPCR)
gene exon III, 23 bp insertion mutation in the Turkish pediatric thrombotic
patients. Thromb Haemost 2002;88:10689.
[88] Galligan L, Livingstone W, Mynett-Johnston L, Smith OP. Prevalence of the 23 bp
endothelial protein C receptor (EPCR) gene insertion in the Irish population.
Thromb Haemost 2002;87:7734.
[89] Grossmann R, Schwender S, Geisen U, Schambeck C, Merati G, Walter U. CBS
844ins68, MTHFR TT677 and EPCR 4031ins23 genotypes in patients with deep-
vein thrombosis. Thromb Res 2002;107:135.
[90] Hermida J, Hurtado V, Villegas-Méndez A, Catto AJ, Philippou H. Identication
and characterization of a natural R96C EPCR variant. J Thromb Haemost 2003;1:
18502.
[91] Pecheniuk NM, Elias DJ, Xu X, Grifn JH. Failure to validate association of gene
polymorphisms in EPCR, PAR-1, FSAP and protein S Tokushima with venous
thromboembolism among Californians of European ancestry. Thromb Haemost
2008;99:4535.
[92] Medina P, Navarro S, Estellés A, Vayá A, Bertina RM, España F. Inuence of the
4600A/G and 4678G/C polymorphisms in the endothelial protein C receptor
(EPCR) gene on the risk of venous thromboembolism in carriers of factor V
Leiden. Thromb Haemost 2005;94:38994.
[93] España F, Zuazu I, Vicente V, Estellés A, Marco P, Aznar J. Quantication of
circulating activated protein C in human plasma by immunoassaysenzyme
levels are proportional to total protein C levels. Thromb Haemost 1996;75:5661.
[94] España F, Vayá A, Mira Y, Medina P, Estellés A, Villa P, et al. Low level of
circulating activated protein C is a risk factor for venous thromboembolism.
Thromb Haemost 2001;86:136873.
[95] Gu JM, Crawley JT, Ferrell G, Zhang F, Li W, Esmon NL, et al. Disruption of the
endothelial cell protein C receptor gene in mice causes placental thrombosis and
early embryonic lethality. J Biol Chem 2002;277:4333543.
[96] Hopmeier P, Puehr inger H, van Trots enburg M, Atamaniuk J, Oberkanins C,
Dossenbach-Glaninger A. Association of endothelial protein C receptor
haplotypes, factor V Leiden and recurrent rst trimester pregnancy loss. Clin
Biochem 2008;41:10224.
[97] Kaare M, Ulander VM, Painter JN, Ahvenainen T, Kaaja R, Aittomaki K. Variations
in the thrombomodulin and endothelial protein C receptor genes in couples with
recurrent miscarriage. Hum Reprod 2007;22:8648.
[98] Medina P, España F, Vos HL, Bertina RM. Functional analysis of SNPs specic for
haplotype 1 (H1) of the gene coding for the Endothelial Protein C Receptor
(EPCR). J Thromb Haemost 2007;5(Suppl 2):P-M-083.
[99] Yamagishi K, Cushman M, Heckbert SR, Tsai MY, Folsom AR. Lack of association of
soluble endothelial protein C receptor and PROCR 6936A/G polymorphism with
the risk of venous thromboembolism in a prospective study. Br J Haematol
2009;145:2216.
415S. Navarro et al. / Thrombosis Research 128 (2011) 410416
[100] SimioniP, Morboeuf O, Tognin G, GavassoS, Tormene D, Woodhams B, et al. Soluble
endothelial protein C receptor (sEPCR) levels and venous thromboembolism in
carriers of two dysfunctional protein C variants. Thromb Res 2006;117:5238.
[101] Navarro S, Medina P, Mira Y, Estellés A, Villa P, Ferrando F, et al. Haplotypes of the
EPCR gene, prothrombin levels, and the risk of venous thrombosis in carriers of
the prothrombin G20210A mutation. Haematologica 2008;93:88591.
[102] Galanaud JP, Cochery-Nouvellon E, Alonso S, Chauleur C, Mercier E, Lissalde-
Lavigne G, et al. Paternal endothelial protein C receptor 219Gly variant as a mild
and limited risk factor for deep vein thrombosis during pregnancy. J Thromb
Haemost 2010;8:70713.
[103] Cochery-Nouvellon E, Chauleur C, Demattei C, Mercier E, Fabbro-Peray P, Mares
P, et al. The A6936G polymorphism of the endothelial protein C receptor gene is
associated with the risk of unexplained foetal loss in Mediterranean European
couples. Thromb Haemost 2009;102:65667.
[104] Lavigne-Lissalde G, Cochery-Nouvellon E, Mercier E, Mares P, Gris JC. High
plasma levels of endothelial protein C receptor are associated with the risk of
unexplained fetal death. J Thromb Haemost 2005;3:3935.
[105] IrelandH, KonstantoulasCJ, Cooper JA, Hawe E,Humphries SE, MatherH, et al. EPCR
Ser219Gly:elevated sEPCR,prothrombinF1 + 2, riskfor coronary heartdisease, and
increased sEPCR shedding in vitro. Atherosclerosis 2005;183:28392.
[106] Qu D, Wang Y, Song Y, Esmon NL, Esmon CT. The Ser219NGly dimorphism of the
endothelial protein C receptor contributes to the higher soluble protein levels
observed in individuals with the A3 haplotype. J Thromb Haemost 2006;4:22935.
[107] Reiner AP,Carty CL, Jenny NS, Nievergelt C, Cushman M, Stearns-KurosawaDJ, et al.
PROC, PROCR and PROS1 polymorphisms, plasma anticoagulant phenotypes, and
risk of cardiovascular disease and mortality in older adults: the Cardiovascular
Health Study. J Thromb Haemost 2008;6:162532.
[108] Tang W, Basu S, Kong X, Pankow JS, Aleksic N, Tan A, et al. Genome-wide
association study identies novel loci for plasma levels of protein C: the ARIC
study. Blood 2010;116:50326.
[109] IrelandHA, Cooper JA, DrenosF, Acharya J, MitchellJP, Bauer KA,et al. FVII, FVIIa,and
downstream markers of extrinsic pathway activation differ by EPCR Ser219Gly
variant in healthy men. Arterioscler Thromb Vasc Biol 2009;29:196874.
[110] Smith NL, Chen MH, Dehghan A, Strachan DP, Basu S, Soranzo N, et al. Novel
associations of multiple genetic loci with plasma levels of factor VII, factor VIII,
and von Willebrand factor: The CHARGE (Cohorts for Heart and Aging Research
in Genome Epidemiology) Consortium. Circulation 2010;121:138292.
[111] van de Water NS, French JK, McDowell J, Browett PJ. The endothelial protein C
receptor (EPCR) 23 bp insert in patients with myocardial infarction. Thromb
Haemost 2001;85:74951.
[112] Medina P, Nav arro S, Corra l J, Zorio E, Roldan V, Estell és A, et al. Endothelial
protein C receptor polymorphisms and risk of myocardial infarction.
Haematologica 2008;93:135863.
[113] Ulu A, Gunal D, Tiras S, Egin Y, Deda G, Akar N. EPCR gene A3 haplotype and
elevated soluble endothelial protein C receptor (sEPCR) levels in Turkish
pediatric stroke patients. Thromb Res 2007;120:4752.
416 S. Navarro et al. / Thrombosis Research 128 (2011) 410416
... Additionally, pre-treatment of samples with phorbol-12-myristate 13-acetate (PMA), TNFα, IL-1β, or LPS induces EPCR shedding ( Figure 2) [60][61][62][63]. sEPCR does not have the transmembrane and cytoplasmic tail domain of mEPCR [64], but it can bind both PC and APC with an affinity similar to mEPCR. Binding PC to sEPCR rather than mEPCR blocks surface interactions with negatively charged phospholipids that are required for the efficient inactivation of factor (F)V and FVIIIa. ...
... High plasma sEPCR can result in a low mEPCR level and reduced APC activity, leading to a pro-thrombotic state in the body [68]. Plasma sEPCR, therefore, has the potential as a marker for hypercoagulable states [64,69]. ...
... TACE cleavage is regulated by Polo-like kinase 2 (PLK2), mitogen-activated protein kinases (MAPKs), and protein kinase C (PKC) [91] and can be induced by pre-treatment of cells with PMA, TNF-α, or IL-1β [60][61][62][63]. Once cleaved, the mEPCR and sEPCR will compete for ligand binding, making fewer mEPCR-ligand functions realised [64]. H1 favours mEPCR compared to the common allele of H2 due to decreased TACE cleavage. ...
Article
Full-text available
Endothelial Protein C Receptor (EPCR) is a key regulator of the activated protein C anti-coagulation pathway due to its role in the binding and activation of this protein. EPCR also binds to other ligands such as Factor VII and X, γδ T-cells, plasmodium falciparum erythrocyte membrane protein 1, and Secretory group V Phospholipases A2, facilitating ligand-specific functions. The functions of EPCR can also be regulated by soluble (s)EPCR that competes for the binding sites of membrane-bound (m)EPCR. sEPCR is created when mEPCR is shed from the cell surface. The propensity of shedding alters depending on the genetic haplotype of the EPCR gene that an individual may possess. EPCR plays an active role in normal homeostasis, anti-coagulation pathways, inflammation, and cell stemness. Due to these properties, EPCR is considered a potential effector/mediator of inflammatory diseases. Rheumatic diseases such as rheumatoid arthritis and systemic lupus erythematosus are autoimmune/inflammatory conditions that are associated with elevated EPCR levels and disease activity, potentially driven by EPCR. This review highlights the functions of EPCR and its contribution to rheumatic diseases.
... This ligand binding type of EPCR is discernible in plasma and restrains both PC activation and activated PC (APC) anticoagulant effect. This would recommend that sEPCR may display a procoagulant activity and might be utilized as a potential clinical marker for a hypercoagulable state [7]. ...
... Elevated sEPCR levels are found in patients with systemic inflammatory diseases such as sepsis and systemic lupus erythematosus, and conditions related to considerable thrombin formation and coagulopathy [7,8]. ...
... Its activation occurs with the involvement of the endothelial cell protein C receptor (EPCR), located mainly on vascular endothelial cells [25]. Recent studies have shown that the endothelial cell protein C receptor plays an important role not only in coagulation and fibrinolysis but also in many important signalling pathways and pathophysiological processes [26]. This receptor has been shown to be involved in many immune reactions, as well as inflammation, apoptosis and cytoprotection [27]. ...
Article
Full-text available
Background: Coronary artery disease is caused by changes in the coronary arteries due to the atherosclerotic process and thrombotic changes. A very important role in the development of the atherosclerotic process in the coronary vessels is played by the inflammatory process and the immune response. Due to the important role of lipids and the coagulation process in the atherosclerotic process, research has also focused on genes affecting lipid metabolism and the coagulation system. Lipoprotein lipase (LPL) is an enzyme that metabolises lipids, hydrolysing triglycerides to produce free fatty acids and glycerol. Protein C (PC) is an essential component of coagulation and fibrinolysis. It is activated on the endothelial surface by the membrane-bound thrombin-thrombomodulin complex. Platelet-derived growth factor (PDGF) has a number of important functions in processes related to fibroblast and smooth muscle cell function. Due to their influence on lipid metabolism and coagulation processes, LPL, PROCR (endothelial cell protein C receptor) and PDGF may affect the atherosclerotic process and, thus, the risk of coronary heart disease. The aim of the study was to examine the associations between the LPL rs264, PROCR rs867186 and PDGF rs974819 gene polymorphisms and the risk of unstable angina and selected clinical parameters. Methods: The study included 232 patients with unstable angina and 144 healthy subjects as the control group. Genotyping was performed using real-time PCR. Results: There were no statistically significant differences in the distribution of the polymorphisms tested between the patients with unstable angina and the control subjects. The results showed associations between the PROCR rs867186 and PDGF rs974819 polymorphisms and some clinical parameters in patients with unstable angina. In patients with the PDGF rs974819 CC genotype, there were increased values for cholesterol and LDL serum levels in comparison with patients with the PDGF rs974819 CT and TT genotypes. In patients with the PROCR rs867186 AA genotype, HDL serum levels were lower than in patients with the GA genotype. Conclusions: The results of our study did not show that the LPL rs264, PROCR rs867186 and PDGF rs974819 gene polymorphisms were significant risk factors for unstable angina in our population. The results of the study suggest that PDGF rs974819 and PROCR rs867186 may be associated with some parameters of lipid metabolism.
Article
Thalassemia is the most common monogenic disorder of red blood cells (RBCs) caused by defects in the synthesis of globin chains. Thalassemia phenotypes have a wide spectrum of clinical manifestations and vary from severe anemia requiring regular blood transfusions to clinically asymptomatic states. Ineffective erythropoiesis and toxicity caused by iron overload are major factors responsible for various complications in thalassemia patients, especially β-thalassemia major (β-TM) patients. Common complications in thalassemia patients include iron overload, thrombosis, cardiac morbidity, vascular dysfunction, inflammation, and organ dysfunction. Extracellular vesicles (EVs) are small membrane vesicles released from various cells' plasma membranes due to activation and apoptosis. Based on studies, EVs play a role in various processes, including clot formation, vascular damage, and pro-inflammatory processes. In recent years, they have also been studied as biomarkers in the diagnosis and prognosis of diseases. Considering the high level of EVs in thalassemia and their role in cellular processes, this study reviews the role of EVs in the common complications of β-thalassemia patients for the first time.
Article
Full-text available
Cardiovascular disease is currently the leading cause of death worldwide. Atherosclerosis is an important pathological basis of cardiovascular disease, and its early diagnosis is of great significance. Urine bears no need nor mechanism to be stable, so it accumulates many small changes and is therefore a good source of biomarkers in the early stages of disease. In this study, ApoE-/- mice were fed a high-fat diet for 5 months. Urine samples from the experimental group and control group (C57BL/6 mice fed a normal diet) were collected at seven time points. Proteomic analysis was used for comparison within the experimental group and for comparison between the experimental group and the control group. The results of the comparison within the experimental group showed a significant difference in the urinary proteome before and after a one-week high-fat diet, and several of the differential proteins have been reported to be associated with atherosclerosis and/or as biomarker candidates. The results of the comparison between the experimental group and the control group indicated that the biological processes enriched by the GO analysis of the differential proteins correspond to the progression of atherosclerosis. The differences in chemical modifications of urinary proteins have also been reported to be associated with the disease. This study demonstrates that urinary proteomics has the potential to sensitively monitor changes in the body and provides the possibility of identifying early biomarkers of atherosclerosis.
Preprint
Full-text available
Cardiovascular disease is currently the leading cause of death worldwide. Atherosclerosis is an important pathological basis of cardiovascular disease, and its early diagnosis is of great significance. Urine is more conducive in the accumulation and response of changes in the physiological state of the body and is not regulated by homeostasis mechanisms, so it is a good source of biomarkers in the early stage of disease. In this study, ApoE-/- mice induced by a high-fat diet for 5 months were used to construct an animal model of atherosclerosis. Urine samples from the experimental group and control group, which were C57BL/6 mice fed a normal diet, were collected at seven time points. Proteomic analysis was used for internalcontrol and intergroup control. Internal control results showed a significant difference in the urinary proteome before and after a 1-week high-fat diet, and several differential proteins have been reported to be associated with atherosclerosis or for use as biomarkers. The results of the intergroup control indicated that the biological process enriched by the GO analysis of the differential proteins corresponded to disease progression. Differences in chemical modifications of urinary proteins have also been reported to be associated with the disease. This study demonstrates that urinary proteomics has the potential to monitor changes in the body sensitively and provides the possibility of identifying early biomarkers of atherosclerosis.
Article
Full-text available
To bridge the gap between organ demand and supply, xenotransplantation has long been considered as a realistic option for end-stage organ failure. Early this year this promise became reality for David Bennett Sr., the first patient whose own failing heart was replaced with a xeno-pig heart. To get here has been a rollercoaster ride of physiological hurdles seemingly impossible to overcome, technological breakthroughs and ethical and safety concerns. It started in 1984, with Stephanie Fae Beauclair, also known as baby Fae, receiving a baboon heart, which allowed her to survive for another 30 days. For ethical reasons primate work was soon abandoned in favour of the pig. But increased phylogenetic distance also brought with it an increased immunological incompatibility. It has been the development of ever more sophisticated genetic engineering tools, which brought down the physiological barriers, enabled humanisation of porcine organs and helped addressing safety concerns. This renewed the confidence in xenotransplantation, brought new funding opportunities and resulted finally in the first in human trial.
Article
Full-text available
Background Klinefelter syndrome (KS) is one of the commonest sex chromosome disorders. Affected males become infertile and highly susceptible to several health problems, including vascular thromboembolism (VTE). The risk of VTE may be exacerbated by an underlying genetically inherited thrombophilia. In this study, we aimed to investigate the genotype and allele frequencies of common gene polymorphisms related to hereditary thrombophilia in infertile males with KS compared to normal, fertile men. Methods Eighty-five infertile males with KS and 75 healthy control males were included in this case-control study. Genetic testing was done using an extended thrombophilia gene panel by Multiplex PCR reverse hybridization method. Results There was an increased frequency of mutant alleles and heterozygous genotypes of FV Leiden, FVH1299R, ProG20210A, MTHFRC677T and PAI-1 4G/5Gthrombophilic gene polymorphisms in KS patients compared to the control group. It was shown that 10.7% of KS patients had the A3 haplotype of the EPCR gene in comparison to 5.3% of control patients.The A3/A3 genotype was found only in KS patients (7.1%). Carriers ofmore than one mutant allele in KS patients exceeded the control (p<0.001). Conclusion A high prevalence of thrombophilic gene polymorphisms and the coexistence of different mutant alleles were evident in infertile KS males. These data highlight the importance of conducting further studies to understand the role of hereditary thrombophilia in predicting venous thrombosis in patients with Klinefelter syndrome.
Article
Presently, no data on the molecular basis of hereditary protein C (PC) deficiency in Spain is available. We analyzed the PC gene (PROC) in 109 patients with symptomatic PC deficiency and in 342 relatives by sequencing the 9 PROC exons and their flanking intron regions. In 93 probands, we found 58 different mutations (26 novel). Thirty-seven consisted of a nucleotide change, mainly missense mutations, 1 was a 6-nucleotide insertion causing the duplication of 2 amino acids, and 4 were deletions of 1, 3, 4, and 16 nucleotides. Nine mutations caused type II deficiencies, with the presence of normal antigen levels but reduced anticoagulant activity. Using a PC level of 70% as lowest normal limit, we found no mutations in 16 probands and 25 relatives with PC levels ≤ 70%. On the contrary, 4 probands and 12 relatives with PC levels > 70% carried the mutation identified in the proband. The spectrum of recurrent mutations in Spain is different from that found in the Netherlands, where the most frequent mutations were p.Gln174* and p.Arg272Cys, and is more similar to that found in France, where the most frequent were p.Arg220Gln and p.Pro210Leu. In our study, p.Val339Met (9 families), p.Tyr166Cys (7), p.Arg220Gln (6), and p.Glu58Lys (5) were the most prevalent. This study confirms the considerable heterogeneity of the genetic abnormality in PC deficiencies, and allowed genetic counseling to those individuals whose PC levels were close to the lower limit of the normal reference range.
Article
Background: It is well known that warfarin inhibits the synthesis of vitamin K-dependent anticoagulants, including thrombin, protein C and S, and factor Xa, leading, paradoxically, to an initial hypercoagulable state. Edoxaban, a direct inhibitor of activated factor X is widely used for the treatment of acute venous thromboembolism (VTE). However, the effect of edoxaban on circulating coagulation factors, in patients with acute VTE, remains unknown. Methods and results: We enrolled 57 patients with acute VTE with/without pulmonary embolism treated with edoxaban (n=37) or warfarin (n=20) in a clinical setting. Before treatment and 2 weeks after treatment, we evaluated thrombotic burden using ultrasound or computed tomography angiography. We also evaluated thrombin generation, represented by prothrombin fragment F1+2; thrombus degradation, represented by D-dimer; and levels of anticoagulants, including protein C, protein S, and antithrombin III. Both edoxaban and warfarin treatment improved thrombotic burden and decreased prothrombin fragment F1+2, and D-dimer. Edoxaban treatment preserved protein C and protein S levels. In contrast, warfarin decreased protein C and protein S levels. Neither treatment affected antithrombin III. Conclusions: Edoxaban improves VTE while preserving protein C and protein S levels, thereby indicating that edoxaban improves thrombotic burden while maintaining levels of anticoagulants.
Article
The endothelial cell protein C/activated protein C receptor (EPCR) is located primarily on the surface of the large vessels of the vasculature. In vitro studies suggest that it is involved in the protein C anticoagulant pathway. We report the organization and nucleotide sequence of the human EPCR gene. It spans approximately 6 kbp of genomic DNA, with a transcription initiation point 79 bp upstream of the translation initiation (Met) codon in close proximity to a TATA box and other promoter element consensus sequences. The human EPCR gene has been localized to 20q11.2 and consists of four exons interrupted by three introns, all of which obey the GT-AG rule. Exon I encodes the 5′ untranslated region and the signal peptide, and exon IV encodes the transmembrane domain, the cytoplasmic tail, and the 3′ untranslated region. Exons II and III encode most of the extracellular region of the EPCR. These exons have been found to correspond to those encoding the 1 and 2 domains of the CD1/major histocompatibility complex (MHC) class I superfamily. Flanking and intervening introns are of the same phase (phase I) and the position of the intervening intron is identically located. Secondary structure prediction for the amino acid sequence of exons II and III corresponds well with the actual secondary structure elements determined for the 1 and 2 domains of HLA-A2 and murine CD1.1 from crystal structures. These findings suggest that the EPCR folds with a β-sheet platform supporting two -helical regions collectively forming a potential binding pocket for protein C/activated protein C.
Article
Purified human factor VIII procoagulant protein (VIII:C) was treated with purified human activated protein C (APC) and the loss of VIII:C activity correlated with proteolysis of the VIII:C polypeptides. APC proteolyzed all VIII:C polypeptides with mol wt = 92,000 or greater, but not the doublet at mol wt = 79–80,000. These results and our previous thrombin activation studies of purified VIII:C, are analogous with similar studies of factor V and form the basis for the following hypothesis: activated VIII:C consists of heavy and light chain polypeptides [mol wt = 92,000 and mol wt = 79–80,000 (or 71–72,000), respectively] which are similar in Mr to the heavy and light chains of activated factor V. Thrombin activates VIII:C and V by generating these polypeptide chains from larger precursors and APC inactivates both molecules by cleavage at a site located in the heavy chain region of activated VIII:C and V.
Article
A family with a history of severe recurrent venous thromboembolic disease was studied to determine if a plasma protein deficiency could account for observed disease. Protein S levels in plasma were determined immunologically using the Laurell rocket technique. The propositus, his mother, his aunt, and his cousin who were clinically affected had 17% to 65% of the control levels of protein S antigen (normal range, 71% to 147%). Since three of these patients were receiving oral anticoagulant therapy, the ratios of protein S to prothrombin, factor X, and protein C in these patients were compared with values for a group of orally anticoagulated controls. These results suggested that protein S is half-normal in all family members with thrombotic disease. Other proteins known to be associated with familial thrombotic disease, including antithrombin III, plasminogen, fibrinogen, and protein C, were normal. Because plasma protein S serves as a cofactor for the anticoagulant activity of activated protein C and because protein C deficiency is associated with recurrent thrombotic disease, it is suggested that recurrent thrombotic disease in this family is the result of an inherited deficiency of protein S.
Article
Background: The endothelial cell protein C receptor (EPCR) enhances protein C activation by the thrombin-thrombomodulin complex. As evidence is accumulating that EPCR is an important component of the protein C anticoagulant pathway, polymorphisms in the EPCR gene might be candidate risk factors predisposing to venous thromboembolism (VTE). Recently, a 23bp insertion in exon 3 of the EPCR gene has been identified, which duplicates the preceding 23 bases and results in a STOP codon downstream from the insertion point. However, the clinical significance of this mutation in VTE remains to be clarified. Methods and Results: In this study we evaluated the EPCR 23bp insertion in 889 patients with documented VTE and in 500 healthy controls. The prevalence of the EPCR insertion among patients was 0.1%, which was not significantly different compared to controls (0.6%, p = 0.1). Conclusions: Our findings showed that the EPCR 23bp insertion is very rare in both patients with VTE and the general population and failed to support an association between the EPCR 23bp insertion and an increased risk of VTE.
Article
Human protein C and activated protein C are shown to bind to endothelium specifically, selectively and saturably (K-d = 30 nM, 7000 sites per cell) in a Ca2+ dependent fashion. Expression cloning revealed a 1.3-kilobase pair cDNA that coded for a novel type 1 transmembrane glycoprotein capable of binding protein C. This protein appears to be a member of the CD1/major histocompatibility complex superfamily. Like thrombomodulin, the receptor involved in protein C activation, the endothelial cell protein C receptor function and message are both down-regulated by exposure of endothelium to tumor necrosis factor. Identification of endothelial cell protein C receptor as a member of the CD1/major histocompatibility complex superfamily provides insights into the role of protein C in regulating the inflammatory response.
Article
The protein C anticoagulant pathway serves as a major system for controlling thrombosis, limiting inflammatory responses, and potentially decreasing endothelial cell apoptosis in response to inflammatory cytokines and ischemia. The essential components of the pathway involve thrombin, thrombomodulin, the endothelial cell protein C receptor (EPCR), protein C, and protein S. Thrombomodulin binds thrombin, directly inhibiting its clotting and cell activation potential while at the same time augmenting protein C (and thrombin activatable fibrinolysis inhibitor [TAFI]) activation. Furthermore, thrombin bound to thrombomodulin is inactivated by plasma protease inhibitors > 20 times faster than free thrombin, resulting in increased clearance of thrombin from the circulation. The inhibited thrombin rapidly dissociates from thrombomodulin, regenerating the anticoagulant surface. Thrombomodulin also has direct anti-inflammatory activity, minimizing cytokine formation in the endothelium and decreasing leukocyte-endothelial cell adhesion. EPCR augments protein C activation approximately 20-fold in vivo by binding protein C and presenting it to the thrombin-thrombomodulin activation complex. Activated protein C (APC) retains its ability to bind EPCR, and this complex appears to be involved in some of the cellular signaling mechanisms that down-regulate inflammatory cytokine formation (tumor necrosis factor, interleukin-6). Once APC dissociates from EPCR, it binds to protein S on appropriate cell surfaces where it inactivates factors Va and VIIIa, thereby inhibiting further thrombin generation. Clinical studies reveal that deficiencies of protein C lead to microvascular thrombosis (purpura fulminans). During severe sepsis, a combination of protein C consumption, protein S inactivation, and reduction in activity of the activation complex by oxidation, cytokine-mediated down-regulation, and proteolytic release of the activation components sets in motion conditions that would favor an acquired defect in the protein C pathway, which in turn favors microvascular thrombosis, increased leukocyte adhesion, and increased cytokine formation. APC has been shown clinically to protect patients with severe sepsis. Protein C and thrombomodulin are in early stage clinical trials for this disease, and each has distinct potential advantages and disadvantages relative to APC.