Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Nutr Res ; 35(11): 939-47, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26439440

ABSTRACT

Polyunsaturated fatty acid metabolism in toddlers is regulated by a complex network of interacting factors. The contribution of maternal genetic and epigenetic makeup to this milieu is not well understood. In a cohort of mothers and toddlers 16 months of age (n = 65 mother-child pairs), we investigated the association between maternal genetic and epigenetic fatty acid desaturase 2 (FADS2) profiles and toddlers' n-6 and n-3 fatty acid metabolism. FADS2 rs174575 variation and DNA methylation status were interrogated in mothers and toddlers, as well as food intake and plasma fatty acid concentrations in toddlers. A multivariate fit model indicated that maternal rs174575 genotype, combined with DNA methylation, can predict α-linolenic acid plasma concentration in all toddlers and arachidonic acid concentrations in boys. Arachidonic acid intake was predictive for its plasma concentration in girls, whereas intake of 3 major n-3 species (eicosapentaenoic, docosapentaenoic, and docosahexaenoic acids) were predictive for their plasma concentrations in boys. FADS2 genotype and DNA methylation in toddlers were not related to plasma concentrations or food intakes, except for CpG8 methylation. Maternal FADS2 methylation was a predictor for the boys' α-linolenic acid intakes. This exploratory study suggests that maternal FADS2 genetic and epigenetic status could be related to toddlers' polyunsaturated fatty acid metabolism.


Subject(s)
DNA Methylation/genetics , Epigenesis, Genetic/genetics , Fatty Acid Desaturases/genetics , Fatty Acids, Omega-3/genetics , Fatty Acids, Unsaturated/genetics , Mothers , Female , Humans , Infant , Male
2.
Nutr Res ; 35(11): 948-55, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26455892

ABSTRACT

Maternal transfer of fatty acids is important to fetal brain development. The prenatal environment may differentially affect the substrates supporting declarative memory abilities, as the level of fatty acids transferred across the placenta may be affected by the maternal fatty acid desaturase 2 (FADS2) rs174575 single nucleotide polymorphism. In this study, we hypothesized that toddler and maternal rs174575 genotype and FADS2 promoter methylation would be related to the toddlers' declarative memory performance. Seventy-one 16-month-old toddlers participated in an imitation paradigm designed to test immediate and long-term declarative memory abilities. FADS2 rs174575 genotype was determined and FADS2 promoter methylation was quantified from blood by bisulfite pyrosequencing for the toddlers and their natural mothers. Toddlers of GG mothers at the FADS2 rs174575 single nucleotide polymorphism did not perform as well on memory assessments as toddlers of CC or CG mothers when controlling for plasma α-linolenic acid and child genotype. Toddler methylation status was related to immediate memory performance, whereas maternal methylation status was related to delayed memory performance. Thus, prenatal experience and maternal FADS2 status have a pervasive, long-lasting influence on the brain development of the offspring, but as the postnatal environment becomes more primary, the offsprings' own biology begins to have an effect.


Subject(s)
Cognition , DNA Methylation/genetics , Epigenesis, Genetic/genetics , Fatty Acid Desaturases/genetics , Fatty Acids, Omega-3/genetics , Mothers , Female , Genotype , Humans , Infant , Male
3.
Int J Dev Neurosci ; 36: 38-44, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24866706

ABSTRACT

Many animal and human studies indicated that dietary ω-3 fatty acids could have beneficial roles on brain development, memory, and learning. However, the exact mechanisms involved are far from being clearly understood, especially for α-linolenic acid (ALA), which is the precursor for the ω-3 elongation and desaturation pathways. This study investigated the alterations induced by different intakes of flaxseed oil (containing 50% ALA), during gestation and lactation, upon the expression of genes involved in neurogenesis, memory-related molecular processes, and DNA methylation, in the brains of mouse offspring at the end of lactation (postnatal day 19, P19). In addition, DNA methylation status for the same genes was investigated. Maternal flaxseed oil supplementation during lactation increased the expression of Mecp2, Ppp1cc, and Reelin, while decreasing the expression of Ppp1cb and Dnmt3a. Dnmt1 expression was decreased by postnatal flaxseed oil supplementation but this effect was offset by ALA deficiency during gestation. Mecp2 DNA methylation was decreased by maternal ALA deficiency during gestation, with a more robust effect in the lactation-deficient group. In addition, linear regression analysis revealed positive correlations between Mecp2, Reelin, and Ppp1cc, between Gadd45b, Bdnf, and Creb1, and between Egr1 and Dnmt1, respectively. However, there were no correlations, in any gene, between DNA methylation and gene expression. In summary, the interplay between ALA availability during gestation and lactation differentially altered the expression of genes involved in neurogenesis and memory, in the whole brain of the offspring at the end of lactation. The Mecp2 epigenetic status was correlated with ALA availability during gestation. However, the epigenetic status of the genes investigated was not associated with transcript levels, suggesting that either the regulation of these genes is not necessarily under epigenetic control, or that the whole brain model is not adequate for the exploration of epigenetic regulation in the context of this study.


Subject(s)
Brain , DNA Methylation/drug effects , Epigenesis, Genetic/drug effects , Methyl-CpG-Binding Protein 2/genetics , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/physiopathology , alpha-Linolenic Acid/toxicity , Analysis of Variance , Animals , Antigens, Differentiation/genetics , Antigens, Differentiation/metabolism , Brain/drug effects , Brain/growth & development , Brain/metabolism , Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/metabolism , Female , Male , Memory/drug effects , Methyl-CpG-Binding Protein 2/metabolism , Mice , Mice, Inbred C57BL , Pregnancy , Reelin Protein , Statistics as Topic
4.
FASEB J ; 28(7): 2970-8, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24671709

ABSTRACT

Effect alleles (alleles with a polymorphism that is associated with the effect being measured) in a small number of single-nucleotide polymorphisms (SNPs) are known to influence the dietary requirement for choline. In this study, we examined a much larger number of SNPs (n=200) in 10 genes related to choline metabolism for associations with development of organ dysfunction (liver or muscle) when 79 humans were fed a low-choline diet. We confirmed that effect alleles in SNPs such as the C allele of PEMT rs12325817 increase the risk of developing organ dysfunction in women when they consume a diet low in choline, and we identified novel effect alleles, such as the C allele of CHKA SNP rs7928739, that alter dietary choline requirements. When fed a low-choline diet, some people presented with muscle damage rather than liver damage; several effect alleles in SLC44A1 (rs7873937, G allele; rs2771040, G; rs6479313, G; rs16924529, A; and rs3199966, C) and one in CHKB (rs1557502, A) were more common in these individuals. This suggests that pathways related to choline metabolism are more important for normal muscle function than previously thought. In European, Mexican, and Asian Americans, and in individuals of African descent, we examined the prevalence of the effect alleles in SNPs that alter choline requirement and found that they are differentially distributed among people of different ethnic and racial backgrounds. Overall, our study has identified novel genetic variants that modulate choline requirements and suggests that the dietary requirement for choline may be different across racial and ethnic groups.-Da Costa, K.-A., Corbin, K. D., Niculescu, M. D., Galanko, J. A., Zeisel, S. H. Identification of new genetic polymorphisms that alter the dietary requirement for choline and vary in their distribution across ethnic and racial groups.


Subject(s)
Choline/metabolism , Ethnicity/genetics , Nutritional Requirements/genetics , Polymorphism, Single Nucleotide/genetics , Adolescent , Adult , Aged , Alleles , Diet , Female , Humans , Male , Middle Aged , Young Adult
5.
Am J Public Health ; 103 Suppl 1: S33-42, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23927503

ABSTRACT

Social and behavioral research in public health is often intimately tied to profound, but frequently neglected, biological influences from underlying genetic, environmental, and epigenetic events. The dynamic interplay between the life, social, and behavioral sciences often remains underappreciated and underutilized in addressing complex diseases and disorders and in developing effective remediation strategies. Using a case-study format, we present examples as to how the inclusion of genetic, environmental, and epigenetic data can augment social and behavioral health research by expanding the parameters of such studies, adding specificity to phenotypic assessments, and providing additional internal control in comparative studies. We highlight the important roles of gene-environment interactions and epigenetics as sources of phenotypic change and as a bridge between the life and social and behavioral sciences in the development of robust interdisciplinary analyses.


Subject(s)
Behavioral Sciences , Choice Behavior , Disease/genetics , Disease/psychology , Epigenomics , Gene-Environment Interaction , Social Sciences , Behavioral Research , Environment , Humans , Phenotype , Psychology , Research Design , Social Environment
6.
J Periodontol ; 84(11): 1606-16, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23368949

ABSTRACT

BACKGROUND: Tumor necrosis factor-α (TNF-α) plays a central role in the molecular pathogenesis of periodontal disease. However, the epigenetic regulation attributable to microbial and inflammatory signals at the biofilm-gingival interface are poorly understood. In this study, the DNA methylation alteration within the TNFA promoter in human gingival biopsies from different stages of periodontal disease is investigated and the regulatory mechanism of TNFA transcription by DNA methylation is explored. METHODS: Gingival biopsies were obtained from 17 patients with chronic periodontitis (CP) and 18 periodontally healthy individuals. Another 11 individuals participated in an experimentally induced gingivitis study, and gingival biopsies were collected at the baseline, induction, and resolution phase. To confirm that TNFA promoter methylation modulated TNFA transcription, THP.1 cells were treated with a DNA methyltransferase inhibitor, 5-Aza-2-deoxycytidine (5-Aza-2dC), and an RAW294.7 cell line transfected with a TNFA promoter-specific luciferase reporter system with or without methylation was used. RESULTS: In gingival biopsies from individuals with severe CP, two individual cytosine-guanine dinucleotides (CpG sites) within the TNFA promoter (at -163 and -161 bp) displayed increased methylation in CP samples compared to those with gingival health (16.1% ± 5.1% versus 11.0% ± 4.6%, P = 0.02 and 19.8% ± 4.1% versus 15.4% ± 3.6%, P = 0.04, respectively). The methylation level at -163 bp was inversely associated with the transcription level of TNFA (P = 0.018). However, no significant difference in the TNFA promoter methylation pattern was observed in samples biopsied during the induction or resolution phase of experimentally induced gingivitis, which represented a reversible periodontal lesion. THP.1 cells treated with 5-Aza-2dC demonstrated a time-dependent increase in TNFA messenger level. It was also found that the luciferase activity decreased 2.6-fold in a construct containing an in vitro methylated TNFA promoter when compared to the unmethylated insert (P = 0.03). CONCLUSION: Although the biopsy samples represented a mixed cell population, the change in promoter methylation status in chronic periodontal disease suggested that DNA methylation may be an important regulatory mechanism in controlling TNFA transcriptional expression in periodontal disease.


Subject(s)
Chronic Periodontitis/genetics , Epigenesis, Genetic/genetics , Tumor Necrosis Factor-alpha/genetics , Adult , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Biopsy , Campylobacter rectus/immunology , Cell Culture Techniques , Cell Line , Chronic Periodontitis/immunology , Chronic Periodontitis/pathology , CpG Islands/genetics , Cross-Sectional Studies , DNA Methylation/genetics , DNA Modification Methylases/antagonists & inhibitors , Decitabine , Female , Gene Expression Regulation/genetics , Gingivitis/genetics , Gingivitis/immunology , Gingivitis/pathology , Humans , Luciferases , Luminescent Agents , Male , Middle Aged , Monocytes/drug effects , Monocytes/immunology , Monocytes/microbiology , Osteoclasts/cytology , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , Transcription, Genetic/genetics , Transfection/methods , Young Adult
8.
FASEB J ; 27(1): 350-8, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22997227

ABSTRACT

Previous studies indicated that the intake of α-linolenic acid (ALA) can alter the concentration of both ω-6 and ω-3 fatty acids in both mother and offspring, with consequences on postnatal brain development. This study describes the association between maternal ALA availability during gestation and lactation, and alterations in the Fads2 DNA methylation in both maternal and offspring livers, at the end of lactation period. Both Fads2 promoter and intron 1 DNA methylation were increased in the groups receiving postnatal flaxseed oil containing 50% ALA (mothers or pups), while bivariate analysis indicated a significant association of the Fads2 epigenetic status in the liver between each mother and its offspring. In addition, Fads2 expression was negatively correlated with promoter methylation at the individual level in maternal livers (P<0.05). This study also indicated that the interplay between ALA availability during gestation and lactation can differentially alter the expression of desaturases and elongases involved in ω-6 and ω-3 metabolic pathways. In summary, when considering the perinatal dietary ALA requirements in mice, both gestation and lactation periods should be considered as having distinct roles in modulating the metabolism of ω-6 and ω-3 fatty acids in maternal mouse livers.


Subject(s)
Epigenesis, Genetic , Liver/metabolism , alpha-Linolenic Acid/administration & dosage , Animals , Base Sequence , DNA Methylation , DNA Primers , Fatty Acid Desaturases/genetics , Fatty Acids/blood , Female , Lactation , Mice , Mice, Inbred C57BL , Pregnancy , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction
9.
Aging Dis ; 3(6): 444-53, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23251850

ABSTRACT

Multiple cues from the environment of our indirect and immediate ancestors, which often persist throughout the prenatal period and adulthood, are shaping our phenotypes through either direct, parent-to-child influences, or transgenerational inheritance. These effects are due to gene-environment interactions, which are intended to be a predictive tool and a mechanism of quick adaptation to the environment, as compared with genetic variations that are inherited over many generations. In certain circumstances the influences induced by the gene-environment interactions can have deleterious effects upon the health status, in the context of a radical change in the environment that does not fit with the predicted conditions, via epigenetic alterations. Conversely the best fit to the expected environment might have a delayed aging process and a longer life span. This review will touch upon the Developmental Origins of Health and Disease (DoHAD) concept, while discussing recent advances in the understanding of metabolic and cognitive disruptions, with a focus on epigenetic factors, their transgenerational effects, and the consequences they might have upon the onset of chronic disease and premature exitus.

10.
Toxicol Appl Pharmacol ; 264(3): 439-50, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22959928

ABSTRACT

Inorganic arsenic (iAs) is a complete transplacental carcinogen in mice. Previous studies have demonstrated that in utero exposure to iAs promotes cancer in adult mouse offspring, possibly acting through epigenetic mechanisms. Humans and rodents enzymatically convert iAs to its methylated metabolites. This reaction requires S-adenosylmethionine (SAM) as methyl group donor. SAM is also required for DNA methylation. Supplementation with folate, a major dietary source of methyl groups for SAM synthesis, has been shown to modify iAs metabolism and the adverse effects of iAs exposure. However, effects of gestational folate supplementation on iAs metabolism and fetal DNA methylation have never been thoroughly examined. In the present study, pregnant CD1 mice were fed control (i.e. normal folate, or 2.2 mg/kg) or high folate diet (11 mg/kg) from gestational day (GD) 5 to 18 and drank water with 0 or 85 ppm of As (as arsenite) from GD8 to 18. The exposure to iAs significantly decreased body weight of GD18 fetuses and increased both SAM and S-adenosylhomocysteine (SAH) concentrations in fetal livers. High folate intake lowered the burden of total arsenic in maternal livers but did not prevent the effects of iAs exposure on fetal weight or hepatic SAM and SAH concentrations. In fact, combined folate-iAs exposure caused further significant body weight reduction. Notably, iAs exposure alone had little effect on DNA methylation in fetal livers. In contrast, the combined folate-iAs exposure changed the CpG island methylation in 2,931 genes, including genes known to be imprinted. Most of these genes were associated with neurodevelopment, cancer, cell cycle, and signaling networks. The canonical Wnt-signaling pathway, which regulates fetal development, was among the most affected biological pathways. Taken together, our results suggest that a combined in utero exposure to iAs and a high folate intake may adversely influence DNA methylation profiles and weight of fetuses, compromising fetal development and possibly increasing the risk for early-onset of disease in offspring.


Subject(s)
Arsenites/toxicity , Epigenomics , Folic Acid/pharmacology , Gene Expression Regulation, Developmental/drug effects , Sodium Compounds/toxicity , Animals , Arsenites/administration & dosage , Female , Fetal Weight/drug effects , Fetus/drug effects , Folic Acid/administration & dosage , Folic Acid/blood , Liver/drug effects , Liver/embryology , Liver/metabolism , Male , Mice , Pregnancy , S-Adenosylhomocysteine/metabolism , S-Adenosylmethionine/metabolism , Sodium Compounds/administration & dosage
11.
Biomol Concepts ; 3(2): 151-60, 2012 Apr.
Article in English | MEDLINE | ID: mdl-25436529

ABSTRACT

Abstract The rapid progress in nutritional epigenetics allowed for a much better understanding of the mechanisms involved in gene-nutrient interactions and the roles that nutrition has in transgenerational inheritance of acquired epigenetic traits. Studies indicated that a considerable number of nutrients or diet types are capable of inducing epimutations. In parallel, the rapid development of genome-wide DNA methylation detection methods allowed for a broader image on how nutrition impacts the epigenetic status in human and animal models. But this increased complexity in the epigenetic field and also brought important challenges that need resolution, or it suggests that some of the initial epigenetic paradigms have to be revisited or reconsidered. The aim of this review is to discuss the inherent challenges that need to be resolved, from both practical and theoretical aspects, stemming from the rapid progress in the field of nutritional epigenetics, with a focus on DNA methylation. Because such challenges are present at every stage of study development, the review systematically discusses the most common issues relevant to DNA methylation in a nutritional context. Various types of challenges and potential bias generators are discussed within study design, sample quality, detection methods, data processing, and statistical and bioinformatic analysis. Additional aspects to be considered include epigenetic heterogeneity of treatment groups, the role of genomic variability in introducing measurement bias and errors in interpretation of changes, and issues related to the final interpretation of results and in assigning functional significance. It is also posited that all these issues will be largely resolved within the next decade.

12.
ILAR J ; 53(3-4): 270-8, 2012.
Article in English | MEDLINE | ID: mdl-23744966

ABSTRACT

Within the last two decades, significant progress has been made in understanding the importance of epigenetic mechanisms in the regulation of gene expression as a consequence of gene-environment interactions. Nutrition, among many other environmental factors, is a key player that can induce epigenetic changes not only in the directly exposed organisms but also in subsequent generations through the transgenerational inheritance of epigenetic traits. This article aims to provide insights into the usefulness of the mouse model for epigenetic studies involving nutrition as well as the inherent limitations when compared with epigenetic phenomena in humans. Mice are one of the most versatile models for nutrition and epigenetic studies because of several features, such as short life-span, relative low cost for generating samples, the existence of well-characterized genetically engineered lines, the detailed sequencing of genomes, and the relative similarity of their metabolic processes to human metabolism. However, several limitations have to be acknowledged, such as the different location of genes on the chromosomes (and hence possibly different consequences of some epigenetic alterations), differences in the epigenetic patterns established during late embryogenesis, and possible epigenetic differences associated with cellular senescence caused by the different structure of telomeres when compared with humans. All these aspects have to be carefully analyzed when deciding whether a mouse model should be considered for a study in nutrition and epigenetics. Consequently, the results obtained from mouse studies should be carefully interpreted regarding their relevance to humans.


Subject(s)
Epigenesis, Genetic/genetics , Nutritional Status , Animals , DNA Methylation/genetics , Gene-Environment Interaction , Humans , Mice
13.
Int J Dev Neurosci ; 29(8): 795-802, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21964326

ABSTRACT

The availability of ω-3 polyunsaturated fatty acids is essential for perinatal brain development. While the roles of docosahexaenoic acid (the most abundant ω-3 species) were extensively described, less is known about the role of α-linolenic acid (ALA), which is the initial molecular species undergoing elongation and desaturation within the ω-3 pathways. This study describes the association between maternal ALA availability during gestation and lactation, and alterations in hippocampal development (dentate gyrus) in the mouse male offspring, at the end of lactation (postnatal day 19, P19). Postnatal ALA supplementation increased cell proliferation (36% more proliferating cells compared to a control group) and early neuronal differentiation, while postnatal ALA deficiency increased cellular apoptosis within the dentate gyrus of suckling pups (61% more apoptotic cells compared to a control group). However, maternal ALA deficiency during gestation prevented the increased neurogenesis induced by postnatal supplementation. Fatty acid analysis revealed that ALA supplementation increased the concentration of the ω-3 species in the maternal liver and serum, but not in the brain of the offspring, excepting for ALA itself. Interestingly, ALA supplementation also increased the concentration of dihomo γ-linolenic acid (a ω-6 species) in the P19 brains, but not in maternal livers or serum. In conclusion, postnatal ALA supplementation enhances neurogenesis in the dentate gyrus of the offspring at postnatal day 19, but its beneficial effects are offset by maternal ALA deficiency during gestation. These results suggest that ALA is required in both fetal and postnatal stages of brain development.


Subject(s)
Dietary Supplements , Hippocampus/growth & development , Lactation/physiology , Pregnancy, Animal/physiology , alpha-Linolenic Acid/metabolism , Animals , Body Weight , Cell Differentiation , Female , Hippocampus/cytology , Male , Mice , Mice, Inbred C57BL , Neurons/cytology , Neurons/physiology , Pregnancy , Random Allocation , alpha-Linolenic Acid/administration & dosage
14.
Curr Opin Clin Nutr Metab Care ; 14(1): 35-40, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21088573

ABSTRACT

PURPOSE OF REVIEW: This review synthesizes recently published information regarding nutrition and its impact upon epigenetically mediated mechanisms involved in longevity and aging. RECENT FINDINGS: Recent studies enriched considerably our understanding of the relationship between aging and gene-nutrient interactions that continuously shape our phenotype. Epigenetic mechanisms play an important role in mediating between the nutrient inputs and the ensuing phenotypic changes throughout our entire life and seem to be responsible, in part, for the biological changes that occur during aging. Less is known about the epigenetic role that nutrients have in directly influencing longevity and aging. However, recent studies clearly indicated that because nutrition modulates epigenetic events associated with various diseases (e.g., cancer, obesity, and diabetes), there is at least an indirect epigenetic link between nutrition and longevity and, therefore, biologic plausibility to hypothesize the epigenetic role of nutrition in altering longevity. Apart from limited human studies, promising animal studies brought us much closer to understanding how nutrition could have such an impact upon longevity and aging. SUMMARY: Complex epigenetic mechanisms are involved in aging and longevity, directly or indirectly via disease mechanisms. Nutrition has a strong impact upon epigenetic processes and, therefore, holds promise in having important roles in regulating longevity and aging.


Subject(s)
Aging/genetics , Diet , Epigenesis, Genetic , Longevity/genetics , Nutrigenomics , Animals , Humans , Phenotype
15.
FASEB J ; 24(1): 184-95, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19752176

ABSTRACT

Maternal choline availability is essential for fetal neurogenesis. Choline deprivation (CD) causes hypomethylation of specific CpG islands in genes controlling cell cycling in fetal hippocampus. We now report that, in C57BL/6 mice, CD during gestational days 12-17 also altered methylation of the histone H3 in E17 fetal hippocampi. In the ventricular and subventricular zones, monomethyl-lysine 9 of H3 (H3K9me1) was decreased by 25% (P<0.01), and in the pyramidal layer, dimethyl-lysine 9 of H3 (H3K9me2) was decreased by 37% (P<0.05). These changes were region specific and were not observed in whole-brain preparations. Also, the same effects of CD on H3 methylation were observed in E14 neural progenitor cells (NPCs) in culture. Changes in G9a histone methyltransferase might mediate altered H3K9me2,1. Gene expression of G9a was decreased by 80% in CD NPCs (P<0.001). In CD, H3 was hypomethylated upstream of the RE1 binding site in the calbindin 1 promoter, and 1 CpG site within the calbindin1 promoter was hypermethylated. REST binding to RE1 (recruits G9a) was decreased by 45% (P<0.01) in CD. These changes resulted in increased expression of calbindin 1 in CD (260%; P<0.05). Thus, CD modulates histone methylation in NPCs, and this could underlie the observed changes in neurogenesis.


Subject(s)
Choline Deficiency/genetics , Choline Deficiency/metabolism , Histones/chemistry , Histones/metabolism , S100 Calcium Binding Protein G/genetics , Animals , Apoptosis , Base Sequence , Binding Sites/genetics , Calbindin 1 , Calbindins , Cells, Cultured , CpG Islands , DNA/genetics , DNA/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Epigenesis, Genetic , Female , Hippocampus/embryology , Hippocampus/metabolism , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Methylation , Mice , Mice, Inbred C57BL , Mitosis , Models, Biological , Pregnancy , Promoter Regions, Genetic , Repressor Proteins/metabolism , S100 Calcium Binding Protein G/metabolism
16.
Int J Dev Neurosci ; 27(7): 627-33, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19695321

ABSTRACT

The importance of maternal nutrition for fetal brain development is increasingly recognized. Previous studies have suggested that maternal obesity or maternal exposure to obesogenic diets may permanently alter brain structure and function in the offspring. To test whether maternal exposure to a high-fat diet, prior and during gestation, alters fetal hippocampal development, we fed 8-week old C57BL/6 females with a high-fat diet (60% calories from fat) for 10 weeks prior to matting and 17 days after. Fetal brains at embryonic day E17 were used to determine developmental changes in the hippocampus. We report that maternal exposure to the high-fat diet induced small for gestational age (SGA) status and fetal resorption. The proliferation of neural progenitors was increased in the neuroepithelium from hippocampus and cortex in fetuses from mothers fed the high-fat diet when compared to controls, but decreased within the dentate gyrus (DG). Apoptosis in the hippocampus was decreased (Ammon's Horn and fimbria). The differentiation of calretinin-positive neurons within the DG was also decreased. These data indicate that, under the influence of a maternal high-fat diet administered prior and during gestation, fetal hippocampal development is altered at embryonic day 17, as indicated by region-specific changes in proliferation of neural precursors, decreased apoptosis, and by decreased neuronal differentiation within the dentate gyrus.


Subject(s)
Diet , Dietary Fats/adverse effects , Fetus , Hippocampus/embryology , Maternal Nutritional Physiological Phenomena , Neurons/physiology , Obesity/complications , Animals , Body Weight , Calbindin 2 , Cell Proliferation , Female , Fetus/anatomy & histology , Fetus/physiology , Hippocampus/cytology , Hippocampus/metabolism , Male , Mice , Mice, Inbred C57BL , Neurons/cytology , Pregnancy , Random Allocation , S100 Calcium Binding Protein G/metabolism , Stem Cells/cytology , Stem Cells/physiology
17.
Am J Trop Med Hyg ; 81(2): 313-6, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19635890

ABSTRACT

We report the case of a 42-year-old woman who was admitted to the hospital for fever, chills, nausea, vomiting, fatigue, myalgia, and general muscle weakness. All these symptoms had occurred 3 weeks after the ingestion of inadequately cooked pork meat, subsequently confirmed to be infested with Trichinella spiralis. Laboratory results showed mild leukocytosis, inflammation, and mild liver and muscle cytolytic syndrome, all suggestive of trichinellosis. Echocardiography showed apical hypokinesis and an apical mass (likely a thrombus). The immunologic assessment for the presence of Trichinella antigens was positive. The outcome was favorable after treatment with an anticoagulant, an antiaggregant, prednisone, and mebendazole. Follow-up controls showed the absence of any symptoms and thrombus, with only mild electrocardiogram modifications still present.


Subject(s)
Coronary Thrombosis/complications , Trichinella spiralis , Trichinellosis/complications , Adult , Animals , Coronary Angiography , Coronary Thrombosis/diagnosis , Diagnosis, Differential , Echocardiography , Electrocardiography , Female , Humans , Ventricular Function, Left
18.
Toxicol Sci ; 107(1): 220-6, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18948303

ABSTRACT

Diethanolamine (DEA) is a common ingredient of personal care products. Dermal administration of DEA diminishes hepatic stores of the essential nutrient choline and alters brain development. We previously reported that 80 mg/kg/day of DEA during pregnancy in mice reduced neurogenesis and increased apoptosis in the fetal hippocampus. This study was designed to establish the dose-response relationships for this effect of DEA. Timed-pregnant C57BL/6 mouse dams were dosed dermally from gestation day 7-17 with DEA at 0 (controls), 5, 40, 60, and 80 mg/kg body/day. Fetuses (embryonic day 17 [E17]) from dams treated dermally with 80 mg/kg body/day DEA had decreased neural progenitor cell mitosis at the ventricular surface of the ventricular zone (hippocampus, 54.1 +/- 5.5%; cortex, 58.9 +/- 6.8%; compared to controls; p < 0.01). Also, this dose of DEA to dams increased rates of apoptosis in E17 fetal hippocampus (to 177.2 +/- 21.5% of control; measured using activated caspase-3; p < 0.01). This dose of DEA resulted in accumulation of DEA and its metabolites in liver and in plasma. At doses of DEA less than 80 mg/kg body/day to dams, there were no differences between treated and control groups. In a small group of human subjects, dermal treatment for 1 month with a commercially available skin lotion containing 1.8 mg DEA per gram resulted in detectable plasma concentrations of DEA and dimethyldiethanolamine, but these were far below those concentrations associated with perturbed brain development in the mouse.


Subject(s)
Ethanolamines/pharmacology , Fetus/drug effects , Hippocampus/drug effects , Administration, Cutaneous , Adult , Analysis of Variance , Animals , Apoptosis/drug effects , Brain/drug effects , Brain/embryology , Brain/metabolism , Caspase 3/blood , Choline/metabolism , Dose-Response Relationship, Drug , Ethanolamines/blood , Ethanolamines/metabolism , Female , Fetus/embryology , Hippocampus/embryology , Hippocampus/metabolism , Humans , Immunohistochemistry , Liver/chemistry , Male , Mice , Microscopy, Confocal , Middle Aged , Mitosis/drug effects , Neurogenesis/drug effects , Pregnancy
19.
Am J Clin Nutr ; 86(1): 230-9, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17616785

ABSTRACT

BACKGROUND: Some humans fed a low-choline diet develop hepatosteatosis, liver and muscle damage, and lymphocyte apoptosis. The risk of developing such organ dysfunction is increased by the presence of single-nucleotide polymorphisms (SNPs) in genes involved in folate and choline metabolism. OBJECTIVE: We investigated whether these changes that occur in the expression of many genes when humans are fed a low-choline diet differ between subjects who develop organ dysfunction and those who do not. We also investigated whether expression changes were dependent on the presence of the SNPs of interest. DESIGN: Thirty-three subjects aged 20-67 y were fed for 10 d a baseline diet containing the recommended adequate intake of choline. They then were fed a low-choline diet for up to 42 d or until they developed organ dysfunction. Blood was collected at the end of each phase, and peripheral lymphocytes were isolated and used for genotyping and for gene expression profiling with the use of microarray hybridization. RESULTS: Feeding a low-choline diet changed the expression of 259 genes, and the profiles of subjects who developed and those who did not develop signs of organ dysfunction differed. Group clustering and gene ontology analyses found that the diet-induced changes in gene expression profiles were significantly influenced by the SNPs of interest and that the gene expression phenotype of the variant gene carriers differed significantly even with the baseline diet. CONCLUSION: These findings support our hypothesis that a person's susceptibility to organ dysfunction when fed a low-choline diet is modulated by specific SNPs in genes involved in folate and choline metabolism.


Subject(s)
Choline Deficiency/blood , Choline Deficiency/genetics , Lymphocytes/physiology , Adult , Aged , Choline/administration & dosage , Choline Deficiency/enzymology , Choline Dehydrogenase/biosynthesis , Choline Dehydrogenase/genetics , Cluster Analysis , DNA/chemistry , DNA/genetics , Female , Gene Expression Profiling , Gene Expression Regulation, Enzymologic , Humans , Lymphocytes/enzymology , Lymphocytes/metabolism , Male , Methylenetetrahydrofolate Dehydrogenase (NADP)/biosynthesis , Methylenetetrahydrofolate Dehydrogenase (NADP)/genetics , Middle Aged , Oligonucleotide Array Sequence Analysis , Phosphatidylethanolamine N-Methyltransferase/biosynthesis , Phosphatidylethanolamine N-Methyltransferase/genetics , Polymorphism, Single Nucleotide
20.
Mol Genet Metab ; 91(2): 165-75, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17409006

ABSTRACT

Over a four-year period, we collected clinical and biochemical data from five Amish children who were homozygous for missense mutations in 5,10-methylenetetrahydrofolate reductase (MTHFR c.1129C>T). The four oldest patients had irreversible brain damage prior to diagnosis. The youngest child, diagnosed and started on betaine therapy as a newborn, is healthy at her present age of three years. We compared biochemical data among four groups: 16 control subjects, eight heterozygous parents, and five affected children (for the latter group, both before and during treatment with betaine anhydrous). Plasma amino acid concentrations were used to estimate changes in cerebral methionine uptake resulting from betaine therapy. In all affected children, treatment with betaine (534+/-222 mg/kg/day) increased plasma S-adenosylmethionine, improved markers of tissue methyltransferase activity, and resulted in a threefold increase of calculated brain methionine uptake. Betaine therapy did not normalize plasma total homocysteine, nor did it correct cerebral 5-methyltetrahydrofolate deficiency. We conclude that when the 5-methyltetrahydrofolate content of brain tissue is low, dietary betaine sufficient to increase brain methionine uptake may compensate for impaired cerebral methionine recycling. To effectively support the metabolic requirements of rapid brain growth, a large dose of betaine should be started early in life.


Subject(s)
Betaine/therapeutic use , Brain Diseases/prevention & control , Methylenetetrahydrofolate Reductase (NADPH2)/deficiency , Adolescent , Adult , Brain/metabolism , Brain Diseases/cerebrospinal fluid , Brain Diseases/metabolism , Child , Child, Preschool , Humans , Infant, Newborn , Methionine/cerebrospinal fluid , Methionine/metabolism , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Methyltransferases/metabolism , Mutation, Missense , Neonatal Screening , S-Adenosylmethionine/blood , S-Adenosylmethionine/cerebrospinal fluid
SELECTION OF CITATIONS
SEARCH DETAIL
...