Skip to main content
British Journal of Pharmacology logoLink to British Journal of Pharmacology
. 2015 Feb 27;172(6):1574–1586. doi: 10.1111/bph.12977

Unravelling the theories of pre-eclampsia: are the protective pathways the new paradigm?

Asif Ahmed 1,, Wenda Ramma 2
PMCID: PMC4354257  PMID: 25303561

Abstract

Pre-eclampsia is a vascular disorder of pregnancy where anti-angiogenic factors, systemic inflammation and oxidative stress predominate, but none can claim to cause pre-eclampsia. This review provides an alternative to the ‘two-stage model’ of pre-eclampsia in which abnormal spiral arteries modification leads to placental hypoxia, oxidative stress and aberrant maternal systemic inflammation. Very high maternal soluble fms-like tyrosine kinase-1 (sFlt-1 also known as sVEGFR) and very low placenta growth factor (PlGF) are unique to pre-eclampsia; however, abnormal spiral arteries and excessive inflammation are also prevalent in other placental disorders. Metaphorically speaking, pregnancy can be viewed as a car with an accelerator and brakes, where inflammation, oxidative stress and an imbalance in the angiogenic milieu act as the ‘accelerator’. The ‘braking system’ includes the protective pathways of haem oxygenase 1 (also referred as Hmox1 or HO-1) and cystathionine-γ-lyase (also known as CSE or Cth), which generate carbon monoxide (CO) and hydrogen sulphide (H2S) respectively. The failure in these pathways (brakes) results in the pregnancy going out of control and the system crashing. Put simply, pre-eclampsia is an accelerator–brake defect disorder. CO and H2S hold great promise because of their unique ability to suppress the anti-angiogenic factors sFlt-1 and soluble endoglin as well as to promote PlGF and endothelial NOS activity. The key to finding a cure lies in the identification of cheap, safe and effective drugs that induce the braking system to keep the pregnancy vehicle on track past the finishing line.

Linked Articles

This article is part of a themed section on Pharmacology of the Gasotransmitters. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2015.172.issue-6

Tables of Links

TARGETS
Nuclear hormone receptorsa2004
PPAR-γ
Catalytic receptorsb2004
VEGFR-1
VEGFR-2
Enzymesc2004
Akt (PKB)
Arginase
CSE
CBS
HO-1
HO-2
MPST
NOS1 (neuronal NOS)
NOS2 (inducible NOS)
NOS3 (endothelial NOS)
LIGANDS
Aspirin IL-6
Asymmetric dimethylarginine (ADMA) L-arginine
BH4 L-NAME
Bilirubin Nitric oxide (NO)
Biliverdin Pravastatin
Bradykinin Sildenafil
cGMP TNF-α
Cystathionine TxA2
Cysteine VCAM-1
DL-propargylglycine VEGF-A
Endothelin-1 (ET-1) Vitamin C
Fibronectin Von Willebrand factor
Homocysteine

These Tables list key protein targets and ligands in this article which are hyperlinked to corresponding entries in http://www.guidetopharmacology.org, the common portal for data from the IUPHAR/BPS Guide to PHARMACOLOGY (Pawson et al., 2014) and are permanently archived in the Concise Guide to PHARMACOLOGY 2013/14 (a,b,cAlexander et al., 2013a,b,c).

Introduction

Pre-eclampsia is a major cause of maternal death worldwide (Lowe et al., 2009). It is characterized by the onset of new hypertension with proteinuria after 20 weeks of gestation. There are no effective pharmacological agents to treat pre-eclampsia. The only solution is the premature termination of the pregnancy. Although maternal symptoms appear to be largely resolved with the delivery of the baby, a growing body of evidence indicates that pre-eclampsia is associated with long-term health issues for both mother and baby (Smith et al., 2001; Bellamy et al., 2007; MacDonald et al., 2007). The human placenta is central to the development of pre-eclampsia. The incidence of pre-eclampsia increases as pregnancy proceeds from singleton to twin, triplets and quadruplets as the mass of placenta increases. Pre-eclampsia also occurs in molar pregnancies further indicating that it is the placenta and not the fetus that causes the condition. This condition is still being debated as a ‘disease of theories’ as we are entering into the second half of the second decade of the 21st century. In this review, we hope to shed new light and provide a clear steer where researchers need to focus if we are to find a cure for this elusive disorder in the first quarter of this century.

Re-evaluation of existing theories of pre-eclampsia

Abnormal spiral artery remodelling was first postulated over five decades ago (Brosens, 1964; 1977) and has been accepted as the underlying cause of pre-eclampsia (Khong et al., 1986; Feinberg et al., 1991; Brosens et al., 2002; Burton et al., 2009). Referred to as the two-stage process (Redman, 1992; Redman and Sargent, 2005; Roberts and Hubel, 2009), for over two decades, most researchers have argued that the development of pre-eclampsia stems from abnormal spiral artery modification leading to placental hypoxia, increase in oxidative stress and aberrant maternal systemic inflammatory responses (Naljayan and Karumanchi, 2013).

Recent studies have shown that such defects are not specific to pre-eclampsia (Lyall et al., 2013), and are also associated with placental abruption, preterm premature rupture of membranes and intrauterine fetal death (Avagliano et al., 2011), indicating that abnormal spiral artery remodelling may be a common underlying contributing factor for abnormal placentation, but is not specific to pre-eclampsia. Additionally, the immune-deficient Rag2−/−c−/− double-knockout mice that lack spiral artery remodelling exhibit no pre–eclampsia-like phenotype. Burke et al. concluded that neither gestational hypertension nor deficient placental growth was an outcome of impaired spiral artery remodelling (Burke et al., 2010). Moreover, pre-eclampsia may occur in twin pregnancy despite normal uterine artery velocity waveform (Rizzo et al., 1993). Finally, a meta-analysis on the performance of first trimester uterine artery Doppler showed similar predictive accuracy for pre-eclampsia and other complications such as stillbirths and abruption (Velauthar et al., 2014). The test had high specificity, but low sensitivity, and this pattern of performance was noticed across various conditions including pre-eclampsia. This once again indicates that the shallow trophoblastic invasion in the spiral arteries resulting in progressive utero-placental ischaemia may relate to a more generalized remodelling pathology and is not the initiator of pre-eclampsia per se (Burke and Karumanchi, 2013).

The concept that abnormal spiral artery remodelling leading to hypoxia to support the two-stage hypothesis is further challenged by other sets of data. Huppertz and colleagues report that failure of endovascular trophoblast invasion does not lead to hypoxia. Rather, all measurements available to date point to increased oxygen levels within the placenta in patients with a failure of spiral artery transformation (Huppertz et al., 2014). Moreover, elevated soluble fms-like tyrosine kinase 1 (sVEGFR-1 also known as sFlt-1) from pre-eclamptic placenta is unlikely to be due to hypoxia per se as pre-eclamptic placenta continues to generate substantially higher levels of sVEGFR-1, even after 24 h of culture ex vivo under atmospheric condition as compared with normal placental explants under identical experimental conditions (Ahmad and Ahmed, 2004). If we accept that the clinical signs of pre-eclampsia are due, in large part, to elevated levels of sVEGFR-1, these findings challenge the long held belief that pre-eclampsia arises because of placental hypoxia. Based on these studies, we argue that the failed remodelling of maternal spiral arteries leading to hypoxia has been misconceived, for many years, as the cause of pre-eclampsia.

Another theory that has long been adopted without convincing clinical evidence is the idea that an elevation in maternal systemic inflammation is the cause of pre-eclampsia (Redman et al., 1999). Although studies in rodents support the systemic maternal inflammatory response and placental ischaemia hypothesis (LaMarca et al., 2007; Cotechini et al., 2014), this is not supported by human cytokine data. In human pregnancy, the elevation in pro-inflammatory status does not seem to precede the onset of pre-eclampsia (Djurovic et al., 2002; Kronborg et al., 2011; Carty et al., 2012). Furthermore, it is not associated with the severity of the disorder as there is no observed significant difference among serum TNF-α and IL-6 levels in patients with mild pre-eclampsia, severe pre-eclampsia and hemolysis, elevated liver enzymes, low platelets syndrome in pre-eclampsia (Ozler et al., 2012). Moreover, pregnant women with elevated systemic inflammation had increased IL-6 levels, but normal angiogenic status without exhibiting symptoms of hypertension or proteinurea (Ramma et al., 2012). More importantly, a prospective longitudinal study of 2600 women with singleton pregnancies who went on to develop pre-eclampsia using multiplex analysis of inflammatory markers and cytokines showed that there was no increase in any of the cytokines before the onset of pre-eclampsia (Carty et al., 2012). In addition, antenatal corticosteroid treatment demonstrated that inflammation is unlikely to be the major contributor to severe pre-eclampsia or useful for therapeutic targeting (Nayeri et al., 2014). These findings weaken the hypothesis that inflammation is the cause of pre-eclampsia.

Is endothelial activation the central phenomenon?

Maternal endothelial dysfunction is the central phenomenon responsible for the clinical signs of pre-eclampsia. Soluble E-selectin, a marker of endothelial cell activation, was found to be significantly higher at 12–16 weeks gestation in women who subsequently developed pre-eclampsia (Carty et al., 2012). One of the theories postulated to be responsible for maternal endothelial dysfunction is the excessive production of free radicals (Wickens et al., 1981; Tsukatani, 1983; Roberts et al., 1989; Davidge, 1998; Hubel, 1999). This theory led obstetricians and scientists to advocate that maternal supplementation with low-dose aspirin or antioxidants could reduce oxidative stress (Wallenburg et al., 1986; Schiff et al., 1990; Kharb, 2000; Poston et al., 2006). These studies were undertaken without clear mechanistic evidence as daily intake of vitamin C, for just 6 weeks, increases the plasma level of 8-oxoadenine, a marker for DNA damage mediated by oxygen radicals (Podmore et al., 1998). Subsequent extensive systematic reviews with meta-analysis revealed that there is little evidence to support the administration of low-dose aspirin or vitamins C and E for either high- or low-risk women to prevent pre-eclampsia (Rossi and Mullin, 2011; Villa et al., 2013). There may be some benefit in the reduction of perinatal death and other adverse perinatal outcomes of low-dose aspirin initiated before 16 weeks of gestation (Roberge et al., 2013).

In the last decade, the hypothesis that pre-eclampsia arise because of the loss of VEGF activity as a result of the ‘increase in the levels of endogenous soluble (s)VEGFR-1 (also known as sFlt-1) that may antagonize the beneficial effects of VEGF’ (Ahmed, 1997), has been validated. A number of studies have consistently shown that the imbalance in anti-angiogenic factors is most strongly associated with the clinical signs of pre-eclampsia and disease severity (Chappell et al., 2002; 2013; Maynard et al., 2003; Ahmad and Ahmed, 2004; Buhimschi et al., 2006; Crispi et al., 2006; Levine et al., 2006; Venkatesha et al., 2006; Ramma et al., 2012). Anti-angiogenic factors, sVEGFR-1 and soluble endoglin (sEng, also known as CD105) are increased before the clinical onset of pre-eclampsia (Levine et al., 2006), and pregnant rodents exposed to high circulating levels of sVEGFR-1 illicit severe pre–eclampsia-like symptoms (Ramma and Ahmed, 2011; Rana et al., 2014).

Metaphorical view of pre-eclampsia

Metaphorically speaking pregnancy can be seen as a car with an accelerator and brakes, where inflammation, oxidative stress and an imbalance in the angiogenic milieu act as ‘accelerators’. It is the failure in the braking systems, the endogenous protective pathway, that result in the ‘accelerator’ going out of control until the system crashes, manifesting itself as pre-eclampsia (http://youtube/vWUCWbKo1dE). The identification of the braking system and how to enhance the brakes when the system starts to fail can restore balance and possibly cure pre-eclampsia. The strategy to identify a cure for pre-eclampsia needs to be centred on identifying cytoprotective pathways, which reduce sVEGFR-1, sEng, oxidative stress and inflammation. The study of enzyme systems, which generate gasotransmitters, has the potential to lead to the discovery of effective treatments for pre-eclampsia. The trial that uses pravastatin to ameliorate early-onset pre-eclampsia (StAmP) is the first such approach underway in the UK; similarly other clinical trials are now taking place in the USA and Australia.

Endogenous cytoprotective pathway

While most laboratories focused on finding the ‘accelerators’ of pre-eclampsia, our laboratory switched our efforts to identify key endogenous protective pathways; the control switch for the ‘accelerators’. The first ‘brake’ system identified was the haem oxygenase (HO)/carbon monoxide (CO) pathway (Ahmed et al., 2000). Recently, another ‘brake’ system discovered was the cystathionine-γ-lyase (CSE also known as Cth) (Wang et al., 2013), which produces hydrogen sulphide (H2S) (Bir and Kevil, 2013). These protective pathways inhibit sVEGFR-1 and sEng release (Cudmore et al., 2007; Wang et al., 2013) making them good candidates for the ‘brake’ theory outlined in Figures 1 and 2.

Figure 1.

Figure 1

Effect of defective protective pathways in pre-eclampsia. This diagram illustrates that the defect in HO-1 (also referred to as Hmox) and CSE (also known as Cth), which generates signalling molecules, CO and H2S, and which results in an increase in sVEGFR-1 and sEng as well as a decrease in PlGF production. These are key factors responsible for vascular dysfunction in pre-eclampsia.

Figure 2.

Figure 2

The accelerator and brake theory of pre-eclampsia. Schematic diagram to illustrate the sequence of events involved in the pathogenesis of pre-eclampsia. The upstream events consist of dysregulation of endogenous protective pathways – ‘the brakes’ – [CSE which generates H2S and HO-1 that produces CO] leading to maternal endothelial activation. As a consequence, there is an increase in anti-angiogenic factors – ‘the accelerator’ – (sVEGFR-1, sEng and soluble E-slectin and a decrease in angiogenic factors PlGF and eNOS, which generates NO). These biochemical changes lead to a generalized endothelial dysfunction, renal injury and generation of reactive oxygen species, which precedes the clinical onset of pre-eclampsia. After 20 weeks of gestation, the clinical symptoms manifest themselves as high BP and proteinurea, which are concurrent with excessive inflammation as indicated by increase in pro-inflammatory cytokines (Th1 cytokine production) and ET-1 release.

Gasotransmitters

Gaseous signalling molecules, such as NO, CO and H2S, have all been hailed as promising molecules with clinical therapeutic potential largely because of their ability to act as vasodilators (Moncada, 1997; Dulak et al., 2008; Bir and Kevil, 2013). The enzyme systems that generate NO, CO and H2S are able to promote placental vasodilatation by regulating placental blood vessel tone in vitro and in vivo (Gude et al., 1990; Myatt et al., 1991; Gonzalez et al., 1995; Learmont and Poston, 1996; Odrcich et al., 1998; Ahmed et al., 2000; Bainbridge et al., 2002; Zhao et al., 2008; Patel et al., 2009; Holwerda et al., 2012; Cindrova-Davies et al., 2013; Wang et al., 2013). The discovery that NO, CO or H2S can promote placental blood flow is not surprising as these molecules have been shown to do this in a number of other vascular beds (Mustafa et al., 2009; Coletta et al., 2012). This property of these gaseous molecules by themselves is of limited therapeutic value in suppressing pre-eclampsia as illustrated by numerous NO studies (Johal et al., 2014). However, it is the additional properties of CO and H2S that makes them an attractive proposition in the search to identify therapies that would prevent pre-eclampsia.

HO/CO system

HO is the rate-limiting enzyme responsible for the degradation of haem in the endoplasmic reticulum to generate equimolar amounts of biliverdin, free iron and CO (Tenhunen et al., 1969). Biliverdin is rapidly reduced to bilirubin, a potent antioxidant, by the cytosolic enzyme biliverdin reductase. CO is a potent vasodilator and also has anti-apoptotic properties. HO exists in two main isoforms, HO-1 and HO-2. The enzyme HO-2 is a 36 kDa protein that is constitutively expressed at high concentrations in the brain, testis and vascular endothelium. The inducible form, HO-1 is a 32 kDa protein that is widely distributed in the body, with high concentrations in the liver and the spleen. In mammalian tissues, HO-1 is induced by its substrate haem and also by heavy metals. Furthermore, stimuli that cause oxidative stress, such as peroxynitrite, modified lipids, hypoxia, hyperoxia, ischaemia/reperfusion, hyperthermia and endotoxic shock, up-regulate the expression of HO-1 (Sikorski et al., 2004). HO-1 via its products inhibits oxidative stress, inflammation and apoptosis (Dulak et al., 2008). A deficiency of HO-1 in humans results in severe and persistent endothelial damage as indicated by the marked elevation in thrombomodulin and von Willebrand factor (Yachie et al., 1999).

HO/CO cytoprotective pathway in pregnancy

HO-1 and CO play a role in the maintenance of uterine quiescence during human pregnancy (Acevedo and Ahmed, 1998) and in modulating the utero-placental circulation (Ahmed et al., 2000; Lyall et al., 2000). More importantly, placental HO protected human placenta from cellular damage (Ahmed et al., 2000). A recent murine study showed that CO acts as a key molecule in successful pregnancy by modulating the uterine NK cells, which result in the promotion of the remodelling of maternal spiral arteries (Linzke et al., 2014). Decreased expression of HO has also been associated with human pregnancy disorders such as recurrent miscarriages (Denschlag et al., 2004), intrauterine growth retardation (Wong et al., 2012) and pre-eclampsia (Ahmed et al., 2000; Barber et al., 2001).

In pre-eclampsia, the functional importance of HO gained traction after the publication, which demonstrated that adenoviral HO-1 overexpression or CO exposure reduced sVEGFR-1 release from endothelial cells, while siRNA-mediated HO-1 knockdown increases sVEGFR-1 release (Cudmore et al., 2007). A small, but significant clinical study subsequently showed that the HO-1 mRNA was decreased in the chorionic villous (fetal placental cells) sampled at 11 weeks of gestation in women who went on to develop pre-eclampsia compared with normal pregnancies (Farina et al., 2008). Also, the addition of pre-eclampsia sera to the trophoblast increases sEng release and inhibits HO-1 expression (Aoki et al., 2012). Further support for HO-1 acting as a negative regulator of anti-angiogenic factors comes from a recent clinical study demonstrating that in samples of villous trophoblast obtained from women between 6 and 11 weeks of gestation undergoing elective abortion, the mRNA levels of HO-1 were significantly increased with gestational age, whereas the mRNA expression of sVEGFR-1 was significantly decreased with increasing gestational age (Miyagami et al., 2013). Finally, a recent study showed that the long allele of a guanine–thymine polymorphism in the HO-1 promoter region is associated with pre-eclampsia (Kaartokallio et al., 2014). Poor vascular compliance maybe associated with long allele. Collectively, these clinical studies reinforce our theory that placental HO-1 is (i) protective (Ahmed et al., 2000) and (ii) acts as a negative regulator of sVEGFR-1 and sEng (Cudmore et al., 2007). These findings support our proposed model of pre-eclampsia as an accelerator–brake defect disorder. Thus, we would argue that a partial loss of HO-1 activity early in gestation maybe responsible for the cascade of events that culminate in pre-eclampsia (Figure 2).

Interestingly another protective pathway appears to be mediated via the HO-1 system. Kenny's laboratory showed that the administration of a PPAR-γ agonist prevented the development of several of the pathophysiological characteristics associated with the reduced uterine perfusion pressure (RUPP) rat model of pre-eclampsia, via a HO-1-dependent pathway (McCarthy et al., 2011). Finally, the pharmacological induction of HO by cobalt protoporphyrin attenuates placental ischaemia-induced hypertension in this RUPP model (George et al., 2011) and the application of CO could normalize BP in gestational hypertensive HO-1+/− pregnant mice (Linzke et al., 2014).

Another hallmark of pre-eclampsia is glomerular endotheliosis, which results in poor filtration and increased protein in the urine (Maynard et al., 2003). Apart from the administration of sVEGFR-1, VEGF neutralizing antibody to non-pregnant rats also results in glomerular endothelial cell damage and proteinuria (Sugimoto et al., 2003). Cancer patients receiving anti-VEGF therapy exhibit pre–eclampsia-like symptoms, suggesting that decreased bioavailability of VEGF causes these symptoms (Kabbinavar et al., 2003). Unfortunately, the effect of loss of HO-1 activity on kidney function in a pre-eclampsia setting is unknown and warrants investigation.

CO paradox in pregnancy

The source of CO produced in the body, including in the human placental chorionic villi, was shown to originate from haem, primarily through the action of HO (Ahmed et al., 2005). Women with pre-eclampsia have a significantly reduced level of CO in their exhaled breath compared with those with healthy pregnancies indicating a decreased in HO activity (Baum et al., 2000; Kreiser et al., 2004). Interestingly, smoking during pregnancy reduces the incidence of pre-eclampsia, despite being associated with spontaneous abortion, stillbirth, preterm labour, fetal growth restriction and placental abruption (Conde-Agudelo et al., 1999). Smokers are also known to have reduced levels of circulating sVEGFR-1 (Levine et al., 2006). The smoking paradox was explained by the experimental observation that exposure to CO reduces endothelial and placental sVEGFR-1 and sEng release (Cudmore et al., 2007). Furthermore, cigarette smoke extract was shown to induce the expression of HO-1 in placental explants (Sidle et al., 2007) and decrease sVEGFR-1 release from placental villous explants without affecting the placental apoptotic status (Mehendale et al., 2007). Taken together, these studies showed that CO from cigarette smoke accounted for the reduced incidence of pre-eclampsia in smokers through the inhibition of sVEGFR-1 release (Cudmore et al., 2007; Farina et al., 2008; Zhao et al., 2009; Ahmed, 2011). The theory put forward in the May 2000 issue of Molecular Medicine demonstrating that HO offers protection against placental cytotoxic damage associated with pre-eclampsia appears to hold true, based on the supporting data, which has emerged over the last decade. HO-1 is one of the survival breaks against pre-eclampsia (Figure 2).

CSE/H2S system

H2S is a gaseous signalling molecule, which promotes vasodilatation (Zhao et al., 2001), exhibits cytoprotective anti-inflammatory properties (Zanardo et al., 2006), protects against cellular damage induced by reperfusion injury (Elrod et al., 2007) or lethal hypoxia (Blackstone and Roth, 2007), and stimulates angiogenesis in the vasculature (Papapetropoulos et al., 2009). Endogenous H2S production is regulated by three enzymes CSE (also known as Cth), cystathionine-β-synthase (CBS) and 3-mercaptopyruvate sulfurtransferase (MPST), and generated from the substrates cystathionine, homocysteine, cysteine and mercaptopyruvate respectively (Bir and Kevil, 2013). CBS is most abundantly expressed in the brain (Kery et al., 1994) whereas CSE is the principal enzyme responsible for the endogenous production of H2S in the vasculature (Yang et al., 2008; Kabil et al., 2011). Administration of the CSE selective inhibitor (DL-propargylglycine, PAG) leads to elevated BP and vascular remodelling in the rat (Yan et al., 2004) and H2S levels are reduced in pulmonary hypertensive rats (Yanfei et al., 2006). Mice lacking CSE develop age-dependent hypertension, severe hyperhomocysteinaemia, and endothelial dysfunction (Yang et al., 2008).

CSE/H2S pathway in pregnancy

The effects of low levels of H2S showed normal fetal growth and development during pregnancy and it increased delivery time in rats (Hayden et al., 1990) suggesting H2S may exhibit tocolytic characteristics. Indeed H2S inhibits spontaneous contractility in isolated pregnant rat uterine (Sidhu et al., 2001) and human myometrial strips (Hu et al., 2011; You et al., 2011; Robinson and Wray, 2012), similar to what was reported for CO (Acevedo and Ahmed, 1998). This is an area worthy of further investigation. Can myometrial-specific changes in HO-1 or CSE influence parturition?

An H2S-producing system exists within the utero-placental unit (Patel et al., 2009; You et al., 2011). For almost 10 years it has been know that both H2S and CO act as vasodilators (Leffler et al., 2006). It is therefore reassuring that perfusion of normal placenta with an H2S donor results in vasorelaxation of preconstricted vasculature (Cindrova-Davies et al., 2013). H2S may act as a vasodilator in the placental circulation as is the case for H2S in a number of other vascular beds.

Recent immunohistochemical localization studies with CSE and CBS reported contradictory findings on placental pathologies. Holwerda and colleagues observed no changes in CSE expression, but a decrease in CBS expression in placenta from severe pre-eclampsia (Holwerda et al., 2012). In contrast, CSE immunoreactivity was reduced in placenta from pregnancies with severe early-onset growth-restriction and pre-eclampsia (Cindrova-Davies et al., 2013; Wang et al., 2013). Real-time PCR confirmed reduced CSE mRNA in pre-eclamptic women and it was associated with decreased levels of plasma H2S (Wang et al., 2013). However, these observational studies provide little insight into the contributing role of this enzyme system or its gaseous product in pre-eclampsia.

Given that the placenta is a highly vascular organ, Wang et al. proposed that the dys-regulation of the CSE/H2S pathway would promote placental abnormalities and contribute to a pre–eclampsia-like condition. Like the HO-1 system, which negatively regulates sVEGFR-1 and sEng (Cudmore et al., 2007), the CSE pathway has been shown to have similar capabilities. Endothelial CSE knockdown by siRNA increased the endogenous release of sVEGFR-1 and sEng while adenoviral-mediated CSE overexpression inhibited their release from endothelial cells. Furthermore, inhibition of CSE activity by administration of PAG to pregnant mice induced hypertension, liver damage, elevated sVEGFR-1 and sEng and promoted abnormal labyrinth vascularization in the placenta and decreased fetal growth. These symptoms were reversed when the inhibitor was supplemented with GYY4137, a slow-releasing H2S-generating compound demonstrating that the effect of CSE inhibitor was due to inhibition of H2S production (Wang et al., 2013). These findings imply that endogenous H2S is required for healthy placental vasculature and a decrease in CSE/H2S activity may contribute to the pathogenesis of pre-eclampsia. Indeed, H2S was also reported to protect against acute myocardial ischaemia/reperfusion injury. Treatment with H2S donor (SG-1002) offers cardioprotection via up-regulation of the VEGF–Akt–NOS3–NO pathway (Kondo et al., 2013).

The importance of H2S in preventing damage is further supported by a recent study showing that H2S attenuates adenovirus-mediated overexpression of sVEGFR-1-induced hypertension and renal damage in non-pregnant Sprague Dawley rats (Holwerda et al., 2014). This study also showed that rat renal VEGF-A mRNA expression increased significantly with intraperitoneal injection of H2S donor, sodium hydrosulphide (NaHS) treatment and NaHS stimulated podocytes in culture to release VEGF (Holwerda et al., 2014). H2S is known to stimulate VEGF; exposure of vascular smooth muscle cells to H2S up-regulates hypoxia inducible factor (HIF)-1α and VEGF protein levels and increased HIF-1α binding activity under hypoxic condition (Liu et al., 2010). Recently, VEGFR-2 was reported as the direct target of H2S and a VEGF receptor inhibitor suppressed angiogenesis induced by H2S (Tao et al., 2013). These findings indicate that H2S promotes angiogenesis via VEGF receptor activation.

Clearly, CSE may also play a key role in placental development as inhibition of CSE activity by PAG inhibited trophoblast invasion in vitro (Wang et al., 2013). Inhibition of CSE activity in early (first trimester) human placental explants obtained from termination of pregnancy results in a marked reduction in placenta growth factor (PlGF) production (Wang et al., 2013). In pre-eclampsia, the maternal circulating level of PlGF is decreased well before the onset of the symptoms (Levine et al., 2004; 2006). Although the significance of this decrease in PlGF remains to be explained in women, studies by Kumasawa et al. and colleagues show that administration of PlGF in lentiviral sVEGFR-1-infected mice depresses the level of sVEGFR-1 and ameliorated hypertension, glomerular endotheliosis and proteinuria in the mice (Kumasawa et al., 2011). The loss of activity in the H2S-producing enzyme CSE may account for the reduction in PlGF in pre-eclampsia (Wang et al., 2013). As PlGF is one of the earliest maternal circulating markers to be reduced in women destined to develop early-onset pre-eclampsia (Chappell et al., 2002; 2013), CSE/H2S activity may be upstream to PlGF and could be an earlier biomarker as well as a key regulator that keeps the level of PlGF sufficiently high to counteract the deleterious effect of sVEGFR-1, which can induce pre–eclampsia-like symptoms if allowed to go unchecked.

Pharmacological studies using a PAG inhibitor have their limitations because of off-target effects and have been shown to inhibit other pyridoxal-5′-phosphate-dependent enzymes (Whiteman et al., 2011); the lack of selectivity for specific isozymes means future studies need to be carried out using gene inactivation approaches by delineating CSE in the placenta and maternal endothelium (Bir and Kevil, 2013).

NOS3/NO system

NO is synthesized from the nonessential amino acid L-arginine (L-arg) by the enzyme NOS. There are three isoforms of NOS: NOS1 (neuronal), NOS2 (inducible) and NOS3 (endothelial). NOS1 and NOS3 are considered constitutive (cNOS). The NO produced in endothelial cells induces vascular smooth muscle relaxation via a cGMP-dependent pathway to promote vasodilatation. Other beneficial effects of NO in the vascular system include inhibition of ET-1 and TxA2 production, platelet aggregation, leukocyte adhesion and smooth muscle proliferation (Lowe, 2000). NO also inhibits production of VCAM-1 and fibronectin as well as low-density lipoprotein oxidation (Ahmed et al., 2009) and is critical for neovascularization (Bussolati et al., 2001; Ahmad et al., 2006).

Loss of endothelial NOS (NOS3) activity is an established contributor to endothelial dysfunction (Heitzer et al., 2001). NO is highly reactive as it has an unpaired electron, thus a dynamic competition between superoxide and lipid radicals for reaction with NO is inevitable. NO only stimulates superoxide-dependent lipid oxidation when the production rate of NO is less than superoxide (Rubbo et al., 2000). In endothelial cells, NOS3 exists in a homodimeric complex that is stabilized by the cofactor BH4. Decreased availability of BH4 results in ‘uncoupling’ of NOS3 activity and increases the production of superoxide (d'Uscio et al., 2001; Bendall et al., 2005).

NOS3/NO pathway in pregnancy

Enzyme expression and localization studies by themselves can be misleading. However, blocking the production of NO by administering a NOS-inhibiting agent produces virtually all the symptoms of pre-eclampsia in pregnant mice and rats suggesting that the NOS pathway is a key player in this disorder (Lowe, 2000). A meta-analysis showed that genetic variations in the NOS3 gene contribute to an increased risk for pre-eclampsia (Dai et al., 2013). Indeed BH4 doubled NOS3 activity in a concentration-dependent manner in homogenates of first trimester and term placenta (Kukor et al., 2000) and uncoupled NOS3 and oxidative stress in a rat model of pregnancy-induced hypertension (Mitchell et al., 2007). However, BH4 concentrations in pre-eclamptic placenta were reported to be comparable with those of normal placenta (Kukor et al., 2000). In non-pregnant mice lacking NOS3, the sVEGFR1-induced pre-eclampsia phenotype in mice is aggravated (Li et al., 2012).

A nested case control study of screening for pre-eclampsia revealed that asymmetric dimethylarginine (ADMA), arginine and homoarginine at 11–13 weeks' gestation did not change in women who went on to develop late-onset pre-eclampsia, but L-arg and homoarginine levels were decreased in those who developed early-onset pre-eclampsia (Khalil et al., 2013). Interestingly, maternal serum ADMA concentration tends to increase during normal pregnancy, but ADMA concentrations in the second trimester were significantly elevated in pregnancies that later developed pre-eclampsia (Rizos et al., 2012).

An important cause of impaired endothelial NO production is the reduced availability of the NOS3 substrate L-arg. A study reported that plasma nitrite and superoxide dismutase activity were significantly lower while arginase activity and plasma endothelin levels were significantly higher in pre-eclamptic women than in normotensive pregnant women (Bernardi et al., 2008). Why arginase activity is increased during pregnancy is unknown, but should be a subject of future investigation. The Davidge laboratory have shown that an increased arginase expression in pre-eclampsia can induce the uncoupling of NOS as a source of superoxide in the maternal vasculature in pre-eclampsia, and suggested that L-arg supplementation in the face of oxidative stress could lead to a further increase in peroxynitrite (Sankaralingam et al., 2010). Thus, this brings into question L-arg supplementation as therapy without clear mechanistic evidence.

Maximum activity of NOS3 is achieved when the residue S1177, which is targeted by Akt, is phosphorylated and residue T497 is dephosphorylated. The constitutively active NOS3 mutant, S1177D enhances NO-mediated relaxation of bradykinin-stimulated porcine coronary arteries (Teupe et al., 2002). Basal production of NO from cells is markedly reduced in cells transfected with T497A and T497A/S1177D (TASD) NOS3 because of the generation of more superoxide anion that interact with NO. In contrast, NO production is augmented in cells transfected with S1177D NOS3 (Lin et al., 2003). Using adenoviruses expressing wild-type NOS3 and S1177D and TASD NOS3 mutants, we discovered that neither the constitutively active NOS3 mutant S1177D nor the TASD NOS3 mutants suppress VEGF-induced sVEGFR-1 release from endothelial cells. These unpublished findings are in marked contrast to studies in which sildenafil citrate reduces the plasma levels of sVEGFR-1 and sEng in L–NAME-treated Sprague Dawley pregnant rats (Ramesar et al., 2011). Sildenafil may exert its effect indirectly. Finally, storkhead box 1 (STOX1) overexpression switches the free radical balance from reactive oxygen species to reactive nitrogen species in the placenta (Doridot et al., 2014), which may deprive maternal NO and compromise blood flow or increase endothelial dysfunction. Could this act as a genetic switch altering vascular function and inhibiting the protective genes such as HO-1?

Conclusion

Women destined to develop early-onset pre-eclampsia could be offered therapies that up-regulate ‘the endogenous cytoprotective pathway’. A cheap and widely available therapy against pre-eclampsia may be on the horizon if pravastatin (3-hydroxy-3-methylglutaryl-coenzyme A reductase inhibitors) proves to be ‘safe’ in randomized controlled trial (RCT) (StAmP). Although statins are contraindicated in pregnancy, the finding that statins inhibited sVEGFR-1 led to the UK regulatory authorities approving the first RCT on statins in pre-eclampsia. Today, there are two additional trials underway in Australia and the United States.

In order to develop additional therapies to tackle pre-eclampsia, basic research is urgently needed to elucidate fully the role of HO-1 and CSE and their metabolites. Murine models do not equate to pre-eclampsia in women, but they can replicate many of the pre-eclampsia symptoms. This offers very useful tools to perform proof of principle experiments to determine the role of specific genes and potential new therapies. H2S or CO may modulate NOS3 activity to prevent pre-eclampsia. It is established that one of the beneficial effects of statins in the prevention of cardiovascular events is their ability to enhance endothelial function by increasing NOS3 expression and activity (Xenos et al., 2005). The success of the StAmP trial may not only depend on its ability to up-regulate HO or inhibit sVEGFR-1, but on the power of statins to inhibit an array of stress factors associated with pre-eclampsia.

This review suggests new investigations with new thinking which challenges the existing dogma, employs the use of sophisticated inducible- and tissue-specific knockdown tools in what we refer to ‘the maternal and fetal vascular complications mouse models of pre-eclampsia’ combined with human clinical-based sample studies. For example, transcription factor STOX1 overexpressing pregnant mice exhibit symptoms of severe pre-eclampsia: gestational hypertension, proteinuria, and elevated plasma levels of sVEGFR-1 (soluble fms-like tyrosine kinase 1) and sEng (Doridot et al., 2013). We propose a new paradigm as outlined here in ‘the accelerator and brake’ hypothesis, which can encompass novel factors (Figure 2). We need to design rigorous tests for a given hypothesis and bring in a team-based approach, which can utilize resources from a range of settings. Studies need to encompass clinical samples, cell culture studies and proof of concept mouse models as outlined earlier. The criticism of mouse models in pre-eclampsia needs to stop, provided the animal data are backed up by human data as illustrated in a number of comprehensive studies (Maynard et al., 2003; Wang et al., 2013). Animal studies in pre-eclampsia should be accepted as powerful scientific tools where specific genes can be delineated in a tissue-specific fashion to address specific questions. We propose this approach for the cytoprotective pathway gasotransmitter enzymes, which we believe holds the greatest promise in the clinic and maybe to the underlining mechanism that is defective in pre-eclampsia. In conclusion, our discovery of the two gasotranmitter enzyme systems as protective pathways to treat or prevent preeclampsia (Figure 1) and the accelerator-brake model of preeclampsia is being used to develop new therapies, which aim to enhance the brakes when the system starts to fail. The StAmP trial is the first such approach to be completed. The future personalized pre-eclampsia therapy is looking promising!

Acknowledgments

We are grateful to the British Heart Foundation and the Medical Research Council for their generous financial support over the years without whose support these ideas would have died an early death. We wish to express our deepest gratitude to all our collaborators for sharing their resources and for stimulating discussions, and finally we wish to thank all the past and present researchers, students and technicians who have worked in our laboratory, without whose efforts, nothing would have been possible.

Glossary

ADMA

asymmetric dimethylarginine

BH4

tetrahydrobiopterin

CBS

cystathionine-β-synthase

CO

carbon monoxide

CSE (also known as Cth)

cystathionine-γ-lyase

H2S

hydrogen sulphide

HO (also know as Hmox)

haem oxygenase

L-arg

L-arginine

PAG

DL-propargylglycine

PlGF

placenta growth factor

RUPP

reduced uterine perfusion pressure

sEng

soluble endoglin

sVEGFR-1 (also known as sFlt-1

soluble fms-like tyrosine kinase-1

StAmP

pravastatin to ameliorate early-onset pre-eclampsia

Conflict of interest

The authors have no conflict of interest to report.

References

  1. Acevedo CH, Ahmed A. Hemeoxygenase-1 inhibits human myometrial contractility via carbon monoxide and is upregulated by progesterone during pregnancy. J Clin Invest. 1998;101:949–955. doi: 10.1172/JCI927. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Ahmad S, Ahmed A. Elevated placental soluble vascular endothelial growth factor receptor-1 inhibits angiogenesis in preeclampsia. Circ Res. 2004;95:884–891. doi: 10.1161/01.RES.0000147365.86159.f5. [DOI] [PubMed] [Google Scholar]
  3. Ahmad S, Hewett PW, Wang P, Al-Ani B, Cudmore M, Fujisawa T, et al. Direct evidence for endothelial vascular endothelial growth factor receptor-1 function in nitric oxide-mediated angiogenesis. Circ Res. 2006;99:715–722. doi: 10.1161/01.RES.0000243989.46006.b9. [DOI] [PubMed] [Google Scholar]
  4. Ahmed A. Heparin-binding angiogenic growth factors in pregnancy. Trophoblast Res. 1997;10:215–258. [Google Scholar]
  5. Ahmed A. New insights into the etiology of preeclampsia: identification of key elusive factors for the vascular complications. Thromb Res. 2011;127S3:S72–S75. doi: 10.1016/S0049-3848(11)70020-2. [DOI] [PubMed] [Google Scholar]
  6. Ahmed A, Rahman M, Zhang X, Acevedo CH, Nijjar S, Rushton I, et al. Induction of placental heme oxygenase-1 is protective against TNFalpha- induced cytotoxicity and promotes vessel relaxation. Mol Med. 2000;6:391–409. [PMC free article] [PubMed] [Google Scholar]
  7. Ahmed A, Fujisawa T, Niu XL, Ahmad S, Al-Ani B, Chudasama K, et al. Angiopoietin-2 confers Atheroprotection in apoE−/− mice by inhibiting LDL oxidation via nitric oxide. Circ Res. 2009;104:1333–1336. doi: 10.1161/CIRCRESAHA.109.196154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Ahmed H, McLaughlin BE, Soong J, Marks GS, Brien JF, Nakatsu K. The source of endogenous carbon monoxide formation in human placental chorionic villi. Cell Mol Biol (Noisy-Le-Grand) 2005;51:447–451. [PubMed] [Google Scholar]
  9. Alexander SPH, Benson HE, Faccenda E, Pawson AJ, Sharman JL, Spedding M, et al. The Concise Guide to PHARMACOLOGY 2013/14: Nuclear hormone receptors. Br J Pharmacol. 2013a;170:1652–1675. doi: 10.1111/bph.12448. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Alexander SPH, Benson HE, Faccenda E, Pawson AJ, Sharman JL, Spedding M, et al. The Concise Guide to PHARMACOLOGY 2013/14: Catalytic receptors. Br J Pharmacol. 2013b;170:1676–1705. doi: 10.1111/bph.12449. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Alexander SPH, Benson HE, Faccenda E, Pawson AJ, Sharman JL, Spedding M, et al. The Concise Guide to PHARMACOLOGY 2013/14: Enzymes. Br J Pharmacol. 2013c;170:1797–1867. doi: 10.1111/bph.12451. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Aoki Y, Yamamoto T, Fumihisa C, Nakamura A, Asanuma A, Suzuki M. Effect on the production of soluble endoglin from human choriocarcinoma cells by preeclampsia sera. Am J Reprod Immunol. 2012;67:413–420. doi: 10.1111/j.1600-0897.2011.01086.x. [DOI] [PubMed] [Google Scholar]
  13. Avagliano L, Bulfamante GP, Morabito A, Marconi AM. Abnormal spiral artery remodelling in the decidual segment during pregnancy: from histology to clinical correlation. J Clin Pathol. 2011;64:1064–1068. doi: 10.1136/jclinpath-2011-200092. [DOI] [PubMed] [Google Scholar]
  14. Bainbridge SA, Farley AE, McLaughlin BE, Graham CH, Marks GS, Nakatsu K, et al. Carbon monoxide decreases perfusion pressure in isolated human placenta. Placenta. 2002;23:563–569. doi: 10.1053/plac.2002.0845. [DOI] [PubMed] [Google Scholar]
  15. Barber A, Robson SC, Myatt L, Bulmer JN, Lyall F. Heme oxygenase expression in human placenta and placental bed: reduced expression of placenta endothelial HO-2 in preeclampsia and fetal growth restriction. FASEB J. 2001;15:1158–1168. doi: 10.1096/fj.00-0376com. [DOI] [PubMed] [Google Scholar]
  16. Baum M, Schiff E, Kreiser D, Dennery PA, Stevenson DK, Rosenthal T, et al. End-tidal carbon monoxide measurements in women with pregnancy-induced hypertension and preeclampsia. Am J Obstet Gynecol. 2000;183:900–903. doi: 10.1067/mob.2000.109047. [DOI] [PubMed] [Google Scholar]
  17. Bellamy L, Casas JP, Hingorani AD, Williams DJ. Pre-eclampsia and risk of cardiovascular disease and cancer in later life: systematic review and meta-analysis. BMJ. 2007;335:974. doi: 10.1136/bmj.39335.385301.BE. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Bendall JK, Alp NJ, Warrick N, Cai S, Adlam D, Rockett K, et al. Stoichiometric relationships between endothelial tetrahydrobiopterin, endothelial NO synthase (eNOS) activity, and eNOS coupling in vivo: insights from transgenic mice with endothelial-targeted GTP cyclohydrolase 1 and eNOS overexpression. Circ Res. 2005;97:864–871. doi: 10.1161/01.RES.0000187447.03525.72. [DOI] [PubMed] [Google Scholar]
  19. Bernardi F, Constantino L, Machado R, Petronilho F, Dal-Pizzol F. Plasma nitric oxide, endothelin-1, arginase and superoxide dismutase in pre-eclamptic women. J Obstet Gynaecol Res. 2008;34:957–963. doi: 10.1111/j.1447-0756.2008.00860.x. [DOI] [PubMed] [Google Scholar]
  20. Bir SC, Kevil CG. Sulfane sustains vascular health: insights into cystathionine gamma-lyase function. Circulation. 2013;127:2472–2474. doi: 10.1161/CIRCULATIONAHA.113.003489. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Blackstone E, Roth MB. Suspended animation-like state protects mice from lethal hypoxia. Shock. 2007;27:370–372. doi: 10.1097/SHK.0b013e31802e27a0. [DOI] [PubMed] [Google Scholar]
  22. Brosens I. A study of the spiral arteries of the decidua basalis in normotensive and hypertensive pregnancies. J Obstet Gynaecol Br Commonw. 1964;71:222–230. doi: 10.1111/j.1471-0528.1964.tb04270.x. [DOI] [PubMed] [Google Scholar]
  23. Brosens IA. Morphological changes in the utero-placental bed in pregnancy hypertension. Clin Obstet Gynaecol. 1977;4:573–593. [PubMed] [Google Scholar]
  24. Brosens JJ, Pijnenborg R, Brosens IA. The myometrial junctional zone spiral arteries in normal and abnormal pregnancies: a review of the literature. Am J Obstet Gynecol. 2002;187:1416–1423. doi: 10.1067/mob.2002.127305. [DOI] [PubMed] [Google Scholar]
  25. Buhimschi CS, Magloire L, Funai E, Norwitz ER, Kuczynski E, Martin R, et al. Fractional excretion of angiogenic factors in women with severe preeclampsia. Obstet Gynecol. 2006;107:1103–1113. doi: 10.1097/01.AOG.0000207698.74104.4f. [DOI] [PubMed] [Google Scholar]
  26. Burke SD, Karumanchi SA. Spiral artery remodeling in preeclampsia revisited. Hypertension. 2013;62:1013–1014. doi: 10.1161/HYPERTENSIONAHA.113.02049. [DOI] [PubMed] [Google Scholar]
  27. Burke SD, Barrette VF, Bianco J, Thorne JG, Yamada AT, Pang SC, et al. Spiral arterial remodeling is not essential for normal blood pressure regulation in pregnant mice. Hypertension. 2010;55:729–737. doi: 10.1161/HYPERTENSIONAHA.109.144253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Burton GJ, Woods AW, Jauniaux E, Kingdom JC. Rheological and physiological consequences of conversion of the maternal spiral arteries for uteroplacental blood flow during human pregnancy. Placenta. 2009;30:473–482. doi: 10.1016/j.placenta.2009.02.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Bussolati B, Dunk C, Grohman M, Kontos CD, Mason J, Ahmed A. Vascular endothelial growth factor receptor-1 modulates vascular endothelial growth factor-mediated angiogenesis via nitric oxide. Am J Pathol. 2001;159:993–1008. doi: 10.1016/S0002-9440(10)61775-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Carty DM, Anderson LA, Freeman DJ, Welsh PI, Brennand JE, Dominiczak AF, et al. Early pregnancy soluble E-selectin concentrations and risk of preeclampsia. J Hypertens. 2012;30:954–959. doi: 10.1097/HJH.0b013e328352573b. [DOI] [PubMed] [Google Scholar]
  31. Chappell LC, Seed PT, Briley A, Kelly FJ, Hunt BJ, Charnock-Jones DS, et al. A longitudinal study of biochemical variables in women at risk of preeclampsia. Am J Obstet Gynecol. 2002;187:127–136. doi: 10.1067/mob.2002.122969. [DOI] [PubMed] [Google Scholar]
  32. Chappell LC, Duckworth S, Seed PT, Griffin M, Myers J, Mackillop L, et al. Diagnostic accuracy of placental growth factor in women with suspected preeclampsia: a prospective multicenter study. Circulation. 2013;128:2121–2131. doi: 10.1161/CIRCULATIONAHA.113.003215. [DOI] [PubMed] [Google Scholar]
  33. Cindrova-Davies T, Herrera EA, Niu Y, Kingdom J, Giussani DA, Burton GJ. Reduced cystathionine gamma-lyase and increased MicroRNA-21 expression are associated with increased vascular resistance in growth-restricted pregnancies: hydrogen sulfide as a placental vasodilator. Am J Pathol. 2013;182:1448–1458. doi: 10.1016/j.ajpath.2013.01.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Coletta C, Papapetropoulos A, Erdelyi K, Olah G, Modis K, Panopoulos P, et al. Hydrogen sulfide and nitric oxide are mutually dependent in the regulation of angiogenesis and endothelium-dependent vasorelaxation. Proc Natl Acad Sci U S A. 2012;109:9161–9166. doi: 10.1073/pnas.1202916109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Conde-Agudelo A, Althabe F, Belizan JM, Kafury-Goeta AC. Cigarette smoking during pregnancy and risk of preeclampsia: a systematic review. Am J Obstet Gynecol. 1999;181:1026–1035. doi: 10.1016/s0002-9378(99)70341-8. [DOI] [PubMed] [Google Scholar]
  36. Cotechini T, Komisarenko M, Sperou A, Macdonald-Goodfellow S, Adams MA, Graham CH. Inflammation in rat pregnancy inhibits spiral artery remodeling leading to fetal growth restriction and features of preeclampsia. J Exp Med. 2014;211:165–179. doi: 10.1084/jem.20130295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Crispi F, Dominguez C, Llurba E, Martin-Gallan P, Cabero L, Gratacos E. Placental angiogenic growth factors and uterine artery Doppler findings for characterization of different subsets in preeclampsia and in isolated intrauterine growth restriction. Am J Obstet Gynecol. 2006;195:201–207. doi: 10.1016/j.ajog.2006.01.014. [DOI] [PubMed] [Google Scholar]
  38. Cudmore M, Ahmad S, Al-Ani B, Fujisawa T, Coxall H, Chudasama K, et al. Negative regulation of soluble Flt-1 and soluble endoglin release by heme oxygenase-1. Circulation. 2007;115:1789–1797. doi: 10.1161/CIRCULATIONAHA.106.660134. [DOI] [PubMed] [Google Scholar]
  39. d'Uscio LV, Baker TA, Mantilla CB, Smith L, Weiler D, Sieck GC, et al. Mechanism of endothelial dysfunction in apolipoprotein E-deficient mice. Arterioscler Thromb Vasc Biol. 2001;21:1017–1022. doi: 10.1161/01.atv.21.6.1017. [DOI] [PubMed] [Google Scholar]
  40. Dai B, Liu T, Zhang B, Zhang X, Wang Z. The polymorphism for endothelial nitric oxide synthase gene, the level of nitric oxide and the risk for pre-eclampsia: a meta-analysis. Gene. 2013;519:187–193. doi: 10.1016/j.gene.2013.01.004. [DOI] [PubMed] [Google Scholar]
  41. Davidge ST. Oxidative stress and altered endothelial cell function in preeclampsia. Semin Reprod Endocrinol. 1998;16:65–73. doi: 10.1055/s-2007-1016254. [DOI] [PubMed] [Google Scholar]
  42. Denschlag D, Marculescu R, Unfried G, Hefler LA, Exner M, Hashemi A, et al. The size of a microsatellite polymorphism of the haem oxygenase 1 gene is associated with idiopathic recurrent miscarriage. Mol Hum Reprod. 2004;10:211–214. doi: 10.1093/molehr/gah024. [DOI] [PubMed] [Google Scholar]
  43. Djurovic S, Clausen T, Wergeland R, Brosstad F, Berg K, Henriksen T. Absence of enhanced systemic inflammatory response at 18 weeks of gestation in women with subsequent pre-eclampsia. BJOG. 2002;109:759–764. doi: 10.1111/j.1471-0528.2002.01330.x. [DOI] [PubMed] [Google Scholar]
  44. Doridot L, Passet B, Mehats C, Rigourd V, Barbaux S, Ducat A, et al. Preeclampsia-like symptoms induced in mice by fetoplacental expression of STOX1 are reversed by aspirin treatment. Hypertension. 2013;61:662–668. doi: 10.1161/HYPERTENSIONAHA.111.202994. [DOI] [PubMed] [Google Scholar]
  45. Doridot L, Chatre L, Ducat A, Vilotte JL, Lombes A, Mehats C, et al. Nitroso-redox balance and mitochondrial homeostasis are regulated by STOX1, a pre-eclampsia-associated gene. Antioxid Redox Signal. 2014;21:819–834. doi: 10.1089/ars.2013.5661. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Dulak J, Deshane J, Jozkowicz A, Agarwal A. Heme oxygenase-1 and carbon monoxide in vascular pathobiology: focus on angiogenesis. Circulation. 2008;117:231–241. doi: 10.1161/CIRCULATIONAHA.107.698316. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Elrod JW, Calvert JW, Morrison J, Doeller JE, Kraus DW, Tao L, et al. Hydrogen sulfide attenuates myocardial ischemia-reperfusion injury by preservation of mitochondrial function. Proc Natl Acad Sci U S A. 2007;104:15560–15565. doi: 10.1073/pnas.0705891104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Farina A, Sekizawa A, de Sanctis P, Purwosunu Y, Okai T, Cha DH, et al. Gene expression in chorionic villous samples at 11 weeks' gestation from women destined to develop preeclampsia. Prenat Diagn. 2008;28:956–961. doi: 10.1002/pd.2109. [DOI] [PubMed] [Google Scholar]
  49. Feinberg RF, Kliman HJ, Cohen AW. Preeclampsia, trisomy 13, and the placental bed. Obstet Gynecol. 1991;78:505–508. [PubMed] [Google Scholar]
  50. George EM, Cockrell K, Aranay M, Csongradi E, Stec DE, Granger JP. Induction of heme oxygenase 1 attenuates placental ischemia-induced hypertension. Hypertension. 2011;57:941–948. doi: 10.1161/HYPERTENSIONAHA.111.169755. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Gonzalez C, Cruz MA, Gallardo V, Lagos M, Varela J, Albornoz J, et al. Nitric oxide and prostaglandin systems inhibition on the isolated perfused human placenta from normal and preeclamptic pregnancies. Gynecol Obstet Invest. 1995;40:244–248. doi: 10.1159/000292345. [DOI] [PubMed] [Google Scholar]
  52. Gude NM, King RG, Brennecke SP. Role of endothelium-derived nitric oxide in maintenance of low fetal vascular resistance in placenta. Lancet. 1990;336:1589–1590. doi: 10.1016/0140-6736(90)93374-x. [DOI] [PubMed] [Google Scholar]
  53. Hayden LJ, Goeden H, Roth SH. Growth and development in the rat during sub-chronic exposure to low levels of hydrogen sulfide. Toxicol Ind Health. 1990;6:389–401. doi: 10.1177/074823379000600303. [DOI] [PubMed] [Google Scholar]
  54. Heitzer T, Schlinzig T, Krohn K, Meinertz T, Munzel T. Endothelial dysfunction, oxidative stress, and risk of cardiovascular events in patients with coronary artery disease. Circulation. 2001;104:2673–2678. doi: 10.1161/hc4601.099485. [DOI] [PubMed] [Google Scholar]
  55. Holwerda KM, Bos EM, Rajakumar A, Ris-Stalpers C, van Pampus MG, Timmer A, et al. Hydrogen sulfide producing enzymes in pregnancy and preeclampsia. Placenta. 2012;33:518–521. doi: 10.1016/j.placenta.2012.02.014. [DOI] [PubMed] [Google Scholar]
  56. Holwerda KM, Burke SD, Faas MM, Zsengeller Z, Stillman IE, Kang PM, et al. Hydrogen sulfide attenuates sFlt1-induced hypertension and renal damage by upregulating vascular endothelial growth factor. J Am Soc Nephrol. 2014;25:717–725. doi: 10.1681/ASN.2013030291. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Hu R, Lu J, You X, Zhu X, Hui N, Ni X. Hydrogen sulfide inhibits the spontaneous and oxytocin-induced contractility of human pregnant myometrium. Gynecol Endocrinol. 2011;27:900–904. doi: 10.3109/09513590.2010.551563. [DOI] [PubMed] [Google Scholar]
  58. Hubel CA. Oxidative stress in the pathogenesis of preeclampsia. Proc Soc Exp Biol Med. 1999;222:222–235. doi: 10.1177/153537029922200305. [DOI] [PubMed] [Google Scholar]
  59. Huppertz B, Weiss G, Moser G. Trophoblast invasion and oxygenation of the placenta: measurements versus presumptions. J Reprod Immunol. 2014;101–102:74–79. doi: 10.1016/j.jri.2013.04.003. [DOI] [PubMed] [Google Scholar]
  60. Johal T, Lees CC, Everett TR, Wilkinson IB. The nitric oxide pathway and possible therapeutic options in pre-eclampsia. Br J Clin Pharmacol. 2014;78:244–257. doi: 10.1111/bcp.12301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Kaartokallio T, Klemetti MM, Timonen A, Uotila J, Heinonen S, Kajantie E, et al. Microsatellite polymorphism in the heme oxygenase-1 promoter is associated with nonsevere and late-onset preeclampsia. Hypertension. 2014;64:172–177. doi: 10.1161/HYPERTENSIONAHA.114.03337. [DOI] [PubMed] [Google Scholar]
  62. Kabbinavar F, Hurwitz HI, Fehrenbacher L, Meropol NJ, Novotny WF, Lieberman G, et al. Phase II, randomized trial comparing bevacizumab plus fluorouracil (FU)/leucovorin (LV) with FU/LV alone in patients with metastatic colorectal cancer. J Clin Oncol. 2003;21:60–65. doi: 10.1200/JCO.2003.10.066. [DOI] [PubMed] [Google Scholar]
  63. Kabil O, Vitvitsky V, Xie P, Banerjee R. The quantitative significance of the transsulfuration enzymes for H2S production in murine tissues. Antioxid Redox Signal. 2011;15:363–372. doi: 10.1089/ars.2010.3781. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Kery V, Bukovska G, Kraus JP. Transsulfuration depends on heme in addition to pyridoxal 5′-phosphate. Cystathionine beta-synthase is a heme protein. J Biol Chem. 1994;269:25283–25288. [PubMed] [Google Scholar]
  65. Khalil AA, Tsikas D, Akolekar R, Jordan J, Nicolaides KH. Asymmetric dimethylarginine, arginine and homoarginine at 11–13 weeks' gestation and preeclampsia: a case-control study. J Hum Hypertens. 2013;27:38–43. doi: 10.1038/jhh.2011.109. [DOI] [PubMed] [Google Scholar]
  66. Kharb S. Vitamin E and C in preeclampsia. Eur J Obstet Gynecol Reprod Biol. 2000;93:37–39. doi: 10.1016/s0301-2115(00)00243-8. [DOI] [PubMed] [Google Scholar]
  67. Khong TY, de Wolf F, Robertson WB, Brosens I. Inadequate maternal vascular response to placentation in pregnancies complicated by pre-eclampsia and by small-for-gestational age infants. Br J Obstet Gynaecol. 1986;93:1049–1059. doi: 10.1111/j.1471-0528.1986.tb07830.x. [DOI] [PubMed] [Google Scholar]
  68. Kondo K, Bhushan S, King AL, Prabhu SD, Hamid T, Koenig S, et al. H(2)S protects against pressure overload-induced heart failure via upregulation of endothelial nitric oxide synthase. Circulation. 2013;127:1116–1127. doi: 10.1161/CIRCULATIONAHA.112.000855. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Kreiser D, Baum M, Seidman DS, Fanaroff A, Shah D, Hendler I, et al. End tidal carbon monoxide levels are lower in women with gestational hypertension and pre-eclampsia. J Perinatol. 2004;24:213–217. doi: 10.1038/sj.jp.7211062. [DOI] [PubMed] [Google Scholar]
  70. Kronborg CS, Gjedsted J, Vittinghus E, Hansen TK, Allen J, Knudsen UB. Longitudinal measurement of cytokines in pre-eclamptic and normotensive pregnancies. Acta Obstet Gynecol Scand. 2011;90:791–796. doi: 10.1111/j.1600-0412.2011.01134.x. [DOI] [PubMed] [Google Scholar]
  71. Kukor Z, Valent S, Toth M. Regulation of nitric oxide synthase activity by tetrahydrobiopterin in human placentae from normal and pre-eclamptic pregnancies. Placenta. 2000;21:763–772. doi: 10.1053/plac.2000.0584. [DOI] [PubMed] [Google Scholar]
  72. Kumasawa K, Ikawa M, Kidoya H, Hasuwa H, Saito-Fujita T, Morioka Y, et al. Pravastatin induces placental growth factor (PGF) and ameliorates preeclampsia in a mouse model. Proc Natl Acad Sci U S A. 2011;108:1451–1455. doi: 10.1073/pnas.1011293108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. LaMarca BD, Ryan MJ, Gilbert JS, Murphy SR, Granger JP. Inflammatory cytokines in the pathophysiology of hypertension during preeclampsia. Curr Hypertens Rep. 2007;9:480–485. doi: 10.1007/s11906-007-0088-1. [DOI] [PubMed] [Google Scholar]
  74. Learmont JG, Poston L. Nitric oxide is involved in flow-induced dilation of isolated human small fetoplacental arteries. Am J Obstet Gynecol. 1996;174:583–588. doi: 10.1016/s0002-9378(96)70432-5. [DOI] [PubMed] [Google Scholar]
  75. Leffler CW, Parfenova H, Jaggar JH, Wang R. Carbon monoxide and hydrogen sulfide: gaseous messengers in cerebrovascular circulation. J Appl Physiol (1985) 2006;100:1065–1076. doi: 10.1152/japplphysiol.00793.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Levine RJ, Maynard SE, Qian C, Lim KH, England LJ, Yu KF, et al. Circulating angiogenic factors and the risk of preeclampsia. N Engl J Med. 2004;350:672–683. doi: 10.1056/NEJMoa031884. [DOI] [PubMed] [Google Scholar]
  77. Levine RJ, Lam C, Qian C, Yu KF, Maynard SE, Sachs BP, et al. Soluble endoglin and other circulating antiangiogenic factors in preeclampsia. N Engl J Med. 2006;355:992–1005. doi: 10.1056/NEJMoa055352. [DOI] [PubMed] [Google Scholar]
  78. Li F, Hagaman JR, Kim HS, Maeda N, Jennette JC, Faber JE, et al. eNOS deficiency acts through endothelin to aggravate sFlt-1-induced pre-eclampsia-like phenotype. J Am Soc Nephrol. 2012;23:652–660. doi: 10.1681/ASN.2011040369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Lin MI, Fulton D, Babbitt R, Fleming I, Busse R, Pritchard KA, Jr, et al. Phosphorylation of threonine 497 in endothelial nitric-oxide synthase coordinates the coupling of L-arginine metabolism to efficient nitric oxide production. J Biol Chem. 2003;278:44719–44726. doi: 10.1074/jbc.M302836200. [DOI] [PubMed] [Google Scholar]
  80. Linzke N, Schumacher A, Woidacki K, Croy BA, Zenclussen AC. Carbon monoxide promotes proliferation of uterine natural killer cells and remodeling of spiral arteries in pregnant hypertensive heme oxygenase-1 mutant mice. Hypertension. 2014;63:580–588. doi: 10.1161/HYPERTENSIONAHA.113.02403. [DOI] [PubMed] [Google Scholar]
  81. Liu X, Pan L, Zhuo Y, Gong Q, Rose P, Zhu Y. Hypoxia-inducible factor-1alpha is involved in the pro-angiogenic effect of hydrogen sulfide under hypoxic stress. Biol Pharm Bull. 2010;33:1550–1554. doi: 10.1248/bpb.33.1550. [DOI] [PubMed] [Google Scholar]
  82. Lowe DT. Nitric oxide dysfunction in the pathophysiology of preeclampsia. Nitric Oxide. 2000;4:441–458. doi: 10.1006/niox.2000.0296. [DOI] [PubMed] [Google Scholar]
  83. Lowe SA, Brown MA, Dekker GA, Gatt S, McLintock CK, McMahon LP, et al. Guidelines for the management of hypertensive disorders of pregnancy 2008. Aust N Z J Obstet Gynaecol. 2009;49:242–246. doi: 10.1111/j.1479-828X.2009.01003.x. [DOI] [PubMed] [Google Scholar]
  84. Lyall F, Barber A, Myatt L, Bulmer JN, Robson SC. Hemeoxygenase expression in human placenta and placental bed implies a role in regulation of trophoblast invasion and placental function. FASEB J. 2000;14:208–219. doi: 10.1096/fasebj.14.1.208. [DOI] [PubMed] [Google Scholar]
  85. Lyall F, Robson SC, Bulmer JN. Spiral artery remodeling and trophoblast invasion in preeclampsia and fetal growth restriction: relationship to clinical outcome. Hypertension. 2013;62:1046–1054. doi: 10.1161/HYPERTENSIONAHA.113.01892. [DOI] [PubMed] [Google Scholar]
  86. MacDonald SE, Walker M, Ramshaw H, Godwin M, Chen XK, Smith G. Hypertensive disorders of pregnancy and long-term risk of hypertension: what do Ontario prenatal care providers know, and what do they communicate? J Obstet Gynaecol Can. 2007;29:705–710. doi: 10.1016/s1701-2163(16)32601-9. [DOI] [PubMed] [Google Scholar]
  87. Maynard SE, Min JY, Merchan J, Lim KH, Li J, Mondal S, et al. Excess placental soluble fms-like tyrosine kinase 1 (sFlt1) may contribute to endothelial dysfunction, hypertension, and proteinuria in preeclampsia. J Clin Invest. 2003;111:649–658. doi: 10.1172/JCI17189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. McCarthy FP, Drewlo S, Kingdom J, Johns EJ, Walsh SK, Kenny LC. Peroxisome proliferator-activated receptor-{gamma} as a potential therapeutic target in the treatment of preeclampsia. Hypertension. 2011;58:280–286. doi: 10.1161/HYPERTENSIONAHA.111.172627. [DOI] [PubMed] [Google Scholar]
  89. Mehendale R, Hibbard J, Fazleabas A, Leach R. Placental angiogenesis markers sFlt-1 and PlGF: response to cigarette smoke. Am J Obstet Gynecol. 2007;197:363. doi: 10.1016/j.ajog.2007.06.025. e1–5. [DOI] [PubMed] [Google Scholar]
  90. Mitchell BM, Cook LG, Danchuk S, Puschett JB. Uncoupled endothelial nitric oxide synthase and oxidative stress in a rat model of pregnancy-induced hypertension. Am J Hypertens. 2007;20:1297–1304. doi: 10.1016/j.amjhyper.2007.08.007. [DOI] [PubMed] [Google Scholar]
  91. Miyagami S, Koide K, Sekizawa A, Ventura W, Yotsumoto J, Oishi S, et al. Physiological changes in the pattern of placental gene expression early in the first trimester. Reprod Sci. 2013;20:710–714. doi: 10.1177/1933719112466309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Moncada S. Nitric oxide in the vasculature: physiology and pathophysiology. Ann N Y Acad Sci. 1997;811:60–67. doi: 10.1111/j.1749-6632.1997.tb51989.x. discussion 67–9. [DOI] [PubMed] [Google Scholar]
  93. Mustafa AK, Gadalla MM, Snyder SH. Signaling by gasotransmitters. Sci Signal. 2009;2:re2. doi: 10.1126/scisignal.268re2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Myatt L, Brewer A, Brockman DE. The action of nitric oxide in the perfused human fetal-placental circulation. Am J Obstet Gynecol. 1991;164:687–692. doi: 10.1016/s0002-9378(11)80047-5. [DOI] [PubMed] [Google Scholar]
  95. Naljayan MV, Karumanchi SA. New developments in the pathogenesis of preeclampsia. Adv Chronic Kidney Dis. 2013;20:265–270. doi: 10.1053/j.ackd.2013.02.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  96. Nayeri UA, Buhimschi IA, Laky CA, Cross SN, Duzyj CM, Ramma W, et al. Antenatal corticosteroids impact the inflammatory rather than the antiangiogenic profile of women with preeclampsia. Hypertension. 2014;63:1285–1292. doi: 10.1161/HYPERTENSIONAHA.114.03173. [DOI] [PubMed] [Google Scholar]
  97. Odrcich MJ, Graham CH, Kimura KA, McLaughlin BE, Marks GS, Nakatsu K, et al. Heme oxygenase and nitric oxide synthase in the placenta of the guinea-pig during gestation. Placenta. 1998;19:509–516. doi: 10.1016/s0143-4004(98)91044-x. [DOI] [PubMed] [Google Scholar]
  98. Ozler A, Turgut A, Sak ME, Evsen MS, Soydinc HE, Evliyaoglu O, et al. Serum levels of neopterin, tumor necrosis factor-alpha and Interleukin-6 in preeclampsia: relationship with disease severity. Eur Rev Med Pharmacol Sci. 2012;16:1707–1712. [PubMed] [Google Scholar]
  99. Papapetropoulos A, Pyriochou A, Altaany Z, Yang G, Marazioti A, Zhou Z, et al. Hydrogen sulfide is an endogenous stimulator of angiogenesis. Proc Natl Acad Sci U S A. 2009;106:21972–21977. doi: 10.1073/pnas.0908047106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Patel P, Vatish M, Heptinstall J, Wang R, Carson RJ. The endogenous production of hydrogen sulphide in intrauterine tissues. Reprod Biol Endocrinol. 2009;7:10. doi: 10.1186/1477-7827-7-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  101. Pawson AJ, Sharman JL, Benson HE, Faccenda E, Alexander SP, Buneman OP, et al. NC-IUPHAR. The IUPHAR/BPS Guide to PHARMACOLOGY: an expert-driven knowledgebase of drug targets and their ligands. Nucl. Acids Res. 2014;42:D1098–1106. doi: 10.1093/nar/gkt1143. (Database Issue): [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Podmore ID, Griffiths HR, Herbert KE, Mistry N, Mistry P, Lunec J. Vitamin C exhibits pro-oxidant properties. Nature. 1998;392:559. doi: 10.1038/33308. [DOI] [PubMed] [Google Scholar]
  103. Poston L, Briley AL, Seed PT, Kelly FJ, Shennan AH. Vitamin C and vitamin E in pregnant women at risk for pre-eclampsia (VIP trial): randomised placebo-controlled trial. Lancet. 2006;367:1145–1154. doi: 10.1016/S0140-6736(06)68433-X. [DOI] [PubMed] [Google Scholar]
  104. Ramesar SV, Mackraj I, Gathiram P, Moodley J. Sildenafil citrate decreases sFlt-1 and sEng in pregnant l-NAME treated Sprague-Dawley rats. Eur J Obstet Gynecol Reprod Biol. 2011;157:136–140. doi: 10.1016/j.ejogrb.2011.03.005. [DOI] [PubMed] [Google Scholar]
  105. Ramma W, Ahmed A. Is inflammation the cause of pre-eclampsia? Biochem Soc Trans. 2011;39:1619–1627. doi: 10.1042/BST20110672. [DOI] [PMC free article] [PubMed] [Google Scholar]
  106. Ramma W, Buhimschi IA, Zhao G, Dulay AT, Nayeri UA, Buhimschi CS, et al. The elevation in circulating anti-angiogenic factors is independent of markers of neutrophil activation in preeclampsia. Angiogenesis. 2012;15:341–348. doi: 10.1007/s10456-012-9261-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  107. Rana S, Karumanchi SA, Lindheimer MD. Angiogenic factors in diagnosis, management, and research in preeclampsia. Hypertension. 2014;63:198–202. doi: 10.1161/HYPERTENSIONAHA.113.02293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  108. Redman CW. Immunological aspects of pre-eclampsia. Baillieres Clin Obstet Gynaecol. 1992;6:601–615. doi: 10.1016/s0950-3552(05)80012-4. [DOI] [PubMed] [Google Scholar]
  109. Redman CW, Sargent IL. Latest advances in understanding preeclampsia. Science. 2005;308:1592–1594. doi: 10.1126/science.1111726. [DOI] [PubMed] [Google Scholar]
  110. Redman CW, Sacks GP, Sargent IL. Preeclampsia: an excessive maternal inflammatory response to pregnancy. Am J Obstet Gynecol. 1999;180:499–506. doi: 10.1016/s0002-9378(99)70239-5. [DOI] [PubMed] [Google Scholar]
  111. Rizos D, Eleftheriades M, Batakis E, Rizou M, Haliassos A, Hassiakos D, et al. Levels of asymmetric dimethylarginine throughout normal pregnancy and in pregnancies complicated with preeclampsia or had a small for gestational age baby. J Matern Fetal Neonatal Med. 2012;25:1311–1315. doi: 10.3109/14767058.2011.632037. [DOI] [PubMed] [Google Scholar]
  112. Rizzo G, Arduini D, Romanini C. Uterine artery Doppler velocity waveforms in twin pregnancies. Obstet Gynecol. 1993;82:978–983. [PubMed] [Google Scholar]
  113. Roberge S, Nicolaides KH, Demers S, Villa P, Bujold E. Prevention of perinatal death and adverse perinatal outcome using low-dose aspirin: a meta-analysis. Ultrasound Obstet Gynecol. 2013;41:491–499. doi: 10.1002/uog.12421. [DOI] [PubMed] [Google Scholar]
  114. Roberts JM, Hubel CA. The two stage model of preeclampsia: variations on the theme. Placenta. 2009;30(Suppl. A):S32–S37. doi: 10.1016/j.placenta.2008.11.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Roberts JM, Taylor RN, Musci TJ, Rodgers GM, Hubel CA, McLaughlin MK. Preeclampsia: an endothelial cell disorder. Am J Obstet Gynecol. 1989;161:1200–1204. doi: 10.1016/0002-9378(89)90665-0. [DOI] [PubMed] [Google Scholar]
  116. Robinson H, Wray S. A new slow releasing, H(2)S generating compound, GYY4137 relaxes spontaneous and oxytocin-stimulated contractions of human and rat pregnant myometrium. PLoS ONE. 2012;7:e46278. doi: 10.1371/journal.pone.0046278. [DOI] [PMC free article] [PubMed] [Google Scholar]
  117. Rossi AC, Mullin PM. Prevention of pre-eclampsia with low-dose aspirin or vitamins C and E in women at high or low risk: a systematic review with meta-analysis. Eur J Obstet Gynecol Reprod Biol. 2011;158:9–16. doi: 10.1016/j.ejogrb.2011.04.010. [DOI] [PubMed] [Google Scholar]
  118. Rubbo H, Batthyany C, Radi R. Nitric oxide-oxygen radicals interactions in atherosclerosis. Biol Res. 2000;33:167–175. doi: 10.4067/s0716-97602000000200017. [DOI] [PubMed] [Google Scholar]
  119. Sankaralingam S, Xu H, Davidge ST. Arginase contributes to endothelial cell oxidative stress in response to plasma from women with preeclampsia. Cardiovasc Res. 2010;85:194–203. doi: 10.1093/cvr/cvp277. [DOI] [PubMed] [Google Scholar]
  120. Schiff E, Barkai G, Ben-Baruch G, Mashiach S. Low-dose aspirin does not influence the clinical course of women with mild pregnancy-induced hypertension. Obstet Gynecol. 1990;76:742–744. doi: 10.1097/00006250-199011000-00002. [DOI] [PubMed] [Google Scholar]
  121. Sidhu R, Singh M, Samir G, Carson RJ. L-cysteine and sodium hydrosulphide inhibit spontaneous contractility in isolated pregnant rat uterine strips in vitro. Pharmacol Toxicol. 2001;88:198–203. doi: 10.1034/j.1600-0773.2001.d01-104.x. [DOI] [PubMed] [Google Scholar]
  122. Sidle EH, Casselman R, Smith GN. Effect of cigarette smoke on placental antioxidant enzyme expression. Am J Physiol Regul Integr Comp Physiol. 2007;293:R754–R758. doi: 10.1152/ajpregu.00505.2006. [DOI] [PubMed] [Google Scholar]
  123. Sikorski EM, Hock T, Hill-Kapturczak N, Agarwal A. The story so far: molecular regulation of the heme oxygenase-1 gene in renal injury. Am J Physiol Renal Physiol. 2004;286:F425–F441. doi: 10.1152/ajprenal.00297.2003. [DOI] [PubMed] [Google Scholar]
  124. Smith GC, Pell JP, Walsh D. Pregnancy complications and maternal risk of ischaemic heart disease: a retrospective cohort study of 129,290 births. Lancet. 2001;357:2002–2006. doi: 10.1016/S0140-6736(00)05112-6. [DOI] [PubMed] [Google Scholar]
  125. Sugimoto H, Hamano Y, Charytan D, Cosgrove D, Kieran M, Sudhakar A, et al. Neutralization of circulating vascular endothelial growth factor (VEGF) by anti-VEGF antibodies and soluble VEGF receptor 1 (sFlt-1) induces proteinuria. J Biol Chem. 2003;278:12605–12608. doi: 10.1074/jbc.C300012200. [DOI] [PubMed] [Google Scholar]
  126. Tao BB, Liu SY, Zhang CC, Fu W, Cai WJ, Wang Y, et al. VEGFR2 functions as an H2S-targeting receptor protein kinase with its novel Cys1045-Cys1024 disulfide bond serving as a specific molecular switch for hydrogen sulfide actions in vascular endothelial cells. Antioxid Redox Signal. 2013;19:448–464. doi: 10.1089/ars.2012.4565. [DOI] [PMC free article] [PubMed] [Google Scholar]
  127. Tenhunen R, Marver HS, Schmid R. Microsomal heme oxygenase. Characterization of the enzyme. J Biol Chem. 1969;244:6388–6394. [PubMed] [Google Scholar]
  128. Teupe C, Richter S, Fisslthaler B, Randriamboavonjy V, Ihling C, Fleming I, et al. Vascular gene transfer of phosphomimetic endothelial nitric oxide synthase (S1177D) using ultrasound-enhanced destruction of plasmid-loaded microbubbles improves vasoreactivity. Circulation. 2002;105:1104–1109. doi: 10.1161/hc0902.104720. [DOI] [PubMed] [Google Scholar]
  129. Tsukatani E. Etiology of EPH-gestosis from the viewpoint of dynamics of vasoactive prostanoid, lipid peroxides and vitamin E. Nihon Sanka Fujinka Gakkai Zasshi. 1983;35:713–720. [PubMed] [Google Scholar]
  130. Velauthar L, Plana MN, Kalidindi M, Zamora J, Thilaganathan B, Illanes SE, et al. First-trimester uterine artery Doppler and adverse pregnancy outcome: a meta-analysis involving 55,974 women. Ultrasound Obstet Gynecol. 2014;43:500–507. doi: 10.1002/uog.13275. [DOI] [PubMed] [Google Scholar]
  131. Venkatesha S, Toporsian M, Lam C, Hanai J, Mammoto T, Kim YM, et al. Soluble endoglin contributes to the pathogenesis of preeclampsia. Nat Med. 2006;12:642–649. doi: 10.1038/nm1429. [DOI] [PubMed] [Google Scholar]
  132. Villa PM, Kajantie E, Raikkonen K, Pesonen AK, Hamalainen E, Vainio M, et al. Aspirin in the prevention of pre-eclampsia in high-risk women: a randomised placebo-controlled PREDO Trial and a meta-analysis of randomised trials. BJOG. 2013;120:64–74. doi: 10.1111/j.1471-0528.2012.03493.x. [DOI] [PubMed] [Google Scholar]
  133. Wallenburg HC, Dekker GA, Makovitz JW, Rotmans P. Low-dose aspirin prevents pregnancy-induced hypertension and pre-eclampsia in angiotensin-sensitive primigravidae. Lancet. 1986;1:1–3. doi: 10.1016/s0140-6736(86)91891-x. [DOI] [PubMed] [Google Scholar]
  134. Wang K, Ahmad S, Cai M, Rennie J, Fujisawa T, Crispi F, et al. Dysregulation of hydrogen sulfide producing enzyme cystathionine gamma-lyase contributes to maternal hypertension and placental abnormalities in preeclampsia. Circulation. 2013;127:2514–2522. doi: 10.1161/CIRCULATIONAHA.113.001631. [DOI] [PubMed] [Google Scholar]
  135. Whiteman M, le Trionnaire S, Chopra M, Fox B, Whatmore J. Emerging role of hydrogen sulfide in health and disease: critical appraisal of biomarkers and pharmacological tools. Clin Sci (Lond) 2011;121:459–488. doi: 10.1042/CS20110267. [DOI] [PubMed] [Google Scholar]
  136. Wickens D, Wilkins MH, Lunec J, Ball G, Dormandy TL. Free radical oxidation (peroxidation) products in plasma in normal and abnormal pregnancy. Ann Clin Biochem. 1981;18:158–162. doi: 10.1177/000456328101800306. [DOI] [PubMed] [Google Scholar]
  137. Wong RJ, Zhao H, Stevenson DK. A deficiency in haem oxygenase-1 induces foetal growth restriction by placental vasculature defects. Acta Paediatr. 2012;101:827–834. doi: 10.1111/j.1651-2227.2012.02729.x. [DOI] [PubMed] [Google Scholar]
  138. Xenos ES, Stevens SL, Freeman MB, Cassada DC, Goldman MH. Nitric oxide mediates the effect of fluvastatin on intercellular adhesion molecule-1 and platelet endothelial cell adhesion molecule-1 expression on human endothelial cells. Ann Vasc Surg. 2005;19:386–392. doi: 10.1007/s10016-005-0011-7. [DOI] [PubMed] [Google Scholar]
  139. Yachie A, Niida Y, Wada T, Igarashi N, Kaneda H, Toma T, et al. Oxidative stress causes enhanced endothelial cell injury in human heme oxygenase-1 deficiency. J Clin Invest. 1999;103:129–135. doi: 10.1172/JCI4165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  140. Yan H, Du J, Tang C. The possible role of hydrogen sulfide on the pathogenesis of spontaneous hypertension in rats. Biochem Biophys Res Commun. 2004;313:22–27. doi: 10.1016/j.bbrc.2003.11.081. [DOI] [PubMed] [Google Scholar]
  141. Yanfei W, Lin S, Junbao D, Chaoshu T. Impact of L-arginine on hydrogen sulfide/cystathionine-gamma-lyase pathway in rats with high blood flow-induced pulmonary hypertension. Biochem Biophys Res Commun. 2006;345:851–857. doi: 10.1016/j.bbrc.2006.04.162. [DOI] [PubMed] [Google Scholar]
  142. Yang G, Wu L, Jiang B, Yang W, Qi J, Cao K, et al. H2S as a physiologic vasorelaxant: hypertension in mice with deletion of cystathionine gamma-lyase. Science. 2008;322:587–590. doi: 10.1126/science.1162667. [DOI] [PMC free article] [PubMed] [Google Scholar]
  143. You XJ, Xu C, Lu JQ, Zhu XY, Gao L, Cui XR, et al. Expression of cystathionine beta-synthase and cystathionine gamma-lyase in human pregnant myometrium and their roles in the control of uterine contractility. PLoS ONE. 2011;6:e23788. doi: 10.1371/journal.pone.0023788. [DOI] [PMC free article] [PubMed] [Google Scholar]
  144. Zanardo RC, Brancaleone V, Distrutti E, Fiorucci S, Cirino G, Wallace JL. Hydrogen sulfide is an endogenous modulator of leukocyte-mediated inflammation. FASEB J. 2006;20:2118–2120. doi: 10.1096/fj.06-6270fje. [DOI] [PubMed] [Google Scholar]
  145. Zhao H, Wong RJ, Doyle TC, Nayak N, Vreman HJ, Contag CH, et al. Regulation of maternal and fetal hemodynamics by heme oxygenase in mice. Biol Reprod. 2008;78:744–751. doi: 10.1095/biolreprod.107.064899. [DOI] [PubMed] [Google Scholar]
  146. Zhao H, Wong RJ, Kalish FS, Nayak NR, Stevenson DK. Effect of heme oxygenase-1 deficiency on placental development. Placenta. 2009;30:861–868. doi: 10.1016/j.placenta.2009.07.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  147. Zhao W, Zhang J, Lu Y, Wang R. The vasorelaxant effect of H(2)S as a novel endogenous gaseous K(ATP) channel opener. EMBO J. 2001;20:6008–6016. doi: 10.1093/emboj/20.21.6008. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from British Journal of Pharmacology are provided here courtesy of The British Pharmacological Society

RESOURCES