Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Physiol ; 14: 1124938, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36935746

RESUMO

Growth and maturation of the fetal gastrointestinal tract near term prepares the offspring for the onset of enteral nutrition at birth. Structural and functional changes are regulated by the prepartum rise in cortisol in the fetal circulation, although the role of the coincident rise in plasma tri-iodothyronine (T3) is unknown. This study examined the effect of hypothyroidism on the structural development of the gastrointestinal tract and the activity of brush-border digestive enzymes in the ovine fetus near term. In intact fetuses studied between 100 and 144 days of gestation (dGA; term ∼145 days), plasma concentrations of T3, cortisol and gastrin; the mucosal thickness in the abomasum, duodenum, jejunum and ileum; and intestinal villus height and crypt depth increased with gestational age. Removal of the fetal thyroid gland at 105-110 dGA suppressed plasma thyroxine (T4) and T3 concentrations to the limit of assay detection in fetuses studied at 130 and 144 dGA, and decreased plasma cortisol and gastrin near term, compared to age-matched intact fetuses. Hypothyroidism was associated with reductions in the relative weights of the stomach compartments and small intestines, the outer perimeter of the intestines, the thickness of the gastric and intestinal mucosa, villus height and width, and crypt depth. The thickness of the mucosal epithelial cell layer and muscularis propria in the small intestines were not affected by gestational age or treatment. Activities of the brush border enzymes varied with gestational age in a manner that depended on the enzyme and region of the small intestines studied. In the ileum, maltase and dipeptidyl peptidase IV (DPPIV) activities were lower, and aminopeptidase N (ApN) were higher, in the hypothyroid compared to intact fetuses near term. These findings highlight the importance of thyroid hormones in the structural and functional development of the gastrointestinal tract near term, and indicate how hypothyroidism in utero may impair the transition to enteral nutrition and increase the risk of gastrointestinal disorders in the neonate.

2.
J Dev Orig Health Dis ; 14(1): 77-87, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-35822505

RESUMO

Prenatal glucocorticoid overexposure causes adult metabolic dysfunction in several species but its effects on adult mitochondrial function remain largely unknown. Using respirometry, this study examined mitochondrial substrate metabolism of fetal and adult ovine biceps femoris (BF) and semitendinosus (ST) muscles after cortisol infusion before birth. Physiological increases in fetal cortisol concentrations pre-term induced muscle- and substrate-specific changes in mitochondrial oxidative phosphorylation capacity in adulthood. These changes were accompanied by muscle-specific alterations in protein content, fibre composition and abundance of the mitochondrial electron transfer system (ETS) complexes. In adult ST, respiration using palmitoyl-carnitine and malate was increased after fetal cortisol treatment but not with other substrate combinations. There were also significant increases in protein content and reductions in the abundance of all four ETS complexes, but not ATP synthase, in the ST of adults receiving cortisol prenatally. In adult BF, intrauterine cortisol treatment had no effect on protein content, respiratory rates, ETS complex abundances or ATP synthase. Activity of citrate synthase, a marker of mitochondrial content, was unaffected by intrauterine treatment in both adult muscles. In the ST but not BF, respiratory rates using all substrate combinations were significantly lower in the adults than fetuses, predominantly in the saline-infused controls. The ontogenic and cortisol-induced changes in mitochondrial function were, therefore, more pronounced in the ST than BF muscle. Collectively, the results show that fetal cortisol overexposure programmes mitochondrial substrate metabolism in specific adult muscles with potential consequences for adult metabolism and energetics.


Assuntos
Hidrocortisona , Mitocôndrias , Gravidez , Feminino , Animais , Ovinos , Hidrocortisona/metabolismo , Mitocôndrias/metabolismo , Músculo Esquelético/metabolismo , Parto , Fosforilação Oxidativa
3.
Biomolecules ; 12(6)2022 05 31.
Artigo em Inglês | MEDLINE | ID: mdl-35740893

RESUMO

In adults, glucocorticoids are stress hormones that act, partly, through actions on mitochondrial oxidative phosphorylation (OXPHOS) to increase energy availability. Before birth, glucocorticoids are primarily maturational signals that prepare the fetus for new postnatal challenges. However, the role of the normal prepartum glucocorticoid rise in preparing mitochondria for the increased postnatal energy demands remains largely unknown. This study examined the effect of physiological increases in the fetal cortisol concentration on cerebral mitochondrial OXPHOS capacity near term (~130 days gestation, term ~145 days gestation). Fetal sheep were infused with saline or cortisol for 5 days at ~0.8 of gestation before the mitochondrial content, respiratory rates, abundance of the electron transfer system proteins and OXPHOS efficiency were measured in their cortex and cerebellum. Cerebral morphology was assessed by immunohistochemistry and stereology. Cortisol treatment increased the mitochondrial content, while decreasing Complex I-linked respiration in the cerebellum. There was no effect on the cortical mitochondrial OXPHOS capacity. Cortisol infusion had regional effects on cerebral morphology, with increased myelination in the cerebrum. The findings demonstrate the importance of cortisol in regulating the cerebral mitochondrial OXPHOS capacity prenatally and have implications for infants born preterm or after glucocorticoid overexposure due to pregnancy complications or clinical treatment.


Assuntos
Glucocorticoides , Hidrocortisona , Animais , Encéfalo/metabolismo , Feminino , Feto/metabolismo , Idade Gestacional , Glucocorticoides/metabolismo , Glucocorticoides/farmacologia , Humanos , Hidrocortisona/farmacologia , Mitocôndrias/metabolismo , Fosforilação Oxidativa , Gravidez , Ovinos
4.
Nutrients ; 14(11)2022 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-35684104

RESUMO

Glucocorticoids have an important role in development of the metabolic phenotype in utero. They act as environmental and maturational signals in adapting feto-placental metabolism to maximize the chances of survival both before and at birth. They influence placental nutrient handling and fetal metabolic processes to support fetal growth, fuel storage and energy production with respect to nutrient availability. More specifically, they regulate the transport, utilization and production of a range of nutrients by the feto-placental tissues that enables greater metabolic flexibility in utero while minimizing any further drain on maternal resources during periods of stress. Near term, the natural rise in fetal glucocorticoid concentrations also stimulates key metabolic adaptations that prepare tissues for the new energy demanding functions after birth. Glucocorticoids, therefore, have a central role in the metabolic communication between the mother, placenta and fetus that optimizes offspring metabolic phenotype for survival to reproductive age. This review discusses the effects of maternal and fetal glucocorticoids on the supply and utilization of nutrients by the feto-placental tissues with particular emphasis on studies using quantitative methods to assess metabolism in rodents and sheep in vivo during late pregnancy. It considers the routes of glucocorticoid overexposure in utero, including experimental administration of synthetic glucocorticoids, and the mechanisms by which these hormones control feto-placental metabolism at the molecular, cellular and systems levels. It also briefly examines the consequences of intrauterine glucocorticoid overexposure for postnatal metabolic health and the generational inheritance of metabolic phenotype.


Assuntos
Glucocorticoides , Placenta , Animais , Feminino , Desenvolvimento Fetal , Feto/metabolismo , Glucocorticoides/metabolismo , Parto , Placenta/metabolismo , Gravidez , Ovinos
5.
Domest Anim Endocrinol ; 78: 106657, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34525421

RESUMO

Hormones have an important role in regulating fetal metabolism in relation to the prevailing nutritional conditions both in late gestation and during the prepartum period as the fetus prepares for birth. In particular, the pancreatic, thyroid and adrenal hormones all affect fetal uptake and utilization of nutrients for oxidative metabolism, tissue accretion and fuel storage. These hormones also influence the fetal metabolic preparations for the nutritional transition from intra- to extra-uterine life. This review discusses the role of insulin, glucagon, thyroxine, tri-iodothyronine, cortisol and the catecholamines in these processes during normal intrauterine conditions and in response to maternal undernutrition with particular emphasis on the sheep fetus. It also considers the metabolic interactions between these hormones and their role in the maturation of key tissues, such as the liver, skeletal muscle and adipose tissue, in readiness for their new metabolic functions after birth. Endocrine regulation of fetal metabolism is shown to be multifactorial and dynamic with a central role in optimizing metabolic fitness for survival both in utero and at birth.


Assuntos
Sistema Endócrino , Feto , Animais , Sistema Endócrino/fisiologia , Feminino , Hidrocortisona/metabolismo , Troca Materno-Fetal , Gravidez , Ovinos , Tiroxina
6.
J Endocrinol ; 252(1): 71-80, 2021 11 24.
Artigo em Inglês | MEDLINE | ID: mdl-34708692

RESUMO

Hormones have an important role in the regulation of fetal growth and development, especially in response to nutrient availability in utero. Using micro-CT and an electromagnetic three-point bend test, this study examined the effect of pancreas removal at 0.8 fraction of gestation on the developing bone structure and mechanical strength in fetal sheep. When fetuses were studied at 10 and 25 days after surgery, pancreatectomy caused hypoinsulinaemia, hyperglycaemia and growth retardation which was associated with low plasma concentrations of leptin and a marker of osteoclast activity and collagen degradation. In pancreatectomized fetuses compared to control fetuses, limb lengths were shorter, and trabecular (Tb) bone in the metatarsi showed greater bone volume fraction, Tb thickness, degree of anisotropy and porosity, and lower fractional bone surface area and Tb spacing. Mechanical strength testing showed that pancreas deficiency was associated with increased stiffness and a greater maximal weight load at fracture in a subset of fetuses studied near term. Overall, pancreas deficiency in utero slowed the growth of the fetal skeleton and adapted the developing bone to generate a more compact and connected structure. Maintenance of bone strength in growth-retarded limbs is especially important in a precocial species in preparation for skeletal loading and locomotion at birth.


Assuntos
Desenvolvimento Ósseo/fisiologia , Desenvolvimento Fetal/fisiologia , Insulina/deficiência , Pancreatopatias/embriologia , Animais , Osso e Ossos/metabolismo , Feminino , Insulina/metabolismo , Pâncreas/metabolismo , Pâncreas/patologia , Pâncreas/cirurgia , Pancreatectomia , Pancreatopatias/complicações , Pancreatopatias/metabolismo , Pancreatopatias/fisiopatologia , Gravidez , Ovinos
7.
FASEB J ; 35(5): e21591, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33891344

RESUMO

Thyroid hormones regulate adult metabolism partly through actions on mitochondrial oxidative phosphorylation (OXPHOS). They also affect neurological development of the brain, but their role in cerebral OXPHOS before birth remains largely unknown, despite the increase in cerebral energy demand during the neonatal period. Thus, this study examined prepartum development of cerebral OXPHOS in hypothyroid fetal sheep. Using respirometry, Complex I (CI), Complex II (CII), and combined CI&CII OXPHOS capacity were measured in the fetal cerebellum and cortex at 128 and 142 days of gestational age (dGA) after surgical thyroidectomy or sham operation at 105 dGA (term ~145 dGA). Mitochondrial electron transfer system (ETS) complexes, mRNA transcripts related to mitochondrial biogenesis and ATP production, and mitochondrial density were quantified using molecular techniques. Cerebral morphology was assessed by immunohistochemistry and stereology. In the cortex, hypothyroidism reduced CI-linked respiration and CI abundance at 128 dGA and 142 dGA, respectively, and caused upregulation of PGC1α (regulator of mitochondrial biogenesis) and thyroid hormone receptor ß at 128 dGA and 142 dGA, respectively. In contrast, in the cerebellum, hypothyroidism reduced CI&II- and CII-linked respiration at 128 dGA, with no significant effect on the ETS complexes. In addition, cerebellar glucocorticoid hormone receptor and adenine nucleotide translocase (ANT1) were downregulated at 128 dGA and 142 dGA, respectively. These alterations in mitochondrial function were accompanied by reduced myelination. The findings demonstrate the importance of thyroid hormones in the prepartum maturation of cerebral mitochondria and have implications for the etiology and treatment of the neurodevelopmental abnormalities associated with human prematurity and congenital hypothyroidism.


Assuntos
Regulação da Expressão Gênica , Hipotireoidismo/complicações , Mitocôndrias/patologia , Doenças Mitocondriais/patologia , Fosforilação Oxidativa , Efeitos Tardios da Exposição Pré-Natal/patologia , Hormônios Tireóideos/deficiência , Animais , Circulação Cerebrovascular , Feminino , Mitocôndrias/metabolismo , Doenças Mitocondriais/etiologia , Doenças Mitocondriais/metabolismo , Gravidez , Ovinos
8.
Thyroid ; 31(6): 861-869, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33126831

RESUMO

Background: The fetal hypothalamic-pituitary-adrenal (HPA) axis plays a key role in the control of parturition and maturation of organ systems in preparation for birth. In hypothyroid fetuses, gestational length may be prolonged and maturational processes delayed. The extent to which the effects of thyroid hormone deficiency in utero on the timing of fetal maturation and parturition are mediated by changes to the structure and function of the fetal HPA axis is unknown. Methods: In twin sheep pregnancies where one fetus was thyroidectomized and the other sham-operated, this study investigated the effect of hypothyroidism on circulating concentrations of adrenocorticotrophic hormone (ACTH) and cortisol, and the structure and secretory capacity of the anterior pituitary and adrenal glands. The relative population of pituitary corticotrophs and the masses of the adrenal zones were assessed by immunohistochemical and stereological techniques. Adrenal mRNA abundances of key steroidogenic enzymes and growth factors were examined by quantitative polymerase chain reaction. Results: Hypothyroidism in utero reduced plasma concentrations of ACTH and cortisol. In thyroid-deficient fetuses, the mass of corticotrophs in the anterior pituitary gland was unexpectedly increased, while the mass of the zona fasciculata and its proportion of the adrenal gland were decreased. These structural changes were associated with lower adrenocortical mRNA abundances of insulin-like growth factor (IGF)-I and its receptor, and key steroidogenic enzymes responsible for glucocorticoid synthesis. The relative mass of the adrenal medulla and its proportion of the adrenal gland were increased by thyroid hormone deficiency in utero, without any change in expression of phenylethanolamine N-methyltransferase or the IGF system. Conclusions: Thyroid hormones are important regulators of the structure and secretory capacity of the pituitary-adrenal axis before birth. In hypothyroid fetuses, low plasma cortisol may be due to impaired adrenocortical growth and steroidogenic enzyme expression, secondary to low circulating ACTH concentration. Greater corticotroph population in the anterior pituitary gland of the hypothyroid fetus indicates compensatory cell proliferation and that there may be abnormal corticotroph capacity for ACTH synthesis and/or impaired hypothalamic input. Suppression of the development of the fetal HPA axis by thyroid hormone deficiency may contribute to the delay in fetal maturation and delivery observed in hypothyroid offspring.


Assuntos
Corticosteroides/metabolismo , Glândulas Suprarrenais/metabolismo , Hormônio Adrenocorticotrópico/metabolismo , Hipotireoidismo Congênito/metabolismo , Corticotrofos/metabolismo , Desenvolvimento Fetal/fisiologia , Doenças Fetais/metabolismo , Tireoidectomia , Glândulas Suprarrenais/patologia , Medula Suprarrenal/metabolismo , Medula Suprarrenal/patologia , Animais , Contagem de Células , Proliferação de Células , Hipotireoidismo Congênito/patologia , Corticotrofos/patologia , Doenças Fetais/patologia , Maturidade dos Órgãos Fetais , Hidrocortisona/sangue , Sistema Hipotálamo-Hipofisário/metabolismo , Fator de Crescimento Insulin-Like I/genética , Sistema Hipófise-Suprarrenal/metabolismo , RNA Mensageiro/metabolismo , Receptor IGF Tipo 1/genética , Ovinos , Tiroxina/deficiência , Tiroxina/metabolismo , Tri-Iodotironina/deficiência , Tri-Iodotironina/metabolismo , Zona Fasciculada/metabolismo , Zona Fasciculada/patologia
9.
J Dev Orig Health Dis ; 12(2): 309-318, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-32489168

RESUMO

Prenatal glucocorticoid overexposure has been shown to programme adult cardiovascular function in a range of species, but much less is known about the long-term effects of neonatal glucocorticoid overexposure. In horses, prenatal maturation of the hypothalamus-pituitary-adrenal axis and the normal prepartum surge in fetal cortisol occur late in gestation compared to other precocious species. Cortisol levels continue to rise in the hours after birth of full-term foals and increase further in the subsequent days in premature, dysmature and maladapted foals. Thus, this study examined the adult cardiovascular consequences of neonatal cortisol overexposure induced by adrenocorticotropic hormone administration to full-term male and female pony foals. After catheterisation at 2-3 years of age, basal arterial blood pressures (BP) and heart rate were measured together with the responses to phenylephrine (PE) and sodium nitroprusside (SNP). These data were used to assess cardiac baroreflex sensitivity. Neonatal cortisol overexposure reduced both the pressor and bradycardic responses to PE in the young adult males, but not females. It also enhanced the initial hypotensive response to SNP, slowed recovery of BP after infusion and reduced the gain of the cardiac baroreflex in the females, but not males. Basal diastolic pressure and cardiac baroreflex sensitivity also differed with sex, irrespective of neonatal treatment. The results show that there is a window of susceptibility for glucocorticoid programming during the immediate neonatal period that alters cardiovascular function in young adult horses in a sex-linked manner.


Assuntos
Sistema Cardiovascular/patologia , Nitroprussiato/toxicidade , Fenilefrina/toxicidade , Animais , Animais Recém-Nascidos , Sistema Cardiovascular/efeitos dos fármacos , Feminino , Cavalos , Masculino , Fatores Sexuais , Vasoconstritores/toxicidade , Vasodilatadores/toxicidade
10.
Thyroid ; 30(6): 794-805, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32070265

RESUMO

Background: Development of adipose tissue before birth is essential for energy storage and thermoregulation in the neonate and for cardiometabolic health in later life. Thyroid hormones are important regulators of growth and maturation in fetal tissues. Offspring hypothyroid in utero are poorly adapted to regulate body temperature at birth and are at risk of becoming obese and insulin resistant in childhood. The mechanisms by which thyroid hormones regulate the growth and development of adipose tissue in the fetus, however, are unclear. Methods: This study examined the structure, transcriptome, and protein expression of perirenal adipose tissue (PAT) in a fetal sheep model of thyroid hormone deficiency during late gestation. Proportions of unilocular (UL) (white) and multilocular (ML) (brown) adipocytes, and UL adipocyte size, were assessed by histological and stereological techniques. Changes to the adipose transcriptome were investigated by RNA sequencing and bioinformatic analysis, and proteins of interest were quantified by Western blotting. Results: Hypothyroidism in utero resulted in elevated plasma insulin and leptin concentrations and overgrowth of PAT in the fetus, specifically due to hyperplasia and hypertrophy of UL adipocytes with no change in ML adipocyte mass. RNA sequencing and genomic analyses showed that thyroid deficiency affected 34% of the genes identified in fetal adipose tissue. Enriched Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) pathways were associated with adipogenic, metabolic, and thermoregulatory processes, insulin resistance, and a range of endocrine and adipocytokine signaling pathways. Adipose protein levels of signaling molecules, including phosphorylated S6-kinase (pS6K), glucose transporter isoform 4 (GLUT4), and peroxisome proliferator-activated receptor γ (PPARγ), were increased by fetal hypothyroidism. Fetal thyroid deficiency decreased uncoupling protein 1 (UCP1) protein and mRNA content, and UCP1 thermogenic capacity without any change in ML adipocyte mass. Conclusions: Growth and development of adipose tissue before birth is sensitive to thyroid hormone status in utero. Changes to the adipose transcriptome and phenotype observed in the hypothyroid fetus may have consequences for neonatal survival and the risk of obesity and metabolic dysfunction in later life.


Assuntos
Tecido Adiposo Marrom/metabolismo , Tecido Adiposo Branco/metabolismo , Hipotireoidismo Congênito/metabolismo , Termogênese/fisiologia , Animais , Modelos Animais de Doenças , Feminino , Insulina/sangue , Leptina/sangue , PPAR gama/metabolismo , Ovinos , Transdução de Sinais/fisiologia , Transcriptoma , Proteína Desacopladora 1/metabolismo
11.
13.
Am J Physiol Regul Integr Comp Physiol ; 314(6): R781-R790, 2018 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-29443548

RESUMO

Widespread expression of leptin and its receptor in developing cartilage and bone suggests that leptin may regulate bone growth and development in the fetus. Using microcomputed tomography, this study investigated the effects of exogenous leptin and leptin receptor antagonism on aspects of bone structure in the sheep fetus during late gestation. From 125 to 130 days of gestation (term ~145 days), chronically catheterized singleton sheep fetuses were infused intravenously for 5 days with either saline (0.9% saline, n = 13), recombinant ovine leptin at two doses (0.6 mg·kg-1·day-1 LEP1, n = 10 or 1.4 mg·kg-1·day-1 LEP2, n = 7), or recombinant superactive ovine leptin receptor antagonist (4.6 mg·kg-1·day-1 SOLA, n = 6). No significant differences in plasma insulin-like growth factor-I, osteocalcin, calcium, inorganic phosphate, or alkaline phosphatase were observed between treatment groups. Total femur midshaft diameter and metatarsal lumen diameter were narrower in male fetuses treated with exogenous leptin. In a fixed length of femur midshaft, total and bone volumes were reduced by the higher dose of leptin; nonbone space volume was lower in both groups of leptin-treated fetuses. Leptin infusion caused increments in femur porosity and connectivity density, and vertebral trabecular thickness. Leptin receptor antagonism decreased trabecular spacing and increased trabecular number, degree of anisotrophy, and connectivity density in the lumbar vertebrae. The increase in vertebral porosity observed following leptin receptor antagonism was greater in the malecompared with female, fetuses. Therefore, leptin may have a role in the growth and development of the fetal skeleton, dependent on the concentration of leptin, sex of the fetus, and bone type examined.


Assuntos
Osso e Ossos/efeitos dos fármacos , Feto/efeitos dos fármacos , Leptina/farmacologia , Receptores para Leptina/antagonistas & inibidores , Animais , Desenvolvimento Ósseo/efeitos dos fármacos , Osso e Ossos/anatomia & histologia , Relação Dose-Resposta a Droga , Feminino , Fêmur/anatomia & histologia , Fêmur/crescimento & desenvolvimento , Desenvolvimento Fetal/efeitos dos fármacos , Idade Gestacional , Fator de Crescimento Insulin-Like I/análise , Masculino , Osteocalcina/sangue , Porosidade , Gravidez , Caracteres Sexuais , Ovinos , Tomografia Computadorizada por Raios X
14.
Physiol Rep ; 5(11)2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28611150

RESUMO

Stress during pregnancy is associated with metabolic dysfunction in the adult offspring in human and other animals. However, little is known about the metabolic effects of pregnancy stress on the mothers and fetuses during pregnancy itself. This study aimed to determine the consequences of the common experimental procedures of injection and single housing in pregnant rats on fetal and maternal hepatic glucogenic capacities. On day (D) 20 of pregnancy, feto-placental weights and the glycogen content and activities of phosphoenolpyruvate carboxykinase (PEPCK) and glucose-6-phosphatase (G6Pase) of fetal and maternal liver were measured in rats pair or single housed from D1 with or without saline injection from D15 to D19. Housing and saline injection both affected hepatic glucogenic capacity. In maternal liver, saline injection but not housing reduced glycogen content and raised G6Pase activity, whereas housing but not treatment increased PEPCK activity. In fetuses, housing and injection interacted in regulating PEPCK activity and reducing hepatic glycogen content and placental weight. Body weight was decreased and hepatic G6Pase increased by injection but not housing in the fetuses. Single-housed dams ate less than those pair-housed near term while saline injection elevated maternal plasma corticosterone concentrations. Thus, single housing and saline injection are both stresses during rat pregnancy that alter feto-placental weight and hepatic glucogenic capacity of the fetuses and dams near term. Routine experimental procedures per se may, therefore, have consequences for offspring hepatic phenotype as well as modifying the outcomes of dietary and other environmental challenges during pregnancy.


Assuntos
Fígado/metabolismo , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Estresse Psicológico/metabolismo , Animais , Glicemia/metabolismo , Feminino , Feto , Glucose/metabolismo , Glucose-6-Fosfatase/metabolismo , Glicogênio/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Fosfoenolpiruvato Carboxiquinase (GTP)/metabolismo , Gravidez , Ratos , Ratos Wistar
15.
J Physiol ; 595(11): 3331-3343, 2017 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-28144955

RESUMO

KEY POINTS: Thyroid hormones are important regulators of growth and maturation before birth, although the extent to which their actions are mediated by insulin and the development of pancreatic beta cell mass is unknown. Hypothyroidism in fetal sheep induced by removal of the thyroid gland caused asymmetric organ growth, increased pancreatic beta cell mass and proliferation, and was associated with increased circulating concentrations of insulin and leptin. In isolated fetal sheep islets studied in vitro, thyroid hormones inhibited beta cell proliferation in a dose-dependent manner, while high concentrations of insulin and leptin stimulated proliferation. The developing pancreatic beta cell is therefore sensitive to thyroid hormone, insulin and leptin before birth, with possible consequences for pancreatic function in fetal and later life. The findings of this study highlight the importance of thyroid hormones during pregnancy for normal development of the fetal pancreas. ABSTRACT: Development of pancreatic beta cell mass before birth is essential for normal growth of the fetus and for long-term control of carbohydrate metabolism in postnatal life. Thyroid hormones are also important regulators of fetal growth, and the present study tested the hypotheses that thyroid hormones promote beta cell proliferation in the fetal ovine pancreatic islets, and that growth retardation in hypothyroid fetal sheep is associated with reductions in pancreatic beta cell mass and circulating insulin concentration in utero. Organ growth and pancreatic islet cell proliferation and mass were examined in sheep fetuses following removal of the thyroid gland in utero. The effects of triiodothyronine (T3 ), insulin and leptin on beta cell proliferation rates were determined in isolated fetal ovine pancreatic islets in vitro. Hypothyroidism in the sheep fetus resulted in an asymmetric pattern of organ growth, pancreatic beta cell hyperplasia, and elevated plasma insulin and leptin concentrations. In pancreatic islets isolated from intact fetal sheep, beta cell proliferation in vitro was reduced by T3 in a dose-dependent manner and increased by insulin at high concentrations only. Leptin induced a bimodal response whereby beta cell proliferation was suppressed at the lowest, and increased at the highest, concentrations. Therefore, proliferation of beta cells isolated from the ovine fetal pancreas is sensitive to physiological concentrations of T3 , insulin and leptin. Alterations in these hormones may be responsible for the increased beta cell proliferation and mass observed in the hypothyroid sheep fetus and may have consequences for pancreatic function in later life.


Assuntos
Proliferação de Células , Doenças Fetais/fisiopatologia , Hiperinsulinismo/fisiopatologia , Hipotireoidismo/fisiopatologia , Células Secretoras de Insulina/fisiologia , Animais , Células Cultivadas , Feminino , Doenças Fetais/sangue , Hiperinsulinismo/sangue , Hiperinsulinismo/etiologia , Hipotireoidismo/sangue , Hipotireoidismo/complicações , Insulina/sangue , Células Secretoras de Insulina/efeitos dos fármacos , Leptina/sangue , Gravidez , Ovinos , Tri-Iodotironina/farmacologia
16.
Endocrinology ; 157(1): 395-404, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26479186

RESUMO

In human and ovine fetuses, glucocorticoids stimulate leptin secretion, although the extent to which leptin mediates the maturational effects of glucocorticoids on pulmonary development is unclear. This study investigated the effects of leptin administration on indices of lung structure and function before birth. Chronically catheterized singleton sheep fetuses were infused iv for 5 days with either saline or recombinant ovine leptin (0.5 mg/kg · d leptin (LEP), 0.5 LEP or 1.0 mg/kg · d, 1.0 LEP) from 125 days of gestation (term ∼145 d). Over the infusion, leptin administration increased plasma leptin, but not cortisol, concentrations. On the fifth day of infusion, 0.5 LEP reduced alveolar wall thickness and increased the volume at closing pressure of the pressure-volume deflation curve, interalveolar septal elastin content, secondary septal crest density, and the mRNA abundance of the leptin receptor (Ob-R) and surfactant protein (SP) B. Neither treatment influenced static lung compliance, maximal lung volume at 40 cmH2O, lung compartment volumes, alveolar surface area, pulmonary glycogen, protein content of the long form signaling Ob-Rb or phosphorylated signal transducers and activators of transcription-3, or mRNA levels of SP-A, C, or D, elastin, vascular endothelial growth factor-A, the vascular endothelial growth factor receptor 2, angiotensin-converting enzyme, peroxisome proliferator-activated receptor γ, or parathyroid hormone-related peptide. Leptin administration in the ovine fetus during late gestation promotes aspects of lung maturation, including up-regulation of SP-B.


Assuntos
Feto/efeitos dos fármacos , Leptina/farmacologia , Pulmão/efeitos dos fármacos , Organogênese/efeitos dos fármacos , Animais , Relação Dose-Resposta a Droga , Feminino , Terapias Fetais , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Infusões Intravenosas , Leptina/administração & dosagem , Leptina/genética , Leptina/farmacocinética , Pulmão/embriologia , Pulmão/metabolismo , Pulmão/fisiologia , Complacência Pulmonar/efeitos dos fármacos , Gravidez , Proteína B Associada a Surfactante Pulmonar/agonistas , Proteína B Associada a Surfactante Pulmonar/genética , Proteína B Associada a Surfactante Pulmonar/metabolismo , RNA Mensageiro/metabolismo , Distribuição Aleatória , Receptores para Leptina/agonistas , Receptores para Leptina/genética , Receptores para Leptina/metabolismo , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/sangue , Proteínas Recombinantes/farmacocinética , Proteínas Recombinantes/farmacologia , Ovinos , Capacidade Pulmonar Total/efeitos dos fármacos
17.
PLoS One ; 10(8): e0136115, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26287800

RESUMO

The effects of endogenous and synthetic glucocorticoids on fetal lung maturation are well-established, although the role of leptin in lung development before birth is unclear. This study examined mRNA and protein levels of the signalling long-form leptin receptor (Ob-Rb) in fetal ovine lungs towards term, and after experimental manipulation of glucocorticoid levels in utero by fetal cortisol infusion or maternal dexamethasone treatment. In fetal ovine lungs, Ob-Rb protein was localised to bronchiolar epithelium, bronchial cartilage, vascular endothelium, alveolar macrophages and type II pneumocytes. Pulmonary Ob-Rb mRNA abundance increased between 100 (0.69 fractional gestational age) and 144 days (0.99) of gestation, and by 2-4-fold in response to fetal cortisol infusion and maternal dexamethasone treatment. In contrast, pulmonary Ob-Rb protein levels decreased near term and were halved by glucocorticoid treatment, without any significant change in phosphorylated signal transducer and activator of transcription-3 (pSTAT3) at Ser727, total STAT3 or the pulmonary pSTAT3:STAT3 ratio. Leptin mRNA was undetectable in fetal ovine lungs at the gestational ages studied. These findings demonstrate differential control of pulmonary Ob-Rb transcript abundance and protein translation, and/or post-translational processing, by glucocorticoids in utero. Localisation of Ob-Rb in the fetal ovine lungs, including alveolar type II pneumocytes, suggests a role for leptin signalling in the control of lung growth and maturation before birth.


Assuntos
Pulmão/embriologia , Pulmão/metabolismo , Receptores para Leptina/genética , Receptores para Leptina/metabolismo , Células Epiteliais Alveolares/metabolismo , Animais , Dexametasona/farmacologia , Feminino , Feto/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Glucocorticoides/farmacologia , Hidrocortisona/sangue , Hidrocortisona/farmacologia , Pulmão/efeitos dos fármacos , Fosforilação , Gravidez , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fator de Transcrição STAT3/química , Fator de Transcrição STAT3/metabolismo , Carneiro Doméstico , Transdução de Sinais
18.
Exp Physiol ; 100(12): 1477-87, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26040783

RESUMO

NEW FINDINGS: What is the topic of this review? This review discusses the role of the glucocorticoids as regulatory signals during intrauterine development. It examines the functional significance of these hormones as maturational, environmental and programming signals in determining offspring phenotype. What advances does it highlight? It focuses on the extensive nature of the regulatory actions of these hormones. It highlights the emerging data that these actions are mediated, in part, by the placenta, other endocrine systems and epigenetic modifications of the genome. Glucocorticoids are important regulatory signals during intrauterine development. They act as maturational, environmental and programming signals that modify the developing phenotype to optimize offspring viability and fitness. They affect development of a wide range of fetal tissues by inducing changes in cellular expression of structural, transport and signalling proteins, which have widespread functional consequences at the whole organ and systems levels. Glucocorticoids, therefore, activate many of the physiological systems that have little function in utero but are vital at birth to replace the respiratory, nutritive and excretory functions previously carried out by the placenta. However, by switching tissues from accretion to differentiation, early glucocorticoid overexposure in response to adverse conditions can programme fetal development with longer term physiological consequences for the adult offspring, which can extend to the next generation. The developmental effects of the glucocorticoids can be direct on fetal tissues with glucocorticoid receptors or mediated by changes in placental function or other endocrine systems. At the molecular level, glucocorticoids can act directly on gene transcription via their receptors or indirectly by epigenetic modifications of the genome. In this review, we examine the role and functional significance of glucocorticoids as regulatory signals during intrauterine development and discuss the mechanisms by which they act in utero to alter the developing epigenome and ensuing phenotype.


Assuntos
Desenvolvimento Fetal/fisiologia , Glucocorticoides/metabolismo , Transdução de Sinais/fisiologia , Útero/metabolismo , Útero/fisiologia , Animais , Feminino , Humanos , Gravidez , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia
19.
Endocrinology ; 156(8): 3038-46, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26039155

RESUMO

Antenatal synthetic glucocorticoids promote fetal maturation in pregnant women at risk of preterm delivery and their mechanism of action may involve other endocrine systems. This study investigated the effect of maternal dexamethasone treatment, at clinically relevant doses, on components of the renin-angiotensin system (RAS) in the pregnant ewe and fetus. From 125 days of gestation (term, 145 ± 2 d), 10 ewes carrying single fetuses of mixed sex (3 female, 7 male) were injected twice im, at 10-11 pm, with dexamethasone (2 × 12 mg, n = 5) or saline (n = 5) at 24-hour intervals. At 10 hours after the second injection, maternal dexamethasone treatment increased angiotensin-converting enzyme (ACE) mRNA levels in the fetal lungs, kidneys, and heart and ACE concentration in the circulation and lungs, but not kidneys, of the fetuses. Fetal cardiac mRNA abundance of angiotensin II (AII) type 2 receptor decreased after maternal dexamethasone treatment. Between the two groups of fetuses, there were no significant differences in plasma angiotensinogen or renin concentrations; in transcript levels of renal renin, or AII type 1 or 2 receptors in the lungs and kidneys; or in pulmonary, renal or cardiac protein content of the AII receptors. In the pregnant ewes, dexamethasone administration increased pulmonary ACE and plasma angiotensinogen, and decreased plasma renin, concentrations. Some of the effects of dexamethasone treatment on the maternal and fetal RAS were associated with altered insulin and thyroid hormone activity. Changes in the local and circulating RAS induced by dexamethasone exposure in utero may contribute to the maturational and tissue-specific actions of antenatal glucocorticoid treatment.


Assuntos
Dexametasona/farmacologia , Feto/efeitos dos fármacos , Exposição Materna , Sistema Renina-Angiotensina/efeitos dos fármacos , Angiotensinogênio/sangue , Animais , Feminino , Desenvolvimento Fetal/efeitos dos fármacos , Feto/metabolismo , Masculino , Peptidil Dipeptidase A/sangue , Gravidez/sangue , Gravidez/efeitos dos fármacos , Gravidez/metabolismo , Receptores de Angiotensina/metabolismo , Renina/sangue , Sistema Renina-Angiotensina/fisiologia , Ovinos
20.
Artigo em Inglês | MEDLINE | ID: mdl-23887107

RESUMO

Hormones are both growth stimulatory and growth inhibitory in utero. They act as environmental and maturational signals in regulating tissue accretion and differentiation during late gestation. They ensure that fetal development is appropriate for the nutrient supply and is optimal for neonatal survival. Growth-stimulatory hormones, such as insulin, the insulin-like growth factors and the thyroid hormones, have anabolic effects on fetal metabolism and increase cellular nutrient uptake and energy production for tissue accretion. Thyroid hormones also have specific effects on tissue differentiation at key developmental milestones. Similarly, leptin appears to affect development of specific fetal tissues and may counterbalance the maturational actions of other hormones near term. Glucocorticoids inhibit growth in utero but are essential for prepartum tissue differentiation in preparation for delivery. They also affect fetal bioavailability of most of the other growth-regulatory hormones. In addition, many of these hormones alter the placental capacity to supply nutrients for fetal growth. In producing a fetoplacental epigenome specific to the prevailing intrauterine environment, hormones interact to produce phenotypical diversity with potential health consequences long after birth.


Assuntos
Sistema Endócrino/fisiologia , Desenvolvimento Fetal/fisiologia , Feto/metabolismo , Hormônios/metabolismo , Somatomedinas/metabolismo , Feminino , Humanos , Placenta , Gravidez
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...