Next Article in Journal
Advances in E-Health
Next Article in Special Issue
Nutritional Strategies for Childhood Obesity Prevention
Previous Article in Journal
Antiplatelet Effects of PCSK9 Inhibitors in Primary Hypercholesterolemia
Previous Article in Special Issue
Sugar Intake: Are All Children Made of Sugar?
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Personalized Nutrition Approach in Pregnancy and Early Life to Tackle Childhood and Adult Non-Communicable Diseases

by
Shaikha Alabduljabbar
,
Sara Al Zaidan
,
Arun Prasath Lakshmanan
and
Annalisa Terranegra
*
Mother and Child Health Program, Research Department, Sidra Medicine, P.O. Box 26999, Doha, Qatar
*
Author to whom correspondence should be addressed.
Life 2021, 11(6), 467; https://doi.org/10.3390/life11060467
Submission received: 31 March 2021 / Revised: 5 May 2021 / Accepted: 11 May 2021 / Published: 24 May 2021
(This article belongs to the Special Issue Pediatric Nutrition for a Healthy Life)

Abstract

:
The development of childhood and adult non-communicable diseases (NCD) is associated with environmental factors, starting from intrauterine life. A new theory finds the roots of epigenetic programming in parental gametogenesis, continuing during embryo development, fetal life, and finally in post-natal life. Maternal health status and poor nutrition are widely recognized as implications in the onset of childhood and adult diseases. Early nutrition, particularly breastfeeding, also plays a primary role in affecting the health status of an individual later in life. A poor maternal diet during pregnancy and lack of breastfeeding can cause a nutrient deficiency that affects the gut microbiota, and acts as a cofactor for many pathways, impacting the epigenetic controls and transcription of genes involved in the metabolism, angiogenesis, and other pathways, leading to NCDs in adult life. Both maternal and fetal genetic backgrounds also affect nutrient adsorption and functioning at the cellular level. This review discusses the most recent evidence on maternal nutrition and breastfeeding in the development of NCD, the potentiality of the omics technologies in uncovering the molecular mechanisms underlying it, with the future prospective of applying a personalized nutrition approach to prevent and treat NCD from the beginning of fetal life.

1. Introduction

Nutrition plays an important role at all life stages—before and during pregnancy, lactation, infancy, and childhood, as well as during adult life. Maternal nutrition has a major impact on the infant, not only because the nutrient exchange through the placenta and breast milk is involved in fetal and infant growth, but it also plays a role in determining the offspring’s risk of developing non-communicable diseases (NCDs) [1,2,3]. NCDs are defined as non-infectious diseases that progress slowly, but become chronic; they usually require long-term treatment. NCDs include cardiovascular disease (CVD), type 2 diabetes (T2D), metabolic syndrome, etc. [1]. Recent studies have investigated the roles of pregnancy and infancy as the most critical stages that influence the risks of NCDs in childhood and adult life. New nutritional recommendations were developed to reduce the burden of NCDs in future generations [4].
In the last few years, the food science have started to analyze the effects of nutrients and dietary behaviors on cellular functions and gene modulation This new approach is defined as precision nutrition, where genetic background as well as microbiota are taken into account, to understand the response to diet and to single nutrients intake, and to tackle metabolic diseases (e.g., NCDs) [5,6].
Since there is limited evidence for personalized nutrition in pregnancy and early life, this review aims to summarize current data and perspectives on the roles of diet, nutrigenetics, gut microbiome, epigenetics, and transcriptomics in pregnancy and breastfeeding, contributing to develop NCDs in the offspring.

2. Maternal Diet and NCDs

Maternal health status and poor nutrition are widely recognized as implications in the onset of childhood and adult diseases [7]. A poor maternal diet can cause a nutrient deficiency that, as mentioned before, acts as a cofactor for many pathways, impacting the control of genes involved in the metabolism, angiogenesis, cognitive development, and other pathways, leading to chronic diseases in adult life. The mechanism is known as the “window of susceptibility” to nutritional programming in the fetal life [8] and refers to vulnerability of the fetus to environmental factors, such as the maternal diet, affecting health outcomes determined by the timing of the exposures [9]. In the 1980s, the paradigm of the Developmental Origins of Health and Disease (DoHAD) was developed. Utilizing multiple studies from different geographical areas, researchers observed how famine affected the following generations [10]. The Dutch famine resulted in low caloric intake, below 1000 Kcal per day, for a certain period of time. Therefore, researchers investigated whether the maternal diet had a role in the offspring developing NCDs [11]. Researchers examined the long-term consequences of starvation on the fetuses after many years. The Dutch famine data showed that the prevalence of coronary artery disease was significantly higher in fetuses exposed to undernutrition compared to non-exposed fetuses [12]. Ravelli et al. found also that exposure to famine during fetal life resulted in the development of glucose intolerance in adulthood [13]. Moreover, adults who were exposed to the Chinese famine during gestational life showed significantly higher systolic and diastolic blood pressure, hypertension, and a higher risk of developing metabolic syndrome, compared to those who were not exposed to the famine [14]. Type 2 diabetes (T2D) was higher in offspring exposed to the Biafran famine [15] and the Ukraine famine, where the analysis showed that the more severe the exposure, the higher the risk [16]. According to prior studies on human and non-human models, authors showed an association between low maternal protein intake during pregnancy and an increase in the systolic blood pressure in the offspring later in life [17,18,19]. On the opposite side, a study on 569 children showed that infants of mothers who followed the Mediterranean diet during gestation period had lower blood pressure during childhood [20]. This finding is supported by another study that used a rat model fed with a high-fat diet during pregnancy; it showed an association with hypertension in the offspring [21]. Moreover, pre-conceptional obesity was found to be a risk factor, creating adverse metabolic effects in the mother and children [22,23]. An obesogenic diet during pregnancy after the fertilization stage can lead to the fetus acquiring morbidities, such as hyperinsulinemia and hypercholesterolemia, which are related to obesity, and increase the disease vulnerability throughout adult life [24].
Researchers are looking at potential mechanisms that explain the link between maternal nutritional status and offspring developing NCDs. The hypothesis is that adaptations occur in cases of nutrient deficiencies in fetuses, in order to give priority to the vital organs, such as the brain, and to maintain normal growth; therefore, the growth and functioning of the non-vital insulin-sensitive organs (e.g., the liver and pancreas) are compromised [25]. This can happen by blood flow redistribution to the organs, as well as reduction in secreting anabolic hormones, such as insulin, with reduced performance of these organs, which can continue later in life, causing disease [26]. Other theories suggest that the impact can start from the oocyte meiosis process until the baby’s birth. Intake of nutrients in the preconception period could affect the oocyte and later the fetal development, as referring to the recent research from Santangelo et al., who drew attention to the role of polyphenol intake in inhibiting the oocytes apoptosis and follicle atresia during ovarian developing stages [27].

3. Breastfeeding and NCDs

Many studies demonstrated the beneficial effects of breastfeeding in reducing the risk of NCDs. It has been shown that breastmilk plays a protective role against obesity [28,29,30,31] compared to formula milk [32]. This might be due to differences in the components in terms of nutrients and hormones of each type of milk. The protein content is higher in formula milk than in breast milk, while breast milk contains the leptin hormone that is lacking in the formula milk [33]. A study showed an association between the high fat and protein content in baby formula and the high secretion of insulin growth factor-type 1 (IGF-1), which consequently stimulated the adipocytes, resulting in weight gain [34]. According to in vitro studies, leptin that is present in the breast milk could possibly affect the growth factors and prevent the formation of the adipocytes [35,36]. Furthermore, breastfeeding has an impact on the protein and calorie intake [37], secretion of insulin [38], size of adipocyte, as well as maintaining balanced fat reserves [39]. Moreover, breastfeeding is suggested to prevent type 2 diabetes (T2D) in adulthood [40,41,42,43]. Different studies indicated that infants consuming formula milk have increased level of insulin compared to breast-fed infants [38,44,45], leading to a change in glucagon and insulin release, which play a role in early development of insulin resistance and T2D. This phenomenon can be explained again by the different compositions between the two types of milk, but also by the amount of formula milk intake that is usually much higher than breast milk [46]. Furthermore, breastfeeding is associated with a reduction of the main risk factors for cardiovascular diseases in adulthood [43,47], such as a high level of cholesterol [43,48], high blood pressure [43,48], and a low density lipoprotein (LDL) level [40]. In addition, high density lipoprotein (HDL) levels in adulthood can be improved by breastfeeding during infancy [49,50]. Finally, because of reduced sodium content in the breast milk, breast-fed infants are at a lower risk of developing hypertension during adult life [51].
These (and other) studies suggest the importance of nutrition during pregnancy and early life in determining the development of NCDs later in adulthood. The molecular mechanisms underlying these events are still far from being fully clarified. The approach of precision nutrition is to identify the molecular mechanisms underlying the effect of diet on the onset on NCD, and for this purpose, it uses the multi-omics approach, which is able to detect changes at a molecular level. The results from this multilevel analysis will elucidate the specific mechanisms of each nutrient on single tissues, cell types, and genes in a personalized medicine fashion.

4. Omics Technologies Applied to the Precision Nutrition in Pregnancy

The science of precision nutrition is relatively new and is taking advantages of all the recent technical advances in the omics technologies, such as genomics, transcriptomics, epigenomics, and microbiome. Precision nutrition merges the traditional diet and nutritional status analyses with multi-omics, enabling the capacity to define the individual response to diet and a personalized treatment for each single patient other than the “one-size-fits-all” approach traditionally used in diet therapy. In the following sections, we will discuss the current knowledge on single omics applications in pregnancy and offspring health status; in particular, we will focus on studies performed on the gut microbiota, nutrigenetics, epigenetics, and transcriptomics.

4.1. Diet and Gut Microbiota in NCD

The human microbiota refers to the community of 10–100 trillion microorganisms living in different sites of the human body, with a majority in the intestinal tract. The term microbiome refers to the genome of the microbiota [52]. Microbial dysbiosis has been shown to be related to some NCDs, such as obesity and autoimmune disorders [53,54,55,56,57,58]. That is because microbiota was found to be involved in host metabolism regulation, immunological responses, as well as in other physiological pathways [54,58,59,60]. Due to the fact that diet [61,62] and breast-feeding [61,63] are some of the environmental factors influencing the gut microbiota composition and abundance [60], maternal diet effects on offspring developing NCDs might be interfered by the microbiome.

4.1.1. Maternal Diet and Gut Microbiota

The New Hampshire Birth Cohort Study data indicated that infant gut microbiota is influenced by maternal diet. The researchers showed that infant gut Streptococcus and Clostridium neonatale were positively and negatively correlated with maternal fish and seafood consumption, respectively. Moreover, a decrease in abundance of genus Bifidobacterium in the infants’ gut was related to increased consumption of fruit by the mothers during pregnancy; however, the authors suggested that the results can be influenced by the mode of the delivery as well [64]. Another study, conducted on primate models, confirmed the role of the maternal diet on the offspring’s microbiota alterations. Two groups of Macaca fuscata animal mothers were given different diets, one was fed with standard chow (CTD), consisting of 13% fat from soya bean oil, and the other group with a high fat diet (HFD), consisting of 36% fat from different sources. The offspring were delivered vaginally and breastfed until 6 to 7 months, when they either maintained their mothers’ diets or switched to the other diet, ending up with four offspring cohorts. This study data illustrated that persistent microbial dysbiosis was shown in the offspring of mothers fed with HFD during gestation, where Epsilonproteobacteria Campylobacter spp. and Helicobacter spp. were depleted and Firmicutes Ruminococcus and Dialister were enriched [65].
Since there is not yet a clear causal pathway of maternal diet during pregnancy affecting the health of the neonates, it was suggested that microbiome may have an influential role. Metabolic diseases, such as obesity and insulin resistance in children, were found to be associated with maternal obesity and dietary intake, correlating with infant gut microbiota alteration [66]. Hansen et al. examined the effect of feeding non-obese diabetic pregnant mice with gluten-free diets on their offspring. The co-authors found that the female offspring showed a decrease in gut bacterial levels of Cyanobacteria and Deferribacteres, as well as a reduction in diabetes incidence. After 4 and 10 weeks from birth, the offspring presented an increase in Verrucomicrobia, Proteobacteria, and TM7 abundances and these bacteria were reported having a preventive role against diabetes development [67]. Similarly, a maternal low protein diet showed an alteration of the male offspring microbiota in early development in which consistent high Roseburia intestinalis levels were shown; this bacterial taxa was reported to be inversely correlated with atherosclerotic lesions [68], as well as its abundance, was low in T2D patients [55]. Another study conducted on mice described the effects of the maternal diet on the offspring’s intestinal dysbiosis as the pregnant dam mouse model fed with a western-style diet caused high abundance of Clostridiales and adverse outcomes, such as autoimmunity [69]. The mechanism of how the maternal diet alters the infant microbiome is still not clear. However, it is thought that the infant obtains the mother’s microbiome through swallowing the amniotic fluid [70] as some studies have demonstrated the similarity of bacterial taxonomies between the placenta and amniotic fluid, and the oral and meconium of the neonate [71,72,73].

4.1.2. Breastfeeding Effect on Offspring Gut Microbiota

Human milk is another factor that impacts the development and maturation of the microbiome of the newborn, which is closely related to the mother’s milk and skin microbiome [74]. The breast milk prebiotic compounds, like the human milk oligosaccharides (HMO), are metabolized by the infant gut microbiome [75], and indorse bacterial community growth [76]. HMOs were found to a have a protective role in the development of obesity and autoimmune diseases in mouse models [77,78]. Based on Savage et al., the abundance of Bifidobacteria, Lactobacillus, and Clostridia were positively correlated with the duration of breastfeeding [79]; when breastfeeding was compared to formula feeding, it was shown to significantly impact the health of the infant by reducing the risk of developing obesity [80,81], with an increase in the breastfeeding duration [82]. Moreover, breastfeeding showed a protective role against type 1 diabetes acting on the gut microbiome [74,83].
Many of these studies have been conducted on non-human models, showing an association between diet during pregnancy, lactation period, and the microbiota alterations, leading to NCDs in their offspring (summarized in Figure 1). Further investigations are needed to validate these findings in human subjects.

4.2. Nutrigenetics Role in Offspring NCD

4.2.1. Maternal Diet and Nutrigenetics of NCD

The interaction between the mother’s nutrition during pregnancy and her genetics background impacts the fetal nutrient source; hence, these alterations could influence fetal growth and development [84,85]. The nutritional genetics, which is called nutrigenetics, refers to a different response to nutrients and dietary factors due to gene variants [86]. We summarize here—below and in Figure 1—the most recent findings in nutrigenetics. An example of the interaction between genes and nutrients leading to a disease is the genetic variants of the methylenetetrahydrofolate reductase (MTHFR) gene with a single nucleotide polymorphism (SNP), in which a cytosine is substituted with a thymine, resulting in the amino acid change [87]. This SNP reduces the enzyme efficiency causing the accumulation of homocysteine in the plasma and, as a consequence, reducing the bioavailability of folate and vitamin B12 [88]. These two latter nutrients play a very important role in cell proliferation and differentiation, thus low availability or intake of folate and vitamin B12 by pregnant women carrying this SNP could affect the embryogenesis [89]. In addition, disturbances of maternal folate and vitamin B12 levels during pregnancy may contribute to the development of T2D and adiposity after 6 years of age, as stated by Yajnik et al. [90]. Maternal over-nutrition, leading to diabetes and obesity, may affect the function of the placenta and the fetal metabolism by nutrients provided through the placenta. Changes in these nutrients possibly provoke the proadipogenesis effect, as well as hyperinsulinemia in the fetus [91]. High levels of lipids in pregnant women could lead to accumulation of lipids in the placenta [92], and the fetal supply of crucial, long chain fatty acids are affected [93]; thus, impacting the growth and development of the fetus [94]. A genome-wide association study(GWAS) that used the largest number of individuals to identify maternal and fetal variants contributing to gestational weight gain (GWG) found some evidence that maternal SNPs may be a factor of GWG more than fetal SNPs. These preliminary data propose that the relation between GWG and the later outcome of the offspring possibly reflect the role of intrauterine environment [95]. A study on 950 African mother–child pairs, aimed to explore whether the genetic predisposition to adulthood obesity and birthweight was related to the mother’s obesity and whether an unbalanced diet could be a contributing factor. The study found that fetuses who were genetically susceptible to obesity showed a significant negative correlation with birthweight when the mother was also at a high genetic risk of obesity [96]. Changes in birthweight were reported to be linked to nutrients availability in the intrauterine environment [97]. In addition to unhealthy diet intake during pregnancy, polymorphisms in the mother and infant genes were shown to affect fetal health negatively [98]. For instance, Fatty Acid Desaturase 1 (FADS1) and Fatty Acid Desaturase 2 (FADS2) genes encode delta-5 and delta-6 desaturase enzymes, which are involved in the metabolism of polyunsaturated fatty acids (PUFA) [99]. Genetic variations in these genes, in mothers, was associated with a decrease in eicosapentaenoic acid (EPA) and arachidonic acid (AA) levels in the breast milk as well as in the baby blood [100,101]. Abnormal levels of maternal omega-3 and omega-6 fatty acids were reported to be correlated with low birth weight (LBW) [102], and this outcome was linked with CVD [103]. Moreover, abnormal activity of these enzymes affects the glucose metabolism, leading to a direct relation with diabetes risk [104]. Research was conducted on newborns affected by maturity-onset diabetes of the young (MODY) to study the association between birthweight and inherited mutations in the glucokinase gene (GCK) that causes a reduction in the pancreatic beta cells sensing to the glucose molecules. The study discovered that the baby birthweight was higher than the average when the mother carried the GCK–MODY gene mutation, because of the high maternal blood glucose level that activated high insulin secretion. The birthweight was reduced in case the fetus carried the mutation. So, it was suggested that alteration in the birthweight reflected the fetal insulin secretion induced by the fetal genotype as well as by the maternal hyperglycemia and her genotype [105]. Other studies, taking into account the birthweight, were conducted with scope to analyze the correlation between T2D risk loci variants, influencing the secretion of the insulin, and the birthweight. These studies concluded that 2 fetal risk alleles at the CDK5 regulatory subunit-associated protein 1-like 1 gene (CDKAL1) and 2 alleles at hematopoietically expressed homeobox/insulin-degrading enzyme (HHEX/IDE) loci were related to a lower birthweight. Interestingly, fetuses who carried the four risk alleles and had reduced birthweights were the ones whose mothers smoked three cigarettes daily in the last three months of pregnancy [106].

4.2.2. Breastfeeding and Nutrigenetics of NCD

Early nutrition could also contribute to the health of newborns. Human breast milk (HBM) shows a protective influence from developing metabolic disorders later in life even in individuals who are genetically susceptible to these diseases [107]. The peroxisome proliferator-activated receptor-γ (PPARγ2) is a transcription factor that is expressed in fat cells and regulates insulin sensitivity. A study found that adolescents who were not breastfed in their early lives and carried the polymorphism PPARγ2 Pro12Ala, had increased BMI, waist circumference, and skinfold thickness compared to the ones who consumed breast milk, regardless of the breastfeeding duration [107]. Similarly, variations in the PPARγ2 gene were linked to a high risk of developing obesity in adulthood [108]. Since this gene ligand is present in the HBM, it was suggested to reduce the transcriptional activity of PPARγ2 in individuals with the polymorphism [109]. Moreover, PPARγ2 deficiency was found to contribute to increased levels of lipid oxidation enzymes in the lactating mammary gland and high levels of oxidized free fatty acids were observed in the offspring of mothers lacking this gene [110].
The impact of the nutrients on the interaction between metabolism and genetic variability is complex and this can explain the scarcity of studies on the genome and diet interaction in pregnant women and the infant susceptibility to NCD. Further research will give a better view on how and what nutrients taken during the gestation period can be affected by the maternal and fetal genetic background, resulting in a disease status in the children, hence the protection from those diseases. As a “one-size fits-all” approach is not recommended to be applied in the precision medicine revolution, then nutritional recommendations will be more valid if they are based on the patient’s nutrigenetics background.

4.3. Epigenomics Role in Offspring NCD

4.3.1. Maternal Diet and Epigenetics of NCD

It has been proven that unhealthy diet contributes to NCDs and health problems [111,112,113]. However, the mechanisms are still not completely understood and seem to involve also the epigenetic modifications, which by definition affect the gene expression without changing the DNA sequence. Epigenetic changes can be established during fetal development and known to get impacted by the environment including maternal nutrition [114] leading to changes in the phenotype, such as NCD [85] (Figure 1). Diet can influence the epigenetics via different mechanisms, including histones modifications, DNA methylation and microRNA (miRNA). Histones modifications occur in the proteins that are bounded with DNA base pairs (histones) by covalent addition or removal of functional groups that alter the chromatin structure, hence affecting the gene expression [115]. DNA methylation is the epigenetic mechanism that transfers a methyl group onto specific cytosine residues of the DNA chain, modifying the DNA helix structure and affecting the binding of the transcription complex to DNA transcription site [116]. The third mechanism of epigenetics is the miRNA regulation. The miRNAs are 18–25 nucleotides non-coding RNAs that modulate the gene expression by binding to the untranslated regions of the mRNA in order to repress the translation of proteins and deteriorate the mRNA [117].
Nutrients may change the structure of chromatin, as lysine and arginine found in the histone N-terminal tails [118]. Moreover, many nutrients work as methyl-donors (e.g., methionine, choline, folate), directly providing the substrates for the methylation reactions; other nutrients, such as vitamin B2, B6, and B12, are cofactors of the enzymes involved in the methylation and, finally, nutrients (e.g., polyphenols) are able to modulate the function of the methylation enzymes [119]. Thus, many findings from different studies indicate that the susceptibility of fetal epigenetic modifications can be due to maternal diet, among other factors.
McKay et al. found that unbalanced concentrations of nutrients intake during pregnancy affect the offspring DNA methylation. High levels of vitamin B12 in the maternal blood was correlated with the reduction in the total level of DNA methylation of the neonate, whether the elevated concentration of serum vitamin B12 in the newborn correlated with decrease methylation levels of the insulin-like growth factor-binding protein 3 (IGFBP-3) gene, responsible for intrauterine growth [120]. Low maternal folic acid intake, as well as pregnant women exposed to famine, also affect the expression of insulin growth factor 2 (IGF-2) gene in the offspring by modification of the DNA methylation levels [84]. According to an epigenome-wide analysis, methylated regions that existed in insulin receptors, and carnitine aminotransferase genes that were associated with glucose homeostasis and lipid metabolism, were identified in adults exposed to famine in their fetal life [24,121,122]. Methylation cofactors, such as folate and zinc derived from food, play a role in developing NCD in the offspring. Imbalance of maternal folate concentration affects the development of the fetus [123] through homocysteine buildup, which subsequently affects the fetus organs, including renal and cardiovascular systems [124]. Zinc deficiency in the intrauterine life may have an epigenetic impact on the methylation of promoters, leading to immune pathogenesis [125], increasing the risk of developing CVD and renal disorders [126]. The role of long chain polyunsaturated fatty acids (LC-PUFAs) during gestation was investigated in the offspring health. Khot et al. aimed to look into this aspect and its epigenetic mechanism [127]. The investigators found that low maternal intake of LCPUFAs affects the DNA methylation patterns on angiogenic factor genes, which promotes vascular dysregulation that turns out in rising the cardiovascular risk [127].
The effect of maternal undernutrition on the offspring’s cholesterol dysregulation via epigenetic mechanisms was firstly demonstrated by Sohi et al. in 2011 [128]. This study used rat models that were fed either a low protein diet (LPD), which consisted of 8% protein, or a control diet, consisting of 20% protein. Both diets contained the same amount of Kcal. The authors showed evidence that low protein intake in gestation and lactation period had a role in increasing the cholesterol level in offspring at day 21 after birth. This was found to correlate with the cholesterol 7α-hydroxylase (Cyp7a1) transcriptional repression because of post-translational histone modifications of Cyp7a1 promoter. Cyp7a1 is a rate limiting enzyme that converts the cholesterol into bile acids to regulate the cholesterol level in the body [129]. High circulating cholesterol is one of the risk factors of cardiovascular diseases [130,131]. Additionally, the expression of the Jumonji domain-containing demethylase (Jmjd2a) gene in utero decreased as a result of the LPD, which may have contributed to the significant increase in histone H3 trimethylation associated with Cyp7a1 promoter [128]. Similarly, in an experimental rat model, it was shown that maternal protein restriction during pregnancy and lactation led to an impaired glucose tolerance via histone acetylation of the liver X receptor α (LXRα), which regulates the gluconeogenesis in the liver [132]. A similar correlation was also found between maternal protein restriction and histone modifications on the glucose transporter type 4 (GLUT4) gene resulting in the overexpression of GLUT4 in the skeletal muscle of the female offspring [133], as well as histone modifications on the GATA binding protein 6 (GATA6) gene, which is associated with cardiovascular and metabolic diseases in adulthood [134]. In another animal study, the DNA methylation and histone acetylation in the placenta and liver of the fetus were affected by the abnormal levels of cholesterol and lipid induced by the maternal diet, causing lipid accumulation in the fetal liver [135].
Nutrition also shows an effect on the levels of miRNA, impacting its function in modulating gene expression [136]. Of particular interest in our discussion, specific miRNAs are involved in the regulation of metabolic processes, such as insulin signaling and glucose metabolism, so miRNA dysregulation can be leading to NCDs, for instance T2D [137,138,139] and CVD [137,140]. Moreover, it was shown that maternal malnutrition can impact the miRNA of the fetus, resulting in modified proteins, contributing to the development of obesity and diabetes after birth [141]. Particularly, it was shown that nutrients can influence the expression of miRNAs regulating the folate-mediated one carbon metabolism, which has a role in the regulation of homocysteine, methionine, as well as the protein methylation [142]. Studies on animals reported that both maternal overeating and low protein consumption during pregnancy were associated with significant miRNA dysregulation in the offspring tissues, such as liver and heart [141,143]. These miRNAs are used as biomarkers for some chronic diseases [144,145], which are extracted from various samples, including breast milk [137,146]. An animal study reported that high caloric intake by obese ewes during the peri-conception period led to elevation in expression of specific liver miR-29b, miR-103, and miR-107 in the offspring [147]. In previous reports, miR29b expression was found increased in the liver [148], kidney [148], and pancreatic tissues [149,150] of diabetic animals and human models [151,152]. Moreover, a high level of expressed miR-29b was involved in repressing glucose uptake by insulin via the inhibition of Akt activity, which indicated insulin resistance [153]. An increased serum level of miR-29a was seen in T1D children [154]. The miR-103 and miR-107 are involved in regulation of insulin sensitivity [155] and were found to be upregulated in ewes offspring, signifying that those lambs developed insulin signaling dysregulation [156]. Another form of miR-29 is miR-29c, which targets elastin and collagen in the aorta and functions in maintaining its elasticity. MiR-29c was observed being inhibited in rats of mothers who were undernourished in the gestation period. Thus, the offspring vascular contractility was affected and hypertension was developed [157,158]. Reports on baboons showed that having an obesogenic diet before and after pregnancy promoted significant changes in the cardiac miRNA levels, which are possibly related to cardiovascular diseases [159]. In this study, cardiac miRNAs were assessed and eight of them were differentially expressed; 55 of them were upregulated, and the rest were downregulated in baboons who were born from mothers under high fat/high fructose (HFD) diets compared to those born from mothers who had normal diets during gestation. Some of these miRNAs were also reported in human mapping and contributed to CVD [159].
Maternal vitamin intake was shown to play a key role in the health of the offspring in the long-term; moreover, vitamins were found to have an impact on the expression of miRNAs [160]. A study showed that low vitamin D concentration in pregnant women contributed to their children developing chronic diseases via the expression of miRNAs involved in multiple metabolic pathways [161]. Moreover, low level maternal vitamin B12 was associated with alteration of miRNA expression, which are involved in adipogenesis and insulin metabolism that may initiate metabolic disorders in the offspring [162]. Other micronutrients showing similar effects were polyphenols. These are considered as antioxidants and were found to modify the miRNA expression linked to glucose metabolism, insulin signaling, oxidative stress, and inflammation, which may lead to diabetes and CVD [138,163]. Polyphenol sources are mainly fruits and drinks made from plants [164]. Rat models were used in a recent study to show the harmful effects of maternal consumption of flavonoids from grape seeds in downregulating the offspring hepatic miR-33a, which plays a role in cholesterol regulation, promoting CVD [165]. A study conducted on obese women showed that diet restrictions (for these women to lose weight before pregnancy) led to reprogramming the lipid metabolisms of the offspring, in which the infants’ DNA methylations in liver genes switched to healthy expressions.

4.3.2. Breastfeeding and Epigenetics of NCD

Researchers reported the association between long duration of breastfeeding and reduction in obesity risk later in life [166] and other studies demonstrated the impact of breastfeeding duration on the infants’ DNA methylation [167,168]. According to Obermann-Borst et al., the long period of breastfeeding lowered the CpG methylation of the leptin (LEP) gene, which is involved in appetite regulation and fat metabolism, and the LEP CpG methylation, was inversely correlated with BMI of children who were at 17 months of age. The reduced methylation of LEP caused an increase in the expression and concentration of leptin, a hormone that plays a role in programming the metabolic pathways. In addition, leptin is one of the breast milk constituents; it was proposed that it would likely contribute to programming the neuroendocrine system via methylation of the LEP promoter, with a protective effect on childhood obesity [167]. Another study, analyzing the LEP gene, found a significant difference between a child’s growth and the LEP methylation level. The authors observed higher LEP CpG3 methylation when children breastfed for 7 to 9 months and a significant reduction in the weight of the children when they were breastfed for 10 to 12 months [169].
Several studies showed that miRNAs have a crucial function in epigenetic regulation processes as well as intercellular communication [170,171,172]. Moreover, miRNAs are involved in regulation and development of the immune system [173]. Approximately 1400 different miRNAs were identified in breast milk [174,175,176]. Melnik and colleagues found an association between miRNA-148a derived from milk with pancreatic beta-cell differentiation, highlighting that breastfeeding can have a potential protective role against T2D [176]. Human (and some mammals) milk contain abundant levels of miR-125a-5p, which regulate oxysterol-binding protein-related protein (ORP), which has a role in lipid metabolism [177]. It was observed that highly expressed miRNA in human breast milk is involved in production and homeostasis of triglycerides and some were linked to regulation of fatty acid biosynthesis genes [173]. The authors of the aforementioned findings also reported other miRNAs that are involved in regulating lactose synthesis, which occurs in the mammary gland, more specifically, regulating the UDP-glucose transporter, as well as the UDP-galactose transporter [173]. There is very limited evidence on maternal dietary patterns affecting the nutritional content of breast milk [178]. According to a study conducted in nursing rats, it reported that an obesogenic diet may lead to changes in specific miRNA levels in the breast milk. Compared to controls, there was an increase in miR-222 concentration and a decrease in miR-200 and miR-26 in those obesogenic diet-fed rats. The study researchers also mentioned that this type of diet changes the levels of bioactive proteins found in the milk, such as increasing leptin and adiponectin concentrations and decreasing irisin levels, which, consequently, influence the offspring’s metabolism [179]. Another study conducted on humans indicated that maternal nutritional status and diet could affect the expression of miRNA present in breast milk. Findings by Zamanillo et al. showed that leptin, adiponectin, and miRNAs decreased throughout the lactation period in normal weight mothers, while they were altered in the overweight/obese mothers. Moreover, a negative correlation was observed between milk miRNA expression and leptin or adiponectin levels in normal-weight mothers, while there was no correlation observed between those in the overweight/obese mothers. Furthermore, the BMI of infants of normal-weight mothers was negatively correlated, with the miRNAs were miR-103, miR-17, miR-181a, miR-222, miR-let7c, and miR-146b [180].
These findings emphasize the role of epigenetics and nutrition in influencing the metabolism of one’s offspring; the findings also show how these epigenetic modifications are reversible, which can be used in disease treatment and prevention. However, more research needs to be done in humans, as the data scarcity can be due to difficult clinical applications on pregnant women to ensure safety [84].

4.4. Nutri-Transcriptomics Role in Offspring NCD

Likewise, the genetic variants and epigenetics are involved in the health outcomes of the offspring due to the nutritional status of the mother; the proteins being translated by those genes can have a crucial role in developing a disease. This concept is called nutri-transcriptomics, as the transcription of the gene depends on the diet, and how nutrients and nutritional status impact the gene expression at the level of mRNA [181].

4.4.1. Maternal Diet and Nutri-Transcriptomics of NCD

A trial study by Al-Garawi et al. investigated the role of vitamin D on gene expression during pregnancy. Thirty pregnant women enrolled in the study were supplemented with vitamin D, and their blood samples were collected in the first and third trimesters. Transcriptional profiles revealed that 5839 genes were significantly differentially expressed, and 14 of them exhibited significant correlation with maternal vitamin D concentrations. Their data suggest that alterations of the gene expression occur during pregnancy and this could be associated with vitamin D supplementation, which contributes toward increasing the concentration of the circulating 25(OH)D precursor [182]. Another study confirming this concept proposed that changes in the gene expression are directly induced by maternal vitamin D intake, which can have an effect on the birth outcome [183]. Other observational studies found that low administration of vitamin D during pregnancy might cause preeclampsia. A study was conducted on female mice models that were divided into two groups, one deficient in vitamin D and the other sufficient with vitamin D; then they were bred with vitamin D-sufficient males. The pregnant mice with low vitamin D concentrations presented high systolic and diastolic blood pressure, and it was continued until 7 days after delivery. Moreover, according to their kidney function analyses, an increase in the mRNA expression for renin and angiotensin II receptors was shown and related to the vitamin deficiency. Surprisingly, when the vitamin D was re-supplemented to the deficient models, it reversed the effect [184]. This was supported by another randomized controlled trial conducted on human pregnant subjects who were supplemented with vitamin D daily throughout their pregnancies. They were classified into two groups, whose subjects with circulating 25(OH)D below 100 nmoles are considered deficient, and above that level were considered sufficient. Their placental mRNAs were analyzed and showed that soluble FMS-like tyrosine kinase 1 (sFlt-1) and vascular endothelial growth factor (VEGF) genes, which have critical functions in the angiogenesis pathway, were significantly downregulated in women who had higher intake of vitamin D (≥100 ng/mL). Thus, maternal vitamin D supplementation may have an impact on the placental gene transcription by downregulation of antiangiogenic factors that can contribute to vascular complications in pregnancy, such as preeclampsia [185]. The relation between an increase of sFlt-1 and preeclampsia was well addressed in many research studies [186,187,188,189]. It was reported that children of preeclamptic mothers were more prone to develop endocrine and metabolic diseases in their first five years of age [190]. In addition, body mass index (BMI) was found higher in males born with preeclamptic mothers compared to those of normotensive pregnancies [191]. Besides this finding, studies on metabolic and inflammatory factors found that the lipid profile differed, and the level of tumor necrosis factor alpha (TNF-α) was higher in the cord blood of children of preeclamptic mothers [192,193,194]. Furthermore, high levels of triglycerides and leptin levels were presented in preeclampsia-exposed children who measured low on the Quantitative Insulin Sensitivity Check Index (QUICKI), which is an assessment used to calculate the insulin resistance degree and the secretory capacity of the pancreatic beta cells [195]. Low levels of QUICKI indicate highly insulin resistance, which is associated with obesity risk and cardiovascular diseases [196].
Animal models were mostly used to study the association between nutrition and transcriptomic background. A significant reduction of hypoxia-inducible factor 1-alpha (HIF-1α) levels was seen in baboon fetuses of HFD-fed mothers, as this factor is known to be crucial for cardiovascular system development [197,198]. Therefore, a reduced level of HIF-1α could precede to abnormal cardiac function during fetal and post-fetal life [159]. In another animal study, two groups of pregnant female mice were assigned to two different diets during conception and lactation periods: one with a normal chow diet (NCD) and the other with an isocaloric low protein diet (LPD). Their findings demonstrated that offspring born from LPD-fed mothers exhibited LBW, glucose intolerance, and a decrease in insulin secretion. The same animals also expressed some of miRNAs at weaning stage, and all of these expressed miRNAs were involved in inflammatory pathways with a high levels of serum pro-inflammatory IL-6 and TNF-α cytokines. In addition, the mRNA and protein expression of these cytokines was a significant high level in those offspring. Hence, these results illustrate that consuming low protein levels during pregnancy affects miRNA expression, which might be linked to chronic inflammation and glucose intolerance in offspring [199].
A study on pigs focused on nutrient intake during pregnancy and its influence on the offspring developing metabolic problems. Cai et al. observed that pig mothers who consumed betaine while they were pregnant gave birth to newborn pigs who presented elevated serum and hepatic betaine contents, with a significant upregulation of the hepatic enzyme glycine N-methyltransferase (GNMT). Moreover, liver cholesterol was higher and the expression of the cholesterol metabolic genes was altered in the neonate piglets. Cholesterol homeostasis is maintained through certain factors, such as 3-hydroxy-3-methylglutaryl CoA reductase (HMGCR) and sterol regulatory element-binding protein-2 (SREBP2) were both downregulated at mRNA level. The enzymes cholesterol-27α-hydroxylase (CYP27α1) and cholesterol-7α-hydroxylase (CYP7α1), which function in bile acid transformation, were upregulated at the mRNA levels [200]. Moreover, the same author conducted another study on the role of betaine in galactose metabolism of piglets. The study reported that neonate piglets exposed to betaine in their fetal lives showed low concentrations of galactose in their blood, which was correlated with a significant downregulation of the galactokinase-1 (GALK1) hepatic gene, involved in galactose breakdown process. This enzyme deficiency was associated with increase galactose in the newborn’s blood, as galactose can pass freely from the mother through the placenta to the fetus, and its elevated level causes liver toxicity. Thus, as betaine is involved in galactose and cholesterol homeostasis, and its deficiency could be associated with lipid disorder and diabetes, this study suggests that betaine can play an effective role in the offspring’s health via genetics, transcriptomics, and epigenetics [201].

4.4.2. Breastfeeding and Nutri-Transcriptomics of NCD

Few research studies have investigated the effects of breastfeeding in nutri-transcriptomics and its role in NCD development. Cheshmeh et al., in a case-control study involving 150 infants, aged between 5 and 6 months, investigated the impact of breastfeeding and formula feeding on the expression of obesity-related genes, which are fat mass and obesity-associated (FTO), carnitine palmitoyltransferase 1A (CPT1A), and peroxisome proliferator-activated receptor-α (PPAR-α). The subjects were categorized into three groups: breastfed only, formula-fed only, and fed with both milks. The study findings showed that infants fed formula milk in either groups exhibited higher weight as well as higher expression of FTO and CPT1A genes, and a lower expression of PPAR-α gene when compared to the exclusive breastfeeding group. The authors concluded that breastfeeding apparently showed a protective effect against obesity, modulating the expression of obesity-related genes [202]. A parallel, multicentric study measured the expression levels of PPARα and CPT1A genes, together with other genes, such as solute carrier family 27 member 2 (SLC27A2), fatty acid synthase (FASN), insulin receptor (INSR), and leptin receptor (LEPR), which are used as transcriptional biomarkers of the metabolic status in children from 2 to 9 years old from eight different European countries. The children who were breastfed showed higher expressions of SLC27A2, FASN, PPARα, and INSR, and were at lower risk to develop obesity. On the other hand, an increase in triglycerides levels was shown in formula-fed children who also had low expression of these genes. According to these data, higher expressions of SLC27A2, FASN, PPARα, and INSR genes in the children’s blood reflected a protective role of breastfeeding, as these genes are indicators of a lower risk of developing insulin resistance and dyslipidemia linked with obesity in children. In addition, these biomarkers are likely to distinguish—among formula-fed children—the ones that are at a high risk of metabolic changes [203].
Thus, transcriptomics is very helpful in terms of identifying the genes that express RNAs that have been modulated by diet, specifically nutrients taken during pregnancy and breastfeeding, and then correlate the physiological and pathological long-term effects on the offspring. The main studies are summarized in Figure 1.

5. Conclusions

Maternal diet and nutritional status, as well as single nutrient intake during the entire course of pregnancy and breastfeeding, were demonstrated to affect fetal molecular pathways, such as lipid and liver profiles, inflammation, and angiogenesis, which contribute to the development of NCDs in childhood and adult life. These pathways are regulated at multiple levels via the gut microbiota, through epigenetic modifications, ending with affecting gene expression and protein function (Figure 2). The application of omics technologies to study the role of maternal diet and breastfeeding on the offspring’s health status is able to detect the modifications at a molecular level. However, more studies are needed to confirm the current findings; moreover, it is recommended to move to an integrated multi-omics approach that is able to dissect the multiple interactions among nutrients, microbiota metabolites, and the mutual effects on genes modifications and expression. Moreover, population-specific and patient-personalized studies will contribute to the development of a personalized nutrition approach to prevent and treat NCD from the fetal life stage.

Author Contributions

A.T. created the review content, Figure 2, and reviewed the draft; S.A. and S.A.Z. wrote the manuscript; S.A. created Figure 1; A.P.L. reviewed and edited the draft. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Sidra Medicine, project # SDR400132.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Acknowledgments

We acknowledged Sidra Medicine for funding the publication.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Brumana, L.; Arroyo, A.; Schwalbe, N.R.; Lehtimaki, S.; Hipgrave, D.B. Maternal and child health services and an integrated, life-cycle approach to the prevention of non-communicable diseases. BMJ Glob. Heal. 2017, 2, e000295. [Google Scholar] [CrossRef] [Green Version]
  2. Jacob, C.M.; Newell, M.; Hanson, M. Narrative review of reviews of preconception interventions to prevent an increased risk of obesity and non-communicable diseases in children. Obes. Rev. 2019, 20, 5–17. [Google Scholar] [CrossRef] [PubMed]
  3. Hanson, M.; Godfrey, K.M.; Lillycrop, K.A.; Burdge, G.C.; Gluckman, P.D. Developmental plasticity and developmental origins of non-communicable disease: Theoretical considerations and epigenetic mechanisms. Prog. Biophys. Mol. Biol. 2011, 106, 272–280. [Google Scholar] [CrossRef] [PubMed]
  4. Koletzko, B.; Godfrey, K.M.; Poston, L.; Szajewska, H.; Van Goudoever, J.B.; De Waard, M.; Brands, B.; Grivell, R.M.; Deussen, A.R.; Dodd, J.M.; et al. Nutrition During Pregnancy, Lactation and Early Childhood and its Implications for Maternal and Long-Term Child Health: The Early Nutrition Project Recommendations. Ann. Nutr. Metab. 2019, 74, 93–106. [Google Scholar] [CrossRef] [PubMed]
  5. Bashiardes, S.; Godneva, A.; Elinav, E.; Segal, E. Towards utilization of the human genome and microbiome for personalized nutrition. Curr. Opin. Biotechnol. 2018, 51, 57–63. [Google Scholar] [CrossRef] [PubMed]
  6. Toro-Martín, D.; Arsenault, B.J.; Despres, J.-P.; Vohl, M.-C. Precision nutrition: A review of personalized nutritional approaches for the prevention and management of metabolic syndrome. Nutrients 2017, 9, 913. [Google Scholar] [CrossRef] [Green Version]
  7. Gluckman, P.D.; Hanson, M.A.; Cooper, C.; Thornburg, K.L. Effect of In Utero and Early-Life Conditions on Adult Health and Disease. N. Engl. J. Med. 2008, 359, 61–73. [Google Scholar] [CrossRef] [Green Version]
  8. Randunu, R.S.; Bertolo, R.F. The Effects of Maternal and Postnatal Dietary Methyl Nutrients on Epigenetic Changes that Lead to Non-Communicable Diseases in Adulthood. Int. J. Mol. Sci. 2020, 21, 3290. [Google Scholar] [CrossRef]
  9. Wright, R.O. Environment, susceptibility windows, development, and child health. Curr. Opin. Pediatr. 2017, 29, 211–217. [Google Scholar] [CrossRef] [Green Version]
  10. Barker, D.J.P. The origins of the developmental origins theory. J. Intern. Med. 2007, 261, 412–417. [Google Scholar] [CrossRef]
  11. Heindel, J.J.; Vandenberg, L.N. Developmental origins of health and disease: A paradigm for understanding disease etiology and prevention. Curr. Opin. Pediatr. 2015, 27, 248. [Google Scholar] [CrossRef] [Green Version]
  12. Roseboom, T.J.; Van Der Meulen, J.H.P.; Osmond, C.; Barker, D.J.P.; Ravelli, A.C.J.; Schroeder-Tanka, J.M.; Van Montfrans, G.A.; Michels, R.P.J.; Bleker, O.P. Coronary heart disease after prenatal exposure to the Dutch famine, 1944. Hear 2000, 84, 595–598. [Google Scholar] [CrossRef] [Green Version]
  13. Ravelli, A.; van der Meulen, J.; Michels, R.; Osmond, C.; Barker, D.; Hales, C.; Bleker, O. Glucose tolerance in adults after prenatal exposure to famine. Lancet 1998, 351, 173–177. [Google Scholar] [CrossRef]
  14. Li, Y.; Jaddoe, V.W.; Qi, L.; He, Y.; Wang, D.; Lai, J.; Zhang, J.; Fu, P.; Yang, X.; Hu, F.B. Exposure to the Chinese Famine in Early Life and the Risk of Metabolic Syndrome in Adulthood. Diabetes Care 2011, 34, 1014–1018. [Google Scholar] [CrossRef] [Green Version]
  15. Hult, M.; Tornhammar, P.; Ueda, P.; Chima, C.; Bonamy, A.-K.E.; Ozumba, B.; Norman, M. Hypertension, Diabetes and Overweight: Looming Legacies of the Biafran Famine. PLoS ONE 2010, 5, e13582. [Google Scholar] [CrossRef] [Green Version]
  16. Lumey, L.H.; Khalangot, M.D.; Vaiserman, A.M. Association between type 2 diabetes and prenatal exposure to the Ukraine famine of 1932–33: A retrospective cohort study. Lancet Diabetes Endocrinol. 2015, 3, 787–794. [Google Scholar] [CrossRef]
  17. Langley-Evans, S.C.; Gardner, D.S.; Welham, S.J. Intrauterine Programming of Cardiovascular Disease by Maternal Nutritional Status. Nutrition 1998, 14, 39–47. [Google Scholar] [CrossRef]
  18. Seckl, J.R. Glucocorticoids, feto-placental 11β-hydroxysteroid dehydrogenase type 2, and the early life origins of adult disease. Steroids 1997, 62, 89–94. [Google Scholar] [CrossRef]
  19. Langley, S.C.; Jackson, A.A. Increased Systolic Blood Pressure in Adult Rats Induced by Fetal Exposure to Maternal Low Protein Diets. Clin. Sci. 1994, 86, 217–222. [Google Scholar] [CrossRef] [Green Version]
  20. Chatzi, L.; Rifas-Shiman, S.L.; Georgiou, V.; Joung, K.E.; Koinaki, S.; Chalkiadaki, G.; Margioris, A.; Sarri, K.; Vassilaki, M.; Vafeiadi, M.; et al. Adherence to the Mediterranean diet during pregnancy and offspring adiposity and cardiometabolic traits in childhood. Pediatr. Obes. 2017, 12, 47–56. [Google Scholar] [CrossRef] [Green Version]
  21. Guberman, C.; Jellyman, J.K.; Han, G.; Ross, M.G.; Desai, M. Maternal high-fat diet programs rat offspring hypertension and activates the adipose renin-angiotensin system. Am. J. Obs. Gynecol. 2013, 209, 262.e1–262.e8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Dubé, E.; Gravel, A.; Martin, C.; Desparois, G.; Moussa, I.; Ethier-Chiasson, M.; Forest, J.-C.; Giguère, Y.; Masse, A.; Lafond, J. Modulation of Fatty Acid Transport and Metabolism by Maternal Obesity in the Human Full-Term Placentabiol. Biol. Reprod. 2012, 87, 14. [Google Scholar] [CrossRef] [PubMed]
  23. Gázquez, A.; Uhl, O.; Ruíz-Palacios, M.; Gill, C.; Patel, N.; Koletzko, B.; Poston, L.; Larqué, E. Placental lipid droplet composition: Effect of a lifestyle intervention (UPBEAT) in obese pregnant women. Biochim. Biophys. Acta Mol. Cell Biol. Lipids 2018, 1863, 998–1005. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Green, L.R.; Hester, R.L. Parental Obesity: Intergenerational Programming and Consequences; Springer: Berlin/Heidelberg, Germany, 2016. [Google Scholar]
  25. Barker, D.J. The malnourished baby and infant: Relationship with Type 2 diabetes. Brit. Med. Bull. 2001, 60, 69–88. [Google Scholar] [CrossRef] [Green Version]
  26. Wells, J.C.K.; Pomeroy, E.; Walimbe, S.R.; Popkin, B.M.; Yajnik, C.S. The Elevated Susceptibility to Diabetes in India: An Evolutionary Perspective. Front. Public Health 2016, 4, 145. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Santangelo, C.; Varì, R.; Scazzocchio, B.; Filesi, C.; Masella, R. Management of reproduction and pregnancy complications in maternal obesity: Which role for dietary polyphenols? BioFactors 2013, 40, 79–102. [Google Scholar] [CrossRef] [PubMed]
  28. Poothullil, J.M. Breastfeeding and Risk of Overweight. JAMA 2001, 286, 1448–1450. [Google Scholar] [CrossRef]
  29. Parsons, T.; Power, C.; Manor, O. Infant feeding and obesity through the lifecourse. Arch. Dis. Child. 2003, 88, 793–794. [Google Scholar] [CrossRef] [Green Version]
  30. Frye, C.S.; Heinrich, J.J. Trends and predictors of overweight and obesity in East German children. Int. J. Obes. 2003, 27, 963–969. [Google Scholar] [CrossRef] [Green Version]
  31. Owen, C.G.; Martin, R.M.; Whincup, P.H.; Smith, G.D.; Cook, D.G. Effect of Infant Feeding on the Risk of Obesity Across the Life Course: A Quantitative Review of Published Evidence. Pediatrics 2005, 115, 1367–1377. [Google Scholar] [CrossRef]
  32. Dietz, W.H. Breastfeeding may help prevent childhood overweight. JAMA 2001, 285, 2506–2507. [Google Scholar] [CrossRef]
  33. Metzger, M.W.; McDade, T.W. Breastfeeding as obesity prevention in the United States: A sibling difference model. Am. J. Hum. Biol. 2009, 22, 291–296. [Google Scholar] [CrossRef]
  34. Stolzer, J.M. Breastfeeding and obesity: A meta-analysis. Open J. Prev. Med. 2011, 1, 88–93. [Google Scholar] [CrossRef] [Green Version]
  35. Hauner, H.; Röhrig, K.; Petruschke, T. Effects of epidermal growth factor (EGF), platelet-derived growth factor (PDGF) and fibroblast growth factor (FGF) on human adipocyte development and function. Eur. J. Clin. Investig. 1995, 25, 90–96. [Google Scholar] [CrossRef]
  36. Petruschke, T.; Röhrig, K.; Hauner, H. Transforming growth factor beta (TGF-beta) inhibits the differentiation of human adipocyte precursor cells in primary culture. Int. J. Obes. Relat. Metab. Disord. J. Int. Assoc. Study Obes. 1994, 18, 532–536. [Google Scholar]
  37. Heinig, M.J.; Nommsen, L.A.; Peerson, J.M.; Lonnerdal, B.; Dewey, K.G. Energy and protein intakes of breast-fed and formula-fed infants during the first year of life and their association with growth velocity: The DARLING Study. Am. J. Clin. Nutr. 1993, 58, 152–161. [Google Scholar] [CrossRef] [Green Version]
  38. Lucas, A.; Blackburn, A.; Aynsley-Green, A.; Sarson, D.; Adrian, T.; Bloom, S. Breast vs bottle: Endocrine responses are different with formula feeding. Lancet 1980, 315, 1267–1269. [Google Scholar] [CrossRef]
  39. Von Kries, R.; Koletzko, B.; Sauerwald, T.; Von Mutius, E.; Barnert, D.; Grunert, V.; Von Voss, H. Breast feeding and obesity: Cross sectional study. BMJ 1999, 319, 147–150. [Google Scholar] [CrossRef] [Green Version]
  40. Ravelli, A.C.J.; Van Der Meulen, J.H.P.; Osmond, C.; Barker, D.J.P.; Bleker, O.P. Infant feeding and adult glucose tolerance, lipid profile, blood pressure, and obesity. Arch. Dis. Child. 2000, 82, 248–252. [Google Scholar] [CrossRef] [Green Version]
  41. Young, T.K.; Martens, P.J.; Taback, S.P.; Sellers, E.A.C.; Dean, H.J.; Cheang, M.; Flett, B. Type 2 diabetes mellitus in children: Prenatal and early infancy risk factors among native Canadians. Arch. Pediatr. Adolesc. Med. 2002, 156, 651–655. [Google Scholar] [CrossRef] [Green Version]
  42. Martin, R.M.; Ebrahi, S.; Griffin, M.; Smith, G.D.; Nicolaides, A.N.; Georgiou, N.; Watson, S.; Feankel, S.; Holly, J.M.P.; Gunnell, D. Breastfeeding and atherosclerosis: Intima-media thickness and plaques at 65-year follow-up of the Boyd Orr cohort. Arterioscleros. Thrombos. Vasc. Biol. 2005, 25, 1482–1488. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Rich-Edwards, J.W.; Stampfer, M.J.; Manson, J.E.; Rosner, B.; Hu, F.B.; Michels, K.B.; Willett, W.C. Breastfeeding During Infancy and the Risk of Cardiovascular Disease in Adulthood. Epidemiology 2004, 15, 550–556. [Google Scholar] [CrossRef] [PubMed]
  44. Aynsley-Green, A. The endocrinology of feeding in the newborn. Baillière’s Clin. Endocrinol. Metab. 1989, 3, 837–868. [Google Scholar] [CrossRef]
  45. Salmenperä, L.; Perheentupa, J.; Siimes, M.A.; Adrian, T.E.; Bloom, S.R.; Aynsley-Green, A. Effects of feeding regimen on blood glucose levels and plasma concentrations of pancreatic hormones and gut regulatory peptides at 9 months of age: Comparison between infants fed with milk formula and infants exclusively breast-fed from birth. J. Pediatr. Gastroenterol. Nutr. 1988, 7, 651–656. [Google Scholar] [CrossRef]
  46. Pettitt, D.J.; Forman, M.R.; Hanson, R.L.; Knowler, W.C.; Bennett, P.H. Breastfeeding and incidence of non-insulin-dependent diabetes mellitus in Pima Indians. Lancet 1997, 350, 166–168. [Google Scholar] [CrossRef]
  47. Martin, R.M.; Ben-Shlomo, Y.; Gunnell, D.; Elwood, P.; Yarnell, J.W.G.; Smith, G.D. Breast feeding and cardiovascular disease risk factors, incidence, and mortality: The Caerphilly study. J. Epidemiol. Community Heal. 2005, 59, 121–129. [Google Scholar] [CrossRef] [Green Version]
  48. Martin, R.M.; Ness, A.R.; Gunnell, D.; Emmett, P.; Smith, D. Does breast-feeding in infancy lower blood pressure in childhood? The Avon Longitudinal Study of Parents and Children (ALSPAC). Circulation 2004, 109, 1259–1266. [Google Scholar] [CrossRef] [Green Version]
  49. Singhal, A.; Cole, T.J.; Fewtrell, M.; Lucas, A. Breastmilk feeding and lipoprotein profile in adolescents born preterm: Follow-up of a prospective randomised study. Lancet 2004, 363, 1571–1578. [Google Scholar] [CrossRef]
  50. Rudnicka, A.R.; Owen, C.G.; Strachan, D.P. The Effect of Breastfeeding on Cardiorespiratory Risk Factors in Adult Life. Pediatrics 2007, 119, e1107–e1115. [Google Scholar] [CrossRef]
  51. Fomon, S.J. Infant Feeding in the 20th Century: Formula and Beikost. J. Nutr. 2001, 131, 409S–420S. [Google Scholar] [CrossRef]
  52. Ursell, L.K.; Metcalf, J.L.; Parfrey, L.W.; Knight, R. Defining the human microbiome. Nutr. Rev. 2012, 70, S38–S44. [Google Scholar] [CrossRef] [Green Version]
  53. Le Chatelier, E.; Nielsen, T.; Qin, J.; Prifti, E.; Hildebrand, F.; Falony, G.; Almeida, M.; Arumugam, M.; Batto, J.-M.; Kennedy, S.; et al. Richness of human gut microbiome correlates with metabolic markers. Nature 2013, 500, 541–546. [Google Scholar] [CrossRef]
  54. Ridaura, V.K.; Faith, J.J.; Rey, F.E.; Cheng, J.; Duncan, A.E.; Kau, A.L.; Griffin, N.W.; Lombard, V.; Henrissat, B.; Bain, J.R.; et al. Gut Microbiota from Twins Discordant for Obesity Modulate Metabolism in Mice. Science 2013, 341, 1241214. [Google Scholar] [CrossRef] [Green Version]
  55. Qin, J.; Li, Y.; Cai, Z.; Li, S.; Zhu, J.; Zhang, F.; Liang, S.; Zhang, W.; Guan, Y.; Shen, D.; et al. A metagenome-wide association study of gut microbiota in type 2 diabetes. Nature 2012, 490, 55–60. [Google Scholar] [CrossRef]
  56. Ley, R.E.; Bäckhed, F.; Turnbaugh, P.; Lozupone, C.A.; Knight, R.D.; Gordon, J.I. Obesity alters gut microbial ecology. Proc. Natl. Acad. Sci. USA 2005, 102, 11070–11075. [Google Scholar] [CrossRef] [Green Version]
  57. Bäckhed, F.; Manchester, J.K.; Semenkovich, C.F.; Gordon, J.I. Mechanisms underlying the resistance to diet-induced obesity in germ-free mice. Proc. Natl. Acad. Sci. USA 2007, 104, 979–984. [Google Scholar] [CrossRef] [Green Version]
  58. Turnbaugh, P.J.; Ley, R.E.; Mahowald, M.A.; Magrini, V.; Mardis, E.R.; Gordon, J.I. An obesity-associated gut microbiome with increased capacity for energy harvest. Nat. Cell Biol. 2006, 444, 1027–1031. [Google Scholar] [CrossRef]
  59. Huttenhower, C. The Human Microbiome Project Consortium. Structure, function and diversity of the healthy human microbiome. Nature 2012, 486, 207. [Google Scholar]
  60. Robertson, R.C.; Kaliannan, K.; Strain, C.R.; Ross, R.P.; Stanton, C.; Kang, J.X. Maternal omega-3 fatty acids regulate offspring obesity through persistent modulation of gut microbiota. Microbiome 2018, 6, 1–14. [Google Scholar] [CrossRef]
  61. Bokulich, N.A.; Chung, J.; Battaglia, T.; Henderson, N.; Jay, M.; Li, H.; Lieber, A.D.; Wu, F.; Perez-Perez, G.I.; Chen, Y.; et al. Antibiotics, birth mode, and diet shape microbiome maturation during early life. Sci. Transl. Med. 2016, 8, 343ra82. [Google Scholar] [CrossRef] [Green Version]
  62. Gilbert, J.A.; Quinn, R.A.; Debelius, J.; Xu, Z.Z.; Morton, J.; Garg, N.; Jansson, J.K.; Dorrestein, R.A.Q.N.G.P.C.; Knight, R. Microbiome-wide association studies link dynamic microbial consortia to disease. Nat. Cell Biol. 2016, 535, 94–103. [Google Scholar] [CrossRef] [PubMed]
  63. Hill, C.J.; Lynch, D.B.; Murphy, K.; Ulaszewska, M.; Jeffery, I.B.; O’Shea, C.A.; Watkins, C.; Dempsey, E.; Mattivi, F.; Tuohy, K.; et al. Evolution of gut microbiota composition from birth to 24 weeks in the INFANTMET Cohort. Microbiome 2017, 5, 1–18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Lundgren, S.N.; Madan, J.C.; Emond, J.A.; Morrison, H.G.; Christensen, B.C.; Karagas, M.R.; Hoen, A.G. Maternal diet during pregnancy is related with the infant stool microbiome in a delivery mode-dependent manner. Microbiome 2018, 6, 1–11. [Google Scholar] [CrossRef] [Green Version]
  65. Ma, J.; Prince, A.L.; Bader, D.; Hu, M.; Ganu, R.; Baquero, K.; Blundell, P.; Harris, R.A.; Frias, A.E.; Grove, K.L.; et al. High-fat maternal diet during pregnancy persistently alters the offspring microbiome in a primate model. Nat. Commun. 2014, 5, 1–11. [Google Scholar] [CrossRef] [PubMed]
  66. Mulligan, C.M.; Friedman, J.E. Maternal modifiers of the infant gut microbiota: Metabolic consequences. J. Endocrinol. 2017, 235, R1–R12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Hansen, C.H.F.; Krych, L.; Buschard, K.; Metzdorff, S.B.; Nellemann, C.; Hansen, L.H.; Nielsen, D.S.; Frokiaer, H.; Skov, S.; Hansen, A.K. A Maternal Gluten-Free Diet Reduces Inflammation and Diabetes Incidence in the Offspring of NOD Mice. Diabetes 2014, 63, 2821–2832. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Kasahara, K.; Krautkramer, K.A.; Org, E.; Romano, K.A.; Kerby, R.L.; Vivas, E.I.; Mehrabian, M.; Denu, J.M.; Bäckhed, F.; Lusis, A.J.; et al. Interactions between Roseburia intestinalis and diet modulate atherogenesis in a murine model. Nat. Microbiol. 2018, 3, 1461–1471. [Google Scholar] [CrossRef] [PubMed]
  69. Myles, I.A.; Fontecilla, N.M.; Janelsins, B.M.; Vithayathil, P.J.; Segre, J.A.; Datta, S.K. Parental Dietary Fat Intake Alters Offspring Microbiome and Immunity. J. Immunol. 2013, 191, 3200–3209. [Google Scholar] [CrossRef]
  70. DiGiulio, D.B.; Romero, R.; Amogan, H.P.; Kusanovic, J.P.; Bik, E.M.; Gotsch, F.; Kim, C.J.; Erez, O.; Edwin, S.; Relman, D.A. Microbial Prevalence, Diversity and Abundance in Amniotic Fluid During Preterm Labor: A Molecular and Culture-Based Investigation. PLoS ONE 2008, 3, e3056. [Google Scholar] [CrossRef]
  71. Collado, M.C.; Rautava, S.; Aakko, J.; Isolauri, E.; Salminen, S. Human gut colonisation may be initiated in utero by distinct microbial communities in the placenta and amniotic fluid. Sci. Rep. 2016, 6, 23129. [Google Scholar] [CrossRef] [Green Version]
  72. Antony, K.M.; Ma, J.; Mitchell, K.B.; Racusin, D.A.; Versalovic, J.; Aagaard, K. The preterm placental microbiome varies in association with excess maternal gestational weight gain. Am. J. Obs. Gynecol. 2015, 212, 653.e1–653.e16. [Google Scholar] [CrossRef] [Green Version]
  73. Aagaard, K.; Ma, J.; Antony, K.M.; Ganu, R.; Petrosino, J.; Versalovic, J. The placenta harbors a unique microbiome. Sci. Transl. Med. 2014, 6, ra65–ra237. [Google Scholar] [CrossRef] [Green Version]
  74. Pannaraj, P.S.; Li, F.; Cerini, C.; Bender, J.M.; Yang, S.; Rollie, A.; Adisetiyo, H.; Zabih, S.; Lincez, P.J.; Bittinger, K.; et al. Association Between Breast Milk Bacterial Communities and Establishment and Development of the Infant Gut Microbiome. JAMA Pediatr. 2017, 171, 647–654. [Google Scholar] [CrossRef] [PubMed]
  75. Rudloff, S.; Kunz, C. Milk Oligosaccharides and Metabolism in Infants. Adv. Nutr. 2012, 3, 398S–405S. [Google Scholar] [CrossRef] [Green Version]
  76. Coppa, G.V.; Bruni, S.; Morelli, L.; Soldi, S.; Gabrielli, O. The first prebiotics in humans: Human milk oligosaccharides. J. Clin. Gastroenterol. 2004, 38, S80–S83. [Google Scholar] [CrossRef]
  77. Xiao, L.; Land, B.V.; Engen, P.A.; Naqib, A.; Green, S.J.; Nato, A.; Leusink-Muis, T.; Garssen, J.; Keshavarzian, A.; Stahl, B.; et al. Human milk oligosaccharides protect against the development of autoimmune diabetes in NOD-mice. Sci. Rep. 2018, 8, 1–15. [Google Scholar] [CrossRef] [Green Version]
  78. Hamilton, M.K.; Ronveaux, C.C.; Rust, B.M.; Newman, J.W.; Hawley, M.; Barile, D.; Mills, D.A.; Raybould, H.E. Prebiotic milk oligosaccharides prevent development of obese phenotype, impairment of gut permeability, and microbial dysbiosis in high fat-fed mice. Am. J. Physiol. Liver Physiol. 2017, 312, G474–G487. [Google Scholar] [CrossRef]
  79. Savage, J.H.; Lee-Sarwar, K.A.; Sordillo, J.E.; Lange, N.E.; Zhou, Y.; O’Connor, G.T.; Sandel, M.; Bacharier, L.B.; Zeiger, R.; Sodergren, E.; et al. Diet during Pregnancy and Infancy and the Infant Intestinal Microbiome. J. Pediatr. 2018, 203, 47–54.e4. [Google Scholar] [CrossRef]
  80. Dieterich, C.M.; Felice, J.P.; O’Sullivan, E.; Rasmussen, K.M. Breastfeeding and Health Outcomes for the Mother-Infant Dyad. Pediatr. Clin. N. Am. 2013, 60, 31–48. [Google Scholar] [CrossRef] [Green Version]
  81. Binns, C.; Lee, M.; Low, W.Y. The Long-Term Public Health Benefits of Breastfeeding. Asia Pac. J. Public Health 2016, 28, 7–14. [Google Scholar] [CrossRef] [Green Version]
  82. Ip, S.; Chung, M.; Raman, G.; Trikalinos, T.A.; Lau, J. A summary of the Agency for Healthcare Research and Quality’s evidence report on breastfeeding in developed countries. Breastfeed. Med. 2009, 4, S17–S30. [Google Scholar] [CrossRef]
  83. Brugman, S.; Visser, J.T.J.; Hillebrands, J.L.; Bos, N.A.; Rozing, J. Prolonged exclusive breastfeeding reduces autoimmune diabetes incidence and increases regulatory T-cell frequency in bio-breeding diabetes-prone rats. Diabetes Metab. Res. Rev. 2009, 25, 380–387. [Google Scholar] [CrossRef] [PubMed]
  84. Cordero, P.; Vinciguerra, M.; Oben, J.A. Maternal Perinatal Nutrition and Offspring Programming. In Principles of Nutrigenetics and Nutrigenomics; Elsevier BV: Amsterdam, The Netherlands, 2020; pp. 121–127. [Google Scholar]
  85. Krishnaveni, G.V.; Srinivasan, K. Maternal Nutrition and Offspring Stress Response—Implications for Future Development of Non-Communicable Disease: A Perspective from India. Front. Psychiatry 2019, 10, 795. [Google Scholar] [CrossRef] [PubMed]
  86. Meiliana, A.; Wijaya, A. Nutrigenetics, Nutrigenomics and Precision Nutrition. Indones. Biomed. J. 2020, 12, 189–200. [Google Scholar] [CrossRef]
  87. Wu, G.; Imhoff-Kunsch, B.; Girard, A.W. Biological Mechanisms for Nutritional Regulation of Maternal Health and Fetal Development. Paediatr. Périnat. Epidemiol. 2012, 26, 4–26. [Google Scholar] [CrossRef] [PubMed]
  88. Gris, J.-C.; Perneger, T.V.; Quéré, I.; Mercier, E.; Fabbro-Peray, P.; Lavigne-Lissalde, G.; Hoffet, M.; Déchaud, H.; Boyer, J.-C.; Ripart-Neveu, S.; et al. Antiphospholipid/antiprotein antibodies, hemostasis-related autoantibodies, and plasma homocysteine as risk factors for a first early pregnancy loss: A matched case-control study. Blood 2003, 102, 3504–3513. [Google Scholar] [CrossRef] [Green Version]
  89. van der Put, N.M.; Gabreels, F.; Stevens, M.B.; Smeitink, A.M.; Trijbels, F.J.M.; Eskes, T.K.A.B.; van den Heuvel, L.; Blom, H.J. A second common mutation in the methylenetetrahydrofolate reductase gene: An additional risk factor for neural-tube defects? Am. J. Hum. Genet. 1998, 62, 1044–1051. [Google Scholar] [CrossRef] [Green Version]
  90. Yajnik, C.S.; Deshpande, S.S.; Jackson, A.A.; Refsum, H.; Rao, S.; Fisher, D.J.; Bhat, D.S.; Naik, S.S.; Coyaji, K.J.; Joglekar, C.V.; et al. Vitamin B12 and folate concentrations during pregnancy and insulin resistance in the offspring: The Pune Maternal Nutrition Study. Diabetology 2007, 51, 29–38. [Google Scholar] [CrossRef] [Green Version]
  91. Leikin-Frenkel, A.I. Perinatal lipid nutrition. In Molecular Nutrition: Mother and Infant; Elsevier: Amsterdam, The Netherlands, 2021; pp. 337–359. [Google Scholar]
  92. Muhlhausler, B.S.; Gugusheff, J.R.; Ong, Z.Y.; Vithayathil, M.A. Pregnancy, obesity and insulin resistance: Maternal overnutrition and the target windows of fetal development. Horm. Mol. Biol. Clin. Investig. 2013, 15, 25–36. [Google Scholar] [CrossRef] [Green Version]
  93. Segura, M.T.; Demmelmair, H.; Krauss-Etschmann, S.; Nathan, P.; Dehmel, S.; Padilla, M.C.; Rueda, R.; Koletzko, B.; Campoy, C. Maternal BMI and gestational diabetes alter placental lipid transporters and fatty acid composition. Placenta 2017, 57, 144–151. [Google Scholar] [CrossRef]
  94. Barbour, L.A.; Hernandez, T.L. Maternal Lipids and Fetal Overgrowth: Making Fat from Fat. Clinical 2018, 40, 1638–1647. [Google Scholar] [CrossRef] [Green Version]
  95. Warrington, N.M. Early Growth Genetics (EGG) Consortium; Richmond, R.; Fenstra, B.; Myhre, R.; Gaillard, R.; Paternoster, L.; Wang, C.A.; Beaumont, R.N.; Das, S.; et al. Maternal and fetal genetic contribution to gestational weight gain. Int. J. Obes. 2018, 42, 775–784. [Google Scholar] [CrossRef] [Green Version]
  96. Shrestha, D.; Rahman, M.L.; Workalemahu, T.; Zhu, C.; Tekola-Ayele, F. Influence of Fetal and Maternal Genetic Susceptibility to Obesity on Birthweight in African Ancestry Populations. Front. Genet. 2018, 9, 511. [Google Scholar] [CrossRef]
  97. Catalano, P.A.; Ehrenberg, H. The short-and long-term implications of maternal obesity on the mother and her offspring. BJOG Int. J. Obs. Gynaecol. 2006, 113, 1126–1133. [Google Scholar] [CrossRef]
  98. Maugeri, A.; Barchitta, M.; Agodi, A. Gene–nutrient interaction: From fetal development to lifelong health. In Molecular Nutrition: Mother and Infant; Elsevier: Amsterdam, The Netherlands, 2021; pp. 299–315. [Google Scholar]
  99. Marquardt, A.; Stöhr, H.; White, K.; Weber, B.H. cDNA Cloning, Genomic Structure, and Chromosomal Localization of Three Members of the Human Fatty Acid Desaturase Family. Genome 2000, 66, 175–183. [Google Scholar] [CrossRef]
  100. Koletzko, B.; Lattka, E.; Zeilinger, S.; Illig, T.; Steer, C. Genetic variants of the fatty acid desaturase gene cluster predict amounts of red blood cell docosahexaenoic and other polyunsaturated fatty acids in pregnant women: Findings from the Avon Longitudinal Study of Parents and Children. Am. J. Clin. Nutr. 2010, 93, 211–219. [Google Scholar] [CrossRef] [Green Version]
  101. Lattka, E.; Koletzko, B.; Zeilinger, S.; Hibbeln, J.R.; Klopp, N.; Ring, S.M.; Steer, C.D. Umbilical cord PUFA are determined by maternal and child fatty acid desaturase (FADS) genetic variants in the Avon Longitudinal Study of Parents and Children (ALSPAC). Br. J. Nutr. 2012, 109, 1196–1210. [Google Scholar] [CrossRef] [Green Version]
  102. van Eijsden, M.; Hornstra, G.; van der Wal, M.F.; Vrijkotte, T.G.M.; Bonsel, G.J. Maternal n− 3, n− 6, and trans fatty acid profile early in pregnancy and term birth weight: A prospective cohort study. Am. J. Clin. Nutr. 2008, 87, 887–895. [Google Scholar] [CrossRef] [Green Version]
  103. Barker, D.J. The developmental origins of well-being. Sci. Well-Being 2005, 359, 58–73. [Google Scholar] [CrossRef] [Green Version]
  104. Kröger, J.; Schulze, M.B. Recent insights into the relation of Δ5 desaturase and Δ6 desaturase activity to the development of type 2 diabetes. Curr. Opin. Lipidol. 2012, 23, 4–10. [Google Scholar] [CrossRef]
  105. Hattersley, A.T.; Beards, F.; Ballantyne, E.; Appleton, M.; Harvey, R.; Ellard, S. Mutations in the glucokinase gene of the fetus result in reduced birth weight. Nat. Genet. 1998, 19, 268–270. [Google Scholar] [CrossRef] [PubMed]
  106. Freathy, R.M.; Bennett, A.J.; Ring, S.M.; Shields, B.; Groves, C.J.; Timpson, N.J.; Weedon, M.N.; Zeggini, E.; Lindgren, C.M.; Lango, H.; et al. Type 2 Diabetes Risk Alleles Are Associated with Reduced Size at Birth. Diabetes 2009, 58, 1428–1433. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Verier, C.; Meirhaeghe, A.; Bokor, S.; Breidenassel, C.; Manior, Y.; Molnar, D.; Artero, E.G.; Nova, E.; De Henauw, S.; Moreno, L.A.; et al. Breast-Feeding Modulates the Influence of the Peroxisome Proliferator–Activated Receptor-γ (PPARG2) Pro12Ala Polymorphism on Adiposity in Adolescents: The Healthy Lifestyle in Europe by Nutrition in Adolescence (HELENA) cross-sectional study. Diabetes Care 2010, 33, 190–196. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Mansoori, A.; Amini, M.; Kolahdooz, F.; Seyedrezazadeh, E. Obesity and Pro12Ala Polymorphism of Peroxisome Proliferator-Activated Receptor-Gamma Gene in Healthy Adults: A Systematic Review and Meta-Analysis. Ann. Nutr. Metab. 2015, 67, 104–118. [Google Scholar] [CrossRef]
  109. Indrio, F.; Martini, S.; Francavilla, R.; Corvaglia, L.; Cristofori, F.; Mastrolia, S.A.; Neu, J.; Rautava, S.; Spena, G.R.; Raimondi, F.; et al. Epigenetic Matters: The Link between Early Nutrition, Microbiome, and Long-term Health Development. Front. Pediatr. 2017, 5, 178. [Google Scholar] [CrossRef]
  110. Wan, Y.; Saghatelian, A.; Chong, L.-W.; Zhang, C.-L.; Cravatt, B.F.; Evans, R.M. Maternal PPARγ protects nursing neonates by suppressing the production of inflammatory milk. Genes Dev. 2007, 21, 1895–1908. [Google Scholar] [CrossRef] [Green Version]
  111. Ronto, R.; Wu, J.H.; Singh, G.M. The global nutrition transition: Trends, disease burdens and policy interventions. Public Heal. Nutr. 2018, 21, 2267–2270. [Google Scholar] [CrossRef] [Green Version]
  112. Sofi, F.; Macchi, C.; Abbate, R.; Gensini, G.F.; Casini, A. Mediterranean diet and health. BioFactors 2013, 39, 335–342. [Google Scholar] [CrossRef]
  113. Zimmet, P.; Alberti, K.G.M.M.; Shaw, J. Global and societal implications of the diabetes epidemic. Nat. Cell Biol. 2001, 414, 782–787. [Google Scholar] [CrossRef]
  114. Weinhold, B. Epigenetics: The Science of Change. Environ. Health Perspect. 2006, 114, A160–A167. [Google Scholar] [CrossRef] [Green Version]
  115. Nowacka-Zawisza, M.; Wiśnik, E. DNA methylation and histone modifications as epigenetic regulation in prostate cancer. Oncol. Rep. 2017, 38, 2587–2596. [Google Scholar] [CrossRef] [Green Version]
  116. Moore, L.; Le, T.; Fan, G. DNA methylation and its basic function. Neuropsychopharmacology 2013, 38, 23–38. [Google Scholar] [CrossRef] [Green Version]
  117. Yao, Q.; Chen, Y.; Zhou, X. The roles of microRNAs in epigenetic regulation. Curr. Opin. Chem. Biol. 2019, 51, 11–17. [Google Scholar] [CrossRef]
  118. Zhang, P.; Torres, K.; Liu, X.; Liu, C.-G.; Pollock, R.E. An Overview of Chromatin-Regulating Proteins in Cells. Curr. Protein Pept. Sci. 2016, 17, 401–410. [Google Scholar] [CrossRef] [Green Version]
  119. Jiménez-Chillarón, J.C.; Díaz, R.; Martínez, D.; Pentinat, T.; Ramón-Krauel, M.; Ribó, S.; Plösch, T. The role of nutrition on epigenetic modifications and their implications on health. Biochim. 2012, 94, 2242–2263. [Google Scholar] [CrossRef]
  120. McKay, J.A.; Groom, A.; Potter, C.; Coneyworth, L.J.; Ford, D.; Mathers, J.C.; Relton, C.L. Genetic and Non-Genetic Influences during Pregnancy on Infant Global and Site Specific DNA Methylation: Role for Folate Gene Variants and Vitamin B 12. PLoS ONE 2012, 7, e33290. [Google Scholar] [CrossRef]
  121. Cordero, P.; Li, J.; Oben, J.A. Epigenetics of obesity: Beyond the genome sequence. Curr. Opin. Clin. Nutr. Metab. Care 2015, 18, 361–366. [Google Scholar] [CrossRef]
  122. Moraru, A.; De Almeida, M.M.; Degryse, J.-M. PALTEM: What Parameters Should Be Collected in Disaster Settings to Assess the Long-Term Outcomes of Famine? Int. J. Environ. Res. Public Health 2018, 15, 857. [Google Scholar] [CrossRef] [Green Version]
  123. van Uitert, E.M.; Steegers-Theunissen, R.P. Influence of maternal folate status on human fetal growth parameters. Molec. Nutr. Food Res. 2013, 57, 582–595. [Google Scholar] [CrossRef]
  124. Welch, G.N., Jr.; Loscalzo, J.G.R.U. Homocysteine, Oxidative Stress, and Vascular Disease. Hosp. Pr. 1997, 32, 81–92. [Google Scholar] [CrossRef]
  125. Wong, C.P.; Rinaldi, N.A.; Ho, E. Zinc deficiency enhanced inflammatory response by increasing immune cell activation and inducing IL6 promoter demethylation. Mol. Nutr. Food Res. 2015, 59, 991–999. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Tomat, A.L.; de los Ángeles Costa, M.; Arranz, C.T. Zinc restriction during different periods of life: Influence in renal and cardiovascular diseases. Nutrients 2011, 27, 392–398. [Google Scholar] [CrossRef] [PubMed]
  127. Khot, V.; Chavan-Gautam, P.; Joshi, S. Proposing interactions between maternal phospholipids and the one carbon cycle: A novel mechanism influencing the risk for cardiovascular diseases in the offspring in later life. Life Sci. 2015, 129, 16–21. [Google Scholar] [CrossRef] [PubMed]
  128. Sohi, G.; Marchand, K.; Revesz, A.; Arany, E.; Hardy, D.B. Maternal Protein Restriction Elevates Cholesterol in Adult Rat Offspring Due to Repressive Changes in Histone Modifications at the Cholesterol 7α-Hydroxylase Promoter. Mol. Endocrinol. 2011, 25, 785–798. [Google Scholar] [CrossRef] [Green Version]
  129. Jelinek, D.F.; Andersson, S.; Slaughter, C.A.; Russell, D.W. Cloning and regulation of cholesterol 7 alpha-hydroxylase, the rate-limiting enzyme in bile acid biosynthesis. J. Biol. Chem. 1990, 265, 8190–8197. [Google Scholar] [CrossRef]
  130. Wilson, P.W.F.; D’Agostino, R.B.; Levy, D.; Belanger, A.M.; Silbershatz, H.; Kannel, W.B. Prediction of Coronary Heart Disease Using Risk Factor Categories. Circulation 1998, 97, 1837–1847. [Google Scholar] [CrossRef] [Green Version]
  131. Mathieu, P.; Pibarot, P.; Després, J.-P. Metabolic Syndrome: The Danger Signal in Atherosclerosis. Vasc. Health Risk Manag. 2006, 2, 285–302. [Google Scholar] [CrossRef] [Green Version]
  132. Vo, T.X.; Revesz, A.; Sohi, G.; Ma, N.; Hardy, D.B. Maternal protein restriction leads to enhanced hepatic gluconeogenic gene expression in adult male rat offspring due to impaired expression of the liver X receptor. J. Endocrinol. 2013, 218, 85–97. [Google Scholar] [CrossRef] [Green Version]
  133. Zheng, S.; Rollet, M.; Pan, Y.-X. Protein restriction during gestation alters histone modifications at the glucose transporter 4 (GLUT4) promoter region and induces GLUT4 expression in skeletal muscle of female rat offspring. J. Nutr. Biochem. 2012, 23, 1064–1071. [Google Scholar] [CrossRef]
  134. Sun, C.; Denisenko, O.; Sheth, B.; Cox, A.; Lucas, E.S.; Smyth, N.R.; Fleming, T.P. Epigenetic regulation of histone modifications and Gata6 gene expression induced by maternal diet in mouse embryoid bodies in a model of developmental programming. BMC Dev. Biol. 2015, 15, 3. [Google Scholar] [CrossRef] [Green Version]
  135. Ramaiyan, B.; Talahalli, R.R. Dietary Unsaturated Fatty Acids Modulate Maternal Dyslipidemia-Induced DNA Methylation and Histone Acetylation in Placenta and Fetal Liver in Rats. Lipids 2018, 53, 581–588. [Google Scholar] [CrossRef]
  136. Hawkins, P.G.; Morris, K.V. RNA and transcriptional modulation of gene expression. Cell Cycle 2008, 7, 602–607. [Google Scholar] [CrossRef] [Green Version]
  137. Ross, S.A.; Davis, C.D. The Emerging Role of microRNAs and Nutrition in Modulating Health and Disease. Annu. Rev. Nutr. 2014, 34, 305–336. [Google Scholar] [CrossRef]
  138. Berná, G.; Oliveras-López, M.J.; Jurado-Ruíz, E.; Tejedo, J.; Bedoya, F.; Soria, B.; Martín, F. Nutrigenetics and Nutrigenomics Insights into Diabetes Etiopathogenesis. Nutrients 2014, 6, 5338–5369. [Google Scholar] [CrossRef] [Green Version]
  139. Zampetaki, A.; Kiechl, S.; Drozdov, I.; Willeit, P.; Mayr, U.; Prokopi, M.; Mayr, A.; Weger, S.; Oberhollenzer, F.; Bonora, E.; et al. Plasma MicroRNA Profiling Reveals Loss of Endothelial MiR-126 and Other MicroRNAs in Type 2 Diabetes. Circ. Res. 2010, 107, 810–817. [Google Scholar] [CrossRef]
  140. Qin, D.-N.; Qian, L.; Hu, D.-L.; Yu, Z.-B.; Han, S.-P.; Zhu, C.; Wang, X.; Hu, X. Effects of miR-19b Overexpression on Proliferation, Differentiation, Apoptosis and Wnt/β-Catenin Signaling Pathway in P19 Cell Model of Cardiac Differentiation In Vitro. Cell Biophys. 2013, 66, 709–722. [Google Scholar] [CrossRef]
  141. Wang, J.; Wu, Z.; Li, D.; Li, N.; Dindot, S.V.; Satterfield, M.C.; Bazer, F.W.; Wu, G. Nutrition, Epigenetics, and Metabolic Syndrome. Antioxid. Redox Signal. 2012, 17, 282–301. [Google Scholar] [CrossRef]
  142. Williams, S.R.; Yang, Q.; Chen, F.; Liu, X.; Keene, K.L.; Jacques, P.; Chen, W.-M.; Weinstein, G.; Hsu, F.-C.; Beiser, A.; et al. Genome-Wide Meta-Analysis of Homocysteine and Methionine Metabolism Identifies Five One Carbon Metabolism Loci and a Novel Association of ALDH1L1 with Ischemic Stroke. PLoS Genet. 2014, 10, e1004214. [Google Scholar] [CrossRef]
  143. Neri, C.; Edlow, A.G. Effects of Maternal Obesity on Fetal Programming: Molecular Approaches. Cold Spring Harb. Perspect. Med. 2016, 6, a026591. [Google Scholar] [CrossRef] [Green Version]
  144. Flowers, E.; Won, G.Y.; Fukuoka, Y. MicroRNAs associated with exercise and diet: A systematic review. Physiol. Genom. 2015, 47, 1–11. [Google Scholar] [CrossRef] [Green Version]
  145. Wagner, A.E.; Piegholdt, S.; Ferraro, M.; Pallauf, K.; Rimbach, G. Food derived microRNAs. Food Funct. 2015, 6, 714–718. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Turchinovich, A.; Samatov, T.R.; Tonevitsky, A.G.; Burwinkel, B. Circulating miRNAs: Cell–cell communication function? Front. Genet. 2013, 4, 119. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Nicholas, L.M.; Rattanatray, L.; MacLaughlin, S.M.; Ozanne, S.E.; Kleemann, D.O.; Walker, S.K.; Morrison, J.L.; Zhang, S.; Muhlhäusler, B.S.; Martin-Gronert, M.S.; et al. Differential effects of maternal obesity and weight loss in the periconceptional period on the epigenetic regulation of hepatic insulin-signaling pathways in the offspring. Faseb. J. 2013, 27, 3786–3796. [Google Scholar] [CrossRef] [PubMed]
  148. Long, J.; Wang, Y.; Wang, W.; Chang, B.H.J.; Danesh, F.R. MicroRNA-29c Is a Signature MicroRNA under High Glucose Conditions That Targets Sprouty Homolog 1, and Its in Vivo Knockdown Prevents Progression of Diabetic Nephropathy. J. Biol. Chem. 2011, 286, 11837–11848. [Google Scholar] [CrossRef] [Green Version]
  149. Bagge, A.; Clausen, T.R.; Larsen, S.; Ladefoged, M.; Rosenstierne, M.W.; Larsen, L.; Vang, O.; Nielsen, J.H.; Dalgaard, L.T. MicroRNA-29a is up-regulated in beta-cells by glucose and decreases glucose-stimulated insulin secretion. Biochem. Biophys. Res. Commun. 2012, 426, 266–272. [Google Scholar] [CrossRef] [Green Version]
  150. Roggli, E.; Gattesco, S.; Caille, D.; Briet, C.; Boitard, C.; Meda, P.; Regazzi, R. Changes in microRNA expression contribute to pancreatic β-cell dysfunction in prediabetic NOD mice. Diabetes 2012, 61, 1742–1751. [Google Scholar] [CrossRef] [Green Version]
  151. Liang, J.; Liu, C.; Qiao, A.; Cui, Y.; Zhang, H.; Cui, A.; Zhang, S.; Yang, Y.; Xiao, X.; Chen, Y.; et al. MicroRNA-29a-c decrease fasting blood glucose levels by negatively regulating hepatic gluconeogenesis. J. Hepatol. 2013, 58, 535–542. [Google Scholar] [CrossRef]
  152. Pandey, A.K.; Verma, G.; Vig, S.; Srivastava, S.; Srivastava, A.K.; Datta, M. miR-29a levels are elevated in the db/db mice liver and its overexpression leads to attenuation of insulin action on PEPCK gene expression in HepG2 cells. Mol. Cell. Endocrinol. 2011, 332, 125–133. [Google Scholar] [CrossRef]
  153. He, A.; Zhu, L.; Gupta, N.; Chang, Y.; Fang, F. Overexpression of Micro Ribonucleic Acid 29, Highly Up-Regulated in Diabetic Rats, Leads to Insulin Resistance in 3T3-L1 Adipocytes. Mol. Endocrinol. 2007, 21, 2785–2794. [Google Scholar] [CrossRef]
  154. Nielsen, L.B.; Wang, C.; Sørensen, K.; Bang-Berthelsen, C.H.; Hansen, L.; Andersen, M.-L.M.; Hougaard, P.; Juul, A.; Zhang, C.-Y.; Pociot, F.; et al. Circulating Levels of MicroRNA from Children with Newly Diagnosed Type 1 Diabetes and Healthy Controls: Evidence That miR-25 Associates to Residual Beta-Cell Function and Glycaemic Control during Disease Progression. Exp. Diabetes Res. 2012, 2012, 1–7. [Google Scholar] [CrossRef] [Green Version]
  155. Trajkovski, M.; Hausser, J.; Soutschek, J.; Bhat, B.; Akin, A.; Zavolan, M.; Heim, M.H.; Stoffel, M. MicroRNAs 103 and 107 regulate insulin sensitivity. Nat. Cell Biol. 2011, 474, 649–653. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Vinciguerra, M.; Sanchez, P.C. Molecular Nutrition: Mother and Infant; Academic Press: Cambridge, MA, USA, 2020. [Google Scholar]
  157. Khorram, O.; Chuang, T.D.; Pearce, W.J. Long-term effects of maternal undernutrition on offspring carotid artery remodeling: Role of miR-29c. J. Dev. Orig. Health Dis. 2015, 6, 342–349. [Google Scholar] [CrossRef] [Green Version]
  158. Khorram, O.; Han, G.; Bagherpour, R.; Magee, T.R.; Desai, M.; Ross, M.G.; Chaudhri, A.A.; Toloubeydokhti, T.; Pearce, W.J. Effect of maternal undernutrition on vascular expression of micro and messenger RNA in newborn and aging offspring. Am. J. Physiol. Integr. Comp. Physiol. 2010, 298, R1366–R1374. [Google Scholar] [CrossRef]
  159. Maloyan, A.; Muralimanoharan, S.; Huffman, S.; Cox, L.A.; Nathanielsz, P.W.; Myatt, L.; Nijland, M.J. Identification and comparative analyses of myocardial miRNAs involved in the fetal response to maternal obesity. Physiol. Genom. 2013, 45, 889–900. [Google Scholar] [CrossRef] [Green Version]
  160. García-Segura, L.; Perezandrade, M.E.; Miranda-Ríos, J. The Emerging Role of MicroRNAs in the Regulation of Gene Expression by Nutrients. J. Nutr. Nutr. 2013, 6, 16–31. [Google Scholar] [CrossRef]
  161. Enquobahrie, D.A.; Williams, M.A.; Qiu, C.; Siscovick, D.S.; Sorensen, T.K. Global maternal early pregnancy peripheral blood mRNA and miRNA expression profiles according to plasma 25-hydroxyvitamin D concentrations. J. Matern. Neonatal Med. 2011, 24, 1002–1012. [Google Scholar] [CrossRef] [Green Version]
  162. Adaikalakoteswari, A.; Vatish, M.; Alam, M.T.; Ott, S.; Kumar, S.; Saravanan, P. Low Vitamin B12 in pregnancy is associated with adipose-derived circulating miRs Targeting PPAR γ and insulin resistance. J. Clin. Endocrinol. Metab. 2017, 102, 4200–4209. [Google Scholar] [CrossRef] [Green Version]
  163. Bladé, C.; Baselga-Escudero, L.; Salvado, M.J.; Arola-Arnal, A. mi RNA s, polyphenols, and chronic disease. Molec. Nutr. Food Res. 2013, 57, 58–70. [Google Scholar] [CrossRef]
  164. Johnson, I.T.; Scalbert, A.J.; Saltmarsh, M. Polyphenols: Antioxidants and beyond. Am. J. Clin. Nutr. 2005, 81, 215S–217S. [Google Scholar]
  165. Del Bas, J.M.; Crescenti, A.; Arola-Arnal, A.; Oms-Oliu, G.; Arola, L.; Caimari, A. Intake of grape procyanidins during gestation and lactation impairs reverse cholesterol transport and increases atherogenic risk indexes in adult offspring. J. Nutr. Biochem. 2015, 26, 1670–1677. [Google Scholar] [CrossRef]
  166. Victora, C.G.; Bahl, R.; Barros, A.J.D.; França, G.V.; Horton, S.; Krasevec, J.; Murch, S.; Sankar, M.J.; Walker, N.; Rollins, N.C.; et al. Breastfeeding in the 21st century: Epidemiology, mechanisms, and lifelong effect. Lancet 2016, 387, 475–490. [Google Scholar] [CrossRef] [Green Version]
  167. Obermann-Borst, S.A.; Eilers, P.H.; Tobi, E.W.; De Jong, F.H.; Slagboom, P.E.; Heijmans, B.T.; Steegers-Theunissen, R.P. Duration of breastfeeding and gender are associated with methylation of the LEPTIN gene in very young children. Pediatr. Res. 2013, 74, 344–349. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  168. Sherwood, W.B.; Bion, V.; Lockett, G.A.; Ziyab, A.H.; Soto-Ramírez, N.; Mukherjee, N.; Kurukulaaratchy, R.J.; Ewart, S.; Zhang, H.; Arshad, S.H.; et al. Duration of breastfeeding is associated with leptin (LEP) DNA methylation profiles and BMI in 10-year-old children. Clin. Epigenetics 2019, 11, 1–10. [Google Scholar] [CrossRef] [PubMed]
  169. Pauwels, S.; Symons, L.; Vanautgaerden, E.-L.; Ghosh, M.; Duca, R.C.; Bekaert, B.; Freson, K.; Huybrechts, I.; Langie, S.A.S.; Koppen, G.; et al. The Influence of the Duration of Breastfeeding on the Infant’s Metabolic Epigenome. Nutrients 2019, 11, 1408. [Google Scholar] [CrossRef] [Green Version]
  170. Wang, L.; Sadri, M.; Giraud, D.; Zempleni, J. RNase H2-Dependent Polymerase Chain Reaction and Elimination of Confounders in Sample Collection, Storage, and Analysis Strengthen Evidence That microRNAs in Bovine Milk Are Bioavailable in Humans. J. Nutr. 2018, 148, 153–159. [Google Scholar] [CrossRef] [Green Version]
  171. Sanwlani, R.; Fonseka, P.; Chitti, S.V.; Mathivanan, S. Milk-Derived Extracellular Vesicles in Inter-Organism, Cross-Species Communication and Drug Delivery. Proteomes 2020, 8, 11. [Google Scholar] [CrossRef]
  172. Cintio, M.; Polacchini, G.; Scarsella, E.; Montanari, T.; Stefanon, B.; Colitti, M. MicroRNA Milk Exosomes: From Cellular Regulator to Genomic Marker. Animals 2020, 10, 1126. [Google Scholar] [CrossRef]
  173. Alsaweed, M.; Hartmann, P.E.; Geddes, D.T.; Kakulas, F. MicroRNAs in Breastmilk and the Lactating Breast: Potential Immunoprotectors and Developmental Regulators for the Infant and the Mother. Int. J. Env.. Res. Public Heal. 2015, 12, 13981–14020. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Tomé-Carneiro, J.; Fernández-Alonso, N.; Tomás-Zapico, C.; Visioli, F.; Iglesias-Gutierrez, E.; Dávalos, A. Breast milk microRNAs harsh journey towards potential effects in infant development and maturation. Lipid encapsulation can help. Pharm. Res. 2018, 132, 21–32. [Google Scholar] [CrossRef] [PubMed]
  175. Zhou, Q.; Li, M.; Wang, X.; Li, Q.; Wang, T.; Zhou, X.; Wang, X.; Gao, X.; Li, X. Immune-related MicroRNAs are Abundant in Breast Milk Exosomes. Int. J. Biol. Sci. 2012, 8, 118–123. [Google Scholar] [CrossRef] [PubMed]
  176. Izumi, H.; Tsuda, M.; Sato, Y.; Kosaka, N.; Ochiya, T.; Iwamoto, H.; Namba, K.; Takeda, Y. Bovine milk exosomes contain microRNA and mRNA and are taken up by human macrophages. J. Dairy Sci. 2015, 98, 2920–2933. [Google Scholar] [CrossRef] [Green Version]
  177. Olkkonen, V.M.; Levine, T.P. Oxysterol binding proteins: In more than one place at one time? Biochem. Cell Biol. 2004, 82, 87–98. [Google Scholar] [CrossRef]
  178. Bravi, F.; Wiens, F.; DeCarli, A.; Pont, A.D.; Agostoni, C.; Ferraroni, M. Impact of maternal nutrition on breast-milk composition: A systematic review. Am. J. Clin. Nutr. 2016, 104, 646–662. [Google Scholar] [CrossRef] [Green Version]
  179. Pomar, C.A.; Sánchez, J.; Palou, A. The Intake of a Cafeteria Diet in Nursing Rats Alters the Breast Milk Concentration of Proteins Important for the Development of Offspring. Nutrients 2020, 12, 2470. [Google Scholar] [CrossRef]
  180. Zamanillo, R.; Sánchez, J.; Serra, F.; Palou, A. Breast Milk Supply of MicroRNA Associated with Leptin and Adiponectin Is Affected by Maternal Overweight/Obesity and Influences Infancy BMI. Nutrients 2019, 11, 2589. [Google Scholar] [CrossRef] [Green Version]
  181. Savini, I.; Gasperi, V.; Catani, V.M. Nutrigenetics. In eLS; John Wiley & Sons, Ltd.: Chichester, UK, 2016. [Google Scholar] [CrossRef]
  182. Al-Garawi, A.; Carey, V.J.; Chhabra, D.; Mirzakhani, H.; Morrow, J.; Lasky-Su, J.; Qiu, W.; Laranjo, N.; Litonjua, A.A.; Weiss, S.T. The Role of Vitamin D in the Transcriptional Program of Human Pregnancy. PLoS ONE 2016, 11, e0163832. [Google Scholar] [CrossRef]
  183. Brehm, J.M.; Celedón, J.C.; Soto-Quiros, M.E.; Avila, L.; Hunninghake, G.M.; Forno, E.; Laskey, D.; Sylvia, J.S.; Hollis, B.W.; Weiss, S.T.; et al. Serum Vitamin D Levels and Markers of Severity of Childhood Asthma in Costa Rica. Am. J. Respir. Crit. Care Med. 2009, 179, 765–771. [Google Scholar] [CrossRef] [Green Version]
  184. Liu, N.Q.; Ouyang, Y.; Bulut, Y.; Lagishetty, V.; Chan, S.Y.; Hollis, B.W.; Wagner, C.; Equils, O.; Hewison, M. Dietary Vitamin D Restriction in Pregnant Female Mice Is Associated With Maternal Hypertension and Altered Placental and Fetal Development. Endocrinology 2013, 154, 2270–2280. [Google Scholar] [CrossRef] [Green Version]
  185. Schulz, E.V.; Cruze, L.; Wei, W.; Gehris, J.; Wagner, C.L. Maternal vitamin D sufficiency and reduced placental gene expression in angiogenic biomarkers related to comorbidities of pregnancy. J. Steroid Biochem. Mol. Biol. 2017, 173, 273–279. [Google Scholar] [CrossRef]
  186. Lu, F.; Longo, M.; Tamayo, E.; Maner, W.; Al-Hendy, A.; Anderson, G.D.; Hankins, G.D.; Saade, G.R. The effect of over-expression of sFlt-1 on blood pressure and the occurrence of other manifestations of preeclampsia in unrestrained conscious pregnant mice. Am. J. Obs. Gynecol. 2007, 196, 396.e1–396.e7. [Google Scholar] [CrossRef]
  187. Moore, A.G.; Young, H.; Keller, J.M.; Ojo, L.R.; Yan, J.; Simas, T.A.M.; Maynard, S.E. Angiogenic biomarkers for prediction of maternal and neonatal complications in suspected preeclampsia. J. Matern. Neonatal Med. 2012, 25, 2651–2657. [Google Scholar] [CrossRef]
  188. Karumanchi, S.; Epstein, F. Placental ischemia and soluble fms-like tyrosine kinase 1: Cause or consequence of preeclampsia? Kidney Int. 2007, 71, 959–961. [Google Scholar] [CrossRef] [Green Version]
  189. Rana, S.; Powe, C.E.; Salahuddin, S.; Verlohren, S.; Perschel, F.H.; Levine, R.J.; Lim, K.-H.; Wenger, J.B.; Thadhani, R.; Karumanchi, S.A. Angiogenic Factors and the Risk of Adverse Outcomes in Women With Suspected Preeclampsia. Circulation 2012, 125, 911–919. [Google Scholar] [CrossRef] [Green Version]
  190. Wu, C.S.; Nohr, E.A.; Bech, B.H.; Vestergaard, M.; Catov, J.M.; Olsen, J. Health of children born to mothers who had preeclampsia: A population-based cohort study. Am. J. Obs. Gynecol. 2009, 201, 269.e1–269.e10. [Google Scholar] [CrossRef]
  191. Washburn, L.; Nixon, P.; Russell, G.; Snively, B.M.; O’Shea, T.M. Adiposity in Adolescent Offspring Born Prematurely to Mothers with Preeclampsia. J. Pediatr. 2013, 162, 912–917. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  192. Rodie, V.; Caslake, M.; Stewart, F.; Sattar, N.; Ramsay, J.; Greer, I.; Freeman, D. Fetal cord plasma lipoprotein status in uncomplicated human pregnancies and in pregnancies complicated by pre-eclampsia and intrauterine growth restriction. Atherosclerosis 2004, 176, 181–187. [Google Scholar] [CrossRef] [PubMed]
  193. Ophir, E.; Dourleshter, G.; Hirsh, Y.; Fait, V.; German, L.; Bornstein, J. Newborns of pre-eclamptic women: A biochemical difference present in utero. Acta Obs. Gynecol. Scand. 2006, 85, 1172–1178. [Google Scholar] [CrossRef]
  194. Guillemette, L.; Lacroix, M.; Allard, C.; Patenaude, J.; Battista, M.-C.; Doyon, M.; Moreau, J.; Ménard, J.; Ardilouze, J.-L.; Perron, P.; et al. Preeclampsia is associated with an increased pro-inflammatory profile in newborns. J. Reprod. Immunol. 2015, 112, 111–114. [Google Scholar] [CrossRef] [PubMed]
  195. Ghosh, S.; Collier, A. Churchill’s Pocketbook of Diabetes E-Book; Elsevier Health Sciences: Amsterdam, The Netherlands, 2021. [Google Scholar]
  196. Mirzaalian, Y.; Nourian, M.; Gholamalizadeh, M.; Doaei, S.; Hatami, M.; Hassanzadeh, A.; Askari, G.; Farahi, R. The association of quantitative insulin sensitivity indices (HOMA-IR and QUICKI) with anthropometric and cardiometabolic indicators in adolescents. Arch. Med. Sci. Atheroscler. Dis. 2019, 4, 32–37. [Google Scholar] [CrossRef] [PubMed]
  197. Iyer, N.V.; Kotch, L.E.; Agani, F.; Leung, S.W.; Laughner, E.; Wenger, R.H.; Gassmann, M.; Gearhart, J.D.; Lawer, A.M.; Yu, A.Y.; et al. Cellular and developmental control of O2 homeostasis by hypoxia-inducible factor 1α. Genes Dev. 1998, 12, 149–162. [Google Scholar] [CrossRef] [Green Version]
  198. Semenza, G.L.; Agani, F.; Iyer, N.; Kotch, L.; Laughner, E.; Leung, S.; Yu, A. Regulation of cardiovascular development and physiology by hypoxia-inducible factor. Ann. N.Y. Acad. Sci. 1999, 874, 262–268. [Google Scholar] [CrossRef]
  199. Zheng, J.; Xiao, X.; Zhang, Q.; Wang, T.; Yu, M.; Xu, J. Maternal Low-Protein Diet Modulates Glucose Metabolism and Hepatic MicroRNAs Expression in the Early Life of Offspring. Nutrients 2017, 9, 205. [Google Scholar] [CrossRef] [Green Version]
  200. Cai, D.; Jia, Y.; Lu, J.; Yuan, M.; Sui, S.; Song, H.; Zhao, R. Maternal dietary betaine supplementation modifies hepatic expression of cholesterol metabolic genes via epigenetic mechanisms in newborn piglets. Br. J. Nutr. 2014, 112, 1459–1468. [Google Scholar] [CrossRef] [Green Version]
  201. Cai, D.; Yuan, M.; Liu, H.; Han, Z.; Pan, S.; Yang, Y.; Zhao, R. Epigenetic and SP1-mediated regulation is involved in the repression of galactokinase 1 gene in the liver of neonatal piglets born to betaine-supplemented sows. Eur. J. Nutr. 2017, 56, 1899–1909. [Google Scholar] [CrossRef]
  202. Cheshmeh, S.; Nachvak, S.M.; Rezvani, N.; Saber, A. Effects of Breastfeeding and Formula Feeding on the Expression Level of FTO, CPT1A and PPAR-α Genes in Healthy Infants. Diabetes Metab. Syndr. Obes. Targets 2020, 13, 2227–2237. [Google Scholar] [CrossRef]
  203. Priego, T.; Sancher, J.; Pico, C.; Ahrens, W.; Bammann, K.; De Henauw, S.; Fraterman, A.; Iacoviello, L.; Lissner, L.; Molnar, D.; et al. Impact of breast-feeding compared to formula-feeding on blood-cell transcript-based potential biomarkers of health in children. Ann. Nutr. Metab. 2013, 63 (Suppl. 1), 225–226. [Google Scholar] [CrossRef]
Figure 1. The effect of mother’s nutrition during pregnancy on the fetus and offspring health has been associated with microbiome, genetics, epigenetics, and transcriptomics in humans and non-human models. A repeated number in the lists indicates the pathway to follow from the mother to fetus and to offspring in each omics separately. Numbers not indicated show that there is no stated association. Arrows: (↑) represents the increase, (↓) represents the decrease, (↑↓) represents the disturbance/imbalance, (→) represents the leading cause.
Figure 1. The effect of mother’s nutrition during pregnancy on the fetus and offspring health has been associated with microbiome, genetics, epigenetics, and transcriptomics in humans and non-human models. A repeated number in the lists indicates the pathway to follow from the mother to fetus and to offspring in each omics separately. Numbers not indicated show that there is no stated association. Arrows: (↑) represents the increase, (↓) represents the decrease, (↑↓) represents the disturbance/imbalance, (→) represents the leading cause.
Life 11 00467 g001
Figure 2. Precision nutrition applied to pregnancy to understand the mechanisms that lead to NCD in childhood and adult life. Created with BioRender.com (accessed on 7 May 2021).
Figure 2. Precision nutrition applied to pregnancy to understand the mechanisms that lead to NCD in childhood and adult life. Created with BioRender.com (accessed on 7 May 2021).
Life 11 00467 g002
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Alabduljabbar, S.; Zaidan, S.A.; Lakshmanan, A.P.; Terranegra, A. Personalized Nutrition Approach in Pregnancy and Early Life to Tackle Childhood and Adult Non-Communicable Diseases. Life 2021, 11, 467. https://doi.org/10.3390/life11060467

AMA Style

Alabduljabbar S, Zaidan SA, Lakshmanan AP, Terranegra A. Personalized Nutrition Approach in Pregnancy and Early Life to Tackle Childhood and Adult Non-Communicable Diseases. Life. 2021; 11(6):467. https://doi.org/10.3390/life11060467

Chicago/Turabian Style

Alabduljabbar, Shaikha, Sara Al Zaidan, Arun Prasath Lakshmanan, and Annalisa Terranegra. 2021. "Personalized Nutrition Approach in Pregnancy and Early Life to Tackle Childhood and Adult Non-Communicable Diseases" Life 11, no. 6: 467. https://doi.org/10.3390/life11060467

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop