Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Nutrients ; 13(11)2021 Oct 28.
Article in English | MEDLINE | ID: mdl-34836113

ABSTRACT

Iron deficiency (ID) anemia is the foremost micronutrient deficiency worldwide, affecting around 40% of pregnant women and young children. ID during the prenatal and early postnatal periods has a pronounced effect on neurodevelopment, resulting in long-term effects such as cognitive impairment and increased risk for neuropsychiatric disorders. Treatment of ID has been complicated as it does not always resolve the long-lasting neurodevelopmental deficits. In animal models, developmental ID results in abnormal hippocampal structure and function associated with dysregulation of genes involved in neurotransmission and synaptic plasticity. Dysregulation of these genes is a likely proximate cause of the life-long deficits that follow developmental ID. However, a direct functional link between iron and gene dysregulation has yet to be elucidated. Iron-dependent epigenetic modifications are one mechanism by which ID could alter gene expression across the lifespan. The jumonji and AT-rich interaction domain-containing (JARID) protein and the Ten-Eleven Translocation (TET) proteins are two families of iron-dependent epigenetic modifiers that play critical roles during neural development by establishing proper gene regulation during critical periods of brain development. Therefore, JARIDs and TETs can contribute to the iron-mediated epigenetic mechanisms by which early-life ID directly causes stable changes in gene regulation across the life span.


Subject(s)
Anemia, Iron-Deficiency/genetics , Epigenesis, Genetic/physiology , Hippocampus/metabolism , Infant Nutritional Physiological Phenomena/genetics , Maternal Nutritional Physiological Phenomena/genetics , Anemia, Iron-Deficiency/complications , Animals , Animals, Newborn , Child Development/physiology , Epigenomics , Female , Hippocampus/growth & development , Humans , Infant , Infant, Newborn , Neurodevelopmental Disorders/genetics , Neurogenesis/physiology , Neuronal Plasticity/physiology , Pregnancy , Prenatal Exposure Delayed Effects/genetics , Synaptic Transmission/physiology
2.
Nutrients ; 13(8)2021 Aug 22.
Article in English | MEDLINE | ID: mdl-34445048

ABSTRACT

This study evaluates the prevalence of autistic behaviors in fragile X syndrome as a function of infant diet. Retrospective survey data from the Fragile X Syndrome Nutrition Study, which included data on infant feeding and caregiver-reported developmental milestones for 190 children with fragile X syndrome enrolled in the Fragile X Online Registry with Accessible Database (FORWARD), were analyzed. Exploratory, sex-specific associations were found linking the use of soy-based infant formula with worse autistic behaviors related to language in females and self-injurious behavior in males. These findings prompt prospective evaluation of the effects of soy-based infant formula on disease comorbidities in fragile X syndrome, a rare disorder for which newborn screening could be implemented if there was an intervention. Gastrointestinal problems were the most common reason cited for switching to soy-based infant formula. Thus, these findings also support the study of early gastrointestinal problems in fragile X syndrome, which may underly the development and severity of disease comorbidities. In conjunction with comorbidity data from the previous analyses of the Fragile X Syndrome Nutrition Study, the findings indicate that premutation fragile X mothers should be encouraged to breastfeed.


Subject(s)
Autism Spectrum Disorder/epidemiology , Feeding Behavior/psychology , Fragile X Syndrome/psychology , Infant Formula/statistics & numerical data , Infant Nutritional Physiological Phenomena/genetics , Adolescent , Autism Spectrum Disorder/genetics , Comorbidity , Female , Fragile X Syndrome/physiopathology , Gastrointestinal Diseases/epidemiology , Gastrointestinal Diseases/genetics , Humans , Infant , Male , Nutrition Surveys , Parents , Prevalence , Retrospective Studies
3.
Nutrients ; 12(11)2020 Oct 29.
Article in English | MEDLINE | ID: mdl-33137917

ABSTRACT

BACKGROUND: Breastfeeding is associated with short and long-term health benefits. Long-term effects might be mediated by epigenetic mechanisms, yet the literature on this topic is scarce. We performed the first epigenome-wide association study of infant feeding, comparing breastfed vs non-breastfed children. We measured DNA methylation in children from peripheral blood collected in childhood (age 7 years, N = 640) and adolescence (age 15-17 years, N = 709) within the Accessible Resource for Integrated Epigenomic Studies (ARIES) project, part of the larger Avon Longitudinal Study of Parents and Children (ALSPAC) cohort. Cord blood methylation (N = 702) was used as a negative control for potential pre-natal residual confounding. RESULTS: Two differentially-methylated sites presented directionally-consistent associations with breastfeeding at ages 7 and 15-17 years, but not at birth. Twelve differentially-methylated regions in relation to breastfeeding were identified, and for three of them there was evidence of directional concordance between ages 7 and 15-17 years, but not between birth and age 7 years. CONCLUSIONS: Our findings indicate that DNA methylation in childhood and adolescence may be predicted by breastfeeding, but further studies with sufficiently large samples for replication are required to identify robust associations.


Subject(s)
Breast Feeding , DNA Methylation/physiology , Eating/genetics , Infant Nutritional Physiological Phenomena/genetics , Longevity/genetics , Adolescent , Bottle Feeding , Child , Child, Preschool , Epigenesis, Genetic , Epigenomics , Female , Fetal Blood/metabolism , Genome-Wide Association Study , Humans , Infant , Infant, Newborn , Longitudinal Studies , Male
4.
Nutrients ; 11(12)2019 Dec 05.
Article in English | MEDLINE | ID: mdl-31817318

ABSTRACT

Despite constant research and public policy efforts, the obesity epidemic continues to be a major public health threat, and new approaches are urgently needed. It has been shown that nutrient imbalance in early life, from conception to infancy, influences later obesity risk, suggesting that obesity could result from "developmental programming". In this review, we evaluate the possibility that early postnatal nutrition programs obesity risk via epigenetic mechanisms, especially DNA methylation, focusing on four main topics: (1) the dynamics of epigenetic processes in key metabolic organs during the early postnatal period; (2) the epigenetic effects of alterations in early postnatal nutrition in animal models or breastfeeding in humans; (3) current limitations and remaining outstanding questions in the field of epigenetic programming; (4) candidate pathways by which early postnatal nutrition could epigenetically program adult body weight set point. A particular focus will be given to the potential roles of breast milk fatty acids, neonatal metabolic and hormonal milieu, and gut microbiota. Understanding the mechanisms by which early postnatal nutrition can promote lifelong metabolic modifications is essential to design adequate recommendations and interventions to "de-program" the obesity epidemic.


Subject(s)
Epigenesis, Genetic/genetics , Infant Nutritional Physiological Phenomena/genetics , Pediatric Obesity/genetics , Animals , Animals, Newborn , Breast Feeding , Cellular Reprogramming/genetics , Child Development , DNA Methylation/genetics , Energy Metabolism/genetics , Female , Follow-Up Studies , Humans , Infant , Infant, Newborn , Male , Maternal Nutritional Physiological Phenomena , Obesity/genetics
5.
Pediatr Res ; 85(6): 822-829, 2019 05.
Article in English | MEDLINE | ID: mdl-30791043

ABSTRACT

BACKGROUND: The aim of this study was to evaluate the direct effects of matrix metalloproteinase (MMP9 rs17577, MMP9 rs17576) and alfa 2 adrenergic receptor (ADRA2A rs553668) gene polymorphisms investigated in mothers and their newborns on maternal weight gain (MWG) during pregnancy and the newborn's birth weight (BW), taking into account the presence of other related factors. METHODS: We performed a cross-sectional study in 197 mother-newborn pairs in an Obstetrics Gynecology Clinic, in order to evaluate the demographic and anthropometric parameters, and gene polymorphism. RESULTS: BW was positively correlated with maternal age (p = 0.021) and the educational level (p = 0.002), and negatively correlated with smoking status in pregnant women (p < 0.001). The MMP9 rs17577 variant genotypes in mothers led to a lower BW (p = 0.049). The mothers with a variant genotype of ADRA2A rs553668 gene polymorphism had newborns with a higher BW (p = 0.030). MWG and gestational age (GesAge) influenced BW (p < 0.05). We noticed that newborns' variant genotype of MMP9 rs17577 was related to a significant increase in BW (p = 0.010), while the newborns who carried the variant genotype of MMP9 rs17576 expressed a negative correlation, decreasing the BW (p = 0.032). CONCLUSION: Our study emphasizes the role of MMP9 rs17577, MMP9 rs17576, and ADRA2A rs553668 SNPs in BW determinism.


Subject(s)
Matrix Metalloproteinase 9/genetics , Nutritional Status/genetics , Receptors, Adrenergic, alpha-2/genetics , Adult , Birth Weight/genetics , Cross-Sectional Studies , Female , Gestational Weight Gain/genetics , Humans , Infant Nutritional Physiological Phenomena/genetics , Infant, Newborn , Male , Maternal Nutritional Physiological Phenomena/genetics , Maternal-Fetal Exchange/genetics , Models, Genetic , Polymorphism, Single Nucleotide , Pregnancy , Romania , Young Adult
6.
J Pediatr Gastroenterol Nutr ; 64(3): 446-453, 2017 03.
Article in English | MEDLINE | ID: mdl-27276431

ABSTRACT

BACKGROUND: Infants who are not breast-fed benefit from formula with both docosahexaenoic acid (C22:6n3) and arachidonic acid (ARA; C20:4n6). The amount of ARA needed to support immune function is unknown. Infants who carry specific fatty acid desaturase (FADS) polymorphisms may require more dietary ARA to maintain adequate ARA status. OBJECTIVE: The aim of the study was to determine whether ARA intake or FADS polymorphisms alter ARA levels of lymphocytes, plasma, and red blood cells in term infants fed infant formula. METHODS: Infants (N = 89) were enrolled in this prospective, double-blind controlled study. Infants were randomized to consume formula containing 17 mg docosahexaenoic acid and 0, 25, or 34 mg ARA/100 kcal for 10 weeks. Fatty acid composition of plasma phosphatidylcholine and phosphatidylethanolamine, total fatty acids of lymphocytes and red blood cells, activation markers of lymphocytes, and polymorphisms in FADS1 and FADS2 were determined. RESULTS: Lymphocyte ARA was higher in the 25-ARA formula group than in the 0- or 34-ARA groups. In plasma, 16:0/20:4 and 18:0/20:4 species of phosphatidylcholine and phosphatidylethanolamine were highest and 16:0/18:2 and 18:0/18:2 were lowest in the 34-ARA formula group. In minor allele carriers of FADS1 and FADS2, plasma ARA content was elevated only at the highest level of ARA consumed. B-cell activation marker CD54 was elevated in infants who consumed formula containing no ARA. CONCLUSIONS: ARA level in plasma is reduced by low ARA consumption and by minor alleles in FADS. Dietary ARA may exert an immunoregulatory role on B-cell activation by decreasing 16:0/18:2 and 18:0/18:2 species of phospholipids. ARA intake from 25 to 34 mg/100 kcal is sufficient to maintain cell ARA level in infants across genotypes.


Subject(s)
Arachidonic Acid/administration & dosage , B-Lymphocytes/metabolism , Fatty Acid Desaturases/genetics , Infant Formula/chemistry , Infant Nutritional Physiological Phenomena/genetics , Lymphocyte Activation , Arachidonic Acid/blood , Biomarkers/blood , Delta-5 Fatty Acid Desaturase , Docosahexaenoic Acids/administration & dosage , Double-Blind Method , Follow-Up Studies , Genetic Markers , Humans , Infant , Infant, Newborn , Intention to Treat Analysis , Polymorphism, Genetic , Prospective Studies
8.
Pediatr Res ; 77(1-2): 263-9, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25314584

ABSTRACT

Arachidonic acid (AA) is supplied together with docosahexaenoic acid (DHA) in infant formulas, but we have limited knowledge about the effects of supplementation with either of these long-chain polyunsaturated fatty acids (LCPUFA) on growth and developmental outcomes. AA is present in similar levels in breast milk throughout the world, whereas the level of DHA is highly diet dependent. Autopsy studies show similar diet-dependent variation in brain DHA, whereas AA is little affected by intake. Early intake of DHA has been shown to affect visual development, but the effect of LCPUFA on neurodevelopment remains to be established. Few studies have found any functional difference between infants supplemented with DHA alone compared to DHA+AA, but some studies show neurodevelopmental advantages in breast-fed infants of mothers supplemented with n-3 LCPUFA alone. It also remains to be established whether the AA/DHA balance could affect allergic and inflammatory outcomes later in life. Disentangling effects of genetic variability and dietary intake on AA and DHA-status and on functional outcomes may be an important step in the process of determining whether AA-intake is of any physiological or clinical importance. However, based on the current evidence we hypothesize that dietary AA plays a minor role on growth and development relative to the impact of dietary DHA.


Subject(s)
Arachidonic Acid/pharmacology , Child Development/drug effects , Dietary Supplements , Genetic Variation , Infant Nutritional Physiological Phenomena/physiology , Perinatal Care/methods , Arachidonic Acid/administration & dosage , Brain/metabolism , Female , Humans , Infant Nutritional Physiological Phenomena/genetics , Infant, Newborn , Maternal-Fetal Exchange/physiology , Milk, Human/chemistry , Pregnancy
9.
Early Hum Dev ; 90 Suppl 2: S23-4, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25220121

ABSTRACT

Epigenetic changes have long-lasting effects on gene expression and are related to, and often induced by, the environment in which early development takes place. In particular, the period of development that extends from pre-conception to early infancy is the period of life during which epigenetic DNA imprinting activity is the most active. Epigenetic changes have been associated with modification of the risk for developing a wide range of adulthood, non-communicable diseases (including cardiovascular diseases, metabolic diseases, diseases of the reproductive system, etc.). This paper reviews the molecular basis of epigenetics, and addresses the issues related to the process of developmental programming of the various areas of human health.


Subject(s)
Epigenesis, Genetic , Infant Nutritional Physiological Phenomena/genetics , Humans , Infant , Infant, Newborn , Nutrigenomics
10.
Proc Nutr Soc ; 71(2): 276-83, 2012 May.
Article in English | MEDLINE | ID: mdl-22390978

ABSTRACT

The huge health burden accompanying obesity is not only attributable to inadequate dietary and sedentary lifestyle habits, since a predisposing genetic make-up and other putative determinants concerning easier weight gain and fat deposition have been reported. Thus, several investigations aiming to understand energy metabolism and body composition maintenance have been performed considering the participation of perinatal nutritional programming and epigenetic processes as well as inflammation phenomena. The Developmental Origins of Health and Disease hypothesis and inheritance-oriented investigations concerning gene-nutrient interactions on energy homoeostasis and metabolic functions have suggested that inflammation could be not only a comorbidity of obesity but also a cause. There are several examples about the role of nutritional interventions in pregnancy and lactation, such as energetic deprivation, protein restriction and excess fat, which determine a cluster of disorders affecting energy efficiency in the offspring as well as different metabolic pathways, which are mediated by epigenetics encompassing the chromatin information encrypted by DNA methylation patterns, histone covalent modifications and non-coding RNA or microRNA. Epigenetic mechanisms may be boosted or impaired by dietary and environmental factors in the mother, intergenerationally or transiently transmitted, and could be involved in the obesity and inflammation susceptibility in the offspring. The aims currently pursued are the early identification of epigenetic biomarkers concerned in individual's disease susceptibility and the description of protocols for tailored dietary treatments/advice to counterbalance adverse epigenomic events. These approaches will allow diagnosis and prognosis implementation and facilitate therapeutic strategies in a personalised 'epigenomically modelled' manner to combat obesity and inflammation.


Subject(s)
Diet , Energy Metabolism , Epigenesis, Genetic , Inflammation , Maternal Nutritional Physiological Phenomena , Obesity/etiology , Prenatal Exposure Delayed Effects , Body Composition , Female , Fetal Development/genetics , Genetic Predisposition to Disease , Humans , Infant , Infant Nutritional Physiological Phenomena/genetics , Inflammation/genetics , Inflammation/metabolism , Lactation , Maternal Nutritional Physiological Phenomena/genetics , Obesity/genetics , Obesity/metabolism , Pregnancy , Prenatal Exposure Delayed Effects/genetics
11.
Nutr Res Rev ; 24(2): 198-205, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22008232

ABSTRACT

A growing number of studies focusing on the developmental origin of health and disease hypothesis have identified links among early nutrition, epigenetic processes and diseases also in later life. Different epigenetic mechanisms are elicited by dietary factors in early critical developmental ages that are able to affect the susceptibility to several diseases in adulthood. The studies here reviewed suggest that maternal and neonatal diet may have long-lasting effects in the development of non-communicable chronic adulthood diseases, in particular the components of the so-called metabolic syndrome, such as insulin resistance, type 2 diabetes, obesity, dyslipidaemia, hypertension, and CVD. Both maternal under- and over-nutrition may regulate the expression of genes involved in lipid and carbohydrate metabolism. Early postnatal nutrition may also represent a vital determinant of adult health by making an impact on the development and function of gut microbiota. An inadequate gut microbiota composition and function in early life seems to account for the deviant programming of later immunity and overall health status. In this regard probiotics, which have the potential to restore the intestinal microbiota balance, may be effective in preventing the development of chronic immune-mediated diseases. More recently, the epigenetic mechanisms elicited by probiotics through the production of SCFA are hypothesised to be the key to understand how they mediate their numerous health-promoting effects from the gut to the peripheral tissues.


Subject(s)
Diet , Epigenesis, Genetic , Gene Expression Regulation , Metabolic Syndrome/genetics , Nutritional Physiological Phenomena/genetics , Pregnancy Complications/genetics , Probiotics/therapeutic use , Female , Gastrointestinal Tract/immunology , Gastrointestinal Tract/microbiology , Humans , Immune System Diseases/prevention & control , Infant Nutritional Physiological Phenomena/genetics , Infant, Newborn , Malnutrition/genetics , Nutritional Status , Pregnancy , Prenatal Exposure Delayed Effects/genetics , Prenatal Nutritional Physiological Phenomena/genetics
12.
Curr Opin Clin Nutr Metab Care ; 13(3): 284-93, 2010 May.
Article in English | MEDLINE | ID: mdl-20375884

ABSTRACT

PURPOSE OF REVIEW: The ways in which epigenetic modifications fix the effects of early environmental events, ensuring sustained responses to transient stimuli, which result into modified gene expression patterns and phenotypes later in life, is a topic of considerable interest. This review focuses on recently discovered mechanisms and calls into question prevailing views about the dynamics, positions and functions of relevant epigenetic marks. RECENT FINDINGS: Animal models, including mice, rats, sheep, pigs and rabbits, remain a vital tool for studying the influence of early nutritional events on adult health and disease. Most epigenetic studies have addressed the long-term effects on a small number of epigenetic marks, at the global or individual gene level, of environmental stressors in humans and animal models. They have demonstrated the existence of a self-propagating epigenetic cycle. In parallel, an increasing number of studies based on high-throughput technologies and focusing on humans and mice have revealed additional complexity in epigenetic processes, by highlighting the importance of crosstalk between the different epigenetic marks. In recent months, a number of studies focusing on the developmental origin of health and disease and metabolic programming have identified links between early nutrition, epigenetic processes and long-term illness. SUMMARY: Despite recent progress, we are still far from understanding how, when and where environmental stressors disturb key epigenetic mechanisms. Thus, identifying the original key marks and their changes throughout development, during an individual's lifetime or over several generations, remains a challenging issue.


Subject(s)
Epigenesis, Genetic , Fetal Development/genetics , Gene Expression Regulation, Developmental , Gene Expression , Infant Nutritional Physiological Phenomena/genetics , Prenatal Nutritional Physiological Phenomena/genetics , Adult , Animals , DNA Methylation , Female , Genetic Predisposition to Disease , Histones , Humans , Infant , Phenotype , Pregnancy
13.
Pediatr Med Chir ; 31(2): 65-71, 2009.
Article in Italian | MEDLINE | ID: mdl-19642498

ABSTRACT

Many adult diseases seem to be associated with early nutrition and the subsequent growth pattern. Epidemiological studies hypotized that babies with intrauterine and/or neonatal growth retardation may be at greater risk of metabolic syndrome later in life. According to the Barker's "thrifty phenotype hypotesis" early malnutrition, whereas inducing physiological compensation by the promotion of early survival, appears to confer greater susceptibility to adults diseases. Epigenetics, that is the interindividual variation in DNA methylation patterns and chromatin remodelling, provide a potential explanation for how environmental factors can modify the risk for development of many common diseases. Beginning from animal models, many studies concerning early nutrition, epigenetic modifications and genes expression have been carried out. Maternal undernutrition during pregnancy, especially in the peri-implantation period, not only causes a prolonged growth retardation but also modifies the programming of biochemical mechanisms related to endocrine-metabolic control. Human studies have demonstrated the role played by IGF-1 as indicator of nutritional status and fetal/postnatal growth retardation. It has been reported that alterations in IGF axis, which predispose to adults diseases, may be due to an alterated epigenetic regulation that can modify IGF expression. Despite the critical inter-relation between early nutrition, growth, development, and subsequent health, there are few data on the influence of early nutrition on the modifications of the epigenetic gear. Furthermore it is hoped for a bigger attention to the early nutrition to prevent the development of diseases later in life.


Subject(s)
Epigenesis, Genetic , Gene Expression Regulation, Developmental , Insulin-Like Growth Factor II/genetics , Insulin-Like Growth Factor I/genetics , Prenatal Nutritional Physiological Phenomena/genetics , Adult , Animals , Evidence-Based Medicine , Female , Fetal Growth Retardation/genetics , Fetal Growth Retardation/prevention & control , Genetic Markers/genetics , Humans , Infant Nutritional Physiological Phenomena/genetics , Infant, Newborn , Malnutrition/genetics , Malnutrition/prevention & control , Metabolic Syndrome/genetics , Metabolic Syndrome/prevention & control , Phenotype , Pregnancy , Risk Factors , Transcription, Genetic
15.
Adv Exp Med Biol ; 646: 95-104, 2009.
Article in English | MEDLINE | ID: mdl-19536667

ABSTRACT

Breast milk is practically the only food eaten during the first months of life in fully breastfed infants and it is assumed to match the nutritional needs during these first months of postnatal life. Breastfeeding compared with infant formula feeding confers protection against several metabolic and physiological changes later on in life and, particularly, against obesity and related medical complications. Recent data from our laboratory, identifying leptin as the first specific compound responsible for these beneficial effects, are reviewed and discussed.


Subject(s)
Breast Feeding , Infant Nutritional Physiological Phenomena/physiology , Leptin/physiology , Maternal Nutritional Physiological Phenomena/physiology , Obesity/prevention & control , Adult , Animals , Female , Humans , Infant , Infant Nutritional Physiological Phenomena/genetics , Infant, Newborn , Leptin/genetics , Leptin/metabolism , Maternal Nutritional Physiological Phenomena/genetics , Obesity/genetics , Rats
16.
Nestle Nutr Workshop Ser Pediatr Program ; 62: 1-9; disucssion 9-12, 2008.
Article in English | MEDLINE | ID: mdl-18626189

ABSTRACT

Genetic research has focused on identifying linkages between polymorphisms and phenotypic traits to explain variations in complex biologies. However, the magnitude of these linkages has not been particularly high. Conversely, the ability of developmental plasticity to generate biological variation from one genotype is well understood, while interest has emerged in the clinical significance of epigenetic processes, particularly those influenced by the external environment. Environmental cues in early development may induce responses that provide adaptive advantage later in life. The benefit of such responses depends on the fidelity of the prediction of the future environment. Life history and physiological changes mediated through epigenetic processes then follow, determining the later phenotype. Developmental mismatch, leading to disease, can arise from discordance between the fetal environment, which is relatively constant across generations, and the postnatal nutritional environment, which can change drastically within and between generations. Metabolic disorders represent the outcome of an individual living in an energetically inappropriate environment. Experimental and clinical evidence suggests that individual capacity to live in a given energetic environment is influenced by developmental factors acting through epigenetic mechanisms. Epigenetic biomarkers may be able to identify a risk of developmental mismatch and thus offer the opportunity for nutritional or other intervention.


Subject(s)
Child Development/physiology , Genetic Variation , Infant Nutritional Physiological Phenomena/physiology , Nutritional Requirements , Prenatal Nutritional Physiological Phenomena/physiology , Chronic Disease/epidemiology , Epigenesis, Genetic , Female , Genetic Linkage , Humans , Infant , Infant Nutritional Physiological Phenomena/genetics , Infant, Newborn , Nutrition Disorders/etiology , Nutrition Disorders/genetics , Pregnancy , Prenatal Exposure Delayed Effects , Prenatal Nutritional Physiological Phenomena/genetics
17.
Nestle Nutr Workshop Ser Pediatr Program ; 62: 239-49; discussion 249-52, 2008.
Article in English | MEDLINE | ID: mdl-18626204

ABSTRACT

Current nutrition recommendations, directed towards populations, are based on estimated average nutrient requirements for a target population and intend to meet the needs of most individuals within that population. They also aim at preventing common diseases such as obesity, diabetes and cardiovascular disease. For infants with specific genetic polymorphisms, e.g. some inborn errors of metabolism, adherence to current recommendations will cause disease symptoms and they need personalized nutrition recommendations. Some other monogenic polymorphisms, e.g. adult hypolactasia, are common but with varying prevalence between ethnic groups and within populations. Ages at onset as well as the degree of the resulting lactose intolerance also vary, making population-based as well as personalized recommendations difficult. The tolerable intake is best set by each individual based on symptoms. For polygenetic diseases such as celiac disease, type-1 diabetes and allergic disease, current knowledge is insufficient to suggest personalized recommendations aiming at primary prevention for all high-risk infants, although it may be justified to provide such recommendations on an individual level should the parents ask for them. New technologies such as nutrigenetics and nutrigenomics are promising tools with which current nutrition recommendations can possibly be refined and the potential of individualized nutrition be explored. It seems likely that in the future it will be possible to offer more subgroups within a population personalized recommendations.


Subject(s)
Chronic Disease/prevention & control , Diet Therapy/standards , Infant Nutrition Disorders/prevention & control , Infant Nutritional Physiological Phenomena/physiology , Nutrigenomics/methods , Ethnicity , Food, Organic , Genetic Predisposition to Disease , Humans , Infant , Infant Nutrition Disorders/genetics , Infant Nutritional Physiological Phenomena/genetics , Infant, Newborn , Nutrition Policy , Nutritional Requirements , Risk Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...