Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 69
Filter
1.
Clin Sci (Lond) ; 135(9): 1103-1126, 2021 05 14.
Article in English | MEDLINE | ID: mdl-33899910

ABSTRACT

Poor maternal nutrition in pregnancy affects fetal development, predisposing offspring to cardiometabolic diseases. The role of mitochondria during fetal development on later-life cardiac dysfunction caused by maternal nutrient reduction (MNR) remains unexplored. We hypothesized that MNR during gestation causes fetal cardiac bioenergetic deficits, compromising cardiac mitochondrial metabolism and reserve capacity. To enable human translation, we developed a primate baboon model (Papio spp.) of moderate MNR in which mothers receive 70% of control nutrition during pregnancy, resulting in intrauterine growth restriction (IUGR) offspring and later exhibiting myocardial remodeling and heart failure at human equivalent ∼25 years. Term control and MNR baboon offspring were necropsied following cesarean-section, and left ventricle (LV) samples were collected. MNR adversely impacted fetal cardiac LV mitochondria in a sex-dependent fashion. Increased maternal plasma aspartate aminotransferase, creatine phosphokinase (CPK), and elevated cortisol levels in MNR concomitant with decreased blood insulin in male fetal MNR were measured. MNR resulted in a two-fold increase in fetal LV mitochondrial DNA (mtDNA). MNR resulted in increased transcripts for several respiratory chain (NDUFB8, UQCRC1, and cytochrome c) and adenosine triphosphate (ATP) synthase proteins. However, MNR fetal LV mitochondrial complex I and complex II/III activities were significantly decreased, possibly contributing to the 73% decreased ATP content and increased lipid peroxidation. MNR fetal LV showed mitochondria with sparse and disarranged cristae dysmorphology. Conclusion: MNR disruption of fetal cardiac mitochondrial fitness likely contributes to the documented developmental programming of adult cardiac dysfunction, indicating a programmed mitochondrial inability to deliver sufficient energy to cardiac tissues as a chronic mechanism for later-life heart failure.


Subject(s)
Fetal Nutrition Disorders/metabolism , Maternal Nutritional Physiological Phenomena , Mitochondria, Heart/metabolism , Adenine Nucleotides/metabolism , Animals , Female , Fetal Nutrition Disorders/pathology , Mitochondria, Heart/ultrastructure , Oxidative Stress , Papio , Pregnancy
2.
Front Endocrinol (Lausanne) ; 11: 591559, 2020.
Article in English | MEDLINE | ID: mdl-33324346

ABSTRACT

Excessive dietary fat intake has extensive impacts on several physiological systems and can lead to metabolic and nonmetabolic disease. In animal models of ingestion, exposure to a high fat diet during pregnancy predisposes offspring to increase intake of dietary fat and causes increase in weight gain that can lead to obesity, and without intervention, these physiological and behavioral consequences can persist for several generations. The hypothalamus is a region of the brain that responds to physiological hunger and fullness and contains orexigenic neuropeptide systems that have long been associated with dietary fat intake. The past fifteen years of research show that prenatal exposure to a high fat diet increases neurogenesis of these neuropeptide systems in offspring brain and are correlated to behavioral changes that induce a pro-consummatory and obesogenic phenotype. Current research has uncovered several potential molecular mechanisms by which excessive dietary fat alters the hypothalamus and involve dietary fatty acids, the immune system, gut microbiota, and transcriptional and epigenetic changes. This review will examine the current knowledge of dietary fat-associated changes in the hypothalamus and the potential pathways involved in modifying the development of orexigenic peptide neurons that lead to changes in ingestive behavior, with a special emphasis on inflammation by chemokines.


Subject(s)
Dietary Fats/adverse effects , Eating , Fetal Nutrition Disorders/pathology , Hypothalamus/pathology , Inflammation Mediators/metabolism , Inflammation/pathology , Prenatal Exposure Delayed Effects/pathology , Animals , Female , Fetal Nutrition Disorders/etiology , Fetal Nutrition Disorders/metabolism , Humans , Hypothalamus/metabolism , Inflammation/etiology , Inflammation/metabolism , Neuropeptides/metabolism , Pregnancy , Prenatal Exposure Delayed Effects/etiology , Prenatal Exposure Delayed Effects/metabolism
3.
Physiol Rep ; 8(3): e14359, 2020 02.
Article in English | MEDLINE | ID: mdl-32026612

ABSTRACT

We hypothesized that late gestation malnutrition differentially affects expandability of adipose tissues to predispose for early postnatal visceral adiposity. Twin-lambs born to dams fed HIGH (150%/110% of required energy/protein, respectively), NORM (100% of requirements) or LOW (50% of NORM) diets during the last trimester were used. Postnatally, lambs were raised on moderate (CONV) or high-carbohydrate-high-fat (HCHF) diets. Adipose tissues were sampled at autopsy at 6 months of age (~puberty) to characterize cellularity, adipocyte cross-sectional area and gene expression patterns. HIGH and LOW compared to NORM lambs had reduced intrinsic (under CONV diet) cellularity in subcutaneous and mesenteric (particularly LOW), and reduced obesity-induced (under HCHF diet) hyperplasia in subcutaneous, mesenteric and perirenal (particularly HIGH) adipose tissues. This corresponded with more pronounced HCHF diet-induced hypertrophy in mesenteric (particularly LOW), perirenal (particularly HIGH) and subcutaneous (particularly HIGH) adipose tissues, and tissue-specific reductions in mRNA expressions for lipid metabolism, angiogenesis and adipose development. Gene expression for inflammation and lipid metabolism markers were increased and decreased, respectively, in HCHF lambs (HCHF lambs became obese) in all tissues. Both prenatal over- and undernutrition predisposed for abdominal adiposity and extreme perirenal hypertrophy due to reduced intrinsic (observed under CONV diet) cellularity and impaired ability of subcutaneous, mesenteric and perirenal adipose tissues to expand by hyperplasia rather than hypertrophy on an obesogenic (HCHF) diet.


Subject(s)
Fetal Nutrition Disorders/metabolism , Intra-Abdominal Fat/metabolism , Lipid Metabolism , Obesity/metabolism , Adiposity , Animals , Diet, High-Fat/adverse effects , Female , Fetal Nutrition Disorders/pathology , Intra-Abdominal Fat/pathology , Male , Obesity/etiology , Obesity/pathology , Sheep
4.
Dev Biol ; 443(1): 78-91, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30189195

ABSTRACT

Maternal undernutrition during pregnancy (MUN) often leads to low birth weight (LBW) neonates that have a reduced total nephron endowment, leaving these neonates susceptible to kidney disease throughout their lives. For reasons unknown, these LBW neonates have impaired kidney development due to a severe reduction in renal SIX2+ stem cells during nephrogenesis. Using a mouse model of MUN, we investigated SIX2+ stem cell reduction in the LBW neonate. Significant upregulation of the protein fetuin-B (measured by PCR and immunoblotting) in the MUN mother's placenta, organs and circulation yielded a 3-fold increase of this protein in the embryonic kidney. Recombinant fetuin-B, administered to healthy pregnant mothers at the concentration equivalent to that in the MUN mother, crossed the placenta and reduced both SIX2+ stem cells by 50% and nephron formation by 66% in embryonic kidneys (measured by immunofluorescence and the physical dissector/fractionator stereological method). Administration of fetuin-B to kidney explants yielded similar reductions in renal SIX2+ stem cells and nephron formation. Fetuin-B treatment of isolated embryonic renal SIX2+ stem cell primary cultures 1) increased NF-kB activity and apoptosis, 2) reduced cell proliferation due to upregulated p21 nuclear activity and subsequent cell cycle arrest, and 3) enhanced generation of reactive oxygen species (measured by fluorescence microscopy). In conclusion, MUN increases fetuin-B in the developing embryonic kidney. The increase in fetuin-B blunts nephrogenesis by reducing SIX2+ stem cells by promoting their apoptosis (via NF-kB upregulation), blunting their proliferative renewal (via p21 upregulation) and enhancing oxidative stress.


Subject(s)
Fetal Nutrition Disorders/metabolism , Fetuin-B/metabolism , Kidney/embryology , Animals , Apoptosis/physiology , Embryonic Stem Cells/metabolism , Female , Fetal Nutrition Disorders/genetics , Homeodomain Proteins/metabolism , Infant, Low Birth Weight/physiology , Kidney/metabolism , Male , Maternal Health , Mice , Nephrons/embryology , Nephrons/metabolism , Oxidative Stress/physiology , Pregnancy , Primary Cell Culture , Transcription Factors/metabolism , Up-Regulation
5.
Mol Metab ; 6(6): 560-573, 2017 06.
Article in English | MEDLINE | ID: mdl-28580286

ABSTRACT

OBJECTIVE: Poor fetal nutrition increases the risk of type 2 diabetes in the offspring at least in part by reduced embryonic ß-cell growth and impaired function. However, it is not entirely clear how fetal nutrients and growth factors impact ß-cells during development to alter glucose homeostasis and metabolism later in life. The current experiments aimed to test the impact of fetal nutrients and growth factors on endocrine development and how these signals acting on mTOR signaling regulate ß-cell mass and glucose homeostasis. METHOD: Pancreatic rudiments in culture were used to study the role of glucose, growth factors, and amino acids on ß-cell development. The number and proliferation of pancreatic and endocrine progenitor were assessed in the presence or absence of rapamycin. The impact of mTOR signaling in vivo on pancreas development and glucose homeostasis was assessed in models deficient for mTOR or Raptor in Pdx1 expressing pancreatic progenitors. RESULTS: We found that amino acid concentrations, and leucine in particular, enhance the number of pancreatic and endocrine progenitors and are essential for growth factor induced proliferation. Rapamycin, an mTORC1 complex inhibitor, reduced the number and proliferation of pancreatic and endocrine progenitors. Mice lacking mTOR in pancreatic progenitors exhibited hyperglycemia in neonates, hypoinsulinemia and pancreatic agenesis/hypoplasia with pancreas rudiments containing ductal structures lacking differentiated acinar and endocrine cells. In addition, loss of mTORC1 by deletion of raptor in pancreatic progenitors reduced pancreas size with reduced number of ß-cells. CONCLUSION: Together, these results suggest that amino acids concentrations and in particular leucine modulates growth responses of pancreatic and endocrine progenitors and that mTOR signaling is critical for these responses. Inactivation of mTOR and raptor in pancreatic progenitors suggested that alterations in some of the components of this pathway during development could be a cause of pancreatic agenesis/hypoplasia and hyperglycemia.


Subject(s)
Amino Acids/deficiency , Cell Differentiation , Embryonic Stem Cells/cytology , Fetal Nutrition Disorders/metabolism , Glucose Metabolism Disorders/metabolism , Insulin-Secreting Cells/cytology , Mechanistic Target of Rapamycin Complex 1/metabolism , Amino Acids/metabolism , Animals , Cell Proliferation , Embryonic Stem Cells/metabolism , Female , Glucose Metabolism Disorders/etiology , Insulin-Secreting Cells/metabolism , Mechanistic Target of Rapamycin Complex 1/genetics , Mice , Mice, Inbred C57BL , Pregnancy , Signal Transduction
6.
J Dev Orig Health Dis ; 8(3): 370-381, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28327211

ABSTRACT

Placental transport of vitamin D and other nutrients (e.g. amino acids, fats and glucose) to the fetus is sensitive to maternal and fetal nutritional cues. We studied the effect of maternal calorific restriction on fetal vitamin D status and the placental expression of genes for nutrient transport [aromatic T-type amino acid transporter-1 (TAT-1); triglyceride hydrolase/lipoprotein uptake facilitator lipoprotein lipase (LPL)] and vitamin D homeostasis [CYP27B1; vitamin D receptor (VDR)], and their association with markers of fetal cardiovascular function and skeletal muscle growth. Pregnant sheep received 100% total metabolizable energy (ME) requirements (control), 40% total ME requirements peri-implantation [PI40, 1-31 days of gestation (dGA)] or 50% total ME requirements in late gestation (L, 104-127 dGA). Fetal, but not maternal, plasma 25-hydroxy-vitamin D (25OHD) concentration was lower in PI40 and L maternal undernutrition groups (P<0.01) compared with the control group at 0.86 gestation. PI40 group placental CYP27B1 messenger RNA (mRNA) levels were increased (P<0.05) compared with the control group. Across all groups, higher fetal plasma 25OHD concentration was associated with higher skeletal muscle myofibre and capillary density (P<0.05). In the placenta, higher VDR mRNA levels were associated with higher TAT-1 (P<0.05) and LPL (P<0.01) mRNA levels. In the PI40 maternal undernutrition group only, reduced fetal plasma 25OHD concentration may be mediated in part by altered placental CYP27B1. The association between placental mRNA levels of VDR and nutrient transport genes suggests a way in which the placenta may integrate nutritional cues in the face of maternal dietary challenges and alter fetal physiology.


Subject(s)
Caloric Restriction/adverse effects , Fetal Nutrition Disorders/metabolism , Malnutrition/metabolism , Maternal-Fetal Exchange/physiology , Prenatal Exposure Delayed Effects/metabolism , Vitamin D/metabolism , Animals , Female , Fetal Nutrition Disorders/etiology , Fetus , Malnutrition/complications , Muscle, Skeletal/metabolism , Pregnancy , Random Allocation , Sheep
7.
Int J Obes (Lond) ; 41(3): 360-371, 2017 03.
Article in English | MEDLINE | ID: mdl-28028317

ABSTRACT

OBJECTIVES: Calorie-restriction during gestation in rats has been seen to produce lasting detrimental effects in the offspring, affecting energy balance control and other related metabolic functions. Our aim was to assess whether leptin supplementation throughout lactation may prevent the dysmetabolic phenotype in adulthood associated with gestational calorie restriction. METHODS: Three groups of male Wistar rats were followed: the offspring of ad libitum fed dams (controls); the offspring of 20% calorie-restricted dams during gestation (CR); and CR rats supplemented with physiological doses of leptin throughout lactation (CR-Leptin). Pups were weaned with a standard diet (SD) until 4 months of age, and then half of the animals of each group were moved to a Western diet (WD) until 6 months of age. Body weight and food intake were recorded. Energy expenditure, locomotive activity, blood parameters, liver triglycerides (TG), and gene expression and specific proteins in liver and white adipose tissue (WAT) were measured in adulthood. RESULTS: Adult CR rats, but not CR-Leptin rats, displayed greater adiposity index and feed efficiency (both under SD) than controls, along with lower locomotive activity and energy expenditure, higher HOMA-IR index and greater circulating TG and leptin levels. CR animals also exhibited increased values of the respiratory exchange ratio and more severe signs of hepatic steatosis under WD than CR-Leptin animals. Gene expression analysis revealed that CR, but not CR-Leptin, animals displayed indicators of lower capacity for WAT expansion, along with decreased lipogenesis and lipolytic capacity under SD, and impaired lipogenic response of the liver to WD feeding, in accordance with diminished insulin sensitivity and WAT leptin signaling. CONCLUSIONS: Oral leptin supplementation in physiological doses throughout lactation in rats prevents most of the detrimental effects on energy homeostasis and metabolic alterations in adulthood caused by inadequate fetal nutrition.


Subject(s)
Animals, Suckling/metabolism , Caloric Restriction , Fetal Nutrition Disorders/metabolism , Lactation/physiology , Leptin/administration & dosage , Leptin/pharmacology , Prenatal Exposure Delayed Effects/metabolism , Prenatal Exposure Delayed Effects/prevention & control , Adipose Tissue, White/metabolism , Adipose Tissue, White/pathology , Administration, Oral , Animals , Disease Models, Animal , Female , Leptin/blood , Male , Obesity/metabolism , Obesity/pathology , Pregnancy , Rats , Rats, Wistar
8.
Nutr Res ; 36(10): 1055-1067, 2016 10.
Article in English | MEDLINE | ID: mdl-27865347

ABSTRACT

Intrauterine growth restriction (IUGR) affects up to 10% of pregnancies and often results in short- and long-term sequelae for offspring. The mechanisms underlying IUGR are poorly understood, but it is known that healthy placentation is essential for nutrient provision to fuel fetal growth, and is regulated by immunologic inputs. We hypothesized that in pregnancy, maternal food restriction (FR) resulting in IUGR would decrease the overall immunotolerant milieu in the placenta, leading to increased cellular stress and death. Our specific objectives were to evaluate (1) key cytokines (eg, IL-10) that regulate maternal-fetal tolerance, (2) cellular processes (autophagy and endoplasmic reticulum [ER] stress) that are immunologically mediated and important for cellular survival and functioning, and (3) the resulting IUGR phenotype and placental histopathology in this animal model. After subjecting pregnant mice to mild and moderate FR from gestational day 10 to 19, we collected placentas and embryos at gestational day 19. We examined RNA sequencing data to identify immunologic pathways affected in IUGR-associated placentas and validated messenger RNA expression changes of genes important in cellular integrity. We also evaluated histopathologic changes in vascular and trophoblastic structures as well as protein expression changes in autophagy, ER stress, and apoptosis in the mouse placentas. Several differentially expressed genes were identified in FR compared with control mice, including a considerable subset that regulates immune tolerance, inflammation, and cellular integrity. In summary, maternal FR decreases the anti-inflammatory effect of IL-10 and suppresses placental autophagic and ER stress responses, despite evidence of dysregulated vascular and trophoblast structures leading to IUGR.


Subject(s)
Autophagy , Endoplasmic Reticulum Stress , Fetal Growth Retardation/etiology , Fetal Nutrition Disorders/etiology , Interleukin-10/metabolism , Placenta , Prenatal Nutritional Physiological Phenomena , Animals , Apoptosis , Blood Vessels , Eating , Energy Intake , Female , Fetal Growth Retardation/metabolism , Fetal Nutrition Disorders/metabolism , Immune Tolerance , Inflammation/etiology , Inflammation/metabolism , Mice, Inbred C57BL , Mothers , Placenta/immunology , Placenta/metabolism , Placenta/pathology , Pregnancy , RNA, Messenger/metabolism , Sequence Analysis, RNA , Trophoblasts
9.
Physiol Behav ; 165: 35-42, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27364433

ABSTRACT

In neonatal rats, hunger and satiety responses occur particularly via dehydration and gastric distention, respectively. The control of food intake in newborns is yet to be fully consolidated, particularly with respect to the participation of the hypothalamic nuclei and their relationship with the serotonergic pathway. Moreover, it is unclear how the environmental stressors in early life, like undernutrition, interfere in these events. Therefore, this study examined the serotonin-system's impact on food intake in rat neonates at postnatal day (P) 10 and P18 and the manner in which protein undernutrition during pregnancy and lactation interferes in this behavior. To accomplish this, Wistar rats were used, nutritionally manipulated by a diet having two protein levels, (8% and 17%) during pregnancy and lactation, to form the Control (n=10) and Low protein groups (n=10). At 10 and 18 postnatal days pups received an acute dose of fenfluramine (3mg/kg) or saline (0.9% NaCl) and subjected to milk consumption testing and then perfused to obtain the brains for the analysis of cell activation of the immunoreactive c-Fos in the hypothalamic and raphe nuclei. At 10days a reduction in weight gain was observed in both groups. On comparison of the neuronal activation for the paraventricular nucleus, an increased activation in response to fenfluramine was observed. At 18days, the weight gain percentage differed between the groups according to the nutritional manipulation, in which the control animals had no significant change while the undernourished presented increased weight gain with the use of fenfluramine. The marking of c-Fos in response to fenfluramine in the hypothalamic and raphe nuclei revealed, an especially lower activation of the PVN, MnR and DR compared intra-group. However when evaluating the effect of undernutrition, marking activation was observed to increase in all the nuclei analyzed, in the hypothalamus and raphe. Data from this study indicate that the action of serotonin via food intake in the neonates may have been delayed by early protein undernutrition.


Subject(s)
Diet, Protein-Restricted/adverse effects , Eating/physiology , Hypothalamus/physiology , Malnutrition/physiopathology , Raphe Nuclei/physiology , Serotonin/metabolism , Animals , Animals, Newborn , Disease Models, Animal , Eating/drug effects , Female , Fenfluramine/pharmacology , Fetal Nutrition Disorders/metabolism , Fetal Nutrition Disorders/physiopathology , Hypothalamus/drug effects , Hypothalamus/growth & development , Lactation , Male , Milk , Neurons/drug effects , Neurons/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Raphe Nuclei/drug effects , Raphe Nuclei/growth & development , Rats, Wistar , Selective Serotonin Reuptake Inhibitors/pharmacology , Weight Gain/drug effects , Weight Gain/physiology
10.
Nutrition ; 32(11-12): 1275-84, 2016.
Article in English | MEDLINE | ID: mdl-27210508

ABSTRACT

OBJECTIVE: The aim of this study was to evaluate the effects of food restriction followed by controlled refeeding on glucose tolerance in pigs exposed to intrauterine malnutrition. METHODS: Pregnant sows (n = 11) were assigned to either a control (C) group or an undernutrition (U) group (75% of C) during gestation. At postnatal 68 d, the offspring (n = 16) were placed on either a cafeteria feeding (CF) group or a food-restricted (FR) group (75% of CF) for 6 wk. After that, all offspring were fed ad libitum until 189 d (dpn189). RESULTS: The results showed that maternal malnutrition induced offspring glucose intolerance, which was demonstrated by increased serum glucose and triacylglycerol content at dpn189, as well as increased area under the blood glucose curve (AUC) during the intravenous glucose tolerance test (i.v.GTT) (P < 0.05). Interestingly, food restriction followed by controlled refeeding further increased serum glucose content at dpn189 and AUC during i.v.GTT in pigs born from U sows (P < 0.05), which was accompanied by catch-up growth during the refeeding period. These changes were associated with increased mRNA levels of hepatic gluconeogenesis (PC, PEPCK) enzymes (P < 0.05), decreased mRNA level of muscle glucose transporter (GLUT4; P = 0.07), and reduced mRNA level of insulin signaling protein (IRS1, P < 0.05) in the liver. CONCLUSIONS: Our results indicate that catch-up growth following food restriction can exacerbate glucose intolerance in offspring exposed to intrauterine malnutrition. This may be caused by increased hepatic gluconeogenesis, decreased muscle glucose transport, and impaired hepatic insulin signaling.


Subject(s)
Fetal Nutrition Disorders/metabolism , Glucose Intolerance/etiology , Prenatal Exposure Delayed Effects/etiology , Animals , Animals, Newborn , Disease Models, Animal , Female , Food Deprivation , Gluconeogenesis/genetics , Glucose Intolerance/genetics , Glucose Intolerance/metabolism , Glucose Transporter Type 4/genetics , Growth , Insulin Receptor Substrate Proteins/genetics , Liver/metabolism , Malnutrition/complications , Malnutrition/metabolism , Muscle, Skeletal/metabolism , Pregnancy , Pregnancy Complications/metabolism , Prenatal Exposure Delayed Effects/genetics , Prenatal Exposure Delayed Effects/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sus scrofa
11.
J Dev Orig Health Dis ; 6(4): 342-9, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26008599

ABSTRACT

The purpose of this study was to examine the hypothesis that excess maternal glucocorticoids in response to maternal undernutrition programs the expression of extracellular matrix (ECM) components potentially by miR-29c. We measured the expression of mRNA (qRT-PCR) and protein (Western blot) for collagen 3A1, collagen 4A5 and matrix metalloproteinase 2 (MMP2) in offspring carotid arteries from three groups of dams: 50% food-restricted in latter half of gestation [maternal undernutrition (MUN)], MUN dams who received metyrapone (MET) (500 mg/ml ) in drinking water from day 10 of gestation to term, and control dams fed an ad libitum diet. The expression of miR-29c was significantly decreased at 3 weeks, 3 months and 9 months in MUN carotid arteries, and these decreases in expression were partially blocked by treatment of dams with MET. The expression pattern of ECM genes that are targets of miR-29c correlated with miR-29c expression. Expression of mRNA was increased for elastin (ELN) and MMP2 mRNA in 3-week MUN carotids; in 9-month carotids there were also significant increases in expression of Col3A1 and Col4A5. These changes in mRNA expression of ECM genes at 3 weeks and 9 months were blocked by MET treatment. Similarly, the expression of ELN and MMP2 proteins at 3 weeks were increased in MUN carotids, and by 9 months there were also increases in expression of Col3A1 and Col4A5, which were blocked by MET in MUN carotids. Overall, the results demonstrate a close correlation between expression of miR-29c and the ECM proteins that are its targets thus supporting our central hypothesis.


Subject(s)
Carotid Arteries/metabolism , Extracellular Matrix Proteins/metabolism , Fetal Nutrition Disorders/metabolism , MicroRNAs/metabolism , Prenatal Nutritional Physiological Phenomena , Animals , Collagen Type III/metabolism , Elastin/metabolism , Extracellular Matrix Proteins/genetics , Female , Glucocorticoids/metabolism , Matrix Metalloproteinase 2/metabolism , Pregnancy , Rats, Sprague-Dawley
12.
Nat Commun ; 5: 5592, 2014 Nov 26.
Article in English | MEDLINE | ID: mdl-25424739

ABSTRACT

Periconceptional diet may persistently influence DNA methylation levels with phenotypic consequences. However, a comprehensive assessment of the characteristics of prenatal malnutrition-associated differentially methylated regions (P-DMRs) is lacking in humans. Here we report on a genome-scale analysis of differential DNA methylation in whole blood after periconceptional exposure to famine during the Dutch Hunger Winter. We show that P-DMRs preferentially occur at regulatory regions, are characterized by intermediate levels of DNA methylation and map to genes enriched for differential expression during early development. Validation and further exploratory analysis of six P-DMRs highlight the critical role of gestational timing. Interestingly, differential methylation of the P-DMRs extends along pathways related to growth and metabolism. P-DMRs located in INSR and CPT1A have enhancer activity in vitro and differential methylation is associated with birth weight and serum LDL cholesterol. Epigenetic modulation of pathways by prenatal malnutrition may promote an adverse metabolic phenotype in later life.


Subject(s)
Antigens, CD/metabolism , DNA Methylation , Fetal Development , Fetal Nutrition Disorders/metabolism , Prenatal Exposure Delayed Effects/metabolism , Receptor, Insulin/metabolism , Starvation , Antigens, CD/genetics , Birth Weight , Carnitine O-Palmitoyltransferase/genetics , Carnitine O-Palmitoyltransferase/metabolism , Epigenesis, Genetic , Female , Fetal Nutrition Disorders/genetics , Humans , Male , Middle Aged , Molecular Sequence Data , Netherlands , Pregnancy , Prenatal Exposure Delayed Effects/genetics , Receptor, Insulin/genetics
13.
Endocrinology ; 155(12): 4808-17, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25264936

ABSTRACT

Maternal undernutrition (UN) is known to cause cardiac hypertrophy, elevated blood pressure, and endothelial dysfunction in adult offspring. Maternal UN may also lead to disturbances in GH regulation in offspring. Because GH plays a key role in cardiac development, we used a model of maternal UN to examine the effects of neonatal GH treatment on cardiac hypertrophy, cardiac micro RNA (miRNA) profiles, and associated gene regulation in adult offspring. Female Sprague-Dawley rats were fed either a standard control diet (CON) or 50% of CON intake throughout pregnancy (UN). From neonatal day 3 until weaning (d 21), CON and UN pups received either saline (S) (CON-S, UN-S) or GH (2.5 µg/g·d) (CON-GH, UN-GH). Heart structure was determined by hematoxylin and eosin staining, and miRNA was isolated from cardiac tissue and miRNA expression analyzed using Cardiovascular miRNA gene Arrays (SABiosciences Ltd). Maternal UN caused marked increases in cardiac hypertrophy and left ventricular cardiomyocyte area, which were reversed by preweaning GH treatment. Systolic blood pressure was increased in UN-S groups and normalized in UN-GH groups (CON-S 121 ± 2 mmHg, CON-GH 115 ± 3 mm Hg, UN-S 146 ± 3 mmHg, and UN-GH 127 ± 2 mmHg). GH treatment during early development facilitated a reversal of pathological changes in offspring hearts caused by UN during pregnancy. Specific cardiac miRNA profiles were exhibited in response to maternal UN, accompanied by up-regulation of the lethal-7 (LET-7) miRNA family in GH-treated offspring. miRNA target analysis revealed a number of genes associated with inflammation and cardiovascular development, which may be involved in the altered cardiac function of these offspring. Up-regulation of the LET-7 family of miRNAs observed in GH groups may mediate the reversal of cardiac hypertrophy observed in adult offspring males of UN mothers.


Subject(s)
Fetal Nutrition Disorders/metabolism , Growth Hormone/therapeutic use , Hypertension/metabolism , Hypertrophy, Left Ventricular/metabolism , MicroRNAs/metabolism , Prenatal Exposure Delayed Effects/metabolism , Animals , Birth Weight/drug effects , Blood Pressure/drug effects , Drug Evaluation, Preclinical , Female , Growth Hormone/pharmacology , Heart/drug effects , Heart/embryology , Hypertension/prevention & control , Hypertrophy, Left Ventricular/prevention & control , Male , Malnutrition , Maternal Nutritional Physiological Phenomena , Myocardium/metabolism , Pregnancy , Rats, Sprague-Dawley
15.
Int J Clin Exp Pathol ; 7(6): 3101-11, 2014.
Article in English | MEDLINE | ID: mdl-25031729

ABSTRACT

Epidemiological studies have demonstrated that intrauterine growth restriction (IUGR) increases the risk for respiratory morbidity from infancy, throughout childhood and into adulthood. Chronic restriction of nutrients causes abnormalities in the airways and lungs of offspring, but whether IUGR adversely impacts fetal pulmonary vascular development and underlying mechanisms remain under investigation. In this study, we investigated the effects of protein malnutrition in utero on pulmonary alveolarization and vascular growth of the fetal lung and placentae. Pregnant rats were feed with an isocaloric low-protein diet (8% protein) until delivery. Placenta and fetal lungs were harvested on 20th day of gestation (term 21 days of gestation). Lung index (lung weight as a percentage of body weight), total DNA and protein, radial alveolar count, arteriolar wall thickness, lung maturity and angiogenic factor VEGF were assessed. The lung was hypoplastic in IUGR fetus, evidenced by reduction in lung weight, DNA and protein content. Protein restriction in utero led to higher glycogen levels, but reduced number of alveoli as confirmed by the measurement of radial alveolar counts. IUGR fetus had significantly reduced VEGF, Flk-1 levels in lung but no changes in Flt-1 mRNA. Furthermore, IUGR was associated with increased lung miR-126-3p levels, which modulated the expression of angiogenic factor. In contrast, with regard to the placenta, IUGR fetus presented with decreased expression of VEGF, with no changes in VEGF receptors and expression-regulating miRNAs. This work suggested that VEGF signaling defect plays an important role in the defective lung development, which may explain the increased incidence of respiratory infections in IUGR patients.


Subject(s)
Fetal Growth Retardation/metabolism , Fetal Growth Retardation/pathology , Pulmonary Alveoli/blood supply , Pulmonary Alveoli/embryology , Pulmonary Alveoli/pathology , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism , Animals , Disease Models, Animal , Female , Fetal Nutrition Disorders/metabolism , Fetal Nutrition Disorders/pathology , Fetus , Immunoblotting , Lung/blood supply , Lung/embryology , Lung/metabolism , Pregnancy , Prenatal Exposure Delayed Effects , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction
17.
Science ; 345(6198): 1255903, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-25011554

ABSTRACT

Adverse prenatal environments can promote metabolic disease in offspring and subsequent generations. Animal models and epidemiological data implicate epigenetic inheritance, but the mechanisms remain unknown. In an intergenerational developmental programming model affecting F2 mouse metabolism, we demonstrate that the in utero nutritional environment of F1 embryos alters the germline DNA methylome of F1 adult males in a locus-specific manner. Differentially methylated regions are hypomethylated and enriched in nucleosome-retaining regions. A substantial fraction is resistant to early embryo methylation reprogramming, which may have an impact on F2 development. Differential methylation is not maintained in F2 tissues, yet locus-specific expression is perturbed. Thus, in utero nutritional exposures during critical windows of germ cell development can impact the male germline methylome, associated with metabolic disease in offspring.


Subject(s)
DNA Methylation , Fetal Nutrition Disorders/metabolism , Prenatal Exposure Delayed Effects , Spermatozoa/metabolism , Animals , Caloric Restriction , Epigenesis, Genetic , Female , Fetal Nutrition Disorders/genetics , Insulin/metabolism , Insulin Secretion , Male , Metabolic Diseases/metabolism , Mice , Mice, Inbred ICR , Nucleosomes/metabolism , Pregnancy , Spermatozoa/physiology
18.
Gerontology ; 60(6): 502-7, 2014.
Article in English | MEDLINE | ID: mdl-24820242

ABSTRACT

BACKGROUND: While malnutrition is an important concern in the developing world, Western countries are experiencing a pandemic of obesity and metabolic diseases. OBJECTIVE: This work reviews the current state of knowledge of the effects of malnutrition during early life on metabolism in older age. METHODS: The impact of early-life determinants on diabetes and related metabolic diseases in later life is elucidated by three different methodological approaches. First, results from animal studies in dietary manipulation models are reviewed. Second, findings from epidemiological studies that often use natural experiments to determine the effects of famines on the health status of the population are discussed. Finally, the relation between maternal or childhood malnutrition and diabetes in adulthood is explored in a big-data study using the entire population of a country across a century. RESULTS: We present overwhelming evidence that the maternal or early childhood nutritional status negatively affects both the short- and long-term health status and development of the offspring, thereby providing starting points to formulate intervention and prevention strategies. In particular, it was found that in the case of early-life exposure to famine, the risk of the offspring to develop type 2 diabetes in older age is up to 125% higher than without famine exposure. CONCLUSION: Due to its inherent complexity, an understanding of the long-term effects of maternal and childhood malnutrition on metabolism in older age necessitates interdisciplinary and big-data approaches. Only then can we hope to prevent chronic diseases at their earliest beginning.


Subject(s)
Child Nutrition Disorders/complications , Fetal Nutrition Disorders/etiology , Infant Nutrition Disorders/complications , Metabolic Diseases/epidemiology , Aged , Child , Child Nutrition Disorders/metabolism , Fetal Nutrition Disorders/metabolism , Humans , Infant , Infant Nutrition Disorders/metabolism
19.
BMC Genomics ; 15: 49, 2014 Jan 21.
Article in English | MEDLINE | ID: mdl-24447410

ABSTRACT

BACKGROUND: Maternal undernutrition leads to an increased risk of metabolic disorders in offspring including obesity and insulin resistance, thought to be due to a programmed thrifty phenotype which is inappropriate for a subsequent richer nutritional environment. In a rat model, both male and female offspring of undernourished mothers are programmed to become obese, however postnatal leptin treatment gives discordant results between males and females. Leptin treatment is able to rescue the adverse programming effects in the female offspring of undernourished mothers, but not in their male offspring. Additionally, in these rats, postnatal leptin treatment of offspring from normally-nourished mothers programmes their male offspring to develop obesity in later life, while there is no comparable effect in their female offspring. RESULTS: We show by microarray analysis of the female liver transcriptome that both maternal undernutrition and postnatal leptin treatment independently induce a similar thrifty transcriptional programme affecting carbohydrate metabolism, amino acid metabolism and oxidative stress genes. Paradoxically, however, the combination of both stimuli restores a more normal transcriptional environment. This demonstrates that "leptin reversal" is a global phenomenon affecting all genes involved in fetal programming by maternal undernourishment and leptin treatment. The thrifty transcriptional programme was associated with pro-inflammatory markers and downregulation of adaptive immune mediators, particularly MHC class I genes, suggesting a deficit in antigen presentation in these offspring. CONCLUSIONS: We propose a revised model of developmental programming reconciling the male and female observations, in which there are two competing programmes which collectively drive liver transcription. The first element is a thrifty metabolic phenotype induced by early life growth restriction independently of leptin levels. The second is a homeostatic set point calibrated in response to postnatal leptin surge, which is able to over-ride the metabolic programme. This "calibration model" for the postnatal leptin surge, if applicable in humans, may have implications for understanding responses to catch-up growth in infants. Additionally, the identification of an antigen presentation deficit associated with metabolic thriftiness may relate to a previously observed correlation between birth season (a proxy for gestational undernutrition) and infectious disease mortality in rural African communities.


Subject(s)
Fetal Nutrition Disorders/genetics , Leptin/pharmacology , Liver/drug effects , Amino Acids/metabolism , Animals , Carbohydrate Metabolism/genetics , Diet , Disease Models, Animal , Female , Fetal Development , Fetal Nutrition Disorders/metabolism , Fetal Nutrition Disorders/pathology , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/metabolism , Inflammation Mediators/metabolism , Liver/metabolism , Male , Obesity/metabolism , Obesity/pathology , Oxidative Stress/genetics , Phenotype , Pregnancy , Rats , Rats, Wistar , Transcriptome/drug effects
20.
Acta Physiol (Oxf) ; 210(1): 215-27, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24224789

ABSTRACT

AIM: Metabolic programming via components of the maternal diet during gestation may play a role in the development of different aspects of the metabolic syndrome. Using a mouse model, we aimed to characterize the role of maternal western-type diet in the development of non-alcoholic fatty liver disease (NAFLD) in the offspring. METHODS: Female mice were fed either a western (W) or low-fat control (L) semisynthetic diet before and during gestation and lactation. At weaning, male offspring were assigned either the W or the L diet, generating four experimental groups: WW, WL, LW and LL offspring. Biochemical, histological and epigenetic indicators were investigated at 29 weeks of age. RESULTS: Male offspring exposed to prenatal and post-weaning western-style diet (WW) showed hepatomegaly combined with accumulation of hepatic cholesterol and triglycerides. This accumulation was associated with up-regulation of de novo lipid synthesis, inflammation and dysregulation of lipid storage. Elevated hepatic transaminases and increased expression of Tnfa, Cd11, Mcp1 and Tgfb underpin the severity of liver injury. Histopathological analysis revealed the presence of advanced steatohepatitis in WW offspring. In addition, alterations in DNA methylation in key metabolic genes (Ppara, Insig, and Fasn) were detected. CONCLUSION: Maternal dietary fat intake during early development programmes susceptibility to liver disease in male offspring, mediated by disturbances in lipid metabolism and inflammatory response. Long-lasting epigenetic changes may underlie this dysregulation.


Subject(s)
Aging/metabolism , Diet, Fat-Restricted , Dietary Fats/metabolism , Fatty Liver/metabolism , Fetal Nutrition Disorders/metabolism , Prenatal Exposure Delayed Effects/metabolism , Animals , Female , Male , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease , Pregnancy
SELECTION OF CITATIONS
SEARCH DETAIL
...