Sei sulla pagina 1di 10

Am J Physiol Heart Circ Physiol 294: H541–H550, 2008.

First published November 30, 2007; doi:10.1152/ajpheart.01113.2007. Invited Review

Pathophysiology of hypertension during preeclampsia: linking placental


ischemia with endothelial dysfunction
Jeffrey S. Gilbert, Michael J. Ryan, Babbette B. LaMarca, Mona Sedeek, Sydney R. Murphy,
and Joey P. Granger
Department of Physiology and Biophysics and Center for Excellence in Cardiovascular-Renal Research, University
of Mississippi Medical Center, Jackson, Mississippi

Gilbert JS, Ryan MJ, LaMarca BB, Sedeek M, Murphy SR, Granger JP.
Pathophysiology of hypertension during preeclampsia: linking placental ischemia
with endothelial dysfunction. Am J Physiol Heart Circ Physiol 294: H541–H550,
2008. First published November 30, 2007; doi:10.1152/ajpheart.01113.2007.—
Studies over the last decade have provided exciting new insights into potential
mechanisms underlying the pathogenesis of preeclampsia. The initiating event in
preeclampsia is generally regarded to be placental ischemia/hypoxia, which in turn
results in the elaboration of a variety of factors from the placenta that generates
profound effects on the cardiovascular system. This host of molecules includes
factors such as soluble fms-like tyrosine kinase-1, the angiotensin II type 1 receptor
autoantibody, and cytokines such as tumor necrosis factor-␣, which generate
widespread dysfunction of the maternal vascular endothelium. This dysfunction
manifests as enhanced formation of factors such as endothelin, reactive oxygen
species, and augmented vascular sensitivity to angiotensin II. Alternatively, the
preeclampsia syndrome may also be evidenced as decreased formation of vasodi-
lators such as nitric oxide and prostacyclin. Taken together, these alterations cause
hypertension by impairing renal pressure natriuresis and increasing total peripheral
resistance. Moreover, the quantitative importance of the various endothelial and
humoral factors that mediate vasoconstriction and elevation of arterial pressure
during preeclampsia remains to be elucidated. Thus identifying the connection
between placental ischemia/hypoxia and maternal cardiovascular abnormalities in
hopes of revealing potential therapeutic regimens remains an important area of
investigation and will be the focus of this review.
kidney; pregnancy; nitric oxide; vascular endothelial growth factor; cytokines

PREECLAMPSIA, a pregnancy-specific syndrome characterized by development of suitable animal models for mechanistic re-
new onset hypertension and proteinuria, is a considerable search of this disease (72). Consequently, it is held by many
obstetric problem and a significant source of maternal and that more effective strategies for prevention and treatment of
neonatal morbidity and mortality (12, 95). Although pre- preeclampsia shall be forthcoming with the recent progress in
eclampsia and related hypertensive disorders of pregnancy developing animal models that allow careful mechanistic in-
continue to affect ⬃8% of all pregnancies, the incidence of vestigation of the underlying pathophysiological mechanisms
preeclampsia has seen a 40% increase in recent years (81). involved in preeclampsia (32).
Moreover, it has recently been recognized that women who
endure preeclampsia are at a greater risk for cardiovascular Placental Ischemia/Hypoxia and the Etiology
disease than nonpreeclamptic women and the men who fa- of Preeclampsia
thered those preeclamptic pregnancies (37). Despite thorough
characterization of the preeclamptic syndrome and a suite of Although the pathophysiology of preeclampsia remains un-
contributing circulating factors (12, 77, 80, 95), the mecha- defined, placental ischemia/hypoxia is widely regarded as a
nisms underlying the pathogenesis of this troubling condition key factor (24, 28, 80). Inadequate trophoblast invasion leading
remain nebulous. Interestingly, it has been proposed that not to incomplete remodeling of the uterine spiral arteries is
only are increased circulating factors responsible for much of considered to be a primary cause of placental ischemia (24).
the preeclamptic syndrome, but they may also predispose the Thus the poorly perfused and hypoxic placenta is thought to
maternal cardiovascular system to subsequent endothelial dys- synthesize and release increased amounts of vasoactive factors
function as the mother ages (20). such as soluble fms-like tyrosine kinase-1 (sFlt-1), cytokines,
The uncertainties regarding the mechanisms of preeclampsia and possibly the angiotensin II (ANG II) type 1 receptor
are at least partially attributable to difficulties faced in the autoantibodies (AT1-AA) (24, 66, 78, 79, 111). Figure 1
illustrates a model by which these and other candidate mole-
Address for reprint requests and other correspondence: J. P. Granger, Dept. of
cules are thought to induce widespread activation/dysfunction
Physiology and Biophysics, Univ. of Mississippi Medical Ctr., 2500 N. State St., of the maternal endothelium in vessels of the kidney and other
Jackson, MS 39216-4505 (e-mail: jgranger@physiology.umsmed.edu). organs that ultimately results in hypertension.
http://www.ajpheart.org 0363-6135/08 $8.00 Copyright © 2008 the American Physiological Society H541
Downloaded from www.physiology.org/journal/ajpheart by ${individualUser.givenNames} ${individualUser.surname} (036.084.226.018) on February 5, 2018.
Copyright © 2008 American Physiological Society. All rights reserved.
Invited Review
H542 PLACENTAL ISCHEMIA AND CARDIOVASCULAR DYSFUNCTION

Fig. 1. Pathways by which reduced uterine perfusion pressure


(RUPP) and placental ischemia may lead to endothelial and
cardiovascular dysfunction during pregnancy. Placental ischemia
results in increased synthesis of soluble fms-like tyrosine kinase-1
(sFlt-1), TNF-␣ and IL-6, angiotensin II type 1 receptor autoan-
tibodies (AT1-AA), and thromboxane (TX). Elevations in these
factors are proposed to result in endothelial dysfunction by
decreases in bioavailable nitric oxide (NO) and increased reactive
oxygen species (ROS) and endothelin-1 (ET-1), which in turn
results in altered renal function, increased total peripheral resis-
tance (TPR), and ultimately hypertension. PlGF, placental growth
factor.

Perhaps the most prominent molecule postulated to play a theless, experimental data investigating the mechanisms under-
key role in the pathogenesis of preeclampsia is sFlt-1. Several lying preeclampsia have been limited because of the difficulty
lines of evidence (15, 41, 42, 47, 50, 51, 74, 93, 103, 104, 117) in performing mechanistic studies in pregnant women. Al-
support the hypothesis that the ischemic placenta contributes to though several animal models have been developed to study
endothelial cell dysfunction in the maternal vasculature by preeclampsia, none to date completely represents the protean
inducing an alteration in the balance of circulating levels of characteristics of the human syndrome. Furthermore, informa-
angiogenic/antiangiogenic factors such as vascular endothelial tion on the mechanisms mediating the long-term increase in
growth factor (VEGF), placental growth factor (PlGF), and vascular resistance and arterial pressure associated with pla-
sFlt-1. Although recent data suggest that circulating sFlt-1 cental ischemia is lacking.
concentrations may presage the clinical onset of preeclamptic Experimental induction of chronic uteroplacental ischemia
symptoms (47, 51, 75, 106), several studies indicate that appears to be a promising animal model to study potential
placental hypoxia and poor placental perfusion may initiate this mechanisms of preeclampsia since reductions in uteroplacental
imbalance of angiogenic factors (61, 71). Nevertheless, it blood flow in a variety of animals lead to a hypertensive state
remains unclear whether impaired placental perfusion initiates that has many of the manifestations present in preeclamptic
preeclamptic symptoms such as hypertension, endothelial dys- women (5–7, 9 –11, 18, 22, 31, 33, 48, 49, 61, 94). It is
function, and increased sFlt-1 or whether inadequate placental important to note that the reduced uterine perfusion pressure
development occurs initially and is followed by a pathological (RUPP) model of placental ischemia-induced hypertension in
rise in sFlt-1 expression and secretion (41). pregnancy most closely relates to severe premature/preterm
Hypertension associated with preeclampsia develops during preeclampsia and not late-onset preeclampsia. The former is
late pregnancy and remits after delivery or termination of the characterized by intrauterine growth restriction, hypertension,
pregnancy, suggesting that the placenta is a central culprit in and proteinuria, whereas the latter is not regarded as mediated
the disease. The foremost hypothesis regarding the initiating by placental ischemia and is usually not associated with intra-
event in preeclampsia is postulated to be reduced placental uterine growth restriction. Thus a considerable strength of the
perfusion that, in turn, leads to widespread dysfunction of the RUPP model is that it allows mechanistic investigation of the
maternal vascular endothelium. Although numerous other fac- preterm ischemic placenta and the factors it elaborates.
tors including genetic, immunological, behavioral, and envi- The relationship between reduced uteroplacental perfusion
ronmental influences have been implicated in the pathogenesis and hypertension during pregnancy has been demonstrated in a
of preeclampsia (95, 96), the main focus of this review is to variety of animals, mostly those with hemochorial placenta-
describe the links between placental ischemia/hypoxia and the tion. Previous studies have shown that chronic RUPP via
cardiovascular dysfunction that is widely recognized as a part partial ligation or placement of silver clips on the lower
of the preeclampsia syndrome. abdominal aorta or uterine arteries results in proteinuric hyper-
Reduced Uterine Perfusion Pressure as a Model to Study the tension in the baboon (18, 19, 61), rhesus monkeys (22, 124),
Pathophysiology of Hypertension During Pregnancy rabbit (5, 59), and the dog (6, 35). Our laboratory has estab-
lished a RUPP model in the pregnant rat (7, 9 –11, 31, 33, 48,
The physiological mechanisms that mediate the alterations 49, 94). In this model, uteroplacental perfusion is reduced by
in cardiovascular and renal function that are requisite for a ⬃40% by the placement of silver clips on the aorta and ovarian
successful pregnancy have been studied in great detail. None- arteries on day 14 of a 21-day gestation (94). On gestation day
AJP-Heart Circ Physiol • VOL 294 • FEBRUARY 2008 • www.ajpheart.org
Downloaded from www.physiology.org/journal/ajpheart by ${individualUser.givenNames} ${individualUser.surname} (036.084.226.018) on February 5, 2018.
Copyright © 2008 American Physiological Society. All rights reserved.
Invited Review
PLACENTAL ISCHEMIA AND CARDIOVASCULAR DYSFUNCTION H543
19, the RUPP animals display increased mean arterial pressure
(MAP), decreased glomerular filtration rate, decreased renal
pressure natriuresis, decreased renal plasma flow and protein-
uria, and endothelial dysfunction (7, 9 –11, 26, 31, 33, 48, 49).
Recently, we demonstrated that these pregnant RUPP rats have
increased total peripheral resistance, decreased cardiac index,
and decreased uteroplacental blood flow (94), indicating that
these animals have marked cardiovascular dysfunction similar
to what is observed in preeclamptic women (11a, 17, 94, 109).
We have also recently shown that RUPP hypertension in the rat
is associated with an imbalance of angiogenic factors, in
particular increased sFlt-1 and decreased VEGF and PlGF (30).
Thus these models of preeclampsia have many of the features
common in preeclamptic women and provide researchers with
a valuable substrate from which to investigate the mechanisms
of hypertension during pregnancy.
The role of a variety of endothelial, autacoid, and hormonal
factors that mediate the cardiovascular and renal dysfunction
produced by chronic reductions in uteroplacental perfusion
pressure will be the primary focus of the remaining portion of
this review. In the present review, we will also focus on the
effects of reductions in uteroplacental perfusion pressure on
vascular endothelial function since factors that emanate from
the ischemic placenta are thought to be responsible for wide-
spread chronic long-term alterations in arterial pressure.

Mediators of Endothelial Dysfunction in Response to


Placental Ischemia
The maternal vascular endothelium appears to be an impor-
tant target of factors that are triggered by placental ischemia/
hypoxia in preeclampsia. The endothelium is a single-cell
lining that covers the luminal side of blood vessels. This
strategic location permits it to signal alterations in hemody-
namics and humoral factors by synthesizing and releasing
vasoactive substances. Thus a critical balance exists between
endothelium-derived relaxing and contracting factors that Fig. 2. Reduced uterine perfusion pressure elicits increased mean arterial
pressure (MAP; A), decreased cardiac index (CI; cardiac output/body weight;
maintain vascular homeostasis. When this delicate balance is B), and increased TPR (C) in late gestation rats. NP, normal pregnant. All data
disrupted, the vasculature is predisposed to vasoconstriction, are expressed as means ⫾ SE. *P ⬍ 0.05. Graph adapted from Sholook et al.
leukocyte adherence, mitogenesis, prooxidation, and vascular (Ref. 94).
inflammation (100, 101). Furthermore, markers of endothelial
dysfunction may serve as predictors of the syndrome in women
that develop preeclampsia since many are often elevated weeks creased in the RUPP rat compared with normal pregnant rats,
before observance of clinical manifestations. shown in Fig. 3 (26). Chronic RUPP in pregnant rats also
Nitric oxide. Substantial evidence indicates that nitric oxide decreases renal protein expression of neuronal NO synthase but
(NO) production is elevated in normal pregnancy and that these not urinary nitrite/nitrate excretion relative to control pregnant
increases appear to play an important role in the renal vasodi- rats (9). Although no difference in urinary nitrite/nitrate excre-
latation of pregnancy (97). In contrast, the role of NO in tion was found between RUPP and control pregnant rats, we
preeclampsia remains enigmatic (25, 76, 91).Taken together, have found that basal and stimulated release of NO from
these studies underscore the need for continued investigation isolated vascular strips were significantly lower in RUPP rats
into the (patho)physiological role of NO in preeclampsia. (13). Although, whether there is a reduction in NO production
Studies from several laboratories indicate that chronic NO in this spontaneous model of pregnancy-induced hypertension
synthase inhibition in pregnant rats produces hypertension remains unclear.
associated with peripheral and renal vasoconstriction, protein- Oxidative stress. During oxidative stress, an imbalance of
uria, intrauterine growth restriction, and increased fetal mor- pro- and antioxidant factors results in endothelial dysfunction,
bidity (23, 43). We have previously reported similar cardio- either by direct actions on the vasculature or through reduc-
vascular perturbations exist in the NO inhibition and RUPP tions in the bioavailability of vasoactive mediators (113).
models, the latter illustrated in Fig. 2 (94). In addition, we have Oxidative stress may mediate endothelial cell dysfunction and
also previously shown that placental ischemia results in the contribute to the pathophysiology of preeclampsia based on
impairment of acetylcholine-mediated vasorelaxation and that evidence of increased prooxidant activity along with decreased
nitrite and nitrate productions in aortic strips are both de- antioxidant protection. During preeclampsia, oxidative stress
AJP-Heart Circ Physiol • VOL 294 • FEBRUARY 2008 • www.ajpheart.org
Downloaded from www.physiology.org/journal/ajpheart by ${individualUser.givenNames} ${individualUser.surname} (036.084.226.018) on February 5, 2018.
Copyright © 2008 American Physiological Society. All rights reserved.
Invited Review
H544 PLACENTAL ISCHEMIA AND CARDIOVASCULAR DYSFUNCTION

hypertension (89), suggesting that there may be other pathways


generating reactive oxygen species in this model. Although
oxidative stress is implicated in the pathogenesis of preeclamp-
sia, it remains unclear whether it is a primary or secondary
mediator of increased blood pressure and deranged renal func-
tion.
Endothelin. Another endothelial-derived factor that may
play a role in preeclampsia is the vasoconstrictor endothelin-1
(ET-1) (63, 67, 102). Since endothelial damage is a known
stimulus for ET-1 synthesis, increases in the production of
endothelin may participate in the pathophysiology of pre-
eclampsia (12). Furthermore, ET-1 is also reported to increase
oxidative stress in placental villi (27). Thus endothelin may
have additional effects on the maternal cardiovascular system
not only by direct actions on the vasculature but also indirectly
via oxidative stress.
Previously, we have investigated the role of endothelin in
mediating RUPP hypertension in conscious, chronically instru-
mented pregnant rats (11). Furthermore, we have shown that
RUPP elicits increased renal cortical and medullary expression
of preproendothelin and that chronic administration of the
selective endothelin type A (ETA) receptor antagonist (ABT-
627, 5 mg䡠kg⫺1 䡠day⫺1 for 10 days) markedly attenuates the
increased mean arterial pressure in these rats (11). In contrast,
ETA receptor blockade had no significant effect on blood
pressure in the normal pregnant animal, suggesting that ET-1
plays an important role in mediating the hypertension produced
by chronic RUPP pregnant rats (11). Furthermore, recent work
in our laboratory has shown that sera from pregnant rats
Fig. 3. Acetylcholine (ACh)-induced relaxation of vascular strips precon-
stricted with phenylephrine (3 ⫻ 10⫺7 mol/l) (A) and the basal and ACh-
exposed to chronic RUPP increase ET-1 production by cul-
induced nitrite/nitrate production (B) in endothelium-intact aortic strips of NP tured endothelial cell and that this increase is mediated via the
and RUPP rats. Adapted from Crews et al. (Ref. 26). AT1 receptor (82). Although the exact mechanism linking
enhanced production of ET-1 to placental ischemia in pregnant
may result from interactions between the maternal component rats or in preeclamptic women remains unknown, possibilities
that may include preexisting conditions such as obesity, dia- include the production of an agonistic AT1-AA and/or in-
betes, and hyperlipidemia and/or the placental component that creased TNF-␣ as we shall describe later.
may involve secretion of lipid peroxides (79). Arachidonic acid metabolites. Several lines of evidence
Several important antioxidants are significantly decreased in suggest that changes in the metabolites of arachidonic acid may
women with preeclampsia. Vitamin C, vitamin A, vitamin E, play a role in mediating the renal dysfunction and increase in
␤-carotene, glutathione levels, and iron-binding capacity are arterial pressure during preeclampsia (114). Significant alter-
lower in the maternal circulation of women with preeclampsia ations in the balance of prostacyclin and thromboxane produc-
than in women with a normal pregnancy. Interestingly, sup- tion occur in women with preeclampsia (46, 68, 114). Thus,
plementation does not appear to ameliorate the incidence of although there is evidence that AA metabolites play a part in
preeclampsia in multicenter clinical trials (73, 83). Reduced preeclampsia, their role is not clearly defined.
superoxide dismutase (SOD) levels and decreased SOD activ- Experimental studies in animals have endeavored to deter-
ity have been reported in neutrophils and placentas of women mine the role of AA metabolites such as thromboxane in
with preeclampsia (113). The decrease in SOD levels and preeclampsia. Such evidence derives from studies indicating
activity in women with preeclampsia is important as diminished that short-term increases in systemic arterial pressure produced
SOD occurs within both the maternal and placental components. by acute RUPP in pregnant dogs can be prevented by throm-
Consequently, there appears to be a decreased total antioxidant boxane receptor antagonism (119). Furthermore, we have pre-
protective capacity in women with preeclampsia. viously reported that urinary excretion of thromboxane B2 was
Experimental data reveal that there is an intimate relation- increased in the RUPP rats compared with normal pregnant rats
ship among reactive oxygen species, NO activity, and blood at day 19 of gestation (56). Additionally, inhibition of cyto-
pressure in rats (85). Thus it is not surprising that there is chrome P-450 enzymes with 1-aminobenzotriazole attenuated
increased oxidative stress in the hypertensive pregnant RUPP the hypertension and increased renal vascular resistance, 20-
rat (87– 89). Whereas antioxidant treatment with vitamins C HETE formation, and cytochrome P-450 4A expression in the
and E does not decrease blood pressure, the SOD mimetic, renal cortex normally observed in the RUPP rat (55). Never-
Tempol, does attenuate RUPP hypertension (88, 89). Similarly, theless, experimental data are limited in this area, and the
apocynin, an NADP(H) oxidase inhibitor, attenuates but does quantitative importance of prostaglandins in mediating long-
not normalize the increased blood pressure observed in RUPP term reduction in renal hemodynamics and increased arterial
AJP-Heart Circ Physiol • VOL 294 • FEBRUARY 2008 • www.ajpheart.org
Downloaded from www.physiology.org/journal/ajpheart by ${individualUser.givenNames} ${individualUser.surname} (036.084.226.018) on February 5, 2018.
Copyright © 2008 American Physiological Society. All rights reserved.
Invited Review
PLACENTAL ISCHEMIA AND CARDIOVASCULAR DYSFUNCTION H545
pressure produced by chronic RUPP in pregnant rats remains
unclear.
Renin-angiotensin system. The renin-angiotensin system
(RAS) plays an important role in the long-term regulation of
renal function and arterial pressure during a variety of physi-
ological and pathophysiological conditions, and pregnancy and
preeclampsia are no exception. During normal pregnancy,
plasma renin concentration, renin activity, and ANG II levels
are all elevated, yet vascular responsiveness to ANG II appears
to be reduced (12, 92). In contrast, during preeclampsia, there
appears to be a marked increase in the sensitivity to ANG II
(92), although the mechanisms underlying these observations
remain unclear.
Recent studies in preeclamptic women have revealed several
exciting findings regarding the RAS. AbdAlla and colleagues
(1, 2) have shown that the AT1 receptor forms heterodimers Fig. 4. Effect of losartan, an AT1 receptor antagonist, on medium endothelin
with the bradykinin B2 receptor and results in enhanced ANG concentration after exposure of human umbilical vein endothelial cells to
II sensitivity. Furthermore, these authors (1, 2) have shown that serum from RUPP rats or NP rats. *P ⬍ 0.05 vs. control pregnant rats. All data
AT1-B2 heterodimers are present in greater abundance in are expressed as means ⫾ SE. Adapted from Roberts et al. (Ref. 82).
preeclamptic women, suggesting that this heterodimerization
may play a part in the long-observed increased ANG II sensi-
tivity in preeclampsia. Another intriguing observation regard- investigated with respect to the manner in which the AT1-AA
ing the involvement of the RAS in the pathophysiology of contribute to the pathophysiology of preeclampsia.
preeclampsia is the demonstration of increased circulating Cytokines. Several groups have suggested that the etiology
concentrations of an agonistic AT1-AA in preeclamptic women of preeclampsia may involve a hypoxia-induced upregulation
(111, 112). Interestingly, the AT1-AA appear to be responsible of placental inflammatory cytokines (24, 78, 116). Although
IL-6 and TNF-␣ are reportedly elevated in preeclamptic
for a variety of effects in several different tissues ranging from
women, the importance of these cytokines in mediating the
increased intracellular Ca2⫹ mobilization to monocyte activa-
cardiovascular and renal dysfunction in response to placental
tion and stimulation of IL-6 production from mesangial cells
ischemia during pregnancy remains unclear. We have previ-
(16, 99, 115, 120). Another effect that has recently been
ously shown that serum levels of TNF-␣ and IL-6 are elevated
attributed to the AT1 receptor is stimulation of sFlt-1 expres-
in RUPP rats and that chronic infusion of TNF-␣ (Fig. 5) or IL-
sion from trophoblast cells but not endothelial cells via cal-
6 into pregnant rats at concentrations similar to what is ob-
cineurin signaling (123). Although these findings potentially served in preeclamptic women increases arterial pressure and
implicate AT1 as a central mediator of several pathways in decreases renal plasma flow and glomerular filtration rate (8,
preeclampsia, both the specific mechanisms that lead to excess 29, 49). Furthermore, we have found that a low-dose infusion
production and the mechanisms whereby AT1-AA increase of TNF-␣ results in decreased renal neuronal NO synthase
blood pressure during pregnancy remain unclear. Conse- expression (8) while also increasing ET-1 mRNA in the kid-
quently, this has become an area of intense interest. ney, placenta, and vasculature (49). Likewise, we have also
Increased vascular responsiveness to ANG II during pre- reported that ET-1 is complicit in the mediation of this form of
eclampsia does not prove it is an important endogenous medi- pregnancy-induced hypertension since the increased MAP in
ator of vasoconstriction or hypertension in experimental mod- response to TNF-␣ is completely abolished in pregnant rats
els of preeclampsia since it could merely reflect low endoge- treated with an ETA receptor antagonist (49). Collectively,
nous ANG II formation. As such, the importance of increased these findings suggest that TNF-␣-induced hypertension in
ANG II to the control of renal function and blood pressure pregnant rats is mediated in part by endothlelin, via ETA
during preeclampsia remains to be determined. Early experi- receptor activation. These studies also suggest that selective
ments in our laboratory indicated that chronic oral administra- ETA receptor antagonists for the treatment of hypertension in
tion of converting enzyme inhibitor enalapril (250 mg/l for 6 women with preeclampsia should receive further attention.
days) decreased MAP to a similar extent in pregnant rats with Another mechanism by which cytokines may contribute to
RUPP and normal pregnant rats, suggesting that the RAS does hypertension during pregnancy is through the modulation of
not play a major role in mediating the hypertension produced sympathetic nerve activity. Previous studies have shown that
by chronic reductions in uterine perfusion pressure in pregnant women with preeclampsia have increased sympathetic tone
rats (7). Nevertheless, we have recently found that AT1 recep- (34, 86). Although few studies have examined the effect of
tor antagonism attenuated the blood pressure response to pla- specific inflammatory cytokines on blood pressure and the
cental ischemia and that RUPP rats have increased circulating regulation of sympathetic activity, one recent experiment has
levels of the AT1-AA (54). As described in Endothelin and demonstrated that an acute forebrain infusion of TNF-␣ in rats
illustrated in Fig. 4, we have also shown that serum from increased arterial pressure, heart rate, and renal sympathetic
pregnant rats exposed to reductions in uterine perfusion en- nerve activity, effects mediated by prostaglandins in the para-
hances endothelin production by endothelial cells via AT1 ventricular nucleus (122). Similarly, another study showed that
receptor activation (82). Although the initial findings by our intracisternal or intravenous infusion of IL-1␤ increases blood
laboratory and others are exciting, there remains much to be pressure in a prostaglandin-dependent manner in rats (121).
AJP-Heart Circ Physiol • VOL 294 • FEBRUARY 2008 • www.ajpheart.org
Downloaded from www.physiology.org/journal/ajpheart by ${individualUser.givenNames} ${individualUser.surname} (036.084.226.018) on February 5, 2018.
Copyright © 2008 American Physiological Society. All rights reserved.
Invited Review
H546 PLACENTAL ISCHEMIA AND CARDIOVASCULAR DYSFUNCTION

resulted in increased arterial pressure and proteinuria and


decreased plasma-free VEGF and PlGF concentrations similar
to that observed in the preeclamptic patients. The authors (66)
also showed that sFlt-1 impaired VEGF- and PlGF-induced
vasorelaxation. Subsequently, similar observations using ade-
novirus transfection have been reported in the mouse (60).
Recently, we have developed a model of increased circulating
sFlt-1 in pregnant rats using recombinant sFlt-1 delivered via
osmotic minipump placed intraperitoneally and found that the
dams are hypertensive, have smaller placentae and fetuses, are
proteinuric, and show evidence of impaired vascular function
on day 18 of gestation (J. P. Granger, unpublished observa-
tions). Although these studies have established the importance
Fig. 5. Changes in MAP in response to TNF-␣ infusion and treatment with an
endothelin type A (ETA) receptor antagonist in NP rats. (*P ⬍ 0.05 vs. NP
of sFlt-1 as an important preeclamptic factor, further studies
rats). All data are expressed as means ⫾ SE. Adapted from LaMarca et al. are needed to elucidate mechanisms governing the expression
(Ref. 49). and actions of this protein.

Whether chronic elevations in inflammatory cytokines contrib-


ute to the increased sympathetic activity during preeclampsia
remains to be determined.
Angiogenic factors. Although VEGF is primarily recognized
for its potent angiogenic and mitogenic effects on endothelial
cells, it has also been recognized as an important contributor to
cell homeostasis, in particular with respect to the balance of
oxidative stress (3, 4). VEGF exerts its actions mainly by two
receptors, VEGF receptor-1 and -2, also known as Flt-1 and the
kinase domain region (Flk/KDR), respectively. A soluble and
endogenously secreted form of Flt-1 is produced mainly in the
placenta by alternative splicing and contains the extracellular
ligand-binding domain but not the transmembrane and cyto-
plasmic portions (21, 44, 45). sFlt-1 disrupts VEGF signaling
either by binding VEGF and PlGF or by forming heterodimers
with the KDR receptor (45). Although sFlt-1 is not a vasocon-
strictor, it does significantly inhibit the dilatory actions of both
VEGF and PlGF in vitro, and chronic elevations in circulating
concentrations result in increased blood pressure (60, 66). An
additional antiangiogenic factor, soluble endoglin (sEng), has
also been revealed as a factor in the pathogenesis of preeclamp-
sia (64, 108). Endoglin is a component of the TGF-␤ receptor
complex and is a hypoxia-inducible protein associated with
cellular proliferation and NO signaling (38, 52). sEng, on the
other hand, has been shown to be antiangiogenic since it is
thought to impair TGF-␤1 binding to cell surface receptors
(38, 108).
Considerable clinical evidence has accumulated that pre-
eclampsia is strongly linked to an imbalance between proan-
giogenic (VEGF and PlGF) and antiangiogenic (sFlt-1) factors
in the maternal circulation (47, 50, 51, 66, 74, 93, 103, 104,
117). Both plasma and amniotic fluid concentrations of sFlt-1
are increased in preeclamptic patients, as well as placental
sFlt-1 mRNA (41, 47, 60, 65, 66, 107, 110). Recently, studies
have reported that increased sFlt-1 may have a predictive value
in diagnosing preeclampsia since concentrations seem to in-
crease before manifestation of overt symptoms (e.g., hyperten-
sion and proteinuria) (47, 51). Similarly, recent clinical evi-
dence also suggests that sEng may also presage the onset of Fig. 6. Plasma concentrations of sFlt-1 (A) were increased in RUPP (n ⫽ 18)
preeclampsia (50). pregnant rats compared with NP (n ⫽ 18) controls at day 19 of pregnancy.
Plasma concentrations of VEGF (B) and PlGF (C) were decreased in RUPP
In an elegantly designed study reported several years ago, (n ⫽ 18) pregnant rats compared with NP (n ⫽ 18) controls at day 19 of
Maynard et al. (66) reported that exogenous administration of pregnancy. All data are expressed as means ⫾ SE. *P ⬍ 0.05. Adapted from
sFlt-1 into pregnant rats via adenovirus-mediated gene transfer Gilbert et al. (Ref. 30).

AJP-Heart Circ Physiol • VOL 294 • FEBRUARY 2008 • www.ajpheart.org


Downloaded from www.physiology.org/journal/ajpheart by ${individualUser.givenNames} ${individualUser.surname} (036.084.226.018) on February 5, 2018.
Copyright © 2008 American Physiological Society. All rights reserved.
Invited Review
PLACENTAL ISCHEMIA AND CARDIOVASCULAR DYSFUNCTION H547
Recently, Li and coworkers (53) showed that VEGF infusion eclampsia rather than resulting from poor placental perfusion
attenuates the increased blood pressure and renal damage (31). Further studies are underway to determine what influence
observed in pregnant rats overexpressing sFlt-1. Thus, from obesity may exert during experimental placental ischemia.
this study, it can be gleaned that sFlt-1 plays a role in the
hypertension and renal dysfunction in preeclampsia; however, Summary
these observations did not shed any light on the matter of Despite being one of the leading causes of maternal death
pathological sFlt-1 overexpression. To this end, we have re- and a major contributor of maternal and perinatal morbidity,
cently demonstrated that uteroplacental ischemia increased the mechanisms responsible for the pathogenesis of preeclamp-
plasma and placental sFlt-1, and this is associated with de- sia remain enigmatic. The initiating event in preeclampsia has
creased VEGF and PlGF in the late gestation pregnant rat (30) been postulated to be reduced uteroplacental perfusion as a
(Fig. 6). Similarly, Makris and colleagues (61) have reported result of abnormal cytotrophoblast invasion of spiral arterioles.
that uteroplacental ischemia increases sFlt-1 in the baboon as Placental ischemia is thought to lead to widespread activation/
well. dysfunction of the maternal vascular endothelium that results
Recent work investigating sEng has furthered progress with in enhanced formation of endothelin, thromboxane, and super-
respect to the role of antiangiogenic factors in preeclampsia oxide, increased vascular sensitivity to ANG II, and decreased
(108). Venkatesha et al. (108) have shown that sEng inhibits formation of vasodilators such as NO and prostacyclin. These
in vitro endothelial cell tube formation to a similar extent as endothelial abnormalities, in turn, cause hypertension by im-
sFlt-1. Furthermore, the authors reported in vivo data in the pairing renal-pressure natriuresis and increasing total periph-
pregnant rat, indicating that an adenovirus-mediated increase eral resistance (summarized in Fig. 1). The quantitative impor-
of sFlt-1 and sEng in concert exacerbated the effects of either tance of the various endothelial and humoral factors in medi-
factor alone and resulted in fetal growth restriction, severe ating vasoconstriction and increased arterial pressure during
hypertension, and nephritic range proteinuria (108). Thus there preeclampsia remains unclear.
is compelling experimental evidence that compliments the clinical Although recent studies support a role for angiogenic fac-
observations that sEng is an important factor in the pathogenesis tors, the AT1-AA, cytokines, and other factors as potential
of preeclampsia. mediators of endothelial dysfunction, finding the link between
Metabolic factors. There are other comorbid conditions that placental ischemia and maternal endothelial and vascular ab-
have been proposed as potential contributors to endothelial normalities remains an important area of investigation. Mi-
dysfunction in preeclampsia. Recent studies have indicated a croarray analysis of genes within the ischemic/hypoxic pla-
relationship between elements of the metabolic syndrome such centa of women with preeclampsia and in animal models of
as elevated serum triglycerides and free fatty acids (40, 57), preeclampsia should provide new insights into novel factors
insulin resistance (14, 36, 62, 90, 118), and glucose intolerance that may provide additional links between placental ischemia/
(39, 98) and the occurrence of preeclampsia. In fact, several hypoxia and hypertension. More effective strategies for the
authors have suggested that insulin resistance may presage the prevention of preeclampsia should be forthcoming once the
manifestation of preeclampsia (98, 118), whereas Thadhani underlying pathophysiological mechanisms that are involved
et al. (105) have proposed that insulin resistance during preg- in preeclampsia are completely understood.
nancy may collude with other conditions such as impaired
angiogenesis to generate a preeclamptic phenotype. GRANTS
Fatty acids may contribute to endothelial dysfunction by This work was supported by National Heart, Lung, and Blood Institute
serving as substrates to generate lipid peroxides that are sig- Grants HL-51971, HL-38499, and HL-90269.
nificantly increased in plasma from women with preeclampsia
(69). Therefore, the generation of free radicals, lipid peroxides, REFERENCES
and reactive oxygen species may be an important mechanism 1. AbdAlla S, Lother H, el Massiery A, Quitterer U. Increased AT1
contributing to endothelial dysfunction in preeclampsia (101). receptor heterodimers in preeclampsia mediate enhanced angiotensin II
Although plasma levels of lipids are increased during normal responsiveness. Nat Med 7: 1003–1009, 2001.
2. AbdAlla S, Lother H, Quitterer U. AT1-receptor heterodimers show
pregnancy, plasma concentrations of both triglyceride-rich li- enhanced G-protein activation and altered receptor sequestration. Nature
poproteins and nonesterified fatty acids are significantly in- 407: 94 –98, 2000.
creased in women that develop preeclampsia relative to normal 3. Abid M, Schoots I, Spokes K, Wu S, Mawhinney C, Aird W. Vascular
pregnant women (57, 58). This significantly increased plasma endothelial growth factor-mediated induction of manganese superoxide
triglycerides in women with preeclampsia correlates with an dismutase occurs through redox-dependent regulation of forkhead and
I␬B/NF-␬B. J Biol Chem 279: 44030 – 44038, 2004.
increased plasma of concentrations low-density lipoproteins 4. Abid M, Tsai J, Spokes K, Deshpande S, Irani K, Aird W. Vascular
(84). The nature of this correlative data has provided difficulty endothelial growth factor induces manganese-superoxide dismutase ex-
in determining a causal effect for abnormal lipid metabolism in pression in endothelial cells by a Rac1-regulated NADPH oxidase-
the pathogenesis of preeclampsia. dependent mechanism. FASEB J 15: 2548 –2550, 2001.
5. Abitbol MM, Gallo GR, Pirani CL, Ober WB. Production of experi-
Because there was no definitive data indicating whether or mental toxemia in the pregnant rabbit. Am J Obstet Gynecol 124:
not metabolic derangements were sequelae or potential con- 460 – 470, 1976.
tributors to placental ischemia, we recently tested this question 6. Abitbol MM, Pirani CL, Ober WB, Driscoll SG, Cohen MW. Pro-
in our RUPP model. Data obtained from the RUPP model duction of experimental toxemia in the pregnant dog. Obstet Gynecol 48:
suggest that metabolic derangements similar to the metabolic 537–548, 1976.
7. Alexander BT, Cockrell K, Cline FD, Llinas MT, Sedeek M, Granger
syndrome X are not a direct consequence of RUPP (31). JP. Effect of angiotensin II synthesis blockade on the hypertensive
Rather, it appears that factors associated with metabolic abnor- response to chronic reductions in uterine perfusion pressure in pregnant
malities may contribute to cardiovascular dysfunction in pre- rats. Hypertension 38: 742–745, 2001.

AJP-Heart Circ Physiol • VOL 294 • FEBRUARY 2008 • www.ajpheart.org


Downloaded from www.physiology.org/journal/ajpheart by ${individualUser.givenNames} ${individualUser.surname} (036.084.226.018) on February 5, 2018.
Copyright © 2008 American Physiological Society. All rights reserved.
Invited Review
H548 PLACENTAL ISCHEMIA AND CARDIOVASCULAR DYSFUNCTION

8. Alexander BT, Cockrell KL, Massey MB, Bennett WA, Granger JP. in the pregnant rat: role of interleukin 6. Hypertension 48: 711–716,
Tumor necrosis factor-alpha-induced hypertension in pregnant rats re- 2006.
sults in decreased renal neuronal nitric oxide synthase expression. Am J 30. Gilbert JS, Babcock SA, Granger JP. Hypertension produced by
Hypertens 15: 170 –175, 2002. reduced uterine perfusion pressure in pregnant rats is associated with
9. Alexander BT, Kassab SE, Miller MT, Abram SR, Reckelhoff JF, increased soluble sFlt-1 expression. Hypertension 50: 1142–1147, 2007.
Bennett WA, Granger JP. Reduced uterine perfusion pressure during 31. Gilbert JS, Dukes M, LaMarca BB, Cockrell K, Babcock SA,
pregnancy in the rat is associated with increases in arterial pressure and Granger JP. Effects of reduced uterine perfusion pressure on blood
changes in renal nitric oxide. Hypertension 37: 1191–1195, 2001. pressure and metabolic factors in pregnant rats. Am J Hypertens 20:
10. Alexander BT, Llinas MT, Kruckeberg WC, Granger JP. L-Arginine 686 – 691, 2007.
attenuates hypertension in pregnant rats with reduced uterine perfusion 32. Granger JP, Alexander BT, Bennett WA, Khalil RA. Pathophysiology
pressure. Hypertension 43: 832– 836, 2004. of pregnancy-induced hypertension. Am J Hypertens 14: 178S–185S,
11. Alexander BT, Rinewalt AN, Cockrell KL, Massey MB, Bennett WA, 2001.
Granger JP. Endothelin type a receptor blockade attenuates the hyper- 33. Granger JP, LaMarca BB, Cockrell K, Sedeek M, Balzi C, Chandler
tension in response to chronic reductions in uterine perfusion pressure. D, Bennett W. Reduced uterine perfusion pressure (RUPP) model for
Hypertension 37: 485– 489, 2001. studying cardiovascular-renal dysfunction in response to placental ischemia.
11a.Anonymous. National High Blood Pressure Education Program Working Methods Mol Med 122: 383–392, 2006.
Group Report on High Blood Pressure in Pregnancy. Am J Obstet 34. Greenwood JP, Scott EM, Stoker JB, Walker JJ, Mary DA. Sympa-
Gynecol 163: 1691–1712, 1990. thetic neural mechanisms in normal and hypertensive pregnancy in
12. August P, Lindheimer M. Pathophysiology of preeclampsia. In: Hyper- humans. Circulation 104: 2200 –2204, 2001.
tension, edited by Laragh J and Brenner BM. New York: Raven, 1995, p. 35. Hodari AA. Chronic uterine ischemia and reversible experimental “toxemia
2407–2426. of pregnancy.” Am J Obstet Gynecol 97: 597– 607, 1967.
13. Barron LA, Giardina JB, Granger JP, Khalil RA. High-salt diet 36. Innes KE, Wimsatt JH, McDuffie R. Relative glucose tolerance and
enhances vascular reactivity in pregnant rats with normal and reduced subsequent development of hypertension in pregnancy. Obstet Gynecol
uterine perfusion pressure. Hypertension 38: 730 –735, 2001. 97: 905–910, 2001.
14. Bartha J, Romero-Carmona R, Torrejon-Cardoso R, Comino-Del- 37. Irgens HU, Reisaeter L, Irgens LM, Lie RT. Long-term mortality of
gado R. Insulin, insulin-like growth factor-1, and insulin resistance in mothers and fathers after pre-eclampsia: population based cohort study.
women with pregnancy-induced hypertension. Am J Obstet Gynecol 187: BMJ 323: 1213–1217, 2001.
735–740, 2002. 38. Jerkic M, Rivas-Elena JV, Prieto M, Carron R, Sanz-Rodriguez F,
15. Bdolah Y, Sukhatme VP, Karumanchi SA. Angiogenic imbalance in Perez-Barriocanal F, Rodriguez-Barbero A, Bernabeu C, Lopez-
the pathophysiology of preeclampsia: newer insights. Semin Nephrol 24: Novoa JM. Endoglin regulates nitric oxide-dependent vasodilatation.
548 –556, 2004. FASEB J 18: 609 – 611, 2004.
16. Bobst SM, Day MC, Gilstrap LC 3rd, Xia Y, Kellems RE. Maternal 39. Joffe G, Esterlitz J, Levine R, Clemens J, Ewell M, Sibai B, Catalano
autoantibodies from preeclamptic patients activate angiotensin receptors P. The relationship between abnormal glucose tolerance and hyperten-
on human mesangial cells and induce interleukin-6 and plasminogen sive disorders of pregnancy in healthy nulliparous women. Am J Obstet
activator inhibitor-1 secretion. Am J Hypertens 18: 330 –336, 2005. Gynecol 179: 1032–1037, 1998.
17. Bosio P, Mckenna P, Conroy R, O’Herlihy C. Maternal central 40. Kaaja R, Tikkanen MJ, Viinikka L, Ylikorkala O. Serum lipopro-
hemodynamics in hypertensive disorders of pregnancy. Obstet Gynecol teins, insulin, and urinary prostanoid metabolites in normal and hyper-
94: 978 –984, 1999. tensive pregnant women. Obstet Gynecol 85: 353–356, 1995.
18. Cavanagh D, Rao PS, Tsai CC, O’Connor TC. Experimental toxemia 41. Karumanchi SA, Bdolah Y. Hypoxia and sFlt-1 in preeclampsia: the
in the pregnant primate. Am J Obstet Gynecol 128: 75– 85, 1977. “chicken-and-egg” question. Endocrinology 145: 4835– 4837, 2004.
19. Cavanagh D, Rao PS, Tung KS, Gaston L. Eclamptogenic toxemia: the 42. Karumanchi SA, Maynard SE, Stillman IE, Epstein FH, Sukhatme
development of an experimental model in the subhuman primate. Am J VP. Preeclampsia: a renal perspective. Kidney Int 67: 2101–2113, 2005.
Obstet Gynecol 120: 183–196, 1974. 43. Kassab S, Miller MT, Hester R, Novak J, Granger JP. Systemic
20. Chambers JC, Fusi L, Malik IS, Haskard DO, De Swiet M, Kooner hemodynamics and regional blood flow during chronic nitric oxide
JS. Association of maternal endothelial dysfunction with preeclampsia. synthesis inhibition in pregnant rats. Hypertension 31: 315–320, 1998.
JAMA 285: 1607–1612, 2001. 44. Kendall RL, Thomas KA. Inhibition of vascular endothelial cell growth
21. Clark DE, Smith SK, He Y, Day KA, Licence DR, Corps AN, factor activity by an endogeneously encoded soluble receptor. Proceed
Lammoglia R, Charnock-Jones DS. A vascular endothelial growth Natl Acad Sci USA 90: 10705–10709, 1993.
factor antagonist is produced by the human placenta and released into the 45. Kendall RL, Wang G, Thomas KA. Identification of a natural soluble
maternal circulation. Biol Reprod 59: 1540 –1548, 1998. form of the vascular endothelial growth factor receptor, FLT-1, and its
22. Combs CA, Katz MA, Kitzmiller JL, Brescia RJ. Experimental heterodimerization with KDR. Biochem Biophys Res Commun 226:
preeclampsia produced by chronic constriction of the lower aorta: vali- 324 –328, 1996.
dation with longitudinal blood pressure measurements in conscious 46. Klockenbusch W, Goecke TW, Krüssel JS, Tutschek BA, Crombach
rhesus monkeys. Am J Obstet Gynecol 169: 215–223, 1993. G, Schrör K. Prostacyclin deficiency and reduced fetoplacental blood
23. Conrad KP. Animal models of pre-eclampsia: do they exist? Fetal flow in pregnancy-induced hypertension and preeclampsia. Gynecol
Medicine Review 2: 67– 88, 1990. Obstet Invest 50: 103–107, 2000.
24. Conrad KP, Benyo DF. Placental cytokines and the pathogenesis of 47. Lam C, Lim KH, Karumanchi SA. Circulating angiogenic factors in
preeclampsia. Am J Reprod Immunol 37: 240 –249, 1997. the pathogenesis and prediction of preeclampsia. Hypertension 46: 1077–
25. Conrad KP, Kerchner LJ, Mosher MD. Plasma and 24-h NOx and 1085, 2005.
cGMP during normal pregnancy and preeclampsia in women on a 48. LaMarca BB, Bennett WA, Alexander BT, Cockrell K, Granger JP.
reduced NOx diet. Am J Physiol Renal Physiol 277: F48 –F57, 1999. Hypertension produced by reductions in uterine perfusion in the pregnant
26. Crews JK, Herrington JN, Granger JP, Khalil RA. Decreased endo- rat: role of tumor necrosis factor-alpha. Hypertension 46: 1022–1025,
thelium-dependent vascular relaxation during reduction of uterine perfu- 2005.
sion pressure in pregnant rat. Hypertension 35: 367–372, 2000. 49. LaMarca BB, Cockrell K, Sullivan E, Bennett W, Granger JP. Role
27. Fiore G, Florio P, Micheli L, Nencini C, Rossi M, Cerretani D, of endothelin in mediating tumor necrosis factor-induced hypertension in
Ambrosini G, Giorgi G, Petraglia F. Endothelin-1 triggers placental pregnant rats. Hypertension 46: 82– 86, 2005.
oxidative stress pathways: putative role in preeclampsia. J Clin Endocri- 50. Levine RJ, Lam C, Qian C, Yu KF, Maynard SE, Sachs BP, Sibai
nol Metab 90: 4205– 4210, 2005. BM, Epstein FH, Romero R, Thadhani R, Karumanchi SA. Soluble
28. Fisher SJ, Roberts JM. Defects in placentation and placental perfusion. endoglin and other circulating antiangiogenic factors in preeclampsia.
In: Chelsey’s Hypertensive Disorders in Pregnancy, edited by Lindhei- N Engl J Med 355: 992–1005, 2006.
mer MD, Roberts JM, and Cunningham FG. Stanford, CT: Appleton & 51. Levine RJ, Maynard SE, Qian C, Lim KH, England LJ, Yu KF,
Lange, 1999, p. 377–394. Schisterman EF, Thadhani R, Sachs BP, Epstein FH, Sibai BM,
29. Gadonski G, LaMarca BB, Sullivan E, Bennett W, Chandler D, Sukhatme VP, Karumanchi SA. Circulating angiogenic factors and the
Granger JP. Hypertension produced by reductions in uterine perfusion risk of preeclampsia. N Engl J Med 350: 672– 683, 2004.

AJP-Heart Circ Physiol • VOL 294 • FEBRUARY 2008 • www.ajpheart.org


Downloaded from www.physiology.org/journal/ajpheart by ${individualUser.givenNames} ${individualUser.surname} (036.084.226.018) on February 5, 2018.
Copyright © 2008 American Physiological Society. All rights reserved.
Invited Review
PLACENTAL ISCHEMIA AND CARDIOVASCULAR DYSFUNCTION H549
52. Li C, Issa R, Kumar P, Hampson IN, Lopez-Novoa JM, Bernabeu C, 73. Poston L, Briley AL, Seed PT, Kelly FJ, Shennan AH. Vitamin C and
Kumar S. CD105 prevents apoptosis in hypoxic endothelial cells. J Cell vitamin E in pregnant women at risk for pre-eclampsia (VIP trial):
Sci 116: 2677–2685, 2003. randomised placebo-controlled trial. Lancet 367: 1145–1154, 2006.
53. Li Z, Zhang Y, Ying Ma J, Kapoun AM, Shao Q, Kerr I, Lam A, 74. Rajakumar A, Michael HM, Rajakumar PA, Shibata E, Hubel CA,
O’Young G, Sannajust F, Stathis P, Schreiner G, Karumanchi SA, Karumanchi SA, Thadhani R, Wolf M, Harger G, Markovic N.
Protter AA, Pollitt NS. Recombinant vascular endothelial growth factor Extra-placental expression of vascular endothelial growth factor recep-
121 attenuates hypertension and improves kidney damage in a rat model tor-1, (Flt-1) and soluble Flt-1 (sFlt-1), by peripheral blood mononuclear
of preeclampsia. Hypertension 50: 686 – 692, 2007. cells (PBMCs) in normotensive and preeclamptic pregnant women.
54. Llinas M, Wallukat G, Dechend R, Mueller DN, Luft FC, Alexander Placenta 26: 563–573, 2005.
BT, LaMarca B, Granger JP. Agonistic autoantibodies to the AT1 75. Rana S, Karumanchi SA, Levine RJ, Venkatesha S, Rauh-Hain JA,
receptor in a rat model of preeclampsia induced by chronic reductions in Tamez H, Thadhani R. Sequential changes in antiangiogenic factors in
uterine perfusion pressure (RUPP) (Abstract). Hypertension 46: 884, early pregnancy and risk of developing preeclampsia. Hypertension 50:
2005. 137–142, 2007.
55. Llinas MT, Alexander BT, Capparelli MF, Carroll MA, Granger JP. 76. Ranta V, Viinikka L, Halmesmaki E, Ylikorkala O. Nitric oxide
Cytochrome P-450 inhibition attenuates hypertension induced by reduc- production with preeclampsia. Obstet Gynecol 93: 442– 445, 1999.
tions in uterine perfusion pressure in pregnant rats. Hypertension 43: 77. Redman CW. Current topic: pre-eclampsia and the placenta. Placenta
623– 628, 2004. 12: 301–308, 1991.
56. Llinas MT, Alexander BT, Seedek M, Abram SR, Crell A, Granger 78. Rinehart BK, Terrone DA, Lagoo-Deenadayalan S, Barber WH,
JP. Enhanced thromboxane synthesis during chronic reductions in uter- Hale EA, Martin JN Jr, Bennett WA. Expression of the placental
ine perfusion pressure in pregnant rats. Am J Hypertens 15: 793–797, cytokines tumor necrosis factor alpha, interleukin 1beta, and interleukin
2002. 10 is increased in preeclampsia. Am J Obstet Gynecol 181: 915–920,
57. Lorentzen B, Drevon CA, Endresen MJ, Henriksen T. Fatty acid 1999.
pattern of esterified and free fatty acids in sera of women with normal and 79. Roberts JM, Taylor RN, Goldfien A. Clinical and biochemical evi-
pre-eclamptic pregnancy. Br J Obstet Gynaecol 102: 530 –537, 1995. dence of endothelial cell dysfunction in the pregnancy syndrome pre-
58. Lorentzen B, Henriksen T. Plasma lipids and vascular dysfunction in eclampsia. Am J Hypertens 4: 700 –708, 1991.
preeclampsia. Semin Reprod Endocrinol 16: 33–39, 1998. 80. Roberts JM, Taylor RN, Musci TJ, Rodgers GM, Hubel CA,
59. Losonczy G, Brown G, Venuto RC. Increased peripheral resistance McLaughlin MK. Preeclampsia: an endothelial cell disorder. Am J
during reduced uterine perfusion pressure hypertension in pregnant Obstet Gynecol 161: 1200 –1204, 1989.
rabbits. Am J Med Sci 303: 233–240, 1992. 81. Roberts JM, Pearson G, Cutler J, Lindheimer M. Summary of the
60. Lu F, Longo M, Tamayo E, Maner W, Al-Hendy A, Anderson GD, NHLBI working group on research on hypertension during pregnancy.
Hankins GD, Saade GR. The effect of over-expression of sFlt-1 on Hypertension 41: 437– 445, 2003.
blood pressure and the occurrence of other manifestations of preeclamp- 82. Roberts L, LaMarca BB, Fournier L, Bain J, Cockrell K, Granger
JP. Enhanced endothelin synthesis by endothelial cells exposed to sera
sia in unrestrained conscious pregnant mice. Am J Obstet Gynecol 196:
from pregnant rats with decreased uterine perfusion. Hypertension 47:
e1– e7, 2007.
615– 618, 2006.
61. Makris A, Thornton C, Thompson J, Thomson S, Martin R, Ogle R,
83. Rumbold AR, Crowther CA, Haslam RR, Dekker GA, Robinson JS,
Waugh R, McKenzie P, Kirwan P, Hennessy A. Uteroplacental isch-
the Study Group ACTS. Vitamins C and E and the risks of preeclamp-
emia results in proteinuric hypertension and elevated sFLT-1. Kidney Int
sia and perinatal complications. N Engl J Med 354: 1796 –1806, 2006.
71: 977–984, 2007.
84. Sattar N, Greer IA, Louden J, Lindsay G, McConnell M, Shepherd
62. Martinez Abundis E, Gonzalez Ortiz M, Quiñones Galvan A, Fer-
J, Packard CJ. Lipoprotein subfraction changes in normal pregnancy:
rannini E. Hyperinsulinemia in glucose-tolerant women with preeclamp-
threshold effect of plasma triglyceride on appearance of small, dense low
sia. A controlled study. Am J Hypertens 9: 610 – 614, 1996. density lipoprotein. J Clin Endocrinol Metab 82: 2483–2491, 1997.
63. Mastrogiannis DS, O’Brien WF, Krammer J, Benoit R. Potential role 85. Schnackenberg CG, Welch WJ, Wilcox CS. Normalization of blood
of endothelin-1 in normal and hypertensive pregnancies. Am J Obstet pressure and renal vascular resistance in shr with a membrane-permeable
Gynecol 165: 1711–1716, 1991. superoxide dismutase mimetic: role of nitric oxide. Hypertension 32:
64. Masuyama H, Nakatsukasa H, Takamoto N, Hiramatsu Y. Correla- 59 – 64, 1998.
tion between soluble endoglin, vascular endothelial growth factor recep- 86. Schobel HP, Fischer T, Heuszer K, Geiger H, Schmieder RE. Pre-
tor-1, and adipocytokines in preeclampsia. J Clin Endocrinol Metab 92: eclampsia—a state of sympathetic overactivity. N Engl J Med 335:
2672–2679, 2007. 1480 –1485, 1996.
65. Maynard SE, Venkatesha S, Thadhani R, Karumanchi SA. Soluble 87. Sedeek MH, Alexander BT, Sholook MM, Chandler DL, Abram SR,
Fms-like tyrosine kinase 1 and endothelial dysfunction in the pathogen- Granger JP. Renal cortical NADPH oxidase and superoxide dismutase
esis of preeclampsia. Pediatr Res 57: 1R–7R, 2005. activity in response to reduced uterine perfusion pressure in pregnant rats
66. Maynard SE, Min JY, Merchan J, Lim KH, Li J, Mondal S, (Abstract). FASEB J 18: A740, 2004.
Libermann TA, Morgan JP, Sellke FW, Stillman IE, Epstein FH, 88. Sedeek MH, Sholook MM, Blazli C, Abram SR, Chandler DL,
Sukhatme VP, Karumanchi SA. Excess placental soluble fms-like Granger JP. Tempol, but not vitamin E & C, decreases the blood
tyrosine kinase 1 (sFlt1) may contribute to endothelial dysfunction, pressure response to a chronic reduction in uterine perfusion pressure in
hypertension, and proteinuria in preeclampsia. J Clin Invest 111: 649 – pregnant rats (Abstract). FASEB J 18: A740, 2004.
658, 2003. 89. Sedeek MH, Wang YP, Granger JP. Increased oxidative stress in a rat
67. McMahon LP, Redman CW, Firth JD. Expression of the three endo- model of preeclampsia (Abstract). Am J Hypertens 17: 142A, 2004.
thelin genes and plasma levels of endothelin in pre-eclamptic and normal 90. Seely EW, Solomon CG. Insulin resistance and its potential role in
gestations. Clin Sci (Lond) 85: 417– 424, 1993. pregnancy-induced hypertension. J Clin Endocrinol Metab 88: 2393–
68. Mills JL, DerSimonian R, Raymond E, Morrow JD, Roberts LJ, 2398, 2003.
Clemens JD, Hauth JC, Catalano P, Sibai B, Curet LB, Levine RJ. 91. Shaamash AH, Elsnosy ED, Makhlouf AM, Zakhari MM, Ibrahim
Prostacyclin and thromboxane changes predating clinical onset of pre- OA, El-Dien HM. Maternal and fetal serum nitric oxide (NO) concen-
eclampsia: a multicenter prospective study. JAMA 282: 356 –362, 1999. trations in normal pregnancy, pre-eclampsia and eclampsia. Int J Gynaecol
69. Murai JT, Muzykanskiy E, Taylor RN. Maternal and fetal modulators Obstet 68: 207–214, 2000.
of lipid metabolism correlate with the development of preeclampsia. 92. Shah DM. Role of the renin-angiotensin system in the pathogenesis of
Metabolism 46: 963–967, 1997. preeclampsia. Am J Physiol Renal Physiol 288: F614 –F625, 2005.
71. Nevo O, Soleymanlou N, Wu Y, Xu J, Kingdom J, Many A, Zamudio 93. Shibata E, Rajakumar A, Powers RW, Larkin RW, Gilmour C,
S, Caniggia I. Increased expression of sFlt-1 in in vivo and in vitro Bodnar LM, Crombleholme WR, Ness RB, Roberts JM, Hubel CA.
models of human placental hypoxia is mediated by HIF-1. Am J Physiol Soluble fms-like tyrosine kinase 1 is increased in preeclampsia but not in
Regul Integr Comp Physiol 291: R1085–R1093, 2006. normotensive pregnancies with small-for-gestational-age neonates: rela-
72. Podjarny E, Losonczy G, Baylis C. Animal models of preeclampsia. tionship to circulating placental growth factor. J Clin Endocrinol Metab
Semin Nephrol 24: 596 – 606, 2004. 90: 4895– 4903, 2005.

AJP-Heart Circ Physiol • VOL 294 • FEBRUARY 2008 • www.ajpheart.org


Downloaded from www.physiology.org/journal/ajpheart by ${individualUser.givenNames} ${individualUser.surname} (036.084.226.018) on February 5, 2018.
Copyright © 2008 American Physiological Society. All rights reserved.
Invited Review
H550 PLACENTAL ISCHEMIA AND CARDIOVASCULAR DYSFUNCTION

94. Sholook MM, Gilbert JS, Sedeek MH, Huang M, Hester RL, 109. Visser W, Wallenburg HC. Central hemodynamic observations in
Granger JP. Systemic hemodynamic and regional blood flow changes in untreated preeclamptic patients. Hypertension 17: 1072–1077, 1991.
response to chronic reductions in uterine perfusion pressure in pregnant 110. Vuorela P, Helske S, Hornig C, Alitalo K, Weich H, Halmesmaki E.
rats. Am J Physiol Heart Circ Physiol 293: H2080 –H2084, 2007. Amniotic fluid-soluble vascular endothelial growth factor receptor-1 in
95. Sibai B, Dekker G, Kupferminc M. Pre-eclampsia. Lancet 365: 785– preeclampsia. Obstet Gynecol 95: 353–357, 2000.
799, 2005. 111. Wallukat G, Homuth V, Fischer T, Lindschau C, Horstkamp B,
96. Sibai B, Ewell M, Levine R, Klebanoff M, Esterlitz J, Catalano P, Jupner A, Baur E, Nissen E, Vetter K, Neichel D, Dudenhausen JW,
Goldenberg R, Joffe G. Risk factors associated with preeclampsia in Haller H, Luft FC. Patients with preeclampsia develop agonistic auto-
healthy nulliparous women. Am J Obstet Gynecol 177: 1003–1010, 1997. antibodies against the angiotensin AT1 receptor. J Clin Invest 103:
97. Sladek SM, Magness RR, Conrad KP. Nitric oxide and pregnancy. 945–952, 1999.
Am J Physiol Regul Integr Comp Physiol 272: R441–R463, 1997. 112. Wallukat G, Neichel D, Nissen E, Homuth V, Luft FC. Agonistic
98. Solomon CG, Graves SW, Greene MF, Seely EW. Glucose intolerance autoantibodies directed against the angiotensin II AT1 receptor in patients
as a predictor of hypertension in pregnancy. Hypertension 23: 717–721, with preeclampsia. Can J Physiol Pharmacol 81: 79 – 83, 2003.
1994. 113. Walsh SW. Maternal-placental interactions of oxidative stress and anti-
99. Stepan H, Faber R, Wessel N, Wallukat G, Schultheiss HP, Walther oxidants in preeclampsia. Semin Reprod Endocrinol 16: 93–104, 1998.
T. Relation between circulating angiotensin II type 1 receptor agonistic 114. Walsh SW. Eicosanoids in preeclampsia. Prostaglandins Leukot Essent
autoantibodies and soluble fms-like tyrosine kinase 1 in the pathogenesis Fatty Acids 70: 223–232, 2004.
of preeclampsia. J Clin Endocrinol Metab 91: 2424 –2427, 2006. 115. Walther T, Wallukat G, Jank A, Bartel S, Schultheiss HP, Faber R,
100. Taylor RN, Roberts JM. Endothelial cell dysfunction. In: Chelsey’s Stepan H. Angiotensin II type 1 receptor agonistic antibodies reflect
Hypertensive Disorder’s of Pregnancy, edited by Lindheimer M, Roberts fundamental alterations in the uteroplacental vasculature. Hypertension
JM, and Cunningham FG. Stanford, CT: Appleton & Lange, 2007, p. 46: 1275–1279, 2005.
395– 429. 116. Wang Y, Walsh SW. TNF alpha concentrations and mRNA expression
101. Taylor RN, de Groot CJ, Cho YK, Lim KH. Circulating factors as are increased in preeclamptic placentas. J Reprod Immunol 32: 157–169,
markers and mediators of endothelial cell dysfunction in preeclampsia. 1996.
Semin Reprod Endocrinol 16: 17–31, 1998. 117. Wolf M, Shah A, Lam C, Martinez A, Smirnakis KV, Epstein FH,
102. Taylor RN, Varma M, Teng NN, Roberts JM. Women with pre- Taylor RN, Ecker JL, Karumanchi SA, Thadhani R. Circulating
eclampsia have higher plasma endothelin levels than women with normal levels of the antiangiogenic marker sFLT-1 are increased in first versus
pregnancies. J Clin Endocrinol Metab 71: 1675–1677, 1990. second pregnancies. Am J Obstet Gynecol 193: 16 –22, 2005.
103. Thadhani R, Mutter WP, Wolf M, Levine RJ, Taylor RN, Sukhatme 118. Wolf M, Sandler L, Munoz K, Hsu K, Ecker JL, Thadhani R. First
VP, Ecker J, Karumanchi SA. First trimester placental growth factor trimester insulin resistance and subsequent preeclampsia: a prospective
and soluble fms-like tyrosine kinase 1 and risk for preeclampsia. J Clin study. J Clin Endocrinol Metab 87: 1563–1568, 2002.
Endocrinol Metab 89: 770 –775, 2004. 119. Woods LL. Importance of prostaglandins in hypertension during reduced
104. Thadhani RI, Johnson RJ, Karumanchi SA. Hypertension during uteroplacental perfusion pressure. Am J Physiol Regul Integr Comp
pregnancy: a disorder begging for pathophysiological support. Hyperten- Physiol 257: R1558 –R1561, 1989.
sion 46: 1250 –1251, 2005. 120. Xia Y, Wen H, Bobst S, Day MC, Kellems RE. Maternal autoantibod-
105. Thadhani R, Ecker JL, Mutter WP, Wolf M, Smirnakis KV, ies from preeclamptic patients activate angiotensin receptors on human
Sukhatme VP, Levine RJ, Karumanchi SA. Insulin resistance and trophoblast cells. J Soc Gynecol Investig 10: 82–93, 2003.
alterations in angiogenesis: additive insults that may lead to preeclamp- 121. Ye S, Mozayeni P, Gamburd M, Zhong H, Campese VM. Interleukin-
sia. Hypertension 43: 988 –992, 2004. 1beta and neurogenic control of blood pressure in normal rats and rats
106. Tranquilli AL, Bezzeccheri V, Giannubilo SR, Scagnoli C, Mazzanti with chronic renal failure 1. Am J Physiol Heart Circ Physiol 279:
L, Garzetti GG. Amniotic vascular endothelial growth factor (VEGF) H2786 –H2796, 2000.
and nitric oxide (NO) in women with subsequent preeclampsia. Eur J 122. Zhang ZH, Wei SG, Francis J, Felder RB. Cardiovascular and renal
Obstet Gynecol Reprod Biol 113: 17–20, 2004. sympathetic activation by blood-borne TNF-alpha in rat: the role of
107. Tsatsaris V, Goffin F, Munaut C, Brichant JF, Pignon MR, Noel A, central prostaglandins 1. Am J Physiol Regul Integr Comp Physiol 284:
Schaaps JP, Cabrol D, Frankenne F, Foidart JM. Overexpression of R916 –R927, 2003.
the soluble vascular endothelial growth factor receptor in preeclamptic 123. Zhou CC, Ahmad S, Mi T, Xia L, Abbasi S, Hewett PW, Sun C,
patients: pathophysiological consequences. J Clin Endocrinol Metab 88: Ahmed A, Kellems RE, Xia Y. Angiotensin II induces soluble fms-like
5555–5563, 2003. tyrosine kinase-1 release via calcineurin signaling pathway in pregnancy.
108. Venkatesha S, Toporsian M, Lam C, Hanai J, Mammoto T, Kim Circ Res 100: 88 –95, 2007.
YM, Bdolah Y, Lim KH, Yuan HT, Libermann TA, Stillman IE, 124. Zhou Y, Chiu K, Brescia RJ, Combs CA, Katz MA, Kitzmiller JL,
Roberts D, D’Amore PA, Epstein FH, Sellke FW, Romero R, Heilbron DC, Fisher SJ. Increased depth of trophoblast invasion after
Sukhatme VP, Letarte M, Karumanchi SA. Soluble endoglin contrib- chronic constriction of the lower aorta in rhesus monkeys. Am J Obstet
utes to the pathogenesis of preeclampsia. Nat Med 12: 642– 649, 2006. Gynecol 169: 224 –229, 1993.

AJP-Heart Circ Physiol • VOL 294 • FEBRUARY 2008 • www.ajpheart.org


Downloaded from www.physiology.org/journal/ajpheart by ${individualUser.givenNames} ${individualUser.surname} (036.084.226.018) on February 5, 2018.
Copyright © 2008 American Physiological Society. All rights reserved.

Potrebbero piacerti anche