Abstract
Choline and DHA are nutrients that, when provided during the first 1000 days from conception to age 2 years, may have beneficial effects on child neurodevelopment as well as related health factors, including birth outcomes and child growth, morbidity, and inflammation. Because these nutrients are found mainly in animal-source foods, they may be lacking in the diets of pregnant and lactating women and young children in low- and middle-income countries, potentially putting children at risk for suboptimal development and health. Prior reviews of these nutrients have mainly focused on studies from high-income countries. Here, a narrative review is presented of studies describing the pre- and postnatal roles of choline, docosahexaenoic acid, and a combination of the 2 nutrients on child neurodevelopment, birth outcomes, growth, morbidity, and inflammation in low- and middle-income countries. More studies are needed to understand the specific, long-term effects of perinatal choline and docosahexaenoic acid intake in various contexts.
Keywords: child growth, choline, DHA, neurodevelopment, pregnancy
INTRODUCTION
The period from conception through the first 2 years after birth (termed the “first 1000 days”) is a time of rapid neurodevelopment when developmental trajectories are malleable to contextual exposures, with outcomes negatively affected by risk factors and positively affected by interventions.1 Nearly 250 million children younger than 5 years in low- and middle-income countries (LMICs) are at risk for not reaching their developmental potential , which can have adverse effects on future schooling, productivity, and health.2 Although many environmental conditions influence neurodevelopment, nutrition is a major component.3 Nutrition also affects factors such as preterm birth, childhood stunting, morbidity, and inflammation, which are common in LMICs and linked to impaired neurodevelopment.4 Choline and docosahexaenoic acid (DHA) are 2 nutrients that may influence child neurodevelopment, as well as birth outcomes, growth, morbidity, and inflammation.
Both choline and DHA can be endogenously produced from precursors; however, it is unlikely that endogenous production is sufficient to meet needs, so recommended intake levels have been established for pregnant and lactating women and young children (Table 1).5,6 Except for DHA requirements for women, these recommendations are based on adequate intake levels and may over- or underestimate needs. Choline recommendations, in particular, are based on few studies and do not consider neurodevelopmental outcomes. Whereas the World Health Organization developed the DHA guidelines with the Food and Agriculture Organization of the United Nations, there are no global guidelines for choline intake.
Table 1.
Recommended intake levels for choline and docosahexaenoic acid in pregnant or lactating women and young children
| Choline (mg/d)a | DHAb | |
|---|---|---|
| Pregnant women | 450 | 200 mg/d |
| Lactating women | 550 | 200 mg/d |
| Infants aged 0–6 mo | 125 | 0.10%–0.18% of total energy |
| Infants aged 7–12 mo | 150 | 10–12 mg/kg |
| Children aged 1–2 y | 200 | 10–12 mg/kg |
Abbreviation: DHA,docosahexaenoic acid.
Adequate intake levels set by the United States Institute of Medicine.5
Average nutrient requirement levels (for women) and adequate intake levels (for infants and children) set by the Food and Agriculture Organization of the United Nations, World Health Organization.6
The main food sources of choline and DHA are of animal origin, such as eggs and fish. Because animal source foods may be expensive,7 intake of choline and DHA may be limited in many LMICs. Processed foods may also provide choline as lecithin; as processed food consumption increases in LMICs, this may become a significant source. Breast milk is a rich source of choline and DHA for young children, although concentrations of both nutrients vary on the basis of maternal intake.8,9 Foods contain different forms of choline (free choline, phosphocholine, glycerophosphocholine, phosphatidylcholine, and sphingomyelin); each form should be included in estimates of total choline intake.10
A lack of representative food composition estimates in national food databases limits assessment of dietary intake; however, it seems intake often falls below recommended levels in LMICs. For example, in a review that reported choline intake in 15 countries, intake among women was lowest in Mexico (263 mg/d) and highest in Sweden (374 mg/d).10 Another study in The Gambia reported even lower intakes of choline (155.2 mg/d) among 62 nonpregnant women of reproductive age.11 Intake of DHA below recommendations is also common in LMICs. According to food balance sheets from 175 countries, per capita availability of DHA among low-income countries was 96 mg/day compared with 184–473 mg/day across high-income countries.12 Using similar data plus breastfeeding rates, the median DHA intake among children aged 6–36 months in LMICs was estimated to be 48.7 mg/day, well below recommendations.13 These nutritional inadequacies may put children at risk for suboptimal development and may be exacerbated by other common health factors in LMICS. These include inadequate intake of other nutrients, such as iron, zinc, and vitamin B12, required for endogenous production of DHA and choline,6,14 as well as conditions like gestational diabetes mellitus and an altered composition of the gut microbiota, which may affect DHA and choline availability, respectively.15,16
The relationship between poor intake and outcomes is clouded by limitations in assessing choline and DHA status. Plasma choline is poorly correlated with intake across a range of intake levels,11,17 and may be affected by plasma volume expansion in pregnancy. Lipid-soluble forms of choline (such as phosphatidylcholine) are influenced by fat metabolism and transport in lipoproteins. For DHA, red blood cell (RBC) concentration is a better marker of habitual exposure than is plasma concentration, although sample collection and storage are more difficult.18
Few reviews have examined choline and DHA together. Choline and DHA are present in many of the same food sources and are hypothesized to have similar effects on neurodevelopment, especially memory and learning. Their metabolism is also linked—phosphatidylcholine molecules can incorporate DHA, as we describe later in this article—and there is evidence that these nutrients work synergistically to promote neurodevelopment.19 Several reviews have focused on either choline or DHA, presenting evidence from predominantly high-income countries.20–23 This review presents the evidence relating choline, DHA, and a combination of the 2 nutrients during the first 1000 days of life to children’s neurodevelopment, birth outcomes, growth, morbidity, and inflammation in the context of LMICs.
METHODS
Literature searches were performed in PubMed using the terms: choline, DHA, docosahexaenoic acid, fish, egg; pregnancy, lactation, complementary feeding, formula, infant; child development, neurodevelopment, cognition, memory, vision, visual; child growth, height, weight, head circumference; preterm, birth; morbidity; diarrhea; enteropathy; inflammation; as well as a list of LMICs based on World Bank income categories in 2019–2020. Abstracts and titles were screened for inclusion. Reference lists were scanned for eligible studies.
Selected papers included observational studies or randomized controlled trials (RCTs) in LMICs. Relevant animal studies were included in discussions of mechanisms; however, only human trials were included in discussions of the evidence in LMICs. All types of interventions were eligible, including supplements, foods, or dietary advice to consume foods rich in choline and/or DHA, and studies could include co-interventions, such as arachidonic acid (ARA) or eicosapentaenoic acid (EPA). Studies published in English by August 11, 2020, were eligible.
The outcomes assessed were neurodevelopment (behavioral and physiological measures), physical growth (height, weight, head circumference, measures of adiposity), birth outcomes (preterm birth/gestational age, birth length, birth weight), morbidity (illnesses such as diarrhea, environmental enteropathy), and biochemical markers of inflammation. Visual development was included with neurodevelopmental outcomes, when available. Morbidity and inflammation data are presented together because of the limited number of studies identified. Outcomes could be measured at any age; however, the initial exposure assessment must have been during pregnancy or the first 2 postnatal years. Articles were organized by the nutrient of interest (choline, DHA, or both) and the timing of exposure (prenatal, postnatal, or across both periods).
Choline
Proposed mechanisms
Choline is an essential micronutrient that is important for early neurodevelopment. In rodent studies, clear improvements were observed in lifelong memory when choline was supplemented during specific pre- and postnatal periods,24–26 in part by altering rates of mitosis and apoptosis of neural progenitor cells in the hippocampus and the cerebral cortex.27,28 These effects are epigenetically mediated through conversion of choline to the methyl donor betaine.29 Betaine donates a methyl group to homocysteine to form methionine and eventually S-adenosyl methionine. These epigenetic changes may affect neurodevelopment in indirect ways, as well. For example, high maternal intake of choline decreases placental expression of cortisol-stimulating genes, with potential effects on learning and memory.30 Choline may also affect development in its role as a precursor of phosphatidylcholine and acetylcholine. Phosphatidylcholine is a major component of cell membranes and a precursor of sphingomyelin, required for myelination of neurons, and the cell-signaling molecule diacylglycerol. Acetylcholine is a neurotransmitter involved in the encoding of new memories in the hippocampus; it is also a neuromodulator that influences neurogenesis and synapse formation.31
Compared with neurodevelopment, there is less mechanistic evidence for choline’s role in birth outcomes, child growth, morbidity, and inflammation. In rodent models, prenatal choline supplementation modulates nutrient transport across the placenta, increasing choline availability and altering glucose and amino acid metabolism.32 As a methyl donor, choline may reduce homocysteine levels, which are associated with adverse pregnancy outcomes,33 and increase vitamin B12 availability in pregnant women.34 Perinatal choline may also influence bone growth and body size. Rodent knockout models without the choline kinase enzyme (which converts choline to phosphocholine) have altered bone formation,35,36 and phosphatidylcholine is required for the production of new cell membranes. Related to morbidity and inflammation, choline supplementation in rodents reduced markers of inflammation after lipopolysaccharide administration during pregnancy.37 Different forms of prenatal choline (eg, free choline or phosphatidylcholine) also may affect development of the offspring immune system.38 Choline’s roles outside of neurodevelopment are active areas of research.
Choline during pregnancy
Designs of reviewed studies
Two RCTs in LMICs have been conducted to study prenatal choline supplementation in human populations. In South Africa, heavy alcohol consumers were randomly assigned to choline (2 g/d) or placebo from mid-pregnancy until delivery (n = 69).39 Although baseline plasma choline concentration was not reported, mean choline intake at baseline was below guidelines for pregnant women (∼ 370 mg/d in both groups). In a trial in Ukraine, researchers also examined the effect of choline among women who consumed alcohol during pregnancy (n = 163); however, this trial enrolled abstaining pregnant women, as well (n = 204).40,41 Women were randomly assigned to a daily multiple micronutrient supplement (MMS) with 750 mg of choline, MMS alone, or standard of care (ie, no provision of supplements) from the first prenatal visit until delivery. The subgroups that received choline were small (n = 19 alcohol consumers and n = 18 alcohol abstainers). Baseline plasma choline levels were similar across groups (∼ 15 µmol/L). The primary outcome of both trials was neurodevelopment during the first year of life. One observational study in China reported associations of maternal plasma choline with birth outcomes.42 We found no studies in LMICs that reported on prenatal choline and infant morbidity or inflammation.
Neurodevelopment
In the South African trial, infants in the choline group had improved eye-blink conditioning, an early marker of learning and memory, than did control infants at 6.5 months; however, this was only significant after removing 4 infants in the choline group whose mothers were considered to have poor adherence. The choline group also had significantly higher mean novelty preference scores on the Fagan Test of Infant Intelligence compared with control children at 12 months (64.5% vs 59.1%; P < 0.05), demonstrating improved visual recognition memory. There were no effects on information processing speed at 6.5 or 12 months.39 In the Ukrainian trial, addition of choline to MMS did not significantly affect Bayley Scales of Infant Development (BSID) II Psychomotor Development Index or Mental Development Index scores at 4–11 months.41 However, infants in the choline group demonstrated improved encoding and memory of visual stimuli, as measured by larger and faster changes in heart rate during habituation and dishabituation tasks at 4–11 months.40
Together, the findings from these 2 studies suggest neurodevelopmental benefits in the first year from prenatal supplementation of choline doses from 750 mg–2 g/d, although this may be primarily generalizable to women who consume alcohol during pregnancy. More studies are required with abstaining women in LMICs and with prolonged follow-up to assess the long-term effects of prenatal choline supplementation. Detecting effects of choline may depend on the neurodevelopmental assessment methods used. Assessments of attention and memory based on eye-blink, eye movements, and heart rate may be more sensitive than assessments based on acquisition of developmental milestones, such as the BSID.
Pregnancy outcomes
In the South African trial, there was no difference between groups in mean gestational age (choline, 38.8 weeks vs control, 38.9 weeks) or incidence of low birth weight (LBW) (25.0% vs 32.3%), although mean birth length was nonsignificantly lower in the choline group (47.2 cm [SD, 3.3] vs 48.9 cm [SD, 3.7]; P < 0.1).39 In Ukraine, birth outcomes were compared by maternal supplementation (MMS vs standard of care; MMS with choline vs MMS alone) and alcohol consumption. Children whose mothers received the MMS with or without choline had significantly higher birth weight compared with the standard-of-care control group, a pattern that was evident among those born to women who consumed alcohol during pregnancy and those born to women who abstained. However, when contrasting the group who received MMS plus choline with MMS alone, birth weight was significantly lower (−126 g among mothers who consumed alcohol, and −171 g among abstaining mothers; P = 0.048).40 In an observational study of 115 pregnant women in China, maternal plasma choline was not associated with birth outcomes, although the choline metabolite betaine was inversely associated with birth weight.42
The scant information available suggests additional prenatal choline may be related to smaller birth size; however, this reflects the findings of only 2 small trials that enrolled women who consumed alcohol during pregnancy, neither of which were designed to investigate birth outcomes. Future studies should explore the link between prenatal choline supplementation and birth size in LMICs.
Child growth
In the South African trial, the control group decreased in weight, length, and head circumference z scores over the first year; in contrast, the choline group experienced catch-up growth in weight percentile and head circumference percentile from birth to 12 months.39 No studies reported on growth after prenatal choline supplementation among women who did not consume alcohol.
Choline from birth to 2 years
Designs of reviewed studies
No trials of early postnatal (0–2 years) choline supplementation in LMICs were identified. Three observational studies reported on the association of choline and growth within this life stage in Malawi,43 Brazil,44 and Bangladesh.45 No studies reported on early postnatal choline supplementation and child neurodevelopment, morbidity, or inflammation in LMICs.
Child growth
In a cross-sectional study of 325 Malawian children aged 12–59 months, researchers observed a difference of 0.41 cm in height per 1 SD difference in serum choline (P < 0.0001), with a larger magnitude in boys (0.60 cm) than in girls (0.19 cm).43 Ratios of betaine to choline and trimethylamine N-oxide to choline, representing choline conversion to metabolites, were also investigated; both ratios were negatively associated with children’s height-for-age z scores (HAZ).
In Brazil, urinary metabolites were measured among 326 children age 6–24 months with weight-for-age z scores (WAZ) of less than −2 or greater than −1. Children with a WAZ less than −2 had lower concentrations of urinary choline metabolites, signifying changes in choline metabolism among underweight children.44 In a metabolomics study of 130 Bangladeshi children, sphingomyelins and phosphatidylcholine species were positively associated with change in HAZ from 9 months to 4 years.45 Overall, observational studies in LMICs provide evidence that serum or urinary markers of choline concentration are positively associated with child growth, although stronger study designs must test this connection.
Choline during the first 1000 days
To our knowledge, no trials or observational studies in LMICs have reported on the association between choline intake or plasma choline concentration spanning pre- and postnatal periods and child neurodevelopment, growth, morbidity, or inflammation.
Limitations and future directions
More information is needed regarding the role of perinatal choline in LMICs on all child health outcomes. Both of the reviewed RCTs enrolled alcohol consumers, and neither assessed dose-response relationships or stratified by baseline choline intake, limiting the ability to refine choline intake recommendations. Given that choline’s influence on neurodevelopment is hypothesized to extend from pregnancy through complementary feeding, potentially up to year 4,25 studies of choline supplementation across this period are needed. Studies should also examine the effects of prenatal choline on birth size, because some studies suggest smaller length or weight after prenatal supplementation.
Docosahexaenoic acid
Proposed mechanisms
DHA is a long-chain polyunsaturated fatty acid (LC-PUFA) highly concentrated in brain and retinal tissues, where it influences neural and visual development. In animal models, perinatal supplementation with DHA improved performance on cognitive tests,23 and prenatal deficiency was associated with poorer cognitive performance.46 Comprehensive reviews of DHA’s mechanisms may be found elsewhere.22 Briefly, increased DHA levels promote neural development, including formation of hippocampal synapses.47 Phospholipid-bound DHA in retinal membranes influences visual signaling pathways by interacting with rhodopsin.48 DHA is also a ligand for cell surface receptors such as GPR120, influencing anti-inflammatory cell-signaling pathways,49 and transcription factors, influencing gene expression in the brain.50,51 DHA is a precursor for a myriad of anti-inflammatory metabolites, including resolvins and neuroprotectins52; in producing these metabolites, DHA blocks metabolism of ARA to pro-inflammatory eicosanoids, including prostaglandins and leukotrienes. Because these metabolites have important physiological functions, balance of DHA and ARA during early life seems necessary for optimal development.53
DHA is well known for its anti-inflammatory actions, including creation of anti-inflammatory eicosanoids, decreased production of inflammatory cytokines, and altered cell signaling.54 These changes affect development of immune function in infants, as well.55 Prenatal DHA supplementation is associated with a more mature infant immune system (characterized by improved oral tolerance and a more balanced T-helper cell 1 and T-helper cell 2 response) in humans.56
LC-PUFAs including DHA are associated with longer gestation and larger birth weight,57 perhaps due to altered production of eicosanoids involved in parturition.58 DHA may also promote prenatal growth via changes in gene expression. Changes in methylation of genes related to fetal growth and development (IGF2/H129) were reported after prenatal DHA supplementation, only among preterm infants or overweight mothers.59 It is unclear if these changes in methylation could affect postnatal growth.
DHA during pregnancy
Designs of reviewed studies
Seven trials examined the effects of prenatal DHA supplementation in LMICs. Neurodevelopment and pregnancy outcomes were the primary focus of the trials identified. In Mexico, the Prenatal DHA (Omega-3 Fatty Acid) Supplements on Infant Growth and Development (POSGRAD) trial randomly assigned 1094 women to 400 mg/d algal DHA vs placebo during the second half of pregnancy.60 The authors of this study reported a range of outcomes, and the study was unique in supplementing DHA alone, without other LC-PUFAs. In Bangladesh, 400 women were randomly assigned to consuming fish oil daily (containing 1.2 g DHA, the largest dose among reviewed trials) vs soy oil capsules daily throughout the third trimester, and neurodevelopmental, growth, and birth outcomes were reported.61 Five RCTs reported only pregnancy outcomes, including a trial in China,62 3 trials in Iran,63–65 and 1 in Egypt.66 Of note, the trial by Ostadrahimi et al63 is included in this discussion of pregnancy outcomes because participating pregnant women were provided prenatal supplementation; however, supplementation continued after birth, and further discussion of this trial is included later in the section on pre- and postnatal DHA. This was also the only trial to report DHA status at baseline.
Seven observational studies were identified, including 2 in Mexico (1 reporting only neurodevelopmental outcomes67 and 1 reporting pregnancy outcomes, child growth, and inflammation68) and 5 in India, which only reported birth outcomes.69–73
Neurodevelopment and visual development
In Bangladesh, there was no improvement in infant BSID-II scores at 10 months after maternal supplementation with a large dose of fish oil.61 Similarly, the POSGRAD study in Mexico reported no group differences in brainstem auditory-evoked potentials at 1–3 months, visual-evoked potentials at 3–6 months,74 BSID-II scores at 18 months,75 or McCarthy Scales of Children’s Abilities scores at 5 years.76 However, compared with control children, prenatal DHA supplementation was associated with improved sustained attention at 5 years as measured by the percentage of children scoring < 40 on the omissions subtest of the Conners’ Kiddie Continuous Performance Test (14.4% vs 25.7%; P < 0.0001).76 No association was found between DHA intake during the third trimester and brainstem auditory-evoked potentials at 1–3 months in an observational study of 76 Mexican women.67
The scant evidence suggests little effect of prenatal DHA on neurodevelopment or visual processing in LMICs. Studies with prolonged follow-up are needed to determine if the delayed benefit to attention reported in the POSGRAD trial is consistent across other studies.
Pregnancy outcomes
Of the 7 RCTs in which the effects of prenatal DHA supplementation on birth outcomes were examined, 3 trials reported significant effects (Table 260,61–66,68–73). In Iran, healthy pregnant women receiving fish oil had fewer LBW infants compared with a control group (0% vs 6.7%; P = 0.02)63; however, similar studies among Iranian women with gestational diabetes reported null effects.64,65 In the Mexican POSGRAD study, there were no differences in birth outcomes between groups except after stratification by gravidity. Among primigravid mothers, prenatal DHA supplementation was associated with heavier babies with larger head circumference and lower risk of LBW and intrauterine growth restriction.60 In Egypt, women with asymmetrical intrauterine growth restriction pregnancy, as measured via ultrasound, were given aspirin with or without omega-3 fatty acids for 6 weeks during the third trimester. The omega-3 group had greater estimated fetal weight gain during the intervention and larger birth weight at delivery, compared with those who received aspirin alone.66
Table 2.
Studies describing the role of prenatal DHA on pregnancy outcomes in low- and middle-income countries
| Reference | Participants | Type of exposure | Timing of exposure measurement | Pregnancy outcomes | Results | ||
|---|---|---|---|---|---|---|---|
| Randomized controlled trials | |||||||
| Ali et al (2017)66 | n = 80 pregnant women in Egypt with asymmetric IUGR | Omega-3 capsules (1 g fish oil) + aspirin (n = 40) vs aspirin alone (n = 40) | From gestational wk 28–30, for 6 wk | Gestational age; birth weight | Increased birth weight in the group receiving omega-3 with aspirin (2022, SD 25 vs 2324, SD 19 g, P < 0.01). | ||
| Jamilian et al (2016)64 | n = 56 pregnant women in Iran with gestational diabetes | Omega-3 pearls (120 mg DHA, 180 mg EPA) (n = 27) vs placebo (n = 27) | From gestational wk 24–28, for 6 wk | Gestational age; birth weight, length, head circumference | No significant differences between groups. | ||
| Olsen et al (2019)62 | n = 5118 pregnant women in China | 2 g fish oil (n = 1706) vs 0.5 g fish oil (n = 1695) vs placebo (n = 1717) | Gestational wk 16–24 until gestational week 37 | Gestational age; preterm birth, early preterm birth, early term birth | No significant differences between groups. | ||
| Ostadrahimi et al (2017)63 | n = 150 pregnant women in Iran | Fish oil (120 mg DHA, 180 mg EPA, 400 mg ALA; n = 75) vs liquid-paraffin placebo (n = 75) | Gestational week 20 until delivery | Gestational age; preterm birth; birth weight, length, and head circumference; LBW | Fewer LBW infants in the fish oil group than in the placebo group (0% vs 6.7%; P = 0.02). | ||
| Ramakrishnan et al (2010)60 | n = 1094 pregnant women in Mexico | 400 mg algal DHA capsules (n = 547) vs placebo (n = 547) | Gestational wk 18–22 until delivery | Gestational age; preterm birth; birth weight, length, and head circumference; IUGR | Among primigravid mothers, birth weight was 99.4 g (95%CI, 5.5–193.4g) heavier, head circumference was 0.5 cm (95%CI, 0.1–0.9 cm) larger, and risk of LBW and IUGR were lower in the DHA group. | ||
| Razavi et al (2017)65 | n = 120 pregnant women in Iran with gestational diabetes | Randomized 1:1:1:1 to omega-3 capsules (240 mg DHA, 360 mg EPA), 50 000 IU vitamin D, both, or control | From gestational wk 24–28, for 6 wk | Gestational age; preterm birth; birth weight, length, head and circumference | No significant differences between groups. | ||
| Tofail et al (2006)61 | n = 400 pregnant women in Bangladesh | 4 g fish oil (1.2 g DHA, 1.8 g EPA) (n = 200) vs soy oil placebo (n = 200) | Gestational week 25 until delivery | Gestational age; preterm birth; birth weight, length, head circumference | No significant differences between groups. | ||
| Observational studies | |||||||
| Al-Hinai et al (2018)68 | n = 236 pregnant women in Mexico | Intake of fatty acids |
|
Birth weight and length; gestational age | Second-trimester DHA intake was negatively associated with birth weight (−0.07 kg per SD [95%CI, −0.12, −0.02]) and length (−0.34 cm [95%CI, −0.59, −0.09]). | ||
| Dhobale et al (2011)72 | n = 102 pregnant women in India, categorized into preterm or term | Placental fatty acids | At delivery | Preterm vs term; birth weight, length, head and chest circumference | Placental DHA level was lower in the preterm group than in the term group (2.05 , SD 0.97 g/100 g fatty acids vs 3.19, SD 0.94 g/100 g fatty acids; P < 0.01). | ||
| Kilari et al (2011)73 | n = 235 pregnant women in India, categorized into LBW or NBW | Maternal and umbilical plasma and RBC fatty acids | At delivery | LBW vs NBW | Higher cord plasma DHA levels in LBW group (P = 0.022). Among female infants, lower plasma and RBC DHA levels in the LBW group (P = 0.031). | ||
| Meher et al (2016)70 | n = 111 pregnant women in India, categorized into LBW or NBW | Maternal and umbilical plasma and RBC fatty acids |
|
LBW vs NBW; birth weight, length, head and chest circumference | Positive associations between maternal RBC DHA levels at 16–20 wk and birth weight (r = 0.222; P = 0.025), and maternal RBC DHA at delivery and baby head circumference (r = 0.241; P = 0.027). | ||
| Meher et al (2016)71 | n = 78 pregnant women in India, categorized into LBW or NBW | Placental fatty acids | At delivery | LBW vs NBW; birth weight, length, head and chest circumference; gestational age | Placental DHA level was lower in the LBW group than in the NBW group (2.18, SD 0.56 g/100g fatty acid vs 2.53, SD 0.78 g/100g fatty acid; P = 0.032). Positive association between placental DHA level and birth weight (r = 0.325; P = 0.011). | ||
| Wadhwani et al (2015)69 | n = 109 pregnant women in India | Maternal and umbilical plasma fatty acids |
|
Birth weight, length, head and chest circumference | Positive association between maternal plasma omega-3 fatty acids at 16–20 wk of gestation and baby chest circumference (r = 0.236; P < 0.05). | ||
Abbreviations: ALA, α-linolenic acid; DHA, docosahexaenoic acid; EPA, eicosapentaenoic acid; IUGR, intrauterine growth restriction; LBW, low birth weight; NBW, normal birth weight; RBC, red blood cell.
Of the 6 observational reports, 5 were from prospective studies in India that followed women through pregnancy and delivery. Two reports noted positive associations between maternal plasma or RBC DHA levels and birth size.69,70 Two others reported lower placental DHA in preterm and LBW babies compared with term and normal weight babies,70,72 although in 1 report, researchers found higher cord plasma levels of DHA among LBW newborns.73 Interestingly, an observational study in Mexico found negative associations between second trimester dietary intake of DHA, EPA, and ARA with birth weight and length.68 The authors suggest this may have been due to concomitant intake of toxins like mercury or substitution of fish in place of other animal source foods, rather than a negative effect of DHA itself.68
Overall, evidence suggests a positive effect of prenatal DHA on birth outcomes, especially birth weight, in LMICs. Several trials had relatively small sample sizes (n = 4 studies with n ≤ 150 participants), perhaps limiting the ability to detect differences in preterm birth or gestational age, although the 2 largest trials (n > 1000 participants)60,62 also reported null effects on these outcomes. More research is needed to understand the context in which DHA, with or without other nutrients in fish oil, may affect birth outcomes. Effects may vary on the basis of maternal characteristics such as gravidity and pregnancy risk; these characteristics should be recorded in future studies.
Child growth
Growth outcomes in LMICs were reported in 3 studies. In Bangladesh, mean weight-for-height z scores (WHZ), WAZ, and HAZ at age 10 months were moderately low (−0.6 to −1.3) and not different between intervention and control groups.61 Among primigravid mothers in the POSGRAD study, children in the DHA group were 0.7 cm longer than control children at age 18 months (95%CI, 0.1–1.3; P = 0.02).77 This effect was lost over time, with no differences in child growth between DHA and control groups at 60 months.78 In an observational study in Mexico, maternal intake of DHA, EPA, and ARA during the second trimester was negatively associated with child height and body mass index (BMI) z score at ages 8–14 years.68 In each of these studies, the relationship of DHA with postnatal growth closely mirrored the relationship found with birth size. Possibly, these results simply reflect altered prenatal growth. Additional studies in LMICs may help uncover relationships between DHA and postnatal growth. Considering the opposing effects on linear growth between the 2 Mexican studies, more information is needed on DHA’s effects specific to height.
Morbidity and inflammation
In the POSGRAD trial, Mexican infants whose mothers were supplemented with DHA had fewer cold symptoms at 1 and 3 months than did control infants (respectively: 37.6% vs 44.6%, P < 0.05; and 37.8% vs 44.1%, P < 0.05).79 At 3 months, the DHA group spent 14% less time sick than did the control group.79 Children in the DHA group also had fewer respiratory symptoms through 18 months of age, but only among children whose mothers were atopic.80 In the observational study in Mexico, there was no association between maternal DHA intake and children’s C-reactive protein level or other markers of metabolic risk at 8–14 years.68 More studies in varied contexts are needed to better understand this relationship.
DHA from birth to 2 years
Designs of reviewed studies
Eight trials provided DHA during the early postnatal period (0–2 years) in LMICs. Generally, trials provided DHA either directly to breastfeeding infants or via inclusion in infant formula or total parenteral nutrition (TPN). Currently, DHA is recommended for inclusion in infant formulas81; however, inclusion is not required and may not occur in some LMICs.82 Many trials focused on neurodevelopment or visual development (Table 383,84–96), including 3 from Turkey,84,87,88 and 1 each from Taiwan,86 The Gambia,89 Ethiopia,83 and Egypt.85 Of these trials, 2 provided fish oil directly,83,89 2 provided fish oil via TPN,84,87 and 3 supplemented infant formula with DHA alone88 or DHA with ARA.85,86 Additionally, a trial in Malawi reported on gut permeability and growth after supplementation with a micronutrient powder and fish oil.97 Half of the trials83,85,89,97 reported DHA status at baseline.
Table 3.
Studies describing the role of postnatal DHA level on neurodevelopment and visual development in low- and middle-income countries
| Reference | Participants | Type of exposure | Timing of exposure | Developmental measure(s) | Child’s age at measurement | Results |
|---|---|---|---|---|---|---|
| Randomized controlled trials | ||||||
| Argaw et al (2019)83 | n = 360 mother-child dyads in Ethiopia | Randomized 1:1:1:1 to maternal intervention (fish oil [215 mg DHA, 285 mg EPA]), infant intervention (169 mg DHA, 331 mg EPA), both, or control | Starting at 6–12 mo old, for 1 y |
|
18–24 mo old | No significant difference across groups. |
| Beken et al (2014)84 | n = 80 VLBW preterm infants (< 32 wk gestation) in Turkey | SMOFlipida (n = 40) vs standard lipid emulsion (n = 40) | Birth until weaning from TPN (mean age, 14 d) |
|
Birth until hospital discharge (mean age, 34 d) | Control group had higher odds of retinopathy of prematurity than the group receiving SMOFlipid (OR, 9.1; 95%CI, 1.9–43.8). No difference between groups in need for laser photocoagulation. |
| El-khayat et al (2007)85 | n = 42 term infants in Egypt with WHZ < −2 | Infant formula supplemented with 0.01 g/100 mL DHA, 0.02 g/100 mL ARA (n = 21) vs standard formula (n = 21) | Starting at 6–25 mo old, for 8 wk | BSID II (MDI and PDI scores) |
|
Larger mean change in MDI and PDI scores in the supplementation group than in control group. Positive correlations between plasma DHA level and MDI (r = 0.52) and PDI (r = 0.50; P < 0.05 for both). |
| Fang et al (2005)86 | n = 27 preterm infants (30–37 wk of gestational age) in Taiwan | Infant formula supplemented with 0.05% DHA, 0.10% ARA (n = 16) vs standard formula (n = 11) | Birth until 6 mo old | Visual acuity: visual evoked potentials, Lea grating acuity cards, Hiding Heidi low-contrast “FACE” cards;BSID (MDI and PDI scores) |
|
No significant differences in visual acuity measures between groups. There was a significant difference in MDI and PDI scores between groups via repeated measures ANOVA, with higher scores in the supplemented group. |
| Ozkan et al (2019)87 | n = 89 preterm infants (<32 wk of gestation) in Turkey | SMOFlipida (n = 42) vs standard lipid emulsion (n = 47) | Birth until weaning from TPN (mean age, 13 d) | Retinopathy of prematurity diagnosis | Birth until hospital discharge (mean age not provided) | No significant difference between groups. |
| Unay et al (2004)88 | n = 54 term newborns in Turkey who received formula, and n = 26 breastfeeding control infants | Infant formula supplemented with 0.5g DHA/100g lipids (n = 28) vs standard formula (n = 26) vs breast milk (n = 26) | Birth until 16 wk old | Brainstem auditory evoked potentials (absolute wave and interpeak latencies describe response to auditory stimuli) |
|
All latencies decreased from birth to 16 wk; the group receiving standard formula had smaller decreases than the DHA-supplemented or breastfed groups (P < 0.05 for all). |
| van der Merwe et al (2013)89 | n = 183 infants in The Gambia | Fish oil containing 200 mg DHA, 300 mg EPA (n = 92) vs olive oil placebo (n = 91) | Starting at 3 mo old, for 6 mo |
|
1 yr old | No significant difference between groups. |
| Observational studies | ||||||
| Fahmida et al (2015)90 | n = 240 children of Sasak ethnicity in Indonesia, categorized by genotype |
|
12–17 mo old | BSID II (MDI score) | 12–17 mo old | Genotype was not significantly associated with MDI score; however, the log DHA-to-EPA ratio was associated with MDI score (β = 1.75; 95%CI, 0.08–3.41). |
| Gharehbaghi et al (2020)91 | n = 341 preterm infants (<2000 g, <34 wk of gestational age) in Iran | SMOFlipida vs standard lipid emulsion | Birth until weaning from TPN (mean age, 14 d) | Retinopathy of prematurity diagnosis | Birth until final follow-up (age not provided) | No significant difference between groups. |
| Henjum et al (2018)92 | n = 320 infants in Nepal | Infant RBC fatty acids | 2–11 mo old |
|
5 y old | No significant association between RBC DHA level and neurodevelopmental scores. |
| Krasevec et al (2002)93 | n = 56 infants in Cuba |
|
2 mo old | Visual acuity measured via Teller acuity cards | 2 mo old | No significant associations between visual acuity scores and fatty acid concentrations. |
| Luxwolda et al (2014)94 | n = 97 infants from 3 tribes in Tanzania plus 15 Dutch infant controls |
|
10–20 wk old | General movement quality measured via Assessment of Motor repertoire | 10–20 wk old | Children in the high-fish-intake tribe had improved observed movement patterns compared with Dutch control children; no difference between tribes. RBC-DHA level was associated with observed movement patterns score (β = 0.304; 95%CI, 0.061, 0.547). |
| Marín et al (2000)95 | n = 28 term, moderately underweight (WAZ between −2 and −3) infants in Argentina |
|
45–90 d old | Full-field flash electroretinography (the b-wave latency describes retinal response to light stimuli) | 45–90 d old | Standard formula group had longer b-wave latencies (mean ± SD: 73.8 ± 7.4 ms) compared with LC-PUFA or breast milk groups (52.0 ± 5.4 and 51.3 ± 1.0). Correlation between infant RBC DHA and b-wave latency (r2 = 0.96; P < 0.0001). |
| Unal et al (2018)96 | n = 227 VLBW preterm infants (25–32 wk of gestational age) in Turkey | SMOFlipida vs standard lipid emulsion | Birth until weaning from TPN (mean age, 7 d) | Retinopathy of prematurity diagnosis | Birth until hospital discharge (mean age, 45 d [fish oil group] and 48 d [control group]; P = 0.317) | No significant difference between groups. |
Abbreviations: ANOVA, analysis of variance;ARA, arachidonic acid; BSID, Bayley Scales of Infant Development; DHA, docosahexaenoic acid; EPA, eicosapentaenoic acid; LC-PUFA, long-chain polyunsaturated fatty acid; MDI, Mental Developmental Index; OR, odds ratio; PDI, Psychomotor Developmental Index; TPN, total parenteral nutrition; VLBW, very low birth weight; WAZ, weight-for-age z score; WHZ, weight-for-height z score.
In contrast to standard emulsions, SMOFlipid (Fresenius Kabi) adds fish oil, medium-chain triglycerides, and higher levels of α-tocopherol.
Ten observational studies described DHA in plasma, RBCs, lipid emulsion or breast milk, and neurodevelopment or growth outcomes across a range of LMICs.
Neurodevelopment and visual development
Of 7 studies in which visual development was measured, only 2 reported a significant relationship between DHA and visual development (Table 3). In a trial in Turkey, researchers reported that addition of fish oil to TPN emulsions reduced risk for retinopathy of prematurity among very-low-birth-weight preterm infants.84 However, null results were reported in 3 similar studies in Turkey and Iran of preterm infants receiving TPN.87,91,96 In Argentina, malnourished infants who consumed standard formula had poorer retinal response to light stimuli compared with those who consumed LC-PUFA–supplemented formula or breast milk95; however, the study was small (n = 28), observational, and researchers did not correct for potentially confounding factors, such as socioeconomic status or maternal education. The 1 study involving healthy, term children in Cuba found null associations between plasma, RBC, or breast milk DHA concentrations and visual development.93
Of 8 studies reporting neurodevelopmental outcomes, 3 RCTs and 2 observational studies reported significant results (Table 3). Among RCTs, the 3 that supplemented infant formula with DHA, with or without other fatty acids, reported significant improvements in neurodevelopment.85,86,88 In both RCTs which directly supplemented breastfeeding infants or lactating women, null results were reported.83,89
An observational study in Indonesia found that although genotype of the FADS gene cluster, involved in endogenous production of LC-PUFAs, was not related to the BSID-II Mental Development Index at ages 12–17 months, the plasma DHA-to-EPA ratio was positively associated with this score.90 In Tanzania, RBC DHA was positively associated with movement patterns at 10–20 weeks of age.94 However, there was no association between RBC DHA in infancy and neurodevelopment at 5 years in children in Nepal.92
Overall, there is little evidence that DHA supplementation improves visual or neural development for healthy, breastfeeding children in LMICs. However, benefits to visual development were seen among malnourished or hospitalized infants, and there is supportive evidence for including DHA in infant formula. There may be a relationship between plasma or RBC DHA and neurodevelopment, limited to specific populations or developmental domains.
Child growth
Seven studies in LMICs included measures of child growth. Among trials, changes in body composition and adiposity were commonly noted. In The Gambia, infants who received fish oil had larger mid-upper arm circumference for their age and triceps skinfold thickness for age compared with control infants.89 In Ethiopia, fish oil provision to breastfeeding infants, but not to lactating women, was associated with increased monthly WHZ gains compared with control infants.98 In a Malawian trial, children aged 12–35 months who received micronutrient powder with fish oil gained more weight over 24 weeks than did control children (1.3 kg vs 1.1 kg; P = 0.01).97 There was no difference in linear growth, and no other anthropometrics were reported. In Taiwan, in a trial of preterm infants, researchers found no differences in child height, weight, or head circumference with DHA-supplemented vs traditional formula; no measures of body composition were reported.86
Authors of 3 observational studies have reported a relationship between DHA in serum or breast milk and child height and weight; none included other anthropometric indices. In Malawi, serum DHA and ARA concentrations were positively associated with HAZ among 400 Malawian children aged 12–59 months.99 In a small sample in China (n = 41), breast milk DHA was positively related to postnatal length gain at 1 month (r = 0.83) and 3 months (r = 0.76; P < 0.01 for both) and weight gain at 3 months (r = 0.46; P < 0.05).100 In the Congo and Burkina Faso, children’s monthly weight gain from birth to 5 months was examined in association with breast milk fatty acid content. Monthly weight gain decreased as the ratio of omega-6 fatty acids to omega-3 fatty acids increased until a cutoff of 15:1, at which point weight gain remained at a steady low.101 Although not specific to DHA, it suggests that a substantial intake of omega 3 fatty acids is needed among lactating women with high intake of omega-6 rich oils to optimize child weight gain.
Although authors of observational studies in LMICs have noted links between DHA and child length and weight, in RCTs, effects on body composition and adiposity are more commonly reported. Few trials included supplementation of preterm infants or lactating women. Studies should investigate these populations and include a variety of anthropometric measures.
Morbidity and inflammation
Five studies reported on morbidity or inflammation related to postnatal DHA in LMICs. In Ethiopia, prevalence of inflammation (based on elevated C-reactive protein levels) and morbidity was not different between groups after supplementing lactating women or infants with fish oil vs a control; the authors suggested this finding may have been due to the low prevalence of inflammation and morbidity in this study compared with others.98 In Malawi, all participants had high ratios (> 0.1) of lactulose to mannitol at baseline, reflecting increased gut permeability, and there was no difference among children who received micronutrient powder with or without fish oil for 24 weeks compared with the control group.97 Similarly, in The Gambia, the average ratio of lactulose to mannitol was 0.22, and nearly half of children had elevated C-reactive protein levels. Children who received fish oil had no differences in lactulose-to-mannitol ratio, inflammatory markers, or morbidity, compared with control children.89
In Turkey, there were no changes in pro- or anti-inflammatory cytokine levels among preterm children randomly assigned to receive fish oil vs standard lipids in TPN. However, there was a lower prevalence of bronchopulmonary dysplasia in the fish oil group, and total antioxidant capacity was higher after 7 days, but not 14 days, of treatment.87 In a similar study in Turkey, provision of fish oil did not reduce morbidity or mortality rate, and total antioxidant capacity was higher in the fish oil group than in the control group, but this difference disappeared after treatment ended.96 Overall, there is little support for an effect of postnatal DHA on inflammation, gut permeability, or morbidity in LMICs.
DHA across the first 1000 days
To our knowledge, only 1 study has described pre- and postnatal DHA provision in an LMIC. In Iran, 150 women were randomly assigned to receive fish oil or liquid-paraffin placebo from 20 weeks’ gestation to 30 days postpartum.102 Pregnancy outcomes are reported in Table 3. Developmentally, there were no differences across the 5 domains of the Ages and Stages Questionnaire at 4 or 6 months, except higher communication scores in the fish oil group at 4 months.102 No differences in infant length, weight, or head circumference were noted between groups from birth to 6 months.102 Morbidity and inflammation data were not reported.
Limitations and future directions
The literature from LMICs suggests positive effects of prenatal DHA supplementation on birth weight and morbidity, with a potential delayed benefit to attention at age 5 years. Additionally, studies in LMICs support the addition of DHA to infant formula for improved neurodevelopment. Across life stages, conclusions have been limited by variations in dose, timing, vehicle, context, and co-interventions. The effects of DHA may vary with baseline DHA status; however, few trials reported this information. Many trials also lacked endline measures of status, relying on maternal report or pill counts for adherence data. Future trials should explore pre- and postnatal supplementation, including to preterm infants or lactating women.
Choline and DHA
Proposed mechanisms
Beyond the individual effects of choline and DHA, the 2 may work together to improve neurodevelopment.19 Among malnourished pigs, addition of dietary DHA, methyl donor nutrients including choline, or both attenuated losses in fetal brain weight compared with controls.103 Combined choline and DHA administration decreases brain inflammation104 and oxidative stress105 in mouse models. In fact, these nutrients may work synergistically: offspring of dams supplemented with choline and DHA had more hippocampal neurons than those given either nutrient alone.106
This synergy reflects the interconnected nature of choline and DHA metabolism. Phosphatidylcholine incorporates DHA via the phosphatidylethanolamine N-methyltransferase pathway and is the main carrier of DHA in plasma, including among preterm infants.107 Lysophosphatidylcholine-DHA is the main form of DHA transported into the brain and eye, via the Mfsd2a transporter.108 Maintenance of phosphatidylcholine-DHA levels is important for neural progenitor cell proliferation.109 Additionally, choline and DHA affect each other’s transport and metabolism. Prenatal choline supplementation increases placental transcript abundance of DHA transporters in mice.32 Likewise, DHA increases choline uptake in retinal cells110 and stimulates production of acetylcholine in cultured cholinergic cells.111 When choline and DHA are provided together, circulating levels of each nutrient increase more than when provided separately.112,113 Although this synergy has been linked to improved neurodevelopment, its relationship with birth outcomes, growth, morbidity, and inflammation is unclear.
Choline and DHA during pregnancy
No studies specific to prenatal choline and DHA were identified in LMICs. Several studies of fish or egg intake were identified and included in this review, because fish and eggs are sources of choline and DHA. Although typically studied for its omega-3 fatty acids, fish also contains choline. (Fish oil, on the other hand, does not.) Eggs also provide these nutrients, although the DHA content varies. Both provide other food components as well, including neuroprotective factors like iodine or iron, and toxins like mercury. To understand the unique effects of choline and DHA, studies specific to these nutrients are needed.
Several observational studies have reported a link between maternal fish or egg consumption and birth outcomes in LMICs. Prospective cohort studies in Iran,114,115 Turkey,116 and India117 linked increased fish intake during pregnancy to decreased odds of LBW, although in 1 study in India, the opposite relationship was reported.118 Risk for preterm birth was also inversely related to fish consumption in Iran114,115 and Pakistan.119 Maternal consumption of eggs was positively associated with birth weight in Iran and India.115,118 We found no studies that reported on neurodevelopment, child growth, morbidity, or inflammation.
Choline and DHA from birth to 2 years
Designs of reviewed studies
Although no studies in LMICs have examined postnatal choline and DHA directly, 9 studies investigated foods containing choline and DHA along with other nutrients. In 3 RCTs, supplements fortified with choline, DHA, and other nutrients were compared with traditional supplemental foods and nonsupplement controls in Guinea-Bissau, South Africa, and Cambodia.120–122 In 2 RCTs, researchers examined the provision of 1 egg/day during the early complementary feeding period (6–15 months) vs a nonintervention control in Ecuador (The Lulun Project) and Malawi (The Mazira Project).123,124 One study in China compared the effects of nutrition education, including recommendations to provide daily egg yolks as an infant’s first food vs a nonintervention control on children’s growth.125 Only 1 trial presented baseline measures of choline and DHA status, in a separate article.126 An observational study in Haiti examined neurodevelopment,127 and 2 in India and Zambia studied growth.128,129
Neurodevelopment
In Guinea-Bissau, children younger than 4 years had improved working memory and better cerebral blood flow with consumption of a supplement containing DHA, choline, and other nutrients, compared with a traditional meal, but there was no difference compared with a common fortified food (Corn Soy Blend++).120 No other domains of development were measured. In the South African trial, a fortified, small-quantity, lipid-based nutrient supplement (SQ-LNS-plus) was associated with improved locomotor development, as measured by the Kilifi Developmental Inventory at 12 months, compared with a nonintervention control.121 The standard SQ-LNS was not different from nonintervention control, suggesting the additional nutrients were responsible for these findings. In the Mazira Project in Malawi, daily egg consumption did not affect children’s memory, attention, language, or personal social scores, but there were fewer children with delayed fine-motor development compared with control children (prevalence ratio, 0.59; 95%CI, 0.38–0.91).130 Children’s egg intake was also associated with motor, but not language, development in an observational study of 583 infants in Haiti; other developmental domains were not measured.127
Together, the limited evidence from LMICs suggests a benefit to neurodevelopment, especially motor development, from postnatal intake of choline- and DHA-containing foods. No studies provided these nutrients to lactating women; this may be an area for future research.
Child growth
Eight studies reported on child growth in LMICs (Table 4120–126,128,129,131). In Guinea-Bissau, the fortified supplement was associated with decreased WAZ, BMI for age, fat tissue accretion, and increased lean tissue accretion compared with the corn-soy blend among children younger than 4 years.120 In Cambodia, consumption of a novel ready-to-use supplemental food with choline, DHA, and other nutrients was associated with increased mid-upper arm circumference compared with that of nonintervention control children, but there were no differences in HAZ, WAZ, or weight-for-length z score.122
Table 4.
Studies describing the role of postnatal foods containing choline and DHA on child growth in low- and middle-income countriesa
| Reference | Participants | Type of exposure | Timing of exposure | Growth measure(s) | Timing of measurement | Results |
|---|---|---|---|---|---|---|
| Randomized controlled trials | ||||||
| Borg et al (2020)122 | n = 485 children from 28 clusters in Cambodia | Fish-based RUSF (n = 128) vs CSB++ (n = 123) vs micronutrient powder (n = 107) vs control (n = 127) | Starting at 6–11 mo old, for 6 mo | HAZ, WAZ, WHZ; MUAC |
|
The fish-based RUSF group had higher MUAC (0.04 cm; 95%CI, 0.01–0.06) than control group but was not different from the CSB++ or micronutrient powder groups. |
| Guldan et al (2000)125 | n = 495 children from 4 townships b | Nutrition education including recommendation of egg yolks for infants (n = 250) vs control (n = 245) | 1 y intervention aimed at pregnant women and infants <1 y old | HAZ, WAZ | Endline (measured infants ages 4–12 mo only) | HAZ (−1.32 vs −1.96; P = 0.022) and WAZ (−1.17 vs −1.93; P = 0.004) were higher in the nutrition education townships than in controls, only among 12-mo-old children. |
| Iannotti et al (2017)123 | n = 163 infants in Ecuador | One egg per day (143.6 mg choline, 30 mg DHA c; n = 83) vs control (n = 80) | Starting at 6–9 mo old, for 6 mo | HAZ, WAZ, WHZ, BMI z score; stunting and underweight |
|
The egg group had increased HAZ, WAZ, WHZ, and BMI z score than the control group. Lower prevalence of stunting and underweight in the egg group than in the control group. |
| Roberts et al (2020)120 | n = 1059 children in Guinea-Bissau | “NEWSUP” d (22.1 mg choline, 534 mg omega-3 fatty acids; n = 368) vs CSB++ (n = 350) vs traditional rice meal (n = 341) | Starting at 15 mo to 7 y old, for 23 wk | HAZ, WAZ, BMI z score; MUAC; lean tissue area; fat tissue area |
|
Among children < 4 y old, the group receiving NEWSUP had decreased WAZ, BMI for age, fat tissue area, and increased lean tissue area compared with the corn-soy blend group. Compared with the control, WAZ and MUAC were decreased. |
| Smuts et al (2019)121 | n = 750 infants in South Africa | SQ-LNS–plus (7.8 mg choline, 75 mg DHA; n = 250) vs SQ-LNS (n = 250) vs control (n = 250) | Starting at 6 mo old, for 6 mo | HAZ, WAZ, WHZ; MUAC; head circumference |
|
Compared with control group, the SQ-LNS–plus group had higher HAZ at 8 mo (effect size: 0.11, 95% CI, 0.01–0.22) and 10 mo (0.16; 95%CI, 0.04–0.27), but not 12 mo (0.09; −0.02, 0.21). |
| Stewart et al (2019)124 | n = 660 infants in Malawi | One egg per day (126 mg choline, 40 mg DHA, cn = 331) vs control (n = 329) | Starting at 6–9 mo old, for 6 mo | HAZ, WAZ, WHZ, HCAZ; stunting, underweight, wasted, small head size |
|
No difference in growth between groups except improved HCAZ (adjusted mean difference: 0.12; 95%CI, 0.49–1.42) and lower prevalence of small head size in the egg group compared with the control group. |
| Observational studies | ||||||
| Aguayo et al (2016)129 | n = 2561 children in India | Feeding practices, including consumption of eggs | 0–23 mo old | HAZ; stunting status | 0–23 mo old | Children ages 6–23 mo who did not consume eggs had increased odds of stunting after adjustment (OR, 2.073; 95%CI, 1.191–3.606). |
| Marinda et al (2018)128 | n = 714 children in Zambia | Feeding practices, including consumption of fish | 6–59 mo old | HAZ, WAZ, WHZ | 6–59 mo old | Among children ages 6–23 mo, there was a positive correlation between fish consumption and HAZ (r = 0.139; P = 0.008). |
Abbreviations: CSB, corn-soy blend; DHA, docosahexaenoic acid; HAZ, height-for-age z score; HCAZ, head circumference-for-age z score; MUAC, mid-upper arm circumference; OR, odds ratio; RUSF, ready-to-use supplementary food; SQ-LNS, small-quantity lipid-based nutrient supplements; WAZ, weight-for-age z score; WHZ, weight-for-height z score.
Townships were not randomly selected.
NEWSUP was a novel food supplement fortified with choline, DHA, and other nutrients, including polyphenols, chromium, and molybdenum.
Researchers have noted an effect on linear growth in several trials. In South Africa, HAZ was higher in the SQ-LNS-plus group than in the control group at ages 8 and 10 months, but not 12 months; the standard SQ-LNS group was not different from the control group.121 Large increases in HAZ (effect size: 0.61; 95%CI, 0.45–0.77) and WAZ (0.61; 95%CI, 0.37–0.77), as well as increases in WHZ and BMI for age, were noted after egg provision in Ecuador.123 However, these effects were absent 2 years later, suggesting a longer intervention may be needed to sustain benefits.132 In Malawi, despite a similar study design, no effects on HAZ, WAZ, or WHZ were reported after egg provision, although head circumference for age was larger in the intervention group.124 This difference in response may be due to the high rates of fish consumption in Malawi124; perhaps eggs improve growth only in the absence of choline- and DHA-containing foods in the usual diet. Baseline stunting rates were also lower in the Malawi study (14%) compared with the study in Ecuador (38%).123,124 In China, 12-month-old children in townships where eggs were recommended for child feeding had larger WAZ and HAZ but not WHZ, compared with children in control townships.125 However, these townships were not randomly selected and received additional messages about other health practices, such as breastfeeding. In India and Zambia, nonconsumption of eggs and fish by children aged 6–23 months was associated with increased risk of stunting.128,129 Overall, these studies suggested a beneficial effect of foods containing choline and DHA on child growth in LMICs, albeit perhaps limited to certain contexts.
Morbidity and inflammation
In South Africa, the SQ-LNS-plus group had decreased longitudinal prevalence of fever, coughing, and wheezing, and increased longitudinal prevalence of diarrhea, vomiting, and rashes and sores compared with the control group. These effects were not specific to choline and DHA, because the standard SQ-LNS group had similar results.121 In the Lulun Project in Ecuador, prevalence of diarrhea in the past 7 days was higher in the egg group than in the control group; however, the data were from parental reports, which the authors speculate may have been biased.123 The Mazira Project has not yet reported child morbidity outcomes. No trial has reported on inflammation.
Choline and DHA across the first 1000 days
No trials in LMICs have reported on pre- and postnatal relationships between choline, DHA, and child neurodevelopment, growth, morbidity, or inflammation.
Limitations and future directions
The literature on perinatal choline and DHA in LMICs is sparse, and no studies assessed the effects of choline and DHA independently of other nutrients. When possible, the specific effects of these nutrients, independent of other dietary factors, should be assessed. Postnatal choline and DHA doses were generally below recommendations; however, improvements to neurodevelopment and growth were evident even at these levels. Given these promising findings, more trials in diverse contexts should be prioritized.
DISCUSSION
Overall, limited data suggest improvements in child development, birth outcomes, growth, morbidity, and inflammation related to perinatal provision of choline, DHA, and a combination of the 2 nutrients in LMICs. There is evidence to suggest that supplementation with these nutrients may be beneficial for pregnant and lactating women and young children. However, more research is needed to address the following questions.
What are the specific long-term effects of choline and DHA during early life in LMICs?
Additional studies are required to understand the effects of varying doses of choline and/or DHA on child health in LMICs. Trials should use high-quality physiological measures of child development, such as eye-movement response time and heart rate, and accurate biomarkers. Measures such as eye tracking are feasible in LMICs133 but may require more funding and training than assessments based on acquisition of developmental milestones. Accurate biomarkers of intake and status will be required across all settings for better measurement of exposure and understanding of biological effects. Controlled feeding trials with varying dosages in multiple arms, although challenging, would provide high-quality evidence and are lacking in LMICs. Studies with prolonged follow-up are needed to understand the long-term impacts on health and productivity.
In what settings would choline and DHA supplementation be beneficial?
Although intake of choline and DHA is thought to be low in many LMICs, this is not the case in all settings. Coastal populations may have substantial intake of fish, regardless of income.12 A useful example of this concept is a comparison between the Mazira and the Lulun Projects. Both trials provided eggs to young children in LMICs, but the results on child growth were strikingly different between populations.123,124 The investigators suggested several possible reasons for this contrast, including differences in background fish intake (high intake near Lake Malawi; low intake in highland Ecuador).124 Indeed, in Malawi, breast milk DHA concentrations among women living near the lake are higher than the global average.134 Especially in areas with adequate intake of animal source foods, choline and DHA may not be limiting nutrients for children’s growth and development. There is a need for more information on population choline and DHA status as well as usual dietary intake. Incorporation of choline and DHA into national nutrition monitoring systems and food composition databases is needed to inform future interventions. Databases should include the 5 chemical forms of choline, which may have variable effects on children’s health,38 as well as betaine, a separate dietary component that may have a choline-sparing effect and is worthy of more research.
How might choline and DHA fit into local, sustainable, and affordable diets?
Considering the perinatal benefits of choline and DHA, efforts to increase maternal and infant intake of these nutrients are needed in LMICs. Breast milk is a good source of these nutrients and should be recommended as the only food for infants up to age 6 months; however, the concentrations in breast milk vary by maternal diet,8,9 and complementary food sources of these nutrients are needed after 6 months. The main food sources of these nutrients are often relatively expensive, and there are concerns about sustainability and environmental issues related to their production. Alternative food products, such as biofortified foods, may be needed to meet global maternal and infant needs affordably and sustainably.
Where food sources are unavailable or inappropriate, supplementation is an option. Choline is required and DHA is recommended for inclusion in infant formula,81 and choline is recommended in prenatal supplements,135 but products meeting these recommendations may not be available or affordable in LMICs. Choline is supplemented as choline salts, such as choline bitartrate, or phosphatidylcholine. DHA is often supplemented as either fish oil or algal oil. Krill oil contains DHA linked to phospholipids including phosphatidylcholine and has similar bioavailability to fish oil; however, it is expensive and has similar sustainability constraints.136
CONCLUSION
More research is needed on the role of choline and DHA during the first 1000 days on child outcomes in LMICs. Dose-response trials are necessary to refine nutrient intake requirements, and measures of population status should be incorporated into national nutrition programs. This would enable better monitoring of global dietary adequacy as well as improved formulation of fortified or supplementary foods. At this time, adequate intake of foods rich in choline and DHA should be recommended for pregnant and lactating women and their young children, including breast milk for infants.
Acknowledgments
Author contributions. M.G.B. performed the literature search and drafted the manuscript. C.P.S. and E.L.P. critically reviewed the manuscript. All authors approved the final version of the manuscript.
Funding/support. C.P.S. and E.L.P. are supported by the Gates Foundation (Grant # OPP1169033) and the US Department of Agriculture National Institute of Food and Agriculture, Hatch projects no. CA-D-NTR-2493-H and no. CA-D-NTR-2092-H.
Declaration of interest. None.
References
- 1. Grantham-McGregor S, Cheung YB, Cueto S, et al. ; International Child Development Steering Group. Developmental potential in the first 5 years for children in developing countries. Lancet. 2007;369:60–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Black MM, Walker SP, Fernald LCH, et al. ; Lancet Early Childhood Development Series Steering Committee. Early childhood development coming of age: science through the life course. Lancet. 2017;389:77–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Prado EL, Dewey KG.. Nutrition and brain development in early life. Nutr Rev. 2014;72:267–284. [DOI] [PubMed] [Google Scholar]
- 4. Bhutta ZA, Guerrant RL, Nelson CA.. Neurodevelopment, nutrition, and inflammation: the evolving global child health landscape. Pediatrics 2017;139:S12–S22. [DOI] [PubMed] [Google Scholar]
- 5.Institute of Medicine. Dietary Reference Intakes for Thiamin, Riboflavin, Niacin, Vitamin B6, Folate, Vitamin B12, Pantothenic Acid, Biotin, and Choline. Washington, DC: National Academy Press; 1998. [PubMed]
- 6.Food and Agriculture Organization of the United Nations. Fats and Fatty Acids in Human Nutrition: Report of an Expert Consultation. Vol 91. Rome, Italy: Food and Agriculture Organization of the United Nations; 2010. [PubMed]
- 7. Headey DD, Alderman HH.. The relative caloric prices of healthy and unhealthy foods differ systematically across income levels and continents. J Nutr. 2019;149:2020–2033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Fischer LM, da Costa KA, Galanko J, et al. Choline intake and genetic polymorphisms influence choline metabolite concentrations in human breast milk and plasma. Am J Clin Nutr. 2010;92:336–346. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Jensen CL, Maude M, Anderson RE, et al. Effect of docosahexaenoic acid supplementation of lactating women on the fatty acid composition of breast milk lipids and maternal and infant plasma phospholipids. Am J Clin Nutr. 2000;71:292s–299s. [DOI] [PubMed] [Google Scholar]
- 10. Wiedeman AM, Barr SI, Green TJ, et al. Dietary choline intake: current state of knowledge across the life cycle. Nutrients 2018;10:1513. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Dominguez-Salas P, Moore SE, Cole D, et al. DNA methylation potential: dietary intake and blood concentrations of one-carbon metabolites and cofactors in rural African women. Am J Clin Nutr. 2013;97:1217–1227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Forsyth S, Gautier S, Salem N.. Global estimates of docosahexaenoic acid and arachidonic acid in developing and developed countries. Ann Nutr Metab. 2016;68:258–267. [DOI] [PubMed] [Google Scholar]
- 13. Forsyth S, Gautier S, Salem N.. Estimated dietary intakes of arachidonic acid and docosahexaenoic acid in infants and young children living in developing countries. Ann Nutr Metab. 2016;69:64–74. [DOI] [PubMed] [Google Scholar]
- 14. Stipanuk M, Caudill MA.. Biochemical, Physiological, and Molecular Aspects of Human Nutrition. 3rd ed. St. Louis: Saunders Elsevier; 2012. [Google Scholar]
- 15. Pagán A, Prieto-Sánchez MT, Blanco-Carnero JE, et al. Materno-fetal transfer of docosahexaenoic acid is impaired by gestational diabetes mellitus. Am J Physiol - Endocrinol Metab 2013;305:826–833. [DOI] [PubMed] [Google Scholar]
- 16. Romano KA, Vivas EI, Amador-Noguez D, et al. Intestinal microbiota composition modulates choline bioavailability from diet and accumulation of the proatherogenic metabolite trimethylamine-N-oxide. MBio 2015;6:1–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Abratte CM, Wang W, Li R, et al. Choline status is not a reliable indicator of moderate changes in dietary choline consumption in premenopausal women. J Nutr Biochem. 2009;20:62–69. [DOI] [PubMed] [Google Scholar]
- 18. Brenna JT, Plourde M, Stark KD, et al. Best practices for the design, laboratory analysis, and reporting of trials involving fatty acids. Am J Clin Nutr. 2018;108:211–227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Mun JG, Legette LL, Ikonte CJ, et al. Choline and DHA in maternal and infant nutrition: synergistic implications in brain and eye health. Nutrients 2019;11:1125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Caudill MA. Pre and postnatal health: evidence of increased choline needs. J Am Diet Assoc. 2010;110:1198–1206. [DOI] [PubMed] [Google Scholar]
- 21. Wallace TC. A comprehensive review of eggs, choline, and lutein on cognition across the life-span. J Am Coll Nutr. 2018;37:269–285. [DOI] [PubMed] [Google Scholar]
- 22. Calder PC. Very long-chain n-3 fatty acids and human health: fact, fiction and the future. Proc Nutr Soc. 2018;77:52–72. [DOI] [PubMed] [Google Scholar]
- 23. 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;82:281–295. [DOI] [PubMed] [Google Scholar]
- 24. Meck WH, Williams CL, Cermak JM, et al. Developmental periods of choline sensitivity provide an ontogenetic mechanism for regulating memory capacity and age-related dementia. Front Integr Neurosci 2008;1:1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Zeisel SH. The fetal origins of memory: the role of dietary choline in optimal brain development. J Pediatr. 2006;149:S131–S136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Blusztajn JK, Slack BE, Mellott TJ.. Neuroprotective actions of dietary choline. Nutrients 2017;9:815. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Albright CD, Tsai AY, Friedrich CB, et al. Choline availability alters embryonic development of the hippocampus and septum in the rat. Dev Brain Res. 1999;113:13–20. [DOI] [PubMed] [Google Scholar]
- 28. Wang Y, Surzenko N, Friday WB, et al. Maternal dietary intake of choline in mice regulates development of the cerebral cortex in the offspring. FASEB J. 2016;30:1566–1578. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Zeisel SH. Choline, other methyl-donors and epigenetics. Nutrients 2017;9:445. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Jiang X, Yan J, West AA, et al. Maternal choline intake alters the epigenetic state of fetal cortisol-regulating genes in humans. FASEB J. 2012;26:3563–3574. [DOI] [PubMed] [Google Scholar]
- 31. Haam J, Yakel JL.. Cholinergic modulation of the hippocampal region and memory function. J Neurochem. 2017;142:111–121. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Kwan ST (Cecilia), King JH, Yan J, et al. Maternal choline supplementation modulates placental nutrient transport and metabolism in late gestation of mouse pregnancy. J Nutr 2017;147:2083–2092. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Nwogu CM, Okunade KS, Adenekan MA, et al. Association between maternal serum homocysteine concentrations in early pregnancy and adverse pregnancy outcomes. Ann Afr Med. 2020;19:113–118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. King JH, Kwan ST (Cecilia), Bae S, et al. Maternal choline supplementation alters vitamin B-12 status in human and murine pregnancy. J Nutr Biochem. 2019;72:108210. [DOI] [PubMed] [Google Scholar]
- 35. Li Z, Wu G, Sher RB, et al. Choline kinase beta is required for normal endochondral bone formation. Biochim Biophys Acta. 2014;1840:2112–2122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Kular J, Tickner JC, Pavlos NJ, et al. Choline kinase β mutant mice exhibit reduced phosphocholine, elevated osteoclast activity, and low bone mass. J Biol Chem. 2015;290:1729–1742. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Zhang M, Han X, Bao J, et al. Choline supplementation during pregnancy protects against gestational lipopolysaccharide-induced inflammatory responses. Reprod Sci. 2018;25:74–85. [DOI] [PubMed] [Google Scholar]
- 38. Lewis ED, Richard C, Goruk S, et al. The form of choline in the maternal diet affects immune develpoment in suckled rat offspring. J Nutr. 2015;146:823–830. [DOI] [PubMed] [Google Scholar]
- 39. Jacobson SW, Carter RC, Molteno CD, et al. Efficacy of maternal choline supplementation during pregnancy in mitigating adverse effects of prenatal alcohol exposure on growth and cognitive function: a randomized, double-blind, placebo-controlled clinical trial. Alcohol Clin Exp Res. 2018;42:1327–1341. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Kable JA, Coles CD, Keen CL, et al. The impact of micronutrient supplementation in alcohol-exposed pregnancies on information processing skills in Ukrainian infants. Alcohol. 2015;49:647–656. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Coles CD, Kable JA, Keen CL, et al. ; the CIFASD. Dose and timing of prenatal alcohol exposure and maternal nutritional supplements: developmental effects on 6-month-old infants. Matern Child Health J. 2015;19:2605–2614. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Du Y-F, Wei Y, Yang J, et al. Maternal betaine status, but not that of choline or methionine, is inversely associated with infant birth weight. Br J Nutr. 2019;121:1279–1286. [DOI] [PubMed] [Google Scholar]
- 43. Semba RD, Zhang P, Gonzalez-Freire M, et al. The association of serum choline with linear growth failure in young children from rural Malawi. Am J Clin Nutr. 2016;104:191–197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Mayneris-Perxachs J, Lima AAM, Guerrant RL, et al. Urinary N-methylnicotinamide and β-aminoisobutyric acid predict catch-up growth in undernourished Brazilian children. Sci Rep. 2016;6:19780. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Moreau GB, Ramakrishnan G, Cook HL, et al. Childhood growth and neurocognition are associated with distinct sets of metabolites. EBioMedicine 2019;44:597–606. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Janssen CIF, Zerbi V, Mutsaers MPC, et al. Impact of dietary n-3 polyunsaturated fatty acids on cognition, motor skills and hippocampal neurogenesis in developing C57BL/6J mice. J Nutr Biochem. 2015;26:24–35. [DOI] [PubMed] [Google Scholar]
- 47. Cao D, Kevala K, Kim J, et al. Docosahexaenoic acid promotes hippocampal neuronal development and synaptic function. J Neurochem. 2009;111:510–521. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Salem N, Litman B, Kim H-Y, et al. Mechanisms of action of docosahexaenoic acid in the nervous system. Lipids 2001;36:945–959. [DOI] [PubMed] [Google Scholar]
- 49. Oh DY, Talukdar S, Bae EJ, et al. GPR120 is an omega-3 fatty acid receptor mediating potent anti-inflammatory and insulin sensitizing effects. Cell 2010;142:687–698. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Schmitz G, Ecker J.. The opposing effects of n-3 and n-6 fatty acids. Prog Lipid Res. 2008;47:147–155. [DOI] [PubMed] [Google Scholar]
- 51. Kitajka K, Sinclair AJ, Weisinger RS, et al. Effects of dietary omega-3 polyunsaturated fatty acids on brain gene expression. Proc Natl Acad Sci USA. 2004;101:10931–10936. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Tassoni D, Kaur G, Weisinger RS, et al. The role of eicosanoids in the brain. Asia Pac J Clin Nutr 2008;17:220–228. [PubMed] [Google Scholar]
- 53. Colombo J, Shaddy DJ, Kerling EH, et al. Docosahexaenoic acid (DHA) and arachidonic acid (ARA) balance in developmental outcomes. Prostaglandins Leukot Essent Fatty Acids. 2017;121:52–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Calder PC. Marine omega-3 fatty acids and inflammatory processes: effects, mechanisms and clinical relevance. Biochim Biophys Acta. 2015;1851:469–484. [DOI] [PubMed] [Google Scholar]
- 55. Gottrand F. Long-chain polyunsaturated fatty acids influence the immune system of infants. J Nutr. 2008;138:1807S–1812S. [DOI] [PubMed] [Google Scholar]
- 56. Richard C, Lewis ED, Field CJ.. Evidence for the essentiality of arachidonic and docosahexaenoic acid in the postnatal maternal and infant diet for the development of the infant’s immune system early in life. Appl Physiol Nutr Metab. 2016;41:461–475. [DOI] [PubMed] [Google Scholar]
- 57. Middleton P, Gomersall J, Gould J, et al. Omega-3 fatty acid addition during pregnancy. Cochrane Database Syst Rev. 2018;11:CD003402. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Allen KGD, Harris MA.. The role of n-3 fatty acids in gestation and parturition. Exp Biol Med (Maywood). 2001;226:498–506. [DOI] [PubMed] [Google Scholar]
- 59. Lee H-S, Barraza-Villarreal A, Biessy C, et al. Dietary supplementation with polyunsaturated fatty acid during pregnancy modulates DNA methylation at IGF2/H19 imprinted genes and growth of infants. Physiol Genomics. 2014;46:851–857. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Ramakrishnan U, Stein AD, Parra-Cabrera S, et al. Effects of docosahexaenoic acid supplementation during pregnancy on gestational age and size at birth: randomized, double-blind, placebo-controlled trial in Mexico. Food Nutr Bull. 2010;31:S108–S116. [DOI] [PubMed] [Google Scholar]
- 61. Tofail F, Kabir I, Hamadani JD, et al. Supplementation of fish-oil and soy-oil during pregnancy and psychomotor development of infants. J Heal Popul Nutr 2006;24:48–56. [PubMed] [Google Scholar]
- 62. Olsen SF, Halldorsson TI, Li M, et al. Examining the effect of fish oil supplementation in Chinese pregnant women on gestation duration and risk of preterm delivery. J Nutr. 2019;149:1942–1951. [DOI] [PubMed] [Google Scholar]
- 63. Ostadrahimi A, Mohammad-Alizadeh S, Mirghafourvand M, et al. The effect of fish oil supplementation on maternal and neonatal outcomes: a triple-blind, randomized controlled trial. J Perinat Med 2017;45:1069–1077. [DOI] [PubMed] [Google Scholar]
- 64. Jamilian M, Samimi M, Kolahdooz F, et al. Omega-3 fatty acid supplementation affects pregnancy outcomes in gestational diabetes: a randomized, double-blind, placebo-controlled trial. J Matern Neonatal Med. 2016;29:669–675. [DOI] [PubMed] [Google Scholar]
- 65. Razavi M, Jamilian M, Samimi M, et al. The effects of vitamin D and omega-3 fatty acids co-supplementation on biomarkers of inflammation, oxidative stress and pregnancy outcomes in patients with gestational diabetes. Nutr Metab (Lond). 2017;14:80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Ali MK, Amin ME, Amin AF, et al. Evaluation of the effectiveness of low-dose aspirin and omega 3 in treatment of asymmetrically intrauterine growth restriction: a randomized clinical trial. Eur J Obstet Gynecol Reprod Biol. 2017;210:231–235. [DOI] [PubMed] [Google Scholar]
- 67. Parra-Cabrera S, Moreno-Macias H, Mendez-Ramirez I, et al. Maternal dietary omega fatty acid intake and auditory brainstem-evoked potentials in Mexican infants born at term: cluster analysis. Early Hum Dev. 2008;84:51–57. [DOI] [PubMed] [Google Scholar]
- 68. Al-Hinai M, Baylin A, Tellez-Rojo MM, et al. Maternal intake of omega-3 and omega-6 polyunsaturated fatty acids during mid-pregnancy is inversely associated with linear growth. J Dev Orig Health Dis. 2018;9:432–441. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Wadhwani NS, Pisal HR, Mehendale SS, et al. A prospective study of maternal fatty acids, micronutrients and homocysteine and their association with birth outcome. Matern Child Nutr. 2015;11:559–573. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Meher A, Randhir K, Mehendale S, et al. Maternal fatty acids and their association with birth outcome: a prospective study. PLoS One. 2016;11:e0147359. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Meher AP, Wadhwani N, Randhir K, et al. Placental DHA and mRNA levels of PPARγ and LXRα and their relationship to birth weight. J Clin Lipidol 2016;10:767–774. [DOI] [PubMed] [Google Scholar]
- 72. Dhobale MV, Wadhwani N, Mehendale SS, et al. Reduced levels of placental long chain polyunsaturated fatty acids in preterm deliveries. Prostaglandins Leukot Essent Fatty Acids. 2011;85:149–153. [DOI] [PubMed] [Google Scholar]
- 73. Kilari A, Mehendale S, Dangat K, et al. Associations of long-chain polyunsaturated fatty acid concentrations with birth outcome in term Indian mothers and their neonates. Am J Hum Biol. 2011;23:319–324. [DOI] [PubMed] [Google Scholar]
- 74. Stein AD, Wang M, Rivera JA, et al. Auditory- and visual-evoked potentials in Mexican infants are not affected by maternal supplementation with 400 mg/d docosahexaenoic acid in the second half of pregnancy. J Nutr. 2012;142:1577–1581. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Ramakrishnan U, Stinger A, DiGirolamo AM, et al. Prenatal docosahexaenoic acid supplementation and offspring development at 18 months: randomized controlled trial. PLoS One. 2015;10:e0120065. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Ramakrishnan U, Gonzalez-Casanova I, Schnaas L, et al. Prenatal supplementation with DHA improves attention at 5 y of age: a randomized controlled trial. Am J Clin Nutr. 2016;104:1075–1082. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Stein AD, Wang M, Martorell R, et al. Growth to age 18 months following prenatal supplementation with docosahexaenoic acid differs by maternal gravidity in Mexico. J Nutr. 2011;141:316–320. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Gonzalez-Casanova I, Stein AD, Hao W, et al. Prenatal supplementation with docosahexaenoic acid has no effect on growth through 60 months of age. J Nutr. 2015;145:1330–1334. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Imhoff-Kunsch B, Stein AD, Martorell R, et al. Prenatal docosahexaenoic acid supplementation and infant morbidity: randomized controlled trial. Pediatrics 2011;128:505–512. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Escamilla-Nuñez MC, Barraza-Villarreal A, Hernańdez-Cadena L, et al. Omega-3 fatty acid supplementation during pregnancy and respiratory symptoms in children. Chest 2014;146:373–382. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Codex Alimentarius Commission. Standard for Infant Formula and Formulas for Special Medical Purposes Intended for Infants. Geneva, Switzerland: Food and Agriculture Organization of the United Nations; 2007.
- 82. Lien EL, Richard C, Hoffman DR.. DHA and ARA addition to infant formula: current status and future research directions. Prostaglandins Leukot Essent Fatty Acids. 2018;128:26–40. [DOI] [PubMed] [Google Scholar]
- 83. Argaw A, Huybregts L, Wondafrash M, et al. Neither n-3 long-chain PUFA supplementation of mothers through lactation nor of offspring in a complementary food affects child overall or social-emotional development: a 2 × 2 factorial randomized controlled trial in rural Ethiopia. J Nutr. 2019;149:505–512. [DOI] [PubMed] [Google Scholar]
- 84. Beken S, Dilli D, Fettah ND, et al. The influence of fish-oil lipid emulsions on retinopathy of prematurity in very low birth weight infants: a randomized controlled trial. Early Hum Dev. 2014;90:27–31. [DOI] [PubMed] [Google Scholar]
- 85. El-khayat H, Shaaban S, Emam EK, et al. Cognitive functions in protein-energy malnutrition: in relation to long chain-polyunsaturated fatty acids. Pak J Biol Sci. 2007;10:1773–1781. [DOI] [PubMed] [Google Scholar]
- 86. Fang P-C, Kuo H-K, Huang C-B, et al. The effect of supplementation of docosahexaenoic acid and arachidonic acid on visual acuity and neurodevelopment in larger preterm infants. Chang Gung Med J. 2005;28:708–715. [PubMed] [Google Scholar]
- 87. Ozkan H, Koksal N, Dorum BA, et al. New-generation fish oil and olive oil lipid for prevention of oxidative damage in preterm infants: single center clinical trial at university hospital in Turkey. Pediatr Int. 2019;61:388–392. [DOI] [PubMed] [Google Scholar]
- 88. Unay B, Sarici S, Ulas U, et al. Nutritional effects on auditory brainstem maturation in healthy term infants. Arch Dis Child Fetal Neonatal Ed. 2004;89:177–179. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. van der Merwe LF, Moore SE, Fulford AJ, et al. Long-chain PUFA supplementation in rural African infants: a randomized controlled trial of effects on gut integrity, growth, and cognitive development. Am J Clin Nutr. 2013;97:45–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Fahmida U, Htet MK, Adhiyanto C, et al. Genetic variants of FADS gene cluster, plasma LC-PUFA levels and the association with cognitive function of under-two-year-old Sasaknese Indonesian children. Asia Pac J Clin Nutr. 2015;24:323–328. [DOI] [PubMed] [Google Scholar]
- 91. Gharehbaghi G, Mohagheghi P, Sedaghat A, et al. Parenteral fish-oil lipid emulsions in retinopathy of prematurity: a retrospective comparative study. J Curr Ophthalmol. 2020;32:69–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Henjum S, Kvestad I, Shrestha M, et al. Erythrocyte DHA and AA in infancy is not associated with developmental status and cognitive functioning five years later in Nepalese children. Nutr J. 2018;17:70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Krasevec JM, Jones PJ, Cabrera-Hernandez A, et al. Maternal and infant essential fatty acid status in Havana, Cuba. Am J Clin Nutr. 2002;76:834–844. [DOI] [PubMed] [Google Scholar]
- 94. Luxwolda MF, Kuipers RS, Rudy Boersma E, et al. DHA status is positively related to motor development in breastfed African and Dutch infants. Nutr Neurosci. 2014;17:97–103. [DOI] [PubMed] [Google Scholar]
- 95. Marín MC, Rey GE, Pedersolí LC, et al. Dietary long-chain fatty acids and visual response in malnourished nursing infants. Prostaglandins Leukot Essent Fatty Acids. 2000;63:385–390. [DOI] [PubMed] [Google Scholar]
- 96. Unal S, Demirel N, Erol S, et al. Effects of two different lipid emulsions on morbidities and oxidant stress statuses in preterm infants: an observational study. J Matern Neonatal Med. 2018;31:850–856. [DOI] [PubMed] [Google Scholar]
- 97. Smith HE, Ryan KN, Stephenson KB, et al. Multiple micronutrient supplementation transiently ameliorates environmental enteropathy in Malawian children aged 12-35 months in a randomized controlled clinical trial. J Nutr. 2014;144:2059–2065. [DOI] [PubMed] [Google Scholar]
- 98. Argaw A, Wondafrash M, Bouckaert KP, et al. Effects of n-3 long-chain PUFA supplementation to lactating mothers and their breastfed children on child growth and morbidity: a 2 × 2 factorial randomized controlled trial in rural Ethiopia. Am J Clin Nutr. 2018;107:454–464. [DOI] [PubMed] [Google Scholar]
- 99. Semba RD, Trehan I, Li X, et al. Low serum omega-3 and omega-6 polyunsaturated fatty acids and other metabolites are associated with poor linear growth in young children from rural Malawi. Am J Clin Nutr. 2017;106:1490–1499. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Xiang M, Lei S, Li T, et al. Composition of long chain polyunsaturated fatty acids in human milk and growth of young infants in rural areas of northern China. Acta Paediatr Int J Paediatr. 2007;88:126–131. [DOI] [PubMed] [Google Scholar]
- 101. Rocquelin G, Tapsoba S, Kiffer J, et al. Human milk fatty acids and growth of infants in Brazzaville (The Congo) and Ouagadougou (Burkina Faso). Public Health Nutr. 2003;6:241–247. [DOI] [PubMed] [Google Scholar]
- 102. Ostadrahimi A, Salehi-pourmehr H, Mohammad-Alizadeh-Charandabi S, et al. The effect of perinatal fish oil supplementation on neurodevelopment and growth of infants: a randomized controlled trial. Eur J Nutr. 2018;57:2387–2397. [DOI] [PubMed] [Google Scholar]
- 103. Lima HK, Lin X, Jacobi SK, et al. Supplementation of maternal diets with docosahexaenoic acid and methylating vitamins impact growth and development of fetuses from malnourished gilts. Curr Dev Nutr 2018;2:1–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Fourrier C, Remus-Borel J, Greenhalgh AD, et al. Docosahexaenoic acid-containing choline phospholipid modulates LPS-induced neuroinflammation in vivo and in microglia in vitro. J Neuroinflammation. 2017;14:170. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105. Almeida PMD, Kamath SU, Shenoy PR, et al. Persistent attenuation of brain oxidative stress through aging in perinatal maternal separated rat pups supplemented with choline and docosahexaenoic acid or Clitoria ternatea aqueous root extract. fn. 2018;56:206–214. [DOI] [PubMed] [Google Scholar]
- 106. Thomas Rajarethnem H, Megur Ramakrishna Bhat K, Jc M, et al. Combined supplementation of choline and docosahexaenoic acid during pregnancy enhances neurodevelopment of fetal hippocampus. Neurol Res Int. 2017;2017:1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107. Bernhard W, Maas C, Shunova A, et al. Transport of long-chain polyunsaturated fatty acids in preterm infant plasma is dominated by phosphatidylcholine. Eur J Nutr. 2018;57:2105–2112. [DOI] [PubMed] [Google Scholar]
- 108. Wong BH, Silver DL.. Mfsd2a: a physiologically important lysolipid transporter in the brain and eye. In: Jiang X-C, ed. Lipid Transfer in Lipoprotein Metabolism and Cardiovascular Disease. Singapore: Springer Singapore; 2020:223–234. [DOI] [PubMed] [Google Scholar]
- 109. da Costa KA, Rai KS, Craciunescu CN, et al. Dietary docosahexaenoic acid supplementation modulates hippocampal development in the Pemt-/- mouse. J Biol Chem. 2010;285:1008–1015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Hyman BT, Spector AA.. Choline uptake in cultured human Y79 retinoblastoma cells: effect of polyunsaturated fatty acid compositional modifications. J Neurochem. 1982;38:650–656. [DOI] [PubMed] [Google Scholar]
- 111. Machová E, Málková B, Lisá V, et al. The increase of choline acetyltransferase activity by docosahexaenoic acid in NG108-15 cells grown in serum-free medium is independent of its effect on cell growth. Neurochem Res. 2006;31:1239–1246. [DOI] [PubMed] [Google Scholar]
- 112. van Wijk N, Balvers M, Cansev M, et al. Dietary crude lecithin increases systemic availability of dietary docosahexaenoic acid with combined intake in rats. Lipids 2016;51:833–846. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Bernhard W, Böckmann K, Maas C, et al. Combined choline and DHA supplementation: a randomized controlled trial. Eur J Nutr. 2020;59:729–739. [DOI] [PubMed] [Google Scholar]
- 114. Fereidooni B, Jenabi E.. The use of omega 3 on pregnancy outcomes: a single-center study. J Pak Med Assoc 2014;64:1363–1365. [PubMed] [Google Scholar]
- 115. Akbari Z, Mansourian M, Kelishadi R.. Relationship of the intake of different food groups by pregnant mothers with the birth weight and gestational age: need for public and individual educational programs. J Edu Health Promot. 2015;4:23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. Canda MT, Sezer O, Demir N.. An audit of seafood consumption awareness during pregnancy and its association with maternal and fetal outcomes in a Turkish population. J Obstet Gynaecol. 2011;31:293–297. [DOI] [PubMed] [Google Scholar]
- 117. Muthayya S, Dwarkanath P, Thomas T, et al. The effect of fish and ω-3 LCPUFA intake on low birth weight in Indian pregnant women. Eur J Clin Nutr. 2009;63:340–346. [DOI] [PubMed] [Google Scholar]
- 118. Malhotra N, Upadhyay RP, Bhilwar M, et al. The role of maternal diet and iron-folic acid supplements in influencing birth weight: evidence from India’s National Family Health Survey. J Trop Pediatr. 2014;60:454–460. [DOI] [PubMed] [Google Scholar]
- 119. Baig SA, Khan N, Baqai T, et al. Preterm birth and its associated risk factors. A study at tertiary care hospitals of Karachi, Pakistan. J Pak Med Assoc 2013;63:414–418. [PubMed] [Google Scholar]
- 120. Roberts SB, Franceschini MA, Silver RE, et al. Effects of food supplementation on cognitive function, cerebral blood flow, and nutritional status in young children at risk of undernutrition: randomized controlled trial. Br Med J 2020;370:m2397. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121. Smuts CM, Matsungo TM, Malan L, et al. Effect of small-quantity lipid-based nutrient supplements on growth, psychomotor development, iron status, and morbidity among 6-to 12-mo-old infants in South Africa: a randomized controlled trial. Am J Clin Nutr. 2019;109:55–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122. Borg B, Sok D, Mihrshahi S, et al. Effectiveness of a locally produced ready-to-use supplementary food in preventing growth faltering for children under 2 years in Cambodia: a cluster randomised controlled trial. Matern Child Nutr. 2020;16:e12896. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123. Iannotti LL, Lutter CK, Stewart CP, et al. Eggs in early complementary feeding and child growth: a randomized controlled trial. Pediatrics 2017;140:e20163459. [DOI] [PubMed] [Google Scholar]
- 124. Stewart CP, Caswell B, Iannotti L, et al. The effect of eggs on early child growth in rural Malawi: the Mazira Project randomized controlled trial. Am J Clin Nutr. 2019;110:1026–1033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. Guldan GS, Fan H-C, Ma X, et al. Culturally appropriate nutrition education improves infant feeding and growth in rural Sichuan, China. J Nutr. 2000;130:1204–1211. [DOI] [PubMed] [Google Scholar]
- 126. Iannotti LL, Lutter CK, Waters WF, et al. Eggs early in complementary feeding increase choline pathway biomarkers and DHA: a randomized controlled trial in Ecuador. Am J Clin Nutr. 2017;106:1482–1389. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127. Iannotti L, Jean Louis Dulience S, Wolff P, et al. Nutrition factors predict earlier acquisition of motor and language milestones among young children in Haiti. Acta Paediatr. 2016;105:e406–e411. [DOI] [PubMed] [Google Scholar]
- 128. Marinda PA, Genschick S, Khayeka-Wandabwa C, et al. Dietary diversity determinants and contribution of fish to maternal and under-five nutritional status in Zambia. PLoS One. 2018;13:e0204009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129. Aguayo VM, Nair R, Badgaiyan N, et al. Determinants of stunting and poor linear growth in children under 2 years of age in India: an in-depth analysis of Maharashtra’s comprehensive nutrition survey. Matern Child Nutr. 2016;12:121–140. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130. Prado E, Maleta K, Caswell B, et al. Early child development outcomes of a randomized trial providing one egg per day to children age 6 to 15 months in Malawi. J Nutr. 2020;150:1933–1942. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131. Caswell B, Arnold C, Lutter C, et al. Impacts of an egg intervention on nutrient intake adequacy among young Malawian children. Matern Child Nutr. 2021;17:e13196. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132. Iannotti LL, Chapnick M, Nicholas J, et al. Egg intervention effect on linear growth no longer present after two years. Matern Child Nutr. 2020;16:e12925. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133. Forssman L, Ashorn P, Ashorn U, et al. Eye-tracking-based assessment of cognitive function in low-resource settings. Arch Dis Child. 2017;102:301–302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134. Yakes Jimenez E, Mangani C, Ashorn P, et al. Breast milk from women living near Lake Malawi is high in docosahexaenoic acid and arachidonic acid. Prostaglandins Leukot Essent Fatty Acids. 2015;95:71–78. [DOI] [PubMed] [Google Scholar]
- 135. Berg S. AMA backs global health experts in calling infertility a disease. American Medical Association News; 2017. Available at: https://www.ama-assn.org/delivering-care/public-health/ama-backs-global-health-experts-calling-infertility-disease. Accessed May 31, 2020.
- 136. Kwantes JM, Grundmann O.. A brief review of krill oil history, research, and the commercial market. J Diet suppl. 2015;12:23–35. [DOI] [PubMed] [Google Scholar]
