Placental development in normal and compromised pregnancies-- a review

Share Embed


Descripción

Placenta (2002), 23, Supplement A, Trophoblast Research, 16, S119–S129 doi:10.1053/plac.2002.0792, available online at http://www.idealibrary.com on

Placental Development in Normal and Compromised Pregnancies— A Review T. R. H. Regnaulta, H. L. Galanb, T. A. Parkera and R. V. Anthonya,c,d a Department of Pediatrics, b Department of Obstetrics and Gynecology, University of Colorado Health Sciences Center, Denver, CO 80262 and c Department of Physiology, Colorado State University, Fort Collins, CO 80523, USA

Intrauterine growth restriction (IUGR) is a significant cause of infant mortality and morbidity. It is now clear that IUGR infants exhibit higher rates of coronary heart disease, type 2-diabetes, hypertension and stroke as adults. Therefore, fetal growth not only impacts the outcome of the perinatal period, but also impacts adult well-being. The etiologies of IUGR are numerous, but are often associated with abnormalities in placental structure and function. The process of implantation and placentation requires the production of a plethora of growth factors, cell-adhesion molecules, extracellular matrix proteins, hormones and transcription factors. Many of these exhibit altered expression within the placenta of IUGR pregnancies. However, it has been difficult to fully assess their role during the development of placental insufficiency (PI) in the human, underscoring the need for animal models. Using an ovine model of PI-IUGR we have observed changes in the expression of vascular endothelial growth factor, placental growth factor, their common receptors, as well as angiopoietin 2 and its receptor, Tie 2. We found that changes in these growth factors can be associated with both acute and chronic changes in placental vascular structure and function. These studies and others are providing needed insight into the developmental chronology of placental insufficiency. Placenta (2002), 23, Supplement A, Trophoblast Research 16, S119–S129  2002 IFPA and Elsevier Science Ltd

INTRODUCTION Intrauterine growth restriction (IUGR) is a significant cause of infant mortality and morbidity, affecting upwards to 8 per cent of all pregnancies (Brar and Rutherford, 1988; Pollack and Divon, 1992). Complications arising during IUGR pregnancies include: (1) intrauterine demise, (2) intrapartum fetal distress, and (3) perinatal asphyxia. In IUGR infants, neonatal complications include: meconium aspiration, metabolic and hematologic disturbances, cognitive dysfunction and cerebral palsy. Furthermore, epidemiological studies provide compelling evidence that IUGR predisposes individuals for coronary heart disease, hypertension, stroke and diabetes during adulthood (Barker et al., 1989, 1990, 1993a,b). While IUGR fetuses can be categorized into symmetric or asymmetric patterns of growth, the majority of cases present an asymmetric pattern of growth, often associated with abnormalities in placental structure and function. The placenta is a multifaceted organ that plays critical roles in maintaining and protecting the developing fetus. These roles include transferring nutrients from the mother to the fetus and waste secretion from the fetus to the mother, acting as a barrier for the fetus against pathogens and the maternal immune system, and serving as an active endocrine organ d

To whom correspondence should be addressed at: ARBL-Foothills Campus, Department of Physiology, Colorado State University, Fort Collins, CO 80523-1683, USA. Tel: 970-491-2586; Fax: 970-4913557; E-mail: [email protected] 0143–4004/02/0AS119+11 $35.00/0

capable of synthesizing and secreting a plethora of hormones, growth factors, cytokine and other bioactive products. A number of placental structural abnormalities have been associated with IUGR pregnancies, including a decrease in villous number, diameter and surface area, as well as a decrease in arterial number, lumen size and branching (Giles, Trudinger and Baird, 1985; Krebs et al., 1996; Lee and Yeh, 1986; Macara et al., 1996; Salafia et al., 1997). All of these abnormalities can adversely affect placental function, and ultimately deprive the developing fetus of the nutrients required for optimal growth.

PLACENTAL DEVELOPMENT The process of implantation and placentation requires the production of numerous angiogenic growth factors, celladhesion molecules, cytokines and growth factors, extracellular matrix metalloproteinases, hormones and transcription factors. During normal human pregnancy, extravillous trophoblast (EVT) cells migrate and invade the spiral artery vessel walls within the decidua and myometrium (Brosens, Robertson and Dixon, 1967; Meekins et al., 1994), but in pregnancies complicated by pre-eclampsia and/or IUGR, this invasion is largely restricted to the decidual portion of the spiral arteries, and failure of these arteries to become low resistance vessels (Meekins et al., 1994; Redline and Patterson, 1995; Roberson,  2002 IFPA and Elsevier Science Ltd

S120

Brosens and Dixon, 1975). A number of factors have been associated with EVT invasion, including angiogenic growth factors and their respective receptors. While it remains unresolved as to the exact mechanism causing deficient trophoblast invasion, angiogenic growth factors are logical candidates as regulatory molecules in placental development and function.

Fetal placenta angiogenesis A number of angiogenic growth factors have been identified in human placenta, including: basic fibroblast growth factor (bFGF; Crescimanno et al., 1995; Shams and Ahmed, 1994), hepatocyte growth factor (HGF; Kilby et al., 1996), placenta growth factor (PlGF; Shore et al., 1997; Vuorela et al., 1997) and vascular endothelial growth factor (VEGF; Ahmed et al., 1995; Sharkey et al., 1993; Shore et al., 1997; Vuorela et al., 1997). The later two (VEGF and PlGF) belong to a family of angiogenic growth factors that include VEGF (also called VEGF-A), PlGF, VEGF-B (Olofsson et al., 1996), VEGF-C (Juokov et al., 1996), VEGF-D (Yamada et al., 1997) and VEGF-E (Lyttle et al., 1994). While VEGF, PlGF, VEGF-B and VEGF-C are all expressed in the human placenta (Vuorela et al., 1997; Dunk and Ahmed, 2001), previous investigations have focused primarily on VEGF-A and PlGF. VEGF-A exists as multiple isoforms (VEGF121, VEGF145, VEGF165, VEGF189 and VEGF206) that result from alternative splicing of the VEGF primary transcript (Dvorak et al., 1995). Similar alternative splicing of the other family members (e.g. PlGF-1 and PlGF-2) also occurs. Beyond the multiple ligands in this family, there are several receptors capable of binding members of this growth factor family. These include: fms-like tyrosine kinase-1 (Flt-1 or VEGFR-1) and a soluble form of VEGFR-1 (sVEGFR-1; Thomas, 1996), kinase insert domain-containing receptor (KDR, Flk-1 or VEGFR-2; Thomas, 1996), fms-like tyrosine kinase-4 (Flt-4 or VEGFR-3; Joukov et al, 1996) and neuropilin-1 and -2 (Gluzman-Poltorak et al., 2000). While VEGF expression occurs in the villous trophoblast (Ahmed et al., 1995; Sharkey et al., 1993; Vuorela et al., 1997), PlGF expression appears confined to villous syncytiotrophoblast (Shore et al., 1997; Vuorela et al., 1997). Both VEGFR-1 and -2 are expressed in placental villous endothelium (Charnock-Jones et al., 1994; Vuckovic et al., 1996), but VEGFR-1 is also expressed in EVT (Ahmed et al., 1995). VEGFR-1 binds homodimers of VEGF, homodimers of PlGF, or VEGF/PlGF heterodimers, whereas VEGFR-2 binds only VEGF homodimers or VEGF/PlGF heterodimers (Thomas, 1996). When VEGF binds VEGFR-2, rather than VEGFR-1, efficient mitogenic and chemotactic responses occur with endothelial cells. However, the lack of a potent mitogenic response from PlGF binding VEGFR-1 led to the suggestion that it is ineffective in mediating endothelial cell mitogenesis (Thomas, 1996). Furthermore, the binding of VEGF/PlGF heterodimers to VEGFR-2 led to the suggestion that PlGF may diminish VEGF stimulated angiogenesis through heterodimer formation (Cao et al., 1996; DiSalvo et al., 1995).

Placenta (2002), Vol. 23, Supplement A, Trophoblast Research, Vol. 16

However, the presence of functional VEGFR-1 receptors on isolated human trophoblast suggests that VEGF or PlGF binding to trophoblast VEGFR-1 may play a role in EVT invasion and differentiation (Ahmed et al., 1995; Shore et al., 1997). Supporting this hypothesis is the fact that early placental development occurs in a hypoxic environment (Kingdom and Kaufmann, 1999), a known stimulator of VEGF expression (Schweiki et al., 1992; Stavri et al., 1995). Indeed, VEGF expression by placental tissues is upregulated by hypoxia (Shore et al., 1997; Wheeler, Elcock and Anthony, 1995) and downregulated by hyperoxia (Shore et al., 1997), whereas the opposite appears to occur with PlGF (Shore et al., 1997; Khaliq et al., 1999). Hypoxic regulation of VEGF expression is mediated at both the transcriptional and post-transcriptional levels (Figure 1). Increased VEGF mRNA stability during low oxygen tension is an important post-transcriptional response (Levy et al., 1998; Shima, Deutsch and D’Amore, 1995; Shih and Claffey, 1999). A hypoxia stability region in the 3 -untranslated region of the human and bovine VEGF mRNA is bound by heterogeneous nuclear ribonuceloprotein L (hnRNP L), stabilizing the mRNA (Shih and Claffey, 1999), thereby increasing its half-life. Additionally, hypoxia inducible factor-1 (HIF-1) is a basic helix-loop-helix transcription factor comprised of two subunits (HIF-1 and HIF-1) that interacts with a ‘hypoxic response element (HRE)’ in genes induced by hypoxia (Wang et al., 1995). HIF-1 is constitutively expressed, whereas HIF-1 is regulated (Figure 1) by targeted proteolysis by the von Hippel-Lindau tumor suppressor gene product (pVHL; Maxwell et al., 1999). In the presence of oxygen, HIF-1 is degraded (Maxwell et al., 1999), but under hypoxic conditions it is stabilized allowing functional HIF-1 to act via the HRE in genes such as VEGF (Figure 1). While the expression of hnRNP L and HIF-1 have not been described in preeclamptic or IUGR placenta, Caniggia et al. (2000a) reported HIF-1 expression to be high at 5–7 weeks of gestation, decreasing around 9 weeks when placental O2 tension is believed to increase, and HIF-1 was absent at 11–14 weeks. Additionally, these investigators found that transforming growth factor 3 (TGF3) expression parallels that of HIF-1, and treatment of placental villous explants with HIF-1 anti-sense oligonucleotides inhibits the expression of both HIF-1 and TGF3. From these results and others, Caniggia et al. (2000b) suggest that the developmental increase in placental O2 tension reduces HIF-1 expression, which in turn reduces TGF3 expression, thereby removing TGF3’s inhibition of trophoblast differentiation along the invasive pathway. They also suggest that a failure in this developmental switch would arrest EVT at an immature stage, resulting in a shallow invasion of the uterus that would lead to reduced utero-placental perfusion. The basic helix-loop-helix transcriptioin factor HIF-2 is also capable of heterodimerizing with HIF-1 to form functional HIF-1 (Figure 1), and was recently found to be in greater abundance in late-gestational placenta from preeclamptic pregnancies (Rajakumar et al., 2001). In these

Regnault et al.: Placental Development in IUGR Pregnancies

Normoxia

S121

HRE

HIF-1α

HIF-1β

VEGF

HIF-1β

HIF-1α

pVHL HIF-2α

Hypoxia

HIF-2α

HRE

VEGF

hnRNP-L Figure 1. Schematic representation of the regulation of VEGF gene transcription and mRNA stability. Hypoxia inducible factor-1 (HIF-1) or HIF-2 combine with HIF-1 to form the functional basic helix-loop-helix transcription factor HIF-1 to stimulate transcription of the VEGF gene, via interaction with the hypoxia response element (HRE). This occurs under hypoxic conditions, but is down regulated under normoxic conditions due to targeted proteolysis of HIF-1 and HIF-2, mediated by the von Hippel-Lindau tumor suppressor gene product (pVHL). Additionally, VEGF is regulated post-transcriptionally through stabilization of its mRNA by interaction with heterogeneous nuclear ribonuceloprotein L (hnRNP L).

studies, the concentration of HIF-1 and HIF-2 mRNA was not altered by pre-eclampsia, just the amount of HIF-2 protein. Such findings indicate that both HIF-1 and HIF-2 are important factors that appear to have a role in abnormal placental development, and that primary regulation of these transcription factors is at the protein level (Rajakumar and Conrad, 2000). Recent evidence that pVHL abundance is greater within hypoxic regions of the placenta, and is increased under hypoxic culture conditions of chorionic villi explants, along with HIF-2 indicates a complex regulation (Figure 1) of HIF-1 and HIF-2 by pVHL (Genbacev et al., 2001). While pVHL targets HIF-1 and HIF-2 for proteosome degradation (Maxwell et al., 1999), it appears that pVHL itself may be finely regulated by hypoxia (Genbacev et al., 2001) such that shuttling of HIF-1 back and forth between the cytoplasm and nucleus, coupled with cytoplasmic targeting for proteolysis, provides the exacting control of hypoxia-mediated gene expression.

Angiogenic growth factors in compromised pregnancies The concept that a lack of EVT invasion of the spiral arteries (Meekins et al., 1994; Redline and Patterson, 1995; Roberson, Brosens and Dixon, 1975) results in placental ischemia and the development of pre-eclampsia and IUGR, led to the investigation of VEGF/PlGF expression in these placental pathologies. Kingdom and Kaufmann (1997) suggested that three distinct types of hypoxia may occur in the feto-placental unit. These include: (1) Pre-placental hypoxia, where the mother, placenta and fetus are hypoxia as may occur at high altitude; (2) Utero-placental hypoxia, where maternal oxygenation is

normal but impaired utero-placental circulation results in placental and fetal hypoxia with preserved end diastolic flow in the umbilical arteries as in pre-eclampsia; and (3) Postplacental hypoxia, where only the fetus is believed to be hypoxic, with reduced, absent or reversed end diastolic flow of the umbilical arteries. Another way of classifying these pregnancies resulting in IUGR, is early- or late-onset IUGR (Kingdom et al., 2000), with late-onset generally encompassing the pre-placental and utero-placental hypoxic groups. In pre-eclamptic pregnancies, placental VEGF was reduced (Cooper et al., 1996; Lyall et al., 1997). These results do not coincide with an expectation of placental hypoxia stimulating VEGF expression. The reason VEGF expression is reduced in these pre-eclamptic pregnancies (Cooper et al., 1996; Lyall et al., 1997) is not readily apparent, but suggests the placenta may not always be able to respond to hypoxic conditions with increased VEGF production. However, supporting the concept that pre-eclamptic placenta are hypoxic is the reduction of PlGF in maternal blood observed in pre-eclamptic pregnancies (Torry et al., 1998; Tidwell et al., 2001). In cases of post-placental hypoxia associated with early-onset IUGR, the placental villi are believed to be exposed to greater oxygen tension or ‘placental hyperoxia’ (Kingdom and Kaufmann, 1997). This concept is supported by the observation that the oxygen content of uterine venous blood is close to arterial values in severe IUGR pregnancies (Pardi et al., 1992), and that VEGF expression is reduced and PlGF expression is increased (Lyall et al., 1997; Khaliq et al., 1999). Regulation of angiogenic growth factor expression by hypoxia or hyperoxia involves numerous levels of control, and to fully understand how growth factors like VEGF and PlGF may act in the development of compromised placental

S122

function, interaction with their receptors and/or other factors must also be considered. For example, we do not know what happens to VEGFR-1 and -2 expression at various times during the development of pre-eclamptic or IUGR pregnancies. In addition to the two more common receptors for VEGF and PlGF, neuropilin-1 and -2 (NP-1, NP-2) are in human umbilical vein endothelial cells (Gluzman-Poltorak et al., 2000), and while they have not been localized to the placenta proper yet, it would not be surprising that they are also involved in placenta vascular regulation. This could add another layer of regulation to the actions of VEGF and PlGF, since NP-1 and NP-2 act as receptors for VEGF165 and PlGF-2 but only NP-2 acts as a receptor for VEGF145 (Gluzman-Poltorak et al., 2000; Migdal et al., 1998). NP-1 not only binds VEGF165, but it also complexes with VEGFR-1 (Fuh, Garcia and deVos, 2000), which led to the hypothesis that VEGFR-1 acts as a negative regulator of angiogenesis by competing for NP-1. To further understand the regulation by endothelial growth factors, one must consider the down-stream effects of VEGF and PlGF interaction with the various receptors that might influence placental circulation. Expression of endothelial nitric oxide synthase (eNOS; Kroll and Waltenberger, 1998) and angiopoietin 2 (Ang2; Oh et al., 1999) are stimulated by VEGF. In late gestation human pregnancies complicated by pre-eclampsia or IUGR, eNOS expression within the syncytiotrophoblast is reported to increase (Myatt et al., 1997), as is the nitric oxide concentration of umbilical venous blood (Lyall et al., 1996). This may be a compensatory response to improve blood flow within the IUGR placenta. However, these data do not fit with the reports of depressed VEGF expression in IUGR placenta (Khaliq et al., 1999; Lyall et al., 1997). This apparent discrepancy between VEGF and eNOS expression may result from the type of IUGR present in these studies; i.e., did the various placentae in these reports exhibit uteroplacental hypoxia or post-placental hypoxia (see discussion above)? Alternatively, different pathways for VEGF and eNOS regulation may be in play. In addition to eNOS, both hypoxia and VEGF stimulate Ang 2 expression (Oh et al., 1999), and Ang 2 stimulates the release of NO from human trophoblast cells in vitro (Dunk et al., 2000). Angiopoietin 1 (Ang1) has been localized in the cyto/syncytiotrophoblast bilayer in firsttrimester human placenta (Dunk et al., 2000), Ang2 was found in the cytotrophoblast layer (Dunk et al., 2000) or in the syncytiotrophoblast (Goldman-Wohl et al., 2000), and their common receptor (Tie2) was found in the cyto/ syncytiotrophoblast bilayer (Dunk et al., 2000) and in endovascular invasive trophoblasts (Goldman-Wohl et al., 2000). While Ang1 acts synergistically with VEGF (Koblizek et al., 1998) to stimulate angiogenesis, Ang 2 is an antagonist to Ang1 and in the absence of VEGF it is thought to destabilize or regress blood vessels (Maisonpierre et al., 1997). At term, in severe IUGR placenta (Dunk et al., 2000), Ang 2 mRNA concentrations were not different from normal placenta, but the amount of Ang 2 protein appeared to be significantly decreased. Reduction in placental Ang 2 (Dunk et al., 2000),

Placenta (2002), Vol. 23, Supplement A, Trophoblast Research, Vol. 16

coupled with reduced VEGF expression and increased PlGF expression (Lyall et al., 1997; Khaliq et al., 1999), fits with the concept of post-placental hypoxia in severe or early-onset IUGR. Unfortunately, the studies examining vascular growth factors and their regulation or function in compromised pregnancies have, by necessity, utilized late gestational or term tissues. While these studies are extremely valuable, they do not provide insight into the role of these factors and receptors in the progressive development of a compromised placenta. Furthermore, with most of the human pregnancies examined we do not know what is being supplied to or being utilized by the utero-placental unit. Consequently, animal models of IUGR are valuable for providing a better understanding of the changes that likely take place in the development and progression of placental insufficiency leading to IUGR.

ANIMAL MODELS OF IUGR A variety of methods exist for creating IUGR pregnancies in several animal species. Of the species used for IUGR studies, the pregnant sheep provides several advantages as an animal model to examine the development and progression of IUGR pregnancies, not the least of which is the ability to instrument both mother and fetus for nutrient transfer and utilization studies. Growth restriction in fetal sheep can be induced by nutrient restriction, nutrient excess, placental embolization with microspheres, limitation of placental implantation sites, umbilical artery ligation, administration of corticosteroids and exposure to elevated environmental temperatures. Of all models of IUGR, sheep exposed to high ambient temperatures develop the most severe IUGR (Alexander and Williams, 1971; Bell, Wilkening and Meschia, 1987; Thureen et al., 1992). Exposure of pregnant ewes to hyperthermic conditions for approximately 80 days (40–120 days post coitus; dpc) results in an IUGR fetus whose placenta is also growth retarded (Bell, Wilkening and Meschia, 1987; Thureen et al., 1992). Similar results are obtained when exposure is for only 55 days (37–93 dpc), with significant reductions in fetal and placental weights (Galan et al., 1999; Regnault et al., 1999). Fetal growth in these pregnancies is asymmetric in nature, as evidenced by greater biparietal diameter/abdominal circumerance ratios (Galan et al., 1999). Furthermore, current evidence indicates that development of IUGR in chronically hyperthermic ewes occurs as a consequence of primary reduction in placental growth in early gestation (Bell et al., 1989; Galan et al., 1999; Thureen et al., 1992), making this an excellent model to examine normal and impaired placental development, i.e., placental insufficiency (PI) that results in IUGR.

Placental development in sheep While structural differences exist between human and sheep placenta, there are important similarities in function and

Regnault et al.: Placental Development in IUGR Pregnancies

functional structure. A primary structural difference in the placenta of these two species occurs as a result of raised areas of non-glandular but well vascularized endometrium, termed caruncles, that are always present within the uterus of sheep (Boyd and Hamilton, 1952; Stegeman, 1974). Intimate association of developing chorion over these raised surfaces establishes the foundation for placental development. In areas of caruncular projections, fetal vascularizatioin of the placenta is derived from the vascularized allantois, following its expansion from the hind-gut at 15 dpc allowing contact and fusion with the chorion (Stegeman, 1974). Early in the fourth week of pregnancy, interdigitation of embryonic and maternal tissues becomes apparent (Boshier, 1969), and in areas of close association between chorion and the caruncular surface, a series of ridges and grooves form which eventually give rise to fetal villi of the cotyledon (Steven, 1975). The maternal caruncles continue to grow and develop deeply branched crypts into which the cotyledonary villi project, elongate and branch forming an apposing network of fetal villi within the maternal crypts (Steven, 1975). Maximum surface area obtained by branching of the villi appears to be regulated by the size of the placentome (caruncle plus cotyledon), which is directly correlated to uterine weight in normal sheep (Stegeman, 1974). While the placenta of sheep can be divided into numerous discreet attachment sites, within each placentome the villous tree is structurally similar to that which arises in the human, as both can be divided into stem, intermediate, and terminal villi (Leiser et al., 1997; Kaufmann, Sen and Schweikhart, 1979; Kaufmann, 1982). Furthermore, the structure of fetal vessels (stem arteries and veins, intermediate arterioles and venules, and terminal capillaries) of sheep and humans are comparable (Leiser et al., 1997), allowing the sheep to serve as a model of fetal placenta vascularization throughout the period of placental development.

PI-IUGR in sheep The natural phenomenon of hyperthermia-induced PI-IUGR in sheep allows the opportunity to study PI-IUGR in a commonly used fetal physiology model. Early studies (Alexander and Williams, 1971) examined the effects of exposure to heat at various stages of gestation on placental and fetal outcomes. Pregnant ewes exposed to hyperthermic conditions in mid-gestation (50–100 dpc), mid-gestation through term (50–150 dpc), as well as only in the final third of gestation (100–150 dpc), resulted in PI-IUGR. The decrease in placental mass associated with hyperthermia during the middle and last third of gestation has been implicated as the underlying cause of IUGR (Galan et al., 1999; Regnault et al., 1999). Indeed, placental size has been linked with size of the fetus late in pregnancy when fetal demands are highest (Mellor and Murray, 1981, 1982), but placental size is a crude indicator that does not address the mechanisms responsible for IUGR. Studies of the PI-IUGR sheep model show that it has characteristics in common with human IUGR. For example,

S123

ewes exposed to hyperthermic conditions from 39–125 dpc had fetal and placental masses roughly one-half of their thermoneutral counterparts (1.6 vs. 3.1 kg; 0.15 vs 0.36 kg, respectively) at 135 dpc (Thureen et al., 1992). These fetuses were hypoxic and hypoglycemic, and the fetal hypoglycemia resulted in a greater maternal : fetal glucose gradient (Thureen et al., 1992). Similar results from human IUGR pregnancies have been reported (Economides and Nicolaides, 1989; Pardi et al., 1993; Nieto-Diaz et al., 1996). Transport of two essential amino acids, leucine and threonine, across the PI-IUGR placenta is also reduced, resulting in the routing of these two amino acids into fetal accretion and away from oxidation (Anderson et al., 1997; Ross et al., 1996). The reduction in transport is not simply a function of placental mass, since transport was still less when normalized on placental weight. A reduction in the fetal : maternal leucine molar percent enrichment has been observed in both sheep (Ross et al., 1996) and human (Marconi et al., 1999) IUGR. Reductions in fetal amino acid concentrations, reduced placental transport, and a reduction in the system A transporter in cases of human IUGR (Glazier et al., 1997; Jansson, Scholtbach and Powell, 1998; Marconi et al., 1999; Norberg, Powell and Jansson, 1998) agree with the results from sheep. Furthermore, reduced umbilical blood flow, and increased umbilical artery pulsatility index and placental vascular resistance have been reported for both human and sheep IUGR pregnancies (Galan et al., 1998; Thureen et al., 1992; Trudinger et al., 1987). Combined, these data highlight important functional similarities between human IUGR and sheep PI-IUGR pregnancies induced by hyperthermia (summarized in Figure 2), and provide evidence of true functional-placental insufficiency. When pregnant ewes are subjected to a hyperthermic (HT) environment (40C for 12 h/d and 35C for 12 h/d; 30–40 per cent relative humidity) or a thermoneutral (TN) environment (20C) beginning at 40 dpc, the HT exposure results in a rapid (within 2–3 d) and sustained increase (HT 39.860.1 vs. TN 39.200.1C; Pc0.001) in core body temperature (CBT) (Regnault et al., 1999). Exposure to this regimen for either 55 or 80 days results in significant reductions in placental and fetal weights near term (Galan et al., 1999). Recently, we have collected tissues from HT and TN pregnancies following treatment for 15 days (55 dpc), 50 days (90 dpc) of HT, or 80 days followed by TN exposure for an additional 15 days (135 dpc). Placental weights tended to be smaller (Pc0.10) by 55 dpc, whereas it took until 90 dpc to see a reduction in fetal weights. Interestingly, at 55 dpc the fetal/placental weight ratio is greater (Pc0.01) in HT pregnancies. The fetal/ placental weight ratio as an indicator of placental function, suggests that placental function may be acutely enhanced in HT pregnancies. Due to the changes in umbilical Dopplar velocity wave forms observed in human IUGR (Krebs et al., 1996; Salafia et al., 1997; Trudinger et al., 1989) and in our sheep model of PI-IUGR (Galan et al., 1998), vascular architecture in HT and TN ewes was examined in the developing placenta by generating vascular erosion casts at 90 dpc (Figure 3). Fetal vessels

S124

Placenta (2002), Vol. 23, Supplement A, Trophoblast Research, Vol. 16

Morphology

Metabolic

Vasculature

Asymmetrical growth

HUMAN IUGR FETUS

OVINE IUGR FETUS

Yes

Yes

Placental weight





Fetal weight





Brain–liver ratio





Glucose gradient





Lactate concentration





Fetal amino acid concentrations





Leucine fetal/maternal MPE





Pulsatility index (umbilical artery)





Fetal hypoxia





Figure 2. Summary of the common features of IUGR pregnancies that have been described in the human and in a sheep model of IUGR induced by hyperthermic exposure of pregnant ewes. MPE=molar per cent enrichment.

from TN placentae (Figure 3, panel B) were relatively straight and organized parallel to one another. However, fetal vessels of HT placentae were coiled and appeared in a more random orientation relative to neighboring vessels (Figure 3, panel C). Additionally, sinusoidal dilations also appeared to be more numerous in HT fetal vessels (Figure 3, panel C). Similar architectural differences are observed in fetal placenta vascular casts of ewes kept at sea level (normoxia) with those of ewes kept at high altitude (hypoxia) between 45 and 140 dpc (Krebs, Longo and Leiser, 1997). Exposure of pregnant ewes to conditions of hypoxia increased the length and degree of coiling of fetal and maternal placental vasculature. These investigators concluded that the increase in vessel length and coiling occurred as an adaptation to supply more oxygen to the fetus. The similarity in these results may indicate that alterations in placental vasculature resulting from exposure of pregnant ewes to hypoxic or hyperthermic conditions may be mediated by the same mechanisms. Using the fetal placenta cotyledons and uterine caruncles collected at 55, 90 and 135 dpc from TN and HT pregnancies, we have characterized the expression of VEGF, PlGF and their receptors (VEGFR-1 and -2). At 55 dpc, there was a significant tissue by treatment interaction for VEGF mRNA expression, as a result of increased expression of cotyledonary VEGF in HT ewes only (Pc0.01). There was no difference in VEGF mRNA expression between treatments for caruncular tissue at 55 dpc, nor was mRNA expression different between treatments or tissue types at 90 dpc. The concentration of PlGF mRNA was not effected by treatment group or tissue type at either gestational age examined. With the 135 dpc tissues, we found a reduction (Pc0.01) in VEGF mRNA within fetal cotyledons, and an increase (Pc0.05) in maternal caruncle PlGF mRNA (summarized in Figure 4). The enhanced expression of VEGF in HT pregnancies at 55 dpc may be indicative of an increased state of hypoxia within the developing HT placenta, and could be involved in driving the altered vasculature presented in Figure 3. However, it is not known if the HT environment creates a more hypoxic

environment for early placental development, or if the increased CBT in HT ewes may directly stimulate VEGF expression. To better understand the functional significance of the changes observed in VEGF expression within HT pregnancies, we examined mRNA concentrations for the VEGF/ PlGF receptors, VEGFR-1 and -2. Both VEGFR-1 and -2 mRNA concentrations in cotyledonary tissue rose significantly with advancing gestational age (Pc0.05), but no effect of gestational age was observed in uterine caruncular tissue. No effect of the HT environment was observed for either receptor at 55 dpc in either fetal placental cotyledons or maternal uterine caruncles, or in maternal uterine caruncles at 90 dpc. However, both VEGFR-1 and -2 mRNA concentration in fetal cotyledons was significantly reduced at 90 dpc. At 135 dpc, only VEGFR-1 mRNA concentration was reduced in fetal cotyledons (Figure 4). Combined (Figure 4), the results of our examination of VEGF, PlGF and their receptors at various time-points in our IUGR pregnancies, point to differing acute and chronic responses to the HT environment (Figure 4). Acute up-regulation of VEGF expression may be a compensatory mechanism aimed at maintaining normal placental development, by enhancing angiogenesis. This appears to be borne out in preliminary histomorphometry data. Within the basal plate region of the placenta, the number of maternal vessels is reduced (30.83.38 vs. 15.02.49; TN vs. HT; Pc0.01). However, within the chorionic plate, the number of fetal vessels is increased (15.61.60 vs. 22.171.66; TN vs. HT; Pc0.05). A reduction in maternal placental angiogenesis may result in fetal placental hypoxia, thereby driving increased VEGF expression and accelerated fetal placental angiogenesis. Interestingly, we have preliminary data indicating that both VEGF and eNOS mRNA concentrations are reduced (Pc0.01) in fetal lung at 55 dpc. This could result if fetal PO2 was increased. Such a scenario might exist if increased fetal placental angiogenesis and vessel number resulted in increased O2 extraction by the fetal placenta. If ‘fetal hyperoxia’ existed during early to mid-gestation, followed by fetal hypoxia

Regnault et al.: Placental Development in IUGR Pregnancies

S125 Acute

Transitional

Chronic

VEGF







PIGF







VEGFR-1







VEGFR-2







Ang 1







Ang 2







Tie 2







VEGF







PIGF







VEGFR-1







VEGFR-2







Ang 1







Ang 2







Tie 2







Fetal Placenta

Maternal Placenta

Figure 4. Summary of changes in placental expression of vascular growth factors and their receptors in control and IUGR ovine pregnancies. Arrows indicate significant changes in the mRNA concentration within either maternal placental or fetal placental tissues, during acute (55 dpc), transitional (90 dpc) or chronic (135 dpc) progression of placental insufficiency. Horizontal lines indicate no significant changes.

Figure 3. Scanning electron micrographs of ovine fetal placental vascular casts. Panel A=low power magnification (100) of fetal placenta vasculature (90 dpc) extending from the intermediate zone (top) to the basal plate zone (bottom). Panel B=Fetal placenta vasculature from a control pregnancy, in the intermediate zone at higher magnification (1000) showing organized outgrowth of vessels which remain relatively straight. Panel C=Fetal placenta vasculature (1000 magnification) from an IUGR pregnancy in the intermediate zone demonstrating an increase in vessel coiling and overall disorganized arrangement of vessels.

(Thureen et al., 1992) during late-gestation, it is easy to envision how fetal organogenesis could be negatively impacted and could lead to fetal hypertension.

In contrast to our results following acute exposure to the HT environment, chronic exposure (50 days), or possibly a failure to adequately compensate, results in down-regulation of both receptors at 90 dpc. The consequences of the later may be an inability to respond to VEGF. The importance of the reduction in VEGFR-1 alone at 135 dpc is not clear. Since it is possible that the measured reduction in VEGFR-1 resulted from a reduction in the expression of the soluble form of VEGFR-1 mRNA, we tested our samples for the presence of sVEGFR-1 mRNA. As yet, we have not been able to detect mRNA encoding sVEGFR-1 within sheep placenta, suggesting that the observed changes result from VEGFR-1 transcripts. If only ‘full-length’ VEGFR-1 mRNA is reduced, this may be a mechanism to drive VEGF binding to VEGFR-2. This would be advantageous in regards to VEGFs ability to interact with a functional receptor, thereby stimulating angiogenesis in an attempt to overcome a deficit in placental vasculature. However, one must keep in mind that fetal placental VEGF mRNA concentration is reduced and maternal placental PlGF concentration is increased at 135 dpc. Consequently, stimulation of angiogenesis through VEGFR-2 may be impaired by a lack of VEGF. Other regulators of placental angiogenesis and placental vascular control have also been analysed in the samples collected at 55, 90 and 135 dpc. Both Ang 1 and Ang 2 mRNA concentration were greater (Pc0.05) in HT fetal cotyledons at 55 dpc, but not at other gestational ages, or in maternal caruncles (Figure 4). Interestingly, Tie2 mRNA concentration in fetal cotyledons was increased at 55 dpc, unaffected by treatment at 90 dpc, and reduced at 135 dpc. Additionally, there was a significant reduction (Pc0.05) in its mRNA

S126

Placenta (2002), Vol. 23, Supplement A, Trophoblast Research, Vol. 16

Control A

IUGR V

PO2 84 mmHg

PO2 52 mmHg

A

PO2 61 mmHg**

PO2 84 mmHg

Placenta

Placenta

PO2 19 mmHg**

PO2 29 mmHg a

V

g

a

g

Uterine blood flow to Umbilical blood flow ratio 2.19 + 0.84 4.38 + 0.48** Figure 5. Schematic representation of utero-placental PO2 in normal and IUGR ovine pregnancies at 135 dpc. IUGR was induced by HT treatment from 40 to 120 dpc, followed by TN conditions until 135 dpc when these measurements were taken. A=maternal femoral artery; V=uterine vein; a=fetal femoral artery; g=umbilical vein. **Pc0.01 between treatment comparisons.

concentration at 135 dpc in maternal caruncles. These results indicate that angiopoietin stimulated angiogenesis or vascular remodeling may be increased acutely in HT pregnancies, but reduced chronically. Immuno-blotting of maternal caruncle and fetal cotyledon tissue protein obtained at 55, 90 and 135 dpc revealed contrasting results. The concentrations of Ang1 and Tie 2 were not effected by treatment, but there was a significant increase in Ang2 at 55 dpc and a decrease at 135 dpc in HT fetal cotyledons. The contrasting results between the mRNA and protein concentration data are not easily explainable, but are similar in many ways with results obtained with human IUGR placenta (Dunk et al., 2000). In those studies, changes in Ang2 tissue concentration without concordant changes in Ang2 mRNA were observed. It is possible that post-transcriptional regulation of both Ang2 and Tie2, such as exists with VEGF (Shih and Claffey, 1999), could explain these discrepancies. Alternatively, disagreement between measures of tissue concentration of mRNA and protein for these growth factors and receptors could result from the fact that they exist in more than one cell type within the placenta (Dunk et al., 2000; Goldman-Wohl et al., 2000). Expression by one cell type may be increased while at the same time it is decreased in another cell type. Such a pattern of expression could easily impact the results of measuring whole tissue concentrations of mRNA or protein, making it difficult to fully ascertain the ‘functional’ amount of a given ligand or receptor. It must also be kept in mind that many things can impact tissue concentration of a given protein, including rate of synthesis, rate of secretion, interaction with extracellular matrix, amount bound by receptor and the turnover rate of bound receptor. Consequently, we believe that tissue concentration of mRNA may give a more accurate assessment of the activity of these systems within the placenta. Our results with VEGF, and the reported relationship between VEGF and endothelial nitric oxide synthase (eNOS) expression (Kroll and Waltenberger, 1998), prompted us to begin examining eNOS in our collected tissues. We have

measured eNOS protein concentration in cotyledons from TN and HT pregnancies at 90 dpc, and localized eNOS in cotyledon cross-sections by immunohistochemistry (Galan et al., 2001). The fetal placental cotyledons had greater (Pc0.01) concentrations of eNOS than did the maternal uterine caruncles, and cotyledon eNOS concentration was reduced by ]50 per cent in the HT pregnancies at 90 dpc (Pc0.05). We have yet to make these comparisons in our 55 and 135 dpc tissues to determine if eNOS expression is altered in the same manner as VEGF and Ang 2. Based on our results so far, we postulate that eNOS expression will be stimulated in the 55 dpc, and that the diminished expression of eNOS at 90 dpc is a function of the reduction in VEGFR-1 or VEGFR-2. Our immunohistochemistry data (Galan et al., 2001) localized eNOS to the vascular endothelium, but also to chorionic binucleate cells (BNCs). These cells are thought to be analogous to human syncytiotrophoblasts, but they are also migratory in nature (Wooding, 1992). It is possible that the BNC source of eNOS may provide a means for the fetal placenta to modify maternal uterine caruncle vascular tone, when the BNCs migrate and fuse with the syncytium thereby releasing their contents towards the maternal vasculature (Wooding, 1992). The reduction in eNOS that we observed in our HT pregnancies is in contrast to the increased eNOS and nitric oxide production by human placenta from IUGR and preeclamptic pregnancies (Lyall et al., 1996; Myatt et al., 1997). However, the human studies utilized late-gestation tissues, and it is possible that we will see similar results with our 135 dpc tissues. This would again highlight that measured alterations in placental development of IUGR pregnancies are gestational age dependent.

FUNCTIONAL SIGNIFICANCE The question remains as to what functional impact the altered expression of vascular growth factors and their receptors may

Regnault et al.: Placental Development in IUGR Pregnancies

have on delivery of O2 and nutrients to the rapidly growing fetus during later gestation. Earlier studies of human and HT-induced sheep IUGR pregnancies demonstrated that these fetuses are hypoxic and hypoglycemic (see discussion above; Figure 2). Yet, it has been suggested (Kingdom and Kaufmann, 1997; Kingdom et al., 2000) that early-onset IUGR may result in ‘placental hyperoxia’ during late gestation. Our observations of growth factor and receptor expression during late-gestation sheep IUGR pregnancies (Figure 4) agree with this concept, and are supported by recent data obtained with these pregnancies (Figure 5). At 135 dpc, our IUGR pregnancies exhibit increased (Pc0.01) uterine venous blood flow and P2. This coupled with reduced umbilical vein blood flow and P2, suggests that a barrier to placental O2 extraction exists during late-gestation in these pregnancies. The extent of this extraction barrier is borne out when one considers the doubling of the uterine vein : umbilical vein blood flow ratio (Figure 5). We believe that this barrier results from the changes exhibited in vascular growth factors and their receptors during the development of these IUGR pregnancies. Furthermore, the combination of our results support the concept of ‘placental hyperoxia’, at least at the maternal-fetal

S127

interface of the placenta, derived from examination of human IUGR placenta (Kingdom and Kaufmann, 1997; Kingdom et al., 2000). In this review, we have tried to highlight a number of the factors that may be involved in altered placental vascular development in both human and sheep IUGR pregnancies. While it is not easy to assign cause and effect from the data discussed, it is likely safe to assume that altered vascular development in the placenta may be a driving force behind many of the hallmarks of IUGR pregnancies. From the summarization of our results from sheep IUGR pregnancies, it appears that altered expression of vascular growth factors and their receptors occurs in a fashion such that acute changes may be quite different from changes observed in late-gestation. Therefore, a thorough understanding of when and why these changes take place may be paramount to our understanding of the development of IUGR-associated placenta. Such understanding will likely be obtained only from use of a consistent animal model, such as ours, in which the ex vivo examination of regulator factor expression can be compared to in vivo fluxes of O2 and nutrients across the placenta.

ACKNOWLEDGEMENTS The authors would like to thank Drs B. deVrijer, S. W. Limesand and R. B. Wilkening, as well as Ms M. Davidsen and A. S. Erickson-Hagen for their contributions to the experimental results and interpretation of those results discussed in this review. This work was supported in part by a grant from the National Institutes of Health (HD 20761).

REFERENCES Ahmed AS, Li XF, Dunk CE, Whittle MJ & Rollason T (1995) Colocalisation of vascular endothelial growth factor and its flt-1 receptor in human placenta. Growth Factors, 12, 235–243. Alexander G & Williams D (1971) Heat stress and development of the conceptus in domestic sheep. J Agric Sci, 76, 53–72. Anderson AH, Fennessey PV, Meschia G, Wilkening RB & Battaglia FC (1997) Placental transport of threonine and its utilization in the normal and growth-restricted fetus. Am J Physiol, 272, E892–E900. Barker DJP, Bull AR, Osmond C & Simmonds SJ (1990) Fetal and placental size and risk of hypertension in adult life. Brit Med J, 301, 259–262. Barker DJP, Gluckman PD, Godfrey KM, Harding JE, Owens JA & Robinson JS (1993a) Fetal nutrition and cardiovascular disease in adult life. Lancet, 341, 938–941. Barker DJP, Hales CN, Fall CHD, Osnond C, Phipps K & Clark PMS (1993b) Type 2 diabetes mellitus, hypertension and hyperlipidaemia: relation to reduced fetal growth. Diabetologia, 36, 62–67. Barker D, Osmond C, Golding J, Kuth D & Wadsworth M (1989) Growth in utero, blood pressure in childhood and adult life, and mortality from cardiovascular disease. Brit Med J, 298, 564–567. Bell AW, McBride BW, Slepetis R, Early RJ & Currie WB (1989) Chronic heat stress and perinatal development in sheep: I. Conceptus and maternal plasma hormones and metabolites. J Anim Sci, 67, 3289–3299. Bell AW, Wilkening RB & Meschia G (1987) Some aspects of placental function in chronically heat-stressed ewes. J Dev Physiol, 9, 17–29. Boshier DP (1969) A histological and histochemical examination of implantation and early placentome formation in sheep. J Reprod Fertil, 19, 51–61. Boyd JD & Hamilton WJ (1952) Cleavage, early development and implantation of the egg. In Marshall’s Physiology of Reproduction (Ed.) Parkes AS, pp. 1–110. London: Longman, Green. Brar HS & Rutherford SE (1988) Classification of intrauterine growth retardation. Sem Perinatol, 12, 2–10.

Brosens I, Robertson WB & Dixon HG (1967) The physiological response to the vessels of the placental bed to normal pregnancy. J Pathol Bacteriol, 93, 569–579. Caniggia I, Mostachfi H, Winter J, Gassmann M, Lye SJ, Kuliszewski & Post M (2000a) Hypoxia-inducible factor-1 mediates the biological effects of oxygen on human trophoblast differentiation through TGF3. J Clin Invest, 105, 577–587. Caniggia I, Winter J, Lye SJ & Post M (2000b) Oxygen and placental development during the first trimester: implications for the pathophysiology of pre-eclampsia. Placenta, 14, S25–30. Cao Y, Chen H, Zhou L, Chiang MK, Anand-Apte B, Weatherbee JA, Wang Y, Fang F, Flanagan JG & Tsang ML (1996) Heterodimers of placenta growth factor/vascular growth factor, endothelial activity, tumor cell expression, and high affinity binding to Flk-1/KDR. J Biol Chem, 271, 3154–3162. Charnock-Jones DS, Sharkey AM, Boocock CA, Ahmed A, Plevin R, Ferrara N & Smith SK (1994) Localization and activation of the receptor for vascular endothelial growth factor on human trophoblast and choriocarcinoma cells. Biol Reprod, 51, 524–530. Cooper JC, Sharkey AM, Charnock-Jones DS, Palmer CR & Smith SK (1996) VEGF mRNA levels in placentae from pregnancies complicated by pre-eclampsia. Br J Obstet Gynaeco, 103, 1191–1196. Crescimanno C, Marzioni D, Persico MG, Vuckovic M, Muhauser J & Castelucci M (1995) Expression of bFGF, PlGF and their receptors in the human placenta. Placenta, 16, A13. DiSalvo J, Bayne ML, Conn G, Kwok PW, Trivedi PG, Soderman DD, Palisi TM, Sullivan KA & Thomas KA (1995) Purification and characterization of a naturally occurring vascular endothelial growth factorplacenta growth factor heterodimer. J Biol Chem, 270, 7717–7723. Dunk C & Ahmed A (2001) Expression of VEGF-C and activation of its receptors VEGFR-2 and VEGFR-3 in trophoblast. Histo Histopathol, 16, 359–375. Dunk C, Shams M, Nijjar S, Rhaman M, Qiu Y, Bussolati B & Ahmed A (2000) Angiopoietin-1 and angiopoietin-2 activate trophoblast Tie-2 to

S128 promote growth and migration during placental development. Am J Pathol, 156, 2185–2199. Dvorak HF, Brown LF, Detmar M & Dvorak AM (1995) Vascular permeability factor/vascular endothelial growth factor, microvascular hyperpermeability and angiogenesis. Am J Physiol, 146, 1029–1039. Economides DL & Nicolaides KH (1989) Blood glucose and oxygen tension levels in small-for-gestational-age fetuses. Am J Obstet Gynecol, 160, 385–389. Fuh G, Garcia C & de Vos AM (2000) The interaction of neuropilin-1 with vascular endothelial growth factor and its receptor Flt-1. J Biol Chem, 275, 26690–26695. Galan HL, Hussey MJ, Barbera A, Ferrazzi E, Chung M, Hobbins JC & Battaglia FC (1999) Relationship of fetal growth to duration of heat stress in an ovine model of placental insufficiency. Am J Obstet Gynecol, 180, 1278–1282. Galan HG, Hussey MJ, Chung M, Chyu JK, Hobbins JC & Battaglia FC (1998) Doppler velocimetry of growth restricted fetuses in an ovine model of placental insufficiency. Am J Obstet Gynecol, 178, 451–456. Galan HL, Regnault TRH, Le Cras TD, Tyson RW, Anthony RV, Wilkening RB & Abman SH (2001) Cotyledon and binucleate cell nitric oxide synthase expression in an ovine model of fetal growth retardation. J App Physiol, 90, 2420–2426. Genbacev O, Krtolica A, Kaelin W & Fisher SJ (2001) Human cytotrophoblast expression of the von Hipple-Lindau protein is downregulated during uterine invasion in situ and upregulated by hypoxia in vitro. Dev Biol, 233, 526–536. Giles WB, Trudinger BJ & Baird P (1985) Fetal umbilical artery flow velocity wafeforms and placental resistance, pathological correlation. Br J Obstet Gynaecol, 92, 31–38. Glazier JD, Cetin I, Perugino G, Ronzoni S, Grey AM, Mahendran D, Marconi AM, Pardi G & Sibley CP (1997) Association between the activity of the system A amino acid transporter in the microvillous plasma membrane of the human placenta and severity of fetal compromise in intrauterine growth restriction. Ped Res, 42, 514–519. Gluzman-Poltorak Z, Cohen T, Herzog Y & Neufeld G (2000) Neuropilin-2 and neuropilin-1 are receptors for the 165-amino acid form of vascular endothelial growth factor (VEGF) and of placenta growth factor-2, but only neuropilin-2 functions as a receptor for the 145-amino acid form of VEGF. J Biol Chem, 275, 18040–18045. Goldman-Wohl DS, Ariel I, Greenfield C, Lavy Y & Yagel S (2000) Tie-2 and angiopoietin-2 expression at the fetal-maternal interface: a receptor ligand model for vascular remodeling. Mol Hum Reprod, 6, 81–87. Jansson T, Scholtbach V & Powell TL (1998) Placental transport of leucine and lysine is reduced in intrauterine growth restriction. Ped Res, 44, 532–537. Joukov V, Pajusola K, Kaipainen A, Chilov D, Lahtinen I, Kukk E, Saksela O, Kalkkinen N & Alitalo K (1996) A novel vascular endothelial growth factor, VEGF-C, is a ligand for the Flt4 (VEGFR-3) and KDR (VEGF-2) receptor tyrosine kinases. EMBO J, 15, 290–298. Kaufmann P (1982) Development and differentiation of the human villous tree. Biblio Anat, 22, 29–39. Kaufmann P, Sen DK & Schweikhart G (1979) Classification of human placental villi. I. Histology. Cell Tissue Res, 200, 409–423. Khaliq A, Dunk C, Jiang J, Shams M, Li XF, Acevedo C, Weich H, Whittle M & Ahmed A (1999) Hypoxia down-regulates placenta growth factor, whereas fetal growth restriction up-regulates placenta growth factor expression: molecular evidence for ‘placental hyperoxia’ in intrauterine growth restriction. Lab Invest, 79, 151–170. Kilby MD, Afford S, Li XF, Strain AJ, Ahmed A & Whittle MJ (1996) Localization of hepatocyte growth factor (HGF) and its receptor (c-met) protein and mRNA in human term placenta. Growth Factors, 13, 133–139. Kingdom J, Huppertz B, Seaward G & Kaufmann P (2000) Development of the placental villous tree and its consequences for fetal growth. Eur J Obstet Gynecol Reprod Biol, 92, 35–43. Kingdom JC & Kaufmann P (1997) Oxygen and placental villous development: origins of fetal hypoxia. Placenta, 18, 613–621. Kingdom JC & Kaufmann P (1999) Oxygen and placental vascular development. Adv Exp Med Biol, 474, 259–275. Koblizek TI, Weiss C, Yancopoulos GD, Deutsch U & Risau W (1998) Angiopoietin-1 induces sprouting angiogenesis in vitro. Curr Biol, 8, 529–532. Krebs C, Longo LD & Leiser R (1997) Term ovine placental vasculature: comparison of sea level and high altitude conditions by corrosion cast and histomorphometry. Placenta, 18, 43–51.

Placenta (2002), Vol. 23, Supplement A, Trophoblast Research, Vol. 16 Krebs C, Macara LM, Leiser R, Bowman AW, Greer IA & Kingdom JCP (1996) Intrauterine growth restriction with absent end-diastolic flow velocity in the umbilical artery is associated with maldevelopment of the placental terminal villous tree. Am J Obstet Gynecol, 175, 1834–1842. Kroll J & Waltenberger J (1998) VEGF-A induces expression of eNOS and iNOS in endothelial cells via VEGF receptor-2 (KDR). Biochem Biophys Res Commun, 252, 743–746. Lee MML & Yeh M (1986) Fetal microcirculation of abnormal human placenta. I. Scanning electron microscopy of placental vascular casts from small for gestational age fetus. Am J Obstet Gynecol, 154, 1133–1139. Leiser R, Krebs C, Ebert B & Dantzer V (1997) Placental corrosion cast studies: a comparison between ruminants and humans. Microsc Res Tech, 38, 76–87. Levy NS, Chung S, Furneaux H & Levy AP (1998) Hypoxic stabilization of vascular endothelial growth factor mRNA by the RNA-binding protein HuR. J Biol Chem, 273, 6417–6423. Lyall F, Greer IA, Young A & Myatt L (1996) Nitric oxide concentrations are increased in the feto-placental circulation in intrauterine growth restriction. Placenta, 17, 165–168. Lyall F, Young A, Boswell F, Kingdom JC & Greer IA (1997) Placental expression of vascular endothelial growth factor in placentae from pregnancies complicated by pre-eclampsia and intrauterine growth restriction does not support placental hypoxia at delivery. Placenta, 18, 269–276. Lyttle DJ, Fraser KM, Fleming SB, Mercer AA & Robinson AJ (1994) Homologs of a vascular endothelial growth factor are encoded by the poxvirus orf virus. J Virol, 68, 84–92. Macara L, Kingdom JC, Kaufmann P, Kohnen G, Hair J, More IA, Lyall F & Greer IA (1996) Structural analysis of placental terminal villi from growth-restricted pregnancies with abnormal umbilical Doppler waveforms. Placenta, 17, 37–48. Maisonpierre PC, Suri C, Jones PF, Bartunkova S, Wiegand SJ, Radziejewski C, Compton D, McClain J, Aldrich TH, Papadopoulos N, Daly TJ, Davis S, Sato TN & Yancopoulos GD (1997) Angiopoietin-2, a natural antagonist for Tie2 that disrupts in vivo angiogenesis. Science, 277, 55–60. Marconi AM, Paolini CL, Stramare L, Cetin I, Fennessey PV, Pardi G & Battaglia FC (1999) Steady state maternal-fetal leucine enrichments in normal and intrauterine growth-restricted pregnancies. Ped Res, 46, 114– 119. Maxwell PH, Wiesener MS, Chang G-W, Clifford SC, Vaux EC, Cockman ME, Wykoff CC, Pugh CW, Maher ER & Ratcliffe PJ (1999) The tumor suppressor protein VHL targets hypoxia-inducible factors for oxygen-dependent proteolysis. Nature, 399, 271–275. Meekins JW, Pijenborg R, Hanssens M, McFadyen IR & VanAsshe A (1994) A study of placental bed spiral arteries and trophoblast invasion in normal and severe pre-eclamptic pregnancies. Br J Obstet Gynaecol, 101, 669–674. Mellor DJ & Murray L (1981) Effects of placental weight and maternal nutrition on the growth rates of individual fetuses in single and twin bearing ewes during late pregnancy. Res Vet Sci, 30, 198–204. Mellor DJ & Murray L (1982) Effects on the rate of increase in fetal girth of refeeding ewes after short periods of sever undernutrition during late pregnancy. Res Vet Sci, 32, 377–382. Migdal M, Huppertz B, Tessler S, Comforti A, Shibuya M, Reich R, Baumann H & Neufeld G (1998) Neuropilin-1 is a placenta growth factor-2 receptor. J Biol Chem, 273, 22272–22278. Myatt L, Eis AL, Brockman DE, Greer IA & Lyall F (1997) Endothelial nitric oxide synthase in placental villous tissue from normal, pre-eclamptic and intrauterine growth restricted pregnancies. Hum Reprod, 12, 167–172. Nieto-Diaz A, Villar J, Matorras-Weinig R & Valenzuela-Ruiz P (1996) Intrauterine growth retardation at term: association between anthropometric and endocrine parameters. Acta Obstet Gynecol Scand, 75, 127–131. Norberg S, Powell TL & Jansson T (1998) Intrauterine growth restriction is associated with a reduced activity of placental taurine transporters. Ped Res, 44, 233–238. Oh H, Takagi H, Suzuma K, Otani A, Matsumura M & Honda Y (1999) Hypoxia and vascular endothelial growth factor selectively up-regulate angiopoietin-2 in bovine microvascular endothelial cells. J Biol Chem, 274, 15732–15739. Olofsson B, Pajusola K, Kaipainen A, von Euler G, Juokov V, Saksela O, Orpana A, Pettersson RF, Alitalo K & Eriksson U (1996) Vascular endothelial growth factor B, a novel growth factor for endothelial cells. Proc Natl Acad Sci, 93, 2576–2581.

Regnault et al.: Placental Development in IUGR Pregnancies Pardi G, Cetin I, Marconi AM, Bozzetti P, Buscaglia M, Makowski EL & Battaglia FC (1992) Venous drainage of the human uterus: respiratory gas studies in normal and fetal growth-retarded pregnancies. Am J Obstet Gynecol, 166, 699–706. Pardi, G, Cetin I, Marconi AM, Lanfranchi A, Ferrazzi E, Bozzetti P, Buscaglia M & Battaglia FC (1993) Diagnostic value of blood sampling in fetuses with growth retardation. N Engl J Med, 328, 692–696. Pollack RN & Divon MY (1992) Intrauterine growth retardation: Definition, classification and etiology. Clin Obstet Gynecol, 35, 99–107. Rajakumar A & Conrad KP (2000) Expression, ontogeny and regulation of hypoxia-inducible transcription factors in the human placenta. Biol Reprod, 63, 559–569. Rajakumar A, Whitelock KA, Weissfeld LA, Daftary AR, Markovic N & Conrad KP (2001) Selective overexpression of the hypoxia-inducible transcription factor, HIF-2a, in placentas from women with preeclampsia. Biol Reprod, 64, 499–506. Redline RW & Patterson P (1995) Pre-eclampsia is associated with an excess of proliferative immature intermediate trophoblast. Human Pathol, 26, 594–600. Regnault TRH, Orbus RJ, Battaglia FC, Wilkening RB & Anthony RV (1999) Altered arterial concentrations of placental hormones during maximal placental growth in a model of placental insufficiency. J Endocrinol, 162, 433–442. Roberson WB, Brosens I & Dixon HG (1975) Uteroplacenta vascular pathology. Eur J Obstet Gynecol Reprod Biol, 5, 47–65. Ross JC, Fennessey PV, Wilkening RB, Battaglia FC & Meschia G (1996) Placental transport and fetal utilization of leucine in a model of fetal growth retardation. Am J Physiol, 270, E491–E503. Salafia CM, Pezzullo JC, Minior VK & Divon MY (1997) Placental pathology of absent and reversed end-diastolic flow in growth-restricted fetuses. Obstet Gynecol, 90, 830–836. Schweiki D, Itin A, Neufeld G, Gitay-Goren H & Keshet E (1992) Vascular endothelial growth factor induced by hypoxia may mediate hypoxia initiated angiogenesis. Nature, 359, 843–845. Shams M & Ahmed A (1994) Localization of mRNA for basic fibroblast growth factor in human placenta. Growth Factors, 11, 105–111. Sharkey AM, Charnock-Jones DS, Boocock CA, Brown KD & Smith SK (1993) Expression of mRNA for vascular endothelial growth factor in human placenta. J Reprod Fertil, 99, 609–615. Shih SC & Claffey KP (1999) Regulation of human vascular endothelial growth factor mRNA stability in hypoxia by heterogenous nuclear ribonucleoprotein L. J Biol Chem, 274, 1359–1365. Shima DT, Deutsch U & D’Amore PA (1995) Hypoxic induction of vascular endothelial growth factor (VEGF) in human epithelial cells is mediated by increases in mRNA stability. FEBS Letters, 370, 203–208.

S129 Shore VH, Wang TH, Wang CL, Torry RJ, Caudle MR & Torry DS (1997) Vascular endothelial growth factor, placenta growth factor and their receptors in isolated human trophoblast. Placenta, 18, 657–665. Stavri G, Zachary I, Baskerville P, Martin J & Erusalimsky J (1995) Basic fibroblast growth factor upregulates the expression of vascular endothelial growth factor in vascular smooth muscle cells. Synergistic interaction with hypoxia. Circulation, 92, 5–8. Stegeman JHJ (1974) Placental development in the sheep and its relation to fetal development. A qualitative and quantitative anatomic and histologic study. Bijdragen Tot De Dierkunde, 44, 3–72. Steven D (1975) Anatomy of the placental barrier. In Comparative placentation. Essays in Structure and Function (Ed.) Steven DH, pp. 25–27. London: Academic Press. Thomas KA (1996) Vascular endothelial growth factor, a potent and selective angiogenic agent. J Biol Chem, 271, 603–606. Thureen PJ, Trembler KA, Meschia G, Makowski EL & Wilkening RB (1992) Placental glucose transport in heat-induced fetal growth retardation. Am J Physiol, 263, R578–R585. Tidwell SC, Ho HN, Chiu WH, Torry RJ & Torry DS (2001) Low maternal serum levels of placenta growth factor as an antecedent of clinical preeclampsia. Am J Obstet Gynecol, 184, 1267–1272. Torry DS, Wang HS, Wang TH, Caudle MR & Torry RJ (1998) Preeclampsia is associated with reduced serum levels of placenta growth factor. Am J Obstet Gynecol, 179, 1539–1544. Trudinger BJ, Cook Cm, Giles WB, Connelly A & Thompson RS (1987) Umbilical artery velocity waveforms in high-risk pregnancy. Randomized controlled trial. Lancet, 1, 188–190. Vuckovic M, Ponting J, Terman BI, Niketic V, Seif MW & Kumar S (1996) Expression of vascular endothelial growth factor receptor, KDR, in human placenta. J Anat, 188, 361–366. Vuorela P, Hatva E, Lymboussaki A, Kaipainen A, Joukov V, Persico MG, Alitalo K & Halmesmaki E (1997) Expression of vascular endothelial growth factor and placenta growth factor in human placenta. Biol Reprod, 56, 489–494. Wang GL, Jiang BH, Rueand EA & Semenza GL (1995) Hypoxia inducible factor 1 is a basic helix-loop-helix PAS heterodimer regulated by cellular O2 tension. Proc Natl Acad. Sci, 92, 5510–5514. Wheeler T, Elcock CL & Anthony FW (1995) Angiogenesis in the placental environment. Placenta, 16, 289–296. Wooding FB (1992) Current topics: the synepitheliochorial placenta of ruminants: binucleate cell fusions and hormone production. Placenta, 13, 101–113. Yamada Y, Nezu J, Shimane M & Hirata Y (1997) Molecular cloning of a novel vascular endothelial growth factor, VEGF-D. Genomics, 42, 483–488.

Lihat lebih banyak...

Comentarios

Copyright © 2017 DATOSPDF Inc.