Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2017 Apr 1.
Published in final edited form as: Curr Hypertens Rep. 2016 Apr;18(5):38. doi: 10.1007/s11906-016-0633-x

Placental ischemia and resultant phenotype in animal models of preeclampsia

Babbette LaMarca, Lorena M Amaral, Ashlyn C Harmon, Denise C Cornelius, Jessica L Faulkner, Mark W Cunningham Jr
PMCID: PMC5127437  NIHMSID: NIHMS790766  PMID: 27076345

Abstract

Preeclampsia is new onset (or worsening of preexisting) hypertension that occurs during pregnancy. It is accompanied by chronic inflammation, intrauterine growth restriction, elevated anti-angiogenic factors, and can occur with or without proteinuria. Although the exact etiology is unknown, it is thought that preeclampsia begins early in gestation with reduced uterine spiral artery remodeling leading to decreased vasculogenesis of the placenta as the pregnancy progresses. Soluble factors, stimulated by the ischemic placenta, shower the maternal vascular endothelium and are thought to cause endothelial dysfunction and to contribute to the development of hypertension during pregnancy. Due to the difficulty in studying such soluble factors in pregnant women, various animal models have been designed. Studies from these models have contributed to a better understanding of how factors released in response to placental ischemia may lead to increased blood pressure and reduced fetal weight during pregnancy. This review will highlight various animal models and the major findings indicating the importance of placental ischemia to lead to the pathophysiology observed in preeclamptic patients.

Keywords: preeclampsia, animal models, inflammation, pregnancy, hypertension

Introduction

Preeclampsia (PE) is a systemic disease that affects the function and health of multiple organs in both the mother and baby. While there is no cure for preeclampsia, if left untreated, a more severe, convulsive form called eclampsia may occur [13]. PE results, in part, from improper placentation and trophoblast invasion during pregnancy, which leads to placental ischemia [4, 5]. Placental ischemia, in turn, promotes a chronic inflammation in the mother that contributes to much of the pathophysiology characterized by increased mean arterial pressure (MAP), endothelial activation and dysfunction, antiangiogenic factors, activated inflammatory cells secreting autoantibodies and inflammatory cytokines, and increased oxidative stress [68]. In order to study the intricate mechanisms by which preeclampsia develops and progresses, animal models that recapitulate the disease are necessary. Knowing that PE may be initiated by placental ischemia with reduction of placental blood flow between 50–70 % [9], animal models with significantly reduced uteroplacental blood flow have been developed [10]. While such animal models of placental ischemia do have their limitations, studies in a variety of species suggest that reduced uterine perfusion pressure (RUPP) models have many features of PE observed in women. These models provide an opportunity for investigators to quantify the relative importance of pathophysiological factors in mediating cardiovascular and renal dysfunction in response to placental ischemia of pregnancy. The models also provide an important tool to test the efficacy of novel therapeutic approaches for the prevention and treatment of preeclampsia.

Preeclampsia is associated with placental ischemia

While the specific mechanisms leading to the development of preeclampsia have yet to be elucidated, it is known that changes in placentation due to poor vascular remodeling in the spiral artery potentially lead to the characteristic pathophysiology of this disease [1113]. Trophoblasts migrate along the spiral arteries and increase the diameter, leading to a lower resistance in the vessels that allows for proper placental perfusion and blood flow to the fetus and results in a healthy pregnancy for the mother and baby [1417]. Maternal blood volume, cardiac output, and heart rate are increased early in pregnancy, but blood pressure is normally decreased, starting in the second trimester, as a result of decreased resistance in the peripheral vasculature [18]. Placental oxygen tension is low in the beginning of pregnancy [19], but PO2 increases around weeks 10–12 of pregnancy as intervillous blood flow increases and supplies the placenta with oxygen and nutrient delivery [20, 21].

The process of trophoblast invasion and spiral artery remodeling during normal pregnancy rely heavily on the maternal immune system [22, 23]. During trophoblast invasion, the decidua, which is the lining of the uterus and forms the maternal portion of the placenta, contains a high number of immune cells that are necessary for the proper physiology of trophoblast migration [2426].

Macrophages, natural killer (NK) cells, dendritic cells, T cells, and T regulatory cells (Tregs) are all present in the decidua and are required for a normal pregnancy. Uterine NK cells (uNK), macrophages, and dendritic cells all cumulatively play an integral role in trophoblast invasion and decidua formation [27, 28], while regulatory T cells and regulatory cytokines ensure proper control and function of these proinflammatory cells and their actions [26, 29, 30]. Together, the immune cells present in the decidua during pregnancy work to ensure proper implantation of the placenta, and to promote trophoblast invasion that is neither shallow nor overly invasive [22, 23]. This is accomplished through their production of cytokines and angiogenic factors necessary for normal pregnancy. Any imbalance in these local immune responses could potentially result in an altered formation of the placenta, which could lead to complications during pregnancy such as preeclampsia or even less desirable, loss of the pregnancy [5, 31].

Pregnancies affected by PE are associated with an improper immune response and milieu of immune cells and cytokines [32, 33]. Placental ischemia has been shown to produce an imbalance in immune function that leads to a chronic inflammation and presents a state during pregnancy that is similar to an autoimmune disease [5, 34, 35]. This immune imbalance consists of increased pro-inflammatory immune cells and cytokines, and decreased regulatory immune cells and cytokines, which is believed to contribute to the overall pathophysiology of PE, [33, 3638]. This alteration in immune balance contributes to production of reactive oxygen species [39, 40], increased endothelin-1 expression [4144], and B-cell production of auto-antibodies to the angiotensin II type 1 (AT1-AA), all of which culminate in the development of hypertension during pregnancy [35, 4549].

Hypertension in response to placental ischemia; the RUPP model

Early procedures that induce a reduction of the uteroplacental blood flow during pregnancy lead to hypertension associated with placental ischemia in pregnant animals. In 1939, researchers clamped the abdominal aorta of pregnant dogs and found that it led to an approximate 50 % decrease in placental blood flow and increased the blood pressure by 25 mmHg [50]. When the clamp was removed, the blood pressure returned to normal. The same procedure was used in non-pregnant dogs, but there was no increase in blood pressure, demonstrating a pregnancy specific pathophysiology. Others have since performed a similar procedure to induce placental ischemia by decreasing uteroplacental perfusion by 40 %, leading to markedly increased systolic blood pressure in conscious pregnant dogs [51]. Endothelial irregularities and placental infarcts were also noted in these studies. Other methods have been successfully implemented in dogs where ligation or banding of the utero-ovarian arteries before pregnancy resulted in a gradual increase in blood pressure after the dogs had become pregnant, with a subsequent marked increase in blood pressure throughout the second and third trimesters [52].

This method has been employed by many other researchers in many different animal species. Researchers have induced a reduction in placental blood flow that led to hypertension in pregnant rats, rabbits, rhesus monkeys, and baboons. Aortic clamping in rabbits was also linked to preeclampsia complications, such as fetal compromise, kidney injury, platelet activation, and intrauterine growth restriction [53, 54]. In the rhesus monkey, a 30 % increase in blood pressure occurred with aortic clamping, which was accompanied by an approximate 60 % reduction in uteroplacental blood flow and subsequent development of proteinuria, increased fetal demise, and intrauterine growth restriction [55, 56]. Banding of the uterine-ovarian arteries in baboons during pregnancy led to increased blood pressure and caused much of the pathophysiology seen in women with preeclampsia [57].

The reduced uterine perfusion pressure (RUPP) rat model of preeclampsia uses a combination of aortic constriction and occlusion of the uterine-ovarian arteries to decrease blood flow to the uterus and create a state of placental ischemia in pregnant rats [58]. These rats develop much of the same pathophysiology as women with preeclampsia. Mechanical induction of placental ischemia is performed in pregnant Sprague-Dawley rats. On day 14 of gestation, restrictive silver clips are placed on the abdominal aorta above the iliac bifurcation and, in order to prevent compensatory blood flow through the ovaries, silver clips are also placed on branches of the ovarian arteries [58, 59]. Blood pressures and tissues are collected on day 19 of gestation. This procedure reduces uteroplacental blood flow by approximately 40 % and increases blood pressure by approximately 20–30 mmHg compared to normal pregnant Sprague-Dawley rats [58]. RUPP rats demonstrate much of the same pathophysiology observed in preeclampsia, including endothelial dysfunction [58, 60, 61], decreased pup weights and litter sizes, intrauterine growth restriction [42, 62], and chronic inflammation associated with the agonistic autoantibodies to the AT1 receptor (AT1-AA) [63]. Full characterization of RUPP animals was completed through many studies that demonstrated that the model sufficiently recapitulates preeclampsia [10]. These animals demonstrate characteristics of endothelial dysfunction with a reduction in nitric oxide (NO) [58] and increased contractility of vascular smooth muscle cells [60]. As seen in women with preeclampsia, RUPP rats also have increased production of AT1-AA [63], which leads to activation of the AngII receptor and contributes to the increases in blood pressure. Similarly to women with preeclampsia, this increase in AngII receptor activity also leads to an increased production in endothelin-1(ET-1) in renal cortices, placentas, and in the vascular circulation [61]. Reactive oxygen species (ROS) in both the placenta and in circulation are increased in RUPP rats versus normal pregnant animals [64]. Importantly, anti-angiogenic factors like soluble fms-like tyrosine kinase-(sFlt-1) and soluble endoglin (sEng) are increased in RUPP animals, which leads to further decreases in the vascularization of the placenta and blood delivery to the fetus, and contributes to the development of hypertension [65, 66].

Placental ischemia in preeclamptic patients is associated with increased inflammatory T cells and inflammatory cytokines. In addition, PE is accompanied by decreased regulatory cells and regulatory cytokines. In concert, such alteration will result in a chronic state of inflammation. Similar immune alterations associated with preeclampsia were validated in RUPP rats to demonstrate a role for placental ischemia in causing these same imbalances in the immune repertoire [67]. An increase in inflammatory CD4+ T cells along with a decrease in T regulatory (Tregs) cells is seen in RUPP rats. Increased inflammatory cytokines such as tumor necrosis factor alpha (TNF-α) and interleukin 6 (IL-6), and interleukin 17 (IL-17) are seen in the circulation of RUPP rats, and regulatory cytokines such as interleukin 10 (IL-10) and interleukin 4 (IL-4) are decreased [43, 68]. Table 1 shows a summary of the similarities between the RUPP rat model and women with preeclampsia. The presence of all of these characteristics in this model indicate that the RUPP model can be a valuable tool for the study of mechanisms stimulated by placental ischemia that lead to characteristics of preeclampsia.

Table 1.

Pathophysiology seen in women with preeclampsia and in response to placental ischemia RUPP Rats

Normal Pregnant Preeclampsia RUPP Rats
Hypertension − − − +++ +++
Endothelial dysfunction − − − +++ +++
Intrauterine growth restriction − − − +++ +++
Inflammation + +++ +++
Agonistic autoantibodies to AT1 − − − +++ +++
Anti-angiogenic factors − − − +++ +++

T Lymphocytes during Preeclampsia

A state of controlled inflammation that exists during a normal, healthy pregnancy is necessary for proper implantation and placentation to occur. A balance that exists between pro-inflammatory and anti-inflammatory cells and cytokines during pregnancy allows for the control of necessary inflammatory responses while preventing excessive inflammation [6972]. When this balance is shifted toward pro-inflammatory responses, and inflammation thus occurs it results in characteristics similar to those observed in preeclampsia [7375].

A role of inflammatory T cells in the pathogenesis of preeclampsia has been established through adoptive transfer studies using Th1-like splenocytes. Zenclussen et al. (2004) demonstrated the importance of inflammatory T cells when they isolated splenocytes from normal pregnant mice, cultured them to promote differentiation into Th-1-like cells that secreted Th-1 specific cytokines [76], and transferred them into normal pregnant mice. Adoptive transfer of these Th-1-like cells provoked symptoms of preeclampsia such as increased blood pressure, alterations in kidney function, and increased inflammatory markers in the decidua. Adoptive transfer of the Th-1-like splenocytes in non-pregnant mice did not result in any changes in immune composition or blood pressure, thus demonstrating that such a response is specific to pregnancy [76].

The Role of CD4+ T cells in Preeclampsia

Later studies further supported a role for Th-1 cells in the development of preeclamptic symptoms. CD4+ T cells from the RUPP rat model of preeclampsia were adoptively transferred into normal pregnant Sprague-Dawley rats. Adoptive transfer of RUPP CD4+ T cells into normal pregnant rats leads to much of the characteristic pathophysiology seen during preeclampsia, such as a significant increase in blood pressure and declines in glomerular filtration, circulating inflammatory cytokines TNF-α, IL-6 and IL-17, anti-angiogenic factor sFlt-1, and AT1-AA compared to normal pregnant recipients of normal pregnant CD4+ T cells, [38], increased ET-1, and oxidative stress. Normal pregnant CD4+ T cells had no effect on blood pressure or circulating factors when injected into normal pregnant rats [38]. Furthermore, the adoptive transfer of RUPP CD4+ T cells into non-pregnant rats did not have any effect on blood pressure or circulating factors. Losartan or Rituximab blocked any AT1-AA-mediated increase of blood pressure in response to inflammatory T cells [77, 78]. These adoptive transfer studies, along with the previously described Th-1-like splenocyte study, indicate a strong role for preeclamptic inflammatory T cells in the development of hypertension during pregnancy, and in promoting the pathophysiology that accompanies the rise in blood pressure.

IL-17 and TH17 cells mediate hypertension, oxidative stress, and AT1-AA during pregnancy

Although clinical studies show an association between preeclampsia and increased TH17 cells and IL-17, their role in mediating the pathophysiology of preeclampsia had not previously been established. Dhillon et al. examined the effect of IL-17 infusion on blood pressure during pregnancy. This study showed that IL-17 caused hypertension and increased TH17 cells, oxidative stress, and AT1-AA [40]. A blockade of the AT1 receptor with losartan attenuated the blood pressure response and placental ROS. Additionally, the depletion of B-cells with Rituximab blunted hypertension and numbers of TH17 cells. Administration of the superoxide dismutase mimetic TEMPOL attenuated the hypertension, decreased placental production of ROS, and significantly decreased circulating AT1-AAs [40]. The effect of tempol on IL-17-induced pathophysiology suggests that the ROS may be an important signaling molecule for B-cells to produce AT1-AAs. This data suggests that IL-17 causes hypertension via placental oxidative stress and activation of the AT1 receptor via the AT1-AA.

Subsequent studies were performed to examine the effect of an IL-17 blockade on the RUPP rat model of PE. A soluble form of the IL-17 receptor C (IL-17RC) was infused from gestational day 14–19 in RUPP rats to block the IL-17 signaling pathway. The IL17 blockade reduced hypertension, intrauterine growth restriction (IUGR), oxidative stress and AT1-AA, and lowered circulating TH17 cells. A significant improvement in pup weight was accompanied by increased placental weight and decreased uterine artery resistance index, indicating the importance of this pathway on the uteroplacental environment [79]. Thus, inhibition of this immune pathway could lower rates of fetal demise as a direct result of decreased placental stress serving as a fetal protective mechanism during PE, and could be a novel therapeutic approach to improving the treatment of PE.

The effect of decreased Tregs in preeclampsia

Along with increased levels of inflammatory T cells, regulatory T cells (Tregs) are decreased during preeclampsia in response to placental ischemia. These cells are identified by the expression of cell surface markers CD4+ CD25+ and their specific internal transcription factor forkhead box protein 3 (Foxp3+) [80]. Tregs are responsible for suppression of responses in the adaptive and innate immune system, and they control immune responses through various mechanisms. Loss of Treg function has been shown to lead to autoimmune diseases and other immunopathology, including maternal loss of tolerance for the fetus during pregnancy [80]. Tregs are increased very early in normal pregnancy and reach their highest levels during the second trimester before decreasing back to normal levels [81]. The peak times for Tregs coincide with important processes like vascular remodeling and trophoblast invasion, which are integral to a healthy pregnancy. Evidence for the contribution of reduced Tregs to the pathophysiology of preeclampsia has been provided by many studies. Tregs have shown to be decreased both in the circulation and decidua of women with preeclampsia, and this decrease is directly proportional to the severity of the disease [82]. Decreased circulating Tregs have also been seen in women who have had multiple miscarriages [72]. Studies have shown that decreases in decidual Tregs cause increased apoptosis in trophoblasts, preventing sufficient invasion of the trophoblast [74, 83]. In addition, an abortion-prone mouse model was found to have decreased numbers of CD4+ CD25+ Tregs in the decidua, but adoptive transfer of Tregs from normal pregnant mice decreased spontaneous abortions [84]. Furthermore, the role of Tregs during pregnancy to provide fetal-maternal tolerance has been demonstrated through adoptive transfer studies where Tregs were depleted prior to injection into T-cell-deficient mice. In this study, allogeneic fetuses, but not syngeneic fetuses, were rejected in the absence of Tregs, and additional studies have also demonstrated that Treg depletion during pregnancy decreases the number of pups surviving to term [85].

As with PE patients, RUPP rats exhibit a decrease in Tregs in response to placental ischemia. We recently demonstrated that adoptive transfer of NP Tregs into RUPP rats prior to placental insult results in lower blood pressure, blunts inflammation and Endothelin-1 expression, normalizes tissue oxidative stress, and attenuates AT1-AA production in response to placental ischemia. We utilized flow cytometry to demonstrate that the adoptive transfer was sufficient to increase circulating Tregs in response to placental ischemia. This suggests that the number of Tregs is important in controlling inflammation and the vasoactive factors that contribute to hypertension during pregnancy, such as oxidative stress, AT1-AA and ET-1. Although no improvement in fetal morbidity was observed with adoptive transfer of NP Tregs into RUPP rats, improving pathophysiology in the mother may indirectly benefit the fetus by increasing time to delivery, thereby giving the fetus more time to develop and mature.

Syncytial knots and PE

Although PE is associated with chronic inflammation, a stimulus for the inflammatory response is unknown. Recent studies suggests that the inflammatory response is triggered by particles, ranging from large deported multinuclear fragments to sub-cellular components, shed from the syncytial surface of the human placenta [86]. Using an in-vitro model, Chamley and co-workers showed that these syncytial knots are shed by an apoptosis-like programmed cell death process, phagocytosed by macrophages, which then establish a tolerogenic response [87]. However, when phagocytosed, necrotic syncytial knots appear to be immunostimulatory. While there is a strong correlation between trophoblast debris and immune activation in preeclampsia, it remains unclear whether these particles could indeed elicit a pathogenic response in vivo. To address this issue, Chamley et al. recently reported that chronic administration of necrotic trophoblast debris increases blood pressure in pregnant rats relative to pregnant controls [88]. This report was the first demonstration that necrotic trophoblast debris can alter blood pressure in vivo and, while the mechanism by which the necrotic trophoblast debris becomes hypertensive remains to be determined, this observation is consistent with the growing body of evidence suggesting that necrotic trophoblast debris can contribute to the hypertension of preeclampsia.

AT1-AA during pregnancy

Angiotensin II type 1 receptor autoantibodies (AT1-AA) are elevated in women with preeclampsia as early as the 2nd trimester of pregnancy, which is when preeclampsia is usually diagnosed. This suggests that AT1-AAs may play a major role in the pathophysiology of preeclampsia. [45, 63]. AT1-AAs remain elevated in postpartum preeclampsia women after 1 year of delivery, indicating they could be a major risk factor for the development of cardiovascular and kidney disease in mothers later in life.[89]. Importantly, in 1973, Gant et al. found that ANGII infusion into pregnant women in their 2nd trimester resulted in a heightened vasoconstrictor and pressor response in women that went on to develop PE compared to those with NP. Due to a lack of research in this area, the mediator of this heightened response remained unknown. With the discovery of AT1-AA several years later, we hypothesized that AT1-AA may be the causal factor for heightened blood pressure and vasoconstrictor response.

To demonstrate a role for AT1-AAs in the pathophysiology of PE, several studies have investigated the treatment of pregnant rodents with the antibody. Isolated human and rodent AT1-AAs administered to rats during pregnancy were shown to increase blood pressure while causing renal pathology, and led to the elevation in several circulating factors associated with preeclampsia, such as sFlt-1, sEng, oxidative stress, endothelin-1, TNF alpha, and IL-6 and endothelial microparticles [10, 9093]. Interestingly, when administered simultaneously to pregnant rats, ANGII and AT1-AA exhibit a greater increase in blood pressure than when either one is administered alone [48, 90]. Chronic administration of ANGII and AT-AAs together increases oxidative stress, endothelin-1 secretion, and renal artery resistive index above that seen with ANGII or AT1-AA alone [48, 90]. These data taken together suggest a synergist effect when both AT1-AAs and ANG II are present together during pregnancy, possibly confirming our hypothesis.

The mechanism of increased ANGII sensitivity in preeclampsia is unknown. However, we hypothesize that AT1-AAs increase ANGII by increasing the AT1R affinity for ANG II. One mechanism by which the AT1-AA can increase ANGII sensitivity is by increasing the dimerization of the ATIR to the vasodepressor bradykinin receptor [94] [95, 96]. Others suggest that it may change the conformation of the AT1R in the membrane of a cell, thus allowing for ANGII to bind more readily to the receptor. Therefore, an increase in ANGII sensitivity in preeclampsia could be due to AT1R receptor dimerization or changes in conformation; however, more studies are needed to verify this hypothesis.

Animal models used to study the role of angiogenic factors

Of particular interest for preeclampsia research is the finding that sFlt-1, positively regulated by hypoxia, is produced by both placental trophoblasts and human placental villous explants in response to low oxygen tension, which is thought to occur in response to placental ischemia [97, 98]. sFlt-1 is the soluble receptor for VEGF and has been shown to interfere with VEGF signaling. While sFlt-1 production is regulated by the hypoxia-inducible factor-1, proof of concept studies using animal model RUPP have demonstrated that placental ischemia can cause increased sFlt-1 during pregnancy. Models of RUPP in both non-human primates and rats have shown significant increases in circulating sFlt-1, and concurrent decreases in bioavailable circulating VEGF [65, 99].

Several experimental models have demonstrated a causative role for sFlt-1 in the pathology of preeclampsia. Viral overexpression of sFlt-1 in pregnant rats led to a preeclampsia-like state, with hypertension, glomerular endotheliosis, and proteinuria [100]. In agreement with the viral expression experiments, direct infusion of sFlt-1 into pregnant mice or rats induces preeclamptic-like symptoms, including hypertension and reduced fetal weight [101104]. Endothelin and oxidative stress are two major effector molecules in sFlt-1 hypertension during pregnancy. Administration of an ETA receptor antagonist or Tempol completely normalized blood pressure [105].

It does not appear that the pathological manifestations seen in sFlt-1-induced PIH are a direct result of circulating sFlt-1 but rather the loss of VEGF. VEGF administration in rats with placental ischemia restored normal blood pressure, renal function, and vascular activity [106]. These and other data from animal models have led to the hypothesis that the ratio between sFlt-1 and VEGF or PlGF is critical to maintaining healthy endothelial function and normal vascular activity.

Genetic models resulting in placental ischemia

BPH/5 mouse

The BPH/5 mouse model is a substrain derivation of the BPH/2 “borderline hypertensive” mouse, and exhibits mildly elevated blood pressure throughout the adult lifespan of the animal. Davisson et al. demonstrated that the BPH/5 model exhibits elevations in blood pressure (up to ~25mmHg) during pregnancy that resolve immediately following delivery. The increase in blood pressure was accompanied by proteinuria, glomerulosclerosis, intrauterine growth restriction, maternal endothelial dysfunction, and increased fetal mortality [107]. Additionally, the BPH/5 mouse has altered cytotrophoblast invasion and placental abnormalities that coincide with increased uterine artery vascular resistance and placental ischemia. As with other models of placental ischemia, both angiogenic imbalance and oxidative stress are associated with the etiology of BPH/5 gestational hypertension. Importantly, the administration of Tempol or the viral delivery of VEGF121 attenuated the hypertension and proteinuria [108, 109]. BPH/5 remains one of the most intriguing models for the identification of new pathogenic factors in a spontaneous animal model.

Genetic modification of the renin/angiotensin system

Though its role in long-term maintenance of blood pressure is well established, the exact pathophysiological role of the RAS in the development of preeclampsia is less clear. During normal pregnancy, plasma renin concentration/activity and ANGII levels are elevated, while vascular sensitivity to ANGII appears to be decreased. In contrast, during preeclampsia a significant increase in ANGII sensitivity is noted, possibly due in part to production of agonistic AT1-AA [110]. To investigate the role of the RAS in PE, several groups have utilized transgenic mouse and rodent strains in which females overexpressing human angiotensinogen (hAGN) are crossed with males overexpressing human renin hREN [111114]. This model has been shown to exhibit hypertension and proteinuria during pregnancy [111, 112]. The Lavoie laboratory uses a related model in which mice that constitutively overexpress both REN and AGN subsequently develop chronic hypertension and fetal growth restriction; this is considered a model of superimposed preeclampsia [114]. Both of these models have proven to be versatile tools for investigating the role of the RAS in the etiology of PE, and are likely to be of great utility in future studies.

Conclusion

Hypertension in response to placental ischemia begins early in gestation with inadequate remodeling of the uterine spiral arteries, leading to decreased vascularity and blood supply to the growing uteroplacental unit. Much of this remodeling is controlled by close interactions between immune cells and the maternal vasculature and the invading cytotrophoblasts. Some unknown event occurs that changes this interaction into a chronic state of immune activation and decreased vascularity of the uteroplacental unit. Secreted factors cause an increase in blood pressure and a decrease in fetal growth. CD4+Tcells are among the activated immune cells that have been shown to play an instrumental role in stimulating agonistic autoantibodies, inflammatory cytokines and anti-angiogenic factors, all known players in the pathogenicity of preeclampsia. Various animal models have demonstrated a role for such factors that are not only elevated in preeclampsia, but play an important role in the clinical presentation of the disease. Through knowledge gained from such animal models, we may be able to develop new chemotherapeutic targets to improve treatment of hypertension secondary to placental ischemia in preeclamptic women.

Footnotes

Compliance with Ethics Guidelines

Human and Animal Rights and Informed Consent

This article does not contain any studies with human or animal subjects performed by any of the authors.

Conflict of Interest

Drs. LaMarca, Amaral, Harmon, Cornelius, Jessica, and Cunningham declare no conflicts of interest.

References

Papers of particular interest, published recently, have been highlighted as:

• Of importance

•• Of major importance

  • 1.Keyes LE, et al. Intrauterine growth restriction, preeclampsia, and intrauterine mortality at high altitude in Bolivia. Pediatr Res. 2003;54(1):20–5. doi: 10.1203/01.PDR.0000069846.64389.DC. [DOI] [PubMed] [Google Scholar]
  • 2.Powe CE, Levine RJ, Karumanchi SA. Preeclampsia, a disease of the maternal endothelium: the role of antiangiogenic factors and implications for later cardiovascular disease. Circulation. 2011;123(24):2856–69. doi: 10.1161/CIRCULATIONAHA.109.853127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Thadhani RI, Johnson RJ, Karumanchi SA. Hypertension during pregnancy: a disorder begging for pathophysiological support. Hypertension. 2005;46(6):1250–1. doi: 10.1161/01.HYP.0000188701.24418.64. [DOI] [PubMed] [Google Scholar]
  • 4.Redman CW. Preeclampsia: a multi-stress disorder. Rev Med Interne. 2011;32(Suppl 1):S41–4. doi: 10.1016/j.revmed.2011.03.331. [DOI] [PubMed] [Google Scholar]
  • 5*.Redman CW, I, Sargent L. Immunology of pre-eclampsia. Am J Reprod Immunol. 2010;63(6):534–43. doi: 10.1111/j.1600-0897.2010.00831.x. [DOI] [PubMed] [Google Scholar]
  • 6.Brown CE, et al. Low-dose aspirin. II. Relationship of angiotensin II pressor responses, circulating eicosanoids, and pregnancy outcome. Am J Obstet Gynecol. 1990;163(6 Pt 1):1853–61. doi: 10.1016/0002-9378(90)90764-x. [DOI] [PubMed] [Google Scholar]
  • 7.Hladunewich M, Karumanchi SA, Lafayette R. Pathophysiology of the clinical manifestations of preeclampsia. Clin J Am Soc Nephrol. 2007;2(3):543–9. doi: 10.2215/CJN.03761106. [DOI] [PubMed] [Google Scholar]
  • 8.Khalil RA, Granger JP. Vascular mechanisms of increased arterial pressure in preeclampsia: lessons from animal models. Am J Physiol Regul Integr Comp Physiol. 2002;283(1):R29–45. doi: 10.1152/ajpregu.00762.2001. [DOI] [PubMed] [Google Scholar]
  • 9.Lunell NO, et al. Uteroplacental blood flow in pregnancy induced hypertension. Scand J Clin Lab Invest Suppl. 1984;169:28–35. doi: 10.3109/00365518409085374. [DOI] [PubMed] [Google Scholar]
  • 10.Li J, LaMarca B, Reckelhoff JF. A model of preeclampsia in rats: the reduced uterine perfusion pressure (RUPP) model. Am J Physiol Heart Circ Physiol. 2012;303(1):H1–8. doi: 10.1152/ajpheart.00117.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Gilbert JS, et al. Pathophysiology of hypertension during preeclampsia: linking placental ischemia with endothelial dysfunction. Am J Physiol Heart Circ Physiol. 2008;294(2):H541–50. doi: 10.1152/ajpheart.01113.2007. [DOI] [PubMed] [Google Scholar]
  • 12.Odegard RA, et al. Preeclampsia and fetal growth. Obstet Gynecol. 2000;96(6):950–5. [PubMed] [Google Scholar]
  • 13.Roberts JM, Escudero C. The placenta in preeclampsia. Pregnancy Hypertens. 2012;2(2):72–83. doi: 10.1016/j.preghy.2012.01.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Ahmed A, et al. Regulation of placental vascular endothelial growth factor (VEGF) and placenta growth factor (PIGF) and soluble Flt-1 by oxygen--a review. Placenta. 2000;21(Suppl A):S16–24. doi: 10.1053/plac.1999.0524. [DOI] [PubMed] [Google Scholar]
  • 15.Rodesch F, et al. Oxygen measurements in endometrial and trophoblastic tissues during early pregnancy. Obstet Gynecol. 1992;80(2):283–5. [PubMed] [Google Scholar]
  • 16.Zhou Y, et al. Preeclampsia is associated with abnormal expression of adhesion molecules by invasive cytotrophoblasts. J Clin Invest. 1993;91(3):950–60. doi: 10.1172/JCI116316. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Zhou Y, et al. Human cytotrophoblasts adopt a vascular phenotype as they differentiate. A strategy for successful endovascular invasion? J Clin Invest. 1997;99(9):2139–51. doi: 10.1172/JCI119387. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Mabie WC, et al. A longitudinal study of cardiac output in normal human pregnancy. Am J Obstet Gynecol. 1994;170(3):849–56. doi: 10.1016/s0002-9378(94)70297-7. [DOI] [PubMed] [Google Scholar]
  • 19.Jauniaux E, Burton GJ. Morphological and biological effects of maternal exposure to tobacco smoke on the feto-placental unit. Early Hum Dev. 2007;83(11):699–706. doi: 10.1016/j.earlhumdev.2007.07.016. [DOI] [PubMed] [Google Scholar]
  • 20.Jauniaux E, et al. Amniotic gas values and acid-base status during acute maternal hyperoxemia and hypoxemia in the early fetal sheep. Am J Obstet Gynecol. 2000;182(3):661–5. doi: 10.1067/mob.2000.103937. [DOI] [PubMed] [Google Scholar]
  • 21.Jauniaux E, et al. In-vivo measurement of intrauterine gases and acid-base values early in human pregnancy. Hum Reprod. 1999;14(11):2901–4. doi: 10.1093/humrep/14.11.2901. [DOI] [PubMed] [Google Scholar]
  • 22.Lala PK, Chakraborty C. Factors regulating trophoblast migration and invasiveness: possible derangements contributing to pre-eclampsia and fetal injury. Placenta. 2003;24(6):575–87. doi: 10.1016/s0143-4004(03)00063-8. [DOI] [PubMed] [Google Scholar]
  • 23.Wells M. The pathology of gestational trophoblastic disease: recent advances. Pathology. 2007;39(1):88–96. doi: 10.1080/00313020601137367. [DOI] [PubMed] [Google Scholar]
  • 24.King A. Uterine leukocytes and decidualization. Hum Reprod Update. 2000;6(1):28–36. doi: 10.1093/humupd/6.1.28. [DOI] [PubMed] [Google Scholar]
  • 25.Trundley A, Moffett A. Human uterine leukocytes and pregnancy. Tissue Antigens. 2004;63(1):1–12. doi: 10.1111/j.1399-0039.2004.00170.x. [DOI] [PubMed] [Google Scholar]
  • 26*.Williams PJ, et al. Decidual leucocyte populations in early to late gestation normal human pregnancy. J Reprod Immunol. 2009;82(1):24–31. doi: 10.1016/j.jri.2009.08.001. [DOI] [PubMed] [Google Scholar]
  • 27.Craven CM, Morgan T, Ward K. Decidual spiral artery remodelling begins before cellular interaction with cytotrophoblasts. Placenta. 1998;19(4):241–52. doi: 10.1016/s0143-4004(98)90055-8. [DOI] [PubMed] [Google Scholar]
  • 28.Hanna J, et al. Decidual NK cells regulate key developmental processes at the human fetal-maternal interface. Nat Med. 2006;12(9):1065–74. doi: 10.1038/nm1452. [DOI] [PubMed] [Google Scholar]
  • 29.Somerset DA, et al. Normal human pregnancy is associated with an elevation in the immune suppressive CD25+ CD4+ regulatory T-cell subset. Immunology. 2004;112(1):38–43. doi: 10.1111/j.1365-2567.2004.01869.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30**.Tilburgs T, et al. Evidence for a selective migration of fetus-specific CD4+CD25bright regulatory T cells from the peripheral blood to the decidua in human pregnancy. J Immunol. 2008;180(8):5737–45. doi: 10.4049/jimmunol.180.8.5737. [DOI] [PubMed] [Google Scholar]
  • 31.Veenstra van Nieuwenhoven AL, Heineman MJ, Faas MM. The immunology of successful pregnancy. Hum Reprod Update. 2003;9(4):347–57. doi: 10.1093/humupd/dmg026. [DOI] [PubMed] [Google Scholar]
  • 32.Freeman DJ, et al. Short- and long-term changes in plasma inflammatory markers associated with preeclampsia. Hypertension. 2004;44(5):708–14. doi: 10.1161/01.HYP.0000143849.67254.ca. [DOI] [PubMed] [Google Scholar]
  • 33.LaMarca B, Cornelius D, Wallace K. Elucidating immune mechanisms causing hypertension during pregnancy. Physiology (Bethesda) 2013;28(4):225–33. doi: 10.1152/physiol.00006.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Cornelius DC, et al. Administration of interleukin-17 soluble receptor C suppresses TH17 cells, oxidative stress, and hypertension in response to placental ischemia during pregnancy. Hypertension. 2013;62(6):1068–73. doi: 10.1161/HYPERTENSIONAHA.113.01514. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Irani RA, et al. Autoantibody-mediated angiotensin receptor activation contributes to preeclampsia through tumor necrosis factor-alpha signaling. Hypertension. 2010;55(5):1246–53. doi: 10.1161/HYPERTENSIONAHA.110.150540. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Prins JR, et al. Preeclampsia is associated with lower percentages of regulatory T cells in maternal blood. Hypertens Pregnancy. 2009;28(3):300–11. doi: 10.1080/10641950802601237. [DOI] [PubMed] [Google Scholar]
  • 37**.Santner-Nanan B, et al. Systemic increase in the ratio between Foxp3+ and IL-17-producing CD4+ T cells in healthy pregnancy but not in preeclampsia. J Immunol. 2009;183(11):7023–30. doi: 10.4049/jimmunol.0901154. [DOI] [PubMed] [Google Scholar]
  • 38.Wallace K, et al. CD4+ T-helper cells stimulated in response to placental ischemia mediate hypertension during pregnancy. Hypertension. 2011;57(5):949–55. doi: 10.1161/HYPERTENSIONAHA.110.168344. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Conrad KP, Benyo DF. Placental cytokines and the pathogenesis of preeclampsia. Am J Reprod Immunol. 1997;37(3):240–9. doi: 10.1111/j.1600-0897.1997.tb00222.x. [DOI] [PubMed] [Google Scholar]
  • 40.Dhillion P, et al. IL-17-mediated oxidative stress is an important stimulator of AT1-AA and hypertension during pregnancy. Am J Physiol Regul Integr Comp Physiol. 2012;303(4):R353–8. doi: 10.1152/ajpregu.00051.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Granger JP, et al. Pathophysiology of hypertension during preeclampsia linking placental ischemia with endothelial dysfunction. Hypertension. 2001;38(3 Pt 2):718–22. doi: 10.1161/01.hyp.38.3.718. [DOI] [PubMed] [Google Scholar]
  • 42.Granger JP, et al. Pathophysiology of preeclampsia: linking placental ischemia/hypoxia with microvascular dysfunction. Microcirculation. 2002;9(3):147–60. doi: 10.1038/sj.mn.7800137. [DOI] [PubMed] [Google Scholar]
  • 43.LaMarca BB, et al. Role of endothelin in mediating tumor necrosis factor-induced hypertension in pregnant rats. Hypertension. 2005;46(1):82–6. doi: 10.1161/01.HYP.0000169152.59854.36. [DOI] [PubMed] [Google Scholar]
  • 44.LaMarca BD, et al. Pathophysiology of hypertension in response to placental ischemia during pregnancy: a central role for endothelin? Gend Med. 2008;5(Suppl A):S133–8. [Google Scholar]
  • 45.Herse F, LaMarca B. Angiotensin II type 1 receptor autoantibody (AT1-AA)-mediated pregnancy hypertension. Am J Reprod Immunol. 2013;69(4):413–8. doi: 10.1111/aji.12072. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.LaMarca B, Wallace K, Granger J. Role of angiotensin II type I receptor agonistic autoantibodies (AT1-AA) in preeclampsia. Curr Opin Pharmacol. 2011;11(2):175–9. doi: 10.1016/j.coph.2011.01.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Parrish MR, et al. Angiotensin II type 1 autoantibody induced hypertension during pregnancy is associated with renal endothelial dysfunction. Gend Med. 2011;8(3):184–8. doi: 10.1016/j.genm.2011.04.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Wenzel K, et al. Angiotensin II type 1 receptor antibodies and increased angiotensin II sensitivity in pregnant rats. Hypertension. 2011;58(1):77–84. doi: 10.1161/HYPERTENSIONAHA.111.171348. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Zhou CC, et al. Autoantibody from women with preeclampsia induces soluble Fms-like tyrosine kinase-1 production via angiotensin type 1 receptor and calcineurin/nuclear factor of activated T-cells signaling. Hypertension. 2008;51(4):1010–9. doi: 10.1161/HYPERTENSIONAHA.107.097790. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Podjarny E, Losonczy G, Baylis C. Animal models of preeclampsia. Semin Nephrol. 2004;24(6):596–606. doi: 10.1016/s0270-9295(04)00131-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Abitbol MM. A simplified technique to produce toxemia in the pregnant dog. Am J Obstet Gynecol. 1981;139(5):526–34. doi: 10.1016/0002-9378(81)90512-3. [DOI] [PubMed] [Google Scholar]
  • 52.Woods LL, Brooks VL. Role of the renin-angiotensin system in hypertension during reduced uteroplacental perfusion pressure. Am J Physiol. 1989;257(1 Pt 2):R204–9. doi: 10.1152/ajpregu.1989.257.1.R204. [DOI] [PubMed] [Google Scholar]
  • 53.Abitbol MM, Driscoll SG, Ober WB. Placental lesions in experimental toxemia in the rabbit. Am J Obstet Gynecol. 1976;125(7):942–8. doi: 10.1016/0002-9378(76)90493-2. [DOI] [PubMed] [Google Scholar]
  • 54.Abitbol MM, et al. Production of experimental toxemia in the pregnant rabbit. Am J Obstet Gynecol. 1976;124(5):460–70. doi: 10.1016/0002-9378(76)90169-1. [DOI] [PubMed] [Google Scholar]
  • 55.Combs CA, et al. Experimental preeclampsia produced by chronic constriction of the lower aorta: validation with longitudinal blood pressure measurements in conscious rhesus monkeys. Am J Obstet Gynecol. 1993;169(1):215–23. doi: 10.1016/0002-9378(93)90171-e. [DOI] [PubMed] [Google Scholar]
  • 56.Zhou Y, et al. Increased depth of trophoblast invasion after chronic constriction of the lower aorta in rhesus monkeys. Am J Obstet Gynecol. 1993;169(1):224–9. doi: 10.1016/0002-9378(93)90172-f. [DOI] [PubMed] [Google Scholar]
  • 57.Cavanagh D, et al. Pregnancy-induced hypertension: development of a model in the pregnant primate (Papio anubis) Am J Obstet Gynecol. 1985;151(7):987–99. doi: 10.1016/0002-9378(85)90679-9. [DOI] [PubMed] [Google Scholar]
  • 58.Crews JK, et al. Decreased endothelium-dependent vascular relaxation during reduction of uterine perfusion pressure in pregnant rat. Hypertension. 2000;35(1 Pt 2):367–72. doi: 10.1161/01.hyp.35.1.367. [DOI] [PubMed] [Google Scholar]
  • 59.Alexander BT, et al. Reduced uterine perfusion pressure during pregnancy in the rat is associated with increases in arterial pressure and changes in renal nitric oxide. Hypertension. 2001;37(4):1191–5. doi: 10.1161/01.hyp.37.4.1191. [DOI] [PubMed] [Google Scholar]
  • 60.Murphy JG, et al. Enhanced [Ca2+]i in renal arterial smooth muscle cells of pregnant rats with reduced uterine perfusion pressure. Am J Physiol Heart Circ Physiol. 2003;284(1):H393–403. doi: 10.1152/ajpheart.00247.2002. [DOI] [PubMed] [Google Scholar]
  • 61.Roberts L, et al. Enhanced endothelin synthesis by endothelial cells exposed to sera from pregnant rats with decreased uterine perfusion. Hypertension. 2006;47(3):615–8. doi: 10.1161/01.HYP.0000197950.42301.dd. [DOI] [PubMed] [Google Scholar]
  • 62.Alexander BT, et al. Tumor necrosis factor-alpha-induced hypertension in pregnant rats results in decreased renal neuronal nitric oxide synthase expression. Am J Hypertens. 2002;15(2 Pt 1):170–5. doi: 10.1016/s0895-7061(01)02255-5. [DOI] [PubMed] [Google Scholar]
  • 63**.Wallukat G, et al. Patients with preeclampsia develop agonistic autoantibodies against the angiotensin AT1 receptor. J Clin Invest. 1999;103(7):945–52. doi: 10.1172/JCI4106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Sedeek M, et al. Role of reactive oxygen species in hypertension produced by reduced uterine perfusion in pregnant rats. Am J Hypertens. 2008;21(10):1152–6. doi: 10.1038/ajh.2008.239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.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. 2007;50(6):1142–7. doi: 10.1161/HYPERTENSIONAHA.107.096594. [DOI] [PubMed] [Google Scholar]
  • 66.Gilbert JS, et al. Hypertension produced by placental ischemia in pregnant rats is associated with increased soluble endoglin expression. Hypertension. 2009;53(2):399–403. doi: 10.1161/HYPERTENSIONAHA.108.123513. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Kupferminc MJ, et al. Tumor necrosis factor-alpha is elevated in plasma and amniotic fluid of patients with severe preeclampsia. Am J Obstet Gynecol. 1994;170(6):1752–7. discussion 1757–9. [PubMed] [Google Scholar]
  • 68.Gadonski G, et al. Hypertension produced by reductions in uterine perfusion in the pregnant rat: role of interleukin 6. Hypertension. 2006;48(4):711–6. doi: 10.1161/01.HYP.0000238442.33463.94. [DOI] [PubMed] [Google Scholar]
  • 69.Heikkinen J, et al. Phenotypic characterization of regulatory T cells in the human decidua. Clin Exp Immunol. 2004;136(2):373–8. doi: 10.1111/j.1365-2249.2004.02441.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Saito S, et al. Th1/Th2/Th17 and regulatory T-cell paradigm in pregnancy. Am J Reprod Immunol. 2010;63(6):601–10. doi: 10.1111/j.1600-0897.2010.00852.x. [DOI] [PubMed] [Google Scholar]
  • 71.Sakaguchi S, et al. Regulatory T cells and immune tolerance. Cell. 2008;133(5):775–87. doi: 10.1016/j.cell.2008.05.009. [DOI] [PubMed] [Google Scholar]
  • 72.Sasaki Y, et al. Decidual and peripheral blood CD4+CD25+ regulatory T cells in early pregnancy subjects and spontaneous abortion cases. Mol Hum Reprod. 2004;10(5):347–53. doi: 10.1093/molehr/gah044. [DOI] [PubMed] [Google Scholar]
  • 73.Lash GE, Robson SC, Bulmer JN. Review: Functional role of uterine natural killer (uNK) cells in human early pregnancy decidua. Placenta. 2010;31(Suppl):S87–92. doi: 10.1016/j.placenta.2009.12.022. [DOI] [PubMed] [Google Scholar]
  • 74.Munoz-Suano A, Hamilton AB, Betz AG. Gimme shelter: the immune system during pregnancy. Immunol Rev. 2011;241(1):20–38. doi: 10.1111/j.1600-065X.2011.01002.x. [DOI] [PubMed] [Google Scholar]
  • 75.Tilburgs T, et al. Expression of NK cell receptors on decidual T cells in human pregnancy. J Reprod Immunol. 2009;80(1–2):22–32. doi: 10.1016/j.jri.2009.02.004. [DOI] [PubMed] [Google Scholar]
  • 76.Zenclussen AC, et al. Introducing a mouse model for pre-eclampsia: adoptive transfer of activated Th1 cells leads to pre-eclampsia-like symptoms exclusively in pregnant mice. Eur J Immunol. 2004;34(2):377–87. doi: 10.1002/eji.200324469. [DOI] [PubMed] [Google Scholar]
  • 77.Novotny SR, et al. Activating autoantibodies to the angiotensin II type I receptor play an important role in mediating hypertension in response to adoptive transfer of CD4+ T lymphocytes from placental ischemic rats. Am J Physiol Regul Integr Comp Physiol. 2012;302(10):R1197–201. doi: 10.1152/ajpregu.00623.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Novotny S, et al. CD4 T Cells Play a Critical Role in Mediating Hypertension in Response to Placental Ischemia. J Hypertens (Los Angel) 2013;2 doi: 10.4172/2167-1095.1000116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Cornelius DC, et al. Administration of Interleukin-17 Soluble Receptor C Suppresses TH17 Cells, Oxidative Stress, and Hypertension in Response to Placental Ischemia During Pregnancy. Hypertension. 2013 doi: 10.1161/HYPERTENSIONAHA.113.01514. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Kleinewietfeld M, Hafler DA. Regulatory T cells in autoimmune neuroinflammation. Immunol Rev. 2014;259(1):231–44. doi: 10.1111/imr.12169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Quinn KH, et al. The unique pathophysiology of early-onset severe preeclampsia: role of decidual T regulatory cells. J Reprod Immunol. 2011;91(1–2):76–82. doi: 10.1016/j.jri.2011.05.006. [DOI] [PubMed] [Google Scholar]
  • 82.Sasaki Y, et al. Proportion of peripheral blood and decidual CD4(+) CD25(bright) regulatory T cells in pre-eclampsia. Clin Exp Immunol. 2007;149(1):139–45. doi: 10.1111/j.1365-2249.2007.03397.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.von Rango U. Fetal tolerance in human pregnancy--a crucial balance between acceptance and limitation of trophoblast invasion. Immunol Lett. 2008;115(1):21–32. doi: 10.1016/j.imlet.2007.09.014. [DOI] [PubMed] [Google Scholar]
  • 84.Zenclussen AC, et al. Abnormal T-cell reactivity against paternal antigens in spontaneous abortion: adoptive transfer of pregnancy-induced CD4+CD25+ T regulatory cells prevents fetal rejection in a murine abortion model. Am J Pathol. 2005;166(3):811–22. doi: 10.1016/S0002-9440(10)62302-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Aluvihare VR, Kallikourdis M, Betz AG. Regulatory T cells mediate maternal tolerance to the fetus. Nat Immunol. 2004;5(3):266–71. doi: 10.1038/ni1037. [DOI] [PubMed] [Google Scholar]
  • 86.Redman CW, et al. Review: Does size matter? Placental debris and the pathophysiology of pre-eclampsia. Placenta. 2012;33(Suppl):S48–54. doi: 10.1016/j.placenta.2011.12.006. [DOI] [PubMed] [Google Scholar]
  • 87.Chamley LW, et al. Trophoblast deportation: just a waste disposal system or antigen sharing? J Reprod Immunol. 2011;88(2):99–105. doi: 10.1016/j.jri.2011.01.002. [DOI] [PubMed] [Google Scholar]
  • 88.Lau SY, et al. Necrotic trophoblast debris increases blood pressure during pregnancy. J Reprod Immunol. 2013;97(2):175–82. doi: 10.1016/j.jri.2012.12.005. [DOI] [PubMed] [Google Scholar]
  • 89.Hubel CA, et al. Agonistic angiotensin II type 1 receptor autoantibodies in postpartum women with a history of preeclampsia. Hypertension. 2007;49(3):612–7. doi: 10.1161/01.HYP.0000256565.20983.d4. [DOI] [PubMed] [Google Scholar]
  • 90.Brewer J, et al. Endothelin-1, oxidative stress, and endogenous angiotensin II: mechanisms of angiotensin II type I receptor autoantibody-enhanced renal and blood pressure response during pregnancy. Hypertension. 2013;62(5):886–92. doi: 10.1161/HYPERTENSIONAHA.113.01648. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.LaMarca B, et al. Hypertension in response to autoantibodies to the angiotensin II type I receptor (AT1-AA) in pregnant rats: role of endothelin-1. Hypertension. 2009;54(4):905–9. doi: 10.1161/HYPERTENSIONAHA.109.137935. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Parrish MR, et al. The effect of immune factors, tumor necrosis factor-alpha, and agonistic autoantibodies to the angiotensin II type I receptor on soluble fms-like tyrosine-1 and soluble endoglin production in response to hypertension during pregnancy. Am J Hypertens. 2010;23(8):911–6. doi: 10.1038/ajh.2010.70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Parrish MR, et al. Hypertension in response to AT1-AA: role of reactive oxygen species in pregnancy-induced hypertension. Am J Hypertens. 2011;24(7):835–40. doi: 10.1038/ajh.2011.62. [DOI] [PubMed] [Google Scholar]
  • 94.Xia Y, Kellems RE. Angiotensin receptor agonistic autoantibodies and hypertension: preeclampsia and beyond. Circ Res. 2013;113(1):78–87. doi: 10.1161/CIRCRESAHA.113.300752. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Quitterer U, Lother H, Abdalla S. AT1 receptor heterodimers and angiotensin II responsiveness in preeclampsia. Semin Nephrol. 2004;24(2):115–9. doi: 10.1016/j.semnephrol.2003.11.007. [DOI] [PubMed] [Google Scholar]
  • 96.AbdAlla S, et al. Increased AT(1) receptor heterodimers in preeclampsia mediate enhanced angiotensin II responsiveness. Nat Med. 2001;7(9):1003–9. doi: 10.1038/nm0901-1003. [DOI] [PubMed] [Google Scholar]
  • 97.Nagamatsu T, et al. Cytotrophoblasts up-regulate soluble fms-like tyrosine kinase-1 expression under reduced oxygen: an implication for the placental vascular development and the pathophysiology of preeclampsia. Endocrinology. 2004;145(11):4838–45. doi: 10.1210/en.2004-0533. [DOI] [PubMed] [Google Scholar]
  • 98.Nevo O, et al. Increased expression of sFlt-1 in in vivo and in vitro models of human placental hypoxia is mediated by HIF-1. Am J Physiol Regul Integr Comp Physiol. 2006;291(4):R1085–93. doi: 10.1152/ajpregu.00794.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Makris A, et al. Uteroplacental ischemia results in proteinuric hypertension and elevated sFLT-1. Kidney Int. 2007;71(10):977–84. doi: 10.1038/sj.ki.5002175. [DOI] [PubMed] [Google Scholar]
  • 100.Maynard SE, et al. Excess placental soluble fms-like tyrosine kinase 1 (sFlt1) may contribute to endothelial dysfunction, hypertension, and proteinuria in preeclampsia. J Clin Invest. 2003;111(5):649–58. doi: 10.1172/JCI17189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Bridges JP, et al. Oxidative stress contributes to soluble fms-like tyrosine kinase-1 induced vascular dysfunction in pregnant rats. Am J Hypertens. 2009;22(5):564–8. doi: 10.1038/ajh.2009.24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Murphy SR, et al. Role of endothelin in mediating soluble fms-like tyrosine kinase 1-induced hypertension in pregnant rats. Hypertension. 2010;55(2):394–8. doi: 10.1161/HYPERTENSIONAHA.109.141473. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Li Z, et al. Recombinant vascular endothelial growth factor 121 attenuates hypertension and improves kidney damage in a rat model of preeclampsia. Hypertension. 2007;50(4):686–92. doi: 10.1161/HYPERTENSIONAHA.107.092098. [DOI] [PubMed] [Google Scholar]
  • 104.Bergmann A, et al. Reduction of circulating soluble Flt-1 alleviates preeclampsia-like symptoms in a mouse model. J Cell Mol Med. 2010;14(6B):1857–67. doi: 10.1111/j.1582-4934.2009.00820.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Murphy S, et al. Role of Endothelin in Mediating Soluble fms-Like Tyrosine Kinase 1 Induced Hypertension in Pregnant Rats. Hypertension. 2010;55:394–398. doi: 10.1161/HYPERTENSIONAHA.109.141473. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Gilbert JS, et al. Recombinant vascular endothelial growth factor 121 infusion lowers blood pressure and improves renal function in rats with placentalischemia-induced hypertension. Hypertension. 2010;55(2):380–5. doi: 10.1161/HYPERTENSIONAHA.109.141937. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Davisson RL, et al. Discovery of a spontaneous genetic mouse model of preeclampsia. Hypertension. 2002;39(2 Pt 2):337–42. doi: 10.1161/hy02t2.102904. [DOI] [PubMed] [Google Scholar]
  • 108.Wallace K, et al. Hypertension in response to CD4(+) T cells from reduced uterine perfusion pregnant rats is associated with activation of the endothelin-1 system. Am J Physiol Regul Integr Comp Physiol. 2012;303(2):R144–9. doi: 10.1152/ajpregu.00049.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Woods AK, et al. Adenoviral delivery of VEGF121 early in pregnancy prevents spontaneous development of preeclampsia in BPH/5 mice. Hypertension. 2011;57(1):94–102. doi: 10.1161/HYPERTENSIONAHA.110.160242. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Shah DM. Role of the renin-angiotensin system in the pathogenesis of preeclampsia. Am J Physiol Renal Physiol. 2005;288(4):F614–25. doi: 10.1152/ajprenal.00410.2003. [DOI] [PubMed] [Google Scholar]
  • 111.Takimoto E, et al. Hypertension induced in pregnant mice by placental renin and maternal angiotensinogen. Science. 1996;274(5289):995–8. doi: 10.1126/science.274.5289.995. [DOI] [PubMed] [Google Scholar]
  • 112.Bohlender J, Ganten D, Luft FC. Rats transgenic for human renin and human angiotensinogen as a model for gestational hypertension. J Am Soc Nephrol. 2000;11(11):2056–61. doi: 10.1681/ASN.V11112056. [DOI] [PubMed] [Google Scholar]
  • 113.Verlohren S, et al. Uterine vascular function in a transgenic preeclampsia rat model. Hypertension. 2008;51(2):547–53. doi: 10.1161/HYPERTENSIONAHA.107.103176. [DOI] [PubMed] [Google Scholar]
  • 114.Falcao S, et al. Mice overexpressing both human angiotensinogen and human renin as a model of superimposed preeclampsia on chronic hypertension. Hypertension. 2009;54(6):1401–7. doi: 10.1161/HYPERTENSIONAHA.109.137356. [DOI] [PubMed] [Google Scholar]

RESOURCES