You are on page 1of 15

REVIEWS

Pre-eclampsia part 1: current understanding


of its pathophysiology
Tinnakorn Chaiworapongsa, Piya Chaemsaithong, Lami Yeo and Roberto Romero
Abstract | Pre-eclampsia is characterized by new-onset hypertension and proteinuria at 20weeks of
gestation. In the absence of proteinuria, hypertension together with evidence of systemic disease (such as
thrombocytopenia or elevated levels of liver transaminases) is required for diagnosis. This multisystemic
disorder targets several organs, including the kidneys, liver and brain, and is a leading cause of maternal
and perinatal morbidity and mortality. Glomeruloendotheliosis is considered to be a characteristic lesion
of pre-eclampsia, but can also occur in healthy pregnant women. The placenta has an essential role in
development of this disorder. Pathogenetic mechanisms implicated in pre-eclampsia include defective deep
placentation, oxidative and endoplasmic reticulum stress, autoantibodies to type1 angiotensin II receptor,
platelet and thrombin activation, intravascular inflammation, endothelial dysfunction and the presence of an
antiangiogenic state, among which an imbalance of angiogenesis has emerged as one of the most important
factors. However, this imbalance is not specific to pre-eclampsia, as it also occurs in intrauterine growth
restriction, fetal death, spontaneous preterm labour and maternal floor infarction (massive perivillous fibrin
deposition). The severity and timing of the angiogenic imbalance, together with maternal susceptibility, might
determinethe clinical presentation of pre-eclampsia. This Review discusses the diagnosis, classification,
clinical manifestations andputative pathogenetic mechanisms of pre-eclampsia.

Chaiworapongsa, T. etal. Nat. Rev. Nephrol. advance online publication 8 July 2014; doi:10.1038/nrneph.2014.102

Introduction
Pre-eclampsia is characterized by new-onset hyper toxinshence the name toxaemia of pregnancy) that,
tension and proteinuria at 20weeks of gestation. 16 when released into the maternal circulation, are respon-
Inthe absence of proteinuria, diagnosis requires the pres- sible for the clinical manifestations of the disease. 13,14
ence of hypertension together with evidence of systemic These soluble factors are thought to cause endothelial
disease (such as thrombocytopenia, elevated levels of liver cell dysfunction,15,16 intravascular inflammation1719 and
transaminases, renal insufficiency, pulmonary oedema activation of the haemostatic system;20 accordingly, pre-
and visual or cerebral disturbances).7,8 This gestation- eclampsia is considered to be primarily a vascular dis
specific syndrome affects 35% of all pregnancies, and order. The clinical manifestations of pre-eclampsia result
is a leading cause of maternal and perinatal morbidity from the involvement of multiple organs, including the
and mortality.17,9 Pre-eclampsia can progress to eclamp- kidneys, liver, brain, heart, lung, pancreas and the vas-
sia, which is characterized by new-onset grand mal seiz culature.2,21 This Review discusses the diagnosis, classifi-
ures and affects 2.78.2 women per 10,000 deliveries.10 cation, clinical manifestations and putative pathogenetic
Complications of pre-eclampsia or eclampsia include mechanisms of pre-eclampsia. Its prediction, preven-
cerebrovascular accidents, liver rupture, pulmonary tion and management will be addressed in part 2 of this
oedema or acute renal failure that can result in maternal Review as a separate article.22
Perinatology Research death.11 Adverse perinatal outcomes of pre-eclampsia and
Branch, Program for eclampsia are attributed largely to preterm delivery, which A historical overview
Perinatal Research and
Obstetrics, Division of
occurs secondary to maternal or fetal complications, Eclampsia was first recognized as a convulsive disorder
Intramural Research, intrauterine growth restriction (IUGR) and fetaldeath.9 of pregnancy; the name is derived from the Greek word
Eunice Kennedy Shriver A placenta, but not the fetus, is required for the devel- eklampsis (meaning lightning), reflecting the sudden
National Institute of
Child Health and opment of pre-eclampsia, as pre-eclampsia can occur onset of convulsions in pregnant women.23 Albuminuria
Human Development, in patients with hydatidiform moles.12 Indeed, the only was reported in patients with eclampsia in 1840,24 and
NIH, 31 Center Drive,
Bethesda, MD 20892,
effective treatment for pre-eclampsia is delivery of approximately 50years later, the presence of hyper
USA and 3990 John R the placenta. The traditional view of the pathogenetic tension was also recognized in such patients.25 The term
Street, Detroit, mechanisms involved in pre-eclampsia is that an ischae pre-eclampsia was subsequently introduced to describe
MI48201, USA (T.C.,
P.C., L.Y., R.R.). mic placenta produces soluble factors (formerly called the state preceding eclampsia.1 In the early 20th century,
differentiating between glomerulonephritis (then known
Correspondence to: T.C.
tchaiwor@ Competing interests as Bright disease) and pre-eclampsia in pregnant women
med.wayne.edu The authors declare no competing interests. was challenging, as both conditions are associated with

NATURE REVIEWS | NEPHROLOGY ADVANCE ONLINE PUBLICATION | 1


2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

Key points estimates of the heritability of pre-eclampsia range from


22% to 47%. 30 Candidate-gene studies have demon-
Diagnosis of pre-eclampsia is based on new-onset hypertension and proteinuria
strated significant associations between DNA variants
at 20weeks of gestation or, in the absence of proteinuria, hypertension
together with evidence of systemic disease and pre-eclampsia for a few genes in the maternal and
Genetic and environmental factors are thought to create conditions leading to fetal genome, including collagen1(I) chain (COL1A1),
defective deep placentation; the injured placenta then releases factors into the IL-1 (IL1A) for the maternal genotype and urokinase
maternal circulation that induce pre-eclampsia plasminogen activator surface receptor (PLAUR) for
Pre-eclampsia is characterized by multiple aetiologies and pathogenetic the fetal genotype.31 Moreover, maternalfetal genotype
mechanisms, a long subclinical phase, fetal involvement, adaptive clinical incompatibility of lymphotoxin (LTA), von Willebrand
manifestations and geneenvironment interactions
factor (VWF) and collagen 2(IV) chain (COL4A2) seem
An imbalance between angiogenic and antiangiogenic factors has emerged as
acentral pathogenetic mechanism in pre-eclampsia
to be a risk factor for pre-eclampsia.32 Other specific DNA
An antiangiogenic state can also be observed in conditions other than pre- variants that increase the risk of pre-eclampsia include
eclampsia, including intrauterine growth restriction, fetal death, spontaneous the Factor V Leiden mutation, mutations in endothelial
preterm labour and maternal floor infarction nitric oxide synthase, human leukocyte antigen and
The severity and timing of the antiangiogenic state, as well as maternal angiotensin-converting enzyme.33 A meta-analysis of 11
susceptibility, might determine the clinical presentation of pre-eclampsia studies, involving 1,297 cases of pre-eclampsia and 1,791
controls, found a modest but significant association
between pre-eclampsia and the single nucleotide poly-
Box 1 | Risk factors for pre-eclampsia192 morphism (SNP) rs1799889 in SERPINE1. Specifically,
Nulliparous women this association involved the 675 4G/5G polymorphism
Extreme maternal age (<20years193 or >35years194) (4G versus 5G) and, for the recessive genetic model
History of pre-eclampsia in previous pregnancy (4G/4G versus 4G/5G+5G/5G), the odds ratio was 1.36
Multi-fetal gestation
(95%CI 1.131.64, P=0.001).34 By contrast, a genome-
Obesity195,196
Family history of pre-eclampsia (mother or sister)
wide association study of 177 cases of pre-eclampsia
Pre-existing medical conditions, including chronic and 166 controls did not identify any variants associated
hypertension, diabetes mellitus, antiphospholipid with pre-eclampsia;35 however, associations with four
syndrome,197,198 thrombophilia, autoimmune disease, SNPs in PSG11 (which encodes pregnancy-specific
renal disease, infertility 1-glycoprotein 11) reached nominal significance.35
Limited sperm exposure
Dangerous father28,199 Diagnosis of pre-eclampsia
Urinary tract infection191,200
Typical presentation
Pre-eclampsia is traditionally diagnosed by new-onset
hypertension and proteinuria.21 The prevention of eclamp- hypertension and proteinuria at 20weeks of gesta-
sia was proposed as a major goal of prenatal care in 1901, tion. Although hypertension is essential according to
which led directly to the current emphasis on detecting all diagnostic criteria, the requirement of proteinuria
early signs of pre-eclampsia.26 As a consequence, pregnant for the diagnosis of pre-eclampsia is a matter of debate.
women now have their blood pressure measured and urine Some professional organizations allow diagnosis of pre-
tested for proteinuria at every antenatal visit. eclampsia on the basis of the presence of new-onset
hypertension together with evidence of systemic involve-
Risk factors ment, such as thrombocytopenia, elevated levels of liver
Pre-eclampsia often occurs in young women having their transaminases, renal insufficiency, pulmonary oedema
first pregnancy. This observation has been attributed to and visual or cerebral disturbances (Box2). 7,8 The
an immune mechanism, as the maternal immune system rationale for omitting proteinuria is that pre-eclampsia
develops tolerance to paternal alloantigens follow- might manifest before glomerular capillary endotheliosis
ingexposure to seminal fluid and/or sperm.27 Prolonged becomes severe enough to induce proteinuria. 36,37
exposure to semen is thought to decrease the risk of Moreover, patients with nonproteinuric pre-eclampsia
developing pre-eclampsia (Box1),27 possibly explain- (with evidence of systemic involvement) are more likely
ing the increased risk of this condition in women with a than those with gestational hypertension to have severe
short interval between first coitus and conception, those hypertension and undergo premature delivery. However,
undergoing assisted reproductive technologies involving women with proteinuric pre-eclampsia are at greater risk
artificial insemination, women using barrier methods than women with nonproteinuric pre-eclampsia to have
of contraception and in multiparous women who have severe hypertension, and deliver premature babies or
changed partner since the previous pregnancy.27 A pater- babies who are small for their gestational age.38
nal component to the risk of pre-eclampsia has also been The gold standard for diagnosis of proteinuria during
proposed, known as the dangerous father hypothesis, pregnancy is total protein 300mg in a 24h urine
according to which men who have fathered a previ- sample. However, this cut-off has not been adequately
ous pregnancy complicated by pre-eclampsia have an validated and might be too high;39 a subsequent study
increased risk of doing so again with a new partner.28 demonstrated that the mean total protein concentration
Familial clustering supports a genetic component to in 24h urine samples from normal pregnant women was
the risk of developing pre-eclampsia.29 In twin studies, 117.0mg (upper limit of 95% CI 259.4mg).39 Urinary

2 | ADVANCE ONLINE PUBLICATION www.nature.com/nrneph


2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

Box 2 | Classification of hypertensive disorders of pregnancy Atypical presentations


Hypertension and proteinuria have been recognized
Chronic hypertension
Blood pressure 140/90mmHg before pregnancy and at <20weeks of
as the essential criteria for diagnosis of pre-eclampsia.
gestation, or diagnosed for the first time during pregnancy and does not However, this multisystemic disease can also present
resolvepostpartum with atypical manifestations, giving rise to its nickname
Pre-eclampsia and eclampsia the great imitator.46 Some patients present with HELLP
Blood pressure 140/90mmHg on two occasions at least 4h apart or syndrome (haemolysis, elevated liver transaminases, low
160/110mmHg within a shorter interval (minutes), at 20weeks of platelets),4749 which is widely regarded as a form of pre-
gestation, in women with previously normal blood pressure and proteinuria* eclampsia, despite it failing to meet conventional diag-
In the absence of proteinuria, new-onset hypertension plus new onset of any of nostic criteria. HELLP syndrome occurs only in pregnant
the following features: serum creatinine concentrations >97mol/l or doubling women, resolves with delivery and is frequently, but not
of serum creatinine concentration in the absence of other renal disease;
always, associated with hypertension and proteinuria.50
elevation of liver transaminases to twice normal concentration; pulmonary
oedema; and cerebral or visual symptoms
Patients can also present with right upper quadrant pain
Eclampsia: seizures in women with pre-eclampsia that cannot be attributed to (attributable to distention of the capsule of Glisson), and
other causes some components of HELLP syndrome, but without
Pre-eclampsia superimposed on chronic hypertension hypertension and proteinuria.50
Women with hypertension (at <20weeks gestation) and new-onset proteinuria*
In women with hypertension and proteinuria* (at <20weeks gestation), Classification
development of any of the following features: sudden increase in proteinuria;* Pre-eclampsia can be classified as early (<34weeks) or
sudden increase in blood pressure in women whose hypertension was late (34weeks), according to gestational age at diagno-
previously well controlled; thrombocytopenia (platelet count <100,000per mm3); sis or delivery (Box3).3,51 Although several other gesta-
and elevated liver transaminase levels
tional age cut-offs have been suggested (such as 32weeks
Gestational hypertension and 36weeks),52 34weeks remains the most commonly
New-onset blood pressure 140/90mmHg detected at 20weeks gestation
used,53,54 presumably as the rate of neonatal morbidities
without proteinuria
Pre-eclampsia does not develop and blood pressure returns to normal by
declines considerably after reaching this gestational time
12weeks postpartum point. For instance, in women with severe pre-eclampsia,
*Defined as urinary protein excretion 300mg/24h, a total protein:creatinine ratio 30mg/ expectant management is no longer considered and
mmol (or 0.3 when both are measured in mg/dl) or a dipstick reading of 1+ (only if other induction of labour is recommended after the 34th week
quantitative methods are not available).7
of gestation. 7 However, whether early and late pre-
eclampsia have different pathogenetic mechanisms or
dipstick tests for proteinuria are considered unreliable are merely gradations of the same underlying condition
owing to variations in protein excretion, activity, diet and remains unclear.55 Pre-eclampsia can also be classified
posture at the time of assessment.39 However, a dipstick according to its severity (Box4);1,56 nonetheless, to avoid
result of 1+ has a positive-predictive value of 8292% conveying a false sense of security to clinicians, some
for predicting 24h urinary protein levels of 300mg.35,36 professional organizations have abandoned the term
By contrast, the negative predictive value of a nega- mild or severe pre-eclampsia in favour of pre-eclampsia
tive or trace dipstick result was low (3460%).40,41 The with or without severe features.7
assessment of proteinuria by protein:creatinine ratios
is recommended in several guidelines, with the most Pathogenetic mechanisms
commonly used cut-off value being 30mg/mmol. 7 Placental ischaemia
However, protein:creatinine ratios can vary depending The central hypothesis governing our understanding of
on the time of day the measurement is taken42 and this pre-eclampsia is that the disorder results from ischae
parameter might, therefore, be unreliable for the diagno- mia of the placenta, which in turn releases factors into
sis of proteinuria.39 Indeed, a recent study examined the the maternal circulation that are capable of inducing
urinary protein:creatinine ratio at three different time the clinical manifestations of the disease. This concept
points (at 0800h, 1200h and 1700h) and showed that emerged from observations that placental infarctions are
the ratios varied throughout the day (mean coefficient common in patients with eclampsia.13 In 1914, research-
of variation 36%). However, no differences among the ers proposed that placental infarctions are due to inter-
three measurements with respect to sensitivity (8996%) ference with the maternal blood supply to the placenta,
and specificity (7578%) to predict proteinuria were and that a necrotic placenta releases products directly
found.43 A systematic review concluded that the spot into the intervillous space and maternal circulation.13
protein:creatinine ratio is a reasonable rule out test Importantly, this hypothesis was supported by studies
for significant proteinuria during pregnancy because in which the injection of placental extracts into guinea
of its high sensitivity and low negative likelihood ratio pigs elicited convulsions with hepatic and renal lesions,
(0.12).44 Of note, although the presence of proteinuria similar to those observed in women who died of eclamp-
can aid in the diagnosis of pre-eclampsia, the severity sia. 13 In 1940, studies in pregnant dogs showed that
of proteinuria has limited prognostic value45 and, con- clamping of the abdominal aorta (which reduced utero-
sequently, severe proteinuria has been removed from placental perfusion by 50%) led to maternal hypertension
the diagnostic criteria for severe pre-eclampsia by some that resolved after release of the clamp (Figure1). This
professionalorganizations.7 hypertensive response was not observed in nonpregnant

NATURE REVIEWS | NEPHROLOGY ADVANCE ONLINE PUBLICATION | 3


2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

Box 3 | Classification of pre-eclampsia reaction against fetal tissue,63,64 shear flow stress caused
by abnormal blood flow in nontransformed spiral arter-
Early pre-eclampsia (<34weeks*)
Uncommon (prevalence 0.38% or 12% of all pre-
ies, a systemic inflammatory response that involves the
eclampsia)201 uterine decidua and maternal genetic predisposition.65
Associated with extensive villous and vascular lesions Atherosis might further impede blood flow to the pla-
of the placenta centa during the third trimester. However, the reader
Higher risk of maternal and fetal complications than should note that failure of physiological transformation
late pre-eclampsia of the spiral arteries is neither specific to pre-eclampsia
Late pre-eclampsia (34weeks*) nor sufficient to cause it,66 as such failure has also been
Majority of all cases of pre-eclampsia (prevalence observed in other obstetric syndromes, including spon-
2.72% or 88% of all pre-eclampsia)201 taneous abortion,67,68 IUGR,69,70 fetal death,68 placental
Minimal placental lesions
abruption,71 preterm labour 72 and preterm premature
Maternal factors (such as metabolic syndrome and
hypertension) have important roles
rupture of membranes.73 The mechanisms responsible
Most cases of eclampsia and maternal death occur for failure of physiological transformation of the spiral
inlate disease arteries have not been fullyelucidated.
*Gestational age at diagnosis or delivery.3,44 Human pregnancy is characterized by deep placen-
tation, in which trophoblast cells in the placental bed
invade not only the decidua, but also one-third of the
dogs.57 A uteroplacental origin of the signal driving this thickness of the myometrium.59,66,74 As invasive tropho
hypertensive response was supported by the observation blasts are responsible for transformation of the spiral
that, after removal of the pregnant uterus, clamping of arteries, some researchers have suggested that an abnor-
the aorta no longer elicited hypertension.57 Subsequent mality in trophoblasts might result in shallow placen
work revealed that uteroplacental blood flow is decreased tation and inadequate transformation of the spiral
in women with pre-eclampsia.58 arteries, leading to pre-eclampsia.75
Other than trophoblast abnormalities, the effects of
Transformation of the spiral arteries shear stress on vascular endothelium can affect remodel
During a normal pregnancy, uterine blood flow increases ling of the spiral arteries. The diameter of the uterine
to enable perfusion of the intervillous space of the pla- artery (especially the proximal portion of vessels that
centa and to support fetal growth. The increased blood are upstream of the branches into the spiral arteries)
flow is achieved by physiological transformation of the increases prior to completion of placentation, possibly as
spiral arteries of the uterus, a process in which tropho- a consequence of the vasodilatatory properties of elevated
blasts invade the arterial wall, destroy the media and levels of oestrogen.76 Blood flow in the uterine arteries
transform the spiral arteries from narrow-diameter also changes during the first few weeks of pregnancy and
to large-diameter vessels, thereby enabling adequate subsequently, haemochorial placentation converts the
perfusion of the placenta (Figures2 and 3a).59 spiral arteries into a low-velocity, high-flow chamber.59
In pre-eclampsia (and eclampsia), the myometrial Decreased downstream resistance accelerates blood flow
segment of the spiral artery fails to undergo physio velocity in afferent (radial and arcuate) arteries, increas-
logical transformation during the second trimester ing shear stress on the arterial wall. The increase in shear
(Figures2 and 3b),60 which is thought to explain the stress stimulates the endothelium to augment its produc-
uteroplacental ischaemia observed in pre-eclampsia. tion of nitric oxide, resulting in vasodilatation, which
Moreover, nontransformed spiral arteries are prone to further lowers uterine vascular resistance, and in turn,
atherosis, characterized by the presence of lipid-laden normalizes shear stress on the arterial wall. The increase
macrophages within the lumen, fibrinoid necrosis of in blood flow resulting from a large lumen (but decreased
the arterial wall and a mononuclear perivascular infil- flow velocity) might stimulate changes in both vascular
trate.61,62 The atherotic lesions resemble atherosclerotic smooth muscle and the extracellular matrix and, in turn,
plaques and are proposed to result from several patho- vascular remodelling.77
physiological mechanisms that include immunological
Hypoxia and trophoblast invasion
Upon arrival of the conceptus to the endometrium, nutri-
Box 4 | Severe features of pre-eclampsia (one or more of these findings)
tion is initially provided by secretions from the endo
Systolic blood pressure 160mmHg, or diastolic blood pressure 110mmHg metrial glands (histiotrophic nutrition).78 Subsequently,
on two occasions at 4h apart while the patient is onbed rest the blastocyst invades the decidua. In the early phases
Platelet count <100,000 per mm3
of implantation, the gestational sac exists in an environ-
Elevated liver enzymes (twice normal concentrations)
Renal insufficiency (serum creatinine concentration >1.1mg/dl or a doubling
ment with low oxygen tension, which favours tropho-
ofserum creatinine concentration) or oliguria (<500ml in 24h) blast proliferation. Trophoblasts anchor the blastocyst to
Pulmonary oedema or cyanosis maternal tissues, and also plug the tips of the spiral arter-
New-onset cerebral or visual disturbances ies within the decidua.79 Eventually, lacunae are formed
Severe persistent right upper quadrant or epigastric pain within the trophoblasts, which subsequently coalesce to
Adapted from the Am. J. Obstet. Gynecol. 205, Sibai, B.M. Evaluation and management of severe create the intervillous space. Opening of the spiral arter-
preeclampsia before 34 weeks gestation, 191198 (2011), with permission fromElsevier.
ies into the intervillous space enables the development of

4 | ADVANCE ONLINE PUBLICATION www.nature.com/nrneph


2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

a Hypertension b No hypertension c Hypertension d No hypertension


Renal artery

Left
kidney

Clamp
Aorta

Blood supply Hysterectomy


to uterus

Pregnant
Uterus uterus

Figure 1 | An experiment supporting the concept that hypertension in pregnancy represents a uteroplacental response to
ischaemia. a | In the Goldblatt model of renovascular hypertension, clamping the renal artery leads to development of
hypertension through renal ischaemia in nonpregnant animals. b | By contrast, clamping the aorta below the renal arteries
does not induce hypertension in nonpregnant animals. c | Aortic clamping in pregnant animals leads to hypertension.
d|After hysterectomy, however, hypertension can no longer be elicited by aortic clamping, suggesting that the ischaemic
pregnant uterus is the source of signals that lead to maternal systemic hypertension. Permission obtained from Semin.
Perinatol. 12, Romero, R. etal. Toxemia: new concepts in an old disease, 302323 Elsevier (1988).

haemochorial placentation, and shifts nutrition from a evidence for the presence of hypoxia in the intervillous
histiotrophic to haematotrophic type.80 space in patients destined to develop pre-eclampsia is
The initial burst of blood into the intervillous space lacking because these measurements cannot be per-
increases oxygen tension, generating oxidative stress formed in ongoing pregnancies. Moreover, expression
on the trophoblasts, which promotes trophoblast dif- of HIF1 is upregulated not only by hypoxia, but also
ferentiation from a proliferative to an invasive pheno- by inflammatory stimuli (for example, thrombin, vaso
type. Differentiated trophoblasts invade deeper into the active peptides, cytokines, such as tumour necrosis factor
decidua, reaching into the superficial myometrium80 and [TNF], and reactive oxygen species [ROS]), especially
facilitating physiological transformation of the spiral arter- those mediated by nuclear factor B (NF-B), as the
ies. Thus, the initial phase of placentation occurs under promoter of HIF1 contains an NFB binding site.88
conditions of relative hypoxia.81 Indeed, hypoxia-inducible Inadequate deep placentation can also result from a
factor (HIF)-1, a marker of cellular oxygen deprivation, decidual defect; optimal decidualization requires proper
is expressed at high levels in trophoblasts. Persistent preconditioning, which is achieved by successive men-
hypoxia or failure to downregulate transforming growth struations.89 Inadequate preconditioning might explain
factor3 (TGF3) expression after 9weeks of gestation the high rate of pre-eclampsia in young women (such as
might result in failure of trophoblasts to differentiate from those aged <20years).89 Another possibility is that defec-
the proliferative to invasive phenotype,82 shallow tropho- tive placentation might be the result of combinations
blast invasion and inadequate transformation of the spiral of factors that affect both decidua and trophoblasts.89
arteries,82 although this hypothesis remains unproven. Indeed, implantation creates conditions in which fetal
Considerable evidence supports a role for hypoxia in cells (carrying paternal antigens) and maternal cells
creating an environment that predisposes to implantation come into contact in the decidua; a role for the immune
disorders, including pre-eclampsia: expression of HIF1 system in normal placentation is, therefore, easy to envi-
and HIF2 protein is increased in the placentas of sion. Successful pregnancy requires that the maternal
women with pre-eclampsia;83 overexpression of HIF1 immune system does not reject the trophoblast.27 Roles
leads to hypertension, proteinuria and fetal growth for natural killer cells in the decidua, HLAC molecules
restriction in mice;84 mice with deletion of the Comt on the fetal trophoblast (its only known polymorphic
gene, which encodes catechol Omethyltransferase (an histocompatibility antigen) and regulatory Tcells have
enzyme that catabolizes estradiol to 2methoxyestradiol), been implicated in the tolerogenic state associated with
develop hypertension and proteinuria when pregnant;85 normal pregnancy, as well as in pre-eclampsia.27
and 2methoxyestradiol, an inhibitor of HIF1, can Maternalfetal immune recognition at the site of pla-
suppress the production of soluble vascular endothelial centation is highly individualized by two polymorphic
growth factor receptor 1 (sVEGFR1), a potent anti gene systems: HLAC molecules of trophoblasts and
angiogenic factor.85 This observation is consistent with their cognate receptors, killer cell immunoglobulin-
the finding that women with pre-eclampsia had lower like receptors (KIRs) of natural killer cells.27 At least
placental COMT protein expression85 and serum levels two haplotype groups of KIR (A and B) and two types
of 2methoxyestradiol85 than healthy controls; however, of HLAC (C1 and C2) are known. HLAC2 interacts
these findings could not be replicated in subsequent with KIRs more strongly than does HLAC1.27,90 Uterine
studies.86,87 An important point to note is that direct natural killer cells release chemokines, angiogenic factors

NATURE REVIEWS | NEPHROLOGY ADVANCE ONLINE PUBLICATION | 5


2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

Normal pregnancy Pre-eclampsia pre-eclampsia. By contrast, KIR AA mothers carrying


HLAC2 fetuses might have an increased s usceptibility
Maternal
blood to pre-eclampsia.27,90

Blood Endoplasmic reticulum stress


flow Narrow spiral arteries create conditions for ischaemia
Decidua
reperfusion injury in the intervillous space. This injury,
in turn, might lead to endoplasmic reticulum (ER) stress,
which regulates cell homeostasis through its involvement
Trophoblast in post-translational modifications and protein folding.91
Decidua
During states of energy crisis (such as hypoxia), the ER
myometrium suspends protein folding (referred to as the unfolded
boundary
protein response, or UPR).92 The UPR can lead to ces-
sation of cell proliferation and, when severe, apoptosis.
Trophoblast apoptosis93 results in the release of micro
particles and nanoparticles into the maternal circula-
tion, which can stimulate an intravascular inflammatory
Myometrium response.94 Evidence supporting the involvement of ER
stress in pre-eclampsia and IUGR includes activation of
Spiral artery
the UPR, as shown by upregulation of factors involved
in several signalling pathways characteristic of UPR
activation following ER stress in the placenta.92,95

Figure 2 | Failure of physiological transformation of the spiral arteries is implicated Oxidative stress
in pre-eclampsia. a | In a normal pregnancy, physiological transformation of the Oxidative stress arises when the production of ROS
myometrial segment of the spiral artery occurs. Trophoblast cells extend to both overwhelms the intrinsic antioxidant defence mecha-
thedecidual segment and one-third of the myometrial segment of the spiral artery.
nisms operating in tissues.96 Oxidative stress is relevant
Both the arterial media and endothelium are destroyed by trophoblasts, converting
the arteries into wide-calibre vessels and increasing the delivery of blood to the
to the pathophysiology of pre-eclampsia, as it induces the
intervillous space. b | In pregnancies affected by pre-eclampsia, a key feature release of proinflammatory cytokines and chemokines as
associated with the failure of physiological transformation of the spiral arteries is well as trophoblast debris.97 The cause of oxidative stress
lack of invasion of the trophoblasts into the myometrial segment of the spiral artery. in the placentas of women with pre-eclampsia is thought
The resulting lack of transformation of blood vessels results in narrow spiral to be intermittent hypoxia and reoxygenation, probably
arteries, a disturbed pattern of blood flow and reduced uteroplacental perfusion. resulting from deficient conversion of the myometrial
Permission obtained from Nature Publishing Group Moffett-King, A. etal. Nat. Rev. segment of the spiral arteries, which contains a contrac-
Immunol. 2, 656663 (2002).
tile portion of the artery.96 Indeed, radiographic studies
of the spiral arteries note these constricted portions at
a b the myometrial segment of the spiral artery just proxi-
mal to the myometrialendometrial junction in both the
human and the rhesus monkey.98,99
Increased ROS exposure can lead to protein carboxy
lation, lipid peroxidation and DNA oxidationall of
which have been observed in placentas of patients with
pre-eclampsia.96 Under pathological conditions, such as
hypoxia or ischaemiareperfusion injury, increased con-
version of xanthine dehydrogenase to xanthine oxidase
Figure 3 | Transformed and nontransformed spiral arteries in the myometrium. promotes the production of uric acid and superoxide
a|Transformed spiral arteries are characterized by the presence of intramural from degraded purines (such as xanthine and hypo
trophoblasts (arrowheads) and fibrinoid degeneration (arrows) of the arterial wall. xanthine).96 Xanthine oxidase expression and activity
b | Nontransformed spiral arteries lack intramural trophoblasts and fibrinoid are increased in invasive trophoblasts obtained from
degeneration, and retain intact arterial contours. Arrowheads indicate the
patients with pre-eclampsia.100 Furthermore, placental
presence of trophoblasts in myometrium, but not in the wall of the spiral artery.
Both images stained with cytokeratin 7 (brown) and periodic acidSchiff (pink), antioxidant mechanisms are impaired in patients with
magnification 200. Permission obtained from the NIH Espinoza, J. etal. pre-eclampsia, as shown by their decreased expression
J.Perinat. Med. 34, 447458 (2006). of superoxide dismutase and glutathione peroxidase
compared with women with normal pregnancies.101
and cytokines that promote trophoblast invasion. These Another important source of free radicals in humans
secretions are increased upon binding of HLAC anti- is free haeme, a pro-oxidant molecule produced daily via
gens to stimulatory KIRs (haplotype B), whereas they are the degradation of circulating red blood cells. Protection
reduced by antigen binding to haplotype A of KIRs. Thus, from free haeme is afforded by the actions of haeme oxy-
KIR BB mothers carrying HLAC1 fetuses might have genase, which converts free haeme first to biliverdin and
the best chance of adequate placentation and avoiding subsequently to bilirubin, releasing free iron and carbon

6 | ADVANCE ONLINE PUBLICATION www.nature.com/nrneph


2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

monoxide.102 Biliverdin and bilirubin have potent anti- stimulate the synthesis of NADPH oxidase, a rate-limiting
oxidant properties, whereas carbon monoxide induces enzyme in the synthesis of ROS, leading to oxidative
vasodilatation, has antiapoptotic properties and promotes stress.115 These autoantibodies can also stimulate tissue
angiogenesis.102 Three isoforms of haeme oxygenase have factor release by monocytes and vascular smooth muscle
been characterized: HO1 (inducible), HO2 (constitu- cells, as well as plasminogen activator inhibitor-1 (PAI1)
tive) and HO3 (unknown function).102 Several lines release by mesangial and trophoblast cells.115 Collectively,
of evidence support a role for haeme oxygenases in the these actions can lead to increased thrombin generation,
pathogenesis of pre-eclampsia. Pregnant Hmox1+/ mice, impaired fibrinolysis and consequently fibrin deposi-
which have a partial deficiency in HO1, demonstrate tion. Anti-AT1-autoabtibodies can also stimulate placen-
hypertension, small placentas and elevated plasma levels tal production of sVEGFR1 and, therefore, lead to an
of sVEGFR1 (all features of pre-eclampsia).103 HO1 and antiangiogenic state.114
HO2 mRNA104 and protein105 expression are decreased in Of interest, reduced uterine perfusion pressure in
women with pre-eclampsia (the decreased protein expres- rats induces production of anti-AT1-autoantibodies,
sion was detected in peri-infarct regions of the placenta).105 sVEGFR1, TNF and endothelin1, as well as hyper-
In trophoblasts obtained from women at 11weeks gesta- tension and proteinuria. 116 Similarly, administration
tion, HO1 mRNA expression was lower in individuals of TNF,116 IL6116 or IL17117 to pregnant rats results in
who went on to develop pre-eclampsia than in those who hypertension, placental oxidative stress and enhanced
had a normal pregnancy.106 mRNA expression of super- AT1 activity, the effects of which can be abrogated
oxide dismutase and catalase are also lower in peripheral byAT1 blockade. Furthermore, anti-AT1 autoantibodies
blood samples from patients with pre-eclampsia than in stimulate deposition of complement C3 in the pla-
samples from normal pregnant women.104 These findings centa and kidneys of pregnant mice, whereas treatment
indicate that pre-eclampsia is associated with a deficiency with a complement C3a receptor antagonist inhibited
of antioxidant enzymes in the placenta and probably autoantibody-induced elevation of sVEGFR1, small
inthe peripheral blood as well.104 Interest in manipulating placenta formation and IUGR.118 These findings suggest
theexpression of HO1 as a potential therapeutic inter- that anti-AT1 autoantibodies mediate hypertension
vention for pre-eclampsia has been fuelled by preclinical during pregnancy through activation of complement C3
studies; pharmacological induction of HO1 expression and production of antiangiogenic factors.118 However, it
(by cobalt protoporphyrin or pravastatin) can attenuate is noteworthy that glomerular capillary endotheliosis, a
hypertension, decrease oxidative stress and reduce serum characteristic renal lesion of pre-eclampsia, has not been
concentrations of sVEGFR1 in rodent models of pre- demonstrated in the reduced uterine perfusion pressure
eclampsia (generated by either reduced uterine perfusion rat model of pre-eclampsia.116
pressure107 or overexpression of sVEGFR1 resulting from A subset of Bcells (CD19+CD5+) might be involved in
adenovirus-mediated sFlt1 gene transfer).108 production of anti-AT1 autoantibodies in pre-eclampsia,
as addition of sera from patients with pre-eclampsia to
Antibodies to type1 angiotensin II receptor these cells increased both the proportion of CD19+CD5+
Normal pregnancy is characterized by reduced vascular cells and anti-AT1 autoantibody activity.119 Moreover, the
responsiveness to angiotensin II.109 However, pregnant proportion of CD19+CD5+ Bcells was also enriched in
women with pre-eclampsia have increased sensitivity peripheral blood from patients with pre-eclampsia versus
to the effects of angiotensin II, a difference that can be that from women with normal pregnancies. By contrast,
detected as early as 24weeks of gestation.109 The mecha- administration of rituximab (an antibody directed against
nisms responsible for the physiological refractoriness to CD20, a co-stimulatory molecule on Bcells) to rats with
angiotensin II in normal pregnancy and the enhanced reduced uterine perfusion pressure reduced anti-AT1
response to it in pre-eclampsia include genetic predispo- autoantibody titres, blood pressure and endothelin1
sition, maladaptive immune responses and environmental levels, as well as depleting Bcells. Adoptive transfer of
triggers.110 A subset of women with pre-eclampsia have CD4+ Tlymphocytes from rats with reduced uterine per-
detectable serum autoantibodies against type1 angio fusion pressure to normal pregnant rats increased blood
tensin II receptor (AT1), 111 and such autoantib odies pressure and anti-AT1 autoantibody activity.119 These
can activate AT1 in endothelial cells, vascular smooth effects were abrogated by administration of losartan or
muscle cells and mesangial cells. In pregnant rats, anti- by Bcell depletion using rituximab.120
AT1 autoantibodies (either produced endogenously Some investigators have proposed that parvov irus
in response to transgenic expression of human renin infection might be a predisposing factor for pre-
and angiotensinogen112 or administered by injection of eclampsia, as the AT1 epitope recognized by anti-AT1
purified anti-AT1 autoantibodies from women with pre- autoantibodies shares a high degree of homology with
eclampsia113) leads to hypertension, proteinuria, glomer- parvovirus B19 capsid proteins.121 However, the pres-
ular capillary endotheliosis and increased production of ence of parvovirus B19 IgG (a marker of previous viral
sVEGFR1114 and soluble endoglin114these effects can be infection) does not correlate with the presence or activity
attenuated by administration of losartan (an AT1-receptor of anti-AT1 autoantibodies.122 Currently, the lack of an
blocker). These observations indicate that proteinuria and immunoassay for this specific AT1 epitope might hinder
renal pathology in rats with anti-AT1 autoantibodiesmight progress in the understanding and implementation of
result from AT1 activation. Anti-AT1autoantibodies can anti-AT1 autoantibody testing in clinical practice.

NATURE REVIEWS | NEPHROLOGY ADVANCE ONLINE PUBLICATION | 7


2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

Intravascular inflammation thrombocytopenia can occur before the development of


Normal pregnancy is characterized by phenotypic and hypertension.49 Several lines of evidence support a role
functional evidence of activation of circulating granulo of platelet activation in pre-eclampsia: increased platelet
cytes and monocytes,17,123 and the magnitude of this size, reduced platelet lifespan, increased maternal plasma
intravascular inflammatory response is increased in levels of platelet factor 4, thromboglobulin and platelet-
patients with pre-eclampsia. 1719 Observational evi- specific proteins stored in granules (and released upon
dence supporting this view includes findings of ele- activation) and increased production of thromboxane B2
vated levels of proinflammatory cytokines in maternal by platelets.20 Platelet activation might also lead to thrombi
blood of pre-eclampsia.124 Experimental studies have in the microcirculation of several target organs.2
also shown that some features of pre-eclampsia, such Since vasospasm and platelet consumption are fea-
as hypertension and placental oxidative stress, can be tures of pre-eclampsia, this disorder was proposed to
elicited by administration of TNF,125 IL6125 or IL17.117 represent an endotheliumplatelet abnormality owing
Although the maternal manifestations of pre-eclampsia to a deficiency in prostacyclin.2 Indeed, prostacyclin has
have been attributed to intravascular inflammation,1719 vasodilatatory effects and inhibits platelet aggregation.2
several studies have demonstrated that patients with Reduced levels of a stable metabolite of prostac yclin
other obstetric syndromes (such as preterm labour in maternal blood133 and urine2 has been reported in
with intact membranes,126 preterm prelabour rupture of association withpre-eclampsia. Moreover, the placentas
membranes,127 IUGR128,129 and pyelonephritis123) have of womenwith pre-eclampsia produce more thrombox-
evidence of intravascular inflammation without hyper- aneA2 than prostacyclin, and thromboxane A2 can induce
tension and proteinuria. Intravascular inflammation is, vasoconstriction and platelet aggregation.134 Asimilar
therefore, a feature of pre-eclampsia, but is not sufficient argument has been made for a role of nitric oxide in
to cause the disorder. The mechanisms responsible for pre-eclampsia, as a deficiency of nitric oxide can cause
intravascular inflammation in pre-eclampsia are thought vasoconstriction and increased platelet aggregation.135
to include increased release of microparticles and nano- Experimental evidence supporting this hypothesis came
particles from the syncytiotrophoblast into the maternal from the observation that administration of nitric oxide
circulation,94 as well as proinflammatory cytokines and blockers (inhibitors of nitric oxide synthase such as or
chemokines released upon activation of NFB in the lnitroarginine methyl ester) to pregnant rats resulted in
context of ER and oxidative stress.97 a pre-eclampsia-like syndrome that included hypertension,
proteinuria, IUGR and g lomerularinjury.136
Endothelial cell activation and/or dysfunction A prominent feature of pre-eclampsia is activation
Endothelial cell activation and/or dysfunction has been of soluble components of the coagulation cascade.137
proposed to be a central feature of pre-eclampsia130 Excessive thrombin generation has been consistently
aconcept that has considerable appeal, as vasospasm demonstrated in pre-eclampsia138 and can be subclini-
isa key component of this disorder. Proteinuria can also cal in nature, or lead to overt disseminated intravascu-
be considered as a manifestation of damage to the fenes- lar coagulation in the presence of placental abruption.
trated glomerular endothelium. In the original observa- Excessive thrombin generation might be related to
tion supporting this hypothesis, sera from patients with endothelial cell dysfunction, platelet activation, chemo
pre-eclampsia (but not from normal pregnant women) taxis of monocytes, proliferation of lymphocytes, neutro
induced the release of 51Cr by human umbilical vein phil activation or excessive generation of tissue factor in
endothelial cells.16,130 Subsequent studies showed that response to the activity of proinflammatory cytokines,
maternal levels of Eselectin and vascular cell adhesion such as IL1 and TNF. Thrombin activation can also lead
protein 1 (VCAM1) were higher in patients with pre- to deposition of fibrin in multiple organ systems, which
eclampsia than in normal pregnant women.131 However, is a major contributor to the pathology of pre-eclampsia.2
the Eselectin findings are not specific for pre-eclampsia, Excessive thrombin generation can be assessed by meas
as elevated levels are also observed in other obstetric uring the concentration of thrombinantithrombin com-
syndromes (including pregnancies with small for gesta- plexes,138 antithrombin III activity 139 or by a thrombin
tional age fetuses).132 Endothelial cell activation and/or generation assay.140 In some patients, pre-eclampsia and/
dysfunction is postulated to be secondary to intravascu- or HELLP syndrome can be considered as thrombotic
lar inflammation.18 Given that other obstetric syndromes microangiopathy-like disorders, in light of the similarities
also feature intravascular inflammation, a clear expla- in clinical presentation and pathology observed in multiple
nation of why this phenomenon would lead to endo organs (thrombocytopenia, haemolysis, endothelial injury,
thelial cell dysfunction in some patients and not others complement activation and deposition of thrombin and/or
is lacking. One possibility is that the different clinical fibrin in arterioles and capillaries of the brain, kidneys and
presentations depend upon the degree of inflammation liver). Examples of other thrombotic microangiopathic
and/or maternal susceptibility to endothelial cell injury. disorders include thrombotic thrombocytopenic purpura
and haemolytic uremic syndrome.141
Platelet and thrombin activation
Pre-eclampsia can be associated with thrombocytopenia An antiangiogenic state
(owing to platelet consumption), which is also associ- Angiogenesis, the formation of new blood vessels from
ated with adverse pregnancy outcomes. 49 Moreover, pre-existing ones, is essential for a successful pregnancy.142

8 | ADVANCE ONLINE PUBLICATION www.nature.com/nrneph


2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

Defective angiogenesis has long been considered as isoform in the placenta and serum of women with pre-
a pathway to pre-eclampsia,143 and rigorous research eclampsia.159 Reduced uteroplacental blood flow,160,161
showed that an antiangiogenic state is involved in the damage to the villous trees,162 syncytial shedding of
pathogenesis of pre-eclampsia. 144 Following on from antiangiogenic factors,162 oxidative stress,103 anti-AT1
observations that patients with cancer who were treated autoantibodies,113 proinflammatory cytokines,114 excess
with antiangiogenic agentssuch as bevacizumab, which thrombin163 and hypoxia164 have all been proposed to be
targets vascular endothelial growth factor (VEGF)-A responsible for the shift in the balance of angiogenic and
developed proteinuria and hypertension,145 an analy- antiangiogenic factors in favour of an antiangiogenic state
sis of differential gene expression patterns (microarray in pre-eclampsia(Figure4).
experiments) showed that mRNA levels of the soluble
VEGF receptor VEGFR1 were higher in the placentas of sVEGFR1 induced hypertension and proteinuria
patients with pre-eclampsia than in those of healthy preg- sVEGFR1 exerts antiangiogenic effects by binding
nant women.144 Subsequently, persuasive evidence was toand inhibiting the biological activity of circulating
gathered in support of the concept that sVEGFR1 has VEGF and PlGF.165 VEGF is important for the maint
a role in the pathogenesis of pre-eclampsia: the median enance of endothelial cell function, especially in fenes-
plasma and/or serum levels of sVEGFR1 are higher in trated endothelium, which is found in brain, liver and
women with pre-eclampsia than in women with normal glomeruli.166 Indeed, knockout of even only a single allele
pregnancies;144,146,147 pre-eclampsia is associated with (Vegfa+/) results in progressive endothelial degeneration,
decreased plasma and/or serum levels of free VEGF and which is fatal at days 1112 in mice.142 Increased availa-
placental growth factor (PlGF);143,144 sera from women bility of sVEGFR1 in pre-eclampsia might also counter
with pre-eclampsia demonstrated antiangiogenic effects act the nitric-oxide-induced vasodilatatory effects of
on endothelial tube formation (a bioassay for angio VEGF, which result in hypertension.144 Indeed, plasma
genesis), and this effect could be reversed by addition nitrite and sVEGFR1 levels are inversely correlated in
of VEGF and PlGF;144 high levels of sVEGFR1 in preg- pre-eclampsia.167 The overall effect of increased produc-
nant animals (achieved by adenovirus-mediated sFlt1 tion of sVEGFR1 is increased maternal vascular tone,
gene transfer) induced hypertension, proteinuria and which maintains uterine perfusion. Moreover, sVEGFR1
glomerular capillary endotheliosis;144 maternal plasma might antagonize the effects of VEGFA, and thereby
concentrations of sVEGFR1 are increased prior to prime endothelial cells to have increased sensitivity to
clinical diagnosis of pre-eclampsia146,148150 and decrease proinflammatory factors such as TNF. These findings
dramatically after delivery;144 maternal plasma levels of suggest the possibility of convergent effects between an
sVEGFR1 are higher in severe pre-eclampsia than in antiangiogenic factor and a proinflammatory cytokine.168
forms of the disease without severe features (previously In addition, sVEGFR1 can induce proteinuria by
termed mild pre-eclampsia), as well as higher in early blocking the effects of VEGFA. Mice with podocyte-
than in late pre-eclampsia;146,147 levels of sVEGFR1 in specific deletion of Veg fa develop glomerular
plasma samples from the uterine vein are significantly capillary endotheliosis, the renal lesion observed in pre-
higher than in samples from the antecubital vein in eclampsia.169 Conversely, overexpression of VEGFA in
patients with pre-eclampsia, but not in normal pregnant podocytes leads to end-stage renal disease, indicating
women;151 and several risk factors for pre-eclampsia are that appropriate expression of VEGFA is essential for
associated with increased plasma and/or serum levels of maintenance of glomerular function.169 Furthermore,
sVEGFR1 (including a history of pre-eclampsia,152 nulli women in the second trimester (2528weeks gestation)
parity,153 multi-fetal gestations,154 diabetes mellitus,155 who subsequently developed pre-eclampsia, and those
chronic hypertension152 and gestationalhypertension156). at the time of diagnosis of pre-eclampsia, have higher
The primary source of the elevated plasma levels numbers of podocytes per milligram of creatinine in
ofsVEGFR1 in pre-eclampsia is the placenta,144 although their urine, identified by immunostaining for podocin
sVEGFR1 can also be produced by peripheral blood after culturing urinary sediments for 24h, than do those
mononuclear cells, macrophages, endothelial cells and with a normal pregnancy.170 In another study, the number
vascular smooth muscle cells.157 Initially, sVEGFR1 of urinary podocytes (identif ied by immunostaining
was reported to be a ~90kDa protein that also existed for podocalyxin) per millilitre of urine correlated with
in a glycosylated form, with a molecular weight ranging total urinary protein, systolic and diastolic blood pres-
100120kDa. Subsequent studies found several vari- sure, and was inversely correlated with both free PlGF
ants of this protein, including 60, 100, 145 and 185kDa and the PlGF to sVEGFR1 ratio at the time of diagnosis
isoforms. The 100kDa isoform (and, to a lesser extent, of pre-eclampsia.170,171 These observations strengthen
the 145kDa isoform) was the main isoform found in the evidence supporting an imbalance between angio-
cultured cytotrophoblasts, peripheral blood mono- genic and antiangiogenic factors and podocyte injury
nuclear cells and uterine microvascular cells, whereas as having a role in development of the clinical features
the 145kDa (along with the 100kDa) isoform was the ofpre-eclampsia.
predominant form in the plasma of women with pre-
eclampsia in one study.158 Another study found that the Role of soluble endoglin
130kDa sVEGFR114 variant (which is expressed specifi- Pregnant rats with sVEGFR1 overexpression (owing
cally in vascular smooth muscle cells) is the predominant to adenovirus-mediated transfection with sFlt1) do not

NATURE REVIEWS | NEPHROLOGY ADVANCE ONLINE PUBLICATION | 9


2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

Genetic factors Abnormal Oxidative stress Increased AT1 Figure 4 | Integrated model of the complex
trophoblast/decidual autoantibodies pathophysiology of pre-eclampsia. Genetic (including
interaction o2
o2 maternalfetal genotype incompatibility) and
o2 environmental (preconception exposure to paternal
ROS antigens) factors disrupt pregnancy-induced
immunomodulation, leading to trophoblast and decidual
pathology, shallow endometrial invasion and failure of
physiological transformation of the spiral arteries
Failure of physiological Defective
(adisorder of deep placentation). The degree of uterine
transformation of deep ischaemia is determined by the severity of the
myometrial segment placentation
of the spiral artery placentation defect and fetal demand on the blood supply.
Obstetric disorders occur when these two factors are
mismatched. The timing and extent of the mismatch
Placental determines the clinical presentation (fetal death, pre-
dysfunction eclampsia with IUGR, IUGR alone and late pre-eclampsia).
Pre-eclampsia occurs as a result of adaptive responses
involving the release of inflammatory cytokines, anti-AT1
autoantibodies, angiogenic and antiangiogenic factors and
syncytiotrophoblast-derived particles into the maternal
Oxidative and Proinflammatory Increased AT1 Syncytiotrophoblast circulation. Collectively, these factors induce leukocyte
endoplasmic cytokines autoantibodies microparticles activation, intravascular inflammation, endothelial cell
reticulum stress and nanoparticles
dysfunction and excessive thrombin generation. The
o2
o2 multiorgan features of pre-eclampsia result from
o2 theconsequences of these processes in different target
organs. Abbreviations: AT1, type1 angiotensin II receptor;
ER, endoplasmic reticulum; ICH, intracerebral
haemorrhage; IUGR, intrauterine growth restriction; PlGF,
placental growth factor; ROS, reactive oxygen species;
s,soluble; VEGF, vascular endothelial growth factor;
VEGFR1, vascular endothelial growth factor receptor 1.
Antiangiogenesis sVEGFR-1
sEndoglin
PIGF endoglin inhibits endothelial tube formation to the
VEGF Angiogenesis
same extent as does sVEGFR1 in invitro assays. 172
Combined administration of adenoviral vectors encod-
Leukocyte and endothelial cell activation
ing endoglin and sFlt1 to pregnant rats generated a
phenotype resembling HELLP syndrome.172 Evidence
for the involvement of soluble endoglin in the patho-
genetic mechanisms of pre-eclampsia includes higher
maternal plasma concentrations of soluble endoglin
inwomen with pre-eclampsia than in healthy pregnant
Proteinuria, Acute IUGR, women, both before 156 and at the time of 172 clinical
renal Pancreatitis fatty liver, fetal
failure liver rupture death diagnosis; levels correlated with severity of disease.156,172
Furthermore, higher maternal serum levels of soluble
Cardiac Haemolytic endoglin were recorded in patients with HELLP syn-
Hypertension failure, Seizure,
ICH, anaemia,
pulmonary
blindness thrombo- drome than in those with pre-eclampsia but without
oedema cytopenia
HELLP syndrome.172 However, subsequent studies found
that plasma levels of soluble endoglin in patients with
HELLP syndrome were either higher than175 or not sig-
display the full spectrum of clinical symptoms observed nificantly different from176 those in patients with pre-
in pre-eclampsia.172 Specifically, these animals develop eclampsia, indicating that soluble endoglin might not
hypertension, proteinuria and glomerular capillary be a specific marker for HELLPsyndrome.
endotheliosis, but not fetal growth restriction, liver
dysfunction or thrombocytopenia.172 A second anti Specificity of the angiogenic balance
angiogenic factor implicated in the pathogenesis of pre- Abnormalities in the profiles of angiogenic and anti
eclampsia is soluble endoglin, a cell-surface co-receptor angiogenic factors are not pathognomonic for pre-
of TGF1 and TGF3 that induces migration and eclampsia. Indeed, such abnormalities are also present
proliferation of endothelial cells.172 Loss-of-function in other conditions, including IUGR,150,177 spontan
mutations in the human ENG gene cause hereditary eous preterm labour,178 fetal death,179,180 maternal floor
haemorrhagic telangiectasia, a disease characterized infarction (massive perivillous fibrin deposition),181
by vascular malformations. 173 Mice lacking endo spontaneous abortion 182 and placental abruption
glin (Eng/) die from defective vascular development, with hypertension.183 Precisely why abnormal angio-
poor vascular smooth muscle development and arrest genic and antiangiogenic profiles are associated with
of endothelial remodelling. 174 Recombinant soluble pre-eclampsia as well as these other complications of

10 | ADVANCE ONLINE PUBLICATION www.nature.com/nrneph


2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

pregnancy remains to be elucidated. We believe that cerebrovascular accident and maternal death. Finally,
the severity and timing of the antiangiogenic state, and the factors that determine pre-eclampsia are likely to
probably also maternal susceptibility, might determine result from a combination of genetic predisposition and
the phenotype. For example, a profoundly antiangio- environmental factors. Patients with urinary tract infec-
genic state could lead to fetal death, with lesser degrees tions or asymptomatic bacteriuria (considered to result
of severity resulting in pre-eclampsia with IUGR, iso- from environmental exposure) during pregnancy are at
lated IUGR or preterm labour with intact membranes. increased risk of developing pre-eclampsia.191 Familial
Moreover, several obstetric conditions that are con- clustering of cases29 and the results of candidate gene
sidered to be risk factors for pre-eclampsiasuch as studies suggest that pre-eclampsia has a genetic com-
molar pregnancy,184 pregnancies with fetal trisomy 13 ponent; 33 however, a geneenvironment interaction
(the FLT1 gene, which encodes VEGFR1, is located on for pre-eclampsia remains to be proven. Pre-eclampsia
chromosome 13)185 and twin-to-twin transfusion syn- also shares many features with atherosclerosis, in which
drome186also have disruption in the balance of angio- solid evidence of geneenvironment interactions exists.
genic and antiangiogenic factors, either in the placenta An attractive possibility is that pre-eclampsia could be
or peripheral blood. considered a pregnancy-specific clinical manifestation
of an increased risk of atherosclerosis. 65 This concept
Great obstetrical syndromes might explain why mothers with pre-eclampsia are at
Obstetric disorders, in contrast to diseases in the non increased risk of cardiovascular disease later in life. Pre-
pregnant state, develop in the context of a unique eclampsia can also be considered to have survival value.
biol ogical situationtwo individuals with different A compromised uterine blood supply can lead to IUGR,
genomes coexisting, one inside the other. The common which is an adaptive mechanism whereby the fetus slows
intere st of mother and fetus is successful reproduc- its growth to avoid outstripping the delivery of oxygen.
tion; however, conflict can occur when the interests of This adaptation alone might be sufficient for the fetus
mother and fetus diverge, perhaps as the result of insults to reach the end of pregnancy. However, when IUGR
(such as infection or a compromised blood supply). We alone is insufficient, signals originating from an ischae
have proposed the term great obstetrical syndromes mic placenta increase the maternal blood p ressure to
to describe common features of important obstetric sustain the fetus and prevent its death.
diseases. These features include multiple aetiologies,
a long subclinical phase, fetal involvement, generally Conclusions
adaptive clinical manifestations and the involvement Pre-eclampsia is a leading cause of maternal and peri-
of geneenvironment interactions.187,188 Although this natal morbidity and mortality. The diagnosis of pre-
concept emerged in the context of preterm labour, it eclampsia is currently based on nonspecific clinical
applies equally to pre-eclampsia. For example, it has symptoms and laboratory tests. Multiple pathogenetic
become increasingly clear that pre-eclampsia is not one mechanisms have been implicated in this disorder,
disorder, but rather different entities recognized by a among which an imbalance between angiogenic and
common phenotype (hypertension and proteinuria), antiangiogenic factors has emerged as one of the most
which represents the manifestation of one or more important. Thus, diagnosis and subclassification of pre-
insults to the uteroplacental unit. Thus, we should not eclampsia based on biomarkers of specific aetiologies
be surprised that multiple pathogenetic mechanisms might be useful for identifying patients at risk, moni-
and predisposing factors have been implicated in pre- toring disease progression and providing effective inter
eclampsia. Patients at risk of pre-eclampsia have an ventions. The roles of anti-AT1 autoantibodies, Bcells
abnormal response to angiotensin II weeks before the and geneenvironment interactions in the pathogenesis
clinical diagnosis can be made,109 and abnormal uterine of pre-eclampsia and other obstetric syndromes should
artery Doppler velocimetry measurements and elevated be investigated. Further understanding of the mecha-
levels of angiogenic factors often manifest months nisms of abnormal placentation could advance current
before a clinical diagnosis of pre-eclampsia. Within this knowledge of the pathogenesis of pre-eclampsia as well
long subclinical phase lies the opportunity for predic- as other disorders, such as fetal growth restriction, still-
tion and prevention of pre-eclampsia. Although pre- birth and some types of spontaneous preterm deliv-
eclampsia is diagnosed by clinical signs in the mother ery. In part2 of this Review, we discuss the prediction,
(hypertension and proteinuria), fetal involvement can prevention and management of pre-eclampsia.22
manifest as IUGR as well as other more-subtle abnor-
malities, such as thrombocytopenia or neutropenia.189 Review criteria
Importantly, hypertension is not the cause of pre-
A search for original research and review articles
eclampsia, but it can be considered an adaptive response published in English between 1840 and 2013 focusing
of the ischaemic uteroplacental unit, which signals to on pre-eclampsia was performed in PubMed, using
the mother the need to maintain perfusion. The resolu- the following search terms, alone or in combination:
tion of maternal hypertension that follows fetal death in pre-eclampsia, toxaemia, pregnancy-induced
some patients with pre-eclampsia supports this view.190 hypertension and eclampsia. The bibliographies of
However, adaptive responses can become maladap- pertinent articles were also examined to identify further
relevant papers.
tive; for instance, a hypertensive crisis can result in

NATURE REVIEWS | NEPHROLOGY ADVANCE ONLINE PUBLICATION | 11


2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

1. Lindheimer, M.D., Roberts, J.M., 20. Kenny, L.C., Baker, P.N. & Cunningham, F.G. in predictor of absent or severe proteinuria. Am. J.
Cunningham,G.C. & Chesley, L. in Chesleys Chesleys Hypertensive Disorders in Pregnancy Obstet. Gynecol. 170, 137141 (1994).
Hypertensive Disorders in Pregnancy (eds (eds Lindheimer, M.D., Roberts, J.M. & 41. Kuo, V.S., Koumantakis, G. & Gallery, E.D.
Lindheimer, M.D., Roberts, J.M. & Cunningham, G.C.) 335351 (Elsevier, 2009). Proteinuria and its assessment in normal and
Cunningham,G.C.), 124 (Elsevier, 2009). 21. Lindheimer, M.D., Roberts, J.M., hypertensive pregnancy. Am. J. Obstet. Gynecol.
2. Romero, R., Lockwood, C., Oyarzun, E. & Cunningham,G.C. & Chesley, L. in Chesleys 167, 723728 (1992).
Hobbins, J.C. Toxemia: new concepts in an old Hypertensive Disorders in Pregnancy (eds 42. Lindow, S.W. & Davey, D.A. The variability of
disease. Semin. Perinatol. 12, 302323 (1988). Lindheimer, M.D., Roberts, J.M. & urinary protein and creatinine excretion in
3. Redman, C.W. & Sargent, I.L. Latest advances Cunningham,G.C.) 2536 (Elsevier, 2009). patients with gestational proteinuric
in understanding preeclampsia. Science 308, 22. Chaiworapongsa, T., Chaemsaithong, P., hypertension. Br. J. Obstet. Gynaecol. 99,
15921594 (2005). Korzeniewski, S.J., Yeo, L. & Romero, R. 869872 (1992).
4. Roberts, J.M. & Gammill, H.S. Preeclampsia: Pre-eclampsia part 2: prediction, prevention 43. Verdonk, K. etal. Variation of urinary protein
recent insights. Hypertension 46, 12431249 andmanagement. Nat. Rev. Nephrol. http:// tocreatinine ratio during the day in women
(2005). dx.doi.org/10.1038/nrneph.2014.103. withsuspected pre-eclampsia. BJOG http://
5. Sibai, B., Dekker, G. & Kupferminc, M. 23. Lindheimer, M.D. W. Benson and Pamela Harer dx.doi.org/10.1111/1471-0528.12803.
Pre-eclampsia. Lancet 365, 785799 (2005). Seminar on History. The History of Preeclampsia 44. Cote, A.M. etal. Diagnostic accuracy of urinary
6. Steegers, E.A., von Dadelszen, P., Duvekot, J.J. and Eclampsia as Seen by a Nephrologist (2012). spot protein:creatinine ratio for proteinuria in
& Pijnenborg, R. Pre-eclampsia. Lancet 376, 24. Lever, J.C. Cases of puerperal convulsions with hypertensive pregnant women: systematic
631644 (2010). remarks. Guys Hosp. Rep. 2, 495517 (1843). review. BMJ 336, 10031006 (2008).
7. American College of Obstetricians and 25. Ballantyne, J.W. Sphygmographic tracings in 45. Thangaratinam, S. etal. Estimation of
Gynecologists Task Force on Hypertension in puerperal eclampsia. Edinburgh Med. J. 30, proteinuria as a predictor of complications of
Pregnancy, Hypertension in Pregnancy [online], 10071020 (1885). pre-eclampsia: a systematic review. BMC Med. 7,
http://www.acog.org/resources_and_ 26. Ballantyne, J.W. A plea for a pro-maternity 10 (2009).
publications/task_force_and_work_group_ hospital. Br. Med. J. 1, 813814 (1901). 46. Goodlin, R.C. Severe pre-eclampsia: another
reports/hypertension_in_pregnancy (2013). 27. Redman, C.W. & Sargent, I.L. Immunology great imitator. Am. J. Obstet. Gynecol. 125,
8. Lowe, S.A. etal. Guidelines for the management ofpre-eclampsia. Am. J. Reprod. Immunol. 63, 747753 (1976).
of hypertensive disorders of pregnancy 2008. 534543 (2010). 47. Weinstein, L. Syndrome of hemolysis, elevated
Aust. NZ J. Obstet. Gynaecol. 49, 242246 (2009). 28. Dekker, G., Robillard, P.Y. & Roberts, C. The liver enzymes, and low platelet count: a severe
9. Hutcheon, J.A., Lisonkova, S. & Joseph, K.S. etiology of preeclampsia: the role of the father. consequence of hypertension in pregnancy.
Epidemiology of pre-eclampsia and the other J.Reprod. Immunol. 89, 126132 (2011). Am.J. Obstet. Gynecol. 142, 159167 (1982).
hypertensive disorders of pregnancy. Best Pract. 29. Chesley, L.C., Annitto, J.E. & Cosgrove, R.A. 48. Romero, R. etal. Clinical significance of liver
Res. Clin. Obstet. Gynaecol. 25, 391403 (2011). The familial factor in toxemia of pregnancy. dysfunction in pregnancy-induced hypertension.
10. Thornton, C., Dahlen, H., Korda, A. & Obstet. Gynecol. 32, 303311 (1968). Am. J. Perinatol. 5, 146151 (1988).
Hennessy,A. The incidence of preeclampsia and 30. Thornton, J.G. & Macdonald, A.M. Twin 49. Romero, R. etal. Clinical significance,
eclampsia and associated maternal mortality in mothers, pregnancy hypertension and pre- prevalence, and natural history of
Australia from population-linked datasets: eclampsia. Br. J. Obstet. Gynaecol. 106, thrombocytopenia in pregnancy-induced
20002008 Am. J. Obstet. Gynecol. 208, 476 570575 (1999). hypertension. Am. J. Perinatol. 6, 3238 (1989).
e471e475 (2013). 31. Goddard, K.A. etal. Candidate-gene association 50. Sibai, B.M. Diagnosis, controversies, and
11. Adu-Bonsaffoh, K., Samuel, O.A. & Binlinla, G. study of mothers with pre-eclampsia, and their management of the syndrome of hemolysis,
Maternal deaths attributable to hypertensive infants, analyzing 775SNPs in 190 genes. elevated liver enzymes, and low platelet count.
disorders in a tertiary hospital in Ghana. Int. J. Hum.Hered. 63, 116 (2007). Obstet. Gynecol. 103, 981991 (2004).
Gynaecol Obstet. 123, 110113 (2013). 32. Parimi, N. etal. Analytical approaches to detect 51. von Dadelszen, P., Magee, L.A. & Roberts, J.M.
12. Acosta-Sison, H. The relationship of hydatidiform maternal/fetal genotype incompatibilities that Subclassification of preeclampsia. Hypertens.
mole to pre-eclampsia and eclampsia; a study increase risk of pre-eclampsia. BMC Med. Genet. Pregnancy 22, 143148 (2003).
of85 cases. Am. J. Obstet. Gynecol. 71, 9, 60 (2008). 52. Crispi, F. etal. Predictive value of angiogenic
12791282 (1956). 33. Ward, K. & Lindheimer, M.D. in Chesleys factors and uterine artery Doppler for early-
13. Young, J. The aetiology of eclampsia and Hypertensive Disorders in Pregnancy (eds versus late-onset pre-eclampsia and intrauterine
albuminuria and their relation to accidental Lindheimer, M.D., Roberts, J.M. & growth restriction. Ultrasound Obstet. Gynecol.
haemorrhage: (an anatomical and experimental Cunningham,G.C.) 5171 (Elsevier, 2009). 31, 303309 (2008).
investigation.). Proc. R. Soc. Med. 7, 307348 34. Zhao, L., Bracken, M.B., Dewan, A.T. & Chen, S. 53. Soto, E. etal. Late-onset preeclampsia is
(1914). Association between the SERPINE1 (PAI1) associated with an imbalance of angiogenic and
14. Page, E.W. On the pathogenesis of pre- 4G/5G insertion/deletion promoter anti-angiogenic factors in patients with and
eclampsia and eclampsia. J. Obstet. Gynaecol polymorphism (rs1799889) and pre-eclampsia: without placental lesions consistent with
Br.Commonw. 79, 883894 (1972). a systematic review and meta-analysis. maternal underperfusion. J. Matern. Fetal
15. Rodgers, G.M., Taylor, R.N. & Roberts, J.M. Mol.Hum. Reprod. 19, 136143 (2013). Neonatal Med. 25, 498507 (2012).
Preeclampsia is associated with a serum factor 35. Zhao, L. etal. Genome-wide association study 54. Parra-Cordero, M. etal. Prediction of early
cytotoxic to human endothelial cells. Am. J. identifies a maternal copy-number deletion in andlate pre-eclampsia from maternal
Obstet. Gynecol. 159, 908914 (1988). PSG11 enriched among preeclampsia patients. characteristics, uterine artery Doppler and
16. Roberts, J.M., Edep, M.E., Goldfien, A. & BMC Pregnancy Childbirth 12, 61 (2012). markers of vasculogenesis during first trimester
Taylor,R.N. Sera from preeclamptic women 36. Morgan, M.A. & Thurnau, G.R. Pregnancy- of pregnancy. Ultrasound Obstet. Gynecol. 41,
specifically activate human umbilical vein induced hypertension without proteinuria: is it 538544 (2013).
endothelial cells invitro: morphological and true preeclampsia? South. Med. J. 81, 210213 55. Ogge, G. etal. Placental lesions associated with
biochemical evidence. Am. J. Reprod. Immunol. (1988). maternal underperfusion are more frequent in
27, 101108 (1992). 37. Barton, J.R., OBrien, J. M., Bergauer, N.K., early-onset than in late-onset preeclampsia.
17. Sacks, G.P., Studena, K., Sargent, K. & Jacques, D.L. & Sibai, B.M. Mild gestational J.Perinat. Med. 39, 641652 (2011).
Redman,C.W. Normal pregnancy and hypertension remote from term: progression and 56. Sibai, B.M. Evaluation and management of
preeclampsia both produce inflammatory outcome. Am. J. Obstet. Gynecol. 184, 979983 severe preeclampsia before 34 weeks
changes in peripheral blood leukocytes akin to (2001). gestation. Am. J. Obstet. Gynecol. 205, 191198
those of sepsis. Am. J. Obstet. Gynecol. 179, 38. Homer, C.S., Brown, M.A., Mangos, G. & (2011).
8086 (1998). Davis,G.K. Non-proteinuric pre-eclampsia: 57. Ogden, E., Hildebrand, G.J. & Page, E.W. Rise
18. Redman, C.W., Sacks, G.P. & Sargent, I.L. anovel risk indicator in women with gestational of blood pressure during ischemia of gravid
Preeclampsia: an excessive maternal hypertension. J. Hypertens. 26, 295302 (2008). uterus. Proc. Soc. Exp. Bio Med. 43, 4951
inflammatory response to pregnancy. Am. J. 39. Lindheimer, M.D. & Kanter, D. Interpreting (1940).
Obstet. Gynecol. 180, 499506 (1999). abnormal proteinuria in pregnancy: the need 58. Lunell, N.O., Nylund, L.E., Lewander, R. &
19. Gervasi, M.T. etal. Phenotypic and metabolic fora more pathophysiological approach. Obstet. Sarby,B. Uteroplacental blood flow in pre-
characteristics of monocytes and granulocytes Gynecol. 115, 365375 (2010). eclampsia measurements with indium113m
in preeclampsia. Am. J. Obstet. Gynecol. 185, 40. Meyer, N.L., Mercer, B.M., Friedman, S.A. & and a computer-linked gamma camera. Clin. Exp.
792797 (2001). Sibai, B.M. Urinary dipstick protein: a poor Hypertens. B 1, 105117 (1982).

12 | ADVANCE ONLINE PUBLICATION www.nature.com/nrneph


2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

59. Brosens, I., Robertson, W.B. & Dixon, H.G. 77. Osol, G. & Moore, L.G. Maternal uterine 94. Redman, C.W. & Sargent, I.L. Microparticles
Thephysiological response of the vessels of vascular remodeling during pregnancy. and immunomodulation in pregnancy and
theplacental bed to normal pregnancy. J. Pathol. Microcirculation 21, 3847 (2014). pre-eclampsia. J. Reprod. Immunol. 76, 6167
Bacteriol. 93, 569579 (1967). 78. Burton, G.J., Watson, A.L., Hempstock, J., (2007).
60. Brosens, I. & Renaer, M. On the pathogenesis Skepper, J.N. & Jauniaux, E. Uterine glands 95. Lian, I.A. etal. Increased endoplasmic reticulum
ofplacental infarcts in pre-eclampsia. provide histiotrophic nutrition for the human stress in decidual tissue from pregnancies
J.Obstet.Gynaecol Br. Commonw. 79, 794799 fetus during the first trimester of pregnancy. complicated by fetal growth restriction with and
(1972). J.Clin. Endocrinol. Metab. 87, 29542959 without pre-eclampsia. Placenta 32, 823829
61. Hertig, A.T. Vascular pathology in hypertensive (2002). (2011).
albuminuric toxemias of pregnancy. Clinics 4, 79. Burton, G.J., Hempstock, J. & Jauniaux, E. 96. Burton, G.J. & Jauniaux, E. Oxidative stress.
10111015 (1945). Nutrition of the human fetus during the first Best Pract Res. Clin. Obstet. Gynaecol. 25,
62. De Wolf, F., Robertson, W.B. & Brosens, I. The trimestera review. Placenta 22 (Suppl. A), 287299 (2011).
ultrastructure of acute atherosis in hypertensive S70S77 (2001). 97. Cindrova-Davies, T., Spasic-Boskovic, O.,
pregnancy. Am. J. Obstet. Gynecol. 123, 164174 80. Jauniaux, E. etal. Onset of maternal arterial Jauniaux, E., Charnock-Jones, D.S. &
(1975). blood flow and placental oxidative stress. Burton,G.J. Nuclear factor-B, p38, and
63. Robertson, W.B., Brosens, I. & Dixon, H.G. Apossible factor in human early pregnancy stress-activated protein kinase mitogen-
Thepathological resonse of the vessels of the failure. Am. J. Pathol. 157, 21112122 activated protein kinase signaling pathways
placental bed to hypertensive pregnancy. (2000). regulate proinflammatory cytokines and
J.Pathol. Bacteriol. 93, 581592 (1967). 81. Genbacev, O., Joslin, R., Damsky, C.H., apoptosis in human placental explants in
64. Labarrere, C.A. Acute atherosis. Polliotti,B.M. & Fisher, S.J. Hypoxia alters early response to oxidative stress: effects of
Ahistopathological hallmark of immune gestation human cytotrophoblast differentiation/ antioxidant vitamins. Am. J. Pathol. 170,
aggression? Placenta 9, 95108 (1988). invasion invitro and models the placental 15111520 (2007).
65. Staff, A.C., Dechend, R. & Redman, C.W. defects that occur in preeclampsia. J. Clin. 98. Martin, C.B. Jr, McGaughey, H.S. Jr, Kaiser,I.H.,
Review: Preeclampsia, acute atherosis of the Invest. 97, 540550 (1996). Donner, M.W. & Ramsey, E.M. Intermittent
spiral arteries and future cardiovascular 82. Caniggia, I. etal. Hypoxia-inducible factor1 functioning of the uteroplacental arteries. Am. J.
disease: two new hypotheses. Placenta mediates the biological effects of oxygen on Obstet. Gynecol. 90, 819823 (1964).
34(Suppl.), S73S78 (2013). human trophoblast differentiation through 99. Burton, G.J., Woods, A.W., Jauniaux, E. &
66. Brosens, I., Pijnenborg, R., Vercruysse, L. & TGF(3). J. Clin. Invest. 105, 577587 Kingdom, J.C. Rheological and physiological
Romero, R. The Great Obstetrical Syndromes (2000). consequences of conversion of the maternal
are associated with disorders of deep 83. Rajakumar, A., Brandon, H.M., Daftary, A., spiral arteries for uteroplacental blood flow
placentation. Am. J. Obstet. Gynecol. 204, Ness,R. & Conrad, K.P. Evidence for the during human pregnancy. Placenta 30, 473482
193201 (2011). functional activity of hypoxia-inducible (2009).
67. Khong, T.Y., Liddell, H.S. & Robertson, W.B. transcription factors overexpressed in 100. Many, A., Hubel, C.A., Fisher, S.J.,
Defective haemochorial placentation as a cause preeclamptic placentae. Placenta 25, 763769 Roberts,J.M. & Zhou, Y. Invasive
of miscarriage: a preliminary study. Br. J. Obstet. (2004). cytotrophoblasts manifest evidence of oxidative
Gynaecol. 94, 649655 (1987). 84. Tal, R. etal. Effects of hypoxia-inducible factor-1 stress in preeclampsia. Am.J. Pathol. 156,
68. Ball, E., Bulmer, J.N., Ayis, S., Lyall, F. & overexpression in pregnant mice: possible 321331 (2000).
Robson,S.C. Late sporadic miscarriage is implications for preeclampsia and intrauterine 101. Vaughan, J.E. & Walsh, S.W. Oxidative
associated with abnormalities in spiral artery growth restriction. Am. J. Pathol. 177, stressreproduces placental abnormalities
transformation and trophoblast invasion. 29502962 (2010). ofpreeclampsia. Hypertens. Pregnancy 21,
J.Pathol. 208, 535542 (2006). 85. Kanasaki, K. etal. Deficiency in catecholO 205223 (2002).
69. Brosens, I.A., Robertson, W.B. & Dixon, H.G. methyltransferase and 2methoxyoestradiol 102. George, E.M. & Granger, J.P. Heme oxygenase
The role of the spiral arteries in the isassociated with pre-eclampsia. Nature 453, in pregnancy and preeclampsia. Curr. Opin.
pathogenesis of preeclampsia. Obstet. Gynecol. 11171121 (2008). Nephrol. Hypertens. 22, 156162 (2013).
Annu. 1, 177191 (1972). 86. Palmer, K. etal. Severe early-onset preeclampsia 103. Zhao, H., Wong, R.J., Kalish, F.S., Nayak, N.R.
70. Khong, T.Y., De Wolf, F., Robertson, W.B. & is not associated with a change in placental & Stevenson, D.K. Effect of heme oxygenase1
Brosens, I. Inadequate maternal vascular catechol Omethyltransferase (COMT) deficiency on placental development. Placenta
response to placentation in pregnancies expression. Am. J. Pathol. 178, 24842488 30, 861868 (2009).
complicated by pre-eclampsia and by (2011). 104. Nakamura, M. etal. Cellular mRNA expressions
smallforgestational age infants. Br. J. Obstet. 87. Seol, H.J., Cho, G.J., Oh, M.J. & Kim, H.J. of anti-oxidant factors in the blood of
Gynaecol. 93, 10491059 (1986). 2methoxyoestradiol levels and placental preeclamptic women. Prenat Diagn. 29,
71. Dommisse, J. & Tiltman, A.J. Placental bed catecholOmethyltransferase expression in 691696 (2009).
biopsies in placental abruption. Br. J. Obstet. patients with late-onset preeclampsia. Arch. 105. Lash, G.E. etal. Relationship between tissue
Gynaecol. 99, 651654 (1992). Gynecol. Obstet. 287, 881886 (2013). damage and heme oxygenase expression in
72. Kim, Y.M. etal. Failure of physiologic 88. Redman, C.W. & Sargent, I.L. Placental stress chorionic villi of term human placenta. Am. J.
transformation of the spiral arteries in patients and pre-eclampsia: a revised view. Placenta Physiol. Heart Circ. Physiol. 284, H160H167
with preterm labor and intact membranes. 30(Suppl. A), S38S42 (2009). (2003).
Am.J.Obstet. Gynecol. 189, 10631069 (2003). 89. Brosens, J.J., Parker, M.G., McIndoe, A., 106. Farina, A. etal. Gene expression in chorionic
73. Kim, Y.M. etal. Failure of physiologic Pijnenborg, R. & Brosens, I.A. A role for villous samples at 11weeks gestation from
transformation of the spiral arteries in the menstruation in preconditioning the uterus for women destined to develop preeclampsia.
placental bed in preterm premature rupture successful pregnancy. Am. J. Obstet. Gynecol. Prenat. Diagn. 28, 956961 (2008).
ofmembranes. Am. J. Obstet. Gynecol. 187, 200, 615.e1615.e6 (2009). 107. George, E.M. etal. Induction of heme
11371142 (2002). 90. Moffett, A. & Hiby, S.E. How does the maternal oxygenase1 attenuates placental ischemia-
74. Pijnenborg, R. & Brosens, I. in Placental Bed immune system contribute to the development induced hypertension. Hypertension 57,
Disorders: Basic Science and its Translation to of preeclampsia? Placenta 28 (Suppl. A), 941948 (2011).
Obstetrics (eds Pijnenborg, R., Brosens, I. & S51S56 (2007). 108. Costantine, M.M. etal. Using pravastatin to
Romero, R.) 97108 (Cambridge University 91. Zhang, K. & Kaufman, R.J. From endoplasmic- improve the vascular reactivity in a mouse model
Press, 2010). reticulum stress to the inflammatory response. of soluble fms-like tyrosine kinase1induced
75. Zhou, Y., Damsky, C.H. & Fisher, S.J. Nature 454, 455462 (2008). preeclampsia. Obstet. Gynecol. 116, 114120
Preeclampsia is associated with failure of 92. Burton, G.J., Yung, H.W., Cindrova-Davies, T. & (2010).
human cytotrophoblasts to mimic a vascular Charnock-Jones, D.S. Placental endoplasmic 109. Gant, N.F., Chand, S., Whalley, P.J. &
adhesion phenotype. One cause of defective reticulum stress and oxidative stress in the MacDonald, P.C. The nature of pressor
endovascular invasion in this syndrome? J. Clin. pathophysiology of unexplained intrauterine responsiveness to angiotensin II in human
Invest. 99, 21522164 (1997). growth restriction and early onset preeclampsia. pregnancy. Obstet. Gynecol. 43, 854 (1974).
76. Rosenfeld, C.R., Roy, T. & Cox, B.E. Placenta 30 (Suppl. A), S43S48 (2009). 110. Dechend, R., Luft, F.C. & Lindheimer, M. in
Mechanisms modulating estrogen-induced 93. Huppertz, B., Kadyrov, M. & Kingdom, J.C. Chesleys Hypertensive Disorders in Pregnancy
uterine vasodilation. Vascul. Pharmacol. 38, Apoptosis and its role in the trophoblast. Am. J. (eds Lindheimer, M.D., Roberts, J.M. &
115125 (2002). Obstet. Gynecol. 195, 2939 (2006). Cunningham, G.C.) 287296 (Elsevier, 2009).

NATURE REVIEWS | NEPHROLOGY ADVANCE ONLINE PUBLICATION | 13


2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

111. Wallukat, G. etal. Patients with preeclampsia ofmembranes. J. Matern. Fetal Neonatal Med. 146. Levine, R.J. etal. Circulating angiogenic factors
develop agonistic autoantibodies against the 11, 171175 (2002). and the risk of preeclampsia. N. Engl. J. Med.
angiotensin AT1 receptor. J. Clin. Invest. 103, 128. Sabatier, F. etal. Neutrophil activation in 350, 672683 (2004).
945952 (1999). preeclampsia and isolated intrauterine growth 147. Chaiworapongsa, T. etal. Evidence supporting
112. Dechend, R. etal. Agonistic autoantibodies to restriction. Am. J. Obstet. Gynecol. 183, arole for blockade of the vascular endothelial
the AT1 receptor in a transgenic rat model of 15581563 (2000). growth factor system in the pathophysiology of
preeclampsia. Hypertension 45, 742746 129. Ogge, G. etal. Leukocytes of pregnant women preeclampsia. Young Investigator Award. Am. J.
(2005). with smallforgestational age neonates have a Obstet. Gynecol. 190, 15411550 (2004).
113. Zhou, C.C. etal. Angiotensin receptor agonistic different phenotypic and metabolic activity from 148. Chaiworapongsa, T. etal. Plasma soluble
autoantibodies induce pre-eclampsia in pregnant those of women with preeclampsia. J. Matern. vascular endothelial growth factor receptor1
mice. Nat. Med. 14, 855862 (2008). Fetal Neonatal Med. 23, 476487 (2010). concentration is elevated prior to the clinical
114. Parrish, M.R. etal. The effect of immune factors, 130. Roberts, J.M. etal. Preeclampsia: diagnosis of pre-eclampsia. J. Matern. Fetal
tumor necrosis factor-alpha, and agonistic anendothelial cell disorder. Am. J. Obstet. Neonatal Med. 17, 318 (2005).
autoantibodies to the angiotensin II typeI Gynecol. 161, 12001204 (1989). 149. Widmer, M. etal. Mapping the theories of
receptor on soluble fms-like tyrosine1 and 131. Chaiworapongsa, T. etal. Soluble adhesion preeclampsia and the role of angiogenic factors:
soluble endoglin production in response to molecule profile in normal pregnancy and pre- a systematic review. Obstet. Gynecol. 109,
hypertension during pregnancy. Am. J. Hypertens. eclampsia. J. Matern. Fetal Neonatal Med. 12, 168180 (2007).
23, 911916 (2010). 1927 (2002). 150. Romero, R. etal. A longitudinal study of
115. Xia, Y. & Kellems, R.E. Angiotensin receptor 132. Bretelle, F. etal. Maternal endothelial soluble angiogenic (placental growth factor) and anti-
agonistic autoantibodies and hypertension: cell adhesion molecules with isolated small for angiogenic (soluble endoglin and soluble
preeclampsia and beyond. Circ. Res. 113, 7887 gestational age fetuses: comparison with pre- vascular endothelial growth factor receptor1)
(2013). eclampsia. BJOG 108, 12771282 (2001). factors in normal pregnancy and patients
116. Li, J., LaMarca, B. & Reckelhoff, J.F. A model of 133. Bussolino, F., Benedetto, C., Massobrio, M. & destined to develop preeclampsia and deliver
preeclampsia in rats: the reduced uterine Camussi, G. Maternal vascular prostacyclin asmall for gestational age neonate. J. Matern.
perfusion pressure (RUPP) model. Am. J. Physiol. activity in pre-eclampsia. Lancet 2, 702 (1980). Fetal Neonatal Med. 21, 923 (2008).
Heart Circ. Physiol. 303, H1H8 (2012). 134. Walsh, S.W. Preeclampsia: an imbalance 151. Bujold, E. etal. Evidence supporting that the
117. Dhillion, P. etal. IL17mediated oxidative stress inplacental prostacyclin and thromboxane excess of the sVEGFR1 concentration in
is an important stimulator of AT1-AA and production. Am. J. Obstet. Gynecol. 152, maternal plasma in preeclampsia has a uterine
hypertension during pregnancy. Am. J. Physiol. 335340 (1985). origin. J. Matern. Fetal Neonatal Med. 18, 916
Regul. Integr Comp. Physiol. 303, R353R358 135. Freedman, J.E. etal. Deficient platelet-derived (2005).
(2012). nitric oxide and enhanced hemostasis in mice 152. Maynard, S.E. etal. Gestational angiogenic
118. Girardi, G., Yarilin, D., Thurman, J.M., lacking the NOSIII gene. Circ. Res. 84, biomarker patterns in high risk preeclampsia
Holers,V.M. & Salmon, J.E. Complement 14161421 (1999). groups. Am. J. Obstet. Gynecol. 53, e1e9
activation induces dysregulation of angiogenic 136. Yallampalli, C. & Garfield, R.E. Inhibition of nitric (2013).
factors and causes fetal rejection and growth oxide synthesis in rats during pregnancy 153. Wolf, M. etal. Circulating levels of the
restriction. J.Exp. Med. 203, 21652175 produces signs similar to those of preeclampsia. antiangiogenic marker sFLT1 are increased in
(2006). Am. J. Obstet. Gynecol. 169, 13161320 (1993). first versus second pregnancies. Am. J. Obstet.
119. Herse, F. & LaMarca, B. Angiotensin II type1 137. Cadroy, Y. etal. Evaluation of six markers of Gynecol. 193, 1622 (2005).
receptor autoantibody (AT1-AA)-mediated haemostatic system in normal pregnancy and 154. Bdolah, Y. etal. Twin pregnancy and the risk
pregnancy hypertension. Am. J. Reprod. Immunol. pregnancy complicated by hypertension or pre- ofpreeclampsia: bigger placenta or relative
69, 413418 (2013). eclampsia. Br. J. Obstet. Gynaecol. 100, ischemia? Am. J. Obstet. Gynecol. 198,
120. Novotny, S.R. etal. Activating autoantibodies 416420 (1993). 428.e1428.e6 (2008).
tothe angiotensin II typeI receptor play an 138. Chaiworapongsa, T. etal. Evidence of invivo 155. Cohen, A. etal. Circulating levels of the
important role in mediating hypertension in generation of thrombin in patients with antiangiogenic marker soluble FMS-like tyrosine
response to adoptive transfer of CD4+ smallforgestational-age fetuses and pre- kinase 1 are elevated in women with
Tlymphocytes from placental ischemic rats. eclampsia. J. Matern. Fetal Neonatal Med. 11, pregestational diabetes and preeclampsia:
Am.J.Physiol. Regul. Integr Comp. Physiol. 302, 362367 (2002). angiogenic markers in preeclampsia and
R11971201 (2012). 139. Kobayashi, T., Tokunaga, N., Sugimura, M., preexisting diabetes. Diabetes Care 30,
121. Herse, F. etal. Prevalence of agonistic Kanayama, N. & Terao, T. Predictive values of 375377 (2007).
autoantibodies against the angiotensin II type1 coagulation/fibrinolysis parameters for the 156. Levine, R.J. etal. Soluble endoglin and other
receptor and soluble fms-like tyrosine kinase 1 termination of pregnancy complicated by severe circulating antiangiogenic factors in
in a gestational age-matched case study. preeclampsia. Semin. Thromb. Hemost. 27, preeclampsia. N. Engl. J. Med. 355, 9921005
Hypertension 53, 393398 (2009). 137141 (2001). (2006).
122. Stepan, H., Wallukat, G., Schultheiss, H.P., 140. Sharma, S.K., Philip, J., Whitten, C.W., 157. Rajakumar, A. etal. Extra-placental expression
Faber, R. & Walther, T. Is parvovirus B19 the Padakandla, U.B. & Landers, D.F. Assessment ofvascular endothelial growth factor receptor1,
cause for autoimmunity against the of changes in coagulation in parturients with (Flt1) and soluble Flt1 (sFlt1), by peripheral
angiotensinII type receptor? J. Reprod. Immunol. preeclampsia using thromboelastography. blood mononuclear cells (PBMCs) in
73, 130134 (2007). Anesthesiology 90, 385390 (1999). normotensive and preeclamptic pregnant
123. Naccasha, N. etal. Phenotypic and metabolic 141. Fakhouri, F., Vercel, C. & Fremeaux-Bacchi, V. women. Placenta 26, 563573 (2005).
characteristics of monocytes and granulocytes Obstetric nephrology: AKI and thrombotic 158. Rajakumar, A. etal. Novel soluble Flt1 isoforms
in normal pregnancy and maternal infection. microangiopathies in pregnancy. Clin. J. Am. Soc. in plasma and cultured placental explants from
Am.J. Obstet. Gynecol. 185, 11181123 (2001). Nephrol. 7, 21002106 (2012). normotensive pregnant and preeclamptic
124. Lau, S.Y. etal. Tumor necrosis factor-, 142. Ferrara, N. etal. Heterozygous embryonic women. Placenta 30, 2534 (2009).
interleukin-6, and interleukin-10 levels are lethality induced by targeted inactivation of the 159. Sela, S. etal. A novel human-specific soluble
altered in preeclampsia: a systematic review VEGF gene. Nature 380, 439442 (1996). vascular endothelial growth factor receptor 1:
andmeta-analysis. Am. J. Reprod. Immunol. 70, 143. Torry, D.S., Wang, H.S., Wang, T.H., Caudle,M.R. celltypespecific splicing and implications to
412427 (2013). & Torry, R.J. Preeclampsia is associated with vascular endothelial growth factor homeostasis
125. McCarthy, F.P., Kingdom, J.C., Kenny, L.C. & reduced serum levels of placenta growth factor. and preeclampsia. Circ. Res. 102, 15661574
Walsh, S.K. Animal models of preeclampsia: Am. J. Obstet. Gynecol. 179, 15391544 (1998). (2008).
uses and limitations. Placenta 32, 413419 144. Maynard, S.E. etal. Excess placental soluble 160. Gilbert, J.S., Babcock, S.A. & Granger, J.P.
(2011). fms-like tyrosine kinase 1 (sFlt1) may contribute Hypertension produced by reduced uterine
126. Gervasi, M.T. etal. Phenotypic and metabolic to endothelial dysfunction, hypertension, and perfusion in pregnant rats is associated with
characteristics of maternal monocytes and proteinuria in preeclampsia. J. Clin. Invest. 111, increased soluble fms-like tyrosine kinase1
granulocytes in preterm labor with intact 649658 (2003). expression. Hypertension 50, 11421147
membranes. Am. J. Obstet. Gynecol. 185, 145. Yang, J.C. etal. A randomized trial of (2007).
11241129 (2001). bevacizumab, an anti-vascular endothelial 161. Makris, A. etal. Uteroplacental ischemia results
127. Gervasi, M.T. etal. Maternal intravascular growth factor antibody, for metastatic renal in proteinuric hypertension and elevated sFLT1.
inflammation in preterm premature rupture cancer. N. Engl. J. Med. 349, 427434 (2003). Kidney Int. 71, 977984 (2007).

14 | ADVANCE ONLINE PUBLICATION www.nature.com/nrneph


2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

162. Rajakumar, A. etal. Transcriptionally active thrombocytopenia from HELLP syndrome. 191. Conde-Agudelo, A., Villar, J. & Lindheimer, M.
syncytial aggregates in the maternal circulation J.Matern. Fetal Neonatal Med. 23, 366370 Maternal infection and risk of preeclampsia:
may contribute to circulating soluble fms-like (2010). systematic review and metaanalysis. Am. J.
tyrosine kinase 1 in preeclampsia. Hypertension 177. Chaiworapongsa, T. etal. The maternal plasma Obstet. Gynecol. 198, 722 (2008).
59, 256264 (2012). soluble vascular endothelial growth factor 192. Leveno, K.J. & Cunningham, F.G. in Chesleys
163. Lockwood, C.J. etal. Thrombin regulates soluble receptor1 concentration is elevated in SGA and Hypertensive Disorders in Pregnancy (eds
fms-like tyrosine kinase1 (sFlt1) expression in the magnitude of the increase relates to Doppler Lindheimer, M.D., Roberts, J.M. &
first trimester decidua: implications for abnormalities in the maternal and fetal Cunningham,G.C.), 389414 (Elsevier, 2009).
preeclampsia. Am. J. Pathol. 170, 13981405 circulation. J. Matern. Fetal Neonatal Med. 21, 193. Saftlas, A.F., Olson, D.R., Franks, A.L.,
(2007). 2540 (2008). Atrash,H.K. & Pokras, R. Epidemiology of
164. Nagamatsu, T. etal. Cytotrophoblasts up- 178. Chaiworapongsa, T. etal. A subset of patients preeclampsia and eclampsia in the United
regulate soluble fms-like tyrosine kinase1 destined to develop spontaneous preterm labor States, 19791986. Am. J. Obstet. Gynecol. 163,
expression under reduced oxygen: an implication has an abnormal angiogenic/anti-angiogenic 460465 (1990).
for the placental vascular development and the profile in maternal plasma: evidence in support 194. Zhang, J., Zeisler, J., Hatch, M.C. &
pathophysiology of preeclampsia. Endocrinology of pathophysiologic heterogeneity of preterm Berkowitz,G. Epidemiology of pregnancy-induced
145, 48384845 (2004). labor derived from a longitudinal study. J.Matern. hypertension. Epidemiol. Rev. 19, 218232
165. Kendall, R.L. & Thomas, K.A. Inhibition of Fetal Neonatal Med. 22, 11221139 (2009). (1997).
vascular endothelial cell growth factor activity by 179. Romero, R. etal. An imbalance between 195. Eskenazi, B., Fenster, L. & Sidney, S.
an endogenously encoded soluble receptor. Proc. angiogenic and anti-angiogenic factors precedes Amultivariate analysis of risk factors for
Natl Acad. Sci. USA 90, 1070510709 (1993). fetal death in a subset of patients: results of a preeclampsia. JAMA 266, 237241 (1991).
166. Esser, S. etal. Vascular endothelial growth factor longitudinal study. J. Matern. Fetal Neonatal Med. 196. Bodnar, L.M., Ness, R.B., Markovic, N. &
induces endothelial fenestrations invitro. J. Cell 23, 13841399 (2010). Roberts, J.M. The risk of preeclampsia rises
Biol. 140, 947959 (1998). 180. Chaiworapongsa, T. etal. Maternal plasma with increasing prepregnancy body mass index.
167. Sandrim, V.C. etal. Nitric oxide formation is concentrations of angiogenic/antiangiogenic Ann. Epidemiol. 15, 475482 (2005).
inversely related to serum levels of factors in the third trimester of pregnancy to 197. Branch, D.W., Silver, R.M., Blackwell, J.L.,
antiangiogenic factors soluble fms-like tyrosine identify the patient at risk for stillbirth at or near Reading, J.C. & Scott, J.R. Outcome of treated
kinase1 and soluble endogline in preeclampsia. term and severe late preeclampsia. Am. J. pregnancies in women with antiphospholipid
Hypertension 52, 402407 (2008). Obstet. Gynecol. 208, 287.e1287.e15 (2013). syndrome: an update of the Utah experience.
168. Cindrova-Davies, T., Sanders, D.A., Burton, G.J. 181. Whitten, A.E. etal. Evidence of an imbalance Obstet. Gynecol. 80, 614620 (1992).
& Charnock-Jones, D.S. Soluble FLT1 sensitizes ofangiogenic/antiangiogenic factors in 198. Lima, F. etal. A study of sixty pregnancies in
endothelial cells to inflammatory cytokines by massiveperivillous fibrin deposition (maternal patients with the antiphospholipid syndrome.
antagonizing VEGF receptor-mediated signalling. floor infarction): a placental lesion associated Clin. Exp. Rheumatol. 14, 131136 (1996).
Cardiovasc. Res. 89, 671679 (2011). with recurrent miscarriage and fetal death. Am.J. 199. Lie, R.T. etal. Fetal and maternal contributions
169. Eremina, V. etal. Glomerular-specific alterations Obstet. Gynecol. 208, 310.e1310.e11 (2013). to risk of pre-eclampsia: population based study.
of VEGFA expression lead to distinct congenital 182. Muttukrishna, S. etal. Soluble Flt1 and PlGF: BMJ 316, 13431347 (1998).
and acquired renal diseases. J. Clin. Invest. 111, new markers of early pregnancy loss? PLoS ONE 200. Schieve, L.A., Handler, A., Hershow, R., Persky, V.
707716 (2003). 6, e18041 (2011). & Davis, F. Urinary tract infection during
170. Craici, I.M. etal. Podocyturia predates 183. Signore, C. etal. Circulating angiogenic factors pregnancy: its association with maternal
proteinuria and clinical features of preeclampsia: and placental abruption. Obstet. Gynecol. 108, morbidity and perinatal outcome. Am. J. Public
longitudinal prospective study. Hypertension 61, 338344 (2006). Health 84, 405410 (1994).
12891296 (2013). 184. Koga, K. etal. Elevated serum soluble fms-like 201. Lisonkova, S. & Joseph, K.S. Incidence of
171. Chen, G. etal. Effects of angiogenic factors, tyrosine kinase 1 (sFlt1) level in women with preeclampsia: risk factors and outcomes
antagonists, and podocyte injury on hydatidiform mole. Fertil. Steril. 94, 305308 associated with early- versus late-onset disease.
development of proteinuria in preeclampsia. (2010). Am. J. Obstet. Gynecol. 209, 544.e541544.e512
Reprod. Sci. 20, 579588 (2013). 185. Bdolah, Y. etal. Circulating angiogenic proteins (2013).
172. Venkatesha, S. etal. Soluble endoglin in trisomy 13. Am. J. Obstet. Gynecol. 194,
contributes to the pathogenesis of 239245 (2006).
Acknowledgements
preeclampsia. Nat. Med. 12, 642649 (2006). 186. Kusanovic, J.P. etal. Twintotwin transfusion
The authors research is supported partly by the
173. McAllister, K.A. etal. Endoglin, a TGF- binding syndrome: an antiangiogenic state? Am. J.
Perinatology Research Branch, Division of
protein of endothelial cells, is the gene for Obstet. Gynecol. 198, 382.e1382.e8 (2008).
IntramuralResearch, Eunice Kennedy Shriver
hereditary haemorrhagic telangiectasia type1. 187. Romero, R. The child is the father of the man.
NationalInstitute of Child Health and Human
Nat. Genet. 8, 345351 (1994). Prenat. Neonat. Med. 1, 811 (1996).
Development, NIH, Department of Health and Human
174. Li, D.Y. etal. Defective angiogenesis in mice 188. Romero, R. Prenatal medicine: the child is the
Services (NICHD/NIH/DHHS) and partly with Federal
lacking endoglin. Science 284, 15341537 father of the man. 1996.J. Matern. Fetal
funding from the NICHD and NIH under contract
(1999). Neonatal Med. 22, 636639 (2009).
no.HHSN275201300006C.
175. Reimer, T. etal. Angiogenic factors and 189. Fraser, S.H. & Tudehope, D.I. Neonatal
acute-phase proteins in serum samples of neutropenia and thrombocytopenia following
preeclampsia and HELLP patients: a matched- maternal hypertension. J. Paediatr. Child Health Author contributions
pair analysis. J. Matern. Fetal Neonatal Med. 26, 32, 3134 (1996). All authors researched data for the article, made
263269 (2013). 190. Sarhanis, P. & Pugh, D.H. Resolution of pre- substantial contributions to discussion of the
176. Young, B. etal. The use of angiogenic eclampsia following intrauterine death of one content, wrote, reviewed and edited the manuscript
biomarkers to differentiate non-HELLP related twin. Br. J. Obstet. Gynaecol. 99, 159160 (1992). before submission.

NATURE REVIEWS | NEPHROLOGY ADVANCE ONLINE PUBLICATION | 15


2014 Macmillan Publishers Limited. All rights reserved

You might also like