You are on page 1of 41

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24

The importance of antioxidant micronutrients in pregnancy Hiten D. Mistry1 & Paula J. Williams2*
1

Division of Womens Health, Maternal and Fetal Research Unit, St. Thomas Hospital, Kings College

London, London, SE1 7UH, UK;


2

Human Genetics, School of Molecular and Medical Sciences, University of Nottingham, Queens Medical

Centre, Nottingham, NG7 2UH, UK .

To whom correspondence should be addressed:

Dr Paula J Williams Human Genetics, School of Molecular Medical Sciences, University of Nottingham, A Floor West Block, Queen's Medical Centre, Nottingham NG7 2UH E- mail: Paula.Williams@nottingham.ac.uk

25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47

Abstract Pregnancy places increased demands on the mother to provide adequate nutrition to the growing conceptus. A number of micronutrients function as essential cofactors for or themselves acting as antioxidants. Oxidative stress is generated during normal placental development however, when supply of antioxidant micronutrients is limited, exaggerated oxidative stress within both the placenta and maternal circulation occurs, resulting in adverse pregnancy outcomes. The present literature review summarises the current understanding of selected micronutrient antioxidants selenium, copper, zinc, manganese and vitamins C & E in pregnancy. To summarise antioxidant activity of selenium is via its incorporation into the glutathione peroxidase enzymes, levels of which have been shown to be reduced in miscarriage and pre-eclampsia. Copper, zinc and manganese are all essential cofactors for superoxide dismutases, which has reduced activity in pathological pregnancy. Larger intervention trials are required to reinforce or refute a beneficial role of micronutrient supplementation in disorders of pregnancies.

Keywords: Pregnancy, antioxidants, micronutrients, vitamins.

48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65 66 67 68 69 70

Introduction Nutrition in Pregnancy The importance of proper nutrition prior to and throughout pregnancy has long been known for optimising the health and wellbeing of both mother and baby [1]. Pregnancy is a period of increased metabolic demands with changes in a womens physiology and the requirements of a growing fetus [2, 3]. Insufficient supplies of essential vitamins and micronutrients can lead to a state of biological competition between the mother and conceptus, which can be detrimental to the health status of both [4] Deficiency of trace elements during pregnancy is closely related to mortality and morbidity in the new born [5]. Deficiencies of specific antioxidant activities associated with the micronutrients selenium, copper, zinc and manganese can result in poor pregnancy outcomes, including fetal growth restriction [6], pre-eclampsia [7] and the associated increased risk of diseases in adulthood, including cardiovascular disease and type 2 diabetes [8-11].

A large body of research has investigated the role of macronutrients in pregnancy and especially the effects of both maternal under and over nutrition on the long term health of the offspring [12, 13]. A number of hypotheses have been suggested to explain the contribution of maternal nutrition during the fetal and embryonic periods and the programming of the cardiovascular and metabolic system of the offspring [14, 15]. In addition to the contribution of macronutrition to successful pregnancy more recently studies have begun to focus on the role of essential specific micronutrients, so called because they are an absolute requirement and are required in only small amounts daily [16].

71 72 73 74 75 76 77 78 79 80 81 82 83 84 85 86 87 88 89 90 91 92 93

The pregnant mother must provide a source of nourishment and gaseous exchange to enable maximal embryonic and fetal growth to occur, whilst at the same time preparing her body for labour and parturition and the later demands of lactation. In addition a careful balance must be maintained between providing immune surveillance to protect the mother from infection, whilst at the same time allowing the implantation and survival of the semi-allogenic conceptus [17, 18]. The development and establishment of the placenta and its circulatory system is crucial in the successful maintenance of both maternal health and also enabling development of the embryo and growth of the fetus.

The establishment of the feto-placental circulation requires the invasion of placental derived extravillous trophoblast from anchoring cytotrophoblast columns through the maternal decidua and into the maternal spiral arteries (Figure 1). During the first 8 weeks of pregnancy trophoblast plugs within the spiral arteries which exist to protect embryonic DNA from damage by oxidative stress [19], are released allowing the onset of placental circulation (Figure 1). The release of trophoblast plugs with flow of blood into the intervillous space leads to the generation of oxidative stress [20]. However, the placenta is armed with antioxidant defences, including the selenium-dependent glutathione peroxidases, thioredoxin reductases, selenoprotein-P and copper/zinc and manganese superoxide dismutases (Cu/Zn and Mn SODs) (Figure 2), which protect the placenta from any undue harm [21-24]. As the extravillous trophoblast cells migrate through the spiral artery wall they are involved in the process of physiological conversion, which serves to enlarge the vessel lumen thereby maximising blood supply to the intervillous space, enabling maximal perfusion of nutrients and gases through the placental

94 95 96 97 98 99 100 101 102 103 104 105 106 107 108 109 110 111 112 113 114 115 116

syncytiotrophoblast. Deficient extravillous trophoblast invasion is associated with reduced spiral artery remodelling leading to reduced blood flow and increased oxidative stress within the placenta and is known to be the primary defect occurring in preeclampsia [25, 26], fetal growth restriction [27, 28] and sporadic miscarriage [29]. Additionally, pre-eclampsia has also been associated with reduced levels of antioxidant enzyme protection causing further placental damage [30-32].

The pathogenesis of adverse pregnancy outcomes including pre-eclampsia and fetal growth restriction [33] and a number of neonatal outcomes [34] have been shown to be associated with oxidative stress. Pre-eclampsia (de novo proteinuric hypertension) is estimated to occur in ~3% of all pregnancies and is a leading cause of maternal and perinatal mortality and morbidity in the Western world [35, 36]; together with other hypertensive disorders of pregnancy, pre-eclampsia is responsible for approximately 60,000 maternal deaths each year [37] and increases perinatal mortality five-fold [38]. Optimal outcome for the mother and child often dictates that the infant is delivered early leading to increased preterm delivery and low infant birthweight rates. Placental and maternal systemic oxidative stress are components of the syndrome [39] and contribute to a generalised maternal systemic inflammatory activation [40]. Placental ischaemiareperfusion injury has been implicated in excessive production of ROS, causing release of placental factors that mediate the inflammatory responses [41].

Fetal growth restriction is associated with increased perinatal mortality and morbidity [42]. The mechanisms are still to be elucidated but a likely factor is placental

117 118 119 120 121 122 123 124 125 126 127 128 129 130 131 132 133 134 135 136 137 138 139

ischemia/hypoxia [43] but it is thought that ischemia-reperfusion injury may contribute to the oxidative stress and could result in the release of reactive oxygen species into the maternal circulation possibly resulting in oxidative DNA damage and may underlie development of fetal growth restriction [44].

The current review focuses specifically on those micronutrients which are associated with antioxidant activity due to the importance of oxidative stress in both normal pregnancy and pathological pregnancy outcomes. However, it is important to highlight that other micronutrients are also important in pregnancy [16] but are outside the scope of the current review.

Methods A number of micronutrients and vitamins are known to serve as antioxidants or be essential cofactors for antioxidant enzymes; these include, selenium, copper, zinc, manganese and vitamins C and E. The present literature review summarises current understanding of the importance of these micronutrient antioxidants in pregnancy. For this review, we included data and relevant information obtained through a search of the PubMed database using free text and Medical Subject Headings terms for all articles published in English from 1971 through 2011 which included the term selenium, zinc, copper, manganese, vitamin E and C and one of the following: pregnancy, preeclampsia or fetal growth restriction. We further included published and unpublished data from our own laboratories and an in- house library of relevant publications. The procedure was concluded by the perusal of the reference sections of all relevant studies or

140 141 142 143 144 145 146 147 148 149 150 151 152 153 154 155 156 157 158 159 160 161 162

reviews, a manual search of key journals and abstracts from the major annual meetings in the field of pregnancy and nutrition and contact with experts on the subject, in an effort to identify relevant unpublished data.

Selenium Selenium is incorporated into proteins to make selenoproteins, including the glutathione peroxidase antioxidant enzymes, thioredoxin reductases and selenoprotein-P [45]. In addition, selenium is essential for the production of active thyroid hormones and is essential for normal thyroid function [46]. Maternal selenium concentrations and glutathione peroxidase activity fall during pregnancy (selenium concentrations in 1st trimester: 65 g/L; 3rd trimester: 50 g/L) [47, 48]. Worldwide differences exist in assessment of selenium requirements, adequacy and intakes (Table 1). Most of these values have been determined from the intake believed necessary to maximise the activity of the antioxidant glutathione peroxidase in plasma [49, 50] and selenium, like vitamins C and E has received much attention in recent years. It has been observed that babies on average have lower selenium concentrations compared to the mother (Maternal selenium 58.4 g/L; Umbilical cord selenium: 42.1 g/L) [32, 51], which is expected as selenium is transported via the placenta across a concentration gradient via an anion exchange pathway, shared with sulphate [52, 53].

Recurrent early pregnancy loss has been associated with reduced serum selenium concentrations compared to healthy controls in two observational studies from UK (mean SD: 54 19 vs. 76 14 g/L respectively) [54] and Turkey (55 17 vs. 81 16 g/L

163 164 165 166 167 168 169 170 171 172 173 174 175 176 177 178 179 180 181 182 183 184 185

respectively) [55]. It has therefore been suggested that reduced selenium concentration results in reduced glutathione peroxidase activity culminating in reduced antioxidant protection of biological membranes and DNA during the early stages of embryonic development [54, 56]. Although speculative, and requiring larger placebo-controlled randomised trials, women with recurrent early pregnancy loss may benefit from optimisation of selenium status.

Recently, our group and others have demonstrated through retrospective studies, the association between low serum selenium concentrations and reduced antioxidant function of the associated antioxidant glutathione peroxidase enzymes in women with preeclampsia (defined a de novo proteinuric hypertension) [32, 57, 58]. It has been suggested that adequate selenium status is important for antioxidant defence and may be a potential factor in women at risk of pre-eclampsia; this hypothesis has been further justified by the reduced expression and activities of glutathione peroxidase found in maternal, fetal and placental samples taken from 25 pre-eclamptic pregnancies, when compared to 27 normal controls in our recent cross-sectional retrospective study [32]. Dawson et al, also completed a retrospective study in the US and reported lower amniotic fluid selenium concentrations in 29 pre-eclamptics delivering between 33-36 weeks gestation compared to 48 gestation- matched controls (10 1 vs. 7 0.7 g/L respectively) [59].

Fetal growth restriction or delivery of a small- for- gestational-age (SGA) is defined as an individualised birth weight ratio below the 10 th percentile [42]. Reports of selenium concentrations with regards to fetal growth restriction are inconsistent. A retrospective

186 187 188 189 190 191 192 193 194 195 196 197 198 199 200 201 202 203 204 205 206 207

study had reported low placental selenium concentrations in 49 mothers affected by fetal growth restriction, compared to 36 healthy normal birth weight controls [60], whereas others have reported higher [61, 62] or unchanged concentrations [63]. Another retrospective study also demonstrated that in 81 SGA babies, infant plasma selenium concentrations to be significantly lower compared to controls [64]. A recent retrospective study by our group on an adolescent cohort [65] found lower maternal plasma selenium concentrations on 28 mothers who delivered SGA babies compared to 143 healthy controls [66]. Further studies are warranted to fully investigate the potential link between selenium deficiency and fetal growth restriction.

Only a small number of selenium supplementation trials during pregnancy have been carried out to date. A prospective, randomised, placebo controlled study of selenium supplementation during and after pregnancy in pregnant women positive for thyroid peroxidase antibodies (TPOAb(+)) who are prone to develop postpartum thyroid dysfunction (PPTD) and permanent hypothyroidism found that supplementation with 200 g/d selenomethionine significantly reduced the incidence of PPTD and permanent hypothyroidism [67]. The authors concluded that selenium supplementation during pregnancy and in the postpartum period reduced thyroid inflammatory activity and the incidence of hypothyroidism, possibly by increasing the selenoproteins glutathione peroxidase or iodothyronine deiodinase activities, thereby contributing in part, to counterbalance the postpartum immunological rebound [67].

208 209 210 211 212 213 214 215 216 217 218 219 220 221 222 223 224 225 226 227 228 229 230

To date there have been a couple of small placebo-controlled randomised control trials on selenium supplementation, reporting lower rates of pre-eclampsia and/or pregnancy induced hypertension in the supplemented groups [68-70]. It must be noted that neither of these studies adequately addressed the role of supplementation on the incidence of preeclampsia. Currently the Selenium in Pregnancy Intervention Trial (SPRINT) is underway in the UK, jointly by the University of Surrey and Oxford. This is a small randomised controlled trial of selenium supplementation (60 g a day). It is not powered to demonstrate clinical benefit but will provide insight into the impact of selenium supplements on laboratory measurements of circulating factors that are relevant to the development of pre-eclampsia. If this is successful a much larger multicentre trial will be needed to analyse clinical benefit.

Coppe r Copper is an essential cofactor for a number of enzymes involved in metabolic reactions, angiogenesis, oxygen transport and antioxidant protection, including catalase, superoxide dismutase (SOD) (Figure 2) and cytochrome oxidase [71]. During pregnancy, plasma copper concentrations significantly increase, returning to normal non-pregnant values after delivery (Mean SD - 1st trimester: 147.6 34.6; 3rd trimester: 204.2 41.8 g/L) [72-74]. The increase in copper with progression of pregnancy could be partly related to synthesis of ceruloplasmin, a major copper binding protein, due to altered levels of oestrogen [74]. Approximately 96% of plasma copper is strongly bound to ceruloplasmin [75], a protein with antioxidant ferroxidase properties [75, 76]. The dietary intake of copper in women aged between 19-24 years is generally below the recommended levels

10

231 232 233 234 235 236 237 238 239 240 241 242 243 244 245 246 247 248 249 250 251 252 253

(Table 1) [77] which may cause problems during pregnancy when requirements increase [78].

Copper is essential for embryonic development [79]. Maternal dietary deficiency can result in both short term consequences including early embryonic death and gross structural abnormalities, as well as long term consequences such as increased r isk of cardiovascular disease risk and reduced fertilisation rates [71, 80]; current recommendations on intakes are summarised in Table 1. Severe copper deficiency can lead to reproductive failure and early embryonic death [81], whereas mild or moderate deficiency has little effect on either the number of live births or neonatal weight [82].

Cu/Zn SOD is an important antioxidant known to be expressed in both maternal and fetal tissues [83]. Copper concentration has been shown to be higher in maternal plasma than in umbilical cord plasma [84-86]. It has been suggested that the placenta acts as a blockade in the transfer of copper from the mother to the fetus [86, 87]. A recent observational Turkish study on 61 placentae from healthy pregnancies between 37 and 40 weeks gestation found that copper concentrations positively correlated with neonatal weight; the authors suggested that copper may have interactive connections in human placenta [88] and this requires further studies to fully elucidate its role. It is known that copper is transferred across the placenta via high affinity copper transporter (CTR1) and have been shown to be expressed early in pregnancy and it is also thought that placental copper transport is related to iron transport but the mechanism is unknown [78].

11

254 255 256 257 258 259 260 261 262 263 264 265 266 267 268 269 270 271 272 273 274 275 276

A small retrospective study in the 1980s reported increased placental copper concentrations in 8 pre-eclamptic women compared to 10 controls (53 vs. 124 g/Kg respectively), suggesting that this increase in pre-eclampsia maybe an exaggerated response of normal pregnancies [89]. Further retrospective studies from Turkey have shown elevation of maternal serum copper levels in pre-eclampsia after clinical onset of the disease compared to controls (mean SD: 159 38 vs. 194 52 g/L respectively) [90, 91]. Moreover, increased amniotic fluid copper concentration from 19 pre-eclamptic women compared to 53 controls has also been reported (mean SD: 19 5 vs. 29 3 g/L respectively) [59]. It is thought that as copper is a redox-active transition metal and can participate in single electron reactions and catalyse the formation of free radicals, including undesirable hydroxyl radicals, it could contribute to oxidative stress characteristic of pre-eclampsia [90]. This illustrates that copper itself appears to act as a pro-oxidant, but when associated in Cu/Zn SOD functions as a n antioxidant. Furthermore, studies have reported increased levels of serum ceruloplasmin in women with pre-eclampsia; this positively correlated with serum malondialdehyde, suggesting an increased production of this antioxidant protein in response to increased lipid peroxidation [90, 92, 93], although further studies are required to confirm this hypothesis. However, it must be remembered that concentrations early in pregnancy have yet to be determined and data on copper status in normal human p regnancies are sparse. This is further hampered by the fact that at present there is no reliable biomarker for copper status, so whether deficiency is a significant public health problem remains unclear [94].

12

277 278 279 280 281 282 283 284 285 286 287 288 289 290 291 292 293 294 295 296 297 298 299

Zinc Zinc is an essential constituent of over 200 metalloenzymes participating in carbohydrate and protein metabolism, nucleic acid synthesis, antioxidant functions (through Cu/Zn SOD; Fig. 2) and other vital functions such cellular division and differentiation, making it essential for successful embryogenesis. [72]. It was estimated in 2002 by the World Health Organisation that suboptimal zinc nutrition effected nearly half the worlds population [95]. During pregnancy, zinc is also used to assist the fetus to develop the brain and also to be an aid to the mother in labour [96]. It has been estimated that the total amount of zinc retained during pregnancy is ~ 100 mg [97]. The requirement of zinc during the third trimester is approximately twice as high as that in non-pregnant women [98]. Plasma zinc concentrations decline as pregnancy progresses (mean SD 1st trimester: 71.3 12.9 g/L; 3rd trimester: 58.5 11.5 g/L) [72, 74, 99, 100]. In addition, a nutrition analysis revealed that in pregnant women the everyday diet intakes includes not more than 50% of the daily requirement of zinc [101].

Alteration in zinc homeostasis may have devastating effects on pregnancy outcome, including prolonged labour, fetal growth restriction, or embryonic or fetal death [102]. Zinc is a trace element with a great importance for fetal growth re striction where it is used in order to improve fetal growth [72]. Goldenberg et al completed a randomised double-blind placebo-controlled trail of zinc supplementation (25 mg per day) during pregnancy from 19 weeks gestation in African-American women (294 in supplemented and 286 in placebo group). Those given zinc supplementation had a significantly greater birth weight and head circumference compared to the placebo group highlighting the

13

300 301 302 303 304 305 306 307 308 309 310 311 312 313 314 315 316 317 318 319 320 321 322

importance of adequate zinc supply during pregnancy [103]. A limitation of many of the randomised controlled trails of zinc supplementation and pregnancy outcome is that they lacked the sample sizes to detect differences [102].

Many of the zinc supplementation studies have been conducted in developing countries where incidence of zinc deficiency is high and these women are often selected as they are less well nourished or have low plasma zinc levels [103, 104]; benefits of supplementation include reduced incidence of pregnancy- induced hypertension or low birth weights [103]. These studies do however suggest the benefits of zinc supplementation in developing countries where zinc deficiency is likely, although for developed countries there is conflicting data as to the benefits [105-108].

Zinc deficiency has been associated with pre-eclampsia since the 1980s [59, 89, 109] including adolescent pregnancies [110]. Placental zinc concentration has also been shown to be lower in pre-eclampsia in cross-sectional retrospective studies with placental zinc values positively correlating with birth weights [89, 111, 112]. More recently lower serum concentrations of zinc in pre-eclampsia compared to controls have been shown in two relatively small retrospective studies from Turkey (mean SD: 10.6 4.4 vs. 12.7 4.1 g/L respectively) [91, 113]; the authors suggested that this may be useful for early diagnosis as lower plasma zinc concentrations have been associated with increased lipid peroxidation in rat studies [114]. Moreover, in a recent retrospective study in India reported reduced serum zinc concentrations in mild and severe pre-eclamptic mothers compared to controls; the authors suggest that the reduction could not only effect the

14

323 324 325 326 327 328 329 330 331 332 333 334 335 336 337 338 339 340 341 342 343 344 345

antioxidant protection but could also contribute to the rise in blood pressure [115]. The lower serum zinc concentrations in mothers who develop pre-eclampsia has been suggested to at least be partly due to reduced oestrogen and zinc binding-protein levels [116]. Zinc is transported across the placenta via active transport from the mother to the fetus [109]. Studies have shown that the fetus has notably higher zinc concentrations compared to the mother, even in cases of pre-eclampsia [109, 117], indicating that the fetus itself, can maintain adequate zinc homeostasis. Amniotic fluid zinc concentrations have also been reported to be decreased in pre-eclamptic women delivering preterm (3336 weeks gestation) in a small retrospective cross-sectional study from the US [59]. It must be noted that as with all these micronutrients, the concentrations early in pregnancy in relation to the development of pregnancy complications remains to be established.

Manganese Manganese is a free element in nature (often in combination with iron), furthermore manganese(II) ions function as cofactors for a number of enzymes; the element is thus a required trace mineral for all known living organisms. Manganese is also an important cofactor for a number of enzymes, including the antioxidant manganese superoxide dismutase (Mn-SOD; Figure 2) which may protect the placenta from oxidative stress by detoxifying superoxide anions [118]. Dietary manganese is the main source of exposure under normal circumstances; current requirements for manganese, although less studied compared to other micronutrients are shown in Table 1 and little is known about the effects of deficiency or excess of manganese on the developing human fetus or pregnancy outcome [119]. This is further hampered by the fact that at present sensitive biomarkers

15

346 347 348 349 350 351 352 353 354 355 356 357 358 359 360 361 362 363 364 365 366 367 368

of manganese exposure and nutritional status are not available other than some estimates from blood concentrations [119].

Circulating whole blood manganese concentrations have been shown to be lower in women with fetal growth restriction compared to healthy controls (mean SD: 16.7 4.8 vs. 19.1 5.9 g/L respectively) indicating that this micronutrient may be important in maintaining fetal growth [120]. This study also found that manganese concentrations were higher in umbilical samples from fetal growth restriction cases compared to controls suggesting that manganese contributes to different effects on birth weight in healthy mothers [120]. Zota et al retrospective study in the US reported a non- linear relationship between manganese concentrations and birth weights in a cohort of 470 full term (delivered at > 37 weeks gestation) infant further indicating the potential affect on fetal growth [121]. A recent small retrospective study of African-American mothers reported reduced umbilical cord whole blood manganese concentrations in neonates born to mothers with pre-eclampsia compared to controls (mean [95% CI]: 2.2 [1.5, 3.2] vs. 3.7 [3.2, 4.2] g/L respectively) [122]. Furthermore, this study found that like other micronutrients umbilical cord blood from smoking mothers had reduced manganese concentrations [122]. Than et al demonstrated increased fetal membrane MnSOD mRNA expression in women with pre-term labour [123]. Manganese is one of the least studied micronutrients and at present no supplementation trial has been published which may reflect the lack of data on manganese concentrations in pregnancy.

16

369 370 371 372 373 374 375 376 377 378 379 380 381 382 383 384 385 386 387 388 389 390 391

Vitamins C and E Vitamin C (ascorbic acid and dihydroascorbic acid) is an essential water-soluble vitamin found widely in fruit and vegetables; it has important roles in co llagen synthesis, wound healing, prevention of anaemia and as an antioxidant as it can quench a variety of reactive oxygen species and reactive nitrogen species in aqueous environments [124]. Vitamin C is commonly included in low doses (<200mg/ day) within multivitamin preparations for pregnancy but has also been given in higher doses (up to 1000mg/ day) as a supplement, alone or in combination with vitamin E [125]. Smoking has been shown to increase oxidative stress and metabolic turnover of vitamin C, thus the requirement for smokers is increased by 35 mg/day [126].

Vitamin E (-tocopherol) is a lipid-soluble vitamin acting with the lipid membrane and with synergistic interactions with vitamin C [127] (Figure 3). Vitamin E functions primarily as a chain-breaking antioxidant that prevents propagation of lipid peroxidation [125, 128]. A considerable interest exists regarding prevention of maternal and perinatal morbidity with vitamins C and E. However, the most recent meta-analysis of ten trials (6533 women) published in 2008 of antioxidant supplementation (including vitamin C and E but also other supplements such as lycopene) showed no difference in the relative risk (RR) of pre-eclampsia (RR 0.73, 95% CI 0.51 to 1.06), preterm birth (before 37 weeks) (RR 1.10, 95% CI 0.99 to 1.22), SGA infants (RR 0.83, 95% CI 0.62 to 1.11) or any baby death (RR 1.12, 95% CI 0.81 to 1.53) [7]. Considerable heterogeneity between the trials was seen reflecting the different supplements studied, the varying risk criteria used for entry into the studies and the study sizes. A couple of subsequent recent

17

392 393 394 395 396 397 398 399 400 401 402 403 404 405 406 407 408 409 410 411 412 413 414

multicentre double-blinded randomised trials of a combination of vitamin C and E [129, 130] also found supplementation did not reduce the rate of pre-eclampsia or gestational hypertension and like the Vitamins In Pre-eclampsia trial in 2006 [131] increased the risk of fetal loss or perinatal death and preterm prelabour rupture of membranes. Another recent multicentre placebo-controlled trial of vitamin C and E in women with type-1 diabetes in pregnancy (DAPIT) also reported no differences in the rates of pre-eclampsia between supplemented or placebo groups [132]. Further investigations are required as the concentrations of these vitamins remain significantly reduced in women with preeclampsia, but in the absence of further evidence, routine supplementation with higher dose vitamin C and E is not recommended as they can be potentially dangerous in high concentrations.

Adolescent pregnancies Another factor to highlight is that pregnancy in adolescent women is a topic of increasing health concern in many countries [133]. Pregnant adolescents are of a greater nutritional risk as they themselves are undergoing an intense process of growth and development [134]. It has been suggested that there is competition for nutrients between a pregnant adolescent and her fetus, as both are in critical stages of growth during the gestational period [65, 135]. As mentioned throughout above, micronutrient deficiencies have been associated with adolescent pregnancies [66, 84, 110]. The recent About Teenage Eating study showed that teenage pregnancy in the UK is associated with decreased consumption of micronutrients and that this is associated with an increased risk of SGA infants [65].

18

415 416 417 418 419 420 421 422 423 424 425 426 427 428 429 430 431 432 433 434 435 436 437

Low micronutrient intake poses a health risk to successful pregnancy in the UK, especially in adolescence. Socioeconomic deprivation in the UK population has been shown to be an independent risk factor for eating less fruit and vegetables and also for increased smoking, which is known to cause further depletion of micronutrients [136]. Although attempts are being made to persuade women of low socioeconomic status of the importance of a balanced diet, the central roles that fruit and vegetables have in this and that healthy food can be low cost and convenient this is often met with resistance [137].

The increase in pregnancies in mothers of older ages highlights another subgro up at increased risk of pregnancy complications. We are not aware of any studies at present examining this important group and future investigations are required especially as some of these micronutrient concentrations decline with increasing age.

Conclusions Increased knowledge about the importance of these specific antioxidant micronutrients and the crucial part that they have in maintaining successful pregnancy and determining both the long and short term health of both mother and baby needs to be addressed and made a key focus for future health strategies in improving pregnancy outcomes. This is particularly important with regards to pre-eclampsia and fetal growth restriction, where oxidative stress is an essential component to the aetiology of these conditions and so these specific antioxidant micronutrient deficiencies may play a contributing role. Only by fully understanding the requirements for micronutrients during pregnancy will we be able to evaluate the potential use of these dietary antioxidant supplements as a way of

19

438 439 440 441 442 443 444 445 446 447 448 449 450 451 452 453 454 455 456 457 458 459 460

preventing pathological pregnancy outcomes. However, it must also be remembered that these antioxidant and other micronutrients can be obtained via a healthy diet thereby negating the need for supplementation. Future strategies foc ussing on providing nutritional guidance specifically to pregnant women will be pivotal in helping to ensure optimal health of both mother and baby.

Key Messages 1) Further research is needed to accurately quantify levels of micronutrients in pregnancy and how levels vary over the course of pregnancy. 2) The actions of antioxidant micronutrients on maternal, fetal and placental health needs to further elucidated. 3) Prenatal guidance needs to be made clear to ensure that women and practioners are aware of the nutritional requirements during pregnancy, and how healthy diet can prevent diseases of pregnancy.

Conflict of Interest: None of the authors have any conflict of interest.

Funding: H.D. Mistry was funded by Tommys Charity (Charity No. 3266897) and P.J. Williams was supported by the Nottingham University Hosp itals Special Trustees Charity (RB17B3).

20

461 462 463 464 465 466 467 468 469 470 471 472 473 474 475 476 477 478 479 480 481 482 483

References [1] Allen LH. Multiple micronutrients in pregnancy and lactation: an overview. Am J

Clin Nutr 2005;81:1206S. [2] Broughton Pipkin F. Maternal Physiology. In: Edmonds DK, editor. Dewhurst's

textbook of Obstetrics and Gynaecology. Oxford: Blackwell Publishing, 2007. [3] Bader RA, Bader ME, Rose DF, Braunwald E. Hemodynamics at rest and during

exercise in normal pregnancy as studies by cardiac catheterization. J Clin Invest 1955;34:1524. [4] King JC. The risk of maternal nutritional depletion and poor outcomes increases

in early or closely spaced pregnancies. J Nutr 2003;133:1732S. [5] Srivastava S, Mehrotra PK, Srivastava SP, Siddiqui MK. Some essential elements

in maternal and cord blood in relation to birth weight and gestational age of the baby. Biol Trace Elem Res 2002;86:97. [6] Fall CH, Yajnik CS, Rao S, Davies AA, Brown N, Farrant HJ. Micronutrients and

fetal growth. J Nutr 2003;133:1747S. [7] Rumbold A, Duley L, Crowther CA, Haslam RR. Antioxidants for preventing pre-

eclampsia. Cochrane Database Syst Rev 2008:CD004227. [8] Bellamy L, Casas JP, Hingorani AD, Williams DJ. Pre-eclampsia and risk of

cardiovascular disease and cancer in later life: systematic review and meta-analysis. BMJ 2007;335:974. [9] Lykke JA, Langhoff-Roos J, Sibai BM, Funai EF, Triche EW, Paidas MJ.

Hypertensive pregnancy disorders and subsequent cardiovascular morbidity and type 2 diabetes mellitus in the mother. Hypertension 2009;53:944.

21

484 485 486 487 488 489 490 491 492 493 494 495 496 497 498 499 500 501 502 503 504 505 506

[10]

Vikse BE, Irgens LM, Leivestad T, Skjaerven R, Iversen BM. Preeclampsia and

the risk of end-stage renal disease. N Engl J Med 2008;359:800. [11] Staff AC, Dechend R, Pijnenborg R. Learning From the Placenta. Acute Atherosis

and Vascular Remodeling in Preeclampsia-Novel Aspects for Atherosclerosis and Future Cardiovascular Health. Hypertension 2010. [12] Armitage JA, Khan IY, Taylor PD, Nathanielsz PW, Poston L. Developmental

programming of the metabolic syndrome by maternal nutritional imbalance: how strong is the evidence from experimental models in mammals? J Physiol 2004;561:355. [13] Armitage JA, Taylor PD, Poston L. Experimental models of developmental

programming: consequences of exposure to an energy rich diet during development. J Physiol 2005;565:3. [14] Solomons NW. Developmental origins of health and disease: concepts, caveats,

and consequences for public health nutrition. Nutr Rev 2009;67 Suppl 1:S12. [15] Wells JC. The thrifty phenotype as an adaptive maternal effect. Biol Rev Camb

Philos Soc 2007;82:143. [16] Cetin I, Berti C, Calabrese S. Role of micronutrients in the periconceptional

period. Hum Reprod Update 2009. [17] Challis JR, Lockwood CJ, Myatt L, Norman JE, Strauss JF, 3rd, Petraglia F.

Inflammation and pregnancy. Reprod Sci 2009;16:206. [18] Hanson LA, Silfverdal SA. The mother's immune system is a balanced threat to

the foetus, turning to protection of the neonate. Acta Paediatr 2009;98:221. [19] Ornoy A. Embryonic oxidative stress as a mechanism of teratogenesis with

special emphasis on diabetic embryopathy. Reprod Toxicol 2007;24:31.

22

507 508 509 510 511 512 513 514 515 516 517 518 519 520 521 522 523 524 525 526 527 528

[20]

Jauniaux E, Watson AL, Hempstock J, Bao YP, Skepper JN, Burton GJ. Onset of

maternal arterial blood flow and placental oxidative stress. A possible factor in human early pregnancy failure. Am J Pathol 2000;157:2111. [21] Wang Y, Walsh SW. Antioxidant activities and mRNA expression of superoxide

dismutase, catalase, and glutathione peroxidase in normal and preeclamptic placentas. J Soc Gynecol Investig 1996;3:179. [22] Vanderlelie J, Venardos K, Clifton VL, Gude NM, Clarke FM, Perkins AV.

Increased biological oxidation and reduced anti-oxidant enzyme activity in pre-eclamptic placentae. Placenta 2005;26:53. [23] Mistry HD, Kurlak LO, Williams PJ, Ramsay MM, Symonds ME, Broughton

Pipkin F. Differential expression and distribution of placental glutathione peroxidases 1, 3 and 4 in normal and preeclamptic pregnancy. Placenta 2010;31:401. [24] Rayman MP. Selenoproteins and human health: Insights from epidemiological

data. Biochim Biophys Acta 2009. [25] Redman CW, Sargent IL. Placental debris, oxidative stress and pre-eclampsia.

Placenta 2000;21:597. [26] Burton GJ, Jauniaux E. Placental oxidative stress: from miscarriage to

preeclampsia. J Soc Gynecol Investig 2004;11:342. [27] Brosens I, Dixon HG, Robertson WB. Fetal growth retardation and the arteries of

the placental bed. Br J Obstet Gynaecol 1977;84:656. [28] Brosens IA, Robertson WB, Dixon HG. The role of the spiral arteries in the

pathogenesis of preeclampsia. Obstet Gynecol Annu 1972;1:177.

23

529 530 531 532 533 534 535 536 537 538 539 540 541 542 543 544 545 546 547 548 549 550 551

[29]

Gupta S, Agarwal A, Banerjee J, Alvarez JG. The role of oxidative stress in

spontaneous abortion and recurrent pregnancy loss: a systematic review. Obstet Gynecol Surv 2007;62:335. [30] Perkins AV. Endogenous anti-oxidants in pregnancy and preeclampsia. Aust N Z

J Obstet Gynaecol 2006;46:77. [31] Nakamura M, Sekizawa A, Purwosunu Y, Okazaki S, Farina A, Wibowo N,

Shimizu H, Okai T. Cellular mRNA expressions of anti-oxidant factors in the blood of preeclamptic women. Prenat Diagn 2009;29:691. [32] Mistry HD, Wilson V, Ramsay MM, Symonds ME, Broughton Pipkin F. Reduced

selenium concentrations and glutathione peroxidase activity in pre-eclamptic pregnancies. Hypertension 2008;52:881. [33] Myatt L, Cui X. Oxidative stress in the placenta. Histochem Cell Biol

2004;122:369. [34] Saugstad OD. Update on oxygen radical disease in neonatology. Curr Opin Obstet

Gynecol 2001;13:147. [35] 2010. [36] [37] [38] Sibai B, Dekker G, Kupferminc M. Pre-eclampsia. Lancet 2005;365:785. Broughton Pipkin F. Risk factors for preeclampsia. N Engl J Med 2001;344:925. Roberts JM, Lain KY. Recent Insights into the pathogenesis of pre-eclampsia. Steegers EA, von Dadelszen P, Duvekot JJ, Pijnenborg R. Pre-eclampsia. Lancet

Placenta 2002;23:359. [39] Poston L. The Role of Oxidative Stress. In: Critchley H, MacLean A, Poston L,

Walker J, editors. Pre-eclampsia. London: RCOG Press, 2004.

24

552 553 554 555 556 557 558 559 560 561 562 563 564 565 566 567 568 569 570 571 572 573 574

[40]

Redman CW, Sargent IL. Pre-eclampsia, the placenta and the maternal systemic

inflammatory response--a review. Placenta 2003;24 Suppl A:S21. [41] Hung TH, Burton GJ. Hypoxia and reoxygenation: a possible mechanism for

placental oxidative stress in preeclampsia. Taiwan J Obstet Gynecol 2006;45:189. [42] Cetin I, Foidart JM, Miozzo M, Raun T, Jansson T, Tsatsaris V, Reik W, Cross J,

Hauguel-de-Mouzon S, Illsley N, Kingdom J, Huppertz B. Fetal growth restriction: a workshop report. Placenta 2004;25:753. [43] Biri A, Bozkurt N, Turp A, Kavutcu M, Himmetoglu O, Durak I. Role of

oxidative stress in intrauterine growth restriction. Gynecol Obstet Invest 2007;64:187. [44] Takagi Y, Nikaido T, Toki T, Kita N, Kanai M, Ashida T, Ohira S, Konishi I.

Levels of oxidative stress and redox-related molecules in the placenta in preeclampsia and fetal growth restriction. Virchows Arch 2004;444:49. [45] [46] Rayman MP. The importance of selenium to human health. Lancet 2000;356:233. Beckett GJ, Arthur JR. Selenium and endocrine systems. J Endocrinol

2005;184:455. [47] Zachara BA, Wardak C, Didkowski W, Maciag A, Marchaluk E. Changes in

blood selenium and glutathione concentrations and glutathione peroxidase activity in human pregnancy. Gynecol Obstet Invest 1993;35:12. [48] Mihailovic M, Cvetkovic M, Ljubic A, Kosanovic M, Nedeljkovic S, Jovanovic I,

Pesut O. Selenium and malondialdehyde content and glutathione peroxidase activity in maternal and umbilical cord blood and amniotic fluid. Biol Trace Elem Res 2000;73:47. [49] Thomson CD. Assessment of requirements for selenium and adequacy of

selenium status: a review. Eur J Clin Nutr 2004;58:391.

25

575 576 577 578 579 580 581 582 583 584 585 586 587 588 589 590 591 592 593 594 595 596

[50]

Rayman MP. Food-chain selenium and human health: emphasis on intake. Br J

Nutr 2008;100:254. [51] Gathwala G, Yadav OP, Singh I, Sangwan K. Maternal and cord plasma selenium

levels in full- term neonates. Indian J Pediatr 2000;67:729. [52] Shennan DB. A study of selenate efflux from human placental microvillus

membrane vesicles. Biosci Rep 1987;7:675. [53] Shennan DB. Selenium (selenate) transport by human placental brush border

membrane vesicles. Br J Nutr 1988;59:13. [54] Barrington JW, Taylor M, Smith S, Bowen-Simpkins P. Selenium and recurrent

miscarriage. J Obstet Gynaecol 1997;17:199. [55] Kocak I, Aksoy E, Ustun C. Recurrent spontaneous abortion and selenium

deficiency. Int J Gynaecol Obstet 1999;65:79. [56] Zachara BA, Dobrzynski W, Trafikowska U, Szymansk i W. Blood selenium and

glutathione peroxidases in miscarriage. BJOG 2001;108:244. [57] Rayman MP, Bode P, Redman CW. Low selenium status is associated with the

occurrence of the pregnancy disease preeclampsia in women from the United Kingdom. Am J Obstet Gynecol 2003;189:1343. [58] Maleki A, Fard MK, Zadeh DH, Mamegani MA, Abasaizadeh S, Mazloomzadeh

S. The Relationship between Plasma Level of Se and Preeclampsia. Hypertens Pregnancy 2010. [59] Dawson EB, Evans DR, Nosovitch J. Third-trimester amniotic fluid metal levels

associated with preeclampsia. Arch Environ Health 1999;54:412.

26

597 598 599 600 601 602 603 604 605 606 607 608 609 610 611 612 613 614 615 616 617

[60]

Klapec T, Cavar S, Kasac Z, Rucevic S, Popinjac A. Selenium in placenta

predicts birth weight in normal but not intrauterine growth restriction pregnancy. J Trace Elem Med Biol 2008;22:54. [61] Osada H, Watanabe Y, Nishimura Y, Yukawa M, Seki K, Sekiya S. Profile of

trace element concentrations in the feto-placental unit in relation to fetal growth. Acta Obstet Gynecol Scand 2002;81:931. [62] Zadrozna M, Gawlik M, Nowak B, Marcinek A, Mrowiec H, Walas S, Wietecha-

Posluszny R, Zagrodzki P. Antioxidants activities and concentration of selenium, zinc and copper in preterm and IUGR human placentas. J Trace Elem Med Biol 2009;23:144. [63] Llanos MN, Ronco AM. Fetal growth restriction is related to placental levels of

cadmium, lead and arsenic but not with antioxidant activities. Reprod Toxicol 2009;27:88. [64] Strambi M, Longini M, Vezzosi P, Berni S, Buoni S. Selenium status, birth

weight, and breast- feeding: pattern in the first month. Biol Trace Elem Res 2004;99:71. [65] Baker PN, Wheeler SJ, Sanders TA, Thomas JE, Hutchinson CJ, Clarke K, Berry

JL, Jones RL, Seed PT, Poston L. A prospective study of micronutrient status in adolescent pregnancy. Am J Clin Nutr 2009;89:1114. [66] Mistry HD, Kurlak LO, Young SY, Briley AL, Broughton Pipkin F, Poston L.

Reduced plasma selenium in adolescent mothers with small for gestational age infants; impact of smoking and ethnicity. Society for Gynecologic Investigation. Orlando: Reproductive Sciences, 2010.

27

618 619 620 621 622 623 624 625 626 627 628 629 630 631 632 633 634 635 636 637 638

[67]

Negro R, Greco G, Mangieri T, Pezzarossa A, Dazzi D, Hassan H. The influence

of selenium supplementation on postpartum thyroid status in pregnant women with thyroid peroxidase autoantibodies. J Clin Endocrinol Metab 2007;92:1263. [68] Han L, Zhou SM. Selenium supplement in the prevention of pregnancy induced

hypertension. Chin Med J (Engl) 1994;107:870. [69] Rumiris D, Purwosunu Y, Wibowo N, Farina A, Sekizawa A. Lower rate of

preeclampsia after antioxidant supplementation in pregnant wome n with low antioxidant status. Hypertens Pregnancy 2006;25:241. [70] Tara F, Rayman MP, Boskabadi H, Ghayour-Mobarhan M, Sahebkar A, Yazarlu

O, Ouladan S, Tavallaie S, Azimi-Nezhad M, Shakeri MT, Teymoori MS, Razavi BS, Oladi M, Ferns G. Selenium supplementation and premature (pre- labour) rupture of membranes: a randomised double-blind placebo-controlled trial. J Obstet Gynaecol 2010;30:30. [71] Gambling L, Andersen HS, McArdle HJ. Iron and copper, and their interactions

during development. Biochem Soc Trans 2008;36:1258. [72] Izquierdo Alvarez S, Castanon SG, Ruata ML, Aragues EF, Terraz PB, Irazabal

YG, Gonzalez EG, Rodriguez BG. Updating of normal levels of copper, zinc and selenium in serum of pregnant women. J Trace Elem Med Biol 2007;21 Suppl 1:49. [73] Huang HM, Leung PL, Sun DZ, Zhu MG. Hair and serum calcium, iron, copper,

and zinc levels during normal pregnancy at three trimesters. Biol Trace Elem Res 1999;69:111.

28

639 640 641 642 643 644 645 646 647 648 649 650 651 652 653 654 655 656 657 658 659 660 661

[74]

Liu J, Yang H, Shi H, Shen C, Zhou W, Dai Q, Jiang Y. Blood copper, zinc,

calcium, and magnesium levels during different duration of pregnancy in Chinese. Biol Trace Elem Res 2010;135:31. [75] Fattah MM, Ibrahim FK, Ramadan MA, Sammour MB. Ceruloplasmin and

copper level in maternal and cord blood and in the placenta in normal pregnancy and in pre-eclampsia. Acta Obstet Gynecol Scand 1976;55:383. [76] Shakour-Shahabi L, Abbasali- Zadeh S, Rashtchi-Zadeh N. Serum level and

antioxidant activity of ceruloplasmin in preeclampsia. Pakistan Journal of biological Sciences 2010;13:321. [77] Institute of Medcine. Dietary Reference Intakes for Vitamin A, Vitamin K,

Arsenic, Boron, Chromium, Copper, Iodine, Iron, Manganese, Molybdenum, Nickel, Silicon, Vanadium, and Zinc.: National Academy Press, 2001. [78] McArdle HJ, Andersen HS, Jones H, Gambling L. Copper and iron transport

across the placenta: regulation and interactions. J Neuroendocrinol 2008;20:427. [79] Kambe T, Weaver BP, Andrews GK. The genetics of essential metal homeostasis

during development. Genesis 2008;46:spcone. [80] Keen CL, Hanna LA, Lanoue L, Uriu-Adams JY, Rucker RB, Clegg MS.

Developmental consequences of trace mineral deficiencies in rodents: acute and longterm effects. J Nutr 2003;133:1477S. [81] Prohaska JR, Brokate B. The timing of perinatal copper deficiency in mice

influences offspring survival. J Nutr 2002;132:3142. [82] Masters DG, Keen CL, Lonnerdal B, Hurley LS. Comparative aspects of dietary

copper and zinc deficiencies in pregnant rats. J Nutr 1983;113:1448.

29

662 663 664 665 666 667 668 669 670 671 672 673 674 675 676 677 678 679 680 681 682

[83]

Ali Akbar S, Nicolaides KH, Brown PR. Measurement of Cu/Zn SOD in placenta,

cultured cells, various fetal tissues, decidua and semen by ELISA. J Obstet Gynaecol 1998;18:331. [84] de Moraes ML, de Faria Barbosa R, Santo RE, da Silva Santos F, de Jesus EF,

Sardinha FL, Tavares do Carmo MD. Maternal-Fetal Distribution of Calcium, Iron, Copper, and Zinc in Pregnant Teenagers and Adults. Biol Trace Elem Res 2010. [85] McArdle HJ, Ashworth CJ. Micronutrients in fetal growth and development. Br

Med Bull 1999;55:499. [86] Krachler M, Rossipal E, Micetic-Turk D. Trace element transfer from the mother

to the newborn--investigations on triplets of colostrum, maternal and umbilical cord sera. Eur J Clin Nutr 1999;53:486. [87] Rossipal E, Krachler M, Li F, Micetic-Turk D. Investigation of the transport of

trace elements across barriers in humans: studies of placental and mammary transfer. Acta Paediatr 2000;89:1190. [88] Ozdemir Y, Borekci B, Levet A, Kurudirek M. Assessment of trace element

concentration distribution in human placenta by wavelength dispersive X-ray fluorescence: effect of neonate weight and maternal age. Appl Radiat Isot 2009;67:1790. [89] Brophy MH, Harris NF, Crawford IL. Elevated copper and lowered zinc in the

placentae of pre-eclamptics. Clin Chim Acta 1985;145:107. [90] Serdar Z, Gur E, Develioglu O. Serum iron and copper status and oxidative stress

in severe and mild preeclampsia. Cell Biochem Funct 2006;24:209.

30

683 684 685 686 687 688 689 690 691 692 693 694 695 696 697 698 699 700 701 702 703

[91]

Kolusari A, Kurdoglu M, Yildizhan R, Adali E, Edirne T, Cebi A, Demir H,

Yoruk IH. Catalase activity, serum trace element and heavy metal concentrations, and vitamin A, D and E levels in pre-eclampsia. J Int Med Res 2008;36:1335. [92] Aksoy H, Taysi S, Altinkaynak K, Bakan E, Bakan N, Kumtepe Y. Antioxidant

potential and transferrin, ceruloplasmin, and lipid peroxidation levels in women with preeclampsia. J Investig Med 2003;51:284. [93] Vitoratos N, Salamalekis E, Dalamaga N, Kassanos D, Creatsas G. Defective

antioxidant mechanisms via changes in serum ceruloplasmin and total iron binding capacity of serum in women with pre-eclampsia. Eur J Obstet Gynecol Reprod Biol 1999;84:63. [94] Arredondo M, Nunez MT. Iron and copper metabolism. Mol Aspects Med

2005;26:313. [95] WHO. The World Health Report 2002: Reducing risks, promoting healthy life.

Geneva: World Health Organisation, 2002. [96] Uriu-Adams JY, Keen CL. Zinc and reproduction: effects of zinc deficiency on

prenatal and early postnatal development. Birth Defects Res B Dev Reprod Toxicol 2010;89:313. [97] Swanson CA, King JC. Zinc and pregnancy outcome. Am J Clin Nutr

1987;46:763. [98] WHO/FAO/IAEA. Trace Elements in Human Nutrition and Health. Geneva:

World Health Organisation, 1996.

31

704 705 706 707 708 709 710 711 712 713 714 715 716 717 718 719 720 721 722 723 724 725

[99]

Ilhan N, Ilhan N, Simsek M. The changes of trace elements, malondialdehyde

levels and superoxide dismutase activities in pregnancy with or without preeclampsia. Clin Biochem 2002;35:393. [100] Groenen PM, Roes EM, Peer PG, Merkus HM, Steegers EA, Steegers-Theunissen RP. Myo- inositol, glucose and zinc concentrations determined in the preconceptional period, during and after pregnancy. Eur J Obstet Gynecol Reprod Biol 2006;127:50. [101] Harley K, Eskenazi B, Block G. The association of time in the US and diet during pregnancy in low- income women of Mexican descent. Paediatr Perinat Epidemiol 2005;19:125. [102] King JC. Determinants of maternal zinc status during pregnancy. Am J Clin Nutr 2000;71:1334S. [103] Goldenberg RL, Tamura T, Neggers Y, Copper RL, Johnston KE, DuBard MB, Hauth JC. The effect of zinc supplementation on pregnancy outcome. JAMA 1995;274:463. [104] Black RE. Micronutrients in pregnancy. Br J Nutr 2001;85 Suppl 2:S193. [105] Garg HK, Singhal KC, Arshad Z. A study of the effect of oral zinc supplementation during pregnancy on pregnancy outcome. Indian J Physiol Pharmacol 1993;37:276. [106] Caulfield LE, Zavaleta N, Figueroa A, Leon Z. Maternal zinc supplementation does not affect size at birth or pregnancy duration in Peru. J Nutr 1999;129:1563. [107] Osendarp SJ, van Raaij JM, Arifeen SE, Wahed M, Baqui AH, Fuchs GJ. A randomized, placebo-controlled trial of the effect of zinc supplementation during

32

726 727 728 729 730 731 732 733 734 735 736 737 738 739 740 741 742 743 744 745 746 747 748

pregnancy on pregnancy outcome in Bangladeshi urban poor. Am J Clin Nutr 2000;71:114. [108] Jonsson B, Hauge B, Larsen MF, Hald F. Zinc supplementation during pregnancy: a double blind randomised controlled trial. Acta Obstet Gynecol Scand 1996;75:725. [109] Kiilholma P, Paul R, Pakarinen P, Gronroos M. Copper and zinc in pre-eclampsia. Acta Obstet Gynecol Scand 1984;63:629. [110] Cherry FF, Bennett EA, Bazzano GS, Johnson LK, Fosmire GJ, Batson HK. Plasma zinc in hypertension/toxemia and other reproductive variables in adolescent pregnancy. Am J Clin Nutr 1981;34:2367. [111] Diaz E, Halhali A, Luna C, Diaz L, Avila E, Larrea F. Newborn birth weight correlates with placental zinc, umbilical insulin- like growth factor I, and leptin levels in preeclampsia. Arch Med Res 2002;33:40. [112] Acikgoz S, Harma M, Mungan G, Can M, Demirtas S. Comparison of angiotensin-converting enzyme, malonaldehyde, zinc, and copper levels in preeclampsia. Biol Trace Elem Res 2006;113:1. [113] Kumru S, Aydin S, Simsek M, Sahin K, Yaman M, Ay G. Comparison of serum copper, zinc, calcium, and magnesium levels in preeclamptic and healthy pregnant women. Biol Trace Elem Res 2003;94:105. [114] Yousef MI, El-Hendy HA, El-Demerdash FM, Elagamy EI. Dietary zinc deficiency induced-changes in the activity of enzymes and the levels of free radicals, lipids and protein electrophoretic behavior in growing rats. Toxicology 2002;175:223. [115] Jain S, Sharma P, Kulshreshtha S, Mohan G, Singh S. The role of calcium, magnesium, and zinc in pre-eclampsia. Biol Trace Elem Res 2010;133:162.

33

749 750 751 752 753 754 755 756 757 758 759 760 761 762 763 764 765 766 767 768 769 770 771

[116] Bassiouni BA, Foda AI, Rafei AA. Maternal and fetal plasma zinc in preeclampsia. Eur J Obstet Gynecol Reprod Biol 1979;9:75. [117] Kiilholma P, Gronroos M, Erkkola R, Pakarinen P, Nanto V. The role of calcium, copper, iron and zinc in preterm delivery and premature rupture of fetal membranes. Gynecol Obstet Invest 1984;17:194. [118] Ademuyiwa O, Odusoga OL, Adebawo OO, Ugbaja RN. Endogenous antioxidant defences in plasma and erythrocytes of pregnant women during different trimesters of pregnancy. Acta Obstet Gynecol Scand 2007:1. [119] Wood RJ. Manganese and birth outcome. Nutr Rev 2009;67:416. [120] Vigeh M, Yokoyama K, Ramezanzadeh F, Dahaghin M, Fakhriazad E, Seyedaghamiri Z, Araki S. Blood manganese concentrations and intrauterine growth restriction. Reprod Toxicol 2008;25:219. [121] Zota AR, Ettinger AS, Bouchard M, Amarasiriwardena CJ, Schwartz J, Hu H, Wright RO. Maternal blood manganese levels and infant birth weight. Epidemiology 2009;20:367. [122] Jones EA, Wright JM, Rice G, Buckley BT, Magsumbol MS, Barr DB, Williams BL. Metal exposures in an inner-city neonatal population. Environ Int 2010;36:649. [123] Than NG, Romero R, Tarca AL, Draghici S, Erez O, Chaiworapongsa T, Kim YM, Kim SK, Vaisbuch E, Tromp G. Mitochondrial manganese superoxide dismutase mRNA expression in human chorioamniotic membranes and its association with labor, inflammation, and infection. J Matern Fetal Neonatal Med 2009;22:1000. [124] Buettner GR. The pecking order of free radicals and antioxidants: lipid peroxidation, alpha-tocopherol, and ascorbate. Arch Biochem Biophys 1993;300:535.

34

772 773 774 775 776 777 778 779 780 781 782 783 784 785 786 787 788 789 790 791 792 793

[125] Poston L, Chappell LC, Seed P, Shennan A. Biomarkers for oxidative stress in pre-eclampsia. Pregnancy Hypertension 2011;1:22. [126] Institute of Medicine. Dietary Reference Intakes for Vitamin C, Vitamin E, Selenium, and Carotenoids. National Academy Press, 2000. p.284. [127] Packer JE, Slater TF, Willson RL. Direct observation of a free radical interaction between vitamin E and vitamin C. Nature 1979;278:737. [128] Burton GW, Foster DO, Perly B, Slater TF, Smith IC, Ingold KU. Biological antioxidants. Philos Trans R Soc Lond B Biol Sci 1985;311:565. [129] Xu H, Perez-Cuevas R, Xiong X, Reyes H, Roy C, Julien P, Smith G, von Dadelszen P, Leduc L, Audibert F, Moutquin JM, Piedboeuf B, Shatenstein B, ParraCabrera S, Choquette P, Winsor S, Wood S, Benjamin A, Walker M, Helewa M, Dube J, Tawagi G, Seaward G, Ohlsson A, Magee LA, Olatunbosun F, Gratton R, Shear R, Demianczuk N, Collet JP, Wei S, Fraser WD. An international trial of antioxidants in the prevention of preeclampsia (INTAPP). Am J Obstet Gynecol 2010;202:239 e1. [130] Roberts JM, Myatt L, Spong CY, Thom EA, Hauth JC, Leveno KJ, Pearson GD, Wapner RJ, Varner MW, Thorp JM, Jr., Mercer BM, Peaceman AM, Ramin SM, Carpenter MW, Samuels P, Sciscione A, Harper M, Smith WJ, Saade G, Sorokin Y, Anderson GB. Vitamins C and E to prevent complications of pregnancy-associated hypertension. N Engl J Med 2010;362:1282. [131] Poston L, Briley AL, Seed PT, Kelly FJ, Shennan AH. Vitamin C and vitamin E in pregnant women at risk for pre-eclampsia (VIP trial): randomised placebo-controlled trial. Lancet 2006;367:1145.

35

794 795 796 797 798 799 800 801 802 803 804 805 806 807 808 809 810 811 812 813 814 815 816

[132] McCance DR, Holmes VA, Maresh MJ, Patterson CC, Walker JD, Pearson DW, Young IS. Vitamins C and E for prevention of pre-eclampsia in women with type 1 diabetes (DAPIT): a randomised placebo-controlled trial. Lancet 2010;376:259. [133] Maia PA, Figueiredo RC, Anastacio AS, Porto da Silveira CL, Donangelo CM. Zinc and copper metabolism in pregnancy and lactation of adolescent women. Nutrition 2007;23:248. [134] American Dietetic Association. Position of the American Dietetic Association: nutrition management of adolescent pregnancy. J Am Diet Assoc 1989;89:104. [135] Naeye RL. Teenaged and pre-teenaged pregnancies: consequences of the fetalmaternal competition for nutrients. Pediatrics 1981;67:146. [136] Amuzu A, Carson C, Watt HC, Lawlor DA, Ebrahim S. Influence of area and individual lifecourse deprivation on health behaviours: findings from the British Women's Heart and Health Study. Eur J Cardiovasc Prev Rehabil 2009;16:169. [137] Hampson SE, Martin J, Jorgensen J, Barker M. A social marketing approach to improving the nutrition of low- income women and children: an initial focus group study. Public Health Nutr 2009;12:1563. [138] Department of Health. Dietary reference values for foodenergy and nutrients for the United Kingdom. Report on Social Subjects no. 41. London, 1991.

36

817 818 819 820 821 822

Table 1: Requirement of micronutrient intakes for selenium, copper, zinc, manganese, vitamin C and vitamin E. RDA: Recommended Dietary Allowance; RNI: Reference Nutrient Intakes and NR: Normative Requirement Estimate. Values taken from 1 Institute of Medicine [77, 126], 2 Department of Health [138] and
3

WHO/FAO/IAEA [98]

Selenium (g/ d) RDA1 Female Adul t Pregnancy Upper limit 400 55 60

Copper (g/ d)

Zinc (mg/d)

Manganese (mg/d)

Vitami n C (mg/d)

Vitami n E (g/ d)

900 1,000

8 11

1.8 2

75 85

15 15

10,000

40

2,000

1,000

Female RNI2 Adul t Pregnancy 60 75 1,200 1,500 7 7 1.4 Female NR3 Adul t Pregnancy 30 1,350 1,150 1 2 40 50 -

823 824 825 826 827

37

828 829 830 831 832 833 834 835 836 837 838 839 840 841 842 843 844 845 846 847 848 849 850

Figure Legends

Figure 1: Diagram of the maternal fetal interface in early pregnancy. Extravillous cytotrophoblast cells migrate away from the cell column of the anchoring villus and invade through the maternal deciduas and inner third of the myometrium in o rder to gain access to maternal blood supply via maternal spiral arteries. Transformation of the maternal spiral arteries occurs as endovascular trophoblast help convert the endothelial cells lining the arteries into an amorphous fibrinoid matrix which is unresponsive to vasoactive stimuli, and serves to enlarge the vascular lumen maximising blood supply to the intervillous space.

Figure 2: Major pathways of reactive oxygen species generation and metabolism. Superoxide can be generated by specialised enzymes, such as the xanthine or NADPH oxidases, or as a byproduct of cellular metabolism, particularly the mitochondrial electron transport chain. Superoxide dismutase (SOD), both Cu/Zn and Mn SOD then converts the superoxide to hydrogen peroxide (H2 O2 ) which has to be rapidly removed from the system. This is generally achieved by catalase or peroxidases, such as the selenium dependent glutathione peroxidases (GPxs) which use reduced glutathione (GSH) as the electron donor.

Figure 3: Synergistic mechanisms of vitamin C (ascorbate) and vitamin E (-tocopherol) to prevent lipid peroxidation. O 2 . oxygen free radical.

38

851 852 853

Figure 1.

854 855 856 857 858 859 860 861 862 863 864 865

39

866 867 868 869 870 871 872 873 874 875 876 877 878 879 880 881 882 883 884 885 886 887 888 889 890 891 892 893 894 895 896 897 898 899 900 901 902

Figure 2.

O2
NAD(P)H Xanthine oxidase NADPH oxidase M itochondrial Electron Transport Chain NAD(P)+

O2 -.
H+

Cu/Zn & Mn SOD Se GPxs H2 O 2GSH GSSG

Catalase

H2 O2

H2 O

Glutathione reductase

40

903 904

Figure 3.

Hydrophilic Ascorbate

Lipophilic -tocopherol

O2

Ascorbate

-tocopherol Plas ma memb rane protein

Lipid pero xide

905 906

41

You might also like