You are on page 1of 8

Pathology - Research and Practice 214 (2018) 7–14

Contents lists available at ScienceDirect

Pathology - Research and Practice


journal homepage: www.elsevier.com/locate/prp

Review

Angiogenic and antiangiogenic factors in preeclampsia T


a b c
Fernanda Rodrigues Helmo , Angela Maria Moed Lopes , Anna Cecília Dias Maciel Carneiro ,
Carolina Guissoni Camposb, Polyana Barbosa Silvab, Maria Luíza Gonçalves dos Reis Monteiroa,
Laura Penna Rochaa, Marlene Antônia dos Reisa, Renata Margarida Etchebehered,

Juliana Reis Machadoa,e, Rosana Rosa Miranda Corrêaa,
a
Discipline of General Pathology, Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
b
Oncology Research Institute, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
c
Discipline of Histology, Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro. Uberaba, Minas Gerais, Brazil
d
Surgical Pathology Service, Clinical Hospital, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
e
Department of General Pathology, Institute of Tropical Pathology and Public Health, Federal University of Goiás, Goiânia, Goiás, Brazil

A R T I C L E I N F O A B S T R A C T

Keywords: Background: Pre-eclampsia is a multifactorial hypertensive disorder that is triggered by placental insufficiency
Preeclampsia and that accounts for up to 15% of maternal deaths. In normal pregnancies, this process depends on the balance
Endothelial dysfunction between the expression of angiogenic factors and antiangiogenic factors, which are responsible for remodeling
Soluble fms-like-tyrosine-kinase receptor the spiral arteries, as well as for neoangiogenesis and fetal development.
Soluble endoglin
Purpose: The aim of this review is to discuss the main scientific findings regarding the role of angiogenic and
Angiogenisis
antiangiogenic factors in the etiopathogenesis of preeclampsia.
Methods: An extensive research was conducted in the Pubmed database in search of scientific manuscripts
discussing potential associations between angiogenic and antiangiogenic factors and preeclampsia. Ninety-one
papers were included in this review.
Results: There is an increased expression of soluble fms-like tyrosine kinase receptor and soluble endoglin in pre-
eclampsia, as well as reduced placental expression of vascular endothelial growth factor and placental growth
factor. Systemic hypertension, proteinuria and kidney injury – such as enlargement and glomerular fibrin de-
posit, capillary occlusion due to edema, and hypertrophy of endocapillary cells – are some of these changes. The
complex etiopathogenesis of preeclampsia instigates research of different biomarkers that allow for the early
diagnosis of this entity, such as vascular endothelial growth factor, placental growth factor, soluble fms-like
tyrosine kinase receptor, soluble endoglin, placental glycoprotein pregnancy-associated plasma protein-A and
protein 13.
Conclusion: Even though it is possible to establish an efficient and effective diagnostic tool, three key principles
must be observed in the management of preeclampsia: prevention, early screening and treatment.

1. Introduction and/or diastolic blood pressure ≥ 90 mm Hg in hypertensive women,


followed by proteinuria ≥ 300 mg/L in urine/24 h [1–3]. Moreover,
Preeclampsia (PE) is a complex hypertensive pregnancy syndrome the presence of thrombocytopenia (platelets < 100,000/mμl), renal
that affects between 2 and 7% of pregnant women after the 20th week, failure (creatinine ≥ 1.1 mg/dl) or impairment of liver function (dou-
and which may appear superimposed on chronic hypertension (CH), for bled normal concentration of oxaloacetic transaminase and pyruvate
instance. It is characterized by systolic blood pressure ≥ 140 mm Hg transaminase), pulmonary edema, as well as cerebral and visual

Abbreviations: AT1-AA, angiotensin II type 1 receptor agonistic autoantibodies; ATF, activating transcription factor; CH, chronic hypertension; dNK, decidual NK cell; ECM, extracellular
matrix; eIF2a, eukaryotic initiation factor 2 subunit a; ER, endoplasmatic reticulum Flk-1 kinase insert domain receptor; Flt-1, fms-like tyrosine kinase receptor; GRP78, glucose-regulated
protein 78; ICAM, intercellular adhesion molecule; IL, interleukin; NOTCH, notch transmembrane receptors; PAPP-A, placental glycoprotein pregnancy-associated plasma protein-A; PE,
preeclampsia; PERK, PKR-like endoplasmic reticulum kinase; PlGF, placental growth factor; PP13, protein 13; sEng, soluble endoglina; sFlt-1, soluble form of Flt-1; sTNF-R, soluble tumor
necrosis factor receptors; TGF-β, transforming growth factor-β; TNF-α, tumor necrosis factor-α; VEGF, vascular endothelial growth factor

Corresponding author at: Instituto de Ciências Biológicas e Naturais/Disciplina de Patologia Geral/Universidade Federal do Triângulo Mineiro, Rua Frei Paulino n° 30, Bairro Nossa
Senhora da Abadia, CEP: 38025-180, Uberaba, Minas Gerais, Brazil.
E-mail address: rosana.correa@uftm.edu.br (R.R.M. Corrêa).

https://doi.org/10.1016/j.prp.2017.10.021
Received 13 March 2017; Received in revised form 23 October 2017; Accepted 25 October 2017
0344-0338/ © 2017 Elsevier GmbH. All rights reserved.
F.R. Helmo et al. Pathology - Research and Practice 214 (2018) 7–14

disturbances, accompanied by hypertension [4] in the absence of pro- circulation that are responsible for causing clinical changes in PE
teinuria, are other criteria for the diagnosis of PE [3,5]. [2,28].
Eclampsia, from the Greek eklampsis (lightning), refers to the sudden In normal pregnancies, cytotrophoblasts (cells outside the blastocyst
onset of persistent headache, photophobia, scotomas, epigastric pain or layer) invade the uterine wall between the 6th and 8th week, destroy
pain in the right upper quadrant, as well as seizures [3,6]. It is note- the muscular middle layer, acquire endothelial cell phenotype, and
worthy that the impairment of hepatic and renal function may lead to transform spiral arteries into large vessels of low vascular resistance in
hemolysis, which is characterized by increased plasma level of lactate order to increase blood flow to the intervillous space and thence to the
dehydrogenase, persistent increase in the level of liver enzymes and in fetus during embryonic development [2,14,28,29]. However, there are
thrombocytopenia, which is called HELLP (hemolysis, elevated liver failures in this process in PE, and the absence of arterial remodeling
enzymes and low platelets) [7]; and which may lead to intense in- promotes lipid deposition in the vessel wall, as well as blood flow
flammatory response, endothelial injury, generalized vascular re- turbulence, platelet aggregation, fibrinoid necrosis, and intense acti-
sistance [8], disseminated intravascular coagulation, hepatic infarction, vation of the inflammatory response [2,14,17].
cerebral vascular disease, premature placental abruption and oliguria Even though the mechanisms of this disorder are not fully known
[2,3], for instance. [2,14], it is believed that poor cytotrophoblast invasion concomitant
Different risk factors for PE have been discussed in the literature, with uteroplacental hypoperfusion may contribute to PE, since multiple
and predisposing factors may include the following: African descent, areas of infarction, sclerotic arterioles, increasing vascular resistance,
obesity, short stature, nutritional deficiencies, previous CH (prevalent and reduced placental perfusion are observed in the placenta
in up to 35% of the cases) or gestational hypertension [9] in previous [28,30,31], thus oxidative stress and IUGR are favored [30,32]. Fur-
pregnancies, heredity, urinary tract infections, diabetes mellitus, auto- thermore, there is a reduction in the area and volume of the intervillous
immune diseases, hydatidiform mole, multiple pregnancy, and fetal space, in the intermediate and terminal villi, both in cases with PE
macrosomia [10–13]. According to Chaiworapongsa et al. [15], other alone and in cases with PE accompanied by IUGR. Remodeling of blood
factors include primiparity in young women, which is associated with vessels in both villi is also significantly reduced [32].
intolerance of the maternal immune system to paternal alloantigens Therefore, placental hypoxia is considered as a triggering factor for
present in the seminal fluid and in the sperm; heredity, represented by the increase in the synthesis of vasoconstrictors such as endothelin and
the presence of genetic variations in collagen α1-chain (I), interleukin- superoxide, as well as the increase in von Willebrand factor antigen,
1α (IL-1α), urokinase-type plasminogen activator receptor; maternal- cellular fibronectin, soluble tissue factor, soluble E-selectin, platelet-
fetal incompatibility of lymphotoxin-α, von Willebrand factor, and α2 derived growth factor, susceptibility to the effects of angiotensin II and
chain of collagen [14]; mutations in factor V Leiden, in human leuko- norepinephrine; placental hypoxia is also a triggering factor for the
cyte antigen, in endothelial nitric oxide synthases, and in angiotensin reduction in nitric oxide synthesis by the maternal vascular en-
converting enzyme [15]. dothelium. This endothelial dysfunction has systemic effects, since
Although little is known about the etiopathogenesis of pre- these factors affect the blood vessels in the liver, brain and kidneys, for
eclampsia, this syndrome has a major impact on maternal and fetal example; in the latter, there is a reduction in the glomerular filtration
health worldwide, since it is responsible for 10–15% of maternal deaths rate as well as impairment of blood pressure regulation [17,28,29].
[16,17], preterm births, intrauterine growth restriction (IUGR), and Abnormalities in placentation in patients with PE appear to be as-
fetal deaths [2]. In developing countries, there are high rates of ma- sociated with changes in different components of the signaling pathway
ternal morbidity and mortality from PE, mainly due to poor prenatal that mediates cytotrophoblast migration/invasion. Studies have showed
care [16]. In Brazil, according to data by the National Health System that the activation of the Notch signaling pathway is associated with the
(SUS, in Portuguese) [18], approximately 76,000 maternal deaths and differentiation and modulation of functions of trophoblast cells through
500,000 perinatal deaths are estimated to be associated with PE every the interaction of the Notch transmembrane receptors (NOTCH1-4) and
year, with an average of three maternal deaths/day due to complica- their respective ligands, DLL1,3,4 and JAG1,2 [29,33]. An experimental
tions arising from the disease [19]. study showed a correlation between the non-expression of the NOTCH-
Therefore, the identification of risk factors and the establishment of 2 receptor and the reduction in the diameter of maternal uterine vessels
criteria for the diagnosis of PE are essential for the effective treatment during placentation, with significant impairment of perfusion [33].
and prevention of complications during pregnancy. Platelet count, Mutations in the gene encoding STOX1 transcription factor may also be
measurement of hepatic enzymes, evaluation of proteinuria and blood associated with the pathogenesis of PE, since the overexpression of this
pressure levels, weight gain monitoring, analysis of kidney and lung factor in choriocarcinoma cells mimics the same transcriptional profile
function, and assessment of previous pregnancy history (e.g. premature observed in PE [29,34].
birth, children born small for gestational age, etc.) must be recorded Endoplasmatic reticulum (ER) stress, characterized by the accu-
during prenatal care. However, it is interesting to note that this entity mulation of unfolded proteins and misfolded proteins within the orga-
may have atypical manifestations as those observed in the HELLP nelle, interfering with functions of synthesis, post-translational folding
syndrome, yet in the absence of proteinuria and hypertension; thereby, and assembly of all secreted and membrane-bound proteins, including
careful research and analysis of signs and symptoms of pregnant women hormones, growth factors, and receptors [35–37]. Loss of homeostasis
are required [2,3]. activates unfolded proteins response, consisting of three main signaling
According to the literature, several mechanisms are associated with routes: PKR-like endoplasmic reticulum kinase (PERK), activating
the development of this multifactorial syndrome, including changes transcription factor 6 (ATF6) and inositol-requiring 1 (Ire1). Under
during placentation [20], oxidative stress [21,22], exacerbated in- normal conditions, these transmembrane proteins become inactive by
flammatory response [23–25], thrombosis, activation of the renin-an- binding glucose-regulated protein 78 (GRP78 or BiP) to its N terminal.
giotensin-aldosterone system [25,26], and endothelial dysfunction However, consumption of GRP78 (Ca2+–dependent chaperone protein)
caused by changes in the angiogenic profile [27]; the latter is associated triggered by accumulation of misfolded proteins promotes dimeriza-
with the pathophysiological changes described in PE. tion, autophosphorylation and activation of PERK and Ire1. Conse-
quently, activation of PERK favors the phosphorylation of eukaryotic
2. Etiopathogenesis of preeclampsia initiation factor 2 subunit a (eIF2a) and translation of activating tran-
scription factor-4 (ATF4), which results in blocking protein translation
The pathophysiology of PE remains largely unknown. Nonetheless, and reducing accumulation in ER. In Golgi complex, ATF6 is cleaved by
this hypertensive disorder is thought to be triggered by placental dys- the transcription factor responsible for the expression of ER chaperone
function, which favors the synthesis of specific factors in the maternal genes. Splices Xbp1 mRNA (originated from activation of Ire

8
F.R. Helmo et al. Pathology - Research and Practice 214 (2018) 7–14

endoribonuclease domain) is responsible for the transcription of genes remodeling, which is called pseudo-vasculogenesis [49–51].
that regulate degradation of misfolded proteins and ER biogenesis. In In normal pregnancies, the expression of different molecules, such
intense ER stress, PERK and ATF6 favor CHOP and caspase-4 (proa- as integrins, cadherins and metalloproteinases, allows the cytotropho-
poptotic proteins) synthesis and apoptosis of the affected cell [35,38]. blast to switch from an epithelial to an endothelial cell phenotype,
Dilatation of ER cistaernae in syncytotrophoblast, greater phos- thereby stimulating the formation of pseudoendothelium in the spiral
phorylation of eIF2a, reduction of protein synthesis, greater apoptosis arteries between the 12th and 20th weeks of embryonic development
and impairment of placentation are some changes triggered by ER stress [49]. Several factors are believed to interfere with this process, such as
in PE [35]. A research indicates that the expression of GRP78, PERK, oxygen tension, and the activation of decidual NK cells and macro-
eIF2a, ATF4, CHOP, and caspase-12 mRNA, as well as their respective phages, for instance. Nonetheless, shifting the balance in the expression
proteins, were significantly higher in human placentas with early and of angiogenic and antiangiogenic factors seems to be the key factor in
late-onset PE, compared with normal placentas [38]. the endothelial dysfunction observed in PE [22,49,50].
In addition to the metabolic pathway disorders in this disease, re- According to the literature, VEGF, PlGF, endoglin, basic fibroblast
search has reported that there may be maternal immune tolerance to growth factor, angiopoietins, and transforming growth factor-β (TGF-β)
fetal antigens during intrauterine development. It is assumed that the are some angiogenic factors associated with the development of the
recognition of fetal antigens by CD4+ T cells and by uterine natural extensive vascular network [17,49]; and the first three factors are
killer [33] cells is fundamental to placental growth. Failures in this particularly relevant to the etiopathogenesis of PE.
process cause miscarriages, impaired placentation, changes in placental VEGF is a cytokine synthesized by macrophages, T cells, tumor cells,
perfusion and in the activation of the immune system throughout and cytotrophoblasts [51] and, thus, it is involved in various physio-
pregnancy [29]. logical and pathological conditions [5]. VEGF isoforms include VEGF-A,
Particular differences are identified in decidual NK cell (dNK) VEGF-B, VEGF-C, VEGF-D and PlGF. Moreover, cells can express three
phenotype in PE. Effective trophoblast invasion is regulated by acti- main types of cellular receptors known as Flt-1 (fms-like tyrosine kinase
vation and inhibition of receptors (KIR, CD94/NKG2, ILT families) in receptor or VEGFR1), Flk-1 (kinase insert domain receptor or VEGFR2)
dNK cells, which favor interaction with trophoblast HLA Class I mole- and Flt-4 (VEGFR3), and they all have an extracellular domain, a
cules (HLA-C/G/E). Evidence shows that the frequency of genotype KIR transmembrane domain and an intracellular tyrosine kinase domain; as
AA associated with the allele HLA-C with C2 epitope, is higher in well as the soluble form of Flt-1 (sFlt-1), in which transmembrane and
pregnancies with impairment of spiral artery remodeling [39,40]. In cytoplasmic domains are absent. In syncytiotrophoblast, sFlt-1 is syn-
addition, dNK cells from women with high resistance index (uterine thesized through a splicing of mRNA of VEGRF-1 (or Flt-1) gene, which
artery Doppler ultrasound) have lower expression of KIR2DL/S1,3,5 encodes proteins without the ability to bind to the VEGF or PlGF within
and LILRB1 (ILT-2); in turn, the reduction of LILRB1 stimulates the cells, but capable of interacting with free growth factors in maternal
greater expression of tumor necrosis factor-α (TNF-α) and the lower circulation [13,52–54]. VEGF-A, -B and PlGF bind to Flt-1; VEGF-A and
expression of CXCL10. There is also greater secretion of endostatin, -C bind to Flk-1; whereas VEGF-C and -D bind to Flt-4. Furthermore,
angiogenin and the soluble IL-2 receptor. These changes interfere in the VEGF-A, VEGF-B and PlGF may also bind to sFlt-1 [5,49,54–56].
ability of dNK cells to regulate trophoblast invasion and migration Therefore, VEGF and its receptors are expressed in different organs,
[40,41]. including the kidneys, the liver and the brain [55]. In the human pla-
An in vitro assay in which endothelial cells form 3D tube-like centa, VEGF is mainly synthesized by cytotrophoblast and Hofbauer
structures was used to investigate effects of dNK on vessel stability. The cells early in the first trimester, whereas Flk-1 and Flt-1 receptors are
results indicate that dNK cells from women with high spiral artery re- expressed throughout the placenta, mainly in cytotrophoblasts. Studies
sistance index had reduced ability to activate endothelial cells, since indicate that the activation of Flk-1 and Flt-1 receptors would trigger
endothelial cells presented lower expression of ICAM-1 and TNF-α, signaling pathways associated with endothelial migration and pro-
compared with endothelial cells activated by dNK from women with liferation, as well as vascular permeability, formation, tubular ramifi-
normal spiral artery resistance index. Consequently, there will also be cation and maintenance of blood vessels; whereas Flt-4 would be re-
impairment in remodeling of uterine arteries [42]. lated to the development of lymphatic vessels. Because sFlt-1 has
The higher levels of syncytiotrophoblast micro-particles in the ma- antiangiogenic properties, when it binds to VEGF and PlGF, it prevents
ternal circulation also seems to contribute to a systemic inflammatory their interactions with Flt-1 and Flk-1 receptors [5,49].
response, leading to increased production of inflammatory mediators PlGF, in turn, has been described to be expressed in four isoforms
[9,29,43,44]. It is important to note that hypoxia associated with pla- (PlGF-1, PlGF-2, PlGF-3 and PlGF-4), which play a key role in vascu-
cental insufficiency causes oxidative stress, which is when there is an logenesis throughout the embryonic development. In the placenta, this
increased synthesis of reactive oxygen species (e.g. peroxynitrite, hy- factor is first detected in the syncytiotrophoblast, and serum levels in-
drogen peroxide, hydroxyl radical) and reduction of antioxidant crease from the 30th week with subsequent decrease in normotensive
synthesis (e.g. superoxide dismutase, glutathione, vitamin C) [21,30]. pregnancies [13].
These reactive oxygen species lead to lipid peroxidation and, hence, Endoglin, also known as CD105, is a membrane-bound protein that
the synthesis of toxic byproducts (lipid peroxide, malondialdehyde, and acts as a coreceptor for TGF-β1 and TGF-β3. It consists of an extra-
lipid hydroperoxide), and severe damage and dysfunction of the vas- cellular domain, a single transmembrane domain, and a short cyto-
cular endothelium, which is associated with increased synthesis of in- plasmic domain and, similarly to Flt-1, it also has a soluble isoform
flammatory mediators and with change in the serum levels of angio- (sEng) that has antiangiogenic properties [5,57,58]. Endoglin has a
genic and antiangiogenic factors [21,45–47], such as soluble receptor little expression in quiescent endothelial cells, but a significant ex-
tyrosine kinase, soluble endoglin, vascular endothelial growth factor pression in stromal cells, hematopoietic stem cells, proliferative en-
(VEGF), and placental growth factor (PlGF) [1,13,21,47,48]. dothelial cells, and in the syncytiotrophoblast [5]; it inhibits apoptosis
and activates the synthesis of endothelial nitric oxide [13]. Therefore, it
3. Angiogenic and antiangiogenics factors is capable of inducing cell migration and proliferation [5,59].
It is believed that in breast and colo-rectal neoplasias sEng is formed
The placenta is an organ that is highly vascularized by maternal and through the action of metalloproteinase-14 in the transmembrane do-
fetal blood vessels, and its development, maturation and functions de- main of endoglin; however maintains its extracellular domain and thus
pend on efficient vasculogenesis (de novo formation of new blood ves- the ability to bind to TGF-β and BMP-9. Metalloproteinase-14 is also
sels), on angiogenesis (the formation of new blood vessels from other expressed in syncytiotrophoblast and in PE acts on endoglin of placental
pre-existing vessels), and on cytotrophoblast invasion for spiral artery cells. Research has shown that the greater expression of inhibitors of

9
F.R. Helmo et al. Pathology - Research and Practice 214 (2018) 7–14

this metalloproteinase attenuates the synthesis of sEng by placenta for sFlt-1 levels, a significant average increase was observed in severe
[5,59,60]. EP (19,070.6 pg/ml) or eclampsia (28,527.9 pg/ml) when compared to
In circulation, both sFlt-1 and sEng interact with their respective normal pregnancies (6483.3 pg/ml). A result similar to that of sFlt-1
binders (VEGF, PlGF and TNF-β), impairing the binding of these pro- was found regarding sEng in severe PE (37.2 pg/ml) or eclampsia
angiogenic molecules to their native endothelial cell-surface receptors. (46.7 pg/ml) in comparison with pregnancies without changes
Thus, elevated serum levels of sFlt-1 and sEng, and reduced serum le- (12.7 pg/ml) [57,79]. It should be noted that in early-onset PE (< 34
vels of PlGF are observed in the PE [53,54,59,61,62]. According to a weeks), the levels of PlGF appear to be significantly lower, with a dif-
research, plasma concentrations of sFlt-1(41.5 ± 15.8 vs ference of 500% in relation to late-onset PE (> 34 weeks); as for sFlt-1,
6.11 ± 3.3 ng/ml) and sEng (86.8 ± 38.3 vs 13.4 ± 6.1 ng/ml) in it has a higher level of early-onset PE, and it may reach a concentration
women with PE were significantly higher in relation to those normo- difference of 600% in comparison with late-onset PE [52,80].
tensive; in contrast, PlGF (96.1 ± 46.4 vs 508.2 ± 332.3 pg/ml) In premature births, whose diagnosis of PE was confirmed, there
presented a significant reduction in relation to normotensive pregnant was a reduction in PlGF levels in the first and second trimesters of
women [62]. pregnancy; whereas sFlt-1 and sEng levels, although lower in the first
Thus, it is clear that placentation is dependent on the balance be- quarter, were substantially higher in the following quarter. Thus, it was
tween the expression of angiogenic and antiangiogenic factors possible to infer that pregnant women whose variation of PlGF was
throughout pregnancy. The expression of VEGF-A, PlGF, endoglin, and minimal or absent between the first and second trimester, and whose
their receptors, Flt-1, Flk-1, sFlt-1 and sEng, appears to be essential for sFlt-1 and sEng levels increased continuously in the second trimester,
the establishment of uteroplacental circulation and for fetal develop- were at high risk for PE [52,53,57,61,73–75,81].
ment [63,64]. Studies have showed that VEGF-A, VEGF-B and PlGF are Studies have already showed that the synthesis of anti-angiogenic
responsible for modulating cytotrophoblast proliferation, differentia- factors can be triggered by several stimuli. For example, the expression
tion and invasion via Flt-1 and Flk-1 receptors; since when promoting of endothelin-1, TNF-α and angiotensin II type 1 receptor agonistic
placental angiogenesis, they also promote the expression of coreceptor autoantibodies (AT1-AA) is increased in PE and it promotes the
neuropilin-1, contributing to the enhancement of VEGF-A activity synthesis of sFlt-1 and sEng, as well as proteinuria and systemic hy-
through interaction with Flk-1. As for endoglin, not only does it con- pertension [82,83]. Hypoxia, in turn, promotes the expression of hy-
tribute to the formation of new blood vessels, but also to the regulation poxia-inducible factor 1α (HIF-1α); and HIF-1α promotes the expres-
of muscle tone [51]. sion of TGF-β3, which acts as an antagonist of cytotrophoblast
There is a higher expression of sFlt-1 in the placenta of PE patients, proliferation [72,82,84,85].
so increased maternal serum concentration of this substance is notice- Other factors have a significant influence on endothelial dysfunc-
able. On the other hand, a lower expression of VEGF-A and PlGF is tion, concomitantly to antiangiogenic factors. mRNA expression of the
observed, since sFlt-1 binds to both factors in the circulation and pre- angiotensin II type 1 receptor and of angiotensin II is increased in
vents interaction with endogenous receptors [53,54,65–68]. In addition chorionic villi, and it is responsible for the reduction of uteroplacental
to these changes, the serum concentration and the mRNA expression of perfusion and vasoconstriction stimulation. Hence, the persistence of
sEng are increased; and because sEng competes with the endogenous hypoxia, syncytiotrophoblast micro-particles in the circulation, and the
receptor when binding to TGF-β, both the vasculogenesis and placental increased expression of TNF-α [82] promote an exacerbated synthesis
angiogenesis of pregnant women with PE are impaired [51,69–72]. Due of IL-6, IL-16 and TNF-α, which also contributes to worsening the en-
to intense endothelial dysfunction, both antiangiogenic factors (Flt-1 dothelial dysfunction present in this disease [25,82]. Endostatin (frag-
and sEng) can be detected in the maternal circulation early in the first ment of collagen XVII), in turn, affects the interaction of VEGF with
trimester of pregnancy, prior to the first clinical signs of disease Filk-1 and, thus, interferes with the intracellular signaling pathways
[51–53,61,73–75]. responsible for cell motility [50,51]. Finally, the increased levels of
Maynard et al. [54] demonstrated that serum levels of sFlt-1 were matrix metalloproteinase-9 in the placenta seems to be associated with
five times higher in pregnant women with PE than in normotensive impaired extracellular matrix (ECM) degradation during angiogenesis,
women; as a consequence, VEGF and PlGF levels were found to be as well as with cytotrophoblast invasion in PE [51].
proportionally reduced in these women [54]. The literature confirms Finally, impaired endothelial integrity culminates in an increased
that there is a sharp increase in sFlt-1 levels between 11 and 5 weeks plasma protein concentration in the interstitial space, and thus accu-
before the first signs of PE [52,53], as well as a substantial reduction in mulation of fluid in the ECM, favoring the swelling of the face and limbs
PlGF and VEGF levels [53], whereas sEng levels increased significantly [2] mainly. Concurrently with this systemic change, there is intense
between 11 and 9 weeks before clinical signs [52,61]. activation of coagulation, reduced tissue perfusion, and increased
Experimental tests have showed that the increased synthesis of sFlt- synthesis of vasoconstrictor agents [69,86].
1 inhibits angiogenic activity due to the blockade of endogenous
functions of VEGF and PlGF, as well as physiological vasodilation. 4. Biomarkers and management of preeclampsia
Furthermore, pregnant and non-pregnant rats were given a dose of
recombinant adenovirus (responsible for the gene encoding of sFlt-1 The complex etiopathogenesis of PE instigates worldwide research
mRNA) developed systemic hypertension, proteinuria and kidney in- on the definition of clinical and laboratory parameters that allow for
jury, such as glomerular enlargement and glomerular fibrin deposits, the early diagnosis of this entity. Because systemic hypertension and
capillary occlusion due to edema, hypertrophy of endocapillary cells, proteinuria are not specific symptoms [5], different biomarkers have
podocytes with protein absorption droplets, and effacement of podocyte been investigated as promising diagnostic tools [69,87,88].
foot processes, hence characterizing glomerular endotheliosis [54]. In the first and second trimesters of pregnancy, the relationship
These findings are in agreement with other studies since in PE alone between sFlt-1/PlGF is regarded as a promising index of the imbalance
or in the concomitant presence of HELLP syndrome, the levels of Flt-1 between angiogenic and antiangiogenic factors [53,75,89,90]. Serum
mRNA increased 27.7 times, and Eng mRNA increased 6.9 times [76]; levels of both factors are already altered in early pregnancy; for in-
and there was a reduction of 85.4% in the expression of VEGF mRNA stance, sFlt-1 can be detected from the fifth week. When it is associated
[77]. Moreover, Strevens et al. [78] found similar changes in renal with changes in plasma concentration of sEng and with Doppler ultra-
biopsies from women with PE [78]. sound of the uterine artery between 20th and 24th week, the predictive
Laboratory tests have confirmed that PlGF serum levels are lower in value of these factors reaches up to 89.5% sensitivity and 95% speci-
pregnancies with severe PE (104 pg/ml) or eclampsia (84.3 pg/ml) in ficity [68,90]; whereas in the presence of abnormal uterine perfusion
comparison with pregnancies without abnormalities (335.6 pg/ml); as there may be 100% sensitivity with 93.3% specificity [66]. It is also

10
F.R. Helmo et al. Pathology - Research and Practice 214 (2018) 7–14

suggested that nulliparous pregnant women who have serum levels of (3.94–5.10) pg/ml] and sTNF-R2 (8698 ± 1602 vs. 6508 ± 1241 pg/
PlGF < 107 pg/ml or < 20 pg/ml on the 26th week are at risk of de- ml) when compared with non-pregnant women [58].
veloping moderate and severe PE [91], respectively. A recent study developed with 4380 offsprings from preeclamptic
In the third quarter, the change in PlGF levels, combined with pregnancies, highlighted the first genome-wide significant suscept-
maternal characteristics, has a sensitivity of 86% for the diagnosis of PE ibility locus (rs4769613), located nearby Flt-1 gene encoding Fms-like
between the 34th and 37th weeks of pregnancy, and 53% from the 37th tyrosine kinase-1. Higher expression of rs4769613 showed strong re-
week; with 10% false positive results [68]. Other studies point out that lation with offspring from pregnancies with late PE or which birth
the combined assessment of sFlt-1 and PlGF has a sensitivity of weights exceeded the tenth centile. Thus, this finding reinforces the
74.1–78%, with a specificity of 67.2–84% [2,92,93]. hereditary risk associated with PE, since a placental isoform of sFlt-1 is
Moreover, measurement of sFlt-1 and PlGF can contribute to the implicated in its etiopathogenesis [102].
identification and interpretation of abnormal uterine artery Doppler Experimental treatments are another promising research area.
ultrasound parameters in GA < 34 weeks [90,94]. Pregnant women According to a multicentric study, 10 mg of pravastatin (a hydrophilic
with increased rates of sFlt-1:PlGF are increasingly at risk of severe statin) helps diminishing PE and premature delivery in pregnant
hypertensive disorders, placental rupture, inotropic support, and en- women at high risk for PE. In this study, placebo group (10 subjects)
dotracheal intubation [94,95], whereas women with high levels of sEng had four women who developed PE and five had preterm deliveries
only present a serious risk of hematoma or hepatic rupture [94]. before 37 weeks compared with none PE cases and one preterm delivery
A prospective observational study conducted in 14 countries, en- in pravastatin group (10 subjects). In addition, a higher concentration
titled PROGNOSIS (Prediction of Short-Term Outcome in Pregnant of PlGF and a reduction of sFlt-1 and sEng concentration was observed
Women with Suspected Preeclampsia Study), investigated the value of in the group receiving pravastatin. It is also noted that five newborns
using the sFlt-1:PlGF ratio for prediction of preeclampsia in short term. from women in placebo group were admitted to an intermediate nur-
The median sFlt-1:PlGF ratio was higher among pregnant women with sery or to neonatal intensive care unit, while in the other group, only
PE or HELLP syndrome (146.4) within the first week or four weeks of two newborns required such attention [103].
evaluation (104.8), when compared to those who did not develop any Another research that evaluated the removal of sFlt-1 by whole-
disorder within the first week (6.3) or within four weeks (5.5). For the blood apheresis using charged dextran sulfate column, pointed out that
single-cutoff model (independent of gestational age; sFlt-1:PIGF pregnant women who received apheresis treatment presented a re-
ratio = 38), the gestational phase model [a model with two cutoff duction in serum levels of sFlt-1. Comparing the mean pre-apheresis
points, one for the earlier gestational phase (24 to < 34 weeks) and one and post-apheresis sFlt-1 concentration, there was a reduction from
for the later gestational phase (≥34 weeks)], and the gestational-week 17,394 (range 7916–35,301) pg/ml to 14,265 (6446–26,259) pg/ml,
model (a model with a cutoff point for each gestational week), the area respectively; an average reduction in sFlt-1 concentration of 18%
under the curve were 89.2%, 90.9%, and 90.5% for 1-week rule out and (7%–28%) per treatment. It is still outstanding a decrease in protein/
86.4%, 86.2%, and 86.2% for 4-week rule in, respectively. Moreover, creatinine (P/C) ratios following apheresis in almost all pregnant
the median cutoff points were 38.2 (1-week rule out) and 37.5 (4-week women. When compared the mean starting P/C ratio and post-treat-
rule in); indicating that the single cutoff point of 38 was suitable for ment values there was a reduction from 6.5 g/g (0.4–16.9 g/g) to 3.2
validation [96]. (0.2–8.6) g/g, respectively; an average reduction in P/C ratio of 44%
In validation phase, the median sFlt-1:PlGF ratio was 87.8 for [104].
pregnant women diagnosed with PE or HELLP syndrome within the first Although in the future it will be possible to establish an efficient and
week of evaluation and 59.4 within four weeks, when compared to effective diagnostic tool, the management of PE needs to follow three
those who did not develop any disorder within the first week (8.0) and key principles: prevention, early screening, and treatment. Therefore,
within four weeks (6.3). Negative predictive value (absence of diag- the identification of and risk calculation for PE is important, since
nosis within 1 week) of 38 or lower for the sFlt-1:PlGF ratio was 99.3%, dietary changes, smoking cessation, comorbidity monitoring, blood
and the positive predictive value (diagnosis of PE, eclampsia or HELLP pressure (BP), and weight gain, for example, can contribute to the
syndrome within 4 weeks) was 36.7%. Thus, the results suggest that the prevention of this condition [105].
use of sFlt-1:PlGF ratio to evaluate proteinuria and blood pressure The evaluation of serum electrolytes (e.g.: sodium, potassium, urea,
contributes to the diagnosis of PE [96]. creatinine, and uric acid), liver transaminases, blood counts (e.g.
It is noteworthy that in both cases there is a higher risk for pre- identification of thrombocytopenia, and bilirubin), 24-h urine (in order
maturity [44,61,97,98] and for deliveries of newborn small for GA to monitor proteinuria), and heart rate and lung function of the mother
[89,95,97,98]. sFlt-1:PlGF has been showed to be inversely correlated should be performed continuously, and so should analysis of cardioto-
with GA at birth, since 86% of pregnant women who gave birth at < 34 cography, fetal growth, amniotic fluid volume, and umbilical artery
weeks had rates higher than 85 [81]. perfusion through Doppler ultrasound [3,105]. Therefore, the im-
Other biomarkers and experimental treatments have a promising portance of high quality prenatal care for pregnancy risk stratification
future according to the literature. The serum level of placental glyco- into low- and high-risk groups.
protein pregnancy-associated plasma protein-A (PAPP-A) and of pla- The introduction of oral anti-hypertensives (e.g.: methyldopa, hy-
cental protein 13 (PP13), associated with Doppler ultrasound of the dralazine, β-blockers, and nifedipine) [105,106] and low-dose acet-
uterine artery, have a predictive value of approximately 70% [87,99] ylsalicylic acid (in high-risk pregnancies after the first trimester) [96]
and 86% [22,87], with 5% and 10% false positive results, respectively; should be taken into account for the control of BP (BP < 160 × 105
the concentration of PAPP-A and PP13 is higher in PE. Similarly, serum mm Hg, mean BP < 125 mm Hg), for the reduction of heart attack risk
levels of fetal hemoglobin, of α1-microglobulin, of activin A, of inhibin in hypertensive emergencies (≥160 × 105 mm Hg), as well as for the
A, and of cellular metabolism products become altered in PE, and ap- increase in placental blood flow [22,105]. In the imminence or signs of
pear to significantly contribute to the progression of the disease seizure, it is recommended to administer magnesium sulfate (MgSO4)
[87,100,101]. [dose of 4 g MgSO4, followed by infusion for 1 g/h up to 24 h after
Laboratory tests indicate elevated levels of urinary prolactin in PE delivery] [17,105,107] and plasma expanders [105,106]. Nonetheless,
accompanied by HELLP syndrome or eclampsia [95]; as well as high delivery is indicated in the presence of fetal distress, when BP is diffi-
plasma levels of malondialdehyde (10.72 ± 2.20 mmol/ml) than in cult to control and clinical parameters and eclampsia deteriorate, since
normotensive women (9.53 ± 2.45 mmol/ml), and positively asso- PE resolves after the delivery of the fetus and the delivery of the pla-
ciated with maternal plasma sFlt-1 levels [47]. It is also observed an centa [105].
increase in plasma levels of sTNF-R1 [29.34 (16.76–55.29) vs. 4.45 Furthermore, postpartum follow-up is essential for monitoring

11
F.R. Helmo et al. Pathology - Research and Practice 214 (2018) 7–14

maternal blood pressure, and for the subsequent confirmation of the Author contributions
resolution of gestational hypertension or diagnosis of systemic hy-
pertension in postpartum mothers, as well as for monitoring the risk of Fernanda Rodrigues Helmo: ‘I declare that I participated in the
renal and cardiovascular diseases [2,28,105]. project development, data collection, manuscript writing/editing and
The literature highlights the increased risk for cardiovascular dis- that I have seen and approved the final version. I have no conflicts of
eases (ex.: coronary death, myocardial infarction, fatal or non-fatal interest’. Angela Maria Moed Lopes: ‘I declare that I participated in
stroke, coronary insufficiency, angina pectoris, transient ischemic at- the project development, data collection, manuscript writing and that I
tack, intermittent claudication or congestive heart failure) among have seen and approved the final version. I have no conflicts of in-
women with previous PE [108–110]. In these women, significant terest’. Anna Cecília Dias Maciel Carneiro: ‘I declare that I partici-
changes in blood pressure, fasting glucose, fasting insulin, high-sensi- pated in the project development, data collection, manuscript writing
tive C-reactive protein, triglycerides and cholesterol were observed and that I have seen and approved the final version. I have no conflicts
about three months postpartum [108]. of interest’. Carolina Guissoni Campos: ‘I declare that I participated in
A research has shown that women diagnosed with isolated PE the project development, data collection, manuscript writing and that I
showed a doubled risk for major coronary events, compared with have seen and approved the final version. I have no conflicts of in-
women without PE; and the risk may triple if PE is accompanied by terest’. Polyana Barbosa Silva: ‘I declare that I participated in the
newborns small for gestational age and/or preterm delivery, or become project development, data collection, manuscript writing and that I
even larger than 4,7 times for women with recurrent PE, and with a risk have seen and approved the final version. I have no conflicts of in-
of dying even 23% higher in relation to women without PE [110]. terest’. Maria Luíza Gonçalves dos Reis Monteiro: ‘I declare that I
It should be noted that a meta-analysis indicates a relative risk of participated in the manuscript writing and that I have seen and ap-
3.70 for the diagnosis of hypertension after PE, compared to women proved the final version. I have no conflicts of interest’. Laura Penna
who did not develop PE. As well as a risk of 2.60 for fatal ischemic heart Rocha: ‘I declare that I participated in the data collection and that I
disease, which can be almost eight times higher if PE develops before have seen and approved the final version. I have no conflicts of in-
37 weeks. It also stands out the relative increased risk for fatal and non- terest’. Marlene Antônia dos Reis: ‘I declare that I participated in the
fatal stroke and venous thromboembolism in women who developed PE management and that I have seen and approved the final version. I have
[111]. no conflicts of interest’. Renata Margarida Etchebehere: ‘I declare
that I participated in the management and that I have seen and ap-
proved the final version. I have no conflicts of interest’. Juliana Reis
5. Conclusion Machado: ‘I declare that I participated in the management and that I
have seen and approved the final version. I have no conflicts of in-
Preeclampsia has a major impact on maternal and fetal health, since terest’. Rosana Rosa Miranda Corrêa: ‘I declare that I participated in
it is responsible for up to 15% of maternal deaths. Even though there the project development, management and that I have seen and ap-
are different predisposing factors, such as African descent, obesity and proved the final version. I have no conflicts of interest’.
previous CH, this hypertensive disorder is believed to be triggered by a
change in the expression of angiogenic and anti-angiogenic factors, Acknowledgements
which appears to be the key factor in the endothelial dysfunction ob-
served in this entity. We thank Conselho Nacional de Desenvolvimento Cientifico e
The expression of VEGF-A, PlGF, endoglin, and their respective re- Tecnológico (CNPq), Coordenação de Aperfeiçoamento de Pessoal de
ceptors Flt-1, Flk-1, sFlt-1 and sEng, is fundamental to the establish- Nível Superior (CAPES), Fundação de Amparo a Pesquisa do Estado de
ment of uteroplacental circulation. Nevertheless, in PE there is an in- Minas Gerais (FAPEMIG), and Fundação de Ensino e Pesquisa de
creased expression of sFlt-1 and sEng by the placenta, a decreased Uberaba (FUNEPU) for financial support.
expression of VEGF-A and PlGF, and, thus, impaired angiogenesis and
vasculogenesis. The imbalance between angiogenic and anti-angiogenic References
factors seems to be triggered by the increased expression of endothelin-
1, TNF-α and AT1-AAs, for instance. [1] E. Eiland, C. Nzerue, M. Faulkner, Preeclampsia 2012, J. Pregnancy 2012 (2012)
Studies have identified promising biomarkers that can promote the 586578.
early diagnosis of this entity in the future, such as VEGF, PlGF, sFlt-1, [2] T. Chaiworapongsa, P. Chaemsaithong, L. Yeo, R. Romero, Pre-eclampsia part 1:
current understanding of its pathophysiology, Nat. Rev. Nephrol. 10 (2014)
sEng, PAPP-A and PP13. However, screening and early treatment may 466–480.
contribute to the prevention of PE. [3] ACOG, Hypertension in Pregnancy, American College of Obstetricians and
Gynecologists (ACOG), Washington, 2013.
[4] A.B. Lerner, J.D. Case, Y. Takahashi, Isolation of melatonin and 5-methoxyindole-
Compliance with ethical standards 3-acetic acid from bovine pineal glands, J. Biol. Chem. 235 (1960) 1992–1997.
[5] Z. Liu, G.B. Afink, P.T. Dijke, Soluble fms-like tyrosine kinase 1 and soluble en-
doglin are elevated circulating anti-angiogenic factors in pre-eclampsia,
Funding Pregnancy Hypertens. 2 (2012) 358–367.
[6] M.D. Lindheimer, W. Benson, P. Harer, The history of preeclampsia and eclampsia
as seen by a nephrologist, Clin. Rev. 31 (2013) 8–12.
This study was funded by Conselho Nacional de Desenvolvimento [7] M. Silasi, B. Cohen, S.A. Karumanchi, S. Rana, Abnormal placentation angiogenic
Cientifico e Tecnológico (CNPq) (grant number 470029/20110), factors, and the pathogenesis of preeclampsia, Obstet. Gynecol. Clin. North Am. 37
Coordenação de Aperfeiçoamento de Pessoal de Nível Superior (CAPES) (2010) 239–253.
[8] A.M. Borzychowski, I.L. Sargent, C.W. Redman, Inflammation and pre-eclampsia,
(grant number PNPD-02604/094), Fundação de Amparo a Pesquisa do Semin. Fetal Neonatal Med. 11 (2006) 309–316.
Estado de Minas Gerais (FAPEMIG) (grant number CDS-APQ-02135- [9] M. Schmidt, B. Hoffmann, D. Beelen, A. Gellhaus, E. Winterhager, R. Kimmig,
14), and Fundação de Ensino e Pesquisa de Uberaba (FUNEPU) (grant S. Kasimir-Bauer, Detection of circulating trophoblast particles in peripheral ma-
ternal blood in preeclampsia complicated pregnancies, Hypertens. Pregnancy 27
number CDS-922/2009). All them from Brazil.
(2008) 131–142.
[10] R. Hong, M. Ruiz-Beltran, Impact of prenatal care on infant survival in Bangladesh,
Matern. Child Health J. 11 (2007) 199–206.
Ethical approval [11] L.O. Walker, L.W. Chesnut, Identifying health disparities and social inequities
affecting childbearing women and infants, J. Obstet. Gynecol. Neonatal Nurs. 39
(2010) 328–338.
This article does not contain any studies with human participants or [12] R.W. Powers, A. Jeyabalan, R.G. Clifton, P. Van Dorsten, J.C. Hauth,
animals performed by any of the authors.

12
F.R. Helmo et al. Pathology - Research and Practice 214 (2018) 7–14

M.A. Klebanoff, M.D. Lindheimer, B. Sibai, M. Landon, M. Miodovnik, Soluble fms- NK cells in pregnancies with impaired vascular remodelling, J. Reprod. Immunol.
Like tyrosine kinase 1 (sFlt1), endoglin and placental growth factor (PlGF) in 119 (2017) 81–84.
preeclampsia among high risk pregnancies, PLoS One 5 (2010) e13263. [41] A.E. Wallace, G.S. Whitley, B. Thilaganathan, J.E. Cartwright, Decidual natural
[13] L.L. Jardim, D.R. Rios, L.O. Perucci, L.P. de Sousa, K.B. Gomes, L.M. Dusse, Is the killer cell receptor expression is altered in pregnancies with impaired vascular
imbalance between pro-angiogenic and anti-angiogenic factors associated with remodeling and a higher risk of pre-eclampsia, J. Leukoc. Biol. 97 (2015) 79–86.
preeclampsia, Clin. Chim. Acta 447 (2015) 34–38. [42] R. Fraser, G.S. Whitley, B. Thilaganathan, J.E. Cartwright, Decidual natural killer
[14] G.E. Lash, H.A. Otun, B.A. Innes, M. Kirkley, L. De Oliveira, R.F. Searle, cells regulate vessel stability: implications for impaired spiral artery remodelling,
S.C. Robson, J.N. Bulmer, Interferon-gamma inhibits extravillous trophoblast cell J. Reprod. Immunol. 110 (2015) 54–60.
invasion by a mechanism that involves both changes in apoptosis and protease [43] A. Moffett, S.E. Hiby, How Does the maternal immune system contribute to the
levels, FASEB J. 20 (2006) 2512–2518. development of pre-eclampsia? Placenta 28 (Suppl. A) (2007) S51–S56.
[15] T. Chaiworapongsa, R. Romero, S.J. Korzeniewski, J.P. Kusanovic, E. Soto, J. Lam, [44] T. Chaiworapongsa, R. Romero, J. Espinoza, E. Bujold, Y. Mee Kim, L.F. Goncalves,
Z. Dong, N.G. Than, L. Yeo, E. Hernandez-Andrade, A. Conde-Agudelo, R. Gomez, S. Edwin, Evidence supporting a role for blockade of the vascular en-
S.S. Hassan, Maternal plasma concentrations of angiogenic/antiangiogenic factors dothelial growth factor system in the pathophysiology of preeclampsia. Young
in the third trimester of pregnancy to identify the patient at risk for stillbirth at or Investigator Award, Am. J. Obstet. Gynecol. 190 (2004) 1541–1547 discussion
near term and severe late preeclampsia, Am. J. Obstet. Gynecol. 208 (2013) 287 1547–1550.
e281–287. e215. [45] S.W. Walsh, Maternal-placental interactions of oxidative stress and antioxidants in
[16] L. Duley, The global impact of pre-eclampsia and eclampsia, Semin. Perinatol. 33 preeclampsia, Semin. Reprod. Endocrinol. 16 (1998) 93–104.
(2009) 130–137. [46] T.H. Hung, G.J. Burton, Hypoxia and reoxygenation: a possible mechanism for
[17] S.E. Maynard, S.A. Karumanchi, Angiogenic factors and preeclampsia, Semin. placental oxidative stress in preeclampsia, Taiwan J. Obstet. Gynecol. 45 (2006)
Nephrol. 31 (2011) 33–46. 189–200.
[18] T.C. Nogueira, C. Lellis-Santos, D.S. Jesus, M. Taneda, S.C. Rodrigues, F.G. Amaral, [47] A.V. Kulkarni, S.S. Mehendale, H.R. Yadav, A.S. Kilari, V.S. Taralekar, S.R. Joshi,
A.M. Lopes, J. Cipolla-Neto, S. Bordin, G.F. Anhê, Absence of melatonin induces Circulating angiogenic factors and their association with birth outcomes in pre-
night-time hepatic insulin resistance and increased gluconeogenesis due to sti- eclampsia, Hypertens. Res. 33 (2010) 561–567.
mulation of nocturnal unfolded protein response, Endocrinology 152 (2011) [48] A. Ahmed, New insights into the etiology of preeclampsia: identification of key
1253–1263. elusive factors for the vascular complications, Thromb. Res. 127 (Suppl. 3) (2011)
[19] Brasil, DATASUS (Ed.), Mortalidade e nascidos vivos: óbitos maternos, Ministério S72–S75.
da Saúde, Brasília, 2015. [49] A.S. Cerdeira, S.A. Karumanchi, Angiogenic factors in preeclampsia and related
[20] R. Pijnenborg, J.M. Bland, W.B. Robertson, I. Brosens, Uteroplacental arterial disorders, Cold Spring Harb. Perspect. Med. 2 (2012).
changes related to interstitial trophoblast migration in early human pregnancy, [50] A.E. Wallace, R. Fraser, S. Gurung, S.S. Goulwara, G.S. Whitley, A.P. Johnstone,
Placenta 4 (1983) 397–413. J.E. Cartwright, Increased angiogenic factor secretion by decidual natural killer
[21] S. Gupta, A. Agarwal, R.K. Sharma, The role of placental oxidative stress and lipid cells from pregnancies with high uterine artery resistance alters trophoblast
peroxidation in preeclampsia, Obstet. Gynecol. Surv. 60 (2005) 807–816. function, Hum. Reprod. 29 (2014) 652–660.
[22] A.F. Tanbe, R.A. Khalil, Circulating and vascular bioactive factors during hy- [51] A. Pratt, F. Da Silva Costa, A.J. Borg, B. Kalionis, R. Keogh, P. Murthi, Placenta-
pertension in pregnancy, Curr. Bioact. Compd. 6 (2010) 60–75. derived angiogenic proteins and their contribution to the pathogenesis of pre-
[23] A. Kharfi, Y. Giguere, V. Sapin, J. Masse, B. Dastugue, J.C. Forest, Trophoblastic eclampsia, Angiogenesis 18 (2015) 115–123.
remodeling in normal and preeclamptic pregnancies: implication of cytokines, [52] R. Romero, J.K. Nien, J. Espinoza, D. Todem, W. Fu, H. Chung, J.P. Kusanovic,
Clin. Biochem. 36 (2003) 323–331. F. Gotsch, O. Erez, S. Mazaki-Tovi, R. Gomez, S. Edwin, T. Chaiworapongsa,
[24] B. Sibai, G. Dekker, M. Kupferminc, Pre-eclampsia, Lancet 365 (2005) 785–799. R.J. Levine, S.A. Karumanchi, A longitudinal study of angiogenic (placental
[25] W. Ramma, I.A. Buhimschi, G. Zhao, A.T. Dulay, U.A. Nayeri, C.S. Buhimschi, growth factor) and anti-angiogenic (soluble endoglin and soluble vascular en-
A. Ahmed, The elevation in circulating anti-angiogenic factors is independent of dothelial growth factor receptor-1) factors in normal pregnancy and patients
markers of neutrophil activation in preeclampsia, Angiogenesis 15 (2012) destined to develop preeclampsia and deliver a small for gestational age neonate,
333–340. J. Matern. Fetal Neonatal Med. 21 (2008) 9–23.
[26] A. Elsheikh, G. Creatsas, G. Mastorakos, S. Milingos, D. Loutradis, S. Michalas, The [53] R.J. Levine, S.E. Maynard, C. Qian, K.H. Lim, L.J. England, K.F. Yu,
renin-aldosterone system during normal and hypertensive pregnancy, Arch. E.F. Schisterman, R. Thadhani, B.P. Sachs, F.H. Epstein, B.M. Sibai, V.P. Sukhatme,
Gynecol. Obstet. 264 (2001) 182–185. S.A. Karumanchi, Circulating angiogenic factors and the risk of preeclampsia, N.
[27] F. Khan, J.J. Belch, M. MacLeod, G. Mires, Changes in endothelial function pre- Engl. J. Med. 350 (2004) 672–683.
cede the clinical disease in women in whom preeclampsia develops, Hypertension [54] S.E. Maynard, J.Y. Min, J. Merchan, K.H. Lim, J. Li, S. Mondal, T.A. Libermann,
46 (2005) 1123–1128. J.P. Morgan, F.W. Sellke, I.E. Stillman, F.H. Epstein, V.P. Sukhatme,
[28] M. Sircar, R. Thadhani, S.A. Karumanchi, Pathogenesis of preeclampsia, Curr. S.A. Karumanchi, Excess placental soluble fms-like tyrosine kinase 1 (sFlt1) may
Opin. Nephrol. Hypertens. 24 (2015) 131–138. contribute to endothelial dysfunction hypertension, and proteinuria in pre-
[29] A.C. Palei, F.T. Spradley, J.P. Warrington, E.M. George, J.P. Granger, eclampsia, J. Clin. Invest. 111 (2003) 649–658.
Pathophysiology of hypertension in pre-eclampsia: a lesson in integrative phy- [55] M. Shibuya, L. Claesson-Welsh, Signal transduction by VEGF receptors in regula-
siology, Acta Physiol. (Oxf.) 208 (2013) 224–233. tion of angiogenesis and lymphangiogenesis, Exp. Cell Res. 312 (2006) 549–560.
[30] C.W. Redman, I.L. Sargent, Latest advances in understanding preeclampsia, [56] A.S. Maharaj, P.A. D’Amore, Roles for VEGF in the adult, Microvasc. Res. 74
Science 308 (2005) 1592–1594. (2007) 100–113.
[31] E. Soto, R. Romero, J.P. Kusanovic, G. Ogge, Y. Hussein, L. Yeo, S.S. Hassan, [57] C.L. Haggerty, M.E. Seifert, G. Tang, J. Olsen, D.C. Bass, S.A. Karumanchi,
C.J. Kim, T. Chaiworapongsa, Late-onset preeclampsia is associated with an im- R.B. Ness, Second trimester anti-angiogenic proteins and preeclampsia, Pregnancy
balance of angiogenic and anti-angiogenic factors in patients with and without Hypertens. 2 (2012) 158–163.
placental lesions consistent with maternal underperfusion, J. Matern. Fetal [58] L.O. Perucci, K.B. Gomes, L.G. Freitas, L.C. Godoi, P.N. Alpoim, M.B. Pinheiro,
Neonatal Med. 25 (2012) 498–507. A.S. Miranda, A.L. Teixeira, L.M. Dusse, L.P. Sousa, Soluble endoglin, transforming
[32] M. Egbor, T. Ansari, N. Morris, C.J. Green, P.D. Sibbons, Morphometric placental growth factor-Beta 1 and soluble tumor necrosis factor alpha receptors in different
villous and vascular abnormalities in early- and late-onset pre-eclampsia with and clinical manifestations of preeclampsia, PLoS One 9 (2014) e97632.
without fetal growth restriction, BJOG 113 (2006) 580–589. [59] S. Venkatesha, M. Toporsian, C. Lam, J. Hanai, T. Mammoto, Y.M. Kim, Y. Bdolah,
[33] N.M. Hunkapiller, M. Gasperowicz, M. Kapidzic, V. Plaks, E. Maltepe, K.H. Lim, H.T. Yuan, T.A. Libermann, I.E. Stillman, D. Roberts, P.A. D'Amore,
J. Kitajewski, J.C. Cross, S.J. Fisher, A role for Notch signaling in trophoblast F.H. Epstein, F.W. Sellke, R. Romero, V.P. Sukhatme, M. Letarte, S.A. Karumanchi,
endovascular invasion and in the pathogenesis of pre-eclampsia, Development 138 Soluble endoglin contributes to the pathogenesis of preeclampsia, Nat. Med. 12
(2011) 2987–2998. (2006) 642–649.
[34] V. Rigourd, C. Chauvet, S.T. Chelbi, R. Rebourcet, F. Mondon, F. Letourneur, [60] T.J. Kaitu’u-Lino, K.R. Palmer, C.L. Whitehead, E. Williams, M. Lappas, S. Tong,
T.M. Mignot, S. Barbaux, D. Vaiman, STOX1 overexpression in choriocarcinoma MMP-14 is expressed in preeclamptic placentas and mediates release of soluble
cells mimics transcriptional alterations observed in preeclamptic placentas, PLoS endoglin, Am. J. Pathol. 180 (2012) 888–894.
One 3 (2008) e3905. [61] R.J. Levine, C. Lam, C. Qian, K.F. Yu, S.E. Maynard, B.P. Sachs, B.M. Sibai,
[35] G.J. Burton, H.W. Yung, Endoplasmic reticulum stress in the pathogenesis of early- F.H. Epstein, R. Romero, R. Thadhani, S.A. Karumanchi, Soluble endoglin and
onset pre-eclampsia, Pregnancy Hypertens. 1 (2011) 72–78. other circulating antiangiogenic factors in preeclampsia, N. Engl. J. Med. 355
[36] D.S. Charnock-Jones, Placental hypoxia, endoplasmic reticulum stress and ma- (2006) 992–1005.
ternal endothelial sensitisation by sFLT1 in pre-eclampsia, J. Reprod. Immunol. [62] P.K. Aggarwal, N. Chandel, V. Jain, V. Jha, The relationship between circulating
114 (2016) 81–85. endothelin-1: soluble fms-like tyrosine kinase-1 and soluble endoglin in pre-
[37] L. Du, F. He, L. Kuang, W. Tang, Y. Li, D. Chen, eNOS/iNOS and endoplasmic eclampsia, Am. J. Pathol. 180 (2012) 888–894.
reticulum stress-induced apoptosis in the placentas of patients with preeclampsia, [63] N. Ferrara, H.P. Gerber, J. LeCouter, The biology of VEGF and its receptors, Nat.
J. Hum. Hypertens. 31 (2017) 49–55. Med. 9 (2003) 669–676.
[38] J. Fu, L. Zhao, L. Wang, X. Zhu, Expression of markers of endoplasmic reticulum [64] M.G. Cardenas-Mondragon, G. Vallejo-Flores, J. Delgado-Dominguez, J.F. Romero-
stress-induced apoptosis in the placenta of women with early and late onset severe Arauz, A. Gomez-Delgado, G. Aguilar-Madrid, J.J. Sanchez-Barriga, J. Marquez-
pre-eclampsia, Taiwan J. Obstet. Gynecol. 54 (2015) 19–23. Acosta, Preeclampsia is associated with lower production of vascular endothelial
[39] A. Moffett, S. Hiby, Influence of activating and inhibitory killer immunoglobulin- growth factor by peripheral blood mononuclear cells, Arch. Med. Res. 45 (2014)
like receptors on predisposition to recurrent miscarriages, Hum. Reprod. 24 561–569.
(2009) 2048–2049. [65] A. Wang, S. Rana, S.A. Karumanchi, Preeclampsia: the role of angiogenic factors in
[40] J.E. Cartwright, L. James-Allan, R.J. Buckley, A.E. Wallace, The role of decidual its pathogenesis, Physiology (Bethesda) 24 (2009) 147–158.

13
F.R. Helmo et al. Pathology - Research and Practice 214 (2018) 7–14

[66] H. Stepan, A. Geipel, F. Schwarz, T. Kramer, N. Wessel, R. Faber, Circulatory so- [91] S.K. Ghosh, S. Raheja, A. Tuli, C. Raghunandan, S. Agarwal, Serum PLGF as a
luble endoglin and its predictive value for preeclampsia in second-trimester potential biomarker for predicting the onset of preeclampsia, Arch. Gynecol.
pregnancies with abnormal uterine perfusion, Am. J. Obstet. Gynecol. 198 (2008) Obstet. 285 (2012) 417–422.
175 e171–176. [92] H. Stepan, A. Unversucht, N. Wessel, R. Faber, Predictive value of maternal an-
[67] L. Govender, I. Mackraj, P. Gathiram, J. Moodley, The role of angiogenic: anti- giogenic factors in second trimester pregnancies with abnormal uterine perfusion,
angiogenic and vasoactive factors in pre-eclamptic African women: early- versus Hypertension 49 (2007) 818–824.
late-onset pre-eclampsia, Cardiovasc. J. Afr. 23 (2012) 153–159. [93] A. Molvarec, A. Szarka, S. Walentin, E. Szucs, B. Nagy, J. Rigo Jr., Circulating
[68] E. Llurba, F. Crispi, S. Verlohren, Update on the pathophysiological implications angiogenic factors determined by electrochemiluminescence immunoassay in re-
and clinical role of angiogenic factors in pregnancy, Fetal Diagn. Ther. 37 (2015) lation to the clinical features and laboratory parameters in women with pre-
81–92. eclampsia, Hypertens. Res. 33 (2010) 892–898.
[69] C. Lam, K.H. Lim, S.A. Karumanchi, Circulating angiogenic factors in the patho- [94] J. Stubert, S. Ullmann, M. Bolz, T. Külz, M. Dieterich, D.U. Richter, T. Reimer,
genesis and prediction of preeclampsia, Hypertension 46 (2005) 1077–1085. Prediction of preeclampsia and induced delivery at < 34 weeks gestation by sFLT-
[70] A.C. Valbuena-Diez, F.J. Blanco, B. Oujo, C. Langa, M. Gonzalez-Nunez, E. Llano, 1 and PlGF in patients with abnormal midtrimester uterine Doppler velocimetry: a
A.M. Pendas, M. Diaz, A. Castrillo, J.M. Lopez-Novoa, C. Bernabeu, Oxysterol- prospective cohort analysis, BMC Pregnancy Childbirth 14 (2014) 1–11.
induced soluble endoglin release and its involvement in hypertension, Circulation [95] A. Leaños-Miranda, I. Campos-Galicia, K.L. Ramírez-Valenzuela, Z.L. Chinolla-
126 (2012) 2612–2624. Arellano, I. Isordia-Salas, Circulating angiogenic factors and urinary prolactin as
[71] A. Goel, S. Rana, Angiogenic factors in preeclampsia: potential for diagnosis and predictors of adverse outcomes in women with preeclampsia, Hypertension 61
treatment, Curr. Opin. Nephrol. Hypertens. 22 (2013) 643–650. (2013) 1118–1125.
[72] S.E. Maynard, S. Venkatesha, R. Thadhani, S.A. Karumanchi, Soluble Fms-like [96] H. Zeisler, E. Llurba, F. Chantraine, M. Vatish, A.C. Staff, M. Sennstrom,
tyrosine kinase 1 and endothelial dysfunction in the pathogenesis of preeclampsia, M. Olovsson, S.P. Brennecke, H. Stepan, D. Allegranza, P. Dilba, M. Schoedl,
Pediatr. Res. 57 (2005) 1R–7R. M. Hund, S. Verlohren, Predictive value of the sFlt-1:PlGF ratio in women with
[73] S. Rana, S.A. Karumanchi, R.J. Levine, S. Venkatesha, J.A. Rauh-Hain, H. Tamez, suspected preeclampsia, N. Engl. J. Med. 374 (2016) 13–22.
R. Thadhani, Sequential changes in antiangiogenic factors in early pregnancy and [97] O. Erez, R. Romero, J. Espinoza, W. Fu, D. Todem, J.P. Kusanovic, F. Gotsch,
risk of developing preeclampsia, Hypertension 50 (2007) 137–142. S. Edwin, J.K. Nien, T. Chaiworapongsa, P. Mittal, S. Mazaki-Tovi, N.G. Than,
[74] L.J. Vatten, A. Eskild, T.I. Nilsen, S. Jeansson, P.A. Jenum, A.C. Staff, Changes in R. Gomez, S.S. Hassan, The change in concentrations of angiogenic and anti-an-
circulating level of angiogenic factors from the first to second trimester as pre- giogenic factors in maternal plasma between the first and second trimesters in risk
dictors of preeclampsia, Am. J. Obstet. Gynecol. 196 (2007) 239 e231–236. assessment for the subsequent development of preeclampsia and small-for-gesta-
[75] A.R. Saxena, E.W. Seely, J.W. Rich-Edwards, L.E. Wilkins-Haug, S.A. Karumanchi, tional age, J. Matern. Fetal Neonatal Med. 21 (2008) 279–287.
T.F. McElrath, First trimester PAPP-A levels correlate with sFlt-1 levels long- [98] L.J. Vatten, B.O. Åsvold, A. Eskild, Angiogenic factors in maternal circulation and
itudinally in pregnant women with and without preeclampsia, BMC Pregnancy preeclampsia with or without fetal growth restriction, Acta Obstet. Gynecol.
Childbirth 13 (2013) 1–9. Scand. 91 (2012) 1388–1394.
[76] Y. Purwosunu, A. Sekizawa, A. Farina, N. Wibowo, K. Koide, S. Okazaki, [99] A.O. Odibo, Y. Zhong, K.R. Goetzinger, L. Odibo, J.L. Bick, C.R. Bower,
M. Nakamura, T. Okai, Evaluation of physiological alterations of the placenta D.M. Nelson, First-trimester placental protein 13 PAPP-A, uterine artery Doppler
through analysis of cell-free messenger ribonucleic acid concentrations of angio- and maternal characteristics in the prediction of pre-eclampsia, Placenta 32
genic factors, Am. J. Obstet. Gynecol. 198 (2008) 124 e121–127. (2011) 598–602.
[77] S.C. Kim, M.J. Park, B.S. Joo, J.K. Joo, D.S. Suh, K.S. Lee, Decreased expressions of [100] A. Reddy, S. Suri, I.L. Sargent, C.W. Redman, S. Muttukrishna, Maternal circu-
vascular endothelial growth factor and visfatin in the placental bed of pregnancies lating levels of activin A, inhibin A, sFlt-1 and endoglin at parturition in normal
complicated by preeclampsia, J. Obstet. Gynaecol. Res. 38 (2012) 665–673. pregnancy and pre-eclampsia, PLoS One 4 (2009) e4453.
[78] H. Strevens, D. Wide-Swensson, A. Hansen, T. Horn, I. Ingemarsson, S. Larsen, [101] A.Y. Pramatirta, J. Mose, J.S. Effendi, S.R. Krisnadi, A.D. Anwar, P.N. Fauziah,
J. Willner, S. Olsen, Glomerular endotheliosis in normal pregnancy and pre- J.I. Gurnadi, D.D. Rihibiha, Correlation between cell-free mRNA expressions and
eclampsia, BJOG 110 (2003) 831–836. PLGF protein level in severe preeclampsia, BMC Res. Notes 8 (2015) 1–6.
[79] E. Vaisbuch, J.E. Whitty, S.S. Hassan, R. Romero, J.P. Kusanovic, D.B. Cotton, [102] R. McGinnis, V. Steinthorsdottir, N.O. Williams, G. Thorleifsson, S. Shooter,
Y. Sorokin, S.A. Karumanchi, Circulating angiogenic and antiangiogenic factors in S. Hjartardottir, S. Bumpstead, L. Stefansdottir, L. Hildyard, J.K. Sigurdsson,
women with eclampsia, Am. J. Obstet. Gynecol. 204 (2011) 152 e151–159. J.P. Kemp, G.B. Silva, L.C.V. Thomsen, T. Jaaskelainen, E. Kajantie, S. Chappell,
[80] F. Crispi, C. Dominguez, E. Llurba, P. Martin-Gallan, L. Cabero, E. Gratacos, N. Kalsheker, A. Moffett, S. Hiby, W.K. Lee, S. Padmanabhan, N.A.B. Simpson,
Placental angiogenic growth factors and uterine artery Doppler findings for V.A. Dolby, E. Staines-Urias, S.M. Engel, A. Haugan, L. Trogstad, G. Svyatova,
characterization of different subsets in preeclampsia and in isolated intrauterine N. Zakhidova, D. Najmutdinova, A.F. Dominiczak, H.K. Gjessing, J.P. Casas,
growth restriction, Am. J. Obstet. Gynecol. 195 (2006) 201–207. F. Dudbridge, J.J. Walker, F.B. Pipkin, U. Thorsteinsdottir, R.T. Geirsson,
[81] S. Rana, C.E. Powe, S. Salahuddin, S. Verlohren, F.H. Perschel, R.J. Levine, D.A. Lawlor, A.C. Iversen, P. Magnus, H. Laivuori, K. Stefansson, L. Morgan,
K.H. Lim, J.B. Wenger, R. Thadhani, S.A. Karumanchi, Angiogenic factors and the Variants in the fetal genome near FLT1 are associated with risk of preeclampsia,
risk of adverse outcomes in women with suspected preeclampsia, Circulation 125 Nat. Genet. 49 (2017) 1255–1260.
(2012) 911–919. [103] M.M. Costantine, K. Cleary, M.F. Hebert, M.S. Ahmed, L.M. Brown, Z. Ren,
[82] E. Laresgoiti-Servitje, N. Gomez-Lopez, The pathophysiology of preeclampsia in- T.R. Easterling, D.M. Haas, L.S. Haneline, S.N. Caritis, R. Venkataramanan,
volves altered levels of angiogenic factors promoted by hypoxia and autoantibody- H. West, M. D’Alton, G. Hankins, E.K.S.N.I.o.C.H.a.H.D.O.-F.P.R.U. Network,
mediated mechanisms, Biol. Reprod. 87 (2012) 36. Safety and pharmacokinetics of pravastatin used for the prevention of pre-
[83] M.R. Parrish, S.R. Murphy, S. Rutland, K. Wallace, K. Wenzel, G. Wallukat, eclampsia in high-risk pregnant women: a pilot randomized controlled trial, Am. J.
S. Keiser, L.F. Ray, R. Dechend, J.N. Martin, J.P. Granger, B. LaMarca, The effect of Obstet. Gynecol. 214 (2016) 20 e21–720. e717.
immune factors tumor necrosis factor-alpha, and agonistic autoantibodies to the [104] R. Thadhani, H. Hagmann, W. Schaarschmidt, B. Roth, T. Cingoez,
angiotensin II type I receptor on soluble fms-like tyrosine-1 and soluble endoglin S.A. Karumanchi, J. Wenger, K.J. Lucchesi, H. Tamez, T. Lindner, A. Fridman,
production in response to hypertension during pregnancy, Am. J. Hypertens. 23 U. Thome, A. Kribs, M. Danner, S. Hamacher, P. Mallmann, H. Stepan, T. Benzing,
(2010) 911–916. Removal of soluble Fms-like tyrosine kinase-1 by dextran sulfate apheresis in
[84] D.S. Torry, H.S. Wang, T.H. Wang, M.R. Caudle, R.J. Torry, Preeclampsia is as- preeclampsia, J. Am. Soc. Nephrol.: JASN 27 (2016) 903–913.
sociated with reduced serum levels of placenta growth factor, Am. J. Obstet. [105] F.A. English, L.C. Kenny, F.P. McCarthy, Risk factors and effective management of
Gynecol. 179 (1998) 1539–1544. preeclampsia, Integr. Blood Press Control 8 (2015) 7–12.
[85] D. Fujita, A. Tanabe, T. Sekijima, H. Soen, K. Narahara, Y. Yamashita, Y. Terai, [106] L. Duley, J. Williams, D.J. Henderson-Smart, Plasma volume expansion for treat-
H. Kamegai, M. Ohmichi, Role of extracellular signal-regulated kinase and AKT ment of women with pre-eclampsia, Cochrane Database Syst. Rev. (2000)
cascades in regulating hypoxia-induced angiogenic factors produced by a tro- CD001805.
phoblast-derived cell line, J. Endocrinol. 206 (2010) 131–140. [107] D. Altman, G. Carroli, L. Duley, B. Farrell, J. Moodley, J. Neilson, D. Smith,
[86] T. Cindrova-Davies, D.A. Sanders, G.J. Burton, D.S. Charnock-Jones, Soluble FLT1 M.T.C. Group, Do women with pre-eclampsia and their babies, benefit from
sensitizes endothelial cells to inflammatory cytokines by antagonizing VEGF re- magnesium sulphate? The Magpie Trial: a randomised placebo-controlled trial,
ceptor-mediated signalling, Cardiovasc. Res. 89 (2011) 671–679. Lancet 359 (2002) 1877–1890.
[87] U.D. Anderson, M.G. Olsson, K.H. Kristensen, B. Akerstrom, S.R. Hansson, Review: [108] J.H. Veerbeek, W. Hermes, A.Y. Breimer, B.B. van Rijn, S.V. Koenen, B.W. Mol,
biochemical markers to predict preeclampsia, Placenta 33 (Suppl) (2012) A. Franx, C.J. de Groot, M.P. Koster, Cardiovascular disease risk factors after early-
S42–S47. onset preeclampsia late-onset preeclampsia, and pregnancy-induced hypertension,
[88] W. Schaarschmidt, S. Rana, H. Stepan, The course of angiogenic factors in early- vs Hypertension 65 (2015) 600–606.
late-onset preeclampsia and HELLP syndrome, J. Perinat. Med. 41 (2013) [109] N.M. Breetveld, C. Ghossein-Doha, S. van Kuijk, A.P. van Dijk, M.J. van der Vlugt,
511–516. W.M. Heidema, R.R. Scholten, M.E. Spaanderman, Cardiovascular disease risk is
[89] E.R. Unal, C.J. Robinson, D.D. Johnson, E.Y. Chang, Second-trimester angiogenic only elevated in hypertensive formerly preeclamptic women, BJOG 122 (2015)
factors as biomarkers for future-onset preeclampsia, Am. J. Obstet. Gynecol. 197 1092–1100.
(2007) 211 e211–214. [110] H.K. Riise, G. Sulo, G.S. Tell, J. Igland, O. Nygard, S.E. Vollset, A.C. Iversen,
[90] J.P. Kusanovic, R. Romero, T. Chaiworapongsa, O. Erez, P. Mittal, E. Vaisbuch, R. Austgulen, A.K. Daltveit, Incident coronary heart disease after preeclampsia:
S. Mazaki-Tovi, F. Gotsch, S.S. Edwin, R. Gomez, L. Yeo, A. Conde-Agudelo, role of reduced fetal growth, preterm delivery, and parity, J. Am. Heart Assoc. 6
S.S. Hassan, A prospective cohort study of the value of maternal plasma con- (2017).
centrations of angiogenic and anti-angiogenic factors in early pregnancy and [111] L. Bellamy, J.P. Casas, A.D. Hingorani, D.J. Williams, Pre-eclampsia and risk of
midtrimester in the identification of patients destined to develop preeclampsia, J. cardiovascular disease and cancer in later life: systematic review and meta-ana-
Matern. Fetal Neonatal Med. 22 (2009) 1021–1038. lysis, BMJ 335 (2007) 974.

14

You might also like