Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
Add more filters










Publication year range
1.
J Endocrinol ; 260(3)2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38109257

ABSTRACT

Adverse environmental conditions before birth are known to programme adult metabolic and endocrine phenotypes in several species. However, whether increments in fetal cortisol concentrations of the magnitude commonly seen in these conditions can cause developmental programming remains unknown. Thus, this study investigated the outcome of physiological increases in fetal cortisol concentrations on glucose-insulin dynamics and pituitary-adrenal function in adult sheep. Compared with saline treatment, intravenous fetal cortisol infusion for 5 days in late gestation did not affect birthweight but increased lamb body weight at 1-2 weeks after birth. Adult glucose dynamics, insulin sensitivity and insulin secretion were unaffected by prenatal cortisol overexposure, assessed by glucose tolerance tests, hyperinsulinaemic-euglycaemic clamps and acute insulin administration. In contrast, prenatal cortisol infusion induced adrenal hypo-responsiveness in adulthood with significantly reduced cortisol responses to insulin-induced hypoglycaemia and exogenous adrenocorticotropic hormone (ACTH) administration relative to saline treatment. The area of adrenal cortex expressed as a percentage of the total cross-sectional area of the adult adrenal gland was also lower after prenatal cortisol than saline infusion. In adulthood, basal circulating ACTH but not cortisol concentrations were significantly higher in the cortisol than saline-treated group. The results show that cortisol overexposure before birth programmes pituitary-adrenal development with consequences for adult stress responses. Physiological variations in cortisol concentrations before birth may, therefore, have an important role in determining adult phenotypical diversity and adaptability to environmental challenges.


Subject(s)
Adrenocorticotropic Hormone , Hydrocortisone , Female , Pregnancy , Animals , Sheep , Hydrocortisone/metabolism , Adrenocorticotropic Hormone/metabolism , Fetus/metabolism , Adrenal Glands/metabolism , Glucose/metabolism , Insulin/metabolism , Gestational Age
2.
Biol Reprod ; 106(4): 814-822, 2022 04 26.
Article in English | MEDLINE | ID: mdl-35040958

ABSTRACT

Mature granulated trophoblast binucleate cells (BNC) have been found in all ruminant placentas examined histologically so far. BNC are normally fairly evenly distributed throughout the fetal villus and all their granules contain a similar variety of hormones and pregnancy associated glycoproteins (PAGs). Only the Giraffe is reported to show a different BNC protein expression, this paper is designed to investigate that. Gold labelled Lectin histochemistry and protein immunocytochemistry were used on deplasticised 1 µm sections of a wide variety of ruminant placentomes with a wide range of antibodies and lectins. In the Giraffe placentomes, even though the lectin histochemistry shows an even distribution of BNC throughout the trophoblast of the placental villi, the protein expression in the BNC granules is limited to the BNC either in the apex or the base of the villi. Placental lactogens and Prolactin (PRL) are present only in basally situated BNC: PAGs only in the apical BNC. PRL is only found in the Giraffe BNC which react with many fewer of the wide range of antibodies used here to investigate the uniformity of protein expression in ruminant BNC. The possible relevance of these differences to ruminant function and evolution is considered to provide a further example of the versatility of the BNC system.


Subject(s)
Giraffes , Placenta , Animals , Female , Lectins/metabolism , Placenta/metabolism , Pregnancy , Prolactin/metabolism , Ruminants/metabolism , Trophoblasts/metabolism
3.
J Endocrinol ; 251(1): 53-68, 2021 08 25.
Article in English | MEDLINE | ID: mdl-34321363

ABSTRACT

In adults, glucocorticoids act to match the supply and demand for energy during physiological challenges, partly through actions on tissue mitochondrial oxidative phosphorylation (OXPHOS) capacity. However, little is known about the role of the natural prepartum rise in fetal glucocorticoid concentrations in preparing tissues for the increased postnatal energy demands. This study examined the effect of manipulating cortisol concentrations in fetal sheep during late gestation on mitochondrial OXPHOS capacity of two skeletal muscles with different postnatal locomotive functions. Mitochondrial content, biogenesis markers, respiratory rates and expression of proteins and genes involved in the electron transfer system (ETS) and OXPHOS efficiency were measured in the biceps femoris (BF) and superficial digital flexor (SDF) of fetuses either infused with cortisol before the prepartum rise or adrenalectomised to prevent this increment. Cortisol infusion increased mitochondrial content, biogenesis markers, substrate-specific respiration rates and abundance of ETS complex I and adenine nucleotide translocator (ANT1) in a muscle-specific manner that was more pronounced in the SDF than BF. Adrenalectomy reduced mitochondrial content and expression of PGC1α and ANT1 in both muscles, and ETS complex IV abundance in the SDF near term. Uncoupling protein gene expression was unaffected by cortisol manipulations in both muscles. Gene expression of the myosin heavy chain isoform, MHCIIx, was increased by cortisol infusion and reduced by adrenalectomy in the BF alone. These findings show that cortisol has a muscle-specific role in prepartum maturation of mitochondrial OXPHOS capacity with important implications for the health of neonates born pre-term or after intrauterine glucocorticoid overexposure.


Subject(s)
Fetus/metabolism , Hydrocortisone/physiology , Mitochondria, Muscle/metabolism , Muscle, Skeletal/metabolism , Oxidative Phosphorylation , Animals , Animals, Newborn , Cell Respiration , Female , Myosin Heavy Chains/metabolism , Organelle Biogenesis , Oxygen Consumption , Pregnancy , Sheep
4.
J Physiol ; 598(12): 2453-2468, 2020 06.
Article in English | MEDLINE | ID: mdl-32087026

ABSTRACT

KEY POINTS: Skeletal muscle energy requirements increase at birth but little is known regarding the development of mitochondria that provide most of the cellular energy as ATP. Thyroid hormones are known regulators of adult metabolism and are important in driving several aspects of fetal development, including muscle fibre differentiation. Mitochondrial density and the abundance of mitochondrial membrane proteins in skeletal muscle increased during late gestation. However, mitochondrial functional capacity, measured as oxygen consumption rate, increased primarily after birth. Fetal hypothyroidism resulted in significant reductions in mitochondrial function and density in skeletal muscle before birth. Mitochondrial function matures towards birth and is dependent on the presence of thyroid hormones, with potential implications for the health of pre-term and hypothyroid infants. ABSTRACT: Birth is a significant metabolic challenge with exposure to a pro-oxidant environment and the increased energy demands for neonatal survival. This study investigated the development of mitochondrial density and activity in ovine biceps femoris skeletal muscle during the perinatal period and examined the role of thyroid hormones in these processes. Muscle capacity for oxidative phosphorylation increased primarily after birth but was accompanied by prepartum increases in mitochondrial density and the abundance of electron transfer system (ETS) complexes I-IV and ATP-synthase as well as by neonatal upregulation of uncoupling proteins. This temporal disparity between prepartum maturation and neonatal upregulation of mitochondrial oxidative capacity may protect against oxidative stress associated with birth while ensuring energy availability to the neonate. Fetal thyroid hormone deficiency reduced oxidative phosphorylation and prevented the prepartum upregulation of mitochondrial density and ETS proteins in fetal skeletal muscle. Overall, the data show that mitochondrial function matures over the perinatal period and is dependent on thyroid hormones, with potential consequences for neonatal viability and adult metabolic health.


Subject(s)
Muscle, Skeletal , Thyroid Hormones , Adult , Animals , Female , Humans , Mitochondria/metabolism , Mitochondria, Muscle/metabolism , Muscle, Skeletal/metabolism , Oxidative Phosphorylation , Oxygen Consumption , Pregnancy , Sheep , Thyroid Hormones/metabolism
5.
Equine Vet J ; 52(2): 165-173, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31721295

ABSTRACT

In many species, the pattern of growth and physiological development in utero has an important role in determining not only neonatal viability but also adult phenotype and disease susceptibility. Changes in fetal development induced by a range of environmental factors including maternal nutrition, disease, placental insufficiency and social stresses have all been shown to induce adult cardiovascular and metabolic dysfunction that often lead to ill health in later life. Compared to other precocious animals, much less is known about the physiological development of the fetal horse or the longer-term impacts on its phenotype of altered development in early life because of its inaccessibility in utero, large size and long lifespan. This review summaries the available data on the normal metabolic, cardiovascular and endocrine development of the fetal horse during the second half of gestation. It also examines the responsiveness of these physiological systems to stresses such as hypoglycaemia and hypotension during late gestation. Particular emphasis is placed on the role of the equine placenta and fetal endocrine glands in mediating the changes in fetal development seen towards term and in response to nutritional and other environmental cues. The final part of the review presents the evidence that the early life environment of the horse can alter its subsequent metabolic, cardiovascular and endocrine phenotype as well as its postnatal growth and bone development. It also highlights the immediate neonatal environment as a key window of susceptibility for programming of equine phenotype. Although further studies are needed to identify the cellular and molecular mechanisms involved, developmental programming of physiological phenotype is likely to have important implications for the health and potential athletic performance of horses, particularly if born with abnormal bodyweight, premature or dysmature characteristics or produced by assisted reproductive technologies, indicative of an altered early life environment.


Subject(s)
Fetal Development , Placenta , Animals , Female , Fetus , Horses , Phenotype , Pregnancy
6.
J Dev Orig Health Dis ; 8(2): 206-215, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27995843

ABSTRACT

In several species, adult metabolic phenotype is influenced by the intrauterine environment, often in a sex-linked manner. In horses, there is also a window of susceptibility to programming immediately after birth but whether adult glucose-insulin dynamics are altered by neonatal conditions remains unknown. Thus, this study investigated the effects of birth weight, sex and neonatal glucocorticoid overexposure on glucose-insulin dynamics of young adult horses. For the first 5 days after birth, term foals were treated with saline as a control or ACTH to raise cortisol levels to those of stressed neonates. At 1 and 2 years of age, insulin secretion and sensitivity were measured by exogenous glucose administration and hyperinsulinaemic-euglycaemic clamp, respectively. Glucose-stimulated insulin secretion was less in males than females at both ages, although there were no sex-linked differences in glucose tolerance. Insulin sensitivity was greater in females than males at 1 year but not 2 years of age. Birth weight was inversely related to the area under the glucose curve and positively correlated to insulin sensitivity at 2 years but not 1 year of age. In contrast, neonatal glucocorticoid overexposure induced by adrenocorticotropic hormone (ACTH) treatment had no effect on whole body glucose tolerance, insulin secretion or insulin sensitivity at either age, although this treatment altered insulin receptor abundance in specific skeletal muscles of the 2-year-old horses. These findings show that glucose-insulin dynamics in young adult horses are sexually dimorphic and determined by a combination of genetic and environmental factors acting during early life.


Subject(s)
Birth Weight , Blood Glucose/metabolism , Glucocorticoids/pharmacology , Insulin/metabolism , Aging , Animals , Animals, Newborn , Female , Horses , Insulin Resistance , Male , Receptor, Insulin/metabolism , Sex Factors
7.
Domest Anim Endocrinol ; 56 Suppl: S121-32, 2016 07.
Article in English | MEDLINE | ID: mdl-27345310

ABSTRACT

Glucocorticoids (GCs) are important environmental and maturational signals during intrauterine development. Toward term, the maturational rise in fetal glucocorticoid receptor concentrations decreases fetal growth and induces differentiation of key tissues essential for neonatal survival. When cortisol levels rise earlier in gestation as a result of suboptimal conditions for fetal growth, the switch from tissue accretion to differentiation is initiated prematurely, which alters the phenotype that develops from the genotype inherited at conception. Although this improves the chances of survival should delivery occur, it also has functional consequences for the offspring long after birth. Glucocorticoids are, therefore, also programming signals that permanently alter tissue structure and function during intrauterine development to optimize offspring fitness. However, if the postnatal environmental conditions differ from those signaled in utero, the phenotypical outcome of early-life glucocorticoid receptor overexposure may become maladaptive and lead to physiological dysfunction in the adult. This review focuses on the role of GCs in developmental programming, primarily in farm species. It examines the factors influencing GC bioavailability in utero and the effects that GCs have on the development of fetal tissues and organ systems, both at term and earlier in gestation. It also discusses the windows of susceptibility to GC overexposure in early life together with the molecular mechanisms and long-term consequences of GC programming with particular emphasis on the cardiovascular, metabolic, and endocrine phenotype of the offspring.


Subject(s)
Fetal Development , Glucocorticoids/metabolism , Livestock/physiology , Uterus/physiology , Animals , Female , Pregnancy
8.
Domest Anim Endocrinol ; 50: 45-9, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25240233

ABSTRACT

The present study tested the hypothesis that overexposure to endogenous glucocorticoids in neonatal life alters the reactivity of the hypothalamic-pituitary-adrenal (HPA) axis in ponies at 1 and 2 yr of age. Newborn foals received saline (0.9% NaCl, n = 8, control) or long-acting adrenocorticotropic hormone (ACTH1-24) (Depot Synacthen 0.125 mg intramuscularly twice daily, n = 9) for 5 d after birth to raise cortisol concentrations 5- to 6-fold. At 1 and 2 yr of age, HPA axis function was assessed by bolus administration of short-acting ACTH1-24 (1 µg/kg intravenous) and insulin (0.5 U/kg intravenous) to induce hypoglycemic on separate days. Arterial blood samples were taken at 5 to 30-min intervals before and after drug administration to measure plasma ACTH and/or cortisol concentrations. There were no differences in the basal plasma ACTH or cortisol concentrations or in the cortisol response to exogenous ACTH1-24 with neonatal treatment or age. At 1 and 2 yr of age, the increment in plasma ACTH but not cortisol at 60 min in response to insulin-induced hypoglycemia was greater in ponies treated neonatally with ACTH than saline (P < 0.05). Neonatal cortisol overexposure induced by neonatal ACTH treatment, therefore, alters functioning of the HPA axis in adult ponies.


Subject(s)
Adrenocorticotropic Hormone/blood , Animals, Newborn , Cosyntropin/pharmacology , Glucocorticoids/pharmacology , Horses/physiology , Pituitary-Adrenal System/physiology , Adrenal Cortex/drug effects , Aging , Animals , Cosyntropin/administration & dosage , Glucocorticoids/administration & dosage , Hydrocortisone/blood
9.
Placenta ; 36 Suppl 1: S50-9, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25524059

ABSTRACT

Hormones have an important role in regulating fetal development. They act as environmental signals and integrate tissue growth and differentiation with relation to nutrient availability. While hormones control the developmental fate of resources available to the fetus, the actual supply of nutrients and oxygen to the fetus depends on the placenta. However, much less is known about the role of hormones in regulating placental development, even though the placenta has a wide range of hormone receptors and produces hormones itself from early in gestation. The placenta is, therefore, exposed to hormones by autocrine, paracrine and endocrine mechanisms throughout its lifespan. It is known to adapt its phenotype in response to environmental cues and fetal demand signals, particularly when there is a disparity between the fetal genetic drive for growth and the nutrient supply. These adaptive responses help to maintain fetal growth during adverse conditions and are likely to depend, at least in part, on the hormonal milieu. This review examines the endocrine regulation of placental phenotype with particular emphasis on the glucocorticoid hormones. It focuses on the availability of placental hormone receptors and on the effects of hormones on the morphology, transport capacity and endocrine function of the placenta.


Subject(s)
Hormones/pharmacology , Placenta/drug effects , Placentation/drug effects , Animals , Female , Fetal Development/drug effects , Hormones/physiology , Humans , Maternal-Fetal Exchange/drug effects , Phenotype , Placenta/physiology , Placental Hormones/pharmacology , Placental Hormones/physiology , Pregnancy , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Cytoplasmic and Nuclear/physiology
10.
Placenta ; 35(5): 305-10, 2014 May.
Article in English | MEDLINE | ID: mdl-24680290

ABSTRACT

INTRODUCTION AND METHODS: Tragulus, the mouse deer, is considered the most primitive ruminant, with a diffuse placenta grossly quite unlike the cotyledonary type of the other ruminants. This immunocytochemical investigation of placental transporters was designed to elucidate possible mechanisms of evolution to the cotyledonary form. RESULTS AND DISCUSSION: Tragulus expresses several of the major transport systems characteristic of the ruminants: the trophoblast binucleate cell (BNC) dynamics, the requirement for two isoforms, GT1 and GT3, for glucose transport, the provision of Aquaporin 3 for water control, and uterine milk and histiotrophic secretion from uterine glands. However whereas the expression of the 9 kD Calcium Binding Protein (9 CBP) for calcium transport in ruminants is restricted to the intercotyledonary trophoblast with its areolae, Tragulus, having no intercotyledonary area, expresses 9 CBP throughout the villus trophoblast. There is some localised development of areolar-like structures in the mid term Tragulus but it is insignificant at term. The strong expression of Glucose Transporter 1 (GT1) in the BNC granules is unique to Tragulus. CONCLUSION: Tragulus relies on essentially similar transport and BNC dynamics as the other ruminants. Thus the evolutionary pressures driving the development of the cotyledonary placenta probably lie in the increase in body size and the consequent need for a larger placental area to ensure sufficient glucose for the fetus. The delivery in Tragulus of GT1 to the maternal facing side may be this species unique solution to maintain the glucose supply.


Subject(s)
Biological Evolution , Placenta/metabolism , Ruminants/metabolism , Trophoblasts/metabolism , Animals , Aquaporins/metabolism , Female , Glycoproteins/metabolism , Immunohistochemistry , Pregnancy , Sodium-Glucose Transport Proteins/metabolism , alpha 1-Antitrypsin/metabolism
11.
J Endocrinol ; 221(3): R87-R103, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24648121

ABSTRACT

The thyroid hormones, thyroxine (T4) and triiodothyronine (T3), are essential for normal growth and development of the fetus. Their bioavailability in utero depends on development of the fetal hypothalamic-pituitary-thyroid gland axis and the abundance of thyroid hormone transporters and deiodinases that influence tissue levels of bioactive hormone. Fetal T4 and T3 concentrations are also affected by gestational age, nutritional and endocrine conditions in utero, and placental permeability to maternal thyroid hormones, which varies among species with placental morphology. Thyroid hormones are required for the general accretion of fetal mass and to trigger discrete developmental events in the fetal brain and somatic tissues from early in gestation. They also promote terminal differentiation of fetal tissues closer to term and are important in mediating the prepartum maturational effects of the glucocorticoids that ensure neonatal viability. Thyroid hormones act directly through anabolic effects on fetal metabolism and the stimulation of fetal oxygen consumption. They also act indirectly by controlling the bioavailability and effectiveness of other hormones and growth factors that influence fetal development such as the catecholamines and insulin-like growth factors (IGFs). By regulating tissue accretion and differentiation near term, fetal thyroid hormones ensure activation of physiological processes essential for survival at birth such as pulmonary gas exchange, thermogenesis, hepatic glucogenesis, and cardiac adaptations. This review examines the developmental control of fetal T4 and T3 bioavailability and discusses the role of these hormones in fetal growth and development with particular emphasis on maturation of somatic tissues critical for survival immediately at birth.


Subject(s)
Fetal Development/physiology , Maternal-Fetal Exchange/physiology , Thyroxine/physiology , Triiodothyronine/physiology , Female , Humans , Hypothalamo-Hypophyseal System/embryology , Hypothalamo-Hypophyseal System/physiology , Models, Biological , Pituitary-Adrenal System/embryology , Pituitary-Adrenal System/physiology , Pregnancy , Thyroxine/metabolism , Triiodothyronine/metabolism
12.
Equine Vet J Suppl ; (41): 38-42, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22594024

ABSTRACT

REASONS FOR PERFORMING THE STUDY: The effects of overexposure to glucocorticoids during early life of the foal on the subsequent HPA programming of the hypothalamic-pituitary-adrenal axis are unknown. OBJECTIVES: To test the hypotheses that excess glucocorticoid exposure in early life subsequently increases both basal plasma concentrations of cortisol and the adrenocortical responsiveness to exogenous adrenocorticotropic hormone (ACTH). METHODS: Foals received either saline (0.9% NaCl, n = 9) or long-acting ACTH (0.125 mg i.m. b.i.d., n = 6) for 5 days from Day 1 to increase endogenous cortisol concentrations. Long-term indwelling catheters were inserted under local anaesthesia into the jugular veins of foals aged 2 and 12 weeks. After recovery, short-acting ACTH1-24 was given as a single i.v. injection (2 microg/kg bwt) and blood samples were taken at 5-30 min intervals before and after ACTH administration to measure plasma cortisol concentrations. RESULTS: Basal plasma cortisol concentrations were higher in ACTH- than in saline-treated foals at age 3 weeks, but not at 13 weeks. There were no significant differences in either the time profile or the area under the cortisol curve in response to ACTH between the 2 groups. CONCLUSIONS: These data suggest that ACTH-induced overexposure to glucocorticoids during early post natal life of the foal does not have a programming effect on HPA axis function at 13 weeks. In foals, the effects of ACTH-induced overexposure to glucocorticoids, if any, may not become apparent until much later in life in a long-lived species such as the horse. POTENTIAL RELEVANCE: These studies suggest that clinical and other stressful conditions that raise plasma cortisol concentrations during early life are unlikely to programme cardiovascular and metabolic function in horses in the short term.


Subject(s)
Animals, Newborn , Cosyntropin/pharmacology , Horses/physiology , Hypothalamo-Hypophyseal System/drug effects , Pituitary-Adrenal System/drug effects , Adrenal Glands/drug effects , Adrenal Glands/physiology , Animals , Female , Hydrocortisone/blood , Hypothalamo-Hypophyseal System/physiology , Male , Pituitary-Adrenal System/physiology
13.
Equine Vet J Suppl ; (41): 130-9, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22594041

ABSTRACT

In adapting to life ex utero, the foal encounters a number of physiological challenges. It has to assume the nutritional, respiratory and excretory functions of the placenta and activate full regulatory control over its own internal environment for the first time. To achieve this, there must be structural and functional changes to a wide range of tissues including several endocrine glands. In most species, including the horse, these maturational changes begin in late gestation and continue into the first few days of neonatal life. Consequently, during this perinatal period, there are major changes in the sensitivity and/or set point of key endocrine axes, which alter the circulating hormone concentrations in the foal. In turn, these endocrine changes are responsible for many of the other physiological adaptations essential for neonatal survival. The perinatal alterations in the hypothalamic-pituitary-adrenal (HPA) axis are particularly important in these processes, although the sympatho-adrenal medullary axis and endocrine pancreas also have key roles in ensuring homeostasis during the multiple novel stimuli experienced at birth. Abnormalities in the perinatal endocrine profile caused by adverse conditions before or after birth may, therefore, lead to maladaptation or aid survival of the newborn foal depending on the specific circumstances. This review examines the perinatal changes in endocrinology in normal and compromised foals and the role of these endocrine changes in the physiological adaptations to extrauterine life with particular emphasis on the HPA axis, adreno-medullary catecholamines and the endocrine pancreas.


Subject(s)
Adaptation, Physiological/physiology , Animals, Newborn/physiology , Horses/physiology , Hypothalamo-Hypophyseal System/physiology , Pituitary-Adrenal System/physiology , Animals
14.
J Endocrinol ; 210(2): 189-98, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21642376

ABSTRACT

Thyroid hormones are important for normal bone growth and development in postnatal life. However, little is known about the role of thyroid hormones in the control of bone development in the fetus. Using computed tomography and mechanical testing, the structure and strength of metatarsal bones were measured in sheep fetuses in which thyroid hormone levels were altered by thyroidectomy or adrenalectomy. In intact fetuses, plasma concentrations of total calcium and the degradation products of C-terminal telopeptides of type I collagen increased between 100 and 144 days of gestation (term 145±2 days), in association with various indices of bone growth and development. Thyroid hormone deficiency induced by thyroidectomy at 105-110 days of gestation caused growth retardation of the fetus and significant changes in metatarsal bone structure and strength when analyzed at both 130 and 144 days of gestation. In hypothyroid fetuses, trabecular bone was stronger with thicker, more closely spaced trabeculae, despite lower bone mineral density. Plasma osteocalcin was reduced by fetal thyroidectomy. Removal of the fetal adrenal gland at 115-120 days of gestation, and prevention of the prepartum rises in cortisol and triiodothyronine, had no effect on bodyweight, limb lengths, metatarsal bone structure or strength, or circulating markers of bone metabolism in the fetuses studied near term. This study demonstrates that hypothyroidism in utero has significant effects on the structure and strength of bone, with different consequences for cortical and trabecular bone.


Subject(s)
Bone Development/physiology , Bone and Bones/physiology , Fetal Development/physiology , Hypothyroidism/physiopathology , Thyroid Hormones/blood , Animals , Bone and Bones/diagnostic imaging , Calcium/blood , Hydrocortisone/blood , Hypothyroidism/blood , Hypothyroidism/diagnostic imaging , Radiography , Sheep , Thyroidectomy , Weight-Bearing/physiology
15.
Am J Physiol Endocrinol Metab ; 300(1): E94-102, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20959526

ABSTRACT

In adults, the adrenal glands are essential for the metabolic response to stress, but little is known about their role in fetal metabolism. This study examined the effects of adrenalectomizing fetal sheep on glucose and oxygen metabolism in utero in fed conditions and after maternal fasting for 48 h near term. Fetal adrenalectomy (AX) had little effect on the rates of glucose and oxygen metabolism by the fetus or uteroplacental tissues in fed conditions. Endogenous glucose production was negligible in both AX and intact, sham-operated fetuses in fed conditions. Maternal fasting reduced fetal glucose levels and umbilical glucose uptake in both groups of fetuses to a similar extent but activated glucose production only in the intact fetuses. The lack of fasting-induced glucogenesis in AX fetuses was accompanied by falls in fetal glucose utilization and oxygen consumption not seen in intact controls. The circulating concentrations of cortisol and total catecholamines, and the hepatic glycogen content and activities of key gluconeogenic enzymes, were also less in AX than intact fetuses in fasted animals. Insulin concentrations were also lower in AX than intact fetuses in both nutritional states. Maternal glucose utilization and its distribution between the fetal, uteroplacental, and nonuterine maternal tissues were unaffected by fetal AX in both nutritional states. Ovine fetal adrenal glands, therefore, have little effect on basal rates of fetal glucose and oxygen metabolism but are essential for activating fetal glucogenesis in response to maternal fasting. They may also be involved in regulating insulin sensitivity in utero.


Subject(s)
Adrenal Cortex Hormones/physiology , Adrenal Glands/physiology , Fetal Nutrition Disorders/physiopathology , Food Deprivation , Gluconeogenesis , Maternal Nutritional Physiological Phenomena , Adrenal Glands/embryology , Adrenalectomy , Algorithms , Animals , Blood Glucose/analysis , Female , Fetal Nutrition Disorders/blood , Glucose/metabolism , Liver/embryology , Liver/enzymology , Liver Glycogen/analysis , Oxygen Consumption , Placenta/metabolism , Pregnancy , Sheep , Umbilical Cord/metabolism , Uterus/metabolism
16.
Soc Reprod Fertil Suppl ; 67: 41-57, 2010.
Article in English | MEDLINE | ID: mdl-21755662

ABSTRACT

The pattern of intrauterine growth and size at birth, in particular, programmes the structure and function of tissues later in life in many species, which has important implications for the incidence of adult-onset generative diseases in human populations. In mammals, the main determinant of intrauterine growth is the placental supply of nutrients which, in turn, depends on the size, morphology, transport characteristics and endocrine function of the placenta. However, compared to somatic tissues, little is known about the developmental programming of the placenta. This review examines the epigenetic regulation of placental phenotype with particular emphasis on the nutrient transfer capacity of the ovine placenta and environmental factors shown to cause developmental programming of other tissues. Overall, the placenta is responsive to environmental factors and uses a number of different strategies to adapt its phenotype to help support fetal growth during adverse intrauterine conditions. It is, therefore, not just a passive conduit for nutrient transfer to the fetus but alters its nutrient supply capacity dynamically to optimise fetal nutrient acquisition. Thus, the placental epigenome provides both a memory of environmental conditions experienced during development and an index of the future well being of the offspring.


Subject(s)
Placentation , Sheep/physiology , Animals , Female , Gene Expression Regulation, Developmental/physiology , Pregnancy
17.
J Endocrinol ; 204(3): 319-29, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20016055

ABSTRACT

Glucocorticoid administration during pregnancy programmes cardiovascular and metabolic functions in the adult offspring. Often, the control procedures are stressful per se and raise maternal glucocorticoid concentrations. This study compared the effects of maternal injection with dexamethasone (dex, 200 microg/kg) or saline with no treatment from 15 to 20 days of rat pregnancy on offspring growth and glucose metabolism. Near term, maternal corticosterone concentrations were higher in the saline-treated dams and lower in the dex-treated dams relative to untreated animals. In both male and female offspring, growth rate was measured for 14 weeks, and glucose tolerance was assessed between 12 and 13 weeks together with body fat content and plasma concentrations of insulin, leptin, and corticosterone between 14 and 15 weeks. Offspring liver was collected at different ages and was analyzed for glycogen content and gluconeogenic enzyme activity. Compared with untreated animals, both dex and saline treatments altered postnatal growth although adult body weight was unaffected. The two treatments had different effects on adult insulin concentrations and on hepatic glycogen content and gluconeogenic enzyme activities both pre- and postnatally. Relative to untreated animals, adult glucose tolerance was improved by maternal saline injection in males but not in females, while it was impaired in female offspring but not in male offspring of the dex-treated dams. Adult glucose tolerance was related to male body fat content but not to female body fat content. Dex and saline treatments of pregnant rats have differential sex-linked effects on the growth and glucose metabolism of their offspring, which indicates that the programming actions of natural and synthetic glucocorticoids may differ.


Subject(s)
Dexamethasone/administration & dosage , Glucocorticoids/administration & dosage , Glucose/metabolism , Growth/drug effects , Maternal-Fetal Exchange , Animals , Animals, Newborn/growth & development , Animals, Newborn/metabolism , Body Weight , Dexamethasone/adverse effects , Female , Glucocorticoids/adverse effects , Male , Pregnancy , Rats , Rats, Wistar , Stress, Physiological
18.
Nutr Metab Cardiovasc Dis ; 19(8): 555-62, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19179059

ABSTRACT

BACKGROUND AND AIM: Low birth weight is associated with an increased incidence of adult glucose intolerance, type 2 diabetes and cardiovascular disease in humans. In pregnant rats, dietary calorie or protein deprivation results in growth retarded pups, which become glucose intolerant adults with abnormal hepatic glucose metabolism and gluconeogenic enzyme activities. However, whether these abnormalities are present before birth remain unknown. METHODS AND RESULTS: This study examined the effects of manipulating dietary protein and carbohydrate intake during rat pregnancy on the fetal and maternal hepatic activities of the gluconeogenic enzymes, glucose-6-phosphatase (G6Pase) and phosphoenolpyruvate carboxykinase (PEPCK). Wistar rats were fed ad libitum with either standard chow throughout pregnancy (25% protein, 57% carbohydrate, n=6) or an isocaloric, low protein, high carbohydrate diet (LPHC, 8% protein, 81% carbohydrate) for different periods of pregnancy (early, 0-10 days, n=6; late, 10-20 days, n=7; throughout, 0-20 days, n=6) before tissue collection at day 20. The LPHC diet had no effect on fetal or placental weights, or on fetal hepatic activities of G6Pase and PEPCK in the early LPHC group. In contrast, fetuses of dams fed the LPHC diet in late or throughout pregnancy had lower body and placental weights, and higher hepatic G6Pase and PEPCK activities than controls. Maternal hepatic G6Pase activity was elevated in all LPHC groups, while maternal PEPCK activity was only increased significantly in the late LPHC group. CONCLUSIONS: Feeding a LPHC diet, particularly during late pregnancy, therefore, up-regulates fetal and maternal hepatic glucogenic capacity.


Subject(s)
Animal Nutritional Physiological Phenomena , Caloric Restriction , Disease Models, Animal , Fetus/metabolism , Gluconeogenesis , Liver/metabolism , Maternal Nutritional Physiological Phenomena , Protein Deficiency/metabolism , Animals , Dietary Carbohydrates/administration & dosage , Dietary Carbohydrates/metabolism , Dietary Proteins/administration & dosage , Dietary Proteins/metabolism , Female , Fetal Weight , Fetus/enzymology , Fetus/physiopathology , Gestational Age , Glucose-6-Phosphatase/metabolism , Glycogen/metabolism , Liver/embryology , Liver/enzymology , Liver/physiopathology , Phosphoenolpyruvate Carboxykinase (GTP)/metabolism , Placenta/metabolism , Placentation , Pregnancy , Protein Deficiency/embryology , Protein Deficiency/physiopathology , Rats , Rats, Wistar
19.
Exp Clin Endocrinol Diabetes ; 116(7): 393-403, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18704835

ABSTRACT

Delivery of viable young requires co-ordination of fetal maturation with the onset of labour at term. In turn, this depends on a cascade of fetal and maternal endocrine events. The sequence of these events is broadly similar in most mammals but there are differences in placental hormone synthesis and in the timing and magnitude of key prepartum endocrine changes between species. In most farm animals, maternal progesterone (P4) levels decline and oestrogen levels increase in the last 5 - 10 days before delivery in response to activation of the fetal hypothalamicpituitary-adrenal (HPA) axis and increased fetal cortisol concentrations. This cortisol surge is also responsible for fetal maturation and increasing uteroplacental prostaglandin (PG) synthesis. In the mare, there is little, if any, P4 in the maternal plasma during late gestation and both progestagens and oestrogens are produced by a feto-placental unit which uses precursors supplied by the fetus to synthesise a range of C21 and C18 steroids, many of which are unique to the horse. Regulation of uterine quiescence and activation is, therefore, complex in the mare near term. Indeed, total progestagen concentrations rise and total oestrogen levels fall in the mare during the last 20 - 30 days of gestation and only show the changes typical of impending parturition in other species in the last 24 - 48 h before delivery. Fetal cortisol concentrations also rise late in gestation in the horse compared to other species. In common with other species, the prepartum endocrine cascade appears to begin in the fetal horse with activation of the fetal HPA axis but, initially, the primary product of the fetal equine adrenal appears to be pregnenolone (P5) and not cortisol. This leads to increased progestagen production by the uteroplacental tissues, which maintains uterine quiescence in the face of increasing uterine stretch caused by the rapidly growing fetus. Very close to term in association with increasing fetal ACTH levels, the fetal equine adrenals appear to switch to producing cortisol. This late cortisol surge induces a period of rapid fetal maturation and may also contribute to increased uteroplacental oestradiol-17 beta and PG production. The fall in P5 availability may reduce uteroplacental progestagen production and lift the block on myometrial contractility. Finally, increased PG secretion activates myometrial contractions, which stimulate oxytocin release via a neuroendocrine reflex. The endocrine regulation of equine parturition, therefore, involves progestagens, oestrogens, PGs and oxytocin as in other species. However, further studies are required to establish the causes and consequences of the rise and fall in maternal progestagens and the extent to which initiation of equine labour depends on the fetal HPA axis.


Subject(s)
Estrogens/physiology , Horses/physiology , Parturition/physiology , Progestins/physiology , Animals , Female , Fetal Development/physiology , Hypothalamo-Hypophyseal System/physiology , Pregnancy
20.
J Neuroendocrinol ; 20(4): 439-50, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18266944

ABSTRACT

Intrauterine programming is the process by which the structure and function of tissues are altered permanently by insults acting during early development. In mammals, the placenta controls intrauterine development by supplying oxygen and nutrients, and by regulating the bioavailability of specific hormones involved in foetal growth and development. Consequently, the placenta is likely to have a key role in mediating the programming effects of suboptimal conditions during development. This review examines placental phenotype in different environmental conditions and places particular emphasis on regulation of placental nutrient transfer capacity and endocrine function by insults known to cause intrauterine programming. More specifically, it examines the effects of a range of environmental challenges on the size, morphology, blood flow and transporter abundance of the placenta and on its rate of consumption and production of nutrients. In addition, it considers the role of hormone synthesis and metabolism by the placenta in matching intrauterine development to the prevailing environmental conditions. The adaptive responses that the placenta can make to compensate for suboptimal conditions in utero are also assessed in relation to the strategies adopted to maximise foetal growth and viability at birth. Environmentally-induced changes in placental phenotype may provide a mechanism for transmitting the memory of early events to the foetus later in gestation, which leads to intrauterine programming of tissue development long after the original insult.


Subject(s)
Embryonic Development/physiology , Placenta/physiology , Animals , Biological Transport/physiology , Endocrine System/physiology , Female , Food , Humans , Membrane Transport Proteins/metabolism , Models, Biological , Organ Size/physiology , Placenta/anatomy & histology , Placenta/blood supply , Placenta/metabolism , Placental Circulation/physiology , Pregnancy , Regional Blood Flow/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...