Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 107
Filter
1.
Nutrients ; 13(12)2021 Nov 26.
Article in English | MEDLINE | ID: mdl-34959795

ABSTRACT

We investigated if supplementing obese mothers (MO) with docosahexaenoic acid (DHA) improves milk long-chain polyunsaturated fatty acid (LCPUFA) composition and offspring anxiety behavior. From weaning throughout pregnancy and lactation, female Wistar rats ate chow (C) or a high-fat diet (MO). One month before mating and through lactation, half the mothers received 400 mg DHA kg-1 d-1 orally (C+DHA or MO+DHA). Offspring ate C after weaning. Maternal weight, total body fat, milk hormones, and milk nutrient composition were determined. Pups' milk nutrient intake was evaluated, and behavioral anxiety tests were conducted. MO exhibited increased weight and total fat, and higher milk corticosterone, leptin, linoleic, and arachidonic acid (AA) concentrations, and less DHA content. MO male and female offspring had higher ω-6/ ω-3 milk consumption ratios. In the elevated plus maze, female but not male MO offspring exhibited more anxiety. MO+DHA mothers exhibited lower weight, total fat, milk leptin, and AA concentrations, and enhanced milk DHA. MO+DHA offspring had a lower ω-6/ω-3 milk intake ratio and reduced anxiety vs. MO. DHA content was greater in C+DHA milk vs. C. Supplementing MO mothers with DHA improves milk composition, especially LCPUFA content and ω-6/ω-3 ratio reducing offspring anxiety in a sex-dependent manner.


Subject(s)
Animals, Newborn/psychology , Behavior, Animal/drug effects , Dietary Supplements , Docosahexaenoic Acids/administration & dosage , Milk/chemistry , Animals , Anxiety/prevention & control , Eating/psychology , Fatty Acids, Omega-3/analysis , Fatty Acids, Omega-6/analysis , Fatty Acids, Unsaturated/analysis , Female , Lactation , Male , Maternal Nutritional Physiological Phenomena/drug effects , Obesity , Pregnancy , Rats , Rats, Wistar , Sex Factors
2.
Nutrients ; 13(11)2021 Nov 18.
Article in English | MEDLINE | ID: mdl-34836384

ABSTRACT

Several studies suggest that the maternal protein content and source can affect the offspring's health. However, the chronic impact of maternal quality and quantity protein restriction, and reversible changes upon rehabilitation, if any, in the offspring, remains elusive. This study examined the effects of maternal low-quality protein (LQP) and low-protein (LP) intake from preconception to post-weaning, followed by rehabilitation from weaning, on body composition, glucose-homeostasis, and metabolic factors in rat offspring. Wistar rats were exposed to normal protein (NP; 20% casein), LQP (20% wheat gluten) or LP (8% casein) isocaloric diets for 7 weeks before pregnancy until lactation. After weaning, the offspring were exposed to five diets: NP, LQP, LQPR (LQP rehabilitated with NP), LP, and LPR (LP rehabilitated with NP) for 16 weeks. Body composition, glucose-homeostasis, lipids, and plasma hormones were investigated. The LQP and LP offspring had lower bodyweight, fat and lean mass, insulin and HOMA-IR than the NP. The LQP offspring had higher cholesterol, T3 and T4, and lower triacylglycerides and glucose, while these were unaltered in LP compared to NP. The majority of the above outcomes were reversed upon rehabilitation. These results suggest that the chronic exposure of rats to maternal LQP and LP diets induced differential adverse effects by influencing body composition and metabolism, which were reversed upon rehabilitation.


Subject(s)
Blood Glucose/metabolism , Body Composition/drug effects , Diet, Protein-Restricted/adverse effects , Dietary Proteins/administration & dosage , Homeostasis/drug effects , Animals , Animals, Newborn/metabolism , Female , Male , Maternal Nutritional Physiological Phenomena/drug effects , Pregnancy , Rats , Rats, Wistar
3.
Nutrients ; 13(9)2021 Aug 31.
Article in English | MEDLINE | ID: mdl-34578953

ABSTRACT

Maternal supplementation during pregnancy with docosahexaenoic acid (DHA) is internationally recommended to avoid postpartum maternal depression in the mother and improve cognitive and neurological outcomes in the offspring. This study was aimed at determining whether this nutritional intervention, in the rat, protects the offspring against the development of obesity and its associated metabolic disorders. Pregnant Wistar rats received an extract of fish oil enriched in DHA or saline (SAL) as placebo by mouth from the beginning of gestation to the end of lactation. At weaning, pups were fed standard chow or a free-choice, high-fat, high-sugar (fc-HFHS) diet. Compared to animals fed standard chow, rats exposed to the fc-HFHS diet exhibited increased body weight, liver weight, body fat and leptin in serum independently of saline or DHA maternal supplementation. Nevertheless, maternal DHA supplementation prevented both the glucose intolerance and the rise in serum insulin resulting from consumption of the fc-HFHS diet. In addition, animals from the DHA-fc-HFHS diet group showed decreased hepatic triglyceride accumulation compared to SAL-fc-HFHS rats. The beneficial effects on glucose homeostasis declined with age in male rats. Yet, the preventive action against hepatic steatosis was still present in 6-month-old animals of both sexes and was associated with decreased hepatic expression of lipogenic genes. The results of the present work show that maternal DHA supplementation during pregnancy programs a healthy phenotype into the offspring that was protective against the deleterious effects of an obesogenic diet.


Subject(s)
Animal Nutritional Physiological Phenomena/drug effects , Diet, High-Fat/adverse effects , Docosahexaenoic Acids/pharmacology , Fatty Liver/prevention & control , Lactation , Animals , Diet, High-Fat/methods , Dietary Supplements , Disease Models, Animal , Docosahexaenoic Acids/administration & dosage , Fatty Liver/etiology , Female , Maternal Nutritional Physiological Phenomena/drug effects , Pregnancy , Rats , Rats, Wistar
4.
Nutrients ; 13(9)2021 Sep 20.
Article in English | MEDLINE | ID: mdl-34579155

ABSTRACT

Developmental programming of chronic diseases by perinatal exposures/events is the basic tenet of the developmental origins hypothesis of adult disease (DOHaD). With consumption of fructose becoming more common in the diet, the effect of fructose exposure during pregnancy and lactation is of increasing relevance. Human studies have identified a clear effect of fructose consumption on maternal health, but little is known of the direct or indirect effects on offspring. Animal models have been utilized to evaluate this concept and an association between maternal fructose and offspring chronic disease, including hypertension and metabolic syndrome. This review will address the mechanisms of developmental programming by maternal fructose and potential options for intervention.


Subject(s)
Fructose/adverse effects , Prenatal Exposure Delayed Effects/chemically induced , Animals , Female , Humans , Hypertension/chemically induced , Maternal Nutritional Physiological Phenomena/drug effects , Metabolic Syndrome/chemically induced , Pregnancy
5.
Nutrients ; 13(7)2021 Jul 20.
Article in English | MEDLINE | ID: mdl-34371987

ABSTRACT

It was previously observed that in a population of a high-income country, dietary multiple micronutrient supplementation in pregnancy was associated with an increased risk of gestational diabetes (GDM) and increased offspring size at birth. In this follow-up study, we investigated whether similar changes are observed with dietary iron supplementation. For this we used the prospective Cambridge Baby Growth Study with records of maternal GDM status, nutrient supplementation, and extensive offspring birth size measurements. Maternal iron supplementation in pregnancy was associated with GDM development (risk ratio 1.67 (1.01-2.77), p = 0.048, n = 677) as well as offspring size and adiposity (n = 844-868) at birth in terms of weight (ß' = 0.078 (0.024-0.133); p = 0.005), head circumference (ß' = 0.060 (0.012-0.107); p = 0.02), body mass index (ß' = 0.067 (0.014-0.119); p = 0.01), and various skinfold thicknesses (ß' = 0.067-0.094; p = 0.03-0.003). In a subset of participants for whom GDM statuses were available, all these associations were attenuated by adjusting for GDM. Iron supplementation also attenuated the associations between multiple micronutrient supplementation and these same measures. These results suggest that iron supplementation may mediate the effects associated with multiple micronutrient supplementation in pregnancy in a high-income country, possibly through the increased risk of developing GDM.


Subject(s)
Birth Weight/drug effects , Dietary Supplements , Iron, Dietary/adverse effects , Maternal Nutritional Physiological Phenomena/drug effects , Micronutrients/adverse effects , Adiposity/drug effects , Adult , Body Mass Index , Diabetes, Gestational/chemically induced , Diabetes, Gestational/physiopathology , Female , Follow-Up Studies , Humans , Infant, Newborn , Iron, Dietary/administration & dosage , Male , Micronutrients/administration & dosage , Pregnancy , Prospective Studies , Skinfold Thickness
6.
Clin Nutr ; 40(9): 5053-5061, 2021 09.
Article in English | MEDLINE | ID: mdl-34455263

ABSTRACT

BACKGROUND: Pregnancy generally alters the balance of maternal metabolism, but the molecular profiles in early pregnancy and associated factors of folate supplementation in pregnant women remains incompletely understood. METHODS: Untargeted metabonomics based on high-performance liquid chromatography-high-resolution mass spectrometry integrated with multivariate metabolic pathway analysis were applied to characterize metabolite profiles and associated factors of folate supplements in early pregnancy. The metabolic baseline of early pregnancy was determined by metabolic analysis of 510 serum samples from 131 non-pregnant and 379 pregnant healthy Chinese women. The pathophysiology of adaptive reactions and metabolic challenges induced by folate supplementation in early pregnancy was further compared between pregnant women with (n = 168) and without (n = 184) folate supplements. RESULTS: Compared with non-pregnant participants, 106 metabolites, majority of which are related to amino acids and lysophosphatidylcholine/phosphatidylcholine, and 13 metabolic pathways were significantly changed in early pregnancy. The supplementation of folate in early pregnancy induced marked changes in N-acyl ethanolamine 22:0, N-acyl taurine 18:2, glycerophosphoserine 44:1 and 8,11,14-eicosatrienoate, proline, and aminoimidazole ribotide levels. CONCLUSIONS: During early pregnancy, the metabolism of amino acids significantly changes to meet the physiological requirements of pregnant women. Folate intake may change glucose and lipid metabolism. These findings provide a comprehensive landscape for understanding the basic characteristics and gestational metabolic networks of early pregnancy and folate supplementation. This study provides a basis for further research into the relationship between metabolic markers and pregnancy diseases. TRIAL REGISTRATION: This study protocol was registered on www.ClinicalTrials.gov, NCT03651934, on August 29, 2018 (prior to recruitment).


Subject(s)
Amino Acids/metabolism , Dietary Supplements , Folic Acid/administration & dosage , Maternal Nutritional Physiological Phenomena/drug effects , Metabolic Networks and Pathways/drug effects , Adult , China , Cross-Sectional Studies , Female , Healthy Volunteers , Humans , Metabolomics , Pregnancy
7.
Am J Physiol Regul Integr Comp Physiol ; 321(3): R364-R376, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34259017

ABSTRACT

Preeclampsia is a spontaneously occurring pregnancy complication diagnosed by new-onset hypertension and end-organ dysfunction with or without proteinuria. This pregnancy-specific syndrome contributes to maternal morbidity and mortality and can have detrimental effects on fetal outcomes. Preeclampsia is also linked to increased risk of maternal cardiovascular disease throughout life. Despite intense investigation of this disorder, few treatment options are available. The aim of this study was to investigate the potential therapeutic effects of maternal l-citrulline supplementation on pregnancy-specific vascular dysfunction in the male C57BL/6J × female C57BL/6J C1q-/- preeclampsia-like mouse model. l-Citrulline is a nonessential amino acid that is converted to l-arginine to promote smooth muscle and blood vessel relaxation and improve nitric oxide (NO)-mediated vascular function. To model a preeclampsia-like pregnancy, female C57BL/6J mice were mated to C1q-/- male mice, and a subset of dams was supplemented with l-citrulline throughout pregnancy. Blood pressure, systemic vascular glycocalyx, and ex vivo vascular function were investigated in late pregnancy, and postpartum at 6 and 10 mo of age. Main findings show that l-citrulline reduced blood pressure, increased vascular glycocalyx volume, and rescued ex-vivo vascular function at gestation day 17.5 in this preeclampsia-like model. The vascular benefit of l-citrulline also extended postpartum, with improved vascular function and glycocalyx measures at 6 and 10 mo of age. l-Citrulline-mediated vascular improvements appear, in part, attributable to NO pathway signaling. Taken together, l-citrulline supplementation during pregnancy appears to have beneficial effects on maternal vascular health, which may have translational implications for improved maternal cardiovascular health.


Subject(s)
Citrulline/pharmacology , Maternal Nutritional Physiological Phenomena/drug effects , Parturition/drug effects , Pre-Eclampsia/drug therapy , Animals , Arginine/blood , Blood Pressure/drug effects , Citrulline/blood , Female , Mice, Inbred C57BL , Placenta/metabolism , Pre-Eclampsia/physiopathology , Pregnancy
8.
Int J Mol Sci ; 22(9)2021 Apr 30.
Article in English | MEDLINE | ID: mdl-33946456

ABSTRACT

Maternal nutritional imbalances, in addition to maternal overweight and obesity, can result in long-term effects on the metabolic health of the offspring, increasing the risk of common non-communicable disorders such as obesity, diabetes and cardiovascular disease. This increased disease risk may also be transmitted across generations. Unfortunately, lifestyle interventions have shown reduced compliancy and limited efficacy. Resveratrol is a natural polyphenolic compound reported to have pleiotropic beneficial actions including a possible protective effect against the metabolic programming induced by poor dietary habits during development. However, studies to date are inconclusive regarding the potential metabolic benefits of maternal resveratrol supplementation during pregnancy and lactation on the offspring. Moreover, the responses to metabolic challenges are suggested to be different in males and females, suggesting that the effectiveness of treatment strategies may also differ, but many studies have been performed only in males. Here we review the current evidence, both in humans and animal models, regarding the possible beneficial effects of maternal resveratrol intake on the metabolic health of the offspring and highlight the different effects of resveratrol depending on the maternal diet, as well as the differential responses of males and females.


Subject(s)
Antioxidants/pharmacology , Maternal Nutritional Physiological Phenomena , Resveratrol/pharmacology , Animals , Antioxidants/administration & dosage , Female , Humans , Infant, Newborn , Lactation , Life Style , Male , Maternal Nutritional Physiological Phenomena/drug effects , Obesity/metabolism , Pregnancy , Resveratrol/administration & dosage
9.
Nutrients ; 13(4)2021 Apr 16.
Article in English | MEDLINE | ID: mdl-33923768

ABSTRACT

Surveillance data have highlighted continued disparities in neural tube defects (NTDs) by race-ethnicity in the United States. Starting in 2016, the Food and Drug Administration (FDA) authorized voluntary folic acid fortification of corn masa flour to reduce the risk of neural tube defects (NTDs) among infants of Hispanic women of reproductive age. To assess the impact of voluntary corn masa fortification, cross-sectional data from the National Health and Nutrition Examination Survey (NHANES) 2011-2018 for Hispanic women of reproductive age with available red blood cell (RBC) folate concentrations were analyzed, with additional analyses conducted among Hispanic women whose sole source of folic acid intake was fortified foods (enriched cereal grain products (ECGP) only), excluding ready-to-eat cereals and supplements. RBC folate concentration (adjusted geometric mean) among Hispanic women of reproductive age did not differ between 2011-2016 and 2017-2018, though RBC folate concentration increased significantly among lesser acculturated Hispanic women consuming ECGP only. Concentrations of RBC folate for those born outside the U.S and residing in the U.S <15 years increased from 894 nmol/L (95% CI: 844-946) in 2011-2016 to 1018 nmol/L (95% CI: 982-1162; p < 0.001) in 2017-2018. Primarily Spanish-speaking Hispanic women of reproductive age who only consumed ECGP saw an increase from 941 nmol/L (95% CI: 895-990) in 2011-2016 to 1034 nmol/L (95% CI: 966-1107; p = 0.03) in 2017-2018. By subpopulation, we observed no significant changes in the proportion at risk of NTDs (<748 nmol/L) and no changes in the model-based estimated NTD rates following voluntary corn masa fortification. This analysis suggests that there is a remaining risk among Hispanics for folate sensitive NTDs, though continued monitoring of folate status in future NHANES data cycles will help inform the long-term efficacy of voluntary fortification of corn masa flour.


Subject(s)
Flour/analysis , Folic Acid/administration & dosage , Food, Fortified/analysis , Hispanic or Latino/statistics & numerical data , Zea mays/chemistry , Acculturation , Adult , Anencephaly/epidemiology , Anencephaly/ethnology , Anencephaly/prevention & control , Cross-Sectional Studies , Erythrocytes/chemistry , Female , Folic Acid/blood , Humans , Infant, Newborn , Male , Maternal Nutritional Physiological Phenomena/drug effects , Maternal Nutritional Physiological Phenomena/ethnology , Nutrition Surveys , Nutritional Status , United States/epidemiology , Young Adult
10.
Life Sci ; 271: 119179, 2021 Apr 15.
Article in English | MEDLINE | ID: mdl-33577849

ABSTRACT

BACKGROUND: Male infertility and reproductive dysfunctions have become major global health problems. Although several causative factors have been attributed to this challenge, of importance are alterations in maternal-foetal environment, diet-induced transcriptional changes and dysregulation in chemical signaling via hypothalamic-gonadal axis. AIM: The present study investigated the impact of maternal high-fat diet (HFD) consumption and the putative role of Quercetin-3-O-rutinoside on reproductive functions of male offspring rats at critical developmental stages with a quest to unravel the underpinned molecular changes. MATERIALS AND METHODS: Fifty-six pregnant rats (previously fed normal diet ND) or 45% HFD) were maintained on supplemented chow (150 mg/kg QR) - ND/QR, HFD/QR throughout gestation. Subsequently, dams (n = 7) and offspring (n = 6) were sacrificed at post-natal day (PND) 21, 28 and 35, respectively, and the blood, placenta, hypothalamus (HT), and testicular samples were processed for molecular analysis of Gonadotropin-releasing hormone (GnRH), Luteinizing hormone (LH), testosterone, chemerin, chemokine-like receptor 1 (CMKLR1), tumour necrosis factor α (TNF-α), interleukin 1ß (IL-1ß) and nuclear factor kappa B (NF-κB). KEY FINDINGS: We observed a significant decrease in GnRH level in the HFD group at PND21 and PND28 in male offspring and treatment with QR significantly reduced GnRH. There was a significant reduction in LH levels in the HFD group at PND 21 in the male offspring accompanied by a significant decrease in testosterone level at PND 28 and PND35 which appears to be age dependent. In the HT, Chemerin and CMKLR1 was significantly upregulated in the HFD group at PND 21 and PND 35 respectively while CMKLR1 was significantly downregulated in the HFD group of the placenta and testis at PND 21. TNF-α, IL-1ß and NF-κB were also expressed in the placenta, HT and testis at PND 21. SIGNIFICANCE: Male fertility is affected by maternal HFD consumption while chemerin, CMKLR1 and TNF-α, may play a significant role in male steroidogenesis. Treatment with QR had little or no ameliorative effect on HFD induced alterations in male reproductive functions.


Subject(s)
Diet, High-Fat/adverse effects , Glucosides/pharmacology , Maternal Nutritional Physiological Phenomena/physiology , Prenatal Exposure Delayed Effects/metabolism , Quercetin/analogs & derivatives , Reproduction/physiology , Testis/metabolism , Animals , Female , Gonadotropin-Releasing Hormone/metabolism , Male , Maternal Nutritional Physiological Phenomena/drug effects , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Quercetin/pharmacology , Rats , Rats, Sprague-Dawley , Reproduction/drug effects , Testis/drug effects , Testosterone/metabolism
11.
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
12.
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
13.
BMC Endocr Disord ; 20(1): 139, 2020 Sep 14.
Article in English | MEDLINE | ID: mdl-32928167

ABSTRACT

BACKGROUND: The infants born to women who are overweight or obese in pregnancy are at an increased risk of being born macrosomic or large for gestational age. Antenatal dietary and lifestyle interventions have been shown to be ineffective at reducing this risk. Our aim was to examine the effects of metformin in addition to a diet and lifestyle intervention on fetal growth and adiposity among women with a BMI above the healthy range. METHODS: Women who had a body mass index ≥25 kg/m2 in early pregnancy, and a singleton gestation, were enrolled in the GRoW trial from three public maternity hospitals in metropolitan Adelaide. Women were invited to have a research ultrasounds at 28 and 36 weeks' gestation at which ultrasound measures of fetal biometry and adiposity were obtained. Fetal biometry z-scores and trajectories were calculated. Measurements and calculations were compared between treatment groups. This secondary analysis was pre-specified. RESULTS: Ultrasound data from 511 women were included in this analysis. The difference in femur length at 36 weeks' gestation was (0.07 cm, 95% CI 0.01-0.14 cm, p = 0.019) and this was was statistically significant, however the magnitude of effect was small. Differences between treatment groups for all other fetal biometry measures, z-scores, estimated fetal weight, and adiposity measures at 28 and 36 weeks' gestation were similar. CONCLUSIONS: The addition of metformin to dietary and lifestyle advice in pregnancy for overweight and obese women has no clinically relevant effect on ultrasound measures of fetal biometry or adiposity. TRIAL REGISTRATION: Australian and New Zealand Clinical Trials Registry ( ACTRN12612001277831 ).


Subject(s)
Adiposity/drug effects , Fetal Development/drug effects , Metformin/pharmacology , Prenatal Care/methods , Adiposity/physiology , Adult , Body Mass Index , Diet , Exercise/physiology , Female , Fetus/drug effects , Fetus/metabolism , Gestational Age , Humans , Life Style , Maternal Nutritional Physiological Phenomena/drug effects , Metformin/therapeutic use , Pregnancy , Pregnancy Complications/prevention & control , Prenatal Exposure Delayed Effects/metabolism , Prenatal Exposure Delayed Effects/physiopathology , Risk Reduction Behavior , Young Adult
14.
Nutrients ; 12(9)2020 Sep 04.
Article in English | MEDLINE | ID: mdl-32899673

ABSTRACT

Recent studies have shown that maternal supplementation with folate and long-chain polyunsaturated fatty acids (LC-PUFAs) during pregnancy may affect children's brain development. We aimed at examining the potential long-term effect of maternal supplementation with fish oil (FO) and/or 5-methyl-tetrahydrofolate (5-MTHF) on the brain functionality of offspring at the age of 9.5-10 years. The current study was conducted as a follow-up of the Spanish participants belonging to the Nutraceuticals for a Healthier Life (NUHEAL) project; 57 children were divided into groups according to mother's supplementation and assessed through functional magnetic resonance imaging (fMRI) scanning and neurodevelopment testing. Independent component analysis and double regression methods were implemented to investigate plausible associations. Children born to mothers supplemented with FO (FO and FO + 5-MTHF groups, n = 33) showed weaker functional connectivity in the default mode (DM) (angular gyrus), the sensorimotor (SM) (motor and somatosensory cortices) and the fronto-parietal (FP) (angular gyrus) networks compared to the No-FO group (placebo and 5-MTHF groups, n = 24) (PFWE < 0.05). Furthermore, no differences were found regarding the neuropsychological tests, except for a trend of better results in an object recall (memory) test. Considering the No-FO group, the aforementioned networks were associated negatively with attention and speed-processing functions. Mother's FO supplementation during pregnancy seems to be able to shape resting-state network functioning in their children at school age and appears to produce long-term effects on children´s cognitive processing.


Subject(s)
Brain/growth & development , Child Development/drug effects , Dietary Supplements , Fish Oils/administration & dosage , Maternal Nutritional Physiological Phenomena/drug effects , Tetrahydrofolates/administration & dosage , Adult , Brain/diagnostic imaging , Child , Female , Follow-Up Studies , Humans , Magnetic Resonance Imaging , Male , Maternal Exposure , Nerve Net/diagnostic imaging , Nerve Net/growth & development , Pregnancy , Principal Component Analysis , Regression Analysis , Rest/physiology
15.
Anim Reprod Sci ; 219: 106531, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32828406

ABSTRACT

In this study there was evaluation of effects of dietary inulin during late gestation on sow physiology, farrowing duration and piglet performance. At day 80 of gestation sows were randomly assigned to four groups:basal diet (CON); or basal diet with 0.8 %; 1.6 %; or 2.4 % inulin. The feeding of the diet with 1.6 % inulin resulted in larger weights of the litter at birth a shorter duration of the farrowing period, lesser average birth interval between piglets, lesser number of piglets dead at birth, and fewer piglets/sow dead at birth (P < 0.05). When sows were fed 0.8 % and 1.6 % IN, there was a larger litter weight at weaning, sow average daily feed intake and piglet average daily gain during lactation compared with values for these variables in the CON group (P <  0.05). Additionally, there was an increase in serum concentration of free fatty acid, total cholesterol, and high-density lipoprotein cholesterol with increasing amounts of inulin in the diet (linear, P <  0.05). Sows fed 1.6 % IN had greater serum concentrations of glucose than those in the CON group (P <  0.05). Furthermore, there was a linear increase in serum activity of total antioxidant capacity, total superoxide dismutase and glutathione peroxidase with increasing amounts of inulin in the diet (P <  0.05). In conclusion, results of the present study indicated feeding inulin during late gestation improved reproductive performance of sows, thus, may be a novel additive for the pig industry in improving efficiency of pork production.


Subject(s)
Animals, Newborn/growth & development , Inulin/pharmacology , Parturition/drug effects , Pregnancy, Animal , Swine/physiology , Animal Feed , Animal Nutritional Physiological Phenomena/drug effects , Animals , Body Composition/drug effects , Diet , Dietary Supplements , Female , Gestational Age , Lactation/physiology , Litter Size/drug effects , Maternal Nutritional Physiological Phenomena/drug effects , Pregnancy , Pregnancy, Animal/drug effects , Weaning
16.
Nutrients ; 12(7)2020 Jul 17.
Article in English | MEDLINE | ID: mdl-32708852

ABSTRACT

The early life period is crucial for the maturation of the intestinal barrier, its immune system, and a life-long beneficial host-microbiota interaction. The study aims to assess the impact of a beneficial dietary (short-chain fructooligosaccharides, scFOS) supplementation vs. a detrimental dietary environment (such as mycotoxin deoxynivalenol, DON) on offspring intestinal immune system developmental profiles. Sows were given scFOS-supplemented or DON-contaminated diets during the last 4 weeks of gestation, whereas force-feeding piglets with DON was performed during the first week of offspring life. Intestinal antigen-presenting cell (APC) subset frequency was analyzed by flow cytometry in the Peyer's patches and in lamina propria and the responsiveness of intestinal explants to toll-like receptor (TLR) ligands was performed using ELISA and qRT-PCR from post-natal day (PND) 10 until PND90. Perinatal exposure with scFOS did not affect the ontogenesis of APC. While it early induced inflammatory responses in piglets, scFOS further promoted the T regulatory response after TLR activation. Sow and piglet DON contamination decreased CD16+ MHCII+ APC at PND10 in lamina propria associated with IFNγ inflammation and impairment of Treg response. Our study demonstrated that maternal prebiotic supplementation and mycotoxin contamination can modulate the mucosal immune system responsiveness of offspring through different pathways.


Subject(s)
Food Contamination/analysis , Immune System/metabolism , Mucous Membrane/metabolism , Mycotoxins/toxicity , Prebiotics/administration & dosage , Animal Feed/analysis , Animal Feed/toxicity , Animal Nutritional Physiological Phenomena , Animals , Animals, Newborn , Cytokines/metabolism , Diet/veterinary , Dietary Supplements , Female , Interferon-gamma/metabolism , Maternal Nutritional Physiological Phenomena/drug effects , Mycotoxins/administration & dosage , Oligosaccharides/administration & dosage , Pregnancy , Pregnancy, Animal/drug effects , Pregnancy, Animal/immunology , Receptors, IgG/metabolism , Swine , Trichothecenes/administration & dosage , Trichothecenes/toxicity
17.
Food Funct ; 11(8): 6889-6898, 2020 Aug 01.
Article in English | MEDLINE | ID: mdl-32691032

ABSTRACT

The study investigated the effect of in-feed administration of dried thyme leaf and celery seed mixture (at 1 : 1 DM basis) compared with salinomycin ionophore on milk production and milk nutritive value of Barki ewes. Thirty ewes (37.5 ± 1.8 kg), divided into 3 treatment groups, were fed: (1) a complete control diet comprising concentrates and fodder maize (Zea mays L.) at 60 : 40 dry matter basis, (2) the control diet plus 20 g of thyme and celery mixture supplementation and (3) the control diet supplemented with 1 g of salinomycin per ewe daily for 90 days. Inclusion of thyme-celery treatment increased (P < 0.05) weight gain, average daily gain, milk yield, milk component yields, and feed efficiency, without affecting milk composition. In addition, the thyme-celery treatment enhanced (P < 0.05) nutrient intake and digestibility, total ruminal volatile fatty acids, branched chain fatty acids, and acetate proportions and decreased ammonia-N concentration. Thyme-celery treatment increased (P < 0.05) serum glucose, thyroxine, and glutamate-pyruvate transaminase concentrations. It is concluded that the thyme and celery mixture (1 : 1 DM basis) at 20 g per lactating ewe daily can replace the salinomycin ionophore. Enhanced feed utilization and lactational performance as well as milk nutritive value for human consumption were observed with the natural additive mixture supplementation.


Subject(s)
Anti-Bacterial Agents/pharmacology , Apium , Dietary Supplements , Plant Extracts/pharmacology , Thymus Plant , Acetates/blood , Alanine Transaminase/blood , Ammonia/blood , Animal Feed/analysis , Animal Nutritional Physiological Phenomena/drug effects , Animals , Blood Glucose/drug effects , Complex Mixtures , Digestion/drug effects , Eating/drug effects , Fatty Acids/blood , Fatty Acids, Volatile/blood , Female , Fermentation/drug effects , Lactation/drug effects , Maternal Nutritional Physiological Phenomena/drug effects , Milk/chemistry , Plant Leaves/chemistry , Seeds/chemistry , Sheep , Stomach, Ruminant/drug effects , Thyroxine/blood
18.
Blood ; 136(19): 2206-2216, 2020 11 05.
Article in English | MEDLINE | ID: mdl-32584957

ABSTRACT

Iron disorders are associated with adverse pregnancy outcomes, yet iron homeostatic mechanisms during pregnancy are poorly understood. In humans and rodents, the iron-regulatory hormone hepcidin is profoundly decreased in pregnant mothers, which is thought to ensure adequate iron availability for transfer across placenta. However, the fetal liver also produces hepcidin, which may regulate fetal iron endowment by controlling placental iron export. To determine the relative contribution of maternal vs embryo hepcidin to the control of embryo iron endowment in iron-sufficient or iron-overloaded mice, we generated combinations of mothers and embryos that had or lacked hepcidin. We found that maternal, but not embryonic, hepcidin determined embryo and placental iron endowment in a healthy pregnancy. We further determined that inflammation can counteract pregnancy-dependent suppression of maternal hepcidin. To establish how essential maternal hepcidin suppression is for embryo iron homeostasis, we mimicked the range of maternal hepcidin activity by administering a hepcidin peptide mimetic to pregnant mice. This also allowed us to determine the effect of isolated maternal hepcidin excess on pregnancy, in the absence of other confounding effects of inflammation. Higher doses of hepcidin agonist caused maternal iron restriction and anemia, lower placenta and embryo weight, embryo anemia, and increased embryo mortality. Low agonist doses did not cause maternal anemia but still adversely affected the embryo, causing anemia, tissue iron deficiency (including in the brain), and decreased weight. Our studies demonstrate that suppression of maternal hepcidin during pregnancy is essential for maternal and embryo iron homeostasis and health.


Subject(s)
Embryo, Mammalian/metabolism , Fetus/metabolism , Hepcidins/pharmacology , Homeostasis , Iron/metabolism , Maternal Nutritional Physiological Phenomena/drug effects , Placenta/drug effects , Animals , Embryo, Mammalian/cytology , Embryo, Mammalian/drug effects , Female , Fetus/drug effects , Maternal-Fetal Exchange , Mice , Mice, Inbred C57BL , Mothers , Placenta/metabolism , Pregnancy , Receptors, Transferrin/metabolism
19.
Int J Food Sci Nutr ; 71(8): 954-964, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32393141

ABSTRACT

Soy consumption and its components, including its protein, are related to the beneficial effects of the lipid profile, decreased insulin resistance and glycaemia. However, the safety of the consumption of products containing phytoestrogens in critical stages of development has been questioned, since they may be associated with endocrine-metabolic dysfunctions in adult life. The purpose is to evaluate the effects of maternal dietary soy protein isolate (SPI) during lactation on the breast milk composition, body composition, lipid and glycaemic profiles, and thyroid hormones of dams and offspring at weaning (21 days) and in adulthood (150 days). Lactating rats were divided into casein control (C) and SPI diet groups. At 150 days, the SPI offspring presented lower body protein mass and total mineral content, higher serum FT4, insulin, TC and TG. Maternal consumption of SPI during lactation programmes the progeny to higher metabolic risk profile.


Subject(s)
Diet , Lactation/drug effects , Maternal Nutritional Physiological Phenomena/drug effects , Soybean Proteins/administration & dosage , Animals , Blood Glucose , Body Composition/drug effects , Female , Insulin/blood , Lipids/blood , Male , Milk, Human , Rats , Rats, Wistar , Weaning
20.
Reprod Sci ; 27(4): 963-976, 2020 04.
Article in English | MEDLINE | ID: mdl-32124397

ABSTRACT

The clinical significance of periconceptional folic acid supplementation (FAS) in the prevention of neonatal neural tube defects (NTDs) has been recognized for decades. Epidemiological data and experimental findings have consistently been indicating an association between folate deficiency in the first trimester of pregnancy and poor fetal development as well as offspring health (i.e., NTDs, isolated orofacial clefts, neurodevelopmental disorders). Moreover, compelling evidence has suggested adverse effects of folate overload during perinatal period on offspring health (i.e., immune diseases, autism, lipid disorders). In addition to several single-nucleotide polymorphisms (SNPs) in genes related to folate one-carbon metabolism (FOCM), folate concentrations in maternal serum/plasma/red blood cells must be considered when counseling FAS. Epigenetic information encoded by 5-methylcytosines (5mC) plays a critical role in fetal development and offspring health. S-adenosylmethionine (SAM), a methyl donor for 5mC, could be derived from FOCM. As such, folic acid plays a double-edged sword role in offspring health via mediating DNA methylation. However, the underlying epigenetic mechanism is still largely unclear. In this review, we summarized the link across DNA methylation, maternal FAS, and offspring health to provide more evidence for clinical guidance in terms of precise FAS dosage and time point. Future studies are, therefore, required to set up the reference intervals of folate concentrations at different trimesters of pregnancy for different populations and to clarify the epigenetic mechanism for specific offspring diseases.


Subject(s)
DNA Methylation/drug effects , Epigenesis, Genetic , Folic Acid/administration & dosage , Maternal Nutritional Physiological Phenomena/genetics , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/genetics , Animals , Dietary Supplements , Female , Fetal Development/drug effects , Folic Acid/adverse effects , Folic Acid Deficiency/genetics , Humans , Maternal Nutritional Physiological Phenomena/drug effects , Pregnancy
SELECTION OF CITATIONS
SEARCH DETAIL
...