Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
1.
Cell Mol Life Sci ; 78(4): 1729-1744, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32734584

ABSTRACT

Maternal nutrition is crucial for the offspring's skeleton development and the onset of osteoporosis later in life. While maternal low protein diet has been shown to regulate bone mass negatively, the effect of a high protein diet (HP) remains unexplored. Here, we found that C57BL/6 mice fed with HP delivered offspring with decreased skeletal mineralization at birth and reduced bone mass throughout their life due to a decline in their osteoblast maturation. A small RNA sequencing study revealed that miR-24-1-5p was highly upregulated in HP group osteoblasts. Target prediction and validation studies identified SMAD-5 as a direct target of miR-24-1-5p. Furthermore, mimic and inhibitor studies showed a negative correlation between miR-24-1-5p expression and osteoblast function. Moreover, ex vivo inhibition of miR-24-1-5p reversed the reduced maturation and SMAD-5 expression in the HP group osteoblasts. Together, we show that maternal HP diminishes the bone mass of the offspring through miR-24-1-5p.


Subject(s)
Bone Development/genetics , Maternal Nutritional Physiological Phenomena/genetics , MicroRNAs/genetics , Smad5 Protein/genetics , Animals , Bone Density , Bone and Bones/metabolism , Cell Differentiation/genetics , Diet, High-Protein/adverse effects , Female , Humans , Maternal Nutritional Physiological Phenomena/physiology , Mice , Osteoblasts/metabolism , Osteogenesis/drug effects , Osteogenesis/genetics , Osteoporosis/genetics , Osteoporosis/pathology
2.
Am J Physiol Endocrinol Metab ; 320(3): E438-E452, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33427054

ABSTRACT

Obesity is a prevailing problem across the globe. Women who are obese have difficulty initiating and sustaining lactation. However, the impact of genetics and diet on breastfeeding outcomes is understudied. Here we explore the effect of diet and genotype on lactation. We utilized the low-density lipoprotein receptor (Ldlr-KO) transgenic mouse model as an obesity and hypercholesterolemia model. Additionally, we used the tryptophan hydroxylase 1 (Tph1-KO) mouse, recently identified as a potential anti-obesogenic model, to investigate if addition of Tph1-KO could ameliorate negative effects of obesity in Ldlr-KO mice. We created a novel transgenic mouse line by combining the Ldlr and Tph1 [double knockout (DKO)] mice to study the interaction between the two genotypes. Female mice were fed a low-fat diet (LFD; 10% fat) or high-fat diet (HFD; 60% fat) from 3 wk of age through early [lactation day 3 (L3)] or peak lactation [lactation day 11 (L11)]. After 4 wk of consuming either LFD or HFD, female mice were bred. On L2 and L10, dams were milked to investigate the effect of diet and genotype on milk composition. Dams were euthanized on L3 or L11. There was no impact of diet or genotype on milk protein or triglycerides (TGs) on L2; however, by L10, Ldlr-KO and DKO dams had increased TG levels in milk. RNA-sequencing of L11 mammary glands demonstrated Ldlr-KO dams fed HFD displayed enrichment of genes involved in immune system pathways. Interestingly, the DKO may alter vesicle budding and biogenesis during lactation. We also quantified macrophages by immunostaining for F4/80+ cells at L3 and L11. Diet played a significant role on L3 (P = 0.013), but genotype played a role at L11 (P < 0.0001) on numbers of F4/80+ cells. Thus the impact of diet and genotype on lactation differs depending on stage of lactation, illustrating complexities of understanding the intersection of these parameters.NEW & NOTEWORTHY We have created a novel mouse model that is focused on understanding the intersection of diet and genotype on mammary gland function during lactation.


Subject(s)
Diet, High-Fat , Lactation , Mammary Glands, Animal/metabolism , Receptors, LDL/genetics , Tryptophan Hydroxylase/genetics , Animals , Dietary Fats/pharmacology , Female , Gene-Environment Interaction , Genotype , Lactation/drug effects , Lactation/genetics , Mammary Glands, Animal/drug effects , Maternal Nutritional Physiological Phenomena/drug effects , Maternal Nutritional Physiological Phenomena/genetics , Mice , Mice, Knockout , Mice, Obese , Obesity/genetics , Obesity/metabolism
3.
Am J Physiol Endocrinol Metab ; 320(4): E786-E796, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33586490

ABSTRACT

We investigated the expression levels of nephroblastoma overexpressed [NOV or CCN3 (cellular communication network factor 3)] in the serum and placenta of pregnant women and of pregnant mice fed a high-fat diet (HFD), and its effect on placental glucose transporter 3 (GLUT3) expression, to examine its role in gestational diabetes mellitus (GDM). NOV/CCN3 expression was increased in the mouse serum during pregnancy. At gestational day 18, NOV/CCN3 protein expression was increased in the serum and placenta of the HFD mice compared with that of mice fed a normal diet. Compared with non-GDM patients, the patients with GDM had significantly increased serum NOV/CCN3 protein expression and placental NOV/CCN3 mRNA expression. Therefore, we hypothesized that NOV/CCN3 signaling may be involved in the pathogenesis of GDM. We administered NOV/CCN3 recombinant protein via intraperitoneal injections to pregnant mice fed HFD or normal diet. NOV/CCN3 overexpression led to glucose intolerance. Combined with the HFD, NOV/CCN3 exacerbated glucose intolerance and caused insulin resistance. NOV/CCN3 upregulates GLUT3 expression and affects the mammalian target of rapamycin (mTOR) pathway in the GDM environment in vivo and in vitro. In summary, our results demonstrate, for the first time, the molecular mechanism of NOV/CCN3 signaling in maternal metabolism to regulate glucose balance during pregnancy. NOV/CCN3 may be a potential target for detecting and treating GDM.NEW & NOTEWORTHY NOV/CCN3 regulates glucose homeostasis in mice during pregnancy. NOV/CCN3 upregulates GLUT3 expression and affects the mTOR pathway in the GDM environment in vivo and in vitro.


Subject(s)
Diet, High-Fat , Glucose Transporter Type 3/genetics , Nephroblastoma Overexpressed Protein/genetics , TOR Serine-Threonine Kinases/metabolism , Animals , Cells, Cultured , Diabetes, Gestational/genetics , Diabetes, Gestational/metabolism , Dietary Fats/pharmacology , Female , Glucose/metabolism , Glucose Intolerance/genetics , Glucose Intolerance/metabolism , Glucose Transporter Type 3/metabolism , Humans , Maternal Nutritional Physiological Phenomena/drug effects , Maternal Nutritional Physiological Phenomena/genetics , Mice , Mice, Inbred C57BL , Nephroblastoma Overexpressed Protein/metabolism , Pregnancy , Signal Transduction/drug effects , Signal Transduction/genetics , TOR Serine-Threonine Kinases/genetics , Up-Regulation/drug effects , Up-Regulation/genetics
4.
Int J Mol Sci ; 22(6)2021 Mar 14.
Article in English | MEDLINE | ID: mdl-33799409

ABSTRACT

Linoleic acid (LA), an n-6 polyunsaturated fatty acid (PUFA), is essential for fetal growth and development. We aimed to investigate the effect of maternal and postnatal high LA (HLA) diet on plasma FA composition, plasma and hepatic lipids and genes involved in lipid metabolism in the liver of adult offspring. Female rats were fed with low LA (LLA; 1.44% LA) or HLA (6.21% LA) diets for 10 weeks before pregnancy, and during gestation/lactation. Offspring were weaned at postnatal day 25 (PN25), fed either LLA or HLA diets and sacrificed at PN180. Postnatal HLA diet decreased circulating total n-3 PUFA and alpha-linolenic acid (ALA), while increased total n-6 PUFA, LA and arachidonic acid (AA) in both male and female offspring. Maternal HLA diet increased circulating leptin in female offspring, but not in males. Maternal HLA diet decreased circulating adiponectin in males. Postnatal HLA diet significantly decreased aspartate transaminase (AST) in females and downregulated total cholesterol, HDL-cholesterol and triglycerides in the plasma of males. Maternal HLA diet downregulated the hepatic mRNA expression of Hmgcr in both male and female offspring and decreased the hepatic mRNA expression of Cpt1a and Acox1 in females. Both maternal and postnatal HLA diet decreased hepatic mRNA expression of Cyp27a1 in females. Postnatal diet significantly altered circulating fatty acid concentrations, with sex-specific differences in genes that control lipid metabolism in the adult offspring following exposure to high LA diet in utero.


Subject(s)
Fatty Acids, Omega-6/metabolism , Leptin/genetics , Linoleic Acid/metabolism , Liver/metabolism , Animals , Diet, High-Fat/adverse effects , Fatty Acids, Omega-6/pharmacology , Female , Humans , Lactation/drug effects , Lactation/genetics , Leptin/metabolism , Linoleic Acid/pharmacology , Lipid Metabolism/genetics , Liver/drug effects , Male , Maternal Nutritional Physiological Phenomena/genetics , Pregnancy , Rats , Sex Characteristics , Triglycerides/blood
5.
Ann Nutr Metab ; 76(6): 431-440, 2020.
Article in English | MEDLINE | ID: mdl-33503637

ABSTRACT

INTRODUCTION AND AIMS: Choline-metabolizing genetic variation may interact with choline intake on fetal programming and pregnancy outcome. This case-control study aims to explore the association of maternal choline consumption and phosphatidylethanolamine N-methyltransferase (PEMT) gene polymorphism rs7946 with preterm birth risk. METHODS: 145 Han Chinese women with preterm delivery and 157 Han Chinese women with term delivery were recruited in Shanghai. Dietary choline intake during pregnancy was assessed using a validated food frequency questionnaire. Additionally, DNA samples were genotyped for PEMT rs7946 (G5465A) with plasma homocysteine (Hcy) levels measured. RESULTS: Compared with the lowest quartile of choline intake, women within the highest consumption quartile had adjusted odds ratio (aOR) for preterm birth of 0.48 (95% confidence interval, CI [0.24, 0.95]). There was a significant interaction between maternal choline intake and PEMT rs7946 (p for interaction = 0.04), where the AA genotype carriers who consumed the energy-adjusted choline <255.01 mg/day had aOR for preterm birth of 3.75 (95% CI [1.24, 11.35]), compared to those with GG genotype and choline intake >255.01 mg/day during pregnancy. Additionally, the greatest elevated plasma Hcy was found in the cases with AA genotype and choline consumption <255.01 mg/day (p < 0.001). CONCLUSION: The AA genotype of PEMT rs7946 may be associated with increased preterm birth in these Han Chinese women with low choline intake during pregnancy.


Subject(s)
Choline/analysis , Maternal Nutritional Physiological Phenomena/genetics , Phosphatidylethanolamine N-Methyltransferase/genetics , Pregnancy Outcome/genetics , Premature Birth/genetics , Adult , Asian People/genetics , Case-Control Studies , China , Diet/statistics & numerical data , Diet Surveys , Eating/genetics , Female , Genotype , Humans , Polymorphism, Genetic/drug effects , Pregnancy
6.
Dokl Biol Sci ; 494(1): 244-247, 2020 Sep.
Article in English | MEDLINE | ID: mdl-33083882

ABSTRACT

In the present study it has been shown for the first time that maternal methyl-enriched diet (choline, betaine, folic acid, vitamin B12, L-methionine, zinc) during perinatal period reduces the expression of genetic absence epilepsy and comorbid depression in adult offspring of WAG/Rij rats. This beneficial effect was more pronounced in males compared with females. It is assumed that epigenetic modifications induced by maternal methyl-enriched diet in the offspring at the early stages of ontogenesis might be a possible mechanism underlying the correction of genetically-based pathologic phenotype in WAG/Rij rats. Results suggest that methyl-enriched diet during perinatal period can be potentially used for mitigation or prevention epileptogenesis and depression-like comorbid disorders in people genetically predisposed to absence epilepsy.


Subject(s)
Depression/genetics , Epilepsy, Absence/genetics , Maternal Nutritional Physiological Phenomena/genetics , Sex Characteristics , Animals , Depression/diet therapy , Depression/pathology , Disease Models, Animal , Epigenesis, Genetic/genetics , Epilepsy, Absence/diet therapy , Epilepsy, Absence/pathology , Female , Humans , Male , Pregnancy , Rats
7.
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
8.
Int J Obes (Lond) ; 42(8): 1431-1444, 2018 08.
Article in English | MEDLINE | ID: mdl-29777232

ABSTRACT

BACKGROUND AND OBJECTIVE: Maternal overnutrition has been implicated in affecting the offspring by programming metabolic disorders such as obesity and diabetes, by mechanisms that are not clearly understood. This study aimed to determine the long-term impact of maternal high-fat (HF) diet feeding on epigenetic changes in the offspring's hypothalamic Pomc gene, coding a key factor in the control of energy balance. Further, it aimed to study the additional effects of postnatal overnutrition on epigenetic programming by maternal nutrition. METHODS: Eight-week-old female Sprague-Dawley rats were fed HF diet or low-fat (LF) diet for 6 weeks before mating, and throughout gestation and lactation. At postnatal day 21, samples were collected from a third offspring and the remainder were weaned onto LF diet for 5 weeks, after which they were either fed LF or HF diet for 12 weeks, resulting in four groups of offspring differing by their maternal and postweaning diet. RESULTS: With maternal HF diet, offspring at weaning had rapid early weight gain, increased adiposity, and hyperleptinemia. The programmed adult offspring, subsequently fed LF diet, retained the increased body weight. Maternal HF diet combined with offspring HF diet caused more pronounced hyperphagia, fat mass, and insulin resistance. The ARC Pomc gene from programmed offspring at weaning showed hypermethylation in the enhancer (nPE1 and nPE2) regions and in the promoter sequence mediating leptin effects. Interestingly, hypermethylation at the Pomc promoter but not at the enhancer region persisted long term into adulthood in the programmed offspring. However, there were no additive effects on methylation levels in the regulatory regions of Pomc in programmed offspring fed a HF diet. CONCLUSION: Maternal overnutrition programs long-term epigenetic alterations in the offspring's hypothalamic Pomc promoter. This predisposes the offspring to metabolic disorders later in life.


Subject(s)
Epigenesis, Genetic/genetics , Hypothalamus/metabolism , Maternal Nutritional Physiological Phenomena/genetics , Overnutrition/genetics , Prenatal Exposure Delayed Effects/genetics , Pro-Opiomelanocortin/genetics , Regulatory Sequences, Nucleic Acid/genetics , Animals , DNA Methylation , Disease Models, Animal , Female , Hypothalamus/chemistry , Obesity/genetics , Obesity/metabolism , Overnutrition/metabolism , Overnutrition/physiopathology , Pregnancy , Pro-Opiomelanocortin/metabolism , Rats , Rats, Sprague-Dawley
9.
Br J Nutr ; 119(10): 1119-1132, 2018 05.
Article in English | MEDLINE | ID: mdl-29759106

ABSTRACT

A total of twenty-four healthy twin-bearing Liuyang black goats were allocated to two trials. In Trial 1, twelve goats received either the control diet (CG, n 6, 100 % feed) or restricted diet (RG, n 6, 60 % feed of CG) from gestation days 26 to 65 after synchronisation. In Trial 2, the remaining goats were randomly and equally divided into two treatments: CG and RG from days 95 to 125 of gestation. Placental traits, fetal weight, serum parameters, nitric oxide (NO), angiogenesis gene expression and cotyledon proteome were measured at the end of each trial. In early pregnancy, the total and relative weights of placenta, uterine caruncle and cotyledon, as well as fetus, were increased (P<0Ā·05) in RG. The NO content in maternal serum was also increased (P<0Ā·05) in RG. In all, fifty differentially expressed proteins were identified in cotyledon. The up-regulated proteins are related to proliferation and fission of trophoblast cell and the placenta angiogenesis. During the late pregnancy trial, placental weight was increased (P<0Ā·05) in RG, but weight of the fetus was decreased (P<0Ā·05). The capillary density in the cotyledon was also decreased (P<0Ā·01). A total of fifty-eight proteins were differentially expressed in cotyledon. The up-regulated proteins in RG are related to placenta formation, blood flow regulation and embryonic development. These results indicated that feed intake restriction during gestation influenced the placental and fetal development in a stage-dependent manner. These findings have important implications for developing novel nutrient management strategies in goat production.


Subject(s)
Food Deprivation/physiology , Gestational Age , Goats/physiology , Maternal Nutritional Physiological Phenomena/physiology , Placenta/physiology , Proteome/analysis , Animals , Female , Fetal Development/genetics , Fetal Weight , Maternal Nutritional Physiological Phenomena/genetics , Neovascularization, Physiologic/genetics , Neovascularization, Physiologic/physiology , Organ Size , Placenta/anatomy & histology , Placenta/blood supply , Placentation/genetics , Pregnancy , Trophoblasts/physiology , Up-Regulation , Uterus/anatomy & histology
10.
Semin Cell Dev Biol ; 43: 85-95, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26135290

ABSTRACT

It is now well established that the environment to which we are exposed during fetal and neonatal life can have a long-term impact on our health. This has been termed the developmental origins of health and disease. Factors known to have such programming effects include intrauterine nutrient availability (determined by maternal nutrition and placental function), endocrine disruptors, toxins and infectious agents. Epigenetic processes have emerged as a key mechanism by which the early environment can permanently influence cell function and metabolism after multiple rounds of cell division. More recently it has been suggested that programmed effects can be observed beyond the first generation and that therefore epigenetic mechanisms could form the basis of transmission of phenotype from parent to child to grandchild and beyond. Here we review the evidence for such processes.


Subject(s)
Environmental Exposure/adverse effects , Epigenesis, Genetic/genetics , Inheritance Patterns/genetics , Cell Division/genetics , Chromatin/genetics , DNA Methylation/genetics , Female , Histones/genetics , Histones/metabolism , Humans , Maternal Nutritional Physiological Phenomena/genetics , MicroRNAs/genetics , Models, Genetic
11.
FASEB J ; 30(4): 1548-56, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26700734

ABSTRACT

Maternal diet during pregnancy influences the later life reproductive potential of female offspring. We investigate the molecular mechanisms underlying the depletion of ovarian follicular reserve in young adult females following exposure to obesogenic diet in early life. Furthermore, we explore the interaction between adverse maternal diet and postweaning diet in generating reduced ovarian reserve. Female mice were exposed to either maternal obesogenic (high fat/high sugar) or maternal control dietin uteroand during lactation, then weaned onto either obesogenic or control diet. At 12 wk of age, the offspring ovarian reserve was depleted following exposure to maternal obesogenic diet (P< 0.05), but not postweaning obesogenic diet. Maternal obesogenic diet was associated with increased mitochondrial DNA biogenesis (copy numberP< 0.05; transcription factor A, mitochondrial expressionP< 0.05), increased mitochondrial antioxidant defenses [manganese superoxide dismutase (MnSOD)P< 0.05; copper/zinc superoxide dismutaseP< 0.05; glutathione peroxidase 4P< 0.01] and increased lipoxygenase expression (arachidonate 12-lipoxygenaseP< 0.05; arachidonate 15-lipoxygenaseP< 0.05) in the ovary. There was also significantly increased expression of the transcriptional regulator NF-κB (P< 0.05). There was no effect of postweaning diet on any measured ovarian parameters. Maternal diet thus plays a central role in determining follicular reserve in adult female offspring. Our observations suggest that lipid peroxidation and mitochondrial biogenesis are the key intracellular pathways involved in programming of ovarian reserve.-Aiken, C. E., Tarry-Adkins, J. L., Penfold, N. C., Dearden, L., Ozanne, S. E. Decreased ovarian reserve, dysregulation of mitochondrial biogenesis, and increased lipid peroxidation in female mouse offspring exposed to an obesogenic maternal diet.


Subject(s)
Lipid Peroxidation/physiology , Mitochondria/metabolism , Ovarian Reserve/physiology , Prenatal Exposure Delayed Effects/physiopathology , Animals , Body Weight/genetics , Body Weight/physiology , Diet, High-Fat/adverse effects , Female , Gene Expression Profiling/methods , Lipid Peroxidation/genetics , Maternal Nutritional Physiological Phenomena/genetics , Maternal Nutritional Physiological Phenomena/physiology , Mice, Inbred C57BL , Mitochondria/genetics , Organ Size , Ovarian Reserve/genetics , Ovary/cytology , Ovary/growth & development , Ovary/metabolism , Pregnancy , Prenatal Exposure Delayed Effects/etiology , Prenatal Exposure Delayed Effects/genetics , Reverse Transcriptase Polymerase Chain Reaction
12.
Horm Metab Res ; 49(1): 64-72, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27701682

ABSTRACT

Acylation of ghrelin is mediated by ghrelin O-acyltansferase (GOAT). Exogenous acylated ghrelin (AG) stimulates growth hormone (GH) and food intake. In non-pregnant (NP) animals, the GOAT-ghrelin-GH axis prevents hypoglycemia caused by caloric restriction (CR). In humans, maternal malnutrition challenges glucose metabolism, which is a key determinant of fetal health. To clarify the role of AG and GH, we compared effects of CR on the GOAT-ghrelin-GH axis in pregnant (P) and NP mice. C57BL/6 wild type (WT) and GOAT knock-out (KO) P and NP mice were freely fed (FF) or subjected to 50% CR for one week. CR was started in P mice on Day 10.5 after conception. We measured body composition, blood glucose, plasma ghrelin and GH, stomach, hypothalamus and pituitary GOAT and ghrelin expression, and liver glycogen content and Pck1 expression. GOAT and AG were undetectable in KO. In NP mice, CR did not affect blood glucose (-1.3 mmol/l, p>0.05) in WT but was lowered (-1.8 mmol/l, p<0.0001) in KO. GH and Pck1 mRNA expression increased in WT but not in KO. In P mice, CR markedly lowered glucose (-2.7 mmol/l; p<0.0001) in WT and caused fatal hypoglycemia in KO, despite similarly elevated GH in WT and KO mice. KO animals are more prone to hypoglycemia than WT. GH, which is high in P animals, does not prevent hypoglycemia caused by CR during pregnancy. Our data suggest a specific role of AG in the regulation of gluconeogenesis to maintain euglycemia during pregnancy when energy availability is limited.


Subject(s)
Acyltransferases/physiology , Caloric Restriction , Carbohydrate Metabolism/physiology , Ghrelin/physiology , Maternal Nutritional Physiological Phenomena , Acylation/genetics , Acyltransferases/genetics , Animals , Carbohydrate Metabolism/genetics , Female , Ghrelin/metabolism , Male , Maternal Nutritional Physiological Phenomena/genetics , Membrane Proteins , Mice , Mice, Inbred C57BL , Mice, Knockout , Pregnancy
13.
Alcohol Clin Exp Res ; 41(11): 1928-1937, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28975627

ABSTRACT

BACKGROUND: Reducing meat consumption is often advised; however, inadvertent nutritional deficiencies during pregnancy may result in residual neurodevelopmental harms to offspring. This study assessed possible effects of maternal diets in pregnancy on adverse substance use among adolescent offspring. METHODS: Pregnant women and their 13-year-old offspring taking part in a prospective birth cohort study, the Avon Longitudinal Study of Parents and Children (ALSPAC), provided Food Frequency Questionnaire data from which dietary patterns were derived using principal components analysis. Multivariable logistic regression models including potential confounders evaluated adverse alcohol, cannabis, and tobacco use of the children at 15Ā years of age. RESULTS: Lower maternal meat consumption was associated with greater problematic substance use among 15-year-old offspring in dose-response patterns. Comparing never to daily meat consumption after adjustment, risks were greater for all categories of problem substance use: alcohol, odds ratio ORĀ =Ā 1.75, 95% CIĀ =Ā (1.23, 2.56), pĀ <Ā 0.001; tobacco use ORĀ =Ā 1.85, 95% CIĀ =Ā (1.28, 2.63), pĀ <Ā 0.001; and cannabis ORĀ =Ā 2.70, 95% CIĀ =Ā (1.89, 4.00), pĀ <Ā 0.001. Given the likelihood of residual confounding, potential causality was evaluated using stratification for maternal allelic variants that impact biological activity of cobalamin (vitamin B12) and iron. Lower meat consumption disproportionally increased the risks of offspring substance misuse among mothers with optimally functional (homozygous) variants (rs1801198) of the gene transcobalamin 2 gene (TCN2) which encodes the vitamin B12 transport protein transcobalamin 2 implicating a causal role for cobalamin deficits. Functional maternal variants in iron metabolism were unrelated to the adverse substance use. Risks potentially attributable to cobalamin deficits during pregnancy include adverse adolescent alcohol, cannabis, and tobacco use (14, 37, and 23, respectively). CONCLUSIONS: Lower prenatal meat consumption was associated with increased risks of adolescent substance misuse. Interactions between TCN2 variant status and meat intake implicate cobalamin deficiencies.


Subject(s)
Dietary Proteins/administration & dosage , Genetic Variation/genetics , Meat , Prenatal Exposure Delayed Effects/genetics , Substance-Related Disorders/genetics , Transcobalamins/genetics , Adolescent , Adult , Cohort Studies , Diet Records , Female , Humans , Longitudinal Studies , Maternal Nutritional Physiological Phenomena/genetics , Pregnancy , Prenatal Exposure Delayed Effects/diagnosis , Prenatal Exposure Delayed Effects/epidemiology , Prospective Studies , Substance-Related Disorders/diagnosis , Substance-Related Disorders/epidemiology
14.
Int J Obes (Lond) ; 40(11): 1768-1775, 2016 11.
Article in English | MEDLINE | ID: mdl-27599633

ABSTRACT

OBJECTIVE: Although intrauterine nutritional stress is known to result in offspring obesity and the metabolic phenotype, the underlying cellular/molecular mechanisms remain incompletely understood. We tested the hypothesis that compared with the controls, the bone marrow-derived mesenchymal stem cells (BMSCs) of the intrauterine growth-restricted (IUGR) offspring exhibit a more adipogenic phenotype. METHODS: A well-established rat model of maternal food restriction (MFR), that is, 50% global caloric restriction during the later-half of pregnancy and ad libitum diet following birth that is known to result in an obese offspring with a metabolic phenotype was used. BMSCs at 3 weeks of age were isolated, and then molecularly and functionally profiled. RESULTS: BMSCs of the intrauterine nutritionally-restricted offspring demonstrated an increased proliferation and an enhanced adipogenic molecular profile at miRNA, mRNA and protein levels, with an overall up-regulated PPARƎĀ³ (miR-30d, miR-103, PPARƎĀ³, C/EPBα, ADRP, LPL, SREBP1), but down-regulated Wnt (LRP5, LEF-1, Ɵ-catenin, ZNF521 and RUNX2) signaling profile. Following adipogenic induction, compared with the control BMSCs, the already up-regulated adipogenic profile of the MFR BMSCs, showed a further increased adipogenic response. CONCLUSIONS: Markedly enhanced adipogenic molecular profile and increased cell proliferation of MFR BMSCs suggest a possible novel cellular/mechanistic link between the intrauterine nutritional stress and offspring metabolic phenotype. This provides new potential predictive and therapeutic targets against these conditions in the IUGR offspring.


Subject(s)
Adipogenesis/physiology , Fetal Growth Retardation/pathology , Maternal Nutritional Physiological Phenomena/physiology , Mesenchymal Stem Cells/metabolism , Peroxisome Proliferator-Activated Receptors/physiology , Wnt Signaling Pathway/physiology , Animals , Animals, Newborn , Caloric Restriction , Cell Differentiation , Disease Models, Animal , Down-Regulation , Female , Fetal Growth Retardation/genetics , Maternal Nutritional Physiological Phenomena/genetics , MicroRNAs , Phenotype , Pregnancy , RNA, Messenger , Rats , Rats, Sprague-Dawley , Up-Regulation
15.
Int J Obes (Lond) ; 39(4): 633-41, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25640766

ABSTRACT

Epigenetic mechanisms are emerging as mediators linking early environmental exposures during pregnancy with programmed changes in gene expression that alter offspring growth and development. There is irrefutable evidence from human and animal studies that nutrient and environmental agent exposures (for example, endocrine disruptors) during pregnancy may affect fetal/newborn development resulting in offspring obesity and obesity-associated metabolic abnormalities (metabolic syndrome). This concept of 'gestational programming' is associated with alterations to the epigenome (nongenomic) rather than changes in the DNA sequence (genomic). Epigenetic alterations induced by suboptimal maternal nutrition/endocrine factors include DNA methylation, histone modifications, chromatin remodeling and/or regulatory feedback by microRNAs, all of which have the ability to modulate gene expression and promote the metabolic syndrome phenotype. Recent studies have shown tissue-specific transcriptome patterns and phenotypes not only in the exposed individual, but also in subsequent progeny. Notably, the transmission of gestational programming effects to subsequent generations occurs in the absence of continued adverse environmental exposures, thus propagating the cycle of obesity and metabolic syndrome. This phenomenon may be attributed to an extrinsic process resulting from the maternal phenotype and the associated nutrient alterations occurring within each pregnancy. In addition, epigenetic inheritance may occur through somatic cells or through the germ line involving both maternal and paternal lineages. Since epigenetic gene modifications may be reversible, understanding how epigenetic mechanisms contribute to transgenerational transmission of obesity and metabolic dysfunction is crucial for the development of novel early detection and prevention strategies for programmed metabolic syndrome. In this review we discuss the evidence in human and animal studies for the role of epigenomic mechanisms in the transgenerational transmission of programmed obesity and metabolic syndrome.


Subject(s)
Maternal Nutritional Physiological Phenomena/genetics , Metabolic Syndrome/genetics , Obesity/genetics , Prenatal Exposure Delayed Effects/genetics , Animals , DNA Methylation , Disease Models, Animal , Disease Susceptibility , Environmental Exposure , Epigenomics , Female , Fetal Development/genetics , Humans , Infant, Newborn , Metabolic Syndrome/epidemiology , Obesity/epidemiology , Phenotype , Pregnancy , Prenatal Exposure Delayed Effects/epidemiology , Prenatal Exposure Delayed Effects/metabolism , Risk
16.
Int J Obes (Lond) ; 39(4): 671-6, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25298277

ABSTRACT

OBJECTIVE: Limited knowledge exists on the long-term implications of maternal gestational weight gain (GWG) on offspring health. Our objective was to examine whether high GWG in normal weight women is associated with adult offspring cardio-metabolic risk factors. METHODS: We used a cohort of 308 Danish women who gave birth in 1988-89 and whose offspring participated in a clinical examination at 20 years of age. Main outcome measures were offspring body mass index (BMI), waist circumference, weight-regulating hormones, blood lipids and glucose metabolism. Associations were assessed using multivariable linear and logistic regression models. RESULTS: A weak positive association was observed between GWG during the first 30 weeks and offspring anthropometry. Each 1-kg increase in maternal GWG was associated with 0.1-kg m(-2) higher (95% confidence interval (CI): 0.01, 0.2) offspring BMI and 10% (95% CI: 0.1%, 20%) higher odds of offspring overweight at the age of 20 years, with similar associations observed in both sexes. However, sex differences were observed for the association between maternal GWG and specific cardio-metabolic risk factors. Hence, a 1-kg increase in GWG was associated with 3.4% (95% CI; 0.8, 6.0%) higher homeostasis model assessment-estimated insulin resistance (HOMA-IR), 3.7% (95% CI: 1.4%, 6.2%) higher insulin and 10.7% (95% CI: 5.7%, 15.9%) higher leptin levels in male offspring. These associations were not observed in females, which may partly be explained by more frequent reports of dieting and physical exercise at follow-up among female offspring. CONCLUSIONS: In normal-weight women, high GWG may have modest long-term implications on offspring cardio-metabolic risk factors at adult age.


Subject(s)
Cardiovascular Diseases/physiopathology , Metabolic Syndrome/physiopathology , Weight Gain/physiology , Adolescent , Adult , Adult Children , Body Mass Index , Cardiovascular Diseases/epidemiology , Cardiovascular Diseases/genetics , Denmark/epidemiology , Female , Follow-Up Studies , Guidelines as Topic , Humans , Infant , Infant, Newborn , Insulin Resistance , Male , Maternal Nutritional Physiological Phenomena/genetics , Metabolic Syndrome/epidemiology , Metabolic Syndrome/genetics , Pregnancy , Prenatal Care , Risk Factors , Surveys and Questionnaires
17.
Pharmacol Res ; 100: 322-34, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26325301

ABSTRACT

Maternal nutrition during pregnancy and lactation influences the offspring's health in the long-term. Indeed, human epidemiological studies and animal model experiments suggest that either an excess or a deficit in maternal nutrition influence offspring development and susceptibility to metabolic disorders. Different epigenetic mechanisms may explain in part the way by which dietary factors in early critical developmental steps might be able to affect the susceptibility to develop metabolic diseases in adulthood. microRNAs are versatile regulators of gene expression and play a major role during tissue homeostasis and disease. Dietary factors have also been shown to modify microRNA expression. However, the role of microRNAs in fetal programming remains largely unstudied. This review evaluates in vivo studies conducted to analyze the effect of maternal diet on the modulation of the microRNA expression in the offspring and their influence to develop metabolic and cardiovascular disease in later life. In overall, the available evidence suggests that nutritional status during pregnancy influence offspring susceptibility to the development of cardiometabolic risk factors, partly through microRNA action. Thus, therapeutic modulation of microRNAs can open up new strategies to combat - later in life - the effects of nutritional insult during critical points of development.


Subject(s)
Heart Diseases/etiology , Heart Diseases/genetics , Maternal Nutritional Physiological Phenomena/genetics , MicroRNAs/genetics , Animals , Female , Humans , Lactation/genetics , Mother-Child Relations , Mothers , Pregnancy
18.
Hum Mol Genet ; 21(9): 2086-101, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22307237

ABSTRACT

In addition to the genetic constitution inherited by an organism, the developmental trajectory and resulting mature phenotype are also determined by mechanisms acting during critical windows in early life that influence and establish stable patterns of gene expression. This is the crux of the developmental origins of health and disease hypothesis that suggests undernutrition during gestation and infancy predisposes to ill health in later life. The hypothesis that periconceptional maternal micronutrient supplementation might affect fetal genome-wide methylation within gene promoters was explored in cord blood samples from offspring of Gambian women enrolled into a unique randomized, double blind controlled trial. Significant changes in the epigenome in cord blood DNA samples were further explored in a subset of offspring at 9 months. Gender-specific changes related to periconceptional nutritional supplementation were identified in cord blood DNA samples, some of which showed persistent changes in infant blood DNA samples. Significant effects of periconceptional micronutrient supplementation were also observed in postnatal samples which were not evident in cord blood. In this Gambian population, the increased death rate of individuals born in nutritionally poor seasons has been related to infection and it is of interest that we identified differential methylation at genes associated with defence against infection and immune response. Although the sample size was relatively small, these pilot data suggest that periconceptional nutrition in humans is an important determinant of newborn whole genome methylation patterns but may also influence postnatal developmental patterns of gene promoter methylation linking early with disease risk.


Subject(s)
Dietary Supplements , Epigenesis, Genetic , Fertilization/genetics , Micronutrients/administration & dosage , Adolescent , Adult , CpG Islands , DNA Methylation , Double-Blind Method , Female , Fetal Blood/metabolism , Gambia , Genomic Imprinting , Humans , Infant , Infant, Newborn , Male , Malnutrition/complications , Malnutrition/diet therapy , Malnutrition/genetics , Maternal Nutritional Physiological Phenomena/genetics , Middle Aged , Pregnancy , Pregnancy Complications/diet therapy , Pregnancy Complications/genetics , Prenatal Exposure Delayed Effects/genetics , Promoter Regions, Genetic , Sex Characteristics , Young Adult
19.
Biol Reprod ; 90(1): 5, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24258211

ABSTRACT

Maternal undernutrition around the time of conception is associated with an increased risk of insulin resistance in adulthood. We determined the effect of maternal undernutrition in the periconceptional period (PCUN, i.e., 60 days prior to 6 days after conception) and the preimplantation period (PIUN, i.e., 0-6 days after conception) on mRNA expression and protein abundance of key insulin-signaling molecules as well as the global microRNA expression in quadriceps muscle of singleton and twin fetal sheep in late gestation. In singleton fetuses, exposure to PCUN resulted in lower protein abundance of PIK3CB (P < 0.01), PRKCZ (P < 0.05), and pPRKCZ (Thr410) (P < 0.05) in skeletal muscle compared to controls. In PIUN singletons, there was a higher protein abundance of IRS1 (P < 0.05), PDPK1 (P < 0.05), and SLC2A4 (P < 0.05) compared to controls. In twins, PCUN resulted in higher protein abundance of IRS1 (P < 0.05), AKT2 (P < 0.05), PDPK1 (P < 0.05), and PRKCZ (P < 0.001), while PIUN also resulted in higher protein abundance of IRS1 (P < 0.05), PRKCZ (P < 0.001), and SLC2A4 (P < 0.05) in fetal muscle compared to controls. There were specific patterns of the types and direction of changes in the expression of 22 microRNAs in skeletal muscle after exposure to PCUN or PIUN and clear differences in these patterns between singleton and twin pregnancies. These findings provide evidence that maternal undernutrition around the time of conception induces changes in the expression of microRNAs, which may play a role in altering the abundance of the key insulin-signaling molecules in skeletal muscle and in the association between PCUN undernutrition and insulin resistance in adult life.


Subject(s)
Fertilization , Fetus/metabolism , Insulin/metabolism , Malnutrition/genetics , Maternal Nutritional Physiological Phenomena , MicroRNAs/genetics , Muscle, Skeletal/metabolism , Animals , Embryonic Development/genetics , Female , Fertilization/physiology , Litter Size , Malnutrition/metabolism , Maternal Nutritional Physiological Phenomena/genetics , MicroRNAs/metabolism , Pregnancy , Prenatal Exposure Delayed Effects/genetics , Prenatal Exposure Delayed Effects/metabolism , Sheep, Domestic , Signal Transduction/genetics
20.
Matern Child Health J ; 18(2): 450-61, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23780476

ABSTRACT

The "Life Course Perspective" proposes that environmental exposures, including biological, physical, social, and behavioral factors, as well as life experiences, throughout the entire life span, influence health outcomes in current and future generations. Nutrition, from preconception to adulthood, encompasses all of these factors and has the potential to positively or negatively shape the individual or population health trajectories and their intergenerational differences. This paper applies the T2E2 model (timing, timeline, equity and environment), developed by Fine and Kotelchuck, as an overlay to examine advances in nutritional science, as well as the complex associations between life stages, nutrients, nutrigenomics, and access to healthy foods, that support the life course perspective. Examples of the application of nutrition to each of the four constructs are provided, as well as a strong recommendation for inclusion of nutrition as a key focal point for all health professionals as they address solutions to optimize health outcomes, both domestically and internationally. The science of nutrition provides strong evidence to support the concepts of the life course perspective. These findings lend urgency to the need to improve population health across the life span and over generations by ensuring ready access to micronutrient-dense foods, opportunities to balance energy intake with adequate physical activity and the need for biological, social, physical, and macro-level environments that support critical phases of human development. Recommendations for the application of the life course perspective, with a focus on the emerging knowledge of nutritional science, are offered in an effort to improve current maternal and child health programs, policies, and service delivery.


Subject(s)
Family Health , Health Status Disparities , Human Development , Nutrigenomics , Nutritional Status/physiology , Social Determinants of Health , Adolescent , Adult , Aged , Child , Child Nutritional Physiological Phenomena/genetics , Child Nutritional Physiological Phenomena/physiology , Child, Preschool , Environment , Epigenesis, Genetic , Female , Food Supply/economics , Food Supply/standards , Humans , Infant , Maternal Nutritional Physiological Phenomena/genetics , Maternal Nutritional Physiological Phenomena/physiology , Nutritional Status/genetics , Preconception Care , Pregnancy , Prenatal Care
SELECTION OF CITATIONS
SEARCH DETAIL