Chapter 1

Prenatal Nutrition and Nutrition in Pregnancy: Effects on Long-Term Growth and Development

A. Imdad*
Z. Lassi**
R. Salaam
Z.A. Bhutta,§
*    Department of Pediatrics, Division of Pediatric Gastroenterology, Vanderbilt University Medical Center, Nashville, TN, United States
**    Robinson Research Institute, The University of Adelaide, Adelaide, SA, Australia
    Division of Women and Child Health, Aga Khan University, Karachi, Pakistan
    Centre for Global Child Health, The Hospital for Sick Children, Toronto, Canada
§    Center of Excellence in Women and Child Health, Aga Khan University, Karachi, Pakistan

Abstract

Optimal nutritional status of the mothers is the most influential nongenetic factor for the healthy development of the fetus and well-being of mothers. Maternal malnutrition, both undernutrition and overnutrition (obesity), may lead to adverse pregnancy outcomes and can have substantial negative effects on growth and development during childhood and excess risk of developing chronic diseases during adult life. In settings of chronic malnutrition, the adverse effects can be transmitted across generations with cumulative negative and intergenerational effects. Malnourished mothers, especially adolescent girls and also multigravidas tend to give birth to low birth weight (LBW) babies (including preterm and small for gestational age babies) with excess risk of stunted growth. Such stunted children grow up to be stunted individuals entering the reproductive period with suboptimal nutritional status and repetition of these risks and adverse outcomes across the life cycle. Observational studies from animal and humans make a strong case of intergenerational effects of maternal malnutrition; however the number of intervention studies with long-term follow is small and few robust conclusions can be drawn at this stage. Further intervention studies or cohorts with key health and nutrition interventions and long-term follow-up are needed to understand the relationships of maternal nutrition and risks of adverse medium to long-term outcomes and risks of chronic disease during childhood and adulthood.

Keywords

prenatal nutrition
intergenerational effect
preconception
folic acid
iodine supplementation
multiple micronutrient supplementation
balanced protein energy supplementation

Introduction

Optimal nutritional status of the mothers is the most influential nongenetic factor for the healthy development of the fetus and well-being of mothers (Bhutta et al., 2013Black et al., 2013a). Maternal malnutrition, both undernutrition and overnutrition (obesity), may lead to adverse pregnancy outcomes (Black et al., 2008Johansson et al., 2014Nohr et al., 2012) and can have substantial negative effects on growth and development during childhood and excess risk of developing chronic diseases during adult life (Villar et al., 2003Victora et al., 2008O’Reilly and Reynolds, 2013). In settings of chronic malnutrition, the adverse effects can be transmitted across generations with cumulative negative and intergenerational effects (Martorell and Zongrone, 2012). Malnourished mothers, especially adolescent girls and also multigravidas tend to give birth to low birth weight (LBW) babies [including preterm and small for gestational age babies (SGA)] with excess risk of stunted growth (Martorell and Zongrone, 2012). Such stunted children grow up to be stunted individuals entering the reproductive period with suboptimal nutritional status and repetition of these risks and adverse outcomes across the life cycle. Fig. 1.1 shows a conceptual framework of relationship of nutrition during pregnancy, infancy, childhood, adolescence, and adulthood.
image
Figure 1.1 Conceptual framework: intergenerational relationship of maternal and child nutrition. (Modified from Martorell, R., Zongrone, A., 2012. Intergenerational influences on child growth and undernutrition. Paediatr. Perinat. Epidemiol. 26 (Suppl. 1), 302–314.)
This chapter will focus on the effect of nutrition before conception (preconception) and during pregnancy and its effects on long-term growth and development. We will first describe the epidemiology of maternal malnutrition, then evidence for effect of maternal nutrition on long-term effects for offspring from observational and experimental studies in animals and humans. In this chapter, the terms “long-term” is used interchangeably with “intergenerational effects” and mainly describes adverse outcomes associated with maternal malnutrition for offspring of index pregnancy or beyond and appear during childhood or adulthood.

Epidemiology

Globally about 12% women are underweight with highest prevalence in Asia and Africa (Black et al., 2013b). On the other hand, prevalence of overweight (BMI ≥25 kg/m) and obesity (BMI ≥30 kg/m) has been rising with rates as high as 70% in the Americas and the Caribbean and about 40% in Africa in year 2008. Currently adolescence makes upto 19% of total population in developing countries and about 12% of industrialized nations. About half of the adolescence females living in low-middle income countries were stunted (height for age Z score < −2) and a major proportion was anemic. Teenage pregnancy and stunting, both increase risk of adverse pregnancy outcomes (Black et al., 2013b).

Evidence from animal studies

Studies in animals show strong evidence that maternal undernutrition has intergenerational effects (Drake and Liu, 2010). Price and coworkers studied rhesus monkeys and observed that descended from large-for-date matrilines exhibited enhanced fetal growth, and that of small-for-date birth weight, exhibited restricted growth (mainly for daughters). These associations were observed across five generations (Price and Coe, 2000Price et al., 1999). Stewart and coworkers conducted experiments in rats with protein-restricted diet and studied effects across twelve generations and compared them to controls. There was inhibitory effect on birth size and adult sizes as well as organ size and this recessive effect increased in every generation (Stewart et al., 1975). Further experiments from the same investigators showed that it took three generation to reverse the effects of maternal malnutrition in terms of size, and still there was some observable effects with regards to lack of learning and behavior (Stewart et al., 1980). Other studies had replicated these observations (Cesani et al., 2014Pucciarelli et al., 2002Warren and Bedi, 1985Zamenhof and van Marthens, 1982). One study in rats assessed effect of maternal malnutrition in index pregnancy and dietary rehabilitation for offspring and studied if this was reversible during early childhood. Results showed that malnourished mother gave birth to small size babies, however, when offspring were fed adequate diet, final size was same for experimental and controls groups (Rogers et al., 2003). In summary, animal studies showed that maternal undernutrition can have intergenerational effects on growth and development and these effects can be reversed with adequate nutritional rehabilitations of mothers and offspring.
Maternal overnutrition has also been studied in several experimental animal studies (Zambrano and Nathanielsz, 2013), including those involving rodents (Gupta et al., 2009Kirk et al., 2009Samuelsson et al., 2008), sheep (Yan et al., 2010), and nonhuman primates (McCurdy et al., 2009). Results from these studies showed that maternal obesity can lead to several adverse effects on offspring’s metabolism and may include increased adult body weight and fat mass, reduced insulin sensitivity, increased blood glucose and triglycerides levels, increased lipid deposition and defects in fatty acids metabolism in adult liver, as well as increased leptin levels, and hypothalamic alterations of appetite-regulating neuropeptides (Zambrano and Nathanielsz, 2013). Most of these studies were observatory and further studies are needed to establish exact mechanisms by which maternal obesity adversely affects the offspring.

The human literature

Evidence From Observational Studies

Effect of Maternal Undernutrition: Evidence From Famine Studies

Experimental studies in humans to assess the effect of restricted diet (undernutrition) to mother are challenging due to ethical considerations. Famine studies are quasiexperimental studies that have assessed affect of maternal malnutrition on offspring.
There were four famine periods in recent past where data were available to assess differential effect of maternal malnutrition. Three of these occur during the Second World War in certain areas of Netherlands (Smith, 1947a,b; Lumey and Stein, 1997), Germany (Dean, 1946) and Leningrad (Antonov, 1947Koupil et al., 2007) and fourth one in China (Huang et al., 2010bSong, 2013). The famine during World War II were of relatively short duration causing acute malnutrition in otherwise healthy populations while in China famine was prolonged (3 years) and occurred in a population with high baseline prevalence of chronic malnutrition. There were about 30–50 million deaths during famine in China (Huang et al., 2010b).
During Dutch famine, there was an acute shortage of food and official rations fell as low as 590 calories a day; resulting in maternal weight loss of as much as 2.5 kg from prefamine levels. This resulted in increased incidence of LBW and inflated perinatal mortality. Interesting to note was that the greatest effects on birth weight were among infants conceived before the onset of famine, but delivered during, the famine (Smith, 1947a,b). Maternal undernutrition had the highest impact during second and third trimester of pregnancy, however the infants who were conceived during famine and whose mothers were exposed through the second trimester but then received adequate nutrition during the third trimester, had normal size at birth. Intrauterine exposure was thought to be associated with increased risk of development of hypertension, obesity, and diabetes mellitus in middle age in offspring (Koupil et al., 2007Huang et al., 2010aStein et al., 2006Hult et al., 2010Ross and Desai, 2005). Similar observations of adverse effects of acute maternal malnutrition were reported from Leningrad and Germany.
Chinese famine was prolonged and had exposure windows that included prenatal as well as postnatal life and negative effects were reported during adult life (Huang et al., 2010a). A study by Huang et al. (2010b) compared birth size of infants born to who were conceived prior to the famine, during the famine and after the famine, and compared with those of women who were never exposed to the famine. Results showed that in rural areas, exposure to famine was associated with increases in offspring weight [71 g, (95% CI 30, 113)], length [0.3 cm, (95% CI 0.0, 0.6 cm)] and BMI [0.1 kg/m2, (95% CI 0.0, 0.2)] at birth, compared to those in prefamine and famine cohorts. In urban areas, however, there was no difference in offspring birth size. The authors proposed that markedly increased mortality in rural areas may have resulted in the selection of hardier mothers with greater growth potential, which becomes expressed in their offspring (Huang et al., 2010b) however other authors speculated that mortality was related to severity of famine and nutritional status of the mother at the time of conception (Song, 2010 2013).
In summary, acute severe maternal malnutrition may adversely affect the birth weight of the fetus especially when the exposure is during the third trimester of the pregnancy. All together, there is an increase risk of perinatal mortality when there is exposure to intrauterine famine and the risk may be as high as 6 times compared to normal, as seen in Dutch famine (Smith, 1947a,b).

Effect of Maternal Overweight and Obesity

Effects of maternal obesity on birth outcomes and its long-term effects during childhood and adulthood are less well studied compared to undernutrition. There are, however, recent epidemiological studies that report a relationship between maternal obesity and health of the offspring later in life (O’Reilly and Reynolds, 2013Drake and Reynolds, 2010Howie et al., 2009Maric-Bilkan et al., 2011Poston et al., 2011Zhang et al., 2011). The most common observations among these studies are that maternal obesity is linked to the development of elements of the metabolic syndrome, cardiovascular and renal disease, hypertension, and cerebral dysfunction as well as type 2 diabetes and obesity later in the life of the offspring (Howie et al., 2009Maric-Bilkan et al., 2011). There is a strong association of high fat diet and maternal obesity to obesity in offspring (Zhang et al., 2011Srinivasan et al., 2006). This risk is further modified by accelerated growth during early childhood (Symonds et al., 2009). There are also data that showed maternal obesity is a risk factor for prematurity, birth asphyxia, congenital anomalies, stillbirths, and infant mortality (Johansson et al., 2014Cnattingius et al., 2013Persson et al., 2014Stothard et al., 2009). Maternal obesity can impact neurodevelopment during childhood as well. A study conducted by center for disease control (CDC) showed that in comparison with children of normal-weight mothers, children born to obese mothers had an increased risk of learning or behavioral disabilities, gross motor delays and low reading scores (Hinkle et al., 2013). There was no affect on physical disabilities, fine motor skills, math skills etc.
The exact mechanisms, by which maternal obesity adversely affects the health of offspring, are not clear. Several metabolic pathways are likely to influence fetal development and neonatal outcomes and may include altered placental nutrient transport, glucose metabolism, excessive fetal exposure to insulin, and state of inflammation (Poston et al., 2011). In terms of long-term effects, “programming” must include permanent changes in cellular structure or function in the offspring in response to the metabolic consequences of maternal obesity (Poston et al., 2011). This may involve hormone dysregulation, hyperinsulinemia, fat deposit during fetal life, and epigenetic modification of DNA (Zambrano and Nathanielsz, 2013Zhang et al., 2011Symonds et al., 2009). The human data in this regard are relatively scarce; however, animal studies had helped explain some of the above listed mechanism (Zambrano and Nathanielsz, 2013). More research is needed to discover the mechanisms in humans and ultimately develop interventions to target these mechanisms to avoid the adverse consequence of maternal obesity.

Effect of Preconception Nutritional Status

Maternal prepregnancy nutritional status also determines the pregnancy outcomes and health of offspring later in life. Women who are underweight before pregnancy have a 32% increased risk of preterm birth, and a 64% increased risk of having small-for-gestational age babies (Dean et al., 2014a). The recent pooled analysis found a scarce evidence of an effect of prepregnancy underweight on stillbirths, LBW, or congenital birth defects; although studies on balanced energy protein supplementation during pregnancy have indicated a significant value (Dean et al., 2014a). However, maternal prepregnancy underweight is associated with reduced risk of hypertensive disorders of pregnancy, preeclampsia, and gestational diabetes mellitus (Dean et al., 2014a). Prepregnancy overweight, on the other hand, increases the risk for hypertensive disorders of pregnancy, gestational diabetes mellitus, and cesarean section at time of delivery (Crane et al., 2013Schummers et al., 2015). Similarly there is increased risk of adverse outcomes for fetus and newborn including large-for-gestational age babies, preterm birth, stillbirth, and neonatal death (Dzakpasu et al., 2015Tabet et al., 2015). There is some evidence to suggest that prepregnancy obesity can have detrimental effect on growth and development of children later in life. A study from Brazil showed increased risk of obesity during childhood for children who were born to mother with high prepregnancy body mass index (Castillo et al., 2015). Another study showed abnormal cardio-metabolic profile of adolescence who were born to mothers with high prepregnancy body mass index and had increased weight gain early in pregnancy (Gaillard et al., 2016). Another study showed significant problems with inattention, hyperactivity, and negative emotionality in 5-year-old children who were born to underweight, overweight, or obese mothers (Rodriguez, 2010).

Evidence From Nutritional Interventional Studies Among Humans

There are multiple nutrition interventions that had been shown to have a positive effect on maternal and infant outcomes (Bhutta et al., 2013); however, long-term effects of these interventions are not well studied. In this section, we present the most recent data on long-term effects of nutrition interventions given during preconception period or pregnancy. We included mainly those nutritional interventions that have been shown to have a positive impact on maternal and birth outcomes and were recommended in Lancet Nutrition series published in 2013. Table 1.1 presents a summary of evidence for these interventions.

Table 1.1

Effect of Nutrition Intervention on Maternal and Birth Outcomes and Long-Term Effect for Offspring

Nutrition intervention Effect on maternal and birth outcomesa Long-term effects for offspring
Folic acid Women of reproductive age Significant effects: NTDs (RR 0.28, 95% CI 0.15–0.52), recurrence of NTDs (RR 0.32, 95% CI 0.17–0.60). Nonsignificant effects: other congenital abnormalities, miscarriages, still births. No intervention study reported data for long-term outcomes for offspring for periconceptional folic acid supplementation.
During pregnancy Significant effects: mean birth weight (MD 135.75, 95% CI 47.85–223.68), incidence of megaloblastic anemia (RR 0.21, 95% CI 0.11–0.38). Nonsignificant effects: preterm birth, still births, mean predelivery hemoglobin, serum folate, red cell folate. One study from Nepal showed reduced risk of microalbuminuria for children aged 6–8 years whose mothers were supplemented with folic acid OR, 0.56; (95% CI, 0.33–0.93) and folic acid + iron + zinc OR, 0.53 (95% CI, 0.32–0.89) groups and a reduced risk of metabolic syndrome in the folic acid group OR, 0.63 (95% CI, 0.41–0.97) compared to control.
Iron and folate supplementation Women of reproductive age Intermittent iron supplementation. Significant effects: anemia (RR 0.73, 95% CI 0.56–0.95), serum hemoglobin concentration (MD 4.58 g/L, 95% CI 2.56–6.59), serum ferritin concentration (MD 8.32, 95% CI 4.97–11.66). Nonsignificant effects: iron deficiency, adverse events, depression. No study available that reported outcomes of iron supplementation during preconception period and assessed growth or neurodevelopment outcomes during childhood or adulthood.
During pregnancy

Daily iron-alone supplementation. Significant effects: low birth weight (RR 0.81, 95% CI 0.68–0.97), birth weight (MD 30.81 g, 95% CI 5.94–55.68), serum hemoglobin concentration at term (MD 8.88 g/L, 95% CI 6.96–10.80), anemia at term (RR 0.30, 95% CI 0.19–0.46), iron deficiency (RR 0.43, 95% CI 0.27–0.66), iron deficiency anemia (RR 0.33, 95% CI 0.16–0.69), side-effects (RR 2.36, 95% CI 0.96–5.82).

Nonsignificant effects: premature delivery, neonatal death, congenital anomalies iron-folate supplementation. Significant effects: birth weight (MD 57.7 g, 95% CI 7.66–107.79), anemia at term (RR 0.34, 95% CI 0.21–0.54), serum hemoglobin concentration at term (MD 16.13 g/L, 95% CI 12.74–19.52). Nonsignificant effects: low birth weight, premature birth, neonatal death, congenital anomalies.

Maternal iron/folate supplementation was shown to improve intellectual functioning of children aged 7–9 years from a study from Nepal (Christian et al., 2010); however another study from Australia did not show an effect of maternal iron supplementation on IQ scores in children at 4 years of age (Zhou et al., 2006). No beneficial effect of iron supplementation during pregnancy for growth during childhood based on four studies.
MMN supplementation during pregnancy

Significant effects: low birth weight (RR 0.88, 95% CI 0.85–0.91), SGA (RR 0.89, 95% CI 0.83–0.96), preterm birth (RR 0.97, 95% CI 0.94–0.99).

Nonsignificant effects: miscarriage, maternal mortality, perinatal mortality, stillbirths, and neonatal mortality. Insufficient data for neurodevelopmental outcomes.

Results from meta-analysis that included 9 trials of maternal MMN supplementation and assessed postnatal growth in children < 5 years of age (Lu et al., 2014). Significant effect on head circumference (effect size = 0.08, 95% CI: 0.00–0.15). There was no evidence of benefits on weight (P = 0.11), height (P = 0.66), weight-for-age z scores (P = 0.34), height-for-age z scores (P = 0.81), and weight-for-height z scores (P = 0.22).

Mixed results for neurodevelopment outcomes.

Calcium supplementation during pregnancy Significant effects: preeclampsia (RR 0.48, 95% CI 0.34–0.67), birth weight 85 g (95% CI 37–133), preterm birth (RR 0.76, 95% CI 0.60–0.97). Nonsignificant effects: perinatal mortality, low birth weight, neonatal mortality. Meta-analysis of two randomized trials and three observational studies (Bergel and Barros, 2007). Maternal calcium supplementation decreased incidence of hypertension [RR = 0.59, 95% CI 0.39–0.90] and systolic blood pressure [MD −1.92 mm Hg, 95% CI −3.14 to −0.71] for children aged 7 years.
Iodine supplementation during pregnancy Significant effects: cretinism at 4 years of age (RR 0.27, 95% CI 0.12–0.60), developmental scores 10–20% higher in young children, birth weight 3.82–6.30% higher. One study reported no effect of maternal Iodine supplementation on neurodevelopment outcomes for children aged upto 15 years (Pharoah and Connolly, 1991).
Maternal supplementation with balanced energy protein Significant effects: SGA (RR 0.66, 95% CI 0.49–0.89), stillbirths (RR 0.62, 95% CI 0.40–0.98, birth weight (MD 73 g, 95% CI 30–117). Mixed results for growth outcomes during childhood. One study from India (Kinra et al., 2008) showed that the adolescent whose mothers received balanced protein energy supplementation during pregnancy had more favorable measures of insulin resistance and arterial stiffness: lower HOMA (homoeostasis model Assessment) score and lower augmentation index (measure of arterial stiffness). There was no evidence of differences in blood pressures from study from India, Gambia, and Guatemala (Webb et al., 2005Kinra et al., 2008Hawkesworth et al., 2009). Three studies reported positive effect of protein supplementation during pregnancy and on neurodevelopment outcomes during childhood especially for children born to malnourished mothers (Tofail et al., 2008Waber et al., 1981Freeman et al., 1980).



a The effect estimates in this column were taken from Lancet Nutrition series 2013 paper 2 (Bhutta et al., 2013).

Multiple Micronutrient Supplementations During Pregnancy

Multiple micronutrient (MMN) supplements during pregnancy have been shown to reduce incidence of LBW and SGA (Haider and Bhutta, 2015). Does this reduction in incidence of LBW and SGA babies translate to better long-term growth and development outcomes during childhood and adulthood? Recent data were available for childhood outcomes from studies that assessed MMN supplements during pregnancy for birth outcomes (Khan et al., 2011Ramakrishnan et al., 2009Stewart et al., 2009aRoberfroid et al., 2012Prado et al., 2012McGrath et al., 2006Wang et al., 2012Hanieh et al., 2013Vaidya et al., 2008Huy et al., 2009). We found 10 such studies conducted in 8 different developing countries as follows: Bangladesh (Khan et al., 2011), Nepal (2 studies) (Stewart et al., 2009aVaidya et al., 2008), China (Wang et al., 2012), Indonesia (Prado et al., 2012), Mexico (Ramakrishnan et al., 2009), Burkina Faso (Roberfroid et al., 2012), Vietnam (2 studies) (Hanieh et al., 2013Huy et al., 2009) and Tanzania (McGrath et al., 2006 Feb). All studies were in rural settings. The study conducted in Tanzania included women with HIV infection (McGrath et al., 2006 Feb). Age range for follow-up was 6 months to 8.5 years. There were 6 studies that had more than one intervention study arm (Khan et al., 2011Stewart et al., 2009aMcGrath et al., 2006Wang et al., 2012Hanieh et al., 2013Huy et al., 2009). All studies used UNIMAP formula except Tanzania trial. The comparison groups included iron/folate supplementation in all the studies except Tanzania trial where control group received placebo. In 8 studies, participants received intervention during pregnancy only while in Mexico trial intervention was given to both mother and children up to 2 years.
Four of the ten studies described above showed that MMN had a positive effect on growth outcomes during childhood. Rest of the studies did not show an overall effect for growth; however, some author reported positive effects for those with better compliance and whose mothers were malnourished at the start of the study (Ramakrishnan et al., 2009). Six randomized trial described effect of MMN during pregnancy on neurodevelopment outcomes during childhood (Prado et al., 2012McGrath et al., 2006Hanieh et al., 2013Tofail et al., 2008Christian et al., 2010Li et al., 2009) and these studies had mixed results. Some of the studies showed positive effects on neurodevelopment, such as motor and cognitive abilities of children; however, the results were not consistent across the studies (Leung et al., 2011). A recent meta-analysis assessed effect of MMN supplements during pregnancy and its effects on growth during childhood. It included nine trials and showed that antenatal MMN supplementation increased child head circumference (effect size = 0.08, 95% CI: 0.00–0.15) compared with supplementation with two micronutrient or less (Lu et al., 2014). There was no evidence of benefits on weight (P = 0.11), height (P = 0.66), weight-for-age z scores (P = 0.34), height-for-age z scores (P = 0.81), and weight-for-height z scores (P = 0.22). This review however only analyzed continuous data and no categorical outcomes like stunting or underweight were analyzed. MMN supplements may not affect average weight or height but may decrease the prevalence of stunting or underweight. Stewart et al. (2009b) reported data from Nepal for effect of micronutrient supplements on blood pressure, BMI, waist circumference, glycated hemoglobin, cholesterol, triglycerides, glucose, insulin, and the urinary microalbumin:creatinine ratio in children 6–8 years. No combination of micronutrient supplement produced differential effect on blood pressure, cholesterol, triglycerides, glucose, and insulin. There was however a reduced risk of microalbuminuria in the folic acid and folic acid + iron + zinc groups and a reduced risk of metabolic syndrome in the folic acid group. There was no effect of MMN on reduction of obesity in children 6–8 years (Stewart et al., 2009a). Another study from Nepal by Devakumar and coworkers reported data on obesity, blood pressure, and lung function of children whose mothers were supplemented with MMN compared to iron-folate at an average age of 8.5 years (Devakumar et al., 2014 2015). There was no differential effect on blood pressure, obesity, or lung function (based on spirometry) between the two groups.
Lipid-based nutrient supplements (LNS) include multiple micronutrient (MMN) fortified semisolid pastes usually prepared from vegetable oil, groundnut paste, milk, sugar, and different concentrations of micronutrients depending on the type of product and the specific nutritional conditions in the target population. The existing evidence base on the impact of LNS during pregnancy is limited. When compared to multiple micronutrient supplementations, the provision of LNS to pregnant women in Burkina Faso resulted in higher birth length. Supplemented women were found to have elevated leptin concentrations and could be attributable to the higher neonatal fat mass (Huybregts et al., 2013). However, the positive effect of prenatal LNS on birth length was not sustained during the postnatal phase suggesting lack of long term impact on child linear growth (Lanou et al., 2014).

Balanced Energy Protein Supplementation During Pregnancy

Previous evidence shows that balanced protein supplementation during pregnancy reduces the incidence of stillbirths, LBW, SGA babies, and increase birth weight (Imdad et al., 2011Kramer and Kakuma, 2003Ota et al., 2015). There have been follow-up studies that reported growth outcomes during childhood from randomized trials that assessed effect of prenatal protein energy supplementation. These studies were conducted in Bangladesh (Khan et al., 2011), Guatemala (Webb et al., 2005), Gambia (Hawkesworth et al., 2008), India (Kinra et al., 2008), Colombia (Waber et al., 1981), and Taiwan (Wohlleb et al., 1983). All the studies provided nutrition supplement as balanced protein energy supplements (i.e., protein provided < 25% of total energy supplements). The individual components of energy supplements vary greatly among the studies. Comparison groups included routine care in five studies (Khan et al., 2011Kinra et al., 2008Hawkesworth et al., 2009Elwood et al., 1981) while in three studies control group also received a supplement (Webb et al., 2005Lanou et al., 2014Wehby et al., 2008). Contents of supplements in comparison groups of these studies had low energy content and mainly consisted on multiple micronutrients. In one of the study, nutrition supplements were continued beyond pregnancy and children were also given the supplements (Webb et al., 2005). Age range of follow-up in included studies ranged from 12 months to 18 years.
The study from Bangladesh did not show an overall effect; however, early prenatal food supplementation lead to reduce prevalence of stunting among boys (Khan et al., 2011). The study by Stein et al. from Guatemala showed that children born to women who received the balanced protein energy supplementation were taller (age-adjusted difference: 0.80 cm; 95% CI: 0.16, 1.44 cm), and this effect was more pronounced in boys than in girls. There was a positive effect on neurodevelopment outcomes during childhood as described in three studies (Tofail et al., 2008Waber et al., 1981Freeman et al., 1980). A study from Gambia reported effect of balanced protein energy supplementation during pregnancy on blood pressure, body composition, fasting blood glucose, and cholesterol of children aged 11–17 years (Hawkesworth et al., 2009). There was no difference between the two study groups except that fasting blood sugars in protein supplementation group were slightly lower than control. Webb et al. (2005) reported data for protein energy supplementation during pregnancy from Guatemala and showed that there was no differential effect between high versus low energy supplements on blood pressure for adults 21–29 years old. A cohort study from India reported follow up data for offspring aged 13–18 years of mothers who were given balanced protein energy supplements during pregnancy compared to control (Kinra et al., 2008). The results showed that adolescents from intervention villages had more favorable measures of insulin resistance and arterial stiffness: lower HOMA (homoeostasis model assessment) score and lower augmentation index (measure of arterial stiffness). There was no evidence of differences in blood pressures and serum lipids.
It is important to note that there was significant heterogeneity in above-mentioned studies in terms of study site, participants, type and duration of supplements, type of comparison, and combined supplementation for mothers and children. This means that study results may not applicable from one site to the other site. In summary, balanced protein energy supplementation during pregnancy has variable effects on growth and development during childhood and might modify risk of chronic disease during adulthood.

Calcium Supplementation During pregnancy

Maternal calcium supplementation is known to have protective effect against gestational hypertensive disorders with small effect on preterm birth in populations with low calcium intake (Hofmeyr et al., 2014). This protective effect is reported to continue during childhood; however, there was small number of studies that reported follow-up data and had significant loss to follow-up (Bergel and Barros, 2007). Growth outcomes during childhood were not affected by calcium supplementation during pregnancy in studies reported from Argentina (Belizan et al., 1997), Australia (Hiller et al., 2007), USA (Hatton et al., 2003), Gambia (Hawkesworth et al., 2010), and Egypt (Abdel-Aleem et al., 2009).

Long Chain Fatty Acids

The most important long chain polyunsaturated fatty acids (LCPUFAs), arachidonic acid (AA), and docosahexaenoic acid (DHA), have a functional and structural role in several key cellular processes (McCann and Ames, 2005). DHA is the most abundant fatty acid in the brain and an important component of brain cell membranes and retina while AA is involved in several pathways of activation through the cell membrane receptors and is a precursor of eicosanoids, products of remarkable physiological activity (Fleith and Clandinin, 2005Innis, 2007). These are considered “essential” because the human body cannot synthesize them.
A review evaluating the impact of LCPUFA supplementation during pregnancy on head circumference of newborn suggests that supplementation was associated with significantly greater head circumference of the infants in the intervention group (Deng et al., 2012). However, the difference was no longer significant according to the sensitivity analysis. Another review suggests significantly greater length of pregnancy, increase in head circumference, and a modest increase in birth weight associated with n-3 LCPUFA supplementation during pregnancy for women with low-risk pregnancies (Szajewska et al., 2006Imhoff-Kunsch et al., 2012).
Existing evidence on the effect of prenatal supplementation of LCPUFAs on long-term growth and development is very limited. An existing review evaluating the effect of n-3 LCPUFAs during the perinatal period on later body composition suggests inconclusive evidence. Two of the included studies in the review showed inverse associations between maternal n-3 LCPUFA intake and children’s later body composition [including lower adiposity, body mass index (BMI) or body weight]; two studies showed direct associations while no effects were observed in the remaining two studies (Rodriguez et al., 2012). Another review suggests no effect of maternal n-3 LCPUFA supplementation during pregnancy and/or lactation on BMI in preschool and school-aged children (Stratakis et al., 2014Muhlhausler et al., 2010). There is no consistent data on the effect of LCPUFA supplementation during pregnancy and/or lactation on neurodevelopment and visual function in children (Dziechciarz et al., 2010). Most of the existing data on LCPUFA supplementation comes from high-income countries. More studies in low- and middle-income country settings are needed to determine any effect of supplementation in resource-poor settings, where intake is likely to be low. Further studies are required in which the intervention is confined to the perinatal period and that are sufficiently powered.

Iron/Folate Supplementation

Iron supplementation during pregnancy reduces anemia during pregnancy and also possibly increases birth weight (Pena-Rosas et al., 2015). Four studies were available that evaluated iron/folate supplementation during pregnancy and reported outcomes during childhood or adulthood. These studies were conducted in Nepal (Stewart et al., 2009a), Niger (Preziosi et al., 1997), Australia (Zhou et al., 2006), and China (Wang et al., 2012). Age range of follow-up was 6 months to 8 years. There was no differential effect of iron supplementation during pregnancy on outcomes during childhood. Maternal iron/folate supplementation was shown to improve intellectual functioning of children aged 7–9 years compared to control from a study from Nepal (Christian et al., 2010); however, a study from Australia did not show any difference in IQ scores for children aged 4 years (Zhou et al., 2006).
Preconception iron deficiency anemia is associated with fetal growth restriction and reduced birth weight. (Ronnenberg et al., 2004). There is currently no evidence for long-term effects of iron folate supplementation to the women of reproductive age; however, there is an ongoing study in Vietnam that addresses this question (Nguyen et al., 2012).

Periconceptional Folic Acid Supplementation

Periconceptional folic acid supplementation have proven to reduce the risk of recurrent and neural tube defects (NTDs), and it reduces overall prevalence of NTDs 41% (Dean et al., 2014a). Currently, no long-term data are available for perinatal folic acid supplementation.

Iodine Supplementation

While trials of supplementation during pregnancy have led to improved maternal and fetal outcomes; these benefits has not been demonstrated during childhood. There was one study that reported outcomes during childhood when iodine was supplemented during pregnancy (Pharoah and Connolly, 1991). This study was conducted in Papua New Guinea and was randomized on individual basis. There were about 192 kids available for assessment as part of follow-up. Methods of randomization and allocation concealment were clearly not described; however, the study was double blind. Follow-up was up to 15 years of age. There was no difference in neurodevelopmental outcomes for children whose mothers were supplemented with iodine compared to control.

Discussion

Observational and experimental studies from animals and humans described earlier showed that nutrition before and during pregnancy could have immediate and long-term effects on growth and development. Preconception nutritional status is as important as nutrition during pregnancy (Dean et al., 2014a). Effect of maternal malnutrition can be transmitted across generations and it may take two or three generation to reverse the effect of chronic maternal malnutrition (Martorell and Zongrone, 2012). Effect of acute malnutrition during pregnancy on offspring can be reversed with nutritional rehabilitation during early childhood (Martorell and Zongrone, 2012).
Growth failure is an indicator of poor nutrient availability at the cellular level and reflects widespread functional impairment. Poor nutrition early in life, as reflected in poor growth, has lifelong adverse consequences, including short stature, diminished work capacity, delayed cognition, less schooling, and reduced incomes (Victora et al., 2008Ramakrishnan et al., 1999). It is important to note that there are multiple factors that play an important role in intergenerational growth failure. These include complex biological process, which are not mutually exclusive and may include factors like shared genetic characteristics, epigenetic effects, programming of metabolic changes (due to maternal malnutrition), and the mechanics of a reduced space for the fetus to grow (due to maternal short stature). There are also sociocultural factors like poverty, socioeconomic status, and food security etc. that play significant role and mostly present in populations with high prevalence of maternal malnutrition (Martorell and Zongrone, 2012Ramakrishnan et al., 1999).
There is an interconnection of malnutrition during pregnancy, childhood, and adolescence and it can perpetuate the so-called “intergenerational cycle of growth failure” (Bhutta et al., 2013Black et al., 2013aMartorell and Zongrone, 2012Ramakrishnan et al., 1999). Young girls who grow poorly become stunted women, and are more likely to have small babies. These babies, if girls, will continue the cycle (Martorell and Zongrone, 2012). There are multiple opportunities to break this cycle; however, it needs a holistic approach with improved socioeconomic, nutrition, and health conditions. A recent review on intervention during preconceptional period proposed that optimal preconception care improve maternal and newborn outcomes like smoking cessation; increased use of folic acid; breastfeeding; greater odds of obtaining antenatal care; and lower rates of neonatal mortality (Dean et al., 2014a,b). The two Lancet series on Maternal and Child Undernutrition had described the burden of problem and recommended nutrition intervention for mothers, newborn, and children based on the best available evidence (Bhutta et al., 2013Black et al., 2008 2013aVictora et al., 2008). The latest series proposed that scaling up of proven interventions in high burden countries would reduce stunting by one-third (Bhutta et al., 2013).

Future trends and research

There is increasing recognition of double burden of malnutrition with overnutrition and undernutrition (Black et al., 2013a). Both of these conditions are related to adverse perinatal outcomes and have long-term consequences for human capital (Victora et al., 2008). In most instances these conditions share common causes and would need special consideration of interventions specifically targeting the first 1000 days of life (Naja et al., 2016). Avoidance of pregnancy during adolescence, healthy body composition at time of conception, optimal weight gain during pregnancy, adequate maternal micronutrient stores, exclusive breast feeding for the first 6 months, appropriate complementary foods during infancy, diets rich fruits and vegetables, and environments that encourage physical activity and play are some of the interventions that would promote linear growth but not fatness (Bhutta et al., 2013Ramakrishnan, 2004Nishtar et al., 2016Victora et al., 2016). Future studies should focus on these elements to assess the efficacy, delivery strategies, and cost of these interventions. The current epidemic of widespread availability and intake of sugared drinks, juices, and junk food must be averted through education and other programs (Nishtar et al., 2016).

Conclusions

Observational studies from animal and humans make a strong case of intergenerational effects of maternal malnutrition; however, the number of intervention studies with long-term follow is small and few robust conclusions can be drawn at this stage. Further intervention studies or cohorts with key health and nutrition interventions and long-term follow-up are needed to understand the relationships of maternal nutrition and risks of adverse medium to long-term outcomes and risks of chronic disease during childhood and adulthood.

Sources of additional information

1. Martorell, R., Zongrone, A., 2012. Intergenerational influences on child growth and undernutrition. Paediatr. Perinat. Epidemiol. 26 (Suppl. 1), 302–314.
2. Martorell, R., Richter, L., et al., 2008. Maternal and child undernutrition: consequences for adult health and human capital. Lancet 371 (9609), 340–357.
3. WHO report prepared by Dr. Hélène Delisle: Programming of chronic disease by impaired fetal nutrition Evidence and implications for policy and intervention strategies, 2002.
4. Ramakrishnan, U., Martorell, R., Schroeder, D.G., Flores, R., 1999. Role of intergenerational effects on linear growth. J. Nutr. 129 (2S Suppl.), 544S–549S.
5. Ross, M.G., Desai, M., 2005. Gestational programming: population survival effects of drought and famine during pregnancy. Am. J. Physiol. Regul. Integr. Comp. Physiol. 288 (1), R25–R33.
6. Wadhwa, P.D., Buss, C., Entringer, S., Swanson, J.M., 2009. Developmental origins of health and disease: brief history of the approach and current focus on epigenetic mechanisms. Semin. Reprod. Med. 27 (5), 358–368.
7. Godfrey, K.M., Barker, D.J., 2000. Fetal nutrition and adult disease. Am. J. Clin. Nutr. 71 (5 Suppl.), 1344S–1352S.

References

Abdel-Aleem H, et al. The effect of calcium supplementation during pregnancy on fetal and infant growth: a nested randomized controlled trial within WHO calcium supplementation trial. J. Matern. Fetal Neonatal Med. 2009;22(2):94100.

Antonov AN. Children born during the siege of Leningrad in 1942. J. Pediatr. 1947;30(3):250259.

Belizan JM, et al. Long-term effect of calcium supplementation during pregnancy on the blood pressure of offspring: follow up of a randomised controlled trial. BMJ. 1997;315(7103):281285.

Bergel E, Barros AJ. Effect of maternal calcium intake during pregnancy on children’s blood pressure: a systematic review of the literature. BMC Pediatr. 2007;7:15.

Bhutta ZA, et al. Evidence-based interventions for improvement of maternal and child nutrition: what can be done and at what cost? Lancet. 2013;382(9890):452477.

Black RE, et al. Maternal and child undernutrition: global and regional exposures and health consequences. Lancet. 2008;371(9608):243260.

Black RE, et al. Maternal and child undernutrition and overweight in low-income and middle-income countries. Lancet. 2013;382(9890):427451.

Black RE, et al. Maternal and child undernutrition and overweight in low-income and middle-income countries. Lancet. 2013;382(9890):427451.

Castillo H, Santos IS, Matijasevich A. Relationship between maternal pre-pregnancy body mass index, gestational weight gain and childhood fatness at 6–7 years by air displacement plethysmography. Matern. Child Nutr. 2015;11(4):606617.

Cesani MF, Oyhenart EE, Pucciarelli HM. Effect of intergenerational chronic undernutrition on ponderal, and linear growth. ISRN Nutr. 2014;2014:453460.

Christian P, et al. Prenatal micronutrient supplementation and intellectual and motor function in early school-aged children in Nepal. JAMA. 2010;304(24):27162723.

Cnattingius S, et al. Maternal obesity and risk of preterm delivery. JAMA. 2013;309(22):23622370.

Crane JM, et al. Maternal and perinatal outcomes of extreme obesity in pregnancy. J. Obstet. Gynaecol. Can. 2013;35(7):606611.

Dean, R.F.A., 1946. The size of the baby at birth and the yield of breast milk. In: Studies of Under-Nutrition: Wuppertal 1946-9., in Her Majesty’s Stationary Office, (Medical Research Council special report series, 275). London. p. 346–378.

Dean SV, et al. Preconception care: nutritional risks and interventions. Reprod. Health. 2014;11(Suppl. 3):S3.

Dean SV, et al. Preconception care: closing the gap in the continuum of care to accelerate improvements in maternal, newborn and child health. Reprod. Health. 2014;11(Suppl. 3):S1.

Deng J, et al. Meta-analysis of effect of n-3 long-chain polyunsaturated fatty acid supplementation of pregnant women on head circumference of newborn infants. Zhonghua yu fang yi xue za zhi. 2012;46(12):11121116.

Devakumar D, et al. Effect of antenatal multiple micronutrient supplementation on anthropometry and blood pressure in mid-childhood in Nepal: follow-up of a double-blind randomised controlled trial. Lancet Glob. Health. 2014;2(11):e654e663.

Devakumar D, et al. Effects of antenatal multiple micronutrient supplementation on lung function in mid-childhood: follow-up of a double-blind randomised controlled trial in Nepal. Eur. Respir. J. 2015;45(6):15661575.

Drake AJ, Liu L. Intergenerational transmission of programmed effects: public health consequences. Trends Endocrinol. Metab. 2010;21(4):206213.

Drake AJ, Reynolds RM. Impact of maternal obesity on offspring obesity and cardiometabolic disease risk. Reproduction. 2010;140(3):387398.

Dzakpasu S, et al. Contribution of prepregnancy body mass index and gestational weight gain to adverse neonatal outcomes: population attributable fractions for Canada. BMC Pregnancy Childbirth. 2015;15:21.

Dziechciarz P, Horvath A, Szajewska H. Effects of n-3 long-chain polyunsaturated fatty acid supplementation during pregnancy and/or lactation on neurodevelopment and visual function in children: a systematic review of randomized controlled trials. J. Am. Coll. Nutr. 2010;29(5):443454.

Elwood PC, et al. Child growth (0–5 years), and the effect of entitlement to a milk supplement. Arch. Dis. Child. 1981;56(11):831835.

Fleith M, Clandinin M. Dietary PUFA for preterm and term infants: review of clinical studies. Crit. Rev. Food Sci. Nutr. 2005;45(3):205229.

Freeman HE, et al. Nutrition and cognitive development among rural Guatemalan children. Am. J. Public Health. 1980;70(12):12771285.

Gaillard R, et al. Associations of maternal prepregnancy body mass index and gestational weight gain with cardio-metabolic risk factors in adolescent offspring: a prospective cohort study. BJOG. 2016;123(2):207216.

Gupta A, et al. Hypothalamic alterations in fetuses of high fat diet-fed obese female rats. J. Endocrinol. 2009;200(3):293300.

Haider BA, Bhutta ZA. Multiple-micronutrient supplementation for women during pregnancy. Cochrane Database Syst. Rev. 2015;11:CD004905.

Hanieh S, et al. The effect of intermittent antenatal iron supplementation on maternal and infant outcomes in rural Viet Nam: a cluster randomised trial. PLoS Med. 2013;10(6):e1001470.

Hatton DC, et al. Gestational calcium supplementation and blood pressure in the offspring. Am. J. Hypertens. 2003;16(10):801805.

Hawkesworth S, et al. Dietary supplementation of rural Gambian women during pregnancy does not affect body composition in offspring at 11–17 years of age. J. Nutr. 2008;138(12):24682473.

Hawkesworth S, et al. Maternal protein-energy supplementation does not affect adolescent blood pressure in The Gambia. Int. J. Epidemiol. 2009;38(1):119127.

Hawkesworth S, et al. Effect of maternal calcium supplementation on offspring blood pressure in 5- to 10-y-old rural Gambian children. Am. J. Clin. Nutr. 2010;92(4):741747.

Hiller JE, et al. Calcium supplementation in pregnancy and its impact on blood pressure in children and women: follow up of a randomised controlled trial. Aust. NZ J. Obstet. Gynaecol. 2007;47(2):115121.

Hinkle SN, et al. Maternal prepregnancy weight status and associations with children’s development and disabilities at kindergarten. Int. J. Obes. (Lond). 2013;37(10):13441351.

Hofmeyr GJ, et al. Calcium supplementation during pregnancy for preventing hypertensive disorders and related problems. Cochrane Database Syst. Rev. 2014;6:CD001059.

Howie GJ, et al. Maternal nutritional history predicts obesity in adult offspring independent of postnatal diet. J. Physiol. 2009;587(Pt. 4):905915.

Huang C, et al. Early life exposure to the 1959–1961 Chinese famine has long-term health consequences. J. Nutr. 2010;140(10):18741878.

Huang C, et al. Bigger babies born to women survivors of the 1959-1961 Chinese famine: a puzzle due to survival selection? J. Dev. Orig. Health Dis. 2010;1(6):412418.

Hult M, et al. Hypertension, diabetes and overweight: looming legacies of the Biafran famine. PLoS One. 2010;5(10):e13582.

Huy ND, et al. An effectiveness trial of multiple micronutrient supplementation during pregnancy in Vietnam: impact on birthweight and on stunting in children at around 2 years of age. Food Nutr. Bull. 2009;30(4 Suppl.):S506S516.

Huybregts L, et al. Prenatal lipid-based nutrient supplements increase cord leptin concentration in pregnant women from rural Burkina Faso. J. Nutr. 2013;143(5):576583.

Imdad A, Yakoob MY, Bhutta ZA. Effect of balanced protein energy supplementation during pregnancy on birth outcomes. BMC Public Health. 2011;11(Suppl. 3):S17.

Imhoff-Kunsch B, et al. Effect of n-3 long-chain polyunsaturated fatty acid intake during pregnancy on maternal, infant, and child health outcomes: a systematic review. Paediatr. Perinat. Epidemiol. 2012;26(s1):91107.

Innis SM. Fatty acids and early human development. Early Hum. Dev. 2007;83(12):761766.

Johansson S, et al. Maternal overweight and obesity in early pregnancy and risk of infant mortality: a population based cohort study in Sweden. BMJ. 2014;349:g6572.

Khan AI, Kabir I, Ekstrom EC, Asling-Monemi K. Effects of prenatal food and micronutrient supplementation on child growth from birth to 54 months of age: a randomized trial in Bangladesh. Nutr. J. 2011;10(134).

Kinra S, Rameshwar Sharma, K.V., Mendu, V.V.R., 2008. Effect of integration of supplemental nutrition with public health programmes in pregnancy and early childhood on cardiovascular risk in rural Indian adolescents: long term follow-up of Hyderabad nutrition trial. BMJ. 337, a605.

Kirk SL, et al. Maternal obesity induced by diet in rats permanently influences central processes regulating food intake in offspring. PLoS One. 2009;4(6):e5870.

Koupil I, et al. Blood pressure, hypertension and mortality from circulatory disease in men and women who survived the siege of Leningrad. Eur. J. Epidemiol. 2007;22(4):223234.

Kramer MS, Kakuma R. Energy and protein intake in pregnancy. Cochrane Database Syst. Rev. 2003;4(4): CD000032.

Lanou H, et al. Prenatal nutrient supplementation and postnatal growth in a developing nation: an RCT. Pediatrics. 2014;133(4):e1001e1008.

Leung BM, Wiens KP, Kaplan BJ. Does prenatal micronutrient supplementation improve children’s mental development? A systematic review. BMC Pregnancy Childbirth. 2011;11:12.

Li Q, et al. Effects of maternal multimicronutrient supplementation on the mental development of infants in rural western China: follow-up evaluation of a double-blind, randomized, controlled trial. Pediatrics. 2009;123(4):e685e692.

Lu WP, et al. Effects of multimicronutrient supplementation during pregnancy on postnatal growth of children under 5 years of age: a meta-analysis of randomized controlled trials. PLoS One. 2014;9(2):e88496.

Lumey LH, Stein AD. Offspring birth weights after maternal intrauterine undernutrition: a comparison within sibships. Am. J. Epidemiol. 1997;146(10):810819.

Maric-Bilkan C, et al. Impact of maternal obesity and diabetes on long-term health of the offspring. Exp. Diab. Res. 2011;2011:163438.

Martorell R, Zongrone A. Intergenerational influences on child growth and undernutrition. Paediatr. Perinat. Epidemiol. 2012;26(Suppl. 1):302314.

McCann JC, Ames BN. Is docosahexaenoic acid, an n–3 long-chain polyunsaturated fatty acid, required for development of normal brain function? An overview of evidence from cognitive and behavioral tests in humans and animals. Am. J. Clin. Nutr. 2005;82(2):281295.

McCurdy CE, et al. Maternal high-fat diet triggers lipotoxicity in the fetal livers of nonhuman primates. J. Clin. Invest. 2009;119(2):323335.

McGrath N, et al. Effect of maternal multivitamin supplementation on the mental and psychomotor development of children who are born to HIV-1-infected mothers in Tanzania. Pediatrics. 2006;117(2):e216e225.

McGrath N, et al. Effect of maternal multivitamin supplementation on the mental and psychomotor development of children who are born to HIV-1-infected mothers in Tanzania. Pediatrics. 2006;117(2):e216e225.

Muhlhausler BS, Gibson RA, Makrides M. Effect of long-chain polyunsaturated fatty acid supplementation during pregnancy or lactation on infant and child body composition: a systematic review. Am. J. Clin. Nutr. 2010;92(4):857863.

Naja F, et al. Study protocol: mother and infant nutritional assessment (MINA) cohort study in Qatar and Lebanon. BMC Pregnancy Childbirth. 2016;16(1):98.

Nguyen PH, et al. Rationale, design, methodology and sample characteristics for the Vietnam pre-conceptual micronutrient supplementation trial (PRECONCEPT): a randomized controlled study. BMC Public Health. 2012;12:898.

Nishtar S, Gluckman P, Armstrong T. Ending childhood obesity: a time for action. Lancet. 2016;387(10021):825827.

Nohr EA, et al. Mortality in infants of obese mothers: is risk modified by mode of delivery? Acta Obstet. Gynecol. Scand. 2012;91(3):363371.

O’Reilly JR, Reynolds RM. The risk of maternal obesity to the long-term health of the offspring. Clin. Endocrinol. (Oxf). 2013;78(1):916.

Ota E, et al. Antenatal dietary education and supplementation to increase energy and protein intake. Cochrane Database Syst. Rev. 2015;6:CD000032.

Pena-Rosas JP, et al. Daily oral iron supplementation during pregnancy. Cochrane Database Syst. Rev. 2015;7:CD004736.

Persson M, et al. Maternal overweight and obesity and risks of severe birth-asphyxia-related complications in term infants: a population-based cohort study in Sweden. PLoS Med. 2014;11(5):e1001648.

Pharoah PO, Connolly KJ. Effects of maternal iodine supplementation during pregnancy. Arch. Dis. Child. 1991;66(1):145147.

Poston L, et al. Obesity in pregnancy: implications for the mother and lifelong health of the child. A consensus statement. Pediatr. Res. 2011;69(2):175180.

Prado EL, et al. Maternal multiple micronutrient supplements and child cognition: a randomized trial in Indonesia. Pediatrics. 2012;130(3):e536e546.

Preziosi P, et al. Effect of iron supplementation on the iron status of pregnant women: consequences for newborns. Am. J. Clin. Nutr. 1997;66(5):11781182.

Price KC, Coe CL. Maternal constraint on fetal growth patterns in the rhesus monkey (Macaca mulatta): the intergenerational link between mothers and daughters. Hum. Reprod. 2000;15(2):452457.

Price KC, Hyde JS, Coe CL. Matrilineal transmission of birth weight in the rhesus monkey (Macaca mulatta) across several generations. Obstet. Gynecol. 1999;94(1):128134.

Pucciarelli HM, et al. Relative food intake of rats submitted to a moderate transgenerational undernutrition. Growth Dev. Aging. 2002;65(2):8393.

Ramakrishnan U. Nutrition and low birth weight: from research to practice. Am. J. Clin. Nutr. 2004;79(1):17.

Ramakrishnan U, et al. Role of intergenerational effects on linear growth. J. Nutr. 1999;129(2S Suppl.):544S549S.

Ramakrishnan U, et al. Multiple micronutrient supplementation during early childhood increases child size at 2 y of age only among high compliers. Am. J. Clin. Nutr. 2009;89(4):11251131.

Roberfroid D, et al. Impact of prenatal multiple micronutrients on survival and growth during infancy: a randomized controlled trial. Am. J. Clin. Nutr. 2012;95(4):916924.

Rodriguez A. Maternal pre-pregnancy obesity and risk for inattention and negative emotionality in children. J. Child Psychol. Psychiatry. 2010;51(2):134143.

Rodriguez G, et al. Effect of n-3 long chain polyunsaturated fatty acids during the perinatal period on later body composition. Br. J. Nutr. 2012;107(S2):S117S128.

Rogers EH, et al. Lack of evidence for intergenerational reproductive effects due to prenatal and postnatal undernutrition in the female CD-1 mouse. Reprod. Toxicol. 2003;17(5):519525.

Ronnenberg AG, et al. Preconception hemoglobin and ferritin concentrations are associated with pregnancy outcome in a prospective cohort of Chinese women. J. Nutr. 2004;134(10):2586.

Ross MG, Desai M. Gestational programming: population survival effects of drought and famine during pregnancy. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2005;288(1):R25R33.

Samuelsson AM, et al. Diet-induced obesity in female mice leads to offspring hyperphagia, adiposity, hypertension, and insulin resistance: a novel murine model of developmental programming. Hypertension. 2008;51(2):383392.

Schummers L, et al. Risk of adverse pregnancy outcomes by prepregnancy body mass index: a population-based study to inform prepregnancy weight loss counseling. Obstet. Gynecol. 2015;125(1):133143.

Smith CA. The effect of wartime starvation in Holland upon pregnancy and its product. Am. J. Obstet. Gynecol. 1947;53(4):599608.

Smith CA. Effects of maternal under nutrition upon the newborn infant in Holland (1944–1945). J. Pediatr. 1947;30(3):229243.

Song S. Mortality consequences of the 1959–1961 great leap forward famine in China: debilitation, selection, and mortality crossovers. Soc. Sci. Med. 2010;71(3):551558.

Song S. Identifying the intergenerational effects of the 1959–1961 chinese great leap forward famine on infant mortality. Econ. Hum. Biol. 2013;11(4):474487.

Srinivasan M, et al. Maternal high-fat diet consumption results in fetal malprogramming predisposing to the onset of metabolic syndrome-like phenotype in adulthood. Am. J. Physiol. Endocrinol. Metab. 2006;291(4):E792E799.

Stein AD, et al. Exposure to a nutrition supplementation intervention in early childhood and risk factors for cardiovascular disease in adulthood: evidence from Guatemala. Am. J. Epidemiol. 2006;164(12):1160.

Stewart RJ, Preece RF, Sheppard HG. Twelve generations of marginal protein deficiency. Br. J. Nutr. 1975;33(2):233253.

Stewart RJ, et al. The effect of rehabilitation at different stages of development of rats marginally malnourished for ten to twelve generations. Br. J. Nutr. 1980;43(3):403412.

Stewart CP, et al. Antenatal supplementation with folic acid + iron + zinc improves linear growth and reduces peripheral adiposity in school-age children in rural Nepal. Am. J. Clin. Nutr. 2009;90(1):132140.

Stewart CP, et al. Antenatal micronutrient supplementation reduces metabolic syndrome in 6- to 8-year-old children in rural Nepal. J. Nutr. 2009;139(8):15751581.

Stothard KJ, et al. Maternal overweight and obesity and the risk of congenital anomalies: a systematic review and meta-analysis. JAMA. 2009;301(6):636650.

Stratakis N, et al. Effect of maternal n-3 long-chain polyunsaturated fatty acid supplementation during pregnancy and/or lactation on adiposity in childhood: a systematic review and meta-analysis of randomized controlled trials. Eur. J. Clin. Nutr. 2014;68(12):12771287.

Symonds ME, et al. Nutritional programming of the metabolic syndrome. Nat. Rev. Endocrinol. 2009;5(11):604610.

Szajewska H, Horvath A, Koletzko B. Effect of n–3 long-chain polyunsaturated fatty acid supplementation of women with low-risk pregnancies on pregnancy outcomes and growth measures at birth: a meta-analysis of randomized controlled trials. Am. J. Clin. Nutr. 2006;83(6):13371344.

Tabet M, et al. Prepregnancy body mass index in a first uncomplicated pregnancy and outcomes of a second pregnancy. Am. J. Obstet. Gynecol. 2015;213(4):548.

Tofail F, et al. Effects of prenatal food and micronutrient supplementation on infant development: a randomized trial from the maternal and infant nutrition interventions, matlab (MINIMat) study. Am. J. Clin. Nutr. 2008;87(3):704711.

Vaidya A, et al. Effects of antenatal multiple micronutrient supplementation on children’s weight and size at 2 years of age in Nepal: follow-up of a double-blind randomised controlled trial. Lancet. 2008;371(9611):492499.

Victora CG, et al. Maternal and child undernutrition: consequences for adult health and human capital. Lancet. 2008;371(9609):340357.

Victora CG, et al. Breastfeeding in the 21st century: epidemiology, mechanisms, and lifelong effect. Lancet. 2016;387(10017):475490.

Villar J, et al. Nutritional interventions during pregnancy for the prevention or treatment of maternal morbidity and preterm delivery: an overview of randomized controlled trials. J. Nutr. 2003;133(5 Suppl. 2):1606S1625S.

Waber DP, et al. Nutritional supplementation, maternal education, and cognitive development of infants at risk of malnutrition. Am. J. Clin. Nutr. 1981;34(Suppl. 4):807813.

Wang W, et al. No effect of maternal micronutrient supplementation on early childhood growth in rural western China: 30 month follow-up evaluation of a double blind, cluster randomized controlled trial. Eur. J. Clin. Nutr. 2012;66(2):261268.

Warren MA, Bedi KS. The effects of a lengthy period of undernutrition on food intake and on body and organ growth during rehabilitation. J. Anat. 1985;141:6575.

Webb AL, et al. Maternal and childhood nutrition and later blood pressure levels in young Guatemalan adults. Int. J. Epidemiol. 2005;34(4):898904.

Wehby GL, et al. The effects of prenatal use of folic acid and other dietary supplements on early child development. Matern. Child Health J. 2008;12(2):180187.

Wohlleb JC, et al. The Bacon Chow study: maternal supplementation and infant growth. Early Hum. Dev. 1983;9(1):7991.

Yan X, et al. Up-regulation of Toll-like receptor 4/nuclear factor-kappaB signaling is associated with enhanced adipogenesis and insulin resistance in fetal skeletal muscle of obese sheep at late gestation. Endocrinology. 2010;151(1):380387.

Zambrano E, Nathanielsz PW. Mechanisms by which maternal obesity programs offspring for obesity: evidence from animal studies. Nutr. Rev. 2013;71(Suppl 1):S42S54.

Zamenhof S, van Marthens E. Effects of prenatal and chronic undernutrition on aging and survival in rats. J. Nutr. 1982;112(5):972977.

Zhang S, et al. Maternal obesity and the early origins of childhood obesity: weighing up the benefits and costs of maternal weight loss in the periconceptional period for the offspring. Exp. Diab. Res. 2011;2011:585749.

Zhou SJ, et al. Effect of iron supplementation during pregnancy on the intelligence quotient and behavior of children at 4 y of age: long-term follow-up of a randomized controlled trial. Am. J. Clin. Nutr. 2006;83(5):11121117.

..................Content has been hidden....................

You can't read the all page of ebook, please click here login for view all page.
Reset
13.58.228.178