Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Theriogenology ; 86(1): 120-9, 2016 Jul 01.
Article in English | MEDLINE | ID: mdl-27173959

ABSTRACT

Although sheep have been widely adopted as an animal model for examining the timing of nutritional interventions through pregnancy on the short- and long-term outcomes, only modest programming effects have been seen. This is due in part to the mismatch in numbers of twins and singletons between study groups as well as unequal numbers of males and females. Placental growth differs between singleton and twin pregnancies which can result in different body composition in the offspring. One tissue that is especially affected is adipose tissue which in the sheep fetus is primarily located around the kidneys and heart plus the sternal/neck region. Its main role is the rapid generation of heat due to activation of the brown adipose tissue-specific uncoupling protein 1 at birth. The fetal adipose tissue response to suboptimal maternal food intake at defined stages of development differs between the perirenal abdominal and pericardial depots, with the latter being more sensitive. Fetal adipose tissue growth may be mediated in part by changes in leptin status of the mother which are paralleled in the fetus. Then, over the first month of life plasma leptin is higher in females than males despite similar adiposity, when fat is the fastest growing tissue with the sternal/neck depot retaining uncoupling protein 1, whereas other depots do not. Future studies should take into account the respective effects of fetal number and sex to provide more detailed insights into the mechanisms by which adipose and related tissues can be programmed in utero.


Subject(s)
Adiposity/physiology , Reproduction/physiology , Sheep/physiology , Animals , Female , Fetal Development , Male , Pregnancy
2.
Diabetes Obes Metab ; 18(1): 72-81, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26434748

ABSTRACT

AIMS: To investigate, for a given energy expenditure (EE) rise, the differential effects of glucagon infusion and cold exposure on brown adipose tissue (BAT) activation in humans. METHODS: Indirect calorimetry and supraclavicular thermography was performed in 11 healthy male volunteers before and after: cold exposure; glucagon infusion (at 23 °C); and vehicle infusion (at 23 °C). All volunteers underwent (18)F-fluorodeoxyglucose ((18)F-FDG) positron emission tomography (PET)/CT scanning with cold exposure. Subjects with cold-induced BAT activation on (18)F-FDG PET/CT (n = 8) underwent a randomly allocated second (18)F-FDG PET/CT scan (at 23 °C), either with glucagon infusion (n = 4) or vehicle infusion (n = 4). RESULTS: We observed that EE increased by 14% after cold exposure and by 15% after glucagon infusion (50 ng/kg/min; p < 0.05 vs control for both). Cold exposure produced an increase in neck temperature (+0.44 °C; p < 0.001 vs control), but glucagon infusion did not alter neck temperature. In subjects with a cold-induced increase in the metabolic activity of supraclavicular BAT on (18)F-FDG PET/CT, a significant rise in the metabolic activity of BAT after glucagon infusion was not detected. Cold exposure increased sympathetic activation, as measured by circulating norepinephrine levels, but glucagon infusion did not. CONCLUSIONS: Glucagon increases EE by a similar magnitude compared with cold activation, but independently of BAT thermogenesis. This finding is of importance for the development of safe treatments for obesity through upregulation of EE.


Subject(s)
Adipose Tissue, Brown/metabolism , Energy Metabolism/drug effects , Glucagon/pharmacokinetics , Adult , Cold Temperature , Controlled Before-After Studies , Fluorodeoxyglucose F18 , Healthy Volunteers , Humans , Male , Positron-Emission Tomography/methods , Random Allocation , Thermogenesis/drug effects , Tomography, X-Ray Computed , Young Adult
3.
J Clin Endocrinol Metab ; 101(1): 59-68, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26513002

ABSTRACT

CONTEXT: Maternal obesity and gestational diabetes mellitus (GDM) can both contribute to adverse neonatal outcomes. The extent to which this may be mediated by differences in placental metabolism and nutrient transport remains to be determined. OBJECTIVE: Our objective was to examine whether raised maternal body mass index (BMI) and/or GDM contributed to a resetting of the expression of genes within the placenta that are involved in energy sensing, oxidative stress, inflammation, and metabolic pathways. METHODS: Pregnant women from Spain were recruited as part of the "Study of Maternal Nutrition and Genetics on the Foetal Adiposity Programming" survey at the first antenatal visit (12-20 weeks of gestation) and stratified according to prepregnancy BMI and the incidence of GDM. At delivery, placenta and cord blood were sampled and newborn anthropometry measured. RESULTS: Obese women with GDM had higher estimated fetal weight at 34 gestational weeks and a greater risk of preterm deliveries and cesarean section. Birth weight was unaffected by BMI or GDM; however, women who were obese with normal glucose tolerance had increased placental weight and higher plasma glucose and leptin at term. Gene expression for markers of placental energy sensing and oxidative stress, were primarily affected by maternal obesity as mTOR was reduced, whereas SIRT-1 and UCP2 were both upregulated. In placenta from obese women with GDM, gene expression for AMPK was also reduced, whereas the downstream regulator of mTOR, p70S6KB1 was raised. CONCLUSIONS: Placental gene expression is sensitive to both maternal obesity and GDM which both impact on energy sensing and could modulate the effect of either raised maternal BMI or GDM on birth weight.


Subject(s)
Body Weight , Diabetes, Gestational/physiopathology , Placenta/physiopathology , Pregnancy Outcome , Adolescent , Adult , Anthropometry , Birth Weight/genetics , Body Mass Index , Diabetes, Gestational/genetics , Energy Intake/genetics , Female , Gene Expression/genetics , Glucose Intolerance/complications , Glucose Intolerance/genetics , Humans , Infant, Newborn , Inflammation/genetics , Inflammation/pathology , Longitudinal Studies , Metabolic Networks and Pathways/genetics , Middle Aged , Obesity/complications , Obesity/genetics , Oxidative Stress , Placenta/metabolism , Pregnancy , Spain/epidemiology , Young Adult
4.
Acta Physiol (Oxf) ; 210(1): 20-30, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23351024

ABSTRACT

AIM: Hypothermia induced by cold exposure at birth is prevented in sheep by the rapid onset of non-shivering thermogenesis in brown adipose tissue (BAT). Changes in adipose tissue composition in early life are therefore essential for survival but also influence adiposity in later life and were thus examined in detail during early development. METHODS: Changes in adipose composition were investigated by immunohistochemistry and qRT-PCR between the period from the first appearance of adipose in the mid gestation foetus, through birth and up to 1 month of age. RESULTS: We identified four distinct phases of development, each associated with pronounced changes in tissue histology and in distribution of the BAT specific uncoupling protein (UCP)1. At mid gestation, perirenal adipose tissue exhibited a dense proliferative, structure marked by high expression of KI-67 but with no UCP1 or visible lipid droplets. By late gestation large quantities of UCP1 were present, lipid storage was evident and expression of BAT-related genes were abundant (e.g. prolactin and ß3 receptors). Subsequently, within 12 h of birth, the depot was largely depleted of lipid and expression of genes such as UCP1, PGC1α, CIDEA peaked. By 30 days UCP1 was undetectable and the depot contained large lipid droplets; however, genes characteristic of BAT (e.g. PRDM16 and BMP7) and most characteristic of white adipose tissue (e.g. leptin and RIP140) were still abundant. CONCLUSION: Adipose tissue undergoes profound compositional changes in early life, of which an increased understanding could offer potential interventions to retain BAT in later life.


Subject(s)
Adipose Tissue, Brown/enzymology , Adipose Tissue, Brown/growth & development , Adiposity/physiology , Aging/physiology , Lipid Metabolism/physiology , Sheep/embryology , Sheep/growth & development , Animals
5.
Reprod Fertil Dev ; 25(5): 728-36, 2013.
Article in English | MEDLINE | ID: mdl-22951182

ABSTRACT

Reduced maternal food intake between early-to-mid gestation results in tissue-specific adaptations in the offspring following juvenile-onset obesity that are indicative of insulin resistance. The aim of the present study was to establish the extent to which renal ectopic lipid accumulation, as opposed to other markers of renal stress, such as iron deposition and apoptosis, is enhanced in obese offspring born to mothers nutrient restricted (NR) throughout early fetal kidney development. Pregnant sheep were fed either 100% (control) or NR (i.e. fed 50% of their total metabolisable energy requirement from 30-80 days gestation and 100% at all other times). At weaning, offspring were made obese and, at approximately 1 year, kidneys were sampled. Triglyceride content, HIF-1α gene expression and the protein abundance of the outer-membrane transporter voltage-dependent anion-selective channel protein (VDAC)-I on the kidney cortex were increased in obese offspring born to NR mothers compared with those born to controls, which exhibited increased iron accumulation within the tubular epithelial cells and increased gene expression of the death receptor Fas. In conclusion, suboptimal maternal nutrition coincident with early fetal kidney development results in enhanced renal lipid deposition following juvenile obesity and could accelerate the onset of the adverse metabolic, rather than cardiovascular, symptoms accompanying the metabolic syndrome.


Subject(s)
Fetal Development/physiology , Insulin Resistance/physiology , Kidney/embryology , Lipids/analysis , Maternal Nutritional Physiological Phenomena/physiology , Obesity/physiopathology , Animals , Blotting, Western , DNA Primers/genetics , Female , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunohistochemistry , Kidney/chemistry , Pregnancy , Real-Time Polymerase Chain Reaction , Sheep , Statistics, Nonparametric , Triglycerides/analysis , Voltage-Dependent Anion Channel 1/metabolism
6.
Diabetologia ; 55(6): 1597-606, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22402988

ABSTRACT

Adipose tissue function changes with development. In the newborn, brown adipose tissue (BAT) is essential for ensuring effective adaptation to the extrauterine environment, and its growth during gestation is largely dependent on glucose supply from the mother to the fetus. The amount, location and type of adipose tissue deposited can also determine fetal glucose homeostasis. Adipose tissue first appears at around mid-gestation. Total adipose mass then increases through late gestation, when it comprises a mixture of white and brown adipocytes. BAT possesses a unique uncoupling protein, UCP1, which is responsible for the rapid generation of large amounts of heat at birth. Then, during postnatal life some, but not all, depots are replaced by white fat. This process can be utilised to investigate the physiological conversion of brown to white fat, and how it is re-programmed by nutritional changes in pre- and postnatal environments. A reduction in early BAT deposition may perpetuate through the life cycle, thereby suppressing energy expenditure and ultimately promoting obesity. Normal fat development profiles in the offspring are modified by changes in maternal diet at defined stages of pregnancy, ultimately leading to adverse long-term outcomes. For example, excess macrophage accumulation and the onset of insulin resistance occur in an adipose tissue depot-specific manner in offspring born to mothers fed a suboptimal diet from early to mid-gestation. In conclusion, the growth of the different fetal adipose tissue depots varies according to maternal diet and, if challenged in later life, this can contribute to insulin resistance and impaired glucose homeostasis.


Subject(s)
Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Fetal Development/physiology , Adipose Tissue, Brown/embryology , Adipose Tissue, White/embryology , Animals , Female , Fetal Development/genetics , Humans , Insulin Resistance , Models, Biological , Pregnancy
8.
Endocrinology ; 152(7): 2816-26, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21558318

ABSTRACT

Fetal growth restriction followed by accelerated postnatal growth contributes to impaired metabolic function in adulthood. The extent to which these outcomes may be mediated centrally within the hypothalamus, as opposed to in the periphery within the digestive tract, remains unknown. In a sheep model, we achieved intrauterine growth restriction experimentally by maternal nutrient restriction (R) that involved a 40% reduction in food intake through late gestation. R offspring were then either reared singly to accelerate postnatal growth (RA) or as twins and compared with controls also reared singly. From weaning, all offspring were maintained indoors until adulthood. A reduced litter size accelerated postnatal growth for only the first month of lactation. Independently from postnatal weight gain and later fat mass, R animals developed insulin resistance as adults. However, restricted accelerated offspring compared with both the control accelerated and restricted restricted offspring ate less and had higher fasting plasma leptin as adults, an adaptation which was accompanied by changes in energy sensing and cell proliferation within the abomasum. Additionally, although fetal restriction down-regulated gene expression of mammalian target of rapamycin and carnitine palmitoyltransferase 1-dependent pathways in the abomasum, RA offspring compensated for this by exhibiting greater activity of AMP-activated kinase-dependent pathways. This study demonstrates a role for perinatal nutrition in the peripheral control of food intake and in energy sensing in the gastric mucosal and emphasizes the importance of diet in early life in regulating energy metabolism during adulthood.


Subject(s)
Caloric Restriction/adverse effects , Energy Metabolism , Fetal Growth Retardation/etiology , Gastric Mucosa/metabolism , Lactation , Maternal Nutritional Physiological Phenomena , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Abomasum/growth & development , Abomasum/metabolism , Abomasum/pathology , Adiposity , Animals , Carnitine O-Palmitoyltransferase/genetics , Carnitine O-Palmitoyltransferase/metabolism , Cell Proliferation , Female , Fetal Growth Retardation/metabolism , Fetal Growth Retardation/pathology , Fetal Growth Retardation/physiopathology , Gastric Mucosa/growth & development , Gastric Mucosa/pathology , Gene Expression Regulation , Insulin Resistance , Leptin/blood , Leptin/genetics , Leptin/metabolism , Male , Pregnancy , RNA, Messenger/metabolism , Random Allocation , Sheep , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Weight Gain
9.
Proc Nutr Soc ; 70(2): 268-75, 2011 May.
Article in English | MEDLINE | ID: mdl-21232171

ABSTRACT

Epidemiological and animal studies have demonstrated that early-life nutrition alters the metabolic responses and generates structural changes in complex tissues, such as the kidneys, which may lead to a reduction in the offspring lifespan. Independently, obesity induces a spontaneous low-grade chronic inflammatory response by modulating several of the major metabolic pathways that ultimately compromise long-term renal health. However, the combined effects of maternal nutrition and early-life obesity in the development of renal diseases are far from conclusive. Previous results, using the ovine model, demonstrated that the combination of a reduction in fetal nutrition and juvenile obesity induced a series of adaptations associated with severe metabolic syndrome in the heart and adipose tissue. Surprisingly, exposure to an obesogenic environment in the kidney of those offspring produced an apparent reduction in glomerulosclerosis in relation to age- and weight-matched controls. However, this reduction in cellular apoptosis was accompanied by a rise in glomerular filtration rate and blood pressure of equal intensity when compared with obese controls. The intention of this review is to explain the adaptive responses observed in this model, based on insights into the mechanism of renal fetal programming, and their potential interactions with some of the metabolic changes produced by obesity.


Subject(s)
Fetal Development , Kidney Diseases/etiology , Kidney/physiopathology , Maternal Nutritional Physiological Phenomena , Metabolic Syndrome/complications , Obesity/complications , Prenatal Exposure Delayed Effects , Adipose Tissue/metabolism , Animals , Female , Humans , Inflammation , Kidney/embryology , Kidney/metabolism , Myocardium/metabolism , Obesity/metabolism , Pregnancy
10.
Reproduction ; 141(1): 119-26, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21045167

ABSTRACT

Maternal nutrition during the period of early organ development can modulate the offspring's ability to metabolise excess fat as young adults when exposed to an obesogenic environment. This study examined the hypothesis that exposing offspring to nutrient restriction coincident with early hepatogenesis would result in endocrine and metabolic adaptations that subsequently lead to increased ectopic lipid accumulation within the liver. Pregnant sheep were fed either 50 or 100% of total metabolisable energy requirements from 30 to 80 days gestation and 100% thereafter. At weaning, offspring were made obese, and at ~1 year of age livers were sampled. Lipid infiltration and molecular indices of gluconeogenesis, lipid metabolism and mitochondrial function were measured. Although hepatic triglyceride accumulation was not affected by obesity per se, it was nearly doubled in obese offspring born to nutrient-restricted mothers. This adaptation was accompanied by elevated gene expression for peroxisome proliferator-activated receptor γ (PPARG) and its co-activator PGC1α, which may be indicative of changes in the rate of hepatic fatty acid oxidation. In contrast, maternal diet had no influence on the stimulatory effect of obesity on gene expression for a range of proteins involved in glucose metabolism and energy balance including glucokinase, glucocorticoid receptors and uncoupling protein 2. Similarly, although gene expressions for the insulin and IGF1 receptors were suppressed by obesity they were not influenced by the prenatal nutritional environment. In conclusion, excess hepatic lipid accumulation with juvenile obesity is promoted by suboptimal nutrition coincident with early development of the fetal liver.


Subject(s)
Animal Nutritional Physiological Phenomena , Fatty Liver/metabolism , Liver/metabolism , Malnutrition/metabolism , Maternal Nutritional Physiological Phenomena , Obesity/metabolism , Prenatal Exposure Delayed Effects , Age Factors , Animals , Disease Models, Animal , Fatty Liver/embryology , Fatty Liver/genetics , Fatty Liver/pathology , Fatty Liver/physiopathology , Female , Gene Expression Regulation, Developmental , Gestational Age , Gluconeogenesis/genetics , Lipid Metabolism/genetics , Liver/embryology , Liver/pathology , Liver/physiopathology , Malnutrition/embryology , Malnutrition/genetics , Malnutrition/physiopathology , Mitochondria, Liver/metabolism , Obesity/embryology , Obesity/genetics , Obesity/pathology , Obesity/physiopathology , PPAR gamma/genetics , Pregnancy , Sheep , Triglycerides/metabolism
11.
Animal ; 4(7): 1075-83, 2010 Jul.
Article in English | MEDLINE | ID: mdl-22444610

ABSTRACT

The maternal nutritional and metabolic environment is critical in determining not only the reproductive success but also the long-term health and viability of the offspring. Changes in maternal diet at defined stages of gestation coincident with different stages of development can have pronounced effects on organ and tissue function in later life. This includes adipose tissue for which differential effects are observed between brown and white adipose tissues. One early, critical window of organ development in the ruminant relates to the period covering uterine attachment, or implantation, and rapid placental growth. During this period, there is pronounced cell division within developing organelles in many fetal tissues, leading to their structural development. In sheep, a 50% global reduction in caloric intake over this specific period profoundly affects placental growth and morphology, resulting in reduced placentome weight. This occurs in conjunction with a lower capacity to inactivate maternal cortisol through the enzyme 11ß-hydroxysteroid dehydrogenase type 2 in response to a decrease in maternal plasma cortisol in early gestation. The birth weight of the offspring is, however, unaffected by this dietary manipulation and, although they possess more fat, this adaptation does not persist into adulthood when they become equally obese as those born to control fed mothers. Subsequently, after birth, further changes in fat development occur which impact on both glucocorticoid action and inflammatory responses. These adaptations can include changes in the relative populations of both brown and white adipocytes for which prolactin acting through its receptor appears to have a prominent role. Earlier when in utero nutrient restricted (i.e. between early-to-mid gestation) offspring are exposed to an obesogenic postnatal environment; they exhibit an exaggerated insulin response, which is accompanied by a range of amplified and thus, adverse, physiological or metabolic responses to obesity. These types of adaptations are in marked contrast to the effect of late gestational nutrient restriction, which results in reduced fat mass at birth. As young adults, however, fat mass is increased and, although basal insulin is unaffected, these offspring are insulin resistant. In conclusion, changes in nutrient supply to either the mother and/or her fetus can have profound effects on a range of metabolically important tissues. These have the potential to either exacerbate, or protect from, the adverse effects of later obesity and accompanying complications in the resulting offspring.

12.
Reproduction ; 139(1): 265-74, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19786398

ABSTRACT

The recent discovery of an association between body composition, energy intake and the fat mass and obesity-associated (FTO) gene represents a promising new therapeutic target in obesity prevention. In a well, pre-established large animal model, we investigated the regulation of FTO gene expression under conditions either leading to obesity or increased risk of obesity related disorders: i) a sedentary 'Western' lifestyle and ii) prenatal exposure to nutrient restriction. Pregnant sheep were either fed to fully meet their nutritional requirements throughout gestation or 50% of this amount from early-to-mid gestation. Following weaning, offspring were either made obese through exposure to a sedentary obesogenic environment or remained lean. A significant positive relationship between placental FTO gene expression and fetal weight was found at 110 days gestation. In both the newborn and adult offspring, the hypothalamus was the major site of FTO gene expression. Hypothalamic FTO gene expression was upregulated by obesity and was further increased by prenatal nutrient restriction. Importantly, we found a strong negative relationship between the hypothalamic FTO gene expression and food intake in lean animals only that may imply FTO as a novel controller of energy intake. In contrast, FTO gene expression in the heart was downregulated in obese offspring born to nutrient restricted mothers. In addition, FTO gene expression was unaffected by obesity or prenatal diet in insulin-dependent tissues, where it changed with age possibly reflecting adaptations in cellular energetic activity. These findings extend information gained from human epidemiology and provide new insights into the regulation of in vivo energy metabolism to prevent obesity.


Subject(s)
Gene Expression Regulation, Developmental , Maternal Nutritional Physiological Phenomena , Overweight/metabolism , Prenatal Exposure Delayed Effects/metabolism , Proteins/genetics , Aging/metabolism , Animals , DNA, Complementary/chemistry , Female , Fetal Weight , Hypothalamus/metabolism , Male , Obesity/prevention & control , Organ Size , Organ Specificity , Placenta/metabolism , Pregnancy , Proteins/chemistry , Proteins/metabolism , RNA, Messenger/metabolism , Sequence Alignment , Sheep, Domestic , Thinness/metabolism
13.
J Endocrinol ; 204(2): 173-9, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19934248

ABSTRACT

Maternal parity influences size at birth, postnatal growth and body composition with firstborn infants being more likely to be smaller with increased fat mass, suggesting that adiposity is set in early life. The precise effect of parity on fat mass and its endocrine sensitivity remains unclear and was, therefore, investigated in the present study. We utilised an established sheep model in which perirenal-abdominal fat mass (the major fat depot in the neonatal sheep) increases approximately 10-fold over the first month of life and focussed on the impact of parity on glucocorticoid sensitivity and adipokine expression in the adipocyte. Twin-bearing sheep of similar body weight and adiposity that consumed identical diets were utilised, and maternal blood samples were taken at 130 days of gestation. One offspring from each twin pair was sampled at 1 day of age, coincident with the time of maximal recruitment of uncoupling protein 1 (UCP1), whilst its sibling was sampled at 1 month, when UCP1 had disappeared. Plasma leptin was lower in nulliparous mothers than in multiparous mothers, and offspring of nulliparous mothers possessed more adipose tissue with increased mRNA abundance of leptin, glucocorticoid receptor and UCP2, adaptations that persisted up to 1 month of age when gene expression for interleukin-6 and adiponectin was also raised. The increase in fat mass associated with firstborn status is therefore accompanied by a resetting of the leptin and glucocorticoid axis within the adipocyte. Our findings emphasise the importance of parity in determining adipose tissue development and that firstborn offspring have an increased capacity for adipogenesis which may be critical in determining later adiposity.


Subject(s)
Adipose Tissue/metabolism , Hormones/blood , Parity , 11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism , 11-beta-Hydroxysteroid Dehydrogenase Type 2/metabolism , Adipokines/genetics , Adipokines/metabolism , Animals , Animals, Newborn , Body Weight , Female , Insulin-Like Growth Factor Binding Protein 3/metabolism , Ion Channels/metabolism , Mitochondrial Proteins/metabolism , PPAR alpha/metabolism , PPAR gamma/metabolism , Pregnancy , RNA, Messenger/metabolism , Receptor, Insulin/metabolism , Receptors, Glucocorticoid/metabolism , Sheep , Uncoupling Protein 1 , Uncoupling Protein 2
14.
Proc Nutr Soc ; 68(3): 321-6, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19490741

ABSTRACT

It is now apparent that one key factor determining the current obesity epidemic within the developed world is the extent to which adipose tissue growth and function can be reset in early life. Adipose tissue can be either brown or white, with brown fat being characterised as possessing a unique uncoupling protein (uncoupling protein 1) that enables the rapid generation of heat by non-shivering thermogenesis. In large mammals this function is recruited at approximately the time of birth, after which brown fat is lost, not normally reappearing again throughout the life cycle. The origin and developmental regulation of brown fat in large mammals is therefore very different from that of small mammals in which brown fat is retained throughout the life cycle and may have the same origin as muscle cells. In contrast, white adipose tissue increases in mass after birth, paralleled by a rise in glucocorticoid action and macrophage accumulation. This process can be reset by changes in the maternal nutritional environment, with the magnitude of response being further determined by the timing at which such a challenge is imposed. Importantly, the long-term response within white adipocytes can occur in the absence of any change in total fat mass. The present review therefore emphasises the need to further understand the developmental regulation of the function of fat through the life cycle in order to optimise appropriate and sustainable intervention strategies necessary not only to prevent obesity in the first place but also to reverse excess fat mass in obese individuals.


Subject(s)
Adipose Tissue/physiopathology , Obesity/physiopathology , Adipocytes/metabolism , Adipose Tissue/growth & development , Adipose Tissue/metabolism , Animals , Female , Humans , Infant, Newborn , Maternal Nutritional Physiological Phenomena , Obesity/metabolism , Pregnancy , Prenatal Exposure Delayed Effects
15.
Reproduction ; 138(3): 601-8, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19525364

ABSTRACT

Nutrient restriction (NR) during critical windows of pregnancy has differential effects on placento-fetal growth and development. Our study, therefore, investigated developmental and metabolic adaptations within the ovine placenta following NR at different critical windows during the first 110 days of gestation (term=147 days). Thus, the effects of NR on cell proliferation, glucocorticoid sensitivity, IGF1 and 2 receptor, peroxisome proliferator-activated receptor gamma (PPARG), and uncoupling protein (UCP)2 gene expression in the placenta were examined. Singleton bearing sheep (n=4-8 per group) were fed either 100% of their total metabolizable energy requirements throughout the study or 50% of this amount between 0-30, 31-65, 66-110, and 0-110 days gestation. A significant reduction in cell proliferation and increased gene expression for the glucocorticoid and IGF2 receptors, PPARG, and UCP2 were detected in placentae sampled from mothers who were nutrient restricted between days 66 and 110 of gestation, only, relative to controls. This window of gestation coincides with the maximum placental growth and the start of exponential growth of the fetus when there are substantially increased metabolic demands on the placenta compared with earlier in gestation. Consequently, increased glucocorticoid sensitivity and suppressed IGF2 action could contribute to a switch in the placenta from proliferation to differentiation, thereby improving its nutrient transfer capacity. Upregulation of PPARG and UCP2 would promote placental fatty acid metabolism thereby limiting glucose utilization. These compensatory placental responses may serve to maintain fetal growth but could result in adverse adaptations such as the early onset of the metabolic syndrome in later life.


Subject(s)
Caloric Restriction/veterinary , Cell Proliferation , Glucocorticoids/pharmacology , Placenta/drug effects , Pregnancy, Animal , Sheep , Animal Feed , Animals , Caloric Restriction/adverse effects , Cell Proliferation/drug effects , Drug Resistance/drug effects , Female , Food , Ion Channels/genetics , Ion Channels/metabolism , Lipid Metabolism/genetics , Maternal Nutritional Physiological Phenomena , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , PPAR gamma/genetics , PPAR gamma/metabolism , Placenta/metabolism , Pregnancy , Pregnancy, Animal/genetics , Pregnancy, Animal/physiology , Sheep/embryology , Sheep/genetics , Sheep/metabolism , Sheep/physiology , Time Factors , Uncoupling Protein 2
16.
Am J Physiol Regul Integr Comp Physiol ; 296(5): R1455-63, 2009 May.
Article in English | MEDLINE | ID: mdl-19244582

ABSTRACT

Maternal nutrient restriction (NR) from early to midgestation has marked effects on endocrine sensitivity and organ function of the resulting offspring. We hypothesized that early NR may reset the expression profile of genes central to myocardial energy metabolism, influencing ectopic lipid deposition and cardiac function in the obese adult offspring. NR offspring were exposed to an "obesogenic" environment, and their cardiac function and molecular indexes of myocardial energy metabolism were assessed to explore the hypothesis that an obese individual's risk of heart disease may be modified after maternal NR. Pregnant sheep were fed 100% (control) or 50% (NR) energy requirement from days 30 to 80 of gestation and 100% energy requirement thereafter. At weaning, offspring were exposed to an obesogenic environment or remained lean. At approximately 1 yr of age, the hemodynamic response of these offspring to hypotension, together with left ventricular expression profiles of fatty acid-binding protein 3 (FABP3), peroxisome proliferator-activated receptor-gamma (PPARgamma) and its coactivator (PGC)-1alpha, acetyl-CoA carboxylase (ACC), AMP-activated protein kinase (AMPK)-alpha(2), and voltage-dependent anion channel 1 (VDAC1), was determined. Obesity produced left ventricular hypertrophy in all animals, with increased ectopic (myocardial) lipid in NR offspring. Obesity per se significantly reduced myocardial transcript expression of PGC-1alpha, AMPKalpha(2), VDAC1, and ACC and increased expression of PPARgamma and FABP3. However, although NR animals were similarly obese, their transcript expression of ACC, PPARgamma, and FABP3 was similar to that of lean animals, indicating altered cardiac energy metabolism. Indeed, blunted tachycardia and an amplified inotropic response to hypotension characterized cardiac function in obese NR offspring. The results suggest that maternal NR during early organogenesis can precipitate an altered myocardial response to hypotension and increased myocardial lipid deposition in the adult offspring after adolescent-onset obesity, potentially rendering these individuals more at risk of early heart failure as they age.


Subject(s)
Aging/physiology , Energy Metabolism/physiology , Heart/physiology , Maternal Nutritional Physiological Phenomena/physiology , Obesity/physiopathology , Pregnancy, Animal/physiology , AMP-Activated Protein Kinases/metabolism , Animals , Atropine/pharmacology , Body Composition/physiology , Catecholamines/metabolism , Disease Models, Animal , Fatty Acid-Binding Proteins/metabolism , Female , Heart Ventricles/metabolism , Lipid Metabolism/drug effects , Male , Muscarinic Antagonists/pharmacology , Nitroprusside/pharmacology , Obesity/metabolism , PPAR gamma/metabolism , Pregnancy , Receptors, Adrenergic/metabolism , Sheep
17.
Endocrinology ; 150(2): 634-41, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18818297

ABSTRACT

The impact of maternal nutrient restriction during early-to-midgestation, a period coinciding with early fetal brain development, on appetite regulation and energy balance in the offspring after juvenile obesity was examined. Pregnant sheep were either fed to meet fully their nutritional requirements throughout gestation or 50% of this amount between 30 and 80 d gestation. After weaning, offspring were either made obese through exposure to a sedentary obesogenic environment or remained lean. Maternal nutrient restriction had no effect on birth weight or subsequent growth. At 1 wk of age, only, gene expression for neuropeptide Y in the hypothalamus was reduced in nutrient-restricted offspring. By 1 yr of age, all O animals had increased plasma leptin, nonesterified fatty acids, and insulin, with the latter effect amplified in NR offspring. Fasting plasma glucose, triglycerides, and cortisol were unaffected by obesity. The entrained reduction in physical activity that led to obesity persisted when all animals were maintained within individual pens. However, NRO offspring exhibited reduced daily food intake and were, therefore, no longer in positive "energy balance." This adaptation was accompanied by elevated hypothalamic gene expression for the melanocortin-4 and insulin receptors, AMP-activated kinase, and acetyl coenzyme A carboxylase alpha. In conclusion, nutrient restriction specifically targeted over the period of early fetal brain development contributes to a profoundly different adaptation in energy balance after juvenile obesity. The extent to which this adaptive response may benefit the offspring or result in an exacerbated risk of type 2 diabetes remains to be established.


Subject(s)
Appetite Regulation/physiology , Caloric Restriction , Fetal Nutrition Disorders/physiopathology , Maternal Nutritional Physiological Phenomena , Obesity/physiopathology , Prenatal Exposure Delayed Effects/physiopathology , Adaptation, Biological/genetics , Age Factors , Animals , Caloric Restriction/veterinary , Crown-Rump Length , Energy Metabolism/physiology , Female , Gene Expression/physiology , Gestational Age , Homeostasis/physiology , Hypothalamus/metabolism , Hypothalamus/physiopathology , Insulin Resistance/genetics , Insulin Resistance/physiology , Maternal Nutritional Physiological Phenomena/physiology , Obesity/etiology , Pregnancy , Sheep , Time Factors
18.
Reproduction ; 135(5): 723-32, 2008 May.
Article in English | MEDLINE | ID: mdl-18304988

ABSTRACT

Maternal cold exposure of pregnant sheep promotes fetal growth, whereas nutrient restriction (NR) can reverse this effect. The present study was designed to establish whether cold exposure induced by winter shearing of the mother at 70 days gestation (term=147 days), with or without NR (induced by a 50% reduction in maternal food intake from 110 days gestation), has specific effects on mRNA abundance of hepatic genes related to growth and liver energy metabolism that could regulate postnatal body and liver growth. Measurements of hepatic gene expression for the GH secretagog receptor-1a (GHSR-1A), peroxisome proliferator-activated receptor (PPAR)alpha, phosphoenolpyruvate carboxykinase (PEPCK), and glucose-6-phosphatase activity together with glycogen content were made in the livers of offspring at 1 and 30 days of age. Maternal NR reduced liver mass at day 1, whereas offspring of cold-exposed mothers had larger livers at day 30 irrespective of maternal diet. Cold exposure resulted in the up-regulation of GHSR-1A mRNA abundance and reduced glucose-6-phosphatase activity at 1, but not 30 days of age, whereas IGF-II mRNA was decreased at 1 and 30 days. PPARalpha mRNA abundance was enhanced, while PEPCK was reduced in 30-day old offspring of cold-exposed mothers. NR caused reductions in IGF-I mRNA and, at 1-day postnatal age, down-regulated GHR, while, at 30 days, reduced GHSR-1A gene expression and hepatic glycogen content. In conclusion, we have shown that maternal cold exposure and NR have different effects on the hepatic GH-IGF and metabolic axis that may contribute to changes in liver growth over the first month of life.


Subject(s)
Cold Temperature , Food Deprivation , Liver/metabolism , Maternal Exposure , Sheep, Domestic/metabolism , Somatomedins/metabolism , Animals , Animals, Newborn/growth & development , Female , Glucose-6-Phosphatase/metabolism , Glycogen/metabolism , Growth Hormone/metabolism , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Insulin-Like Growth Factor II/metabolism , PPAR alpha/metabolism , Pregnancy , RNA, Messenger/analysis
19.
Organogenesis ; 4(3): 188-94, 2008 Jul.
Article in English | MEDLINE | ID: mdl-19279732

ABSTRACT

Maternal dexamethasone administration promotes fetal maturation such that thermoregulation is improved following premature delivery and is thus comparable with a full term birth. In the present study we determined the impact of dexamethasone on both the mothers' metabolic status together with adipose tissue function in the newborn. Glucocorticoid action, adipokine gene expression and mitochondrial protein abundance were measured in perirenal adipose tissue of neonatal sheep that were born into either a warm (30 degrees C) or cool (15 degrees C) ambient temperature at 140 days of gestation (dGA; term approximately 147 dGA), either two days after maternal dexamethasone administration, or at 146 dGA for controls. Dexamethasone administration resulted in a reduction in maternal food intake in conjunction with raised plasma cortisol and free triiodothyronine. In offspring of dexamethasone administered mothers, plasma cortisol was lower and non-esterified fatty acids (NEFA) higher than controls. Glucocorticoid receptor (GR), 11beta-hydroxysteroid dehydrogenase (11beta-HSD1), interleukin-6 and uncoupling protein (UCP)1 and 2 mRNA together with voltage dependent anion channel, cytochrome c protein and UCP1 abundance were all increased by dexamethasone administration and being born into a cool ambient temperature. Gene expression of tumor necrosis factor alpha, adiponectin and peroxisome proliferator-activated receptor transcription factor gamma were unaffected by dexamethasone. The abundance of mRNA for the GR, 11beta-HSD1, UCP1 and 2 mRNA together with each protein were positively correlated to plasma NEFA and negatively correlated to plasma cortisol. In conclusion, despite reduced maternal food intake dexamethasone promotes maturation of glucocorticoid action and mitochondrial protein abundance in the newborn, an adaptation dependent on delivery temperature.

20.
Reproduction ; 134(6): 823-30, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18042639

ABSTRACT

This study investigated the developmental and nutritional programming of two important mitochondrial proteins, namely voltage-dependent anion channel (VDAC) and cytochrome c, in the sheep kidney, liver and lung. The effect of maternal nutrient restriction between early and mid-gestation (i.e. 28- to 80-day gestation, the period of maximal placental growth) on the abundance of these proteins was also examined in fetal and juvenile offspring. Fetuses were sampled at 80 and 140 days of gestation (term approximately 147 days), and postnatal animals at 1 and 30 days and 6 months of age. The abundance of VDAC peaked at 140 days of gestation in the lung, compared with 1 day after birth in the kidney and liver, whereas cytochrome c abundance was greatest at 140 days of gestation in the liver, 1 day after birth in the kidney and 6 months of age in lungs. This differential ontogeny in mitochondrial protein abundance between tissues was accompanied with very different tissue-specific responses to changes in maternal food intake. In the liver, maternal nutrient restriction only increased mitochondrial protein abundance at 80 days of gestation, compared with no effect in the kidney. In contrast, in the lung mitochondrial protein, abundance was raised near to term, whereas VDAC abundance was decreased by 6 months of age. These findings demonstrate the tissue-specific nature of mitochondrial protein development that reflects differences in functional adaptation after birth. The divergence in mitochondrial response between tissues to maternal nutrient restriction early in pregnancy further reflects these differential ontogenies.


Subject(s)
Fetal Development/physiology , Food Deprivation , Mitochondrial Proteins/metabolism , Prenatal Exposure Delayed Effects , Sheep/embryology , Aging/physiology , Animals , Animals, Newborn , Biomarkers/analysis , Cytochromes c/analysis , Cytochromes c/metabolism , Female , Fetus/chemistry , Fetus/metabolism , Gestational Age , Kidney/chemistry , Kidney/embryology , Kidney/metabolism , Liver/chemistry , Liver/embryology , Liver/metabolism , Lung/chemistry , Lung/embryology , Lung/metabolism , Mitochondrial Proteins/analysis , Pregnancy , Prenatal Nutritional Physiological Phenomena , Reverse Transcriptase Polymerase Chain Reaction , Sheep/metabolism , Voltage-Dependent Anion Channels/analysis , Voltage-Dependent Anion Channels/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...