9
REPRODUCTION © 2017 Society for Reproduction and Fertility DOI: 10.1530/REP-16-0647 ISSN 1470–1626 (paper) 1741–7899 (online) Online version via www.reproduction-online.org REVIEW Vitamin D deficiency and impaired placental function: potential regulation by glucocorticoids? Nathanael Yates 1 , Rachael C Crew 2 and Caitlin S Wyrwoll 2 1 School of Animal Biology, and 2 School of Human Sciences, The University of Western Australia, Perth, Australia Correspondence should be addressed to C S Wyrwoll; Email: [email protected] Abstract Maternal vitamin D deficiency has been implicated in a range of pregnancy complications including preeclampsia, preterm birth and intrauterine growth restriction. Some of these adverse outcomes arise from alterations in placental function. Indeed, vitamin D appears critical for implantation, inflammation, immune function and angiogenesis in the placenta. Despite these associations, absence of the placental vitamin D receptor in mice provokes little effect. Thus, interactions between maternal and fetal compartments are likely crucial for instigating adverse placental changes. Indeed, maternal vitamin D deficiency elicits changes in glucocorticoid-related parameters in pregnancy, which increase placental and fetal glucocorticoid exposure. As in utero glucocorticoid excess has a well-established role in eliciting placental dysfunction and fetal growth restriction, this review proposes that glucocorticoids are an important consideration when understanding the impact of vitamin D deficiency on placental function and fetal development. Reproduction (2017) 153 R163–R171 Introduction As the sole conduit between maternal and fetal systems, the placenta mediates fetal exposure to nutritional, hormonal and other physiological cues throughout gestation. In addition to facilitating the direct exchange of oxygen and nutrients between mother and fetus, the placenta functions in a specialized endocrine capacity to produce hormones essential for maternal physiological adaptations to pregnancy (Evain-Brion & Malassine 2003). As such, a healthy placenta is vital for normal fetal growth and development, whereas impaired placental function disrupts fetal growth outcomes and may also result in long-term health complications in offspring via developmental programming mechanisms (Burton et al. 2016, Sferruzzi-Perri & Camm 2016). Maternal vitamin D deficiency has become a significant problem in modern day obstetrics; the rates of vitamin D deficiency have increased in recent decades (Looker et al. 2008), and it is estimated that between 18% and 84% of pregnant women worldwide are vitamin D deficient (Dawodu & Wagner 2007). Although vitamin D has traditionally been associated with bone health through its regulation of calcium and phosphate absorption, it also plays central roles in cellular proliferation and differentiation (Samuel & Sitrin 2008), vascular function (Tare et al. 2011, Ni et al. 2014) and immune regulation (Prietl et al. 2013). As a consequence of these pleiotropic functions, vitamin D metabolism is important for a range of key developmental events, including decidualization (Halhali et al. 1991), modulation of maternal immune function (Tamblyn et al. 2015) and fetal bone formation (Young et al. 2012). Accordingly, maternal vitamin D deficiency has been linked to pregnancy complications such as preeclampsia (Bodnar et al. 2007, Achkar et al. 2015), bacterial vaginosis (Bodnar et al. 2009), preterm birth (Qin et al. 2016) and gestational diabetes mellitus (Burris & Camargo 2014). Moreover, gestational vitamin D deficiency is associated with intrauterine growth restriction (IUGR) in infants (Gernand et al. 2014, Chen et al. 2015a) and adverse postnatal health outcomes in offspring, including increased rates of asthma (Zosky et al. 2014), hypertension (Tare et al. 2011) and impaired neurodevelopment (Whitehouse et al. 2012, Eyles et al. 2013, Hawes et al. 2015, Pet & Brouwer- Brolsma 2016). There are several forms of vitamin D, but vitamin D 3 is the naturally occurring form in animals. To elicit biological effects, vitamin D, which is obtained through the diet or synthesized in the skin via UVB exposure must be converted to its active form, 1α 25-dihydroxyvitamin D (1,25(OH) 2 D). To achieve this, circulating vitamin D is first converted to 25-hydroxyvitamin D (25(OH)D) by hepatic 25-hydroxylase, which is then hydroxylated to 1,25(OH) 2 D via the enzymatic activity of 1α-hydroxylase, encoded by the CYP27B1 Downloaded from Bioscientifica.com at 04/28/2022 11:03:52PM via free access

Vitamin D deficiency and impaired placental function

  • Upload
    others

  • View
    1

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Vitamin D deficiency and impaired placental function

PROOF ONLYREPRODUCTION

© 2017 Society for Reproduction and Fertility DOI: 10.1530/REP-16-0647ISSN 1470–1626 (paper) 1741–7899 (online) Online version via www.reproduction-online.org

REVIEW

Vitamin D deficiency and impaired placental function: potential regulation by glucocorticoids?

Nathanael Yates1, Rachael C Crew2 and Caitlin S Wyrwoll2

1School of Animal Biology, and 2School of Human Sciences, The University of Western Australia, Perth, Australia

Correspondence should be addressed to C S Wyrwoll; Email: [email protected]

Abstract

Maternal vitamin D deficiency has been implicated in a range of pregnancy complications including preeclampsia, preterm birth and intrauterine growth restriction. Some of these adverse outcomes arise from alterations in placental function. Indeed, vitamin D appears critical for implantation, inflammation, immune function and angiogenesis in the placenta. Despite these associations, absence of the placental vitamin D receptor in mice provokes little effect. Thus, interactions between maternal and fetal compartments are likely crucial for instigating adverse placental changes. Indeed, maternal vitamin D deficiency elicits changes in glucocorticoid-related parameters in pregnancy, which increase placental and fetal glucocorticoid exposure. As in utero glucocorticoid excess has a well-established role in eliciting placental dysfunction and fetal growth restriction, this review proposes that glucocorticoids are an important consideration when understanding the impact of vitamin D deficiency on placental function and fetal development.Reproduction (2017) 153 R163–R171

Introduction

As the sole conduit between maternal and fetal systems, the placenta mediates fetal exposure to nutritional, hormonal and other physiological cues throughout gestation. In addition to facilitating the direct exchange of oxygen and nutrients between mother and fetus, the placenta functions in a specialized endocrine capacity to produce hormones essential for maternal physiological adaptations to pregnancy (Evain-Brion & Malassine 2003). As such, a healthy placenta is vital for normal fetal growth and development, whereas impaired placental function disrupts fetal growth outcomes and may also result in long-term health complications in offspring via developmental programming mechanisms (Burton et al. 2016, Sferruzzi-Perri & Camm 2016).

Maternal vitamin D deficiency has become a significant problem in modern day obstetrics; the rates of vitamin D deficiency have increased in recent decades (Looker  et  al. 2008), and it is estimated that between 18% and 84% of pregnant women worldwide are vitamin D deficient (Dawodu & Wagner 2007). Although vitamin D has traditionally been associated with bone health through its regulation of calcium and phosphate absorption, it also plays central roles in cellular proliferation and differentiation (Samuel & Sitrin 2008), vascular function (Tare  et  al. 2011, Ni  et  al. 2014) and immune regulation (Prietl  et  al. 2013). As a consequence of these pleiotropic functions,

vitamin D metabolism is important for a range of key developmental events, including decidualization (Halhali et al. 1991), modulation of maternal immune function (Tamblyn et al. 2015) and fetal bone formation (Young  et  al. 2012). Accordingly, maternal vitamin D deficiency has been linked to pregnancy complications such as preeclampsia (Bodnar et al. 2007, Achkar et al. 2015), bacterial vaginosis (Bodnar et al. 2009), preterm birth (Qin et al. 2016) and gestational diabetes mellitus (Burris & Camargo 2014). Moreover, gestational vitamin D deficiency is associated with intrauterine growth restriction (IUGR) in infants (Gernand  et  al. 2014, Chen et al. 2015a) and adverse postnatal health outcomes in offspring, including increased rates of asthma (Zosky et al. 2014), hypertension (Tare et al. 2011) and impaired neurodevelopment (Whitehouse  et  al. 2012, Eyles  et  al. 2013, Hawes  et  al. 2015, Pet & Brouwer-Brolsma 2016).

There are several forms of vitamin D, but vitamin D3 is the naturally occurring form in animals. To elicit biological effects, vitamin D, which is obtained through the diet or synthesized in the skin via UVB exposure must be converted to its active form, 1α 25-dihydroxyvitamin D (1,25(OH)2D). To achieve this, circulating vitamin D is first converted to 25-hydroxyvitamin D (25(OH)D) by hepatic 25-hydroxylase, which is then hydroxylated to 1,25(OH)2D via the enzymatic activity of 1α-hydroxylase, encoded by the CYP27B1

10.1530/REP-16-0647

Downloaded from Bioscientifica.com at 04/28/2022 11:03:52PMvia free access

Page 2: Vitamin D deficiency and impaired placental function

PROOF ONLYN Yates and othersR164

Reproduction (2017) 153 R163–R171 www.reproduction-online.org

gene (Norman 2008). Although the latter conversion typically occurs in the kidney, the placenta also exhibits abundant expression and activity of 1α-hydroxylase (Zehnder et al. 2002), which likely indicates a functional importance for local placental vitamin D metabolism during gestation (Fig. 1). Circulating 1,25(OH)2D elicits genomic effects by binding to the vitamin D receptor (VDR), which dimerizes with the retinoid X receptor (RXR) and induces subsequent gene transcription through vitamin D response element (VDRE) binding to target gene promoter regions (Haussler et al. 2013). However, as with many steroid hormones, vitamin D can also exert rapid non-genomic effects, likely via VDR located within the plasma membrane (Norman  et  al. 2002, Menegaz  et  al. 2011). VDR is also abundantly expressed in placental tissue, thus allowing for localized placental 1,25(OH)2D binding and subsequent gene transcription (Shahbazi  et  al. 2011). Interestingly, the precursor 25-(OH)D readily crosses the placenta; however, 1,25(OH)2D is unable to be transported across the placenta, and thus is found in relatively low levels in fetal circulation (Kovacs 2008).

A recent study shows that placental VDR expression is downregulated in pregnancies complicated by

fetal growth restriction (Nguyen  et  al. 2015b) and is associated with abnormal trophoblast expression of cell-cycle regulatory genes in vitro (Nguyen  et  al. 2015a). This indicates that placental vitamin D metabolism regulates fetal growth and development; however, relatively little is known of the mechanisms behind such regulatory processes, and whether they may be compromised during vitamin D-deficient pregnancies. Interestingly, recent evidence shows that placental-specific ablation of Vdr expression does not have overt effects on placental phenotype or fetal growth outcomes (Wilson  et  al. 2015). This suggests that vitamin D homeostasis during pregnancy likely involves communication between maternal, placental and fetal compartments. Indeed, recent research has shown an interaction between vitamin D pathways and glucocorticoids (‘stress’ hormones) in the context of the placenta, brain, lung and other tissues (Obradovic et al. 2006, Hidalgo  et  al. 2011, Chambers  et  al. 2015, Tesic  et  al. 2015, Kassi  et  al. 2016). The current review aims to discuss the potential mechanisms by which placental function may be implicated in the adverse outcomes associated with vitamin D-deficient pregnancies. The key focus of this review is to propose

Figure 1 Vitamin D metabolic pathways during pregnancy across maternal, fetal and placental compartments. Vitamin D is absorbed into maternal circulation via UVB-catalyzed synthesis in the skin or dietary intake (1) and is then converted to 25(OH)D via hepatic 25-hydroxylase (2). This is subsequently hydroxylated to the active form of the metabolite in various tissues, including the kidney and placenta (3). The precursor 25(OH)D can also directly cross the placenta (6), where it is hydroxylated to the active metabolite in the fetal kidney (4). The active 1,25(OH)D then elicits genomic effects through binding to the vitamin D receptor (VDR) in target tissues, including the placenta (5). This induces the transcription of target genes via vitamin D response element (VDRE), thus enabling the regulation of a range of important processes during gestation (6).

Downloaded from Bioscientifica.com at 04/28/2022 11:03:52PMvia free access

Page 3: Vitamin D deficiency and impaired placental function

PROOF ONLYVitamin D, placenta and glucocorticoids R165

www.reproduction-online.org Reproduction (2017) 153 R163–R171

that the involvement of factors extrinsic to vitamin D pathways, particularly the action of glucocorticoids, is crucial for eliciting placental effects.

Vitamin D deficiency and placental development and function

Vitamin D metabolic pathways in the placenta may facilitate the transport of calcium and phosphate across the placenta, enabling fetal skeletal development. Changes in fetal femoral development with maternal vitamin D insufficiency can be observed as early as 19-week gestation using 3D ultrasound (Mahon  et  al. 2010) and such changes in bone structure may occur well into early childhood (Javaid et al. 2006). Although vitamin D may have a role in fetal skeletal development, other dominant pregnancy hormones such as prolactin, estradiol and lactogen are also key to this process (Kirby et al. 2013). There is evidence that the increased maternal intestinal calcium absorption that occurs in pregnancy and calcium supply to the fetus is regulated by mechanisms independent of the typical vitamin D pathway. Thus, maternal vitamin D deficiency is associated with reduced fetal cord blood calcium and bone mineralization in some (Cockburn  et  al. 1980, Maghbooli et al. 2007), but not all (Javaid et al. 2006) studies. Indeed, a systematic review of vitamin D supplementation in pregnancy found limited evidence for an association between maternal vitamin D status and offspring birthweight, bone mass or serum calcium concentrations (Harvey et al. 2014). Furthermore, mice with genetic removal of Vdr (which therefore eliminates 1,25(OH)2D3 signaling) do not exhibit differences in placental calcium transfer or fetal skeletal mineralization (Kovacs et al. 2005). This raises the possibility that the key function of vitamin D in pregnancy is beyond calcium/bone effects. Indeed, maternal vitamin D deficiency is implicated in pregnancy complications associated with placental dysfunction such as preeclampsia, preterm birth and IUGR. This, in conjunction with the presence of a local vitamin D metabolic pathway, implies that vitamin D has a critical role in placental development and function. So what are the underlying biological mechanisms that contribute to the effects of vitamin D on placental development and function? The key mechanisms include effects on implantation, inflammation, immune function and angiogenesis.

Implantation, inflammation and immune function

Vitamin D has potent effects on immune responses (Liu  et  al. 2006) and influences both the innate and adaptive arms of the immune system (Hewison 2011). Immune adaptations are vital for successful pregnancy outcome and vitamin D likely acts to promote implantation due to its role in inflammatory

pathways and immune function. Thus, administration of 1,25(OH)2D3 in rats promotes decidualization (Halhali  et  al. 1991), and vitamin D likely has a key role to play in the immune function of the decidua (for an extensive review see Tamblyn et al. 2015), given its important immunomodulatory effects on trophoblasts. Indeed, vitamin D metabolites enhance extravillous trophoblast invasion in vitro (Chan et al. 2015) and act in a paracrine manner to suppress cytokine secretion in uterine natural killer cells (Evans  et  al. 2006) and villous trophoblast (Díaz  et  al. 2009, Noyola-Martínez et al. 2013).

In light of this, it seems that the anti-inflammatory actions of vitamin D likely play a critical role in the prevention of placental pathologies. Primary human trophoblast cell cultures treated with antiphospholipid antibodies (as a model of antiphospholipid syndrome, an autoimmune disorder associated with pregnancy loss and preeclampsia) demonstrate a dampened inflammation response in the presence of vitamin D (Gysler et al. 2015), whereas vitamin D treatment inhibits trophoblast expression and secretion of IL-10 in placental tissue obtained from both healthy and pre-eclamptic pregnancies (Barrera et al. 2012). The actions of vitamin D extend into enhancing antibacterial responses to Escherichia coli in trophoblast culture (Liu et al. 2009). Genetic ablation of Cyp27b1 stimulates placental inflammation when challenged with LPS; yet, in wild-type placentas, the presence of vitamin D metabolites suppresses the inflammatory response to LPS (Liu et al. 2011). Placental LPS-induced inflammation can be inhibited by pre-administration of vitamin D, preventing IUGR, which appears at least partly due to increasing interactions of the VDR and the nuclear factor kappa B p65 subunit (Chen  et al. 2015b). Therefore, it appears that vitamin D has generally positive effects in regulating placental immune function and providing protective effects against inflammation and bacterial infection.

Placental angiogenesis and nutrient transport

There is some evidence for maternal vitamin D deficiency altering placental vascular development. Thus, dietary vitamin D-restricted mice exhibited decreased vascular diameter (Liu  et  al. 2013) and decreased fetal capillary length and volume (Tesic et al. 2015) in the labyrinth zone, which is the highly vascular zone of the rodent placenta responsible for nutrient and waste exchange. Mechanistic evidence for vitamin D effects on angiogenesis have been provided by studies in endothelial colony-forming cells. Thus, vitamin D promotes the formation of capillary-like structures in conjunction with an upregulation of angiogenic factor vascular endothelial growth factor (Vegf) (Grundmann et al. 2012). These observed changes in Vegf are supported by the observation that maternal

Downloaded from Bioscientifica.com at 04/28/2022 11:03:52PMvia free access

Page 4: Vitamin D deficiency and impaired placental function

PROOF ONLYN Yates and othersR166

Reproduction (2017) 153 R163–R171 www.reproduction-online.org

vitamin D deficiency is associated with a reduction of Vegf in the highly vascular labyrinth zone of mouse placenta (Tesic et al. 2015).

In addition to altered angiogenic pathways, there is also an association between vitamin D status and placental nutrient transport. Indeed, Cleal and coworkers (Cleal  et  al. 2015) found that human placental gene expression of amino acid transporters correlated with maternal vitamin status as well as vitamin D-binding protein. This implies that while placental vitamin D is important for supplying amino acids to the fetus, delivery of vitamin D to the placenta is also crucial for this process (Cleal et al. 2015). Supporting associations have also been found in mechanistic studies using primary human trophoblast cell culture, whereby 1,25-dihydroxy vitamin D3 was shown to regulate system A amino acid transporter activity via increased expression of the SNAT2 amino acid transporter (Chen et al. 2017).

The specific role of placental vitamin D receptor

So how many of these observed effects of vitamin D on placental development and function are mediated via binding of the vitamin D receptor in the placenta? Initial studies in Vdr-knockout (Vdr−/−) dams demonstrated decreased fertility and a reduction in fetal weight in comparison to heterozygous (Vdr−/−) dams (Kovacs et al. 2005); however, the role of the placenta in this study was not assessed. Elegant investigations of placentas from Vdr+/− matings have proven to be more definitive in aiding our understanding of the significance of placental Vdr, as this eliminates maternal pathophysiology and generates each of the three possible placental–fetal genotypes within the one pregnancy (Vdr+/+, Vdr+/− and Vdr−/−). Thus, in response to an LPS challenge to simulate maternal infection, inflammatory markers were upregulated in the Vdr−/− placentas in comparison to wild type (Vdr+/+) (Liu et al. 2011), further implicating a role for vitamin D as a regulator of placental inflammation. Microarray and morphology comparisons of heterozygous-bred Vdr+/+ and Vdr−/− placentas by Wilson and coworkers (Wilson et al. 2015) have revealed that although there were some differences in gene expression, this did not manifest in altered placental morphology or placental function, as fetal and placental weights were unaltered. Of those genes that did change, Cyp24a1, which encodes the enzyme responsible for the catabolism of 1,25(OH)2D3, was robustly downregulated in Vdr−/− placentas, whereas other altered genes are implicated in pathways such as autophagy, cell signaling and cytoskeletal modifications and mammalian target of rapamycin (mTOR) signaling. It should be noted that, in the studies mentioned (Liu et al. 2011, Wilson et al. 2015), whole placental gene expression was assessed

and therefore, whether there are differential gene expression responses between the junctional and labyrinth zone of Vdr−/− placentas is yet to be established. On the whole, however, the lack of a robust placental phenotype in heterozygous-bred Vdr−/− placentae highlights that maternal vitamin D status and the associated physiological response is critical for causing the placental phenotype, as opposed to direct actions of vitamin D on the placental vitamin D receptor.

The potential physiological responses to maternal vitamin D deficiency are myriad. Our group has recently begun to focus on the potential interactions between vitamin D and glucocorticoids during pregnancy; however, there are many other key candidates of interest. Given the multifaceted role of vitamin D in immune pathways, there remains much work to be done elucidating how maternal vitamin D status influences appropriate immune and inflammation response in pregnancy. Furthermore, the observation that mTOR expression is altered in Vdr−/− placentas is of particular interest as placental mTOR signaling plays a pivotal role as a maternal nutrient sensor and thus is critically implicated in fetal growth and development (for a review see Dimasuay  et  al. 2016). Herein, we discuss how vitamin D deficiency alters glucocorticoid-related pathways in the maternal, fetal and placental compartments and how this may elicit the observed changes in placental development and function.

Glucocorticoids and pregnancy

Glucocorticoids regulate many physiological pathways including cellular differentiation, immunoregulation, inflammation and metabolism. During pregnancy, activity of the hypothalamic–pituitary–adrenal (HPA) axis markedly increases and accordingly, free glucocorticoids (cortisol and corticosterone in humans and rodents respectively) rise. This upregulation of glucocorticoids enables the mother to meet the metabolic demands of pregnancy (Seckl & Holmes 2007, Wharfe  et al. 2016) and plays an essential role in the maturation of fetal tissues in preparation for birth (Cole et al. 1995a,b, Diaz et al. 1998, Surbek et al. 2012). The issue arises, however, when fetal glucocorticoid exposure is excessive as this results in placental dysfunction, fetal growth restriction and altered organ development (Benediktsson  et  al. 1993, Lindsay  et  al. 1996a, Nyirenda et al. 1998, Ain et al. 2005, Hewitt et al. 2006, Holmes  et  al. 2006, Wyrwoll  et  al. 2009, Cuffe  et  al. 2011, Vaughan  et  al. 2012). Furthermore, the effects of early-life glucocorticoid exposure on development resonates throughout the lifespan with adverse effects on adult health outcomes including cardiometabolic and neuropsychiatric disorders (Benediktsson  et  al. 1993, Lindsay  et  al. 1996a,b,

Downloaded from Bioscientifica.com at 04/28/2022 11:03:52PMvia free access

Page 5: Vitamin D deficiency and impaired placental function

PROOF ONLYVitamin D, placenta and glucocorticoids R167

www.reproduction-online.org Reproduction (2017) 153 R163–R171

Nyirenda et al. 1998, Sugden et al. 2001, Holmes et al. 2006, Wyrwoll et al. 2006).

Given the potentially detrimental effects of glucocorticoid excess, it is critical that fetal exposure to glucocorticoids is tightly regulated. Indeed, fetal glucocorticoid levels remain up to 10-fold lower than those of maternal levels during the majority of gestation (Beitins et al. 1973, Edwards et al. 1993). This marked differential arises largely due to the presence of the enzyme 11β-hydroxysteroid dehydrogenase type 2 (Hsd11b2) in placental and fetal tissues. Hsd11b2 catalyzes the conversion of cortisol and corticosterone into their inactive forms, cortisone and 11-dehydrocorticosterone respectively. In humans, Hsd11b2 is highly expressed in the syncytiotrophoblast (Stewart et al. 1995) at the interface between maternal and fetal circulations. Similar patterns of Hsd11b2 expression are seen within the labyrinth zone of the rodent placenta (Brown et al. 1996, Burton et al. 1996). Expression of placental Hsd11b2 decreases at the end of gestation, presumably to facilitate fetal organ maturation (Burton et al. 1996).

Environmental perturbation in pregnancy can increase fetal glucocorticoid exposure by altering placental transfer of glucocorticoids. 10–20% of maternal glucocorticoids reach the fetus unaltered (Benediktsson  et  al. 1997), and therefore, stressors (ie. psychological stress, dietary insults, inflammation and hypoxia) that increase maternal glucocorticoid levels can increase fetal glucocorticoid exposure. Furthermore, these stressors generally reduce placental Hsd11b2 expression and/or activity and thus increase fetal glucocorticoid exposure (Langley-Evans et al. 1996, Mairesse et al. 2007, Seckl & Holmes 2007, Vieau et al. 2007). Therefore, given the high incidence of maternal vitamin D deficiency in pregnancy, could this act as a physiological stressor and exert effects on pathways involved in fetal glucocorticoid exposure?

Vitamin D and glucocorticoid interactions

The possibility of vitamin D and glucocorticoid pathway interactions are a relatively unexplored consideration; however, it seems likely from other studies that there is scope for substantial cross-talk between these two hormones. Indeed, vitamin D exerts antagonistic effects to the exposure of the synthetic glucocorticoid dexamethasone in the hippocampus. Thus, dexamethasone increases viable cells, decreases apoptotic cells and reduces neurite outgrowth in hippocampal cell culture with each change mitigated by concomitant administration of vitamin D (Obradovic et al. 2006). In addition, vitamin D appears to downregulate glucocorticoid receptor levels (Obradovic et al. 2006). Similar observations have been made in a squamous cell carcinoma model whereby dexamethasone

induces de novo transcription of the vitamin D receptor in a glucocorticoid receptor-dependent manner (Hidalgo  et  al. 2011). Furthermore, vitamin D also appears to have direct effects on the glucocorticoid sensitivity of peripheral blood mononuclear cells, with evidence of direct downregulation of glucocorticoid receptor expression and inhibition of GR translocation to the nucleus (Kassi et al. 2016). Moreover, in patients with steroid-resistant asthma, vitamin D administration beneficially alters the clinical response to glucocorticoids (Chambers et al. 2015).

So what occurs to glucocorticoid-related parameters in models of gestational vitamin D deficiency? There are multiple pathways that influence fetal exposure to glucocorticoids with vitamin D deficiency (Fig.  2). Maternal vitamin D deficiency in a mouse model of pregnancy elevates circulating maternal glucocorticoids as well as adrenal weight, indicating possible chronic

Figure 2 Proposed effects of developmental vitamin D deficiency on glucocorticoid metabolism and levels in the mother and fetus. (A) In gestational vitamin D deficiency, maternal circulating active glucocorticoids (GCs) and GC release in response to stress is elevated, likely increasing placental GC exposure and transport. (B) In the placenta, 11β-Hsd2 (a key enzyme that inactivates GCs) is decreased by vitamin D deficiency, which decreases the conversion of active GCs to inactive forms. (C) In the fetus, the combination of increased maternal GC levels and decreased GC inactivation due to vitamin D deficiency leads to increased fetal GC exposure. (D) Ultimately, vitamin D-deficient fetuses exhibit a likely increase in GC exposure in the brain, as indicated by increases in the GC-responsive gene Tsc22d3.

Downloaded from Bioscientifica.com at 04/28/2022 11:03:52PMvia free access

Page 6: Vitamin D deficiency and impaired placental function

PROOF ONLYN Yates and othersR168

Reproduction (2017) 153 R163–R171 www.reproduction-online.org

HPA activation (Fig.  2A, Tesic  et  al. 2015). HPA axis hyperactivation and altered feedback is supported by studies in rats showing that vitamin D-deficient pregnant rats have exaggerated serum corticosterone levels in response to restraint stress (Eyles et al. 2006). In addition to elevated maternal glucocorticoids, vitamin D deficiency also modifies placental Hsd11b2 expression. Thus, in mouse pregnancy at E14.5 (approximately 2/3 of mouse gestation), maternal vitamin D deficiency results in a 32% reduction in Hsd11b2 expression in placentas of male, but not female fetuses (Tesic  et  al. 2015). This reduction in Hsd11b2 presumably increases the placental transfer of glucocorticoids across to the fetus. Although this has not been directly measured, this notion is supported by the observation that the glucocorticoid-sensitive gene Tsc22d3 is increased in fetal brains at E14.5 and E17.5 from vitamin D-deficient dams (Tesic  et  al. 2015). This implies that some of the well-documented effects of early-life vitamin D deficiency on adverse adult health outcomes may in part, be attributed to fetal glucocorticoid exposure. Indeed by E14.5, glucocorticoid receptors are expressed in many organ systems including the brain, heart, lung, kidney and adrenal glands (Thompson et al. 2004, Rog-Zielinska  et  al. 2013), and therefore, many aspects of fetal development can be disturbed by excessive glucocorticoid exposure.

Although the direct level of fetal exposure to glucocorticoids remains unclear, the reduced placental Hsd11b2 (Tesic  et  al. 2015), elevated maternal circulating glucocorticoids (Tesic et al. 2015), increased maternal HPA axis response to stress (Eyles et al. 2006) and upregulated Tsc22d3 in the brains of deficient offspring (Tesic et al. 2015) all point to increased fetal exposure to glucocorticoids in vitamin D-deficient mothers. Reinforcing this notion, are the observations that vitamin D deficiency in mouse pregnancy perturbs placental vascular development (Liu  et  al. 2013, Tesic et al. 2015) recapitulating (albeit to a lesser extent) that seen in rat and mouse models of glucocorticoid excess (Hewitt  et  al. 2006, Wyrwoll  et  al. 2009, Vaughan  et  al. 2013). Although rodent offspring from mothers vitamin D deficient until parturition show normal glucocorticoid restraint response (Eyles  et  al. 2006), we have recent evidence to suggest this may not be the case when mothers and offspring are deficient until weaning (unpublished data). This supports a growing body of evidence suggesting that the timing and duration of vitamin D deficiency are important in determining effects on offspring (Burne et al. 2004a,b). Extensive work is required, however, to tease apart the interactions between vitamin D and glucocorticoids in the context of pregnancy. Indeed, it will be of great interest to see whether similar concomitant effects of vitamin D, glucocorticoids and their respective receptors occur in placental cell lines as they do in other cell-based studies.

Conclusions

Although the diverse effects of vitamin D on placental function may explain many complications associated with vitamin D deficiency in pregnancy, these effects are likely reliant on factors beyond immediate vitamin D pathways. There remains considerable work to be done in teasing apart the exact mechanisms, but the effects of vitamin D deficiency on glucocorticoid metabolism may be an important component for eliciting adverse placental effects. In the wider context, it is tempting to propose that vitamin D supplementation may improve multiple aspects of placental function and maternal physiology. However, there are considerable challenges around appropriate human levels of vitamin D (Lucas & Neale 2014), and thus, this remains an important ongoing area of research.

Declaration of interest

The authors declare that there is no conflict of interest that could be perceived as prejudicing the impartiality of this review.

Funding

This research did not receive any specific grant from any funding agency in the public, commercial or not-for-profit sector.

ReferencesAchkar  M, Dodds  L, Giguère  Y, Forest  J-C, Armson  BA, Woolcott  C,

Agellon  S, Spencer  A & Weiler  HA 2015 Vitamin D status in early pregnancy and risk of preeclampsia. American Journal of Obstetrics and Gynecology 212 511.e511–511.e517. (doi:10.1016/j.ajog.2014.11.009)

Ain R, Canham LN & Soares MJ 2005 Dexamethasone-induced intrauterine growth restriction impacts the placental prolactin family, insulin-like growth factor-II and the Akt signaling pathway. Journal of Endocrinology 185 253–263. (doi:10.1677/joe.1.06039)

Barrera  D, Noyola-Martínez  N, Avila  E, Halhali  A, Larrea  F & Díaz  L 2012 Calcitriol inhibits interleukin-10 expression in cultured human trophoblasts under normal and inflammatory conditions. Cytokine 57 316–321. (doi:10.1016/j.cyto.2011.11.020)

Beitins IZ, Bayard F, Ances IG, Kowarski A & Migeon CJ 1973 The metabolic clearance rate, blood production, interconversion and transplacental passage of cortisol and cortisone in pregnancy near term. Pediatric Research 7 509–519. (doi:10.1203/00006450-197305000-00004)

Benediktsson  R, Lindsay  R, Noble  J, Seckl  J & Edwards  C 1993 Glucocorticoid exposure in utero: new model for adult hypertension. Lancet 341 339–341. (doi:10.1016/0140-6736(93)90138-7)

Benediktsson R, Calder AA, Edwards CR & Seckl JR 1997 Placental 11β‐hydroxysteroid dehydrogenase: a key regulator of fetal glucocorticoid exposure. Clinical Endocrinology 46 161–166. (doi:10.1046/j.1365-2265.1997.1230939.x)

Bodnar LM, Catov JM, Simhan HN, Holick MF, Powers RW & Roberts JM 2007 Maternal vitamin D deficiency increases the risk of preeclampsia. Journal of Clinical Endocrinology and Metabolism 92 3517–3522. (doi:10.1210/jc.2007-0718)

Bodnar LM, Krohn MA & Simhan HN 2009 Maternal vitamin D deficiency is associated with bacterial vaginosis in the first trimester of pregnancy. Journal of Nutrition 139 1157–1161. (doi:10.3945/jn.108.103168)

Downloaded from Bioscientifica.com at 04/28/2022 11:03:52PMvia free access

Page 7: Vitamin D deficiency and impaired placental function

PROOF ONLYVitamin D, placenta and glucocorticoids R169

www.reproduction-online.org Reproduction (2017) 153 R163–R171

Brown R, Diaz R, Robson A, Kotelevtsev Y, Mullins J, Kaufman M & Seckl J 1996 The ontogeny of 11 beta-hydroxysteroid dehydrogenase type 2 and mineralocorticoid receptor gene expression reveal intricate control of glucocorticoid action in development. Endocrinology 137 794–797. (doi:10.1210/en.137.2.794)

Burne THJ, Becker A, Brown J, Eyles DW, Mackay-Sim A & McGrath JJ 2004a Transient prenatal vitamin D deficiency is associated with hyperlocomotion in adult rats. Behavioural Brain Research 154 549–555. (doi:10.1016/j.bbr.2004.03.023)

Burne  THJ, Féron  F, Brown  J, Eyles  DW, McGrath  JJ & Mackay-Sim  A 2004b Combined prenatal and chronic postnatal vitamin D deficiency in rats impairs prepulse inhibition of acoustic startle. Physiology and Behavior 81 651–655. (doi:10.1016/j.physbeh.2004.03.004)

Burris  HH & Camargo  CA 2014 Vitamin D and gestational diabetes mellitus. Current Diabetes Reports 14 451–451. (doi:10.1007/s11892-013-0451-3)

Burton PJ, Smith RE, Krozowski ZS & Waddell B 1996 Zonal distribution of 11 beta-hydroxysteroid dehydrogenase types 1 and 2 messenger ribonucleic acid expression in the rat placenta and decidua during late pregnancy. Biology of Reproduction 55 1023–1028. (doi:10.1095/biolreprod55.5.1023)

Burton  GJ, Fowden  AL & Thornburg  KL 2016 Placental origins of chronic disease. Physiological Reviews 96 1509. (doi:10.1152/physrev.00029.2015)

Chambers  ES, Nanzer  AM, Pfeffer  PE, Richards  DF, Timms  PM, Martineau  AR, Griffiths  CJ, Corrigan  CJ & Hawrylowicz  CM 2015 Distinct endotypes of steroid-resistant asthma characterized by IL-17A high and IFN-γ high immunophenotypes: potential benefits of calcitriol. Journal of Allergy and Clinical Immunology 136 628–637.e624. (doi:10.1016/j.jaci.2015.01.026)

Chan SY, Susarla R, Canovas D, Vasilopoulou E, Ohizua O, McCabe CJ, Hewison M & Kilby MD 2015 Vitamin D promotes human extravillous trophoblast invasion in vitro. Placenta 36 403–409. (doi:10.1016/j.placenta.2014.12.021)

Chen Y-H, Fu L, Hao J-H, Yu Z, Zhu P, Wang H, Xu Y-Y, Zhang C, Tao F-B & Xu D-X 2015a Maternal vitamin D deficiency during pregnancy elevates the risks of small for gestational age and low birth weight infants in Chinese population. Journal of Clinical Endocrinology and Metabolism 100 1912–1919. (doi:10.1210/jc.2014-4407)

Chen YH, Yu Z, Fu L, Wang H, Chen X, Zhang C, Lv ZM & Xu DX 2015b Vitamin D3 inhibits lipopolysaccharide-induced placental inflammation through reinforcing interaction between vitamin D receptor and nuclear factor kappa B p65 subunit. Scientific Reports 5 10871. (doi:10.1038/srep10871)

Chen  Y-Y, Powell  TL & Jansson  T 2017 1,25-Dihydroxy vitamin D3 stimulates system A amino acid transport in primary human trophoblast cells. Molecular and Cellular Endocrinology 442 90–97. (doi:10.1016/j.mce.2016.12.009)

Cleal  JK, Day  PE, Simner  CL, Barton  SJ, Mahon  PA, Inskip  HM, Godfrey KM, Hanson MA, Cooper C, Lewis RM  et al. 2015 Placental amino acid transport may be regulated by maternal vitamin D and vitamin D-binding protein: results from the Southampton Women’s Survey. British Journal of Nutrition 113 1903–1910. (doi:10.1017/S0007114515001178)

Cockburn  F, Belton  NR, Purvis  RJ, Giles  MM, Brown  JK, Turner  TL, Wilkinson EM, Forfar JO, Barrie WJ, McKay GS  et al. 1980 Maternal vitamin D intake and mineral metabolism in mothers and their newborn infants. British Medical Journal 281 11–14. (doi:10.1136/bmj.281.6232.11)

Cole  TJ, Blendy  JA, Monaghan  AP, Krieglstein  K, Schmid  W, Aguzzi  A, Fantuzzi  G, Hummler  E, Unsicker  K & Schütz  G 1995a Targeted disruption of the glucocorticoid receptor gene blocks adrenergic chromaffin cell development and severely retards lung maturation. Genes and Development 9 1608–1621. (doi:10.1101/gad.9.13.1608)

Cole  TJ, Blendy  JA, Monaghan  AP, Schmid  W, Aguzzi  A & Schütz  G 1995b Molecular genetic analysis of glucocorticoid signaling during mouse development. Steroids 60 93–96. (doi:10.1016/0039-128X(94)00009-2)

Cuffe  JSM, Dickinson H, Simmons DG & Moritz KM 2011 Sex specific changes in placental growth and MAPK following short term maternal dexamethasone exposure in the mouse. Placenta 32 981–989. (doi:10.1016/j.placenta.2011.09.009)

Dawodu  A & Wagner  CL 2007 Mother‐child vitamin D deficiency: an international perspective. Archives of Disease in Childhood 92 737–740. (doi:10.1136/adc.2007.122689)

Díaz L, Noyola-Martínez N, Barrera D, Hernández G, Avila E, Halhali A & Larrea F 2009 Calcitriol inhibits TNF-α-induced inflammatory cytokines in human trophoblasts. Journal of Reproductive Immunology 81 17–24. (doi:10.1016/j.jri.2009.02.005)

Diaz R, Brown RW & Seckl JR 1998 Distinct ontogeny of glucocorticoid and mineralocorticoid receptor and 11β-hydroxysteroid dehydrogenase types I and II mRNAs in the fetal rat brain suggest a complex control of glucocorticoid actions. Journal of Neuroscience 18 2570–2580.

Dimasuay KG, Boeuf P, Powell TL & Jansson T 2016 Placental responses to changes in the maternal environment determine fetal growth. Frontiers in physiology 7 12. (doi:10.3389/fphys.2016.00012)

Edwards CRW, Benediktsson R, Lindsay RS & Seckl JR 1993 Dysfunction of placental glucocorticoid barrier: link between fetal environment and adult hypertension? Lancet 341 355–357. (doi:10.1016/0140-6736(93)90148-A)

Evain-Brion  D & Malassine  A 2003 Human placenta as an endocrine organ. Growth Hormone and IGF Research 13 S34–S37. (doi:10.1016/S1096-6374(03)00053-4)

Evans KN, Nguyen L, Chan J, Innes BA, Bulmer JN, Kilby MD & Hewison M 2006 Effects of 25-hydroxyvitamin D3 and 1,25-dihydroxyvitamin D3 on cytokine production by human decidual cells. Biology of Reproduction 75 816–822. (doi:10.1095/biolreprod.106.054056)

Eyles DW, Rogers F, Buller K, McGrath JJ, Ko P, French K & Burne THJ 2006 Developmental vitamin D (DVD) deficiency in the rat alters adult behaviour independently of HPA function. Psychoneuroendocrinology 31 958–964. (doi:10.1016/j.psyneuen.2006.05.006)

Eyles  DW, Burne  THJ & McGrath  JJ 2013 Vitamin D, effects on brain development, adult brain function and the links between low levels of vitamin D and neuropsychiatric disease. Frontiers in Neuroendocrinology 34 47–64. (doi:10.1016/j.yfrne.2012.07.001)

Gernand AD, Simhan HN, Caritis S & Bodnar LM 2014 Maternal vitamin D status and small-for-gestational-age offspring in women at high risk for preeclampsia. Obstetrics and Gynecology 123 40–48. (doi:10.1097/AOG.0000000000000049)

Grundmann M, Haidar M, Haß S, Hubel CA & Versen-Höynck Fv 2012 OS063. Vitamin D promotes endothelial progenitor cell differentiation and upregulates VEGF. Pregnancy Hypertension 2 211. (doi:10.1016/j.preghy.2012.04.064)

Gysler SM, Mulla MJ, Stuhlman M, Sfakianaki AK, Paidas MJ, Stanwood NL, Gariepy A, Brosens JJ, Chamley LW & Abrahams VM 2015 Vitamin D reverses aPL-induced inflammation and LMWH-induced sFlt-1 release by human trophoblast. American Journal of Reproductive Immunology 73 242–250. (doi:10.1111/aji.12301)

Halhali A, Acker GM & Garabedian M 1991 1,25-Dihydroxyvitamin D3 induces in vivo the decidualization of rat endometrial cells. Journal of Reproduction and Fertility 91 59–64. (doi:10.1530/jrf.0.0910059)

Harvey NC, Holroyd C, Ntani G, Javaid K, Cooper P, Moon R, Cole Z, Tinati  T, Godfrey  K & Dennison  E 2014 Vitamin D supplementation in pregnancy: a systematic review. Health Technology Assessment 18 1–190.

Haussler MR, Whitfield GK, Kaneko I, Haussler CA, Hsieh D, Hsieh J-C & Jurutka PW 2013 Molecular mechanisms of vitamin D action. Calcified Tissue International 92 77–98. (doi:10.1007/s00223-012-9619-0)

Hawes  JE, Tesic  D, Whitehouse  AJ, Zosky  GR, Smith  JT & Wyrwoll  CS 2015 Maternal vitamin D deficiency alters fetal brain development in the BALB/c mouse. Behavioural Brain Research 286 192–200. (doi:10.1016/j.bbr.2015.03.008)

Hewison M 2011 Vitamin D and innate and adaptive immunity. Vitamins and hormones 86 23–62. (doi:10.1016/b978-0-12-386960-9.00002-2)

Hewitt DP, Mark PJ & Waddell BJ 2006 Glucocorticoids prevent the normal increase in placental vascular endothelial growth factor expression and placental vascularity during late pregnancy in the rat. Endocrinology 147 5568–5574. (doi:10.1210/en.2006-0825)

Hidalgo  AA, Deeb  KK, Pike  JW, Johnson  CS & Trump  DL 2011 Dexamethasone enhances 1α,25-dihydroxyvitamin D3 effects by increasing vitamin D receptor transcription. Journal of Biological Chemistry 286 36228–36237. (doi:10.1074/jbc.M111.244061)

Holmes MC, Abrahamsen CT, French KL, Paterson JM, Mullins JJ & Seckl JR 2006 The mother or the fetus? 11β-Hydroxysteroid dehydrogenase type 2

Conclusions

Although the diverse effects of vitamin D on placental function may explain many complications associated with vitamin D deficiency in pregnancy, these effects are likely reliant on factors beyond immediate vitamin D pathways. There remains considerable work to be done in teasing apart the exact mechanisms, but the effects of vitamin D deficiency on glucocorticoid metabolism may be an important component for eliciting adverse placental effects. In the wider context, it is tempting to propose that vitamin D supplementation may improve multiple aspects of placental function and maternal physiology. However, there are considerable challenges around appropriate human levels of vitamin D (Lucas & Neale 2014), and thus, this remains an important ongoing area of research.

Declaration of interest

The authors declare that there is no conflict of interest that could be perceived as prejudicing the impartiality of this review.

Funding

This research did not receive any specific grant from any funding agency in the public, commercial or not-for-profit sector.

ReferencesAchkar  M, Dodds  L, Giguère  Y, Forest  J-C, Armson  BA, Woolcott  C,

Agellon  S, Spencer  A & Weiler  HA 2015 Vitamin D status in early pregnancy and risk of preeclampsia. American Journal of Obstetrics and Gynecology 212 511.e511–511.e517. (doi:10.1016/j.ajog.2014.11.009)

Ain R, Canham LN & Soares MJ 2005 Dexamethasone-induced intrauterine growth restriction impacts the placental prolactin family, insulin-like growth factor-II and the Akt signaling pathway. Journal of Endocrinology 185 253–263. (doi:10.1677/joe.1.06039)

Barrera  D, Noyola-Martínez  N, Avila  E, Halhali  A, Larrea  F & Díaz  L 2012 Calcitriol inhibits interleukin-10 expression in cultured human trophoblasts under normal and inflammatory conditions. Cytokine 57 316–321. (doi:10.1016/j.cyto.2011.11.020)

Beitins IZ, Bayard F, Ances IG, Kowarski A & Migeon CJ 1973 The metabolic clearance rate, blood production, interconversion and transplacental passage of cortisol and cortisone in pregnancy near term. Pediatric Research 7 509–519. (doi:10.1203/00006450-197305000-00004)

Benediktsson  R, Lindsay  R, Noble  J, Seckl  J & Edwards  C 1993 Glucocorticoid exposure in utero: new model for adult hypertension. Lancet 341 339–341. (doi:10.1016/0140-6736(93)90138-7)

Benediktsson R, Calder AA, Edwards CR & Seckl JR 1997 Placental 11β‐hydroxysteroid dehydrogenase: a key regulator of fetal glucocorticoid exposure. Clinical Endocrinology 46 161–166. (doi:10.1046/j.1365-2265.1997.1230939.x)

Bodnar LM, Catov JM, Simhan HN, Holick MF, Powers RW & Roberts JM 2007 Maternal vitamin D deficiency increases the risk of preeclampsia. Journal of Clinical Endocrinology and Metabolism 92 3517–3522. (doi:10.1210/jc.2007-0718)

Bodnar LM, Krohn MA & Simhan HN 2009 Maternal vitamin D deficiency is associated with bacterial vaginosis in the first trimester of pregnancy. Journal of Nutrition 139 1157–1161. (doi:10.3945/jn.108.103168)

Downloaded from Bioscientifica.com at 04/28/2022 11:03:52PMvia free access

Page 8: Vitamin D deficiency and impaired placental function

PROOF ONLYN Yates and othersR170

Reproduction (2017) 153 R163–R171 www.reproduction-online.org

null mice provide evidence for direct fetal programming of behavior by endogenous glucocorticoids. Journal of Neuroscience 26 3840–3844. (doi:10.1523/JNEUROSCI.4464-05.2006)

Javaid MK, Crozier SR, Harvey NC, Gale CR, Dennison EM, Boucher BJ, Arden  NK, Godfrey  KM & Cooper  C 2006 Maternal vitamin D status during pregnancy and childhood bone mass at age 9 years: a longitudinal study. Lancet 367 36–43. (doi:10.1016/S0140-6736(06)67922-1)

Kassi  E, Nasiri-Ansari  N, Spilioti  E, Kalotychou  V, Apostolou  PE, Moutsatsou  P & Papavassiliou  AG 2016 Vitamin D interferes with glucocorticoid responsiveness in human peripheral blood mononuclear target cells. Cellular and Molecular Life Sciences 73 4341–4354. (doi:10.1007/s00018-016-2281-3)

Kirby BJ, Ma Y, Martin HM, Buckle Favaro KL, Karaplis AC & Kovacs CS 2013 Upregulation of calcitriol during pregnancy and skeletal recovery after lactation do not require parathyroid hormone. Journal of Bone and Mineral Research 28 1987–2000. (doi:10.1002/jbmr.1925)

Kovacs CS 2008 Vitamin D in pregnancy and lactation: maternal, fetal, and neonatal outcomes from human and animal studies. Journal of Clinical Nutrition 88 520S–528S.

Kovacs CS, Woodland ML, Fudge NJ & Friel JK 2005 The vitamin D receptor is not required for fetal mineral homeostasis or for the regulation of placental calcium transfer in mice. American Journal of Physiology: Endocrinology and Metabolism 289 E133–E144. (doi:10.1152/ajpendo.00354.2004)

Langley-Evans  S, Phillips  G, Benediktsson  R, Gardner  D, Edwards  C, Jackson  A & Seckl  J 1996 Protein intake in pregnancy, placental glucocorticoid metabolism and the programming of hypertension in the rat. Placenta 17 169–172. (doi:10.1016/S0143-4004(96)80010-5)

Lindsay R, Lindsay R, Waddell B & Seckl J 1996a Prenatal glucocorticoid exposure leads to offspring hyperglycaemia in the rat: studies with the 11 b-hydroxysteroid dehydrogenase inhibitor carbenoxolone. Diabetologia 39 1299–1305. (doi:10.1007/s001250050573)

Lindsay R, Lindsay RM, Edwards CRW & Seckl JR 1996b Inhibition of 11β-hydroxysteroid dehydrogenase in pregnant rats and the programming of blood pressure in the offspring. Hypertension 27 1200–1204. (doi:10.1161/01.HYP.27.6.1200)

Liu  PT, Stenger  S, Li  H, Wenzel  L, Tan  BH, Krutzik  SR, Ochoa  MT, Schauber  J, Wu  K & Meinken  C 2006 Toll-like receptor triggering of a vitamin D-mediated human antimicrobial response. Science 311 1770–1773. (doi:10.1126/science.1123933)

Liu N, Kaplan AT, Low J, Nguyen L, Liu GY, Equils O & Hewison M 2009 Vitamin D induces innate antibacterial responses in human trophoblasts via an intracrine pathway. Biology of Reproduction 80 398–406. (doi:10.1095/biolreprod.108.073577)

Liu  NQ, Kaplan  AT, Lagishetty  V, Ouyang  YB, Ouyang  Y, Simmons  CF, Equils O & Hewison M 2011 Vitamin D and the regulation of placental inflammation. Journal of Immunology 186 5968–5974. (doi:10.4049/jimmunol.1003332)

Liu NQ, Ouyang Y, Bulut Y, Lagishetty V, Chan SY, Hollis BW, Wagner C, Equils O & Hewison M 2013 Dietary vitamin D restriction in pregnant female mice is associated with maternal hypertension and altered placental and fetal development. Endocrinology 154 2270–2280. (doi:10.1210/en.2012-2270)

Looker  AC, Pfeiffer  CM, Lacher  DA, Schleicher  RL, Picciano  MF & Yetley EA 2008 Serum 25-hydroxyvitamin D status of the US population: 1988–1994 versus 2000–2004. American Journal of Clinical Nutrition 88 1519–1527. (doi:10.3945/ajcn.2008.26182)

Lucas R & Neale R 2014 What is the optimal level of vitamin D? Separating the evidence from the rhetoric. Australian Family Physician 43 119.

Maghbooli  Z, Hossein-Nezhad  A, Shafaei  AR, Karimi  F, Madani  FS & Larijani B 2007 Vitamin D status in mothers and their newborns in Iran. BMC Pregnancy and Childbirth 7 1. (doi:10.1186/1471-2393-7-1)

Mahon  P, Harvey  N, Crozier  S, Inskip  H, Robinson  S, Arden  N, Swaminathan  R, Cooper  C & Godfrey  K 2010 Low maternal vitamin D status and fetal bone development: cohort study. Journal of Bone and Mineral Research 25 14–19. (doi:10.1359/jbmr.090701)

Mairesse  J, Lesage  J, Breton  C, Bréant  B, Hahn  T, Darnaudéry  M, Dickson SL, Seckl J, Blondeau B & Vieau D 2007 Maternal stress alters endocrine function of the feto-placental unit in rats. American Journal of Physiology: Endocrinology and Metabolism 292 E1526–E1533. (doi:10.1152/ajpendo.00574.2006)

Menegaz  D, Mizwicki  MT, Barrientos-Duran  A, Chen  N, Henry  HL & Norman AW 2011 Vitamin D receptor (VDR) regulation of voltage-gated chloride channels by ligands preferring a VDR-alternative pocket (VDR-AP). Molecular Endocrinology 25 1289–1300. (doi:10.1210/me.2010-0442)

Nguyen  T, Yong  H, Borg  A, Brennecke  S & Murthi  P 2015a Decreased placental vitamin D receptor contributes to aberrant cell-cycle gene regulation in human idiopathic fetal growth restriction. Placenta 36 A44. (doi:10.1016/j.placenta.2015.07.318)

Nguyen T, Yong H, Chollangi T, Borg A, Brennecke S & Murthi P 2015b Placental vitamin D receptor expression is decreased in human idiopathic fetal growth restriction. Journal of Molecular Medicine 93 795–805. (doi:10.1007/s00109-015-1267-1)

Ni W, Watts SW, Ng M, Chen S, Glenn DJ & Gardner DG 2014 Elimination of vitamin D receptor in vascular endothelial cells alters vascular function. Hypertension 64 1290. (doi:10.1161/HYPERTENSIONAHA.114.03971)

Norman AW 2008 From vitamin D to hormone D: fundamentals of the vitamin D endocrine system essential for good health. American Journal of Clinical Nutrition 88 491S–499S. (doi:10.3945/ajcn.2008.27049)

Norman AW, Olivera CJ, Barreto Silva  FR & Bishop  JE 2002 A specific binding protein/receptor for 1alpha,25-dihydroxyvitamin D(3) is present in an intestinal caveolae membrane fraction. Biochemical and Biophysical Research Communications 298 414–419. (doi:10.1016/S0006-291X(02)02482-8)

Noyola-Martínez N, Díaz L, Avila E, Halhali A, Larrea F & Barrera D 2013 Calcitriol downregulates TNF-α and IL-6 expression in cultured placental cells from preeclamptic women. Cytokine 61 245–250. (doi:10.1016/j.cyto.2012.10.001)

Nyirenda  MJ, Lindsay  RS, Kenyon  CJ, Burchell  A & Seckl  JR 1998 Glucocorticoid exposure in late gestation permanently programs rat hepatic phosphoenolpyruvate carboxykinase and glucocorticoid receptor expression and causes glucose intolerance in adult offspring. Journal of Clinical Investigation 101 2174. (doi:10.1172/JCI1567)

Obradovic D, Gronemeyer H, Lutz B & Rein T 2006 Cross-talk of vitamin D and glucocorticoids in hippocampal cells. Journal of Neurochemistry 96 500–509. (doi:10.1111/j.1471-4159.2005.03579.x)

Pet MA & Brouwer-Brolsma EM 2016 The impact of maternal vitamin D status on offspring brain development and function: a systematic review. Advances in Nutrition 7 665–678. (doi:10.3945/an.115.010330)

Prietl B, Treiber G, Pieber TR & Amrein K 2013 Vitamin D and immune function. Nutrients 5 2502–2521. (doi:10.3390/nu5072502)

Qin L-L, Lu F-G, Yang S-H, Xu H-L & Luo B-A 2016 Does maternal vitamin D deficiency increase the risk of preterm birth: a meta-analysis of observational studies. Nutrients 8 301. (doi:10.3390/nu8050301)

Rog-Zielinska  EA, Thomson  A, Kenyon  CJ, Brownstein  DG, Moran  CM, Szumska D, Michailidou Z, Richardson J, Owen E, Watt A  et al. 2013 Glucocorticoid receptor is required for foetal heart maturation. Human Molecular Genetics 22 3269–3282. (doi:10.1093/hmg/ddt182)

Samuel  S & Sitrin  MD 2008 Vitamin D’s role in cell proliferation and differentiation. Nutrition Reviews 66 S116. (doi:10.1111/j.1753-4887.2008.00094.x)

Seckl JR & Holmes MC 2007 Mechanisms of disease: glucocorticoids, their placental metabolism and fetal ‘programming’ of adult pathophysiology. Nature Clinical Practice Endocrinology and Metabolism 3 479–488. (doi:10.1038/ncpendmet0515)

Sferruzzi-Perri  AN & Camm  EJ 2016 The programming power of the placenta. Frontiers in Physiology 7 33. (doi:10.3389/fphys.2016.00033)

Shahbazi M, Jeddi-Tehrani M, Zareie M, Salek-Moghaddam A, Akhondi MM, Bahmanpoor M, Sadeghi MR & Zarnani AH 2011 Expression profiling of vitamin D receptor in placenta, decidua and ovary of pregnant mice. Placenta 32 657–664. (doi:10.1016/j.placenta.2011.06.013)

Stewart PM, Rogerson FM & Mason J 1995 Type 2 11 beta-hydroxysteroid dehydrogenase messenger ribonucleic acid and activity in human placenta and fetal membranes: its relationship to birth weight and putative role in fetal adrenal steroidogenesis. Journal of Clinical Endocrinology and Metabolism 80 885–890. (doi:10.1210/jcem.80.3.7883847)

Sugden  MC, Langdown  ML, Munns  MJ & Holness  MJ 2001 Maternal glucocorticoid treatment modulates placental leptin and leptin receptor expression and materno-fetal leptin physiology during late pregnancy, and elicits hypertension associated with hyperleptinaemia in the early-growth-retarded adult offspring. European Journal of Endocrinology 145 529–539. (doi:10.1530/eje.0.1450529)

Downloaded from Bioscientifica.com at 04/28/2022 11:03:52PMvia free access

Page 9: Vitamin D deficiency and impaired placental function

PROOF ONLYVitamin D, placenta and glucocorticoids R171

www.reproduction-online.org Reproduction (2017) 153 R163–R171

Surbek D, Drack G, Irion O, Nelle M, Huang D & Hoesli I 2012 Antenatal corticosteroids for fetal lung maturation in threatened preterm delivery: indications and administration. Archives of Gynecology and Obstetrics 286 277–281. (doi:10.1007/s00404-012-2339-x)

Tamblyn J, Hewison M, Wagner C, Bulmer J & Kilby M 2015 Immunological role of vitamin D at the maternal–fetal interface. Journal of Endocrinology 224 R107–R121. (doi:10.1530/JOE-14-0642)

Tare  M, Emmett  SJ, Coleman  HA, Skordilis  C, Eyles  DW, Morley  R & Parkington HC 2011 Vitamin D insufficiency is associated with impaired vascular endothelial and smooth muscle function and hypertension in young rats. Journal of Physiology 589 4777–4786. (doi:10.1113/jphysiol.2011.214726)

Tesic D, Hawes JE, Zosky GR & Wyrwoll CS 2015 Vitamin D deficiency in BALB/c mouse pregnancy increases placental transfer of glucocorticoids. Endocrinology 156 3673–3679. (doi:10.1210/en.2015-1377)

Thompson  A, Han  VKM & Yang  K 2004 Differential expression of 11β-hydroxysteroid dehydrogenase types 1 and 2 mRNA and glucocorticoid receptor protein during mouse embryonic development. Journal of Steroid Biochemistry and Molecular Biology 88 367–375. (doi:10.1016/j.jsbmb.2003.12.014)

Vaughan OR, Sferruzzi-Perri AN & Fowden AL 2012 Maternal corticoste-rone regulates nutrient allocation to fetal growth in mice. Journal of Physiology 590 5529–5540. (doi:10.1113/jphysiol.2012.239426)

Vaughan OR, Sferruzzi-Perri AN, Coan PM & Fowden AL 2013 Adaptations in placental phenotype depend on route and timing of maternal dexamethasone administration in mice. Biology of Reproduction 89 80. (doi:10.1095/biolreprod.113.109678)

Vieau  D, Sebaai  N, Léonhardt  M, Dutriez-Casteloot  I, Molendi-Coste  O, Laborie  C, Breton  C, Deloof  S & Lesage  J 2007 HPA axis programming by maternal undernutrition in the male rat offspring. Psychoneuroendocrinology 32 S16–S20. (doi:10.1016/j.psyneuen.2007.03.014)

Wharfe  MD, Wyrwoll  CS, Waddell  BJ & Mark  PJ 2016 Pregnancy suppresses the daily rhythmicity of core body temperature and adipose metabolic gene expression in the mouse. Endocrinology 157 3320–3331. (doi:10.1210/en.2016-1177)

Whitehouse AJO, Holt BJ, Serralha M, Holt PG, Kusel MMH & Hart PH 2012 Maternal serum vitamin d levels during pregnancy and offspring

neurocognitive development. Pediatrics 129 485–493. (doi:10.1542/peds.2011-2644)

Wilson RL, Buckberry S, Spronk F, Laurence  JA, Leemaqz S, O’Leary S, Bianco-Miotto  T, Du  J, Anderson  PH & Roberts  CT 2015 Vitamin D receptor gene ablation in the conceptus has limited effects on placental morphology, function and pregnancy outcome. PLoS ONE 10 e0131287. (doi:10.1371/journal.pone.0131287)

Wyrwoll CS, Mark PJ, Mori TA, Puddey IB & Waddell BJ 2006 Prevention of programmed hyperleptinemia and hypertension by postnatal dietary ω-3 fatty acids. Endocrinology 147 599–606. (doi:10.1210/en.2005-0748)

Wyrwoll CS, Seckl  JR & Holmes MC 2009 Altered placental function of 11β-hydroxysteroid dehydrogenase 2 knockout mice. Endocrinology 150 1287–1293. (doi:10.1210/en.2008-1100)

Young BE, McNanley TJ, Cooper EM, McIntyre AW, Witter F, Harris ZL & O’Brien  KO 2012 Maternal vitamin D status and calcium intake interact to affect fetal skeletal growth in utero in pregnant adolescents. American Journal of Clinical Nutrition 95 1103–1112. (doi:10.3945/ajcn.111.023861)

Zehnder  D, Evans  KN, Kilby  MD, Bulmer  JN, Innes  BA, Stewart  PM & Hewison M 2002 The ontogeny of 25-hydroxyvitamin D 3 1α-hydroxylase expression in human placenta and decidua. American Journal of Pathology 161 105–114. (doi:10.1016/S0002-9440(10)64162-4)

Zosky  GR, Hart  PH, Whitehouse  AJO, Kusel  MM, Ang  W, Foong  RE, Chen L, Holt PG, Sly PD & Hall GL 2014 Vitamin D deficiency at 16 to 20 weeks’ gestation is associated with impaired lung function and asthma at 6 years of age. Annals of the American Thoracic Society 11 571–577. (doi:10.1513/AnnalsATS.201312-423OC)

Received 30 November 2016First decision 9 January 2017Revised manuscript received 22 January 2017Accepted 30 January 2017

Downloaded from Bioscientifica.com at 04/28/2022 11:03:52PMvia free access