You are on page 1of 13

Kidney International, Vol. 67 (2005), pp.

21012113

PERSPECTIVES IN RENAL MEDICINE

Preeclampsia: A renal perspective


S. ANANTH KARUMANCHI, SHARON E. MAYNARD, ISAAC E. STILLMAN, FRANKLIN H. EPSTEIN,
and VIKAS P. SUKHATME
Renal Division and Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston,
Massachusetts; Department of Obstetrics & Gynecology, Beth Israel Deaconess Medical Center and Harvard Medical School,
Boston, Massachusetts; and Department of Pathology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston,
Massachusetts

Preeclampsia: A renal perspective. Preeclampsia is a syndrome


that affects 5% of all pregnancies, producing substantial maternal and perinatal morbidity and mortality. The aim of this review
is to summarize our current understanding of the pathogenesis
of preeclampsia with special emphasis on the recent discovery
that circulating anti-angiogenic proteins of placental origin may
play an important role in the pathogenesis of proteinuria and
hypertension of preeclampsia.

Preeclampsia, the syndrome of hypertension and proteinuria that heralds the seizures of eclampsia, remains
one of the great mysteries in the field of obstetrics.
Although our understanding of the pathophysiology
of preeclampsia has increased over the past 50 years,
it is quite incomplete, and management remains supportive: close observation, treatment with antihypertensive agents and magnesium sulfate, and if progressive
signs and symptoms occur, urgent delivery of the fetus.
Preeclampsia is still one of the leading causes of maternal
and neonatal mortality in the world.
Preeclampsia is characterized by a constellation of
signs and symptoms, including the new onset of hypertension and proteinuria during the last trimester of
pregnancy, usually associated with edema and hyperuricemia [1, 2]. It occurs only in the presence of the
placenta, even when there is no fetus (as in hydatidiform mole) and remits dramatically postpartum [3]. The
placenta in preeclampsia is usually abnormal, with evidence of hypoperfusion and ischemia. Although these
placental changes are neither universal nor specific for
preeclampsia, there appears to be a correlation between
Key words: pregnancy, HELLP syndrome, angiogenesis, pseudovasculogenesis, VEGF, PlGF, soluble Flt-1, soluble VEGFR-1, proteinuria,
edema.
Received for publication February 9, 2004
and in revised for April 23, 2004
Updated on November 23, 2004
Accepted on January 12, 2005

C

2005 by the International Society of Nephrology

the severity of the disease and the extent of placental abnormalities. The clinical findings of severe preeclampsia
are unified by the presence of systemic endothelial dysfunction and microangiopathy, in which the target organ
may be the brain (seizures or eclampsia), the liver [the
hemolysis, elevated liver function tests, and low platelet
count (HELLP) syndrome], or the kidney (glomerular
endotheliosis and proteinuria). Severe preeclampsia is
also associated with small for gestational age (SGA) fetuses. Because of the strong clinical and experimental evidence of early placental involvement and dysfunction of
the maternal endothelium, it is currently believed that
preeclampsia has its origin in disordered vascular development of the placenta, which in turn leads to widespread
maternal vascular endothelial effects [4, 5] (Fig. 1). This
review will discuss mechanisms underlying the clinical
manifestations of preeclampsia. In addition, we will describe evidence suggesting that placental secretion of an
antiangiogenic protein may contribute to the endothelial
dysfunction of preeclampsia.

RISK FACTORS FOR THE DEVELOPMENT OF


PREECLAMPSIA
The risk factors for the development of preeclampsia
are listed in Table 1 [1, 57]. Preeclampsia is more
common not only in first pregnancies, but also in multigravidas who have a new partner, suggesting that prior
exposure to paternal antigens may be protective [8, 9].
However, recent evidence from a large Norwegian birth
registry suggests that prolonged interpregnancy interval,
rather than primipaternity, accounts for this increase in
risk [10], though why this occurs is unclear. Although
preeclampsia is traditionally not considered to be a
genetic disease, it is clear that genetic factors contribute
to the susceptibility to preeclampsia [6]. A family history
(mother, sister, or both) of preeclampsia is associated
with a fourfold increased risk for preeclampsia [6]. Other
epidemiologic studies suggest that paternal genetic
contributions to the zygotic genotype in addition to
2101

2102

Karumanchi et al: Preeclampsia and angiogenic factors

Table 1. Risk factors for the development of preeclampsia


Family history of preeclampsia
Nulliparity
Multiple gestation
Molar pregnancies
Older maternal age
Obesity
Preexisting hypertension
Chronic renal disease
Diabetes mellitus
Thrombotic vascular disease

maternal genes may contribute susceptibility to


preeclampsia [11]. Polymorphisms of genes involved in
the regulation of blood pressure or coagulation, such as
renin, angiotensinogen (T235), endothelial nitric oxide
synthase (eNOS), prothrombin, factor V Leiden, and
methyltetrahydrofolate (MTHFR), though promising in
early studies [1215], have not been confirmed in larger
studies [1620]. Genome-wide scanning of Icelandic
families revealed a significant locus on chromosome 2p13
[logarithm of the odds (LOD) score 4.70] [21]. More
recently, the 2p locus was confirmed in a study of patients
from New Zealand and Australia [22]. Finally, a Dutch
study reported linkage of HELLP syndrome, but not
preeclampsia, with a locus on 12q, suggesting that genetic
factors important in HELLP (hemolysis, elevated liver
function tests, low platelets) syndrome may be distinct
from those in preeclampsia [23]. Women with trisomy 13
fetuses have a higher incidence of preeclampsia, regardless of parity, suggesting that a gene on chromosome 13
may be important in preeclampsia [24]. An activating
mineralocorticoid receptor mutation was described in a
rare group of patients who have only pregnancy-induced
hypertension without proteinuria [25]. The incidence of
preeclampsia is also higher in women who live at high
altitudes and in the third world, suggesting that hypoxia
and/or hitherto unknown environmental factors may
also contribute to the development of preeclampsia [26,
27].
CLINICAL MANIFESTATIONS AND THEIR
PATHOPHYSIOLOGIC UNDERPINNINGS
Hypertension
While in normal pregnancy peripheral vascular resistance and blood pressure are decreased, in preeclampsia these changes are reversed. Increased peripheral
vascular resistance, rather than increased cardiac output,
is the chief cause of hypertension [28]. Sympathetic activation is noted in preeclampsia as it is in other forms
of hypertension, a conclusion supported by electrical
recordings of sympathetic nerve impulses [29] and by
reports of increased concentrations of circulating catecholamines [30]. Sympathetic activation may be respon-

sible for the increase in cardiac output noted by some in


the early stages of preeclampsia [31]. Preeclampsia is also
notable for an exaggerated response to angiotensin II,
catecholamines, and other hypertensive stimuli when
compared to normal pregnant controls [32, 33]. One
group has reported that this response may precede the
onset of overt hypertension by weeks to months [34]
(a finding disputed by others [35]), reminiscent of the
exaggerated response to vasoconstrictors described in
normotensive relatives of patients with essential hypertension [36].
Although total plasma volume has been reported to
be low in preeclampsia [37], there may be increased
effective circulating volume as evidenced by suppressed renin and aldosterone [38, 39] and elevated brain
natriuretic hormone [40] relative to normal pregnancy.
These findings are reminiscent of the fall in plasma volume and rise in blood pressure produced by infusion
of vascoconstrictors suggesting that peripheral vasoconstriction, especially of small venules, shifts blood to the
arteries and central veins while elevating capillary pressure [41]. Blood pressure rises, and renin and aldosterone
levels fall as a secondary phenomenon.
Generalized vascular constriction is universally
present in preeclampsia, at least compared to the physiologic vasodilation of normal pregnancy [28]. There is
substantial evidence that this may be due to endothelial
dysfunction. A myriad of markers for endothelial activation and dysfunction, including endothelin, cellular
fibronectin, plasminogen activator inhibitor-1 (PAI-1),
and von Willebrands factor, are altered in preeclampsia.
Women with preeclampsia have enhanced responsiveness to vasopressors as compared with normal pregnant
women. Women with a history of preeclampsia exihibit
evidence of impaired endothelial-dependent vasorelaxation as measured by brachial artery flowmediated
dilatation up to 3 years after delivery, implying these
changes in the maternal endothelium may be more than
transient [42, 43]. Alterations of endothelial function
have been noted in preeclamptic vessels examined in
vitro, supporting the hypothesis that endothelial dysfunction may underlie the hypertension of preeclampsia
[4, 5]. The increased incidence of preeclampsia in women
with chronic diseases such as diabetes and hypertension
also suggests some factor in the maternal milieu may also
lend susceptibility to preeclampsia. In addition to increased vascular reactivity, the vasoconstriction appears
to be mediated at least in part by alterations in local
concentrations of several vasoactive molecules, including
the vasoconstrictors norepinephrine, endothelin, and
perhaps thromboxane, and the vasodilators prostacyclin
and perhaps nitric oxide.
Prostaglandin I 2 (PGI 2 ) (prostacyclin), a circulating vasodilator produced primarily by the endothelial and smooth muscle layers of blood vessels, is

Karumanchi et al: Preeclampsia and angiogenic factors

Genetic
factors

Environmental
factors

2103

Other

Placental
dysfunction

Circulating
factor(s)

Vascular endothe lial dysfunction

Hypertension

Glomerular
endotheliosis
proteinuria edema
renal insufficiency

Headache
Cerebral edema
Seizures

LFTs
HELLP syndrome

Fig. 1. Placental dysfunction and endothelial


dysfunction in the pathogenesis of preeclampsia. Placental dysfunction, triggered by poorly
understood mechanisms, which may include
genetic, immunologic, and environmental factors, plays an early and primary role in the
development of preeclampsia. The diseased
placenta in turn secretes a factor(s) into
the maternal circulation, causing systemic
endothelial cell dysfunction. Most of the
manifestations of preeclampsia, including
hypertension, proteinuria (glomerular endotheliosis), seizures (cerebral edema and/or
vasospasm), and the hemolysis, elevated
liver function tests, and low platelet count
(HELLP) syndrome can be attributed to vascular and endothelial effects.
Fig. 2. Glomerular endotheliosis. (A) Normal human glomerulus (hematoxylin and
eosin stain). (B) Human preeclamptic
glomerulus (hematoxylin and eosin stain). A
33-year-old woman with twin gestation and
severe preeclampsia at 26 weeks gestation
with urine protein/creatinine ratio of 26 at
the time of biopsy. (C) Electron microscopy
of glomerulus of the above patient described
in (B). Note occlusion of capillary lumen cytoplasm and expansion of the subendothelial
space with some electron-dense material.
Podocyte cytoplasms show protein resorption
droplets and relatively intact foot processes
(original magnification 1500). (D) Control
rat glomerulus (hematoxylin and eosin
stain). Note normal cellularity and open
capillary loops. (E) Soluble Flt-1treated rat
(hematoxylin and eosin stain). Note occlusion
of capillary loops by swollen cytoplasm with
minimal increase in cellularity. (F) Electron
microscopy of sFlt-1treated rat. Note occlusion of capillary loops by swollen cytoplasm
with relative preservation of podocyte foot
processes (original magnification 2500). All
light micrographs taken at identical original
magnification of 40).

2104

Karumanchi et al: Preeclampsia and angiogenic factors

increased in normal pregnancy [44]. In preeclampsia,


prostacyclin production is lower than in normal pregnancy well before the onset of hypertension and proteinuria [45, 46]. Endothelial cells incubated with serum from
women with preeclampsia (vs. serum from normotensive
pregnant women) produce less prostacyclin in vitro, suggesting the presence of a circulating factor suppressing
prostacyclin synthesis [47]. Prostacyclin concentrations
are normal in pregnant women with chronic hypertension but without preeclampsia [48], providing further
circumstantial evidence that decreased prostacyclin is a
contributing cause of preeclampsia rather than a consequence of hypertension.
Thromboxane A 2 (TXA 2 ) is a potent vasoconstrictor synthesized by endothelial cells, activated platelets,
macrophages, and other organs. Urinary excretion of
TXA 2 metabolites has been reported to be increased in
preeclampsia by some [49] but not all investigators [46,
50]. TXA 2 production appears to parallel the severity of
preeclampsia, being higher in patients with coagulopathy
and pronounced platelet activation [50]. Some attempts
to prevent preeclampsia in high risk patients by inhibiting
platelet thromboxane synthesis using aspirin were successful but others found no benefit, either in low risk or
high risk populations [51, 52].
There is some evidence that nitric oxide, a vascular
smooth muscle relaxant synthesized by vascular endothelium, may also mediate the generalized vasodilation of
normal pregnancy [53, 54]. In rodent experiments, nitric oxide inhibition during pregnancy induces hypertension [5557] and proteinuria [58] and reverses the
norepinephrine and angiotensin II resistance characteristic of pregnancy [59, 60]. Thus, damage to vascular endothelium with decreased nitric oxide production might
contribute to vascular constriction in preeclampsia. Although some human studies have reported decreased
nitric oxide production in preeclampsia [6163], others
report nitric oxide production to be unchanged [64, 65]
or even increased [66, 67]. Unfortunately, studies of nitric oxide production are difficult due to its extremely
short circulating half-life and other challenges to its
measurement.
Endothelin-1, a potent vasoconstrictor that can be released by vascular endothelial cells in response to injury [68], has also been implicated in the hypertension of
preeclampsia. Reports of plasma endothelin concentrations in preeclampsia have been mixed, with most [6971]
but not all [50] investigators reporting a modest increase
in circulating concentrations. The release of endothelin
by cultured endothelial cells is enhanced by exposure
to plasma from preeclamptic patients, as compared with
plasma from women with normal pregnancies [72]. Limited animal data suggest that hypertension induced by
endothelin-1 administration during pregnancy produces
proteinuria [73].

Renal dysfunction, proteinuria, and renal pathology


In normal pregnancy, glomerular filtration rate (GFR)
as measured by inulin clearance and renal plasma flow
(para-aminohippurate clearance) increases by 40% to
60% during the first trimester [74, 75], resulting in a fall in
serum markers of renal clearance, including blood urea
nitrogen (BUN), creatinine, and uric acid. In preeclampsia, both GFR and renal plasma flow decrease by 30%
to 40% compared with normal pregnancy of the same
duration [76, 77]. Rarely, prolonged renal hypoperfusion
with resulting acute tubular necrosis can occur in severe
preeclampsia. It is important to note that in preeclampsia, BUN and creatinine often remain in the normal range
for nonpregnant women despite a significant decrease in
GFR from the high level of normal pregnancy.
Proteinuria may rarely precede hypertension but usually accompanies or follows it. After pregnancy is terminated, proteinuria commonly disappears within 3 to 8
weeks, but occasionally persists for months. The quantity
of protein excreted in the urine varies widely from less
than a gram to 8 to 10 g per day. The urinary sediment is
usually bland; red blood cells and cellular casts are rare.
Preeclampsia is the leading cause of nephrotic syndrome
during pregnancy. Recent data suggest that a loss of both
size and charge selectivity of the glomerular barrier contribute to the development of albuminuria [77].
Preeclampsia is associated with a characteristic
glomerular lesion, glomerular capillary endotheliosis (Fig. 2A to C) [7880]. By light microscopy, the
glomeruli are enlarged and the glomerular capillary lumen is bloodless due to endothelial and mesangial cell
swelling and hypertrophy. While these changes may be
focally present in other conditions (such as abruptio placentae), only in preeclampsia is endotheliosis so prominent and widespread. Glomerular cellularity is at most
slightly increased. Mesangial interposition may occur in
severe cases or in the healing stages. Glomerular visceral epithelial cells (podocytes) usually appear swollen
with periodic acid-Schiff (PAS)-positive hyaline droplets.
Immunofluorescence may reveal deposition of fibrin
or fibrinogen derivatives particularly in biopsies done
within 2 weeks postpartum [81]. Electron microscopy
is important to confirm endotheliosis and the loss of
endothelial fenenstrae. Remarkably, despite heavy proteinuria the podocyte foot processes are relatively preserved [82]. Glomerular subendothelial and occasional
mesangial electron-dense deposits can be seen. These
likely relate to fibrin or related breakdown products. Mild
glomerular endotheliosis has been noted in up to 50%
of patients with pregnancy-induced hypertension without proteinuria [83], suggesting that pregnancy-induced
hypertension may in some cases reflect an earlier or
milder form of the same syndrome. Glomerular enlargement and endothelial swelling usually disappear within
8 weeks of delivery, coinciding with resolution of the

Karumanchi et al: Preeclampsia and angiogenic factors

hypertension and proteinuria. Focal segmental glomerulosclerosis (FSGS) can accompany the generalized
glomerular endotheliosis of preeclampsia in 50% or more
of cases [84, 85].
Edema
Although the presence of edema is not necessary for
the diagnosis, sudden weight gain, with edema of the
feet, hands, and face, is a common presenting symptom in preeclampsia [32]. Salt loads are excreted more
slowly than in normal pregnancy [86, 87]. The edema of
preeclampsia is unlike that in congestive heart failure,
hepatic cirrhosis, and nephrotic syndrome, where low
effective circulating volume leads to high plasma renin
and aldosterone concentrations and secondary renal
sodium retention (underfill edema) [88]. Instead, renin
and aldosterone concentrations are suppressed relative
to normal pregnancy [39]. The edema of preeclampsia
thus resembles the overfill edema of acute glomerulonephritis or of acute ischemic renal failure in which
GFR is decreased out of proportion to the drop in renal
plasma flow and in which glomerular-tubular imbalance
has been invoked as a cause of salt retention.
Decreased GFR, increased capillary permeability, and
hypoalbuminemia may contribute to the edema formation. Albumin-bound Evans blue dye disappears
more quickly from the intravascular space in preeclamptic women compared with normal pregnant women,
suggesting endothelial permeability is increased [89].
However, this phenomenon is also seen in non-pregnant
patients with heart failure and the nephrotic syndrome
[90, 91]. Hypoalbuminemia is common and may contribute to edema. However, correction of hypoalbuminemia with albumin infusions in preeclampsia patients
usually does not produce a diuresis, suggesting that the
hypoalbuminemia alone is not responsible for the edema
[92].
Hyperuricemia
The association between preeclampsia and elevated
serum uric acid was first noted more than 80 years
ago [93]. Serum uric acid levels are usually elevated in
preeclampsia, and the degree of uric acid elevation has
been correlated with the severity of proteinuria [94], renal
pathologic changes [83], maternal morbidity [95], and fetal demise [96]. Often, the elevation of uric acid precedes
the onset of proteinuria and fall in GFR [97]. Although
it has been proposed that uric acid generation may be increased in preeclampsia as a result of tissue ischemia [98],
most evidence suggests that decreased renal clearance is
the more important mechanism [99]. Similar decreases in
uric acid clearance have been noted by infusions of vasoconstrictors in humans [100]. It has recently been sug-

2105

gested that hyperuricemia may also directly contribute


to vascular damage and hypertension [101, 102].
Coagulopathy and HELLP syndrome
Preeclampsia is sometimes complicated by consumptive coagulopathy and thrombotic microangiopathy culminating in the HELLP syndrome. HELLP syndrome
develops in approximately 10% to 20% of women with
severe preeclampsia. The tendency of blood to coagulate
is increased in normal pregnancy at least in part because
of changes in the circulating concentrations of coagulant
factors. In the indolent microangiopathy of preeclampsia, it is mainly the factors that are synthesized in the
vascular endothelium that are abnormal. These include
prostacyclins (PGI 2 ), von Willebrand factor, thrombomodulin, cellular fibronectin, and PAI-1 [103106]. Even
in the absence of overt coagulopathy, serum and urine fibrin degradation products are increased, implicating subclinical activation of the coagulation cascade.
These changes are not simply epiphenomena in response to hypertension. Plasma concentrations of cellular fibronectin may be increased weeks before the onset
of hypertension [107]. Exposure of cultured endothelial
cells to serum from women with preeclampsia triggers
increased cellular filbronectin [108, 109] and thrombomodulin [110] release compared with serum from normotensive pregnant women, again suggesting that a
factor in preeclamptic serum is directly activating
endothelial cells. It is interesting to note that other
endothelial disorders, including thrombotic thrombocytopenic purpura/hemolytic uremic syndrome, vasculitis,
and disseminated intravascular coagulation can produce
similar alterations in markers of endothelial activation
[111113]. There is also evidence that platelet activation
is present in preeclampsia as a consequence of endothelial damage. Markers of platelet activation such as beta
thromboglobulin have been found to be elevated prior to
the onset of clinical disease [114]. Furthermore, adhesion
molecules such as soluble P-selectin, soluble E-selectin,
and soluble vascular cell adhesion molecule-1 (VCAM-1)
that reflect endothelial, platelet, and leukocyte activation
have also been reported to be elevated in preeclampsia
[115]. The severe thrombocytopenia that is occasionally
seen is likely due to consumption during intravascular
coagulation.
Neurologic abnormalities
Preeclampsia is occasionally complicated by the development of seizures (eclampsia). Other neurologic symptoms include headache, blurred vision, and temporary
loss of vision. The neurologic changes in eclampsia and
preeclampsia have been attributed to cerebral edema and
vasoconstriction. Brain edema by magnetic resonance

2106

Karumanchi et al: Preeclampsia and angiogenic factors

(MR) imaging and abnormalities in circulating endothelial markers correlate closely with the occurrence of
eclampsia, suggesting that endothelial damage and disruption of the blood-brain barrier may be an underlying
mechanism [116]. The cerebral edema of eclampsia involves predominantly the posterior portions of the white
matter, and has been referred to as reversible posterior leukoencephalopathy syndrome (RPLS) [117]. Interestingly, patients with thrombotic thrombocytopenic
purpura (TTP), another disorder with microangiopathy
also have features of RPLS on MR imaging examinations [118], as do some patients with hypertensive encephalopathy.
ABNORMAL PLACENTATION IN
PREECLAMPSIA
It was 1939, when E.W. Page first proposed that
placenta plays a central role in preeclampsia [3]. Placentas from severe preeclamptic pregnancies classically
have numerous infarcts, sclerotic narrowing of arteries and arterioles, and fibrin deposition and thrombosis.
Physiologic studies have confirmed that uteroplacental
blood flow is diminished and uterine vascular resistance is
increased in preeclamptic women. In addition, placental
ischemia induced by mechanical constriction of the uterine arteries or aorta produces hypertension, proteinuria,
and, variably, glomerular endotheliosis, in a variety of
species [119121]. Placental ischemia may be contributed
to by a decrease in placental production of nitric oxide
[122]. However, placental ischemia alone, as in intrauterine growth restriction, does not appear to be sufficient
to produce preeclampsia. Thus, although uteroplacental
ischemia is an important trigger of preeclampsia, the response to this ischemiaeither at the placental or the
maternal systemic levelmust be variable.
Evidence suggests that placental ischemia in
preeclampsia occurs as a result of defective placental vascular remodeling. Early in normal placental
development, cytotrophoblast cells, fetal in origin, attach
to the uterine deciduas via anchoring villi (see Fig. 3).
Floating villi, containing fetal vessels which participate in
nutrient exchange, are bathed in maternal blood which
fills the intravillous space via uterine spiral arteries.
During placental vasculogenesis, a small percentage of
cytotrophoblasts in the anchoring villi break through
the syncytium and migrate into the endometrium.
These extravillous trophoblasts invade the uterine
spiral arteries of the myometrium, a process that peaks
at about 12 weeks gestation. By 18 to 20 weeks, the
endometrial and superficial myometrial segments of
the spiral arteries are lined by cells of cytotrophoblast
origin, with transformation of these vessels from small
resistance vessels to flaccid, high-caliber capacitance
vessels [123, 124]. This dramatic transformation allows

the increase in placental blood flow needed to sustain


the fetus through the pregnancy.
The characteristic pathologic placental lesion in severe
preeclampsia is diminished endovascular invasion by cytotrophoblasts and failure of uterine spiral arteriolar remodeling [123, 125]. Cytotrophoblast invasion is limited
to the proximal decidua, and the myometrial segments
of the spiral arteries remain narrow and undilated [126],
resulting in uterine hypoperfusion. Zhou et al [127, 128]
have shown that invasive cytotrophoblasts down-regulate
the expression of adhesion molecules characteristic of
their epithelial cell origin and adopt a cell-surface adhesion phenotype typical of endothelial cells, a process
referred to as pseudovasculogenesis. They hypothesized
and confirmed that in preeclampsia, cytotrophoblast cells
fail to undergo this switching of cell-surface integrins
and adhesion molecules [129]. This work suggests that
cytotrophoblast differentiation is abnormal in severe
preeclampsia, and may be an early defect that eventually
leads to placental ischemia. Others have demonstrated
that hypoxia-inducible factor-1 (HIF-1) is up-regulated in
preeclampsia and have suggested that HIF-1 target genes
such as transforming growth factor-beta 3 (TGF-b3) may
block the cytotrophoblast invasion [130132]. More recently, heparin-binding epidermal growth factor (EGF)like growth factor (HB-EGF) has been demonstrated to
be decreased in preeclamptic cytotrophoblasts; however,
its role in cytotrophoblast differentiation and invasion is
unclear [133]. Interestingly, uteroplacental ischemia produced in monkeys by aortic constriction late in gestation
does not produce the defective placental cytotrophoblast
invasion seen in preeclampsia, though it is associated with
proteinuria and hypertension [134].

THE PLACENTAL FACTOR IN PREECLAMPSIA


All of the clinical manifestations of preeclampsia can
be attributed to endothelial cell dysfunction leading to
end-organ damage and hypoperfusion. Based on this
observation, it has been suggested that there may be
a circulating factor, likely placental in origin, which
affects systemic endothelial endothelial cell function
and leads to the clinical syndrome of preeclampsia [4,
135]. Intense investigation has focused on the search
for a placental factor which might induce the maternal
syndrome. Although many candidate factors, including
tumor necrosis factor-a (TNF-a), interleukin (IL)-6,
IL-1a, IL-1b, Fas ligand, neurokinin B, and asymmetric
dimethy L-arginine (ADMA) have been suggested, none
so far has been proven to be etiologic [1, 2, 5, 136, 137].
Recent data suggest that increased syncytiotrophoblast
shedding as a consequence of placental apoptosis may
contribute to endothelial dysfunction [138140]; however, there is no in vivo evidence so far that circulating

2107

Karumanchi et al: Preeclampsia and angiogenic factors

Placenta Anchoring villus


cytotrophoblast
column
Cytotrophoblast
stem cells

Decidua

Myometrium

Cytotrophoblast
Tunica media
smooth muscle

Syncytiotrophoblast
Maternal blood

Bloo

d flow

Spiral
artery

Floating villus
Cytotrophoblast Endovascular
cytotrophoblast
Placenta Anchoring villus
cytotrophobast
column
Cytotrophoblast
stem cells

Decidua

Myometrium

Cytotrophoblast
Maternal
endothelial cells
Syncytiotrophoblast
Maternal blood
Fetal blood
vessel

Less
blood
flow

Maternal
endothelial
cells

Tunica media
vascular smooth
muscle layer

Spiral artery

Cytotrophoblast

Floating villus

Fig. 3. Abnormal placentation in preeclampsia. Exchange of oxygen, nutrients, and waste


products between the fetus and the mother
depends on adequate placental perfusion by
maternal vessels. In normal placental development, invasive cytotrophoblasts of fetal
origin invade the maternal spiral arteries,
transforming them from small-caliber resistance vessels to high-caliber capacitance vessels capable of providing placental perfusion
adequate to sustain the growing fetus. During
the process of vascular invasion, the cytotrophoblasts differentiate from an epithelial phenotype to an endothelial phenotype, a process
referred to as pseudovasculogenesis (upper
panel). In preeclampsia, cytotrophoblasts fail
to adopt an invasive endothelial phenotype.
Instead, invasion of the spiral arteries is shallow and they remain small caliber, resistance
vessels (lower panel). This may result in the
placental ischemia.

Anticoagulant and
vasodilatory factors

VEGF

PIGF

Healthy endothelial cell

FLT-1
Healthy
placenta

Blood vessel

sFLT-1
Procoagulant and
vasoconstricting factors

Dysfunctional endothelial cell


Blood vessel
Preeclampsia
placenta

Fig. 4. Soluble Flt-1 (sFlt-1) causes endothelial dysfunction by antagonizing vascular endothelial growth factor (VEGF) and placental growth
factor (PlGF). There is mounting evidence that VEGF, and possibly PlGF, are required to maintain endothelial health in several tissues including
the kidney and perhaps the placenta. In normal pregnancy, the placenta produces modest concentrations of VEGF, PlGF, and soluble Flt-1. In
preeclampsia, excess placental soluble Flt-1 binds circulating VEGF and PlGF and prevents their interaction with endothelial cell-surface receptors.
This results in endothelial cell dysfunction, including decreased prostacyclin, nitric oxide production and release of procoagulant proteins such as
von Willebrand factor, endothelin, cellular fibronectin, and thrombomodulin.

2108

Karumanchi et al: Preeclampsia and angiogenic factors

placental debris induces preeclampsia-like features. Recently, preeclampsia-like features were noted in p57kip2 (a
cell-cycle inhibitor) knockout mice, however, the mechanisms responsible for this phenotype are unclear [141].
Some [142], but not all [143], investigators have found
markers of oxidative stress to be elevated in women
with preeclampsia, suggesting that generation of reactive
oxygen-free radicals may contribute to endothelial dysfunction [2]. Decreased intake of the antioxidant vitamin
C and low circulating ascorbic acid concentrations are
associated with an increased risk of preeclampsia [144].
A small randomized controlled trial found that antioxidant therapy decreased the incidence of preeclampsia,
suggesting that oxidative stress may be a cause, rather
than a consequence, of the syndrome [145]. Additional
studies are needed to test this hypothesis.
As noted earlier, increased sensitivity to the vasopressor effects of angiotensin is a well-established feature
of preeclampsia. Two recent advances have illuminated
possible mechanisms for this phenomenon. AbdAlla
et al [146] reported increased angiotensin-1/bradykinin
B2 receptor heterodimers in women with preeclampsia,
and showed that such heterodimers can result in enhanced angiotensin II responsiveness [146]. In a similar
vein, Wallukat et al [147] noted increased concentrations
of agonistic antibodies to the angiotensin II type 1 (AT1) receptor in women with preeclampsia. These autoantibodies may induce the production of reactive oxygen
species and block cytotrophoblast invasion in vitro [148,
149], thus accounting for several of the clinical features of
preeclampsia. Further work is needed to establish if these
alterations are etiologic or epiphenomenona that might
be encountered in other examples of microangiopathy.
Recently, gene expression profiling has been used to
search for candidate factors produced by the placenta in
preeclampsia. Using this approach, we found that placental sFlt-1 mRNA (soluble fms-like tyrosine kinase-1) is
up-regulated in preeclampsia [150]. sFlt-1 is a splice variant of the vascular endothelial growth factor (VEGF))
receptor Flt-1, lacking the transmembrane and cytoplasmic domains. sFlt is made in large amounts by the
placenta and is released into the maternal circulation
[151153]. sFlt-1 acts as a potent VEGF and placental growth factor (PlGF) antagonist by binding these
molecules in the circulation [154]. Circulating sFlt-1 concentrations are increased in women with established
preeclampsia [150, 155157]. Consistent with the antagonistic effect of sFlt-1, free (or unbound) VEGF and
free (or unbound) PlGF concentrations are decreased in
preeclamptic women at disease presentation and even
before the onset of clinical symptoms [150, 158, 159] (see
Fig. 4). When administered to pregnant and nonpregnant
rats, sFlt-1 produces a syndrome of hypertension, proteinuria, and glomerular endotheliosis that mimics the
human syndrome of preeclampsia (Fig. 2D to F) [150].

Recently, our laboratory as well as others have observed


that there is a marked rise in circulating sFlt-1 concentration beginning about 5 to 6 weeks before the onset of
clinical preeclampsia, accompanied by decreases in the
circulating free PlGF and VEGF [160, 161]. Moreover,
alterations in these circulating angiogenic proteins correlated with disease severity, earlier onset of preeclampsia,
and the birth of an infant small for gestational age. Finally,
we have also recently reported that decreased urinary
concentrations of free PlGF during midgestation predict the subsequent development of clinical preeclampsia
[162]. These data lend further support to the hypothesis that circulating sFlt-1 may have a pathogenic role in
preeclampsia.
The possibility that antagonism of VEGF and PlGF
might play a role in preeclampsia has sound physiologic
underpinnings (Fig. 4). In addition to being a potent
promoter of angiogenesis, VEGF is known to induce
nitric oxide and vasodilatory prostacyclins in endothelial cells, decreasing vascular tone and blood pressure
[163]. There is evidence from animal models that VEGF
is important in maintaining glomerular endothelial cell
health and healing [164, 165], and its absence induces proteinuria and glomerular endotheliosis [166, 167]. In antiangiogenic oncology trials, antagonism of VEGF using
neutralizing antibodies and VEGF receptor inhibitors
can produce headaches, hypertension, proteinuria, and
coagulopathy in human subjects [168170]. Furthermore,
exogenous VEGF and PlGF can reverse the antiangiogenic properties of preeclamptic serum, as assessed by in
vitro angiogenesis assays [150]. Thus, the antiangiogenic
effects of sFlt-1 may account for many of the manifestations of preeclampsia, including the unique glomerular effects. Some tissues targeted in preeclampsia (i.e.,
renal glomeruli, hepatic sinusoids) have fenestrated endothelia, which are necessary for physiologic diffusion of
water and solutes. It has been shown that VEGF, which
is expressed constitutively in these organs, is necessary
to induce and maintain the health of the fenestrated endothelium [167, 171]. Thus, it is intuitive that tissues with
fenestrated endothelium, especially the renal glomerulus, might be particularly susceptible to damage by sFlt1mediated VEGF blockade.
How placental dysfunction is related to placental sFlt1 production, and why placental perfusion is deranged
in preeclampsia remains unknown. Recent data using
in vitro primary cytotrophoblast cultures suggest that
placental hypoxia may play an important role in upregulating sFlt-1 production [172]. It remains to be shown
if sFlt-1 can induce other features of preeclampsia such
as the HELLP syndrome or if additional synergistic factors are needed. Additionally, the Flt-1 locus at 13q12 has
not been localized within the genomic region identified
by genetic linkage studies for preeclampsia. However, if
the Flt-1 locus contributed to preeclampsia, one could

Karumanchi et al: Preeclampsia and angiogenic factors

hypothesize that patients who carry trisomy 13 fetuses


should have a 50% increased risk of preeclampsia. Two
case-control studies done several years ago did in fact
demonstrate higher incidence of preeclampsia in mothers who carry trisomy 13 fetuses as compared to other trisomies or control pregnant patients [24, 173]. Although
sFlt-1 levels were not measured in the patients with trisomy 13, these data are consistent with the hypothesis that
elevated production of sFlt-1 may lead to preeclampsia.
IMPACT OF PREECLAMPSIA ON LONG-TERM
MATERNAL HEALTH
Women with hypertensive disorders of pregnancy have
an increased long-term incidence of dyslipidemias, insulin
resistance, and cardiovascular diseases [174176]. Sibai,
el-Nazer, and Gonzalez-Ruiz [177] reported an 18-fold increase in the incidence of chronic salt-sensitive hypertension in women with a history of preeclampsia. It has been
speculated that this late-onset hypertension may play a
role in the eightfold increase in cardiovascular mortality
that is also observed in these patients [176]. However, it is
difficult to determine causality, because many of the risk
factors for preeclampsia (such as diabetes mellitus, hypertension, and renal insufficiency) predispose to hypertension and cardiovascular disease as well. It has also been
suggested that subtle renal injury caused by preeclampsia
can eventually lead to the development of chronic hypertension [178, 179]. It is tempting to speculate that the longterm cardiovascular complications noted in some patients
who have had preeclampsia may be due to a chronic antiangiogenic state resulting from polymorphisms in genes
such as sFlt-1. Additionally, patients with preeclampsia
are said to have a decreased long-term incidence of malignancy [180], a provocative observation disputed by some
[181], that may suggest that the antiangiogenic state of
preeclampsia may reflect a permanent state of the maternal milieu.
CONCLUSION
Preeclampsia is a systemic disorder characterized by
widespread endothelial dysfunction. Substantial animal
and human experimental evidence fulfilling Kochs postulates [182] supports the hypothesis that placental sFlt1,
an antiangiogenic protein, may be etiologic. Several important questions remain to be explored, and many of
these can now be approached experimentally. Why do
certain conditions (see Table 1) predispose to preeclampsia? Are there additional synergistic factors which might
induce/predispose to HELLP syndrome? What is the relationship of sFlt-1 with other postulated mediators of
the maternal syndrome such as hyperuricemia, ADMA,
or antibodies to angiotensin II AT-1 receptor? What
governs regulation of sFlt1 transcription and splicing,

2109

and why is it abnormal in preeclampsia? Is placental ischemia a cause or a consequence of excessive placental sFlt1? Large genetic trials such as GOPEC (Genetics
of Preeclampsia) [6], ongoing proteomic/genomic studies of tissue and blood specimens from patients with
preeclampsia, and further characterization of the sFlt-1
induced animal model of preeclampsia may help to answer these questions.
The implications of these advances on our clinical
management of preeclampsia may be profound. Large,
prospective studies are needed to describe in detail
the changes in sFlt1, VEGF, and PlGF that precede
preeclampsia onset; measurement of these factors in
pregnant women may prove to be useful diagnostically. Pharmacologic intervention aimed at restoring
sFlt1/PlGF/VEGF balance may prevent or modify the
course of preeclampsia. Safely prolonging pregnancy
by only a few weeks could substantially reduce maternal and neonatal morbidity and mortality resulting
from preeclampsia, the worlds most common glomerular
disease.
ACKNOWLEDGMENTS
We would like to thank Ben Sachs and Kee-Hak Lim of the Department of Obstetrics and Gynecology at the Beth Israel Deaconess
Medical Center (BIDMC) and Marshall Lindheimer of the University of Chicago for helpful discussions and strong support. This work
was supported by NIH grants (DK02825, DK64255, and DK65997),
the American Society of Nephrology Carl W. Gottschalk award, and
BIDMC seed funds to S.A.K. S.A.K, S.E.M, and V.P.S are listed as coinventors in a patent filed by BIDMC for the use of angiogenesis-related
proteins for the diagnosis and treatment of preeclampsia. Therapeutic
claims of the patent have been licensed by BIDMC to Scios, Inc, CA.
Reprint requests to S. Ananth Karumanchi, M.D., Beth Israel Deaconess Medical Center, Renal Division, 330 Brookline Avenue, Dana
517, Boston, MA 02215.
E-mail: sananth@bidmc.harvard.edu

REFERENCES
1. WALKER JJ: Pre-eclampsia. Lancet 356:12601265, 2000
2. ROBERTS JM, COOPER DW: Pathogenesis and genetics of preeclampsia. Lancet 357:5356, 2001
3. PAGE EW: The relation between hydatid moles, relative ischemia
of the gravid uterus and the placental origin of eclampsia. Am J
Obstet Gynecol 37:291293, 1939
4. ROBERTS JM, TAYLOR RN, MUSCI TJ, et al: Preeclampsia: An endothelial cell disorder. Am J Obstet Gynecol 161:12001204, 1989
5. ROBERTS JM: Preeclampsia: What we know and what we do not
know. Semin Perinatol 24:2428, 2000
6. LACHMEIJER AM, DEKKER GA, PALS G, et al: Searching for
preeclampsia genes: The current position. Eur J Obstet Gynecol
Reprod Biol 105:94113, 2002
7. THADHANI R, STAMPFER MJ, HUNTER DJ, et al: High body mass
index and hypercholesterolemia: risk of hypertensive disorders of
pregnancy. Obstet Gynecol 94:543550, 1999
8. ROBILLARD PY, HULSEY TC, ALEXANDER GR, et al: Paternity patterns and risk of preeclampsia in the last pregnancy in multiparae.
J Reprod Immunol 24:112, 1993
9. TRUPIN LS, SIMON LP, ESKENAZI B: Change in paternity: A risk
factor for preeclampsia in multiparas. Epidemiology 7:240244,
1996

2110

Karumanchi et al: Preeclampsia and angiogenic factors

10. SKJARVEN R, WILCOX AJ, LIE RT: The interval between pregnancies
and the risk of preeclampsia. N Engl J Med 346:3338, 2002
11. ESPLIN MS, FAUSETT MB, FRASER A, et al: Paternal and maternal
components of the predisposition to preeclampsia. N Engl J Med
344:867872, 2001
12. WARD K, HATA A, JEUNEMAITRE X, et al: A molecular variant of
angiotensinogen associated with preeclampsia. Nat Genet 4:5961,
1993
13. ARNGRIMSSON R, HAYWARD C, NADAUD S, et al: Evidence for a
familial pregnancy-induced hypertension locus in the eNOS-gene
region. Am J Hum Genet 61:354362, 1997
14. DIZON-TOWNSON DS, NELSON LM, EASTON K, WARD K: The factor V
Leiden mutation may predispose women to severe preeclampsia.
Am J Obstet Gynecol 175:902905, 1996
15. SOHDA S, ARINAMI T, HAMADA H, et al: Methylenetetrahydrofolate
reductase polymorphism and pre-eclampsia. J Med Genet 34:525
526, 1997
16. ARNGRIMSSON R, GEIRSSON RT, COOKE A, et al: Renin gene restriction fragment length polymorphisms do not show linkage with
preeclampsia and eclampsia. Acta Obstet Gynecol Scand 73:1013,
1994
17. LADE JA, MOSES EK, GUO G, et al: The eNOS gene: A candidate
for the preeclampsia susceptibility locus? Hypertens Preg 18:8193,
1999
18. LACHMEIJER AM, ARNGRIMSSON R, BASTIAANS EJ, et al: Mutations in the gene for methylenetetrahydrofolate reductase, homocysteine levels, and vitamin status in women with a history of
preeclampsia. Am J Obstet Gynecol 184:394402, 2001
19. OSHAUGHNESSY KM, FU B, FERRARO F, et al: Factor V Leiden and
thermolabile methylenetetrahydrofolate reductase gene variants
in an East Anglian preeclampsia cohort. Hypertension 33:1338
1341, 1999
20. GUO G, WILTON AN, FU Y, et al: Angiotensinogen gene variation
in a population case-control study of preeclampsia/eclampsia in
Australians and Chinese. Electrophoresis 18:16461649, 1997
21. ARNGRIMSSON R, SIGURARDTTIR S, FRIGGE ML, et al: A genomewide scan reveals a maternal susceptibility locus for pre-eclampsia
on chromosome 2p13. Hum Mol Genet 8:17991805, 1999
22. MOSES EK, LADE JA, GUO G, et al: A genome scan in families from
Australia and New Zealand confirms the presence of a maternal
susceptibility locus for pre-eclampsia, on chromosome 2. Am J
Hum Genet 67:15811585, 2000
23. LACHMEIJER AM, ARNGRIMSSON R, BASTIAANS EJ, et al: A genomewide scan for preeclampsia in the Netherlands. Eur J Hum Genet
9:758764, 2001
24. TUOHY JF, JAMES DK: Pre-eclampsia and trisomy 13. Br J Obstet
Gynaecol 99:891894, 1992
25. GELLER DS, FARHI A, PINKERTON N, et al: Activating mineralocorticoid receptor mutation in hypertension exacerbated by pregnancy.
Science 289:119123, 2000
26. KEYES LE, ARMAZA JF, NIERMEYER S, et al: Intrauterine growth restriction, preeclampsia, and intrauterine mortality at high altitude
in Bolivia. Pediatr Res 16:16, 2003
27. CHESLEY LC: History and epidemiology of preeclampsiaeclampsia. Clin Obstet Gynecol 27:801820, 1984
28. WALLENBURG HCS: Hemodynamics of hypertensive pregnancy, in
Handbook of Hypertension, edited byRubin PC, New York, Elsevier Science, 1988, pp 66101
29. SCHOBEL HP, FISCHER T, HEUSZER K, et al: PreeclampsiaA
state of sympathetic overactivity. N Engl J Med 335:14801485,
1996
30. MANYONDA IT, SLATER DM, FENSKE C, et al: A role for noradrenaline in pre-eclampsia: Towards a unifying hypothesis for the
pathophysiology. Br J Obstet Gynaecol 105:641648, 1998
31. EASTERLING TR, BENEDETTI TJ, SCHMUCKER BC, MILLARD SP: Maternal hemodynamics in normal and preeclamptic pregnancies: A
longitudinal study. Obstet Gynecol 76:10611069, 1990
32. CHESLEY L: Hypertensive Disorders of Pregnancy, New York, NY,
Appleton Century Crafts, 1978
33. VICTOR R, MARK A: The sympathetic nervous system in human
hypertension, in Hypertension: Pathophysiology, Diagnosis, and
Management, edited byLaragh J, Brenner B, 2nd ed., New York,
NY, Raven Press, 1995, pp 865887

34. GANT NF, DALEY GL, CHAND S, et al: A study of angiotensin II


pressor response throughout primigravid pregnancy. J Clin Invest
52:26822689, 1973
35. KYLE PM, BUCKLEY D, KISSANE J, et al: The angiotensin sensitivity
test and low-dose aspirin are ineffective methods to predict and
prevent hypertensive disorders in nulliparous pregnancy. Am J
Obstet Gynecol 173:865872, 1995
36. LUFT FC, MILLER JZ, COHEN SJ, et al: Heritable aspects of salt
sensitivity. Am J Cardiol 61:1H6H, 1988
37. REDMAN CW: Maternal plasma volume and disorders of pregnancy.
Br Med J (Clin Res Ed) 288:955956, 1984
38. TAPIA HR, JOHNSON CE, STRONG CG: Renin-angiotensin system in
normal and in hypertensive disease of pregnancy. Lancet 2:847
850, 1972
39. AUGUST P, LENZ T, ALES KL, et al: Longitudinal study of the
renin-angiotensin-aldosterone system in hypertensive pregnant
women: deviations related to the development of superimposed
preeclampsia. Am J Obstet Gynecol 163:16121621, 1990
40. OKUNO S, HAMADA H, YASUOKA M, et al: Brain natriuretic peptide
(BNP) and cyclic guanosine monophosphate (cGMP) levels in normal pregnancy and preeclampsia. J Obstet Gynaecol Res 25:407
410, 1999
41. FINNERLY FAJ, BUCHOLZ JH, GUILLAUDEU RL: The blood volume
and plasma proteins during levarterenol-induced hypertension. J
Clin Invest 37:425429, 1958
42. AGATISA PK, NESS RB, ROBERTS JM, et al: Impairment of endothelial function in women with a history of preeclampsia: an indicator
of cardiovascular risk. Am J Physiol Heart Circ Physiol 286:H1389
H1393, 2004
43. CHAMBERS JC, FUSI L, MALIK IS, et al: Association of maternal
endothelial dysfunction with preeclampsia. JAMA 285:16071612,
2001
44. GOODMAN RP, KILLAM AP, BRASH AR, BRANCH RA: Prostacyclin
production during pregnancy: Comparison of production during
normal pregnancy and pregnancy complicated by hypertension.
Am J Obstet Gynecol 142:817822, 1982
45. FITZGERALD DJ, ENTMAN SS, MULLOY K, FITZGERALD GA: Decreased prostacyclin biosynthesis preceding the clinical manifestation of pregnancy-induced hypertension. Circulation 75:956963,
1987
46. MILLS JL, DERSIMONIAN R, RAYMOND E, et al: Prostacyclin and
thromboxane changes predating clinical onset of preeclampsia: A
multicenter prospective study. JAMA 282:356362, 1999
47. BAKER PN, DAVIDGE ST, BARANKIEWICZ J, ROBERTS JM: Plasma
of preeclamptic women stimulates and then inhibits endothelial
prostacyclin. Hypertension 27:5661, 1996
48. MOUTQUIN JM, LINDSAY C, ARIAL N, et al: Do prostacyclin and
thromboxane contribute to the protective effect of pregnancies
with chronic hypertension? A preliminary prospective longitudinal
study. Am J Obstet Gynecol 177:14831490, 1997
49. FITZGERALD DJ, ROCKI W, MURRAY R, et al: Thromboxane A 2
synthesis in pregnancy-induced hypertension. Lancet 335:751754,
1990
50. PAARLBERG KM, DE JONG CL, VAN GEIJN HP, et al: Vasoactive mediators in pregnancy-induced hypertensive disorders: a longitudinal
study. Am J Obstet Gynecol 179:15591564, 1998
51. CARITIS S, SIBAI B, HAUTH J, et al: Low-dose aspirin to prevent
preeclampsia in women at high risk. N Engl J Med 338:701705,
1998
52. CLASP: A randomised trial of low-dose aspirin for the prevention and treatment of pre-eclampsia among 9364 pregnant women.
CLASP (Collaborative Low-dose Aspirin Study in Pregnancy)
Collaborative Group. Lancet 343:619629, 1994
53. WILLIAMS DJ, VALLANCE PJ, NEILD GH, et al: Nitric oxide-mediated
vasodilation in human pregnancy. Am J Physiol 272:H748H752,
1997
54. CONRAD KP, KERCHNER LJ, MOSHER MD: Plasma and 24-h NO(x)
and cGMP during normal pregnancy and preeclampsia in women
on a reduced NO(x) diet. Am J Physiol 277:F48F57, 1999
55. MOLNAR M, HERTELENDY F: N omega-nitro-L-arginine, an inhibitor
of nitric oxide synthesis, increases blood pressure in rats and reverses the pregnancy-induced refractoriness to vasopressor agents.
Am J Obstet Gynecol 166:15601567, 1992

Karumanchi et al: Preeclampsia and angiogenic factors

56. BUHIMSCHI I, YALLAMPALLI C, CHWALISZ K, GARFIELD RE: Preeclampsia-like conditions produced by nitric oxide inhibition: effects of l-arginine, d-arginine and steroid hormones. Hum Reprod
10:27232730, 1995
57. YALLAMPALLI C, GARFIELD RE: Inhibition of nitric oxide synthesis in rats during pregnancy produces signs similar to those of
preeclampsia. Am J Obstet Gynecol 169:13161320, 1993
58. BAYLIS C, MITRUKA B, DENG A: Chronic blockade of nitric oxide
synthesis in the rat produces systemic hypertension and glomerular
damage. J Clin Invest 90:278281, 1992
59. MARTINEZ-ORGADO J, GONZALEZ R, ALONSO MJ, SALAICES M: Impairment of fetal endothelium-dependent relaxation in a rat model
of preeclampsia by chronic nitric oxide synthase inhibition. J Soc
Gynecol Investig 11:8288, 2004
60. MAEDA T, YOSHIMURA T, OHSHIGE A, et al: Nitric oxide affects angiotensin II pressor response: Possible mechanism of
attenuated pressor response during pregnancy and etiology of
pregnancy-induced hypertension. Gynecol Obstet Invest 49:8487,
2000
61. RANTA V, VIINIKKA L, HALMESMAKI E, YLIKORKALA O: Nitric oxide
production with preeclampsia. Obstet Gynecol 93:442445, 1999
62. SHAAMASH AH, ELSNOSY ED, MAKHLOUF AM, et al: Maternal and
fetal serum nitric oxide (NO) concentrations in normal pregnancy,
pre-eclampsia and eclampsia. Int J Gynaecol Obstet 68:207214,
2000
63. SMARASON AK, ALLMAN KG, YOUNG D, REDMAN CW: Elevated levels of serum nitrate, a stable end product of nitric oxide, in women
with pre-eclampsia. Br J Obstet Gynaecol 104:538543, 1997
64. DAVIDGE ST, STRANKO CP, ROBERTS JM: Urine but not plasma nitric oxide metabolites are decreased in women with preeclampsia.
Am J Obstet Gynecol 174:10081013, 1996
65. SILVER RK, KUPFERMINC MJ, RUSSELL TL, et al: Evaluation of nitric
oxide as a mediator of severe preeclampsia. Am J Obstet Gynecol
175:10131017, 1996
66. SELIGMAN SP, BUYON JP, CLANCY RM, et al: The role of nitric oxide
in the pathogenesis of preeclampsia. Am J Obstet Gynecol 171:944
948, 1994
67. GARMENDIA JV, GUTIERREZ Y, BLANCA I, et al: Nitric oxide in different types of hypertension during pregnancy. Clin Sci (Lond)
93:413421, 1997
68. BATTISTINI B, DUSSAULT P: The many aspects of endothelins in
ischemia-reperfusion injury: emergence of a key mediator. J Invest
Surg 11:297313, 1998
69. SLOWINSKI T, NEUMAYER HH, STOLZE T, et al: Endothelin system in
normal and hypertensive pregnancy. Clin Sci (Lond) 103 (Suppl
48):446S449S, 2002
70. SCHIFF E, BEN-BARUCH G, PELEG E, et al: Immunoreactive circulating endothelin-1 in normal and hypertensive pregnancies. Am J
Obstet Gynecol 166:624628, 1992
71. CLARK BA, HALVORSON L, SACHS B, EPSTEIN FH: Plasma endothelin levels in preeclampsia: Elevation and correlation with uric acid
levels and renal impairment. Am J Obstet Gynecol 166:962968,
1992
72. SCALERA F, SCHLEMBACH D, BEINDER E: Production of vasoactive
substances by human umbilical vein endothelial cells after incubation with serum from preeclamptic patients. Eur J Obstet Gynecol
Reprod Biol 99:172178, 2001
73. GREENBERG SG, BAKER RS, YANG D, CLARK KE: Effects of continuous infusion of endothelin-1 in pregnant sheep. Hypertension
30:15851590, 1997
74. DAVISON JM, DUNLOP W: Renal hemodynamics and tubular function normal human pregnancy. Kidney Int 18:152161, 1980
75. SIMS E, KRANTZ K: Serial studies of renal function during pregnancy
and the puerperium in normal women. J Clin Invest 37:17641774,
1958
76. LAFAYETTE RA, DRUZIN M, SIBLEY R, et al: Nature of glomerular
dysfunction in pre-eclampsia. Kidney Int 54:12401249, 1998
77. MORAN P, BAYLIS PH, LINDHEIMER MD, DAVISON JM: Glomerular
ultrafiltration in normal and preeclamptic pregnancy. J Am Soc
Nephrol 14:648652, 2003
78. SPARGO B: The renal lesions in preeclampsia, in Hypertension in
Pregnancy, edited byLindheimer M, New York, Wiley Medical,
1976, pp 129137

2111

79. FISHER KA, LUGER A, SPARGO BH, LINDHEIMER MD: Hypertension in pregnancy: clinical-pathological correlations and remote
prognosis. Medicine (Baltimore) 60:267276, 1981
80. POLLAK V, NETTLES J: The kidney in toxemia of pregnancy; a clinical
and pathological study based on renal biopsies. Medicine 39:469
474, 1960
81. MORRIS R, VASSALDI P, BILLER F, MCCLUSKEY R: Immunofluorescent studies of renal biopsies in the diagnosis of toxemia of pregnancy. Obstet Gynecol 24:3246, 1964
82. MAUTNER W, CHURG J, GRISHMANN E, DACHS S: Preeclamptic
nephropathy. An electron microscopic study. Lab Invest 11:518
530, 1962
83. NOCHY BP, HINGLAIS N, FREUND M, et al: Renal lesions in the hypertensive syndromes of pregnancy: Immunomorphological and
ultrastructural studies in 114 cases. Clinl Nephrol 13:155162, 1980
84. GABER LW, SPARGO BH: Pregnancy-induced nephropathy: The significance of focal segmental glomerulosclerosis. Am J Kidney Dis
9:317323, 1987
85. HEATON JM, TURNER DR: Persistent renal damage following preeclampsia: A renal biopsy study of 13 patients. J Pathol 147:121
126, 1985
86. STRAUSS M: Observations on the etiology of the toxemia of
pregnancy-II. Production of acute exacerbations of toxemia by
sodium salts in pregnant women with hypoproteinemia. Am J Med
Sci 6:772783, 1937
87. GALLERY ED, BROWN MA: Control of sodium excretion in human
pregnancy. Am J Kidney Dis 9:290295, 1987
88. SCHRIER RW: Pathogenesis of sodium and water retention in highoutput and low-output cardiac failure, nephrotic syndrome, cirrhosis, and pregnancy (2). N Engl J Med 319:11271134, 1988
89. BROWN MA, ZAMMIT VC, LOWE SA: Capillary permeability and extracellular fluid volumes in pregnancy-induced hypertension. Clin
Sci (Lond) 77:599604, 1989
90. GALATIUS S, BENT-HANSEN L, WROBLEWSKI H, et al: Plasma disappearance of albumin and impact of capillary thickness in idiopathic
dilated cardiomyopathy and after heart transplantation. Circulation 102:319325, 2000
91. ROSTOKER G, BEHAR A, LAGRUE G: Vascular hyperpermeability in
nephrotic edema. Nephron 85:194200, 2000
92. ORLOFF J, WELT L, STOWE L: The effects of concentrated salt-poor
albumin on the metabolism of water and electrolytes in nephrosis
and toxemia of pregnancy. J Clin Invest 29:770780, 1950
93. SLEMMONS J, BOGERT L: The uric acid content of maternal and fetal
blood. J Biol Chem 32:6369, 1917
94. REDMAN CW, BONNAR J: Plasma urate changes in pre-eclampsia.
Br Med J 1:14841485, 1978
95. MARTIN JN, Jr., MAY WL, MAGANN EF, et al: Early risk assessment
of severe preeclampsia: Admission battery of symptoms and laboratory tests to predict likelihood of subsequent significant maternal
morbidity. Am J Obstet Gynecol 180:14071414, 1999
96. SAGEN N, HARAM K, NILSEN ST: Serum urate as a predictor of
fetal outcome in severe pre-eclampsia. Acta Obstet Gynecol Scand
63:7175, 1984
97. GALLERY ED, GYORY AZ: Glomerular and proximal renal tubular function in pregnancy-associated hypertension: A prospective
study. Eur J Obstet Gynecol Reprod Biol 9:312, 1979
98. MANY A, HUBEL CA, ROBERTS JM: Hyperuricemia and xanthine
oxidase in preeclampsia, revisited. Am J Obstet Gynecol 174:288
291, 1996
99. SCHAFFER N, DILL L, CADDEN J: Uric acid clearance in normal
pregnancy and preeclampsia. J Clin Invest 22:201206, 1943
100. FERRIS TF, GORDEN P: Effects of angiotensin and norepinephrine
upon urate clearance in man. Am J Med 44:359365, 1968
101. WATANABE S, KANG DH, FENG L, et al: Uric acid, hominoid evolution, and the pathogenesis of salt-sensitivity. Hypertension 40:355
360, 2002
102. KANG DH, FINCH J, NAKAGAWA T, et al: Uric acid, endothelial dysfunction and pre-eclampsia: Searching for a pathogenetic link. J
Hypertens 22:229235, 2004
103. TAYLOR RN, CROMBLEHOLME WR, FRIEDMAN SA, et al: High plasma
cellular fibronectin levels correlate with biochemical and clinical
features of preeclampsia but cannot be attributed to hypertension
alone. Am J Obstet Gynecol 165:895901, 1991

2112

Karumanchi et al: Preeclampsia and angiogenic factors

104. FRIEDMAN SA, SCHIFF E, EMEIS JJ, et al: Biochemical corroboration


of endothelial involvement in severe preeclampsia. Am J Obstet
Gynecol 172:202203, 1995
105. HSU CD, IRIYE B, JOHNSON TR, et al: Elevated circulating thrombomodulin in severe preeclampsia. Am J Obstet Gynecol 169:148
149, 1993
106. ESTELLES A, GILABERT J, AZNAR J, et al: Changes in the plasma
levels of type 1 and type 2 plasminogen activator inhibitors in normal pregnancy and in patients with severe preeclampsia. Blood
74:13321338, 1989
107. CHAVARRIA ME, LARA-GONZALEZ L, GONZALEZ-GLEASON A, et al:
Maternal plasma cellular fibronectin concentrations in normal and
preeclamptic pregnancies: A longitudinal study for early prediction of preeclampsia. Am J Obstet Gynecol 187:595601, 2002
108. ROBERTS JM, EDEP ME, GOLDFIEN A, TAYLOR RN: Sera from
preeclamptic women specifically activate human umbilical vein endothelial cells in vitro: Morphological and biochemical evidence.
Am J Reprod Immunol 27:101108, 1992
109. TAYLOR RN, CASAL DC, JONES LA, et al: Selective effects of
preeclamptic sera on human endothelial cell procoagulant protein
expression. Am J Obstet Gynecol 165:17051710, 1991
110. KOBAYASHI H, SADAKATA H, SUZUKI K, et al: Thrombomodulin
release from umbilical endothelial cells initiated by preeclampsia
plasma-induced neutrophil activation. Obstet Gynecol 92:425430,
1998
111. BOEHME MW, SCHMITT WH, YOUINOU P, et al: Clinical relevance of
elevated serum thrombomodulin and soluble E-selectin in patients
with Wegeners granulomatosis and other systemic vasculitides.
Am J Med 101:387394, 1996
112. GANDO S, NAKANISHI Y, KAMEUE T, NANZAKI S: Soluble thrombomodulin increases in patients with disseminated intravascular coagulation and in those with multiple organ dysfunction syndrome
after trauma: role of neutrophil elastase. J Trauma 39:660664,
1995
113. MORI Y, WADA H, OKUGAWA Y, et al: Increased plasma thrombomodulin as a vascular endothelial cell marker in patients with
thrombotic thrombocytopenic purpura and hemolytic uremic syndrome. Clin Appl Thromb Hemost 7:59, 2001
114. BALLEGEER VC, SPITZ B, DE BAENE LA, et al: Platelet activation
and vascular damage in gestational hypertension. Am J Obstet
Gynecol 166:629633, 1992
115. CHAIWORAPONGSA T, ROMERO R, YOSHIMATSU J, et al: Soluble adhesion molecule profile in normal pregnancy and pre-eclampsia. J
Matern Fetal Neonatal Med 12:1927, 2002
116. SCHWARTZ RB, FESKE SK, POLAK JF, et al: Preeclampsia-eclampsia:
Clinical and neuroradiographic correlates and insights into the
pathogenesis of hypertensive encephalopathy. Radiology 217:371
376, 2000
117. HINCHEY J, CHAVES C, APPIGNANI B, et al: A reversible posterior
leukoencephalopathy syndrome. N Engl J Med 334:494500, 1996
118. BAKSHI R, SHAIKH ZA, BATES VE, KINKEL PR: Thrombotic thrombocytopenic purpura: brain CT and MRI findings in 12 patients.
Neurology 52:12851288, 1999
119. KUMAR D: Chronic placetal ischemia in relation to toxemias of
pregnancy. A preliminary report. Am J Obstet Gynecol 84:1323
1329, 1962
120. COMBS CA, KATZ MA, KITZMILLER JL, BRESCIA RJ: Experimental
preeclampsia produced by chronic constriction of the lower aorta:
Validation with longitudinal blood pressure measurements in conscious rhesus monkeys. Am J Obstet Gynecol 169:215223, 1993
121. PODJARNY E, BAYLIS C, LOSONCZY G: Animal models of preeclampsia. Semin Perinatol 23:213, 1999
122. NORIS M, TODESCHINI M, CASSIS P, PASTA F, et al: l-arginine depletion in preeclampsia orients nitric oxide synthase toward oxidant
species. Hypertension 43:614622, 2004
123. BROSENS IA, ROBERTSON WB, DIXON HG: The role of the spiral
arteries in the pathogenesis of preeclampsia. Obstet Gynecol Annu
1:177191, 1972
124. DE WOLF F, DE WOLF-PEETERS C, BROSENS I, ROBERTSON WB: The
human placental bed: Electron microscopic study of trophoblastic invasion of spiral arteries. Am J Obstet Gynecol 137:5870,
1980
125. ROBERTSON WB, BROSENS I, DIXON HG: The pathological response

126.

127.
128.
129.

130.

131.

132.

133.
134.
135.
136.
137.

138.

139.
140.

141.
142.
143.
144.
145.
146.
147.

of the vessels of the placental bed to hypertensive pregnancy. J


Pathol Bacteriol 93:581592, 1967
MEEKINS JW, PIJNENBORG R, HANSSENS M, et al: A study of placental bed spiral arteries and trophoblast invasion in normal and
severe pre-eclamptic pregnancies. Br J Obstet Gynaecol 101:669
674, 1994
ZHOU Y, DAMSKY CH, CHIU K, et al: Preeclampsia is associated
with abnormal expression of adhesion molecules by invasive cytotrophoblasts. J Clin Invest 91:950960, 1993
ZHOU Y, FISHER SJ, JANATPOUR M, et al: Human cytotrophoblasts
adopt a vascular phenotype as they differentiate. A strategy for
successful endovascular invasion? J Clin Invest 99:21392151, 1997
ZHOU Y, DAMSKY CH, FISHER SJ: Preeclampsia is associated with
failure of human cytotrophoblasts to mimic a vascular adhesion
phenotype. One cause of defective endovascular invasion in this
syndrome? J Clin Invest 99:21522164, 1997
CANIGGIA I, GRISARU-GRAVNOSKY S, KULISZEWSKY M, et al: Inhibition of TGF-beta 3 restores the invasive capability of extravillous
trophoblasts in preeclamptic pregnancies. J Clin Invest 103:1641
1650, 1999
CANIGGIA I, MOSTACHFI H, WINTER J, et al: Hypoxia-inducible
factor-1 mediates the biological effects of oxygen on human
trophoblast differentiation through TGFbeta(3). J Clin Invest
105:577587, 2000
RAJAKUMAR A, WHITELOCK KA, WEISSFELD LA, et al: Selective
overexpression of the hypoxia-inducible transcription factor, HIF2alpha, in placentas from women with preeclampsia. Biol Reprod
64:499506, 2001
LEACH RE, ROMERO R, KIM YM, et al: Pre-eclampsia and expression of heparin-binding EGF-like growth factor. Lancet 360:1215
1219, 2002
ZHOU Y, CHIU K, BRESCIA RJ, et al: Increased depth of trophoblast
invasion after chronic constriction of the lower aorta in rhesus
monkeys. Am J Obstet Gynecol 169:224229, 1993
FERRIS TF: Pregnancy, preeclampsia, and the endothelial cell. N
Engl J Med 325:14391440, 1991
PAGE NM, WOODS RJ, GARDINER SM, et al: Excessive placental
secretion of neurokinin B during the third trimester causes preeclampsia. Nature 405:797800, 2000
SAVVIDOU MD, HINGORANI AD, TSIKAS D, et al: Endothelial dysfunction and raised plasma concentrations of asymmetric dimethylarginine in pregnant women who subsequently develop preeclampsia. Lancet 361:15111517, 2003
SMARASON AK, SARGENT IL, STARKEY PM, REDMAN CW: The effect
of placental syncytiotrophoblast microvillous membranes from
normal and pre-eclamptic women on the growth of endothelial
cells in vitro. Br J Obstet Gynaecol 100:943949, 1993
REDMAN CW, SARGENT IL: The pathogenesis of pre-eclampsia.
Gynecol Obstet Fertil 29:518522, 2001
COCKELL AP, LEARMONT JG, SMARASON AK, et al: Human placental syncytiotrophoblast microvillous membranes impair maternal
vascular endothelial function. Br J Obstet Gynaecol 104:235240,
1997
KANAYAMA N, TAKAHASHI K, MATSUURA T, et al: Deficiency in
p57Kip2 expression induces preeclampsia-like symptoms in mice.
Mol Hum Reprod 8:11291135, 2002
HUBEL CA: Oxidative stress in the pathogenesis of preeclampsia.
Proc Soc Exp Biol Med 222:222235, 1999
REGAN CL, LEVINE RJ, BAIRD DD, et al: No evidence for lipid peroxidation in severe preeclampsia. Am J Obstet Gynecol 185:572
578, 2001
ZHANG C, WILLIAMS MA, KING IB, et al: Vitamin C and the risk of
preeclampsiaresults from dietary questionnaire and plasma assay.
Epidemiology 13:409416, 2002
CHAPPELL LC, SEED PT, BRILEY AL, et al: Effect of antioxidants
on the occurrence of pre-eclampsia in women at increased risk: A
randomised trial. Lancet 354:810816, 1999
ABDALLA S, LOTHER H, EL MASSIERY A, QUITTERER U: Increased
AT(1) receptor heterodimers in preeclampsia mediate enhanced
angiotensin II responsiveness. Nat Med 7:10031009, 2001
WALLUKAT G, HOMUTH V, FISCHER T, et al: Patients with preeclampsia develop agonistic autoantibodies against the angiotensin AT1
receptor. J Clin Invest 103:945952, 1999

Karumanchi et al: Preeclampsia and angiogenic factors

148. XIA Y, WEN H, BOBST S, et al: Maternal autoantibodies from


preeclamptic patients activate angiotensin receptors on human trophoblast cells. J Soc Gynecol Investig 10:8293, 2003
149. DECHEND R, VIEDT C, MULLER DN, et al: AT1 receptor agonistic
antibodies from preeclamptic patients stimulate NADPH oxidase.
Circulation 107:16321639, 2003
150. MAYNARD SE, MIN JY, MERCHAN J, et al: Excess placental soluble
fms-like tyrosine kinase 1 (sFlt1) may contribute to endothelial
dysfunction, hypertension, and proteinuria in preeclampsia. J Clin
Invest 111:649658, 2003
151. KENDALL RL, THOMAS KA: Inhibition of vascular endothelial cell
growth factor activity by an endogenously encoded soluble receptor. Proc Natl Acad Sci USA 90:1070510709, 1993
152. CLARK DE, SMITH SK, HE Y, et al: A vascular endothelial growth
factor antagonist is produced by the human placenta and released
into the maternal circulation. Biol Reprod 59:15401548, 1998
153. ZHOU Y, MCMASTER M, WOO K, et al: Vascular endothelial growth
factor ligands and receptors that regulate human cytotrophoblast
survival are dysregulated in severe preeclampsia and hemolysis,
elevated liver enzymes, and low platelets syndrome. Am J Pathol
160:14051423, 2002
154. BANKS RE, FORBES MA, SEARLES J, et al: Evidence for the existence
of a novel pregnancy-associated soluble variant of the vascular
endothelial growth factor receptor, Flt-1. Mol Hum Reprod 4:377
386, 1998
155. KOGA K, OSUGA Y, YOSHINO O, et al: Elevated serum soluble vascular endothelial growth factor receptor 1 (sVEGFR-1) levels in
women with preeclampsia. J Clin Endocrinol Metab 88:23482351,
2003
156. TSATSARIS V, GOFFIN F, MUNAUT C, et al: Overexpression of the
soluble vascular endothelial growth factor receptor in preeclamptic patients: Pathophysiological consequences. J Clin Endocrinol
Metab 88:55555563, 2003
157. CHAIWORAPONGSA T, ROMERO R, ESPINOZA J, et al: Evidence supporting a role for blockade of the vascular endothelial growth factor system in the pathophysiology of preeclampsia. Young Investigator Award. Am J Obstet Gynecol 190:15411547, 2004
158. TAYLOR RN, GRIMWOOD J, TAYLOR RS, et al: Longitudinal serum
concentrations of placental growth factor: evidence for abnormal
placental angiogenesis in pathologic pregnancies. Am J Obstet Gynecol 188:177182, 2003
159. POLLIOTTI BM, FRY AG, SALLER DN, et al: Second-trimester maternal serum placental growth factor and vascular endothelial growth
factor for predicting severe, early-onset preeclampsia. Obstet Gynecol 101:12661274, 2003
160. LEVINE RJ, MAYNARD SE, QIAN C, et al: Circulating angiogenic
factors and the risk of preeclampsia. N Engl J Med 350:672683,
2004
161. HERTIG A, BERKANE N, LEFEVRE G, et al: Maternal serum sFlt1 concentration is an early and reliable predictive marker of preeclampsia. Clin Chem 50:17021703, 2004
162. LEVINE RJ, THADHANI R, QIAN C, et al: Urinary placental growth
factor and risk of preeclampsia. JAMA 293:7785, 2005
163. HE H, VENEMA VJ, GU X, et al: Vascular endothelial growth factor
signals endothelial cell production of nitric oxide and prostacyclin
through flk-1/KDR activation of c-Src. J Biol Chem 274:25130
25135, 1999
164. MASUDA Y, SHIMIZU A, MORI T, et al: Vascular endothelial
growth factor enhances glomerular capillary repair and accelerates

165.
166.

167.
168.

169.
170.

171.
172.

173.
174.
175.
176.
177.

178.
179.
180.
181.
182.

2113

resolution of experimentally induced glomerulonephritis. Am J


Pathol 159:599608, 2001
OSTENDORF T, KUNTER U, EITNER F, et al: VEGF(165) mediates
glomerular endothelial repair. J Clin Invest 104:913923, 1999
SUGIMOTO H, HAMANO Y, CHARYTAN D, et al: Neutralization of
circulating vascular endothelial growth factor (VEGF) by antiVEGF antibodies and soluble VEGF receptor 1 (sFlt-1) induces
proteinuria. J Biol Chem 278:1260512608, 2003
EREMINA V, SOOD M, HAIGH J, et al: Glomerular-specific alterations
of VEGF-A expression lead to distinct congenital and acquired
renal diseases. J Clin Invest 111:707716, 2003
KABBINAVAR F, HURWITZ HI, FEHRENBACHER L, et al: Phase
II, randomized trial comparing bevacizumab plus fluorouracil
(FU)/leucovorin (LV) with FU/LV alone in patients with metastatic
colorectal cancer. J Clin Oncol 21:6065, 2003
YANG JC, HAWORTH L, SHERRY RM, et al: A randomized trial of
bevacizumab, an anti-vascular endothelial growth factor antibody,
for metastatic renal cancer. N Engl J Med 349:427434, 2003
KUENEN BC, LEVI M, MEIJERS JC, et al: Analysis of coagulation cascade and endothelial cell activation during inhibition of vascular
endothelial growth factor/vascular endothelial growth factor receptor pathway in cancer patients. Arterioscler Thromb Vasc Biol
22:15001505, 2002
ESSER S, WOLBURG K, WOLBURG H, et al: Vascular endothelial
growth factor induces endothelial fenestrations in vitro. J Cell Biol
140:947959, 1998
NAGAMATSU T, FUJII T, KUSUMI M, et al: Cytotrophoblasts
up-regulate soluble fms-like tyrosine kinase-1 expression under reduced oxygen: an implication for the placental vascular
development and the pathophysiology of preeclampsia. Endocrinology 145:48384845, 2004
BOYD PA, LINDENBAUM RH, REDMAN C: Pre-eclampsia and trisomy
13: A possible association. Lancet 2:425427, 1987
LAIVUORI H, TIKKANEN MJ, YLIKORKALA O: Hyperinsulinemia 17
years after preeclamptic first pregnancy. J Clin Endocrinol Metab
81:29082911, 1996
HUBEL CA, SNAEDAL S, NESS RB, et al: Dyslipoproteinaemia in
postmenopausal women with a history of eclampsia. Br J Obstet
Gynaecol 107:776784, 2000
IRGENS HU, REISAETER L, IRGENS LM, LIE RT: Long term mortality of mothers and fathers after pre-eclampsia: population based
cohort study. Br Med J 323:12131217, 2001
SIBAI BM, EL-NAZER A, GONZALEZ-RUIZ A: Severe preeclampsiaeclampsia in young primigravid women: Subsequent pregnancy
outcome and remote prognosis. Am J Obstet Gynecol 155:1011
1016, 1986
EPSTEIN FH: Late vascular effects of toxemia of pregnancy. New
Engl J Med 271:391395, 1964
JOHNSON RJ, HERRERA-ACOSTA J, SCHREINER GF, RODRIGUEZITURBE B: Subtle acquired renal injury as a mechanism of saltsensitive hypertension. N Engl J Med 346:913923, 2002
VATTEN LJ, ROMUNDSTAD PR, TRICHOPOULOS D, SKJAERVEN R: Preeclampsia in pregnancy and subsequent risk for breast cancer. Br
J Cancer 87:971973, 2002
MOGREN I, STENLUND H, HOGBERG U: Long-term impact of reproductive factors on the risk of cervical, endometrial, ovarian and
breast cancer. Acta Oncol 40:849854, 2001
CHIEN KR: Meeting Kochs postulates for calcium signaling in cardiac hypertrophy. J Clin Invest 105:13391342, 2000

You might also like