HHS Public Access: The Etiology of Preeclampsia

Download as pdf or txt
Download as pdf or txt
You are on page 1of 44

HHS Public Access

Author manuscript
Am J Obstet Gynecol. Author manuscript; available in PMC 2023 February 01.
Author Manuscript

Published in final edited form as:


Am J Obstet Gynecol. 2022 February ; 226(2 Suppl): S844–S866. doi:10.1016/j.ajog.2021.11.1356.

The etiology of preeclampsia


Eunjung JUNG, MD1,2, Roberto ROMERO, MD, DMedSci1,3,4,5,6, Lami YEO, MD1,2, Nardhy
GOMEZ-LOPEZ, PhD1,2,7, Piya CHAEMSAITHONG, MD, PhD1,2,8, Adithep JAOVISIDHA,
MD8, Offer EREZ, MD1,2,9
1Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division
of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human
Development, National Institutes of Health, US Department of Health and Human Services
Author Manuscript

(NICHD/NIH/DHHS), Bethesda, MD, and Detroit, MI.


2Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit,
MI.
3Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI.
4Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI.
5Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI.
6Detroit Medical Center, Detroit, MI.
7Department of Biochemistry, Microbiology and Immunology, Wayne State University School of
Medicine, Detroit, MI.
Author Manuscript

8Department of Obstetrics and Gynecology, Faculty of Medicine, Ramathibodi Hospital, Mahidol


University, Bangkok, Thailand.
9Department of Obstetrics and Gynecology, HEmek Medical Center, Afula, Israel.

Abstract
A fundamental task in medicine is the understanding of the causes of diseases. Preeclampsia
and eclampsia, an enigmatic and elusive disorder, have been labeled the “disease of theories.”
Preeclampsia is one of the “great obstetrical syndromes” in which multiple and sometimes
overlapping pathologic processes activate a common pathway composed of endothelial cell
activation, intravascular inflammation, and syncytiotrophoblast stress. This article addresses the
potential etiologies, or causal explanations, for preeclampsia. The role of uteroplacental ischemia
Author Manuscript

is well established, based upon a solid body of clinical and experimental evidence. A causal role
for microorganisms has gained recognition through the realization that periodontal disease and

Correspondence: Roberto Romero, MD, D.Med.Sci., Perinatology Research Branch, NICHD/NIH/DHHS, Hutzel Women’s Hospital,
3990 John R, 4 Brush, Detroit, Michigan 48201, USA, Telephone: (313) 993-2700, [email protected].
Dr. Romero has contributed to this work as part of his official duties as an employee of the United States Federal Government.
Disclosure: The authors declare no conflicts of interest.
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our
customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review
of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered
which could affect the content, and all legal disclaimers that apply to the journal pertain.
JUNG et al. Page 2

maternal gut dysbiosis are linked to atherosclerosis, thus possibly to a subset of patients with
Author Manuscript

preeclampsia. The recent reports indicating that SARS-CoV-2 infection appears to be causally
linked to preeclampsia are reviewed as well as the mechanisms whereby this viral infection
can cause this syndrome. Particular etiological factors, such as the breakdown of maternal-fetal
immune tolerance, thought to account for the excess of preeclampsia in primipaternity and
egg donation—may operate, in part, through uteroplacental ischemia, while another, such as
placental aging, may operate largely through syncytiotrophoblast stress. This article also examines
the nature of the association between gestational diabetes mellitus and maternal obesity in
preeclampsia. The various roles of autoimmunity, fetal diseases, and endocrine disorders are also
discussed. A greater understanding of the etiologic factors of preeclampsia is essential to improve
treatment and prevention.

Condensation:
Author Manuscript

This article describes the causal risk factors, or etiology, of preeclampsia.

Keywords
Angiotensin receptor II; atherosclerosis; autoantibodies; Ballantyne syndrome; body mass index;
COVID-19; Cushing’s syndrome; endothelial cell dysfunction; genetic incompatibility; gestational
diabetes mellitus; hydatidiform mole; hydrops fetalis; hyperaldosteronism; hyperparathyroidism;
hypertension; infection; inflammation; insulin resistance; intestinal dysbiosis; maternal anti-
fetal rejection; metabolic syndrome; mirror syndrome; molar pregnancy; obesity; placental
aging; placental ischemia; placental lesions of maternal vascular malperfusion; primipaternity;
proteinuria; SARS-CoV-2; sleep-disordered breathing; sleep disorders; snoring; tolerance

Introduction
Author Manuscript

A fundamental task in medicine is the understanding of the causes of diseases.


Preeclampsia and eclampsia, the enigmatic, elusive disorders of pregnancy, have been
labeled the “diseases of theories”. Preeclampsia and eclampsia are among the “great
obstetrical syndromes” in which multiple and sometimes overlapping pathologic processes
activate a common pathway that leads to the clinical recognition of these disorders.
Just as the syndrome of preterm labor is recognized by the clinical manifestations
of activation of the common pathway of parturition (ie increased uterine contractility,
cervical remodeling, and membrane and decidual activation), so are preeclampsia and
eclampsia. Their common pathway consists of endothelial cell activation, intravascular
inflammation, and syncytiotrophoblast stress, and the diagnosis has traditionally rested
in the detection of hypertension and proteinuria, although professional organizations had
Author Manuscript

recently recommended that the diagnosis can be made in the absence of proteinuria when
patients have evidence of multi-systemic involvement.

A rich body of literature describes the risk factors for preeclampsia and eclampsia (ie the
conditions that increase the likelihood of these syndromes but are not necessarily causal).
However, the elucidation of etiologic, or causal, factors is necessary to successfully treat
and prevent disease. This article reviews the evidence linking these etiologic factors with
preeclampsia and eclampsia, which is summarized in Figure 1.

Am J Obstet Gynecol. Author manuscript; available in PMC 2023 February 01.


JUNG et al. Page 3

Uteroplacental ischemia
Author Manuscript

The principal mechanism of disease implicated in the etiology of preeclampsia and


eclampsia is uteroplacental ischemia. In 1914, James Young proposed that interference of
the uterine blood supply to the placenta would lead to placental infarctions that, in turn,
would release toxins into the maternal circulation, thus causing eclampsia.1 This theory
was based on the demonstration that placental infarctions were observed in patients with
eclampsia and on animal studies that showed subcutaneous injections of extracts of the
autolyzed human placenta into guinea pigs elicited convulsions, hepatic focal necrosis, and
renal lesions, similar to those observed in women who died of eclampsia.1 Dixon and
Taylor reported that intravenous injections of extracts of fresh human placenta induced an
increase in blood pressure in cats, rabbits, and dogs, resembling the effects of adrenaline.2
Therefore, the search for the causes of preeclampsia and eclampsia and for the identity of
the circulating “toxins” began more than 100 years ago.
Author Manuscript

Additional evidence supporting a role for uteroplacental ischemia came from studies by
Ogden, Hildebrand, and Page who reported that clamping of the abdominal aorta below the
renal arteries (to avoid renovascular hypertension) in dogs reduced uteroplacental perfusion;
furthermore, this response was followed by maternal hypertension that resolved after the
clamp was released.3 Because this hypertensive response was not observed in non-pregnant
animals, the investigators concluded that the signals responsible for hypertension must have
originated within the gravid uterus.3 Another observation buttressed this interpretation: after
removal of the pregnant uterus, the clamping of the aorta did not elicit hypertension (Figure
2).3, 4

Subsequent studies strengthened this concept, demonstrating that placental ischemia,


generated by placing silver clips in the uterine arteries of pregnant dogs, led to hypertension
Author Manuscript

and proteinuria.5 Two lines of evidence supported the view that placental ischemia rather
than uterine ischemia was key. First, patients with an abdominal pregnancy were known to
develop preeclampsia given that the implantation site was outside of the uterus.6 Second,
the placement of a Z suture through the placenta to generate ischemia resulted in the
development of hypertension and proteinuria (Figure 3A-3C).7 Moreover, the existence of
a circulating “toxin,” supported by the observation that the administration of blood from a
rabbit with experimental placental ischemia caused by the Z suture, resulted in hypertension
in non-pregnant animals (Figure 3D).7

The first in vivo evidence indicating that women with preeclampsia had decreased maternal-
placental blood flow was reported by McClure Browne and Veall,8 who described the
injection of radioactive sodium into the choriodecidual space of women with a normal
Author Manuscript

pregnancy and in those affected by preeclampsia. The investigators noted that the blood flow
at term was 600 mL/minute, but it was substantially lower in patients with preeclampsia.
These observations have been confirmed with different radioactive tracers in subsequent
studies.9-13 For example, Lunell et al 13 reported that uteroplacental blood flow was reduced
by 50% in patients with preeclampsia and that the reduction was greater in those with severe
preeclampsia than in those with mild disease.

Am J Obstet Gynecol. Author manuscript; available in PMC 2023 February 01.


JUNG et al. Page 4

The role of placental ischemia in the pathogenesis of preeclampsia is now well established.
Author Manuscript

Indeed, the most frequently implemented animal model of the syndrome is chronic reduction
of uteroplacental perfusion in pregnant rats, generated by the placement of a constriction
clip around the aorta below the renal arteries and before the origin of the uterine arteries at
14 days of gestation.14 In addition, this effect led to a reduction in placental blood flow by
approximately 40%, an increase in arterial blood pressure by 20-25 mm Hg by day 19 of
pregnancy, increased vascular resistance, decreased cardiac output and glomerular filtration
rate, and the frequent appearance of proteinuria.14 This model recapitulated two of the many
findings of preeclampsia: (1) an increase in circulating concentrations of soluble vascular
endothelial growth factor receptor-1 (sVEGFR-1; also known as soluble fms-like tyrosine
kinase 1 [sFlt-1]) and endoglin, and (2) a rise in the concentrations of pro-inflammatory
cytokines, such as tumor necrosis factor alpha (TNF-α) and interleukin-6 (IL-6). A similar
non-human primate model of preeclampsia, developed in pregnant baboons by selective
Author Manuscript

ligation of one uterine artery, led to the development of hypertension, proteinuria, and
increased production of sFlt-1.15 Importantly, the administration of short interfering RNAs,
which silence three of the sFlt-1 mRNA isoforms, suppressed sFlt-1 overexpression and
reduced hypertension and proteinuria.16 These studies suggest that the soluble factor, or
“toxin,” responsible for hypertension is, at least in part, sFlt-1. The reader is referred to the
review by Bakrania, George, and Granger17 in this Supplement for more details about the
model and the pathophysiologic events caused by uteroplacental ischemia.

What is the cause of placental ischemia in women with preeclampsia? The traditional
explanation has been that a defect in placentation leads to ischemia18, 19 but more recently,
a dysfunctional maternal cardiovascular system has been implicated.20 The developmental
abnormalities include failure of physiologic transformation of the spiral arteries which are
characterized by a narrow diameter and retention of the muscle in the media of the vessel
Author Manuscript

wall.18, 19 The persistence of the muscular coat is thought to make the vessels susceptible to
the effect of vasoconstrictive agents. In addition, arteries affected by failure of physiologic
transformation are more likely to develop atherosis (Figure 4), which also narrows the vessel
lumen and further compromises placental perfusion.21-26 Atherosis is a lesion specific to the
spiral arteries, which is equivalent to the atherosclerotic lesions observed in the coronary
arteries. Figure 5 illustrates the typical lesions of atherosis in the spiral arteries and shows
lipid-laden macrophages with the deposition of fat droplets detected with oil red-O stain. A
systematic review and meta-analysis showed that placental lesions consistent with maternal
vascular malperfusion (eg placental infarction, failure of physiologic transformation of the
spiral arteries, acute atherosis) is four-fold to seven-fold higher in patients with preeclampsia
than in those with a normal pregnancy.27 A comprehensive review of the failure of
physiological transformation and spiral artery atherosis by Staff et al 28 is available as
Author Manuscript

part of this Supplement. Atherosis is not specific to preeclampsia, and this type of lesion
has been reported in other pregnancy-related conditions, such as spontaneous abortion,29
preterm labor,30 preterm prelabor rupture of the membranes,31 fetal growth restriction,32, 33
and fetal death.25, 29 We have proposed that placental ischemia is a major mechanism of
disease in various obstetrical syndromes and that the timing, severity, and duration of the
insult may explain the clinical occurrence of different obstetrical syndromes.34

Am J Obstet Gynecol. Author manuscript; available in PMC 2023 February 01.


JUNG et al. Page 5

The following evidence supports a causal link between placental ischemia and preeclampsia:
Author Manuscript

(1) experimentally induced ischemia in several animal models leads to hypertension and
proteinuria;3, 5, 7 (2) uterine blood flow is lower in patients with preeclampsia than in
normal pregnant women;8, 11, 13 (3) placental histopathologic lesions indicative of ischemia
(often referred to as maternal vascular malperfusion) are frequent, consistent findings
in preeclampsia and eclampsia; 27 (4) the failure of physiologic transformation of the
spiral arteries and atherosis are typical features of preeclampsia;28, 35 (5) the pulsatility
index of the uterine artery (a parameter to assess resistance to flow) is higher in patients
with preeclampsia than in normal pregnant women; this finding can be observed during
the midtrimester of pregnancy, weeks or even months before the development of the
disease;36, 37 (6) the maternal plasma placental growth factor (PlGF)/sFlt-1 ratio, a non-
invasive marker of lesions of maternal vascular malperfusion, is elevated at the time of the
onset of disease and prior to the development of preeclampsia;38-44 and (7) a blockade of
sFlt-1 mRNA reduces hypertension and proteinuria.16 The frequency of placental lesions
Author Manuscript

of maternal vascular malperfusion, of abnormalities in the uterine artery Doppler, and of


alterations of biomarkers (eg PlGF/sFlt-1) is higher in preterm preeclampsia than in term
preeclampsia, suggesting that ischemia plays a different role in early-onset versus late-onset
preeclampsia.45-48

The case for ischemia as an etiologic factor could be even more persuasive if the treatment
of ischemia could prevent the occurrence of preeclampsia. This evidence is difficult to
generate in pregnant humans. Nonetheless, it can be argued that the efficacy of aspirin in
reducing the rate of preterm preeclampsia49 is achieved through the prevention of arterial
thrombosis in the spiral arteries and intervillous space, as this is the proposed mechanism
for aspirin in the prevention of myocardial infarction in atherosclerosis.50 This interpretation
would also explain the lack of efficacy of aspirin in preventing preeclampsia at term, given
Author Manuscript

that ischemia seems to play a lesser role.

Maternal infection
Maternal infection has been implicated in the etiology for preeclampsia and eclampsia
since the beginning of the 20th century. Albert51 proposed that the “toxins” responsible
for eclampsia were the product of putrefactive changes in the uterine cavity caused by the
action of bacteria (“a latent microbic endometritis”). Indeed, a microorganism ‘Bacillus
eclampsiae’ was proposed to be the cause.52, 53 This view progressively fell out of favor
because preeclampsia and eclampsia do not present the typical features, eg a fever, of
an infectious disease. Nonetheless, the idea that microorganisms may be involved in the
genesis of preeclampsia and eclampsia recurs in the literature every few years, and it has
reemerged recently based on the relationship between periodontal disease, urinary tract
Author Manuscript

infection, SARS-CoV-2 infection, and maternal gut dysbiosis.

Periodontal disease—The best evidence to support a relationship between


microorganisms and preeclampsia derives from studies of periodontal disease, which
increases the risk of developing this pregnancy-related syndrome (OR 1.76; 95% confidence
interval (CI) 1.43-2.18).54 The term periodontal disease refers to an inflammatory condition
caused by immune dysfunction initiated by bacteria within the oral cavity.55 The spectrum

Am J Obstet Gynecol. Author manuscript; available in PMC 2023 February 01.


JUNG et al. Page 6

of disease ranges from gingivitis (inflammation of the soft tissues only) to the destruction
Author Manuscript

of the connective tissue attachment and alveolar bone, which can eventually lead to
tooth loss.55 Bacteria in the periodontal space can be released during dental procedures
or in the course of severe disease, leading to a systemic inflammatory response that
can cause damage and seed sites in the cardiovascular system.56 Indeed, strong evidence
indicates that periodontal disease is a risk factor for atherosclerotic cardiovascular diseases,
including atherosclerosis, coronary artery disease, stroke, and atrial fibrillation.57, 58 In brief,
such evidence denotes that 1) microorganisms found in the periodontal space can cause
bacteremia;59 2) bacteria from the oral cavity have been found in atheromatous plaques;60
and 3) periodontal infections can induce vascular lesions in the aorta and coronary
arteries.57, 61 In an animal model of hyperlipidemia, ie apolipoprotein E null mice, an oral
infection with Porphyromonas gingivalis led to plaque in the aorta (Figure 6).61 Similar
findings have been reported in an integrin β6 null mice model with polymicrobial infection
and periodontal pathogens.62 The etiologic role of periodontal disease in preeclampsia is
Author Manuscript

predicated on the same mechanisms linking periodontal disease and atherosclerosis.

Offenbacher, Beck, Boggess, and Murtha63 have provided clinical, epidemiologic, and
experimental evidence that periodontal disease is causally linked to preeclampsia. For
example, women with periodontal disease who have an elevated C-reactive protein (CRP)
concentration are at increased risk for the development of preeclampsia compared to those
without periodontal disease (adjusted relative risk (aRR) 5.8; 95% CI 1.2–26.9).63 An
elevated CRP would indicate that periodontal disease has led to a systemic inflammatory
process, thus it provides a link between periodontal disease and preeclampsia.

Urinary tract infection—The relationship between microbial colonization of the maternal


urinary tract and preeclampsia has also been reported. A systematic review noted that
Author Manuscript

urinary tract infections are associated with preeclampsia (OR 1.57; 95% CI 1.45-1.70).54
However, the case definitions have been broad and have included pyelonephritis,
lower urinary tract infections, and asymptomatic bacteriuria as a group.64, 65 When
subgroup analysis is performed, evidence for the association with preeclampsia either
weakens or disappears. We have doubts that asymptomatic bacteriuria, which is not
associated with a systemic inflammatory response,66 could cause preeclampsia. Isolated
cases have documented instances in which preeclampsia is associated with malaria,67-69
cytomegalovirus (CMV),70, 71 human immunodeficiency virus,72, 73 but the evidence is
insufficient to support causality.

Experimental observations supporting a causal relationship between infection and systemic


inflammation and preeclampsia indicate that intravenous low-dose endotoxin administration
Author Manuscript

in pregnant rats on day 14 of gestation results in the development of high blood pressure,
proteinuria, low platelet count, and glomerular fibrinogen deposits.74 Bacterial endotoxin is
a method utilized to induce a systemic inflammatory response and the activation of thrombin
through the release of tissue factor. These mechanisms are implicated in the pathogenesis of
preeclampsia.

SARS-CoV-2 Infection—Early during the COVID-19 pandemic, it was recognized


that a subset of non-pregnant patients developed hypertension,75-77 proteinuria,78-81

Am J Obstet Gynecol. Author manuscript; available in PMC 2023 February 01.


JUNG et al. Page 7

thrombocytopenia,82, 83 and elevated liver enzymes,84, 85 which resemble preeclampsia and


Author Manuscript

hemolysis, elevated liver enzymes, and low platelet count (HELLP) syndrome. A recent
meta-analysis demonstrated that SARS-CoV-2 infection during pregnancy is associated with
a significant increase in the odds of developing preeclampsia (OR 1.58; 95% CI 1.39-1.8),
preeclampsia with severe features (OR 1.76; 95% CI 1.18-2.63), eclampsia (OR 1.97; 95%
CI 1.01-3.84), and HELLP syndrome (OR 2.01; 95% CI 1.48-2.97).86 In addition, there is a
dose-response relationship between SARS-CoV-2 infection and the subsequent development
of preeclampsia (Figure 7).87 Patients with severe COVID-19 had a 5-fold greater risk of
preeclampsia than those with asymptomatic COVID-19.87 The median interval between
maternal SARS-CoV-2 infection and the subsequent development of preeclampsia is 3.8
weeks (interquartile range, 0.29-11.5).88

One mechanism whereby SARS-CoV-2 infection can be causally linked to preeclampsia


has been proposed to involve endothelial dysfunction. Indeed, SARS-CoV-2 can infect
Author Manuscript

endothelial cells that normally express angiotensin-converting enzyme 2 (ACE2), one of


the cell entry receptors for the virus, leading to endothelitis.89, 90 Endothelial infection
can induce the activation of thrombin, intravascular inflammation (ie a cytokine storm),
and damage of the microvasculature in target organs; this susceptibility ultimately leads
to the multi-systemic nature of the syndrome, which includes not only renal involvement
but also central nervous system dysfunction and seizures.91-93 Therefore, an infectious
process, which targets the endothelium, could lead to a syndrome similar to preeclampsia
and eclampsia (Figure 8).

Of interest, pregnant women with COVID-19 infection who developed preeclampsia-like


symptoms: their recovery from SARS-CoV-2 infection, followed by the resolution of
hypertension, occurred without the delivery of the fetus and placenta.94 It remains an
Author Manuscript

open question whether preeclampsia, after SARS-CoV-2 infection, may or may not require
placental involvement. Serum and plasma concentrations of sFlt-1, a marker of endothelial
dysfunction, were elevated in non-pregnant subjects with COVID-19.95 This finding is
consistent with another study in pregnant women with severe COVID-19 who presented
an elevated maternal plasma concentration of sFlt-1 and a high sFlt-1/PlGF ratio.96
Genetic susceptibility may explain why some women with COVID-19 infection develop
preeclampsia but others do not.97, 98

Maternal intestinal dysbiosis—The human gut microbiota plays an important role in


host nutrition, harvesting of energy, and immune responses to potential pathogens.99-103
Normal pregnancy represents a state in which a major reorganization of energy distribution
(harvesting, storage, and expenditure) is related to the need to support the growth and
Author Manuscript

development of the fetus and placenta. Remodeling of the gut microbiota during normal
human pregnancy, reported for the third trimester of pregnancy, indicated an overall increase
in Proteobacteria and Actinobacteria and a reduction in microbial richness.104 When the
intestinal content from normal pregnant women in the third trimester was administered to
germ-free mice, it increased adiposity and insulin resistance,105 which have been attributed
to the proinflammatory effects of the gut microbiota.104

Am J Obstet Gynecol. Author manuscript; available in PMC 2023 February 01.


JUNG et al. Page 8

Gut dysbiosis, or an imbalance among the human gut’s microbial communities, is now
implicated in the development of atherosclerosis,106, 107 hypertension,108, 109 proteinuria,110
Author Manuscript

cardiometabolic syndrome,111, 112 and recently, preeclampsia.113-117 A causal link between


gut dysbiosis and cardiovascular disease has been derived from an observation that the
transplant of fecal material from hypertensive, non-pregnant, human subjects to germ-free
mice led to hypertension.108 Similarly, fecal transplants from mice prone to atherosclerosis
can transmit the condition to susceptible mice (eg apolipoprotein E null mice).118 This effect
has been attributed, at least in part, to trimethylamine-N-oxide (TMAO), a bacteria-derived
metabolite from choline and carnitine that is present in gut dysbiosis and has been shown to
accelerate the development of atherosclerosis.107, 118

Changes in the human gut microbiota have been reported in preeclampsia,114, 115, 117, 119
which persist 6 weeks postpartum.120 The changes include a reduction in microbial
burden with Firmicutes, Clostridia, Clostridiales, and Ruminococcus and an increase
Author Manuscript

in Bacteroidetes, Proteobacteria, Actinobacteria, Bacteroidia, Gammaproteobacteria, and


Enterobacteriaceae (Figure 9A).119 Moreover, the study provides evidence to support
causality: fecal transplants from pregnant women with preeclampsia into mice led to the
development of the syndrome.115 The experimental paradigm required the administration of
fecal material from pregnant women with and without preeclampsia to non-pregnant mice
that had received antibiotics for 5 days (to change gut flora); the mice were housed in
pathogen-free facilities.115 Six weeks after the first fecal inoculation, mice were mated.115
Pregnant mice that had received the fecal microbiota transplantation from mothers with
preeclampsia developed high systolic blood pressure and proteinuria, and they delivered
less live pups than mice that receive fecal microbiota transplantation from women with
normotensive pregnancy (Figure 9B).115 Collectively, this evidence suggests a role for
gut dysbiosis in preeclampsia. However, further investigation into the precise mechanisms
Author Manuscript

that may explain this phenomenon as well as the susceptibility of some pregnant women
compared to others is necessary.

Gestational diabetes mellitus, maternal obesity, and the metabolic syndrome


Gestational diabetes mellitus—Gestational diabetes mellitus is an independent risk
factor for preeclampsia, after adjusting for confounders.121, 122 In a retrospective study of
647,392 pregnancies, women with gestational diabetes had an increased risk of preeclampsia
(adjusted odds ratio (aOR) 1.29; 95% CI 1.19-1.41).121 Preexisting diabetes mellitus had
also been linked to the development of preeclampsia: a systematic review reported that
diabetes mellitus prior to pregnancy was associated with an increased risk of preeclampsia
(aRR 3.56; 95% CI 2.54-4.99).123 Diabetes mellitus is considered to be strongly associated
with late-onset rather than early-onset preeclampsia (early onset: aOR 1.87, 95% CI
Author Manuscript

1.6-2.18; late onset: aOR 2.46, 95% CI 2.32-2.61).124 The Hyperglycemia and Adverse
Pregnancy Outcome (HAPO) Study Cooperative Research Group reported a significant
positive association between the degree of maternal hyperglycemia and preeclampsia: the
odds ratio for each 1-SD increase in glucose concentrations (fasting, one hour, and two
hours after a 75mg glucose tolerance test) ranged from 1.21 to 1.28.125

Am J Obstet Gynecol. Author manuscript; available in PMC 2023 February 01.


JUNG et al. Page 9

A causal role was strengthened by the observation that the treatment of gestational diabetes
mellitus with diet,126 insulin,127, 128 and metformin129-132 reduces the risk of preeclampsia.
Author Manuscript

Two randomized clinical trials have shown that the treatment of gestational diabetes mellitus
with insulin reduces the risk of preeclampsia (adjusted treatment effect: 0.70; 95% CI
0.51-0.95).127, 128 Metformin is associated with a reduced risk of preeclampsia (RR 0.68;
95% CI 0.48–0.95)131 and prolongation of gestation in women with preterm preeclampsia
(median, 18 days).133 Prenatal exercise has also been reported to decrease the rate of
gestational diabetes mellitus by 38% and preeclampsia by 41% based on the results of a
systematic review and meta-analysis.134

Collectively, the evidence suggests a causal relationship between gestational diabetes


mellitus and preeclampsia given the consistency of association, its magnitude, the temporal
relationship, and the efficacy of interventions, such as insulin and metformin, in reducing the
rate of preeclampsia.
Author Manuscript

Maternal obesity—Obesity, defined by a body mass index (BMI) of 30.0 kg/m2 or more,
is strongly associated with preeclampsia.135-137 A meta-analysis of 29 prospective cohort
studies (1,980,761 participants and 67,075 cases of preeclampsia) showed that maternal
obesity was significantly associated with the development of preeclampsia (aOR 2.93; 95%
CI 2.58-3.33), and the risk was even higher in severe obesity (BMI ≥ 35 kg/m2; aOR 4.14;
95% CI 3.61-4.75).138 A similar finding was reported overweight women (BMI: 26.1-29.0
kg/m2) who had an increased risk of preeclampsia in comparison to women with a normal
BMI (RR 1.57; 95% CI 1.49-1.64).139

Moreover, a dose-dependent relationship was found between pre-pregnancy BMI and the
risk of preeclampsia in both nulliparous and parous women in a large epidemiological
Author Manuscript

study (41,000 nulliparous and 29,000 multiparous) (Figures 10).140 Data from the USA
Collaborative Perinatal Project indicated similar results for Caucasian and for African-
American women (severe preeclampsia: African American, OR 3.2, 95% CI 2.5-5.0;
Caucasian, OR 3.4, 95% CI 2.1-5.6).141 Most studies considered that obesity predisposes
mainly to late-onset preeclampsia.123, 141 However, a recent population-based study, which
used US vital statistics data and included 15.8 million women (48,007 cases of early-onset
disease and 777,715 cases of late-onset disease), demonstrated that maternal obesity is
associated with an increased risk of both early-onset and late-onset disease (eg BMI 40
kg/m2 or greater: early-onset disease, aRR 2.18, 95% CI 2.12–2.24; late-onset disease: aRR
3.93, 95% CI 3.91–3.96).142 In addition, preconceptional maternal weight loss, either with
lifestyle modification or by bariatric surgery, was shown to be effective in reducing the risk
of preeclampsia (OR 0.67; 95% CI 0.51-0.88).143
Author Manuscript

Metabolic syndrome—The term “metabolic syndrome” refers to a cluster of


metabolic abnormalities that includes central obesity, insulin resistance, atherogenic
dyslipidemia, and hypertension.144, 145 This syndrome is strongly associated with systemic
inflammation, oxidative stress, and endothelial dysfunction, all of which are features of
preeclampsia.146-149 A retrospective cohort study of 212,463 women showed that the
presence of pre-pregnancy metabolic syndrome was associated with an increased risk of
preeclampsia (aOR 1.48; 95% CI 1.26-1.74).150 Preeclampsia is also a risk factor for the

Am J Obstet Gynecol. Author manuscript; available in PMC 2023 February 01.


JUNG et al. Page 10

subsequent development of metabolic syndrome after delivery.151-154 Moreover, bariatric


Author Manuscript

surgery performed before pregnancy as a treatment for metabolic syndrome was associated
with a lower rate of preeclampsia or eclampsia (aOR 0.2; 95% CI 0.09-0.44).155

The mechanisms whereby insulin resistance predisposes to the development of preeclampsia


are related to intravascular inflammation and endothelial cell dysfunction,156, 157 which is
the common pathway of the syndrome. Nonetheless, insulin resistance is neither required
nor sufficient for the development of preeclampsia. The reason why some, but not other,
patients with insulin resistance develop preeclampsia is unknown.

Sleep disorders
Sleep-disordered breathing, a term encompassing obstructive sleep apnea, snoring, periodic
episodes of hypoxia, central apnea, and sleep hypopnea, during pregnancy is a risk factor
of preeclampsia. A systematic review and meta-analysis of 120 studies, which included
Author Manuscript

a total of 58,123,250 pregnant women, indicated that maternal sleep disturbances during
pregnancy were associated with an increased risk of preeclampsia (OR 2.80; 95% CI
2.38-3.30);158 these disturbances included subjective sleep-disordered breathing (OR 3.52;
95% CI 2.58-4.79), obstructive sleep apnea (OR 2.36; 95% CI 2.00-2.79), and the restless
legs syndrome (OR 1.83; 95% CI 1.04-3.21).158

Snoring is defined as the vibration of respiratory structures resulting from turbulent


flow when the upper airway narrows during sleep.159 It is more common in pregnant
women than in non-pregnant women (14%-23% vs 4%)160, 161 and increases the risk of
hypertension,162 and new-onset snoring during pregnancy is associated with preeclampsia
(OR 1.59; 95% CI 1.06-2.37).163 Evidence supporting a causal relationship between snoring
and preeclampsia indicated that treatment with nasal continuous positive airway pressure
Author Manuscript

(CPAP) improved blood pressure in women with preeclampsia.164, 165 Edwards et al164
reported that autosetting nasal CPAP administered through sleep resulted in a marked
reduction in blood pressure (before treatment: mean systolic 146 mm Hg and mean diastolic
92 mm Hg; after treatment: mean systolic 128 mm Hg and mean diastolic 73 mm Hg).
Recently, Bourjeily et al166 reported that pregnant women with obstructive sleep apnea
presented a higher maternal plasma sFlt-1/PlGF concentration ratio than those in a control
group and that maternal sFlt-1 concentrations were decreased after CPAP treatment.167

In normal pregnancy, blood pressure has a circadian rhythm and is at its highest during
the daytime. A reversed diurnal blood-pressure rhythm (ie the nocturnal blood pressure
was higher than the diurnal blood pressure) has been reported in preeclampsia.168, 169
Sleep architecture, using polysomnography, has shown that patients with preeclampsia
Author Manuscript

had altered sleeping patterns,170 specifically spending more time in slow-wave sleep, in
comparison to those with a normal pregnancy (43 ± 3 vs 21 ± 2; p < 0.001).170 One possible
explanation for this finding is that cytokines, such as IL-1 and TNF-α, can increase the
amount of slow-wave sleep,170 as they are elevated in the maternal circulation of those
with preeclampsia.171-173 Collectively, the proposed mechanisms linking sleep disorders and
preeclampsia involve intravascular inflammation and endothelial cell dysfunction.

Am J Obstet Gynecol. Author manuscript; available in PMC 2023 February 01.


JUNG et al. Page 11

Molar pregnancy
Author Manuscript

A hydatidiform mole, a gestational trophoblastic disease characterized by abnormal


proliferation of trophoblast and hydropic changes of the chorionic villi, is associated with
preeclampsia174 and sometimes presents before 20 weeks of gestation.175-178 The frequency
of preeclampsia in patients with a hydatidiform mole ranges from 27% to 40% and is
probably higher in patients who are left untreated until the second trimester.175-178

The mechanism whereby a complete hydatidiform mole causes preeclampsia involves


the excess production of sFlt-1. Maternal serum sFlt-1 concentrations were 2-fold to
3-fold higher in patients with a hydatidiform mole than in those with a gestational age-
matched control;179 an increased expression of sFlt-1 within the molar tissue has also been
reported.180 Moreover, placentas with a hydatidiform mole had an increased expression of
LIGHT (TNFSF14, or TNF superfamily member 14), which was co-localized with sFlt-1
in the molar tissue.181 Chorionic villi in the molar tissue are edematous or hydropic, which
Author Manuscript

are often avascular or display markedly reduced vessel density.175 These villus capillary
changes may lead to excessive production of sFlt-1 that enters maternal circulation and,
consequently, to the development of the preeclampsia syndrome.

Fetal diseases
Specific fetal diseases associated with the development of preeclampsia include 1)
Ballantyne or mirror syndrome;225, 226 2) Trisomy 13 or triploidy;227, 228 and 3) a unique
complication of multiple gestations (eg twin-to-twin transfusion syndrome or selective
fetal growth restriction).229 Ballantyne or mirror syndrome reflects the simultaneously
edematous state of the mother, fetus, and placenta (also called triple edema).230, 231
This syndrome has been observed in fetal hydrops caused by rhesus isoimmunization,230
CMV,232 or parvovirus 19 infections,233 Ebstein’s anomaly,234 aneurysms of the vein
Author Manuscript

of Galen,235 fetal supraventricular tachycardia,236, 237 and placental chorioangioma.238


In mirror syndrome, the mother may develop proteinuria, hypertension, and even severe
preeclampsia.225, 226, 234, 239 The frequency of hypertension in patients with mirror
syndrome is about 60%.225, 226, 234, 239 The reversal of preeclampsia and Ballantyne
syndrome can occur after intrauterine transfusion in parvovirus-induced hydrops without
delivery.240 All patients with mirror syndrome presented an increased maternal plasma
concentration of sFlt-1 (above the 95th percentile for gestational age).241 In addition, it
has been shown that the maternal sFlt-1 concentration returns to normal after intrauterine
transfusion.242, 243

Another example of preeclampsia with fetal disease is Trisomy 13 or triploidy.227, 228, 244 A
case control study of 25 cases with Trisomy 13 showed that the incidence of preeclampsia
Author Manuscript

was significantly higher than detected in the normal karyotype control (44% vs 8%,
p=0.001).245 Women with a Trisomy 13 pregnancy have a higher serum concentration of
sFlt-1/PlGF ratio,246 and the placentas had an increased staining for sFlt-1 when compared
to euploid as well as Trisomy 21.247 Interestingly, sFlt-1 is located on chromosome 13,
suggesting the possibility that an extra copy of chromosome 13 may lead to increased
production of sFlt-1.244, 247

Am J Obstet Gynecol. Author manuscript; available in PMC 2023 February 01.


JUNG et al. Page 12

Multiple gestations with twin-to-twin transfusion syndrome or selective fetal growth


restriction can also predispose to the development of preeclampsia.229 Resolution
Author Manuscript

of hypertension and proteinuria generally occur after the selective termination of


pregnancy248-251 or after the fetal death of one twin.252 An improvement in angiogenic
and anti-antiangiogenic profiles has also been demonstrated after selective termination or
fetal demise of one twin.253, 254 Collectively, these observations indicate that preeclampsia
can resolve without delivery and suggest that fetal compromise may induce the syndrome in
some cases.

Autoimmune mechanisms: a role for antibodies against angiotensin II type I receptor


Preeclampsia is not traditionally considered an autoimmune disorder. However, a role
for autoimmune mechanisms of disease has been investigated for several decades, given
that patients with a systemic autoimmune disease, such as systemic lupus erythematosus
Author Manuscript

(SLE) or antiphospholipid syndrome (APS), were at an increased risk for preeclampsia


(SLE: RR 1.91, 95% CI 1.44-2.53; APS: RR 1.81, 95% CI 1.33-2.45).255-258 Similarly,
several studies have shown that some specific autoantibodies, such as anti-β2 glycoprotein-I
(ab2GPI), anti-cardiolipin antibodies (aCL), or lupus anti-coagulant (LA), are associated
with preeclampsia.256, 259 However, the most compelling case for a role of an autoimmune
mechanism is exemplified by antibodies against the angiotensin II receptor.

Nearly two decades ago, Wallukat et al260 reported that women with preeclampsia
had antibodies that bind to the angiotensin II type I receptor (AT1-AA). Subsequently,
substantial evidence has accumulated that supports a role for AT1-AA in the pathogenesis
of preeclampsia. Such evidence comprises the following findings: 1) 80% of women with
preeclampsia have increased concentrations of AT1-AA in maternal serum at the time of
diagnosis;261 2) the concentration of AT1-AA in maternal serum correlates with the severity
Author Manuscript

of hypertension and proteinuria;262 and 3) the administration of AT1-AA (either produced


endogenously in response to transgenic expression of human renin and angiotensinogen,
or administered by an injection of purified AT1-AA from women with preeclampsia) to
pregnant rats led to hypertension, proteinuria, glomerular capillary endotheliosis,263 and the
increased production of sFlt-1 as well as soluble endoglin,264 which can be attenuated by
the administration of an AT1-receptor blocker (Losartan). However, one longitudinal study,
conducted to determine whether the concentration of AT1-AA is increased before the onset
of preeclampsia, did not produce such a result.265

What causes the production of AT1-AA? Placental ischemia after a reduction in uterine
perfusion pressure has been shown to increase the concentration of serum AT1-AA.266 Also,
the inhibition of AT1-AA by the administration of an epitope-binding peptide (‘n7AAc’)
Author Manuscript

can reduce maternal blood pressure and plasma concentrations of endothelin-1, sFlt-1,
and isoprostanes, a marker of oxidative stress.267 Moreover, the administration of the
inflammatory mediators TNF-α,266, 268 IL-6,268, 269 and IL-17270 to pregnant rats can
induce preeclampsia and enhance AT1-AA activity, which is abrogated by an AT1-AA
blockade.267

Am J Obstet Gynecol. Author manuscript; available in PMC 2023 February 01.


JUNG et al. Page 13

Placental Aging
Author Manuscript

Cells and organisms have a finite lifespan. Human cells in culture can multiply (mitosis)
a limited number of times (Hayflick limit) before they stop dividing and, subsequently,
undergo programmed cell death, or apoptosis.271 The placenta is also thought to have a pre-
specified lifetime, and as age advances, the functional capacity of placental cells declines.
Placental aging has been described for several decades,272, 273 and premature aging has been
implicated as a mechanism of disease for obstetric outcomes, eg preeclampsia,274, 275 fetal
growth restriction,274, 276 fetal death,277 and preterm labor.278

Cytotrophoblasts are replicating cells, but the syncytiotrophoblast is generated by fusion


and, therefore, is in a state of senescence (ie defined as the inability to multiply). Evidence
supporting the concept that senescence is present in the normal syncytiotrophoblast
includes the following findings: 1) high lysosomal activity indicated by staining with
Author Manuscript

beta-galactosidase; 2) increased expression of cyclin-dependent kinase, p53, p21, p16; and


3) the presence of syncytial knots, proposed to be equivalent to senescence-associated
heterochromatic foci, which are clusters of chromatin in the nucleus of senescent
cells.279-281 Cindrova-Davies et al 280 demonstrated that markers of senescence increase
in normal placentas as a function of advancing gestational age.

Placentas of patients with preeclampsia exhibit greater expression of p53, p21, and p16,
shorter telomeres,274 and reduced telomerase activity.282, 283 Redman et al 279 proposed
that with advancing gestational age some patients develop a “twilight placenta,” a condition
in which the organ is affected by senescence. When placental growth reaches its limits
at term, terminal villi become overcrowded with diminished intervillous pore size, leading
to respiratory failure of the placenta (Figure 12) and contributing to intervillous hypoxia
and syncytiotrophoblast stress.284-286 A twilight placenta has been invoked as a potential
Author Manuscript

cause for late pregnancy problems such as late-onset preeclampsia,46, 287 unexplained term
stillbirth, or fetal death in prolonged pregnancy.277, 283, 288 At present, there is not an easy
or practical way to determine placental age; however, recent studies have identified an
epigenetic clock for placental dating,289 which may be used to examine the role of placental
aging in pregnancy complications in the future.

Breakdown of maternal-fetal immune tolerance


Viviparity involves the temporal coexistence of two individuals (mother and fetus) with
different genetic makeup. The placenta and fetus together are viewed as the most successful
semi-allograft (with maternal and paternal contributions), and the mechanisms responsible
for the immune tolerance have been the subject of investigation for decades.182-187
Two typical features of the adaptive immune system are memory and specificity.188-190
Author Manuscript

A role for the immune system has been proposed because preeclampsia is more
common in primigravidas than in parous women and in multigravidas with different
fathers (primipaternity) than in subsequent pregnancies with the same father.191, 192
The predilection for primigravidas has been attributed to a memory-like phenomenon
that protects mothers against paternal antigens in subsequent pregnancies.193 The higher
frequency of preeclampsia in multigravidas whose pregnancy is from a different father
supports the concept that the immune phenomenon is specific to paternally derived

Am J Obstet Gynecol. Author manuscript; available in PMC 2023 February 01.


JUNG et al. Page 14

antigens.194, 195 Moreover, this syndrome is more common in pregnancies that occur from
Author Manuscript

an egg donation in which the placenta and fetus represent a full allograft rather than a
semi-allograft.196

The placenta and fetus express both paternal and maternal antigens; they are a
semiallograft.197-199 The syncytiotrophoblast is in direct contact with maternal blood and
the decidua, thus the maternal immune system is exposed to paternal antigens expressed
by the fetus.182 Immune tolerance is a requirement for a successful pregnancy,185, 200, 201
and a breakdown of tolerance leads to maternal anti-fetal rejection, placental damage, and
pregnancy complications that may include preeclampsia. Placental lesions, which represent
manifestations of maternal anti-fetal rejection, are villitis of unknown etiology,202-205
massive perivillous fibrin deposition,206, 207 chronic chorioamnionitis,208-210 and chronic
deciduitis of the placental basal plate.209 The frequency of chronic villitis is higher in
preeclampsia than in normal pregnancy (25% vs 17%).203, 211 Massive perivillous fibrin
Author Manuscript

deposition was present in 20% of patients with preeclampsia.212

Maternal-fetal genetic incompatibility has been implicated in preeclampsia. Placentation is


regulated by interactions between members of the killer cell immunoglobulin-like receptors
(KIRs) expressed by decidual natural killer (dNK) cells and trophoblast human leukocyte
antigen-C (HLA-C) molecules (Figure 11).213-216 Extravillous trophoblasts are specialized
fetal cells that invade the decidua basalis, where they come into direct contact with
maternal immune cells, eg dNK cells, macrophages, T cells, B cells, and dendritic cells.217
Extravillous trophoblasts do not express major T-cell ligands such as HLA-A and HLA-B
molecules. Instead, these fetal cells express HLA-C, HLA-E, and HLA-G.218 The latter two
are largely monomorphic, while HLA-C is polymorphic, and therefore will vary according
to the genetic makeup of the father.219 On the other hand, the dNK cells express the
Author Manuscript

receptors for HLA-C proteins; these receptors are called KIRs.220 Both maternal KIR and
fetal HLA-C genes are highly polymorphic, and the interaction between HLA-C and KIR
has a unique role in placentation through the facilitation of trophoblast invasion of the
decidua and the physiologic transformation of the spiral artery. The molecular machinery
implicated in this process involves chemokines (CXCL-10 and CXCL8, or IL-8) that can
attract trophoblasts expressing their receptors (CXCR3 and CXCR1), metalloproteinases,
and growth factors, including the angiogenic factors (PlGF, VEGF, etc.).221, 222 Moffett
et al 213-215 has shown that normal pregnancy is more common when a mother has
the KIR BB genotype and the fetus has HLA-C1 genes, whereas preeclampsia is more
frequent when a mother has the KIR AA genotype with additional fetal HLA-C2 genes
(HLA-C2 vs HLA-C1 in KIR AA mothers: 45% vs 20%; OR 2.38; 95% CI 1.45-3.90). By
contrast, Staff et al 223 reported that fetal HLA-C2 combined with maternal KIR-BB was
Author Manuscript

associated with placental lesions of acute atherosis. Patients with preeclampsia and acute
atherosis presented this specific genetic combination in 60% of cases.223 The mechanisms
whereby a breakdown of maternal-fetal immune tolerance leads to preeclampsia appear to
involve defective placentation, an example of the convergence of an immune disorder with
uteroplacental ischemia. Such can be the case for other etiologic factors in preeclampsia. For
further details of immunological mechanisms of preeclampsia, the reader is referred to the
review article by Robillard et al 224 in this Supplement.

Am J Obstet Gynecol. Author manuscript; available in PMC 2023 February 01.


JUNG et al. Page 15

Endocrine disorders
Author Manuscript

An association among several endocrine disorders and preeclampsia has been


reported,290-297 yet the evidence for causality is the weakest among those reviewed in
this article. Two explanations are plausible: the low frequency of endocrine disorders in
pregnancy and the paucity of experimental evidence from animal studies of endocrine
disorders. These investigations have not focused on the effect of such disorders in the
development of preeclampsia. Hence, this section will review the evidence of an association
and the proposed mechanism for activation of the common pathway.

Hyperparathyroidism—Normal pregnancy is characterized by changes in maternal


calcium hemostasis, which are thought to occur to meet fetal demands. By term, the fetus
has 30g of calcium, 20g of phosphorus, and 0.8g of magnesium, and 80% of calcium is
deposited during the third trimester.298 The evidence that links altered calcium hemostasis
Author Manuscript

to preeclampsia is as follows: 1) hypocalcemia is a risk factor for preeclampsia (OR 7.63;


95% CI 1.64-35.37);290 2) low vitamin D concentration in early pregnancy is associated
with a 5-fold increased risk for preeclampsia;299 3) hyperparathyroidism is commonly
found in pregnant women with low calcium and vitamin D;300, 301 4) patients diagnosed
with parathyroid adenoma are at an increased risk for preeclampsia (OR 6.89; 95% CI
2.30-20.58);291 5) the absence of a vitamin D prophylaxis in the mother’s childhood is
associated with a subsequent higher risk of preeclampsia;302 and 6) the administration of
vitamin D combined with calcium or a multivitamin supplement has been shown to reduce
blood pressure and the frequency of preeclampsia in a randomized trial.303, 304 The proposed
mechanisms whereby primary hyperparathyroidism leads to preeclampsia are thought to
involve endothelial cell damage, increased insulin resistance, left ventricular hypertrophy,
and dyslipidemia.305, 306
Author Manuscript

Cushing’s syndrome, aldosteronism, pheochromocytoma, and paraganglioma


—Cushing’s syndrome is associated with an increased risk of preeclampsia (aOR 2.20;
95% CI 1.18–4.41).292, 293 In non-pregnant subjects, the chronic glucocorticoid excess
of Cushing’s syndrome is commonly associated with an increased cardiometabolic risk
through an increase in pro-inflammatory adipokine production, alteration of coagulation and
thrombosis factor, platelet function, and endothelial activation.307-309

Primary aldosteronism is the most common form of endocrine hypertension, and it can be
caused by aldosterone-producing adenoma or bilateral adrenal hyperplasia.310, 311 Increased
aldosterone secretion suppresses renin activity, leading to hypertension, hypokalemia,
and hypernatremia.312, 313 Twenty-five percent of women with this disorder develop
preeclampsia.294, 295
Author Manuscript

Pheochromocytoma and paraganglioma are rare catecholamine-producing tumors, with


a reported frequency of 1/54,000 pregnancies.314 An excess of catecholamine released
by these tumors can elicit signs or symptoms similar to preeclampsia (eg hypertension,
headache, and proteinuria), making the diagnosis of pheochromocytoma during pregnancy
difficult.314, 315 A recent meta-analysis that included 204 pregnant patients with
pheochromocytoma paraganglioma showed that 20% of patients are initially misdiagnosed

Am J Obstet Gynecol. Author manuscript; available in PMC 2023 February 01.


JUNG et al. Page 16

as preeclampsia.316 Maternal and fetal mortality are as high as 28% and 27%, respectively,
when there is a delay in diagnosis and treatment.316, 317 Only a few studies have reported
Author Manuscript

that patients with pheochromocytoma developed superimposed preeclampsia;296, 297


therefore, the existence of such a relationship is not clear yet.316

Conclusion
Multiple and sometimes overlapping insults can induce an adaptive homeostatic vascular
response in pregnancy, which can be recognized clinically by the presence of hypertension
and proteinuria (or other signs of multi-systemic involvement). When this response becomes
maladaptive, it can cause target organ damage and becomes potentially life-threatening
to the mother and fetus. This article has reviewed the etiological factors responsible for
preeclampsia and eclampsia.
Author Manuscript

At present, the classification of the syndrome is largely based on the gestational age at
the time of diagnosis (early-onset vs late-onset preeclampsia). Early-onset preeclampsia is
associated with defective placentation, while late-onset preeclampsia appears to be related
to the mismatch between maternal perfusion and feto-placental demands, together with a
maternal predisposition to cardiovascular disease. However, it is necessary to identify the
fundamental causes of the vascular dysfunction responsible for preeclampsia and to develop
comprehensive predictive models and preventive interventions. This review highlights the
multiple etiologies of the syndrome of preeclampsia. We propose that multiple etiologic
factors converge to cause endothelial cell dysfunction, intravascular inflammation, and
syncytiotrophoblast stress. The recognition that a viral infection as a result of SARS-CoV-2
infection can induce preeclampsia brings challenging questions of whether preeclampsia is
a pregnancy-specific disorder caused by the placenta and cured only by delivery. Further
Author Manuscript

research is required to assess the relative contribution of each cause and the effect of
interventions to prevent this syndrome. A greater understanding of the differences in the
etiology of each subtype of preeclampsia and the pathophysiology of other great obstetrical
syndromes are required in order to improve the understanding of this elusive disease.

Funding:
This research was supported, in part, by the Perinatology Research Branch, Division of Obstetrics and Maternal-
Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and
Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/
DHHS); and, in part, with Federal funds from NICHD/NIH/DHHS under Contract No. HHSN275201300006C.
This research was also supported by the Wayne State University Perinatal Initiative in Maternal, Perinatal and Child
Health (NG-L).

References
Author Manuscript

1. Young J The etiology of eclampsia and albuminuria and their relation to accidental hemorrhage.
Transactions Edinburgh Obstetrical Society 1914;39:153–202. [PubMed: 29612444]
2. Dixon WE, Taylor FE. An Epidiascopic Demonstration on "The Physiological Action of the
Placenta.". Proc R Soc Med 1908;1:11–3.
3. Ogden E, Hildebrand G, Page EW. Rise of blood pressure during ischemia of the gravid uterus. Proc
Soc Exp Biol Med 1940;43:49–51.

Am J Obstet Gynecol. Author manuscript; available in PMC 2023 February 01.


JUNG et al. Page 17

4. Chaiworapongsa T, Chaemsaithong P, Yeo L, Romero R. Pre-eclampsia part 1: current


understanding of its pathophysiology. Nature reviews Nephrology 2014;10:466–80. [PubMed:
Author Manuscript

25003615]
5. Kumar D Chronic placental ischemia in relation to toxemias of pregnancy: a preliminary report. Am
J Obstet Gynecol 1962;84:1323–29.
6. Piering WF, Garancis JG, Becker CG, Beres JA, Lemann J Jr. Preeclampsia related to a functioning
extrauterine placenta: report of a case and 25-year follow-up. Am J Kidney Dis 1993;21:310–3.
[PubMed: 8447308]
7. Berger M, Cavanagh D. Toxemia of pregnancy. The hypertensive effect of acute experimental
placental ischemia. Am J Obstet Gynecol 1963;87:293–305. [PubMed: 14072393]
8. Browne JC, Veall N. The maternal placental blood flow in normotensive and hypertensive women. J
Obstet Gynaecol Br Emp 1953;60:141–7. [PubMed: 13053276]
9. Janisch H, Leodolter S. Results of placental circulation measurements in hazard pregnancies. Z
Geburtshilfe Perinatol 1973;177:74–80. [PubMed: 4198070]
10. Olkkonen HS, Suonio S, Haring P. Determination of placental blood flow by external monitoring
of 113In. Nuklearmedizin 1976;15:168–72. [PubMed: 980794]
Author Manuscript

11. Lippert TH, Cloeren SE, Fridrich R. Assessment of uteroplacental hemodynamics in complicated
pregnancy. Int J Gynaecol Obstet 1978;16:274–80. [PubMed: 35387]
12. Käär K, Jouppila P, Kuikka J, Luotola H, Toivanen J, Rekonen A. Intervillous blood flow in normal
and complicated late pregnancy measured by means of an intravenous 133Xe method. Acta Obstet
Gynecol Scand 1980;59:7–10. [PubMed: 6992498]
13. Lunell NO, Nylund LE, Lewander R, Sarby B. Uteroplacental blood flow in pre-eclampsia
measurements with indium-113m and a computer-linked gamma camera. Clin Exp Hypertens
B 1982;1:105–17. [PubMed: 7184662]
14. Alexander BT, Kassab SE, Miller MT, et al. Reduced uterine perfusion pressure during pregnancy
in the rat is associated with increases in arterial pressure and changes in renal nitric oxide.
Hypertension 2001;37:1191–5. [PubMed: 11304523]
15. Makris A, Thornton C, Thompson J, et al. Uteroplacental ischemia results in proteinuric
hypertension and elevated sFLT-1. Kidney Int 2007;71:977–84. [PubMed: 17377512]
16. Turanov AA, Lo A, Hassler MR, et al. RNAi modulation of placental sFLT1 for the treatment of
Author Manuscript

preeclampsia. Nat Biotechnol 2018.


17. Bakrania BA, George EM, Granger JP. Animal models of preeclampsia: investigating
pathophysiology and therapeutic targets. Am J Obstet Gynecol 2021.
18. Brosens I [Spiraled arterioles of the decidua basalis in the hypertensive complications of
pregnancy. Anatomoclinical study]. Bull Soc R Belge Gynecol Obstet 1963;33:61–72. [PubMed:
14015832]
19. Brosens I A STUDY OF THE SPIRAL ARTERIES OF THE DECIDUA BASALIS IN
NORMOTENSIVE AND HYPERTENSIVE PREGNANCIES. J Obstet Gynaecol Br Commonw
1964;71:222–30. [PubMed: 14138383]
20. Melchiorre K, Giorgione V, Thilaganathan B. The placenta and preeclampsia: villain or victim?
Am J Obstet Gynecol 2021.
21. Hertig A Vascular pathology in hypertensive albuminuric toxemias of pregnancy. Clinics
1945;4:602.
22. De Wolf F, Robertson WB, Brosens I. The ultrastructure of acute atherosis in hypertensive
pregnancy. Am J Obstet Gynecol 1975;123:164–74. [PubMed: 1163579]
Author Manuscript

23. Staff AC, Johnsen GM, Dechend R, Redman CW. Preeclampsia and uteroplacental acute atherosis:
immune and inflammatory factors. J Reprod Immunol 2014;101-102:120–6. [PubMed: 24119981]
24. Kim YM, Chaemsaithong P, Romero R, et al. Placental lesions associated with acute atherosis. J
Matern Fetal Neonatal Med 2015;28:1554–62. [PubMed: 25183023]
25. Labarrere CA, DiCarlo HL, Bammerlin E, et al. Failure of physiologic transformation of spiral
arteries, endothelial and trophoblast cell activation, and acute atherosis in the basal plate of the
placenta. Am J Obstet Gynecol 2017;216:287 e1–87 e16. [PubMed: 28034657]

Am J Obstet Gynecol. Author manuscript; available in PMC 2023 February 01.


JUNG et al. Page 18

26. McMaster-Fay R Uteroplacental vascular syndromes: Theories, hypotheses and controversies.


Clinical Obstetrics, Gynecology and Reproductive Medicine 2018;4.
Author Manuscript

27. Falco ML, Sivanathan J, Laoreti A, Thilaganathan B, Khalil A. Placental histopathology


associated with pre-eclampsia: systematic review and meta-analysis. Ultrasound Obstet Gynecol
2017;50:295–301. [PubMed: 28436167]
28. Staff AC, Fjeldstad HE, Fosheim IK, et al. Failure of physiological transformation and spiral artery
atherosis: their roles in preeclampsia. Am J Obstet Gynecol 2021.
29. Kim YM, Chaemsaithong P, Romero R, et al. The frequency of acute atherosis in normal
pregnancy and preterm labor, preeclampsia, small-for-gestational age, fetal death and midtrimester
spontaneous abortion. J Matern Fetal Neonatal Med 2015:1–9.
30. Kim YM, Bujold E, Chaiworapongsa T, et al. Failure of physiologic transformation of the
spiral arteries in patients with preterm labor and intact membranes. Am J Obstet Gynecol
2003;189:1063–9. [PubMed: 14586356]
31. Kim YM, Chaiworapongsa T, Gomez R, et al. Failure of physiologic transformation of the spiral
arteries in the placental bed in preterm premature rupture of membranes. Am J Obstet Gynecol
2002;187:1137–42. [PubMed: 12439491]
Author Manuscript

32. Labarrere C, Alonso J, Manni J, Domenichini E, Althabe O. Immunohistochemical findings in


acute atherosis associated with intrauterine growth retardation. Am J Reprod Immunol Microbiol
1985;7:149–55. [PubMed: 3893171]
33. Khong TY. Acute atherosis in pregnancies complicated by hypertension, small-for-gestational-age
infants, and diabetes mellitus. Arch Pathol Lab Med 1991;115:722–5. [PubMed: 2064534]
34. Romero R, Kusanovic JP, Chaiworapongsa T, Hassan SS. Placental bed disorders in preterm
labor, preterm PROM, spontaneous abortion and abruptio placentae. Best Pract Res Clin Obstet
Gynaecol 2011;25:313–27. [PubMed: 21388889]
35. Espinoza J, Romero R, Mee Kim Y, et al. Normal and abnormal transformation of the spiral
arteries during pregnancy. J Perinat Med 2006;34:447–58. [PubMed: 17140293]
36. Gallo DM, Poon LC, Akolekar R, Syngelaki A, Nicolaides KH. Prediction of preeclampsia by
uterine artery Doppler at 20-24 weeks' gestation. Fetal Diagn Ther 2013;34:241–7. [PubMed:
24192614]
37. Khalil A, Garcia-Mandujano R, Maiz N, Elkhouli M, Nicolaides KH. Longitudinal changes in
Author Manuscript

uterine artery Doppler and blood pressure and risk of pre-eclampsia. Ultrasound Obstet Gynecol
2014;43:541–7. [PubMed: 24265180]
38. Levine RJ, Lam C, Qian C, et al. Soluble endoglin and other circulating antiangiogenic factors in
preeclampsia. N Engl J Med 2006;355:992–1005. [PubMed: 16957146]
39. Romero R, Nien JK, Espinoza J, et al. 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. [PubMed: 18175241]
40. Erez O, Romero R, Espinoza J, et al. The change in concentrations of angiogenic and anti-
angiogenic factors in maternal plasma between the first and second trimesters in risk assessment
for the subsequent development of preeclampsia and small-for-gestational age. J Matern Fetal
Neonatal Med 2008;21:279–87. [PubMed: 18446652]
41. Kusanovic JP, Romero R, Chaiworapongsa T, et al. A prospective cohort study of the value of
maternal plasma concentrations of angiogenic and anti-angiogenic factors in early pregnancy and
midtrimester in the identification of patients destined to develop preeclampsia. J Matern Fetal
Author Manuscript

Neonatal Med 2009;22:1021–38. [PubMed: 19900040]


42. Chaiworapongsa T, Romero R, Savasan ZA, et al. Maternal plasma concentrations of angiogenic/
anti-angiogenic factors are of prognostic value in patients presenting to the obstetrical triage area
with the suspicion of preeclampsia. J Matern Fetal Neonatal Med 2011;24:1187–207. [PubMed:
21827221]
43. Vaisbuch E, Whitty JE, Hassan SS, et al. Circulating angiogenic and antiangiogenic factors in
women with eclampsia. Am J Obstet Gynecol 2011;204:152.e1–9. [PubMed: 21062661]
44. Chaiworapongsa T, Romero R, Korzeniewski SJ, et al. Plasma concentrations of angiogenic/anti-
angiogenic factors have prognostic value in women presenting with suspected preeclampsia to

Am J Obstet Gynecol. Author manuscript; available in PMC 2023 February 01.


JUNG et al. Page 19

the obstetrical triage area: a prospective study. J Matern Fetal Neonatal Med 2014;27:132–44.
[PubMed: 23687930]
Author Manuscript

45. Espinoza J, Romero R, Nien JK, et al. Identification of patients at risk for early onset and/or severe
preeclampsia with the use of uterine artery Doppler velocimetry and placental growth factor. Am J
Obstet Gynecol 2007;196:326.e1–13. [PubMed: 17403407]
46. Ogge G, Chaiworapongsa T, Romero R, et al. Placental lesions associated with maternal
underperfusion are more frequent in early-onset than in late-onset preeclampsia. J Perinat Med
2011;39:641–52. [PubMed: 21848483]
47. Rana S, Schnettler WT, Powe C, et al. Clinical characterization and outcomes of preeclampsia with
normal angiogenic profile. Hypertens Pregnancy 2013;32:189–201. [PubMed: 23725084]
48. Orabona R, Donzelli CM, Falchetti M, Santoro A, Valcamonico A, Frusca T. Placental histological
patterns and uterine artery Doppler velocimetry in pregnancies complicated by early or late
pre-eclampsia. Ultrasound Obstet Gynecol 2016;47:580–5. [PubMed: 26511592]
49. Rolnik DL, Wright D, Poon LC, et al. Aspirin versus Placebo in Pregnancies at High Risk for
Preterm Preeclampsia. N Engl J Med 2017;377:613–22. [PubMed: 28657417]
50. Relman AS. Aspirin for the primary prevention of myocardial infarction. N Engl J Med
Author Manuscript

1988;318:245–6. [PubMed: 3275897]


51. Albert. Archiv für Gynaekologie 1901;Ixiii:488
52. Loudon I Some historical aspects of toxaemia of pregnancy. A review. BJOG 1991;98:853–58.
53. DeLee J Theories of eclampsia. Am J Obstet 1905;51:325–30.
54. Conde-Agudelo A, Villar J, Lindheimer M. Maternal infection and risk of preeclampsia: systematic
review and metaanalysis. Am J Obstet Gynecol 2008;198:7–22. [PubMed: 18166297]
55. Williams RC. Periodontal disease. N Engl J Med 1990;322:373–82. [PubMed: 2405268]
56. Sanz M, Del Castillo AM, Jepsen S, et al. Periodontitis and Cardiovascular Diseases. Consensus
Report. Global heart 2020;15:1. [PubMed: 32489774]
57. Herrera D, Molina A, Buhlin K, Klinge B. Periodontal diseases and association with
atherosclerotic disease. Periodontol 2000 2020;83:66–89. [PubMed: 32385870]
58. Sen S, Redd K, Trivedi T, et al. Periodontal Disease, Atrial Fibrillation and Stroke. Am Heart J
2021;235:36–43. [PubMed: 33503409]
59. Hajishengallis G, Chavakis T. Local and systemic mechanisms linking periodontal disease and
Author Manuscript

inflammatory comorbidities. Nat Rev Immunol 2021;21:426–40. [PubMed: 33510490]


60. Velsko IM, Chukkapalli SS, Rivera MF, et al. Active invasion of oral and aortic tissues by
Porphyromonas gingivalis in mice causally links periodontitis and atherosclerosis. PLoS One
2014;9:e97811. [PubMed: 24836175]
61. Lalla E, Lamster IB, Hofmann MA, et al. Oral infection with a periodontal pathogen accelerates
early atherosclerosis in apolipoprotein E-null mice. Arterioscler Thromb Vasc Biol 2003;23:1405–
11. [PubMed: 12816879]
62. Velsko IM, Chukkapalli SS, Rivera-Kweh MF, et al. Periodontal pathogens invade gingiva and
aortic adventitia and elicit inflammasome activation in αvβ6 integrin-deficient mice. Infect Immun
2015;83:4582–93. [PubMed: 26371120]
63. Ruma M, Boggess K, Moss K, et al. Maternal periodontal disease, systemic inflammation, and risk
for preeclampsia. Am J Obstet Gynecol 2008;198:389.e1–5. [PubMed: 18295179]
64. Easter SR, Cantonwine DE, Zera CA, Lim KH, Parry SI, McElrath TF. Urinary tract infection
during pregnancy, angiogenic factor profiles, and risk of preeclampsia. Am J Obstet Gynecol
Author Manuscript

2016;214:387.e1–7. [PubMed: 26450405]


65. Yan L, Jin Y, Hang H, Yan B. The association between urinary tract infection during pregnancy and
preeclampsia: A meta-analysis. Medicine (Baltimore) 2018;97:e12192. [PubMed: 30200124]
66. Tambyah PA, Maki DG. Catheter-associated urinary tract infection is rarely symptomatic: a
prospective study of 1,497 catheterized patients. Arch Intern Med 2000;160:678–82. [PubMed:
10724054]
67. Sartelet H, Rogier C, Milko-Sartelet I, Angel G, Michel G. Malaria associated pre-eclampsia in
Senegal. Lancet 1996;347:1121.

Am J Obstet Gynecol. Author manuscript; available in PMC 2023 February 01.


JUNG et al. Page 20

68. Ndao CT, Dumont A, Fievet N, Doucoure S, Gaye A, Lehesran JY. Placental malarial infection as a
risk factor for hypertensive disorders during pregnancy in Africa: a case-control study in an urban
Author Manuscript

area of Senegal, West Africa. Am J Epidemiol 2009;170:847–53. [PubMed: 19679749]


69. Mruma HA, McQuillan R, Norrie J. The association of malaria infection and gestational
hypertension in Africa: Systematic review and meta-analysis. Journal of global health
2020;10:020417. [PubMed: 33110577]
70. Carreiras M, Montagnani S, Layrisse Z. Preeclampsia: a multifactorial disease resulting from the
interaction of the feto-maternal HLA genotype and HCMV infection. Am J Reprod Immunol
2002;48:176–83. [PubMed: 12443029]
71. von Dadelszen P, Magee LA, Krajden M, et al. Levels of antibodies against cytomegalovirus and
Chlamydophila pneumoniae are increased in early onset pre-eclampsia. BJOG 2003;110:725–30.
[PubMed: 12892683]
72. Mattar R, Amed AM, Lindsey PC, Sass N, Daher S. Preeclampsia and HIV infection. Eur J Obstet
Gynecol Reprod Biol 2004;117:240–1. [PubMed: 15541864]
73. Nourollahpour Shiadeh M, Riahi SM, Khani S, et al. Human Immunodeficiency Virus and risk of
pre-eclampsia and eclampsia in pregnant women: A meta-analysis on cohort studies. Pregnancy
Author Manuscript

Hypertens 2019;17:269–75. [PubMed: 31487651]


74. Faas MM, Schuiling GA, Baller JF, Visscher CA, Bakker WW. A new animal model for
human preeclampsia: ultra-low-dose endotoxin infusion in pregnant rats. Am J Obstet Gynecol
1994;171:158–64. [PubMed: 8030692]
75. Somani SS, Richter F, Fuster V, et al. Characterization of Patients Who Return to Hospital
Following Discharge from Hospitalization for COVID-19. J Gen Intern Med 2020;35:2838–44.
[PubMed: 32815060]
76. Xiong S, Liu L, Lin F, et al. Clinical characteristics of 116 hospitalized patients with COVID-19 in
Wuhan, China: a single-centered, retrospective, observational study. BMC Infect Dis 2020;20:787.
[PubMed: 33092539]
77. Huang C, Huang L, Wang Y, et al. 6-month consequences of COVID-19 in patients discharged
from hospital: a cohort study. The Lancet 2021;397:220–32.
78. Nadim MK, Forni LG, Mehta RL, et al. COVID-19-associated acute kidney injury: consensus
report of the 25th Acute Disease Quality Initiative (ADQI) Workgroup. Nature reviews
Nephrology 2020;16:747–64. [PubMed: 33060844]
Author Manuscript

79. Zahid U, Ramachandran P, Spitalewitz S, et al. Acute Kidney Injury in COVID-19 Patients:
An Inner City Hospital Experience and Policy Implications. Am J Nephrol 2020;51:786–96.
[PubMed: 33011717]
80. Martinez-Rojas MA, Vega-Vega O, Bobadilla NA. Is the kidney a target of SARS-CoV-2? Am J
Physiol Renal Physiol 2020;318:F1454–f62. [PubMed: 32412303]
81. Cheng Y, Luo R, Wang K, et al. Kidney disease is associated with in-hospital death of patients with
COVID-19. Kidney Int 2020;97:829–38. [PubMed: 32247631]
82. Rahi MS, Jindal V, Reyes SP, Gunasekaran K, Gupta R, Jaiyesimi I. Hematologic disorders
associated with COVID-19: a review. Ann Hematol 2021;100:309–20. [PubMed: 33415422]
83. Castro RA, Frishman WH. Thrombotic Complications of COVID-19 Infection: A Review. Cardiol
Rev 2021;29:43–47. [PubMed: 32947478]
84. Li J, Fan JG. Characteristics and Mechanism of Liver Injury in 2019 Coronavirus Disease. Journal
of clinical and translational hepatology 2020;8:13–17. [PubMed: 32274341]
85. Saviano A, Wrensch F, Ghany MG, Baumert TF. Liver Disease and Coronavirus Disease 2019:
Author Manuscript

From Pathogenesis to Clinical Care. Hepatology 2021;74:1088–100. [PubMed: 33332624]


86. Conde-Agudelo A, Romero R. SARS-COV-2 infection during pregnancy and risk of preeclampsia:
a systematic review and meta-analysis. Am J Obstet Gynecol 2021.
87. Lai J, Romero R, Tarca AL, et al. SARS-COV-2 and the subsequent development of preeclampsia
and preterm birth: evidence of a dose response relationship supporting causality. Am J Obstet
Gynecol 2021.
88. Rosenbloom JI, Raghuraman N, Carter EB, Kelly JC. Coronavirus disease 2019 infection
and hypertensive disorders of pregnancy. Am J Obstet Gynecol 2021;224:623–24. [PubMed:
33675794]

Am J Obstet Gynecol. Author manuscript; available in PMC 2023 February 01.


JUNG et al. Page 21

89. Varga Z, Flammer AJ, Steiger P, et al. Endothelial cell infection and endotheliitis in COVID-19.
The Lancet 2020;395:1417–18.
Author Manuscript

90. Ackermann M, Verleden SE, Kuehnel M, et al. Pulmonary Vascular Endothelialitis, Thrombosis,
and Angiogenesis in Covid-19. N Engl J Med 2020;383:120–28. [PubMed: 32437596]
91. Su H, Yang M, Wan C, et al. Renal histopathological analysis of 26 postmortem findings of
patients with COVID-19 in China. Kidney Int 2020;98:219–27. [PubMed: 32327202]
92. Sharifian-Dorche M, Huot P, Osherov M, et al. Neurological complications of coronavirus
infection; a comparative review and lessons learned during the COVID-19 pandemic. J Neurol
Sci 2020;417:117085. [PubMed: 32871412]
93. Thakur V, Ratho RK, Kumar P, et al. Multi-Organ Involvement in COVID-19: Beyond Pulmonary
Manifestations. Journal of clinical medicine 2021;10.
94. Mendoza M, Garcia-Ruiz I, Maiz N, et al. Pre-eclampsia-like syndrome induced by severe
COVID-19: a prospective observational study. BJOG 2020;127:1374–80. [PubMed: 32479682]
95. Giardini V, Carrer A, Casati M, Contro E, Vergani P, Gambacorti-Passerini C. Increased sFLT-1/
PlGF ratio in COVID-19: A novel link to angiotensin II-mediated endothelial dysfunction. Am J
Hematol 2020;95:E188–e91. [PubMed: 32472588]
Author Manuscript

96. Espino-y-Sosa S, Martinez-Portilla RJ, Torres-Torres J, et al. Novel Ratio Soluble Fms-like
Tyrosine Kinase-1/Angiotensin-II (sFlt-1/ANG-II) in Pregnant Women Is Associated with Critical
Illness in COVID-19. Viruses 2021;13:1906. [PubMed: 34696336]
97. Lee JW, Lee IH, Sato T, Kong SW, Iimura T. Genetic variation analyses indicate conserved
SARS-CoV-2-host interaction and varied genetic adaptation in immune-response factors in modern
human evolution. Dev Growth Differ 2021.
98. Choudhary S, Sreenivasulu K, Mitra P, Misra S, Sharma P. Role of Genetic Variants and Gene
Expression in the Susceptibility and Severity of COVID-19. Ann Lab Med 2021;41:129–38.
[PubMed: 33063674]
99. Backhed F, Ding H, Wang T, et al. The gut microbiota as an environmental factor that regulates fat
storage. Proc Natl Acad Sci U S A 2004;101:15718–23. [PubMed: 15505215]
100. Ley RE, Turnbaugh PJ, Klein S, Gordon JI. Microbial ecology: human gut microbes associated
with obesity. Nature 2006;444:1022–3. [PubMed: 17183309]
101. Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER, Gordon JI. An obesity-associated
Author Manuscript

gut microbiome with increased capacity for energy harvest. Nature 2006;444:1027–31. [PubMed:
17183312]
102. Jia W, Li H, Zhao L, Nicholson JK. Gut microbiota: a potential new territory for drug targeting.
Nature reviews Drug discovery 2008;7:123–9. [PubMed: 18239669]
103. Hooper LV, Littman DR, Macpherson AJ. Interactions between the microbiota and the immune
system. Science 2012;336:1268–73. [PubMed: 22674334]
104. Koren O, Goodrich JK, Cullender TC, et al. Host remodeling of the gut microbiome and
metabolic changes during pregnancy. Cell 2012;150:470–80. [PubMed: 22863002]
105. Kimura I, Miyamoto J, Ohue-Kitano R, et al. Maternal gut microbiota in pregnancy influences
offspring metabolic phenotype in mice. Science 2020;367.
106. Jonsson AL, Bäckhed F. Role of gut microbiota in atherosclerosis. Nat Rev Cardiol 2017;14:79–
87. [PubMed: 27905479]
107. Sanchez-Rodriguez E, Egea-Zorrilla A, Plaza-Díaz J, et al. The Gut Microbiota and Its
Implication in the Development of Atherosclerosis and Related Cardiovascular Diseases.
Nutrients 2020;12.
Author Manuscript

108. Li J, Zhao F, Wang Y, et al. Gut microbiota dysbiosis contributes to the development of
hypertension. Microbiome 2017;5:14. [PubMed: 28143587]
109. Kim S, Goel R, Kumar A, et al. Imbalance of gut microbiome and intestinal epithelial barrier
dysfunction in patients with high blood pressure. Clin Sci (Lond) 2018;132:701–18. [PubMed:
29507058]
110. Kanbay M, Onal EM, Afsar B, et al. The crosstalk of gut microbiota and chronic kidney
disease: role of inflammation, proteinuria, hypertension, and diabetes mellitus. Int Urol Nephrol
2018;50:1453–66. [PubMed: 29728993]

Am J Obstet Gynecol. Author manuscript; available in PMC 2023 February 01.


JUNG et al. Page 22

111. Leshem A, Horesh N, Elinav E. Fecal Microbial Transplantation and Its Potential Application in
Cardiometabolic Syndrome. Front Immunol 2019;10:1341. [PubMed: 31258528]
Author Manuscript

112. Kazemian N, Mahmoudi M, Halperin F, Wu JC, Pakpour S. Gut microbiota and cardiovascular
disease: opportunities and challenges. Microbiome 2020;8:36. [PubMed: 32169105]
113. Dunlop AL, Mulle JG, Ferranti EP, Edwards S, Dunn AB, Corwin EJ. Maternal Microbiome and
Pregnancy Outcomes That Impact Infant Health: A Review. Adv Neonatal Care 2015;15:377–85.
[PubMed: 26317856]
114. Hampton T Do Gut Bacteria Play a Role in Preeclampsia? JAMA 2020;323:2120–21. [PubMed:
32484518]
115. Chen X, Li P, Liu M, et al. Gut dysbiosis induces the development of pre-eclampsia through
bacterial translocation. Gut 2020;69:513–22. [PubMed: 31900289]
116. Beckers KF, Sones JL. Maternal microbiome and the hypertensive disorder of pregnancy,
preeclampsia. Am J Physiol Heart Circ Physiol 2020;318:H1–h10. [PubMed: 31626558]
117. Altemani F, Barrett HL, Gomez-Arango L, et al. Pregnant women who develop preeclampsia
have lower abundance of the butyrate-producer Coprococcus in their gut microbiota. Pregnancy
Hypertens 2021;23:211–19. [PubMed: 33530034]
Author Manuscript

118. Gregory JC, Buffa JA, Org E, et al. Transmission of atherosclerosis susceptibility with gut
microbial transplantation. J Biol Chem 2015;290:5647–60. [PubMed: 25550161]
119. Wang J, Gu X, Yang J, Wei Y, Zhao Y. Gut Microbiota Dysbiosis and Increased Plasma LPS and
TMAO Levels in Patients With Preeclampsia. Frontiers in cellular and infection microbiology
2019;9:409. [PubMed: 31850241]
120. Lv LJ, Li SH, Li SC, et al. Early-Onset Preeclampsia Is Associated With Gut Microbial
Alterations in Antepartum and Postpartum Women. Frontiers in cellular and infection
microbiology 2019;9:224. [PubMed: 31297341]
121. Schneider S, Freerksen N, Röhrig S, Hoeft B, Maul H. Gestational diabetes and preeclampsia--
similar risk factor profiles? Early Hum Dev 2012;88:179–84. [PubMed: 21890288]
122. Weissgerber TL, Mudd LM. Preeclampsia and diabetes. Curr Diab Rep 2015;15:9. [PubMed:
25644816]
123. Duckitt K, Harrington D. Risk factors for pre-eclampsia at antenatal booking: systematic review
of controlled studies. BMJ 2005;330:565. [PubMed: 15743856]
Author Manuscript

124. Lisonkova S, Joseph KS. Incidence of preeclampsia: risk factors and outcomes associated
with early-versus late-onset disease. Am J Obstet Gynecol 2013;209:544.e1–44.e12. [PubMed:
23973398]
125. Metzger BE, Lowe LP, Dyer AR, et al. Hyperglycemia and adverse pregnancy outcomes. N Engl
J Med 2008;358:1991–2002. [PubMed: 18463375]
126. Kinshella MW, Omar S, Scherbinsky K, et al. Maternal Dietary Patterns and Pregnancy
Hypertension in Low- and Middle-Income Countries: A Systematic Review and Meta-analysis.
Adv Nutr 2021.
127. Crowther CA, Hiller JE, Moss JR, McPhee AJ, Jeffries WS, Robinson JS. Effect of treatment
of gestational diabetes mellitus on pregnancy outcomes. N Engl J Med 2005;352:2477–86.
[PubMed: 15951574]
128. Landon MB, Spong CY, Thom E, et al. A multicenter, randomized trial of treatment for mild
gestational diabetes. N Engl J Med 2009;361:1339–48. [PubMed: 19797280]
129. Romero R, Erez O, Huttemann M, et al. Metformin, the aspirin of the 21st century: its role
in gestational diabetes mellitus, prevention of preeclampsia and cancer, and the promotion of
Author Manuscript

longevity. Am J Obstet Gynecol 2017;217:282–302. [PubMed: 28619690]


130. Kalafat E, Sukur YE, Abdi A, Thilaganathan B, Khalil A. Metformin for prevention of
hypertensive disorders of pregnancy in women with gestational diabetes or obesity: systematic
review and meta-analysis of randomized trials. Ultrasound Obstet Gynecol 2018;52:706–14.
[PubMed: 29749110]
131. Alqudah A, McKinley MC, McNally R, et al. Risk of pre-eclampsia in women taking metformin:
a systematic review and meta-analysis. Diabet Med 2018;35:160–72. [PubMed: 29044702]

Am J Obstet Gynecol. Author manuscript; available in PMC 2023 February 01.


JUNG et al. Page 23

132. Tarry-Adkins JL, Ozanne SE, Aiken CE. Impact of metformin treatment during pregnancy
on maternal outcomes: a systematic review/meta-analysis. Sci Rep 2021;11:9240. [PubMed:
Author Manuscript

33927270]
133. Cluver CA, Hiscock R, Decloedt EH, et al. Use of metformin to prolong gestation in preterm pre-
eclampsia: randomised, double blind, placebo controlled trial. BMJ 2021;374:n2103. [PubMed:
34551918]
134. Davenport MH, Ruchat SM, Poitras VJ, et al. Prenatal exercise for the prevention of gestational
diabetes mellitus and hypertensive disorders of pregnancy: a systematic review and meta-
analysis. Br J Sports Med 2018;52:1367–75. [PubMed: 30337463]
135. Sebire NJ, Jolly M, Harris JP, et al. Maternal obesity and pregnancy outcome: a study of 287,213
pregnancies in London. Int J Obes Relat Metab Disord 2001;25:1175–82. [PubMed: 11477502]
136. Roberts JM, Bodnar LM, Patrick TE, Powers RW. The Role of Obesity in Preeclampsia.
Pregnancy Hypertens 2011;1:6–16. [PubMed: 21532964]
137. Chaemsaithong P, Leung TY, Sahota D, et al. Body mass index at 11-13 weeks' gestation and
pregnancy complications in a Southern Chinese population: a retrospective cohort study. J Matern
Fetal Neonatal Med 2019;32:2056–68. [PubMed: 29301455]
Author Manuscript

138. Wang Z, Wang P, Liu H, et al. Maternal adiposity as an independent risk factor for pre-eclampsia:
a meta-analysis of prospective cohort studies. Obes Rev 2013;14:508–21. [PubMed: 23530552]
139. Conde-Agudelo A, Belizan JM. Risk factors for pre-eclampsia in a large cohort of Latin
American and Caribbean women. BJOG 2000;107:75–83. [PubMed: 10645865]
140. Catov JM, Ness RB, Kip KE, Olsen J. Risk of early or severe pre-eclampsia related to pre-
existing conditions. Int J Epidemiol 2007;36:412–9. [PubMed: 17255351]
141. Bodnar LM, Catov JM, Klebanoff MA, Ness RB, Roberts JM. Prepregnancy body mass index
and the occurrence of severe hypertensive disorders of pregnancy. Epidemiology 2007;18:234–9.
[PubMed: 17237733]
142. Bicocca MJ, Mendez-Figueroa H, Chauhan SP, Sibai BM. Maternal Obesity and the
Risk of Early-Onset and Late-Onset Hypertensive Disorders of Pregnancy. Obstet Gynecol
2020;136:118–27. [PubMed: 32541276]
143. Schenkelaars N, Rousian M, Hoek J, Schoenmakers S, Willemsen S, Steegers-Theunissen R.
Preconceptional maternal weight loss and hypertensive disorders in pregnancy: a systematic
Author Manuscript

review and meta-analysis. Eur J Clin Nutr 2021.


144. Hotamisligil GS. Inflammation and metabolic disorders. Nature 2006;444:860–7. [PubMed:
17167474]
145. Kaur J A comprehensive review on metabolic syndrome. Cardiol Res Pract 2014;2014:943162.
[PubMed: 24711954]
146. Bakker W, Eringa EC, Sipkema P, van Hinsbergh VW. Endothelial dysfunction and diabetes: roles
of hyperglycemia, impaired insulin signaling and obesity. Cell Tissue Res 2009;335:165–89.
[PubMed: 18941783]
147. Funk SD, Yurdagul A Jr., Orr AW. Hyperglycemia and endothelial dysfunction in atherosclerosis:
lessons from type 1 diabetes. Int J Vasc Med 2012;2012:569654. [PubMed: 22489274]
148. Stekkinger E, Scholten R, van der Vlugt MJ, van Dijk AP, Janssen MC, Spaanderman ME.
Metabolic syndrome and the risk for recurrent pre-eclampsia: a retrospective cohort study. BJOG
2013;120:979–86. [PubMed: 23464593]
149. Spradley FT. Metabolic abnormalities and obesity's impact on the risk for developing
preeclampsia. Am J Physiol Regul Integr Comp Physiol 2017;312:R5–r12. [PubMed: 27903516]
Author Manuscript

150. Cho GJ, Park JH, Shin SA, Oh MJ, Seo HS. Metabolic syndrome in the non-pregnant state
is associated with the development of preeclampsia. Int J Cardiol 2016;203:982–6. [PubMed:
26625326]
151. Hubel CA. Dyslipidemia, iron, and oxidative stress in preeclampsia: assessment of maternal and
feto-placental interactions. Semin Reprod Endocrinol 1998;16:75–92. [PubMed: 9654610]
152. Pouta A, Hartikainen AL, Sovio U, et al. Manifestations of metabolic syndrome after
hypertensive pregnancy. Hypertension 2004;43:825–31. [PubMed: 14981067]
153. Forest JC, Girouard J, Massé J, et al. Early occurrence of metabolic syndrome after hypertension
in pregnancy. Obstet Gynecol 2005;105:1373–80. [PubMed: 15932832]

Am J Obstet Gynecol. Author manuscript; available in PMC 2023 February 01.


JUNG et al. Page 24

154. Smith GN, Pudwell J, Walker M, Wen SW. Risk estimation of metabolic syndrome at one and
three years after a pregnancy complicated by preeclampsia. J Obstet Gynaecol Can 2012;34:836–
Author Manuscript

41. [PubMed: 22971452]


155. Bennett WL, Gilson MM, Jamshidi R, et al. Impact of bariatric surgery on hypertensive disorders
in pregnancy: retrospective analysis of insurance claims data. BMJ 2010;340:c1662. [PubMed:
20388692]
156. Jonk AM, Houben AJ, de Jongh RT, Serné EH, Schaper NC, Stehouwer CD. Microvascular
dysfunction in obesity: a potential mechanism in the pathogenesis of obesity-associated insulin
resistance and hypertension. Physiology (Bethesda) 2007;22:252–60. [PubMed: 17699878]
157. Muniyappa R, Sowers JR. Role of insulin resistance in endothelial dysfunction. Rev Endocr
Metab Disord 2013;14:5–12. [PubMed: 23306778]
158. Lu Q, Zhang X, Wang Y, et al. Sleep disturbances during pregnancy and adverse maternal
and fetal outcomes: A systematic review and meta-analysis. Sleep Med Rev 2021;58:101436.
[PubMed: 33571887]
159. Dalmasso F, Prota R. Snoring: analysis, measurement, clinical implications and applications. Eur
Respir J 1996;9:146–59. [PubMed: 8834348]
Author Manuscript

160. Loube DI, Poceta JS, Morales MC, Peacock MD, Mitler MM. Self-reported snoring in pregnancy.
Association with fetal outcome. Chest 1996;109:885–9. [PubMed: 8635365]
161. Franklin KA, Holmgren PA, Jönsson F, Poromaa N, Stenlund H, Svanborg E. Snoring, pregnancy-
induced hypertension, and growth retardation of the fetus. Chest 2000;117:137–41. [PubMed:
10631211]
162. Hu FB, Willett WC, Colditz GA, et al. Prospective study of snoring and risk of hypertension in
women. Am J Epidemiol 1999;150:806–16. [PubMed: 10522651]
163. O'Brien LM, Bullough AS, Owusu JT, et al. Pregnancy-onset habitual snoring,
gestational hypertension, and preeclampsia: prospective cohort study. Am J Obstet Gynecol
2012;207:487.e1–9. [PubMed: 22999158]
164. Edwards N, Blyton DM, Kirjavainen T, Kesby GJ, Sullivan CE. Nasal continuous positive airway
pressure reduces sleep-induced blood pressure increments in preeclampsia. Am J Respir Crit
Care Med 2000;162:252–7. [PubMed: 10903250]
165. Whitehead C, Tong S, Wilson D, Howard M, Walker SP. Treatment of early-onset preeclampsia
Author Manuscript

with continuous positive airway pressure. Obstet Gynecol 2015;125:1106–9. [PubMed:


25774926]
166. Bourjeily G, Curran P, Butterfield K, Maredia H, Carpenter M, Lambert-Messerlian G. Placenta-
secreted circulating markers in pregnant women with obstructive sleep apnea. J Perinat Med
2015;43:81–7. [PubMed: 24846956]
167. Daly A, Robertson A, Bobek G, Middleton S, Sullivan C, Hennessy A. Sleep disordered breathing
controlled by CPAP and sFlt-1 in a pregnant patient with chronic hypertension: Case report and
literature review. Obstetric medicine 2018;11:32–34. [PubMed: 29636812]
168. Redman CW, Beilin LJ, Bonnar J. Reversed diurnal blood pressure rhythm in hypertensive
pregnancies. Clin Sci Mol Med Suppl 1976;3:687s–89s. [PubMed: 1071709]
169. Beilin LJ, Deacon J, Michael CA, et al. Diurnal rhythms of blood pressure, plasma renin activity,
angiotensin II and catecholamines in normotensive and hypertensive pregnancies. Clin Exp
Hypertens B 1983;2:271–93. [PubMed: 6347445]
170. Edwards N, Blyton CM, Kesby GJ, Wilcox I, Sullivan CE. Pre-eclampsia is associated with
marked alterations in sleep architecture. Sleep 2000;23:619–25. [PubMed: 10947029]
Author Manuscript

171. Sacks GP, Studena K, Sargent K, Redman CW. Normal pregnancy and preeclampsia both produce
inflammatory changes in peripheral blood leukocytes akin to those of sepsis. Am J Obstet
Gynecol 1998;179:80–6. [PubMed: 9704769]
172. Redman CW, Sacks GP, Sargent IL. Preeclampsia: an excessive maternal inflammatory response
to pregnancy. Am J Obstet Gynecol 1999;180:499–506. [PubMed: 9988826]
173. Gervasi MT, Chaiworapongsa T, Pacora P, et al. Phenotypic and metabolic characteristics of
monocytes and granulocytes in preeclampsia. Am J Obstet Gynecol 2001;185:792–7. [PubMed:
11641653]

Am J Obstet Gynecol. Author manuscript; available in PMC 2023 February 01.


JUNG et al. Page 25

174. Vassilakos P, Riotton G, Kajii T. Hydatidiform mole: two entities. A morphologic and cytogenetic
study with some clinical consideration. Am J Obstet Gynecol 1977;127:167–70. [PubMed:
Author Manuscript

188340]
175. Kohorn EI. Molar pregnancy: presentation and diagnosis. Clin Obstet Gynecol 1984;27:181–91.
[PubMed: 6705310]
176. Soto-Wright V, Bernstein M, Goldstein DP, Berkowitz RS. The changing clinical presentation of
complete molar pregnancy. Obstet Gynecol 1995;86:775–9. [PubMed: 7566847]
177. Jauniaux E Partial moles: from postnatal to prenatal diagnosis. Placenta 1999;20:379–88.
[PubMed: 10419802]
178. Zilberman Sharon N, Maymon R, Melcer Y, Jauniaux E. Obstetric outcomes of twin pregnancies
presenting with a complete hydatidiform mole and coexistent normal fetus: a systematic review
and meta-analysis. BJOG 2020;127:1450–57. [PubMed: 32339446]
179. Koga K, Osuga Y, Tajima T, et al. Elevated serum soluble fms-like tyrosine kinase 1 (sFlt1) level
in women with hydatidiform mole. Fertil Steril 2010;94:305–8. [PubMed: 19269632]
180. Kanter D, Lindheimer MD, Wang E, et al. Angiogenic dysfunction in molar pregnancy. Am J
Obstet Gynecol 2010;202:184.e1–5. [PubMed: 19922899]
Author Manuscript

181. Iriyama T, Wang G, Yoshikawa M, et al. Increased LIGHT leading to sFlt-1 elevation underlies
the pathogenic link between hydatidiform mole and preeclampsia. Sci Rep 2019;9:10107.
[PubMed: 31300808]
182. Moffett A, Loke C. Immunology of placentation in eutherian mammals. Nat Rev Immunol
2006;6:584–94. [PubMed: 16868549]
183. Leslie M. Immunology. Fetal immune system hushes attacks on maternal cells. Science
2008;322:1450–1.
184. Burlingham WJ. A lesson in tolerance--maternal instruction to fetal cells. N Engl J Med
2009;360:1355–7. [PubMed: 19321873]
185. Rowe JH, Ertelt JM, Xin L, Way SS. Pregnancy imprints regulatory memory that sustains anergy
to fetal antigen. Nature 2012;490:102–6. [PubMed: 23023128]
186. Betz AG. Immunology: Tolerating pregnancy. Nature 2012;490:47–8. [PubMed: 23038465]
187. Williams Z Inducing tolerance to pregnancy. N Engl J Med 2012;367:1159–61. [PubMed:
22992082]
Author Manuscript

188. Colvin RB, Smith RN. Antibody-mediated organ-allograft rejection. Nat Rev Immunol
2005;5:807–17. [PubMed: 16175181]
189. Alegre ML, Florquin S, Goldman M. Cellular mechanisms underlying acute graft rejection: time
for reassessment. Curr Opin Immunol 2007;19:563–8. [PubMed: 17720467]
190. Kim IK, Bedi DS, Denecke C, Ge X, Tullius SG. Impact of innate and adaptive immunity on
rejection and tolerance. Transplantation 2008;86:889–94. [PubMed: 18852649]
191. Robillard PY, Dekker G, Chaouat G, Hulsey TC, Saftlas A. Epidemiological studies on
primipaternity and immunology in preeclampsia--a statement after twelve years of workshops. J
Reprod Immunol 2011;89:104–17. [PubMed: 21543120]
192. Robillard PY, Dekker G, Chaouat G, Elliot MG, Scioscia M. High incidence of early onset
preeclampsia is probably the rule and not the exception worldwide. 20th anniversary of the
reunion workshop. A summary. J Reprod Immunol 2019;133:30–36. [PubMed: 31176084]
193. Gamliel M, Goldman-Wohl D, Isaacson B, et al. Trained Memory of Human Uterine NK Cells
Enhances Their Function in Subsequent Pregnancies. Immunity 2018;48:951–62.e5. [PubMed:
Author Manuscript

29768178]
194. Trupin LS, Simon LP, Eskenazi B. Change in paternity: a risk factor for preeclampsia in
multiparas. Epidemiology 1996;7:240–4. [PubMed: 8728435]
195. Li DK, Wi S. Changing paternity and the risk of preeclampsia/eclampsia in the subsequent
pregnancy. Am J Epidemiol 2000;151:57–62. [PubMed: 10625174]
196. Perni SC, Predanik M, Cho JE, Baergen RN. Placental pathology and pregnancy outcomes in
donor and non-donor oocyte in vitro fertilization pregnancies. J Perinat Med 2005;33:27–32.
[PubMed: 15841610]

Am J Obstet Gynecol. Author manuscript; available in PMC 2023 February 01.


JUNG et al. Page 26

197. Koch CA, Platt JL. Natural mechanisms for evading graft rejection: the fetus as an allograft.
Springer Semin Immunopathol 2003;25:95–117. [PubMed: 12955462]
Author Manuscript

198. Chaouat G, Petitbarat M, Dubanchet S, Rahmati M, Ledée N. Tolerance to the foetal allograft?
Am J Reprod Immunol 2010;63:624–36. [PubMed: 20367624]
199. Erlebacher A Mechanisms of T cell tolerance towards the allogeneic fetus. Nat Rev Immunol
2013;13:23–33. [PubMed: 23237963]
200. Erlebacher A Why isn't the fetus rejected? Curr Opin Immunol 2001;13:590–3. [PubMed:
11544009]
201. Trowsdale J, Betz AG. Mother's little helpers: mechanisms of maternal-fetal tolerance. Nat
Immunol 2006;7:241–6. [PubMed: 16482172]
202. Knox WF, Fox H. Villitis of unknown aetiology: its incidence and significance in placentae from
a British population. Placenta 1984;5:395–402. [PubMed: 6522353]
203. Labarrere C, Althabe O. Chronic villitis of unknown etiology and maternal arterial lesions in
preeclamptic pregnancies. European Journal of Obstetrics and Gynecology and Reproductive
Biology 1985;20:1–11. [PubMed: 4029472]
204. Kim MJ, Romero R, Kim CJ, et al. Villitis of unknown etiology is associated with a
Author Manuscript

distinct pattern of chemokine up-regulation in the feto-maternal and placental compartments:


implications for conjoint maternal allograft rejection and maternal anti-fetal graft-versus-host
disease. J Immunol 2009;182:3919–27. [PubMed: 19265171]
205. Benzon S, Zekić Tomaš S, Benzon Z, Vulić M, Kuzmić Prusac I. Involvement of T
lymphocytes in the placentae with villitis of unknown etiology from pregnancies complicated
with preeclampsia. J Matern Fetal Neonatal Med 2016;29:1055–60. [PubMed: 25812675]
206. Whitten AE, Romero R, Korzeniewski SJ, et al. Evidence of an imbalance of angiogenic/
antiangiogenic factors in massive perivillous fibrin deposition (maternal floor infarction): a
placental lesion associated with recurrent miscarriage and fetal death. Am J Obstet Gynecol
2013;208:310 e1–10 e11. [PubMed: 23333548]
207. Romero R, Whitten A, Korzeniewski SJ, et al. Maternal floor infarction/massive perivillous fibrin
deposition: a manifestation of maternal antifetal rejection? Am J Reprod Immunol 2013;70:285–
98. [PubMed: 23905710]
208. Lee J, Romero R, Dong Z, et al. Unexplained fetal death has a biological signature of maternal
Author Manuscript

anti-fetal rejection: chronic chorioamnionitis and alloimmune anti-human leucocyte antigen


antibodies. Histopathology 2011;59:928–38. [PubMed: 22092404]
209. Kim CJ, Romero R, Chaemsaithong P, Kim JS. Chronic inflammation of the placenta: definition,
classification, pathogenesis, and clinical significance. Am J Obstet Gynecol 2015;213:S53–69.
[PubMed: 26428503]
210. Maymon E, Romero R, Bhatti G, et al. Chronic inflammatory lesions of the placenta are
associated with an up-regulation of amniotic fluid CXCR3: A marker of allograft rejection. J
Perinat Med 2017.
211. Stanek J Histological Features of Shallow Placental Implantation Unify Early-Onset and Late-
Onset Preeclampsia. Pediatr Dev Pathol 2019;22:112–22. [PubMed: 30301442]
212. Devisme L, Chauvière C, Franquet-Ansart H, et al. Perinatal outcome of placental massive
perivillous fibrin deposition: a case-control study. Prenat Diagn 2017;37:323–28. [PubMed:
28152557]
213. Hiby SE, Walker JJ, O'Shaughnessy KM, et al. Combinations of maternal KIR and fetal HLA-C
genes influence the risk of preeclampsia and reproductive success. J Exp Med 2004;200:957–65.
Author Manuscript

[PubMed: 15477349]
214. Hiby SE, Apps R, Chazara O, et al. Maternal KIR in combination with paternal HLA-C2 regulate
human birth weight. J Immunol 2014;192:5069–73. [PubMed: 24778445]
215. Nakimuli A, Chazara O, Hiby SE, et al. A KIR B centromeric region present in Africans
but not Europeans protects pregnant women from pre-eclampsia. Proc Natl Acad Sci U S A
2015;112:845–50. [PubMed: 25561558]
216. Colucci F The role of KIR and HLA interactions in pregnancy complications. Immunogenetics
2017;69:557–65. [PubMed: 28695287]

Am J Obstet Gynecol. Author manuscript; available in PMC 2023 February 01.


JUNG et al. Page 27

217. Faas MM, de Vos P. Uterine NK cells and macrophages in pregnancy. Placenta 2017;56:44–52.
[PubMed: 28284455]
Author Manuscript

218. Papúchová H, Meissner TB, Li Q, Strominger JL, Tilburgs T. The Dual Role of HLA-C in
Tolerance and Immunity at the Maternal-Fetal Interface. Front Immunol 2019;10.
219. Parham P, Moffett A. Variable NK cell receptors and their MHC class I ligands in immunity,
reproduction and human evolution. Nat Rev Immunol 2013;13:133–44. [PubMed: 23334245]
220. 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. [PubMed: 19368562]
221. Hanna J, Goldman-Wohl D, Hamani Y, et al. Decidual NK cells regulate key developmental
processes at the human fetal-maternal interface. Nat Med 2006;12:1065–74. [PubMed:
16892062]
222. Karimi K, Blois SM, Arck PC. The upside of natural killers. Nat Med 2008;14:1184–5. [PubMed:
18989299]
223. Johnsen GM, Storvold GL, Drabbels JJM, et al. The combination of maternal KIR-B and fetal
Author Manuscript

HLA-C2 is associated with decidua basalis acute atherosis in pregnancies with preeclampsia. J
Reprod Immunol 2018;129:23–29. [PubMed: 30055414]
224. Robillard P-Y, Dekker G, Scioscia M, Saito S. Progress in the understanding of the
pathophysiology of immunological maladaptation related to early onset preeclampsia and
metabolic syndrome related to late onset preeclampsia. Am J Obstet Gynecol 2021;Accepted
for publication.
225. John AH, Duncan AS. The maternal syndrome associated with hydrops fetalis. J Obstet Gynaecol
Br Commonw 1964;71:61–5. [PubMed: 14117239]
226. Gedikbasi A, Oztarhan K, Gunenc Z, et al. Preeclampsia due to fetal non-immune hydrops: mirror
syndrome and review of literature. Hypertens Pregnancy 2011;30:322–30. [PubMed: 21174577]
227. Broekhuizen FF, Elejalde R, Hamilton PR. Early-onset preeclampsia, triploidy and fetal hydrops.
J Reprod Med 1983;28:223–6. [PubMed: 6854556]
228. Dotters-Katz SK, Hardisty E, Campbell E, Vora N. Trisomy 13-confined placental mosaicism:
is there an increased risk of gestational hypertensive disorders? Prenat Diagn 2017;37:938–39.
Author Manuscript

[PubMed: 28671725]
229. Giorgione V, Bhide A, Bhate R, Reed K, Khalil A. Are Twin Pregnancies Complicated by Weight
Discordance or Fetal Growth Restriction at Higher Risk of Preeclampsia? Journal of clinical
medicine 2020;9.
230. O'Driscoll DT. A fluid retention syndrome associated with severe iso-immunization to the rhesus
factor. J Obstet Gynaecol Br Emp 1956;63:372–4. [PubMed: 13332452]
231. Kaiser IH. Ballantyne and triple edema. Am J Obstet Gynecol 1971;110:115–20. [PubMed:
4995588]
232. Rana S, Venkatesha S, DePaepe M, Chien EK, Paglia M, Karumanchi SA. Cytomegalovirus-
induced mirror syndrome associated with elevated levels of circulating antiangiogenic factors.
Obstet Gynecol 2007;109:549–52. [PubMed: 17267891]
233. Brochot C, Collinet P, Provost N, Subtil D. Mirror syndrome due to parvovirus B19 hydrops
complicated by severe maternal pulmonary effusion. Prenat Diagn 2006;26:179–80. [PubMed:
16463296]
234. Braun T, Brauer M, Fuchs I, et al. Mirror syndrome: a systematic review of fetal associated
Author Manuscript

conditions, maternal presentation and perinatal outcome. Fetal Diagn Ther 2010;27:191–203.
[PubMed: 20357423]
235. Ordorica SA, Marks F, Frieden FJ, Hoskins IA, Young BK. Aneurysm of the vein of Galen: a new
cause for Ballantyne syndrome. Am J Obstet Gynecol 1990;162:1166–7. [PubMed: 2187346]
236. Sherer DM, Sadovksy E, Menashe M, Mordel N, Rein AJ. Fetal ventricular tachycardia
associated with nonimmunologic hydrops fetalis. A case report. J Reprod Med 1990;35:292–4.
[PubMed: 2325045]
237. Midgley DY, Harding K. The Mirror Syndrome. European Journal of Obstetrics & Gynecology
and Reproductive Biology 2000;88:201–02. [PubMed: 10690681]

Am J Obstet Gynecol. Author manuscript; available in PMC 2023 February 01.


JUNG et al. Page 28

238. Dorman SL, Cardwell MS. Ballantyne syndrome caused by a large placental chorioangioma. Am
J Obstet Gynecol 1995;173:1632–3. [PubMed: 7503218]
Author Manuscript

239. Allarakia S, Khayat HA, Karami MM, et al. Characteristics and management of mirror syndrome:
a systematic review (1956-2016). J Perinat Med 2017;45:1013–21. [PubMed: 28315852]
240. Duthie SJ, Walkinshaw SA. Parvovirus associated fetal hydrops: reversal of pregnancy induced
proteinuric hypertension by in utero fetal transfusion. Br J Obstet Gynaecol 1995;102:1011–3.
[PubMed: 8652468]
241. Espinoza J, Romero R, Nien JK, et al. A role of the anti-angiogenic factor sVEGFR-1 in
the 'mirror syndrome' (Ballantyne's syndrome). J Matern Fetal Neonatal Med 2006;19:607–13.
[PubMed: 17118734]
242. Stepan H, Faber R. Elevated sFlt1 level and preeclampsia with parvovirus-induced hydrops. N
Engl J Med 2006;354:1857–58. [PubMed: 16641410]
243. Goa S, Mimura K, Kakigano A, et al. Normalisation of angiogenic imbalance after intrauterine
transfusion for mirror syndrome caused by parvovirus B19. Fetal Diagn Ther 2013;34:176–9.
[PubMed: 23711762]
244. Kakigano A, Mimura K, Kanagawa T, et al. Imbalance of angiogenic factors and avascular
Author Manuscript

edematous cystic villi in a trisomy 13 pregnancy: a case report. Placenta 2013;34:628–30.


[PubMed: 23611482]
245. Tuohy JF, James DK. Pre-eclampsia and trisomy 13. Br J Obstet Gynaecol 1992;99:891–4.
[PubMed: 1450137]
246. Bdolah Y, Palomaki GE, Yaron Y, et al. Circulating angiogenic proteins in trisomy 13. Am J
Obstet Gynecol 2006;194:239–45. [PubMed: 16389038]
247. Silasi M, Rana S, Powe C, et al. Placental expression of angiogenic factors in Trisomy 13. Am J
Obstet Gynecol 2011;204:546.e1–4.
248. Heyborne KD, Chism DM. Reversal of Ballantyne syndrome by selective second-trimester fetal
termination. A case report. J Reprod Med 2000;45:360–2. [PubMed: 10804498]
249. Audibert F, Salomon LJ, Castaigne-Meary V, Alves K, Frydman R. Selective termination of
a twin pregnancy as a treatment of severe pre-eclampsia. BJOG 2003;110:68–9. [PubMed:
12504939]
250. Heyborne KD, Porreco RP. Selective fetocide reverses preeclampsia in discordant twins. Am J
Author Manuscript

Obstet Gynecol 2004;191:477–80. [PubMed: 15343224]


251. Okby R, Mazor M, Erez O, Beer-Weizel R, Hershkovitz R. Reversal of mirror syndrome after
selective feticide of a hydropic fetus in a dichorionic diamniotic twin pregnancy. J Ultrasound
Med 2015;34:351–3. [PubMed: 25614410]
252. Sarhanis P, Pugh DH. Resolution of pre-eclampsia following intrauterine death of one twin. Br J
Obstet Gynaecol 1992;99:159–60. [PubMed: 1554671]
253. Hladunewich MA, Steinberg G, Karumanchi SA, et al. Angiogenic factor abnormalities and fetal
demise in a twin pregnancy. Nature reviews Nephrology 2009;5:658–62. [PubMed: 19855426]
254. Fox CE, Lash GE, Pretlove SJ, Chan BC, Holder R, Kilby MD. Maternal plasma and amniotic
fluid angiogenic factors and their receptors in monochorionic twin pregnancies complicated
by twin-to-twin transfusion syndrome. Ultrasound Obstet Gynecol 2010;35:695–701. [PubMed:
20069664]
255. Bundhun PK, Soogund MZ, Huang F. Impact of systemic lupus erythematosus on maternal
and fetal outcomes following pregnancy: A meta-analysis of studies published between years
2001-2016. J Autoimmun 2017;79:17–27. [PubMed: 28256367]
Author Manuscript

256. Saccone G, Berghella V, Maruotti GM, et al. Antiphospholipid antibody profile based obstetric
outcomes of primary antiphospholipid syndrome: the PREGNANTS study. Am J Obstet Gynecol
2017;216:525.e1–25.e12. [PubMed: 28153662]
257. Liu L, Sun D. Pregnancy outcomes in patients with primary antiphospholipid syndrome:
A systematic review and meta-analysis. Medicine (Baltimore) 2019;98:e15733. [PubMed:
31096533]
258. Dong Y, Yuan F, Dai Z, Wang Z, Zhu Y, Wang B. Preeclampsia in systemic lupus erythematosus
pregnancy: a systematic review and meta-analysis. Clin Rheumatol 2020;39:319–25. [PubMed:
31754886]

Am J Obstet Gynecol. Author manuscript; available in PMC 2023 February 01.


JUNG et al. Page 29

259. Lee RM, Brown MA, Branch DW, Ward K, Silver RM. Anticardiolipin and anti-beta2-
glycoprotein-I antibodies in preeclampsia. Obstet Gynecol 2003;102:294–300. [PubMed:
Author Manuscript

12907102]
260. Wallukat G, Homuth V, Fischer T, et al. Patients with preeclampsia develop agonistic
autoantibodies against the angiotensin AT1 receptor. J Clin Invest 1999;103:945–52. [PubMed:
10194466]
261. Yang X, Wang F, Chang H, et al. Autoantibody against AT1 receptor from preeclamptic patients
induces vasoconstriction through angiotensin receptor activation. J Hypertens 2008;26:1629–35.
[PubMed: 18622242]
262. Siddiqui AH, Irani RA, Blackwell SC, Ramin SM, Kellems RE, Xia Y. Angiotensin receptor
agonistic autoantibody is highly prevalent in preeclampsia: correlation with disease severity.
Hypertension 2010;55:386–93. [PubMed: 19996068]
263. Zhou CC, Zhang Y, Irani RA, et al. Angiotensin receptor agonistic autoantibodies induce pre-
eclampsia in pregnant mice. Nat Med 2008;14:855–62. [PubMed: 18660815]
264. Parrish MR, Murphy SR, Rutland S, et al. The effect of immune factors, tumor necrosis factor-
alpha, and agonistic autoantibodies to the angiotensin II type I receptor on soluble fms-like
Author Manuscript

tyrosine-1 and soluble endoglin production in response to hypertension during pregnancy. Am J


Hypertens 2010;23:911–6. [PubMed: 20431529]
265. Aggarwal S, Sunderland N, Thornton C, Xu B, Hennessy A, Makris A. A longitudinal analysis
of angiotensin II type 1 receptor antibody and angiogenic markers in pregnancy. Am J Obstet
Gynecol 2017;216:170.e1–70.e8. [PubMed: 27793555]
266. 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;52:1168–72. [PubMed: 18852381]
267. Cunningham MW Jr., Castillo J, Ibrahim T, et al. AT1-AA (Angiotensin II Type 1 Receptor
Agonistic Autoantibody) Blockade Prevents Preeclamptic Symptoms in Placental Ischemic Rats.
Hypertension 2018;71:886–93. [PubMed: 29555668]
268. Li J, LaMarca B, Reckelhoff JF. A model of preeclampsia in rats: the reduced uterine perfusion
pressure (RUPP) model. Am J Physiol Heart Circ Physiol 2012;303:H1–8. [PubMed: 22523250]
269. Lamarca B, Speed J, Ray LF, et al. Hypertension in response to IL-6 during pregnancy: role
of AT1-receptor activation. International journal of interferon, cytokine and mediator research
Author Manuscript

2011;2011:65–70.
270. Dhillion P, Wallace K, Herse F, et al. IL-17-mediated oxidative stress is an important stimulator
of AT1-AA and hypertension during pregnancy. Am J Physiol Regul Integr Comp Physiol
2012;303:R353–8. [PubMed: 22718806]
271. Hayflick L, Moorhead PS. The serial cultivation of human diploid cell strains. Exp Cell Res
1961;25:585–621. [PubMed: 13905658]
272. Martin BJ, Spicer SS. Ultrastructural features of cellular maturation and aging in human
trophoblast. J Ultrastruct Res 1973;43:133–49. [PubMed: 4349958]
273. Burstein R, Frankel S, Soule SD, Blumenthal HT. Aging of the placenta: autoimmune theory of
senescence. Am J Obstet Gynecol 1973;116:271–6.
274. Biron-Shental T, Sukenik-Halevy R, Sharon Y, et al. Short telomeres may play a role in
placental dysfunction in preeclampsia and intrauterine growth restriction. Am J Obstet Gynecol
2010;202:381.e1–7. [PubMed: 20350645]
275. Mayne BT, Leemaqz SY, Smith AK, Breen J, Roberts CT, Bianco-Miotto T. Accelerated placental
Author Manuscript

aging in early onset preeclampsia pregnancies identified by DNA methylation. Epigenomics


2017;9:279–89. [PubMed: 27894195]
276. Sultana Z, Maiti K, Dedman L, Smith R. Is there a role for placental senescence in the genesis of
obstetric complications and fetal growth restriction? Am J Obstet Gynecol 2018;218:S762–s73.
[PubMed: 29275823]
277. Maiti K, Sultana Z, Aitken RJ, et al. Evidence that fetal death is associated with placental aging.
Am J Obstet Gynecol 2017;217:441 e1–41 e14. [PubMed: 28645573]

Am J Obstet Gynecol. Author manuscript; available in PMC 2023 February 01.


JUNG et al. Page 30

278. Gomez-Lopez N, Romero R, Plazyo O, et al. Preterm labor in the absence of acute histologic
chorioamnionitis is characterized by cellular senescence of the chorioamniotic membranes. Am J
Author Manuscript

Obstet Gynecol 2017;217:592.e1–92.e17. [PubMed: 28847437]


279. Redman CWG, Staff AC, Roberts JM. Syncytiotrophoblast stress in preeclampsia: the
convergence point for multiple pathways. Am J Obstet Gynecol 2021.
280. Cindrova-Davies T, Fogarty NME, Jones CJP, Kingdom J, Burton GJ. Evidence of oxidative
stress-induced senescence in mature, post-mature and pathological human placentas. Placenta
2018;68:15–22. [PubMed: 30055665]
281. Higuchi S, Miyamoto T, Kobara H, et al. Trophoblast type-specific expression of senescence
markers in the human placenta. Placenta 2019;85:56–62. [PubMed: 31327484]
282. Londero AP, Orsaria M, Marzinotto S, et al. Placental aging and oxidation damage in a tissue
micro-array model: an immunohistochemistry study. Histochem Cell Biol 2016;146:191–204.
[PubMed: 27106773]
283. Cox LS, Redman C. The role of cellular senescence in ageing of the placenta. Placenta
2017;52:139–45. [PubMed: 28131318]
284. Mayhew TM, Barker BL. Villous trophoblast: morphometric perspectives on growth,
Author Manuscript

differentiation, turnover and deposition of fibrin-type fibrinoid during gestation. Placenta


2001;22:628–38. [PubMed: 11504531]
285. Redman CW, Sargent IL, Staff AC. IFPA Senior Award Lecture: making sense of pre-eclampsia -
two placental causes of preeclampsia? Placenta 2014;35 Suppl:S20–5. [PubMed: 24477207]
286. Serov AS, Salafia CM, Brownbill P, Grebenkov DS, Filoche M. Optimal villi density for maximal
oxygen uptake in the human placenta. J Theor Biol 2015;364:383–96. [PubMed: 25261730]
287. Muralimanoharan S, Maloyan A, Mele J, Guo C, Myatt LG, Myatt L. MIR-210 modulates
mitochondrial respiration in placenta with preeclampsia. Placenta 2012;33:816–23. [PubMed:
22840297]
288. Smith R, Maiti K, Aitken RJ. Unexplained antepartum stillbirth: a consequence of placental
aging? Placenta 2013;34:310–3. [PubMed: 23452441]
289. Tekola-Ayele F, Zeng X, Ouidir M, et al. DNA methylation loci in placenta associated with
birthweight and expression of genes relevant for early development and adult diseases. Clin
Epigenetics 2020;12:78. [PubMed: 32493484]
Author Manuscript

290. Okoror CEM, Enabudoso EJ, Okoror OT, Okonkwo CA. Serum calcium-magnesium ratio in
women with pre-eclampsia at a tertiary hospital in Nigeria. Int J Gynaecol Obstet 2020;149:354–
58. [PubMed: 32167585]
291. Hultin H, Hellman P, Lundgren E, et al. Association of parathyroid adenoma and pregnancy with
preeclampsia. J Clin Endocrinol Metab 2009;94:3394–9. [PubMed: 19531594]
292. Caimari F, Valassi E, Garbayo P, et al. Cushing's syndrome and pregnancy outcomes: a systematic
review of published cases. Endocrine 2017;55:555–63. [PubMed: 27704478]
293. Baghlaf HA, Badeghiesh AM, Suarthana E, Dahan MH. The effect of Cushing's syndrome on
pregnancy complication rates: analysis of more than 9 million deliveries. J Matern Fetal Neonatal
Med 2021:1–7.
294. Landau E, Amar L. Primary aldosteronism and pregnancy. Ann Endocrinol (Paris) 2016;77:148–
60. [PubMed: 27156905]
295. Quartermaine G, Lambert K, Rees K, et al. Hormone-secreting adrenal tumours cause severe
hypertension and high rates of poor pregnancy outcome; a UK Obstetric Surveillance System
study with case control comparisons. BJOG 2018;125:719–27. [PubMed: 28872770]
Author Manuscript

296. Oliva R, Angelos P, Kaplan E, Bakris G. Pheochromocytoma in pregnancy: a case series and
review. Hypertension 2010;55:600–6. [PubMed: 20083723]
297. Dusitkasem S, Herndon BH, Paluzzi D, Kuhn J, Small RH, Coffman JC. From Bad to Worse:
Paraganglioma Diagnosis during Induction of Labor for Coexisting Preeclampsia. Case reports in
anesthesiology 2017;2017:5495808. [PubMed: 28197344]
298. Kovacs CS. Maternal Mineral and Bone Metabolism During Pregnancy, Lactation, and Post-
Weaning Recovery. Physiol Rev 2016;96:449–547. [PubMed: 26887676]

Am J Obstet Gynecol. Author manuscript; available in PMC 2023 February 01.


JUNG et al. Page 31

299. Bodnar LM, Catov JM, Simhan HN, Holick MF, Powers RW, Roberts JM. Maternal vitamin
D deficiency increases the risk of preeclampsia. J Clin Endocrinol Metab 2007;92:3517–22.
Author Manuscript

[PubMed: 17535985]
300. Scholl TO, Chen X, Stein TP. Vitamin D, secondary hyperparathyroidism, and preeclampsia. Am
J Clin Nutr 2013;98:787–93. [PubMed: 23885046]
301. Wheeler BJ, Taylor BJ, de Lange M, et al. A Longitudinal Study of 25-Hydroxy Vitamin D
and Parathyroid Hormone Status throughout Pregnancy and Exclusive Lactation in New Zealand
Mothers and Their Infants at 45° S. Nutrients 2018;10.
302. Hyppönen E, Hartikainen AL, Sovio U, Järvelin MR, Pouta A. Does vitamin D supplementation
in infancy reduce the risk of pre-eclampsia? Eur J Clin Nutr 2007;61:1136–9. [PubMed:
17268418]
303. Marya RK, Rathee S, Manrow M. Effect of calcium and vitamin D supplementation on toxaemia
of pregnancy. Gynecol Obstet Invest 1987;24:38–42. [PubMed: 3623260]
304. Palacios C, Kostiuk LK, Peña-Rosas JP. Vitamin D supplementation for women during pregnancy.
The Cochrane database of systematic reviews 2019;7:Cd008873. [PubMed: 31348529]
305. Nilsson IL, Aberg J, Rastad J, Lind L. Left ventricular systolic and diastolic function and exercise
Author Manuscript

testing in primary hyperparathyroidism-effects of parathyroidectomy. Surgery 2000;128:895–


902. [PubMed: 11114621]
306. Hagström E, Lundgren E, Rastad J, Hellman P. Metabolic abnormalities in patients with
normocalcemic hyperparathyroidism detected at a population-based screening. Eur J Endocrinol
2006;155:33–9. [PubMed: 16793947]
307. Trementino L, Arnaldi G, Appolloni G, et al. Coagulopathy in Cushing's syndrome.
Neuroendocrinology 2010;92 Suppl 1:55–9. [PubMed: 20829619]
308. Isidori AM, Minnetti M, Sbardella E, Graziadio C, Grossman AB. Mechanisms in endocrinology:
The spectrum of haemostatic abnormalities in glucocorticoid excess and defect. Eur J Endocrinol
2015;173:R101–13. [PubMed: 25987566]
309. Pivonello R, De Martino MC, Iacuaniello D, et al. Metabolic Alterations and Cardiovascular
Outcomes of Cortisol Excess. Front Horm Res 2016;46:54–65. [PubMed: 27212264]
310. Monticone S, Burrello J, Tizzani D, et al. Prevalence and Clinical Manifestations of Primary
Aldosteronism Encountered in Primary Care Practice. J Am Coll Cardiol 2017;69:1811–20.
Author Manuscript

[PubMed: 28385310]
311. Käyser SC, Deinum J, de Grauw WJ, et al. Prevalence of primary aldosteronism in primary care:
a cross-sectional study. Br J Gen Pract 2018;68:e114–e22. [PubMed: 29335324]
312. Hollenberg NK. Aldosterone in the development and progression of renal injury. Kidney Int
2004;66:1–9. [PubMed: 15200407]
313. Liang W, Chen C, Shi J, et al. Disparate effects of eplerenone, amlodipine and telmisartan on
podocyte injury in aldosterone-infused rats. Nephrol Dial Transplant 2011;26:789–99. [PubMed:
20729265]
314. Lenders JWM, Langton K, Langenhuijsen JF, Eisenhofer G. Pheochromocytoma and Pregnancy.
Endocrinol Metab Clin North Am 2019;48:605–17. [PubMed: 31345526]
315. Mohamed Ismail NA, Abd Rahman R, Abd Wahab N, Muhammad R, Nor Azmi K.
Pheochromocytoma and pregnancy: a difficult and dangerous ordeal. Malays J Med Sci
2012;19:65–8. [PubMed: 22977377]
316. Langton K, Tufton N, Akker S, et al. Pregnancy and phaeochromocytoma/paraganglioma:
clinical clues affecting diagnosis and outcome - a systematic review. BJOG 2021;128:1264–72.
Author Manuscript

[PubMed: 33342020]
317. Kamari Y, Sharabi Y, Leiba A, Peleg E, Apter S, Grossman E. Peripartum hypertension from
pheochromocytoma: a rare and challenging entity. Am J Hypertens 2005;18:1306–12. [PubMed:
16202853]

Am J Obstet Gynecol. Author manuscript; available in PMC 2023 February 01.


JUNG et al. Page 32
Author Manuscript
Author Manuscript
Author Manuscript

Figure 1.
Multiple etiologies implicated in preeclampsia. Uteroplacental ischemia, maternal infection
and inflammation (eg periodontal disease, urinary tract infection, COVID-19), maternal
intestinal dysbiosis, maternal obesity, sleep disorders, hydatidiform mole, fetal diseases (eg
hydrops fetalis, viral infection, Trisomy 13, and unique complications of multiple gestation),
autoimmune disorders, placental aging, breakdown of maternal-fetal immune tolerance, and
endocrine disorders (eg hyperparathyroidism, Cushing’s syndrome, hyperaldosteronism).
Author Manuscript

Am J Obstet Gynecol. Author manuscript; available in PMC 2023 February 01.


JUNG et al. Page 33
Author Manuscript
Author Manuscript

Figure 2.
A diagrammatic depiction of experiments demonstrating that uterine ischemia in pregnant
animals, but not in non-pregnant animals, can cause hypertension. A. In the Goldblatt
model of renovascular hypertension, clamping the renal artery leads to development of
hypertension through renal ischemia and the release of renin in non-pregnant animals.
Author Manuscript

B. By contrast, clamping the aorta below the renal arteries does not induce hypertension
in non-pregnant animals. C. Clamping of the aorta in pregnant animals below the renal
arteries leads to hypertension. D. The hypertension disappears after a hysterectomy has
been performed; this suggests that the source of the signals leading to maternal systemic
hypertension are derived from the gravid uterus. Modified from Chaiworapongsa T. et al.
Pre-eclampsia part 1: current understanding of its pathophysiology. Nat Rev Nephrol. 2014
Aug; 10 (8):466-80.
Author Manuscript

Am J Obstet Gynecol. Author manuscript; available in PMC 2023 February 01.


JUNG et al. Page 34
Author Manuscript
Author Manuscript

Figure 3.
Experimental demonstration that placental ischemia causes maternal hypertension and that a
soluble factor in the blood of an animal with placental ischemia can induce hypertension in a
non-pregnant animal. A. The Z suture is placed in the uterus to generate placental ischemia.
B. Placental ischemia causes hypertension. Rabbit A had undergone a bilateral nephrectomy;
Author Manuscript

therefore, the kidney is not a cause of the hypertension. After Z sutures were placed through
several placentas, hypertension developed. C. Gross evidence that the suture has caused
a placental infarction. The pale portion of the placenta with the arrow represents a large
infarction. A control placenta of the same animal is illustrated on the right. D. Transfusions
of blood from Rabbit A (a pregnant rabbit with placental ischemia) to a non-pregnant
rabbit (Rabbit B) caused hypertension; this suggests that a circulating factor generated after
placental ischemia is present in the maternal blood and that it can induce hypertension in a
non-pregnant rabbit. Modified from Berger M. and Cavanagh D. Toxemia of pregnancy: The
hypertensive effect of acute experimental placental ischemia. Am J Obstet Gynecol. 1963
Oct 1;87:293-305.
Author Manuscript

Am J Obstet Gynecol. Author manuscript; available in PMC 2023 February 01.


JUNG et al. Page 35
Author Manuscript
Author Manuscript
Author Manuscript

Figure 4.
A. Diagram of maternal blood supply to the placenta. The spiral arteries undergo
physiologic changes in normal pregnancy (grey). In preeclampsia, the myometrial segment
of the spiral artery fails to undergo physiological transformation (blue), which is thought to
Author Manuscript

explain the uteroplacental ischemia observed in preeclampsia. Moreover, non-transformed


spiral arteries are prone to atherosis (yellow), characterized by the presence of lipid-laden
macrophages within the lumen. Placental basal plate spiral arteries with hematoxylin-eosin
stain (B, C, and D). B. Transformed spiral arteries are characterized by the presence of
intramural trophoblasts (arrowheads) and fibrinoid degeneration (arrows) of the arterial wall.
C. Non-transformed spiral arteries lack intramural trophoblasts and fibrinoid degeneration,
and they retain smooth muscle. Arrowheads indicate the presence of trophoblasts in

Am J Obstet Gynecol. Author manuscript; available in PMC 2023 February 01.


JUNG et al. Page 36

myometrium, but not in the wall of the spiral artery. D. Acute atherosis in decidual spiral
Author Manuscript

artery. Many lipid-laden macrophages (arrows) are seen in the spiral artery with the lack
of invasion of the trophoblast (arrowhead) into myometrial segment of the spiral artery.
Images (B, C, and D) stained with cytokeratin 7 (brown) and periodic acid–Schiff (pink),
original magnification ×200. Immunohistochemistry of placental basal plate spiral arteries
(E, F, and G). E. Presence of endothelium (arrow, blue) in vessels with normal trophoblastic
invasion, original magnification × 640. F. spiral artery with presence of endothelium
(blue, arrowhead) and smooth muscle cells (green, arrow), original magnification ×640.
G. Atherosis lesions show the presence of numerous macrophages CD36-reactive (red
reactivity, blue arrow) and smooth muscle cells in the vessel wall (green reactivity, yellow
arrow), original magnification x400. *lumen of spiral artery. Modified from McMaster-Fay
RA. Uteroplacental vascular syndromes: theories, hypotheses and controversies. Clin Obstet
Gynecol Reprod Med 2018;4:1–5. Espinoza J et al. Normal and abnormal transformation
Author Manuscript

of the spiral arteries during pregnancy. J Perinat Med. 2006;34(6):447-58. Labarrere CA et


al. Failure of physiologic transformation of spiral arteries, endothelial and trophoblast cell
activation, and acute atherosis in the basal plate of the placenta. Am J Obstet Gynecol. 2017
Mar;216(3):287.e1-287.e16.
Author Manuscript
Author Manuscript

Am J Obstet Gynecol. Author manuscript; available in PMC 2023 February 01.


JUNG et al. Page 37
Author Manuscript
Author Manuscript
Author Manuscript

Figure 5.
Acute atherosis on oil-red O staining. Fat droplets (arrows) in the non-transformed spiral
artery are stained red. *Lumen of spiral artery. Cover, Am J Obstet Gynecol. November
2014 Yeon Mee Kim, Roberto Romero.
Author Manuscript

Am J Obstet Gynecol. Author manuscript; available in PMC 2023 February 01.


JUNG et al. Page 38
Author Manuscript
Author Manuscript

Figure 6.
Periodontal diseases and association with atherosclerotic disease. A. Bacteria found in the
periodontal space can enter the bloodstream (bacteremia) and eventually the heart, resulting
in atherosclerotic plaque in the heart blood vessels. Compared with uninfected control
(B), periodontal infection with Porphyromonas gingivalis causes atherosclerotic aortic arch
plaques (C, arrow) in apoE-null mice. Scale bar =1 mm. Oil red O staining of cryosections
at the aortic sinus shows few small fatty streaks (control, D), whereas atherosclerotic lesions
were greater in number and size (arrow) in infected animals (E). Scale bar=50 μm. Modified
Author Manuscript

from Hajishengallis G et al. Local and systemic mechanisms linking periodontal disease
and inflammatory comorbidities. Nat Rev Immunol. 2021 Jul;21(7):426-440 and Lalla
E. et al, Oral infection with a periodontal pathogen accelerates early atherosclerosis in
apolipoprotein E-null mice. Arterioscler Thromb Vasc Biol. 2003 Aug 1;23(8):1405-11.
Author Manuscript

Am J Obstet Gynecol. Author manuscript; available in PMC 2023 February 01.


JUNG et al. Page 39
Author Manuscript
Author Manuscript
Author Manuscript

Figure 7.
Association between SARS-CoV-2 infection severity and subsequent development of
preeclampsia. The most severe COVID-19, the greater the risk of preeclampsia.
Modified from Lai J et al. SARS-COV-2 and the subsequent development of preeclampsia
and preterm birth: evidence of a dose response relationship supporting causality. Am J
Author Manuscript

Obstet Gynecol. 2021 Aug 26:S0002-9378(21)00947-9.

Am J Obstet Gynecol. Author manuscript; available in PMC 2023 February 01.


JUNG et al. Page 40
Author Manuscript
Author Manuscript
Author Manuscript

Figure 8.
Placental syncytiotrophoblast stress induces excessive sFlt-1 into the maternal circulation.
sFlt-1 binds to free PlGF or VEGF (proangiogenic factors) with high affinity, thus
preventing their interaction with their cell-surface receptors (i.e. VEGR-1) on the endothelial
cells, leading to endothelial dysfunction. SARS-CoV-2 also targets the endothelium which
Author Manuscript

normally express angiotensin-converting-enzyme 2 (ACE-2), one of the cell entry receptors


for the virus, leading to endothelitis, which induces intravascular inflammation (i.e. cytokine
storm) and endothelial dysfunction.
ACE2; angiotensin-converting enzyme 2, COVID-19; Coronavirus disease 2019, SARS-
CoV-2; severe acute respiratory syndrome coronavirus 2, sFlt-1; Soluble Fms-Like Tyrosine
Kinase-1, VEGFR-1; vascular endothelial growth factor receptor-1

Am J Obstet Gynecol. Author manuscript; available in PMC 2023 February 01.


JUNG et al. Page 41
Author Manuscript
Author Manuscript

Figure 9.
(A) Gut microbiota in patients with preeclampsia (red) and those with normal pregnancy
(blue). The significantly different genera (Firmicutes, Bacteroidetes, Proteobacterias, and
Actinobacteria) at the phylum level in the two groups. The boxes represent the interquartile
range (IQR) between first and third quartiles and the line inside represents the median.
*p < 0.05. (B) Maternal intestinal dysbiosis and preeclampsia. Mice that received fecal
microbiota from patients with preeclampsia (red) had higher systolic blood pressure than
mice that received fecal microbiota from normotensive pregnant women (blue) or those
that received the phosphate-buffered saline (control, grey). Modified from Wang J et al.
Gut Microbiota Dysbiosis and Increased Plasma LPS and TMAO Levels in Patients with
Preeclampsia. Front Cell Infect Microbiol. 2019 Dec 3;9:409 and Chen X. et al. Gut
Author Manuscript

dysbiosis induces the development of pre-eclampsia through bacterial translocation. Gut.


2020 Mar;69(3):513-522.
Author Manuscript

Am J Obstet Gynecol. Author manuscript; available in PMC 2023 February 01.


JUNG et al. Page 42
Author Manuscript
Author Manuscript
Author Manuscript

Figure 10.
Probability of preeclampsia according to the pre-pregnancy body mass index in both
nulliparous (top line, red) and multiparous (bottom line, blue) women. Modified from Catov
JM et al. Risk of early or severe pre-eclampsia related to pre-existing conditions. Int J
Epidemiol. 2007 Apr;36(2):412-9.
Author Manuscript

Am J Obstet Gynecol. Author manuscript; available in PMC 2023 February 01.


JUNG et al. Page 43
Author Manuscript
Author Manuscript

Figure 11.
Author Manuscript

Interactions between maternal KIR and fetal HLA-C at the site of placentation. If the mother
has a KIR BB genotype, which binds to trophoblast HLA-C1 molecules, this activates
dNK cells, producing increased levels of cytokines such as GM-CSF that can enhance
placentation. In contrast, when the mother has a KIR AA genotype and fetal HLA-C2
alleles, this inhibits dNK cells, leading to defective placentation.
KIR; killer cell immunoglobulin like receptors, HLA; human leukocyte antigen,
dNK; decidual natural killer cells, GM-CSF; granulocyte-macrophage colony-stimulating
factor. Modified from https://www.ivi-rmainnovation.com/maternal-killer-immunoglobulin-
receptors-predictive-live-birth-rate/
Author Manuscript

Am J Obstet Gynecol. Author manuscript; available in PMC 2023 February 01.


JUNG et al. Page 44
Author Manuscript
Author Manuscript
Author Manuscript

Figure 12.
A. Optimal villi density for maximal oxygen uptake in the human placenta. Placentas with
low villi density (rarefied villi) have low oxygen uptake, as fetal villi are rare. By contrast, in
placentas with high villous density (villous overcrowding), there is no space for intervillous
Author Manuscript

space for oxygen exchange. The optimal villi density for the oxygen uptake was 0.47 ± 0.06,
calculated from histomorphometrical data for villi and intervillous volumes of placentas.
B. Cross-section of the placenta: 1) rarefied villi in preeclampsia; 2) normal placenta; and
3) villous overcrowding in diabetes mellitus. H&E stained and scale represents 50 μm.
Modified from Serov AS et al. Optimal villi density for maximal oxygen uptake in the
human placenta. J Theor Biol. 2015 Jan 7;364:383-96.

Am J Obstet Gynecol. Author manuscript; available in PMC 2023 February 01.

You might also like