Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 Oct 1.
Published in final edited form as: Hypertension. 2013 Jul 29;62(4):10.1161/HYPERTENSIONAHA.113.00588. doi: 10.1161/HYPERTENSIONAHA.113.00588

RECENT ADVANCES IN THE UNDERSTANDING OF THE PATHOPHYSIOLOGY OF PREECLAMPSIA

Junie P Warrington 1, Eric M George 1, Ana C Palei 1, Frank T Spradley 1, Joey P Granger 1
PMCID: PMC3856636  NIHMSID: NIHMS514582  PMID: 23897068

Introduction

Preeclampsia, a pregnancy specific disorder, is typically defined as new-onset hypertension presenting after the 20th week of gestation with proteinuria. The overall prevalence in the United States is 3-8% with higher incidence in specific ethnic subpopulations, notably African-Americans. Preeclampsia is a major source of maternal and neonatal morbidity and mortality. Moreover, women who endure preeclampsia are at greater risk for cardiovascular diseases later in life. Studies published in Hypertension and other journals over the last few years have not only provided new insights into potential mechanisms underlying the pathophysiology of preeclampsia but also into the identification of potential biomarkers for the early diagnosis of preeclampsia. While numerous factors have been implicated in the pathophysiology of preeclampsia, the main focus of this Recent Advances article is to review recent studies that link placental ischemia, endothelial and vascular dysfunction, and hypertension in preeclampsia.

Biomarkers for the diagnosis of preeclampsia

Despite intense research into the identification of molecular markers of preeclampsia, a reliable and accurate marker for the early diagnosis of preeclampsia remains elusive. A number of recent papers have suggested markers that could prove useful in the diagnosis of the disorder. With the growing interest in autoimmunity in preeclampsia, several studies have looked at immune factors in the maternal circulation as possible biomarkers. For instance, Siddiqui et al found widespread (~95%) presence of agonistic autoantibodies to the angiotensin type-1 receptor (AT1-AA). Additionally, the levels of these autoantibodies correlated well with the degree of disease severity, suggesting potential utility in diagnosis 1. In a related study, Jensen et al found that CD19(+)CD5(+) B-cell populations, a potential source for the AT1-AA, are significantly elevated in the maternal circulation of preeclamptic patients during late gestation when compared to normal pregnant controls at the same gestational stage 2.

One of the most active areas of research in preeclampsia is angiogenic imbalance, principally driven by the production of soluble fms-like tyrosine kinase-1 (sFlt-1), the soluble VEGF and placental growth factor (PlGF) receptor antagonist. Since the initial studies in which sFlt-1 was proposed as a biomarker for the diagnosis of preeclampsia 3, a number of groups have investigated the utility of angiogenic factors as reliable diagnostic markers. Two recent studies have lent credence to this approach. In the first study, Ohkuchi et al found that the sFlt-1/PlGF ratio was a useful component for predicting preeclampsia when measured at 26-31 weeks of gestation 4. Likewise, Perni et al examined angiogenic factors in patients who had preexisting hypertension with superimposed preeclampsia, and found higher circulating levels of sFlt-1 prior to the 20th week of gestation in patients with preeclampsia versus pregnant women who had preexisting hypertension but did not develop preeclampsia 5. While these studies, along with other recent work, suggest that angiogenic balance could be a reliable marker of preeclampsia and allow detection prior to the onset of patient symptoms, others have argued that angiogenic factors may be better markers of intra-uterine growth restriction without the required presence of preeclampsia. However, Romero and colleagues recently reported that changes in the maternal plasma concentration of soluble endoglin (s-Eng), sFlt-1, and PlGF precede the clinical presentation of preeclampsia, but only changes in s-Eng and PlGF precede the delivery of a small for gestational age neonate 6.

In a recent expert opinion article in Hypertension, Staff et al argued that the classic definitions of preeclampsia, based on concepts that are now more than 50 years old, have become outdated and that the definition could be modernized to take account of our current understanding of disease pathophysiology 7. They proposed that the definition of preeclampsia includes the placental biomarker PlGF. They also suggested that the definition may need to be expanded in the future to include other factors such as the anti-angiogenic factors, sFlt, and soluble endoglin 7. Sugulle et al also recently reported that the mid-region-proANP may represent another novel biomarker in preeclampsia. Changes in MR-proANP likely reflect the hemodynamic and cardiovascular alterations and endothelial dysfunction in preeclampsia. The authors also suggested that MR-proANP could represent a supplement to the well-established biomarkers of preeclampsia of predominantly placental origin 8.

Another potential approach to the early diagnosis of preeclampsia is through a multifactorial molecular screen. To this end, Kenny et al utilized mass-spectrometry based prospective metabolomic screening of plasma from preeclamptic patients at 15 weeks of gestation to identify 14 distinct metabolites, which when measured in concert, provided positive predictive value for the development of preeclampsia 9. Meanwhile, Carty et al utilized mass spectrometry-based proteomics to examine the urinary protein profile of preeclamptic patients. The authors identified a panel of 50 peptides including uromodulin, collagen fragments, and fibrinogen, which at gestational week 28 were associated with the development of preeclampsia 10. Taken together, these studies suggest that a combination of immune and angiogenic factors, possibly coupled to mass spec-based profiling of proteins and small molecule metabolites, could provide a definitive diagnostic tool to identify patients at high risk for the development of preeclampsia.

Abnormal implantation and vasculogenesis in preeclampsia

To ensure adequate oxygen and nutrient delivery to the developing uteroplacental unit during normal pregnancy, fetally-derived cytotrophoblasts invade and remodel the maternal uterine spiral arteries. This complex process results in the conversion of the high-resistance, small-diameter spiral arteries into high-capacitance, low-resistance vessels. During preeclampsia, it is thought that poor cytotrophoblast invasion leads to abnormal vascular remodeling and inadequate oxygen delivery to the developing uteroplacental unit. The exact mechanisms responsible for the abnormal placental trophoblast invasion and vascular remodeling in preeclampsia are unclear, but a number of recent studies have increased our understanding of potential mechanisms that may lead to maladaptations. For example, Hering et al recently reported that local tissue Ang II stimulates trophoblast invasion in vivo in the rat and in vitro in human cells. The authors suggested that upregulation of tissue Ang II in the maternal part of the placenta, represents an important growth factor for trophoblast migration and invasion, and abnormalities in this system may play a role in the pathophysiology of preeclampsia 11.

The Notch signaling pathway is thought to play an important role in vasculogenesis by modulating differentiation and function during cell-cell contact. The main pathway consists of four transmembrane receptors (NOTCH1-4) and five ligands (DLL1/3/4 and JAG1/2). Binding of receptors and ligands on adjacent cells triggers serial proteolytic cleavage of the receptor, releasing the Notch intracellular domain that subsequently translocates to the nucleus to bind to transcription factors and induce downstream targets. Supporting a role for Notch signaling in vascular remodeling is a recent report by Hunkapiller et al where the absence of Notch2 in mice is associated with reduced vessel diameter and placental perfusion 12. Additional findings that peri- and endo-vascular cytotrophoblasts often fail to express the Notch ligand, JAG1, in preeclampsia provide further evidence that defects in Notch signaling may be an important part of the pathogenesis of this pregnancy complication 12.

Another recently described molecular pathway implicated in placental vascular development is the transcription factor storkhead box 1 (STOX1), a member of the winged helix transcription factor family. STOX1 was originally implicated in an epidemiological study that suggested increased rates of STOX1 mutation in women who experienced preeclampsia 13. A recent report from Doridot et al. demonstrated that transgenic overexpression of STOX1 in the mouse leads to a phenotype that mimics preeclampsia in several key ways, most notably, a dramatic rise in systolic blood pressure during gestation, proteinuria and elevated maternal circulating levels of sFlt-1 and soluble endoglin 14. While these data are intriguing, much work remains to be done to elucidate the causative and symptomatic role of STOX1 in the development of preeclampsia.

Angiogenic factors

There is strong evidence that an imbalance between pro- and anti-angiogenic factors exist in preeclampsia favoring the anti-angiogenic agents, such as sFlt-1. sFlt-1 is a soluble splice variant of the vascular endothelial growth factor receptor-1 (VEGFR-1), that is able to prevent the actions of pro-angiogenic molecules, such as VEGF and PlGF in target tissues 15, 16. Additional alternatively spliced Flt-1 transcripts have been identified and Jebbink et al found that preeclamptic placentas have about 3-fold higher expression of all mRNA variants compared with controls, with a slight shift to the sFLT-1_v1 17.

Alterations in angiogenic factors are detectable before the clinical diagnosis of preeclampsia 4, 18, 19, including chronic hypertensive women who developed subsequently superimposed preeclampsia 5, 20, and women after 5-8 years of delivery 21. Besides single determinations of plasma sFlt-1 concentration and sFlt-1/PlGF ratio, Woodham et al suggest that the combination of mid-pregnancy 25 hydroxyvitamin D (25(OH)D) level with sFlt-1/PlGF ratio is a better tool to predict the development of severe preeclampsia 22. Siddiqui et al also found that the titer of AT1-AA is significantly correlated with sFlt-1 1. Interestingly, a pilot study in extracorporeal removal of sFlt-1 from women with very preterm preeclampsia revealed that dextran sulfate apheresis can lower circulating sFlt-1 levels, reduce proteinuria, and stabilize blood pressure without adverse maternal and fetal outcomes 23.

Recent in vitro studies have focused on mechanisms whereby sFlt-1, which is strongly bound to the extracellular matrix, gains access to the maternal circulation. Al-Ani et al demonstrated in HUVECs that this process may occur through activation of endothelial proteinase-activated receptor-2 (PAR-2) leading to increased sFlt-1 release 24. Gould et al reported that urotensin-II (U-II), a vasoconstrictor and pro-angiogenic agent elevated in preeclampsia, is associated with increased sFlt-1 secretion from placental explants under hypoxic conditions. In addition, the authors showed that hypoxia augments the levels of U-II receptors in syncytiotrophoblasts, and the U-II receptor expression is upregulated in preeclamptic placentas compared with controls 25. Another study from Karumanchi’s group revealed that the syncytiotrophoblast layer forms abundant “knots” enriched with sFlt-1 during preeclampsia. The easy detachment of these knots from the placenta produces free, transcriptionally active syncytial aggregates that can become an autonomous source of sFlt-1 delivery into the circulation 26.

Placental ischemia in rats, as a result of reduced uterine perfusion pressure (RUPP), results in increased sFlt-1 and decreased free VEGF levels in the circulation 27-29. In addition to the RUPP model, the BPH/5 mouse and other published models have disrupted angiogenic balance 25-29. A number of studies have recently demonstrated that restoration of free VEGF levels in animal models of preeclampsia results in significant improvement in renal function and blood pressure. Gilbert et al reported that chronic infusion of VEGF(121) into placental ischemic rats restores their glomerular filtration rate and endothelial function, resulting in a reduction of the high blood pressure associated with placental ischemia 30. Using BPH/5 mice, Woods et al found that adenoviral-mediated delivery of VEGF(121) prevented the late-gestational elevation of blood pressure and proteinuria, and reduced the incidence of fetal reabsorptions 31. These interesting results suggest that VEGF(121) may be a candidate pharmacotherapy for preeclampsia.

In an attempt to elucidate the mechanisms whereby the enhanced sFlt-1 production seen in preeclampsia leads to hypertension during pregnancy, Murphy et al showed that exogenous sFlt-1 administration into pregnant rats results in high arterial blood pressure and elevated renal preproendothelin mRNA expression32. The simultaneous administration of a selective endothelin type A receptor antagonist brought the blood pressure to normal pregnant range, suggesting that endothelin-1, via endothelin type A receptor, is an important mediator of the hypertension secondary to excess sFlt-1 in pregnancy 32. Li et al also recently reported that reduced maternal eNOS/nitric oxide exacerbates the sFlt1-related preeclampsia-like phenotype through activation of the endothelin system 33. Other processes in which angiogenic factors may play a role include: relaxin-induced vasodilatation during normal pregnancy 34 and increased sensitivity to angiotensin-II during postpartum of preeclamptic women with low-sodium balance 35.

Immune Factors and Inflammation in Preeclampsia

The immune system and inflammatory factors are thought to play an important role in the pathophysiology of preeclampsia. Redman and colleagues have suggested that the inflammatory response is triggered by placental debris or particles, ranging from large deported multinuclear fragments to sub-cellular components, shed from the syncytial surface of the human placenta 36. Utilizing nanoparticle tracking analysis to measure the size and concentration of syncytiotrophoblast vesicles obtained by placental perfusion, they recently found that vesicles range in size from 50nm to 1μm with the majority being <500nm. The authors speculated that changes in the number and size of beneficial syncytiotrophoblast exosomes and harmful microvesicles may be important in the maternal syndrome of preeclampsia.

Maternal immune tolerance involves crucial interactions between regulatory CD4+ T cells and uterine natural killer cells recognizing and accepting the fetal antigens and facilitating placental growth. Abnormalities in this process have been proposed to lead to poor placentation, reduced placental perfusion and stress, and chronic immune activation. Preeclamptic women 37 and placental ischemic rats have a decrease in circulating regulatory CD4+ T cells 38. T helper 17 cells, which are upregulated in a variety of autoimmune disorders, are also increased in preeclamptic women and in placental ischemic rats 38. While these data support the hypothesis that hypertension in response to placental ischemia represents a shift from the normal anti-inflammatory state of pregnancy to a pro-inflammatory state, the quantitative importance of CD4+ T cells and T helper 17 cells in the pathophysiology of preeclampsia remains to be determined.

Another area related to the immune component of preeclampsia is research on the agonistic autoantibodies to the angiotensin Type 1 receptor (AT1-AA). These autoantibodies, isolated over a decade ago in preeclamptic women, have been studied more intensively recently, including their identification in the circulation of rats undergoing placental ischemia. LaMarca and colleagues recently reported the role of B-lymphocyte activation in the production of AT1-AA in placental ischemic rats 39. They found that B-cell depletion resulted in reductions in AT1-AA and was associated with decreases in blood pressure and tissue ET-1 levels. Wenzel et al recently generated and purified activating antibodies against the AT1 receptor (AT1-AB) by immunizing rabbits against the AFHYESQ epitope of the second extracellular loop, which is the binding epitope of endogenous activating AT1-AA from patients with preeclampsia. They reported that the AT1-ABs increase Ang II sensitivity, possibly explaining the increased Ang II sensitivity observed in preeclamptic patients. They also found that passive transfer of AT1-ABs alone into pregnant rats did not induce a preeclamptic syndrome. However, the combination of AT1-ABs and Ang II produced hypertension, proteinuria, abnormal placental vascular remodeling, and intrauterine growth restriction. Additionally, AT1-AB plus Ang II induced hypoxia inducible factor (HIF-1), which was associated with arteriolosclerotic lesions in the spiral arteries and altered trophoblast invasion. Finally, they also found that the AT1-AB enhanced Ang II induced ET-1 expression 40. Taken together, these data demonstrate the important roles that B-lymphocyte activation and AT1-AA play in the pathophysiology of hypertension in response to placental ischemia.

Pro- and anti-inflammatory cytokines are also involved in the pathogenesis of preeclampsia. Pro-inflammatory cytokines such as TNF and IL-6 are elevated in preeclamptic women and in placental ischemic rats. These pro-inflammatory cytokines have not only been shown to stimulate the production of reactive oxygen species, endothelin-1, and the AT1-AA, but also play an important role in increasing blood pressure in response to placental ischemia in pregnant rats. Loss of anti-inflammatory actions may also be important in preeclampsia. Chatterjee et al reported that absence of interleukin 10 exacerbated the hypertension and endothelial dysfunction in a mouse model of preeclampsia caused by Toll-like receptor 3 activation during pregnancy 41. Moreover, they reported that exogenous IL-10 treatment had beneficial effects on endothelial function in the model and suggested that IL-10 therapy may be beneficial for women with preeclampsia.

Heme Oxygenase in Pregnancy and Preeclampsia

One system that has received great attention in recent years is the heme oxygenase-1 (HO-1) system which has been shown to be important for normal placental vascular function, 42-44. Induction of HO-1 has been shown to be a potent anti-hypertensive agent in several forms of experimental hypertension 45-49. The precise mechanisms that govern HO-1 antihypertensive effects are still under investigation, but are hypothesized to function in part through the actions of its biologically active metabolites carbon monoxide (CO), a potent vasodilator and inhibitor of sFlt-1 production, and bilirubin, which has powerful antioxidant properties 45, 50.

Several recent papers have suggested a potential role of the HO-1 system in the management of preeclampsia in both in vivo animal models and in the human population. Wikström et al investigated the effect of cigarette smoking on the incidence of preeclampsia in a Swedish population 51. Women who smoked in mid to late gestation had up to 50% lower incidence of preeclampsia when compared to women using no tobacco products or snuff alone, with degree of use positively correlating with greater protection. From this, the authors concluded that the combustion products of tobacco, which include CO, are responsible for the decreased incidence of preeclampsia in this population. In a subsequent study, we investigated the effects of HO-1 induction in the RUPP rodent model of placental ischemia-induced hypertension 29. Induction of HO-1 by Cobalt Protoporphyrin led to a ~50% attenuation of the RUPP-induced hypertension on gestational day 19. More interestingly, this was accompanied by improvements in angiogenic balance in both the placenta itself and an increase in bioavailable VEGF in the maternal circulation. In addition, the placental oxidative stress, characteristic of both preeclamptic patients and the RUPP rat, was significantly attenuated by HO-1 induction, presumably by enhanced bilirubin production. As a result, the increase in the production of vascular preproendothelin-1 seen in this model was significantly reduced. Following studies also demonstrated attenuation of sFlt-1 induced hypertension, again accompanied by enhanced free VEGF in the maternal circulation and suppression of vascular preproendothelin-1 expression 52.

Two related studies by McCarthy et al also suggest a possible role for HO-1 in both the development and treatment of preeclampsia. Using the rodent RUPP model, McCarthy et al demonstrated that the peroxisome proliferator-activated receptor-γ (PPAR-γ) agonist rosiglitizone could ameliorate many of the pathological manifestations in response to placental ischemia, including hypertension and vascular dysfunction. Furthermore, the beneficial effects of rosiglitizone treatment were dependent on HO-1, as co-administration of the HO inhibitor, SnMP, significantly blocked rosigilitzone’s effects 53. In a subsequent study, the same group found that administration of a PPAR-γ antagonist in rodents during pregnancy mimicked several features of preeclampsia, including hypertension, endothelial dysfunction, and fetal growth restriction. Interestingly, and perhaps counterintuitive to the earlier work, PPAR-γ antagonism resulted in a significant increase in the levels of circulating and placental HO-1, despite worsening angiogenic balance and hypertension 54.

While some of the studies described above provide potential support for the HO-1 system as a therapeutic target in preeclampsia, more work is still needed. In this respect, statins have been shown to stimulate HO-1 expression and inhibit sFlt-1 release in vivo and in vitro; thus, they have the potential to ameliorate early onset preeclampsia. The StAmP trial is currently addressing this potential therapeutic intervention and if positive, statins may be adopted as a therapeutic intervention to prolong affected pregnancies 55.

Endothelial and vascular dysfunction in preeclampsia

The vascular endothelium has many important functions including control of smooth muscle tone through release of vasoconstrictor and vasodilatory substances, and regulation of anti-coagulation, anti-platelet and fibrinolysis functions via release of different soluble factors. The maternal vascular endothelium appears to be an important target for factors that are triggered by placental ischemia/hypoxia in preeclampsia 29, 30, 38, 39, 53. An important link between placental ischemia and vascular function was established in a recent study showing that treatment of small mesenteric arteries from normal pregnant rats with serum from placental ischemic rats attenuates endothelial-dependent relaxation via reduced nitric oxide synthase NOS function 56. NO, not only causes vasodilation, but also has anti-inflammatory and anti-oxidant actions in blood vessels 57, 58. Interestingly, there is increased endothelial adherence and infiltration of neutrophils in omental arteries from preeclamptic compared to normal pregnant women 59. This study showed that reactive oxygen species (ROS) derived from neutrophils augment angiotensin (Ang)II-induced vasoconstriction in omental arteries from normal pregnant women and this effect was exacerbated in endothelium-denuded artery preparations. These data suggest that reduced endothelial NOS function in preeclampsia increases susceptibility to inflammation-induced vascular dysfunction. The need for proper anti-inflammatory mechanisms in normal pregnancy was further demonstrated by the finding that aortic endothelial dysfunction and hypertension in pregnant mice have concomitant deficiency of IL-10 and activation of TLR3 41,60. Reduced action of the transcription factor peroxisome-proliferator activated receptor PPAR-γ is found in RUPP rats 53, 54, 59. Because IL-10 and PPAR-γ positively regulate endothelial NOS 61,62, failure in these pathways may play a role in the pathophysiology of preeclampsia.

Recent studies identified potential pathways in vascular smooth muscle cells (VSMCs) that may fail and promote preeclampsia. Given the importance of adequate placental perfusion, it was demonstrated in uterine arteries from normal, near-term pregnant versus non-pregnant sheep that myogenic tone is reduced; this was mediated by increased expression and activity of large conductance Ca2+-activated K+ (BKCa) channels 63. This ex vivo study showed that 17β-estradiol increases BKCa channel activity in isolated uterine arterial VSMCs. Likewise, the novel ovarian hormone relaxin, which promotes Ca2+ efflux from uterine smooth muscle 64, mediates reduced myogenic tone of renal arteries during pregnancy 34, 65, 66. Also related to the importance of reduced myogenic tone in pregnancy, Xiao et al demonstrated that actin polymerization is reduced in ovine uterine arteries in normal-pregnant sheep 67 whereas activity and expression of RhoA kinase (ROCK), which facilitates Ca2+ sensitization of the contractile apparatus and actin polymerization 68, is increased in omental arteries from preeclamptic women 69. Whether reduced sex hormone regulation of intracellular Ca2+ promotes ROCK activation to induce the known increase in uterine artery resistance in RUPP 70, 71 is not defined.

Cerebrovascular changes in pregnancy and preeclampsia

As discussed earlier, it is widely established that preeclampsia contributes to gross maternal vascular dysfunction. Specifically, cerebrovascular abnormalities play a significant role in the pathogenesis of preeclampsia/eclampsia. An astounding 39% of all preeclampsia/eclampsia deaths are due to cerebrovascular events with cerebral hemorrhage contributing 35%, cerebral edema 3%, and cerebral embolus 1% of preeclampsia-related deaths 72. Neurological symptoms such as headaches, blurred vision, nausea, drowsiness, and seizures are commonly reported in preeclamptic patients 73. Furthermore, women with preeclampsia/eclampsia are at increased risk of developing stroke during pregnancy and the postpartum year 74. Presently, the etiology of preeclampsia-induced cerebrovascular abnormalities is still poorly understood.

Magnetic resonance imaging and computed tomography scans reveal abnormalities consistent with edema in preeclamptic patients 75, 76. Edema forms from either increased water transport into cells (cytotoxic edema) or through the disruption of the blood brain barrier (BBB – vasogenic edema). The BBB, formed by the close association of endothelial cells, smooth muscle cells or pericytes (capillaries), and astrocytes, regulates the transport of substances between the blood and brain tissue. Increased permeability of the cerebral vessels has been reported in both normal pregnancy and preeclampsia. For example, plasma from normal pregnant and preeclamptic women increases permeability of cerebral vessels in an ex vivo model 77. This study suggests that pregnancy itself induces changes in the cerebral vasculature, which may be exacerbated in the presence of increased arterial pressure, characteristic of preeclampsia.

Cerebral blood flow is highly regulated and kept relatively constant even with fluctuations in blood pressure. Acute increases in blood pressure activate the vascular myogenic response, protecting neuronal tissue from damage. Women with severe preeclampsia have increased cerebral blood flow and perfusion pressure 78 and our lab recently demonstrated impaired myogenic tone in the middle cerebral arteries and cerebral edema in placental ischemic rats 79. These conditions of increased cerebral blood flow and impaired myogenic reactivity render preeclamptic patients susceptible to neurological complications with acute increases in blood pressure. This concept is supported by studies demonstrating that during acute hypertension, pregnancy decreases vascular resistance and increases cerebral blood flow 78, 80, resulting in a rightward shift in the autoregulatory curve 81, and cerebral edema.

While the myogenic response is important during sudden, acute elevations in blood pressure, vascular remodeling occurs during chronic elevations of blood pressure. Pregnancy is associated with outward remodeling of penetrating arterioles and increased cerebral capillary density 80 while hypertension induces inward remodeling of posterior cerebral arteries 82,83. Importantly, pregnancy reverses 84 or prevents 85 the inward remodeling of cerebral arteries in hypertensive animals. Together, these studies suggest that failure of the cerebral vasculature to remodel in the presence of decreased vascular resistance and increased cerebral blood flow, results in the transmission of pressure to the cerebral micro-vessels causing BBB disruption and edema.

Cerebral arteries (pial arteries) are innervated by both sympathetic and parasympathetic nerves. Although nerve innervation is thought to play a minimal role in the control of cerebral blood flow, nerve density on posterior cerebral arteries increases during pregnancy and decreases during pregnancy accompanied by hypertension 82. Interestingly, plasma from pregnant rats induces neuronal hyperexcitability and increased microglial activation through TNF alpha activity 86 thereby demonstrating that circulating factors can exert effects on neurons if they come into direct contact as may occur during cerebral hemorrhage or micro-bleeds. Because preeclampsia is associated with increased circulating factors such as vasoactive peptides and inflammatory cytokines, increased cerebral perfusion pressure along with impaired myogenic reactivity, could result in BBB disruption, cerebral edema, and/or excitability of neurons resulting in the neurological symptoms of preeclampsia.

Concluding Remarks

Despite recent advances in identifying the factors involved in the pathophysiology of preeclampsia, there are still many unanswered questions. The initiating events that eventually manifest the disorder remain to be fully elucidated. While abnormal spiral artery remodeling is thought to play a critical role, the underlying cellular and molecular mechanisms remain obscure. As seen in Figure 1, abnormal cytotrophoblast invasion and spiral artery remodeling is proposed to lead to ischemia/hypoxia-mediated increases in sFlt-1, AT1-AA, and inflammatory cytokines, resulting in subsequent detrimental effects on several downstream targets. The decrease in pro-angiogenic factors such as VEGF, and PlGF and the increased production of reactive oxygen species lead to maternal endothelial dysfunction. As a result, increased endothelin (ET-1) production and the decreased bioavailability of NO, lead to the characteristic widespread vascular abnormalities in various organs such as the kidneys and brain, hypertension, and proteinuria observed in this disorder. Currently, the relative importance of each of these factors needs to be ascertained, and the network of cross regulation that governs them needs to be fully elucidated. Unfortunately, the lack of effective pharmacologic therapeutic approaches for the management of preeclampsia is a serious health concern in clinical obstetrics. The identification of new therapies that focus on manipulating these newly defined pathologic pathways is a critical area for future research.

Figure 1. Schematic representation of the proposed initiating events and factors in the pathophysiology of preeclampsia.

Figure 1

sFlt-1 – soluble fms-like tyrosine kinase −1, VEGF – vascular endothelial growth factor, PIGF – placental growth factor, ET-1 – endothelin-1, NO – nitric oxide, RBF – renal blood flow, GFR – glomerular filtration rate.

Acknowledgments

Sources of Funding: This work was supported by NIH grants HL051971, HL108618, and 1T32HL105324

Footnotes

Conflict of Interest/Disclosure: None

This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Siddiqui AH, Irani RA, Blackwell SC, Ramin SM, Kellems RE, Xia Y. Angiotensin receptor agonistic autoantibody is highly prevalent in preeclampsia: Correlation with disease severity. Hypertension. 2010;55:386–393. doi: 10.1161/HYPERTENSIONAHA.109.140061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Jensen F, Wallukat G, Herse F, Budner O, El-Mousleh T, Costa SD, Dechend R, Zenclussen AC. Cd19+cd5+ cells as indicators of preeclampsia. Hypertension. 2012;59:861–868. doi: 10.1161/HYPERTENSIONAHA.111.188276. [DOI] [PubMed] [Google Scholar]
  • 3.Levine RJ, Maynard SE, Qian C, Lim KH, England LJ, Yu KF, Schisterman EF, Thadhani R, Sachs BP, Epstein FH, Sibai BM, Sukhatme VP, Karumanchi SA. Circulating angiogenic factors and the risk of preeclampsia. N Engl J Med. 2004;350:672–683. doi: 10.1056/NEJMoa031884. [DOI] [PubMed] [Google Scholar]
  • 4.Ohkuchi A, Hirashima C, Matsubara S, Takahashi K, Matsuda Y, Suzuki M. Threshold of soluble fms-like tyrosine kinase 1/placental growth factor ratio for the imminent onset of preeclampsia. Hypertension. 2011;58:859–866. doi: 10.1161/HYPERTENSIONAHA.111.174417. [DOI] [PubMed] [Google Scholar]
  • 5.Perni U, Sison C, Sharma V, Helseth G, Hawfield A, Suthanthiran M, August P. Angiogenic factors in superimposed preeclampsia: A longitudinal study of women with chronic hypertension during pregnancy. Hypertension. 2012;59:740–746. doi: 10.1161/HYPERTENSIONAHA.111.181735. [DOI] [PubMed] [Google Scholar]
  • 6.Romero R, Nien JK, Espinoza J, Todem D, Fu W, Chung H, Kusanovic JP, Gotsch F, Erez O, Mazaki-Tovi S, Gomez R, Edwin S, Chaiworapongsa T, Levine RJ, Karumanchi SA. A longitudinal study of angiogenic (placental growth factor) and anti-angiogenic (soluble endoglin and soluble vascular endothelial growth factor receptor-1) factors in normal pregnancy and patients destined to develop preeclampsia and deliver a small for gestational age neonate. J Matern Fetal Neonatal Med. 2008;21:9–23. doi: 10.1080/14767050701830480. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Staff AC, Benton SJ, von Dadelszen P, Roberts JM, Taylor RN, Powers RW, Charnock-Jones DS, Redman CW. Redefining preeclampsia using placenta-derived biomarkers. Hypertension. 2013;61:932–942. doi: 10.1161/HYPERTENSIONAHA.111.00250. [DOI] [PubMed] [Google Scholar]
  • 8.Sugulle M, Herse F, Hering L, Mockel M, Dechend R, Staff AC. Cardiovascular biomarker midregional proatrial natriuretic peptide during and after preeclamptic pregnancies. Hypertension. 2012;59:395–401. doi: 10.1161/HYPERTENSIONAHA.111.185264. [DOI] [PubMed] [Google Scholar]
  • 9.Kenny LC, Broadhurst DI, Dunn W, Brown M, North RA, McCowan L, Roberts C, Cooper GJ, Kell DB, Baker PN. Robust early pregnancy prediction of later preeclampsia using metabolomic biomarkers. Hypertension. 2010;56:741–749. doi: 10.1161/HYPERTENSIONAHA.110.157297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Carty DM, Siwy J, Brennand JE, Zurbig P, Mullen W, Franke J, McCulloch JW, Roberts CT, North RA, Chappell LC, Mischak H, Poston L, Dominiczak AF, Delles C. Urinary proteomics for prediction of preeclampsia. Hypertension. 2011;57:561–569. doi: 10.1161/HYPERTENSIONAHA.110.164285. [DOI] [PubMed] [Google Scholar]
  • 11.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. 2010;56:311–318. doi: 10.1161/HYPERTENSIONAHA.110.150961. [DOI] [PubMed] [Google Scholar]
  • 12.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. 2011;138:2987–2998. doi: 10.1242/dev.066589. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.van Dijk M, Mulders J, Poutsma A, Könst AA, Lachmeijer AM, Dekker GA, Blankenstein MA, Oudejans CB. Maternal segregation of the dutch preeclampsia locus at 10q22 with a new member of the winged helix gene family. Nat Genet. 2005;37:514–519. doi: 10.1038/ng1541. [DOI] [PubMed] [Google Scholar]
  • 14.Doridot L, Passet B, Méhats C, Rigourd V, Barbaux S, Ducat A, Mondon F, Vilotte M, Castille J, Breuiller-Fouché M, Daniel N, le Provost F, Bauchet AL, Baudrie V, Hertig A, Buffat C, Simeoni U, Germain G, Vilotte JL, Vaiman D. Preeclampsia-like symptoms induced in mice by fetoplacental expression of stox1 are reversed by aspirin treatment. Hypertension. 2013;61:662–668. doi: 10.1161/HYPERTENSIONAHA.111.202994. [DOI] [PubMed] [Google Scholar]
  • 15.Lam C, Lim KH, Karumanchi SA. Circulating angiogenic factors in the pathogenesis and prediction of preeclampsia. Hypertension. 2005;46:1077–1085. doi: 10.1161/01.HYP.0000187899.34379.b0. [DOI] [PubMed] [Google Scholar]
  • 16.Lindheimer MD, Romero R. Emerging roles of antiangiogenic and angiogenic proteins in pathogenesis and prediction of preeclampsia. Hypertension. 2007;50:35–36. doi: 10.1161/HYPERTENSIONAHA.107.089045. [DOI] [PubMed] [Google Scholar]
  • 17.Jebbink J, Keijser R, Veenboer G, van der Post J, Ris-Stalpers C, Afink G. Expression of placental flt1 transcript variants relates to both gestational hypertensive disease and fetal growth. Hypertension. 2011;58:70–76. doi: 10.1161/HYPERTENSIONAHA.110.164079. [DOI] [PubMed] [Google Scholar]
  • 18.Banek CT, Gilbert JS. Approaching the threshold for predicting preeclampsia: Monitoring angiogenic balance during pregnancy. Hypertension. 2011;58:774–775. doi: 10.1161/HYPERTENSIONAHA.111.175992. [DOI] [PubMed] [Google Scholar]
  • 19.Rana S, Powe CE, Salahuddin S, Verlohren S, Perschel FH, Levine RJ, Lim KH, Wenger JB, Thadhani R, Karumanchi SA. Angiogenic factors and the risk of adverse outcomes in women with suspected preeclampsia. Circulation. 2012;125:911–919. doi: 10.1161/CIRCULATIONAHA.111.054361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Garovic VD. The role of angiogenic factors in the prediction and diagnosis of preeclampsia superimposed on chronic hypertension. Hypertension. 2012;59:555–557. doi: 10.1161/HYPERTENSIONAHA.111.184192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Kvehaugen AS, Dechend R, Ramstad HB, Troisi R, Fugelseth D, Staff AC. Endothelial function and circulating biomarkers are disturbed in women and children after preeclampsia. Hypertension. 2011;58:63–69. doi: 10.1161/HYPERTENSIONAHA.111.172387. [DOI] [PubMed] [Google Scholar]
  • 22.Woodham PC, Brittain JE, Baker AM, Long DL, Haeri S, Camargo CA, Jr., Boggess KA, Stuebe AM. Midgestation maternal serum 25-hydroxyvitamin d level and soluble fms-like tyrosine kinase 1/placental growth factor ratio as predictors of severe preeclampsia. Hypertension. 2011;58:1120–1125. doi: 10.1161/HYPERTENSIONAHA.111.179069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.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. 2011;124:940–950. doi: 10.1161/CIRCULATIONAHA.111.034793. [DOI] [PubMed] [Google Scholar]
  • 24.Al-Ani B, Hewett PW, Cudmore MJ, Fujisawa T, Saifeddine M, Williams H, Ramma W, Sissaoui S, Jayaraman PS, Ohba M, Ahmad S, Hollenberg MD, Ahmed A. Activation of proteinase-activated receptor 2 stimulates soluble vascular endothelial growth factor receptor 1 release via epidermal growth factor receptor transactivation in endothelial cells. Hypertension. 2010;55:689–697. doi: 10.1161/HYPERTENSIONAHA.109.136333. [DOI] [PubMed] [Google Scholar]
  • 25.Gould PS, Gu M, Liao J, Ahmad S, Cudmore MJ, Ahmed A, Vatish M. Upregulation of urotensin ii receptor in preeclampsia causes in vitro placental release of soluble vascular endothelial growth factor receptor 1 in hypoxia. Hypertension. 2010;56:172–178. doi: 10.1161/HYPERTENSIONAHA.110.152074. [DOI] [PubMed] [Google Scholar]
  • 26.Rajakumar A, Cerdeira AS, Rana S, Zsengeller Z, Edmunds L, Jeyabalan A, Hubel CA, Stillman IE, Parikh SM, Karumanchi SA. Transcriptionally active syncytial aggregates in the maternal circulation may contribute to circulating soluble fms-like tyrosine kinase 1 in preeclampsia. Hypertension. 2012;59:256–264. doi: 10.1161/HYPERTENSIONAHA.111.182170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.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;50:1142–1147. doi: 10.1161/HYPERTENSIONAHA.107.096594. [DOI] [PubMed] [Google Scholar]
  • 28.Gilbert JS, Gilbert SA, Arany M, Granger JP. Hypertension produced by placental ischemia in pregnant rats is associated with increased soluble endoglin expression. Hypertension. 2009;53:399–403. doi: 10.1161/HYPERTENSIONAHA.108.123513. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.George EM, Cockrell K, Aranay M, Csongradi E, Stec DE, Granger JP. Induction of heme oxygenase 1 attenuates placental ischemia-induced hypertension. Hypertension. 2011;57:941–948. doi: 10.1161/HYPERTENSIONAHA.111.169755. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Gilbert JS, Verzwyvelt J, Colson D, Arany M, Karumanchi SA, Granger JP. Recombinant vascular endothelial growth factor 121 infusion lowers blood pressure and improves renal function in rats with placentalischemia-induced hypertension. Hypertension. 2010;55:380–385. doi: 10.1161/HYPERTENSIONAHA.109.141937. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Woods AK, Hoffmann DS, Weydert CJ, Butler SD, Zhou Y, Sharma RV, Davisson RL. Adenoviral delivery of vegf121 early in pregnancy prevents spontaneous development of preeclampsia in bph/5 mice. Hypertension. 2011;57:94–102. doi: 10.1161/HYPERTENSIONAHA.110.160242. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Murphy SR, LaMarca BB, Cockrell K, Granger JP. 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]
  • 33.Li F, Hagaman JR, Kim HS, Maeda N, Jennette JC, Faber JE, Karumanchi SA, Smithies O, Takahashi N. Enos deficiency acts through endothelin to aggravate sflt-1-induced pre-eclampsia-like phenotype. J Am Soc Nephrol. 2012;23:652–660. doi: 10.1681/ASN.2011040369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.McGuane JT, Danielson LA, Debrah JE, Rubin JP, Novak J, Conrad KP. Angiogenic growth factors are new and essential players in the sustained relaxin vasodilatory pathway in rodents and humans. Hypertension. 2011;57:1151–1160. doi: 10.1161/HYPERTENSIONAHA.110.165027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Saxena AR, Karumanchi SA, Brown NJ, Royle CM, McElrath TF, Seely EW. Increased sensitivity to angiotensin ii is present postpartum in women with a history of hypertensive pregnancy. Hypertension. 2010;55:1239–1245. doi: 10.1161/HYPERTENSIONAHA.109.147595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Redman CW, Tannetta DS, Dragovic RA, Gardiner C, Southcombe JH, Collett GP, Sargent IL. 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]
  • 37.Buurma A, Cohen D, Veraar K, Schonkeren D, Claas FH, Bruijn JA, Bloemenkamp KW, Baelde HJ. Preeclampsia is characterized by placental complement dysregulation. Hypertension. 2012;60:1332–1337. doi: 10.1161/HYPERTENSIONAHA.112.194324. [DOI] [PubMed] [Google Scholar]
  • 38.Wallace K, Richards S, Dhillon P, Weimer A, Edholm ES, Bengten E, Wilson M, Martin JN, Jr., LaMarca B. Cd4+ t-helper cells stimulated in response to placental ischemia mediate hypertension during pregnancy. Hypertension. 2011;57:949–955. doi: 10.1161/HYPERTENSIONAHA.110.168344. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.LaMarca B, Wallace K, Herse F, Wallukat G, Martin JN, Jr., Weimer A, Dechend R. Hypertension in response to placental ischemia during pregnancy: Role of b lymphocytes. Hypertension. 2011;57:865–871. doi: 10.1161/HYPERTENSIONAHA.110.167569. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Wenzel K, Rajakumar A, Haase H, Geusens N, Hubner N, Schulz H, Brewer J, Roberts L, Hubel CA, Herse F, Hering L, Qadri F, Lindschau C, Wallukat G, Pijnenborg R, Heidecke H, Riemekasten G, Luft FC, Muller DN, Lamarca B, Dechend R. Angiotensin ii type 1 receptor antibodies and increased angiotensin ii sensitivity in pregnant rats. Hypertension. 2011;58:77–84. doi: 10.1161/HYPERTENSIONAHA.111.171348. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Chatterjee P, Chiasson VL, Kopriva SE, Young KJ, Chatterjee V, Jones KA, Mitchell BM. Interleukin 10 deficiency exacerbates toll-like receptor 3-induced preeclampsia-like symptoms in mice. Hypertension. 2011;58:489–496. doi: 10.1161/HYPERTENSIONAHA.111.172114. [DOI] [PubMed] [Google Scholar]
  • 42.Acevedo CH, Ahmed A. Hemeoxygenase-1 inhibits human myometrial contractility via carbon monoxide and is upregulated by progesterone during pregnancy. J Clin Invest. 1998;101:949–955. doi: 10.1172/JCI927. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Ahmed A, Rahman M, Zhang X, Acevedo CH, Nijjar S, Rushton I, Bussolati B, St John J. Induction of placental heme oxygenase-1 is protective against tnfalpha-induced cytotoxicity and promotes vessel relaxation. Mol Med. 2000;6:391–409. [PMC free article] [PubMed] [Google Scholar]
  • 44.Ahmed A, Cudmore MJ. Can the biology of vegf and haem oxygenases help solve pre-eclampsia? Biochem Soc Trans. 2009;37:1237–1242. doi: 10.1042/BST0371237. [DOI] [PubMed] [Google Scholar]
  • 45.Cao J, Inoue K, Li X, Drummond G, Abraham NG. Physiological significance of heme oxygenase in hypertension. Int J Biochem Cell Biol. 2009;41:1025–1033. doi: 10.1016/j.biocel.2008.10.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Botros FT, Schwartzman ML, Stier CT, Jr., Goodman AI, Abraham NG. Increase in heme oxygenase-1 levels ameliorates renovascular hypertension. Kidney Int. 2005;68:2745–2755. doi: 10.1111/j.1523-1755.2005.00745.x. [DOI] [PubMed] [Google Scholar]
  • 47.Sabaawy HE, Zhang F, Nguyen X, ElHosseiny A, Nasjletti A, Schwartzman M, Dennery P, Kappas A, Abraham NG. Human heme oxygenase-1 gene transfer lowers blood pressure and promotes growth in spontaneously hypertensive rats. Hypertension. 2001;38:210–215. doi: 10.1161/01.hyp.38.2.210. [DOI] [PubMed] [Google Scholar]
  • 48.Yang L, Quan S, Nasjletti A, Laniado-Schwartzman M, Abraham NG. Heme oxygenase-1 gene expression modulates angiotensin ii-induced increase in blood pressure. Hypertension. 2004;43:1221–1226. doi: 10.1161/01.HYP.0000126287.62060.e6. [DOI] [PubMed] [Google Scholar]
  • 49.Vera T, Kelsen S, Stec DE. Kidney-specific induction of heme oxygenase-1 prevents angiotensin ii hypertension. Hypertension. 2008;52:660–665. doi: 10.1161/HYPERTENSIONAHA.108.114884. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Cudmore M, Ahmad S, Al-Ani B, Fujisawa T, Coxall H, Chudasama K, Devey LR, Wigmore SJ, Abbas A, Hewett PW, Ahmed A. Negative regulation of soluble flt-1 and soluble endoglin release by heme oxygenase-1. Circulation. 2007;115:1789–1797. doi: 10.1161/CIRCULATIONAHA.106.660134. [DOI] [PubMed] [Google Scholar]
  • 51.Wikstrom AK, Stephansson O, Cnattingius S. Tobacco use during pregnancy and preeclampsia risk: Effects of cigarette smoking and snuff. Hypertension. 2010;55:1254–1259. doi: 10.1161/HYPERTENSIONAHA.109.147082. [DOI] [PubMed] [Google Scholar]
  • 52.George EM, Arany M, Cockrell K, Storm MV, Stec DE, Granger JP. Induction of heme oxygenase-1 attenuates sflt-1-induced hypertension in pregnant rats. Am J Physiol Regul Integr Comp Physiol. 2011;301:R1495–1500. doi: 10.1152/ajpregu.00325.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.McCarthy FP, Drewlo S, Kingdom J, Johns EJ, Walsh SK, Kenny LC. Peroxisome proliferator-activated receptor-gamma as a potential therapeutic target in the treatment of preeclampsia. Hypertension. 2011;58:280–286. doi: 10.1161/HYPERTENSIONAHA.111.172627. [DOI] [PubMed] [Google Scholar]
  • 54.McCarthy FP, Drewlo S, English FA, Kingdom J, Johns EJ, Kenny LC, Walsh SK. Evidence implicating peroxisome proliferator-activated receptor-gamma in the pathogenesis of preeclampsia. Hypertension. 2011;58:882–887. doi: 10.1161/HYPERTENSIONAHA.111.179440. [DOI] [PubMed] [Google Scholar]
  • 55.Ahmed A. New insights into the etiology of preeclampsia: Identification of key elusive factors for the vascular complications. Thromb Res. 2011;127(Suppl 3):S72–75. doi: 10.1016/S0049-3848(11)70020-2. [DOI] [PubMed] [Google Scholar]
  • 56.Walsh SK, English FA, Johns EJ, Kenny LC. Plasma-mediated vascular dysfunction in the reduced uterine perfusion pressure model of preeclampsia: A microvascular characterization. Hypertension. 2009;54:345–351. doi: 10.1161/HYPERTENSIONAHA.109.132191. [DOI] [PubMed] [Google Scholar]
  • 57.Gao L, Siu KL, Chalupsky K, Nguyen A, Chen P, Weintraub NL, Galis Z, Cai H. Role of uncoupled endothelial nitric oxide synthase in abdominal aortic aneurysm formation: Treatment with folic acid. Hypertension. 2012;59:158–166. doi: 10.1161/HYPERTENSIONAHA.111.181644. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Lob HE, Vinh A, Li L, Blinder Y, Offermanns S, Harrison DG. Role of vascular extracellular superoxide dismutase in hypertension. Hypertension. 2011;58:232–239. doi: 10.1161/HYPERTENSIONAHA.111.172718. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Mishra N, Nugent WH, Mahavadi S, Walsh SW. Mechanisms of enhanced vascular reactivity in preeclampsia. Hypertension. 2011;58:867–873. doi: 10.1161/HYPERTENSIONAHA.111.176602. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Lai Z, Kalkunte S, Sharma S. A critical role of interleukin-10 in modulating hypoxia-induced preeclampsia-like disease in mice. Hypertension. 2011;57:505–514. doi: 10.1161/HYPERTENSIONAHA.110.163329. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Kleinhenz JM, Kleinhenz DJ, You S, Ritzenthaler JD, Hansen JM, Archer DR, Sutliff RL, Hart CM. Disruption of endothelial peroxisome proliferator-activated receptor-gamma reduces vascular nitric oxide production. American journal of physiology. Heart and circulatory physiology. 2009;297:H1647–1654. doi: 10.1152/ajpheart.00148.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Zemse SM, Chiao CW, Hilgers RH, Webb RC. Interleukin-10 inhibits the in vivo and in vitro adverse effects of tnf-alpha on the endothelium of murine aorta. American journal of physiology. Heart and circulatory physiology. 2010;299:H1160–1167. doi: 10.1152/ajpheart.00763.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Hu XQ, Xiao D, Zhu R, Huang X, Yang S, Wilson S, Zhang L. Pregnancy upregulates large-conductance ca(2+)-activated k(+) channel activity and attenuates myogenic tone in uterine arteries. Hypertension. 2011;58:1132–1139. doi: 10.1161/HYPERTENSIONAHA.111.179952. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Downing SJ, Hollingsworth M. Antagonism of relaxin by glibenclamide in the uterus of the rat in vivo. British journal of pharmacology. 1991;104:71–76. doi: 10.1111/j.1476-5381.1991.tb12387.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Conrad KP. Unveiling the vasodilatory actions and mechanisms of relaxin. Hypertension. 2010;56:2–9. doi: 10.1161/HYPERTENSIONAHA.109.133926. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Novak J, Danielson LA, Kerchner LJ, Sherwood OD, Ramirez RJ, Moalli PA, Conrad KP. Relaxin is essential for renal vasodilation during pregnancy in conscious rats. The Journal of clinical investigation. 2001;107:1469–1475. doi: 10.1172/JCI11975. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Xiao D, Huang X, Yang S, Longo LD, Zhang L. Pregnancy downregulates actin polymerization and pressure-dependent myogenic tone in ovine uterine arteries. Hypertension. 2010;56:1009–1015. doi: 10.1161/HYPERTENSIONAHA.110.159137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Zhang W, Du L, Gunst SJ. The effects of the small gtpase rhoa on the muscarinic contraction of airway smooth muscle result from its role in regulating actin polymerization. American journal of physiology. Cell physiology. 2010;299:C298–306. doi: 10.1152/ajpcell.00118.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Zhang W, Wu Y, Du L, Tang DD, Gunst SJ. Activation of the arp2/3 complex by n-wasp is required for actin polymerization and contraction in smooth muscle. American journal of physiology. Cell physiology. 2005;288:C1145–1160. doi: 10.1152/ajpcell.00387.2004. [DOI] [PubMed] [Google Scholar]
  • 70.Verlohren S, Niehoff M, Hering L, Geusens N, Herse F, Tintu AN, Plagemann A, LeNoble F, Pijnenborg R, Muller DN, Luft FC, Dudenhausen JW, Gollasch M, Dechend R. Uterine vascular function in a transgenic preeclampsia rat model. Hypertension. 2008;51:547–553. doi: 10.1161/HYPERTENSIONAHA.107.103176. [DOI] [PubMed] [Google Scholar]
  • 71.Verlohren S, Geusens N, Morton J, Verhaegen I, Hering L, Herse F, Dudenhausen JW, Muller DN, Luft FC, Cartwright JE, Davidge ST, Pijnenborg R, Dechend R. Inhibition of trophoblast-induced spiral artery remodeling reduces placental perfusion in rat pregnancy. Hypertension. 2010;56:304–310. doi: 10.1161/HYPERTENSIONAHA.110.153163. [DOI] [PubMed] [Google Scholar]
  • 72.MacKay AP, Berg CJ, Atrash HK. Pregnancy-related mortality from preeclampsia and eclampsia. Obstet Gynecol. 2001;97:533–538. doi: 10.1016/s0029-7844(00)01223-0. [DOI] [PubMed] [Google Scholar]
  • 73.Chakravarty A, Chakrabarti SD. The neurology of eclampsia: Some observations. Neurol India. 2002;50:128–135. [PubMed] [Google Scholar]
  • 74.Tang CH, Wu CS, Lee TH, Hung ST, Yang CY, Lee CH, Chu PH. Preeclampsia-eclampsia and the risk of stroke among peripartum in taiwan. Stroke. 2009;40:1162–1168. doi: 10.1161/STROKEAHA.108.540880. [DOI] [PubMed] [Google Scholar]
  • 75.Demirtaş O, Gelal F, Vidinli BD, Demirtaş LO, Uluç E, Baloğlu A. Cranial mr imaging with clinical correlation in preeclampsia and eclampsia. Diagn Interv Radiol. 2005;11:189–194. [PubMed] [Google Scholar]
  • 76.Manfredi M, Beltramello A, Bongiovanni LG, Polo A, Pistoia L, Rizzuto N. Eclamptic encephalopathy: Imaging and pathogenetic considerations. Acta Neurol Scand. 1997;96:277–282. doi: 10.1111/j.1600-0404.1997.tb00284.x. [DOI] [PubMed] [Google Scholar]
  • 77.Amburgey OA, Chapman AC, May V, Bernstein IM, Cipolla MJ. Plasma from preeclamptic women increases blood-brain barrier permeability: Role of vascular endothelial growth factor signaling. Hypertension. 2010;56:1003–1008. doi: 10.1161/HYPERTENSIONAHA.110.158931. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Choi SJ, Lee JE, Oh SY, Kim GM, Cho YS, Lee KH, Kim JH, Roh CR. Maternal cerebral blood flow and glucose metabolism in pregnancies complicated by severe preeclampsia. Hypertens Pregnancy. 2012;31:177–188. doi: 10.3109/10641955.2010.507839. [DOI] [PubMed] [Google Scholar]
  • 79.Ryan MJ, Gilbert EL, Glover PH, George EM, Masterson CW, McLemore GR, LaMarca B, Granger JP, Drummond HA. Placental ischemia impairs middle cerebral artery myogenic responses in the pregnant rat. Hypertension. 2011;58:1126–1131. doi: 10.1161/HYPERTENSIONAHA.111.181453. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Cipolla MJ, Sweet JG, Chan SL. Cerebral vascular adaptation to pregnancy and its role in the neurological complications of eclampsia. J Appl Physiol. 2011;110:329–339. doi: 10.1152/japplphysiol.01159.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Euser AG, Cipolla MJ. Cerebral blood flow autoregulation and edema formation during pregnancy in anesthetized rats. Hypertension. 2007;49:334–340. doi: 10.1161/01.HYP.0000255791.54655.29. [DOI] [PubMed] [Google Scholar]
  • 82.Aukes AM, Vitullo L, Zeeman GG, Cipolla MJ. Pregnancy prevents hypertensive remodeling and decreases myogenic reactivity in posterior cerebral arteries from dahl salt-sensitive rats: A role in eclampsia? Am J Physiol Heart Circ Physiol. 2007;292:H1071–1076. doi: 10.1152/ajpheart.00980.2006. [DOI] [PubMed] [Google Scholar]
  • 83.Cipolla MJ, Bishop N, Vinke RS, Godfrey JA. Ppar{gamma} activation prevents hypertensive remodeling of cerebral arteries and improves vascular function in female rats. Stroke. 2010;41:1266–1270. doi: 10.1161/STROKEAHA.109.576942. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Cipolla MJ, Smith J, Bishop N, Bullinger LV, Godfrey JA. Pregnancy reverses hypertensive remodeling of cerebral arteries. Hypertension. 2008;51:1052–1057. doi: 10.1161/HYPERTENSIONAHA.107.100545. [DOI] [PubMed] [Google Scholar]
  • 85.Cipolla MJ, DeLance N, Vitullo L. Pregnancy prevents hypertensive remodeling of cerebral arteries: A potential role in the development of eclampsia. Hypertension. 2006;47:619–626. doi: 10.1161/01.HYP.0000196948.15019.28. [DOI] [PubMed] [Google Scholar]
  • 86.Cipolla MJ, Pusic AD, Grinberg YY, Chapman AC, Poynter ME, Kraig RP. Pregnant serum induces neuroinflammation and seizure activity via tnfα. Exp Neurol. 2012;234:398–404. doi: 10.1016/j.expneurol.2012.01.005. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES