ArticlePDF AvailableLiterature Review

Endoglin in angiogenesis and vascular diseases

Authors:

Abstract and Figures

Endoglin is a transmembrane auxillary receptor for transforming growth factor-beta (TGF-beta) that is predominantly expressed on proliferating endothelial cells. Endoglin deficient mice die during midgestation due to cardiovascular defects. Mutations in endoglin and activin receptor-like kinase 1 (ALK1), an endothelial specific TGF-beta type I receptor, have been linked to hereditary hemorrhagic telangiectasia (HHT), an autosomal dominant vascular dysplasia characterized by telangiectases and arteriovenous malformations. Endoglin heterozygote mice develop HHT-like vascular abnormalities, have impaired tumor and post-ischemic angiogenesis and demonstrate an endothelial nitric oxide synthase-dependent deterioration in the regulation of vascular tone. In pre-eclampsia, placenta-derived endoglin has been shown to be strongly upregulated and high levels of soluble endoglin are released into the circulation. Soluble endoglin was found to cooperate with a soluble form of vascular endothelial growth factor receptor 1 in the pathogenesis of pre-eclampsia by inducing endothelial cell dysfunction. Endoglin is highly expressed in tumor-associated endothelium, and endoglin antibodies have been successfully used to target activated endothelial cells and elicit anti-angiogenic effects in tumor mouse models. These exciting advances provide opportunities for the development of new therapies for diseases with vascular abnormalities.
Content may be subject to copyright.
ORIGINAL PAPER
Endoglin in angiogenesis and vascular diseases
Peter ten Dijke Æ Marie-Jose
´
Goumans Æ
Evangelia Pardali
Received: 30 January 2008 / Accepted: 31 January 2008 / Published online: 19 February 2008
Ó Springer Science+Business Media B.V. 2008
Abstract Endoglin is a transmembrane auxillary receptor
for transforming growth factor-b (TGF-b) that is predom-
inantly expressed on proliferating endothelial cells.
Endoglin deficient mice die during midgestation due to
cardiovascular defects. Mutations in endoglin and activin
receptor-like kinase 1 (ALK1), an endothelial specific
TGF-b type I receptor, have been linked to hereditary
hemorrhagic telangiectasia (HHT), an autosomal dominant
vascular dysplasia characterized by telangiectases and
arteriovenous malformations. Endoglin heterozygote mice
develop HHT-like vascular abnormalities, have impaired
tumor and post-ischemic angiogenesis and demonstrate an
endothelial nitric oxide synthase-dependent deterioration in
the regulation of vascular tone. In pre-eclampsia, placenta-
derived endoglin has been shown to be strongly upregu-
lated and high levels of soluble endoglin are released into
the circulation. Soluble endoglin was found to cooperate
with a soluble form of vascular endothelial growth factor
receptor 1 in the pathogenesis of pre-eclampsia by inducing
endothelial cell dysfunction. Endoglin is highly expressed
in tumor-associated endothelium, and endoglin antibodies
have been successfully used to target activated endothelial
cells and elicit anti-angiogenic effects in tumor mouse
models. These exciting advances provide opportunities for
the development of new therapies for diseases with vas-
cular abnormalities.
Keywords Angiogenesis BMP Endothelial cells
Hereditary hemorrhagic telangiectasia
Signal transduction Smad TGF-b
Introduction
Endoglin (also known as CD105) was initially identified by
a monoclonal antibody (44G4) raised against a pre-B
lymphoblastic HOON cell line [1]. Its cDNA was isolated
in 1990 and predicted that the encoded endoglin protein is a
type I integral membrane glycoprotein (Fig. 1)[2]. This
and subsequent studies showed that endoglin is highly
expressed on proliferating vascular endothelial cells [25].
Since its identification as an accessory receptor for TGF-b
[6] and the link between endoglin haplo-insufficiency and
HHT [7], its role in (tumor) angiogenesis [712] and the
pathological role of soluble endoglin in pre-eclampsia [13],
the interest in endoglin has strongly increased. It is now
evident that endoglin has a pivotal function in vascular
development and disease [14].
TGF-b1 is the prototype of a family of multifunctional
proteins, which includes three TGF-b isoforms (i.e., TGF-
b1, -b2 and -b3), activins and bone morphogenetic proteins
(BMPs), which are involved in many different patho-
physiological processes, including development, wound
healing, cancer, fibrosis, vascular, and immune diseases
[14, 15]. The importance of TGF-b signaling pathway in
vascular morphogenesis was revealed by the targeted
inactivation of TGF-b
signaling components in mice,
which die at midgestation during embryogenesis due to
disrupted vasculogenesis in the yolk sac [14]. TGF-b
family members signal via two related single transmem-
brane spanning type I and type II receptors endowed with
serine/threonine kinase activity [16, 17]. Each ligand has a
P. ten Dijke (&) M.-J. Goumans E. Pardali
Department of Molecular Cell Biology, Leiden University
Medical Center, Building 2, Room R-02-022, Postzone S-1-P,
Postbus 9600, 2300 RC Leiden, The Netherlands
e-mail: p.ten_dijke@lumc.nl
123
Angiogenesis (2008) 11:79–89
DOI 10.1007/s10456-008-9101-9
specific set of type II and type I receptors with which it
interacts. In most cases TGF-b interacts with TGF-b type II
receptor (TbRII) and TGF-b type I receptor (TbRI), also
termed activin receptor-like kinase 5 (ALK5) [18]. In
endothelial cells TGF-b can also signal via ALK1 [19].
Activins signal via activin type II receptors (ActRII) and
ALK4 [20, 21], and BMPs transduce their effects through
BMP type II receptor (BMPRII) and ActRIIs and ALK1, -
2, -3 and -6 (Fig. 2)[2225]. Upon ligand-induced heter-
omeric complex formation, the type II constitutively active
kinase trans-phosphorylates the type I receptor on serine
and threonine residues located in the juxtamembrane
region [26]. The activated type I receptor propagates the
signal into the cell by phosphorylating specific receptor-
regulated (R-) Smads at two carboxy-terminal serine resi-
dues [27, 28]. Whereas TGF-b and activins in most cases
signal via R-Smad2 and Smad3, BMPs activate R-Smad1,
Smad5 and Smad8 [2932]. Activated R-Smads form het-
eromeric complexes with common mediator (Co-)Smad,
i.e., Smad4 in mammals, which accumulate in the nucleus.
There they can bind to DNA (in)directly and act as tran-
scription factor complexes together with other transcription
factors and co-activators and co-repressors [3234].
Endoglin is a TGF-b type III auxiliary receptor (TbRIII)
[6] that is not directly involved in signaling, but modulates
signaling responses of multiple members of the TGF-b
family [35]. In addition, endoglin is involved in TGF-b
independent signaling [36]. Moreover, two endoglin splice
variants exist which can exert opposite effects [37, 38].
Furthermore, a soluble form of endoglin (sEnd) has been
found, most likely generated by proteolytic shedding, which
antagonizes the membrane bound form [13]. Taken toge-
ther, multiple layers of complexity exist by which the
function of endoglin is regulated. In this review, we discuss
the latest advances in our understanding of its mechanism of
action and function in angiogenesis and vascular diseases.
Structure
Human endoglin is a homo-dimeric protein of 658 amino
acid residues that contains an extracellular domain, a single
transmembrane domain and a short intracellular domain
(Fig. 1)[2]. Endoglin is structurally related to betaglycan
[39, 40], another TbRIII, with high similarity in the trans-
membrane and intracellular regions. Both endoglin and
betaglycan are glycoproteins with N-linked and O-linked
glycans; however, endoglin lacks the glycosamino-glycan
(GAG) chains that are characteristic for betaglycan. Both
type III receptors can form heteromeric complexes with
TbRII, but whereas endoglin requires TbRII for binding to
TGF-b1 and -b3, betaglycan alone can bind all three TGF-b
isoforms [41, 42]. Endoglin and betaglycan are usually
expressed as homodimers on the cell surface, which in the
case of endoglin are linked by disulfide bridges. Heteromeric
complexes between endoglin and betaglycan have been
observed in microvascular endothelial cells [43]. Besides the
membrane-bound form, both type III receptors can occur in
soluble form. High circulating levels of sEnd are detected in
patients with cancer [44, 45] or pre-eclampsia [13, 46].
Betaglycan can be shed by membrane-type metalloprotease
1 (MT1-MMP [47] and sEnd is likely also generated by
proteolytic cleavage of the membrane-bound form.
The extracellular domain of human endoglin contains an
Arg-Gly-Asp (RGD) tripeptide sequence that is an inter-
action motif for integrins. However, this sequence is not
conserved in mouse pig and rat [2, 48]. In its extracellular
A
B
Fig. 1 Schematic representation of endoglin structure. (a) Endoglin
is a disulphide-linked dimeric protein with large extracellular domain,
single transmembrane domain and short intracellular region. The
extracellular domain consists of an orphan domain and a ZP domain.
The RGD binding motif that is present in human endoglin is
indicated. Interaction partners and the (potential) regulatory effects
that are mediated through these interactions are indicated (b). The two
splice forms of endoglin, i.e., long (L) and short (S) differ in their
intracellular regions. The intracellular region lacks an enzymatic
motif but is rich in serine and threonine residues that can be
phosphorylated by TbRII, ALK1 and ALK5. At the C-terminus of L-
endoglin a PDZ interaction motif (SMA) is present. Abbreviations:
ALK, activin receptor-like kinase; eNOS, endothelial nitric oxide
synthase; MAPK, mitogen activated protein kinase; TbR, TGF-b
receptor; ZP, Zona Pellucida
80 Angiogenesis (2008) 11:79–89
123
region endoglin harbors a zona pellucida (ZP) domain of
approximately 260 amino acid residues, a feature shared
with betaglycan [49]. The ZP domain is involved in en-
doglin oligomerization and in ligand (in)dependent
heteromeric interactions with the TGF-b receptors TbRII
and ALK5 [50]. Using single-particle electron microscopy,
the three-dimensional structure of the extracellular domain
of endoglin was determined at 25 A
˚
resolution. The two
monomers are positioned in an anti-parallel fashion with
each monomer consisting of three well-defined domains
[51], i.e., one domain with unknown structural homology
and a ZP domain with bipartite structure (Fig. 1).
The intracellular domain of endoglin lacks an enzymatic
motif but contains many serine and threonine residues of
which certain residues are phosphorylated by TGF-b
receptor kinases (see below) [52]. At its C-terminus en-
doglin contains a PDZ interaction motif (SerSerMetAla).
There are two splice isoforms, termed Long (L)- and Short
(S)-endoglin, which differ in their intracellular part; L and
S have cytoplasmic tails of 47 and 14 amino acids,
respectively, and have only 7 juxtamembrane amino acids
in common [53]. The L form is most abundantly expressed
and is also the predominant form in endothelial cells while
the S-form is expressed in liver and lung at significant
levels [37]. Both forms can bind TGF-b [53], and differ-
ently regulate TGF-b-induced responses. L-endoglin
enhanced the TGF-b/ALK1 pathway, while S-endoglin
promoted the TGF-b/ALK5 route upon ectopic expression
in myoblasts [38]. Forced expression of S-endoglin in
vascular endothelium in mice results in reduced tumor
growth and neovascularization and suggest that S-endoglin
(in contrast to L-endoglin) has anti-angiogenic activity.
S-endoglin may elicit this effect by inhibiting the formation
of L–L homodimers as L-and S-forms have been shown to
form heterodimers [37].
Cellular and tissue distribution
Whereas endoglin is expressed at low to non-detectable
levels in resting endothelial cells within normal tissues, it is
highly expressed in vascular endothelial cells in sites of
Fig. 2 TGF-b family signaling pathways in endothelial cells.
Signaling by TGF-b family members, which includes TGF-bs,
activins and BMPs, occurs via specific cell surface type I and type
II receptors that are endowed with serine/threonine kinase activity.
Accessory receptors endoglin and betaglycan modulate TGF-b family
signaling via type I and type II receptors. Soluble endoglin and
betaglycan can sequester ligand and thereby inhibit receptor binding.
In most cells TGF-b signals via TbRII and ALK5. In endothelial cells
(depicted here) it signals also via another type I receptor ALK1.
Activins signal via ActRII and ALK4. BMPs signal via BMPRII and
ActRII and type I receptors ALK1, ALK2, ALK3 and ALK6. The
type I receptors act downstream of type II receptor and determine the
signaling specificity of the receptor complex. Activated type I
receptors initiate intracellular signaling by phosphorylating specific
R-Smads. Activation of ALK1, ALK23, ALK3 and ALK6 leads to
phosphorylation of Smad1, Smad5 and Smad8, and Smad2 and
Smad3 are phosphorylated by ALK4, ALK5 and ALK7. Activated R-
Smads assemble with Smad4 in heteromeric complexes that accu-
mulate in the nucleus. There these complexes regulate specific gene
expression responses by binding to DNA together with other DNA
binding transcription factors. Abbreviatons: ActR, activin receptor;
BMP, bone morphogenetic protein; BMPR, BMP receptor; sEnd,
soluble endoglin; transforming growth factor-b;TbR, TGF-b recep-
tor; TF, transcription factor
Angiogenesis (2008) 11:79–89 81
123
active angiogenesis during embryogenesis, in inflamed
tissues, and within and surrounding tumors [35]. After
ischemia-reperfusion injury and myocardial infarction,
endoglin expression is upregulated in the ischemic area and
border zone [54]. Besides hypoxia, TGF-b, BMP9, and
constitutively active (ca)ALK1 also potently stimulate
endoglin expression [5557], whereas TNFa inhibits en-
doglin expression in endothelial cells [58]. The endoglin
promoter contains multiple Sp1 binding sites that are
shown to play a critical role for basal transcription [59].
Hypoxia and TGF-b cooperate in endoglin promoter acti-
vation [60]. This appears to be mediated via a multi-protein
complex consisting of Smad3/4, Sp1 and HIF1a/b bound to
their cognate DNA binding elements.
Despite the fact that endoglin is considered to be an
endothelial specific marker, several other cell types have
been shown to express endoglin. For example, endoglin is
present on monocytes and upregulated during the mono-
cyte-macrophage transition [61]. Endoglin was also found
to have a crucial role in monocyte-mediated vascular repair
[54]. In addition, endoglin is expressed in syncytiotroph-
oblasts of term placenta [62] and in pre-eclampsia its
expression is highly elevated [13, 46]. While endoglin
in normal smooth muscle cells is low, its expression is
strongly upregulated in vascular smooth muscle cells
in atherosclerosis [63]. During development endoglin
is expressed in a subset of neural crest stem cells and is
required for myogenic differentiation [64]. Moreover, en-
doglin is expressed in adult bone marrow hematopoietic
stem cells (HSCs) [65] and is a functional marker that
defines long-term repopulating hematopoietic stem cells
[66]. Using the differentiation of ES cells into embryoid
bodies as an assay system, endoglin was shown to be
required for efficient myelopoiesis and definitive erythro-
poiesis [67]. In addition, overexpression or knockdown of
endoglin in hematopoietic stem cells revealed that, while
endoglin is not required for engraftment and reconstituting
capacities, it regulates adult erythroid development [68].
Furthermore, endoglin expression is also found in cer-
tain tumor cells, including primary and metastatic lesions
of melanoma [69] and ovary [70] and prostate cancer cells
[71]. In prostate cancer cells, endoglin suppresses cell
adhesion, motility and invasion by activating the TGF-b/
ALK2/Smad1 pathway [72, 73], in a manner that is remi-
niscent of TGF-b/ALK1/Smad1 signaling in endothelial
cells [19]. Consistent with the notion that endoglin
expression is attenuated during prostate cancer progression,
it has been postulated that endoglin has a tumor suppressor
role in prostate cancer. Endoglin is also expressed in epi-
dermal keratinocytes [74]. In a multistage model for mouse
skin carcinogenesis using wild type and endoglin hetero-
zygous mice, endoglin was shown to have a dual role by
inhibiting benign tumor formation, but accelerating the
malignant conversion in skin carcinogenesis. Endoglin
haplo-insufficieny may elicit these effects by enhancing the
tumor suppressing and inhibiting the tumor promoting
effects of TGF-b/ALK5 signaling in tumor cells [74].
Interplay with TGF-b and other signal transduction
pathways
Besides the TGF-b dependent interaction of endoglin with
TbRII [42], endoglin can also interact with TbRII and type
I receptors ALK1 and ALK5 in a ligand independent
manner and both extra- and intracellular domains contrib-
ute to this interaction [50, 75]. Endoglin can bind several
other ligands besides TGF-b, including activins and BMPs
and can interact with activin type II receptors [76]. Inter-
estingly, BMP9 can bind endoglin with high affinity
independently from type I or type II receptors (Fig. 2)[56].
In endothelial cells TGF-b can signal via two distinct
type I receptor pathways, i.e., ALK1 and ALK5 [18].
Whereas TGF-b/ALK1 signaling stimulates cell prolifera-
tion and migration of endothelial cells, TGF-b/ALK5
signaling inhibits these responses (Fig. 2)[19]. Recent
studies have revealed an intricate interplay between the two
signaling pathways, and with endoglin. ALK5/Smad2/3
inhibits ALK1-Smad1/5 signaling and vice versa, and
ALK5 is required for efficient TGF-b/ALK1 signaling [77].
Forced overexpression of endoglin inhibits TGF-b/ALK5
signaling [78, 79] and TGF-b-induced growth inhibition
[42, 61, 80]. Conversely, inhibiting endoglin function by
specific knockdown or treatment with neutralizing anti-
bodies inhibits TGF-b/ALK1 signaling, and (in) directly
potentiates TGF-b/ALK5 signaling [8083
]. Establishment
of endothelial cell lines from endoglin deficient mouse
embryos (mouse embryonic endothelial cells, MEECs)
demonstrated that endothelial cells deficient in endoglin do
not proliferate efficiently, possibly because TGF-b/ALK1
signaling is attenuated and TGF-b/ALK5 signaling is
enhanced. Surviving cells were found to have downregu-
lated ALK5 expression, possibly caused by an adaptive
response to overcome the increased TGF-b/ALK5 induced
growth arrest [80]. Similar results were obtained with pri-
mary cultures of outgrown endothelial cells from the blood
of HHT1 patients [84]. However, in another study using
MEECs derived from endoglin null embryos, it was dem-
onstrated that endoglin is not required for TGF-b-induced
Smad1/5 activation and that endoglin may regulate TGF-b
receptor affinity [85].
The intracellular region of endoglin can be phosphory-
lated by TbRII and type I receptors [52]. TbRII and ALK5
preferentially phosphorylate Ser634 and Ser635, which
greatly facilitate subsequent phosphorylation by caALK1
on threonine residues (Fig. 1). The PDZ-binding motif
82 Angiogenesis (2008) 11:79–89
123
appears to negatively regulate phosphorylation as its
removal greatly increases the serine phosphorylation level
by all three receptor kinases. The endoglin phosphorylation
status influences its subcellular distribution and may
modulate endoglin-mediated effects on endothelial cell
adhesion and proliferation.
Endoglin can localize to caveolae [86]. Interestingly,
caveolin-1, the major protein component in caveolae, was
recently shown to interact and functionally cooperate with
TGF-b/ALK1 signaling in endothelial cells [87]. Specific
knockdown of caveolin-1 or caveolae disruption by cho-
lesterol depletion attenuated ALK1 signaling. This in
contrast to TGF-b/ALK5 signaling for which caveolin-1
has an inhibitory effect [88]. Interestingly, cavaolin-1
knock-out mice develop vascular abnormalities with
increased endothelial permeability [89].
Several studies have suggested that endoglin has func-
tions that are independent from TGF-b family signaling
(Fig. 1). The endoglin-induced inhibition of apoptosis in
endothelial cells subjected to hypoxic stress has been
suggested to occur independently of TGF-b [55]. Endoglin
has also been shown to induce activation of MAP kinases
[78]. Endoglin interacts with zyxin and ZRP-1, LIM
domain containing proteins which are concentrated in focal
adhesions [90, 91]. Via these interactions endoglin may
control cell migration independently of TGF-b. The en-
doglin cytoplasmic tail interacts with b-arrestin2 in
endothelial cells [92]. This interaction results in endoglin
internalization in endosomal vesicles and was shown to
inhibit TGF-b-induced ERK activation and migration in
endothelial cells. Using the cytoplasmic tail as bait in yeast
two hybrid interaction screen, Tctex2b, a dynein light
chain (DLC) family member, was identified [93]. Tctex2b
was also found to interact with TbRII. Ectopic expression
of Tctex2b had an inhibitory effect on TGF-b-induced
transcriptional reporter activity. Endoglin, TbRII, and
Tctex2b colocalize in the plasmamembrane. DLC protein
family members are involved in retrograde protein trans-
port, but whether the function of endoglin is regulated and
directed by this remains to be investigated.
Vascular development
Endoglin expression is elevated during alterations in vas-
cular structure as they occur during embryogenesis,
inflammation, and wound healing [94, 95]. The link
between mutations in the endoglin gene and the vascular
disorder HHT [7] also indicates an important role for en-
doglin in maintaining normal vascular architecture (see
discussion below). Mice deficient in endoglin die 10.5 days
p.c. and fail to form mature blood vessels in the yolk sac
[810
]. Vessels do form, but are dilated and fragile, and
easily rupture. The endoglin deficient embryos are much
more fragile and smaller than their wild type litter mates.
Most embryos lack vitelline vessels, which connect the
yolk sac with the embryo proper. This may also explain the
pericardial effusion, indicative of a circulation defect that
is observed in most mutant embryos. This phenotype of
endoglin deficient embryos is reminiscent of that of TGF-
b1, TbRII, ALK5, and ALK1 [14], and suggests a func-
tional link of endoglin with these other TGF-b receptors
during extra-embryonic vascular development. In contrast
to the lack of vascular networks in the yolk sac of endoglin
deficient mice, the vasculature in the embryo proper was
normal with the exception of the heart [810]. Endoglin
null mice were found to have several cardiac defects;
almost all endoglin deficient embryos demonstrated
enlarged cardiac ventricles and dilated outflow tracts and
the atrioventricular canal endocardium failed to undergo
mesenchymal transformation and to form cushions. These
defects in heart valve formation in the endoglin knockouts
might also be related to perturbation of TGF-b signaling.
TGF-b ligands and signaling receptors are also expressed
in the developing heart [96] and defects in heart looping
and morphogenesis have been reported in transgenic mice
with misexpressed TGF-b receptor [97].
TGF-b has an important role in vascular morphogenesis,
and is involved in endothelial cell function and differen-
tiation of pericytes and smooth muscle cells [98]. Endoglin
mutant mice also demonstrate defects in both endothelial
and smooth muscle cell function [810]. Its predominant
expression in endothelial cells suggests that the primary
defect is endothelial and that defects in smooth muscle cell
differentiation are secondary. Consistent with this notion,
endoglin deficient mice have decreased paracrine TGF-b
signaling from endothelial cells to neighboring mesothelial
cells in the yolk sac [99]. This may lead to defective
mesothelial cell differentiation into pericytes or vascular
smooth muscle cells. As a result the vessels are weak and
susceptible to damage and hemorrhage. However, recent
studies also demonstrate that endoglin may have a cell
autonomous role in vascular smooth muscle cells. Ectopic
expression of endoglin in neural crest cells resulted in
pronounced hemorrhage in cranial vessels and in the
pericardial cavity due to aberrant smooth muscle cells in
the vascular wall [64].
Mice heterozygotes for endoglin serve as a good model
for HHT [9]. They have dilated and fragile blood vessels,
telangiectases, and nosebleeds, which are the clinical
manifestations that are also observed in HHT patients.
Some strains of mice are more affected than others, sug-
gesting the existence of modifier genes. In addition, shear,
blood pressure, and inflammation products may contribute
to disease heterogeneity. This has also been suggested to be
the case for HHT patients [100]. Endoglin heterozygous
Angiogenesis (2008) 11:79–89 83
123
mice have reduced eNOS expression and NO synthesis-
dependent vasodilation is impaired [86, 101]. Endothelial
cells derived from endoglin heterozygous mice showed a
significantly attenuated proliferation, migration, increased
collagen production, and mitigated NO synthase expression
and VEGF secretion. Compared to wild type animals, mice
heterozygous for endoglin showed impaired angiogenesis
as observed by a delayed reperfusion following hindlimb
ischemia [11]. In addition, when matrigel plugs were
implanted in endoglin heterozygote mice to measure
endothelial cell outgrowth and invasion into the extracel-
lular matrix in vivo, it was found that they had significantly
less vascular structures compared to wild type mice [11].
Hereditary hemorrhagic telangiectasia (HHT)
HHT, also termed Osler–Weber–Rendu disease, is an
autosomal dominant vascular disorder with an incidence of
about 1 in 10,000 [100, 102]. Characteristic clinical features
include small dilated bloodvessels (telangiectases) and
arteriovenous malformations (AVMs) in the vasculature of
lung, liver, and brain. The disease manifests itself often
during puberty with spontaneous recurrent nose bleeds.
Later in life the intensity of the nosebleeds increase and also
AVMs become larger and problematic. However, onset and
clinical manifestations of HHT are heterogeneous between
different individuals and even within families [100, 102].
Two HHT variants, i.e., HHT1 and HHT2, have been
described that have been linked to mutations in endoglin
and ALK1, respectively. HHT1 differs from HHT2 in an
earlier onset, stronger prevalence and higher frequency of
pulmonary AVMs. In HHT2 the gastrointestinal bleeding
and liver involvement appears to occur frequently than in
HHT1 families [100, 102]. Recently, another gene in the
TGF-b signaling pathway has been implicated in HHT;
Smad4 is mutated in a subset of HHT patients with juvenile
polyposis, lacking mutations in endoglin and ALK1 [103].
This syndrome is now termed JP-HHT.
Mutations in endoglin include deletions, insertions and
missense mutations, and splice site changes, and in about
80% of the cases lead to premature stop codons and trun-
cated endoglin proteins [100, 102]. It has been suggested
that these mutant proteins may achieve a dominant nega-
tive effect by disrupting normal endoglin function.
However, mutated endoglin proteins were found to be
expressed at low levels, mediated by nonsense mediated
mRNA decay. In addition, mutated proteins may be mis-
folded and not stable and/or do not reach the cell surface
and are thereby unable to form heterodimers with normal
endoglin. Indeed, patients with endoglin mutations have
significantly lower endoglin levels in peripheral blood
monocytes compared to control group [100]. Haplo-
insufficiency, therefore, likely provides the explanation for
the mechanism underlying HHT1. The lower levels of
endoglin expression leads to a dysfunction in TGF-b signal
transduction. Whether other non-TGF-b signaling path-
ways are affected as well by lowered endoglin expression
is not clear.
As already mentioned before, studies in mouse models
suggest that defective paracrine TGF-b signaling of endo-
thelial cells to adjacent pericytes/smooth muscle cells lead
to an inefficient maturation of these cells [99]. This makes
the vessels highly susceptible to rupture, a main charac-
teristic of HHT. Interestingly, HHT patients with endoglin
mutations have been found to have low circulating levels of
TGF-b [104]. Several mechanisms have been suggested
through which the AVMs develop. During development
there may be a loss of arterial and venous identity whereby
the normal separation between veins and arteries are dis-
rupted. In addition, defective vascular remodeling and
dilatation may occur following local inflammation. More-
over, cells that form the capillary endothelial bed that
separates the arteries and veins may be removed by
apoptosis in response to hypoxic stress [14].
Pre-eclampsia
Pre-eclampsia is characterized by the onset of high blood
pressure and significant amounts of protein in the urine in the
third trimester of pregnancy [105]. It affects both the fetus
and the mother and occurs in about 5% of the pregnancies.
Severe pre-eclampsia leads to appearance of Hemolysis
Elevated Liver enzymes and Low Platelets (HELLP) syn-
drome, seizures and/or fetal growth restriction, and can
result in death [105, 106]. The endothelial dysfunction in
pre-eclampsia is thought to be caused by circulating factors
that are released from the placenta [107, 108]. Circulating
levels of placenta-derived soluble form of vascular endo-
thelial growth factor receptor (VEGFR)1 (also known as
sFlt), which sequesters the pro-angiogenic proteins placenta
growth factor (PlGF) and VEGF, are increased before onset
and correlate with the severity of pre-eclampsia [109].
Interestingly, a 65 kDa sEnd is increased in sera of pregnant
women, and strongly elevated in pre-eclamptic patients.
SEnd levels also correlate with disease severity [46].
Whereas forced expression of soluble endoglin increased
vascular permeability and induced a modest increase in
blood pressure without significant proteinuria, ectopic
expression of both sFlt and sEnd in pregnant rats induced the
hallmarks of severe pre-eclampsia [13].
In vitro studies on endothelial cell lines showed that
sEnd interferes with TGF-b signaling and eNOS activation
and thereby causes endothelial dysfunction [13]. The sol-
uble form of endoglin functions likely as a scavenger of
84 Angiogenesis (2008) 11:79–89
123
specific circulating TGF-b family ligands, such as TGF-b
or BMP9 [13, 14]. sEnd and sFlt act differently and may
therefore cooperate to induce the endothelial cell dys-
function [13]. As sEnd levels increase 2–3 months before
the onset of pre-eclampsia, sEnd (and sFlt and PlGF) levels
may be used as diagnostic marker to prioritize patients and
thereby prevent pre-eclampsia-induced death [46]. Inhibi-
tion of the putative protease involved in endoglin shedding
may be of therapeutic benefit in the treatment of pre-
eclampsia.
Whereas in pre-eclampsia there is an increase in sEnd,
patients suffering from an acute myocardial infarction were
found to have a significantly lower level of sEnd in their
serum [110]. Interestingly, patients that died had signifi-
cantly lower levels of sEnd than those that survived. Low
levels of sEnd may therefore be used as a prognostic
marker after acute myocardial infarction.
Anti-angiogenic activity of endoglin antibodies
Tumors depend on angiogenesis to grow beyond a few
cubic millimeters; the exchange of oxygen and nutrients
with carbon dioxide and waste products cannot rely any
longer on diffusion, but require blood vessels. Also
metastasis, the spreading and colonization of the primary
tumor to distant organs, requires tumor angiogenesis [111].
Anti-angiogenic therapy, in which proliferating endothelial
cells within and surrounding tumor associated vessels are
selectively targeted, has been shown (in combination with
chemotherapy) to induce tumor regression and inhibit
metastasis [112].
Endoglin is expressed in tumor associated endothelium
in many solid cancers, including breast, prostate, and cer-
vical cancer [113116]. Intra-tumor microvessel density as
determined by anti-endoglin staining and circulating levels
of soluble endoglin have prognostic significance in cancer.
Antibodies that specifically detect endoglin have success-
fully been used for tumor imaging and endoglin antibodies
coupled with toxins [117] or radioactivity [118] have been
used with favorable outcome for vascular targeting [119,
120]. Monoclonal antibodies to endoglin can inhibit pro-
liferation of endothelial cells [121] and have been shown to
have anti-angiogenic effects [122]. Moreover, immuno-
therapy with the extracellular domain of porcine endoglin
was found to induce cytotoxic T lymphocyte (CTL)-med-
iated cytotoxicity and inhibits both endothelial cell
proliferation and tumor growth in mouse cancer models
[123, 124]. However, the suitability of endoglin as a target
for anti-angiogenic therapy for cancer has been questioned;
endoglin expression has also been observed in the endo-
thelial cells of normal tissues [125, 126]. Differences may
prevail between different antibodies, and testing of
different endoglin antibodies to select the best one for
antibody-based therapeutic approaches has been proposed
[119, 126].
The anti-angiogenic effects of sEnd may be used to
interfere with angiogenically active tumors. Besides en-
doglin, also the signaling TGF-b receptors can be targeted
to achieve anti-angiogenic effects. In this respect, it is of
interest to note that treatment of mice with a small mole-
cule ALK5 kinase inhibitor decreased the pericyte
coverage of the endothelial vessels, and in particular those
ones present in the tumor neovasculature. This activity was
used to stimulate a greater accumulation of anticancer
nanocarriers in tumors [127].
Concluding remarks
Endoglin plays a critical role in angiogenesis and dysreg-
ulation of its expression and/or activity has been implicated
in multiple vascular diseases, most notably HHT, pre-
eclampsia and tumor angiogenesis. While much needs to be
investigated on the molecular mechanisms that underlie its
role in vascular development and disease, the recent
advances provide ample opportunities for better diagnosis
and development of new therapies for diseases with vas-
cular abnormalities.
Acknowledgments Our studies on endoglin are supported by Dutch
Cancer Society (UL-2005-3371) and EU grants Angiotargeting and
Tumor-Host-Genomics.
References
1. Quackenbush EJ, Gougos A, Baumal R, Letarte M (2006) Dif-
ferential localization within human kidney of five membrane
proteins expressed on acute lymphoblastic leukemia cells. J
Immunol 136:118–124
2. Gougos A, Letarte M (1990) Primary structure of endoglin, an
RGD-containing glycoprotein of human endothelial cells. J Biol
Chem 265:8361–8364
3. Burrows FJ, Derbyshire EJ, Tazzari PL, Amlot P, Gazdar AF,
King SW, Letarte M, Vitetta ES, Thorpe PE (1995) Up-regu-
lation of endoglin on vascular endothelial cells in human solid
tumors: implications for diagnosis and therapy. Clin Cancer Res
1:1623–1634
4. Miller DW, Graulich W, Karges B, Stahl S, Ernst M, Ra-
maswamy A, Sedlacek HH, Mu
¨
ller R, Adamkiewicz J (1999)
Elevated expression of endoglin, a component of the TGF-b-
receptor complex, correlates with proliferation of tumor endo-
thelial cells. Int J Cancer 81:568–572
5. Fonsatti E, Jekunen AP, Kairemo KJ, Coral S, Snellman M,
Nicotra MR, Natali PG, Altomonte M, Maio M (2000) Endoglin
is a suitable target for efficient imaging of solid tumors: in vivo
evidence in a canine mammary carcinoma model. Clin Cancer
Res 6:2037–2043
6. Cheifetz S, Bello
´
n T, Cale
´
s C, Vera S, Bernabeu C, Massague
´
J,
Letarte M (1992) Endoglin is a component of the transforming
Angiogenesis (2008) 11:79–89 85
123
growth factor-b receptor system in human endothelial cells. J
Biol Chem 267:19027–19030
7. McAllister KA, Grogg KM, Johnson DW, Gallione CJ, Baldwin
MA, Jackson CE, Helmbold EA, Markel DS, McKinnon WC,
Murrell J et al (1994) Endoglin, a TGF-b binding protein of
endothelial cells, is the gene for hereditary haemorrhagic tel-
angiectasia type 1. Nat Genet 8:345–351
8. Li DY, Sorensen LK, Brooke BS, Urness LD, Davis EC, Taylor
DG, Boak BB, Wendel DP (1999) Defective angiogenesis in
mice lacking endoglin. Science 284:1534–1537
9. Bourdeau A, Dumont DJ, Letarte M (1999) A murine model of
hereditary hemorrhagic telangiectasia. J Clin Invest 104:1343–
1351
10. Arthur HM, Ure J, Smith AJ, Renforth G, Wilson DI, Torsney E,
Charlton R, Parums DV, Jowett T, Marchuk DA, Burn J, Dia-
mond AG (2000) Endoglin, an ancillary TGFb receptor, is
required for extraembryonic angiogenesis and plays a key role in
heart development. Dev Biol 217:42–53
11. Jerkic M, Rodrı
´
guez-Barbero A, Prieto M, Toporsian M, Peri-
cacho M, Rivas-Elena JV, Obreo J, Wang A, Pe
´
rez-Barriocanal
F, Are
´
valo M, Bernabe
´
u C, Letarte M, Lo
´
pez-Novoa JM (2006)
Reduced angiogenic responses in adult Endoglin heterozygous
mice. Cardiovasc Res 69:845–854
12. Du
¨
wel A, Eleno N, Jerkic M, Arevalo M, Bolan
˜
os JP, Bernabeu
C, Lo
´
pez-Novoa JM (2007) Reduced tumor growth and angio-
genesis in endoglin-haploinsufficient mice. Tumour Biol 28:1–8
13. Venkatesha S, Toporsian M, Lam C, Hanai J, Mammoto T, Kim
YM, Bdolah Y, Lim KH, Yuan HT, Libermann TA, Stillman IE,
Roberts D, D’Amore PA, Epstein FH, Sellke FW, Romero R,
Sukhatme VP, Letarte M, Karumanchi SA (2006) Soluble en-
doglin contributes to the pathogenesis of preeclampsia. Nat Med
2:642–649
14. ten Dijke P, Arthur HM (2007) Extracellular control of TGFb
signalling in vascular development and disease. Nat Rev Mol
Cell Biol 8:857–869
15. Blobe GC, Schiemann WP, Lodish HF (2000) Role of trans-
forming growth factor b in human disease. N Engl J Med
342:1350–1358
16. Heldin CH, Miyazono K, ten Dijke P (1997) TGF-b signalling
from cell membrane to nucleus through SMAD proteins. Nature
390:465–471
17. Schmierer B, Hill CS (2007) TGFb-SMAD signal transduction:
molecular specificity and functional flexibility. Nat Rev Mol
Cell Biol 8:970–982
18. Franze
´
n P, ten Dijke P, Ichijo H, Yamashita H, Schulz P, Heldin
CH, Miyazono K (1993) Cloning of a TGFb type I receptor that
forms a heteromeric complex with the TGFb type II receptor.
Cell 75:681–692
19. Goumans MJ, Valdimarsdottir G, Itoh S, Rosendahl A, Sideras
P, ten Dijke P (2002) Balancing the activation state of the
endothelium via two distinct TGF-b type I receptors. EMBO J
21:1743–1753
20. Mathews LS, Vale WW (1991) Expression cloning of an activin
receptor, a predicted transmembrane serine kinase. Cell 65:973–
982
21. Attisano L, Wrana JL, Montalvo E, Massague
´
J (1996) Acti-
vation of signalling by the activin receptor complex. Mol Cell
Biol 16:1066–1073
22. ten Dijke P, Yamashita H, Sampath TK, Reddi AH, Estevez M,
Riddle DL, Ichijo H, Heldin C-H, Miyazono K (1994) Identi-
fication of type I receptors for osteogenic protein-1 and bone
morphogenetic protein-4. J Biol Chem 269:16985–16988
23. Liu F, Ventura F, Doody J, Massague
´
J (1995) Human type II
receptor for bone morphogenic proteins (BMPs): extension of
the two-kinase receptor model to the BMPs. Mol Cell Biol
15:3479–3486
24. Rosenzweig BL, Imamura T, Okadome T, Cox GN, Yamashita
H, ten Dijke P, Heldin CH, Miyazono K (1995) Cloning and
characterization of a human type II receptor for bone morpho-
genetic proteins. Proc Natl Acad Sci USA 92:7632–7636
25. Yamashita H, ten Dijke P, Huylebroeck D, Sampath TK, An-
dries M, Smith JC, Heldin C-H, Miyazono K (1995) Osteogenic
protein-1 binds to activin type II receptors and induces certain
activin-like effects. J Cell Biol 130:217–226
26. Wrana JL, Attisano L, Wieser R, Ventura F, Massague
´
J (1994)
Mechanism of activation of the TGF-b receptor. Nature
370:341–347
27. Abdollah S, Macı
´
as-Silva M, Tsukazaki T, Hayashi H, Attisano
L, Wrana JL (1997) TbRI phosphorylation of Smad2 on Ser465
and Ser467 is required for Smad2-Smad4 complex formation
and signaling. J Biol Chem 272:27678–27685
28. Souchelnytskyi S, Tamaki K, Engstro
¨
m U, Wernstedt C, ten
Dijke P, Heldin C-H (1997) Phosphorylation of Ser465 and
Ser467 in the C terminus of Smad2 mediates interaction with
Smad4 and is required for transforming growth factor-b sig-
naling. J Biol Chem 272:28107–28115
29. Hoodless PA, Haerry T, Abdollah S, Stapleton M, O’Connor
MB, Attisano L, Wrana JL (1986) MADR1, a MAD-related
protein that functions in BMP2 signaling pathways. Cell
85:489–500
30. Eppert K, Scherer SW, Ozcelik H, Pirone R, Hoodless P,
Kim H, Tsui LC, Bapat B, Gallinger S, Andrulis IL,
Thomsen GH, Wrana JL, Attisano L (1996) MADR2 maps
to 18q21 and encodes a TGFb-regulated MAD-related pro-
tein that is functionally mutated in colorectal carcinoma. Cell
86:543–552
31. Zhang Y, Feng X, We R, Derynck R (1996) Receptor-associated
Mad homologues synergize as effectors of the TGF-b response.
Nature 383:168–172
32. Lagna G, Hata A, Hemmati-Brivanlou A, Massague
´
J (1996)
Partnership between DPC4 and SMAD proteins in TGF-b sig-
nalling pathways. Nature 383:832–836
33. Liu F, Pouponnot C, Massague
´
J (1997) Dual role of the Smad4/
DPC4 tumor suppressor in TGFb-inducible transcriptional
complexes. Genes Dev 11:3157–3167
34. Feng XH, Zhang Y, Wu RY, Derynck R (1998) The tumor
suppressor Smad4/DPC4 and transcriptional adaptor CBP/p300
are coactivators for smad3 in TGF-b-induced transcriptional
activation. Genes Dev 12:2153–2163
35. Lastres P, Letamendı
´
a A, Zhang H, Rius C, Almendro N, Raab
U, Lo
´
pez LA, Langa C, Fabra A, Letarte M, Bernabe
´
u C (1996)
Endoglin modulates cellular responses to TGF-b1. J Cell Biol
133:1109–1121
36. Lebrin F, Deckers M, Bertolino P, tn Dijke P (2005) TGF-b
receptor function in the endothelium. Cardiovasc Res 65:599–
608
37. Pe
´
rez-Go
´
mez E, Eleno N, Lo
´
pez-Novoa JM, Ramirez JR, Ve-
lasco B, Letarte M, Bernabe
´
u C, Quintanilla M (2005)
Characterization of murine S-endoglin isoform and its effects on
tumor development. Oncogene 24:4450–4461
38. Velasco S, Alvarez-Mun
˜
oz P, Pericacho M, ten Dijke P, Ber-
nabeu C, Lopez-Novoa JM, Rodriguez-Barbero A (2008) L- and
S-endoglin differentially modulate transforming growth factor
b1 signaling mediated by ALK1 and ALK5 in L6E9 myoblasts.
J Cell Sci (in press)
39. Wang XF, Lin HY, Ng-Eaton E, Downward J, Lodish HF,
Weinberg RA (1991) Expression cloning and characterization of
the TGF-b
type III receptor. Cell 67:797–805
40. Lo
´
pez-Casillas F, Cheifetz S, Doody J, Andres JL, Lane WS,
Massague
´
J (1991) Structure and expression of the membrane
proteoglycan betaglycan, a component of the TGF-b receptor
system. Cell 67:785–795
86 Angiogenesis (2008) 11:79–89
123
41. Lo
´
pez-Casillas F, Wrana JL, Massague
´
J (1993) Betaglycan
presents ligand to the TGFb signaling receptor. Cell 73:1435–
1444
42. Letamendı
´
a A, Lastres P, Botella LM, Raab U, Langa C, Ve-
lasco B, Attisano L, Bernabeu C (1998) Role of endoglin in
cellular responses to transforming growth factor-b. A compar-
ative study with betaglycan. J Biol Chem 273:33011–33019
43. Wong SH, Hamel L, Chevalier S, Philip A (2000) Endoglin
expression on human microvascular endothelial cells association
with betaglycan and formation of higher order complexes with
TGF-b signalling receptors. Eur J Biochem 267:5550–5560
44. Calabro
`
L, Fonsatti E, Bellomo G, Alonci A, Colizzi F, Sigalotti
L, Altomonte M, Musolino C, Maio M (2003) Differential levels
of soluble endoglin (CD105) in myeloid malignancies. J Cell
Physiol 194:171–175
45. Li C, Guo B, Wilson PB, Stewart A, Byrne G, Bundred N,
Kumar S (2000) Plasma levels of soluble CD105 correlate with
metastasis in patients with breast cancer. Int J Cancer 89:122–
126
46. Levine RJ, Lam C, Qian C, Yu KF, Maynard SE, Sachs BP,
Sibai BM, Epstein FH, Romero R, Thadhani R, Karumanchi SA;
CPEP Study Group (2006) Soluble endoglin and other circu-
lating antiangiogenic factors in preeclampsia. N Engl J Med
355:992–1005
47. Velasco-Loyden G, Arribas J, Lo
´
pez-Casillas F (2004) The
shedding of betaglycan is regulated by pervanadate and medi-
ated by membrane type matrix metalloprotease-1. J Biol Chem
279:7721–7733
48. Yamashita H, Ichijo H, Grimsby S, More
´
n A, ten Dijke P, Mi-
yazono K (1994) Endoglin forms a heteromeric complex with
the signaling receptors for transforming growth factor-b. J Biol
Chem 269:1995–2001
49. Bork P, Sander C (1992) A large domain common to sperm
receptors (Zp2 and Zp3) and TGF-b type III receptor. FEBS Lett
300:237–240
50. Guerrero-Esteo M, Sanchez-Elsner T, Letamendia A, Bernabeu
C (2002) Extracellular and cytoplasmic domains of endoglin
interact with the transforming growth factor-b receptors I and II.
J Biol Chem 277:29197–29209
51. Llorca O, Trujillo A, Blanco FJ, Bernabeu C (2007) Structural
model of human endoglin, a transmembrane receptor responsi-
ble for hereditary hemorrhagic telangiectasia. J Mol Biol
365:694–705
52. Koleva RI, Conley BA, Romero D, Riley KS, Marto JA, Lux A,
Vary CP (2006) Endoglin structure and function: Determinants
of endoglin phosphorylation by transforming growth factor-b
receptors. J Biol Chem 281:25110–25123
53. Bello
´
n T, Corbı
´
A, Lastres P, Cale
´
s C, Cebria
´
n M, Vera S,
Cheifetz S, Massague J, Letarte M, Bernabe
´
u C (1993) Identi-
fication and expression of two forms of the human transforming
growth factor-b-binding protein endoglin with distinct cyto-
plasmic regions. Eur J Immunol 23:2340–2345
54. van Laake LW, van den Driesche S, Post S, Feijen A, Jansen
MA, Driessens MH, Mager JJ, Snijder RJ, Westermann CJ,
Doevendans PA, van Echteld CJ, ten Dijke P, Arthur HM,
Goumans MJ, Lebrin F, Mummery CL (2006) Endoglin has a
crucial role in blood cell-mediated vascular repair. Circulation
114:2288–2297
55. Li C, Issa R, Kumar P, Hampson IN, Lopez-Novoa JM, Ber-
nabeu C, Kumar S (2003) CD105 prevents apoptosis in hypoxic
endothelial cells. J Cell Sci 116:2677–2685
56. Scharpfenecker M, van Dinther M, Liu Z, van Bezooijen RL,
Zhao Q, Pukac L, Lo
¨
wik CW, ten Dijke P (2007) BMP-9 signals
via ALK1 and inhibits bFGF-induced endothelial cell prolifer-
ation and VEGF-stimulated angiogenesis. J Cell Sci 120:964–
972
57. Ota T, Fujii M, Sugizaki T, Ishii M, Miyazawa K, Aburatani H,
Miyazono K (2002) Targets of transcriptional regulation by two
distinct type I receptors for transforming growth factor-b in human
umbilical vein endothelial cells. J Cell Physiol 193:299–318
58. Li C, Guo B, Ding S, Rius C, Langa C, Kumar P, Bernabeu C,
Kumar S (2003) TNFa down-regulates CD105 expression in
vascular endothelial cells: a comparative study with TGFb1.
Anticancer Res 23:1189–1196
59. Botella LM, Sa
´
nchez-Elsner T, Rius C, Corbı
´
A, Bernabe
´
uC
(2001) Identification of a critical Sp1 site within the endoglin
promoter and its involvement in the transforming growth factor-
b stimulation. J Biol Chem 276:34486–34494
60. Sa
´
nchez-Elsner T, Botella LM, Velasco B, Langa C, Bernabe
´
u
C (2002) Endoglin expression is regulated by transcriptional
cooperation between the hypoxia and transforming growth fac-
tor-b pathways. J Biol Chem 277:43799–43808
61. Lastres P, Bellon T, Caban
˜
as C, Sanchez-Madrid F, Acevedo A,
Gougos A, Letarte M, Bernabeu C (1992) Regulated expression
on human macrophages of endoglin, an Arg-Gly-Asp-containing
surface antigen. Eur J Immunol 22:393–397
62. St-Jacques S, Forte M, Lye SJ, Letarte M (1994) Localization of
endoglin, a transforming growth factor-b binding protein, and of
CD44 and integrins in placenta during the first trimester of
pregnancy. Biol Reprod 51:405–413
63. Conley BA, Smith JD, Guerrero-Esteo M, Bernabeu C, Vary CP
(2000) Endoglin, a TGF-b receptor-associated protein, is
expressed by smooth muscle cells in human atherosclerotic
plaques. Atherosclerosis 153:323–335
64. Mancini ML, Verdi JM, Conley BA, Nicola T, Spicer DB,
Oxburgh LH, Vary CP (2007) Endoglin is required for myogenic
differentiation potential of neural crest stem cells. Dev Biol
308:520–533
65. Chen CZ, Li M, de Graaf D, Monti S, Go
¨
ttgens B, Sanchez MJ,
Lander ES, Golub TR, Green AR, Lodish HF (2002) Identifi-
cation of endoglin as a functional marker that defines long-term
repopulating hematopoietic stem cells. Proc Natl Acad Sci USA
99:15468–15473
66. Chen CZ, Li L, Li M, Lodish HF (2003) The endoglin(positive)
sca-1(positive) rhodamine(low) phenotype defines a near-
homogeneous population of long-term repopulating hemato-
poietic stem cells. Immunity 19:525–533
67. Perlingeiro RC (2007) Endoglin is required for hemangioblast
and early hematopoietic development. Development 34:3041–
3048
68. Moody JL, Singbrant S, Karlsson G, Blank U, Aspling M,
Flygare J, Bryder D, Karlsson S (2007) Endoglin is not critical
for hematopoietic stem cell engraftment and reconstitution but
regulates adult erythroid development. Stem Cells 25:2809–2819
69. Altomonte M, Montagner R, Fonsatti E, Colizzi F, Cattarossi I,
Brasoveanu LI, Nicotra MR, Cattelan A, Natali PG, Maio M
(1996) Expression and structural features of endoglin (CD105),
a transforming growth factor b1 and b3 binding protein, in
human melanoma. Br J Cancer 74:1586–1591
70. Henriksen R, Gobl A, Wilander E, Oberg K, Miyazono K, Funa
K (1995) Expression and prognostic significance of TGF-b
isotypes, latent TGF-b1 binding protein, TGF-b type I and type
II receptors, and endoglin in normal ovary and ovarian neo-
plasms. Lab Invest 73:213–220
71. Jovanovic B, Huang S, Liu YQ, Naguib KN, Bergan RC (2001)
Am J PharmacoGenomics 1:145–152
72. Liu Y, Jovanovic B, Pins M, Lee C, Bergan RC (2002) Over
expression of endoglin in human prostate cancer suppresses cell
detachment, migration and invasion. Oncogene 21:8272–8281
73. Craft CS, Romero D, Vary CP, Bergan RC (2007) Endoglin
inhibits prostate cancer motility via activation of the ALK2-
Smad1 pathway. Oncogene 26:7240–7250
Angiogenesis (2008) 11:79–89 87
123
74. Quintanilla M, Ramirez JR, Pe
´
rez-Go
´
mez E, Romero D, Ve-
lasco B, Letarte M, Lo
´
pez-Novoa JM, Bernabe
´
u C (2003)
Expression of the TGF-b coreceptor endoglin in epidermal
keratinocytes and its dual role in multistage mouse skin carci-
nogenesis. Oncogene 22:5976–5985
75. Lux A, Attisano L, Marchuk DA (1999) Assignment of trans-
forming growth factor b1 and b3 and a third new ligand to the
type I receptor ALK-1. J Biol Chem 274:9984–9992
76. Barbara NP, Wrana JL, Letarte M (1999) Endoglin is an
accessory protein that interacts with the signaling receptor
complex of multiple members of the transforming growth fac-
tor-beta superfamily. J Biol Chem 274:584–594
77. Goumans MJ, Valdimarsdottir G, Itoh S, Lebrin F, Larsson J,
Mummery C, Karlsson S, ten Dijke P (2003) Activin receptor-
like kinase (ALK)1 is an antagonistic mediator of lateral TGFb/
ALK5 signaling. Mol Cell 12:817–828
78. Guo B, Slevin M, Li C, Parameshwar S, Liu D, Kumar P,
Bernabeu C, Kumar S (2004) CD105 inhibits transforming
growth factor-b-Smad3 signalling. Anticancer Res 24:1337–
1345
79. Scherner O, Meurer SK, Tihaa L, Gressner AM, Weiskirchen R
(2007) Endoglin differentially modulates antagonistic trans-
forming growth factor-b1 and BMP-7 signaling. J Biol Chem
282:13934–13943
80. Lebrin F, Goumans MJ, Jonker L, Carvalho RL, Valdimarsdottir
G, Thorikay M, Mummery C, Arthur HM, ten Dijke P (2004)
Endoglin promotes endothelial cell proliferation and TGF-b/
ALK1 signal transduction. EMBO J 23:4018–4028
81. Li C, Hampson IN, Hampson L, Kumar P, Bernabeu C, Kumar S
(2000) CD105 antagonizes the inhibitory signaling of trans-
forming growth factor b1 on human vascular endothelial cells.
FASEB J 14:55–64
82. Blanco FJ, Santibanez JF, Guerrero-Esteo M, Langa C, Vary CP,
Bernabeu C (2005) Interaction and functional interplay between
endoglin and ALK-1, two components of the endothelial trans-
forming growth factor-b receptor complex. J Cell Physiol
204:574–584
83. She X, Matsuno F, Harada N, Tsai H, Seon BK (2004) Synergy
between anti-endoglin (CD105) monoclonal antibodies and
TGF-b in suppression of growth of human endothelial cells. Int J
Cancer 108:251–257
84. Fernandez-L A, Sanz-Rodriguez F, Zarrabeitia R, Pe
´
rez-Molino
A, Hebbel RP, Nguyen J, Bernabe
´
u C, Botella LM (2005) Blood
outgrowth endothelial cells from Hereditary Haemorrhagic
Telangiectasia patients reveal abnormalities compatible with
vascular lesions. Cardiovasc Res 68:235–248
85. Pece-Barbara N, Vera S, Kathirkamathamby K, Liebner S, Di
Guglielmo GM, Dejana E, Wrana JL, Letarte M (2005) En-
doglin null endothelial cells proliferate faster and are more
responsive to transforming growth factor b1 with higher affinity
receptors and an activated ALK1 pathway. J Biol Chem
280:27800–27808
86. Toporsian M, Gros R, Kabir MG, Vera S, Govindaraju K,
Eidelman DH, Husain M, Letarte M (2005) A role for endoglin
in coupling eNOS activity and regulating vascular tone revealed
in hereditary hemorrhagic telangiectasia. Circ Res 96:684–692
87. Santibanez JF, Blanco FJ, Garrido-Martin EM, Sanz-Rodriguez
F, Del Pozo MA, Bernabeu C (2008) Caveolin-1 interacts and
cooperates with the transforming growth factor-b type I receptor
ALK1 in endothelial caveolae. Cardiovasc Res (in press)
88. Razani B, Zhang XL, Bitzer M, von Gersdorff G, Bo
¨
ttinger EP,
Lisanti MP (2001) Caveolin-1 regulates transforming growth
factor (TGF)-b/SMAD signaling through an interaction with the
TGF-b type I receptor. J Biol Chem 276:6727–6738
89. Drab M, Verkade P, Elger M, Kasper M, Lohn M, Lauterbach B,
Menne J, Lindschau C, Mende F, Luft FC, Schedl A, Haller H,
Kurzchalia TV (2001) Loss of caveolae, vascular dysfunction,
and pulmonary defects in caveolin-1 gene-disrupted mice. Sci-
ence 293:2449–2452
90. Sanz-Rodriguez F, Guerrero-Esteo M, Botella LM, Banville D,
Vary CP, Bernabe
´
u C (2004) Endoglin regulates cytoskeletal
organization through binding to ZRP-1, a member of the Lim
family of proteins. J Biol Chem 271:32858–32868
91. Conley BA, Koleva R, Smith JD, Kacer D, Zhang D, Bernabe
´
u
C, Vary CP (2004) Endoglin controls cell migration and com-
position of focal adhesions: function of the cytosolic domain. J
Biol Chem 279:27440–27449
92. Lee NY, Blobe GC (2007) The interaction of endoglin with
beta-arrestin2 regulates transforming growth factor-b-mediated
ERK activation and migration in endothelial cells. J Biol Chem
282:21507–21517
93. Meng Q, Lux A, Holloschi A, Li J, Hughes JM, Foerg T,
McCarthy JE, Heagerty AM, Kioschis P, Hafner M, Garland JM
(2006) Identification of Tctex2b, a novel dynein light chain
family member that interacts with different transforming growth
factor-b receptors. J Biol Chem 281:37069–37080
94. Torsney E, Charlton R, Parums D, Collis M, Arthur HM (2002)
Inducible expression of human endoglin during inflammation
and wound healing in vivo. Inflamm Res 51:464–470
95. Jonker L, Arthur HM (2002) Endoglin expression in early
development is associated with vasculogenesis and angiogene-
sis. Mech Dev 110:193–196
96. Millan FA, Denhez F, Kondaiah P, Akhurst RJ (1991) Embry-
onic gene expression patterns of TGFb1, b2 and b3 suggest
different developmental functions in vivo. Development
111:131–143
97. Charng MJ, Frenkel PA, Lin Q, Yamada M, Schwartz RJ, Olson
EN, Overbeek P, Schneider MD (1998) A constitutive mutation
of ALK5 disrupts cardiac looping and morphogenesis in mice.
Dev Biol 199:72–79
98. Hirschi KK, Rohovsky SA, D’Amore PA (1998) PDGF, TGF-b,
and heterotypic cell-cell interactions mediate endothelial cell-
induced recruitment of 10T1/2 cells and their differentiation to a
smooth muscle fate. J Cell Biol 141:805–814
99. Carvalho RL, Jonker L, Goumans MJ, Larsson J, Bouwman P,
Karlsson S, ten Dijke P, Arthur HM, Mummery CL (2004)
Defective paracrine signalling by TGFb in yolk sac vasculature
of endoglin mutant mice: a paradigm for hereditary haemor-
rhagic telangiectasia. Development 131:6237–6247
100. Abdalla SA, Letarte M (2006) Hereditary haemorrhagic telan-
giectasia: current views on genetics and mechanisms of disease.
J Med Genet 43:97–110
101. Jerkic M, Rivas-Elena JV, Prieto M, Carro
´
n R, Sanz-Rodrı
´
guez
F, Pe
´
rez-Barriocanal F, Rodrı
´
guez-Barbero A, Bernabe
´
uC,
Lo
´
pez-Novoa JM (2004) Endoglin regulates nitric oxide-
dependent vasodilatation. FASEB J 18:609–611
102. Letteboer TG, Mager JJ, Snijder RJ, Koeleman BP, Lindhout D,
Ploos van Amstel JK, Westermann CJ (2006) Genotype-phe-
notype relationship in hereditary haemorrhagic telangiectasia. J
Med Genet 43:371–377
103. Gallione CJ, Repetto GM, Legius E, Rustgi AK, Schelley SL,
Tejpar S, Mitchell G, Drouin E, Westermann CJ, Marchuk DA
(2004) A combined syndrome of juvenile polyposis and hered-
itary haemorrhagic telangiectasia associated with mutations in
MADH4 (SMAD4). Lancet 363:852–859
104. Letarte M, McDonald ML, Li C, Kathirkamathamby K, Vera S,
Pece-Barbara N, Kumar S (2005) Reduced endothelial secretion
and plasma levels of transforming growth factor-b1 in patients
with hereditary hemorrhagic telangiectasia type 1. Cardiovasc
Res 68:155–164
105. Sibai B, Dekker G, Kupferminc M (2005) Pre-eclampsia. Lancet
365:785–799
88 Angiogenesis (2008) 11:79–89
123
106. Baxter JK, Weinstein L (2004) HELLP syndrome: the state of
the art. Obstet Gynecol Surv 59:838–845
107. Roberts JM, Taylor RN, Musci TJ, Rodgers GM, Hubel CA,
McLaughlin MK (1989) Preeclampsia: an endothelial cell dis-
order. Am J Obstet Gynecol 161:1200–1204
108. Fisher SJ (2004) The placental problem: linking abnormal
cytotrophoblast differentiation to the maternal symptoms of
preeclampsia. Reprod Biol Endocrinol 2:53
109. Levine RJ, Maynard SE, Qian C, Lim KH, England LJ, Yu KF,
Schisterman EF, Thadhani R, Sachs BP, Epstein FH, Sibai BM,
Sukhatme VP, Karumanchi SA (2004) Circulating angiogenic
factors and the risk of preeclampsia. N Engl J Med 350:672–683
110. Cruz-Gonzalez I, Pabo
´
n P, Rodrı
´
guez-Barbero A, Martı
´
n-
Moreiras J, Pericacho M, Sa
´
nchez PL, Ramirez V, Sa
´
nchez-
Ledesma M, Martı
´
n-Herrero F, Jime
´
nez-Candil J, Maree AO,
Sa
´
nchez-Rodrı
´
guez A, Martı
´
n-Luengo C, Lo
´
pez-Novoa JM
(2008) Identification of serum endoglin as a novel prognostic
marker after acute myocardial infarction. J Cell Mol Med (in
press)
111. Carmeliet P (2005) Angiogenesis in life, disease and medicine.
Nature 438:932–936
112. Hurwitz H, Fehrenbacher L, Novotny W, Cartwright T, Hains-
worth J, Heim W, Berlin J, Baron A, Griffing S, Holmgren E,
Ferrara N, Fyfe G, Rogers B, Ross R, Kabbinavar F (2004)
Bevacizumab plus irinotecan, fluorouracil, and leucovorin for
metastatic colorectal cancer. N Engl J Med 350:2335–2342
113. Wikstro
¨
m P, Lissbrant IF, Stattin P, Egevad L, Bergh A (2002)
Endoglin (CD105) is expressed on immature blood vessels and
is a marker for survival in prostate cancer. Prostate 51:268–275
114. Beresford MJ, Harris AL, Ah-See M, Daley F, Padhani AR,
Makris A (2006) The relationship of the neo-angiogenic marker,
endoglin, with response to neoadjuvant chemotherapy in breast
cancer. Br J Cancer 95:1683–1688
115. El-Gohary YM, Silverman JF, Olson PR, Liu YL, Cohen JK,
Miller R, Saad RS (2007) Endoglin (CD105) and vascular
endothelial growth factor as prognostic markers in prostatic
adenocarcinoma. Am J Clin Pathol 127:572–579
116. Brewer CA, Setterdahl JJ, Li MJ, Johnston JM, Mann JL,
McAsey ME (2000) Endoglin expression as a measure of
microvessel density in cervical cancer. Obstet Gynecol 96:224–
228
117. Seon BK, Matsuno F, Haruta Y, Kondo M, Barcos M (1997)
Long-lasting complete inhibition of human solid tumors in SCID
mice by targeting endothelial cells of tumor vasculature with
antihuman endoglin immunotoxin. Clin Cancer Res 3:1031–
1044
118. Bredow S, Lewin M, Hofmann B, Marecos E, Weissleder R
(2000) Imaging of tumour neovasculature by targeting the TGF-
b binding receptor endoglin. Eur J Cancer 36:675–681
119. Fonsatti E, Altomonte M, Nicotra MR, Natali PG, Maio M
(2003) Endoglin (CD105): a powerful therapeutic target on
tumor-associated angiogenetic blood vessels. Oncogene
22:6557–6563
120. Duff SE, Li C, Garland JM, Kumar S (2003) CD105 is important
for angiogenesis: evidence and potential applications. FASEB J
17:984–992
121. Maier JA, Delia D, Thorpe PE, Gasparini G (1997) In vitro
inhibition of endothelial cell growth by the antiangiogenic drug
AGM-1470 (TNP-470) and the anti-endoglin antibody TEC-11.
Anticancer Drugs 8:238–244
122. Takahashi N, Haba A, Matsuno F, Seon BK (2001) Antiangio-
genic therapy of established tumors in human skin/severe
combined immunodeficiency mouse chimeras by anti-endoglin
(CD105) monoclonal antibodies, and synergy between anti-en-
doglin antibody and cyclophosphamide. Cancer Res 61:7846–
7754
123. Tan GH, Wei YQ, Tian L, Zhao X, Yang L, Li J, He QM, Wu Y,
Wen YJ, Yi T, Ding ZY, Kan B, Mao YQ, Deng HX, Li HL,
Zhou CH, Fu CH, Xiao F, Zhang XW (2004) Active immuno-
therapy of tumors with a recombinant xenogeneic endoglin as a
model antigen. Eur J Immunol 34:2012–2021
124. Jiao JG, Li YN, Wang H, Liu Q, Cao JX, Bai RZ, Huang FY
(2006) A plasmid DNA vaccine encoding the extracellular
domain of porcine endoglin induces anti-tumour immune
response against self-endoglin-related angiogenesis in two liver
cancer models. Dig Liver Dis 38:578–587
125. Balza E, Castellani P, Zijlstra A, Neri D, Zardi L, Siri A (2001)
Lack of specificity of endoglin expression for tumor blood
vessels. Int J Cancer 94:579–585
126. Seon BK (2002) Expression of endoglin (CD105) in tumor
blood vessels. Int J Cancer 99:310–311
127. Kano MR, Bae Y, Iwata C, Morishita Y, Yashiro M, Oka M,
Fujii T, Komuro A, Kiyono K, Kaminishi M, Hirakawa K,
Ouchi Y, Nishiyama N, Kataoka K, Miyazono K (2007)
Improvement of cancer-targeting therapy, using nanocarriers for
intractable solid tumors by inhibition of TGF-b signaling. Proc
Natl Acad Sci USA 104:3460–3465
Angiogenesis (2008) 11:79–89 89
123
... This approach led to the identification of ENG, a downstream protein regulated by HDAC6. ENG, a glycosylated membrane protein, is specifically expressed in endothelial cells and is part of the TGF-β receptor superfamily [50]. It predominantly binds to the ligands BMP9 and BMP10 [51,52]. ...
... It predominantly binds to the ligands BMP9 and BMP10 [51,52]. ENG is known to play a crucial role in maintaining vascular integrity [50,53]. Homozygous mutations in mice result in embryonic lethality, and certain point mutations in human ENG are linked to hereditary hemorrhagic telangiectasia (HHT) [50]. ...
... ENG is known to play a crucial role in maintaining vascular integrity [50,53]. Homozygous mutations in mice result in embryonic lethality, and certain point mutations in human ENG are linked to hereditary hemorrhagic telangiectasia (HHT) [50]. However, the relationship between ENG, its ligand BMP9, and angiogenesis has been ambiguous, with conflicting reports regarding BMP9 ′ s role in an-giogenesis [54][55][56][57][58]. ...
Article
Full-text available
Histone deacetylase 6 (HDAC6) plays a crucial role in the acetylation of non-histone proteins and is notably implicated in angiogenesis, though its underlying mechanisms were previously not fully understood. This study conducted transcriptomic and proteomic analyses on vascular endothelial cells with HDAC6 knockdown, identifying endoglin (ENG) as a key downstream protein regulated by HDAC6. This protein is vital for maintaining vascular integrity and plays a complex role in angiogenesis, particularly in its interaction with bone morphogenetic protein 9 (BMP9). In experiments using human umbilical vein endothelial cells (HUVECs), the pro-angiogenic effects of BMP9 were observed, which diminished following the knockdown of HDAC6 and ENG. Western blot analysis revealed that BMP9 treatment increased SMAD1/5/9 phosphorylation, a process hindered by HDAC6 knockdown, correlating with reduced ENG expression. Mechanistically, our study indicates that HDAC6 modulates ENG transcription by influencing promoter activity, leading to increased acetylation of transcription factor SP1 and consequently altering its transcriptional activity. Additionally, the study delves into the structural role of HDAC6, particularly its CD2 domain, in regulating SP1 acetylation and subsequently ENG expression. In conclusion, the present study underscores the critical function of HDAC6 in modulating SP1 acetylation and ENG expression, thereby significantly affecting BMP9-mediated angiogenesis. This finding highlights the potential of HDAC6 as a therapeutic target in angiogenesis-related processes.
... Interestingly, our NanoString nCounter and immunoblot analysis showed that TGFβ was overexpressed in SUM102-MerTK clones ( Figure 4). TGFβ is a co-receptor of ENG and essential for angiogenesis as well as tumor vascularization [54,55]. Sakamoto et al. revealed that TGFβ induced overexpression of ENG in angiosarcoma cells [52], and we demonstrated here that MerTK induces overexpression of both ENG and TGFβ. ...
Article
Full-text available
Triple-negative breast cancer (TNBC) is characterized by the absence of the estrogen receptor, progesterone receptor, and receptor tyrosine kinase HER2 expression. Due to the limited number of FDA-approved targeted therapies for TNBC, there is an ongoing need to understand the molecular underpinnings of TNBC for the development of novel combinatorial treatment strategies. This study evaluated the role of the MerTK receptor tyrosine kinase on proliferation and invasion/metastatic potential in TNBC. Immunohistochemical analysis demonstrated MerTK expression in 58% of patient-derived TNBC xenografts. The stable overexpression of MerTK in human TNBC cell lines induced an increase in proliferation rates, robust in vivo tumor growth, heightened migration/invasion potential, and enhanced lung metastases. NanoString nCounter analysis of MerTK-overexpressing SUM102 cells (SUM102-MerTK) revealed upregulation of several signaling pathways, which ultimately drive cell cycle progression, reduce apoptosis, and enhance cell survival. Proteomic profiling indicated increased endoglin (ENG) production in SUM102-MerTK clones, suggesting that MerTK creates a conducive environment for increased proliferative and metastatic activity via elevated ENG expression. To determine ENG’s role in increasing proliferation and/or metastatic potential, we knocked out ENG in a SUM102-MerTK clone with CRISPR technology. Although this ENG knockout clone exhibited similar in vivo growth to the parental SUM102-MerTK clone, lung metastasis numbers were significantly decreased ~4-fold, indicating that MerTK enhances invasion and metastasis through ENG. Our data suggest that MerTK regulates a unique proliferative signature in TNBC, promoting robust tumor growth and increased metastatic potential through ENG upregulation. Targeting MerTK and ENG simultaneously may provide a novel therapeutic approach for TNBC patients.
... Two solitary LTR10A elements located in introns of endoglin (ENG) and placenta-specific CSF1R transcripts respectively could promote the expression of the two genes as enhancers in both EVT and SynT cell pools (Frost et al., 2023) (Supplementary Table S1). CSF1/CSF1R signaling could promote the growth, proliferation and migration of human trophoblast (Frost et al., 2023), and soluble ENG has been reported to be involved in the pathogenesis of preeclampsia by inducing endothelial cell dysfunction (ten Dijke et al., 2008). Another recent study has demonstrated that several LTR families (such as MER41A, MER50 and LTR8B) adjacent to genes upregulated specially in SynT tend to have increased H3K27ac and decreased H3K9me3 occupancy in SynT relative to human trophoblast stem cells (hTSCs) (Yu et al., 2023). ...
Article
Full-text available
Human endogenous retroviruses (HERVs) are derived from the infection and integration of exogenetic retroviruses. HERVs account for 8% of human genome, and the majority of HERVs are solitary LTRs (solo-LTRs) due to homologous recombination. Multiple findings have showed that solo-LTRs could provide an enormous reservoir of transcriptional regulatory sequences involved in diverse biological processes, especially carcinogenesis and cancer development. The link between solo-LTRs and human diseases still remains poorly understood. This review focuses on the regulatory modules of solo-LTRs, which contribute greatly to the diversification and evolution of human genes. More importantly, although inactivating mutations, insertions and deletions have been identified in solo-LTRs, the inherited regulatory elements of solo-LTRs initiate the expression of chimeric lncRNA transcripts, which have been reported to play crucial roles in human health and disease. These findings provide valuable insights into the evolutionary and functional mechanisms underlying the presence of HERVs in human genome. Taken together, in this review, we will present evidences showing the regulatory and encoding capacity of solo-LTRs as well as the significant impact on various aspects of human biology.
... Mutations in ENG lead to hereditary hemorrhagic telangiectasia (HHT), an autosomal dominant vascular disease 31,32 . ENG dysfunction contributes to tumor associated angiogenesis and inflammation [33][34][35][36] . Moreover, tumor-associated angiogenesis is induced by angiogenic factors, including VEGF and ENG, which are upregulated under hypoxic conditions [37][38][39][40] . ...
Article
Full-text available
Endothelial cells express neuropilin 1 (NRP1), endoglin (ENG) and vascular endothelial growth factor receptor 2 (VEGFR2), which regulate VEGF-A-mediated vascular development and angiogenesis. However, the link between complex formation among these receptors with VEGF-A-induced signaling and biology is yet unclear. Here, we quantify surface receptor interactions by IgG-mediated immobilization of one receptor, and fluorescence recovery after photobleaching (FRAP) measurements of the mobility of another coexpressed receptor. We observe stable ENG/NRP1, ENG/VEGFR2, and NRP1/VEGFR2 complexes, which are enhanced by VEGF-A. ENG augments NRP1/VEGFR2 interactions, suggesting formation of tripartite complexes bridged by ENG. Effects on signaling are measured in murine embryonic endothelial cells expressing (MEEC+/+) or lacking (MEEC-/-) ENG, along with NRP1 and/or ENG overexpression or knockdown. We find that optimal VEGF-A-mediated phosphorylation of VEGFR2 and Erk1/2 requires ENG and NRP1. ENG or NRP1 increase VEGF-A-induced sprouting, becoming optimal in cells expressing all three receptors, and both processes are inhibited by a MEK1/2 inhibitor. We propose a model where the maximal potency of VEGF-A involves a tripartite complex where ENG bridges VEGFR2 and NRP1, providing an attractive therapeutic target for modulation of VEGF-A signaling and biological responses.
... Eng экспрессируется зрелыми эндотелиальными клетками, гладкомышечными клетками сосудов, фибробластами в периваскулярной строме артерий, звездчатыми клетками печени, активированными моноцитами и макрофагами. Однако преобладание экспрессии эндоглина отмечается именно в клетках сосудистого русла [28]. ...
Article
Full-text available
Introduction. Endothelial dysfunction is a key link in the atherosclerotic process and therefore the study of specific markers may provide new opportunities for the development of therapeutic strategies for its correction. Aim of this work was to study the role of new biomarkers of endothelial dysfunction (endocan and endoglin) in early detection and prognosis of cardiovascular pathology. Material and methods. Meta-analysis of the literature sources in the scientific electronic bibliographic databases PubMed and Social Sciences Citation Index was carried out. Results and discussion. The meta-analysis demonstrated diagnostic and prognostic significance of endoglin and endocan in pathology of the cardiovascular system. Conclusion. Having reviewed the publications of leading scientific journals, it is clear that endothelium plays an important role both as a trigger and a catalyst in many cardiovascular diseases. The data presented in this review demonstrates the important role of new biomarkers of endothelial dysfunction, such as endocan and endoglin, in early diagnosis and determining the prognosis of cardiovascular pathology. Further exploration of options for their interpretation may be crucial for developing new therapeutic and diagnostic strategies
... CD105, a cell membrane glycoprotein, is the most important marker for MSCs [52]; however, CD105 acts as an accessory receptor for TGF-β [53][54][55], found overexpressed in actively proliferating endothelial cells, and it is considered as a powerful marker of new vessel formation. Together with CD34, CD105 can also be considered a marker for early stages of vascularization. ...
Article
Full-text available
Among all strategies directed at developing new tools to support re-vascularization of damaged tissues, the use of pro-angiogenic soluble factors, derived from mesenchymal stem cells (MSCs), appears a promising approach for regenerative medicine. Here, we compared the feasibility of two devices, generated by coupling soluble factors of human dental pulp mesenchymal stem cells (DPSCs), with a nanostructured scaffold, to support angiogenesis once transplanted in mice. DPSCs were obtained from impacted wisdom tooth removal, usually considered surgical waste material. After 28 days, we verified the presence of active blood vessels inside the scaffold through optical and scansion electron microscopy. The mRNA expression of surface antigens related to macrophage polarization (CD68, CD80, CD86, CD163, CD206), as well as pro-angiogenic markers (CD31, CD34, CD105, Angpt1, Angpt2, CDH5) was evaluated by real-time PCR. Our results demonstrate the capability of DPSC–scaffold and DPSC soluble factors–scaffold to support angiogenesis, similarly to adipose stem cells, whereas the absence of blood vessels was found in the scaffold grafted alone. Our results provide evidence that DPSC-conditioned medium can be proposed as a cell-free preparation able to support angiogenesis, thus, providing a relevant tool to overcome the issues and restrictions associated with the use of cells.
Article
Physiological embryogenesis and adult tissue homeostasis are regulated by transforming growth factor‐β (TGF‐β), an evolutionarily conserved family of secreted polypeptide factors, acting in an autocrine and paracrine manner. The role of TGF‐β in inflammation, fibrosis, and cancer is complex and sometimes even contradictory, exhibiting either inhibitory or promoting effects depending on the stage of the disease. Under pathological conditions, especially fibrosis and cancer, overexpressed TGF‐β causes extracellular matrix deposition, epithelial–mesenchymal transition, cancer‐associated fibroblast formation, and/or angiogenesis. In this review article, we have tried to dive deep into the mechanism of action of TGF‐β in inflammation, fibrosis, and carcinogenesis. As TGF‐β and its downstream signaling mechanism are implicated in fibrosis and carcinogenesis blocking this signaling mechanism appears to be a promising avenue. However, targeting TGF‐β carries substantial risk as this pathway is implicated in multiple homeostatic processes and is also known to have tumor‐suppressor functions. There is a need for careful dosing of TGF‐β drugs for therapeutic use and patient selection.
Preprint
Full-text available
Endothelial cells express neuropilin 1 (NRP1), endoglin (ENG) and vascular endothelial growth factor receptor 2 (VEGFR2), which regulate VEGF-A-mediated vascular development and angiogenesis. However, the link between complex formation among these receptors with VEGF-A-induced signaling and biology was unclear. Here, we quantified surface receptor interactions by IgG-mediated immobilization of one receptor, and fluorescence recovery after photobleaching (FRAP) measurements of the mobility of another coexpressed receptor. We observed stable ENG/NRP1, ENG/VEGFR2, and NRP1/VEGFR2 complexes, which were enhanced by VEGF-A. ENG augmented NRP1/VEGFR2 interactions, suggesting formation of tripartite complexes bridged by ENG. Effects on signaling were measured in murine embryonic endothelial cells expressing (MEEC+/+) or lacking (MEEC-/-) ENG, along with NRP1 overexpression or knockdown. Optimal VEGF-A-mediated phosphorylation of VEGFR2 and Erk1/2 required ENG and NRP1. ENG or NRP1 increased VEGF-A-induced sprouting, becoming optimal in cells expressing all three receptors, and both processes were inhibited by a MEK1/2 inhibitor. We propose a model where the maximal potency of VEGF-A involves a tripartite complex where ENG bridges VEGFR2 and NRP1, providing an attractive therapeutic target for modulation of VEGF-A signaling and biological responses.
Article
Full-text available
CD105 (endoglin), a receptor for transforming growth factor (TGF) β1 and β3 in vascular endothelial cells, is highly up‐regulated in blood vessels of tissues where neovascularisation occurs. It modulates endothelial‐mesenchymal signalling and is essential for angiogenesis. Indeed, CD105 knock‐out mice die from malvascularisation by 11.5 day p.c. In the present study CD105, TGFβ1 and CD105/TGFβ1 complexes were quantified in plasma samples from 77 healthy individuals and 92 patients with early stage breast cancer prior to any treatment. When compared with normal controls, both CD105 and CD105/TGFβ1 complex levels were significantly elevated in breast cancer patients, whereas TGFβ1 levels were lower in cancer patients. The most important finding to emerge was that CD105 levels were significantly increased in patients who developed distant metastasis compared with disease‐free patients. While there was no significant difference between CD105 levels in controls compared to disease‐free patients, it was significantly higher in patients with metastatic disease. Thus patients who had died following local relapse or distant metastases possessed the highest levels of CD105. Neither CD105/TGFβ1 complex nor TGFβ1 levels correlated with tumour progression. Our data indicate that CD105 might be a valuable novel angiogenic marker for identifying breast cancer patients who are at high risk of developing metastasis. Int. J. Cancer (Pred. Oncol.) 89:122–126, 2000. © 2000 Wiley‐Liss, Inc.
Article
Full-text available
Human endoglin is a dimeric protein that binds transforming growth factor-beta (TGF-beta). A porcine cDNA clone for endoglin was obtained from a porcine uterus cDNA library. The deduced sequence of the primary translated product of endoglin consists of 643 amino acids with a high sequence identity (96%) to human endoglin in the transmembrane and intracellular domains, but with a lower sequence similarity (66%) in the extracellular domain. In contrast to human endoglin, porcine endoglin has no Arg-Gly-Asp tripeptide in its sequence. Antibodies, raised against a peptide corresponding to the intracellular domain of porcine endoglin, immunoprecipitated an 84-kDa protein under reducing condition and a 130-kDa protein under nonreducing condition in porcine aortic endothelial cells. Porcine endoglin bound TGF-beta1 and -beta3 efficiently, but TGF-beta2 less efficiently. Endoglin was found to be coimmunoprecipitated with TGF-beta receptors type I and/or II by the endoglin antibodies or by TGF-beta receptor II antibodies in the presence of ligand. Thus, endoglin and TGF-beta receptors I and/or II most likely formed a heteromeric receptor complex. Endoglin was phosphorylated on serine residue(s), which did not change after stimulation by TGF-beta1. These results revealed that endoglin is a phosphorylated protein which forms a heteromeric complex with signaling receptors for TGF-beta.
Article
Full-text available
Endoglin, a dimeric membrane glycoprotein expressed at high levels on human vascular endothelial cells, shares regions of sequence identity with beta-glycan, a major binding protein for transforming growth factor-beta (TGF-beta) that co-exists with TGF-beta receptors I and II in a variety of cell lines but is low or absent in endothelial cells. We have examined whether endoglin also binds TGF-beta and demonstrate here that the major TGF-beta1-binding protein co-existing with TGF-beta receptors I and II on human umbilical vein endothelial cells is endoglin, as determined by specific immunoprecipitation of endoglin affinity-labeled with I-125-TGF-beta. Furthermore, endoglin ectopically expressed in COS cells binds TGF-beta1. Competition affinity-labeling experiments showed that endoglin binds TGF-beta1 (K(D) approximately 50 pM) and TGF-beta3 with high affinity but fails to bind TGF-beta2. This difference in affinity of endoglin for the TGF-beta isoforms is in contrast to beta-glycan which recognizes all three isoforms. TGF-beta however is binding with high affinity to only a small fraction of the available endoglin molecules, suggesting that some rate-limiting event is required to sustain TGF-beta binding to endoglin.
Article
Pre-eclampsia is a major cause of maternal mortality (15-20% in developed countries) and morbidities (acute and long-term), perinatal deaths, preterm birth, and intrauterine growth restriction. Key findings support a causal or pathogenetic model of superficial placentation driven by immune maladaptation, with subsequently reduced concentrations of angiogenic growth factors and increased placental debris in the maternal circulation resulting in a (mainly hypertensive) maternal inflammatory response. The final phenotype, maternal pre-eclamptic syndrome, is further modulated by pre-existing maternal cardiovascular or metabolic fitness. Currently, women at risk are identified on the basis of epidemiological and clinical risk factors, but the diagnostic criteria of pre-eclampsia remain unclear, with no known biomarkers. Treatment is still prenatal care, timely diagnosis, proper management, and timely delivery. Many interventions to lengthen pregnancy (eg, treatment for mild hypertension, plasma-volume expansion, and corticosteroid use) have a poor evidence base. We review findings on the diagnosis, risk factors, and pathogenesis of pre-eclampsia and the present status of its prediction, prevention, and management.
Article
Article
Endoglin is an integral membrane glycoprotein that binds transforming growth factor-beta 1 (TGF beta 1) with high affinity and is predominantly expressed on human endothelial cells. Characterization of this homodimeric protein from human term placenta has shown that it is particularly abundant on the syncytiotrophoblast. Immunofluorescence staining of sections of first trimester placenta now reveals that endoglin is found at even higher levels on the syncytiotrophoblast of samples ranging from 6 to 12 wk of gestation. Very low levels are observed on the undifferentiated cytotrophoblast cells that can be identified by their expression of the alpha 6 beta 4 integrin, a receptor for laminin. Within the villi, blood vessels and stromal cells are negative for endoglin but positive for alpha 1 beta 1 integrin, a receptor for collagens and laminin. Stromal cells also express CD44, a hyaluronic acid receptor. Of particular interest is the up-regulation of endoglin expression in the transition from polarized undifferentiated to non-polarized intermediate cytotrophoblasts (CTB) as the cells align in columns to invade the uterus. This occurs in parallel with the acquisition of alpha 5 beta 1 integrin (fibronectin receptor) and precedes the loss of alpha 6 beta 4 integrin. CD44 and alpha 1 beta 1 integrin are noticeably absent from the CTB within the columns but are expressed at very high levels throughout the placental bed. Endoglin is undetectable within the decidua; thus, intermediate CTB that have invaded the placental bed express alpha 5 beta 1 integrin and cytokeratins but not endoglin.(ABSTRACT TRUNCATED AT 250 WORDS)
Article
Endoglin is a transforming growth factor–β (TGF-β) binding protein expressed on the surface of endothelial cells. Loss-of-function mutations in the human endoglin gene ENGcause hereditary hemorrhagic telangiectasia (HHT1), a disease characterized by vascular malformations. Here it is shown that by gestational day 11.5, mice lacking endoglin die from defective vascular development. However, in contrast to mice lacking TGF-β, vasculogenesis was unaffected. Loss of endoglin caused poor vascular smooth muscle development and arrested endothelial remodeling. These results demonstrate that endoglin is essential for angiogenesis and suggest a pathogenic mechanism for HHT1.