You are on page 1of 9

ARTICLES

Soluble endoglin contributes to the pathogenesis


© 2006 Nature Publishing Group http://www.nature.com/naturemedicine

of preeclampsia
Shivalingappa Venkatesha1,6, Mourad Toporsian2,6, Chun Lam1,6, Jun-ichi Hanai1, Tadanori Mammoto1,
Yeon M Kim3, Yuval Bdolah1, Kee-Hak Lim1, Hai-Tao Yuan1, Towia A Libermann1, Isaac E Stillman1,
Drucilla Roberts4, Patricia A D’Amore5, Franklin H Epstein1, Frank W Sellke1, Roberto Romero3,
Vikas P Sukhatme1, Michelle Letarte2 & S Ananth Karumanchi1

Preeclampsia is a pregnancy-specific hypertensive syndrome that causes substantial maternal and fetal morbidity and mortality.
Maternal endothelial dysfunction mediated by excess placenta-derived soluble VEGF receptor 1 (sVEGFR1 or sFlt1) is emerging
as a prominent component in disease pathogenesis. We report a novel placenta-derived soluble TGF-b coreceptor, endoglin (sEng),
which is elevated in the sera of preeclamptic individuals, correlates with disease severity and falls after delivery. sEng inhibits
formation of capillary tubes in vitro and induces vascular permeability and hypertension in vivo. Its effects in pregnant rats are
amplified by coadministration of sFlt1, leading to severe preeclampsia including the HELLP (hemolysis, elevated liver enzymes,
low platelets) syndrome and restriction of fetal growth. sEng impairs binding of TGF-b1 to its receptors and downstream signaling
including effects on activation of eNOS and vasodilation, suggesting that sEng leads to dysregulated TGF-b signaling in the
vasculature. Our results suggest that sEng may act in concert with sFlt1 to induce severe preeclampsia.

Preeclampsia complicates 5% of all pregnancies worldwide and is a of preeclampsia, suggesting that excess circulating sFlt1 may have a
major cause of maternal, fetal and neonatal mortality, especially in causal role in preeclampsia4. sFlt1-treated animals, however, do not
developing nations. This condition is characterized by the onset of develop hemolysis and thrombocytopenia, signs observed in the
hypertension and proteinuria in the third trimester of pregnancy1. HELLP syndrome, a subtype of severe preeclampsia2,12. Therefore,
Severe preeclampsia can lead to the appearance of the HELLP we hypothesized that other placenta-derived soluble factors may act
syndrome, seizures (eclampsia) and/or restriction of fetal growth1,2. in concert with sFlt1 to cause endothelial dysfunction, resulting in
The placenta has a central role in preeclampsia, as evidenced by rapid severe preeclampsia.
disappearance of disease symptoms after delivery. The clinical man- Endoglin (Eng) or CD105, a cell-surface coreceptor for trans-
ifestations of preeclampsia reflect widespread endothelial dysfunction, forming growth factor (TGF)-b1 and TGF-b3 isoforms, is highly
resulting in vasoconstriction, end-organ ischemia and increased expressed in endothelial cells and syncytiotrophoblasts and
vascular permeability1. Therefore, it has been suggested that modulates actions of TGF-b1 and TGF-b3 (refs. 13–15). Mutations
placenta-derived circulating factor(s) may induce the endothelial in the gene encoding Eng, ENG, are the underlying cause of
defects leading to preeclampsia3. hereditary hemorrhagic telangiectasia type 1 (HHT1), an autosomal
We and others recently showed that high levels of circulating dominant disorder characterized by arteriovenous malformations
sFlt1 (soluble fms-like tyrosine kinase, also known as soluble VEGF and focal loss of capillaries16. Eng–/– mice die at midgestation
receptor 1) of placental origin may contribute to the pathogenesis of because of defective angiogenesis and cardiovascular development,
preeclampsia4–7. sFlt1 binds to and neutralizes the proangiogenic whereas Eng+/– mice develop signs of HHT17,18. Recently, it was
actions of VEGF and placental growth factor (PlGF). High concentra- found that Eng localizes to caveolae, where it can associate
tions of circulating sFlt1 along with decreased free VEGF and free PlGF with endothelial nitric oxide synthase (eNOS) and regulate its activity
are seen not only during preeclampsia but before the onset of clinical and local vascular tone19. These data suggest the involvement
symptoms4,8–11. Overexpression of sFlt1 in rats leads to hypertension, of Eng not only in cardiovascular development but also in
proteinuria and glomerular endotheliosis, the classical manifestations vascular homeostasis.

1Center for Vascular Biology, Departments of Medicine, Obstetrics and Gynecology, Surgery, and Pathology, Beth Israel Deaconess Medical Center and Harvard Medical
School, 330 Brookline Avenue, Boston, Massachusetts 02215, USA. 2Cancer Research Program, Hospital for Sick Children, and The Heart and Stroke Foundation
Richard Lewar Centre of Excellence, University of Toronto, 555 University Avenue, Toronto, Ontario M5G 1X8, Canada. 3Perinatology Research Branch, National
Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, and Wayne State
University School of Medicine, 3980 John R, Detroit, Michigan 48201, USA. 4Department of Pathology, Massachusetts General Hospital and Harvard Medical School,
55 Fruit Street, Boston, Massachusetts 02114, USA. 5Schepens Eye Research Institute and Harvard Medical School, 20 Staniford Steet, Boston, Massachusetts
02114, USA. 6These authors contributed equally to this work. Correspondence should be addressed to S.A.K. (sananth@bidmc.harvard.edu).
Received 9 March; accepted 5 May; published online 4 June 2006; corrected after print 16 June 2006; doi:10.1038/nm1429

642 VOLUME 12 [ NUMBER 6 [ JUNE 2006 NATURE MEDICINE


ARTICLES

Figure 1 Expression of ENG mRNA and Eng


a b

ia

ia

ia
si
in placentae of normal and preeclamptic

ps

ps

ps
p
am

am

am

am
Normal Preeclampsia

al

Pr l
a
pregnancies. (a) Northern blot analysis for ENG

cl

cl

cl

cl
m

m
ee

ee

ee

ee
or

or
Pr

Pr

Pr
N

N
mRNA from normal and preeeclamptic placentae
5 kb
with their associated gestational ages (GA).
ENG
2 kb 18S ribosomal RNA levels were used as loading
controls. (b) Double immunofluorescence staining
18S of Eng (red) and smooth muscle actin (green) is
shown for preeclamptic placentae and corres-
GA (weeks) 31.3 33.0 30.1 39.2 38.1 38.5
25.6 weeks GA 25.2 weeks GA ponding control placentae derived from individuals
c Normal Preeclampsia with preterm labor. Original magnification,
© 2006 Nature Publishing Group http://www.nature.com/naturemedicine

200. (c) Representative western blot of Eng


(kDa)
90
immunoprecipitates shows full-length Eng
Eng
65
(90 kDa) and a smaller fragment (65 kDa), which
sEng
is more noticeable in preeclamptic placentae.
The normal and preeclamptic samples represent
44 β-actin
individual women. Equal loading for the lysates
GA (weeks) 39.4 30.1 34.1 38.1 28.0 33.0 40.5 weeks GA 41.6 weeks GA used for immunoprecipitation was confirmed by
actin western blots on the same lysates.

eNOS is a Ca2+/calmodulin-regulated nitric oxide (NO) synthase soluble form of Eng (sEng) may be produced by the placenta.
that can be activated by fluid shear stress and neurohumoral stimuli. Expression of sEng in preeclamptic placentae was fourfold higher
Endothelium-derived NO is a potent vasorelaxant that contributes to than in normal pregnancy (n ¼ 10 per group, P o 0.01). Although
the regulation of systemic blood pressure, vascular permeability and both Eng and sEng were upregulated in preeclampsia, quantification
angiogenesis. Indeed, the effects of VEGF on angiogenesis and vascular of the sEng /Eng ratio in these placental specimens showed no
tone are partly mediated by activation of eNOS, through Akt- significant difference between normal and preeclamptic samples,
dependent phosphorylation of eNOS Ser1177 (ref. 20) and increased suggesting that both Eng and sEng are proportionally increased
association of eNOS with Hsp90 (ref. 21). Our recent demonstration in preeclampsia.
that placenta-derived sFlt1 in sera of preeclamptic individuals can
inhibit angiogenesis and induce hypertension4 may in fact reflect Elevated serum sEng correlates with disease severity
impairment of VEGF-dependent activation of eNOS. The regulation After immunoblotting Eng immunoprecipitates for Eng from
of eNOS activity is complex, involving dynamic regulation of its sera of preeclamptic individuals, we observed a single 65 kDa band
phosphorylation status. While phosphorylation at Ser1177 is indica- (Fig. 2a). This protein was present at much lower levels in the sera of
tive of agonist-induced eNOS activation, it is preceded by depho- normal pregnant women and barely detectable in nonpregnant
sphorylation at Thr495. Coordination of these reactions determines women. Purification from sera of preeclamptic individuals and
the activity of eNOS in endothelial cells22. analysis by mass spectrometry showed several Eng-specific peptides
We now report a novel placenta-derived 65 kDa soluble form of Eng ranging from Gly27 to Arg393 (Fig. 2b), indicating a soluble
(sEng) present in sera of pregnant women, elevated in preeclamptic form (sEng) corresponding to the N-terminal region of the full-
individuals and increased with disease severity. sEng cooperated with length protein.
sFlt1 to induce endothelial dysfunction in vitro and severe preeclamp- Our finding of elevated sEng concentrations in sera of preeclamptic
sia-like illness in vivo. We show that sEng interferes with individuals suggested that this may serve as diagnostic or prognostic
TGF-b1 signaling and eNOS activation in endothelial cells, thereby marker for the disease. Quantification by ELISA of serum sEng
disrupting key homeostatic mechanisms necessary for maintenance concentrations showed a two- and threefold increase in preterm and
of vascular health. term pregnancy, respectively, compared to nonpregnant states
(Fig. 2c), suggesting a role for sEng during normal pregnancy.
RESULTS Concentrations of sEng were three-, five- and tenfold higher in
Upregulation of Eng in preeclamptic placenta individuals with mild preeclampsia, severe preeclampsia and HELLP
To identify novel placenta-derived factors involved in preeclampsia, we syndrome, respectively, compared to gestational age–matched preterm
performed gene expression profiling of placental tissue from pregnant controls (Fig. 2c). The clinical characteristics of these individuals are
women with or without preeclampsia using Affymetrix U95A micro- shown in Supplementary Table 1 online. Concentrations of sEng in
array chips. In addition to marked upregulation of mRNA encoding pregnant individuals correlated with those of sFlt1 (R2 ¼ 0.56) except
sFlt1 and Flt1 in preeclamptic compared to normal gestational age- in the HELLP syndrome group, in which the level of sEng was higher
matched placentae4, we noted a fourfold increase in ENG mRNA than that of sFlt1. Blood samples from a subset of women obtained
which was confirmed by northern blot analysis (Fig. 1a). Unlike 48 h after placental delivery showed a 70% reduction in mean sEng
sFlt1, however, no shorter, alternately spliced ENG mRNA was circulating levels in preeclamptic compared to normal pregnant
detected. Immunostaining of placental sections confirmed enhanced women (Fig. 2d). Although these data suggest that the placenta is a
expression of Eng, particularly on the syncytiotrophoblasts of pre- major source of sEng during pregnancy, other sources such as
eclamptic placentae at 25 and 40 weeks relative to age-matched control maternal vasculature cannot be ruled out.
pregnancies (Fig. 1b). Western blot analysis of Eng immunoprecipi-
tates of normal and preeclamptic placentae showed the expected Effects of sEng on endothelial function
90 kDa Eng monomer, characteristic of the integral plasma membrane TGF-b and VEGF are crucial factors in angiogenesis. Recombinant
protein in both groups, albeit at higher levels in preeclamptic samples sEng inhibited endothelial tube formation on Matrigel in vitro
(Fig. 1c). We also observed a smaller 65 kDa band, suggesting that a to the same extent as sFlt1. Moreover, sEng potentiated the

NATURE MEDICINE VOLUME 12 [ NUMBER 6 [ JUNE 2006 643


ARTICLES

a Recomb.
sEng H
Non-
Normal Preeclampsia
b Recomb.
sEng Fraction # c *#
pregnant 100 sEng
(kDa) (kDa) 3 4 5 6

Concentration in serum (ng/ml)


116 116 sFlt1

82 82 80
sEng 64 sEng *
64 *
49 49 60 *
37
20 †† *
*
sEng level (units of optical density)

** 40

16
1 GASCSLSPTS LAETVHCDLQ PVGPERGEVT YTTSQVSKGC VAQAPNAILE 20
51 VHVLFLEFPT GPSQLELTLQ ASKQNGTWPR EVLLVLSVNS SVFLHLQALG
101
1 IPLHLAYNSS LVTFQEPPGV NTTELPSFPK TQILEWAAER GPITSAAELN
© 2006 Nature Publishing Group http://www.nature.com/naturemedicine

12
151 DPQSILLRLG QAQGSLSFCM LEASQDMGRT LEWRPRTPAL VRGCHLEGVA
201 GHKEAHILRV LPGHSAGPRT VTVKVELSCA PGDLDAVLIL QGPPYVSWLI 0
251 DANHNMQIWT TGEYSFKIFP EKNIRGFKLP DTPQGLLGEA RMLNASIVAS Non- Normal Normal Mild Severe HELLP
8 ## 301 FVELPLASIV SLHASSCGGR LQTSPAPIQT TPPKDTCSPE LLMSLIQTKC pregnant term preterm PE PE
351 ADDAMTLVLK KELVAHLKCT ITGLTFWDPS CEAEDRGDKF VLRSAYSSCG
401 MQVSASMISN EAVVNILSSS SPQRKKVHCL NMDSLSFQLG LYLSPHFLQA
4 451
501
SNTIEPGQQS
GNCVSLLSPS
FVQVRVSPSV
PEGDPRFSFL
SEFLLQLDSC
LHFYTVPIPK
HLDLGPEGGT
TGTLSCTVAL
VELIQGRAAK
RPKTGSQDQE
d
551 VHRTVFMRLN IISPDLSGCT SKGLVLPAVL GITFGAFLIG ALLTAALWYI
T=0

sEng concentration in serum (ng/ml)


601 YSHTRSPSKR EPVVAVAAPA SSESSSTNHS IGSTQSTPCS TSSMA 100
0 T = 48
Non- Normal Preeclampsia
pregnant 80
Figure 2 Increased sEng levels in sera from individuals with preeclampsia. (a) Representative western
blot and graph of Eng immunoprecipitates showing higher level of a soluble 65kDa fragment (sEng) in 60
sera of preeclamptic women (at 28, 33 and 32.2 weeks) compared to normal pregnant women (at 34.1,
28.2 and 39.4 weeks; **P o 0.01) and nonpregnant women (wwP o 0.01; n ¼ 3/group). sEng levels in 40
*
normal pregnant women were higher than in nonpregnant individuals (##P o 0.01). HUVEC (H) extracts
and recombinant human sEng served as positive controls (90 kDa and 75 kDa, respectively). Individuals 20
matched for gestational ages were randomly chosen from normal (term and pre-term) and preeclampsia *
(severe and HELLP) groups described in Supplementary Table 1. (b) sEng was purified from the serum of 0
Normal Preeclampsia
preeclamptic individuals. Fractions 4 and 5 eluted from the 44G4-IgG (Eng-specific) Sepharose, were
run on SDS-PAGE under reducing conditions and tested by western blot using a polyclonal antibody to Eng. The eluted fractions were subjected to mass
spectrometry analysis (three runs), and the peptides identified are in boldface and underlined in the sequence of human Eng. The underlined amino acids
562–586 represent the transmembrane domain of human cell-surface Eng. Sera from ten random individuals with preeclampsia (severe and HELLP) in
Supplementary Table 1 were pooled. (c) ELISA results for sEng and sFlt1 in sera of individuals with varying degrees of preeclampsia, control pregnancies
and four nonpregnant healthy volunteers. *P o 0.05 compared to preterm controls, #P o 0.05 compared to severe preeclampsia. All individuals described
in Supplementary Table 1 were analyzed. (d) ELISA results for sEng in a subset of pregnant individuals (normal, n ¼ 6; preeclampsia, n ¼ 11) described in
c with blood drawn before (0–12 h) or after (48 h) delivery. *P o 0.05 as compared to T ¼ 0 samples. All individuals described in Supplementary Table 1
from whom we were able to obtain a serum specimen 48 h after delivery were analyzed.

antiangiogenic actions of sFlt1, as seen by a larger reduction in the than that observed with sFlt1 (Table 1). Proteinuria was modest in
number of capillary-like structures in the presence of both proteins sEng-treated rats, but severe in the sFlt1-treated group. The
(Fig. 3a). This suggests that sEng and sFlt1 block the proangio- sFlt1+sEng group showed nephrotic-range proteinuria, severe hyper-
genic effects of TGF-b1 and VEGF, respectively, present in Matrigel. tension and biochemical evidence of HELLP syndrome (elevated
Capillary permeability is increased in pre-
eclampsia23, and preeclamptic individuals are
more prone to develop peripheral and pul- a b
monary edema. Therefore, we tested the role
Control
of sEng and sFlt1 in microvascular perme- sEng
#
ability using the Evans blue assay in BALB/c * sFlt1
mice pretreated with adenovirus expressing 0.8 sEng+sFlt1

sFlt1, sEng, sFlt1+sEng or mouse Fc protein


**
(optical units of density)

*
Absorbance 620 nm

Control: 139,249 sEng: 75,515 0.6


as a negative control. Capillary permeability * *
was increased by sEng and sFlt1 in the lungs, *
0.4
liver and kidneys (Fig. 3b). Notably, the
combination of sEng and sFlt1 showed an
0.2
additive effect in the liver, indicating that
these soluble receptors may act in concert to
0.0
disrupt endothelial integrity and induce con- Lung Liver Kidney
sFlt1: 74,570 sEng+sFlt1: 38,989
siderable vascular damage and leak.
Figure 3 sEng inhibits capillary formation and increases vascular permeability. (a) Angiogenesis assays
In vivo effects of sEng and sFlt1 were done using human umbilical vein endothelial cells (HUVECs) in growth factor–reduced Matrigel
To induce the clinical features of preeclamp- and performed in the presence of 1 mg of recombinant sEng, sFlt1 or both, and endothelial tube
sia in pregnant rats, we used adenoviral lengths quantified. A representative experiment (n ¼ 4) is shown, with average tube lengths (in pixels)
expression of sEng and sFlt1, alone or in indicated below the panels. Tube length results were quantified, showing a 62.2% ± 3.9, 62% ± 4.1
and 39.7% ± 4.2 reduction in sEng, sFlt1 and sFlt1+sEng, respectively (P o 0.01 versus controls).
combination. At 17–18 d of pregnancy, (b) Microvascular permeability assessed by Evans blue leakage in mice injected with adenovirus
hemodynamic and biochemical data indi- containing Fc (Control), sEng, sFlt1 or sFlt1+sEng. Leakage of Evans blue was quantified in the various
cated that sEng induced a significant change organs. Data represent a mean of four independent experiments. *P o 0.05 compared to controls,
in mean arterial pressure (MAP), albeit lower #P o 0.05 compared to sFlt1 alone.

644 VOLUME 12 [ NUMBER 6 [ JUNE 2006 NATURE MEDICINE


ARTICLES

Table 1 Hemodynamic and biochemical data in pregnant rats

Groups n MAPa (mmHg) Urine albumin/creatinineb (mg/mg) Platelet count ( 1,000/ml) LDH (U/L) AST (U/L) Fetal weightb (g)

Control (CMV) 6 83 ± 5 186 ± 94 1,098 ± 75 156 ± 32 54 ± 4 2.1 ± 0.5


sEng 6 104 ± 6* 432 ± 249 1,195 ±78 188 ± 46 110 ± 13* 1.6 ± 0.4
sFlt1 6 117 ± 7* 2,295 ± 867* 1,131 ± 91 172 ± 53 94 ± 4* 1.75 ± 0.4
sFlt1+sEng 6 121 ± 9* 9,029 ± 4,043* 615 ± 67* 1,952 ± 784* 210 ± 92* 0.75 ± 0.3*

Data are presented as mean ± s.e.m. *P o 0.05 compared to control group. aMAP ¼ diastolic pressure + 1/3 pulse pressure.bFetal weight is the average weight of the litter for each group.
Expression of sFlt1 and sEng were first confirmed in rat plasma by western blots (Supplementary Fig. 3) and circulating concentrations quantified using commercially available ELISA kits. Mean
plasma concentrations of sFlt1 in the control, sEng, sFlt1 and sFlt1+sEng groups were 0.64 ng/ml, 0.66 ng/ml, 249 ng/ml and 204 ng/ml, respectively. Similarly, the concentrations of sEng in
© 2006 Nature Publishing Group http://www.nature.com/naturemedicine

these four groups were 0.39 ng/ml, 129 ng/ml, 0.37 ng/ml and 123 ng/ml, respectively.

lactate dehydrogenase (LDH) and aspartate aminotransferase (AST), sEng together into nonpregnant rats, suggesting that the phenotype in
and decreased platelet counts). We observed restriction of fetal growth pregnant rats resulted from a direct effect on the maternal vessels and
in litters born to the sFlt1+sEng group, probably related to the does not require the placenta (data not shown).
placental vascular ischemia and damage (Table 1).
Renal histology by light and electron microscopy showed sEng inhibits TGF-b–mediated NOS-dependent vasodilation
focal endotheliosis in the sEng group compared to the control Given the known effect of VEGF on reducing vascular reactivity
group (Fig. 4a and Supplementary Fig. 1 online). Severe glomerular through activation of eNOS and the recent demonstration that Eng
endotheliosis was observed in the sFlt1 and the sFlt1+sEng groups modulates eNOS-dependent vasomotor activity19, we assessed the
(Fig. 4a and Supplementary Fig. 1). We observed extensive vascular hemodynamic effects of TGF-b isoforms and sEng on isolated rat
damage of the placenta, including infarction at the maternal-fetal renal microvessels. Both TGF-b1 and TGF-b3 induced a dose-depen-
junction in the sFlt1+sEng group, but not in control rats or in those dent increase in arterial diameter (Fig. 5a), whereas TGF-b2, which is
treated with either agent (Fig. 4b). We noted diffuse inflammation in not a ligand for Eng, did not produce significant vasodilation (o2%
the giant-cell layer (corresponding to human invasive trophoblasts) at 0.1 and 1 mg/ml). sEng significantly attenuated the effects of TGF-b1
in the sFlt1 and sEng groups, and increased inflammation in the and TGF-b3 (Fig. 5a). This acute effect of TGF-b1 and TGF-b3
sFlt1+sEng group compared to either the sFlt1 or sEng groups. Liver isoforms on vascular tone has not been previously recognized to our
histology showed signs of ischemia and areas of necrosis in the knowledge and was also seen in mesenteric vessels (Supplementary
sFlt1+sEng group, similar to those seen in individuals with the Fig. 2 online). VEGF and TGF-b1 had additive effects on vasodilation,
HELLP syndrome (Fig. 4c). Evidence of hemolysis in the sFlt1+sEng which were blocked by the combination of sEng and sFlt1 at concen-
group was confirmed in peripheral blood smears, in which we trations noted in individuals with preeclampsia (Fig. 5b). L-NAME
observed schistocytes and reticulocytosis (Fig. 4d). We also saw blocked the vasodilation mediated by TGF-b1 and VEGF, indicating a
signs of severe maternal vascular damage after injection of sFlt1 and NOS-dependent response (Fig. 5b). These data suggest that circulating

Figure 4 Renal, placental and hepatic


histological changes and peripheral blood
a
Control sEng sFlt1 sFlt1+sEng

smears in pregnant rats after sEng and sFlt1


treatment. (a) Renal histology (plastic
sections) of control, sEng, sFlt1 and sFlt1+sEng
groups. sEng-treated rats showed mild focal
endotheliosis (arrowhead, b) not seen in the
control glomeruli. sFlt1-injected rats showed
moderate to severe endotheliosis with complete b
occlusion of capillary lumens. sFlt1+sEng-treated
rats showed extremely swollen glomeruli and
marked endotheliosis with protein resorption
droplets in the podocytes. Scale bar, 50 mm.
(b) Placental histology (H&E stain) of control,
sEng, sFlt1 and sFlt1+sEng groups. Both the c
sEng- and sFlt1-treated rats showed diffuse
inflammation (arrowheads) at the maternal-
fetal junction that was not seen in controls.
There was hemorrhagic infarction and
fibrinoid necrosis with lumen obstruction
of a maternal vessel (arrow) in the decidua
of the sFlt1+sEng-treated placenta. Scale bar,
d
200 mm. (c) Liver histology in the control,
sEng, sFlt1 and sFlt1+sEng groups. Ischemic
changes with multifocal necrosis (arrowhead)
were noted in the sFlt1+sEng group. Control
group and rats given sEng or sFlt1 showed no
changes. Scale bar, 200 mm. (d) Peripheral
blood smear (Wright stain) of control, sEng, sFlt1 and sFlt1+sEng groups. A representative smear in the sFlt1+sEng group showed active schistocytes
(arrowheads) and reticulocytosis (arrows). No hemolysis was seen in the other groups.

NATURE MEDICINE VOLUME 12 [ NUMBER 6 [ JUNE 2006 645


ARTICLES

Vehicle C sEng
a TGF-β1
b c

Percent change in vascular diameter


125
Percent change in vascular diameter

TGF-β3 16 [I ] TGF-β1 6 12 25 50 100 100 6 12 25 50 100


25
TGF-β1 + sEng (pM)
TGF-β3 + sEng 12 80 kDa TβRII
20
8
15
4 Vehicle *

Percent maximal TβRII binding


* 100 sEng
10
** * 0
80 *
** –4
5 60
* –8 40
0
© 2006 Nature Publishing Group http://www.nature.com/naturemedicine

0.2 2 20 200 VEGF + – + + + –


Concentration (ng/ml) 20
TGF-β1 – + + + – +
sFlt1 – – – + – – 0
sEng – – – + – –
0 25 50 75 100
L-NAME – – – – + + 125
[I ] TGF-β1 (pM)
d 25

* e
Relative light intensity units

20 Vehicle
100 sEng

Percent phospho Thr495 /


Vehicle sEng
15 TGF-β1 (pM) 0 125 250 125 250 90

total eNOS level


(kDa) eNOS 80
140 pThr495
10 70
eNOS * *
140 pSer1177 60
5
50
eNOS
140 Total 40
0
0 0 15 15 TGF-β1 (ng/ml) 0
0 125 250
0 100 0 100 sEng (ng/ml) TGF-β1 (pM)

Figure 5 Recombinant sEng attenuates TGF-b1 binding and has effects on vasodilation through activation of eNOS. (a) Microvascular reactivity of rat renal
microvessels was measured in the presence of TGF-b1 or TGF-b3 from 200 pg/ml to 200 ng/ml, and with or without recombinant sEng at 1 mg/ml (n ¼ 4
per experiment; *P o 0.05 as compared to TGF-b1 or TGF-b3 alone). (b) Microvascular responses of renal microvessels to VEGF, TGF-b1 (1 ng/ml each) or
the combination of the two. The effects of 100 ng/ml each of sFlt1 and sEng on the combined response are shown (n ¼ 4 experiments). Also shown is the
blocking effect of L-NAME on TGF-b1– and VEGF-stimulated responses. (c) Representative autoradiogram and graph showing a dose-dependent increase in
[I125]TGF-b1 (6–100 pM) binding to TbRII on mouse endothelial cells. Treatment with 5 nM recombinant sEng significantly reduced binding at 50 pM and
100 pM (*P o 0.05 compared to untreated group). Competition with 40 excess cold TGF-b1 in cells treated with 100 pM [I125]TGF-b1 abolished receptor
binding and served as background control (C). (d) Graph showing significantly increased TGF-b-induced activation of the Smad 2/3-dependent CAGA-Luc
reporter construct transfected in human umbilical vein endothelial cells (HUVECs) and inhibition by treatment with sEng (n ¼ 3 experiments, **P o 0.01
compared to sEng untreated group). (e) Representative western blots and graph (n ¼ 4) showing significant dephosphorylation at eNOS Thr495 after
treatment with TGF-b1 and attenuation by sEng (*P o 0.05 compared to untreated). Phosphorylation was unchanged at Ser1177 and total levels of eNOS
remained constant throughout the experiments.

sFlt1 and sEng may oppose the physiological NO-dependent vasodi- mesenteric resistance vessels, we explored its immediate effects on
latation elicited by these angiogenic growth factors, contributing to the eNOS activation. Although TGF-b1 had no effect on phosphorylation
development of hypertension seen in preeclampsia. of eNOS Ser1177, it induced a significant (P o 0.01 versus baseline
control) dephosphorylation at Thr495 (Fig. 5e), suggesting that
sEng inhibits TGF-b1 binding and signaling in endothelial cells TGF-b regulates the phosphorylation status of a key residue involved
Given that endoglin is a coreceptor for TGF-b1 and TGF-b3 isoforms, in eNOS activation. This effect was significantly attenuated by
we hypothesized that sEng acts by interfering with binding of sEng (Fig. 5e).
cell-surface receptors. Preincubating radiolabeled TGF-b1 with recom-
binant sEng significantly reduced its binding to TGF-b receptor DISCUSSION
type II (TbRII) at both 50 and 100 pM (Fig. 5c). Thus, sEng Maternal endothelial dysfunction caused by placenta-derived soluble
competes for TGF-b1 binding to its receptors on endothelial cells. factors such as sFlt1 is emerging as a major component in the
To test whether this leads to impaired signaling, we assessed the pathogenesis of preeclampsia26,27. We report a novel soluble form of
activity of a CAGA-Luc reporter construct in human endothelial Eng of placental origin that is present in the sera of pregnant women,
cells. TGF-b1 induced the activation of the Smad 2/3-dependent elevated in preeclamptic individuals and correlated with disease
CAGA-Luc reporter, and this response was abolished by treatment severity. In addition to its potential use as a biomarker of preeclamp-
with sEng (Fig. 5d). sia, we have shown that sEng disrupts formation of endothelial tubes
in vitro and induces vascular permeability and hypertension in vivo.
sEng blocks TGF-b1–mediated activation of eNOS Notably, sEng can act in concert with sFlt1 to amplify endothelial
Although previous studies have shown that chronic exposure of dysfunction and induce clinical signs of severe preeclampsia, including
endothelial cells to TGF-b induces expression of the eNOS gene and development of the HELLP syndrome and restriction of fetal growth.
protein24,25, the immediate effects of TGF-b1 on the activation We show that TGF-b1 induces vasorelaxation through activation of
of eNOS have not yet been characterized. Given our findings that eNOS by triggering dephosphorylation of Thr495, providing a novel
TGF-b1 induces NOS-dependent vasorelaxation in both renal and mechanism for endothelium-dependent vasoregulation. sEng

646 VOLUME 12 [ NUMBER 6 [ JUNE 2006 NATURE MEDICINE


ARTICLES

interferes with TGF-b receptor binding and downstream signaling in may be responsible for hypertension noted in preeclampsia. Their
endothelial cells, and attenuates eNOS activation. We propose that the effects in decreasing activation of eNOS may also account for the
contributions of sEng and sFlt1 to the pathogenesis of maternal increased vascular permeability observed in preeclampsia37. Another
preeclampsia are, at least in part, related to their inhibition of molecule that may be central to the pathogenesis of the procoagulant
VEGF and TGF-b stimulation of endothelial-dependent NO activation state and thrombocytopenia in sFlt1+sEng-treated rats is the antith-
and vasomotor effects. rombotic factor prostacyclin (PGI2). Both VEGF and TGF-b1 stimu-
Increased Eng immunoreactivity has been reported in plasma of late production of PGI238–40, and clinical studies have shown
individuals with angiogenic tumors28, but its nature has remained decreased endothelial production of PGI2 even before the onset of
elusive. We have now characterized a circulating form of Eng observed clinical preeclampsia41. As it is known that TGF-b1 induces produc-
in normal pregnancy and upregulated in preeclampsia. The absence of tion of VEGF by pericytes42, it is also possible that sEng interferes
© 2006 Nature Publishing Group http://www.nature.com/naturemedicine

alternate splice variants in placenta and the partial peptide sequence of with VEGF functions indirectly through impaired production of
purified sEng suggest that it is an N-terminal cleavage product of full- VEGF by pericytes.
length Eng. Given that betaglycan, another TGF-b coreceptor with The placentae of women with severe preeclampsia are characterized
partial sequence identity to Eng, can be shed by membrane-type by shallow invasion of the cytotrophoblasts and impaired spiral
metalloprotease-1 (MT1-MMP)29, we speculate that sEng may be arteriolar remodeling. During normal placentation, cytotrophoblasts
generated by a similar mechanism. undergo a program of pseudovasculogenesis by acquiring the
We show that sEng prevents binding of TGF-b1 to TbRII on endothelial markers VE-cadherin and avb3 integrin, a process
endothelial cells, leading to decreased signaling. As circulating TGF- impaired in preeclampsia43,44. This impaired placentation and accom-
b1 is complexed with latency-associated peptide and latent TGF-b1 panying ischemia are thought to be the primary events leading to the
binding protein, it cannot bind its receptors unless activated. It is elaboration of soluble factors into the circulation. Treatment of villous
therefore likely that sEng only inhibits the effects of TGF-b1 locally, explant cultures with ENG antisense oligonucleotide enhanced tro-
where active TGF-b1 is generated. Hence sEng would not impact phoblast outgrowth and migration45, suggesting that cell-surface Eng
circulating concentrations of TGF-b1. When circulating active TGF-b1 negatively regulates this process. We speculate that sEng is produced
was measured in our cohort of pregnant women, it was undetectable by the placenta as a compensatory mechanism to limit the effects of
during pregnancy, confirming that the majority of circulating TGF-b1 cell-surface Eng. In preeclampsia, excessive production of cell-surface
is inactive. Although several clinical studies reported modest changes endoglin would lead to increased sEng in the maternal circulation,
in total circulating TGF-b1 in preeclampsia30–32, our data do not show which in turn may be responsible for the clinical manifestations
significant differences between normal and preeclamptic pregnancies of preeclampsia.
(35.95 ng/ml versus 37.74 ng/ml in serum, respectively). Eng is also Our findings on the role of sEng in preeclampsia have important
capable of binding other ligands of the TGF-b family, such as activins diagnostic and therapeutic implications. Analogous to sFlt1 (ref. 9),
and BMPs33, although no functional role of Eng in mediating their circulating sEng starts rising 6–10 weeks before clinical symptoms of
effects has been described. preeclampsia (S.A.K. & R.R., unpublished observations). We posit that
In this study, we identified a direct effect of TGF-b1 and TGF-b3 on a predictive test measuring sEng, sFlt1 and PlGF in the serum will have
vascular reactivity, consistent with the known specificity of cell-surface enhanced sensitivity and specificity, and provide a powerful tool in the
Eng for these isoforms, thereby suggesting its essential role in TGF-b– prevention of preeclampsia-induced mortality. In addition, neutraliz-
dependent vasodilatation. This effect is NOS dependent and supports ing antibodies to sEng and sFlt1 may be beneficial in the treatment of
our findings that Eng interacts with eNOS and stabilizes its activa- preeclampsia. Therapeutic monoclonal antibodies targeting surface
tion19. Together, these data suggest a crucial role for Eng in linking Eng are currently being developed for the treatment of metastatic
TGF-b receptor activation to synthesis of NO. We have found that cancers46. Our data suggests that sEng may be another way to
TGF-b1 dephosphorylates eNOS at Thr495, a regulatory step necessary interfere with angiogenically active tumors that express vast amounts
to increase the Ca2+ sensitivity and enzyme activity and preventing of TGF-b. Similar strategies have been employed in the VEGF
uncoupling and production of eNOS-derived superoxide. Although signaling cascade using VEGF-trap, a modified sFlt1 that is currently
activation of eNOS also involves a coordinated increase in phosphor- in cancer clinical trials47.
ylation of Ser1177, this was not observed in response to TGF-b1. In summary, we have shown that excess circulating concentrations
TGF-b may specifically modulate dephosphorylation of Thr495 and of sEng and sFlt1 in individuals with preeclampsia contribute to the
synergize with factors such as VEGF, which can activate eNOS by pathogenesis of hypertension, proteinuria, glomerular endotheliosis,
phosphorylating Ser1177. We observed that TGF-b and VEGF HELLP syndrome and restriction of fetal growth—all hallmarks of
have additive effects on NOS-dependent vasodilatation that are com- severe preeclampsia. Understanding the regulation of sEng and sFlt1
pletely reversed by sEng and sFlt1 at concentrations seen in individuals production in placenta and their effects on systemic and placental
with preeclampsia. vascular function should lead to better insights into their roles during
Our functional studies suggest that sEng and sFlt1 act in concert to normal pregnancy and the pathogenesis, prediction, treatment and
induce vascular damage and HELLP syndrome by interfering with prevention of preeclampsia.
TGF-b1 and VEGF signaling, respectively. One molecule likely to be
involved in the pathogenesis of hypertension34 and central to both
VEGF and TGF-b1 signaling is NO. Several studies support the METHODS
Reagents. We purchased recombinant human sEng (1–587 amino acids,
hypothesis that decreased biologically available NO is central to the
corresponding to the extracellular region of cell-surface Eng), human sFlt1,
pathogenesis of preeclampsia35. The inhibition of eNOS activation by mouse sEng and sFlt1, human TGF-b1 and TGF-b3, mouse VEGF-164 and
both sFlt1 and sEng suggests a molecular basis for the elevated MAP. human VEGF-165 from R&D Systems. Mouse monoclonal antibody to human
Furthermore, as both TGF-b1 and VEGF stimulate expression of Eng (clone P4A4) and polyclonal antibody (H-300) to the N-terminal region of
eNOS25,36, it is likely that sEng and sFlt1 inhibit expression of human Eng were from Santa Cruz. ELISA kits for human and mouse sFlt1, and
eNOS chronically and that their excess circulating concentrations human recombinant sEng were obtained from R&D systems.

NATURE MEDICINE VOLUME 12 [ NUMBER 6 [ JUNE 2006 647


ARTICLES

Subjects. All clinical studies were approved by the Beth Israel Deaconess 850). We detected schistocytes by staining peripheral blood smears with Wright
Medical Center Committee on Clinical Investigations where all subjects were stain. Rats were killed, litters counted and individual placentae and fetuses
recruited and provided informed consent. Clinical information is described in weighed. We used harvested kidneys and placentae for histopathology and
Supplementary Table 1. Preeclampsia was defined by the American College of electron microscopy as previously described4.
Obstetricians and Gynecologists criteria48. Individuals with HELLP syndrome
are presented as a separate severe preeclamptic group. HELLP syndrome was TGF-b1 binding to endothelial cells. We affinity-labeled confluent endothelial
diagnosed when individuals had thrombocytopenia (o100,000 cells/ml) and cell monolayers for 4 h at 41C with increasing concentrations of [I125]TGF-b1
increased LDH (4600 IU/L) and AST (470 IU/L). Healthy pregnant women (DuPont NEN) after preincubation with or without 2.5 nM recombinant sEng
were included as controls; eight individuals with pre-term labor served as or cold TGF-b1 (40-fold excess), washed them and cross-linked them with
gestational age controls. Blood specimens from four nonpregnant healthy disuccinimidyl suberate (Pierce). We solubilized cells in buffer containing 1%
individuals in the age groups (30–45 years) were also included. Placental Triton X-100 and separated extracts under reducing conditions by SDS-PAGE
© 2006 Nature Publishing Group http://www.nature.com/naturemedicine

samples were obtained immediately after delivery. The placental samples used on 4–12% gels13. We visualized levels of [I125]TGF-b1 bound to TbRII with a
in the study were randomly chosen from the preeclampsia (mild, severe, STORM Phosphorimager and quantified them by densitometry using Image-
HELLP) and controls (term and pre-term) groups described in Supplementary Quant software.
Table 1, and the only criterion used for selection was that the controls were
matched in gestational age to the individuals with preeclampsia. Serum TGF-b1–dependent activation of Smad2/3. We transfected human umbilical
was collected at the time of delivery (0–12 h before) and at 48 h (±8 h) vein endothelial cells (HUVECs, Clonetics; passage 3) with (CAGA)12-Luc
plasmid (from K. Miyazano, University of Tokyo) in the absence or presence of
after delivery.
TGF-b1 and/or sEng and measured luciferase activity as previously described50.
Northern blots, ELISA, western blots, immunohistochemistry and immu-
Microvascular reactivity experiments. We performed microvascular reactivity
noprecipitation. We performed northern blots, ELISA, western blots, immu-
experiments as previously described4 using rat renal or mesenteric microvessels
nohistochemistry and immunoprecipitation as described in Supplementary
Methods online. (internal diameter, 70–150 mm). In all experimental groups, we examined the
relaxation responses of kidney microvessels after precontraction with U46619
Purification of sEng and analysis by mass spectrometry. We sequentially (thromboxane A2 agonist) to 40–60% of their baseline diameter at a distending
applied serum (10 ml) from preeclamptic individuals onto CM Affi-gel blue pressure of 40 mmHg. Once the steady-state tone was reached, we examined
and protein A Sepharose (Bio-Rad) columns to remove albumin and immu- the responses to TGF-b1, TGF-b3 or VEGF in a standardized order. We
noglobulins, respectively. We slowly applied the flow-through to a 2.5 ml administered all drugs extraluminally. When required, we pretreated vessels
column of monoclonal antibody 44G4 IgG to human Eng, conjugated to with sEng, sFlt1 or 10–5 M L-NAME for 30 min.
Sepharose. We eluted bound fractions with 0.02 M diethylamine (pH 11.4) and
immediately neutralized them with 1 M Tris pH 7.8. We pooled fractions 4 and TGF-b1–dependent phosphoregulation of eNOS. We incubated confluent
5 with elevated absorbance at 280 nm, reduced them with 10 mM DTT for 1 h mouse endothelial cells in serum-free media for 2 h. We stimulated cells with
at 57 1C and alkylated them with 0.055 M iodoacetomide. We then completely TGF-b1 in the presence and absence of sEng (100 ng/ml) for 15 min and
digested the samples with trypsin (1:100). We resuspended the lyophilized extracted proteins in 2% SDS buffer supplemented with 1 mM Na8VO4, 10 mM
sample in 0.1% trifluoroacetic acid and injected it into a CapLC (Waters) high- Na4P2O7, 25 mM NaF and protease inhibitors (Roche Molecular Biochemicals).
performance liquid chromatography instrument. We separated peptides using a We quantified proteins and immunoblotted them with polyclonal antibodies to
75 mm Nano Series column (LC Packings) and analyzed them using a Qstar XL eNOS Thr495, Ser1177 or monoclonal antibody specific for total eNOS.
MS/MS system. We searched data using the Mascot search engine (Matrix Statistical analysis. Results are presented as mean ± s.e.m. and comparisons
Science) against the human protein database NCBInr. between multiple groups were made by analysis of variance using ANOVA.
Endothelial tube assays and microvascular permeability experiments. We Significant differences are reported when P o 0.05.
performed endothelial tube assays and microvascular permeability experiments Note: Supplementary information is available on the Nature Medicine website.
as described in Supplementary Methods.
ACKNOWLEDGMENTS
Generation of adenoviruses. Adenovirus vectors containing sFlt1 and Fc have We thank all the staff in the Department of Obstetrics at the Beth Israel
been previously described4,49. To generate sEng adenovirus, we used the Adeasy Deaconess Medical Center for help with patient identification and recruitment.
Kit (Stratagene). Briefly, we amplified human sEng (Thr27–Leu586) corre- We thank B. Furie’s laboratory for help with measurement of platelet counts in
sponding to the complete extracellular region with PCR using human cDNA rats, J. Min and the Physiology Core laboratory for help with blood pressure
encoding full-length Eng clone (Invitrogen) as template and the following measurements, J. Li for vascular reactivity experiments, R. Mulligan for help with
primers: forward, 5¢-ACGAAGCTTGAAACAGTCCATTGTGACCTT-3¢ and the production of sFlt1 adenoviruses, L. Zhang for technical assistance with mass
reverse, 5-’TTAGATATCTGGCCT TTGCTTGTGCAACC-3¢. We subcloned spectrometry, B. Sachs, R. Levine, R. Thadhani, J. Flier, W. Aird and S. Pennathur
for discussions. This work was funded by US National Institutes of Health grants
amplified PCR fragments into pShuttle-CMV vector (Stratagene) and con-
DK064255 and HL079594 to S.A.K., Department of Medicine, Obstetrics and
firmed expression by western blotting. We amplified the confirmed sEng clone Gynecology seed funds to S.A.K. and V.P.S., and Heart and Stroke Foundation
in 293T cells and purified it on a CsCl2 density gradient (Qbiogene). Ad-CMV of Ontario grant T5016 to M.L.
(control) was obtained from Qbiogene.
AUTHOR CONTRIBUTIONS
Rat model of preeclampsia. All animal protocols were approved by the Beth S.V.: mRNA and protein expression studies of Eng/sEng, generation and
Israel Deaconess Medical Center Institutional Animal Care and Use Commit- expression of sEng adenoviruses, all animal studies; substantial contribution to
tee. We intravenously injected pregnant Sprague-Dawley rats with 2  109 writing, editing and generation of figures. M.T.: biochemical characterization of
plaque-forming units of Ad-CMV (control), Ad-sFlt1, Ad-sEng or Ad-sFlt1+Ad sEng, TGF-b effects on eNOS dephosphorylation, studies of sEng on TGF-b
sEng. We injected females at day 8 or 9 of pregnancy and measured intracarotid effects; substantial contribution to writing and editing of manuscript and
MAP at 17–18 d under anesthesia as previously described4. We confirmed levels generation of figures. C.L.: enrollment and collection of all human material,
ELISA studies on human samples, in vitro angiogenesis assays and biochemical
of circulating sFlt-1 and sEng by western blotting and quantified them using
studies of urine and plasma obtained from animals. J.H.: TGF-b–Smad promoter
commercially available ELISA kits (R&D Systems). We measured urinary studies. T.M.: vascular permeability studies. Y.M.K.: immunohistochemistry of
albumin by standard dipstick and quantified it using the Nephrat ELISA kit human placental samples. Y.B.: animal studies. K.H.L.: enrollment and collection
(Exocell). We measured urinary creatinine using the Metra creatinine assay kit of all human material, expertise in clinical preeclampsia. H.Y.: in vitro
(Quidel Corp). We measured AST and LDH using commercial kits from angiogenesis assays. T.A.L.: Affymetrix microarray experiments. I.E.S.: all rat
Thermo Electron. We estimated platelet counts by hemocytometry (Hemavet histological studies, including electron microscopy. D.R.: rat placental histological

648 VOLUME 12 [ NUMBER 6 [ JUNE 2006 NATURE MEDICINE


ARTICLES

studies. P.A.D.: provided expertise in vascular biology and TGF-b signaling. 21. Garcia-Cardena, G. et al. Dynamic activation of endothelial nitric oxide synthase by
F.H.E.: provided expertise in clinical preeclampsia. F.W.S.: all microvascular Hsp90. Nature 392, 821–824 (1998).
reactivity experiments. R.R.: immunohistochemistry of human placental samples; 22. Fleming, I., Fisslthaler, B., Dimmeler, S., Kemp, B.E. & Busse, R. Phosphorylation of
provided expertise in preeclampsia. V.P.S.: interpretation of microarray Thr(495) regulates Ca(2+)/calmodulin-dependent endothelial nitric oxide synthase
activity. Circ. Res. 88, E68–E75 (2001).
experiments, microvascular permeability studies; provided overall expertise in
23. Brown, M.A., Zammit, V.C. & Lowe, S.A. Capillary permeability and extracellular fluid
vascular biology and preeclampsia; final editing of the manuscript. M.L.: provided volumes in pregnancy-induced hypertension. Clin. Sci. (Lond.) 77, 599–604
expertise on Eng, analysis and interpretation of studies involving biochemical (1989).
characterization of sEng, TGF-b effects on eNOS dephosphorylation, studies 24. Inoue, N. et al. Molecular regulation of the bovine endothelial cell nitric oxide synthase
of sEng on TGF-b effects; substantially contributed to writing and editing of by transforming growth factor-beta 1. Arterioscler. Thromb. Vasc. Biol. 15, 1255–1261
manuscript. S.A.K: principal investigator of the study; overall design and (1995).
concept, analysis and interpretation of all data, drafting and final editing 25. Saura, M. et al. Smad2 mediates transforming growth factor-b induction of endothelial
of the manuscript. nitric oxide synthase expression. Circ. Res. 91, 806–813 (2002).
© 2006 Nature Publishing Group http://www.nature.com/naturemedicine

26. Bdolah, Y., Sukhatme, V.P. & Karumanchi, S.A. Angiogenic imbalance in the
pathophysiology of preeclampsia: newer insights. Semin. Nephrol. 24, 548–556
COMPETING INTERESTS STATEMENT (2004).
The authors declare competing financial interests (see the Nature Medicine website 27. Redman, C.W. & Sargent, I.L. Latest advances in understanding preeclampsia. Science
for details). 308, 1592–1594 (2005).
28. Li, C. et al. Plasma levels of soluble CD105 correlate with metastasis in patients with
Published online at http://www.nature.com/naturemedicine/ breast cancer. Int. J. Cancer 89, 122–126 (2000).
Reprints and permissions information is available online at http://npg.nature.com/ 29. Velasco-Loyden, G., Arribas, J. & Lopez-Casillas, F. The shedding of betaglycan is
reprintsandpermissions/ regulated by pervanadate and mediated by membrane type matrix metalloprotease-1.
J. Biol. Chem. 279, 7721–7733 (2004).
30. Benian, A., Madazli, R., Aksu, F., Uzun, H. & Aydin, S. Plasma and placental levels
1. Sibai, B., Dekker, G. & Kupferminc, M. Pre-eclampsia. Lancet 365, 785–799 of interleukin-10, transforming growth factor-b1, and epithelial-cadherin in pre-
(2005). eclampsia. Obstet. Gynecol. 100, 327–331 (2002).
2. Weinstein, L. Syndrome of hemolysis, elevated liver enzymes, and low platelet count: a 31. Muy-Rivera, M. et al. Transforming growth factor-b1 (TGF-b1) in plasma is associated
severe consequence of hypertension in pregnancy. Am. J. Obstet. Gynecol. 142, with preeclampsia risk in Peruvian women with systemic inflammation. Am. J.
159–167 (1982). Hypertens. 17, 334–338 (2004).
3. Roberts, J.M. et al. Preeclampsia: an endothelial cell disorder. Am. J. Obstet. Gynecol. 32. Hennessy, A. et al. Transforming growth factor-b 1 does not relate to hypertension in
161, 1200–1204 (1989). pre-eclampsia. Clin. Exp. Pharmacol. Physiol. 29, 968–971 (2002).
4. Maynard, S.E. et al. Excess placental soluble fms-like tyrosine kinase 1 (sFlt1) may 33. Barbara, N.P., Wrana, J.L. & Letarte, M. Endoglin is an accessory protein that interacts
contribute to endothelial dysfunction, hypertension, and proteinuria in preeclampsia. with the signaling receptor complex of multiple members of the transforming growth
J. Clin. Invest. 111, 649–658 (2003). factor-beta superfamily. J. Biol. Chem. 274, 584–594 (1999).
5. Zhou, Y. et al. Vascular endothelial growth factor ligands and receptors that regulate 34. Shesely, E.G. et al. Elevated blood pressures in mice lacking endothelial nitric oxide
human cytotrophoblast survival are dysregulated in severe preeclampsia and hemolysis, synthase. Proc. Natl. Acad. Sci. USA 93, 13176–13181 (1996).
elevated liver enzymes, and low platelets syndrome. Am. J. Pathol. 160, 1405–1423 35. Lowe, D.T. Nitric oxide dysfunction in the pathophysiology of preeclampsia. Nitric
(2002). Oxide 4, 441–458 (2000).
6. Ahmad, S. & Ahmed, A. Elevated placental soluble vascular endothelial growth 36. Papapetropoulos, A., Garcia-Cardena, G., Madri, J.A. & Sessa, W.C. Nitric oxide
factor receptor-1 inhibits angiogenesis in preeclampsia. Circ. Res. 95, 884–891 production contributes to the angiogenic properties of vascular endothelial growth
(2004). factor in human endothelial cells. J. Clin. Invest. 100, 3131–3139 (1997).
7. Chaiworapongsa, T. et al. Evidence supporting a role for blockade of the vascular 37. Predescu, D., Predescu, S., Shimizu, J., Miyawaki-Shimizu, K. & Malik, A.B. Con-
endothelial growth factor system in the pathophysiology of preeclampsia. Young stitutive eNOS-derived nitric oxide is a determinant of endothelial junctional integrity.
Investigator Award. Am. J. Obstet. Gynecol. 190, 1541–1547; discussion Am. J. Physiol. Lung Cell. Mol. Physiol. 289, L371–L381 (2005).
1547–1550 (2004). 38. Tatsumi, M., Kishi, Y., Miyata, T. & Numano, F. Transforming growth factor-beta(1)
8. Taylor, R.N. et al. Longitudinal serum concentrations of placental growth factor: restores antiplatelet function of endothelial cells exposed to anoxia-reoxygenation
evidence for abnormal placental angiogenesis in pathologic pregnancies. Am. J. Obstet. injury. Thromb. Res. 98, 451–459 (2000).
Gynecol. 188, 177–182 (2003). 39. Ristimaki, A., Ylikorkala, O. & Viinikka, L. Effect of growth factors on human vascular
9. Levine, R.J. et al. Circulating angiogenic factors and the risk of preeclampsia. N. Engl. endothelial cell prostacyclin production. Arteriosclerosis 10, 653–657 (1990).
J. Med. 350, 672–683 (2004). 40. He, H. et al. Vascular endothelial growth factor signals endothelial cell production of
10. Chaiworapongsa, T. et al. Plasma soluble vascular endothelial growth factor receptor-1 nitric oxide and prostacyclin through flk-1/KDR activation of c-Src. J. Biol. Chem. 274,
concentration is elevated prior to the clinical diagnosis of pre-eclampsia. J. Matern. 25130–25135 (1999).
Fetal Neonatal Med. 17, 3–18 (2005). 41. Mills, J.L. et al. Prostacyclin and thromboxane changes predating clinical onset of
11. Hertig, A. et al. Maternal serum sFlt1 concentration is an early and reliable predictive preeclampsia: a multicenter prospective study. J. Am. Med. Assoc. 282, 356–362
marker of preeclampsia. Clin. Chem. 50, 1702–1703 (2004). (1999).
12. Romero, R. et al. Clinical significance, prevalence, and natural history of thrombocy- 42. Darland, D.C. et al. Pericyte production of cell-associated VEGF is differentiation-
topenia in pregnancy-induced hypertension. Am. J. Perinatol. 6, 32–38 (1989). dependent and is associated with endothelial survival. Dev. Biol. 264, 275–288
13. Cheifetz, S. et al. Endoglin is a component of the transforming growth factor-beta (2003).
receptor system in human endothelial cells. J. Biol. Chem. 267, 19027–19030 43. Zhou, Y. et al. Human cytotrophoblasts adopt a vascular phenotype as they differenti-
(1992). ate. A strategy for successful endovascular invasion? J. Clin. Invest. 99, 2139–2151
14. Gougos, A. et al. Identification of distinct epitopes of endoglin, an RGD-containing (1997).
glycoprotein of endothelial cells, leukemic cells, and syncytiotrophoblasts. Int. 44. Zhou, Y., Damsky, C.H. & Fisher, S.J. Preeclampsia is associated with failure of human
Immunol. 4, 83–92 (1992). cytotrophoblasts to mimic a vascular adhesion phenotype. One cause of defective
15. St-Jacques, S., Forte, M., Lye, S.J. & Letarte, M. Localization of endoglin, a endovascular invasion in this syndrome? J. Clin. Invest. 99, 2152–2164 (1997).
transforming growth factor-beta binding protein, and of CD44 and integrins in placenta 45. Caniggia, I., Taylor, C.V., Ritchie, J.W., Lye, S.J. & Letarte, M. Endoglin regulates
during the first trimester of pregnancy. Biol. Reprod. 51, 405–413 (1994). trophoblast differentiation along the invasive pathway in human placental villous
16. McAllister, K.A. et al. Endoglin, a TGF-b binding protein of endothelial cells, is the explants. Endocrinology 138, 4977–4988 (1997).
gene for hereditary haemorrhagic telangiectasia type 1. Nat. Genet. 8, 345–351 46. Fonsatti, E., Altomonte, M., Arslan, P. & Maio, M. Endoglin (CD105): a target for anti-
(1994). angiogenetic cancer therapy. Curr. Drug Targets 4, 291–296 (2003).
17. Bourdeau, A., Dumont, D.J. & Letarte, M. A murine model of hereditary hemorrhagic 47. Holash, J. et al. VEGF-Trap: a VEGF blocker with potent antitumor effects. Proc. Natl.
telangiectasia. J. Clin. Invest. 104, 1343–1351 (1999). Acad. Sci. USA 99, 11393–11398 (2002).
18. Li, D.Y. et al. Defective angiogenesis in mice lacking endoglin. Science 284, 48. ACOG practice bulletin. Diagnosis and management of preeclampsia and eclampsia.
1534–1537 (1999). Number 33, January 2002. Obstet. Gynecol. 99, 159–167 (2002).
19. Toporsian, M. et al. A role for endoglin in coupling eNOS activity and regulating 49. Kuo, C.J. et al. Comparative evaluation of the antitumor activity of antiangiogenic
vascular tone revealed in hereditary hemorrhagic telangiectasia. Circ. Res. 96, proteins delivered by gene transfer. Proc. Natl. Acad. Sci. USA 98, 4605–4610
684–692 (2005). (2001).
20. Dimmeler, S., Dernbach, E. & Zeiher, A.M. Phosphorylation of the endothelial nitric 50. Akiyoshi, S. et al. c-Ski acts as a transcriptional co-repressor in transforming growth
oxide synthase at ser-1177 is required for VEGF-induced endothelial cell migration. factor-beta signaling through interaction with smads. J. Biol. Chem. 274, 35269–
FEBS Lett. 477, 258–262 (2000). 35277 (1999).

NATURE MEDICINE VOLUME 12 [ NUMBER 6 [ JUNE 2006 649


E R R ATA A N D C O R R I G E N D A

Erratum: Proton NMR analysis of plasma is a weak predictor of coronary artery


disease
H L Kirschenlohr, J L Griffin, S C Clarke, R Rhydwen, A A Grace, P M Schofield, K M Brindle & J C Metcalfe
Nat. Med. 12, 705–710 (2006); published online 28 May 2006; corrected after print 19 June 2006

In the version of this article initially published, the layout of the data in Tables 1 and 2 is incorrect.
The error has been corrected in the HTML and PDF versions of the article.
© 2006 Nature Publishing Group http://www.nature.com/naturemedicine

Erratum: Mig6 is a negative regulator of EGF receptor–mediated skin


morphogenesis and tumor formation
I Ferby, M Reschke, O Kudlacek, P Knyazev, G Pantè, K Amann, W Sommergruber, N Kraut, A Ullrich, R Fässler & R Klein
Nat. Med. 12, 568–573 (2006); published online 30 April 2006; corrected after print 16 June 2006

In the version of this article initially published, Errfi1 was incorrectly referred to as Erffi1 in several instances, and in Figure 1h the middle panels
of the immunoblot were labeled Egf instead of Hgf.
The errors have been corrected in the HTML and PDF versions of the article.

Corrigendum: Soluble endoglin contributes to the pathogenesis of


preeclampsia
S Venkatesha, M Toporsian, C Lam, J Hanai, T Mammoto, Y M Kim, Y Bdolah, K-H Lim, H-T Yuan, T A Libermann, I E Stillman, D Roberts,
P A D’Amore, F H Epstein, F W Sellke, R Romero, V P Sukhatme, M Letarte & S Ananth Karumanchi
Nat. Med. 12, 642–649 (2006)

Due to an editing error, some reference citations in the text are incorrect. In particular, the second citation of ref. 22 (on p. 644) should be ref. 23
and refs. 23–49 should be refs. 24–50.
This has been corrected in the HTML and PDF versions of the article.

862 VOLUME 12 | NUMBER 7 | JULY 2006 NATURE MEDICINE

You might also like