Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Mar 15.
Published in final edited form as: Compr Physiol. 2020 Dec 9;11(1):1315–1349. doi: 10.1002/cphy.c200008

Preeclampsia: Linking Placental Ischemia with Maternal Endothelial and Vascular Dysfunction

Bhavisha A Bakrania 1,2, Frank T Spradley 1,2,3, Heather A Drummond 1,2, Babbette LaMarca 1,4, Michael J Ryan 1,2, Joey P Granger 1,2,*
PMCID: PMC7959189  NIHMSID: NIHMS1676706  PMID: 33295016

Abstract

Preeclampsia (PE), a hypertensive disorder, occurs in 3% to 8% of pregnancies in the United States and affects over 200,000 women and newborns per year. The United States has seen a 25% increase in the incidence of PE, largely owing to increases in risk factors, including obesity and cardiovascular disease. Although the etiology of PE is not clear, it is believed that impaired spiral artery remodeling of the placenta reduces perfusion, leading to placental ischemia. Subsequently, the ischemic placenta releases antiangiogenic and pro-inflammatory factors, such as cytokines, reactive oxygen species, and the angiotensin II type 1 receptor autoantibody (AT1-AA), among others, into the maternal circulation. These factors cause widespread endothelial activation, upregulation of the endothelin system, and vasoconstriction. In turn, these changes affect the function of multiple organ systems including the kidneys, brain, liver, and heart. Despite extensive research into the pathophysiology of PE, the only treatment option remains early delivery of the baby and importantly, the placenta. While premature delivery is effective in ameliorating immediate risk to the mother, mounting evidence suggests that PE increases risk of cardiovascular disease later in life for both mother and baby. Notably, these women are at increased risk of hypertension, heart disease, and stroke, while offspring are at risk of obesity, hypertension, and neurological disease, among other complications, later in life. This article aims to discuss the current understanding of the diagnosis and pathophysiology of PE, as well as associated organ damage, maternal and fetal outcomes, and potential therapeutic avenues.

Introduction

Hypertensive disorders of pregnancy, including preeclampsia (PE), are the leading cause of premature births and maternal/fetal morbidity and mortality (282). In addition to elevated blood pressure, PE is characterized by proteinuria and/or other system disturbances caused primarily by the release of placental factors into the maternal circulation (25, 163, 184, 188, 215, 360). The American College of Obstetrics and Gynecology (ACOG) recommends diagnosis based on the following criteria: new-onset hypertension after 20 weeks of gestation and one of the following: proteinuria, thrombocytopenia, renal insufficiency, impaired liver function, pulmonary edema, and cerebral or visual symptoms (Table 1) (282).

Table 1.

Diagnostic Criteria for Preeclampsia Based on American College of Obstetrics and Gynecology Guidelines

Hypertension ≥ 140/90 mmHg on two occasions at least 4 h apart
or
≥160/110 mmHg on two occasions within minutes
The new onset of hypertension and one of the following can be used for diagnosis:
Proteinuria ≥300 mg/24 h (or this amount extrapolated from a timed collection)
or
Protein/creatine (each mL/dL) ratio ≥ 0.3
Dipstick reading of 1 + (used only if other measures are unavailable)
Thrombocytopenia Platelet count < 100,000/μL
Renal insufficiency Serum creatine ≥1.1 mg/dL
or
Doubling of serum creatine in the absence of other renal disease
Impaired liver function Twice the normal blood concentration of liver transaminases
Pulmonary edema
Cerebral or visual symptoms

PE can present with varying features and a combination of symptoms based on time of onset, severity, and any preexisting diseases in the mother. Fetal growth restriction is a well-described feature of PE but is no longer a diagnostic criterion based on ACOG guidelines. The International Society for the Study of Hypertension (ISSHP) continues to recognize fetal growth restriction as a diagnostic feature, which likely contributes to some discrepancy in the incidence of disease (370). Women with preexisting hypertension and presenting with other de novo features of PE are diagnosed with superimposed PE. The diagnostic criteria outlined in Table 1 can also be used to differentiate between PE and gestational hypertension, which presents after 20 weeks of gestation but is not usually associated with disturbances in other organ systems. In severe cases, women with PE experience HELLP syndrome (hemolysis, elevated liver enzymes, and low platelet count), which can be considered a subtype of PE (282). HELLP syndrome is a severe but relatively rare complication of PE that necessitates delivery in 70% of cases, of which 15% are preterm (6, 56, 347, 348). Based on the ACOG guidelines, the terms “mild PE” and “severe PE” should no longer be used but rather “PE with or without severe features”, as detailed in Table 2.

Table 2.

Severe Features of Preeclampsia Based on American College of Obstetrics and Gynecology Guidelines (282)

Any of the following findings are considered severe features of PE
  • Blood pressure ≥160/110 on two occasions at least 4 h apart while patient is on bed rest (unless antihypertensive therapy is administered)

  • Thrombocytopenia (platelet count <100,000/μL)

  • Impaired liver function (twice normal blood concentration of liver transaminases), severe persistent right upper quadrant pain or epigastric pain, unaccounted for alternative diagnosis and/or unresponsive to medication

  • Progressive renal insufficiency (serum creatine ≥1.1 mg/dL or doubling of serum creatine in the absence of other renal disease)

  • Pulmonary edema

  • New-onset cerebral or visual disturbances

Timing of PE Onset and Symptoms

Timing of PE onset can play a role in the degree whereby maternal symptoms arise in this disorder. Early-onset PE typically occurs <34 weeks of gestation, which is associated with more severe symptoms, and often necessitates preterm delivery that carries a much greater risk of fetal death (195). Late-onset PE develops >34 weeks of gestation and does not usually require early delivery. Outcomes for both early- and late-onset PE can vary; however, it is widely accepted that early-onset PE carries more adverse risk for both mother and baby during gestation and postpartum (370). In particular, incidence of growth restriction, hemolysis, neurological, and cardiorespiratory complications are far more common in early-onset PE (370). Although PE typically occurs during gestation, some cases of postpartum PE have been reported and are diagnosed by onset of hypertension and organ disturbances similar to gestational PE (84, 120, 278, 282). Reports of de novo postpartum PE have occurred between 48 h and 12 weeks postpartum despite the absence of the placenta by this time. It is possible that subclinical PE was present in these patients during gestation or that vascular and organ dysfunction was mild during gestation and is exacerbated and diagnosed postpartum. Postpartum PE is typically not associated with headaches and should not be mistaken for eclampsia. Eclampsia has more severe and potentially terminal outcomes for the patient due to seizures (306).

There is currently no therapy that can prevent or cure PE. Low-dose aspirin (60–80 mg) has been reported to delay onset of PE and improve perinatal outcomes (282) but is still only used in high-risk patients. Current management regimens for PE involve use of pregnancy-safe antihypertensive pharmacotherapy. Identifying mechanistic pathways that can be targeted to prevent and/or treat PE is an active area of research that will be discussed later in this article.

Historical Perspective

Some of the first records of convulsions during child birth date back to 3000 b.c. and were not isolated to a single geographical area with reports from China, India, Europe, and Africa (54). Observations made from that time included convulsions, presenting almost exclusively in first pregnancies or death, in about one-third of the women. We now know that these convulsions were most likely eclampsia—new onset of seizures during or shortly after pregnancy. The association between eclampsia, edema, and proteinuria, linking eclampsia to some form of renal disease, was made in the 1840s; however, blood pressure measurement was not commonplace as it is today (52, 54, 285). Still, physicians recognized that delivery reduced convulsions and improved disease state in the mother (54). In the early 1900s, following the invention of the inflatable armband to measure blood pressure, physicians quickly noticed that pregnant women with eclampsia had markedly increased blood pressure. In fact, blood pressure was increased prior to delivery and preceded development of convulsions and seizures, hence the origin of the term “preeclampsia” (52, 285). In the 1970s, Brosens et al. (51) found that spiral artery remodeling is deficient in PE and proposed that perhaps hypertension is a compensatory mechanism to ensure sufficient nutrient delivery to the baby (50, 264). A significant advance in the understanding of the pathogenesis of PE was made in the late 1980s, when Roberts, Taylor, and Redman published that endothelial dysfunction plays a central role in the maternal syndrome (277, 283, 284). They noted that symptoms associated with PE are consistent with endothelial cell injury, particularly in the renal glomerulus, liver, and brain.

In the 1990s, it was established that an uncomplicated first pregnancy reduces the likelihood that a subsequent pregnancy will be complicated by PE, assuming the same male partner. If the female changes partner, she carries the same risk as a primiparous woman, suggesting this is an immunological disease (208, 287) that could be triggered by the immunological properties of semen. Subsequent studies confirmed that natural killer (NK) cells, which play a role in regulating placentation and spiral artery remodeling, could recognize complexes of fetal origin from the paternal contribution as foreign and trigger an immune response (70, 189, 343).

It is now well established that PE is a multifaceted and multiorgan disease. While there is certainly an immunological component, there is also evidence for maternal, genetic, and environmental contributors to the etiology of PE. In the early 2000s, it was established that antiangiogenic factors in the maternal circulation contribute to the clinical manifestation of PE (215), and that these antiangiogenic and pro-inflammatory factors are released in response to placental ischemia. The specific role of these factors and their impact on different organ systems will be discussed in this article. Despite profound advances in our understanding of the pathogenesis of PE, a definitive etiology and treatment strategy remains elusive.

Epidemiology

Incidence of PE

Hypertensive pregnancy disorders, including PE, affect more than 10% of pregnancies and are a major contributor of maternal and fetal morbidity and mortality. PE alone impacts 3% to 8% of pregnancies (22, 88, 151, 346). Worldwide, PE is associated with 70,000 to 80,000 maternal deaths and >500,000 infant mortalities per year (87, 146, 243, 282). There is a notable racial disparity in prevalence of this disease. In one study, hospitalizations with PE delivery (per 100) were 3.4 among African-American women compared to 1.7 among Caucasian women (331). In the United States and most developed countries, the incidence of late-onset PE accounts for 90% of cases, as compared to 70% of cases in developing countries (54, 286). However, rates of PE with severe features have increased dramatically in the past 40 years (Figure 1) (22). While changes to the diagnostic criteria may have impacted the number of patients diagnosed with PE, the overall rise in the incidence of PE is most likely due to a significant increase in the risk factors associated with the disease, such as obesity and preexisting cardiovascular and metabolic diseases (358).

Figure 1. Rates of preeclampsia over the past decades.

Figure 1

This figure illustrates the rates of preeclampsia (PE) in 1980 and 2010. These rates may be influenced by the change in definition of PE over time but are also certainly affected by the global increase in risk factors for this disease, including obesity and preexisting cardiovascular disease. Notably, rates of severe PE have significantly increased since 1980. Adapted from Ananth CV, et al., 2013 (22).

Risk factors for PE

A number of risk factors have been identified that contribute to the development of PE, including but not limited to, genetic susceptibility, primiparity, history of PE, preexisting cardiovascular-renal disease, multiple gestations, obesity, and advanced age (Table 3) (40, 276, 282). In addition, risk for early-onset PE in the United States has been linked to African-American race and chronic hypertension, while late-onset PE is associated with younger maternal age, multifetal pregnancy, and diabetes mellitus (195, 325).

Table 3.

Risk Factors for the Development of Preeclampsia Based on the American College of Obstetrics and Gynecology Guidelines (282)

Risk factor Additional references
Primiparity (86, 266)
Previous preeclamptic pregnancy (266)
Chronic hypertension, renal disease or both (266)
History of thrombophilia (41, 97)
Multifetal pregnancy (86)
In vitro fertilization (64, 210, 353)
Family history of preeclampsia (94, 266)
Type I or Type II diabetes mellitus (86)
Obesity (216, 318, 376)
System lupus erythematosus (54, 62)
Advanced maternal age (>40 years) (22, 86)

Some evidence suggests a genetic predisposition to PE (44, 148, 172) and increased susceptibility to PE (63, 94, 123, 218). This concept was first postulated in the 1960s when Chesley, Annitto, and Cosgrove noted that close relatives, including sisters, daughters, and even granddaughters, presented to the hospital with PE and eclampsia (61). To date, the most supportive data suggests a susceptibility locus near the FLT1 gene in neonates from PE pregnancies, which encodes the protein sFlt-1—a pathologic antiangiogenic factor in PE (54, 218). These findings highlight the importance of considering fetal and maternal genetics in the development of PE, particularly because the fetus dictates the genotype of the placenta. Also, there is evidence to suggest that the male partner could play a role in the development of PE. One study showed that advanced paternal age (>45 years old) further increases the likelihood of PE by 14%, based on assessment of >40 million births (168).

PE is characterized by a robust inflammatory and hypercoagulable state, thus, preexisting conditions that inappropriately activate these systems likely increase risk for this disorder. Autoimmune diseases, such as systemic lupus erythematosus and antiphospholipid syndrome (54, 62), and blood disorders such as thrombophilia (41, 97) significantly increase the risk for PE. Overwhelming data also exists to support that cardiovascular and metabolic diseases, including obesity, are major risk factors for PE (318). In an analysis of live births between 1989 and 2005, one study found that increasing body mass index (BMI, kg/m2) is associated with greater risk for both early- and late-onset PE significantly (216). Additionally, the incidence of PE is increased when obesity is combined with other risk factors such as advanced maternal age, nulliparity, and diabetes mellitus (216). In another study of prepregnancy obesity (BMI >30) in primiparous women, incidence of PE was 14.5% and superimposed PE was 2.6% compared to women with BMI <30. Furthermore, severity of PE in these women was greater when the degree of obesity is greater (322, 376). It is important to note that not all women with obesity develop PE; thus, understanding the difference between these two cohorts may be important in unraveling the pathophysiology of this disease.

In contrast to factors that increase the risk of PE, improved maternal nutrition has been associated with reduced incidence (75). Specifically, greater intake of vegetables and plant-based foods (47), dietary fiber (268), and essential micronutrients (217, 372) has been inversely related to the development of PE, particularly when these behaviors are initiated preconception and in early pregnancy. Supplementation of antioxidant nutrients and coefficients, such as vitamin D and magnesium, to restore low levels seen in PE is an active area of research (75, 173, 377). However, clinical studies have had conflicting results attributed to variation in timing of intervention, dose, severity of disease, and sample size, among other factors (75, 80, 145, 166, 222, 334).

Maternal Hemodynamic Adaptations During Normal Pregnancy and PE

The systemic hemodynamic changes that occur during the course of a normal pregnancy have been extensively reviewed (13, 68, 117, 132, 296, 330, 340). During the typical 40 weeks of gestation, extracellular fluid volume increases and systemic vascular resistance decreases, leading to greater venous return to the right atrium. In turn, this causes an increase in stroke volume and maternal cardiac output (by approximately 40%–50%), which typically reaches its peak during the middle of the third trimester (Figure 2) (37, 288, 295). During pregnancy, increased circulating levels of progesterone, prostaglandins, and relaxin are most likely responsible for decreased vascular resistance through the production of nitric oxide (NO), which is a gaseous molecule produced by healthy endothelial cells. A single layer of endothelial cells forms the endothelium and lines the lumen of all blood vessels. NO stimulates pathways in the underlying smooth muscle to promote vasorelaxation (263, 312). It is important to note that, although systemic resistance decreases during pregnancy, mean arterial pressure (MAP) is only slightly reduced (5–10 mmHg) or remains constant because of the concomitant increase in cardiac output. Furthermore, reduced MAP in normal pregnant women is associated with greater reductions in diastolic pressure rather than systolic, which reaches its lowest point by midgestation (28, 233). During the third trimester, pressure begins to rise toward prepregnancy levels (Figure 2). Ultimately, these hemodynamic changes create an environment that meets the metabolic demands of both the mother and baby.

Figure 2. Hemodynamic changes during normal pregnancy.

Figure 2

This figure illustrates the gradual increase of cardiac output (CO; circles) and a gradual decrease in mean arterial pressure (MAP; squares) over the first and second trimesters of normal pregnancy. Data are presented as mean ± SEM. Adapted from Moutquin JM, et al., 1985 (233).

Studies have also focused on changes that occur in specific organ systems over the course of a normal pregnancy. During pregnancy, uterine blood flow is markedly increased and accounts for approximately 25% of cardiac output (340). Uterine artery blood flow can be measured by Doppler ultrasound to assess whether there is adequate blood supply to the fetus. During a normal pregnancy, uterine artery resistance is low, and the Doppler pattern exhibits a high peak flow during systole that rapidly falls during diastole (Figure 3) (119, 219). As discussed below, impaired uterine hemodynamic function has important functional consequences during preeclamptic pregnancy.

Figure 3. Uterine artery resistance in humans and an animal model of superimposed preeclampsia.

Figure 3

This figure illustrates the difference between Doppler waveforms (velocity) during normal pregnancy and preeclampsia (PE) in (A) humans and (B) rats. In normal pregnancy, resistance in uterine arteries is low as evidenced by high peak during systole followed by a fall during diastole. In contrast, Doppler waveforms from PE pregnancies have a peak during systole followed by a sharp fall during diastole. The arrows in the figure indicate the characteristic notch found in PE and the rat model of superimposed PE (Dahl S Rat). Adapted from McLeod L, 2008 (219) and Gillis EE, et al., 2015 (119).

The kidneys have a central role in the chronic control of blood pressure through sodium and water homeostasis. Given that the kidneys receive 20% of the cardiac output, dilation of the renal vasculature during pregnancy is an important contributor to reduced systemic vascular resistance, which, in part, is controlled by elevated circulating levels of progesterone. Another important hormone involved in renal vasodilation, during early gestation, is relaxin, which is produced by the corpus luteum and stimulated by human chorionic gonadotropin (73, 92, 106). Decreased vascular resistance caused by these hormonal factors results in a compensatory activation of the renin angiotensin aldosterone system and subsequent increase in extracellular fluid volume associated with pregnancy (296). In addition, due to vasodilation of both the afferent and efferent arterioles during pregnancy, there is a significant increase in renal plasma flow and glomerular filtration rate, by approximately 30% to 50%, and minimal changes in glomerular capillary pressure (224). These renal adaptations during normal pregnancy are important for maintaining plasma volume, blood pressure, and overall body fluid homeostasis.

In contrast to the hemodynamic changes expected in normal healthy pregnancy, PE is associated with increased MAP and total peripheral resistance along with reduced cardiac output, renal plasma flow, and glomerular filtration. In addition, uterine blood flow is significantly reduced in PE, which can be observed using Doppler ultrasound. Specifically, peak flow during systole is reduced, followed by a sharp fall and a more pronounced notch in the descending wave form (Figure 3). The differences between Doppler waveforms in normal pregnancy versus PE indicate a marked decrease in blood flow in PE (particularly during diastole) and greater uterine artery resistive index (UARI). Among the consequences of inadequate uterine blood flow is poor perfusion of the placenta and insufficient nutrient supply to the fetus. The underlying mechanisms and impact of placental ischemia for the mother will be discussed in detail in the subsequent sections, including experimental animal models that have been utilized to study and expand our understanding of PE.

Animal models: A critical tool in understanding the pathogenesis of PE

Our current understanding of the pathogenesis of PE would be limited without the use of appropriate animal models. In fact, this multisystem disease necessitates the use of the integrated systems that whole-animal models provide. Such studies coupled to in vitro and ex vivo preparations have greatly expanded our knowledge of PE. The reduced uterine perfusion pressure (RUPP) model is a well-established animal model of PE that has been extensively studied and utilized in multiple species, including nonhuman primate (UPI; uteroplacental ischemia) (23, 59), dog (143), rabbit (4), mice (154), and the rat (16, 34, 116, 159, 185, 192, 291, 299, 329). In this model in the rat, placental ischemia is induced by placing silver clips on the abdominal aorta and branches of the ovarian artery to reduce blood flow to the uterine horns, placenta, and fetuses by approximately 40%. This procedure mimics the impaired uterine hemodynamics and placental ischemia-induced production of many of the circulating factors that occur in women with PE (117). While the RUPP model has been a powerful tool to understand the impact of placental factors on the development of PE and the associated hemodynamic changes, it is not without limitations. Due to the surgical restriction of blood flow, the RUPP model cannot be used in studies to pharmacologically promote uterine blood flow. The hypertensive Dahl Salt-Sensitive rat has been shown to spontaneously develop a PE-like phenotype, including mimicking the altered uterine artery Doppler waves in PE patients (Figure 3), without the need of surgical manipulation of the uterine vessels. It represents an excellent model of superimposed PE (119).

Infusion of individual mediators (such as chronic infusion of a particular placental-ischemic factor to mimic circulating levels encountered in PE) allows for assessment of its specific impact on symptoms of PE. For example, infusion of specific pro-inflammatory factors such as tumor necrosis factor-α (TNF-α) and angiotensin II type 1 receptor autoantibody (AT1-AA) increases blood pressure in the pregnant rodents and has widespread implications in the mother (182, 183). Other examples of animal models include genetic mouse models, such as BPH/5, which is mildly hypertensive while nonpregnant and exhibits a PE-like phenotype during pregnancy (79). Rat models overexpressing components of the renin angiotensin system have also been shown to produce a PE-like phenotype during pregnancy (139). The L-NAME (L-NG-nitro arginine methyl ester) model, where an NO synthase inhibitor is administered, produces elevated blood pressure, proteinuria, and reduced fetal weight (53). Another model involves administration of extremely low doses of bacterial lipopolysaccharide (LPS) to pregnant mice and rats to induce a pro-inflammatory profile similar to PE. The LPS model is associated with increased blood pressure, proteinuria, low platelet count, glomerular injury, and brain abnormalities (96). The critical impact that these animal models and others have had on our understanding of PE will be discussed throughout this article.

Pathogenesis of Placental Ischemia in PE

Evidence supports theories of genetic, environmental, and autoimmune origins for the development of PE. While the etiology is not completely understood, it is generally accepted that the pathogenesis of PE occurs in two phases; (1) abnormal placentation and impaired spiral artery remodeling and (2) subsequent clinical manifestations of the disorder. To understand the mechanisms by which abnormal placentation leads to PE, it is important to first understand the anatomy, development, and function of a healthy human placenta and what we have learned about mechanisms of placentation from studies in rodent uteroplacental tissue. We then discuss disturbances to these pathways that lead to lack of spiral artery remodeling in PE.

Placental vascular anatomy

The mature placenta is a complex organ composed of fetal and maternal tissue of varying cell types and three different layers. The outermost layer is the maternal decidua. The decidua is the uterine endometrial lining that has been transformed during implantation and pregnancy. Rodent and human placentas are similar, making these animals useful in the study of pregnancy (Figure 4). In both cases, proper development of the decidua, which contains the endometrial vessels, is critical (157, 276). The spiral arteries, an important target for vascular remodeling during pregnancy, are found in this layer. The innermost, or “fetal”, layer is termed the labyrinth in rodents and the villous layer in humans. This layer is composed of specialized trophoblasts that form a large surface area important in nutrient/gas exchange between the fetus and mother. The middle layer is a transitional, or junctional, layer between the “maternal” and “fetal” portions of the placenta. One difference in vascular anatomy between rodents and humans is that in the human, the spiral arteries drain directly into the intervillous spaces. However, in the rat, the spiral arteries converge onto one or more canals that pass through the junctional zone to the base of the villous layer and then branch out to feed the sinusoid/intervillous spaces where nutrient/gas exchange between maternal and fetal circulations occurs (Figure 4). Following, deoxygenated blood moves through the labyrinth, converging on smaller channels that cross the junctional zone and deliver blood to lacunae or small pools of blood in the decidual layer and then to maternal uterine veins.

Figure 4. Illustrations of mature placenta in rodents and humans.

Figure 4

This figure illustrates the differences between the (A) rodent and (B) human placenta. Rodent models of preeclampsia are valuable tools in understanding the pathogenesis of this maternal disorder. The decidua on the “maternal side” can be seen housing the spiral arteries in both placentas. The “fetal side,” closest to the umbilical cord, in the rodent is termed the labyrinth, and the villous layer in the humans. Villi form large surface areas where nutrient/gas exchange can occur between mother and baby. In terms of vascular anatomy, human spiral arteries drain directly into the intervillous space to surround the villi and allow for this nutrient/waste exchange to occur. In rodents, spiral arteries converge into one or more canals that pass through the junctional zone to the base of the villous layer and then branch out to feed the sinusoid/intervillous spaces. Adapted from Rai and Cross, 2014 (273).

Cytotrophoblast cell differentiation and proliferation

During the luteal phase of the menstrual cycle, blood flow is increased to the endometrium via the spiral arteries to become receptive to implantation of the early embryo (blastocyst). The blastocyst consists of an inner cell mass, which will become the embryo, and an outer trophectoderm layer of stem cells. In humans and rats, the trophectoderm gives rise to various trophoblast cell lineages that form the placental layers and line the maternal vessels. However, the names of these cells differ between human and rat (314). In humans, the trophectoderm gives rise to cytotrophoblasts and the primitive syncytium (107). The latter assists in early invasion of the blastocyst into the uterus and early formation of individual lacunae that fill with blood from uterine capillaries, which will eventually become the continuous intervillous space where maternal blood surrounds the placental villi (58). Proliferation of cytotrophoblasts in the initial stages of placental formation occurs in very low oxygen tensions. The introduction and delivery of oxygenated maternal blood flow generates oxidative stress and activates signaling pathways in the differentiation of these cells from a proliferative to an invasion phenotype (177, 368). Proliferative cytotrophoblasts push through the primitive syncytium to form the placental villi (176). The villous cytotrophoblasts fuse with the preexisting syncytium to form the multinucleated syncytiotrophoblasts that line the intervillous space to serve as the interface between maternal blood and the underlying villi (386). The proliferative cytotrophoblasts also create columns to anchor the placenta to the decidua. From these anchoring structures, cytotrophoblasts can differentiate into one of two phenotypes; invasive extravillous trophoblasts that reside in the interstitium to interact with decidual stromal cells, such as decidual natural killer (dNK) cells (as detailed below); or endovascular trophoblasts that invade and remodel the maternal spiral arteries (Figure 5). In rodents, the extravillous trophoblast cells are termed trophoblast giant cells (193). Distinct subtypes of trophoblast giant cells, classified by their location, function, and gene expression, line the lumen of vessels including the central canal, maternal blood sinusoids, and spiral arteries (310, 311). It is important to mention these cells in rats, because rodents have proven to be useful tools to study mechanisms of trophoblast invasion of the placenta and the dramatic uteroplacental vascular remodeling that occurs during normal pregnancy (313, 320). Cytotrophoblasts and leukocytes, namely dNK cells, make up the bulk of the placenta and work in concert to regulate appropriate spiral artery remodeling.

Figure 5. Illustrations of spiral artery remodeling during normal pregnancy and attenuation of this process in preeclampsia.

Figure 5

Spiral arteries are depicted during nonpregnancy (left panel), mid-to-late gestation during normal pregnancy (middle panel), and preeclampsia (PE) (right panel). Spiral arteries originate in the myometrium and proceed through the endometrium. Placentation during normal pregnancy is noted by decidualization and significant changes to stromal cells of the uterus, and spiral artery remodeling, which involves invasion of fetal-derived cytotrophoblast cells. Early trophoblast lineages fuse to form the overlying and multinucleated syncytiotrophoblast layer that progressively invades the uterus and forms the barrier between maternal blood and the fetal capillaries. These capillaries are developed via vasculogenesis and are housed within the placental villi. Villi are floating or anchored to the uterine wall. The cytotrophoblasts within the anchoring villous column break through the syncytium and eventually depolarize into extravillous (interstitial) trophoblast cells. The latter cells invade and replace the vascular smooth muscle and endothelium of the spiral arteries of the endometrium and the first third of the myometrium while becoming endovascular trophoblast cells. The spiral arteries are remodeled to form conduits promoting blood flow toward the intervillous space to surround and deliver oxygen and nutrients to the placental villi. Attenuations in this process promote placental ischemia and PE. Recent research indicates that resident decidual natural killer cells secrete factors that promote trophoblast function and remodel the spiral arteries, which is reduced in PE. SpA, spiral artery; ArcA, arcuate artery; RadA, radial artery; ES, endometrial stroma; EC, endothelial cells; VSMC, vascular smooth muscle cell; UE, uterine epithelium; EVT, extravillous trophoblast; dNK, decidual natural killer cell; EndoT, endovascular trophoblast; SynT, syncytial trophoblast; FC, fetal capillary.

Cytotrophoblast-mediated spiral artery remodeling

Early in pregnancy, spiral arteries are high resistance, low capacitance vessels. During normal pregnancy, these arteries undergo trophoblast-mediated remodeling to become high capacitance, low resistance vessels that support proper growth of the fetal-placental unit (60). Within the maternal vascular spaces, the trophoblast cells promote development of the placental vasculature in at least two processes: (i) vasculogenic mimicry and (ii) vascular invasion and endothelial mimicry. Vasculogenic mimicry describes how trophoblast cells can create de novo vascular tubes. This process occurs with the formation of the canals, sinusoids/villous space, channels, and lacunae. Vascular invasion and endothelial mimicry describe how trophoblast cells invade into and displace the resident endothelial cells and then alter their morphology to appear more like an endothelial cell (273). This process occurs in spiral arteries in which trophoblasts migrate from the anchoring villi and invade the vasculature intraluminally, termed endovascular invasion, or extraluminally where they colonize the vascular smooth muscle layer (367). In in vitro studies, extravillous trophoblasts promote remodeling in non-placental spiral arteries isolated from women at Caesarean section by inducing endothelial and smooth muscle apoptosis through the cell death surface receptor Fas and its interaction with Fas ligand (27). The Fas receptor belongs to the TNF family of receptors and is a classical mode for induction of cell death by cytotoxic immune cells (361). Migration, invasion, and recolonization of vascular cells by trophoblasts is a progressive process that begins around midgestation in rodents and toward the end of the first trimester humans (290). This phenomenon results in normal remodeling of the uterine spiral arteries. Trophoblast-driven vascular remodeling to widen the spiral arteries is considered an important mechanism to increase blood flow to the growing fetus. During pregnancy, spiral arteries widen from a diameter of 200 μm to 2 mm by term (368) and increasing placental blood flow 4.5 to 10-fold reaching approximately 500 to 600 mL/min at term in humans (24, 280).

Hormones, immune cells, and spiral artery remodeling

Several pregnancy hormones and growth factors drive cytotrophoblast invasion and remodeling. Progesterone derived from decidual stromal cells stimulates increases in vascular endothelial growth factor (VEGF)-A and matrix metalloproteinase (MMP)-2 and −9 (109, 174). VEGF serves as a chemoattractant for first-trimester trophoblast cells and promotes migration (187). Relaxin has antiapoptotic effects on extravillous trophoblast cells (197). In a study in baboons, early pregnancy was associated with low levels of estrogen, which permitted cytotrophoblast invasion into spiral arteries (10, 45). When estradiol levels were increased threefold, cytotrophoblast invasion was significantly reduced, suggesting that lower levels of estrogen that exist in early pregnancy could be important to allow proper cytotrophoblast invasion.

dNK cell activity has been well described in pregnancy. These cells differ from systemic NK cells in that they have immunomodulatory properties and are only marginally cytolytic (276). dNK cells recognize maternally and paternally derived major histocompatibility complexes (MHC). Extravillous trophoblasts of fetal origin express human leukocyte antigen-C (HLA-C) MHC, which combine and produced a unique complex with the maternal killer cell Ig-like receptor (KIR) MHC (228). The introduction of these allogenic HLA-C MHC from the fetus challenges the maternal immune system. Genetic studies have shown that pregnancies with PE are associated with specific KIR-HLA combinations, and that these PE-related combinations could cause inappropriate secretion of chemokines/cytokines by dNK cells and ultimately impact trophoblast invasion (137, 141, 228). Proper placentation and spiral artery remodeling have been shown to be dependent on the cooperation between dNK cells and extravillous trophoblasts (228, 276). dNK cells have been shown to play a major role in cytotrophoblast regulation and invasion (137, 178) by releasing proinvasive factors that stimulate activation of MMP-2 and MMP-9. In early human pregnancy, uterine NK cells are the predominant source of MMP-2 (242). MMPs are important for their role in breaking down components of the extracellular matrix (ECM), thereby removing physical barriers for trophoblast migration. In addition to their role in cytotrophoblast invasion, dNK cells are important secretors of cytokines, growth factors, and angiogenic factors, such as VEGF and placental growth factor (PlGF), and do not express the aggressive cytotoxic phenotype (95). Studies have shown that cytolytic NK cell populations decrease during normal pregnancy, while regulatory NK cells increase, not only in the decidua (dNK cells) but also in the periphery; however, the opposite is true in PE (294).

Impaired cytotrophoblast migration and invasion in PE

Phase 1 in the pathogenesis of PE is hallmarked by abnormal placentation. Specifically, evidence supports a lack of proper trophoblast-mediated spiral artery remodeling in PE. Histological samples of placenta from PE pregnancies frequently depict shallow invasion and incomplete remodeling of these vessels compared to placenta from healthy pregnancies (49), where approximately 100 to 150 spiral arteries undergo complete remodeling (204). Human placental bed biopsies from PE patients indicate that abnormal vascular anatomy is characterized by a lack of decidual and myometrial vascular wall destruction and reductions in endovascular and intramural trophoblast cell counts (203, 204). Human placental tissues from PE patients show a reduction in MMP-2 and MMP-9 levels (265), suggesting a decreased trophoblast invasive/migratory ability (382).

Factors that impact dysregulation of cytotrophoblast function in PE is an active area of research. One study by Zhou et al. (384) showed that cytotrophoblasts do not appropriately transform to express the appropriate vascular adhesion molecules, including integrin and cadherin, during invasion in PE. Another source of cytotrophoblast dysfunction could be uncontrolled oxidative stress. Reactive oxygen species (ROS) production is critical in the signaling of several processes in normal pregnancy, from implantation to proper growth of the fetoplacental unit. However, ROS-generating enzymes are excessively increased, and antioxidant enzymes are decreased in both trophoblasts and placentas from PE pregnancies (385). There are multiple mechanisms whereby ROS are produced throughout PE, attributed mostly to hypoxia, inflammation, and reperfusion injury (121). The placenta undergoes prominent ischemia-reperfusion injury, resulting in ROS formation and endoplasmic reticulum (ER) stress. ER stress has been cited to trigger decidual cell and cytotrophoblast apoptosis in PE (194). Upregulation of proapoptotic transcription factors as a result of ER stress has been shown to negatively regulate transcription of the proangiogenic factor, PlGF, in PE (227). Nuclear factor-kappa B (NF-κB), a redox-sensitive transcription factor, has also been implicated in the early stages of PE by impairing spiral artery remodeling and inducing trophoblast apoptosis (1, 26). In response to high levels of ROS, placental mitochondria become dysfunctional and may undergo degradation (fission) or rescue and repair (fusion) controlled by specific hydrolyzing proteins. Studies show dysregulation of profission and profusion factors in PE compared to normal pregnancy and also in PE with and without severe features (31, 101, 355). However, the full implications of these findings remain unclear. Antiphospholipid syndrome, which is linked to miscarriages and increased risk of PE, is caused by production of antiphospholipid antibodies. Recently, a study found that these antibodies are internalized by syncytiotrophoblasts, and that they specifically target mitochondria and induce ROS generation (354, 387). ROS also cause upregulation of proapoptotic pathways in mitochondria (257), and mitochondrial dysfunction itself leads to further ROS production (235). Ultimately, these disturbances lead to abnormal cytotrophoblast invasion, impaired spiral artery remodeling, poor perfusion of the placenta, and restricted oxygen and nutrient/waste exchange to the fetus.

Experimental animal studies linking trophoblast migration and invasion to spiral artery remodeling

Genetic-manipulation studies in mice have implicated the importance of cytotrophoblast-mediated spiral artery remodeling in pregnancy. Although the number of studies on this topic is too expansive for this article, a few are addressed. Trophoblasts expressing the Tpbpa gene are progenitors of trophoblasts that line most of the maternal vasculature and the sole progenitor of trophoblasts lining the spiral arteries. Ablation of Tpbpa-expressing trophoblasts, using genetically modified animals, decreases the number of trophoblasts lining the maternal placental vasculature, including the spiral arteries, which correlates to a reduction in maternal spiral artery diameters (147). As a likely consequence, these mice had reduced maternal blood volume within the labyrinth or villous layer, the placental layer where maternal-fetal gas and nutrient exchange occurs, consistent with placental insufficiency. All pups with a loss of this specific population of trophoblasts were not viable before the end of the second trimester.

Some studies have gone further to assess the effects of genetic manipulation on trophoblast migration/invasion and spiral artery remodeling. Notch 2 is an important paracrine signaling receptor for cell movements (270). Knockout of Notch 2 in Tpbpa-expressing trophoblast cells significantly reduced trophoblast invasion of spiral arteries. Moreover, altered spiral artery trophoblast invasion was associated with reduced canal diameters and placental perfusion, a key feature in PE, directly linking trophoblast migration and invasion with vascular remodeling (150). Therefore, it is probable to hypothesize that these trophoblast-specific modifications and subsequent abnormalities in placenta vascular remodeling would lead to PE-like symptoms such as hypertension in these mice, particularly because it has been shown that Notch 2 is reduced in placentas from women with PE (383). In another study, adoptive transfer of NK cells from Rag2−/− mice, which lack T or B lymphocytes, into BALB/c-Rag2−/−Il2−/− mice, which lack T, B, and NK cells, corrected the uterine vascular defects found in the latter mouse (144), suggesting that NK cells play a critical role in spiral artery remodeling.

Uterine blood flow is reduced up to 50% in patients with PE (280). As such, markers of placental ischemia and hypoxia, such as hypoxia inducing factors (HIF-1α and HIF-2α), are elevated in PE placental tissue (274). In mice, selective overexpression of HIF-1α in placenta is capable of inducing hypertension, a consequence of placental ischemia (155). In humans, the terminal villi of placentas associated with fetal growth retardation have elongated villous capillaries and inconspicuous loops, suggesting a larger surface area (180). This latter finding would be consistent with placental insufficiency, that is, inefficient nutrient/waste exchange between mother and fetus. In summary, these studies suggest that the pathogenesis of PE is mediated by a lack of trophoblast migration/invasiveness and spiral artery remodeling. The resulting fetal-placental ischemia induces the release of vasoactive factors from the placenta. Since these processes begin early in pregnancy, it is not surprising that circulating placenta factors in response to placental ischemia and vascular resistance are evident much earlier than the clinical manifestation of PE.

Clinical Manifestations of PE

Phase 2 in the pathogenesis of PE is the clinical manifestation of the maternal syndrome. Incomplete development of spiral arteries leads to placental ischemia and the subsequent release of pro-inflammatory and antiangiogenic factors into the maternal circulation. There is also evidence to suggest that PE is associated with increased complement levels (54, 261), which contributes to the excessively hypercoagulable state of this disease. In particular, mutations in the central complement gene, C3, were found to be associated with PE with severe features (153).

  • Reactive oxygen species in PE. Oxidative stress plays a prominent role in the maternal syndrome. In phase 2, ROS are generated in a number of ways, including cellular injury and other biological processes, such as activation of the angiotensin II (Ang II) type 1 (AT1) receptor. In vitro studies have also shown that uterine arteries from rats that are isolated and incubated with serum from PE patients show greater superoxide staining than those incubated with serum from normal pregnant women. This can also be observed in vessels isolated from RUPP rats (165, 299). Ultimately, the increase in prooxidant molecules in PE causes organ injury and leads to endothelial dysfunction by directly inducing vasoconstriction, stimulating production of vasoactive factors, or inhibiting mechanisms for vasodilation.

  • Immune factors in PE. The most well-studied immune factors that are increased in association with PE are the agonistic AT1-AA, TNF-α, interleukin (IL)-6, IL-17, T helper (Th)-1 cells, and Th-17 cells. In fact, infusion of AT1-AA, TNF-α, or IL-17 individually each increases MAP in pregnant rats (182, 183, 342). Also, blockade of the AT1-AA (Figure 6) or TNF-α in the RUPP rat model of PE has been shown to reduce blood pressure, improve angiogenic balance, and decrease expression of vasoconstrictive factors, such as endothelin-1 (ET-1) (71, 183). AT1-AA infusion into rats is associated with increases in the ET-1 precursor, prepro-ET, in the renal cortex and placenta (182, 236).

Figure 6. Angiotensin II Type 1 receptor autoantibodies are increased in preeclampsia.

Figure 6

Panel (A) illustrates that levels of the agonistic angiotensin II type 1 receptor autoantibody (AT1-AA) are significantly increased in serum from preeclamptic women. For these data, spontaneous beating of neonatal rat cardiomyocytes in response to serum exposure was used. Panel (B) shows that when the AT1-AA, which is elevated in the reduced uterine perfusion pressure (RUPP) rat model of preeclampsia (PE), is inhibited (with “n7AAc”), mean arterial pressure (MAP) is reduced, suggesting that AT1-AA has a role in increasing blood pressure in PE. Data are mean + SEM. *P < 0.05. Adapted from Wallukat G, et al., 2008 (359) and Cunningham MW, et al., 2018 (71).

Animal models of PE have been used to assess specific immune cell types that are known to be involved in the pathogenesis of PE. Cornelius et al. showed that Th-17 cells isolated from the spleen of RUPP rats, cultured and injected interperitoneally into normal pregnant rats, induced a PE-like phenotype (66, 357). Furthermore, increasing the population of T regulatory cells (Tregs) in RUPP rats significantly reduces MAP, circulating levels of IL-17 and TNF-α, renal ROS, placental ROS, and prepro-endothelin-1 (prepro-ET-1) (65, 152). NF-κB is activated by Ang II, which is elevated in PE and in turn further increases release of pro-inflammatory cytokines and chemokines. PE is associated with an excess of these pro-inflammatory factors and a deficiency of IL-10 derived from Tregs. IL-10 plays an important role in normal pregnancy in regulating the polarization of T cells to a Th-2 phenotype over the pro-inflammatory Th-1 phenotype (293). Interestingly, supplementation of IL-10 alone in the RUPP rat is sufficient to attenuate MAP and pro-inflammatory factors (138). As discussed previously, NK cells have been shown to play a significant role in PE. Depletion of cytolytic NK cell in the RUPP rat lowers MAP, intrauterine growth restriction (IUGR), and inflammation (159). These data suggest that the increased pro-inflammatory immune cell populations play a deleterious role in PE. These factors all act to cause pathological activation of the maternal endothelium (Figure 7) as well as directly impacting multiple organ systems.

Figure 7. Role of immune factors in the development of endothelial dysfunction in preeclampsia.

Figure 7

This figure illustrates that pro-inflammatory T- and B-cell activation results in elevated prohypertensive cytokines and the angiotensin II type 1 receptor autoantibody (AT1-AA). These factors cause endothelial dysfunction by increasing endothelin-1 (ET-1) production and reducing nitric oxide (NO) bioavailability.

Endothelial dysfunction is a hallmark feature in PE

Endothelial dysfunction is an integral part of PE pathophysiology, specifically during the clinical manifestation of this disease. To understand endothelial dysfunction in PE, it is helpful to understand that during normal conditions, the endothelium (innermost layer of vessels composed of endothelial cells) produces factors that act to maintain vascular tone. Healthy pregnancy is associated with an increase in artery diameter and reductions in total peripheral resistance in early gestation, in part due to an increase in endothelial NO-induced vasodilation. In contrast, ET-1, another endothelium-derived factor, is the most potent pressor (or vasoconstrictor) known. ET-1 has been shown to be significantly elevated during PE pregnancy compared to normal pregnant women and correlates with the severity of the disease in some cases (113, 211). Furthermore, elevations in endothelium-derived vasoconstrictors, including ET-1, Ang II, and thromboxane A2, occur weeks prior to clinical onset of PE (122, 226, 339). Reduced peripheral resistance along with adequate perfusion of maternal organs and the uteroplacental unit are all vital for healthy pregnancy. Therefore, it is important to remember that these vasorelaxing and vasoconstricting factors are produced in a delicate balance, and aberrations to this system can have widespread implications. In the 1990s, in vitro studies confirmed that serum from PE women contained high levels of ET-1 (333), indicating that endothelial dysfunction could be the cause of hypertension and organ damage in this disease. Sera from both PE patients and animal models have been used to show that exposure to circulating placental factors induces ET-1 production (35, 183, 271). In addition, vessels from animal models of PE have been used to demonstrate resistance to mediators of vasorelaxation (acetylcholine; Figure 8) and increased sensitivity to vasoconstrictors (54).

Figure 8. Endothelial dysfunction in placental ischemic rats.

Figure 8

(A) This figure illustrates blunted endothelial-dependent vasorelaxation in response to increasing concentrations of acetylcholine (Ach) in phenylephrine (Phe)-constricted aortic tissue isolated from rats with reduced uterine perfusion pressure (RUPP). (B) ACh at these concentrations induces NO production in normal pregnancy, and this effect is attenuated in aortic strips from placental ischemic rats. Data are mean ± SEM. *P < 0.05 versus pregnant. Adapted from Barron LA, et al., 2001 (39).

There are a number of ways that ET-1 production (and its precursor big ET-1) is increased in PE. Reduced NO production has been shown to inhibit ET-1 production (167). Placental release of both ROS and TNF-α into the maternal circulation has been shown to induce ET-1 production. MMP-1 and MMP-2, which are increased in PE, cleave the ET-1 precursor, big ET-1, to active ET-1 within systemic blood vessels (3). In addition, MMP-1 activates the protease-activated receptor 1 (PAR-1) and mediates the release of ET-1 in endothelial cells (246). Another study found elevated endothelin-converting enzyme, which converts big ET-1 to vasoactive ET-1, in the maternal circulation of PE patients (9). It is most likely a combination of these pathways that pathologically increases vascular and circulating levels of ET-1 in PE women.

ET-1 acts on two G-protein-coupled receptors on the cell surface: ET-1 Type A (ETA) and Type B (ETB), which are localized to vascular smooth muscle cells and endothelial cells, respectively. Activation of the ETA receptor causes vasoconstriction and cell proliferation, while the ETB receptor facilitates vasodilation and natriuresis via NO and prostacyclin, which are significantly reduced in PE (85, 212). ETA antagonism has been studied in a number of animal models of PE and is associated with reductions in MAP (Table 4; Figure 9) and ET-1 precursors in some cases (11, 182, 236). While the role of the ETA receptor in PE is well established, less is known about the role of ETB. However, studies have shown that ETB expression is reduced in the RUPP rat, which could at least in part explain reduced ETB-dependent vasorelaxation (136). Another study showed that ETB-deficient pregnant rats have increased blood pressure during pregnancy (317). Interestingly, chronic infusion of the antiangiogenic factor, sFlt-1, into pregnant rats significantly increased prepro-ET in the renal cortex (182, 236).

Table 4.

Summary of Studies that Inhibit Key Pathways and Placental Factors in Animal Models of Preeclampsia

Intervention Model/Animal MAP References
sFlt-1 inhibition
 PlGF supplementation RUPP—rat (323)
UPI—nonhuman primate (206)
 VEGF supplementation RUPP—rat (198)
ROS inhibition RUPP—rat (299)
AT1-AA inhibition RUPP—rat (71)
TNF-α inhibition RUPP—rat (183)
ETA antagonism RUPP—rat (11)
sFlt-1 infusion—rat (236)
AT1-AA infusion—rat (182)
TNF-α infusion—rat (186)

PlGF, placental growth factor; VEGF, vascular endothelial growth factor; RUPP, reduced uterine perfusion pressure; UPI, uteroplacental ischemia; ROS, reactive oxygen species; AT1-AA, angiotensin II type 1 autoantibody; TNF-α, tumor necrosis factor-α; ETA, endothelin type A receptor.

Figure 9. Endothelin type A receptor blockade reduces blood pressure in animal models of preeclampsia.

Figure 9

This figure illustrates that endothelin type A (ETA) antagonism reduces mean arterial pressure (MAP) in (A) the reduced uterine perfusion pressure (RUPP) model and (B) the sFlt-1 infusion (sFlt-1) model of preeclampsia. Data are mean + SEM. *P < 0.05 versus pregnant. **P < 0.05 versus sFlt-1/RUPP. Adapted from Murphy S, et al., 2010 (236) and Alexander B, et al., 2001 (11).

Angiogenic imbalance and endothelial dysfunction in PE

One of the most intensely studied characteristic factors involved in endothelial dysfunction in PE is sFlt-1 and the angiogenic imbalance that exists in this disease. VEGF is a critical player in normal pregnancy, not only in the development of new vessels and angiogenesis but also in maintaining endothelial integrity. PlGF, a member of the VEGF family, is another angiogenic factor released by the placenta that serves to maintain proper endothelial function during pregnancy. These angiogenic factors bind to vascular endothelial growth factor receptors (VEGFRs) (267). VEGF binds to VEGFR-1 (Flt-1) and VEGFR-2 (Flk-1), whereas PlGF binds exclusively to Flt-1 (98). The soluble form of Flt-1, called sFlt-1, is increased in the circulation of women with PE, where it antagonizes free VEGF and free PlGF. Depletion in the bioavailability of these circulating growth factors is at the detriment of the endothelium and blood pressure regulation (Figure 10). This was demonstrated by sFlt-1 overexpression and infusion into animal models, resulting in increased MAP and a PE-like phenotype (215, 236). Furthermore, PlGF administration has been shown to reduce blood pressure in the RUPP rat and a nonhuman primate model of PE, while VEGF administration has been shown to improve blood pressure in the RUPP model (198, 206, 323). It is important to note that normal pregnancy is associated with low levels of sFlt-1 that gradually increase toward term. Throughout gestation, sFlt-1 levels do not exceed the levels of VEGF and PlGF. Endothelial dysfunction occurs because PE is associated with uncontrolled excess of sFlt-1 that surpasses and antagonizes the available angiogenic factors. Increased circulating levels of sFlt-1 and reduced PlGF levels have been documented in women with PE and, in fact, prior to the onset of clinical symptoms. In 2003, Maynard et al. (215) showed that elevated levels of sFlt-1 are present in PE patients and to a greater extent in PE with severe features. These data were accompanied by decreased circulating free levels of VEGF and PlGF in PE, with much lower concentrations in plasma from women with PE with severe features (Figure 10). Studies also support the presence of a susceptibility locus near the FLT1 gene in neonates from PE pregnancies, suggesting that sFlt-1 is not simply released in response to placental ischemia but rather provides a genetic basis for this disease. Another antiangiogenic factor that is observed to be increased in PE is soluble endoglin (sEng). It is produced by the placenta and has been shown to reduce binding of transforming growth factor-β1 (TGF-β1), which is also important in cell proliferation, vasodilation, and angiogenesis.

Figure 10. Circulating levels of angiogenic factors in preeclampsia.

Figure 10

These data depict the antiangiogenic environment that exists in preeclampsia (PE), whereby (A) sFlt-1 is increased and (B) free vascular endothelial growth factor (VEGF) and (C) placental growth factor (PlGF) are decreased. Importantly, these changes are exaggerated as the severity of PE increases. Data are mean ± SEM. *P < 0.05 versus Mild. **P < 0.05 versus Severe. Adapted from Maynard SE, et al., 2003 (215).

Similar to the ROS and inflammatory factors discussed above, antiangiogenic factors act to dysregulate the NO-soluble guanylate cyclase (sGC)-cyclic guanosine monophosphate (cGMP) signaling pathway, which results in reduced vasorelaxation. One of the major outcomes of endothelium-bound VEGFR activation is the synthesis of NO, which diffuses to the vascular smooth muscle cell and binds to sGC to facilitate the conversion of the second messenger cGMP. cGMP is involved in a number of processes, including cell proliferation and vasodilation. Specifically, cGMP activates protein kinase G (PKG), which phosphorylates voltage-gated potassium channels and promotes relaxation (18, 297). In addition, increased cGMP levels inhibit calcium channel activity, blocking influx of calcium into the cell (156). In the absence of VEGFR stimulation, NO production is reduced, which has been reported in PE women in a number of studies (200, 212). Furthermore, increased levels of ROS in PE act on any available NO to produce peroxynitrite, an unstable, highly reactive, and damaging molecule. Studies show that sGC oxidation by ROS reduces signaling activity and cGMP production, diminishing vasodilatory activity. Studies in animal models have shown that improving NO signaling, by blocking the degradation of cGMP, attenuates hypertension (114, 118, 335). In addition, blockade of cGMP degradation is associated with reductions in circulating sFlt-1 (275) and increases in PlGF (161). Recent studies have investigated the link between the antiangiogenic environment and ET-1 in pregnant women and found that those with higher sFlt-1/PlGF ratio had greater levels of ET-1 (7, 352). All of the factors described in the sections above ultimately drive increases in ET-1 and decreases in NO bioavailability (Figure 11).

Figure 11. Factors that impact endothelial dysfunction in preeclampsia.

Figure 11

This figure illustrates the development of endothelial dysfunction and hypertension in preeclampsia. The release of factors from the ischemic placenta, such as antiangiogenic factors, reactive oxygen species (ROS), matrix metalloproteinases (MMPs), angiotensin II type 1 receptor autoantibody (AT1-AA), and pro-inflammatory cytokines, play a central role in depleting nitric oxide (NO) bioavailability and increasing endothelin-1 (ET-1) production, which drives this prohypertensive pathway.

Another pertinent factor to consider in relation to increased oxidative stress and endothelial dysfunction are lipid peroxides. Circulating small, dense low-density lipoproteins (LDLs) have been shown to be increased in PE, which provide fatty acid substrates for lipid peroxidation (234). Oxidized low density lipoprotein (oxLDL) is increased in PE and binds to endothelial receptors, such as LOX-1 (lectin-like oxidized LDL receptor-1), to internalize and metabolize oxLDL particles. LOX-1 expression has been shown to be increased in response to TNF-α, IL-1, oxLDL, and Ang II, all of which are increased in PE (162, 252, 305). Increased LOX-1 activity is associated with increases in superoxide and peroxynitrite via activation of NADPH oxidase. Increased expression and activity of LOX-1 has been found to play a role in a number of pregnancy- and non-pregnancy-related diseases and thus is thought to contribute to vascular endothelial dysfunction in PE (316).

Sympathetic nerve activity in PE

The sympathetic nervous system tracks and innervates the adventitia of almost every blood vessel (57), and increased sympathetic nerve activity (SNA) is known to promote vasoconstriction and mediate the development of hypertension (102). However, research concerning this system has been relatively understudied in the field of pregnancy and PE. Much of what is currently known about this system in hypertensive pregnancies has been derived from associational studies using surrogate markers of SNA, including low frequency/high frequency (LF/HF) of heart rate variability (91, 231, 374), altered autonomic regulation of heart rate during orthostasis (191, 327), reduced baroreflex sensitivity (230, 281, 366), and circulating and uterine levels of vasoconstrictive substances such as catecholamines and neuropeptide Y (NPY) (2, 104, 169, 175, 248, 255). These markers are higher in PE versus control normotensive pregnancies and point to SNA being elevated in the former group of women, as reviewed by Spradley (319) and Reyes et al. (279). Far fewer studies have attempted to directly link SNA to hypertension in PE, but a study dating back to 1948 provided evidence that SNA drives increased blood pressure in superimposed PE (259). Those authors demonstrated that splanchnicectomy lowered blood pressure in women with superimposed PE. Since then, other investigations have detected that muscle (M)SNA is elevated in women with PE (124, 125, 298). These nerve-recording experiments are important but remain associational in nature and have not fully provided a cause-or-effect relationship between SNA and the pathogenesis of hypertension in PE.

As discussed throughout this article, the pathogenesis of PE is thought to originate from abnormal spiral artery remodeling and placental ischemia with subsequent release of prohypertensive factors such as sFlt-1. This antiangiogenic factor quenches the vasoprotective and antihypertensive actions of VEGF and PlGF (215), which ultimately causes endothelial dysfunction, vasoconstriction, and resultant hypertension. Preliminary experiments in rodents have implied that increased sympathetic drive leads to placental ischemia and alterations in circulating angiogenic factors. Pregnant rats subjected to a combination of chronic sonic, overpopulation, and immobilization stressors have reduced placental levels of the angiogenic factor, PlGF, and increased blood pressure (67). Such chronic stress-induced increases in SNA and blood pressure are supported by other studies and also been detected in rats encountering cold and fasting stress (164, 170, 328). Further evidence that neurological mechanisms initiate the development of PE is that acutely activating sympathetic centers within the brain using Kurotu’s electrodes increased blood pressure in pregnant rats (205). Pregnant women destined to develop PE have increased blood pressure response to sympathetic outflow elicited by the cold-pressor test (369). This test has been shown to activate central pathways within the rostral ventrolateral medulla (RVLM), which increases sympathetic outflow and blood pressure in rats (240). Blockade of oxidative stress within the RVLM significantly reduced blood pressure in an animal model of PE, where desoxycorticosterone acetate (DOCA) is administered to pregnant rats (373). Based on these collective studies, it is feasible to hypothesize that elevated sympathetic drive promotes uteroplacental vasoconstriction, as there are increased nerve fibers observed in the placental bed myometrium in women with PE (99). This sympathetic signaling pathway would then incite placental ischemia and release of soluble placental factors, such as sFlt-1, to quench bioavailable angiogenic factors and cause maternal hypertension.

It is also likely that increased SNA is involved in the downstream hypertensive responses to placental ischemia. The involvement of SNA and its effector target, the adrenergic receptors, in placental ischemia-induced systemic vasoconstriction and hypertension had not been studied until Spradley et al. (321) tested the hypothesis that adrenergic receptor blockade would attenuate hypertension in the RUPP model of PE. They found that adrenergic receptor blockers significantly attenuated placental ischemia-induced hypertension and reported a lesser blood pressure response in normal pregnant control rats. Other researchers have provided suggestive evidence that SNA is increased in RUPP rats by finding that baroreflex sensitivity of blood pressure is reduced in this model of PE (142); this would implicate increased sympathetic drive to the systemic circulation. Indeed, Hines et al. found that the baroreflex was reset to a higher set point (i.e., reduced sensitivity) along with increased renal (R)SNA. It could be that the elevated SNA coupled to the endothelial dysfunction reduces the ability of the endothelium to buffer sympathetic flow-directed activation of adrenergic receptor-mediated vasoconstriction in PE. Overall, it is possible that elevated SNA is not only involved in the maintenance of vasoconstriction and increased blood pressure in PE but also the initiation of upstream placental ischemia and release of factors that cause endothelial dysfunction.

Multiorgan dysfunction in PE

In addition to widespread systemic endothelial dysfunction, PE is associated with multiorgan dysfunction and injury. Localized endothelial dysfunction itself is a major contributor to end-organ damage. Here, we discuss some of the major organ systems affected by this disease.

The kidney in PE

Normal pregnancy is associated with marked increases in renal hemodynamics. Renal plasma flow and glomerular filtration rate increase approximately 40% to 60% compared to nonpregnant women (181, 332). The clinical renal features of PE include reductions in renal hemodynamics, proteinuria, podocyturia, and glomerular endotheliosis that is identified by endothelial swelling and glomerular capillary narrowing (Figure 12) (214, 241). Excessive levels of sFlt-1 in PE are believed to cause glomerular membrane abnormalities by inhibiting VEGF binding to its receptors on glomerular endothelial cell and podocytes (214, 241). In support of this concept are animal studies demonstrating that increasing plasma sFlt-1 levels in pregnant mice or rats, via direct infusion of sFlt-1 or injection of adenovirus expressing the sFlt messenger RNA, results in an increase in blood pressure accompanied by proteinuria (Figure 13) and glomerular endotheliosis (Figure 14) (93, 215, 236, 326). Interestingly, anti-VEGF therapies given to nonpregnant rats or mice also cause glomerular endothelial injury and proteinuria (326). Glomerular disease characterized by proteinuria and glomerular endotheliosis has also been reported in a podocyte-specific VEGF knockout mouse (93). Collectively, these animal studies support the concept that sFlt-1-induced VEGF deficiency can produce a renal phenotype seen in women with PE.

Figure 12. The development of renal dysfunction in preeclampsia.

Figure 12

This figure illustrates the development of renal injury and dysfunction in preeclampsia, resulting in hypertension and proteinuria. Endothelial cell dysfunction and damage in the kidney leads to increased renal vascular resistance, impaired pressure natriuresis, hypertension, and proteinuria.

Figure 13. Elevated sFlt-1 induces a preeclampsia-like phenotype in rats.

Figure 13

This figure illustrates that rats injected with sFlt-1 (via sFlt-1-adenovirus construct) develop (A) elevated mean arterial pressure (MAP) and (B) proteinuria, as measured by urine albumin/creatinine, compared to controls (Fc). Data are presented as mean + SEM. *P < 0.05. Adapted from Maynard SE, et al., 2003 (215).

Figure 14. Elevated sFlt-1 causes fibrin deposition in the glomeruli of rats.

Figure 14

This figure illustrates fibrin deposition within the glomeruli of rats treated with sFlt-1 (sFlt-1) compared to control rats (Fc). Electron microscopy (EM) from an sFlt1-treated rat confirms glomerular injury. Immunofluorescence (IF) for fibrin shows foci of fibrin deposition in glomeruli of sFlt1 rats. The IF images were collected at 40×, and the EM images were collected at 2400× magnification. Adapted from Maynard SE, et al., 2003 (215).

Animal models used to examine the pathophysiology of PE have also been reported to exhibit reduced renal plasma flow and glomerular filtration rate. In RUPP rats, renal plasma flow (<23%) and glomerular filtration rate (<40%) are decreased as well as reductions in NO synthase (NOS1 isoform) (16). A twofold increase in plasma TNF-α in pregnant rats produces significant reductions in renal function that is associated with significant decreases in renal NOS1 protein expression (14, 185). Inhibition of NOS1 in pregnant rats at midgestation results in decreases in renal plasma flow and glomerular filtration rate (5, 196). These data suggest that NOS1 plays a role in mediating the increases in renal hemodynamics that occur during pregnancy and may mediate the reduction in renal hemodynamics in response to placental ischemia. Other factors such as increases in sFlt-1, ET-1, and ROS may also contribute to the reduction in renal hemodynamics.

The liver in PE

Liver dysfunction in PE is caused by increased resistance within the hepatic vascular bed, resulting in ischemia, and elevated circulating liver enzymes, or transaminases. HELLP syndrome occurs in approximately 12% of patients with PE and is characterized by the presence of hemolysis, elevated liver aminotransferases, and low platelet counts. Importantly, transaminases can be elevated in PE patients both with and without HELLP syndrome (272). Another factor contributing to liver dysfunction is believed to be fibrin deposition within the hepatic sinusoids, resulting in sinusoidal obstruction, vasospasm of the liver vascular bed, and liver ischemia, leading to large hematomas, capsular tears, and intraperitoneal hemorrhage (225, 232). Histological changes in the liver in HELLP syndrome may include periportal abnormalities, such as with hemorrhage, sinusoidal fibrin deposition, and hepatocyte necrosis (225, 304).

The brain in PE

While PE can affect multiple organs, the brain is particularly vulnerable. Cerebrovascular dysfunction during PE can lead to acute and long-term cerebral complications in the mother. Acute cerebral complications include cerebral edema, stroke, or even eclampsia (378). Approximately 40% of maternal deaths that result from PE are attributed to cerebrovascular abnormalities (276). The underpinning mechanisms have yet to be fully elucidated; however, they are usually associated with endothelial cell injury and cerebrovascular overperfusion or in some cases vasospasm. Using angiography imaging, diffuse or multifocal segmental vascular narrowing has been observed in women having PE with severe features, which is consistent with some of the forms of cerebrovascular abnormalities listed above (378).

There is also substantial evidence in both PE patients and experimental animal models of PE that impaired regulation of cerebral blood flow, increased blood brain barrier (BBB) permeability, and cerebral edema play an important role in the acute and long-term consequences of cerebrovascular abnormalities. Overperfusion of the brain, due to decreased cerebrovascular resistance, is one mechanism whereby the BBB is disrupted to cause vasogenic edema and neurologic symptoms (Figure 15). In addition, reduced vascular resistance leads to diminished hypertensive remodeling of cerebral arteries during PE and may expose the maternal brain to significantly elevated blood pressure (135). Supporting this concept are studies reporting that pregnancy prevents hypertensive inward remodeling in animal models (135). Another potential mechanism contributing to the increased risk for cerebrovascular disease in women with PE is altered cerebral vascular autoregulatory index resulting from impaired vascular myogenic tone (Figure 16). Some, but not all studies, have reported impaired cerebral blood flow autoregulation in patients with PE; this may be related to the difficulty in assessing cerebral blood flow in pregnant women.

Figure 15. The development of cerebral dysfunction in preeclampsia.

Figure 15

Hypertension and placental factors in preeclampsia cause cerebrovascular dysfunction, resulting in impaired cerebral blood flow autoregulation, vessel rupture, and blood brain barrier permeability, leading to hemorrhagic shock and cerebral edema.

Figure 16. Autoregulation in human preeclampsia.

Figure 16

This figure illustrates middle cerebral artery blood flow autoregulation in normal and preeclamptic pregnancy at 35 to 37 weeks of gestation and shows that the autoregulatory index is impaired in preeclampsia. Data are mean + SD. *P < 0.05. Adapted from Van Veen TR, et al., 2013 (349).

As cerebrovascular function is difficult to monitor in pregnancies already complicated by PE, animal models have been useful tools in understanding the underlying mechanisms. Placental ischemia in the RUPP model leads to increased BBB permeability (Figure 17) and cerebral edema. In these studies, impaired cerebral blood flow regulation was particularly evident at higher perfusion pressures (Figure 18), as well as vascular myogenic reactivity (Figure 19) (291, 363). Furthermore, impaired vascular myogenic response in the RUPP rat is associated with reduced beta epithelial sodium channel (βENaC) expression in cerebral vessels. βENaC proteins form non-voltage-gated Na+ channels and are members of the degenerin protein family. They are expressed in vascular smooth muscle cells and involved in promoting vascular tone induced by changes in pressure. The importance of the altered βENaC proteins and cerebral hemodynamics on BBB function is currently unknown, but there is compelling evidence that placental factors may have a direct effect on cerebral vessels to enhance BBB permeability (135).

Figure 17. Blood-brain permeability in placental ischemic rats.

Figure 17

This figure illustrates increased blood-brain permeability in (A) cerebrum and (B) anterior cerebrum of reduced uterine perfusion pressure (RUPP)-operated rats, as measured by extravasation of Evans blue dye. Data are presented as mean + SEM. *P < 0.05. Adapted from Warrington JP, et al., 2015 (363).

Figure 18. Cerebral blood flow and autoregulation index in placental ischemic rats.

Figure 18

This figure illustrates that (A) cerebral blood flow (CBF), as measured by laser Doppler flowmetry, and (B) autoregulation are significantly impaired in placental ischemic rats (reduced uterine perfusion pressure; RUPP), as evidenced by increases in blood flow at elevated mean arterial pressures (MAP). Data are mean ± SEM. *P < 0.05 versus pregnant, **P < 0.01 versus pregnant. Adapted from Warrington JP, et al., 2014 (365).

Figure 19. Vascular myogenic tone in placental ischemic rats.

Figure 19

This figure illustrates that the relationship between middle cerebral artery tone and pressure is blunted in reduced uterine perfusion pressure (RUPP) rats compared to nonpregnant and normal pregnant rats. Data are presented as mean ± SEM. Adapted from Ryan MJ, et al., 2011 (291).

While placental ischemia leads to a number of cerebrovascular abnormalities that are also seen in women with PE, the various factors linking placental ischemia and cerebrovascular dysfunction are not fully known. Recent studies have implicated AT1-AA and TNF-α, which are both elevated in PE women and in the RUPP model. Warrington et al. (364) reported that the AT1 receptor plays a role in the development of hypertension and impaired cerebral blood flow autoregulatory function in placental ischemic rats. In addition, the AT1-AA impaired cerebral blood flow autoregulation in pregnancy. These findings indicate that activation of the AT1 receptor, in part due to circulating AT1-AA, may be involved in the cerebral vasculature dysfunction in PE. In another study, Warrington and colleagues showed that TNF-α infusion into pregnant rats, to levels reported in women with PE, increases brain water content, and that TNF-α blockade with Etanercept reduced brain water content and BBB permeability (363). Furthermore, a recent study by Duncan et al. (89) demonstrated that excess TNF-α in once normal pregnant rats also reduces cerebrovascular βENaC expression and impairs cerebral blood flow regulation, suggesting that TNF-α may contribute to cerebral edema, increased BBB permeability, and cerebral blood flow regulation. Taken together, these studies indicate that placental factors, particularly AT1-AA and TNF-α, stimulate degenerin-mediated vascular myogenic constriction and cerebral blood flow regulation.

The heart in PE

Pregnancy presents a significant challenge to cardiac function during the course of gestation, with cardiac output increasing 40% to 50% to meet the metabolic demands and support growth of the fetal-placental unit. Cardiac hypertrophy and associated increases in vascular density occur during normal pregnancy (55) and typically reverses within 7 days postpartum. Conversely, PE, particularly with low birth weight offspring, is typically associated with pathological hypertrophy, reduced ejection fraction, and reduced cardiac output. Numerous studies now highlight the prevalence of cardiac dysfunction in this cohort (83, 129, 221, 302, 309). Cardiac dysfunction in PE was historically thought to be the sole consequence of the hemodynamic forces of hypertension. However, recent studies have indicated a more severe cardiac phenotype in PE compared to gestational hypertension (302), suggesting that cardiac dysfunction in PE could be a result of circulating placental factors and occurs independently of hypertension (Figure 20).

Figure 20. Global longitudinal strain in humans and placental ischemic rats.

Figure 20

This figure illustrates the development of cardiac global longitudinal strain (GLS) in humans and in the reduced uterine perfusion pressure (RUPP) rat model of preeclampsia (PE). Importantly, Panel (A) also shows significantly reduced GLS in PE compared to nonproteinuric hypertension (gestational hypertension), suggesting more than hypertension (potentially placental factors) plays a role. In animal models, cardiac dysfunction typically only emerges after 2 weeks in hypertensive models; therefore, data in the RUPP rat in Panel (B) also suggests that placental factors could play a role in the development of cardiac dysfunction in PE. The lower (25th percentile) and upper (75th percentile) limits of each box represent the interquartile range; the line across the box indicates the median. The lowest and highest values within 1.5 interquartile ranges below and above the interquartile range are indicated by the whiskers. Dots represent values that lie outside this range. *P < 0.05. Adapted from Shahul S, et al., 2012 (302) and Bakrania BA, et al., 2019 (34).

Diastolic dysfunction, based on predefined professional recommendations, occurs significantly more frequently in PE patients compared to gestational hypertensive pregnancies (220, 339). Systolic dysfunction, measured by global longitudinal strain (GLS), or deformation of the heart, has been reported to be reduced despite preserved ejection fraction (302). Incidentally, GLS is a sensitive measure that can detect subtle and subclinical cardiac dysfunction. Decreased GLS indicates stiffening of the ventricle and fibrosis and is therefore associated with age or disease. Similarly, the Tei Index, which is the ratio between contraction and relaxation time intervals, also typically increases under similar circumstances and is increased in PE women (45). A number of studies have investigated whether cardiac abnormalities are related to severity of PE. In a study of 192 patients, women with prepregnancy cardiac remodeling as a result of chronic hypertension were more likely to develop cardiac complications during pregnancy (350). Another study reported that eccentric ventricular hypertrophy, which is typically associated with physiological remodeling and exercise, was most associated with adverse outcomes such as admission to neonatal intensive care unit, low birth weight, and use of ventilator for the first 24 h (21). Similar to previous publications, ejection fraction was preserved in these patients, suggesting that current methods of monitoring cardiac dysfunction in PE using crude measures such as ejection fraction may not be conducive in this cohort.

The development of cardiac dysfunction in PE is not well understood; however, placental ischemic factors have been implicated. Animal studies show that the RUPP rat exhibits reduced ejection fraction and GLS during pregnancy (Figure 17) as well as apoptosis, histological measures of fibrosis, and reductions in vascular density (34, 131). Importantly, cardiac dysfunction only develops in animal models of hypertension after 2 weeks; however, these changes in the RUPP rat are observed after only 5 days of placental ischemia, suggesting that placental factors could be contributing to these cardiac abnormalities. The antiangiogenic properties of sFlt-1 could impact cardiac function in PE. Overexpression of sFlt-1 has been shown to induce cardiac dysfunction in mice. Mice with knockout of PCG-1α, which is a pro-angiogenic transcription factor, exhibit systolic dysfunction within 3 weeks (258), suggesting that an antiangiogenic environment is sufficient to induce cardiac dysfunction, independent of elevated blood pressure. Moreover, clinical studies show that the impaired cardiac angiogenesis compares to the levels of circulating sFlt-1.

Circulating Activin A, a profibrotic factor and member of the transforming growth factor TGF-β family, is produced by the placenta, is significantly increased during pregnancy, and correlates with cardiac dysfunction in PE (301). Shahul et al. showed that levels of Activin A with clinical characteristics of PE were the most accurate predictor of abnormal GLS, suggesting that Activin A has a direct role in mediating cardiac dysfunction in PE. Activin A infusion into mice also results in significant impairment to cardiac function (289). Despite previous theories that cardiac dysfunction is a consequence of chronic hypertension, these data suggest that placental factors could have a direct impact on the heart in PE.

Fetal Outcomes

A number of circumstances during pregnancy, such as maternal undernutrition, alcohol consumption, gestational hypertension, maternal obesity, preexisting cardiovascular disease, and PE, can impact fetal outcomes during childhood and adulthood (Figure 21). Pregnancies complicated with hypertension are at the highest overall risk of fetal mortality (134). Of those, PE is the leading cause of perinatal morbidity and mortality. As the only current therapy for PE is delivery of the fetus and placenta, premature birth is particularly common in this disease, which poses severe and long-term risks for the baby. PE is also associated with IUGR and small for gestational age (SGA) births. In addition to the immediate risks of underdevelopment, more recent studies have shown significantly increased risk for the development of cardiovascular disease during child- and adulthood in SGA offspring. In 1990, British epidemiologist Dr. David Barker postulated that the high prevalence of low birth weight babies and subsequent disease during adulthood in the same geographical area was due to the programming of disease during fetal life (38). What is now commonly referred to as the Barker Hypothesis, or the developmental origins of fetal programming, has since been widely studied and considered a critical time frame for therapeutic intervention to improve long-term outcomes on fetal outcomes (15).

Figure 21. Factors involved in long-term cardiovascular risk in offspring from preeclamptic pregnancies.

Figure 21

This figure illustrates how fetal programming leads to increased risk of cardiovascular, metabolic, and neurological disease in infants and adults born from preeclamptic pregnancies. SNS, sympathetic nervous system; RAAS, renin angiotensin aldosterone system. Adapted from Alexander BT, et al., 2015 (15).

Impact of the prenatal environment on fetal outcomes

Placental insufficiency, such as is present in PE, is thought to play a central role in programing cardiovascular disease in offspring. This insufficiency occurs when the placenta cannot appropriately deliver amino acids, oxygen, and nutrients to the growing fetus. In the case of PE, this is due to impaired spiral artery remodeling, which leads to poor perfusion of the placenta and placental ischemia. Although it is widely accepted that placental insufficiency is an initiating event in the programing of disorder, the exact mechanisms are not completely understood. Barker postulated a “brain-sparing” effect where, in the presence of restricted nutrients, blood flow is redistributed away from other organs as an adaptive mechanism for survival. As a result, other major organs remain underdeveloped, resulting in changes to structure and physiology, thereby ultimately “programming” the development of cardiovascular risk and disease. The brain-sparing effect can be observed by enlarged cranial circumference compared to body circumference in 70% to 80% of offspring. Hemodynamically, brain sparing can be detected by reduced middle cerebral artery resistance in the offspring in utero, allowing for increased perfusion. While this compensatory mechanism most likely increases survival, it does not improve the long-term cardiovascular and metabolic risk in later life (103, 140). It is important to note that not all preclinical studies where cardiovascular risk is increased in offspring are associated with maternal hypertension (as is present in PE), whereas this risk is consistently found in models of placental insufficiency, suggesting it is placenta insufficiency in PE that is the key factor in the programming of long-term cardiovascular disease in these offspring.

Postnatal and long-term outcomes for offspring

PE is most notably associated with IUGR, which leads to low birth weight and development of cardiovascular, metabolic, and neurological diseases (Table 5) (201). Some studies in infants born at term suggest that exposure to PE in utero is adequate to lead to these disturbances and are not due solely to preterm delivery (239). Increased risk of cardiovascular disease in offspring exposed to PE has been relatively well established. Studies show a significantly increased risk of elevated blood pressure in childhood through to young adulthood (12, 19, 82, 249). A number of studies have found increased risk of congenital heart disease (CHD) in PE offspring (29, 46, 48, 375), with one study estimating an eightfold greater risk for offspring of PE women. In that study, 30% of infants from PE pregnancies were diagnosed with CHD by day 7 of life and 21% at day 28, as compared to 12% and 3%, respectively, in infants born from healthy pregnancies (375). Some studies have also showed increased prevalence of metabolic disease in children and adults exposed to PE pregnancies, such as insulin insensitivity, obesity, and type 2 diabetes mellitus (19, 160, 300).

Table 5.

Long-Term Risks for Mother and Baby Following a Preeclamptic Pregnancy

Risk factor References
Long term risks for offspring
 Hypertension (12, 19, 82, 249)
 Congenital heart disease (29, 46, 48, 375)
 Insulin sensitivity (19, 300)
 Obesity (19)
 Type 2 diabetes mellitus (19, 160)
 Autism spectrum disorder (55, 207, 356)
 Attention deficit/hyperactivity disorder (133, 308)
 Epilepsy (72, 90, 130, 238)
 Stroke (72, 90, 130, 238)
 Depression (344)
Long term risks for mother
 Hypertension (43, 338)
 Ischemic heart disease (43)
 Cardiac dysfunction (220, 301)
 Heart failure (190, 229, 371)
 Coronary heart disease (190, 371)
 Chronic kidney disease (199, 214, 241)
 Stroke (43, 190, 338)
 Cognitive impairment (378)
 Alzheimer’s disease (338)

Despite the brain-sparing effect observed in pregnancies with placental insufficiency, several preclinical and clinical studies have reported neurodevelopmental impairment in offspring. Clinical studies have demonstrated increased autism spectrum disorder and attention deficit/hyperactivity disorder, as well as cognitive impairments, such as reduced academic performance. In addition, infants born from PE pregnancies are at an increased risk of developing neurological diseases such as epilepsy and stroke (72, 90, 130, 238). In another study, adults from PE pregnancies exhibited much greater tendencies for depressive symptoms (344).

The RUPP model has been useful in understanding some of the cardiovascular aberrations that occur in infants from PE pregnancies. Interestingly, male offspring from RUPP pregnancies develop hypertension, similar to the human population. Female offspring exhibit age-dependent increases in blood pressure, which can be attenuated by androgen receptor blockade or estradiol supplementation in association with reduced AT1a receptor activity, implicating androgen-mediated activation of the AT1 receptor in the development of PE-induced hypertension in offspring (7678). Studies have shown that renal oxidative stress, renal expression of renin, angiotensinogen, and angiotensin-converting enzyme activity, are increased in male IUGR offspring. Furthermore, blockade of the renin angiotensin system (126), renal denervation (251), or chronic antioxidant administration (250) attenuates the development of hypertension in RUPP offspring.

Long-term Risk for Mother

In 2011, the American Heart Association released guidelines for the prevention of cardiovascular disease and recognized PE as a risk factor. Indeed, several studies now highlight the profound long-term effect of a PE pregnancy on the mother, and in doing so, negate theories of this being a disease that is cured at delivery (Table 5). A meta-analysis of over 20 studies determined that the long-term risk of developing hypertension and ischemic heart disease is increased four- and twofold, respectively (43). In addition, the strong association between PE and subsequent risk for hypertension could potentially account for incidence of chronic kidney disease in formerly PE patients in the long term, as they are both known risk factors for renal injury.

While some studies have found little association between previous PE and later chronic kidney disease, others reported strong associations, with up to ninefold increases in risk of chronic kidney disease (199, 214, 241). Little is known with regard to the underpinning causes of long-term renal disease in PE, but a number of animal models have been reported to have sustained decline in renal function. Paauw et al. (254) reported that placental ischemia in pregnant rats is associated with a decline in renal function and persistent cardiac dysfunction 8 weeks postpartum that was independent of blood pressure. Turbeville et al. (345) showed in a model of superimposed PE that pregnancy exaggerates postpartum renal damage despite no difference in blood pressure long term. Utilization of these animal models may provide a better understanding of the mechanisms linking PE and long-term chronic kidney disease as well as could help in developing preventive, diagnostic, and therapeutic strategies for women who develop chronic kidney disease after a preeclamptic pregnancy.

As seizures during pregnancy are a severe complication of PE, it is not surprising that this disease has long-term implications on the brain. Studies showed that the long-term consequences of cerebrovascular impairment include persistent white matter lesions and chronic cognitive abnormalities in mothers with a history of PE (378). White matter lesions are thought to be a result of posterior reversible encephalopathy syndrome, where edema is thought to compress the cerebral tissue causing ischemia and cell death (379). PE itself is a prominent risk factor for long-term cerebrovascular disease risk, but whether white matter lesions increase this risk further is not clear. It is important to note that not all women who experience later cerebrovascular damage experienced seizures during pregnancy. However, white matter lesions are particularly associated with early-onset PE (30).

Mounting evidence indicates that cardiac dysfunction is present in women long after delivery. In a meta-analysis of 22 studies and 6.4 million women, PE was associated with a fourfold increase in the development of postpartum heart failure after pregnancy and a twofold increased risk of coronary heart disease, stroke, and death due to cardiovascular disease (371). Another study showed persistent diastolic and systolic dysfunction 12 months following parturition in the absence of hypertension (220). The same study reported that PE patients were more likely to develop essential hypertension within 2 years of giving birth (220). A 2019 study of 149,712 PE patients observed 18,624 first-time cardiovascular events including heart failure, stroke, and cardiovascular deaths over a 9-year period after PE pregnancy (190). A recent report highlighting the high rate of pregnancy-related heart failure admissions also showed that postpartum heart failure hospitalizations increased by 7% between 2001 and 2006, which have since continued to steadily rise (229). The same report showed that, in over 50 million hospitalizations, almost 40% of women were also patients diagnosed with PE or superimposed PE. Although a number of placental factors have been implicated in the development of cardiac dysfunction during PE, Activin A is the only factor that has been studied to date and found to correlate with long-term systolic dysfunction (301). These large cohort studies suggest that cardiac dysfunction is independent of hypertension during PE, and that elevated circulating placental factors may have a long-term impact on the heart.

While the direct cause of long-term disease in women with a history of PE is not well understood, some factors have been shown to remain increased in this cohort, despite the fact that the placenta is removed and thus can no longer contribute to the circulating milieu. Ang II, AT1-AA, and Activin A have all been shown to remain increased in women with a history of PE (149, 301, 324), suggesting that these factors continue to act on the maternal endothelium and organ systems years after delivery. In addition, a second PE pregnancy has been shown to further increase risk of cardiovascular disease. Some theories of long-term cardiovascular disease in PE are centered around the concept that pregnancy is a physiological “stress test” and that the subsequent cardiovascular disease that is seen in these women was simply dormant or subclinical previously. Nevertheless, this is unlikely because the studies discussed above suggest that new damage occurs during a PE pregnancy.

Clinical Management of PE

Predicting the development of PE

Many studies have attempted to predict the onset of PE (245). There is no single test that can definitively identify women who will develop this disorder; however, measurement of sFlt-1/PlGF ratio has been extensively studied with promising results. Currently, the most accurate predictive measurement of early-onset PE is the sFlt-1/PlGF ratio accompanied by uterine artery Doppler assessment (307). The PROGNOSIS (Prediction of Short-term Outcome in Pregnant Women with Suspected Preeclampsia Study) trial found that women with an sFlt-1/PlGF ratio ≤38 did not develop PE in the subsequent week (380). Furthermore, repeated measurements of this ratio improved risk stratification and could rule out onset of PE for up to 4 weeks (381). A recent article reported that aspirin treatment was effective in reducing the sFlt-1/PlGF ratio in women who were considered to be at risk of developing PE during their first trimester (213).

While efforts to predict PE could improve management of this disease, there is still no effective intervention to prevent its onset. However, a number of micronutrients, particularly those that have been shown to be depleted in PE women, have been studied in this area of research. Selenium, an essential micronutrient, represents a promising candidate, particularly because it is a nonpharmacological option and can be administered as a dietary supplement. It has been shown to improve mitochondrial function, in vitro (171, 269), and has been associated with reduced incidence of PE in a number of studies (262, 372). Other micronutrients, such as magnesium, which is an important antioxidant coefficient, had little to no effect in women at high or low risk of developing PE (80, 81). This area of research requires further investigation and highlights the importance of basic research for the development of appropriate therapeutic options.

Current therapeutic avenues

Currently, treatment for PE is limited to managing symptoms and in severe cases premature delivery of the fetus, which poses significant risk to both mother and baby. Aspirin therapy in high-risk women, betamethasone (<34 weeks), and magnesium sulfate in women at risk for developing eclampsia are the only medications currently administered for management of this disease. In addition, blood pressure monitoring is important both during pregnancy and following delivery, in case the patient develops de novo postpartum PE (276). Antihypertensive therapy is carefully regulated during pregnancy. Based on ACOG guidelines, mild to moderate hypertension (<160/110 mmHg) should not be treated pharmacologically, because there is no evidence that reducing hypertension will improve disease progression or outcomes. In circumstances where antihypertensives must be administered, the recommended therapy is labetalol (beta blocker), nifedipine (calcium channel blocker), or methyldopa (adrenergic receptor blocker) (282).

Despite tremendous advances in understanding the pathophysiology of PE, therapeutic options remain limited. One of the primary reasons for this is the posed risk of teratogenic effects on the fetus. In fact, this further highlights the necessary use of animal models to study PE and potential therapeutic targets. A summary of preclinical and clinical trials that will be discussed in this section can be found in Table 6.

Table 6.

Summary of Clinical Studies Being Researched for the Treatment of Preeclampsia

Therapy Preclinical studies in animal models of PE Clinical trials
NO precursors/donors ↓ MAP, ↔ pup weight (17) ↔ MAP, trend toward reduced incidence of PE and neonatal complications with L-arginine treatment (244)
↓ MAP, ↓ prepro-ET-1 (237) ↓ MAP, ↓ umbilical artery PI, ↔ uterine artery PI (128)
↓ MAP, ↓ proteinuria, ↑ fetal weight, ↓ sFlt-1, ↑ VEGF (253) ↓ MAP, ↓ uterine artery PI, ↓ umbilical artery PI ↔ fetal heart rate, ↔ fetal MCA PI (202)
↓ Diastolic blood pressure, ↔ fetal heart rate (209)
Sildenafil ↓ MAP, ↔ sFlt-1, ↔ pup weight (114) STRIDER trial (260):
↓ MAP, ↓ proteinuria, ↓ kidney injury, ↓ TNF-α, ↓ ET-1, ↓ UARI, ↑ pup weight (118) ↔ Angiogenic factors, ↔ fetal death, ↔ birth weight, ↔ ability to prolong pregnancy, any adverse effects not attributed to Sildenafil (303)
↓ sFlt-1, ↓ sEng (275) ↔ Fetal outcomes, no adverse outcomes (127).
↓ MAP in offspring (335)
PlGF administration ↓ MAP, ↓ sFlt-1, ↓ oxidative stress, ↑ GFR, ↔ pup weight (323)
↓ MAP, ↓ proteinuria, ↓ sFlt-1 mRNA, ↔ sFlt-1 (206)
sFlt-1 removal ↓ sFlt-1, transient ↓ MAP, transient ↓ proteinuria (337)
↓ sFlt-1, transient ↓ MAP, ↓ proteinuria, gestations prolonged by 8 or 15 days following 1 or multiple treatments, respectively, no adverse effects (336)
Statins ↓ MAP, ↓ sFlt-1, ↑ VEGF, ↑ pup weight, ↓ oxidative stress (42) StAmP trial:
↓ MAP in dams and offspring, ↓ sFt-1, ↑ PlGF, ↓ cardiac fibrosis and hypertrophy during pregnancy and postpartum, improved endothelial function in dams and offspring, ↓ ET-1, ↓ oxidative stress, ↑ NO, ↓ glomerular injury during pregnancy and postpartum, ↓ IL-6 and TNF-α (108) ↔ Fetal cholesterol levels, no adverse effects (69)
Activation of antiapoptotic pathway (292) ↔ sFlt-1, gestation prolonged by 4 days, no adverse effects related to pravastatin (8)
↑ Cardiac output postpartum, ↓ cardiac fibrosis postpartum, ↓ cardiac hypertrophy postpartum (179)
Vitamin D ↓ MAP, ↓ sFlt-1, ↓ IL-6, ↔ TNF-α, ↓ AT1-AA, ↓ PPET-1 (100) Meta-analysis of 30 clinical trials—little to no effect to treat PE and IUGR overall (256)
↓ MAP, ↓ proteinuria, ↓ oxidative stress, ↓ ER stress, ↓ apoptosis, ↑ pup weight, ↓ renal injury, ↓ cardiac remodeling (341)
↓ sFlt-1, ↓ TNF-α, ↑ VEGF, ↓ oxidative stress (315)
↓ MAP, ↓ CD4+ T cells, (74)
Multimineral-vitamin D reduced incidence of PE (32)
Vitamin B ↓ sFlt-1, ↓ proteinuria, prolongs gestation, ↑ pup weight (54)
↓ MAP, ↓ proteinuria, ↓ glomerular injury, ↑ pup weight, ↓ HIF1α (105)
HO-1 ↓ MAP, ↑ VEGF, ↔ pup weight, ↓ oxidative stress, ↔ PPET-1 (112)
↓ MAP, ↔ sFlt-1, ↔ VEGF, ↔ pup weight, ↔ TNF-α (115)
↓ MAP, ↑ VEGF, ↔ sFLt-1, ↓ PPET-1 (111)
ETA antagonist ↓ MAP (11, 182, 236)
↓ MAP, ↓ UARI (329)

PE, preeclampsia; ↑, increase; ↓, decrease; ↔, no change; MAP, mean arterial pressure; PI, pulsatility index; MCA, middle cerebral artery; TNF-α, tumor necrosis factor-α; ET-1, endothelin-1; UARI, uterine artery resistive index; sEng, soluble endoglin; STRIDER, Sildenafil TheRapy In Dismal prognosis Early-onset fetal growth Restriction; GFR, glomerular filtration rate; VEGF, vascular endothelial growth factor; PlGF, placental growth factor; NO, nitric oxide; IL-6, interleukin-6; StAmP, Statins to Ameliorate Preeclampsia; AT1-AA, angiotensin II type 1 receptor autoantibody; PPET-1, prepro-endothelin-1; ER, endoplasmic reticulum; CD4+, cluster of differentiation 4 positive; IUGR, intrauterine growth restriction; HIF1 α, hypoxia-inducible factor 1α.

The NO-sGC-cGMP signaling pathway has garnered much interest in the treatment of PE. NO donors and precursors stimulate this pathway and are a well-established therapy for cardiovascular disease. For PE, NO donors/precursors have been successful in preclinical models (17, 237, 253) but yield mixed findings in clinical trials (128, 158, 202, 209, 244). These studies involve administration of L-arginine (an NO precursor) or a molecule that will release, or “donate,” NO molecules. Preclinical studies in the RUPP (17), sFlt1-infusion (237), and L-NAME model (253), where NO synthesis is suppressed, found that MAP is reduced and angiogenic balance is improved in response to l-arginine. In some clinical studies, MAP and maternal vascular resistance are improved, but most show minimal improvement in parameters for infants (128, 202, 244). One explanation for these findings is that the high levels of ROS that are present in PE may result in oxidation of the exogenous NO to produce peroxynitrite and thus have minimal effect on the downstream vasoprotective pathways such as cGMP signaling.

More recent studies focused on blocking cGMP degradation as a mechanism to sustain NO-pathway activity. A number of preclinical studies showed profound improvements to MAP and other characteristics in the mother, including increased pup weight in response to Sildenafil administration (a phosphodiesterase type 5 inhibitor) (114, 118, 275, 335). It is important to acknowledge that in preclinical studies in animal models, increases in pup weight is a crude measure that indicates improved flow and implies better fetal outcomes, but this can only truly be determined through long-term studies in the offspring. In contrast to the findings of preclinical studies, recent clinical trials did not show any benefit, including prolonging gestation or improving IUGR. A 2017 randomized trial for Sildenafil was halted following mortality in 11 offspring due to pulmonary disease. It is important to note that these deaths were not directly attributed to Sildenafil (127, 303). A novel class of drug, sGC stimulators and activators, increases cGMP by promoting sGC activity. In addition to reducing bioavailable NO, ROS can oxidize sGC and inhibit its function, leading to further downregulation of this pathway. In the RUPP rat, sGC activators significantly reduce MAP and endothelial function by increasing cGMP production (36).

Another important avenue to attenuate the clinical manifestation of PE is to improve angiogenic imbalance. One possibility could be to administer PlGF, which has been successful in preclinical models (206, 323). Another method could be to reduce circulating sFlt-1. Thadhani et al. showed that removal of sFlt-1 from serum using an apheresis device delayed delivery by an average of 8 days with a single treatment and a maximum of 21 days (range 11–21) with multiple treatments. This is compared to induction of delivery by 3 to 4 days in the control group. Interestingly, following each apheresis treatment, a reduction in sFlt-1 was associated with a transient decrease in blood pressure, which was followed by a gradual increase as circulating sFlt-1 levels rose again likely due to the ischemic placenta continuing to secrete this factor. Still, these findings represent a hopeful avenue for the treatment of PE (336).

Statins are a commonly prescribed therapy for lowering cholesterol in cardiovascular disease. In addition to this well-established effect, statins have also been shown to have anti-inflammatory properties and protect endothelial function (247). Numerous studies in rodent models of PE have demonstrated the potential beneficial effects of pravastatin therapy in PE. Garrett et al. (108) found that pravastatin improved placental flow and reduced placental insufficiency, resulting in decreased release of vasoactive factors, and in fact, protected both mother and offspring from long-term adverse cardiovascular outcomes. Another study in a mouse model of PE showed that pravastatin activates antiapoptotic pathways in the placenta (292). In RUPP rats treated with pravastatin, MAP and oxidative stress were reduced and angiogenic balance was improved (42). Interestingly, one study reported that statin treatment in another rat model of PE led to amelioration of cardiac fibrosis and capillary rarefaction and improved cardiac function 28 days after pregnancy. Animals in this study were treated during pregnancy (gestational day 15) and postpartum (28 days postpartum) (179). Currently, statins are not recommended during pregnancy; however, a pilot clinical study to test safety and pharmacokinetics did not report any adverse effects in the mother or offspring (69). This pilot study did not report any cholesterol-lowering effects in the fetus. A randomized trial of early-onset PE patients did not find any evidence that pravastatin reduced plasma sFlt-1 or length of gestation (8). As neither of these studies found any adverse effects attributed to pravastatin, this drug continues to be intensively researched.

Several studies have shown that some vitamins are deficient in PE women and have anti-inflammatory and antioxidant properties (33). The RUPP model has been used to examine vitamin D2 and vitamin D3 and demonstrated that both were successful in reducing blood pressure, Th-17 cells, ET-1, sFlt-1, and AT1-AA; however, these vitamins did not have any effect to improve fetal or placental weight (74, 100, 341). In another model of PE that is induced by blocking NO production, vitamin D supplementation reduced TNF-α and sFlt-1 (315). Vitamin B3 (nicotinamide) also has anti-inflammatory properties and significantly reduces MAP, oxidative stress, glomerular endotheliosis, and fetal growth restriction in numerous mouse models of PE (54, 105). Clinically, vitamin supplementation to treat PE has had mixed findings. There are studies suggesting that multimineral-vitamin D during pregnancy can reduce the risk of PE (32). In contrast, one meta-analysis of 30 clinical trials and 7,289 women reported that, while vitamin D supplementation above the current recommendations during pregnancy is safe and may reduce risk of gestational diabetes, overall, it has little to no effect on the risk of PE and IUGR (256).

In addition to the above therapies that have reached clinical trials, a number of interventions have been tested in preclinical models with promising results. ETA antagonism has been tested in rat models of PE and is associated with decreases in MAP and improvement in the angiogenic balance (11, 182, 236). Progesterone is a hormone that typically increases over the course of normal pregnancy and is produced primarily by the placenta. In PE, levels of progesterone do not increase at the same rate, and in fact, its administration is used to prevent spontaneous preterm birth (223). In the RUPP model, progesterone treatment for only 24 h using 17-α-hydroxyprogesterone caproate improves hypertension and suppresses inflammation while improving uterine artery blood flow and litter size (20, 351). Another molecule that has been shown to be reduced in PE and has anti-inflammatory, antiapoptotic, and angiogenic properties is heme-oxygenase-1 (HO-1). RUPP rats treated with CoPP (a known HO-1 inducer) have attenuated hypertension and reduced endothelial dysfunction (112). Further studies in these animals suggested that HO-1 improves cytotrophoblast migration by increasing βENaC expression (362), and that CoPP has a direct effect in blocking TNF-α-induced ET-1 production from glomerular endothelial cells (35). HO-1 treatment also has similar effects in sFlt-1- and TNF-α-infusion models of PE (111, 115). During the development and pathogenesis of PE, both mother and baby are exposed to high levels of oxidative stress where the HO-1 pathway could also be protective. Hypoxia modifies phosphorylation of proteins that regulate survival and injury. Indeed, one study showed that HO-1 treatment following placental ischemia promotes prosurvival responses (110) through activation of ERK and the MAPK pathway. While these interventions have not yet reached clinical trials, the success of preclinical studies, such as those described above, remains invaluable in the search for an appropriate treatment for PE.

Conclusions

PE is a complex, multifaceted disease during pregnancy. It develops in two phases (1) impaired cytotrophoblast migration and uterine invasion causing deficient perfusion of the placenta and (2) the release of factors from the ischemic placenta into the maternal circulation, leading to endothelial and vasculature dysfunction and the maternal clinical syndrome (Figure 22). Throughout this article, we have discussed the many factors and pathways playing a critical role in the development of PE. Specifically, antiangiogenic factors and pro-inflammatory cytokines are at the center of PE pathogenesis in phase 2 by inducing endothelial dysfunction. Endothelial dysfunction is associated with reduced NO availability and upregulation of the endothelin system. The brain, kidneys, liver, and heart are particularly vulnerable to the circulating placental factors associated with PE. Moreover, some adverse effects during PE have been shown to persist postpartum. Thus, PE also poses significant risk for long-term diseases in both mother and her baby. Notably, women and infants born from PE pregnancies are at increased odds for cardiovascular, renal, cerebrovascular, and/or neurological diseases. It is thought that the adverse environment during preeclamptic pregnancies impacts these offspring well into adulthood (50+ years). These findings highlight the need for novel cures to treat the immediate and chronic effects of this maternal disorder. Currently, pharmacological treatment for PE is limited to managing symptoms through antihypertensives, betametasome, and magnesium sulfate. Preclinical studies in animal models have been instrumental not only in understanding the pathophysiology of PE but also in the search for novel therapeutic options for the treatment of this disease. These studies have shown that blockade of excessive placental ischemic factors, such as the antiangiogenic factor, sFlt-1, can improve maternal outcomes. Attenuation of the endothelin system and stimulation of the NO-sGC-cGMP pathway are among other targets that could be important in the treatment of PE. In a clinical trial, clearance of the antiangiogenic factor, sFlt-1, increased length of gestation, substantiating additional larger trials. Basic science and clinical studies investigating the treatment of PE remain an active and promising area of research.

Figure 22. Schematic of the pathogenesis of preeclampsia.

Figure 22

This figure illustrates the pathways involved in the pathophysiology of preeclampsia (PE), from abnormal placentation to the development of hypertension and end-organ dysfunction and injury. Phase 1 of the pathogenesis of PE involves impaired and shallow cytotrophoblast invasion, resulting in impaired spiral artery remodeling, poor placental perfusion, and placental ischemia. Phase 2 is clinical manifestation, which presents >20 weeks of gestation. As a result of placental ischemia, several factors are released into the maternal circulation causing widespread endothelial dysfunction, hypertension, and multiorgan dysfunction. It is important to note that hypertension is the only symptom that occurs in every incidence of PE and is typically coupled with other disturbances/organ dysfunction. NF-κB, nuclear factor-kappa B; Th, T helper cell; NK, natural killer cell; dNK, decidual natural killer cell; HLA-C, human leukocyte antigen-C; MMP, matrix metalloproteinases; C3, complement 3; SNPs, single nucleotide polymorphisms; FLT-1, fms-like tyrosine kinase-1; sFlt-1, soluble Flt-1; sEng, soluble endoglin; VEGF, vascular endothelial growth factor; PlGF, placental growth factor; ROS, reactive oxygen species; AT1-AA, angiotensin II Type 1 receptor autoantibody; Tregs, T regulatory cells; IL, interleukin; TNF-α, tumor necrosis factor-α; ET, endothelin; NO, nitric oxide; IUGR, intrauterine growth restriction.

Didactic Synopsis.

Major teaching points

  • Incidence, diagnosis criteria, and risk factors for pre-eclampsia (PE)
    • Major contributor of maternal and fetal morbidity and mortality
    • New-onset hypertension with new onset of one or more other features
  • Understand the pathophysiology of PE
    • Phase 1: Shallow and reduced cytotrophoblast invasion, resulting in impaired uterine spiral artery remodeling and placental ischemia
    • Phase 2: Clinical manifestations resulting from release of placental factors into the maternal circulation
  • PE is associated with multiorgan dysfunction and injury

  • Pathologies in offspring from preeclamptic pregnancies
    • Intrauterine growth restriction
    • Increased risk of cardiovascular, metabolic, and neurological diseases
  • Risk of cardiovascular disease is significantly increased in women with a history of PE

  • There is currently no definitive treatment to cure PE other than early delivery of baby and importantly, the placenta

  • Animal models are instrumental in understanding mechanisms of pathophysiology and therapeutic options for PE

Acknowledgements

This work was supported by the American Association under grant number 18POST33990293 (BAB) and the National Institutes of Health under grant numbers P01HL051971 (BAB, JPG), P20GM104357 (BAB, JPG), R00HL130577 (FTS), P20GM121334 (FTS, BBL, HAD), R01HD067541 (BBL), R01HL136684 (HAD, MJR, JPG), T32HL105324 (JPG), and U54GM115428 (JPG).

References

  • 1.Aban M, Cinel L, Arslan M, Dilek U, Kaplanoglu M, Arpaci R, Dilek S. Expression of nuclear factor-kappa B and placental apoptosis in pregnancies complicated with intrauterine growth restriction and preeclampsia: An immunohistochemical study. Tohoku J Exp Med 204: 195–202, 2004. [DOI] [PubMed] [Google Scholar]
  • 2.Abboud TK, Artal R, Henriksen EH, Earl S, Kammula RK. Effects of spinal anesthesia on maternal circulating catecholamines. Am J Obstet Gynecol 142: 252–254, 1982. [DOI] [PubMed] [Google Scholar]
  • 3.Abdalvand A, Morton JS, Bourque SL, Quon AL, Davidge ST. Matrix metalloproteinase enhances big-endothelin-1 constriction in mesenteric vessels of pregnant rats with reduced uterine blood flow. Hypertension 61: 488–493, 2013. [DOI] [PubMed] [Google Scholar]
  • 4.Abitbol MM, Gallo GR, Pirani CL, Ober WB. Production of experimental toxemia in the pregnant rabbit. Am J Obstet Gynecol 124: 460–470, 1976. [DOI] [PubMed] [Google Scholar]
  • 5.Abram SR, Alexander BT, Bennett WA, Granger JP. Role of neuronal nitric oxide synthase in mediating renal hemodynamic changes during pregnancy. Am J Phys Regul Integr Comp Phys 281: R1390–R1393, 2001. [DOI] [PubMed] [Google Scholar]
  • 6.Abramovici D, Friedman SA, Mercer BM, Audibert F, Kao L, Sibai BM. Neonatal outcome in severe preeclampsia at 24 to 36 weeks’ gestation: Does the HELLP (hemolysis, elevated liver enzymes, and low platelet count) syndrome matter? Am J Obstet Gynecol 180: 221–225, 1999. [DOI] [PubMed] [Google Scholar]
  • 7.Aggarwal PK, Chandel N, Jain V, Jha V. The relationship between circulating endothelin-1, soluble fms-like tyrosine kinase-1 and soluble endoglin in preeclampsia. J Hum Hypertens 26: 236–241, 2012. [DOI] [PubMed] [Google Scholar]
  • 8.Ahmed A, Williams DJ, Cheed V, Middleton LJ, Ahmad S, Wang K, Vince AT, Hewett P, Spencer K, Khan KS, Daniels JP, Barber K, Kilby M, Knox E, Sellman T, Trinham P, Tuffnell D, Jones V, Syson J, Shah N, Deeks L, Carter W, Dorman E, Thomas S, Harrington D, Higgins N, Wilmott-Powell M, Simpson N, Dolby V, Bricker L, Walkinshaw S, Houghton G, Longworth H, Williamson C, Dhanjal M, Noori M, Machirori M, Howard R, Murray R, Weist S, Denison F, Crawford I, Robson S, Allan C, Myers J, Bernatavicius G, Moorhead L, Chappell L, Nelson-Piercy C, Williams D, Daley R, Rosas M, Greer I, Vitek L, Shennan A, Marlow N, Barnard AM, Thornton J, Rennie J, Peacock J, Gao Smith F, Hyde C, Crawford I, Cudmore M, Furmston A, Fulcher L, Homer L, Howman A, Hilken N, Brown S. Pravastatin for early-onset pre-eclampsia: A randomised, blinded, placebo-controlled trial. BJOG 127: 478–488, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Ajne G, Wolff K, Fyhrquist F, Carlström K, Hemsén-Mörtberg A, Nisell H. Endothelin converting enzyme (ECE) activity in normal pregnancy and preeclampsia. Hypertens Pregnancy 22: 215–224, 2003. [DOI] [PubMed] [Google Scholar]
  • 10.Albrecht ED, Bonagura TW, Burleigh DW, Enders AC, Aberdeen GW, Pepe GJ. Suppression of extravillous trophoblast invasion of uterine spiral arteries by estrogen during early baboon pregnancy. Placenta 27: 483–490, 2006. [DOI] [PubMed] [Google Scholar]
  • 11.Alexander B, Rinewalt A, Cockrell K, Massey M, Bennett W, Granger J. Endothelin type a receptor blockade attenuates the hypertension in response to chronic reductions in uterine perfusion pressure. Hypertension 37: 485–489, 2001. [DOI] [PubMed] [Google Scholar]
  • 12.Alexander BT. The impact of nutritional insults during fetal life on blood pressure. J Nutr Sci Vitaminol 6: S5–S6, 2015. [DOI] [PubMed] [Google Scholar]
  • 13.Alexander BT, Bennett WA, Khalil RA, Granger JP. Preeclampsia: Linking placental ischemia with cardiovascular-renal dysfunction. News Physiol Sci 16: 282–286, 2001. [DOI] [PubMed] [Google Scholar]
  • 14.Alexander BT, Cockrell K, Cline FD, Granger JP. Inducible nitric oxide synthase inhibition attenuates renal hemodynamics during pregnancy. Hypertension 39: 586–590, 2002. [DOI] [PubMed] [Google Scholar]
  • 15.Alexander BT, Dasinger JH, Intapad S. Fetal programming and cardiovascular pathology. Compr Physiol 5: 997–1025, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Alexander BT, Kassab SE, Miller MT, Abram SR, Reckelhoff JF, Bennett WA, Granger JP. Reduced uterine perfusion pressure during pregnancy in the rat is associated with increases in arterial pressure and changes in renal nitric oxide. Hypertension 37: 1191–1195, 2001. [DOI] [PubMed] [Google Scholar]
  • 17.Alexander BT, Llinas MT, Kruckeberg WC, Granger JP. L-Arginine attenuates hypertension in pregnant rats with reduced uterine perfusion pressure. Hypertension 43: 832–836, 2004. [DOI] [PubMed] [Google Scholar]
  • 18.Alioua A, Huggins JP, Rousseau E. PKG-Iα phosphorylates the α-subunit and upregulates reconstituted GK(Ca) channels from tracheal smooth muscle. Am J Phys Lung Cell Mol Phys 268: L1057–L1063, 1995. [DOI] [PubMed] [Google Scholar]
  • 19.Alsnes IV, Vatten LJ, Fraser A, Bjørngaard JH, Rich-Edwards J, Romundstad PR, Åsvold BO. Hypertension in pregnancy and offspring cardiovascular risk in young adulthood: Prospective and sibling studies in the HUNT Study (Nord-Trøndelag Health Study) in Norway. Hypertension 69: 591–598, 2017. [DOI] [PubMed] [Google Scholar]
  • 20.Amaral LM, Cornelius DC, Harmon A, Moseley J, Martin JN, LaMarca B. 17-Hydroxyprogesterone caproate significantly improves clinical characteristics of preeclampsia in the reduced uterine perfusion pressure rat model. Hypertension 65: 225–231, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Ambia AM, Morgan JL, Wells CE, Roberts SW, Sanghavi M, Nelson DB, Cunningham FG. Perinatal outcomes associated with abnormal cardiac remodeling in women with treated chronic hypertension. Am J Obstet Gynecol 218: 519.e1–519.e7, 2018. [DOI] [PubMed] [Google Scholar]
  • 22.Ananth CV, Keyes KM, Wapner RJ. Pre-eclampsia rates in the United States, 1980–2010: Age-period-cohort analysis. BMJ 347: f6564, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Andrew Combs C, Katz MA, Kitzmiller JL. Experimental preeclampsia produced by chronic constriction of the lower aorta: Validation with longitudinal blood pressure measurements in conscious rhesus monkeys. Am J Obstet Gynecol 169: 215–223, 1993. [DOI] [PubMed] [Google Scholar]
  • 24.Ankichetty SP, Chin KJ, Chan VW, Sahajanandan R, Tan H, Grewal A, Perlas A. Regional anesthesia in patients with pregnancy induced hypertension. J Anaesthesiol Clin Pharmacol 29: 435–444, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Aouache R, Biquard L, Vaiman D, Miralles F. Oxidative stress in preeclampsia and placental diseases. Int J Mol Sci 19: 1496, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Armistead B, Kadam L, Drewlo S, Kohan-Ghadr HR. The role of NFκB in healthy and preeclamptic placenta: Trophoblasts in the spotlight. Int J Mol Sci 21, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Ashton SV, Whitley GSJ, Dash PR, Wareing M, Crocker IP, Baker PN, Cartwright JE. Uterine spiral artery remodeling involves endothelial apoptosis induced by extravillous trophoblasts through Fas/FasL interactions. Arterioscler Thromb Vasc Biol 25: 102–108, 2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Atkins AFJ, Watt JM, Milan P, Davies P, Crawford JS. A longitudinal study of cardiovascular dynamic changes throughout pregnancy. Eur J Obstet Gynecol Reprod Biol 12: 215–224, 1981. [DOI] [PubMed] [Google Scholar]
  • 29.Auger N, Fraser WD, Healy-Profitós J, Arbour L. Association between preeclampsia and congenital heart defects. JAMA - J Am Med Assoc 314: 1588–1598, 2015. [DOI] [PubMed] [Google Scholar]
  • 30.Aukes AM, De Groot JC, Wiegman MJ, Aarnoudse JG, Sanwikarja GS, Zeeman GG. Long-term cerebral imaging after pre-eclampsia. BJOG 119: 1117–1122, 2012. [DOI] [PubMed] [Google Scholar]
  • 31.Ausman J, Abbade J, Ermini L, Farrell A, Tagliaferro A, Post M, Caniggia I. Ceramide-induced BOK promotes mitochondrial fission in preeclampsia. Cell Death Dis 9: 298, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Azami M, Azadi T, Farhang S, Rahmati S, Pourtaghi K. The effects of multi mineral-vitamin D and vitamins (C+E) supplementation in the prevention of preeclampsia: An RCT. Int J Reprod Biomed 15: 273–278, 2017. [PMC free article] [PubMed] [Google Scholar]
  • 33.Baker A, Haeri S, Carmargo C, Espinola J, Stuebe A. A nested casecontrol study of midgestation vitamin D deficiency and risk of severe preeclampsia. J Clin Endocrinol Metab 95: 5105–5109, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Bakrania BA, Hall ME, Shahul S, Granger JP. The Reduced Uterine Perfusion Pressure (RUPP) rat model of preeclampsia exhibits impaired systolic function and global longitudinal strain during pregnancy. Pregnancy Hypertens 18: 169–172, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Bakrania BA, Spradley FT, Satchell SC, Rimoldi J, Stec D, Gadepalli R, Granger J. Heme oxygenase-1 is a potent inhibitor of placental ischemia-mediated endothelin-1 production in cultured human glomerular endothelial cells. Am J Phys Regul Integr Comp Phys 314: R427–R432, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Bakrania BA, Spradley FT, Travis AB, Sandner P, Granger JP. Soluble guanylate cyclase activators improve vascular function and attenuate placental ischemia-induced hypertension. Hypertension 72: A013, 2018. [Google Scholar]
  • 37.Bamfo JEAK, Kametas NA, Nicolaides KH, Chambers JB. Maternal left ventricular diastolic and systolic long-axis function during normal pregnancy. Eur J Echocardiogr 8: 360–368, 2007. [DOI] [PubMed] [Google Scholar]
  • 38.Barker DJP. The fetal and infant origins of adult disease. Br Med J 301: 1111, 1990. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Barron LA, Giardina JB, Granger JP, Khalil RA. High-salt diet enhances vascular reactivity in pregnant rats with normal and reduced uterine perfusion pressure. Hypertension 38: 730–735, 2001. [DOI] [PubMed] [Google Scholar]
  • 40.Bartsch E, Medcalf KE, Park AL, Ray JG, Al-Rubaie ZTA, Askie LM, Berger H, Blake J, Graves L, Kingdom JC, Lebovic G, Lord SJ, Maguire JL, Mamdani MM, Meloche J, Urquia ML, Van Wagner V. Clinical risk factors for pre-eclampsia determined in early pregnancy: Systematic review and meta-analysis of large cohort studies. BMJ 353: i1753, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Batashki I, Milchev N, Markova D, Uchikova E, Gǔrova A. Thrombophilia and pregnancy. Akush Ginekol (Sofiia) 45: 38–41, 2006. [PubMed] [Google Scholar]
  • 42.Bauer AJ, Banek CT, Needham K, Gillham H, Capoccia S, Regal JF, Gilbert JS. Pravastatin attenuates hypertension, oxidative stress, and angiogenic imbalance in rat model of placental ischemia-induced hypertension. Hypertension 61: 1103–1110, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Bellamy L, Casas J-P, Hingorani AD, Williams DJ. Pre-eclampsia and risk of cardiovascular disease and cancer in later life: Systematic review and meta-analysis. BMJ 335: 1–12, 2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Benny P, Yamasato K, Yunits B, Zhu X, Ching T, Garmire LX, Berry MJ, Towner D. Maternal cardiovascular-related single nucleotide polymorphisms, genes, and pathways associated with early-onset preeclampsia. PLoS One 14: 1–11, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Bonagura TW, Pepe GJ, Enders AC, Albrecht ED. Suppression of extravillous trophoblast vascular endothelial growth factor expression and uterine spiral artery invasion by estrogen during early baboon pregnancy. Endocrinology 149: 5078–5087, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Boyd HA, Basit S, Behrens I, Leirgul E, Bundgaard H, Wohlfahrt J, Melbye M, Øyen N. Association between fetal congenital heart defects and maternal risk of hypertensive disorders of pregnancy in the same pregnancy and across pregnancies. Circulation 136: 39–48, 2017. [DOI] [PubMed] [Google Scholar]
  • 47.Brantsæter AL, Haugen M, Samuelsen SO, Torjusen H, Trogstad L, Alexander J, Magnus P, Meltzer HM. A dietary pattern characterized by high intake of vegetables, fruits, and vegetable oils is associated with reduced risk of preeclampsia in nulliparous pregnant Norwegian women. J Nutr 139: 1162–1168, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Brodwall K, Leirgul E, Greve G, Vollset SE, Holmstrøm H, Tell GS, Øyen N. Possible common aetiology behind maternal preeclampsia and congenital heart defects in the child: A cardiovascular diseases in Norway Project Study. Paediatr Perinat Epidemiol 30: 76–85, 2016. [DOI] [PubMed] [Google Scholar]
  • 49.Brosens I, Pijnenborg R, Vercruysse L, Romero R. The “great Obstetrical Syndromes” are associated with disorders of deep placentation. Am J Obstet Gynecol 204: 193–201, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Brosens I, Renaer M. On the pathogenesis of placental infarcts in pre-eclampsia. J Obstet Gynaecol Br Commonw 79: 794–799, 1972. [DOI] [PubMed] [Google Scholar]
  • 51.Brosens IA, Robertson WB, Dixon HG. The role of the spiral arteries in the pathogenesis of preeclampsia. Obstet Gynecol Annu 1: 177–191, 1972. [PubMed] [Google Scholar]
  • 52.Brown M Pre-eclampsia: A case of nerves? Lancet 349: 297–289, 1997. [DOI] [PubMed] [Google Scholar]
  • 53.Buhimschi I, Yallampalli C, Chwalisz K, Garfield RE. Pre-eclampsia-like conditions produced by nitric oxide inhibition: Effects of l-arginine, d-arginine and steroid hormones. Hum Reprod 10: 2723–2730, 1995. [DOI] [PubMed] [Google Scholar]
  • 54.Bunney PE, Zink AN, Holm AA, Billington CJ, Kotz CM. Orexin activation counteracts decreases in nonexercise activity thermogenesis (NEAT) caused by high-fat diet. Physiol Behav 176: 139–148, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Burstyn I, Sithole F, Zwaigenbaum L. Autism spectrum disorders, maternal characteristics and obstetric complications among singletons born in Alberta, Canada. Chronic Dis Can 30: 125–134, 2010. [PubMed] [Google Scholar]
  • 56.Cadoret F, Guerby P, Cavaignac-Vitalis M, Vayssiere C, Parant O, Vidal F. Expectant management in HELLP syndrome: Predictive factors of disease evolution. J Matern-Fetal Neonatal Med, 2020. DOI: 10.1080/14767058.2019.1702956. [DOI] [PubMed] [Google Scholar]
  • 57.Carmeliet P Blood vessels and nerves: Common signals, pathways and diseases. Nat Rev Genet 4: 710–720, 2003. [DOI] [PubMed] [Google Scholar]
  • 58.Carter AM, Enders AC, Pijnenborg R. The role of invasive trophoblast in implantation and placentation of primates. Philos Trans R Soc B Biol Sci 370, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Cavanagh D, Rao PS, Tung KSK, Gaston L. Eclamptogenic toxemia: The development of an experimental model in the subhuman primate. Am J Obstet Gynecol 120: 183–196, 1974. [DOI] [PubMed] [Google Scholar]
  • 60.Chaiworapongsa T, Chaemsaithong P, Yeo L, Romero R. Pre-eclampsia: Current understanding of its pathophysiology. Nat Rev Nephrol 10: 466–480, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Chesley LC, Annitto JE, Cosgrove RA. The familial factor in toxemia of pregnancy. Obstet Gynecol 32: 303–311, 1968. [PubMed] [Google Scholar]
  • 62.Clowse MEB, Jamison M, Myers E, James AH. A national study of the complications of lupus in pregnancy. Am J Obstet Gynecol 199: 127.e1–127.e6, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Cnattingius S, Reilly M, Pawitan Y, Lichtenstein P. Maternal and fetal genetic factors account for most of familial aggregation of preeclampsia: A population-based swedish cohort study. Am J Med Genet 130A: 365–371, 2004. [DOI] [PubMed] [Google Scholar]
  • 64.Conrad KP. Evidence for corpus luteal and endometrial origins of adverse pregnancy outcomes in women conceiving with or without assisted reproduction. Obstet Gynecol Clin N Am 47: 163–181, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Cornelius DC, Amaral LM, Harmon A, Wallace K, Thomas AJ, Campbell N, Scott J, Herse F, Haase N, Moseley J, Wallukat G, Dechend R, LaMarca B. An increased population of regulatory T cells improves the pathophysiology of placental ischemia in a rat model of preeclampsia. Am J Phys Regul Integr Comp Phys 309: R884–R891, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Cornelius DC, Amaral LM, Wallace K, Campbell N, Thomas AJ, Scott J, Herse F, Wallukat G, Dechend R, LaMarca B. Reduced uterine perfusion pressure T-helper 17 cells cause pathophysiology associated with preeclampsia during pregnancy. Am J Phys Regul Integr Comp Phys 311: R1192–R1199, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Corr̂a IP, Ruano R, Takiuti NH, Francisco RPV, Bevilacqua E, Zugaib M. Expression of angiogenic factors in placenta of stressed rats. Reprod Fertil Dev 24: 851–858, 2012. [DOI] [PubMed] [Google Scholar]
  • 68.Costantine MM. Physiologic and pharmacokinetic changes in pregnancy. Front Pharmacol 5: 65, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Costantine MM, Cleary K, Hebert MF, Ahmed MS, Brown LM, Ren Z, Easterling TR, Haas DM, Haneline LS, Caritis SN, Venkataramanan R, West H, D’Alton M, Hankins G. Safety and pharmacokinetics of pravastatin used for the prevention of preeclampsia in high-risk pregnant women: A pilot randomized controlled trial. Am J Obstet Gynecol 214: 720.e1–720.e17, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Croy BA, Ashkar AA, Minhas K, Greenwood JD. Can murine uterine natural killer cells give insights into the pathogenesis of preeclampsia? J Soc Gynecol Investig 7: 12–20, 2000. [DOI] [PubMed] [Google Scholar]
  • 71.Cunningham MW, Castillo J, Ibrahim T, Cornelius DC, Campbell N, Amaral L, Vaka VR, Usry N, Williams JM, LaMarca B. AT1-AA (angiotensin II type 1 receptor agonistic autoantibody) blockade prevents preeclamptic symptoms in placental ischemic rats. Hypertension 71: 886–893, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Dang F, Croy BA, Stroman PW, Figueiró-Filho EA. Impactos da pré-eclâmpsia no cérebro de nascituros. Rev Bras Ginecol e Obstet 38: 416–422, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Danielson LA, Kercher LJ, Conrad KP. Impact of gender and endothelin on renal vasodilation and hyperfiltration induced by relaxin in conscious rats. Am J Phys Regul Integr Comp Phys 279: R1298–R1304, 2000. [DOI] [PubMed] [Google Scholar]
  • 74.Darby MM, Wallace K, Cornelius D, Chatman KT, Janae N, Martin JN, Purser CA, Baker RC, Owens MT, Lamarca BB. Vitamin D supplementation suppresses hypoxia-stimulated placental cytokine secretion, hypertension and CD4+ T cell stimulation in response to placental ischemia. Med J Obstet Gynecol 1: 1–11, 2014. [PMC free article] [PubMed] [Google Scholar]
  • 75.Dasinger J, Abais-Buttad J, DL M. Influences of environmental factors during preeclampsia. Am J Phys Regul Integr Comp Phys. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Dasinger JH, Intapad S, Rudsenske BR, Davis GK, Newsome AD, Alexander BT. Chronic blockade of the androgen receptor abolishes age-dependent increases in blood pressure in female growth-restricted rats. Hypertension 67: 1281–1290, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Davis GK, Intapad S, Newsome AD, Coats LE, Bamrick DR, Ojeda NB, Alexander BT. Androgen receptor blockade differentially regulates blood pressure in growth-restricted versus ovarian deficient rats. Hypertension 74: 975–982, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Davis GK, Newsome AD, Cole AB, Ojeda NB, Alexander BT. Chronic estrogen supplementation prevents the increase in blood pressure in female intrauterine growth-restricted offspring at 12 months of age. Hypertension 73: 1128–1136, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Davisson RL, Hoffmann DS, Butz GM, Aldape G, Schlager G, Merrill DC, Sethi S, Weiss RM, Bates JN. Discovery of a spontaneous genetic mouse model of preeclampsia. Hypertension 39: 337–342, 2002. [DOI] [PubMed] [Google Scholar]
  • 80.de Araújo CAL, de Sousa Oliveira L, de Gusmão IMB, Guimarães A, Ribeiro M, Alves JGB. Magnesium supplementation and preeclampsia in low-income pregnant women - A randomized double-blind clinical trial. BMC Pregnancy Childb 20: 1–6, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.de Araújo CAL, Ray JG, Figueiroa JN, Alves JG. BRAzil magnesium (BRAMAG) trial: A double-masked randomized clinical trial of oral magnesium supplementation in pregnancy. BMC Pregnancy Childb 20: 4–10, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.De Jong F, Monuteaux MC, Van Elburg RM, Gillman MW, Belfort MB. Systematic review and meta-analysis of preterm birth and later systolic blood pressure. Hypertension 59: 226–234, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Di Pasquo E, Ghi T, Dall’Asta A, Angeli L, Fieni S, Pedrazzi G, Frusca T. Maternal cardiac parameters can help in differentiating the clinical profile of preeclampsia and in predicting progression from mild to severe forms. Am J Obstet Gynecol 221: 633.e1–633.e9, 2019. [DOI] [PubMed] [Google Scholar]
  • 84.Ditisheim A, Sibai B, Tatevian N. Placental findings in postpartum preeclampsia: A comparative retrospective study. Am J Perinatol, 2019, Epub ahead of print. [DOI] [PubMed] [Google Scholar]
  • 85.Downing I, Shepherd GL, Lewis PJ. Reduced prostacyclin production in pre-eclampsia. Lancet 316: 1374, 1980. [DOI] [PubMed] [Google Scholar]
  • 86.Duckitt K, Harrington D. Risk factors for pre-eclampsia at antenatal booking: Systematic review of controlled studies. Br Med J 330: 565–567, 2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Duley L. Maternal mortality associated with hypertensive disorders of pregnancy in Africa, Asia, Latin America and the Caribbean. BJOG 99: 547–553, 1992. [DOI] [PubMed] [Google Scholar]
  • 88.Duley L. Pre-eclampsia and the hypertensive disorders of pregnancy. Br Med Bull 67: 161–176, 2003. [DOI] [PubMed] [Google Scholar]
  • 89.Duncan JW, Younes ST, Hildebrandt E, Ryan MJ, Granger JP, Drummond HA. Tumor necrosis factor-α impairs cerebral blood flow in pregnant rats: Role of vascular β-epithelial Na + channel. Am J Physiol Circ Physiol 318: H1018–H1027, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Ehrenstein V, Rothman KJ, Pedersen L, Hatch EE, Sørensen HT. Pregnancy-associated hypertensive disorders and adult cognitive function among danish conscripts. Am J Epidemiol 170: 1025–1031, 2009. [DOI] [PubMed] [Google Scholar]
  • 91.Ekholm EMK, Tahvanainen KUO, Metsala T. Heart rate and blood pressure variabilities are increased in pregnancy-induced hypertension. Am J Obstet Gynecol 177: 1208–1212, 1997. [DOI] [PubMed] [Google Scholar]
  • 92.Elharrar V, Nakaya Y, Varro A, Surawicz B. Recovery of V(max) and of action potential duration in canine cardiac purkinje fibers. Fed Proc 43: 419, 1984. [Google Scholar]
  • 93.Eremina V, Sood M, Haigh J, Nagy A, Lajoie G, Ferrara N, Gerber HP, Kikkawa Y, Miner JH, Quaggin SE. Glomerular-specific alterations of VEGF-A expression lead to distinct congenital and acquired renal diseases. J Clin Invest 111: 707–716, 2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Esplin MS, Fausett MB, Fraser A, Kerber R, Mineau G, Carrillo J, Varner MW. Paternal and maternal components of the predisposition to preeclampsia. N Engl J Med 344: 867–872, 2001. [DOI] [PubMed] [Google Scholar]
  • 95.Faas MM, de Vos P. Uterine NK cells and macrophages in pregnancy. Placenta 56: 44–52, 2017. [DOI] [PubMed] [Google Scholar]
  • 96.Faas MM, Schuiling GA, Baller JFW, Visscher CA, Bakker WW. A new animal model for human preeclampsia: Ultra-low-dose endotoxin infusion in pregnant rats. Am J Obstet Gynecol 171: 158–164, 1994. [DOI] [PubMed] [Google Scholar]
  • 97.Facchinetti F, Marozio L, Frusca T, Grandone E, Venturini P, Tiscia GL, Zatti S, Benedetto C. Maternal thrombophilia and the risk of recurrence of preeclampsia. Am J Obstet Gynecol 200: 46.e1–46.e5, 2009. [DOI] [PubMed] [Google Scholar]
  • 98.Failla CM. Placenta growth factor. Encycl Cancer 269: 3590–3592, 2017. [Google Scholar]
  • 99.Fan R, Qiu L, Huang X, Chen Z, Xu H, Zhou C, Zhang X. Increased nerve fibers in placental bed myometrium in women with preeclampsia. Reprod Sci 18: 1262–1266, 2011. [DOI] [PubMed] [Google Scholar]
  • 100.Faulkner JL, Cornelius DC, Amaral LM, Harmon AC, Cunningham MW, Darby MM, Ibrahim T, Thomas DAS, Herse F, Wallukat G, Dechend R, Lamarca B. Vitamin D supplementation improves pathophysiology in a rat model of preeclampsia. Am J Phys Regul Integr Comp Phys 310: R346–R354, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Fisher JJ, Bartho LA, Perkins AV, Holland OJ. Placental mitochondria and reactive oxygen species in the physiology and pathophysiology of pregnancy. Clin Exp Pharmacol Physiol 47: 176–184, 2020. [DOI] [PubMed] [Google Scholar]
  • 102.Fisher JP, Paton JFR. The sympathetic nervous system and blood pressure in humans: Implications for hypertension. J Hum Hypertens 26: 463–475, 2012. [DOI] [PubMed] [Google Scholar]
  • 103.Fleiss B, Wong F, Brownfoot F, Shearer IK, Baud O, Walker DW, Gressens P, Tolcos M. Knowledge gaps and emerging research areas in intrauterine growth restriction-associated brain injury. Front Endocrinol (Lausanne) 10: 1–24, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Fried G, Hökfelt T, Lundberg JM, Terenius L, Hamberger L. Neuropeptide Y and noradrenaline in human uterus and myometrium during normal and pre-eclamptic pregnancy. Hum Reprod 1: 359–364, 1986. [DOI] [PubMed] [Google Scholar]
  • 105.Fushima T, Sekimoto A, Oe Y, Sato E, Ito S, Sato H, Takahashi N. Nicotinamide ameliorates a preeclampsia-like condition in mice with reduced uterine perfusion pressure. Am J Physiol Ren Physiol 312: F366–F372, 2017. [DOI] [PubMed] [Google Scholar]
  • 106.Gagliardi CL, Goldsmith LT, Saketos M, Weiss G, Schmidt CL. Human chorionic gonadotropin stimulation of relaxin secretion by luteinized human granulosa cells. Fertil Steril 58: 314–320, 1992. [PubMed] [Google Scholar]
  • 107.Gamage TKJB, Chamley LW, James JL. Stem cell insights into human trophoblast lineage differentiation. Hum Reprod Update 23: 77–103, 2016. [DOI] [PubMed] [Google Scholar]
  • 108.Garrett N, Pombo J, Umpierrez M, Clark JE, Simmons M, Girardi G. Pravastatin therapy during preeclampsia prevents long-term adverse health effects in mice. JCI Insight 3: 1–16, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Gellersen B, Reimann K, Samalecos A, Aupers S, Bamberger AM. Invasiveness of human endometrial stromal cells is promoted by decidualization and by trophoblast-derived signals. Hum Reprod 25: 862–873, 2010. [DOI] [PubMed] [Google Scholar]
  • 110.George EM, Arany I. Induction of heme oxygenase-1 shifts the balance from proinjury to prosurvival in the placentas of pregnant rats with reduced uterine perfusion pressure. Am J Phys Regul Integr Comp Phys 302: R620–R626, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.George EM, Arany M, Cockrell K, Storm M, Stec DE, Granger JP. Induction of heme oxygenase-1 attenuates sFlt-1-induced hypertension in pregnant rats. Am J Phys Regul Integr Comp Phys 301: R1495–R1500, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.George EM, Cockrell K, Aranay M, Csongradi E, Stec DE, Granger JP. Induction of heme oxygenase 1 attenuates placental ischemia-induced hypertension. Hypertension 57: 941–948, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.George EM, Granger JP. Linking placental ischemia and hypertension in preeclampsia: Role of endothelin 1. Hypertension 60: 507–511, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.George EM, Palei AC, Dent EA, Granger JP. Sildenafil attenuates placental ischemia-induced hypertension. Am J Phys Regul Integr Comp Phys 305: R397–R403, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.George EM, Stout JM, Stec DE, Granger JP. Heme oxygenase induction attenuates TNF-α-induced hypertension in pregnant rodents. Front Pharmacol 6: 1–6, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Gilbert JS, Babcock SA, Granger JP. Hypertension produced by reduced uterine perfusion in pregnant rats is associated with increased soluble fms-like tyrosine kinase-1 expression. Hypertension 50: 1142–1147, 2007. [DOI] [PubMed] [Google Scholar]
  • 117.Gilbert JS, Ryan MJ, LaMarca BB, Sedeek M, Murphy SR, Granger JP. Pathophysiology of hypertension during preeclampsia: Linking placental ischemia with endothelial dysfunction. Am J Physiol Heart Circ Physiol 294: H541–H550, 2008. [DOI] [PubMed] [Google Scholar]
  • 118.Gillis EE, Mooney JN, Garrett MR, Granger JP, Sasser JM. Sildenafil treatment ameliorates the maternal syndrome of preeclampsia and rescues fetal growth in the Dahl Salt-Sensitive rat. Hypertension 67: 647–653, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Gillis EE, Williams JM, Garrett MR, Mooney JN, Sasser JM. The Dahl Salt-Sensitive rat is a spontaneous model of superimposed preeclampsia. Am J Phys Regul Integr Comp Phys 309: R62–R70, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Giwa A, Nguyen M. Late onset postpartum preeclampsia 3 months after delivery. Am J Emerg Med 35: 1582.e1–1582.e3, 2017. [DOI] [PubMed] [Google Scholar]
  • 121.Görlach A, Dimova EY, Petry A, Martínez-Ruiz A, Hernansanz-Agustín P, Rolo AP, Palmeira CM, Kietzmann T. Reactive oxygen species, nutrition, hypoxia and diseases: Problems solved? Redox Biol 6: 372–385, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Goulopoulou S Maternal vascular physiology in preeclampsia. Hypertension 70: 1066–1073, 2017. [DOI] [PubMed] [Google Scholar]
  • 123.Gray KJ, Kovacheva VP, Mirzakhani H, Bjonnes AC, Almoguera B, DeWan AT, Triche EW, Saftlas AF, Hoh J, Bodian DL, Klein E, Huddleston KC, Ingles SA, Lockwood CJ, Hakonarson H, McElrath TF, Murray JC, Wilson ML, Norwitz ER, Karumanchi SA, Bateman BT, Keating BJ, Saxena R. Gene-centric analysis of preeclampsia identifies maternal association at PLEKHG1. Hypertension 72: 408–416, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Greenwood JP, Scott EM, Walker JJ, Stoker JB, Mary DASG. The magnitude of sympathetic hyperactivity in pregnancy-induced hypertension and preeclampsia. Am J Hypertens 16: 194–199, 2003. [DOI] [PubMed] [Google Scholar]
  • 125.Greenwood JP, Stoker JB, Walker JJ, Mary DASG. Sympathetic nerve discharge in normal pregnancy and pregnancy-induced hypertension. J Hypertens 16: 617–624, 1998. [DOI] [PubMed] [Google Scholar]
  • 126.Grigore D, Ojeda NB, Robertson EB, Dawson AS, Huffman CA, Bourassa EA, Speth RC, Brosnihan KB, Alexander BT. Placental insufficiency results in temporal alterations in the renin angiotensin system in male hypertensive growth restricted offspring. Am J Phys Regul Integr Comp Phys 293: R804–R811, 2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Groom KM, McCowan LM, Mackay LK, Lee AC, Gardener G, Unterscheider J, Sekar R, Dickinson JE, Muller P, Reid RA, Watson D, Welsh A, Marlow J, Walker SP, Hyett J, Morris J, Stone PR, Baker PN. STRIDER NZAus: A multicentre randomised controlled trial of sildenafil therapy in early-onset fetal growth restriction. BJOG 126: 997–1006, 2019. [DOI] [PubMed] [Google Scholar]
  • 128.Grunewald C, Kublickas M, Carlström K, Lunell NO, Nisell H. Effects of nitroglycerin on the uterine and umbilical circulation in severe preeclampsia. Obstet Gynecol 86: 600–604, 1995. [DOI] [PubMed] [Google Scholar]
  • 129.Guirguis GF, Aziz MM, Boccia Liang C, Williams SF, Apuzzio JJ, Bilinski R, Mornan AJD, Shah LP. Is preeclampsia an independent predictor of diastolic dysfunction? A retrospective cohort study. Pregnancy Hypertens 5: 359–361, 2015. [DOI] [PubMed] [Google Scholar]
  • 130.Gumusoglu SB, Chilukuri ASS, Santillan DA, Santillan MK, Stevens HE. Neurodevelopmental outcomes of prenatal preeclampsia exposure. Trends Neurosci 43: 253–268, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Gutkowska J, Granger JP, Lamarca BB, Danalache BA, Wang D, Jankowski M. Changes in cardiac structure in hypertension produced by placental ischemia in pregnant rats: Effect of tumor necrosis factor blockade. J Hypertens 29: 1203–1212, 2011. [DOI] [PubMed] [Google Scholar]
  • 132.Hall ME, George EM, Granger JP. The heart during pregnancy. Rev Española Cardiol (English Ed) 64: 1045–1050, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Halmøy A, Klungsøyr K, Skjærven R, Haavik J. Pre- and perinatal risk factors in adults with attention-deficit/hyperactivity disorder. Biol Psychiatry 71: 474–481, 2012. [DOI] [PubMed] [Google Scholar]
  • 134.Hammad IA, Meeks H, Fraser A, Theilen LH, Esplin MS, Smith KR, Varner MW. Risks of cause-specific mortality in offspring of pregnancies complicated by hypertensive disease of pregnancy. Am J Obstet Gynecol 222: 75.e1–75.e9, 2020. [DOI] [PubMed] [Google Scholar]
  • 135.Hammer ES, Cipolla MJ. Cerebrovascular dysfunction in preeclamptic pregnancies. Curr Hypertens Rep 17: 1–13, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Handy DE, Castro R, Loscalzo J. Epigenetic modifications: Basic mechanisms and role in cardiovascular disease. Circulation 123: 2145–2156, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Hanna J, Goldman-Wohl D, Hamani Y, Avraham I, Greenfield C, Natanson-Yaron S, Prus D, Cohen-Daniel L, Arnon TI, Manaster I, Gazit R, Yutkin V, Benharroch D, Porgador A, Keshet E, Yagel S, Mandelboim O. Decidual NK cells regulate key developmental processes at the human fetal-maternal interface. Nat Med 12: 1065–1074, 2006. [DOI] [PubMed] [Google Scholar]
  • 138.Harmon A, Cornelius D, Amaral L, Paige A, Herse F, Ibrahim T, Wallukat G, Faulkner J, Moseley J, Dechend R, LaMarca B. IL-10 supplementation increases Tregs and decreases hypertension in the RUPP rat model of preeclampsia. Hypertens Pregnancy 34: 291–306, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Hering L, Herse F, Geusens N, Verlohren S, Wenzel K, Staff AC, Brosnihan KB, Huppertz B, Luft FC, Muller DN, Pijnenborg R, Cartwright JE, Dechend R. Effects of circulating and local uteroplacental angiotensin ii in rat pregnancy. Hypertension 56: 311–318, 2010. [DOI] [PubMed] [Google Scholar]
  • 140.Hernandez-Andrade E, Figueroa-Diesel H, Jansson T, Rangel-Nava H, Gratacos E. Changes in regional fetal cerebral blood flow perfusion in relation to hemodynamic deterioration in severely growth-restricted fetuses. Ultrasound Obstet Gynecol 32: 71–76, 2008. [DOI] [PubMed] [Google Scholar]
  • 141.Hiby SE, Walker JJ, O’Shaughnessy KM, Redman CWG, Carrington M, Trowsdale J, Moffett A. Combinations of maternal KIR and fetal HLA-C genes influence the risk of preeclampsia and reproductive success. J Exp Med 200: 957–965, 2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Hines T, Beauchamp D, Rice C. Baroreflex control of sympathetic nerve activity in hypertensive pregnant rats with reduced uterine perfusion. Hypertens Pregnancy 26: 303–314, 2007. [DOI] [PubMed] [Google Scholar]
  • 143.Hodari AA. Chronic uterine ischemia and reversible experimental “toxemia of pregnancy”. Am J Obstet Gynecol 97: 597–607, 1967. [DOI] [PubMed] [Google Scholar]
  • 144.Hofmann AP, Gerber SA, Croy BA. Uterine natural killer cells pace early development of mouse decidua basalis. Mol Hum Reprod 20: 66–76, 2014. [DOI] [PubMed] [Google Scholar]
  • 145.Hofstee P, Cuffe JSM, Perkins AV. Analysis of selenoprotein expression in response to dietary selenium deficiency during pregnancy indicates tissue specific differential expression in mothers and sex specific changes in the fetus and offspring. Int J Mol Sci 21: 1–19, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Hogberg U. The World Health Report 2005: “Make every mother and child count” - including Africans. Scand J Public Health 33: 409–411, 2005. [DOI] [PubMed] [Google Scholar]
  • 147.Hu D, Cross JC. Ablation of Tpbpa-positive trophoblast precursors leads to defects in maternal spiral artery remodeling in the mouse placenta. Dev Biol 358: 231–239, 2011. [DOI] [PubMed] [Google Scholar]
  • 148.Huang X, Li Z, Lei J, Wang D, Zhang Y. Genetic polymorphism in DGCR8 is associated with late onset of preeclampsia. BMC Med Genet 20: 1–6, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Hubel CA, Wallukat G, Wolf M, Herse F, Rajakumar A, Roberts JM, Markovic N, Thadhani R, Luft FC, Dechend R. Agonistic angiotensin II type 1 receptor autoantibodies in postpartum women with a history of preeclampsia. Hypertension 49: 612–617, 2007. [DOI] [PubMed] [Google Scholar]
  • 150.Hunkapiller NM, Gasperowicz M, Kapidzic M, Plaks V, Maltepe E, Kitajewski J, Cross JC, Fisher SJ. A role for Notch signaling in trophoblast endovascular invasion and in the pathogenesis of pre-eclampsia. Development 138: 2987–2998, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Hutcheon J, Lisonkova S, Joseph K. Epidemiology of pre-eclapmsia and other hypertensive disorders of pregnancy. Best Pract Res Clin Obstet Gynaecol 25: 391–403, 2011. [DOI] [PubMed] [Google Scholar]
  • 152.Ibrahim T, Przybyl L, Harmon AC, Amaral LM, Faulkner JL, Cornelius DC, Cunningham MW, Hünig T, Herse F, Wallukat G, Dechend R, LaMarca B. Proliferation of endogenous regulatory T cells improve the pathophysiology associated with placental ischaemia of pregnancy. Am J Reprod Immunol 78, 2017. DOI: 10.1111/aji.12724. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Inkeri Lokki A, Kaartokallio T, Holmberg V, Onkamo P, Koskinen LLE, Saavalainen P, Heinonen S, Kajantie E, Kere J, Kivinen K, Pouta A, Villa PM, Hiltunen L, Laivuori H, Meri S. Analysis of complement C3 gene reveals susceptibility to severe Preeclampsia. Front Immunol 8: 1–10, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Intapad S, Warrington JP, Spradley FT, Palei AC, Drummond HA, Ryan MJ, Granger JP, Alexander BT. Reduced uterine perfusion pressure induces hypertension in the pregnant mouse. Am J Phys Regul Integr Comp Phys 307: R1353–R1357, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Iriyama T, Wang W, Parchim NF, Song A, Blackwell SC, Sibai BM, Kellems RE, Xia Y. Hypoxia-independent upregulation of placental hypoxia inducible factor-1α gene expression contributes to the pathogenesis of preeclampsia. Hypertension 65: 1307–1315, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Ishikawa T, Hume JR, Keef KD. Regulation of Ca2+ channels by cAMP and cGMP in vascular smooth muscle cells. Circ Res 73: 1128–1137, 1993. [DOI] [PubMed] [Google Scholar]
  • 157.Jansen CHJR, Kastelein AW, Kleinrouweler CE, Van Leeuwen E, De Jong KH, Pajkrt E, Van Noorden CJF. Development of placental abnormalities in location and anatomy. Acta Obstet Gynecol Scand, 2020, Epub ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Johal T, Lees CC, Everett TR, Wilkinson IB. The nitric oxide pathway and possible therapeutic options in pre-eclampsia. Br J Clin Pharmacol 78: 244–257, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Jr C, Gnam A, Ibrahim T, Lamarca B, Denise C. Reduced uterine perfusion pressure. Clin Sci 131: 2753–2762, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Kajantie E, Osmond C, Eriksson JG. Gestational hypertension is associated with increased risk of type 2 diabetes in adult offspring: The Helsinki Birth Cohort Study. Am J Obstet Gynecol 216: 281.e1–281.e7, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Kakigano A, Mimura K, Kanagawa T, Nakayama M, Kanayama T, Fujita S, Kinugasa-Taniguchi Y, Endo M, Tomimatsu T, Kimura T. Imbalance of angiogenic factors and avascular edematous cystic villi in a trisomy 13 pregnancy: A case report. Placenta 34: 628–630, 2013. [DOI] [PubMed] [Google Scholar]
  • 162.Kakutani M, Masaki T, Sawamura T. A platelet – endothelium interaction mediated by lectin- like oxidized low-density lipoprotein receptor-1. PNAS 97: 360–364, 2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Kalantar F, Rajaei S, Heidari AB, Mansouri R, Rashidi N, Izad MH, Mirahmadian M. Serum levels of tumor necrosis factor-α, interleukin-15 and interleukin-10 in patients with pre-eclampsia in comparison with normotensive pregnant women. Iran J Nurs Midwifery Res 18: 463–466, 2013. [PMC free article] [PubMed] [Google Scholar]
  • 164.Kanayama N, Tsujimura R, She L, Maehara K, Terao T. Cold-induced stress stimulates the sympathetic nervous system, causing hypertension and proteinuria in rats. J Hypertens 15: 383–389, 1997. [DOI] [PubMed] [Google Scholar]
  • 165.Kao CK, Morton JS, Quon AL, Reyes LM, Lopez-jaramillo P, Davidge ST. Mechanism of vascular dysfunction due to circulating factors in women with preeclampsia. Clin Sci 130: 539–549, 2016. [DOI] [PubMed] [Google Scholar]
  • 166.Kasture V, Sundrani D, Dalvi S, Swamy M, Kale A, Joshi S. Maternal omega-3 fatty acids and vitamin E improve placental angiogenesis in late-onset but not early-onset preeclampsia. Mol Cell Biochem 461: 159–170, 2019. [DOI] [PubMed] [Google Scholar]
  • 167.Kelly LK, Wedgwood S, Steinhorn RH, Black SM. Nitric oxide decreases endothelin-1 secretion through the activation of soluble guanylate cyclase. Am J Phys Lung Cell Mol Phys 286: L984–L991, 2004. [DOI] [PubMed] [Google Scholar]
  • 168.Khandwala YS, Baker VL, Shaw GM, Stevenson DK, Lu Y, Eisenberg ML. Association of paternal age with perinatal outcomes between 2007 and 2016 in the United States: Population based cohort study. BMJ 363: k4372, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Khatun S, Kanayama N, Belayet HM, Bhuiyan AB, Jahan S, Begum A, Kobayashi T, Terao T. Increased concentrations of plasma neuropeptide Y in patients with eclampsia and preeclampsia. Am J Obstet Gynecol 182: 896–900, 2000. [DOI] [PubMed] [Google Scholar]
  • 170.Khatun S, Kanayama N, Belayet HM, Masui M, Sugimura M, Kobayashi T, Terao T. Induction of preeclampsia like phenomena by stimulation of sympathetic nerve with cold and fasting stress. Eur J Obstet Gynecol Reprod Biol 86: 89–97, 1999. [DOI] [PubMed] [Google Scholar]
  • 171.Khera A, Vanderlelie JJ, Holland O, Perkins AV. Overexpression of endogenous anti-oxidants with selenium supplementation protects trophoblast cells from reactive oxygen species-induced apoptosis in a Bcl-2-dependent manner. Biol Trace Elem Res 177: 394–403, 2017. [DOI] [PubMed] [Google Scholar]
  • 172.Khidri FF, Waryah YM, Ali FK, Shaikh H, Ujjan ID, Waryah AM. MTHFR and F5 genetic variations have association with preeclampsia in Pakistani patients: A case control study. BMC Med Genet 20: 1–12, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Kiely ME, Wagner CL, Roth DE. Vitamin D in pregnancy: Where we are and where we should go. J Steroid Biochem Mol Biol 201: 105669, 2020. [DOI] [PubMed] [Google Scholar]
  • 174.Kim M, Park HJ, Seol JW, Jang JY, Cho YS, Kim KR, Choi Y, Lydon JP, Demayo FJ, Shibuya M, Ferrara N, Sung HK, Nagy A, Alitalo K, Koh GY. VEGF-A regulated by progesterone governs uterine angiogenesis and vascular remodelling during pregnancy. EMBO Mol Med 5: 1415–1430, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Kjeldsen SE, Eide I, Aakesson I, Oian P, Maltau JM, Lande K, Gjesdal K. Increased arterial adrenaline is highly correlated to blood pressure and in vivo platelet function in pre-eclampsia. J Hypertens 3: s93–s95, 1985. [PubMed] [Google Scholar]
  • 176.Knöfler M, Haider S, Saleh L, Pollheimer J, Gamage TKJB, James J. Human placenta and trophoblast development: Key molecular mechanisms and model systems. Cell Mol Life Sci 76: 3479–3496, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Knöfler M, Pollheimer J. Human placental trophoblast invasion and differentiation: A particular focus on Wnt signaling. Front Genet 4: 190, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Koopman LA, Kopcow HD, Rybalov B, Boyson JE, Orange JS, Schatz F, Masch R, Lockwood CJ, Schachter AD, Park PJ, Strominger JL. Human decidual natural killer cells are a unique NK cell subset with immunomodulatory potential. J Exp Med 198: 1201–1212, 2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Kräker K, O’Driscoll JM, Schütte T, Herse F, Patey O, Golic M, Geisberger S, Verlohren S, Birukov A, Heuser A, Müller DN, Thilaganathan B, Dechend R, Haase N. Statins reverse postpartum cardiovascular dysfunction in a rat model of preeclampsia. Hypertension 75: 202–210, 2020. [DOI] [PubMed] [Google Scholar]
  • 180.Krebs C, Macara LM, Leiser R, Bowman AW, Greer IA, Kingdom JCP. Intrauterine growth restriction with absent end-diastolic flow velocity in the umbilical artery is associated with maldevelopment of the placental terminal villous tree. Am J Obstet Gynecol 175: 1534–1542, 1996. [DOI] [PubMed] [Google Scholar]
  • 181.Krutzén E, Olofsson P, Bäck SE, Nilsson-Ehle P. Glomerular filtration rate in pregnancy: A study in normal subjects and in patients with hypertension, preeclampsia and diabetes. Scand J Clin Lab Invest 52: 387–392, 1992. [DOI] [PubMed] [Google Scholar]
  • 182.LaMarca B, Parrish M, Ray LF, Murphy SR, Roberts L, Glover P, Wallukat G, Wenzel K, Cockrell K, Martin JN, Ryan MJ, Dechend R. Hypertension in response to autoantibodies to the angiotensin II type i receptor (AT1-AA) in pregnant rats: Role of endothelin-1. Hypertension 54: 905–909, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.LaMarca B, Speed J, Fournier L, Babcock SA, Berry H, Cockrell K, Granger JP. Hypertension in response to chronic reductions in uterine perfusion in pregnant rats effect of tumor necrosis factor-α blockade. Hypertension 52: 1161–1167, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.LaMarca B, Wallace K, Granger J. Role of angiotensin II type i receptor agonistic autoantibodies (AT1-AA) in preeclampsia. Curr Opin Pharmacol 11: 175–179, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.LaMarca BBD, Bennett WA, Alexander BT, Cockrell K, Granger JP. Hypertension produced by reductions in uterine perfusion in the pregnant rat: Role of tumor necrosis factor-alpha. Hypertension 46: 1022–1025, 2005. [DOI] [PubMed] [Google Scholar]
  • 186.LaMarca BBD, Cockrell K, Sullivan E, Bennett W, Granger JP. Role of endothelin in mediating tumor necrosis factor-induced hypertension in pregnant rats. Hypertension 46: 82–86, 2005. [DOI] [PubMed] [Google Scholar]
  • 187.Lash GE, Cartwright JE, Whitley GSJ, Trew AJ, Baker PN. The effects of angiogenic growth factors on extravillous trophoblast invasion and motility. Placenta 20: 661–667, 1999. [DOI] [PubMed] [Google Scholar]
  • 188.Lau S, Guild S, Barrett C, Chen Q, McCowan L, Jordan V, Chamley L. Tumor necrosis factor-alpha, interleukin-6, and interleukin-10 levels are altered in preeclampsia: A systematic review and meta-analysis. Am J Reprod Immunol 70: 412–427, 2013. [DOI] [PubMed] [Google Scholar]
  • 189.Le Bouteiller P, Tabiasco J. Killers become builders during pregnancy. Nat Med 12: 991–992, 2006. [DOI] [PubMed] [Google Scholar]
  • 190.Leon LJ, McCarthy FP, Direk K, Gonzalez-Izquierdo A, Prieto-Merino D, Casas JP, Chappell L. Preeclampsia and cardiovascular disease in a large UK pregnancy cohort of linked electronic health records a CAL-IBER study. Circulation 140: 1050–1060, 2019. [DOI] [PubMed] [Google Scholar]
  • 191.Lewinsky RM, Riskin-Mashiah S. Autonomic imbalance in preeclampsia: Evidence for increased sympathetic tone in response to the supinepressor test. Obstet Gynecol 91: 935–939, 1998. [DOI] [PubMed] [Google Scholar]
  • 192.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 303: H1–H8, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Li M, Yee D, Magnuson TR, Smithies O, Caron KM. Reduced maternal expression of adrenomedullin disrupts fertility, placentation, and fetal growth in mice. J Clin Invest 116: 2653–2662, 2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Lian IA, Løset M, Mundal SB, Fenstad MH, Johnson MP, Eide IP, Bjørge L, Freed KA, Moses EK, Austgulen R. Increased endoplasmic reticulum stress in decidual tissue from pregnancies complicated by fetal growth restriction with and without pre-eclampsia. Placenta 32: 823–829, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Lisonkova S, Joseph KS. Incidence of preeclampsia: Risk factors and outcomes associated with early-versus late-onset disease. Am J Obstet Gynecol 209: 544.e1–544.e12, 2013. [DOI] [PubMed] [Google Scholar]
  • 196.Liu R NOS1 in kidney macula densa controls the development of hypertension during pregnancy. J Hypertens 36: e28, 2018. [Google Scholar]
  • 197.Lodhi RSZ, Nakabayashi K, Suzuki K, Yamada AY, Hazama R, Ebina Y, Yamada H. Relaxin has anti-apoptotic effects on human trophoblast-derived HTR-8/SV neo cells. Gynecol Endocrinol 29: 1051–1054, 2013. [DOI] [PubMed] [Google Scholar]
  • 198.Logue OC, Mahdi F, Chapman H, George EM, Bidwell GL. A maternally sequestered, biopolymer-stabilized Vascular Endothelial Growth Factor (VEGF) chimera for treatment of preeclampsia. J Am Heart Assoc 6: e007216, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Lopes van Balen VA, Spaan JJ, Cornelis T, Spaanderman MEA. Prevalence of chronic kidney disease after preeclampsia. J Nephrol 30: 403–409, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Lowe DT. Nitric oxide dysfunction in the pathophysiology of preeclampsia. Nitric Oxide 4: 441–458, 2000. [DOI] [PubMed] [Google Scholar]
  • 201.Lu HQ, Hu R. Lasting effects of intrauterine exposure to preeclampsia on offspring and the underlying mechanism. Am J Perinatol Reports 09: e275–e291, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Luzi G, Caserta G, Iammarino G, Clerici G, Di Renzo GC. Nitric oxide donors in pregnancy: Fetomaternal hemodynamic effects induced in mild pre-eclampsia and threatened preterm labor. Ultrasound Obstet Gynecol 14: 101–109, 1999. [DOI] [PubMed] [Google Scholar]
  • 203.Lyall F, Bulmer JN, Duffie E, Cousins F, Theriault A, Robson SC. Human trophoblast invasion and spiral artery transformation: The role of PECAM-1 in normal pregnancy, preeclampsia, and fetal growth restriction. Am J Pathol 158: 1713–1721, 2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Lyall F, Robson SC, Bulmer JN. Spiral artery remodeling and trophoblast invasion in preeclampsia and fetal growth restriction relationship to clinical outcome. Hypertension 62: 1046–1054, 2013. [DOI] [PubMed] [Google Scholar]
  • 205.Maeda K. Preeclampsia is caused by continuous sympathetic center excitation due to an enlarged pregnant uterus. J Perinat Med 42: 233–237, 2014. [DOI] [PubMed] [Google Scholar]
  • 206.Makris A, Yeung KR, Lim SM, Sunderland N, Heffernan S, Thompson JF, Iliopoulos J, Killingsworth MC, Yong J, Xu B, Ogle RF, Thadhani R, Karumanchi SA, Hennessy A. Placental growth factor reduces blood pressure in a uteroplacental ischemia model of preeclampsia in nonhuman primates. Hypertension 67: 1263–1272, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Mann JR, McDermott S, Bao H, Hardin J, Gregg A. Pre-eclampsia, birth weight, and autism spectrum disorders. J Autism Dev Disord 40: 548–554, 2010. [DOI] [PubMed] [Google Scholar]
  • 208.Marti JJ, Herrmann U. Immunogestosis: A new etiologic concept of “essential” EPH gestosis, with special consideration of the primigravid patient. Preliminary report of a clinical study. Am J Obstet Gynecol 128: 489–493, 1977. [DOI] [PubMed] [Google Scholar]
  • 209.Martínez-Abundis E, González-Ortiz M, Hernández-Salazar F, Huerta-J-Lucas MT. Sublingual isosorbide dinitrate in the acute control of hypertension in patients with severe preeclampsia. Gynecol Obstet Investig 50: 39–42, 2000. [DOI] [PubMed] [Google Scholar]
  • 210.Masoudian P, Nasr A, De Nanassy J, Fung-Kee-Fung K, Bainbridge SA, El Demellawy D. Oocyte donation pregnancies and the risk of preeclampsia or gestational hypertension: A systematic review and metaanalysis. Am J Obstet Gynecol 214: 328–339, 2016. [DOI] [PubMed] [Google Scholar]
  • 211.Mastrogiannis DS, O’Brien WF, Krammer J, Benoit R. Potential role of endothelin-1 in normal and hypertensive pregnancies. Am J Obstet Gynecol 165: 1711–1716, 1991. [DOI] [PubMed] [Google Scholar]
  • 212.Matsubara K, Higaki T, Matsubara Y, Nawa A. Nitric oxide and reactive oxygen species in the pathogenesis of preeclampsia. Int J Mol Sci 16: 4600–4614, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213.Mayer-Pickel K, Kolovetsiou-Kreiner V, Stern C, Münzker J, Eberhard K, Trajanoski S, Lakovschek I-C, Ulrich D, Csapo B, Lang U, Obermayer-Pietsch B, Cervar-Zivkovic M. Effect of low-dose aspirin on soluble FMS-like tyrosine kinase 1/placental growth factor (sFlt-1/PlGF ratio) in pregnancies at high risk for the development of preeclampsia. J Clin Med 8: 1429, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214.Maynard SE, Karumanchi SA. Angiogenic factors and preeclampsia. Semin Nephrol 31: 33–46, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215.Maynard SE, Min J, Merchan J, Lim K, Li J, Mondal S, Libermann TA, Morgon JP, Sellke FW, Stillman IE, Epstein FH, Sukhatme VP, Karumanchi SA. Excess placental soluble fms-like tyrosine kinase (sFlt-1) may contribute to endothelial dysfunction, hypertension and proteinuria in preeclampsia. J Clin Invest 111: 649–658, 2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 216.Mbah AK, Kornosky JL, Kristensen S, August EM, Alio AP, Marty PJ, Belogolovkin V, Bruder K, Salihu HM. Super-obesity and risk for early and late pre-eclampsia. BJOG 117: 997–1004, 2010. [DOI] [PubMed] [Google Scholar]
  • 217.McAlpine JM, McKeating DR, Vincze L, Vanderlelie JJ, Perkins AV. Essential mineral intake during pregnancy and its association with maternal health and birth outcomes in South East Queensland, Australia. Nutr Metab Insights 12: 117863881987944, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 218.McGinnis R, Steinthorsdottir V, Williams NO, Thorleifsson G, Shooter S, Hjartardottir S, Bumpstead S, Stefansdottir L, Hildyard L, Sigurdsson JK, Kemp JP, Silva GB, Thomsen LCV, Jääskeläinen T, Kajantie E, Chappell S, Kalsheker N, Moffett A, Hiby S, Lee WK, Padmanabhan S, Simpson NAB, Dolby VA, Staines-Urias E, Engel SM, Haugan A, Trogstad L, Svyatova G, Zakhidova N, Najmutdinova D, Dominiczak AF, Gjessing HK, Casas JP, Dudbridge F, Walker JJ, Pipkin FB, Thorsteinsdottir U, Geirsson RT, Lawlor DA, Iversen AC, Magnus P, Laivuori H, Stefansson K, Morgan L. Variants in the fetal genome near FLT1 are associated with risk of preeclampsia. Nat Genet 49: 1255–1260, 2017. [DOI] [PubMed] [Google Scholar]
  • 219.McLeod L How useful is uterine artery Doppler ultrasonography in predicting pre-eclampsia and intrauterine growth restriction? CMAJ 178: 727–729, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220.Melchiorre K, Sutherland GR, Liberati M, Thilaganathan B. Preeclampsia is associated with persistent postpartum cardiovascular impairment. Hypertension 58: 709–715, 2011. [DOI] [PubMed] [Google Scholar]
  • 221.Melchiorre K, Thilaganathan B. Maternal cardiac function in preeclampsia. Curr Opin Obstet Gynecol 23: 440–447, 2011. [DOI] [PubMed] [Google Scholar]
  • 222.Menezes de Oliveira AC, Albuquerque Santos A, Rodrigues Bezerra A, Machado Tavares MC, Rocha de Barros AM, Costa Ferreira R. Intake of antioxidant nutrients and coefficients of variation in pregnant women with preeclampsia. Rev Port Cardiol 35: 469–476, 2016. [DOI] [PubMed] [Google Scholar]
  • 223.Merlob P, Stahl B, Klinger G. 17-Hydroxyprogesterone caproate for prevention of recurrent spontaneous preterm birth. Reprod Toxicol 33: 15–19, 2012. [DOI] [PubMed] [Google Scholar]
  • 224.Miasiro N, Paiva ACM. Effects of endothelin-1 and endothelin-3 on the isolated guinea pig ileum: Role of Na+ ions and endothelin receptor subtypes. J Cardiovasc Pharmacol 20: S37–S40, 1992. [DOI] [PubMed] [Google Scholar]
  • 225.Mikolasevic I, Filipec-Kanizaj T, Jakopcic I, Majurec I, Brncic-Fischer A, Sobocan N, Hrstic I, Stimac T, Stimac D, Milic S. Liver disease during pregnancy: A challenging clinical issue. Med Sci Monit 24: 4080–4090, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226.Mills JL, DerSimonian R, Raymond E, Morrow JD, Roberts LJ, Clemens JD, Hauth JC, Catalano P, Sibai B, Curet LB, Levine RJ. Prostacyclin and thromboxane changes predating clinical onset of preeclampsia: A multicenter prospective study. J Am Med Assoc 282: 356–362, 1999. [DOI] [PubMed] [Google Scholar]
  • 227.Mizuuchi M, Cindrova-Davies T, Olovsson M, Charnock-Jones DS, Burton GJ, Yung HW. Placental endoplasmic reticulum stress negatively regulates transcription of placental growth factor via ATF4 and ATF6β: Implications for the pathophysiology of human pregnancy complications. J Pathol 238: 550–561, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228.Moffett A, Colucci F, Moffett A, Colucci F. Uterine NK cells: Active regulators at the maternal-fetal interface. J Clin Invest 124: 1872–1879, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229.Mogos MF, Piano MR, McFarlin BL, Salemi JL, Liese KL, Briller JE. Heart failure in pregnant women. Circ Heart Fail 11: e004005, 2018. [DOI] [PubMed] [Google Scholar]
  • 230.Molino P, Veglio F, Cat Genova G, Melchio R, Benedetto C, Chiarolini L, Rabbia F, Grosso T, Mulatero P, Chiandussi L. Baroreflex control of heart rate is impaired in pre-eclampsia. J Hum Hypertens 13: 179–183, 1999. [DOI] [PubMed] [Google Scholar]
  • 231.Moors S, Staaks KJJ, Westerhuis MEMH, Dekker LRC, Verdurmen KMJ, Oei SG, van Laar JOEH. Heart rate variability in hypertensive pregnancy disorders: A systematic review. Pregnancy Hypertens 20: 56–68, 2020. [DOI] [PubMed] [Google Scholar]
  • 232.Morisco F, Bruno R, Bugianesi E, Burra P, Calvaruso V, Cannoni A, Caporaso N, Caviglia GP, Ciancio A, Fargion S, Federico A, Floreani A, Gaeta GB, Guarino M, Invernizzi P, Licata A, Loguercio C, Mazzella G, Petraglia F, Primignani M, Rodriguez-Castro K, Smedile A, Valenti L, Vannic E, Vannuccini S, Voltolini C, Villa E. AISF position paper on liver disease and pregnancy. Dig Liver Dis 48: 120–137, 2016. [DOI] [PubMed] [Google Scholar]
  • 233.Moutquin JM, Rainville C, Giroux L, Raynauld P, Amyot G, Bilodeau R, Pelland N. A prospective study of blood pressure in pregnancy: Prediction of preeclampsia. Am J Obstet Gynecol 151: 191–196, 1985. [DOI] [PubMed] [Google Scholar]
  • 234.Murai JT, Muzykanskiy E, Taylor RN. Maternal and fetal modulators of lipid metabolism correlate with the development of preeclampsia. Metabolism 46: 963–967, 1997. [DOI] [PubMed] [Google Scholar]
  • 235.Murphy MP. How mitochondria produce reactive oxygen species. Biochem J 417: 1–13, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 236.Murphy S, LaMarca B, Cockrell K, Granger J. Role of endothelin in mediating soluble fms-like tyrosine kinase-1 (sFlt-1)-induced hypertension in pregnant rats Sydney. Hypertension 55: 394–398, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 237.Murphy SR, LaMarca B, Cockrell K, Arany M, Granger JP. L-Arginine supplementation abolishes the blood pressure and endothelin response to chronic increases in plasma sFlt-1 in pregnant rats. Am J Phys Regul Integr Comp Phys 302: R259–R263, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 238.Nahum Sacks K, Friger M, Shoham-Vardi I, Sergienko R, Spiegel E, Landau D, Sheiner E. Long-term neuropsychiatric morbidity in children exposed prenatally to preeclampsia. Early Hum Dev 130: 96–100, 2019. [DOI] [PubMed] [Google Scholar]
  • 239.Nahum Sacks K, Friger M, Shoham-Vardi I, Spiegel E, Sergienko R, Landau D, Sheiner E. Prenatal exposure to preeclampsia as an independent risk factor for long-term cardiovascular morbidity of the offspring. Pregnancy Hypertens 13: 181–186, 2018. [DOI] [PubMed] [Google Scholar]
  • 240.Nakamura T, Kawabe K, Sapru HN. Cold pressor test in the rat: Medullary and spinal pathways and neurotransmitters. Am J Physiol Heart Circ Physiol 295: H1780–H1787, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 241.Naljayan MV, Karumanchi SA. New developments in the pathogenesis of preeclampsia. Adv Chronic Kidney Dis 20: 265–270, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 242.Naruse K, Lash GE, Innes BA, Otun HA, Searle RF, Robson SC, Bulmer JN. Localization of matrix metalloproteinase (MMP)-2, MMP-9 and tissue inhibitors for MMPs (TIMPs) in uterine natural killer cells in early human pregnancy. Hum Reprod 24: 553–561, 2009. [DOI] [PubMed] [Google Scholar]
  • 243.Navaratnam K, Alfirevic Z, Baker PN, Gluud C, Grüttner B, Kublickiene K, Zeeman G, Kenny LC. A multi-centre phase IIa clinical study of predictive testing for preeclampsia: Improved pregnancy outcomes via early detection (IMPROvED). BMC Pregnancy Childb 13: 226, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 244.Neri I, Monari F, Sgarbi L, Berardi A, Masellis G, Facchinetti F. L-Arginine supplementation in women with chronic hypertension: Impact on blood pressure and maternal and neonatal complications. J Matern-Fetal Neonatal Med 23: 1456–1460, 2010. [DOI] [PubMed] [Google Scholar]
  • 245.North RA, McCowan LME, Dekker GA, Poston L, Chan EHY, Stewart AW, Black MA, Taylor RS, Walker JJ, Baker PN, Kenny LC. Clinical risk prediction for pre-eclampsia in nulliparous women: Development of model in international prospective cohort. BMJ 342: d1875, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 246.Nugent WH, Mishra N, Strauss JF, Walsh SW. Matrix metalloproteinase 1 causes vasoconstriction and enhances vessel reactivity to angiotensin ii via protease-activated receptor 1. Reprod Sci 23: 542–548, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 247.Oesterle A, Laufs U, Liao JK. Pleiotropic effects of statins on the cardiovascular system. Circ Res 120: 229–243, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 248.Oian P, Lande K, Kjeldsen S, Gjesdal K, Aakesson I, Eide I, Maltau J. Enhanced platelet release reaction related to arterial plasma adrenaline and blood pressure in pre-eclampsia. BJOG 93: 548–553, 1986. [DOI] [PubMed] [Google Scholar]
  • 249.Ojeda NB, Grigore D, Alexander BT. Developmental programming of hypertension: Insight from animal models of nutritional manipulation. Hypertension 52: 44–50, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 250.Ojeda NB, Hennington BS, Williamson DT, Hill ML, Betson NEE, Sartori-Valinotti JC, Reckelhoff JF, Royals TP, Alexander BT. Oxidative stress contributes to sex differences in blood pressure in adult growth-restricted offspring. Hypertension 60: 114–122, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 251.Ojeda NB, Johnson WR, Dwyer TM, Alexander BT. Early renal denervation prevents development of hypertension in growth-restricted offspring. Clin Exp Pharmacol Physiol 34: 1212–1216, 2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 252.Oka K, Sawamura T, Kikuta KI, Itokawa S, Kume N, Kita T, Masaki T. Lectin-like oxidized low-density lipoprotein receptor 1 mediates phagocytosis of aged/apoptotic cells in endothelial cells. Proc Natl Acad Sci U S A 95: 9535–9540, 1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 253.Oludare GO, Jinadu HD, Aro OO. L-Arginine attenuates blood pressure and reverses the suppression of angiogenic risk factors in a rat model of preeclampsia. Pathophysiology 25: 389–395, 2018. [DOI] [PubMed] [Google Scholar]
  • 254.Paauw ND, Joles JA, Spradley FT, Bakrania B, Zellenger ZK, Franx A, Verhaar MC, Granger JP, Lely AT. Exposure to placental ischemia impairs postpartum maternal renal and cardiac function in rats. Am J Phys Regul Integr Comp Phys 312: R664–R670, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 255.Paiva SPC, Veloso CA, Campos FFC, Carneiro MM, Tilan JU, Wang H, Umans JG, Zukowska Z, Kitlinska J. Elevated levels of neuropeptide Y in preeclampsia: A pilot study implicating a role for stress in pathogenesis of the disease. Neuropeptides 55: 127–135, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 256.Palacios C, Trak-Fellermeier MA, Martinez RX, Lopez-Perez L, Lips P, Salisi JA, John JC, Peña-Rosas JP. Regimens of vitamin D supplementation for women during pregnancy. Cochrane Database Syst Rev 10: CD013446, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 257.Pantham P, Viall CA, Chen Q, Kleffmann T, Print CG, Chamley LW. Antiphospholipid antibodies bind syncytiotrophoblast mitochondria and alter the proteome of extruded syncytial nuclear aggregates. Placenta, 2015. DOI: 10.1016/j.placenta.2015.10.006. [DOI] [PubMed] [Google Scholar]
  • 258.Patten IS, Rana S, Shahul S, Rowe GC, Jang C, Liu L, Hacker MR, Rhee JS, Mitchell J, Mahmood F, Hess P, Farrell C, Koulisis N, Khankin EV, Burke SD, Tudorache I, Bauersachs J, del Monte F, Hilfiker-Kleiner D, Karumanchi SA, Arany Z. Cardiac angiogenic imbalance leads to peripartum cardiomyopathy. Nature 485: 333–338, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 259.Peet M, Isberg E, Bassett R. Toxemia superimposed upon prepregnant hypertension treated by. Surg Gynecol Obstet 86: 673–679, 1948. [PubMed] [Google Scholar]
  • 260.Pels A, Kenny LC, Alfirevic Z, Baker PN, von Dadelszen P, Gluud C, Kariya CT, Mol BW, Papageorghiou A, van Wassenaer-Leemhuis AG, Ganzevoort JW, Groom KM, McCowan LM, Stone PR, Lee A, Mackay L, Oyston C, Gardener G, Khashan A, Eustace J, Dempsey E, Jackson R, Dickinson J, Gill A, Muller P, Sekar R, Reid RA, Unterschneider J, Welsh A, Marlow J, Hyett J, Walker S, Morris J, Watson D, McKinlay C, Harris S, Lim KI, Lalji S, Magee LA, Lee T, Li J, Hutfield A, Ansermino M, Robinson W, Singer J, Synnes AR, Burrows J, Audibert F, Bujold E, Piedboeuf B, Davidge ST, Seaward GR, Dwinnell S, Gagnon R, Gaudet L, Young C, Murphy D, Daly S, McAuliffe F, Malone F, Breathnach F, O’Donoghue K, Al-Nasiry S, de Boer MA, de Groot CJM, Sueters M, Derks JB, van Drongelen J, Duvekot HJ, Elvan-Taspinar A, van Eyck J, van Laar J, Morssink LP, Price L, Astor A, Hardman L, Sharp A, Turner M, Cameron A, Draper E, Clarke P, McKelvey A, Masson G, Aquilin J, Johnstone E, Bugg G, Howe D, Patni S, Mousa H, Russell H, Hannon T, Kilby M, David A, Cohen K, Impey L, Stock S, Poon L, Pasupath D, Khalil A. STRIDER (Sildenafil TheRapy in dismal prognosis early onset fetal growth restriction): An international consortium of randomised placebo-controlled trials. BMC Pregnancy Childb 17: 440, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 261.Penning M, Chua JS, Van Kooten C, Zandbergen M, Buurma A, Schutte J, Bruijn JA, Khankin EV, Bloemenkamp K, Karumanchi SA, Baelde H. Classical complement pathway activation in the kidneys of women with preeclampsia. Hypertension 66: 117–125, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 262.Perkins AV, Vanderlelie JJ. Multiple micronutrient supplementation and birth outcomes: The potential importance of selenium. Placenta Suppl 1: s61–s65, 2016. [DOI] [PubMed] [Google Scholar]
  • 263.Perper JA, Patterson GT, Backner JS. Face imaging reconstructive morphography. A new method for physiognomic reconstruction. Am J Forensic Med Pathol 9: 126–138, 1988. [DOI] [PubMed] [Google Scholar]
  • 264.Pijnenborg R, Dixon G, Robertson WB, Brosens I. Trophoblastic invasion of human decidua from 8 to 18 weeks of pregnancy. Placenta 1: 3–19, 1980. [DOI] [PubMed] [Google Scholar]
  • 265.Plaks V, Rinkenberger J, Dai J, Flannery M, Sund M, Kanasaki K, Ni W, Kalluri R, Werb Z. Matrix metalloproteinase-9 deficiency phenocopies features of preeclampsia and intrauterine growth restriction. Proc Natl Acad Sci U S A 110: 11109–11114, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 266.Poon LCY, Kametas NA, Chelemen T, Leal A, Nicolaides KH. Maternal risk factors for hypertensive disorders in pregnancy: A multivariate approach. J Hum Hypertens 24: 104–110, 2010. [DOI] [PubMed] [Google Scholar]
  • 267.Powe CE, Levine RJ, Karumanchi SA. Preeclampsia, a disease of the maternal endothelium: The role of antiangiogenic factors and implications for later cardiovascular disease. Circulation 123: 2856–2869, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 268.Qiu C, Coughlin KB, Frederick IO, Sorensen TK, Williams MA. Dietary fiber intake in early pregnancy and risk of subsequent preeclampsia. Am J Hypertens 21: 903–909, 2008. [DOI] [PubMed] [Google Scholar]
  • 269.Radenkovic F, Holland O, Vanderlelie JJ, Perkins AV. Selective inhibition of endogenous antioxidants with Auranofin causes mitochondrial oxidative stress which can be countered by selenium supplementation. Biochem Pharmacol 146: 42–52, 2017. [DOI] [PubMed] [Google Scholar]
  • 270.Radke AL, Reynolds LE, Melo RCN, Dvorak AM, Weller PF, Spencer LA. Mature human eosinophils express functional Notch ligands mediating eosinophil autocrine regulation. Blood 113: 3092–3101, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 271.Rahman R, Murthi P, Singh H, Gurusinghe S, Mockler JC, Lim R, Wallace EM. The effects of hydroxychloroquine on endothelial dysfunction. Pregnancy Hypertens 6: 259–262, 2016. [DOI] [PubMed] [Google Scholar]
  • 272.Rahman TM. Severe hepatic dysfunction in pregnancy. QJM 95: 343–357, 2002. [DOI] [PubMed] [Google Scholar]
  • 273.Rai A, Cross JC. Development of the hemochorial maternal vascular spaces in the placenta through endothelial and vasculogenic mimicry. Dev Biol 387: 131–141, 2014. [DOI] [PubMed] [Google Scholar]
  • 274.Rajakumar A, Brandon HM, Daftary A, Ness R, Conrad KP. Evidence for the functional activity of hypoxia-inducible transcription factors overexpressed in preeclamptic placentae. Placenta 25: 763–769, 2004. [DOI] [PubMed] [Google Scholar]
  • 275.Ramesar SV, MacKraj I, Gathiram P, Moodley J. Sildenafil citrate decreases sFlt-1 and sEng in pregnant l-NAME treated Sprague-Dawley rats. Eur J Obstet Gynecol Reprod Biol 157: 136–140, 2011. [DOI] [PubMed] [Google Scholar]
  • 276.Rana S, Lemoine E, Granger J, Karumanchi SA. Preeclampsia: Pathophysiology, challenges, and perspectives. Circ Res 124: 1094–1112, 2019. [DOI] [PubMed] [Google Scholar]
  • 277.Redman CWG, Sacks GP, Sargent IL. Preeclampsia: An excessive maternal inflammatory response to pregnancy. Am J Obstet Gynecol 180: 499–506, 1999. [DOI] [PubMed] [Google Scholar]
  • 278.Redman EK, Hauspurg A, Hubel CA, Roberts JM, Jeyabalan A. Clinical course, associated factors, and blood pressure profile of delayedonset postpartum preeclampsia. Obstet Gynecol 134: 995–1001, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 279.Reyes LM, Usselman CW, Davenport MH, Steinback CD. Sympathetic nervous system regulation in human normotensive and hypertensive pregnancies. Hypertension 71: 793–803, 2018. [DOI] [PubMed] [Google Scholar]
  • 280.Reynolds LP, Caton JS, Redmer DA, Grazul-Bilska AT, Vonnahme KA, Borowicz PP, Luther JS, Wallace JM, Wu G, Spencer TE. Evidence for altered placental blood flow and vascularity in compromised pregnancies. J Physiol 572: 51–58, 2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 281.Rigó J, Visontai Z, Mersich B, Kollai M. Reduced baroreceptor sensitivity and elasticity of the carotid artery in a preeclamptic patient. Gynecol Obstet Investig 60: 84–86, 2005. [DOI] [PubMed] [Google Scholar]
  • 282.Roberts JM, Druzin M, August PA, Gaiser RR, Bakris G, Granger JP, Barton JR, Jeyabalan A, Bernstein IA, Johnson DD, Karamanchi SA, Spong CY, Lindheiner MD, Tsingas E, Owens MY, Martin JN Jr, Saade GR, Sibai BM. Hypertension in pregnancy. Report of the American College of Obstetricians and Gynecologists’ Task Force on Hypertension in Pregnancy. Obstet Gynecol 122 (5): 1122–1131, 2013. [DOI] [PubMed] [Google Scholar]
  • 283.Roberts JM, Redman CWG. Pre-eclampsia: More than pregnancy-induced hypertension. Lancet 341: 1447–1451, 1993. [DOI] [PubMed] [Google Scholar]
  • 284.Roberts JM, Taylor RN, Musci TJ, Rodgers GM, Hubel CA, McLaughlin MK. Preeclampsia: An endothelial cell disorder. Am J Obstet Gynecol 161: 1200–1204, 1989. [DOI] [PubMed] [Google Scholar]
  • 285.Robillard P-Y, Dekker G, Chaouat G, Le Bouteiller P, Scioscia M, Hulsey TC. Preeclampsia and the 20th century: “Le siècle des Lumières”. Pregnancy Hypertens 13: 107–109, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 286.Robillard PY, Dekker G, Iacobelli S, Chaouat G. An essay of reflection: Why does preeclampsia exist in humans, and why are there such huge geographical differences in epidemiology? J Reprod Immunol 114: 44–47, 2016. [DOI] [PubMed] [Google Scholar]
  • 287.Robillard PY, Périanin J, Janky E, Miri EH, Hulsey TC, Papiernik E. Association of pregnancy-induced hypertension with duration of sexual cohabitation before conception. Lancet 344: 973–975, 1994. [DOI] [PubMed] [Google Scholar]
  • 288.Robson SC, Hunter S, Boys RJ, Dunlop W. Serial study of factors influencing changes in cardiac output during human pregnancy. Am J Physiol Heart Circ Physiol 256: H1060–H1065, 1989. [DOI] [PubMed] [Google Scholar]
  • 289.Roh JD, Hobson R, Chaudhari V, Quintero P, Yeri A, Benson M, Xiao C, Zlotoff D, Bezzerides V, Houstis N, Platt C, Damilano F, Lindman BR, Elmariah S, Biersmith M, Lee SJ, Seidman CE, Seidman JG, Gerszten RE, Lach-Trifilieff E, Glass DJ, Rosenzweig A. Activin type II receptor signaling in cardiac aging and heart failure. Sci Transl Med 11: 1–16, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 290.Rossant J, Cross JC. Placental development: Lessons from mouse mutants. Nat Rev Genet 2: 538–548, 2001. [DOI] [PubMed] [Google Scholar]
  • 291.Ryan MJ, Gilbert EL, Glover PH, George EM, Warren Masterson C, McLemore GR, Lamarca B, Granger JP, Drummond HA. Placental ischemia impairs middle cerebral artery myogenic responses in the pregnant rat. Hypertension 58: 1126–1131, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 292.Saad AF, Diken ZM, Kechichian TB, Clark SM, Olson GL, Saade GR, Costantine MM. Pravastatin effects on placental prosurvival molecular pathways in a mouse model of preeclampsia. Reprod Sci 23: 1593–1599, 2016. [DOI] [PubMed] [Google Scholar]
  • 293.Saito S, Sakai M. Th1/Th2 balance in preeclampsia. J Reprod Immunol 59: 161–173, 2003. [DOI] [PubMed] [Google Scholar]
  • 294.Saito S, Shiozaki A, Sasaki Y, Nakashima A, Shima T, Ito M. Regulatory T cells and regulatory natural killer (NK) cells play important roles in feto-maternal tolerance. Semin Immunopathol 29: 115–122, 2007. [DOI] [PubMed] [Google Scholar]
  • 295.San-Frutos L, Engels V, Zapardiel I, Perez-Medina T, Almagro-Martinez J, Fernandez R, Bajo-Arenas JM. Hemodynamic changes during pregnancy and postpartum: A prospective study using thoracic electrical bioimpedance. J Matern-Fetal Neonatal Med 24: 1333–1340, 2011. [DOI] [PubMed] [Google Scholar]
  • 296.Sanghavi M, Rutherford JD. Cardiovascular physiology of pregnancy. Circulation 130: 1003–1008, 2014. [DOI] [PubMed] [Google Scholar]
  • 297.Schmid J, Müller B, Heppeler D, Gaynullina D, Kassmann M, Gagov H, Mladenov M, Gollasch M, Schubert R. The unexpected role of calcium-activated potassium channels: Limitation of no-induced arterial relaxation. J Am Heart Assoc 7: e007808, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 298.Schobel HP, Fischer T, Heuszer K, Geiger H, Schmieder RE. Preeclampsia - A state of sympathetic overactivity. N Engl J Med 335: 1480–1485, 1996. [DOI] [PubMed] [Google Scholar]
  • 299.Sedeek M, Gilbert JS, Lamarca BB, Sholook M, Derrick L, Wang Y, Granger JP. Role of reactive oxygen specied in hypertension produced by reduced uterine perfusion pressure in pregnant rats. Am J Hypertens 21: 1152–1156, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 300.Seppä S, Voutilainen R, Tenhola S. Markers of insulin sensitivity in 12-year-old children born from preeclamptic pregnancies. J Pediatr 167: 125–130, 2015. [DOI] [PubMed] [Google Scholar]
  • 301.Shahul S, Ramadan H, Nizamuddin J, Mueller A, Patel V, Dreixler J, Tung A, Lang RM, Weinert L, Nasim R, Chinthala S, Rana S. Activin A and late postpartum cardiac dysfunction among women with hypertensive disorders of pregnancy novelty and significance. Hypertension 72: 188–193, 2018. [DOI] [PubMed] [Google Scholar]
  • 302.Shahul S, Rhee J, Hacker MR, Gulati G, Mitchell JD, Hess P, Mahmood F, Arany Z, Rana S, Talmor D. Subclinical left ventricular dysfunction in preeclamptic women with preserved left ventricular ejection fraction: A 2D speckle-tracking imaging study. Circ Cardiovasc Imaging 5: 734–739, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 303.Sharp A, Cornforth C, Jackson R, Harrold J, Turner MA, Kenny LC, Baker PN, Johnstone ED, Khalil A, von Dadelszen P, Papageorghiou AT, Alfirevic Z, Agarwal U, Willis E, Mammarella S, Masson G, Aquilina J, Greco E, Higgins S, Vinayagam D, Shaw L, Stephens L, Howe D, Rand A, Patni S, Mousa T, Rabab A, Russell H, Hannon T, Fenn A, Kilby M, Selman T, David A, Spencer R, Cohen K, Breeze A, McKelvey A, Impey L, Loannou C, Stock S, Poon L, Pasupathy D, Webster L, Bugg G. Maternal sildenafil for severe fetal growth restriction (STRIDER): A multicentre, randomised, placebo-controlled, double-blind trial. Lancet Child Adolesc Health 2: 90, 2018. [DOI] [PubMed] [Google Scholar]
  • 304.Shekhar S, Diddi G. Liver disease in pregnancy. Taiwan J Obstet Gynecol 54: 475–482, 2015. [DOI] [PubMed] [Google Scholar]
  • 305.Shimaoka T, Kume N, Minami M, Hayashida K, Sawamura T, Kita T, Yonehara S. LOX-1 supports adhesion of gram-positive and gramnegative bacteria. J Immunol 166: 5108–5114, 2001. [DOI] [PubMed] [Google Scholar]
  • 306.Sibai BM. Etiology and management of postpartum hypertension-preeclampsia. Am J Obstet Gynecol 206: 470–475, 2012. [DOI] [PubMed] [Google Scholar]
  • 307.Sibai BM, Koch MA, Freire S, Pinto e Silva JL, Rudge MVC, Martins-Costa S, Bartz J, de Barros Santos C, Cecatti JG, Costa R, Ramos JG, Spinnato JA. Serum inhibin A and angiogenic factor levels in pregnancies with previous preeclampsia and/or chronic hypertension: Are they useful markers for prediction of subsequent preeclampsia? Am J Obstet Gynecol 199: 268.e1–268.e9, 2008. [DOI] [PubMed] [Google Scholar]
  • 308.Silva D, Colvin L, Hagemann E, Bower C. Environmental risk factors by gender associated with attention-deficit/hyperactivity disorder. Pediatrics 133: e14–e22, 2014. [DOI] [PubMed] [Google Scholar]
  • 309.Silverberg O, Park AL, Cohen E, Fell DB, Ray JG. Premature cardiac disease and death inwomen whose infantwas preterm and small for gestational age a retrospective cohort study. JAMA Cardiol 3: 247–251, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 310.Simmons DG, Fortier AL, Cross JC. Diverse subtypes and developmental origins of trophoblast giant cells in the mouse placenta. Dev Biol 304: 567–578, 2007. [DOI] [PubMed] [Google Scholar]
  • 311.Simmons DG, Natale DRC, Begay V, Hughes M, Leutz A, Cross JC. Early patterning of the chorion leads to the trilaminar trophoblast cell structure in the placental labyrinth. Development 135: 2083–2091, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 312.Sladek SM, Magness RR, Conrad KP. Nitric oxide and pregnancy. Am J Phys Regul Integr Comp Phys 272: R441–R463, 1997. [DOI] [PubMed] [Google Scholar]
  • 313.Soares MJ, Chakraborty D, Karim Rumi MA, Konno T, Renaud SJ. Rat placentation: An experimental model for investigating the hemochorial maternal-fetal interface. Placenta 33: 233–243, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 314.Soares MJ, Varberg KM, Iqbal K. Hemochorial placentation: Development, function, and adaptations. Biol Reprod 99: 196–211, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 315.Song J, Li Y, An R. Vitamin D restores angiogenic balance and decreases tumor necrosis factor-α in a rat model of pre-eclampsia. J Obstet Gynaecol Res 43: 42–49, 2017. [DOI] [PubMed] [Google Scholar]
  • 316.Spaans F, Quon A, Rowe SR, Morton JS, Kirschenman R, Sawamura T, Tannetta DS, Sargent IL, Davidge ST. Alterations in vascular function by syncytiotrophoblast extracellular vesicles via lectin-like oxidized low-density lipoprotein receptor-1 in mouse uterine arteries. Clin Sci 132: 2369–2381, 2018. [DOI] [PubMed] [Google Scholar]
  • 317.Spradley F. Exaggerated placental ischemia-induced hypertension in endothelin receptor type B (ETB)-deficient pregnant rats S independent of increased SFLT-1 or ros levels. Hypertension 70: A037, 2017. [Google Scholar]
  • 318.Spradley FT. Metabolic abnormalities and obesity’s impact on the risk for developing preeclampsia. Am J Phys Regul Integr Comp Phys 312: R5–R12, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 319.Spradley FT. Sympathetic nervous system control of vascular function and blood pressure during pregnancy and preeclampsia. J Hypertens 37: 476–487, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 320.Spradley FT, Ge Y, Granger JP, Chade AR. Utero-placental vascular remodeling during late gestation in Sprague-Dawley rats. Pregnancy Hypertens 20: 36–43, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 321.Spradley FT, Ge Y, Haynes BP, Granger JP, Anderson CD. Adrenergic receptor blockade attenuates placental ischemia-induced hypertension. Phys Rep 6: e13814, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 322.Spradley FT, Palei AC, Granger JP. Increased risk for the development of preeclampsia in obese pregnancies: Weighing in on the mechanisms. Am J Phys Regul Integr Comp Phys 309: R1326–R1343, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 323.Spradley FT, Tan AY, Joo WS, Daniels G, Kussie P, Karumanchi SA, Granger JP. Placental growth factor administration abolishes placental ischemia-induced hypertension. Hypertension 67: 740–747, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 324.Stanhewicz AE, Jandu S, Santhanam L, Alexander LM. Increased angiotensin II sensitivity contributes to microvascular dysfunction in women who have had preeclampsia. Hypertension 70: 382–389, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 325.Sugerman HJ. Effect of obesity on incidence of preeclampsia. Am J Obstet Gynecol 210: 375, 2014. [DOI] [PubMed] [Google Scholar]
  • 326.Sugimoto H, Hamanog Y, Charytan D, Cosgrove D, Kieran M, Sudhakar A, Kalluri R. Neutralization of circulating vascular endothelial growth factor (VEGF) by anti-VEGF antibodies and soluble VEGF receptor 1 (sFlt-1) induces proteinuria. J Biol Chem 278: 12605–12608, 2003. [DOI] [PubMed] [Google Scholar]
  • 327.Swansburg ML, Brown CA, Hains SM, Smith GN, Kisilevsky BS. Maternal cardiac autonomic function and fetal heart rate in preeclamptic compared to normotensive pregnancies. Can J Cardiovasc Nurs 15: 42–52, 2005. [PubMed] [Google Scholar]
  • 328.Takiuti NH, Kahhale S, Zugaib M. Stress in pregnancy: A new Wistar rat model for human preeclampsia. Am J Obstet Gynecol 186: 544–550, 2002. [DOI] [PubMed] [Google Scholar]
  • 329.Tam Tam KB, George E, Cockrell K, Arany M, Speed J, Martin JN, Lamarca B, Granger JP. Endothelin type A receptor antagonist attenuates placental ischemiainduced hypertension and uterine vascular resistance. Am J Obstet Gynecol 204: 330.e1–330.e4, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 330.Tan EK, Tan EL. Alterations in physiology and anatomy during pregnancy. Best Pract Res Clin Obstet Gynaecol 27: 791–802, 2013. [DOI] [PubMed] [Google Scholar]
  • 331.Tanaka M, Jaamaa G, Kaiser M, Hills E, Soim A, Zhu M, Shcherbatykh IY, Samelson R, Bell E, Zdeb M, McNutt LA. Racial disparity in hypertensive disorders of pregnancy in New York state: A 10-year longitudinal population-based study. Am J Public Health 97: 163–170, 2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 332.Tangren JS, Wan Md Adnan WAH, Powe CE, Ecker J, Bramham K, Hladunewich MA, Ankers E, Karumanchi SA, Thadhani R. Risk of preeclampsia and pregnancy complications in women with a history of acute kidney injury. Hypertension 72: 451–459, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 333.Taylor RN, Varma M, Teng NNH, Roberts JM. Women with preeclampsia have higher plasma endothelin levels than women with normal pregnancies. J Clin Endocrinol Metab 71: 1675–1677, 1990. [DOI] [PubMed] [Google Scholar]
  • 334.Tenório MB, Ferreira RC, Moura FA, Bueno NB, Goulart MOF, Oliveira ACM. Oral antioxidant therapy for prevention and treatment of preeclampsia: Meta-analysis of randomized controlled trials. Nutr Metab Cardiovasc Dis 28: 865–876, 2018. [DOI] [PubMed] [Google Scholar]
  • 335.Terstappen F, Spradley FT, Bakrania BA, Clarke SM, Joles JA, Paauw ND, Garrett MR, Lely AT, Sasser JM. Prenatal sildenafil therapy improves cardiovascular function in fetal growth restricted offspring of dahl salt-sensitive rats. Hypertension 73: 1120–1127, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 336.Thadhani R, Hagmann H, Schaarschmidt W, Roth B, Cingoez T, Karumanchi SA, Wenger J, Lucchesi KJ, Tamez H, Lindner T, Fridman A, Thome U, Kribs A, Danner M, Hamacher S, Mallmann P, Stepan H, Benzing T. Removal of soluble fms-like tyrosine kinase-1 by dextran sulfate apheresis in preeclampsia. J Am Soc Nephrol 27: 903–913, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 337.Thadhani R, Kisner T, Hagmann H, Bossung V, Noack S, Schaarschmidt W, Jank A, Kribs A, Cornely OA, Kreyssig C, Hemphill L, Rigby AC, Khedkar S, Lindner TH, Mallmann P, Stepan H, Karumanchi SA, Benzing T. Pilot study of extracorporeal removal of soluble Fms-like tyrosine kinase 1 in preeclampsia. Circulation 124: 940–950, 2011. [DOI] [PubMed] [Google Scholar]
  • 338.Theilen LH, Fraser A, Hollingshaus MS, Schliep KC, Varner MW, Smith KR, Esplin MS. All-cause and cause-specific mortality after hypertensive disease of pregnancy. Obstet Gynecol 128: 238–244, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 339.Thilaganathan B, Kalafat E. Cardiovascular system in preeclampsia and beyond. Hypertension 73: 522–531, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 340.Thornburg KL, Jacobson SL, Giraud GD, Morton MJ. Hemodynamic changes in pregnancy. Semin Perinatol 24: 11–14, 2000. [DOI] [PubMed] [Google Scholar]
  • 341.Tian X, Ma S, Wang Y, Hou L, Shi Y, Yao M, Wang X, Zhang H, Jiang L. Effects of placental ischemia are attenuated by 1,25-dihydroxyvitamin D treatment and associated with reduced apoptosis and increased autophagy. DNA Cell Biol 35: 1–12, 2015. [DOI] [PubMed] [Google Scholar]
  • 342.Travis OK, White D, Pierce WA, Ge Y, Stubbs CY, Spradley FT, Williams JM, Cornelius DC. Chronic infusion of interleukin-17 promotes hypertension, activation of cytolytic natural killer cells, and vascular dysfunction in pregnant rats. Phys Rep 7: e14038, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 343.Tremellen KP, Seamark RF, Robertson SA. Seminal transforming growth factor β1, stimulates granulocyte-macrophage colonystimulating factor production and inflammatory cell recruitment in the Murine Uterus1. Biol Reprod 58: 1217–1225, 1998. [DOI] [PubMed] [Google Scholar]
  • 344.Tuovinen S, Räikkönen K, Kajantie E, Pesonen AK, Heinonen K, Osmond C, Barker DJP, Eriksson JG. Depressive symptoms in adulthood and intrauterine exposure to pre-eclampsia: The Helsinki birth cohort study. BJOG 117: 1236–1242, 2010. [DOI] [PubMed] [Google Scholar]
  • 345.Turbeville HR, Taylor EB, Garrett MR, Didion SP, Ryan MJ, Sasser JM. Superimposed preeclampsia exacerbates postpartum renal injury despite lack of long-term blood pressure difference in the dahl salt-sensitive rat. Hypertension 73: 650–658, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 346.Uzan J, Carbonnel M, Piconne O, Asmar R, Ayoubi JM. Pre-eclampsia: Pathophysiology, diagnosis, and management. Vasc Health Risk Manag 7: 467–474, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 347.Van Pampas MG, Wolf H, Ilsen A, Treffers PE. Maternal outcome following temporizing management of the (H)ELLP syndrome. Hypertens Pregnancy 19: 211–220, 2000. [DOI] [PubMed] [Google Scholar]
  • 348.Van Pampus MG, Wolf H, Westenberg SM, Van Der Post JAM, Bonsel GJ, Treffers PE. Maternal and perinatal outcome after expectant management of the HELLP syndrome compared with pre-eclampsia without HELLP syndrome. Eur J Obstet Gynecol Reprod Biol 76: 31–36, 1998. [DOI] [PubMed] [Google Scholar]
  • 349.Van Veen TR, Panerai RB, Haeri S, Griffioen AC, Zeeman GG, Belfort MA. Cerebral autoregulation in normal pregnancy and preeclampsia. Obstet Gynecol 122: 1064–1069, 2013. [DOI] [PubMed] [Google Scholar]
  • 350.Vasapollo B, Novelli GP, Gagliardi G, Farsetti D, Valensise H. Pregnancy complications in chronic hypertensive patients are linked to pre-pregnancy maternal cardiac function and structure. Am J Obstet Gynecol 223 (3): 425.e1–425.e13, 2020. [DOI] [PubMed] [Google Scholar]
  • 351.Veillon EW, Keiser SD, Parrish MR, Bennett W, Cockrell K, Ray LF, Granger JP, Martin JN, LaMarca B. 17-Hydroxyprogesterone blunts the hypertensive response associated with reductions in uterine perfusion pressure in pregnant rats. Am J Obstet Gynecol 201: 324.e1–324.e6, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 352.Verdonk K, Saleh L, Lankhorst S, Smilde JEI, Van Ingen MM, Garrelds IM, Friesema ECH, Russcher H, Van Den Meiracker AH, Visser W, Danser AHJ. Association studies suggest a key role for endothelin-1 in the pathogenesis of preeclampsia and the accompanying renin-angiotensin-aldosterone system suppression. Hypertension 65: 1316–1323, 2015. [DOI] [PubMed] [Google Scholar]
  • 353.Verson-Hoynck F, Schaub A, Chi Y-Y, Chiu K-H, Liu J, Lingis M, Williams R, Rhoton-Vlasak A, Nichols W, Fleischmann R, Zhang W, Winn V, Segal M, Conrad K, Baker V. Increased preeclampsia risk and reduced aortic compliance with in vitro fertilization cycles in the absence of a corpus luteum. Hypertension 73: 640–649, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 354.Viall CA, Chen Q, Liu B, Hickey A, Snowise S, Salmon JE, Stone PR, Chamley LW. Antiphospholipid antibodies internalised by human syncytiotrophoblast cause aberrant cell death and the release of necrotic trophoblast debris. J Autoimmun 47: 45–57, 2013. [DOI] [PubMed] [Google Scholar]
  • 355.Vishnyakova PA, Volodina MA, Tarasova NV, Marey MV, Tsvirkun DV, Vavina OV, Khodzhaeva ZS, Kan NE, Menon R, Vysokikh MY, Sukhikh GT. Mitochondrial role in adaptive response to stress conditions in preeclampsia. Sci Rep 30: 32410, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 356.Walker CK, Krakowiak P, Baker A, Hansen RL, Ozonoff S, Hertz-Picciotto I. Preeclampsia, placental insufficiency, and autism spectrum disorder or developmental delay. JAMA Pediatr 169: 154–162, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 357.Wallace K, Richards S, Dhillon P, Weimer A, Edholm ES, Bengten E, Wilson M, Martin JN, Lamarca B. CD4+ T-helper cells stimulated in response to placental ischemia mediate hypertension during pregnancy. Hypertension 57: 949–955, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 358.Wallis AB, Saftlas AF, Hsia J, Atrash HK. Secular trends in the rates of preeclampsia, eclampsia, and gestational hypertension, United States, 1987–2004. Am J Hypertens 21: 521–526, 2008. [DOI] [PubMed] [Google Scholar]
  • 359.Wallukat G, Homuth V, Fischer T, Lindschau C, Horstkamp B, Jüpner A, Baur E, Nissen E, Vetter K, Neichel D, Dudenhausen JW, Haller H, Luft FC. Patients with preeclampsia develop agonistic autoantibodies against the angiotensin AT1 receptor. J Clin Invest 103: 945–952, 1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 360.Wang A, Rana S, Karumanchi SA. Preeclampsia: The role of angiogenic factors in its pathogenesis. Physiology (Bethesda) 24: 147–158, 2009. [DOI] [PubMed] [Google Scholar]
  • 361.Waring P, Müllbacher A. Cell death induced by the Fas/Fas ligand pathway and its role in pathology. Immunol Cell Biol 77: 312–317, 1999. [DOI] [PubMed] [Google Scholar]
  • 362.Warrington JP, Coleman K, Skaggs C, Hosick PA, George EM, Stec DE, Ryan MJ, Granger JP, Drummond HA. Heme oxygenase-1 promotes migration and β-epithelial Na+ channel expression in cytotrophoblasts and ischemic placentas. Am J Phys Regul Integr Comp Phys 306: R641–R646, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 363.Warrington JP, Drummond HA, Granger JP, Ryan MJ. Placental ischemia-induced increases in brain water content and cerebrovascular permeability: Role of TNF-α. Am J Phys Regul Integr Comp Phys 309: R1425–R1431, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 364.Warrington JP, Fan F, Duncan J, Cunningham MW, Lamarca BB, Dechend R, Wallukat G, Roman RJ, Drummond HA, Granger JP, Ryan MJ. The angiotensin II type I receptor contributes to impaired cerebral blood flow autoregulation caused by placental ischemia in pregnant rats. Biol Sex Differ 10: 4–11, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 365.Warrington JP, Fan F, Murphy SR, Roman RJ, Drummond HA, Granger JP, Ryan MJ. Placental ischemia in pregnant rats impairs cerebral blood flow autoregulation and increases blood-brain barrier permeability. Phys Rep 2: 1–9, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 366.Wasserstrum N, Kirshon B, Rossavik IK, Willis RS, Moise KJ, Cotton DB. Implications of sino-aortic baroreceptor reflex dysfunction in severe preeclampsia. Obstet Gynecol 74: 34–39, 1989. [PubMed] [Google Scholar]
  • 367.Whitley GSJ, Cartwright JE. Trophoblast-mediated spiral artery remodelling: A role for apoptosis. J Anat 215: 21–26, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 368.Whitley GSJ, Cartwright JE. Cellular and molecular regulation of spiral artery remodelling: Lessons from the cardiovascular field. Placenta 31: 465–474, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 369.Woisetschläger C, Waldenhofer U, Bur A, Herkner H, Kiss H, Binder M, Laggner AN, Hirschl MM. Increased blood pressure response to the cold pressor test in pregnant women developing pre-eclampsia. J Hypertens 18: 399–403, 2000. [DOI] [PubMed] [Google Scholar]
  • 370.Wójtowicz A, Zembala-Szczerba M, Babczyk D, Kołodziejczyk-Pietruszka M, Lewaczyńska O, Huras H. Early-and late-onset preeclampsia: A comprehensive cohort study of laboratory and clinical findings according to the new ISHHP criteria. Int J Hypertens 2019: 4108271, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 371.Wu P, Haththotuwa R, Kwok CS, Babu A, Kotronias RA, Rushton C, Zaman A, Fryer AA, Kadam U, Chew-Graham CA, Mamas MA. Preeclampsia and future cardiovascular health: A systematic review and meta-analysis. Circ Cardiovasc Qual Outcomes 10: e003497, 2017. [DOI] [PubMed] [Google Scholar]
  • 372.Xu M, Guo D, Gu H, Zhang L, Lv S. Selenium and preeclampsia: A systematic review and meta-analysis. Biol Trace Elem Res 171: 283–292, 2016. [DOI] [PubMed] [Google Scholar]
  • 373.Yan JQ, Huang F, Hao F, Su XL, Meng Q, Xu MJ. Oxidative stress in the rostral ventrolateral medulla contributes to cardiovascular regulation in preeclampsia. Front Physiol 4: 772, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 374.Yang CCH, Chao TC, Kuo TBJ, Yin CS, Chen HI. Preeclamptic pregnancy is associated with increased sympathetic and decreased parasympathetic control of HR. Am J Physiol Heart Circ Physiol 278: 47–44, 2000. [DOI] [PubMed] [Google Scholar]
  • 375.Yilgwan CS, Pam VC, Ige OO, Golit WN, Anzaku S, Imade GE, Yilgwan G, Mutihir JT, Sagay AS, Odili A, Zoakah AI, Bode-Thomas F, Simon MA. Profile of congenital heart disease in infants born following exposure to preeclampsia. PLoS One 15: e0229987, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 376.Young OM, Twedt R, Catov JM. Pre-pregnancy maternal obesity and the risk of preterm preeclampsia in the American primigravida. Obesity 24: 1226–1229, 2016. [DOI] [PubMed] [Google Scholar]
  • 377.Yusuf H, Subih HS, Obeidat BS, Sharkas G. Associations of macro and micronutrients and antioxidants intakes with preeclampsia: A casecontrol study in Jordanian pregnant women. Nutr Metab Cardiovasc Dis 29: 458–466, 2019. [DOI] [PubMed] [Google Scholar]
  • 378.Zeeman GG, Cipolla MJ, Cunningham FG. Cerebrovascular (patho) physiology in preeclampsia/eclampsia. In: Lindheimer MD, Roberts JM, Cunningham FG, editors. Chesley’s Hypertensive Disorders of Pregnancy, (3rd ed). Cambridge, Massachusetts, USA: Academic Press, 2009, p. 227–247. [Google Scholar]
  • 379.Zeeman GG, Fleckenstein JL, Twickler DM, Gary Cunningham F. Cerebral infarction in eclampsia. Am J Obstet Gynecol 190: 714–720, 2004. [DOI] [PubMed] [Google Scholar]
  • 380.Zeisler H, Llurba E, Chantraine F, Vatish M, Staff AC, Sennström M, Olovsson M, Brennecke SP, Stepan H, Allegranza D, Dilba P, Schoedl M, Hund M, Verlohren S. Predictive value of the sFlt-1:PlGF ratio in women with suspected preeclampsia. N Engl J Med 374: 13–22, 2016. [DOI] [PubMed] [Google Scholar]
  • 381.Zeisler H, Llurba E, Chantraine FJ, Vatish M, Staff AC, Sennström M, Olovsson M, Brennecke SP, Stepan H, Allegranza D, Schoedl M, Grill S, Hund M, Verlohren S. Soluble fms-like tyrosine kinase-1 to placental growth factor ratio: Ruling out pre-eclampsia for up to 4 weeks and value of retesting. Ultrasound Obstet Gynecol 53: 367–375, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 382.Zhang H, Long Q, Ling L, Gao A, Li H, Lin Q. Elevated expression of KiSS-1 in placenta of preeclampsia and its effect on trophoblast. Reprod Biol 11: 99–115, 2011. [DOI] [PubMed] [Google Scholar]
  • 383.Zhao WX, Huang TT, Jiang M, Feng R, Lin JH. Expression of Notch family proteins in placentas from patients with early-onset severe preeclampsia. Reprod Sci 21: 716–723, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 384.Zhou Y, Damsky CH, Fisher SJ. Preeclampsia is associated with failure of human cytotrophoblasts to mimic a vascular adhesion phenotype: One cause of defective endovascular invasion in this syndrome? J Clin Invest 99: 2152–2164, 1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 385.Zhuang B, Luo X, Rao H, Li Q, Shan N, Liu X, Qi H. Oxidative stress-induced C/EBPβ inhibits β-catenin signaling molecule involving in the pathology of preeclampsia. Placenta 36: 839–846, 2015. [DOI] [PubMed] [Google Scholar]
  • 386.Zsengellér ZK, Rajakumar A, Hunter JT, Salahuddin S, Rana S, Stillman IE, Ananth Karumanchi S. Trophoblast mitochondrial function is impaired in preeclampsia and correlates negatively with the expression of soluble fms-like tyrosine kinase 1. Pregnancy Hypertens 6: 313–319, 2016. [DOI] [PubMed] [Google Scholar]
  • 387.Zussman R, Xu LY, Damani T, Groom KM, Chen Q, Seers B, Viall CA, Chamley LW, Hickey A. Antibodies can specifically target placental mitochondria and induce ROS production. J Autoimmun 111: 102437, 2020. [DOI] [PubMed] [Google Scholar]

Further Reading

  1. Alexander BT, Dasinger JH, Intapad S. Fetal programming and cardiovascular pathology. Compr Physiol 5: 997–1025, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Granger JP, Spradley FT, Bakrania BA. The endothelin system: A critical player in the pathophysiology of preeclampsia. Curr Hypertens Rep 20: 32, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Hammer ES, Cipolla MJ. Cerebrovascular dysfunction in preeclamptic pregnancies. Curr Hypertens Rep 17: 64, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Jim B, Karumanchi SA. Preeclampsia: Pathogenesis, prevention and long-term complications. Semin Nephrol 37: 386–397, 2017. [DOI] [PubMed] [Google Scholar]
  5. Lu HQ, Hu R. Lasting effects of intrauterine exposure to preeclampsia on oand the underlying mechanism. Am J Perinatol Reports 09: e275–e291, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Rana S, Lemoine E, Granger JP, Karumanchi SA. Preeclampsia: Pathophysiology, challenges, and perspectives. Circ Res 124: 1094–1112, 2019. [DOI] [PubMed] [Google Scholar]
  7. Wang A, Rana S, Karumanchi SA. Preeclampsia: The role of angiogenic factors in its pathogenesis. Physiology 24: 147–158, 2009. [DOI] [PubMed] [Google Scholar]

RESOURCES