Uploaded by User89395

Patofisiologi preeklampsia - AHA

advertisement
Compendium on the Pathophysiology and
Treatment of Hypertension
Preeclampsia
Pathophysiology, Challenges, and Perspectives
Sarosh Rana, Elizabeth Lemoine, Joey P. Granger, S. Ananth Karumanchi
Abstract: Hypertensive disorders of pregnancy—chronic hypertension, gestational hypertension, and preeclampsia—
are uniquely challenging as the pathology and its therapeutic management simultaneously affect mother and
fetus, sometimes putting their well-being at odds with each other. Preeclampsia, in particular, is one of the most
feared complications of pregnancy. Often presenting as new-onset hypertension and proteinuria during the third
trimester, preeclampsia can progress rapidly to serious complications, including death of both mother and fetus.
While the cause of preeclampsia is still debated, clinical and pathological studies suggest that the placenta is central
to the pathogenesis of this syndrome. In this review, we will discuss the current evidence for the role of abnormal
placentation and the role of placental factors such as the antiangiogenic factor, sFLT1 (soluble fms-like tyrosine
kinase 1) in the pathogenesis of the maternal syndrome of preeclampsia. We will discuss angiogenic biomarker
assays for disease-risk stratification and for the development of therapeutic strategies targeting the angiogenic
pathway. Finally, we will review the substantial long-term cardiovascular and metabolic risks to mothers and
children associated with gestational hypertensive disorders, in particular, preterm preeclampsia, and the need for
an increased focus on interventional studies during the asymptomatic phase to delay the onset of cardiovascular
disease in women. (Circ Res. 2019;124:1094-1112. DOI: 10.1161/CIRCRESAHA.118.313276.)
Key Words: biomarkers ◼ blood pressure ◼ cardiovascular disease ◼ preeclampsia ◼ pregnancy
Downloaded from http://ahajournals.org by on February 7, 2021
H
ypertensive disorders are a common complication of
pregnancy that put women and their fetuses at disproportionate risk for further complications, as well as life-long
sequelae. Ranging in severity, hypertensive disorders of pregnancy include chronic hypertension—systolic blood pressure
(BP) ≥140 mm Hg or diastolic BP ≥90 mm Hg that predates
the onset of pregnancy; gestational hypertension—hypertension diagnosed after 20 weeks gestation without concurrent
proteinuria; preeclampsia-eclampsia—classically, new-onset
hypertension with new-onset proteinuria; and chronic hypertension with superimposed preeclampsia—chronic hypertension with new-onset proteinuria or other signs/symptoms of
preeclampsia after 20 weeks or chronic proteinuria with newonset hypertension.1 With the greatest morbidity and mortality,
preeclampsia affects 5% to 7% of all pregnant women but is
responsible for over 70 000 maternal deaths and 500 000 fetal
deaths worldwide every year. In the United States, it is a leading
cause of maternal death, severe maternal morbidity, maternal
intensive care admissions, cesarean section, and prematurity.2–4
Delivery can resolve most signs and symptoms; however,
preeclampsia can persist after delivery and, in some cases,
can develop de novo in the postpartum period.1,5 De novo
or persistent postpartum preeclampsia has emerged as an
important risk factor for peripartum morbidity in the United
States.6 Hypertensive disorders of pregnancy and in particular preterm preeclampsia is also associated with substantial
risk for cardiovascular disease (CVD) and cerebrovascular
disease in the long-term.7,8
Epidemiology and Clinical Definition
of Preeclampsia
Risk Factors for Preeclampsia
Risk factors for the development of preeclampsia have been
studied extensively (Table 1). Major risk factors include a
history of preeclampsia, chronic hypertension, pregestational diabetes mellitus, antiphospholipid syndrome, and obesity, among others.9 Other risk factors include advanced
maternal age, nulliparity, history of chronic kidney disease,
and use of assisted reproductive technologies. Relatively
rare risk factors are a family history of preeclampsia and
mother carrying a trisomy 13 fetus.10,11 Genetic susceptibility to preeclampsia has been extensively studied.12,13 A 2017
genome-wide association study analysis of neonates from
4380 cases of preeclampsia and 310 238 controls found a
genome-wide susceptibility locus (rs4769613; P=5.4×10−11)
near the FLT1 (FMS-like tyrosine kinase 1) gene, the protein
From the Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, University of Chicago, IL (S.R.); Harvard Medical School,
Boston, MA (E.L.); Department of Physiology, University of Mississippi Medical Center, Jackson (J.P.G.); Departments of Medicine, Obstetrics and
Gynecology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA (S.A.K.); and Department of Medicine, Cedars-Sinai
Medical Center, Los Angeles, CA (E.L., S.A.K.).
Correspondence to S. Ananth Karumanchi, MD, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90004. Email sananth.
[email protected]
© 2019 American Heart Association, Inc.
Circulation Research is available at https://www.ahajournals.org/journal/res
1094
DOI: 10.1161/CIRCRESAHA.118.313276
Rana et al Preeclampsia and Vascular Disease 1095
Nonstandard Abbreviations and Acronyms
ACOG
Arrb1
ASPRE
Downloaded from http://ahajournals.org by on February 7, 2021
American College of Obstetrics and Gynecology
β-arrestin-1
aspirin for evidence-based preeclampsia prevention trial
AT1-AA
Angiotensin II type 1 receptor autoantibodies
AT1-B2
Angiotensin II type 1 receptor and Bradykinin B2
receptor complex
BP
blood pressure
COMT
catechol-O-methyl transferase
CVD
cardiovascular disease
DV
decidual vasculopathy
ER
endoplasmic reticulum
ET-1
endothelin-1
FLT1
fms-like tyrosine kinase
HELLP syndrome
Hemolysis Elevated Liver enzymes Low Platelets
syndrome
HIF
hypoxia-inducible factors
HO
heme oxygenase
hsFLT1
human soluble FMS-like tyrosine kinase 1
KIR
killer cell Ig-like receptors
IL
interleukin
MHC
major histocompatibility complex
PAPP-A
pregnancy-associated plasma protein A
PGC-1a
proangiogenic transcriptional cofactor- 1alpha
PlGF
placental growth factor
PPCM
peripartum cardiomyopathy
PPV
positive predictive value
RNAi
RNA interference technology
ROS
reactive oxygen species
RR
relative risk
RUPP
reduction in uterine perfusion pressure rodent model
sENG
soluble endoglin
sFLT1
soluble fms-like tyrosine kinase 1
SGA
small for gestational age
StAmP
Statins to ameliorate early onset preeclampsia trial
STRIDER
Sildenafil for severe fetal growth restriction trial
TF
transcription factor
TGF-β1
transforming growth factor β1
Th1/Th2
T helper type 1/type 2 cells
TNF
Tumor necrosis factor
uNK
uterine natural killer cell
UPR
unfolded protein response
VEGF
vascular endothelial growth factor
Table 1.
Risk Factors for Preeclampsia
Major Risk Factors
Prior preeclampsia (RR, 8.4; 95% CI, 7.1–9.9)
Chronic hypertension (RR, 5.1; 95% CI, 4.0–6.5)
Pregestational diabetes mellitus (RR, 3.7; 95% CI, 3.1–4.3)
Multiple gestation (RR, 2.9; 95% CI, 2.6–3.1)
Prepregnancy BMI >30 (RR, 2.8; 95% CI, 2.6–3.1)
Antiphospholipid syndrome (RR, 2.8; 95% CI, 1.8–4.3)
Other risk factors
Systemic lupus erythematosus (RR, 2.5; 95% CI, 1.0–6.3)
History of stillbirth (RR, 2.4; 95% CI, 1.7–3.4)
Prepregnancy BMI >25 (RR, 2.1; 95% CI, 2.0–2.2)
Nulliparity (RR, 2.1; 95% CI, 1.9–2.4)
Prior placental abruption (RR, 2.0; 95% CI, 1.4–2.7)
Assisted reproductive technology (RR, 1.8; 95% CI, 1.6–2.1)
Chronic kidney disease (RR, 1.8; 95% CI, 1.5–2.1)
Advanced maternal age >35 (RR, 1.2; 95% CI, 1.1–1.3)
Genetic susceptibility (mother, father)
Rare risk factors
Family history of preeclampsia
Trisomy 13 fetus
Relative Risk provided from meta-analyses by Ray et al.9 BMI indicates body
mass index; and RR, relative risk.
Clinical Definition of Preeclampsia
Classically, the American College of Obstetrics and
Gynecology (ACOG) defines preeclampsia as the presence
of hypertension and proteinuria occurring after 20 weeks of
gestation in a previously normotensive patient. However, a
significant proportion of women develop systemic manifestations of preeclampsia—such as low platelets or elevated liver
enzymes—before the hallmark of proteinuria is detectable,16,17
resulting in delayed diagnoses. The evolving understanding
of preeclampsia as a heterogeneous hypertensive disorder of
pregnancy led to ACOG’s hypertension 2013 task force to revise the definition of preeclampsia to include the presence of
severe features with or without proteinuria and to exclude degree of proteinuria as a criterion of severe features (Table 2).1
These criteria were confirmed more recently in an update of
the ACOG’s practice guidelines.18
Pathogenesis of Preeclampsia
product of which is a well-established pathogenetic factor in preeclampsia.14 rs4769613 has a higher frequency in
late-onset preeclampsia, exerts effects only in the fetal—not
maternal—genome, and has no difference in transmission
of disease association with parental sex inheritance, making the effects of imprinting unlikely. However, maternal
genetic susceptibility may play a role as well. A multiethnic maternal preeclampsia genome-wide association study
discovered a genome-wide susceptibility locus at rs9478812
(P=5.90×10−7), an intronic region of protein PLEKHGI implicated in BP regulation.15
A placental disease, preeclampsia progresses in 2 stages: (1) abnormal placentation early in the first trimester followed by (2) a
“maternal syndrome in the later second and third trimesters characterized by an excess of antiangiogenic factors19,20 (Figure 1).
While the mechanism of abnormal placentation is controversial,
animal models have demonstrated that uteroplacental ischemia
drives the hypertensive, multi-organ failure response observed
in the maternal preeclamptic syndrome21 (stage 2). A number of
theories have been proposed for the placental dysfunction observed in stage 1, including oxidative stress, abnormal natural
killer cells (NKs) at the maternal-fetal interface, and genetic and
1096 Circulation Research March 29, 2019
Table 2.
Clinical Definition of Preeclampsia1
Preeclampsia
Elevated blood pressure
Systolic ≥140 mm Hg or diastolic ≥90 mm Hg, 2 occasions, 4 h apart
in previously normotensive woman
AND Proteinuria
≥300 mg/24 hour urine collection
or protein/creatinine ≥0.3
or dipstick reading =1+
OR severe features*
Severe features
Systolic blood pressure ≥160 mm Hg or diastolic ≥110 mm Hg, 2
occasions, 4 h apart on bedrest
Thrombocytopenia (<100 000 μL)
Liver function tests 2× normal or severe persistent right upper quadrant
or epigastric pain
Serum creatinine concentration >1.1 mg/dL or doubling of creatinine in
the absence of other renal disease
Pulmonary edema
New-onset cerebral or visual symptoms
Downloaded from http://ahajournals.org by on February 7, 2021
environmental factors, though none have conclusive evidence in
humans. However, substantive evidence supports the idea that
the diseased placenta leads to release of soluble toxic factors in
the maternal circulation that result in inflammation, endothelial
dysfunction, and maternal systemic disease.19,20,22
Stage 1: Abnormal Placentation, Trophoblast
Invasion, and the Maternal-Fetal Interface
During normal placental implantation, cytotrophoblasts migrate into the maternal uterine spiral arteries, forming vascular
sinuses at the fetal-maternal interface to provide nutrition to
the fetus. In normal pregnancy, this invasion progresses deeply
into the spiral artery to the level of the myometrium,23,24 which
leads to extensive remodeling of the maternal spiral arterioles
into high capacitance, high flow vessels.23 In placentas destined
to develop preeclampsia, cytotrophoblasts fail to transform
from the proliferative epithelial subtype to the invasive endothelial subtype which causes incomplete remodeling of the
spiral artery.25 Inadequate spiral arteriolar remodeling leads to
narrow maternal vessels, and relative placental ischemia.26 The
narrow spiral arteries are prone to atherosis—characterized by
the presence of lipid-laden macrophages within the lumen, fibrinoid necrosis of the arterial wall, and a mononuclear perivascular infiltrate,27 leading to further compromise in placental
flow. In humans, placental ischemia can be noninvasively identified using uterine artery Doppler studies. During normal
pregnancy, uterine artery Doppler studies have confirmed robust systolic and diastolic uterine arterial flows; in contrast,
women with preeclampsia have significant impairment of diastolic flow with a characteristic notch in the waveform that
antedates clinical signs and symptoms of preeclampsia.28,29
These findings suggest that an abnormality in the trophoblasts
themselves may result in shallow placentation and inadequate
transformation of the spiral arteries, leading to placental ischemia and the maternal syndrome of preeclampsia.25
Atherosclerotic changes in maternal radial arteries that
supply the decidua—as opposed to the spiral arteries—are also
observed in preeclampsia.30,31 Decidual vasculopathy (DV) is a
lesion common to disorders of placental insufficiency, including intrauterine growth restriction and preeclampsia, and combines (1) acute atherotic lesions with (2) medial hypertrophy
and perivascular lymphocytes (Figure 2). Within preeclampsia
phenotypes, the presence of DV is associated with worse clinical outcome, higher diastolic BP, worse renal function, and
perinatal fetal death.32 Histologically, normal third-trimester
decidual vessels are characterized by flat endothelium and a
loss of medial smooth muscle, while preeclamptic decidua
show signs of loose, edematous endothelium, hypertrophy of
the vessel media, and loss of smooth muscle modifications (as
seen in atherosclerosis), characterizing DV.30 Correlated with
clinical diagnosis, DV has the highest association with preeclampsia with small for gestational age (SGA) and a lesser but
significant association with SGA with doppler abnormalities,
suggesting a pathogenetic similarity between SGA with doppler abnormalities and preeclampsia with SGA at the level of
the decidua.30 Overall, there is significant evidence that decidual vessels demonstrate secondary atherosclerotic changes in
preeclampsia. Further studies are needed to determine if these
changes are representative of the maternal systemic endothelial damage secondary to pathological changes such as hypertension or if DV contributes to the pathogenesis of stage 1.30
In addition to uteroplacental insufficiency, epidemiological
studies suggest that poor uterine decidualization—the stromal
transformation of uterine endometrium to prepare for implantation—may affect the development of preeclampsia. Global
transcriptional profiling of chorionic villus samples points to insufficient or defective decidualization in pregnancies that were
later complicated by severe preeclampsia.33 Endometrial stromal
cells from nonpregnant donors with a history of severe preeclampsia fail to decidualize in vitro and are transcriptionally inert, suggesting baseline genetic abnormalities or genetic modifications.34
Finally, global transcriptional profiling of decidual tissue from
the women with preeclampsia also revealed defects in gene expression. These cells failed to redecidualize in culture, and their
conditioned medium failed to support cytotrophoblast invasion,
suggesting that decidual cells may be an important contributor
to downregulated cytotrophoblast invasion in preeclampsia.34
The in vitro studies are confirmed ex vivo in histological studies of preeclampsia showing shallow placentation.31 Given the
mounting evidence of fetal and maternal abnormalities in preeclampsia, defective placentation might be the result of combinations of factors that affect both trophoblast and decidua.35,36
Hypoxia and Trophoblast Invasion
Upregulation of hypoxia-inducible transcription factors (TFs)
and hypoxia-related gene signatures in the placenta suggest
that hypoxia is central to the pathogenesis of preeclampsia.37
In the early phases of implantation, the gestational sac exists
in a low oxygen tension environment, favoring trophoblast
proliferation. Before the invasion, the proliferating trophoblasts anchor the blastocyst to maternal tissues and plug the
tips of the spiral arteries within the decidua.38 Eventually these
Rana et al Preeclampsia and Vascular Disease 1097
Figure 1. Schematic of the pathogenesis of preeclampsia. Genetic factors, immunologic factors, other maternal factors cause placental dysfunction which
in turn leads to the release of antiangiogenic factors (such as sFLT1 [soluble fms-like tyrosine kinase 1] and sENG [soluble endoglin]) and other inflammatory
mediators to induce preeclampsia.
Downloaded from http://ahajournals.org by on February 7, 2021
trophoblastic-spiral artery plugs collapse, forming an intervillous space. The newly formed sinuses allow for the arrival of
maternal blood, increasing oxygen tension, generating oxidative stress, and promoting trophoblast differentiation from
a proliferative to an invasive phenotype that will invade and
remodel the spiral arteries.39 HIF (Hypoxia-inducible factors)1α and -2α, markers of cellular oxygen deprivation, are expressed at high levels in proliferative trophoblasts and in the
placentas of women with preeclampsia.40 Overexpression
of HIF-1α in pregnant mice is associated with hypertension,
proteinuria, and fetal growth restriction in mice41 and may
result in failure of trophoblastic differentiation from the proliferative to the invasive phenotype.42 Furthermore, inhibition
of HIF-1α by 2-methoxyestradiol, a metabolite of estradiol
that destabilizes HIF-1α, suppresses the production of sFLT1
(soluble fms-like tyrosine kinase 1), a potent antiangiogenic
factor known to contribute to the maternal syndrome.43 HIF1α expression is regulated by many factors in addition to hypoxia, therefore, isolating the dysregulated signal upstream is
challenging.23,44 Magnetic resonance imaging has been used
to assess the placental perfusion fraction—an estimate of the
fraction of perfused tissue by volume—as a marker of uterine
flow or placental function.45–47 In a Swedish study of 35 women
with singleton pregnancies (13 with preeclampsia), Sohlberg
et al47 found a smaller placental perfusion fraction associated
with fetal growth restriction, as well as abnormalities in maternal and fetal vessel doppler flow, neonatal weight, and plasma
markers, including higher levels of sFLT1. Novel blood oxygen level-dependent magnetic resonance imaging responses
promise new, noninvasive, in vivo techniques for assessing
maternal-fetal interface oxygen tension48,49 and may be useful
for mapping areas of placental pathology and insufficiency.50
Oxidative Stress
While low oxygen tension followed by maternal blood flow
oxygenation results in normal placentation, intermittent hypoxia and reoxygenation caused by poor spiral artery invasion
may cause oxidative stress. At the molecular level, preeclamptic placentas show an imbalance of reactive oxygen species
Figure 2. Decidual vasculopathy. Placental bed of the uterus with
decidual vasculopathy in the third trimester. Vessels show chronic injury
with endothelial fragmentation and detachment (arrow) and fibrinoid
necrosis (**) of the vessel wall. Scale bar is 100 μm. Reprinted from Hecht
et al30 with permission. Copyright ©2016, Elsevier.
1098 Circulation Research March 29, 2019
Downloaded from http://ahajournals.org by on February 7, 2021
(ROS)-generating enzymes and antioxidants. In the ex vivo
preeclamptic trophoblast, ROS-producing enzyme expression and activity are increased51 and inhibit the Wnt/β-catenin
signaling pathway that promotes trophoblast invasiveness.52
Oxidative stress may also promote the transcription of antiangiogenic factors such as sFLT1.53 In humans, placental
antioxidant mechanisms are impaired in patients with preeclampsia, as shown by their decreased expression of superoxide dismutase and glutathione peroxidase compared with
women with normal pregnancies.54 However, treatment with
the antioxidants Vitamin E and Vitamin C did not alter disease
in women with preeclampsia, suggesting that ROS may be
less integral to the pathway of the human syndrome.55,56
ROS may derive from mitochondrial stress. Zsengellér et
al57 demonstrated decreased activity of the mitochondrial electron transport chain (ETC) enzyme cytochrome C oxidase in
the syncytiotrophoblast cells of preeclamptic placentas, which
correlated with increased placental sFLT1 expression. Based
on evidence that hydrogen sulfide donors inhibit HIF-1α,58
Covarrubias et al59 demonstrated that AP39, a mitochondrialtargeting hydrogen sulfide donor, pretreatment could decrease
sFLT1 expression in human syncytiotrophoblasts and increase
cytochrome C oxidase activity in a dose-dependent fashion in
normal and preeclamptic placentas, preventing the release of
ROS and the subsequent stabilization of HIF-1α.60 Recently
published studies with mitochondrial antioxidants in animal
models of preeclampsia have also been promising.61
Another possible source of oxidative stress is endoplasmic
reticulum stress caused by ischemia-reperfusion injury.62,63
Endoplasmic reticulum stress has been observed in the decidua and placentas of patients with fetal growth restriction
and preeclampsia and triggers decidual cell and cytotrophoblast apoptosis through the activation of the UPR (unfolded
protein response).64,65 PERK (PKR-like endoplasmic reticulm
kinase), a transmembrane kinase that decreases the translational burden of the endoplasmic reticulum and upregulates
proapoptotic TFs, has emerged as the leading signaling pathway implicated in preeclampsia.64,65 Interestingly, a recent
study suggests that synergy between ATF4 (activating transcription factor 4), a TF downstream of PERK, and ATF6, a
TF regulator of misfolded proteins in endoplasmic reticulum
homeostasis,65,66 negatively regulate the transcription of PlGF
(placental growth factor), an proangiogenic factor central to
the pathogenesis of preeclampsia.67
Heme Oxygenase and Other Enzyme Abnormalities
There is growing evidence that heme oxygenase (HO), the
heme degradation catalyst, has an important role in the vascular function of the mother and the fetus, as well as in placental development and function.68–70 Three isoforms of HO
have been characterized,68,71 with HO-2 playing a role in spiral artery invasion72 and HO-1 highly expressed in noninvasive
trophoblastic phenotypes.73 Treatment of the reduced uterine
perfusion pressure (RUPP) rodent model with CoPP (cobalt
protoporphyrin), an inducer of HO-1, decreased BP and resulted in a proangiogenic shift in the VEGF (vascular endothelial
growth factor)/sFLT1 ratio in the placenta.74 These preclinical
studies have fueled interest in manipulating the expression of
HO-1 as a potential therapeutic intervention for preeclampsia.
Another influence on trophoblastic invasion and spiral artery remodeling may come from corin, a transmembrane enzyme that locally activates atrial natriuretic peptide through
zymogen modification. Corin acts primarily in heart tissue,
however, Cui et al75 found significantly decreased uterine-localized corin mRNA and protein levels as well as several corin
gene mutations in preeclamptic patients. The group then created a knockout corin rodent model as well as transgenic crosses
that retained isolated cardiac corin activity. Both phenotypes
mimicked the hallmarks of preeclampsia, independently of
preexisting hypertension or cardiac-derived atrial natriuretic
peptide. However, in human studies, the evidence is mixed,
as systemic levels of corin and its target—atrial natriuretic
peptide—are upregulated during preeclampsia76,77 not downregulated as would be expected based on the animal studies.
In earlier studies, women with hypertensive disorders of
pregnancy were found to have lower levels of placental catechol-O-methyl transferase (COMT) enzyme,78 while women
with normal gestations were found to have increasing concentrations of 2-methoxyestradiol, the COMT breakdown product of estradiol.79 Based on these observations, Kanasaki et
al43 characterized a COMT knockout rodent model that reproduced the hallmarks of preeclampsia. However, COMT alterations are not a feature of severe early-onset preeclampsia in
humans.80 Given that COMT is decreased in many hypertensive disorders of pregnancy,78 more evidence is required to implicate COMT in the pathogenesis of preeclampsia rather than
a risk factor for all gestational hypertensive disorders.
NK Cells and Impaired Placentation
The uterine NK (uNK) is well characterized in decidualization
physiology81 and may play a role in the abnormal placentation observed in preeclampsia. Unlike peripheral NKs, uNK
is not cytotoxic.82,83 Rather, in the decidua, uNK cells regulate the depth of placentation, spiral artery remodeling, and
trophoblastic invasion.84,85 As the main immunologic player
interacting at the allogenic maternal-fetal cell interface,81
uNKs recognize self-major histocompatibility complexes
(MHCs) derived from the maternal contribution and nonselfallogenic MHCs from the paternal genotype. Specifically,
uNK express KIR (killer cell Ig-like receptors),86 while fetal invasive extravillous trophoblasts express the main KIR
ligand, polymorphic HLA-C (human leukocyte antigen-C)
MHCs.87 Because of independent segregation of maternal
KIR and HLA loci86 and the paternal contribution to extravillous trophoblast HLA-C, every pregnancy results in a unique
combination of KIR (maternal) and HLA-C (fetal) which may
affect the success of placentation.83 Mouse models selected
for nonmatched maternal uNK and paternal MHC molecules
in otherwise genetically identical parents have demonstrated
that allogenicity may promote decidual artery dilation, spiral
artery remodeling, more efficient placentas, and larger fetal
weights.88 In other words, inhibition of the uNK response by
MHC-self recognition may lead to defective artery remodeling.89 Furthermore, certain maternal KIR haplotypes (uNK)
appear protective against preeclampsia while others confer
risk.83,90–92 However, the presence of the risk-associated haplotype is insufficient for disease, suggesting an additional environmental or genetic hit.
Rana et al Preeclampsia and Vascular Disease 1099
Stage 2: Pathogenesis of the Maternal Syndrome
Downloaded from http://ahajournals.org by on February 7, 2021
Imbalance in Circulating Angiogenic Factors
More than a decade ago, several groups identified elevated
levels of the antiangiogenic protein sFLT1 in placentas collected from women with a clinical diagnosis of preeclampsia.93,94 sFLT1 is a soluble protein that exerts antiangiogenic
effects by binding to and inhibiting the biological activity of
proangiogenic proteins VEGF and PlGF95 (Figure 3). VEGF
is important for the maintenance of endothelial cell function,
especially in fenestrated endothelium, which is found in the
brain, liver, and glomeruli, the primary organs affected by preeclampsia.96 A member of the VEGF family, PlGF is important in angiogenesis and selectively binds to VEGFR1/sFLT1
not VEGFR2.97 Several findings implicated sFLT1 in the pathogenesis of preeclampsia: sFLT1 protein levels were high in
maternal plasma or serum94,98; sFLT1 mRNA expression was
high in preeclamptic placentas99; and injecting exogenous
sFLT1 into rodents led to hypertension, proteinuria, glomerular endotheliosis (a hallmark of preeclampsia seen in renal
biopsy), as well as several other preeclamptic features94,100;
treatment of cancer patients with anti-VEGF drugs results in
hypertension and proteinuria101,102; depletion of sFLT1 in preeclamptic plasma using antibodies reverses the antiangiogenic
phenotype in cell culture studies93; lowering sFLT1 or antagonizing sFLT1 in animal models of preeclampsia improves
clinical symptoms103–105 and, spontaneous resolution of clinical signs and symptoms of preeclampsia, when sFLT1 levels
are lowered by 50% or more by treatment of the underlying
placental conditions such as fetal hydrops or removal of diseased placenta in multiple pregnancies.106,107 In addition to the
elevated sFLT1 levels, circulating levels of free PlGF were reduced in women with preeclampsia, suggesting an imbalance
of antiangiogenic and proangiogenic proteins.94,108
The availability of robust immunoassays for angiogenic
factors led to a number of clinical studies measuring the antiangiogenic/proangiogenic markers in large cohorts of human
pregnancies showing that sFLT1 levels are high and free PlGF
is low at the time of clinical diagnosis of preeclampsia as well
as several weeks before the diagnosis.108,109 Angiogenic factor
abnormalities in the plasma correlated with severity of presentation, predicting disease, and adverse outcomes.110,111 Early
studies called into question the utility of angiogenic factors
in prediction of preeclampsia.112 However, a recent multisite,
blinded randomized trial for the use of aspirin for prevention
of preeclampsia used several physiological and biochemical
parameters, including PlGF, with a detection rate of 90% at
a fixed false positive rate of 5% for preeclampsia, suggesting
that the markers may be used algorithmically for early diagnosis.113 Such a strategy for prediction of the syndrome in early
pregnancy would identify women who are at risk for developing the disease and who may benefit from preventative interventions such as aspirin. Early diagnosis would also reduce
anxiety and unnecessary interventions in women at otherwise
low risk of developing preeclampsia.
Another antiangiogenic protein that has also been extensively studied in preeclampsia is soluble endoglin (sENG), an
endogenous TGF-β1 (transforming growth factor β1) inhibitor114 (Figure 3). sENG is elevated in the sera of preeclamptic
women 2 months before the onset of clinical signs of preeclampsia, correlates with disease severity, and falls after
delivery.109,115 In pregnant rats, it appears to potentiate the
vascular effects of sFLT1 to induce a severe preeclampsialike state, including the development of thrombocytopenia
and fetal growth restriction,116,117 and, in combination with
sFLT1, appears to induce cerebral edema resembling the reversible posterior leukoencephalopathy seen in patients with
eclampsia.118,119
Inflammatory Cytokines and Immune Cell Alterations
It is well-established that preeclampsia is a proinflammatory
state, but the culpable cells have yet to be fully elucidated.
Syncytial knots120,121 are allogenic nano to microvesicles shed
from apoptotic or activated trophoblasts121 that have been
identified in the lungs120,122 and plasma of normal pregnancies and in increased amounts in preeclampsia.122–124 Rich in
sFLT1 and endoglin,125,126 syncytiotrophoblast microvesicles
and exosomes may instigate an inflammatory response. In
vitro, syncytiotrophoblast microvesicles activate cultured peripheral blood mononuclear cells, causing a release of proinflammatory cytokines124,127 that is even more robust when
exposed to peripheral blood mononuclear cells from pregnant patients.127 However, in vitro data are not consistent, as
microvesicles induced by an alternative mechanism are not
proinflammatory.128,129
IL (Interleukin)-10—a cytokine that induces the differentiation of the T cell into the Th (T helper type)2 phenotype—
stands out in the literature as an important mitigator of the
maternal syndrome by neutralizing proinflammatory cytokines, AT1-AA (angiotensin II receptor 1 autoantibodies),
placental ROS, and ET-1 (endothelin-1).130 Many cell types
in preeclamptic patients demonstrate a dysregulation in the
balance of IL-10 and proinflammatory cytokines,131–133 including uterine and circulating NKs134 and peripheral blood mononuclear cells. Studies of peripheral blood mononuclear cells
of preeclamptic women had reduced IL-10 secretion,135–138
which may lead to failure of T-cell differentiation. Commonly
referred to as Th2 polarization, normal pregnancy is characterized by a shift in T-cell phenotype towards Th2 relative to
Th1.139,140 Multiple studies have reported an aberrant shift towards the Th1 phenotype in preeclampsia, resulting in insufficient trophoblast invasion.141 Furthermore, a preeclamptic-like
syndrome can be induced in normal pregnant rats with transfer
of CD4+ cells obtained from RUPP models.122
Preeclampsia is also associated with elevated complement levels142,143 and with genetic mutations in C3.144 In animal models, complement inhibition restores spiral artery
capacitance145 and decreases sFLT1 production,146 and a C1q
knockout mouse model mimics preeclamptic features.147,148
However, complement dysregulation is most severe in the
form of severe preeclampsia called hemolysis elevated liver
enzymes low platelets (HELLP) syndrome. HELLP syndrome
has been shown to share a genetic mutation with149 and has a
similar presentation to atypical hemolytic uremic syndrome,
a disease thought to be caused by uncontrolled complement
activation.150–152 Interestingly, many of the same complement
pathway mutations found in hemolytic uremic syndrome are
also associated with preeclampsia.142,150 Further evidence to
1100 Circulation Research March 29, 2019
Figure 3. sFLT1 (soluble fms-like tyrosine kinase 1) and sENG (soluble endoglin) causes endothelial dysfunction by antagonizing VEGF (vascular
endothelial growth factor) and TGF (transforming growth factor)-β1 signaling. There is mounting evidence that VEGF and TGF-β1 are required to
maintain endothelial health in several tissues including the kidney and perhaps the placenta. During normal pregnancy, vascular homeostasis is maintained
by physiological levels of VEGF and TGF-β1 signaling in the vasculature. In preeclampsia, excess placental secretion of sFLT1 and sENG (2 endogenous
circulating antiangiogenic proteins) inhibits VEGF and TGF-β1 signaling respectively in the vasculature. This results in endothelial cell dysfunction, including
decreased prostacyclin, nitric oxide production, and release of procoagulant proteins. Reprinted from Powe et al114 with permission. Copyright ©2011, the
American Heart Association.
Downloaded from http://ahajournals.org by on February 7, 2021
the pathogenic link between atypical hemolytic uremic syndrome and HELLP, a patient presenting with early-onset severe preeclampsia and HELLP was able to delay delivery by
17 days after treatment with Eculizumab,153 an Food and Drug
Administration-approved C5 inhibitor used to treat atypical
hemolytic uremic syndrome 154,155 including in pregnancy.156
The use of Eculizumab as an effective treatment for a severe
form of preeclampsia is promising, however, HELLP is diagnostically difficult to distinguish from atypical hemolytic
uremic syndrome,157 likely because of the overlap in complement pathology.
Renin-Angiotensin Pathway
There is evidence for alterations in the renin-angiotensinaldosterone system in the pathogenesis of preeclampsia.158
Several studies show enhanced angiotensin II sensitivity during and before the onset of preeclampsia despite reduced circulating renin and angiotensin II during preeclampsia when
compared with normal pregnancy.159,160 One potential mechanism for the increased angiotensin II sensitivity is the presence
of circulating autoantibodies to AT1 in the sera of preeclamptic women.161,162 In preclinical studies, autoantibodies to AT1
reproduce many of the hallmark characteristics of preeclampsia: vasoconstriction through activation of ET-1163; endothelial
cell necrosis and apoptosis in human umbilical vein endothelial cells164; stimulation of tissue factor production contributing to hypercoagulation165; reduction of trophoblast invasion
in human cell culture models166; and increased production of
ROS in culture models.167 Produced in response to placental ischemia and systemic inflammation,168 anti-AT1-AA can
also stimulate placental production of antiangiogenic factors
sFLT1 and sENG.169 Finally, CD19+CD5+ cells, as well as
anti-AT1-AA activity, are elevated in the sera of preeclamptic
patients, implicating B lymphocytes as an immune player.170
These findings suggest that anti-AT1-AA made by a subpopulation of CD19+CD5+ in response to placental ischemia and
systemic inflammation may contribute to the hypertension and
production of antiangiogenic factors that characterize the maternal syndrome.
Recent preclinical studies on the hypersensitivity of the
AT1 receptor when complexed with the bradykinin B2 receptor provide compelling evidence for another model for
the activation of the renin-angiotensin-aldosterone system in
the setting of downregulated renin.171 Using a new transgenic
mouse model with maternal, systemically upregulated smooth
muscle AT1-B2 complexes, the group was able to replicate
the preeclampsia syndrome, with pregnant animals developing hypertension, proteinuria, low platelets, increased sFLT1,
AT1-AA, and ET-1, smaller litter sizes, intrauterine growth
restriction, lower renin levels, and a decreased placental labyrinth layer. Furthermore, when heteromerized with B2, ATI
seems independently sensitive to angiotensin II and mechanostimulation, which, the authors suggest, may evolve with an
increase in fetal-placental mass regardless of renin activity.
The preeclamptic-like transgenic mice were then rescued with
lentiviral administration of an inactivation-resistant variant of
Arrb1 (β-arrestin-1), G-protein coupled receptor-associated
protein that desensitizes the AT1 receptor leading to signal
dampening. In ex vivo studies of human placentas, the group
found significantly elevated levels of inactivated (phosphorylated) levels of Arrb1 in preeclamptic placentas compared
with normotensive placentas, as well as increased AT1-B2
complex formation on the vessels of the basal plate of a preeclamptic placenta. Though the AT1-B2 model does appear
to replicate the maternal syndrome well, only a small number of human placentas were analyzed as part of this study.
Rana et al Preeclampsia and Vascular Disease 1101
Additional human studies are required to assess applicability
of this model to the biology of preeclampsia.
Elevated levels of an oxidized form of angiotensinogen
that is more readily cleaved by renin have also been implicated in the pathogenesis of the hypertension observed in preeclampsia.172 However, robust assays to measure this modified
form of angiotensinogen in the blood are needed to characterize a role for oxidized angiotensinogen in preeclampsia.
Finally, in animal models, elevated levels of circulating sFLT1
were sufficient to induce angiotensin II sensitivity by interfering with endothelial nitric oxide production.173
Downloaded from http://ahajournals.org by on February 7, 2021
Sympathetic Nervous System
While a major emphasis of study in the pathogenesis of preeclampsia has been on the link between placental factors and
maternal endothelial dysfunction, several studies have implicated the sympathetic nervous system in the pathogenesis
of preeclampsia.174,175 Schobel et al174 observed that muscle
sympathetic nerve activity is elevated in women with preeclampsia over normal pregnant and hypertensive, nonpregnant
control women. Women with preeclampsia also have reduced
baroreflex sensitivity and greater antihypertensive responses
to nonselective adrenergic receptor blockade.176,177 Studies
using experimental animal models supports that sympathetic
nerve activity is increased in preeclampsia. Placental ischemia-induced hypertension in the RUPP rat model is associated with a hypertensive shift in baroreceptor control on renal
sympathetic nerve activity, 178 and a recent study found that
adrenergic receptor blockade markedly attenuates placental
ischemia-induced hypertension.179 Collectively, studies from
humans and animal models suggest that an intact sympathetic
nervous system may be important in eliciting the full hypertensive response to factors released in response to placental
ischemia.
Lessons From Animal Models
One of the many challenges of the study of preeclampsia is
its reproducibility in animal models. Spontaneous preeclampsia is unique to human gestation. Therefore, animal models
approximate rather than replicate the disease, predominantly
through evidence of systolic hypertension, renal endotheliosis, proteinuria, and, at times, fetal growth restriction and production of antiangiogenic factors.180 The RUPP rodent model,
produced by clipping the abdominal aorta and the main uterine arteries of pregnant Sprague-Dawley rats,181 is most commonly used and presents with elevated mean arterial pressure,
greater vascular reactivity to α-adrenergic agonists181 and increased production of AT1-AA, ROS, and sFLT1 and sENG
as observed in preeclamptic women.182 Other animals, including pregnant rabbits, rhesus monkeys, and baboons, have also
been used to model preeclampsia by inducing uteroplacental
insufficiency.182 To replicate the placenta as the inherently
pathogenic organ rather than as secondary to uterine ischemia, Kumasawa et al104 implanted mice with transgenic blastocysts with trophectodermal layers expressing human sFLT1
via lentiviral vectors. Human sFLT1 levels increased in the
maternal serum as gestation progressed, with corresponding
development of the hallmarks of preeclampsia. However, the
rodents did not develop the full spectrum of severe features as
in humans. Genetic rodent models, such as the BPH/5 model,
have been used as well. Mildly hypertensive at baseline, the
BPH/5 model demonstrates elevated mean arterial pressure,
proteinuria, and progressive glomerular damage in late gestation and delivers significantly smaller litters than controls.183
The previously discussed COMT knockout rodent model similarly presents with systolic hypertension, elevated sFLT1, and
proteinuria that decline after parturition, however, there is no
evidence of systemic vascular damage or fetal growth restriction, suggesting that this model approximates gestational hypertension or mild preeclampsia.43 The C1q knockout mouse
model relies on the observation of the complement factor C1q
at the decidual endothelium-trophoblast interface184 and was
found to have small litters, intrauterine growth restriction, elevated BP, proteinuria, elevated levels of sFLT1, and evidence
of endothelial dysfunction.147,148 Systemic delivery of arginine vasopressin was also sufficient to induce preeclampsialike state in pregnant mice, but lack of placental hypoxia and
sFLT1 upregulation in this model suggest that it may be more
relevant for term preeclampsia that is often characterized by
modest levels of antiangiogenic biomarkers.185 Older rodent
models manipulate the renin-angiotensin system (placental
renin and maternal angiotensinogen) to model pregnancy-induced hypertension,186 however, the relevance of this model to
human disease is questionable as humans with preeclampsia
are characterized by suppressed renin and angiotensin II when
compared with normal pregnancy.
Primarily designed on the observation that placental ischemia induces systemic hypertension, the described animal
models have been useful for characterizing the molecular environment of uteroplacental hypoperfusion as well as a variety
of cytokines and proteins released into the maternal circulation. Animal models can be used to extend correlation data
observed in humans into cause and effect relationships, as has
been seen in sFLT1 and sENG manipulation in RUPP models, and are critical tools for assessing toxicity and efficacy
in novel therapeutics. Of course, animal models are imperfect
for this human-specific disease, as they fail to demonstrate
thrombocytopenia, HELLP syndrome, eclamptic seizures,
and other signs and symptoms that define the severe features
of preeclampsia in humans. Perhaps, the imperfect overlap
in presentation stems from the fact that the primary driver
of preeclampsia, insufficient trophoblast invasion and failure
of spiral artery remodeling, does not occur naturally in other
species and has yet to be modeled accurately. In fact, rodent
trophoblasts minimally invade the spiral arteries, as decidualization occurs after implantation,187 and therefore, may be
an inherently insufficient model species. Furthermore, human
gestation is significantly longer than in rodents, and exposes
pregnant women to much larger doses of circulating toxins,
such as sFLT1, possibly leading to severe features. In fact,
when sFLT1 and sENG levels are concurrently pushed to
exaggerated levels, severe features do occur in animal models.108–110,113 Despite their limitations, experimental studies in
animal models allow investigators to test directly whether
certain factors found in women with preeclampsia can indeed lead to hypertension and other manifestations of the
syndrome. Further investigation is necessary with models that
better approximate human gestation physiology and length to
isolate the primary pathogenic factors.
1102 Circulation Research March 29, 2019
Downloaded from http://ahajournals.org by on February 7, 2021
Maternal Contribution to Disease
Epidemiological studies suggest that several prepregnancy
maternal characteristics increase risk for preeclampsia.188
Interest is growing around obesity and diabetes mellitus as
risk factors (relative risk [RR], ≈3.5 each)189 in light of data
suggesting Metformin, the biguanide first-line therapy for
type 2 diabetes mellitus, may decrease sFLT1.190 In a recent
meta-analysis of 15 randomized controlled trials reporting
the incidence of hypertensive disorders of pregnancy with
Metformin use, Kalafat et al191 found a reduced risk of hypertensive disorders of pregnancy (RR, 0.56; 95% CI, 0.37–0.85)
but a nonsignificantly reduced risk of preeclampsia (RR, 0.74;
95% CI, 0.09–6.28). However, authors admit the low quality
of the evidence and the clinical heterogeneity of the included
studies, limiting conclusions. Prepregnancy vascular dysfunction, such as in women with chronic hypertension, not only
jeopardizes placental perfusion but may also enhance the placental response to ischemia as well as the vascular response
to antiangiogenic factors such as sFLT1. Therefore, baseline
host characteristics may put women at risk of preeclampsia,
even at physiological elevations of antiangiogenic factors
and cytokines. A history of acute kidney injury before pregnancy, despite apparent full recovery, is also associated with
increased risk of pregnancy complications.192 Interestingly, a
shorter interval between the AKI episode and the pregnancy
were associated with higher risks of preeclampsia. This suggests that subclinical renal dysfunction may interfere with the
hemodynamic adaptation of normal pregnancy, which in turn
may lead to impaired placental perfusion and preeclampsia.192
Translation of Preeclampsia
Biology in the Clinic
Biomarkers for Diagnosis, Prediction,
and Prognosis
The ACOG committee opinion issued in 2015 and reaffirmed
in 2017 does not recommend screening to predict preeclampsia beyond obtaining an appropriate medical history.18,193
Because of the lack of adequate screening methods and the
severe sequelae of the disease, all women suspected of preeclampsia undergo resource intensive testing, often requiring
several-day hospitalizations for further investigation. Other
methods of screening have been investigated, including a metabolomic pathways and combined metabolomic-proteomic
data approaches.194–197 A 2017 head to head comparison of the
Fetal Medicine Foundation algorithm-based screening method
(a combination of maternal factors, mean arterial pressure,
uterine-artery pulsatility index, and PlGF) demonstrated superiority to the screening methods currently recommended
by National Institute for Health and Care Excellence and
ACOG.198 Using a similar screening algorithm with the addition of maternal serum PAPP-A (pregnancy-associated plasma
protein-A), the ASPRE trial (aspirin for evidence-based preeclampsia prevention) screened women in the first trimester
to identify those at high risk for preeclampsia.199 High-risk
women were then randomized to receive 150 mg of aspirin
or placebo daily until 36 weeks gestation. Daily low-dose aspirin use in high-risk women was associated with a significantly lower incidence of preterm preeclampsia than placebo,
and detection rate of preterm preeclampsia was 76.7%
(138/180)—43.1% for term preeclampsia—with a false positive rate of 9.1%. These results suggest that early screening
with plasma biomarkers and imaging studies allows for early
intervention and possible prevention of disease.
Perhaps most promising among screening methods is the
use during the third trimester of combined biomarkers—such
as sFLT1, sEng, and PlGF—with high sensitivity and specificity for early diagnosis and prognosis of preeclampsia.200
Substantial evidence already demonstrates encouraging test
characteristics of biomarker assays in this population.201,202 In
a study of over 600 women undergoing initial evaluation of
preeclampsia, an sFLT1/PlGF ratio of ≥85 correlated with diagnosis of preeclampsia and predicted adverse outcomes and
delivery within 2 weeks among women presenting <34 weeks
gestation. Furthermore, the biomarker ratio performed better
than all other currently available tests for prediction of preeclampsia.110 A follow-up study of 402 patients presenting with
preterm singleton pregnancies by the same group showed that
an sFLT1/PlGF ratio >85 had a PPV of 59% in all patients and
74% among patients presenting <34 weeks for developing preeclampsia with severe features within 2 weeks.203 Patients with
preeclampsia with normal angiogenic profile have fewer adverse outcomes suggesting that the angiogenic form of preeclampsia is clinically more important.111 Other groups have
shown similar results. In a multicenter study of women with
suspected preeclampsia, authors showed a reverse association
of free PlGF with gestational age at delivery.204 In a recent multisite study of angiogenic factors, authors concluded that an
sFLT1/PlGF ratio of ≤38 had an negative predictive value of
99.9% for ruling out preeclampsia within 1 week.205 A follow-up
study from the same cohort showed an negative predictive value
of 95% within 4 weeks.206 Overall, serum or plasma angiogenic
factors appear to be a reliable risk-stratification method among
women with suspected preeclampsia, especially for preterm
preeclampsia, allowing for appropriate management.
Recent studies have also suggested that by reducing false
positive rates of diagnosis and consequent unnecessary hospitalizations, risk stratification with biomarkers is economical.
Assays using these plasma biomarkers are available for clinical use in Europe, Canada, Africa, and Asia, and their clinical
success has led to the incorporation of angiogenic factors in
the definition of preeclampsia in the British National Institute
for Health and Care Excellence and national German guidelines.207 Finally, a study in Mozambique showed that measurement of PlGF is feasible in resource poor environments.
The authors concluded that low PlGF in women with suspected preeclampsia was associated with increased transfers
to higher levels of care and increased maternal and perinatal
risks.208 Looking forward, biomarker assays are a cost-effective and reliable screening method that could be lifesaving,
even in areas of limited expertise and resources.
Though not part of any formal diagnostic criteria, hyperuricemia is classically a biomarker indicating progression of
gestational or chronic hypertension to preeclampsia and of
risk for fetal and maternal complications such as SGA.209–212
However, in all-comers, uric acid levels do not predict development of preeclampsia.213–215 Evidence about the contribution of uric acid to the pathogenesis of preeclampsia is mixed,
Rana et al Preeclampsia and Vascular Disease 1103
though general consensus suggests that levels are elevated
secondary to renal injury and decreased excretion.216
Treatment and Management of Preeclampsia
Downloaded from http://ahajournals.org by on February 7, 2021
Current management of preeclampsia in the developed world
includes preconception counseling, perinatal BP control and
monitoring, prenatal aspirin therapy in high-risk women, betamethasone for patients <34 weeks, parenteral magnesium
sulfate, and careful follow-up of postpartum BPs.1 Timely
delivery of the fetus and placenta remains the only definitive
treatment. Even among patients who did not show antenatal signs of preeclampsia, surveillance continues postpartum
because of the rising incidence of postpartum preeclampsia.
Preeclampsia without severe features can be managed expectantly with twice-weekly maternal and fetal monitoring until
37 weeks in the absence of labor, rupture of membranes, vaginal bleeding, or abnormal antepartum testing.1,18 In women
with preeclampsia with severe features at <34 weeks, expectant management can be attempted based on strict inclusion
criteria and with appropriate resources. In these patients, careful attention should be given to worsening maternal and fetal
well-being and delivery is indicated at any time with deterioration of maternal and fetal status. ACOG currently does not
recommend pharmacological treatment of mild to moderate
range hypertension (systolic <160 mm Hg or diastolic <110
mm Hg) in the setting of preeclampsia,1 as it does not appear
to attenuate risk of disease progression and may increase the
risk of fetal growth restriction.217 In a multinational randomized trial for control of BPs enrolling women with chronic and
gestational hypertension, authors found lower prevalence of
severe hypertension (≥160/110 mm Hg) among patients with
tight control of BPs during pregnancy (40.6% versus 27.5%).
There was no difference in development of preeclampsia and
perinatal outcomes were similar.218 A large randomized trial is
underway in the United States to evaluate treatment of moderate hypertension during pregnancy (URL: http://www.clinicaltrials.gov. Unique identifier: NCT02299414). Treatment of
severe hypertension requires pharmacological therapy with labetalol, nifedipine, or methyldopa,1 however, recent evidence
from animal studies suggest that amlodipine may be superior
to nifedipine because of its inducing effects on Arrb1 and subsequent downregulation of the AT1-B2 receptor complex,171
though more clinical evidence is needed. Though current clinical management is limited, innovative medical therapies are
on the horizon.
Small molecules have had variable success in treating hypertensive disorders of pregnancy. Early studies, including the
1998 National Institutes of Health trial, did not show improved
outcomes in women at high risk of preeclampsia when treated
with low dose (60 mg) of aspirin compared with placebo.219
However, with improved risk-stratification methods, Rolnik et
al199 demonstrated a lower incidence of preterm preeclampsia
in high-risk women treated with 150 mg of aspiring compared
with placebo. Aspirin is now recommended for the prevention
of preeclampsia in high-risk women.1
Sildenafil, a phosphodiesterase type 5 inhibitor, was initially thought to be a promising way to reduce placental ischemia given its ability to potentiate the actions of nitric
oxide, causing uterine vasodilation. However, in the 2018
Maternal STRIDER trial (Sildenafil for Severe Fetal Growth
Restriction), sildenafil did not prolong pregnancy or improve
pregnancy outcomes compared with placebo in women with
severe intrauterine growth restriction in the late second to
third trimester.220 A 2017 randomized controlled trial of sildenafil therapy in fetal growth restriction did not show any
benefit in attenuating disease compared with placebo221 and
was prematurely halted when 11 neonates in the sildenafil arm
died secondary to pulmonary disease.222 However, other small
molecules that effect nitric oxide production are still under
investigation. For example, statins continue to show promise
in animal models and in human ex vivo tissue samples.223–225
Likely because of its stimulatory effects on HO and improved
vascular function,224 pravastatin improves fetal and maternal outcomes in patients with antiphospholipid syndrome226
and mitigates disease in patients with severe features225 and
with evidence of placental vascular pathology.227 StAmP trial
(Statins to Ameliorate Early Onset Preeclampsia), a double-blind, multicenter, randomized controlled trial looking at
the effects of Pravastatin on serum biomarkers in preeclampsia, has completed recruitment and is now in the data-analysis
phase (ISRCTN 23410175). Metformin, an insulin sensitizer
and the first-line treatment of type 2 diabetes mellitus, is associated with reduced incidence of hypertensive disorders in
pregnancy,191 as well as decreased levels of circulating sFLT1
and sENG in vitro.228 Metformin has been shown to be safe in
pregnancy, with no significant difference in neonatal outcomes
when compared with insulin in the treatment of gestational diabetes mellitus,229 making Metformin an attractive and readily
available therapy. Finally, ouabain, a cardiac glycoside, has
been shown to inhibit sFLT1 mRNA and protein expression
through the HIF-1α/heat-shock protein 27 pathway in human
cytotrophoblasts and explant cultures as well as in rat models without any fetal adverse effects.230 Further investigation
is necessary to further characterize ouabain as a therapeutic.
Most small molecules target the prevention of preeclampsia in high-risk women, however, no therapy currently exists
for the treatment of preeclampsia to delay preterm delivery.
Adapting apheresis technology safely and commonly used
in pregnant women with familial hypercholesterolemia,231
Thadhani et al232 used a negatively charged dextran sulfate cellulose column to remove positively charged sFLT1 from the
serum of 3 women with preterm (<32 weeks) preeclampsia,
resulting in dose-dependent delays in delivery. To minimize
the side effect profile—namely, transient hypotension—and
improve the efficiency of removal, the same group repeated
the pilot study with an apheresis device that incorporated
plasma separation for sFLT1 removal.233 Eleven women with
very preterm preeclampsia were treated with dextran sulfate
column apheresis with a mean reduction in sFLT1 of 18% per
treatment and an average of 8 days (range 2–11) of extended
gestation with a single treatment and an average of 15 days
(range 11–21) extended gestation with multiple treatments,
compared with an average of 3 to 4 days in controls. There
were no additional adverse events in neonates treated with
apheresis compared with normal and preeclamptic controls
born comparably preterm. Direct therapies for the treatment
of preeclampsia would revolutionize the management of this
highly morbid disease, and the results from these pilot studies
1104 Circulation Research March 29, 2019
show immense promise, with refinement of the specificity of
the apheretic column for known preeclamptic markers.
Other promising therapies for preeclampsia include recombinant human PlGF and RNA interference technology-based
methods. A ligand specific to VEGFR1/Flt1, recombinant
human PlGF has the potential to scavenge excess circulating
sFLT1 without the additional side effects of vascular permeability and edema associated with VEGFR2. Baboon and rodent
studies have shown promising results, lowering BP, proteinuria, and sFLT1 mRNA.234,235 However, the treatment may be
limited given that only a small percentage of total body sFLT1
is circulating236 and, therefore, amenable to ligand interaction.
RNA interference technology provides a cheaper alternative
to recombinant therapies. Silencing sequences of RNA specific to all 3 isoforms of circulating sFLT1 mRNA have been
shown to decrease sFLT1, BP, and proteinuria in pregnant baboon models with a single dose.237 However, given the limitations of animal models of preeclampsia and the regulatory
implications of developing oligonucleotide therapies, significant further study is required before clinical trials.
Preeclampsia and CVD
Short-Term Cardiovascular Complications in the
Postpartum Period
Downloaded from http://ahajournals.org by on February 7, 2021
Postpartum hypertension (defined as hypertension >48 hours
or more after delivery) occurring either de novo or in the setting of preeclampsia has emerged as an important risk factor for significant morbidity in women in the United States.238
While the pathogenesis of this syndrome is still being elucidated, clinical studies have suggested that postpartum hypertension may share similar plasma angiogenic profiles as women
with antepartum preeclampsia and, therefore, may represent a
group of women with subclinical preeclampsia or unresolved
preeclampsia.5 In the peripartum period, preeclampsia is associated with an increased risk of peripartum cardiomyopathy (PPCM) that can progress to chronic heart failure, cardiac
transplantation, or death.239,240 Given that VEGF pathway inhibitors in oncology patients also induce cardiomyopathy,241
elevated levels of antiangiogenic factors may play a role in
the development of PPCM. Using a PGC (proangiogenic transcriptional cofactor)-1α knockout mouse model and human
serum studies, Patten et al240 showed that repeatedly elevated
levels of sFLT1 and prolactin cleavage fragments were synergistically associated with PPCM and that both angiogenic
factor imbalance and prolactin fragmentation are regulated
by PGC-1α. Authors proposed a 2-hit hypothesis for the development of PPCM: (1) an increase in antiangiogenic factors such as sFLT1 that causes cardiac dysfunction as seen
in preeclampsia and (2) an independent decrease in defenses
against antiangiogenic factors in the heart as seen in low levels
of cardiac PGC-1α expression. Thus, inhibition or removal of
sFLT1 could attenuate PPCM in preeclamptic women with an
environmental or genetic predisposition to disease.
In the short-term, neonates may also present with
vascular effects secondary to the elevated levels of antiangiogenic factors. Though hypertension and proteinuria
observed in the mother are not observed in the fetus, elevated levels of sFLT1 have been observed in the amniotic
fluid in women with preeclampsia.242 Epidemiological studies have shown an increased incidence of bronchopulmonary dysplasia243,244 and decreased incidence of retinopathy of
prematurity (adjusted odds ratio 0.65; 95% CI, 0.49–0.86
for early preterm),245 2 diseases of prematurity associated
with neoangiogenesis. Intraamniotic injections of sFlt1 in
rat models disrupt pulmonary angiogenesis and increase
the incidence of bronchopulmonary dysplasia,244 while administration of anti-sFLT1 monoclonal antibody increases
alveolar counts and pulmonary vessel density in the premature pups of preeclampsia rat models.246 Together, these
epidemiological and animal studies imply that sFLT1 affects fetal angiogenesis at mucous membranes exposed to
amniotic fluid.
Preeclampsia and Long-Term CVD
There is accumulating evidence that preeclampsia-eclampsia
predisposes to long-term cardiovascular risk247,248 including
risk of hypertension, peripheral arterial disease (RR, 1.87;
0.94–3.73), coronary artery disease (overall CVD RR: 2.3;
1.95–2.78), cerebrovascular disease (RR, 2.03; 1.54–2.67),
congestive heart failure, vascular dementia (hazard ratio, 3.46;
1.97–6.10), and death (RR, 2.29; 1.73–3.04).248–250 Potential
explanations for the association between preeclampsia and
CVD are debated. It has been proposed that endothelial damage caused by preeclampsia persists beyond postpartum recovery, increasing the risk of CVD,247 and that the risk of
future CVD increases with multiple episodes of preeclampsia.248 Alternatively, an unfavorable cardiovascular risk profile
characterized by higher levels of glucose, cholesterol, hypertension, and abdominal obesity may contribute to the development of preeclampsia and later, CVD.249
The odds of a fatal outcome from CVD have been shown
to be greater than odds of diagnosis (odds ratio =2.89; 95%
CI, 1.71–4.89 and odds ratio =2.01; 95% CI, 1.68–2.41; respectively)250 in preeclampsia, suggesting that women may
die from the sequelae of CVD without first being diagnosed.
Furthermore, meta-regression reveals a graded relationship
between the severity of preeclampsia-eclampsia and the risk
of cardiac disease (mild: RR 2.00, 1.83–2.19; moderate: RR
2.99, 2.51–3.58; and severe: RR 5.36, 3.96–7.27; P<0.0001),
implicating a dose-response to the severity of preeclampsia.249 Studies are underway to find biomarkers and subclinical echocardiographic measures, such as global longitudinal
strain,251–253 to provide early risk assessment and stratification
in women with preeclampsia.
The question remains if there is an opportunity for intervention in asymptomatic women with a history of preeclampsia. A study of 2 tertiary medical centers in the Netherlands254
found that a total of 42% of women with a history of preeclampsia had significant CVD risk factors compared with
14.3% among women with a history of an uncomplicated
pregnancy. In the United States, a 2018 study using the
Nurses’ Health Study II suggested that hypertensive disorders
of pregnancy were associated with 10-year CVD risk overall,
independent of established CVD risk factors. Globally there is
increasing evidence that a history of preeclampsia should be
considered in CVD risk stratification.7
Rana et al Preeclampsia and Vascular Disease 1105
Preeclampsia is also associated with 4.7-fold risk for subsequent end-stage renal disease.255 Based on sibling studies, Vikse
et al256 reported that familial aggregation of risk factors could not
explain the increased end-stage renal disease risk and, therefore,
concluded that preeclampsia per se may lead to long-term kidney damage. Women with preeclampsia are also at a 4-fold risk
of stroke257 and 3-fold risk of vascular dementia later in life.258
Additional studies are needed to elucidate the pathogenesis of
these long-term complications in women with preeclampsia.
Decades of literature observe a relationship between lifelong CVD risk and growth restriction in utero, such as seen
in the fetuses of preeclamptic pregnancies. First described
by Barker and Osmond,259 the so-called Barker Hypothesis
suggests that lack of early nutrition, growth restriction, and
the uterine environment increase susceptibility to other risks
for CVD. Indeed, children born to women with preeclampsia have an increased risk of CVD.260 Neonates may also be
at risk for pulmonary hypertension into their teenage years
and beyond.261 Pulmonary amniotic sFLT1 exposure leading
to decreased pulmonary angiogenesis has been proposed as
a mechanism for this long-term lung injury,246 though the sequelae of prematurity or systemic sFLT1 exposure246 cannot
be excluded. Current research seeks to further characterize
CVD in children born to preeclamptic mothers.262
Conclusions
Downloaded from http://ahajournals.org by on February 7, 2021
Preeclampsia is a leading cause of maternal morbidity and
mortality worldwide, whose only definitive treatment—delivery of the fetus and placenta—carries significant morbidity
and mortality for the neonate. Though the mortality of preeclampsia-eclampsia has decreased significantly in the United
States because of increased antenatal surveillance and early
interventions, the postpartum and lifelong sequelae of preeclampsia have risen in number and significance. Whether the
risk predates and confounds preeclampsia or is a result thereof,
we now know that women with a history of preeclampsia are
at increased risk for CVD250,263 and dementia258 later in life,
including acutely fatal myocardial infarction without the progressive, forewarning symptoms of the acute coronary syndrome.263 An increased focus on interventional studies during
the postpartum, asymptomatic phase is imperative to minimize the risk of CVD and its potentially fatal complications.
Improved risk stratification and therapeutics are other areas
of opportunity with several innovations on the horizon. While
ongoing work in placental oxidative stress and the maternal
immune response demonstrate intriguing insights, perhaps
the most promising area under investigation is the maternal
angiogenic factor imbalance and its effects on vascular function. Ongoing development of methods of risk stratification
using ratios of proangiogenic factors—such as PlGF—and antiangiogenic factors—such as sFLT1 and sENG—have high
detection rates for preterm preeclampsia when incorporated
into algorithms with other predictive elements264 and have
shown high negative predictive value as an isolated assay.
Removal of antiangiogenic proteins through plasma apheresis
significantly prolongs gestation compared with controls and
attenuates the symptoms of preeclampsia,232,233 and recombinant human PlGF and siRNA have shown promising results
in animal models,234,235,237 suggesting possible therapeutic
alternatives to preterm delivery. Most importantly, risk-stratification methods using antiangiogenic factors have already
proven to be safe, efficient, cost-effective, and economical,
making them possibilities for improving care in countries that
carry the highest mortality from preeclampsia.
Disclosures
S. Rana reports serving as a consultant for Roche Diagnostics and
Thermofisher and has received research funding from Roche and
Siemens. Dr Karumanchi is colisted as coinventors on patents related to preeclampsia biomarkers and therapies that are held at
Beth Israel Deaconess Medical Center. He has a financial interest in Aggamin LLC and also reports serving as a consultant to
Thermofisher.He has received research funding from Siemens. The
other authors report no conflicts.
References
1. American College of Obstetricians and Gynecologists; Task Force on
Hypertension in Pregnancy. Hypertension in pregnancy. Report of the
American College of Obstetricians and Gynecologists’ Task Force on
Hypertension in Pregnancy. Obstet Gynecol. 2013;122:1122–1131. doi:
10.1097/01.AOG.0000437382.03963.88
2. Hogan MC, Foreman KJ, Naghavi M, Ahn SY, Wang M, Makela
SM, Lopez AD, Lozano R, Murray CJ. Maternal mortality for 181
countries, 1980-2008: a systematic analysis of progress towards
Millennium Development Goal 5. Lancet. 2010;375:1609–1623. doi:
10.1016/S0140-6736(10)60518-1
3. Wanderer JP, Leffert LR, Mhyre JM, Kuklina EV, Callaghan WM,
Bateman BT. Epidemiology of obstetric-related ICU admissions in
Maryland: 1999-2008*. Crit Care Med. 2013;41:1844–1852. doi:
10.1097/CCM.0b013e31828a3e24
4. Kuklina EV, Ayala C, Callaghan WM. Hypertensive disorders and severe
obstetric morbidity in the United States. Obstet Gynecol. 2009;113:1299–
1306. doi: 10.1097/AOG.0b013e3181a45b25
5. Goel A, Maski MR, Bajracharya S, Wenger JB, Zhang D, Salahuddin
S, Shahul SS, Thadhani R, Seely EW, Karumanchi SA, Rana S.
Epidemiology and mechanisms of de novo and persistent hypertension in the postpartum period. Circulation. 2015;132:1726–1733. doi:
10.1161/CIRCULATIONAHA.115.015721
6. Bernstein PS, Martin JN Jr, Barton JR, Shields LE, Druzin ML, Scavone
BM, Frost J, Morton CH, Ruhl C, Slager J, Tsigas EZ, Jaffer S, Menard
MK. National partnership for maternal safety: consensus bundle on severe hypertension during pregnancy and the postpartum period. Obstet
Gynecol. 2017;130:347–357. doi: 10.1097/AOG.0000000000002115
7. Coutinho T, Lamai O, Nerenberg K. Hypertensive disorders of pregnancy and cardiovascular diseases: current knowledge and future
directions. Curr Treat Options Cardiovasc Med. 2018;20:56. doi:
10.1007/s11936-018-0653-8
8. Tooher J, Thornton C, Makris A, Ogle R, Korda A, Hennessy A.
All hypertensive disorders of pregnancy increase the risk of future cardiovascular disease. Hypertension. 2017;70:798–803. doi:
10.1161/HYPERTENSIONAHA.117.09246
9. Bartsch E, Medcalf KE, Park AL, Ray JG; High Risk of Pre-eclampsia
Identification Group. Clinical risk factors for pre-eclampsia determined
in early pregnancy: systematic review and meta-analysis of large cohort
studies. BMJ. 2016;353:i1753. doi: 10.1136/bmj.i1753
10. Boyd PA, Lindenbaum RH, Redman C. Pre-eclampsia and trisomy 13: a
possible association. Lancet. 1987;2:425–427.
11. Cincotta RB, Brennecke SP. Family history of pre-eclampsia as a predictor
for pre-eclampsia in primigravidas. Int J Gynaecol Obstet. 1998;60:23–27.
12. Cnattingius S, Reilly M, Pawitan Y, Lichtenstein P. Maternal and fetal genetic factors account for most of familial aggregation of preeclampsia: a population-based Swedish cohort study. Am J Med Genet A.
2004;130A:365–371. doi: 10.1002/ajmg.a.30257
13. Esplin MS, Fausett MB, Fraser A, Kerber R, Mineau G, Carrillo
J, Varner MW. Paternal and maternal components of the predisposition to preeclampsia. N Engl J Med. 2001;344:867–872. doi:
10.1056/NEJM200103223441201
14. McGinnis R, Steinthorsdottir V, Williams NO, et al; FINNPEC
Consortium; GOPEC Consortium. Variants in the fetal genome near FLT1
are associated with risk of preeclampsia. Nat Genet. 2017;49:1255–1260.
doi: 10.1038/ng.3895
1106 Circulation Research March 29, 2019
Downloaded from http://ahajournals.org by on February 7, 2021
15. Gray KJ, Kovacheva VP, Mirzakhani H, et al. Gene-centric analysis of
preeclampsia identifies maternal association at PLEKHG1. Hypertension.
2018;72:408–416. doi: 10.1161/HYPERTENSIONAHA.117.10688
16. Morgan MA, Thurnau GR. Pregnancy-induced hypertension without proteinuria: is it true preeclampsia? South Med J. 1988;81:210–213.
17. Barton JR, O’brien JM, Bergauer NK, Jacques DL, Sibai BM. Mild gestational hypertension remote from term: progression and outcome. Am J
Obstet Gynecol. 2001;184:979–983. doi: 10.1067/mob.2001.112905
18. ACOG practice bulletin no. 202 summary: gestational hypertension and
preeclampsia. Obstet Gynecol. 2019;133:211–214.
19. Redman CW, Sargent IL. Latest advances in understanding preeclampsia.
Science. 2005;308:1592–1594. doi: 10.1126/science.1111726
20. Romero R, Chaiworapongsa T. Preeclampsia: a link between trophoblast
dysregulation and an antiangiogenic state. J Clin Invest. 2013;123:2775–
2777. doi: 10.1172/JCI70431
21. Palei AC, Spradley FT, Warrington JP, George EM, Granger JP.
Pathophysiology of hypertension in pre-eclampsia: a lesson in integrative physiology. Acta Physiol (Oxf). 2013;208:224–233. doi:
10.1111/apha.12106
22. Roberts JM, Taylor RN, Musci TJ, Rodgers GM, Hubel CA, McLaughlin
MK. Preeclampsia: an endothelial cell disorder. Am J Obstet Gynecol.
1989;161:1200–1204.
23. Brosens I, Robertson WB, Dixon HG. The physiological response of
the vessels of the placental bed to normal pregnancy. J Pathol Bacteriol.
1967;93:569–579. doi: 10.1002/path.1700930218
24. Brosens I, Pijnenborg R, Vercruysse L, Romero R. The “Great Obstetrical
Syndromes” are associated with disorders of deep placentation. Am J
Obstet Gynecol. 2011;204:193–201. doi: 10.1016/j.ajog.2010.08.009
25. Zhou Y, Damsky CH, Fisher SJ. Preeclampsia is associated with failure
of human cytotrophoblasts to mimic a vascular adhesion phenotype. One
cause of defective endovascular invasion in this syndrome? J Clin Invest.
1997;99:2152–2164. doi: 10.1172/JCI119388
26. Brosens I, Renaer M. On the pathogenesis of placental infarcts in preeclampsia. J Obstet Gynaecol Br Commonw. 1972;79:794–799.
27. De Wolf F, Robertson WB, Brosens I. The ultrastructure of acute atherosis
in hypertensive pregnancy. Am J Obstet Gynecol. 1975;123:164–174.
28. Lin S, Shimizu I, Suehara N, Nakayama M, Aono T. Uterine artery
Doppler velocimetry in relation to trophoblast migration into the myometrium of the placental bed. Obstet Gynecol. 1995;85:760–765.
29. North RA, Ferrier C, Long D, Townend K, Kincaid-Smith P. Uterine
artery Doppler flow velocity waveforms in the second trimester for the
prediction of preeclampsia and fetal growth retardation. Obstet Gynecol.
1994;83:378–386.
30. Hecht JL, Zsengeller ZK, Spiel M, Karumanchi SA, Rosen S. Revisiting
decidual vasculopathy. Placenta. 2016;42:37–43. doi: 10.1016/j.placenta.
2016.04.006
31. Stanek J. Histological features of shallow placental implantation unify early-onset and late-onset preeclampsia [published online October 9, 2018].
Pediatr Dev Pathol. doi: 10.1177/1093526618803759
32. Stevens DU, Al-Nasiry S, Bulten J, Spaanderman ME. Decidual vasculopathy in preeclampsia: lesion characteristics relate to disease severity and perinatal outcome. Placenta. 2013;34:805–809. doi: 10.1016/j.
placenta.2013.05.008
33. Rabaglino MB, Post Uiterweer ED, Jeyabalan A, Hogge WA, Conrad KP.
Bioinformatics approach reveals evidence for impaired endometrial maturation before and during early pregnancy in women who developed preeclampsia. Hypertension. 2015;65:421–429.
34. Garrido-Gomez T, Dominguez F, Quiñonero A, Diaz-Gimeno P, Kapidzic
M, Gormley M, Ona K, Padilla-Iserte P, McMaster M, Genbacev O,
Perales A, Fisher SJ, Simón C. Defective decidualization during and
after severe preeclampsia reveals a possible maternal contribution to
the etiology. Proc Natl Acad Sci USA. 2017;114:E8468–E8477. doi:
10.1073/pnas.1706546114
35. Brosens JJ, Parker MG, McIndoe A, Pijnenborg R, Brosens IA. A role
for menstruation in preconditioning the uterus for successful pregnancy.
Am J Obstet Gynecol. 2009;200:615.e1–615.e6. doi: 10.1016/j.ajog.
2008.11.037
36. Gray KJ, Saxena R, Karumanchi SA. Genetic predisposition to preeclampsia is conferred by fetal DNA variants near FLT1, a gene involved in
the regulation of angiogenesis. Am J Obstet Gynecol. 2018;218:211–218.
doi: 10.1016/j.ajog.2017.11.562
37. Soleymanlou N, Jurisica I, Nevo O, Ietta F, Zhang X, Zamudio S, Post
M, Caniggia I. Molecular evidence of placental hypoxia in preeclampsia. J Clin Endocrinol Metab. 2005;90:4299–4308. doi: 10.1210/jc.
2005-0078
38. Burton GJ, Hempstock J, Jauniaux E. Nutrition of the human fetus during the first trimester–a review. Placenta. 2001;22(suppl A):S70–S77. doi:
10.1053/plac.2001.0639
39. Jauniaux E, Watson AL, Hempstock J, Bao YP, Skepper JN, Burton GJ.
Onset of maternal arterial blood flow and placental oxidative stress.
A possible factor in human early pregnancy failure. Am J Pathol.
2000;157:2111–2122. doi: 10.1016/S0002-9440(10)64849-3
40. Rajakumar A, Brandon HM, Daftary A, Ness R, Conrad KP. Evidence
for the functional activity of hypoxia-inducible transcription factors overexpressed in preeclamptic placentae. Placenta. 2004;25:763–769. doi:
10.1016/j.placenta.2004.02.011
41. Tal R, Shaish A, Barshack I, Polak-Charcon S, Afek A, Volkov A, Feldman
B, Avivi C, Harats D. Effects of hypoxia-inducible factor-1alpha overexpression in pregnant mice: possible implications for preeclampsia and
intrauterine growth restriction. Am J Pathol. 2010;177:2950–2962. doi:
10.2353/ajpath.2010.090800
42. Caniggia I, Mostachfi H, Winter J, Gassmann M, Lye SJ, Kuliszewski
M, Post M. Hypoxia-inducible factor-1 mediates the biological effects of
oxygen on human trophoblast differentiation through TGFbeta(3). J Clin
Invest. 2000;105:577–587. doi: 10.1172/JCI8316
43. Kanasaki K, Palmsten K, Sugimoto H, Ahmad S, Hamano Y, Xie L, Parry S,
Augustin HG, Gattone VH, Folkman J, Strauss JF, Kalluri R. Deficiency in
catechol-O-methyltransferase and 2-methoxyoestradiol is associated with
pre-eclampsia. Nature. 2008;453:1117–1121. doi: 10.1038/nature06951
44. Redman CW, Sargent IL. Placental stress and pre-eclampsia: a revised view.
Placenta. 2009;30(suppl A):S38–S42. doi: 10.1016/j.placenta.2008.11.021
45. Derwig I, Lythgoe DJ, Barker GJ, Poon L, Gowland P, Yeung R, Zelaya
F, Nicolaides K. Association of placental perfusion, as assessed by magnetic resonance imaging and uterine artery Doppler ultrasound, and its
relationship to pregnancy outcome. Placenta. 2013;34:885–891. doi:
10.1016/j.placenta.2013.07.006
46. Sohlberg S, Mulic-Lutvica A, Lindgren P, Ortiz-Nieto F, Wikström
AK, Wikström J. Placental perfusion in normal pregnancy and early
and late preeclampsia: a magnetic resonance imaging study. Placenta.
2014;35:202–206. doi: 10.1016/j.placenta.2014.01.008
47. Sohlberg S, Mulic-Lutvica A, Olovsson M, Weis J, Axelsson O, Wikström
J, Wikström AK. Magnetic resonance imaging-estimated placental perfusion in fetal growth assessment. Ultrasound Obstet Gynecol. 2015;46:700–
705. doi: 10.1002/uog.14786
48. Sørensen A, Peters D, Fründ E, Lingman G, Christiansen O, Uldbjerg
N. Changes in human placental oxygenation during maternal hyperoxia
estimated by blood oxygen level-dependent magnetic resonance imaging (BOLD MRI). Ultrasound Obstet Gynecol. 2013;42:310–314. doi:
10.1002/uog.12395
49. Sinding M, Peters DA, Poulsen SS, Frøkjær JB, Christiansen OB,
Petersen A, Uldbjerg N, Sørensen A. Placental baseline conditions modulate the hyperoxic BOLD-MRI response. Placenta. 2018;61:17–23. doi:
10.1016/j.placenta.2017.11.002
50. Luo J, Abaci Turk E, Bibbo C, et al. In vivo quantification of placental
insufficiency by BOLD MRI: a human study. Sci Rep. 2017;7:3713. doi:
10.1038/s41598-017-03450-0
51. Many A, Hubel CA, Fisher SJ, Roberts JM, Zhou Y. Invasive cytotrophoblasts manifest evidence of oxidative stress in preeclampsia. Am J Pathol.
2000;156:321–331. doi: 10.1016/S0002-9440(10)64733-5
52. Zhuang B, Luo X, Rao H, Li Q, Shan N, Liu X, Qi H. Oxidative
stress-induced C/EBPβ inhibits β-catenin signaling molecule involving in the pathology of preeclampsia. Placenta. 2015;36:839–846. doi:
10.1016/j.placenta.2015.06.016
53. Huang QT, Wang SS, Zhang M, Huang LP, Tian JW, Yu YH, Wang ZJ,
Zhong M. Advanced oxidation protein products enhances soluble Fmslike tyrosine kinase 1 expression in trophoblasts: a possible link between
oxidative stress and preeclampsia. Placenta. 2013;34:949–952. doi:
10.1016/j.placenta.2013.06.308
54. Vaughan JE, Walsh SW. Oxidative stress reproduces placental abnormalities of preeclampsia. Hypertens Pregnancy. 2002;21:205–223. doi:
10.1081/PRG-120015848
55. Poston L, Briley AL, Seed PT, Kelly FJ, Shennan AH; Vitamins in Preeclampsia (VIP) Trial Consortium. Vitamin C and vitamin E in pregnant
women at risk for pre-eclampsia (VIP trial): randomised placebo-controlled
trial. Lancet. 2006;367:1145–1154. doi: 10.1016/S0140-6736(06)68433-X
56. Roberts JM, Myatt L, Spong CY, et al; Eunice Kennedy Shriver National
Institute of Child Health and Human Development Maternal-Fetal
Medicine Units Network. Vitamins C and E to prevent complications of
pregnancy-associated hypertension. N Engl J Med. 2010;362:1282–1291.
doi: 10.1056/NEJMoa0908056
Rana et al Preeclampsia and Vascular Disease 1107
Downloaded from http://ahajournals.org by on February 7, 2021
57. Zsengellér ZK, Rajakumar A, Hunter JT, Salahuddin S, Rana S, Stillman
IE, Ananth Karumanchi S. Trophoblast mitochondrial function is impaired
in preeclampsia and correlates negatively with the expression of soluble
fms-like tyrosine kinase 1. Pregnancy Hypertens. 2016;6:313–319. doi:
10.1016/j.preghy.2016.06.004
58. Kai S, Tanaka T, Daijo H, Harada H, Kishimoto S, Suzuki K, Takabuchi S,
Takenaga K, Fukuda K, Hirota K. Hydrogen sulfide inhibits hypoxia- but
not anoxia-induced hypoxia-inducible factor 1 activation in a von hippellindau- and mitochondria-dependent manner. Antioxid Redox Signal.
2012;16:203–216. doi: 10.1089/ars.2011.3882
59. Covarrubias AE, Lecarpentier E, Lo A, Salahuddin S, Gray KJ, Karumanchi
SA, Zsengeller ZK. AP39, a modulator of mitochondrial bioenergetics,
reduces anti-angiogenic response and oxidative stress in hypoxia-exposed trophoblasts: relevance for preeclampsia pathogenesis. Am J Pathol.
2019;189:104–114.
60. Chandel NS, McClintock DS, Feliciano CE, Wood TM, Melendez JA,
Rodriguez AM, Schumacker PT. Reactive oxygen species generated at
mitochondrial complex III stabilize hypoxia-inducible factor-1alpha during hypoxia: a mechanism of O2 sensing. J Biol Chem. 2000;275:25130–
25138. doi: 10.1074/jbc.M001914200
61. Vaka VR, McMaster KM, Cunningham MW Jr, Ibrahim T, Hazlewood R,
Usry N, Cornelius DC, Amaral LM, LaMarca B. Role of mitochondrial dysfunction and reactive oxygen species in mediating hypertension in the reduced uterine perfusion pressure rat model of preeclampsia. Hypertension.
2018;72:703–711. doi: 10.1161/HYPERTENSIONAHA.118.11290
62. Yung HW, Calabrese S, Hynx D, Hemmings BA, Cetin I, Charnock-Jones
DS, Burton GJ. Evidence of placental translation inhibition and endoplasmic reticulum stress in the etiology of human intrauterine growth restriction. Am J Pathol. 2008;173:451–462. doi: 10.2353/ajpath.2008.071193
63. Yung HW, Korolchuk S, Tolkovsky AM, Charnock-Jones DS, Burton
GJ. Endoplasmic reticulum stress exacerbates ischemia-reperfusion-induced apoptosis through attenuation of Akt protein synthesis in human choriocarcinoma cells. FASEB J. 2007;21:872–884. doi:
10.1096/fj.06-6054com
64. Lian IA, Løset M, Mundal SB, Fenstad MH, Johnson MP, Eide IP, Bjørge
L, Freed KA, Moses EK, Austgulen R. Increased endoplasmic reticulum
stress in decidual tissue from pregnancies complicated by fetal growth
restriction with and without pre-eclampsia. Placenta. 2011;32:823–829.
doi: 10.1016/j.placenta.2011.08.005
65. Fu J, Zhao L, Wang L, Zhu X. Expression of markers of endoplasmic reticulum stress-induced apoptosis in the placenta of women with early and
late onset severe pre-eclampsia. Taiwan J Obstet Gynecol. 2015;54:19–23.
doi: 10.1016/j.tjog.2014.11.002
66. Du L, He F, Kuang L, Tang W, Li Y, Chen D. eNOS/iNOS and endoplasmic
reticulum stress-induced apoptosis in the placentas of patients with preeclampsia. J Hum Hypertens. 2017;31:49–55. doi: 10.1038/jhh.2016.17
67. Mizuuchi M, Cindrova-Davies T, Olovsson M, Charnock-Jones DS,
Burton GJ, Yung HW. Placental endoplasmic reticulum stress negatively
regulates transcription of placental growth factor via ATF4 and ATF6β:
implications for the pathophysiology of human pregnancy complications.
J Pathol. 2016;238:550–561. doi: 10.1002/path.4678
68. George EM, Granger JP. Heme oxygenase in pregnancy and preeclampsia. Curr Opin Nephrol Hypertens. 2013;22:156–162. doi: 10.1097/MNH.
0b013e32835d19f7
69. Cudmore M, Ahmad S, Al-Ani B, Fujisawa T, Coxall H, Chudasama K, Devey
LR, Wigmore SJ, Abbas A, Hewett PW, Ahmed A. Negative regulation of soluble Flt-1 and soluble endoglin release by heme oxygenase-1. Circulation.
2007;115:1789–1797. doi: 10.1161/CIRCULATIONAHA.106.660134
70. George EM, Colson D, Dixon J, Palei AC, Granger JP. Heme oxygenase-1 attenuates hypoxia-induced sFlt-1 and oxidative stress in placental
villi through its metabolic products CO and bilirubin. Int J Hypertens.
2012;2012:486053. doi: 10.1155/2012/486053
71. Origassa CS, Câmara NO. Cytoprotective role of heme oxygenase-1 and
heme degradation derived end products in liver injury. World J Hepatol.
2013;5:541–549. doi: 10.4254/wjh.v5.i10.541
72. McCaig D, Lyall F. Inhibitors of heme oxygenase reduce invasion of human primary cytotrophoblast cells in vitro. Placenta. 2009;30:536–538.
doi: 10.1016/j.placenta.2009.03.004
73. Bilban M, Haslinger P, Prast J, Klinglmüller F, Woelfel T, Haider S, Sachs
A, Otterbein LE, Desoye G, Hiden U, Wagner O, Knöfler M. Identification
of novel trophoblast invasion-related genes: heme oxygenase-1 controls motility via peroxisome proliferator-activated receptor gamma.
Endocrinology. 2009;150:1000–1013. doi: 10.1210/en.2008-0456
74. George EM, Cockrell K, Aranay M, Csongradi E, Stec DE, Granger
JP. Induction of heme oxygenase 1 attenuates placental ischemia-
75.
76.
77.
78.
79.
80.
81.
82.
83.
84.
85.
86.
87.
88.
89.
90.
91.
92.
93.
94.
95.
induced hypertension. Hypertension. 2011;57:941–948. doi: 10.1161/
HYPERTENSIONAHA.111.169755
Cui Y, Wang W, Dong N, et al. Role of corin in trophoblast invasion and
uterine spiral artery remodelling in pregnancy. Nature. 2012;484:246–250.
doi: 10.1038/nature10897
Gu Y, Thompson D, Xu J, Lewis DF, Morgan JA, Cooper DB,
McCathran CE, Wang Y. Aberrant pro-atrial natriuretic peptide/corin/
natriuretic peptide receptor signaling is present in maternal vascular endothelium in preeclampsia. Pregnancy Hypertens. 2018;11:1–6. doi:
10.1016/j.preghy.2017.12.001
Miyazaki J, Nishizawa H, Kambayashi A, Ito M, Noda Y, Terasawa
S, Kato T, Miyamura H, Shiogama K, Sekiya T, Kurahashi H, Fujii T.
Increased levels of soluble corin in pre-eclampsia and fetal growth restriction. Placenta. 2016;48:20–25. doi: 10.1016/j.placenta.2016.10.002
Barnea ER, MacLusky NJ, DeCherney AH, Naftolin F. Catechol-omethyl transferase activity in the human term placenta. Am J Perinatol.
1988;5:121–127. doi: 10.1055/s-2007-999669
Berg D, Sonsalla R, Kuss E. Concentrations of 2-methoxyoestrogens
in human serum measured by a heterologous immunoassay with an
125I-labelled ligand. Acta Endocrinol (Copenh). 1983;103:282–288.
Palmer K, Saglam B, Whitehead C, Stock O, Lappas M, Tong S. Severe
early-onset preeclampsia is not associated with a change in placental catechol O-methyltransferase (COMT) expression. Am J Pathol.
2011;178:2484–2488. doi: 10.1016/j.ajpath.2011.02.029
Bulmer JN, Williams PJ, Lash GE. Immune cells in the placental bed. Int
J Dev Biol. 2010;54:281–294. doi: 10.1387/ijdb.082763jb
Koopman LA, Kopcow HD, Rybalov B, Boyson JE, Orange JS, Schatz F,
Masch R, Lockwood CJ, Schachter AD, Park PJ, Strominger JL. Human decidual natural killer cells are a unique NK cell subset with immunomodulatory potential. J Exp Med. 2003;198:1201–1212. doi: 10.1084/jem.20030305
Moffett A, Colucci F. Uterine NK cells: active regulators at the maternalfetal interface. J Clin Invest. 2014;124:1872–1879. doi: 10.1172/JCI68107
Moffett-King A. Natural killer cells and pregnancy. Nat Rev Immunol.
2002;2:656–663. doi: 10.1038/nri886
Lash GE, Schiessl B, Kirkley M, Innes BA, Cooper A, Searle RF, Robson
SC, Bulmer JN. Expression of angiogenic growth factors by uterine natural killer cells during early pregnancy. J Leukoc Biol. 2006;80:572–580.
doi: 10.1189/jlb.0406250
Parham P, Moffett A. Variable NK cell receptors and their MHC class I ligands in immunity, reproduction and human evolution. Nat Rev Immunol.
2013;13:133–144. doi: 10.1038/nri3370
Apps R, Murphy SP, Fernando R, Gardner L, Ahad T, Moffett A. Human
leucocyte antigen (HLA) expression of primary trophoblast cells and placental cell lines, determined using single antigen beads to characterize
allotype specificities of anti-HLA antibodies. Immunology. 2009;127:26–
39. doi: 10.1111/j.1365-2567.2008.03019.x
Madeja Z, Yadi H, Apps R, Boulenouar S, Roper SJ, Gardner L, Moffett A,
Colucci F, Hemberger M. Paternal MHC expression on mouse trophoblast
affects uterine vascularization and fetal growth. Proc Natl Acad Sci USA.
2011;108:4012–4017. doi: 10.1073/pnas.1005342108
Kieckbusch J, Gaynor LM, Moffett A, Colucci F. MHC-dependent inhibition of uterine NK cells impedes fetal growth and decidual vascular remodelling. Nat Commun. 2014;5:3359. doi: 10.1038/ncomms4359
Hiby SE, Walker JJ, O’shaughnessy KM, Redman CW, Carrington M,
Trowsdale J, Moffett A. Combinations of maternal KIR and fetal HLA-C
genes influence the risk of preeclampsia and reproductive success. J Exp
Med. 2004;200:957–965. doi: 10.1084/jem.20041214
Hiby SE, Apps R, Sharkey AM, et al. Maternal activating KIRs protect
against human reproductive failure mediated by fetal HLA-C2. J Clin
Invest. 2010;120:4102–4110. doi: 10.1172/JCI43998
Hiby SE, Regan L, Lo W, Farrell L, Carrington M, Moffett A. Association
of maternal killer-cell immunoglobulin-like receptors and parental HLA-C
genotypes with recurrent miscarriage. Hum Reprod. 2008;23:972–976.
doi: 10.1093/humrep/den011
Ahmad S, Ahmed A. Elevated placental soluble vascular endothelial
growth factor receptor-1 inhibits angiogenesis in preeclampsia. Circ Res.
2004;95:884–891. doi: 10.1161/01.RES.0000147365.86159.f5
Maynard SE, Min JY, Merchan J, Lim KH, Li J, Mondal S, Libermann TA,
Morgan JP, Sellke FW, Stillman IE, Epstein FH, Sukhatme VP, Karumanchi
SA. Excess placental soluble fms-like tyrosine kinase 1 (sFlt1) may contribute to endothelial dysfunction, hypertension, and proteinuria in preeclampsia. J Clin Invest. 2003;111:649–658. doi: 10.1172/JCI17189
Kendall RL, Thomas KA. Inhibition of vascular endothelial cell growth
factor activity by an endogenously encoded soluble receptor. Proc Natl
Acad Sci USA. 1993;90:10705–10709.
1108 Circulation Research March 29, 2019
Downloaded from http://ahajournals.org by on February 7, 2021
96. Esser S, Wolburg K, Wolburg H, Breier G, Kurzchalia T, Risau W.
Vascular endothelial growth factor induces endothelial fenestrations in
vitro. J Cell Biol. 1998;140:947–959.
97. De Falco S. The discovery of placenta growth factor and its biological
activity. Exp Mol Med. 2012;44:1–9. doi: 10.3858/emm.2012.44.1.025
98. Koga K, Osuga Y, Yoshino O, Hirota Y, Ruimeng X, Hirata T, Takeda
S, Yano T, Tsutsumi O, Taketani Y. Elevated serum soluble vascular endothelial growth factor receptor 1 (sVEGFR-1) levels in women with
preeclampsia. J Clin Endocrinol Metab. 2003;88:2348–2351. doi:
10.1210/jc.2002-021942
99. Tsatsaris V, Goffin F, Munaut C, Brichant JF, Pignon MR, Noel A,
Schaaps JP, Cabrol D, Frankenne F, Foidart JM. Overexpression of
the soluble vascular endothelial growth factor receptor in preeclamptic
patients: pathophysiological consequences. J Clin Endocrinol Metab.
2003;88:5555–5563. doi: 10.1210/jc.2003-030528
100. Lu F, Longo M, Tamayo E, Maner W, Al-Hendy A, Anderson GD, Hankins
GD, Saade GR. The effect of over-expression of sFlt-1 on blood pressure and the occurrence of other manifestations of preeclampsia in unrestrained conscious pregnant mice. Am J Obstet Gynecol. 2007;196:396.
e1–396.e7; discussion 396.e7. doi: 10.1016/j.ajog.2006.12.024
101. Launay-Vacher V, Deray G. Hypertension and proteinuria: a class-effect
of antiangiogenic therapies. Anticancer Drugs. 2009;20:81–82. doi:
10.1097/CAD.0b013e3283161012
102. Vigneau C, Lorcy N, Dolley-Hitze T, Jouan F, Arlot-Bonnemains Y,
Laguerre B, Verhoest G, Goujon JM, Belaud-Rotureau MA, RiouxLeclercq N. All anti-vascular endothelial growth factor drugs can induce
‘pre-eclampsia-like syndrome’: a RARe study. Nephrol Dial Transplant.
2014;29:325–332. doi: 10.1093/ndt/gft465
103. Bergmann A, Ahmad S, Cudmore M, Gruber AD, Wittschen P,
Lindenmaier W, Christofori G, Gross V, Gonzalves ACh, Gröne HJ,
Ahmed A, Weich HA. Reduction of circulating soluble Flt-1 alleviates preeclampsia-like symptoms in a mouse model. J Cell Mol Med.
2010;14:1857–1867. doi: 10.1111/j.1582-4934.2009.00820.x
104. Kumasawa K, Ikawa M, Kidoya H, Hasuwa H, Saito-Fujita T, Morioka Y,
Takakura N, Kimura T, Okabe M. Pravastatin induces placental growth
factor (PGF) and ameliorates preeclampsia in a mouse model. Proc Natl
Acad Sci USA. 2011;108:1451–1455. doi: 10.1073/pnas.1011293108
105. Li Z, Zhang Y, Ying Ma J, Kapoun AM, Shao Q, Kerr I, Lam A,
O’Young G, Sannajust F, Stathis P, Schreiner G, Karumanchi SA,
Protter AA, Pollitt NS. Recombinant vascular endothelial growth factor 121 attenuates hypertension and improves kidney damage in a
rat model of preeclampsia. Hypertension. 2007;50:686–692. doi:
10.1161/HYPERTENSIONAHA.107.092098
106. Hladunewich MA, Steinberg G, Karumanchi SA, Levine RJ, Keating
S, Kingdom J, Keunen J. Angiogenic factor abnormalities and fetal
demise in a twin pregnancy. Nat Rev Nephrol. 2009;5:658–662. doi:
10.1038/nrneph.2009.154
107. Stepan H, Faber R. Elevated sFlt1 level and preeclampsia with parvovirus-induced hydrops. N Engl J Med. 2006;354:1857–1858. doi:
10.1056/NEJMc052721
108. Levine RJ, Maynard SE, Qian C, Lim KH, England LJ, Yu KF,
Schisterman EF, Thadhani R, Sachs BP, Epstein FH, Sibai BM,
Sukhatme VP, Karumanchi SA. Circulating angiogenic factors and
the risk of preeclampsia. N Engl J Med. 2004;350:672–683. doi:
10.1056/NEJMoa031884
109. Romero R, Nien JK, Espinoza J, Todem D, Fu W, Chung H, Kusanovic JP,
Gotsch F, Erez O, Mazaki-Tovi S, Gomez R, Edwin S, Chaiworapongsa
T, Levine RJ, Karumanchi SA. A longitudinal study of angiogenic (placental growth factor) and anti-angiogenic (soluble endoglin and soluble
vascular endothelial growth factor receptor-1) factors in normal pregnancy and patients destined to develop preeclampsia and deliver a small
for gestational age neonate. J Matern Fetal Neonatal Med. 2008;21:9–
23. doi: 10.1080/14767050701830480
110. Rana S, Powe CE, Salahuddin S, Verlohren S, Perschel FH,
Levine RJ, Lim KH, Wenger JB, Thadhani R, Karumanchi SA.
Angiogenic factors and the risk of adverse outcomes in women
with suspected preeclampsia. Circulation. 2012;125:911–919. doi:
10.1161/CIRCULATIONAHA.111.054361
111. Rana S, Schnettler WT, Powe C, Wenger J, Salahuddin S, Cerdeira AS,
Verlohren S, Perschel FH, Arany Z, Lim KH, Thadhani R, Karumanchi
SA. Clinical characterization and outcomes of preeclampsia with normal
angiogenic profile. Hypertens Pregnancy. 2013;32:189–201. doi:
10.3109/10641955.2013.784788
112. Kleinrouweler CE, Wiegerinck MM, Ris-Stalpers C, Bossuyt PM, van
der Post JA, von Dadelszen P, Mol BW, Pajkrt E; EBM CONNECT
113.
114.
115.
116.
117.
118.
119.
120.
121.
122.
123.
124.
125.
126.
127.
128.
129.
130.
131.
Collaboration. Accuracy of circulating placental growth factor, vascular endothelial growth factor, soluble fms-like tyrosine kinase
1 and soluble endoglin in the prediction of pre-eclampsia: a systematic review and meta-analysis. BJOG. 2012;119:778–787. doi:
10.1111/j.1471-0528.2012.03311.x
Poon LC, Kametas NA, Maiz N, Akolekar R, Nicolaides KH. Firsttrimester prediction of hypertensive disorders in pregnancy. Hypertension.
2009;53:812–818. doi: 10.1161/HYPERTENSIONAHA.108.127977
Powe CE, Levine RJ, Karumanchi SA. Preeclampsia, a disease of the
maternal endothelium: the role of antiangiogenic factors and implications for later cardiovascular disease. Circulation. 2011;123:2856–2869.
doi: 10.1161/CIRCULATIONAHA.109.853127
Levine RJ, Lam C, Qian C, Yu KF, Maynard SE, Sachs BP, Sibai
BM, Epstein FH, Romero R, Thadhani R, Karumanchi SA; CPEP
Study Group. Soluble endoglin and other circulating antiangiogenic
factors in preeclampsia. N Engl J Med. 2006;355:992–1005. doi:
10.1056/NEJMoa055352
Venkatesha S, Toporsian M, Lam C, et al. Soluble endoglin contributes
to the pathogenesis of preeclampsia. Nat Med. 2006;12:642–649. doi:
10.1038/nm1429
Wallace K, Morris R, Kyle PB, Cornelius D, Darby M, Scott J, Moseley
J, Chatman K, Lamarca B. Hypertension, inflammation and T lymphocytes are increased in a rat model of HELLP syndrome. Hypertens
Pregnancy. 2014;33:41–54. doi: 10.3109/10641955.2013.835820
Maharaj AS, Walshe TE, Saint-Geniez M, Venkatesha S, Maldonado
AE, Himes NC, Matharu KS, Karumanchi SA, D’Amore PA. VEGF and
TGF-beta are required for the maintenance of the choroid plexus and ependyma. J Exp Med. 2008;205:491–501. doi: 10.1084/jem.20072041
Wallace K, Bean C, Bowles T, Spencer SK, Randle W, Kyle PB,
Shaffery J. Hypertension, anxiety, and blood-brain barrier permeability
are increased in postpartum severe preeclampsia/hemolysis, elevated
liver enzymes, and low platelet count syndrome rats. Hypertension.
2018;72:946–954. doi: 10.1161/HYPERTENSIONAHA.118.11770
Askelund KJ, Chamley LW. Trophoblast deportation part I: review
of the evidence demonstrating trophoblast shedding and deportation during human pregnancy. Placenta. 2011;32:716–723. doi:
10.1016/j.placenta.2011.07.081
Pantham P, Askelund KJ, Chamley LW. Trophoblast deportation part
II: a review of the maternal consequences of trophoblast deportation.
Placenta. 2011;32:724–731. doi: 10.1016/j.placenta.2011.06.019
Attwood HD, Park WW. Embolism to the lungs by trophoblast. J Obstet
Gynaecol Br Commonw. 1961;68:611–617.
Knight M, Redman CW, Linton EA, Sargent IL. Shedding of syncytiotrophoblast microvilli into the maternal circulation in pre-eclamptic pregnancies. Br J Obstet Gynaecol. 1998;105:632–640.
Germain SJ, Sacks GP, Sooranna SR, Soorana SR, Sargent IL, Redman
CW. Systemic inflammatory priming in normal pregnancy and preeclampsia: the role of circulating syncytiotrophoblast microparticles. J
Immunol. 2007;178:5949–5956.
Guller S, Tang Z, Ma YY, Di Santo S, Sager R, Schneider H. Protein
composition of microparticles shed from human placenta during placental perfusion: Potential role in angiogenesis and fibrinolysis in preeclampsia. Placenta. 2011;32:63–69. doi: 10.1016/j.placenta.2010.10.011
Chang X, Yao J, He Q, Liu M, Duan T, Wang K. Exosomes from
women with preeclampsia induced vascular dysfunction by delivering sFlt (Soluble Fms-Like Tyrosine Kinase)-1 and sEng (Soluble
Endoglin) to endothelial cells. Hypertension. 2018;72:1381–1390. doi:
10.1161/HYPERTENSIONAHA.118.11706
Southcombe J, Tannetta D, Redman C, Sargent I. The immunomodulatory
role of syncytiotrophoblast microvesicles. PLoS One. 2011;6:e20245.
doi: 10.1371/journal.pone.0020245
Gupta AK, Rusterholz C, Huppertz B, Malek A, Schneider H, Holzgreve
W, Hahn S. A comparative study of the effect of three different syncytiotrophoblast micro-particles preparations on endothelial cells. Placenta.
2005;26:59–66. doi: 10.1016/j.placenta.2004.04.004
O’Brien M, Baczyk D, Kingdom JC. Endothelial dysfunction in severe
preeclampsia is mediated by soluble factors, rather than extracellular
vesicles. Sci Rep. 2017;7:5887. doi: 10.1038/s41598-017-06178-z
Harmon A, Cornelius D, Amaral L, Paige A, Herse F, Ibrahim T,
Wallukat G, Faulkner J, Moseley J, Dechend R, LaMarca B. IL-10 supplementation increases Tregs and decreases hypertension in the RUPP
rat model of preeclampsia. Hypertens Pregnancy. 2015;34:291–306. doi:
10.3109/10641955.2015.1032054
Weel IC, Baergen RN, Romão-Veiga M, Borges VT, Ribeiro VR, Witkin
SS, Bannwart-Castro C, Peraçoli JC, De Oliveira L, Peraçoli MT.
Rana et al Preeclampsia and Vascular Disease 1109
132.
133.
134.
135.
136.
137.
138.
139.
Downloaded from http://ahajournals.org by on February 7, 2021
140.
141.
142.
143.
144.
145.
146.
147.
148.
149.
Association between placental lesions, cytokines and angiogenic factors
in pregnant women with preeclampsia. PLoS One. 2016;11:e0157584.
doi: 10.1371/journal.pone.0157584
Xu J, Gu Y, Sun J, Zhu H, Lewis DF, Wang Y. Reduced CD200 expression is associated with altered Th1/Th2 cytokine production in placental
trophoblasts from preeclampsia. Am J Reprod Immunol. 2018;79:1. doi:
10.1111/aji.12763
Peixoto AB, Araujo Júnior E, Ribeiro JU, Rodrigues DB, Castro
EC, Caldas TM, Rodrigues Júnior V. Evaluation of inflammatory
mediators in the deciduas of pregnant women with pre-eclampsia/
eclampsia. J Matern Fetal Neonatal Med. 2016;29:75–79. doi:
10.3109/14767058.2014.987117
Darmochwal-Kolarz D, Rolinski J, Leszczynska-Goarzelak B, Oleszczuk
J. The expressions of intracellular cytokines in the lymphocytes of preeclamptic patients. Am J Reprod Immunol. 2002;48:381–386.
Chen W, Qian L, Wu F, Li M, Wang H. Significance of tolllike receptor 4 signaling in peripheral blood monocytes of preeclamptic patients. Hypertens Pregnancy. 2015;34:486–494. doi:
10.3109/10641955.2015.1077860
Medeiros LT, Peraçoli JC, Bannwart-Castro CF, Romão M, Weel
IC, Golim MA, de Oliveira LG, Kurokawa CS, Medeiros Borges VT,
Peraçoli MT. Monocytes from pregnant women with pre-eclampsia are
polarized to a M1 phenotype. Am J Reprod Immunol. 2014;72:5–13. doi:
10.1111/aji.12222
Campos-Cañas J, Romo-Palafox I, Albani-Campanario M, HernándezGuerrero C. An imbalance in the production of proinflammatory and
anti-inflammatory cytokines is observed in whole blood cultures of
preeclamptic women in comparison with healthy pregnant women.
Hypertens Pregnancy. 2014;33:236–249. doi: 10.3109/10641955.
2013.858744
Cristofalo R, Bannwart-Castro CF, Magalhães CG, Borges VT, Peraçoli
JC, Witkin SS, Peraçoli MT. Silibinin attenuates oxidative metabolism
and cytokine production by monocytes from preeclamptic women. Free
Radic Res. 2013;47:268–275. doi: 10.3109/10715762.2013.765951
Saito S, Sakai M. Th1/Th2 balance in preeclampsia. J Reprod Immunol.
2003;59:161–173.
Wegmann TG, Lin H, Guilbert L, Mosmann TR. Bidirectional cytokine interactions in the maternal-fetal relationship: is successful pregnancy a TH2 phenomenon? Immunol Today. 1993;14:353–356. doi:
10.1016/0167-5699(93)90235-D
Sowmya S, Sri Manjari K, Ramaiah A, Sunitha T, Nallari P, Jyothy A,
Venkateshwari A. Interleukin 10 gene promoter polymorphisms in women
with early-onset pre-eclampsia. Clin Exp Immunol. 2014;178:334–341.
doi: 10.1111/cei.12402
Regal JF, Burwick RM, Fleming SD. The complement system and
preeclampsia. Curr Hypertens Rep. 2017;19:87. doi: 10.1007/
s11906-017-0784-4
Derzsy Z, Prohászka Z, Rigó J Jr, Füst G, Molvarec A. Activation of
the complement system in normal pregnancy and preeclampsia. Mol
Immunol. 2010;47:1500–1506. doi: 10.1016/j.molimm.2010.01.021
Lokki AI, Kaartokallio T, Holmberg V, Onkamo P, Koskinen LLE,
Saavalainen P, Heinonen S, Kajantie E, Kere J, Kivinen K, Pouta A,
Villa PM, Hiltunen L, Laivuori H, Meri S. Analysis of complement
C3 gene reveals susceptibility to severe preeclampsia. Front Immunol.
2017;8:589. doi: 10.3389/fimmu.2017.00589
Gelber SE, Brent E, Redecha P, Perino G, Tomlinson S, Davisson
RL, Salmon JE. Prevention of defective placentation and pregnancy
loss by blocking innate immune pathways in a syngeneic model
of placental insufficiency. J Immunol. 2015;195:1129–1138. doi:
10.4049/jimmunol.1402220
Qing X, Redecha PB, Burmeister MA, Tomlinson S, D’Agati VD,
Davisson RL, Salmon JE. Targeted inhibition of complement activation
prevents features of preeclampsia in mice. Kidney Int. 2011;79:331–339.
doi: 10.1038/ki.2010.393
Agostinis C, Bulla R, Tripodo C, Gismondi A, Stabile H, Bossi
F, Guarnotta C, Garlanda C, De Seta F, Spessotto P, Santoni A,
Ghebrehiwet B, Girardi G, Tedesco F. An alternative role of C1q in
cell migration and tissue remodeling: contribution to trophoblast invasion and placental development. J Immunol. 2010;185:4420–4429. doi:
10.4049/jimmunol.0903215
Singh J, Ahmed A, Girardi G. Role of complement component C1q in
the onset of preeclampsia in mice. Hypertension. 2011;58:716–724. doi:
10.1161/HYPERTENSIONAHA.111.175919
Fang CJ, Fremeaux-Bacchi V, Liszewski MK, Pianetti G, Noris M,
Goodship TH, Atkinson JP. Membrane cofactor protein mutations in
150.
151.
152.
153.
154.
155.
156.
157.
158.
159.
160.
161.
162.
163.
164.
165.
166.
167.
168.
169.
atypical hemolytic uremic syndrome (aHUS), fatal Stx-HUS, C3 glomerulonephritis, and the HELLP syndrome. Blood. 2008;111:624–632. doi:
10.1182/blood-2007-04-084533
Kavanagh D, Richards A, Atkinson J. Complement regulatory genes and
hemolytic uremic syndromes. Annu Rev Med. 2008;59:293–309. doi:
10.1146/annurev.med.59.060106.185110
Zipfel PF, Misselwitz J, Licht C, Skerka C. The role of defective complement control in hemolytic uremic syndrome. Semin Thromb Hemost.
2006;32:146–154. doi: 10.1055/s-2006-939770
Zipfel PF, Skerka C. Complement dysfunction in hemolytic
uremic syndrome. Curr Opin Rheumatol. 2006;18:548–555. doi:
10.1097/01.bor.0000240370.47336.ae
Burwick RM, Feinberg BB. Eculizumab for the treatment of preeclampsia/HELLP syndrome. Placenta. 2013;34:201–203. doi:
10.1016/j.placenta.2012.11.014
Legendre CM, Licht C, Muus P, et al. Terminal complement inhibitor
eculizumab in atypical hemolytic-uremic syndrome. N Engl J Med.
2013;368:2169–2181. doi: 10.1056/NEJMoa1208981
Fakhouri F, Hourmant M, Campistol JM, Cataland SR, Espinosa M,
Gaber AO, Menne J, Minetti EE, Provôt F, Rondeau E, Ruggenenti P,
Weekers LE, Ogawa M, Bedrosian CL, Legendre CM. Terminal complement inhibitor eculizumab in adult patients with atypical hemolytic
uremic syndrome: a single-arm, open-label trial. Am J Kidney Dis.
2016;68:84–93. doi: 10.1053/j.ajkd.2015.12.034
Demir E, Yazici H, Ozluk Y, Kilicaslan I, Turkmen A. Pregnant woman
with atypical hemolytic uremic syndrome delivered a healthy newborn
under eculizumab treatment. Case Rep Nephrol Dial. 2016;6:143–148.
doi: 10.1159/000454946
Gupta M, Feinberg BB, Burwick RM. Thrombotic microangiopathies of
pregnancy: differential diagnosis. Pregnancy Hypertens. 2018;12:29–34.
doi: 10.1016/j.preghy.2018.02.007
Irani RA, Xia Y. The functional role of the renin-angiotensin system
in pregnancy and preeclampsia. Placenta. 2008;29:763–771. doi:
10.1016/j.placenta.2008.06.011
Brown MA, Wang J, Whitworth JA. The renin-angiotensin-aldosterone
system in pre-eclampsia. Clin Exp Hypertens. 1997;19:713–726. doi:
10.3109/10641969709083181
Gant NF, Daley GL, Chand S, Whalley PJ, MacDonald PC. A study of
angiotensin II pressor response throughout primigravid pregnancy. J Clin
Invest. 1973;52:2682–2689. doi: 10.1172/JCI107462
Wallukat G, Homuth V, Fischer T, Lindschau C, Horstkamp B, Jüpner
A, Baur E, Nissen E, Vetter K, Neichel D, Dudenhausen JW, Haller H,
Luft FC. Patients with preeclampsia develop agonistic autoantibodies
against the angiotensin AT1 receptor. J Clin Invest. 1999;103:945–952.
doi: 10.1172/JCI4106
Zhou CC, Zhang Y, Irani RA, Zhang H, Mi T, Popek EJ, Hicks MJ, Ramin
SM, Kellems RE, Xia Y. Angiotensin receptor agonistic autoantibodies
induce pre-eclampsia in pregnant mice. Nat Med. 2008;14:855–862. doi:
10.1038/nm.1856
LaMarca B, Parrish M, Ray LF, Murphy SR, Roberts L, Glover P,
Wallukat G, Wenzel K, Cockrell K, Martin JN Jr, Ryan MJ, Dechend R.
Hypertension in response to autoantibodies to the angiotensin II type I
receptor (AT1-AA) in pregnant rats: role of endothelin-1. Hypertension.
2009;54:905–909. doi: 10.1161/HYPERTENSIONAHA.109.137935
Yang X, Wang F, Lau WB, Zhang S, Zhang S, Liu H, Ma XL.
Autoantibodies isolated from preeclamptic patients induce endothelial dysfunction via interaction with the angiotensin II AT1 receptor.
Cardiovasc Toxicol. 2014;14:21–29. doi: 10.1007/s12012-013-9229-8
Dechend R, Homuth V, Wallukat G, Kreuzer J, Park JK, Theuer J,
Juepner A, Gulba DC, Mackman N, Haller H, Luft FC. AT(1) receptor
agonistic antibodies from preeclamptic patients cause vascular cells to
express tissue factor. Circulation. 2000;101:2382–2387.
Xia Y, Wen H, Bobst S, Day MC, Kellems RE. Maternal autoantibodies from preeclamptic patients activate angiotensin receptors on human
trophoblast cells. J Soc Gynecol Investig. 2003;10:82–93.
Dechend R, Viedt C, Müller DN, et al. AT1 receptor agonistic antibodies from preeclamptic patients stimulate NADPH oxidase. Circulation.
2003;107:1632–1639. doi: 10.1161/01.CIR.0000058200.90059.B1
LaMarca B, Wallukat G, Llinas M, Herse F, Dechend R, Granger JP.
Autoantibodies to the angiotensin type I receptor in response to placental
ischemia and tumor necrosis factor alpha in pregnant rats. Hypertension.
2008;52:1168–1172. doi: 10.1161/HYPERTENSIONAHA.108.120576
Parrish MR, Murphy SR, Rutland S, Wallace K, Wenzel K, Wallukat G,
Keiser S, Ray LF, Dechend R, Martin JN, Granger JP, LaMarca B. The
effect of immune factors, tumor necrosis factor-alpha, and agonistic
1110 Circulation Research March 29, 2019
170.
171.
172.
173.
174.
175.
176.
177.
178.
179.
Downloaded from http://ahajournals.org by on February 7, 2021
180.
181.
182.
183.
184.
185.
186.
187.
188.
189.
190.
autoantibodies to the angiotensin II type I receptor on soluble fms-like tyrosine-1 and soluble endoglin production in response to hypertension during
pregnancy. Am J Hypertens. 2010;23:911–916. doi: 10.1038/ajh.2010.70
Herse F, LaMarca B. Angiotensin II type 1 receptor autoantibody
(AT1-AA)-mediated pregnancy hypertension. Am J Reprod Immunol.
2013;69:413–418. doi: 10.1111/aji.12072
Quitterer U, Fu X, Pohl A, Bayoumy KM, Langer A, AbdAlla S. Betaarrestin1 prevents preeclampsia by downregulation of mechanosensitive
AT1-B2 receptor heteromers. Cell. 2019;176:318.e19–333.e19. doi:
10.1016/j.cell.2018.10.050
Zhou A, Carrell RW, Murphy MP, Wei Z, Yan Y, Stanley PL, Stein
PE, Broughton Pipkin F, Read RJ. A redox switch in angiotensinogen modulates angiotensin release. Nature. 2010;468:108–111. doi:
10.1038/nature09505
Burke SD, Zsengellér ZK, Khankin EV, et al. Soluble fms-like tyrosine kinase 1 promotes angiotensin II sensitivity in preeclampsia. J Clin
Invest. 2016;126:2561–2574. doi: 10.1172/JCI83918
Schobel HP, Fischer T, Heuszer K, Geiger H, Schmieder RE.
Preeclampsia – a state of sympathetic overactivity. N Engl J Med.
1996;335:1480–1485. doi: 10.1056/NEJM199611143352002
Reyes LM, Usselman CW, Davenport MH, Steinback CD. Sympathetic
nervous system regulation in human normotensive and hypertensive pregnancies. Hypertension. 2018;71:793–803. doi: 10.1161/
HYPERTENSIONAHA.117.10766
Molino P, Veglio F, Genova GC, Melchio R, Benedetto C, Chiarolini L,
Rabbia F, Grosso T, Mulatero P, Chiandussi L. Baroreflex control of heart
rate is impaired in pre-eclampsia. J Hum Hypertens. 1999;13:179–183.
Cleary KL, Siddiq Z, Ananth CV, Wright JD, Too G, DʼAlton ME,
Friedman AM. Use of antihypertensive medications during delivery hospitalizations complicated by preeclampsia. Obstet Gynecol.
2018;131:441–450. doi: 10.1097/AOG.0000000000002479
Hines T, Beauchamp D, Rice C. Baroreflex control of sympathetic nerve activity in hypertensive pregnant rats with reduced
uterine perfusion. Hypertens Pregnancy. 2007;26:303–314. doi:
10.1080/10641950701415598
Spradley FT, Ge Y, Haynes BP, Granger JP, Anderson CD. Adrenergic
receptor blockade attenuates placental ischemia-induced hypertension.
Physiol Rep. 2018;6:e13814. doi: 10.14814/phy2.13814
Aubuchon M, Schulz LC, Schust DJ. Preeclampsia: animal models
for a human cure. Proc Natl Acad Sci USA. 2011;108:1197–1198. doi:
10.1073/pnas.1018164108
Crews JK, Herrington JN, Granger JP, Khalil RA. Decreased endothelium-dependent vascular relaxation during reduction of uterine perfusion
pressure in pregnant rat. Hypertension. 2000;35:367–372.
LaMarca B, Amaral LM, Harmon AC, Cornelius DC, Faulkner JL,
Cunningham MW Jr. Placental ischemia and resultant phenotype in
animal models of preeclampsia. Curr Hypertens Rep. 2016;18:38. doi:
10.1007/s11906-016-0633-x
Davisson RL, Hoffmann DS, Butz GM, Aldape G, Schlager G, Merrill
DC, Sethi S, Weiss RM, Bates JN. Discovery of a spontaneous genetic
mouse model of preeclampsia. Hypertension. 2002;39:337–342.
Bulla R, Agostinis C, Bossi F, Rizzi L, Debeus A, Tripodo C, Radillo O,
De Seta F, Ghebrehiwet B, Tedesco F. Decidual endothelial cells express
surface-bound C1q as a molecular bridge between endovascular trophoblast and decidual endothelium. Mol Immunol. 2008;45:2629–2640. doi:
10.1016/j.molimm.2007.12.025
Sandgren JA, Deng G, Linggonegoro DW, et al. Arginine vasopressin
infusion is sufficient to model clinical features of preeclampsia in mice.
JCI Insight. 2018;3.
Takimoto E, Ishida J, Sugiyama F, Horiguchi H, Murakami K, Fukamizu
A. Hypertension induced in pregnant mice by placental renin and maternal angiotensinogen. Science. 1996;274:995–998.
Zhang J, Chen Z, Smith GN, Croy BA. Natural killer cell-triggered vascular transformation: maternal care before birth? Cell Mol Immunol.
2011;8:1–11. doi: 10.1038/cmi.2010.38
Hutcheon JA, Lisonkova S, Joseph KS. Epidemiology of pre-eclampsia
and the other hypertensive disorders of pregnancy. Best Pract Res Clin
Obstet Gynaecol. 2011;25:391–403. doi: 10.1016/j.bpobgyn.2011.01.006
Stepan H, Kuse-Föhl S, Klockenbusch W, Rath W, Schauf B, Walther
T, Schlembach D. Diagnosis and treatment of hypertensive pregnancy
disorders. guideline of DGGG (S1-Level, AWMF registry no. 015/018,
December 2013). Geburtshilfe Frauenheilkd. 2015;75:900–914. doi:
10.1055/s-0035-1557924
Kaitu’u-Lino TJ, Brownfoot FC, Beard S, Cannon P, Hastie R, Nguyen
TV, Binder NK, Tong S, Hannan NJ. Combining metformin and
191.
192.
193.
194.
195.
196.
197.
198.
199.
200.
201.
202.
203.
204.
205.
206.
207.
esomeprazole is additive in reducing sFlt-1 secretion and decreasing endothelial dysfunction - implications for treating preeclampsia. PLoS One.
2018;13:e0188845. doi: 10.1371/journal.pone.0188845
Kalafat E, Sukur YE, Abdi A, Thilaganathan B, Khalil A. Metformin for
the prevention of hypertensive disorders of pregnancy in women with
gestational diabetes and obesity: a systematic review and meta-analysis.
Ultrasound Obstet Gynecol. 2018;52:706–714.
Tangren JS, Wan Md Adnan WAH, Powe CE, Ecker J, Bramham K,
Hladunewich MA, Ankers E, Karumanchi SA, Thadhani R. Risk of
preeclampsia and pregnancy complications in women with a history of acute kidney injury. Hypertension. 2018;72:451–459. doi:
10.1161/HYPERTENSIONAHA.118.11161
Committee opinion no. 638: first-trimester risk assessment for earlyonset preeclampsia. Obstet Gynecol. 2015;126:e25–e27.
Odibo AO, Goetzinger KR, Odibo L, Cahill AG, Macones GA, Nelson
DM, Dietzen DJ. First-trimester prediction of preeclampsia using
metabolomic biomarkers: a discovery phase study. Prenat Diagn.
2011;31:990–994. doi: 10.1002/pd.2822
Bahado-Singh RO, Syngelaki A, Akolekar R, Mandal R, Bjondahl TC,
Han B, Dong E, Bauer S, Alpay-Savasan Z, Graham S, Turkoglu O,
Wishart DS, Nicolaides KH. Validation of metabolomic models for prediction of early-onset preeclampsia. Am J Obstet Gynecol. 2015;213:530.
e1–530.e10. doi: 10.1016/j.ajog.2015.06.044
Kelly RS, Croteau-Chonka DC, Dahlin A, et al. Integration of metabolomic and transcriptomic networks in pregnant women reveals biological pathways and predictive signatures associated with preeclampsia.
Metabolomics. 2017;13:7.
Bahado-Singh R, Poon LC, Yilmaz A, Syngelaki A, Turkoglu O, Kumar
P, Kirma J, Allos M, Accurti V, Li J, Zhao P, Graham SF, Cool DR,
Nicolaides K. Integrated proteomic and metabolomic prediction of term
preeclampsia. Sci Rep. 2017;7:16189. doi: 10.1038/s41598-017-15882-9
O’Gorman N, Wright D, Poon LC, et al. Multicenter screening for preeclampsia by maternal factors and biomarkers at 11-13 weeks’ gestation: comparison with NICE guidelines and ACOG recommendations.
Ultrasound Obstet Gynecol. 2017;49:756–760. doi: 10.1002/uog.17455
Rolnik DL, Wright D, Poon LC, et al. Aspirin versus placebo in pregnancies at high risk for preterm preeclampsia. N Engl J Med. 2017;377:613–
622. doi: 10.1056/NEJMoa1704559
Agrawal S, Cerdeira AS, Redman C, Vatish M. Meta-analysis and
systematic review to assess the role of soluble FMS-like tyrosine
kinase-1 and placenta growth factor ratio in prediction of preeclampsia: the SaPPPhirE Study. Hypertension. 2018;71:306–316. doi:
10.1161/HYPERTENSIONAHA.117.10182
Chaiworapongsa T, Romero R, Korzeniewski SJ, Cortez JM, Pappas A,
Tarca AL, Chaemsaithong P, Dong Z, Yeo L, Hassan SS. Plasma concentrations of angiogenic/anti-angiogenic factors have prognostic value in
women presenting with suspected preeclampsia to the obstetrical triage
area: a prospective study. J Matern Fetal Neonatal Med. 2014;27:132–
144. doi: 10.3109/14767058.2013.806905
Chaiworapongsa T, Chaemsaithong P, Korzeniewski SJ, Yeo L, Romero
R. Pre-eclampsia part 2: prediction, prevention and management. Nat
Rev Nephrol. 2014;10:531–540. doi: 10.1038/nrneph.2014.103
Rana S, Salahuddin S, Mueller A, Berg AH, Thadhani RI, Karumanchi
SA. Angiogenic biomarkers in triage and risk for preeclampsia
with severe features. Pregnancy Hypertens. 2018;13:100–106. doi:
10.1016/j.preghy.2018.05.008
Chappell LC, Duckworth S, Seed PT, Griffin M, Myers J, Mackillop L,
Simpson N, Waugh J, Anumba D, Kenny LC, Redman CW, Shennan
AH. Diagnostic accuracy of placental growth factor in women with
suspected preeclampsia: a prospective multicenter study. Circulation.
2013;128:2121–2131. doi: 10.1161/CIRCULATIONAHA.113.003215
Zeisler H, Llurba E, Chantraine F, Vatish M, Staff AC, Sennström M,
Olovsson M, Brennecke SP, Stepan H, Allegranza D, Dilba P, Schoedl
M, Hund M, Verlohren S. Predictive value of the sFlt-1:PlGF ratio in
women with suspected preeclampsia. N Engl J Med. 2016;374:13–22.
doi: 10.1056/NEJMoa1414838
Zeisler H, Llurba E, Chantraine FJ, Vatish M, Staff AC, Sennstrom M,
Olovsson M, Brennecke SP, Stepan H, Allegranza D, Schoedl M, Grill S,
Hund M, Verlohren S. The sFlt-1/PlGF Ratio: ruling out pre-eclampsia
for up to 4 weeks and the value of retesting [published online July 16,
2018]. Ultrasound Obstet Gynecol. doi: 10.1002/uog.19178
Stepan H, Herraiz I, Schlembach D, Verlohren S, Brennecke S, Chantraine
F, Klein E, Lapaire O, Llurba E, Ramoni A, Vatish M, Wertaschnigg D,
Galindo A. Implementation of the sFlt-1/PlGF ratio for prediction and
diagnosis of pre-eclampsia in singleton pregnancy: implications for
Rana et al Preeclampsia and Vascular Disease 1111
208.
209.
210.
211.
212.
213.
214.
215.
216.
217.
Downloaded from http://ahajournals.org by on February 7, 2021
218.
219.
220.
221.
222.
223.
224.
225.
226.
clinical practice. Ultrasound Obstet Gynecol. 2015;45:241–246. doi:
10.1002/uog.14799
Manriquez Rocha B, Mbofana F, Loquiha O, Mudenyanga C,
Ukah UV, Magee LA, von Dadelszen P. Early diagnosis of preeclampsia using placental growth factor: an operational pilot study in
Maputo, Mozambique. Pregnancy Hypertens. 2018;11:26–31. doi:
10.1016/j.preghy.2017.12.005
Bellomo G, Venanzi S, Saronio P, Verdura C, Narducci PL.
Prognostic significance of serum uric acid in women with gestational hypertension. Hypertension. 2011;58:704–708. doi: 10.1161/
HYPERTENSIONAHA.111.177212
Vyakaranam S, Bhongir AV, Patlolla D, Chintapally R. Study of
serum uric acid and creatinine in hypertensive disorders of pregnancy. Int J Med Sci Public Health. 2015;4:1424–1428. doi:
10.5455/ijmsph.2015.15042015294
Wu Y, Xiong X, Fraser WD, Luo ZC. Association of uric acid with progression to preeclampsia and development of adverse conditions in gestational hypertensive pregnancies. Am J Hypertens. 2012;25:711–717.
doi: 10.1038/ajh.2012.18
August P, Helseth G, Cook EF, Sison C. A prediction model for
superimposed preeclampsia in women with chronic hypertension during pregnancy. Am J Obstet Gynecol. 2004;191:1666–1672. doi:
10.1016/j.ajog.2004.03.029
Weerasekera DS, Peiris H. The significance of serum uric acid, creatinine
and urinary microprotein levels in predicting pre-eclampsia. J Obstet
Gynaecol. 2003;23:17–19.
Massé J, Forest JC, Moutquin JM, Marcoux S, Brideau NA, Bélanger
M. A prospective study of several potential biologic markers for early
prediction of the development of preeclampsia. Am J Obstet Gynecol.
1993;169:501–508.
Chen Q, Lau S, Tong M, Wei J, Shen F, Zhao J, Zhao M. Serum uric
acid may not be involved in the development of preeclampsia. J Hum
Hypertens. 2016;30:136–140. doi: 10.1038/jhh.2015.47
Lam C, Lim KH, Kang DH, Karumanchi SA. Uric acid and preeclampsia.
Semin Nephrol. 2005;25:56–60.
Magee LA, Duley L. Oral beta-blockers for mild to moderate
hypertension during pregnancy. Cochrane Database Syst Rev. 2003:
CD002863.
Magee LA, von Dadelszen P, Rey E, et al. Less-tight versus tight control
of hypertension in pregnancy. N Engl J Med. 2015;372:407–417. doi:
10.1056/NEJMoa1404595
Caritis S, Sibai B, Hauth J, Lindheimer MD, Klebanoff M, Thom
E, VanDorsten P, Landon M, Paul R, Miodovnik M, Meis P, Thurnau
G. Low-dose aspirin to prevent preeclampsia in women at high risk.
National Institute of Child Health and Human Development Network of
Maternal-Fetal Medicine Units. N Engl J Med. 1998;338:701–705. doi:
10.1056/NEJM199803123381101
Sharp A, Cornforth C, Jackson R, Harrold J, Turner MA, Kenny LC,
Baker PN, Johnstone ED, Khalil A, von Dadelszen P, Papageorghiou AT,
Alfirevic Z, STRIDER group. Maternal sildenafil for severe fetal growth
restriction (STRIDER): a multicentre, randomised, placebo-controlled,
double-blind trial. Lancet Child Adolesc Health. 2018;2:93–102. doi:
10.1016/S2352-4642(17)30173-6
Pels A, Kenny LC, Alfirevic Z, Baker PN, von Dadelszen P, Gluud C,
Kariya CT, Mol BW, Papageorghiou AT, van Wassenaer-Leemhuis
AG, Ganzevoort W, Groom KM; international STRIDER Consortium.
STRIDER (Sildenafil TheRapy in dismal prognosis early onset fetal
growth restriction): an international consortium of randomised placebo-controlled trials. BMC Pregnancy Childbirth. 2017;17:440. doi:
10.1186/s12884-017-1594-z
Hawkes N. Trial of Viagra for fetal growth restriction is halted after baby
deaths. BMJ. 2018;362:k3247. doi: 10.1136/bmj.k3247
Girardi G. Pravastatin to treat and prevent preeclampsia. Preclinical
and clinical studies. J Reprod Immunol. 2017;124:15–20. doi:
10.1016/j.jri.2017.09.009
Ramma W, Ahmed A. Therapeutic potential of statins and the induction
of heme oxygenase-1 in preeclampsia. J Reprod Immunol. 2014;101102:153–160. doi: 10.1016/j.jri.2013.12.120
Brownfoot FC, Tong S, Hannan NJ, Binder NK, Walker SP,
Cannon P, Hastie R, Onda K, Kaitu’u-Lino TJ. Effects of pravastatin on human placenta, endothelium, and women with severe preeclampsia. Hypertension. 2015;66:687–697; discussion 445. doi:
10.1161/HYPERTENSIONAHA.115.05445
Lefkou E, Mamopoulos A, Dagklis T, Vosnakis C, Rousso D,
Girardi G. Pravastatin improves pregnancy outcomes in obstetric
227.
228.
229.
230.
231.
232.
233.
234.
235.
236.
237.
238.
239.
240.
241.
242.
243.
244.
antiphospholipid syndrome refractory to antithrombotic therapy. J Clin
Invest. 2016;126:2933–2940. doi: 10.1172/JCI86957
Chaiworapongsa T, Romero R, Korzeniewski SJ, Chaemsaithong P,
Hernandez-Andrade E, Segars JH, DeCherney AH, McCoy MC, Kim
CJ, Yeo L, Hassan SS. Pravastatin for the prevention of adverse pregnancy outcome: preeclampsia and more? J Matern Fetal Neonatal Med.
2017;30:3. doi: 10.3109/14767058.2015.1129779
Brownfoot FC, Hastie R, Hannan NJ, Cannon P, Tuohey L, Parry LJ,
Senadheera S, Illanes SE, Kaitu’u-Lino TJ, Tong S. Metformin as a
prevention and treatment for preeclampsia: effects on soluble fmslike tyrosine kinase 1 and soluble endoglin secretion and endothelial
dysfunction. Am J Obstet Gynecol. 2016;214:356.e1–356.e15. doi:
10.1016/j.ajog.2015.12.019
Spaulonci CP, Bernardes LS, Trindade TC, Zugaib M, Francisco RP.
Randomized trial of metformin vs insulin in the management of gestational diabetes. Am J Obstet Gynecol. 2013;209:34.e1–34.e7. doi:
10.1016/j.ajog.2013.03.022
Rana S, Rajakumar A, Geahchan C, Salahuddin S, Cerdeira AS, Burke
SD, George EM, Granger JP, Karumanchi SA. Ouabain inhibits placental
sFlt1 production by repressing HSP27-dependent HIF-1α pathway.
FASEB J. 2014;28:4324–4334. doi: 10.1096/fj.14-252684
Klingel R, Göhlen B, Schwarting A, Himmelsbach F, Straube R.
Differential indication of lipoprotein apheresis during pregnancy. Ther
Apher Dial. 2003;7:359–364.
Thadhani R, Kisner T, Hagmann H, et al. Pilot study of extracorporeal removal of soluble fms-like tyrosine kinase 1 in preeclampsia. Circulation.
2011;124:940–950. doi: 10.1161/CIRCULATIONAHA.111.034793
Thadhani R, Hagmann H, Schaarschmidt W, et al. Removal of soluble
Fms-like tyrosine kinase-1 by dextran sulfate apheresis in preeclampsia.
J Am Soc Nephrol. 2016;27:903–913. doi: 10.1681/ASN.2015020157
Makris A, Yeung KR, Lim SM, Sunderland N, Heffernan S, Thompson
JF, Iliopoulos J, Killingsworth MC, Yong J, Xu B, Ogle RF, Thadhani
R, Karumanchi SA, Hennessy A. Placental growth factor reduces
blood pressure in a uteroplacental ischemia model of preeclampsia
in nonhuman primates. Hypertension. 2016;67:1263–1272. doi:
10.1161/HYPERTENSIONAHA.116.07286
Spradley FT, Tan AY, Joo WS, Daniels G, Kussie P, Karumanchi SA,
Granger JP. Placental growth factor administration abolishes placental
ischemia-induced hypertension. Hypertension. 2016;67:740–747. doi:
10.1161/HYPERTENSIONAHA.115.06783
Weissgerber TL, Rajakumar A, Myerski AC, Edmunds LR, Powers RW,
Roberts JM, Gandley RE, Hubel CA. Vascular pool of releasable soluble
VEGF receptor-1 (sFLT1) in women with previous preeclampsia and
uncomplicated pregnancy. J Clin Endocrinol Metab. 2014;99:978–987.
doi: 10.1210/jc.2013-3277
Turanov AA, Lo A, Hassler MR, et al. RNAi modulation of placental
sFLT1 for the treatment of preeclampsia [published online November 19,
2018]. Nat Biotechnol. doi: 10.1038/nbt.4297
Sibai BM. Etiology and management of postpartum hypertension-preeclampsia. Am J Obstet Gynecol. 2012;206:470–475. doi:
10.1016/j.ajog.2011.09.002
Pearson GD, Veille JC, Rahimtoola S, Hsia J, Oakley CM, Hosenpud
JD, Ansari A, Baughman KL. Peripartum cardiomyopathy: National
Heart, Lung, and Blood Institute and Office of Rare Diseases (National
Institutes of Health) workshop recommendations and review. JAMA.
2000;283:1183–1188.
Patten IS, Rana S, Shahul S, et al. Cardiac angiogenic imbalance
leads to peripartum cardiomyopathy. Nature. 2012;485:333–338. doi:
10.1038/nature11040
Uraizee I, Cheng S, Moslehi J. Reversible cardiomyopathy associated
with sunitinib and sorafenib. N Engl J Med. 2011;365:1649–1650. doi:
10.1056/NEJMc1108849
Wang CN, Chang SD, Peng HH, Lee YS, Chang YL, Cheng PJ, Chao
AS, Wang TH, Wang HS. Change in amniotic fluid levels of multiple
anti-angiogenic proteins before development of preeclampsia and intrauterine growth restriction. J Clin Endocrinol Metab. 2010;95:1431–1441.
doi: 10.1210/jc.2009-1954
Hansen AR, Barnés CM, Folkman J, McElrath TF. Maternal preeclampsia predicts the development of bronchopulmonary dysplasia. J
Pediatr. 2010;156:532–536. doi: 10.1016/j.jpeds.2009.10.018
Tang JR, Karumanchi SA, Seedorf G, Markham N, Abman SH. Excess
soluble vascular endothelial growth factor receptor-1 in amniotic fluid
impairs lung growth in rats: linking preeclampsia with bronchopulmonary dysplasia. Am J Physiol Lung Cell Mol Physiol. 2012;302:L36–
L46. doi: 10.1152/ajplung.00294.2011
1112 Circulation Research March 29, 2019
Downloaded from http://ahajournals.org by on February 7, 2021
245. Yu XD, Branch DW, Karumanchi SA, Zhang J. Preeclampsia and retinopathy of prematurity in preterm births. Pediatrics. 2012;130:e101–
e107. doi: 10.1542/peds.2011-3881
246. Wallace B, Peisl A, Seedorf G, Nowlin T, Kim C, Bosco J, Kenniston
J, Keefe D, Abman SH. Anti-sFlt-1 therapy preserves lung alveolar and vascular growth in antenatal models of bronchopulmonary dysplasia. Am J Respir Crit Care Med. 2018;197:776–787. doi:
10.1164/rccm.201707-1371OC
247. Newstead J, von Dadelszen P, Magee LA. Preeclampsia and future cardiovascular risk. Expert Rev Cardiovasc Ther. 2007;5:283–294. doi:
10.1586/14779072.5.2.283
248. Brouwers L, van der Meiden-van Roest AJ, Savelkoul C, Vogelvang
TE, Lely AT, Franx A, van Rijn BB. Recurrence of pre-eclampsia
and the risk of future hypertension and cardiovascular disease: a systematic review and meta-analysis. BJOG. 2018;125:1642–1654. doi:
10.1111/1471-0528.15394
249. Berends AL, de Groot CJ, Sijbrands EJ, Sie MP, Benneheij SH, Pal R,
Heydanus R, Oostra BA, van Duijn CM, Steegers EA. Shared constitutional risks for maternal vascular-related pregnancy complications and
future cardiovascular disease. Hypertension. 2008;51:1034–1041. doi:
10.1161/HYPERTENSIONAHA.107.101873
250. McDonald SD, Malinowski A, Zhou Q, Yusuf S, Devereaux PJ.
Cardiovascular sequelae of preeclampsia/eclampsia: a systematic review and meta-analyses. Am Heart J. 2008;156:918–930. doi:
10.1016/j.ahj.2008.06.042
251. Shahul S, Ramadan H, Nizamuddin J, Mueller A, Patel V, Dreixler J,
Tung A, Lang RM, Weinert L, Nasim R, Chinthala S, Rana S. Activin
A and late postpartum cardiac dysfunction among women with hypertensive disorders of pregnancy. Hypertension. 2018;72:188–193. doi:
10.1161/HYPERTENSIONAHA.118.10888
252. Shahul S, Ramadan H, Mueller A, Nizamuddin J, Nasim R, Lopes
Perdigao J, Chinthala S, Tung A, Rana S. Abnormal mid-trimester
cardiac strain in women with chronic hypertension predates superimposed preeclampsia. Pregnancy Hypertens. 2017;10:251–255. doi:
10.1016/j.preghy.2017.10.009
253. Shahul S, Rhee J, Hacker MR, Gulati G, Mitchell JD, Hess P, Mahmood
F, Arany Z, Rana S, Talmor D. Subclinical left ventricular dysfunction
in preeclamptic women with preserved left ventricular ejection fraction: a 2D speckle-tracking imaging study. Circ Cardiovasc Imaging.
2012;5:734–739. doi: 10.1161/CIRCIMAGING.112.973818
254. Breetveld NM, Ghossein-Doha C, van Kuijk SM, van Dijk AP, van der
Vlugt MJ, Heidema WM, van Neer J, van Empel V, Brunner-La Rocca
HP, Scholten RR, Spaanderman ME. Prevalence of asymptomatic heart
failure in formerly pre-eclamptic women: a cohort study. Ultrasound
Obstet Gynecol. 2017;49:134–142. doi: 10.1002/uog.16014
255. Vikse BE, Irgens LM, Leivestad T, Skjaerven R, Iversen BM.
Preeclampsia and the risk of end-stage renal disease. N Engl J Med.
2008;359:800–809. doi: 10.1056/NEJMoa0706790
256. Vikse BE, Irgens LM, Karumanchi SA, Thadhani R, Reisæter AV,
Skjærven R. Familial factors in the association between preeclampsia
and later ESRD. Clin J Am Soc Nephrol. 2012;7:1819–1826. doi:
10.2215/CJN.01820212
257. Bushnell C, Chireau M. Preeclampsia and stroke: risks during and after pregnancy. Stroke Res Treat. 2011;2011:858134. doi:
10.4061/2011/858134
258. Basit S, Wohlfahrt J, Boyd HA. Pre-eclampsia and risk of dementia
later in life: nationwide cohort study. BMJ. 2018;363:k4109. doi:
10.1136/bmj.k4109
259. Barker DJ, Osmond C. Infant mortality, childhood nutrition, and ischaemic heart disease in England and Wales. Lancet. 1986;1:1077–1081.
260. Davis EF, Lazdam M, Lewandowski AJ, Worton SA, Kelly B, Kenworthy
Y, Adwani S, Wilkinson AR, McCormick K, Sargent I, Redman C, Leeson
P. Cardiovascular risk factors in children and young adults born to preeclamptic pregnancies: a systematic review. Pediatrics. 2012;129:e1552–
e1561. doi: 10.1542/peds.2011-3093
261. Jayet PY, Rimoldi SF, Stuber T, Salmòn CS, Hutter D, Rexhaj E,
Thalmann S, Schwab M, Turini P, Sartori-Cucchia C, Nicod P, Villena M,
Allemann Y, Scherrer U, Sartori C. Pulmonary and systemic vascular dysfunction in young offspring of mothers with preeclampsia. Circulation.
2010;122:488–494. doi: 10.1161/CIRCULATIONAHA.110.941203
262. Hoodbhoy Z, Hasan BS, Mohammed N, Chowdhury D. Impact of preeclampsia on the cardiovascular health of the offspring: a cohort study protocol. BMJ Open. 2018;8:e024331. doi: 10.1136/bmjopen-2018-024331
263. Brown MC, Best KE, Pearce MS, Waugh J, Robson SC, Bell R.
Cardiovascular disease risk in women with pre-eclampsia: systematic review and meta-analysis. Eur J Epidemiol. 2013;28:1–19. doi:
10.1007/s10654-013-9762-6
264. Rolnik DL, Wright D, Poon LCY, et al. ASPRE trial: performance of
screening for preterm pre-eclampsia. Ultrasound Obstet Gynecol.
2017;50:492–495. doi: 10.1002/uog.18816
Download