You are on page 1of 13

JOURNAL OF THE AMERICAN COLLEGE OF CARDIOLOGY VOL. 76, NO.

14, 2020

ª 2020 BY THE AMERICAN COLLEGE OF CARDIOLOGY FOUNDATION

PUBLISHED BY ELSEVIER

THE PRESENT AND FUTURE

JACC STATE-OF-THE-ART REVIEW

Preeclampsia—Pathophysiology and
Clinical Presentations
JACC State-of-the-Art Review

Christopher W. Ives, MD,a Rachel Sinkey, MD,b,c Indranee Rajapreyar, MD,d Alan T.N. Tita, MD, PHD,b,c
Suzanne Oparil, MDd

ABSTRACT

Preeclampsia is a hypertensive disorder of pregnancy. It affects 2% to 8% of pregnancies worldwide and causes sig-
nificant maternal and perinatal morbidity and mortality. Hypertension and proteinuria are the cornerstone of the disease,
though systemic organ dysfunction may ensue. The clinical syndrome begins with abnormal placentation with subsequent
release of antiangiogenic markers, mediated primarily by soluble fms-like tyrosine kinase-1 (sFlt-1) and soluble endoglin
(sEng). High levels of sFlt-1 and sEng result in endothelial dysfunction, vasoconstriction, and immune dysregulation,
which can negatively impact every maternal organ system and the fetus. This review comprehensively examines the
pathogenesis of preeclampsia with a specific focus on the mechanisms underlying the clinical features. Delivery is the
only definitive treatment. Low-dose aspirin is recommended for prophylaxis in high-risk populations. Other treatment
options are limited. Additional research is needed to clarify the pathophysiology, and thus, identify potential therapeutic
targets for improved treatment and, ultimately, outcomes of this prevalent disease.
(J Am Coll Cardiol 2020;76:1690–702) © 2020 by the American College of Cardiology Foundation.

P reeclampsia is a hypertensive disorder of


pregnancy (HDP). It impacts 2% to 8% of
all pregnancies and is a major cause of
maternal and perinatal morbidity and mortality
cornerstone of the syndrome and is often, but not
always, accompanied by proteinuria. Severe forms
of preeclampsia can be complicated by renal, car-
diac, pulmonary, hepatic, and neurological dysfunc-
(1–3). In the United States, HDP were responsible tion; hematologic disturbances; fetal growth
for 212 (7%) of approximately 3,000 pregnancy- restriction; stillbirth; and maternal death (3,7)
related deaths between 2011 and 2015 (4). Pre- (Table 1). Underlying mechanisms contributing to
eclampsia is a complex disease process originating the pathophysiology of preeclampsia are poorly un-
at the maternal–fetal interface that affects derstood, though this is an active area of interna-
multiple organ systems (5,6). Hypertension is the tional research (5).

From the aTinsley Harrison Internal Medicine Residency Program, Department of Medicine, University of Alabama at Birmingham,
Birmingham, Alabama; bDivision of Maternal-Fetal Medicine, Department of Obstetrics & Gynecology, University of Alabama at
Birmingham, Birmingham, Alabama; cCenter for Women’s Reproductive Health, University of Alabama at Birmingham, Bir-
Listen to this manuscript’s mingham, Alabama; and the dDivision of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham,
audio summary by Birmingham, Alabama. Dr. Sinkey has received a grant from GestVision. Dr. Oparil has received grants and nonfinancial support
Editor-in-Chief from NIH/National Heart, Lung, and Blood Institute during the conduct of the study; has received personal fees from Preventric
Dr. Valentin Fuster on Diagnostics, Inc.; has received personal fees and other from CinCor Pharma Inc. outside the submitted work; and is Editor-in-Chief
JACC.org. of Current Hypertension Reports (Springer Science Business Media LLC). All other authors have reported that they have no
relationships relevant to the contents of this paper to disclose.
The authors attest they are in compliance with human studies committees and animal welfare regulations of the authors’
institutions and Food and Drug Administration guidelines, including patient consent where appropriate. For more information,
visit the JACC author instructions page.

Manuscript received May 8, 2020; revised manuscript received July 14, 2020, accepted August 3, 2020.

ISSN 0735-1097/$36.00 https://doi.org/10.1016/j.jacc.2020.08.014


JACC VOL. 76, NO. 14, 2020 Ives et al. 1691
OCTOBER 6, 2020:1690–702 Pathophysiology of Preeclampsia

dysfunction, a vasoconstrictive state, oxida- ABBREVIATIONS


HIGHLIGHTS AND ACRONYMS
tive stress, and microemboli that contribute
 Preeclampsia, a hypertensive disorder of to the involvement of multiple organ sys-
Ang II = angiotensin II
pregnancy, affects 2% to 8% of pregnant tems, and thus, the clinical features of pre-
AT1R = angiotensin II receptor
women and causes considerable eclampsia (8,9,12). It is also likely that pre-
type 1
mortality. existing endothelial stress, such as
BP = blood pressure
increased sympathetic nervous system tone
 Pre-existing cardiovascular disease likely HDP = hypertensive disorders
from reduced intravascular volume, may of pregnancy
plays a role in the development of
further predispose to development of pre-
IL = interleukin
preeclampsia.
eclampsia (2).
RAAS = renin-angiotensin-
 Delivery is the only definitive treatment. In addition to endothelial dysfunction, aldosterone system

Low-dose aspirin is recommended for immunologic aberrations contribute to the sEng = soluble endoglin
prophylaxis in high-risk women. preeclampsia phenotype. In normal preg-
sFlt = soluble fms-like tyrosine
nancy, T helper cells shift toward the anti- kinase
 Further research is needed to identify
inflammatory Th2 phenotype, which helps Stat3 = signal transducer and
therapies that reduce maternal and
to neutralize proinflammatory cytokines, activator or transcription3
neonatal morbidity and mortality.
angiotensin II type 1 receptor (AT1R) auto- TGF = transforming growth

antibodies, placental reactive oxygen species, factor

PREECLAMPSIA OVERVIEW and endothelin-1 (2). However, in pre- VEGF = vascular endothelial
growth factor
eclampsia, T helper cells shift toward the Th1
Preeclampsia is defined as new-onset hypertension phenotype, increasing release of proinflammatory
and new-onset end-organ damage, including pro- cytokines such as interleukin (IL)-12 and IL-18, and
teinuria, after 20 weeks of gestation (Table 2) (3,7). decreasing IL-10, which leads to apoptosis and
The pathophysiology of this complex process in- reduced trophoblast invasion (13). Increased
volves multiple organ systems and is summarized in CD19 þCD5 þ B lymphocytes may contribute to pro-
the Central Illustration. The clinical syndrome begins duction of antiangiogenic factors. Uterine natural
with abnormal trophoblast invasion before many killer cells, which differ from peripheral natural killer
women know they are pregnant, and long before cells, are likely involved, because inhibition of uter-
clinical manifestations of the disease become ine natural killer cells may lead to defective spiral
apparent (6,8). During normal implantation, tropho- artery remodeling. Syncytial knots, vesicles that shed
blasts invade the decidualized endometrium, leading from trophoblasts, may stimulate an inflammatory
to spiral artery remodeling and obliteration of the response in the placenta (2). LIN28 is an RNA binding
tunica media of myometrial spiral arteries, allowing protein that affects cell metabolism, differentiation,
increased blood flow to the placenta, all independent growth, and invasion. Two paralogs exist: LIN28A and
of maternal vasomotor changes (9). In preeclampsia, LIN28B. LIN28B is increased in extravillous tropho-
trophoblasts fail to adopt an endothelial phenotype, blasts/placenta in normal pregnancy. In preeclamp-
which leads to impaired trophoblast invasion and sia, levels are decreased in the placenta, suggesting a
incomplete spiral artery remodeling (6). The resultant role in preeclampsia by reducing trophoblast differ-
placental ischemia leads to an increase in angiogenic entiation and invasion, and by promoting inflamma-
markers such as soluble fms-like tyrosine kinase-1 tion (14). Elevated complement levels in
(sFlt-1) and soluble endoglin (sEng) (6,10). sFlt-1 has preeclampsia result in complement system dysregu-
been proposed as an underlying mechanism to lation and additional increases in sFlt-1 (2). Women
explain disease in the maternal and fetal units. sFlt-1 with preeclampsia have reduced histocompatibility
binds to and decreases levels of vascular endothelial complex human leukocyte antigen-G and -E, also
growth factor (VEGF) and placental growth factor, suggestive of immune imbalance (13).
which are important mediators of endothelial cell Multiple genetic components have been implicated
function, especially in fenestrated endothelium in the pathogenesis of preeclampsia. Mutations in
(brain, liver, glomeruli) (6,8,10,11). Thus, endothelial complement component 3 are associated with pre-
dysfunction develops in maternal vasculature (10). eclampsia, which may partly account for complement
sEng is a cell surface coreceptor that binds to and system dysregulation (2). Corin, a cardiac protein that
decreases levels of transforming growth factor (TGF)- activates atrial natriuretic peptide, has also been
b, which normally induces migration and prolifera- localized to uterine tissue, and mutations in corin
tion of endothelial cells (8,11). These factors associated with preeclampsia have been identified
mediate downstream effects that create endothelial (2,15). Global transcriptional profiling of chorionic
1692 Ives et al. JACC VOL. 76, NO. 14, 2020

Pathophysiology of Preeclampsia OCTOBER 6, 2020:1690–702

T A B L E 1 Highlights of Clinical Features of Preeclampsia

Clinical Feature Underlying Abnormalities Clinical Consequences

Hypertension Increased SVR and afterload Heart failure


Decreased CO and intravascular volumes Pulmonary edema
Activation of RAAS, ET-1, SNS Renal dysfunction
AT1R down-regulated, placental hypoxia, and AT1R autoantibodies Neurological injury
Increased vasoconstrictors, decreased vasodilators
Increased sFlt-1 and sEng, oxidative stress
Proteinuria Glomerular endotheliosis Hypertension
Disruption of filtration barrier Ischemic heart disease
Increased tubular permeability Stroke
Chronic kidney disease
End-stage renal disease
Renal dysfunction Decreased RBF and GFR Hypertension
Glomerular endotheliosis Chronic kidney disease
Increased tissue factor expression End-stage renal disease
Thrombotic microangiopathy
Neurological abnormalities Headache: loss of fenestrae on choroid plexus, periventricular edema, Seizures
vasogenic edema in posterior cerebral circulation PRES
Visual disturbances: retinopathy, retinal detachment, cortical blindness, Permanent blindness
central serous chorioretinopathy, hypertensive retinopathy, diabetic
retinopathy
Eclampsia Unknown (potentially vasogenic or cytotoxic edema) Permanent neurological dysfunction
Cardiac dysfunction Increased SVR, afterload Heart failure
Concentric LV hypertrophy, LA enlargement Peripartum cardiomyopathy
Increased RVSP, increased LV filling pressures, LV diastolic dysfunction,
Pulmonary edema Increased vascular permeability Acute hypoxemic respiratory failure
Cardiac dysfunction
Corticosteroids/tocolytics
Iatrogenic volume overload
Hepatic dysfunction Hepatic microcirculatory deterioration, hepatocellular injury Liver failure, hepatic rupture
Hematologic dysfunction Procoagulant state Thrombocytopenia, DIC
Fetal growth restriction Incomplete spiral artery remodeling Fetal growth <10th percentile
Decidual vasculopathy
Uterine and placental dysfunction

This table depicts the key aspects underlying the pathophysiology of each clinical feature of preeclampsia. Subsequent clinical consequences that can be observed with each
feature are listed.
AT1R ¼ angiotensin II receptor type 1; CO ¼ cardiac output; DIC ¼ disseminated intravascular coagulation; ET ¼ endothelin; GFR ¼ glomerular filtration rate; LA ¼ left atrial;
LV ¼ left ventricular; PRES ¼ posterior reversible encephalopathy syndrome; RAAS ¼ renin-angiotensin-aldosterone system; RBF ¼ renal blood flow; RVSP ¼ right ventricular
systolic pressure; sEng ¼ soluble endoglin; sFlt ¼ soluble fms-like tyrosine kinase; SNS ¼ sympathetic nervous system; SVR ¼ systemic vascular resistance.

villus samples of women with preeclampsia suggests mutations of placental growth factor, sFlt-1, or sEng
a genetic susceptibility to preeclampsia (2,16). Endo- genes have been identified to date (13). However,
metrial stromal cells from nonpregnant women with a next-generation sequencing, or massive parallel
history of severe preeclampsia failed to decidualize sequencing, enables affordable analysis of large
in vitro and were transcriptionally inert, and decidual genomic regions and is a promising tool for studying
tissue from women with preeclampsia revealed de- genetic influence on preeclampsia (20).
fects in gene expression (17). Long-chain L-3 Lastly, certain acquired risk factors have been
hydroxyacyl-CoA dehydrogenase deficiency, a disor- shown to increase risk of preeclampsia. Obesity (body
der of fatty acid metabolism, has been associated with mass index >30) and diabetes each carries a relative
preeclampsia (18). One study (19) found that women risk increase of 3.5 (2). Chronic hypertension, chronic
who develop preeclampsia are more likely to carry kidney disease, obstructive sleep apnea, pre-
loss-of-function mutations in 43 genes associated gestational diabetes, systemic lupus erythematosus,
with both idiopathic dilated and peripartum cardio- antiphospholipid syndrome, rheumatoid arthritis,
myopathy. The TTN gene, which encodes the sarco- maternal age over 35 years, nulliparity, multifetal
meric protein titin, had the highest frequency of gestations, fetal hydrops, hydatidiform moles, and
mutations (19). Genome-wide association studies assisted reproductive technologies are also associated
have identified both single nucleotide polymorphism with preeclampsia (2,3,13,21–23). Though abnormal
rs4769613, near the FMS-like tyrosine kinase 1 gene, placentation may drive the more immediate devel-
and Rs9478812, within protein PLEKHGI, which is opment of preeclampsia, the preceding information
implicated in blood pressure (BP) regulation, to be suggests an underlying role of pre-existing cardio-
associated with preeclampsia (2). No causal vascular and other organ dysfunction before
JACC VOL. 76, NO. 14, 2020 Ives et al. 1693
OCTOBER 6, 2020:1690–702 Pathophysiology of Preeclampsia

T A B L E 2 Diagnostic Criteria for Preeclampsia

Always necessary. . .
Hypertension
 SBP $140 mm Hg or DBP $90 mm Hg on 2 occasions at least 4 h apart after 20 weeks’ gestation in a woman with previously normal BP
 SBP $160 mm Hg or DBP $110 mm Hg on 1 occasion
. . .And 1 of the following
Proteinuria
 $300 mg per 24-h urine collection (or extrapolated from timed collection), or
 Protein/creatinine ratio of $0.3 mg/dl, or
 Dipstick reading of 2þ (used only when other methods not available)
OR any 1 of the following (in the absence of proteinuria)
Thrombocytopenia
 Platelet count <100,000/mm 3
Renal insufficiency
 Serum creatinine concentration >1.1 mg/dl or a doubling of serum creatinine concentration in the absence of other renal disease
Impaired liver function
 Elevated concentration of liver transaminases to 2 normal
 Severe persistent right upper quadrant or epigastric pain unresponsive to medication
Pulmonary edema
 Diagnosed by physical examination or chest x-ray
Neurological signs
 New-onset headache unresponsive to medication and not accounted for by alternative diagnoses or visual symptoms
 Visual disturbances
Fetal growth restriction*
 Estimated fetal weight <10th percentile

Adapted from ACOG Practice Bulletin No. 202 (3) and ISSHP recommendations (7). *Included in ISSHP definition of preeclampsia though not in ACOG definition.
ACOG ¼ American College of Obstetricians and Gynecologists; BP ¼ blood pressure; DBP ¼ diastolic blood pressure; ISSHP ¼ International Society for the Study of Hypertension
in Pregnancy; SBP ¼ systolic blood pressure.

conception. Additionally, preeclampsia is associated and aldosterone are lower in preeclampsia compared
with long-term consequences following delivery, with normal pregnancy (though still higher than
particularly cardiovascular effects. Studies demon- nonpregnant individuals), and sensitivity to Ang II
strate prior occurrence of preeclampsia to be associ- and norepinephrine is increased (6). The reasons for
ated with higher rates of hypertension, ischemic hypertensive complications in preeclampsia despite
heart disease/recurrent acute coronary syndrome, lower levels of components of RAAS are 2-fold. First,
heart failure, stroke, and death (24–28). Further study in normal pregnancy, AT1R is down-regulated by
into abnormalities in cardiovascular structure and reactive oxygen species. In preeclampsia, AT1R com-
function may identify potential areas for clinical plexes with bradykinin receptor B2 to form a
intervention to reduce disease risk and burden. The heterodimer that enhances the pressor effects of
following discussion reviews the current under- Ang II (29). Second, placental hypoxia contributes to
standing of the pathophysiology of each clinical production of circulating antibodies to AT1R, which in
manifestation of preeclampsia (highlights in Table 1). turn increases vasoconstriction via activation of
endothelin-1, increased sensitivity to circulating
HYPERTENSION Ang II, and increased placental production of sFlt-1
and sEng (30).
Hypertension in pregnancy is defined as elevated Down-regulation of heme oxygenase-1 results in
systolic BP $140 mm Hg and/or diastolic decreased carbon monoxide generation, which then
BP $90 mm Hg on 2 measurements 4 h apart at rest leads to further increases in sFlt-1 and sEng release
(3). Hypertension is a necessary diagnostic criterion (31). sFlt-1 increases peripheral vascular resistance,
for the preeclampsia syndrome (3). Compared with which in turn increases BP (32). Activation of the
normotensive pregnant control patients, hyperten- sympathetic nervous system, RAAS, and endothelin-1
sion in preeclampsia occurs in the setting of increased in an attempt to correct the relative hemoconcentra-
systemic vascular resistance and afterload, and tion in preeclampsia increases vasoconstrictors, such
decreased cardiac output and intravascular volumes. as thromboxane A1 and endothelins, and decreases
Multiple factors contribute to this change. Conduit vasodilators, such as prostacyclin and nitric oxide,
artery compliance decreases, and the usual fall in creating intense vasoconstriction (3,31,33). These de-
nocturnal BP is mitigated or obliterated (11). Despite rangements result in endothelial dysfunction and
activation of the renin-angiotensin-aldosterone sys- attenuate endothelium-dependent vasodilation due
tem (RAAS), levels of renin, angiotensin II (Ang II), to oxidative stress (9,11,31). An observational study
1694 Ives et al. JACC VOL. 76, NO. 14, 2020

Pathophysiology of Preeclampsia OCTOBER 6, 2020:1690–702

C E NT R AL IL L U STR AT IO N Pathogenesis of Preeclampsia

Ives, C.W. et al. J Am Coll Cardiol. 2020;76(14):1690–702.

Acquired, genetic, and immune risk factors contribute to early placental dysfunction (Stage 1). Placental dysfunction results in release of anti-angiogenic factors,
leading to later multiorgan dysfunction (Stage 2). Solid arrows represent progression of disease. Dashed arrows represent SNS effect on respective organs.
Ang II ¼ angiotensin II; ER ¼ endoplasmic reticulum; HA ¼ headache; HIF ¼ hypoxia-inducible transcription factor; HIF ¼ hypoxia-inducible transcription factor;
NO ¼ nitric oxide; PlGF ¼ placental growth factor; PRES ¼ posterior reversible encephalopathy syndrome; RAAS ¼ renin-angiotensin-aldosterone system;
sEng ¼ soluble endoglin; sFlt ¼ soluble fms-like tyrosine kinase; SNS ¼ sympathetic nervous system; TGF ¼ transforming growth factor; VEGF ¼ vascular endothelial
growth factor.

(34) of 205 women with preeclampsia in the preeclampsia (8,11,31,32). Glomerular endotheliosis is
Netherlands found that hypertension can take up to 2 characterized by swollen, vacuolated endothelial
years to resolve following delivery. Both the severity cells with fibrils, swollen mesangial cells, sub-
of preeclampsia and time to delivery were positively endothelial deposits of protein reabsorbed from the
associated with time to resolution (34). Hypertension glomerular filtrate, and tubular casts (3). There is an
in the setting of preeclampsia contributes to the enlarged bloodless glomerulus with an obliterated
development of target organ damage, including heart capillary lumen (usually not accompanied by promi-
failure, pulmonary edema, renal dysfunction and nent capillary thrombi as in thrombotic micro-
acute kidney injury, and neurological injury and angiopathy) (31). High sFlt-1 levels inhibit podocyte-
stroke (3,8,35). specific VEGF, disturbing the glomerular filtration
barrier and resulting in formation of fenestrae
PROTEINURIA contributing to proteinuria (36). The damage to
podocytes is largely responsible for the proteinuria.
Proteinuria in the setting of preeclampsia is the result Slit diaphragm proteins (nephrin, podocin, synapto-
of increased renal tubular permeability to most large- podin, podocalyxin) play a key role in maintaining
molecular-weight proteins, such as albumin, glob- glomerular barrier integrity, and detection of these
ulin, transferrin, and hemoglobin (3). High circulating proteins in the urine precedes the clinical features of
sFlt-1 and decreased nitric oxide are both involved in preeclampsia by several weeks, indicating that dam-
mediating renal tubular injury in the setting of pre- age to these proteins leads to the development of
eclampsia (6,9). sFlt-1 inhibition of VEGF also causes further proteinuria (31). Urine samples from pre-
glomerular endothelial injury, a process termed eclamptic patients have increased levels of proteins
glomerular endotheliosis that is pathognomonic for involved in the coagulation, complement, RAAS, and
JACC VOL. 76, NO. 14, 2020 Ives et al. 1695
OCTOBER 6, 2020:1690–702 Pathophysiology of Preeclampsia

cell adhesion pathways (37). Proteins detected in the aura are associated with preeclampsia (40). Second-
urine of preeclamptic patients are also often mis- ary headache accounts for 35% of headache in preg-
folded (9). It is likely that imbalance between proan- nancy. HDPs, most commonly preeclampsia, are the
giogenic and antiangiogenic factors leads to podocyte most frequent cause of secondary headache and
injury and that this damage contributes to the higher become more common as gestational age increases
risk of later hypertension, chronic kidney disease, (40,41). Use of nifedipine for severe hypertension and
ischemic heart disease, stroke, persistent proteinuria, intravenous magnesium sulfate for eclampsia pro-
and end-stage renal disease seen in patients with phylaxis may also cause headache (41). The charac-
preeclampsia (31). Proteinuria can take up to 2 years teristic preeclampsia headache is progressive,
to resolve following preeclampsia, with the severity bilateral (frontal or occipital), pulsating/throbbing,
of preeclampsia and time to delivery positively associated with visual changes, worse with higher BP,
associated with time to resolution (34). aggravated by physical activity, and unresponsive to
over-the-counter medications (40,41). Symptoms can
RENAL DYSFUNCTION
also be vague and typical of tension-type headache
(41). The characteristic posterior reversible encepha-
Renal dysfunction in preeclampsia is defined as
lopathy syndrome headache is bilateral, occipital,
serum creatinine >1.1 mg/dl or a doubling of baseline
dull, and with no prodrome (40). One theory of the
creatinine (38). Renal blood flow and glomerular
pathophysiology of headache in preeclampsia is that
filtration rate are often decreased in preeclampsia
blocking VEGF and TGF- b leads to loss of fenestrae on
(11). Biopsy changes in these patients include diffuse
the choroid plexus, resulting in endothelial cell
fibrin deposition, endothelial swelling, loss of podo-
instability and periventricular edema (5). These
cytes, and loss of capillary space (glomerular endo-
changes may then precipitate seizures and posterior
theliosis) (6,8). Dysregulation of the glomerular
reversible encephalopathy syndrome, defined by
filtration apparatus occurs in the setting of glomer-
neurological abnormalities with neuroimaging find-
ular endotheliosis (6). In normal pregnancy,
ings of vasogenic edema in the distribution of the
increased tissue factor release from the maternal
posterior cerebral circulation (5,42).
decidua and placenta shifts endothelial cells to a
Visual disturbance in preeclampsia may be due to
procoagulant balance (36). Increased proin-
retinopathy, retinal detachment, or cortical blind-
flammatory cytokines in preeclampsia further stimu-
ness, which typically resolves following delivery
late tissue factor expression by endothelial cells and
(35,43). Central serous chorioretinopathy occurs as
leukocytes (8,39). Damaged endothelial cells then
fluid accumulates behind the retina, leading to
further induce clotting and lose anticoagulant ability
detachment (44). It is thought to arise from hormonal
as prostaglandin and nitric oxide levels decrease,
fluctuations, such as progesterone level changes (45).
leading to thrombotic microangiopathy in the kidneys
Hypertensive retinopathy is a condition of retinal
(36). Increased toll-like receptor 4 leads to increased
microvascular damage secondary to elevated blood
inflammatory cytokines, which in turn increase both
pressure (46). It results from severe vascular spasm in
placental and renal dysfunction (10). Electrolyte ab-
the setting of the angiogenic imbalance of pre-
normalities occur as urinary calcium decreases due to
eclampsia (45,46). In central serous chorioretinop-
increased tubular calcium reabsorption (3). Reduction
athy and hypertensive retinopathy, delivery results
in intravascular volumes in preeclampsia increases
in spontaneous resolution of subretinal fluid, with
sodium and free-water retention (3). Lastly, the same
generally good outcomes, so these conditions are not
mechanisms that trigger hypertension, particularly
emergent indications for delivery (45). By contrast,
involving sFlt-1 and the RAAS system, predispose to
diabetic retinopathy can progress quickly in preg-
renal dysfunction and acute kidney injury, which in-
nancy and up to 1 year postpartum, so close moni-
crease later risk of hypertension, chronic kidney dis-
toring and treatment with laser photocoagulation
ease, and end-stage renal disease (8,29,31).
after progression to severe pre-proliferative diabetic
NEUROLOGICAL DYSFUNCTION retinopathy is recommended (44,45). However,
regression to a prior state of retinopathy can occur in
Preeclampsia may lead to multiple neurological the postpartum period (44). Retinal artery occlusion
problems, including headache, visual disturbances, can also occur and is associated with Protein S defi-
seizure, posterior reversible encephalopathy syn- ciency, elevated factor VIII, and primary anti-
drome, and hemorrhagic stroke (35,38). Multiple phospholipid antibody syndrome (45). Cortical
variants of primary headache including tension type blindness is vision loss due to lesions of the occipital
headache, migraine without aura, and migraine with cortex, possibly due to cerebral edema. It generally
1696 Ives et al. JACC VOL. 76, NO. 14, 2020

Pathophysiology of Preeclampsia OCTOBER 6, 2020:1690–702

resolves in hours to days (44). Though some visual Identification of an imaging finding or reliable
disturbances are temporary, others can result in per- biomarker may provide insight into the otherwise
manent visual disturbance or blindness despite unknown mechanism of eclampsia and may allow
prompt clinical recognition and management. clinicians to target at-risk patients for closer moni-
toring and/or prophylactic treatment.
ECLAMPSIA
CARDIAC DYSFUNCTION
Eclampsia is defined as new-onset tonic-clonic, focal,
or multifocal seizures in the setting of HDP in the Cardiac output increases by 30% to 50% in the first 2
absence of other causes (3). Although progesterone trimesters of normal pregnancy then plateaus after
raises the seizure threshold, estrogen lowers the 20 weeks. The increase in cardiac output is accom-
seizure threshold via down-regulation of gamma plished by a heart rate increase, 50% increase in
aminobutyric acid (8). A Cochrane review of medica- plasma volume, and to compensate for the increased
tions for preeclampsia found that intravenous mag- intravascular volume, transient left ventricular
nesium sulfate reduced the risk of eclampsia by 59%, eccentric hypertrophy. These changes fully recover
superior to phenytoin (47). It is unknown why mag- postpartum (8). In preeclampsia, the increased after-
nesium sulfate works or why it is more effective than load from higher vascular resistance created by
other medications, though it appears to be through impaired placentation leads to worsening of left
mechanisms other than anticonvulsant properties. ventricular remodeling, resulting in mild-to-
The mechanism may be related to mitigating the moderate isolated left ventricular diastolic dysfunc-
endothelial injury underlying preeclampsia (48). tion with concentric left ventricular hypertrophy (8).
Patients with eclampsia who undergo magnetic Whereas eccentric remodeling is a physiological
resonance imaging typically have findings suggestive compensatory response, concentric remodeling in-
of posterior reversible encephalopathy syndrome. dicates an abnormal cardiac response to the increased
However, these findings were seen in areas of the systemic vascular resistance seen in preeclampsia.
brain other than the posterior cerebrum (42). One study (50) examined echocardiographic findings
Eclampsia can develop with systolic BP <160 mm Hg, in women with acute preeclampsia and found higher
suggesting that it may not be BP alone that drives right ventricular systolic pressures, increased left
eclampsia but also endothelial dysfunction (42). atrial size, increased left ventricular wall thickness,
There are 2 proposed pathogenic mechanisms: vaso- diastolic dysfunction, and increased left ventricular
genic and cytotoxic edema. Vasogenic edema may filling pressures. Another study (51) used cardiovas-
result from a sudden rise in BP over 150 mm Hg, cular magnetic resonance imaging and found that
which increases cerebral blood flow, causing hyper- postpartum women with preeclampsia had left atrial
perfusion and edema as intrinsic mechanisms to enlargement compared with the control group of
autoregulate cerebral perfusion are overwhelmed. postpartum women without preeclampsia. HDP in-
Cytotoxic edema is thought to be due to profound crease the risk of peripartum cardiomyopathy, a
vasospasm from excessive cerebrovascular regulation serious complication of pregnancy (52,53). Peri-
in an attempt to correct overperfusion. It has been partum cardiomyopathy is defined as reduced left
suggested that damage from vasogenic edema may be ventricular ejection fraction (<45%) toward the end
reversible, whereas cytotoxic edema may be irre- of pregnancy or in the first few months postpartum
versible, resulting in permanent neurological and is a diagnosis of exclusion (8,54). It occurs in 1 in
dysfunction (42). 1,000 pregnancies worldwide and in 1 in about 3,000
New multidisciplinary studies are investigating live births in the United States (53,54).
novel methods of diagnosing neurological dysfunc- The balance between antioxidant capacity and
tion in preeclampsia (49). Magnetic resonance imag- oxidative stress is upset in peripartum cardiomyopa-
ing can reveal vasogenic edema, thought to occur as a thy, which is worsened by preeclampsia, likely due to
result of a combination of vasoconstriction, impaired the higher levels of sFlt-1 (8,54,55). The antioxidant
autoregulation, and endothelial dysfunction, as well signaling pathway, which is protective in normal
as posterior reversible encephalopathy syndrome. pregnancies, involves mitochondrial superoxide dis-
Additionally, biomarkers, such as S100 calcium- mutase 2 and includes peroxisome proliferator-
binding protein B, neuron-specific enolase, neuro- activated receptor g coactivator 1-a and signal trans-
filament light chain, and tau, are being studied as ducer and activator of transcrition3 (Stat3) (54).
possible diagnostic techniques to identify patients at Peroxisome proliferator-activated receptor g coac-
risk of developing preeclampsia/eclampsia (49). tivator 1- a , a transcriptional coactivator important in
JACC VOL. 76, NO. 14, 2020 Ives et al. 1697
OCTOBER 6, 2020:1690–702 Pathophysiology of Preeclampsia

F I G U R E 1 Pathophysiology of Cardiac Dysfunction in Preeclampsia

Preexisting CVD and subsequent impaired placentation drive an angiogenic imbalance and disrupt normal cardiac changes in pregnancy.
Concentric LVH and/or PPCM may develop, which increase risk for later CVD complications. CO ¼ cardiac output; CVD ¼ cardiovascular
disease; ET ¼ endothelin; HR ¼ heart rate; LA ¼ left atrial; LV ¼ left ventricular; LVH ¼ left ventricular hypertrophy; PPARg ¼ peroxisome
proliferator-activated receptor g coactivator; PPCM ¼ peripartum cardiomyopathy; RVSP ¼ right ventricular systolic pressure; sEng ¼ soluble
endoglin; sFlt ¼ soluble fms-like tyrosine kinase; Stat3 ¼ signal transducer and activator of transcrition3; SVR ¼ systemic vascular resis-
tance; VEGF ¼ vascular endothelial growth factor.

mitochondrial biogenesis, is highly expressed in the vasorelaxation, worsening endothelial dysfunction


heart and is also important in angiogenesis, as it in- (54). Female mice with a cardiomyocyte-specific
creases VEGF (55). Mice lacking peroxisome deletion of Stat3 develop peripartum cardiomyopa-
proliferator-activated receptor g coactivator 1-a have thy (56). Additionally, Stat3 levels have been found to
been shown to have abnormal cardiac energy re- be decreased in preeclamptic placentas of humans
serves, suggesting involvement in the pathogenesis (Figure 1) (58). Given the high prevalence and asso-
of peripartum cardiomyopathy (55). Stat3 is a protein ciated morbidity of cardiac dysfunction in pre-
that up-regulates antioxidative enzymes, promotes eclampsia, further study of the underlying
myocardial angiogenesis, and can mediate car- mechanisms, and thus, potential preventative stra-
diomyocyte hypertrophy (56,57). Expression and tegies and novel therapies is warranted to reduce
activation of Stat3 is decreased in the placentas of morbidity in afflicted women.
preeclampsia rat models (57). Further, Stat3 deletion
in rat models leads to increased cathepsin D, which PULMONARY EDEMA
cleaves the proangiogenic 23-kDa prolactin protein
into the antiangiogenic 16-kDa prolactin (54,56); 16- Pulmonary edema in preeclampsia is rare (0.6% to
kDa prolactin disrupts capillaries and blocks 0.7%) (59) but carried significant morbidity and
1698 Ives et al. JACC VOL. 76, NO. 14, 2020

Pathophysiology of Preeclampsia OCTOBER 6, 2020:1690–702

mortality in 1 study (60). It can occur antepartum or dysfunction in preeclampsia and to assess its long-
postpartum (60). Generally, there are 4 components term effects.
of pulmonary edema in preeclampsia: increased
HEMATOLOGIC DISTURBANCE
vascular permeability, cardiac dysfunction, cortico-
steroids/tocolytics, and iatrogenic volume overload.
The most common hematologic disturbances are
The increased vascular permeability results from
thrombocytopenia and disseminated intravascular
endothelial damage of preeclampsia coupled with the
coagulopathy, a disruption of the clotting cascade
decreased colloid osmotic pressure of pregnancy (60).
leading to intravascular coagulation accompanied by
The combination of diastolic dysfunction and
secondary fibrinolysis (8). Thrombocytopenia
increased vascular resistance increase hydrostatic
(platelets <100,000) is likely due to increased platelet
forces in the pulmonary vasculature (8,60). Admin-
activation, aggregation, and consumption (3,38).
istration of corticosteroids to women with pre-
Pregnancy is a procoagulant state because of in-
eclampsia to reduce neonatal complications of
creases in fibrinogen and other clotting factors, and
prematurity can result in significant maternal pul-
decreases in anticoagulants (Protein C and S) (8). It
monary edema, although by an unknown mechanism
has been hypothesized that syncytiotrophoblasts
(61,62). However, these patients are also generally
shed extracellular vessels that augment platelet
receiving tocolytics or magnesium sulfate, making it
activation, leading to release of soluble factors and
difficult to decipher which predisposes to pulmonary
extracellular vesicles, which might contribute to
edema (60,61). Many women with preeclampsia
placental and systemic microvascular ischemia (64).
receive large volumes of intravenous fluids, leading
An imbalance in angiogenic factors, specifically sFlt-1
to volume overload, especially with mobilization of
and sEng, may also be involved (65). Another theory
fluids postpartum (60). Further, the edema may be
is that sudden activation of the vascular endothelial
worsened by renal retention of salt and water (31).
cascade leads to the release of von Willebrand factor
Though rare, the morbidity and mortality associated
multimers that bind platelets, causing excessive
with pulmonary edema in preeclampsia necessitates a
platelet aggregation and subsequent thrombus for-
deeper understanding of its pathogenesis and an
mation in the microcirculation, resulting in
attempt to limit iatrogenic insults which may pro-
consumptive thrombocytopenia, hemolytic anemia,
mote its development.
and hepatic dysfunction (8,65). This phenomenon
usually resolves 6 to 7 days postpartum (8). Decreased
nitric oxide levels also contribute to qualitative
HEPATIC DYSFUNCTION
platelet dysfunction (9). Possible mechanisms of
disseminated intravascular coagulopathy include:
Hepatic dysfunction in preeclampsia is defined as
consumption coagulopathy, hepatic injury (decreased
transaminases $2 the upper limit of normal and
clotting factor production), and/or systemic maternal
persistent severe right upper quadrant or epigastric
inflammatory response (8). It is important to monitor
tenderness (38). Aspartate aminotransferase is usu-
women with preeclampsia for hematologic abnor-
ally higher than alanine aminotransferase in pre-
malities to support them through this potentially
eclampsia, as aspartate aminotransferase is related to
deadly feature of preeclampsia.
periportal necrosis (3). However, alanine amino-
transferase should not be tested in isolation, because FETAL GROWTH RESTRICTION/
at least 1 case has been reported of alanine amino- FETAL IMPLICATIONS
transferase deficiency in the setting of a HDP diag-
nosis (63). Preeclampsia also causes elevation in Preeclampsia leads to uterine and placental
lactate dehydrogenase and alterations in hepatic dysfunction, which causes fetal growth restriction,
synthetic function, resulting in abnormalities in pro- defined as an estimated fetal weight <10th percentile
thrombin time, partial thromboplastin time, and for gestational age (66). As spiral arteries fail to
fibrinogen (3). Liver failure and hepatic rupture can develop appropriately and lead to incomplete pseu-
occur. Both are related to endothelial dysfunction, dovasculogenesis, placental vascular insults such as
which causes hepatic microcirculatory deterioration placental infarcts occur (3,5,11). Incomplete spiral
and hepatocellular necrosis (8). Decreased expression artery remodeling leads to atherosis of maternal
of endothelial nitric oxide synthase resulting from radial arteries (lipid-laden macrophages in the lumen,
sFlt-1 antagonism of VEGF also leads to liver fibrinoid necrosis in the wall, and mononuclear peri-
dysfunction and thrombocytopenia (8). Further study vascular infiltrate) (2). Decidual vasculopathy,
is needed to better detect and quantify hepatic including loose, edematous endothelium,
JACC VOL. 76, NO. 14, 2020 Ives et al. 1699
OCTOBER 6, 2020:1690–702 Pathophysiology of Preeclampsia

F I G U R E 2 Interventions to Reduce Preeclampsia

No Benefit
Bed rest
Benefit Potential
Fish oil
Delivery Pravastatin
Folic acid
Aspirin Metformin
Garlic
Calcium* Exercise
Sodium restriction
Vitamins C, D, E

Overview of evidence of treatments to reduce risk of preeclampsia. Interventions with proven benefit are delivery for treatment, and aspirin
and calcium for prophylaxis. Pravastatin, metformin, and exercise are currently being investigated and are showing promise. *Only in
nutritional deficiency in low-middle income countries.

hypertrophy of the vessel media, loss of smooth PREVENTION


muscle modifications, and up-regulation of hypoxia-
inducible transcription factor-1 a , results from these A concerted effort has been made to reduce the
changes (2). Structural changes of the glycocalyx and burden of preeclampsia (Figure 2). Unfortunately, bed
hyaluronic acid are also seen (10). Inhibition of TGF-b rest, fish oil, folic acid, garlic, sodium restriction, vi-
by sEng also leads to impaired endothelial vasodila- tamins C and E, and vitamin D studies have not
tion (5). These changes cause impaired diastolic translated to clinical benefit (68–74). The only defin-
placental flow on ultrasound and placental ischemia itive treatment for preeclampsia is delivery, but daily
(2,10). The resultant ischemia can lead to decidual aspirin is recommended by the American College of
and placental endoplasmic reticulum stress and Obstetricians and Gynecologists (75), the United
further oxidative stress (2). States Preventive Services Task Force (76), and the
Biopsies of preeclamptic placentas confirm International Society for the Study of Hypertension in
shallow invasion of trophoblasts and failure of spi- Pregnancy (7) for high-risk women after 12 weeks
ral artery remodeling. Evaluation of the decidua gestation to reduce their risk of preeclampsia.
shows decreased prolactin and insulin-like growth Aspirin is proposed to reduce preeclampsia risk via
factor binding protein (both decidualization inhibition of cyclooxygenase-1 and cyclooxygenase-2,
markers). Decidual cells in vitro have also been which contribute to prostaglandin biosynthesis and
shown to fail to promote cytotrophoblast invasion subsequent endothelial dysfunction (75,76). A recent
(possibly due to reduced nitric oxide) (13). Women Cochrane review (77) summarized evidence from 60
with uncomplicated pregnancies who develop trials including 36,716 women and found an 18%
adverse outcomes in later pregnancies are more reduced risk of preeclampsia with aspirin prophylaxis
likely to have maternal vascular malperfusion le- (relative risk: 0.82,; 95% confidence interval: 0.77 to
sions in the placentas from the initial, uncompli- 0.88).
cated pregnancy. These placentas are also more Exercise is recommended by the International So-
likely to have a decidual vasculopathy (67). Taken ciety for the Study of Hypertension in Pregnancy for
together, these factors contribute to fetal growth preeclampsia prevention, because they cite a low risk
restriction that commonly occurs in pregnancies of adverse events and potential benefit (7). The So-
complicated by preeclampsia. ciety recommends following the protocol of a
1700 Ives et al. JACC VOL. 76, NO. 14, 2020

Pathophysiology of Preeclampsia OCTOBER 6, 2020:1690–702

previous study (78) of 50 min at least 3 days per week CONCLUSIONS


of exercise, which was associated with reduced
weight gain and incidence of HDP (7). However, a Despite the proven benefit of aspirin and other
systematic review and meta-analysis (79) found ex- promising therapies under development for its pre-
ercise in overweight or obese women to reduce vention, preeclampsia remains a leading cause of
weight gain, but not incidence of HDP. Another morbidity and mortality in the developing and
strategy to reduce preeclampsia is calcium supple- developed world. Shallow trophoblast invasion and
mentation, though this applies mainly to women with incomplete transformation of spiral arteries during
nutritional deficiency or in low- to middle-income placentation are the hallmarks of preeclampsia.
countries (7,80). A meta-analysis of 13 trials (15,730 Recent evidence supporting shared risk factors for
women) reported a significant reduction in pre- preeclampsia and cardiovascular disease suggest a
eclampsia with calcium supplementation, with the role for pre-existing cardiovascular disease in the
greatest effect among women with low-baseline cal- development of HDP (87). The physiological stress of
cium intake (80). A 2018 meta-analysis update preeclampsia may then more promptly unmask car-
including 27 trials (18,064 women) confirmed similar diovascular disease in women with a history of the
results (81). A proposed mechanism is that hypocal- disorder. In acknowledgement of these associations,
cemia may stimulate parathyroid hormone or renin the American Heart Association recently issued a
release, which may increase intracellular calcium in statement (88) that provides guidance on cardiovas-
vascular smooth muscle, resulting in vasoconstriction cular conditions in women of reproductive age and
and higher blood pressure (80,81). Calcium supple- urges clinicians to maintain an understanding of
mentation may reduce parathyroid release, thereby cardiovascular disease during pregnancy. Little is
reducing intracellular calcium and smooth muscle known about the mechanisms underlying most clin-
contractility (80,81). Similarly, calcium supplemen- ical features of preeclampsia, particularly super-
tation may reduce uterine smooth muscle contrac- imposed eclampsia, hepatic dysfunction, hematologic
tility and potentially improve uteroplacental blood disturbances, and cardiac remodeling and dysfunc-
flow, preventing preterm labor and delivery (80,81). tion. Nonetheless, the clinical relevance of this dis-
Pravastatin has been proposed as adjunctive ther- ease process is well-documented, given the
apy to reduce the risk of preeclampsia. Data from a substantial effect on maternal and perinatal
mouse model of preeclampsia demonstrated that pra- morbidity and mortality, in both the short and
vastatin prevented vascular dysfunction (82). Another long term. Many ongoing research activities are
study in a rat model of preeclampsia (83) showed that actively seeking to better understand this common
pravastatin reversed postpartum cardiac dysfunction, disorder that impacts 2% to 8% of pregnancies.
suggesting a potential role for secondary prevention Opportunities for future research include defining
of cardiovascular disease following preeclampsia in the roles of pre-existing diseases in the pathogenesis
humans. Preliminary human data did not identify of preeclampsia, elucidating immunologic predis-
safety concerns (84), and a clinical trial (85) is positions and genetic etiologies (such as LIN28B), and
ongoing to investigate the role of pravastatin in the further refining the link between preeclampsia
prevention of preeclampsia in high-risk pregnancy and short- and long-term cardiovascular diseases.
women (Pravastatin for Prevention of Preeclampsia; Meanwhile, prompt diagnosis, close observation, and
NCT01717586). Proposed mechanisms of pravastatin delivery when indicated are the mainstays of treat-
benefit include reversal of angiogenic imbalance, ment to reduce maternal and fetal morbidity and
improvement of endothelial function, and prevention mortality.
of injury from oxidation and inflammation (85).
Metformin has also been proposed to reduce the ACKNOWLEDGMENT The authors thank Mikako
risk of preeclampsia. Preliminary data from a ran- Kawai, who assisted with the Central Illustration and
domized controlled trial (86) of metformin for the Figure 1 design.
prevention of large-for-gestational age fetuses in
obese pregnant women without diabetes are prom- ADDRESS FOR CORRESPONDENCE: Dr. Christopher

ising. Although the primary outcome of reduction in W. Ives, Tinsley Harrison Internal Medicine Resi-
neonatal birthweight was negative, metformin was dency Program, Department of Medicine, University
associated with a 76% reduction in the incidence of of Alabama at Birmingham, 1720 2nd Avenue
preeclampsia (3.0% vs. 11.3%; odds ratio: 0.24,; 95% South, ZRB 1034, Birmingham, Alabama 35294-0007.
confidence interval: 0.10 to 0.61) (86). Further study E-mail: cives@uabmc.edu. Twitter: @UABCardiology,
is warranted. @UAB_CWRH, @uabmedicine.
JACC VOL. 76, NO. 14, 2020 Ives et al. 1701
OCTOBER 6, 2020:1690–702 Pathophysiology of Preeclampsia

REFERENCES

1. Goel A, Maski MR, Bajracharya S, et al. Epide- severe preeclampsia reveals a possible maternal vascular dysfunction. Hypertens Res 2017;40:
miology and mechanisms of de novo and persis- contribution to the etiology. Proc Natl Acad Sci U S 305–10.
tent hypertension in the postpartum period. A 2017;114:E8468–77.
33. Nilsson PM, Viigimaa M, Giwercman A,
Circulation 2015;132:1726–33.
18. Jebbink J, Wolters A, Fernando F, Afink G, van Cifkova R. Hypertension and reproduction. Curr
2. Rana S, Lemoine E, Granger J, Karumanchi SA. der Post J, Ris-Stalpers C. Molecular genetics of Hypertens Rep 2020;22:29.
Preeclampsia. Circ Res 2019;124:1094–112. preeclampsia and HELLP syndrome—a review.
34. Berks D, Steegers EA, Molas M, Visser W.
Biochim Biophys Acta 2012;1822:1960–9.
3. ACOG Practice Bulletin No. 202: Gestational Resolution of hypertension and proteinuria after
Hypertension and Preeclampsia. Obstet Gynecol 19. Gammill HS, Chettier R, Brewer A, et al. Car- preeclampsia. Obstet Gynecol 2009;114:1307–14.
2019;133:e1–25. diomyopathy and preeclampsia. Circulation 2018;
35. Adekomi AD, Moodley J, Naicker T. Neuro-
4. Petersen EE, Davis NL, Goodman D, et al. Vital 138:2359–66.
pathological complications associated with hy-
signs: pregnancy-related deaths, United States, 20. Laissue P, Vaiman D. Exploring the molecular pertensive disorders of pregnancy. Hypertens
2011-2015, and strategies for prevention, 13 aetiology of preeclampsia by massive parallel Pregnancy 2019;38:171–5.
states, 2013-2017. MMWR Morb Mortal Wkly Rep sequencing of DNA. Curr Hypertens Rep 2020;22:
36. Moghaddas Sani H, Zununi Vahed S,
2019;68:423–9. 31.
Ardalan M. Preeclampsia: a close look at renal
5. Jim B, Karumanchi SA. Preeclampsia: patho- 21. Sanapo L, Bublitz MH, Bourjeily G. Sleep dysfunction. Biomed Pharmacother 2019;109:
genesis, prevention, and long-term complications. disordered breathing, a novel, modifiable risk 408–16.
Semin Nephrol 2017;37:386–97. factor for hypertensive disorders of pregnancy.
37. Ding W, Qiu B, Cram DS, et al. Isobaric tag for
6. Sircar M, Thadhani R, Karumanchi SA. Patho- Curr Hypertens Rep 2020;22:28.
relative and absolute quantitation based quanti-
genesis of preeclampsia. Curr Opin Nephrol 22. Burwick RM, Pilliod RA, Dukhovny SE, tative proteomics reveals unique urinary protein
Hypertens 2015;24:131–8. Caughey AB. Fetal hydrops and the risk of severe profiles in patients with preeclampsia. J Cell Mol
7. Brown MA, Magee LA, Kenny LC, et al. The preeclampsia. J Matern Fetal Neonatal Med 2019; Med 2019;23:5822–6.
hypertensive disorders of pregnancy: ISSHP clas- 32:961–5.
38. Wilkerson RG, Ogunbodede AC. Hypertensive
sification, diagnosis & management recommen- 23. Soto-Wright V, Bernstein M, Goldstein DP, disorders of pregnancy. Emerg Med Clin North Am
dations for international practice. Pregnancy Berkowitz RS. The changing clinical presentation 2019;37:301–16.
Hypertens 2018;13:291–310. of complete molar pregnancy. Obstet Gynecol
39. Lockwood CJ, Krikun G, Caze R, Rahman M,
8. Pankiewicz K, Szczerba E, Maciejewski T, 1995;86:775–9.
Buchwalder LF, Schatz F. Decidual cell-expressed
Fijalkowska A. Non-obstetric complications in 24. Brouwers L, van der Meiden-van Roest AJ, tissue factor in human pregnancy and its involve-
preeclampsia. Prz Menopauzalny 2019;18:99–109. Savelkoul C, et al. Recurrence of pre-eclampsia ment in hemostasis and preeclampsia-related
9. Phipps E, Prasanna D, Brima W, Jim B. Pre- and the risk of future hypertension and cardio- angiogenesis. Ann N Y Acad Sci 2008;1127:67–72.
eclampsia: updates in pathogenesis, definitions, and vascular disease: a systematic review and meta-
analysis. BJOG 2018;125:1642–54. 40. Negro A, Delaruelle Z, Ivanova TA, et al.
guidelines. Clin J Am Soc Nephrol 2016;11:1102–13.
Headache and pregnancy: a systematic review.
10. El-Sayed AAF. Preeclampsia: a review of the 25. Ghossein-Doha C, van Neer J, Wissink B, et al. J Headache Pain 2017;18:106.
pathogenesis and possible management strategies Pre-eclampsia: an important risk factor for
asymptomatic heart failure. Ultrasound Obstet 41. Bushman ET, Varner MW, Digre KB. Headaches
based on its pathophysiological derangements.
Gynecol 2017;49:143–9. through a woman’s life. Obstet Gynecol Surv
Taiwan J Obstet Gynecol 2017;56:593–8.
2018;73:161–73.
11. Mustafa R, Ahmed S, Gupta A, Venuto RC. 26. Grand’Maison S, Pilote L, Schlosser K,
Stewart DJ, Okano M, Dayan N. Clinical features 42. Wagner SJ, Acquah LA, Lindell EP, et al. Posterior
A comprehensive review of hypertension in preg-
and outcomes of acute coronary syndrome in reversible encephalopathy syndrome and eclampsia:
nancy. J Pregnancy 2012;2012:105918.
women with previous pregnancy complications. pressing the case for more aggressive blood pressure
12. Spradley FT. Sympathetic nervous system control. Mayo Clin Proc 2011;86:851–6.
Can J Cardiol 2017;33:1683–92.
control of vascular function and blood pressure
during pregnancy and preeclampsia. J Hypertens 27. Theilen LH, Fraser A, Hollingshaus MS, et al. 43. Sutton ALM, Harper LM, Tita ATN. Hyperten-
2019;37:476–87. All-cause and cause-specific mortality after hy- sive disorders in pregnancy. Obstet Gynecol Clin
pertensive disease of pregnancy. Obstet Gynecol North Am 2018;45:333–47.
13. Malik A, Jee B, Gupta SK. Preeclampsia: disease
2016;128:238–44. 44. Schultz KL, Birnbaum AD, Goldstein DA.
biology and burden, its management strategies
with reference to India. Pregnancy Hypertens 28. Theilen LH, Meeks H, Fraser A, Esplin MS, Ocular disease in pregnancy. Curr Opin Oph-
2019;15:23–31. Smith KR, Varner MW. Long-term mortality risk thalmol 2005;16:308–14.
and life expectancy following recurrent hyper- 45. Park YJ, Park KH, Woo SJ. Clinical features of
14. Canfield J, Arlier S, Mong EF, et al. Decreased
tensive disease of pregnancy. Am J Obstet Gyne- pregnancy-associated retinal and choroidal dis-
LIN28B in preeclampsia impairs human tropho-
col 2018;219:107.e1–6. eases causing acute visual disturbance. Korean J
blast differentiation and migration. FASEB J 2019;
33:2759–69. 29. Irani RA, Xia Y. The functional role of the Ophthalmol 2017;31:320–7.
renin-angiotensin system in pregnancy and pre- 46. Ramirez-Montero C, Lima-Gomez V,
15. Cui Y, Wang W, Dong N, et al. Role of corin in
eclampsia. Placenta 2008;29:763–71. Anguiano-Robledo L, Hernandez-Campos ME,
trophoblast invasion and uterine spiral artery
remodelling in pregnancy. Nature 2012;484: 30. Wallukat G, Homuth V, Fischer T, et al. Pa- Lopez-Sanchez P. Preeclampsia as predisposing
246–50. tients with preeclampsia develop agonistic auto- factor for hypertensive retinopathy: participation
antibodies against the angiotensin AT1 receptor. by the RAAS and angiogenic factors. Exp Eye Res
16. Rabaglino MB, Post Uiterweer ED,
J Clin Invest 1999;103:945–52. 2020;193:107981.
Jeyabalan A, Hogge WA, Conrad KP. Bioinformat-
ics approach reveals evidence for impaired endo- 31. Armaly Z, Jadaon JE, Jabbour A, Abassi ZA. 47. Duley L, Gulmezoglu AM, Henderson-Smart DJ,
metrial maturation before and during early Preeclampsia: novel mechanisms and potential Chou D. Magnesium sulphate and other anticon-
pregnancy in women who developed preeclamp- therapeutic approaches. Front Physiol 2018;9:973. vulsants for women with pre-eclampsia. Cochrane
sia. Hypertension 2015;65:421–9. Database Syst Rev 2010;2010:CD000025.
32. Tomimatsu T, Mimura K, Endo M, Kumasawa K,
17. Garrido-Gomez T, Dominguez F, Quinonero A, Kimura T. Pathophysiology of preeclampsia: an 48. Lucas MJ, Leveno KJ, Cunningham FG.
et al. Defective decidualization during and after angiogenic imbalance and long-lasting systemic A comparison of magnesium sulfate with
1702 Ives et al. JACC VOL. 76, NO. 14, 2020

Pathophysiology of Preeclampsia OCTOBER 6, 2020:1690–702

phenytoin for the prevention of eclampsia. N Engl during pregnancy. Eur J Obstet Gynecol Reprod 78. Barakat R, Pelaez M, Cordero Y, et al. Exercise
J Med 1995;333:201–5. Biol 2015;191:141–2. during pregnancy protects against hypertension
and macrosomia: randomized clinical trial. Am J
49. Bergman L, Torres-Vergara P, Penny J, et al. 64. Cines DB, Levine LD. Thrombocytopenia in
Obstet Gynecol 2016;214:649.e1–8.
Investigating maternal brain alterations in pre- pregnancy. Blood 2017;130:2271–7.
eclampsia: the need for a multidisciplinary effort. 79. Syngelaki A, Sequeira Campos M, Roberge S,
65. Jodkowska A, Martynowicz H, Kaczmarek-
Curr Hypertens Rep 2019;21:72. Andrade W, Nicolaides KH. Diet and exercise for
Wdowiak B, Mazur G. Thrombocytopenia in preg-
preeclampsia prevention in overweight and obese
50. Vaught AJ, Kovell LC, Szymanski LM, et al. nancy: pathogenesis and diagnostic approach.
pregnant women: systematic review and meta-
Acute cardiac effects of severe pre-eclampsia. Postepy Hig Med Dosw (Online) 2015;69:1215–21.
analysis. J Matern Fetal Neonatal Med 2019;32:
J Am Coll Cardiol 2018;72:1–11. 66. ACOG Practice Bulletin No. 204: Fetal Growth 3495–501.
51. Birukov A, Wiesemann S, Golic M, et al. Restriction. Obstet Gynecol 2019;133:e97–109.
80. Hofmeyr GJ, Lawrie TA, Atallah AN, Duley L,
Myocardial evaluation of post-preeclamptic 67. Hauspurg A, Redman EK, Assibey-Mensah V, Torloni MR. Calcium supplementation during
women by CMR: is early risk stratification et al. Placental findings in non-hypertensive term pregnancy for preventing hypertensive disorders
possible? J Am Coll Cardiol Img 2020;13:1291–3. pregnancies and association with future adverse and related problems. Cochrane Database Syst
52. Behrens I, Basit S, Lykke JA, et al. Hyperten- pregnancy outcomes: a cohort study. Placenta Rev 2014;6:CD001059.
sive disorders of pregnancy and peripartum car- 2018;74:14–9.
81. Hofmeyr GJ, Lawrie TA, Atallah AN,
diomyopathy: a nationwide cohort study. PLoS 68. Bodnar LM, Catov JM, Simhan HN, Holick MF, Torloni MR. Calcium supplementation during
One 2019;14:e0211857. Powers RW, Roberts JM. Maternal vitamin D pregnancy for preventing hypertensive disorders
53. Bello N, Rendon ISH, Arany Z. The relationship deficiency increases the risk of preeclampsia. J Clin and related problems. Cochrane Database Syst
between pre-eclampsia and peripartum cardio- Endocrinol Metab 2007;92:3517–22. Rev 2018;10:CD001059.
myopathy: a systematic review and meta-analysis. 69. Duley L, Henderson-Smart D. Reduced salt 82. Fox KA, Longo M, Tamayo E, et al. Effects of
J Am Coll Cardiol 2013;62:1715–23. intake compared to normal dietary salt, or high pravastatin on mediators of vascular function in a
54. Pfeffer TJ, Hilfiker-Kleiner D. Pregnancy and intake, in pregnancy. Cochrane Database Syst Rev mouse model of soluble Fms-like tyrosine kinase-
heart disease: pregnancy-associated hypertension 2000;1999:CD001687. 1-induced preeclampsia. Am J Obstet Gynecol
and peripartum cardiomyopathy. Curr Probl Car- 70. Meher S, Abalos E, Carroli G. Bed rest with or 2011;205:366.e1–5.
diol 2018;43:364–88. without hospitalisation for hypertension during 83. Kraker K, O’Driscoll JM, Schutte T, et al. Sta-
55. Patten IS, Rana S, Shahul S, et al. Cardiac pregnancy. Cochrane Database Syst Rev 2005;4: tins reverse postpartum cardiovascular dysfunc-
angiogenic imbalance leads to peripartum cardio- CD003514. tion in a rat model of preeclampsia. Hypertension
myopathy. Nature 2012;485:333–8. 71. Meher S, Duley L. Garlic for preventing pre- 2020;75:202–10.
eclampsia and its complications. Cochrane Data- 84. Costantine MM, Cleary K, Hebert MF, et al.
56. Hilfiker-Kleiner D, Kaminski K, Podewski E,
base Syst Rev 2006;2006:CD006065. Safety and pharmacokinetics of pravastatin used
et al. A cathepsin D-cleaved 16 kDa form of pro-
lactin mediates postpartum cardiomyopathy. Cell 72. Rumbold A, Duley L, Crowther C, Haslam R. for the prevention of preeclampsia in high-risk
2007;128:589–600. Antioxidants for preventing pre-eclampsia. pregnant women: a pilot randomized controlled
Cochrane Database Syst Rev 2005;4:CD004227. trial. Am J Obstet Gynecol 2016;214:720.e1–17.
57. Zhang Z, Wang X, Wang J, Zhang L. The
decreased expression of Stat3 and p-Stat3 in 73. Wen SW, White RR, Rybak N, et al. Effect of 85. Costantine MM, Cleary K. Eunice Kennedy
preeclampsia-like rat placenta. J Mol Histol 2018; high dose folic acid supplementation in pregnancy Shriver National Institute of Child Health and Hu-
49:175–83. on pre-eclampsia (FACT): double blind, phase III, man Development Obstetric–Fetal Pharmacology
randomised controlled, international, multicentre Research Units Network. Pravastatin for the pre-
58. Zhang Z, Yang X, Zhang L, et al. Decreased
trial. BMJ 2018;362:k3478. vention of preeclampsia in high-risk pregnant
expression and activation of Stat3 in severe pre-
women. Obstet Gynecol 2013;121 Pt 1:349–53.
eclampsia. J Mol Histol 2015;46:205–19. 74. Zhou SJ, Yelland L, McPhee AJ, Quinlivan J,
Gibson RA, Makrides M. Fish-oil supplementation 86. Syngelaki A, Nicolaides KH, Balani J, et al.
59. Altman D, Carroli G, Duley L, et al. Do women
in pregnancy does not reduce the risk of gesta- Metformin versus placebo in obese pregnant
with pre-eclampsia, and their babies, benefit from
tional diabetes or preeclampsia. Am J Clin Nutr women without diabetes mellitus. N Engl J Med
magnesium sulphate? The Magpie Trial: a rando-
2012;95:1378–84. 2016;374:434–43.
mised placebo-controlled trial. Lancet 2002;359:
1877–90. 75. ACOG Committee Opinion No. 743: Low-Dose 87. Thilaganathan B, Kalafat E. Cardiovascular
Aspirin Use During Pregnancy. Obstet Gynecol system in preeclampsia and beyond. Hypertension
60. Sciscione AC, Ivester T, Largoza M, Manley J,
2018;132:e44–52. 2019;73:522–31.
Shlossman P, Colmorgen GH. Acute pulmonary
edema in pregnancy. Obstet Gynecol 2003;101: 76. LeFevre ML, U.S. Preventive Services Task 88. Mehta LS, Warnes CA, Bradley E, et al. Car-
511–5. Force. Low-dose aspirin use for the prevention diovascular considerations in caring for pregnant
of morbidity and mortality from preeclampsia: U. patients: a scientific statement from the American
61. Crowley PA. Antenatal corticosteroid therapy: Heart Association. Circulation 2020;141:
S. Preventive Services Task Force recommenda-
a meta-analysis of the randomized trials, 1972 to e884–903.
tion statement. Ann Intern Med 2014;161:
1994. Am J Obstet Gynecol 1995;173:322–35.
819–26.
62. Myles TD. Steroids: plenty of benefits, but not
77. Duley L, Meher S, Hunter KE, Seidler AL,
without risk. Obstet Gynecol 2011;117:429–30. KEY WORDS hypertension, hypertensive
Askie LM. Antiplatelet agents for preventing pre-
disorders of pregnancy, maternal morbidity,
63. Sinkey RG, Smith KL. The first known case of eclampsia and its complications. Cochrane Data-
pathogenesis, pregnancy
alanine aminotransferase deficiency diagnosed base Syst Rev 2019;2019:CD004659.

You might also like