Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
FASEB J ; 35(5): e21591, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33891344

RESUMEN

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.


Asunto(s)
Regulación de la Expresión Génica , Hipotiroidismo/complicaciones , Mitocondrias/patología , Enfermedades Mitocondriales/patología , Fosforilación Oxidativa , Efectos Tardíos de la Exposición Prenatal/patología , Hormonas Tiroideas/deficiencia , Animales , Circulación Cerebrovascular , Femenino , Mitocondrias/metabolismo , Enfermedades Mitocondriales/etiología , Enfermedades Mitocondriales/metabolismo , Embarazo , Ovinos
2.
Am J Physiol Regul Integr Comp Physiol ; 314(6): R781-R790, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29443548

RESUMEN

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.


Asunto(s)
Huesos/efectos de los fármacos , Feto/efectos de los fármacos , Leptina/farmacología , Receptores de Leptina/antagonistas & inhibidores , Animales , Desarrollo Óseo/efectos de los fármacos , Huesos/anatomía & histología , Relación Dosis-Respuesta a Droga , Femenino , Fémur/anatomía & histología , Fémur/crecimiento & desarrollo , Desarrollo Fetal/efectos de los fármacos , Edad Gestacional , Factor I del Crecimiento Similar a la Insulina/análisis , Masculino , Osteocalcina/sangre , Porosidad , Embarazo , Caracteres Sexuales , Ovinos , Tomografía Computarizada por Rayos X
3.
J Physiol ; 595(11): 3331-3343, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28144955

RESUMEN

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.


Asunto(s)
Proliferación Celular , Enfermedades Fetales/fisiopatología , Hiperinsulinismo/fisiopatología , Hipotiroidismo/fisiopatología , Células Secretoras de Insulina/fisiología , Animales , Células Cultivadas , Femenino , Enfermedades Fetales/sangre , Hiperinsulinismo/sangre , Hiperinsulinismo/etiología , Hipotiroidismo/sangre , Hipotiroidismo/complicaciones , Insulina/sangre , Células Secretoras de Insulina/efectos de los fármacos , Leptina/sangre , Embarazo , Ovinos , Triyodotironina/farmacología
4.
Exp Physiol ; 100(12): 1477-87, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26040783

RESUMEN

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.


Asunto(s)
Desarrollo Fetal/fisiología , Glucocorticoides/metabolismo , Transducción de Señal/fisiología , Útero/metabolismo , Útero/fisiología , Animales , Femenino , Humanos , Embarazo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Efectos Tardíos de la Exposición Prenatal/fisiopatología
5.
J Physiol ; 597(16): 4125-4126, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31271219
7.
Curr Opin Clin Nutr Metab Care ; 16(3): 298-309, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23340010

RESUMEN

PURPOSE OF REVIEW: Size at birth is critical in determining life expectancy with both small and large neonates at risk of shortened life spans. This review examines the hormonal and nutritional drivers of intrauterine growth with emphasis on the role of foetal hormones as nutritional signals in utero. RECENT FINDINGS: Nutrients drive intrauterine growth by providing substrate for tissue accretion, whereas hormones regulate nutrient distribution between foetal oxidative metabolism and mass accumulation. The main hormonal drivers of intrauterine growth are insulin, insulin-like growth factors and thyroid hormones. Together with leptin and cortisol, these hormones control cellular nutrient uptake and the balance between accretion and differentiation in regulating tissue growth. They also act indirectly via the placenta to alter the materno-foetal supply of nutrients and oxygen. By responding to nutrient and oxygen availability, foetal hormones optimize the survival and growth of the foetus with respect to its genetic potential, particularly during adverse conditions. However, changes in the intrauterine growth of individual tissues may alter their function permanently. SUMMARY: In both normal and compromised pregnancies, intrauterine growth is determined by multiple hormonal and nutritional drivers which interact to produce a specific pattern of intrauterine development with potential lifelong consequences for health.


Asunto(s)
Desarrollo Fetal , Feto/metabolismo , Fenómenos Fisiologicos Nutricionales Maternos , Estado Nutricional , Hormonas Tiroideas/metabolismo , Disponibilidad Biológica , Femenino , Feto/embriología , Humanos , Insulina/metabolismo , Leptina/metabolismo , Oxígeno/administración & dosificación , Oxígeno/farmacocinética , Placenta/embriología , Placenta/metabolismo , Placentación , Embarazo , Somatomedinas/metabolismo
8.
J Dev Orig Health Dis ; 14(1): 77-87, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-35822505

RESUMEN

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.


Asunto(s)
Hidrocortisona , Mitocondrias , Embarazo , Femenino , Animales , Ovinos , Hidrocortisona/metabolismo , Mitocondrias/metabolismo , Músculo Esquelético/metabolismo , Parto , Fosforilación Oxidativa
9.
Front Physiol ; 14: 1124938, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36935746

RESUMEN

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.

10.
Domest Anim Endocrinol ; 78: 106657, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34525421

RESUMEN

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.


Asunto(s)
Sistema Endocrino , Feto , Animales , Sistema Endocrino/fisiología , Femenino , Hidrocortisona/metabolismo , Intercambio Materno-Fetal , Embarazo , Ovinos , Tiroxina
11.
Nutrients ; 14(11)2022 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-35684104

RESUMEN

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.


Asunto(s)
Glucocorticoides , Placenta , Animales , Femenino , Desarrollo Fetal , Feto/metabolismo , Glucocorticoides/metabolismo , Parto , Placenta/metabolismo , Embarazo , Ovinos
12.
Biomolecules ; 12(6)2022 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-35740893

RESUMEN

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.


Asunto(s)
Glucocorticoides , Hidrocortisona , Animales , Encéfalo/metabolismo , Femenino , Feto/metabolismo , Edad Gestacional , Glucocorticoides/metabolismo , Glucocorticoides/farmacología , Humanos , Hidrocortisona/farmacología , Mitocondrias/metabolismo , Fosforilación Oxidativa , Embarazo , Ovinos
13.
Am J Physiol Gastrointest Liver Physiol ; 300(2): G253-63, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21088234

RESUMEN

The inflammasome is a multiprotein complex whose formation is triggered when a NOD-like receptor binds a pathogen ligand, resulting in activated caspase-1, which converts certain interleukins (IL-1ß, IL-18, and IL-33) to their active forms. There is currently no information on regulation of this system around the time of birth. We employed transcript profiling of fetal rat intestinal and lung RNA at embryonic days 16 (E16) and 20 (E20) with out-of-sample validation using quantitative RT-PCR. Transcript profiling and quantitative RT-PCR demonstrated that transcripts of core components of the NOD-like receptor Nlrp6 inflammasome (Nlrp6, Pycard, Caspase-1) and one of its substrates, IL-18, were increased at E20 compared with E16 in fetal intestine and not lung. Immunohistochemistry demonstrated increased Pycard in intestinal epithelium. Western blotting demonstrated that IL-18 was undetectable at E16, clearly detectable at E20 in its inactive form, and detectable postnatally in both its inactive and active form. Dramatic upregulation of IL-18 was also observed in the fetal sheep jejunum in late gestation (P = 0.006). Transcription factor binding analysis of the rat array data revealed an overrepresentation of nuclear transcription factor binding sites peroxisome proliferator-activated receptor γ (PPAR-γ) and retinoid X receptor-α and chicken ovalbumin upstream promoter transcription factor 1 in the region 1,000 bp upstream of the transcription start site. Rosiglitazone, a PPAR-γ agonist, more than doubled levels of NLRP6 mRNA in human intestinal epithelial (Caco2) cells. These observations provide the first evidence, to our knowledge, linking activity of PPAR-γ to expression of a NOD-like receptor and adds to a growing body of evidence linking pattern recognition receptors of the innate immune system and intestinal colonization.


Asunto(s)
Embrión de Mamíferos/metabolismo , Desarrollo Embrionario , Inflamasomas/metabolismo , Interleucina-8/metabolismo , Intestinos/embriología , Pulmón/embriología , Receptores de Angiotensina/metabolismo , Receptores de Vasopresinas/metabolismo , Animales , Sitios de Unión , Línea Celular Tumoral , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica , Edad Gestacional , Humanos , Inmunidad Innata/genética , Inmunohistoquímica , Inflamasomas/genética , Mucosa Intestinal/embriología , Análisis por Micromatrices , PPAR gamma/metabolismo , ARN Mensajero/metabolismo , Ratas/embriología , Ratas Wistar , Receptores de Angiotensina/genética , Receptores de Vasopresinas/genética , Reproducibilidad de los Resultados , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Ovinos/embriología , Factores de Transcripción/metabolismo
14.
J Endocrinol ; 252(1): 71-80, 2021 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-34708692

RESUMEN

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.


Asunto(s)
Desarrollo Óseo/fisiología , Desarrollo Fetal/fisiología , Insulina/deficiencia , Enfermedades Pancreáticas/embriología , Animales , Huesos/metabolismo , Femenino , Insulina/metabolismo , Páncreas/metabolismo , Páncreas/patología , Páncreas/cirugía , Pancreatectomía , Enfermedades Pancreáticas/complicaciones , Enfermedades Pancreáticas/metabolismo , Enfermedades Pancreáticas/fisiopatología , Embarazo , Ovinos
15.
J Dev Orig Health Dis ; 12(2): 309-318, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-32489168

RESUMEN

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.


Asunto(s)
Sistema Cardiovascular/patología , Nitroprusiato/toxicidad , Fenilefrina/toxicidad , Animales , Animales Recién Nacidos , Sistema Cardiovascular/efectos de los fármacos , Femenino , Caballos , Masculino , Factores Sexuales , Vasoconstrictores/toxicidad , Vasodilatadores/toxicidad
16.
Thyroid ; 31(6): 861-869, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33126831

RESUMEN

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.


Asunto(s)
Corticoesteroides/metabolismo , Glándulas Suprarrenales/metabolismo , Hormona Adrenocorticotrópica/metabolismo , Hipotiroidismo Congénito/metabolismo , Corticotrofos/metabolismo , Desarrollo Fetal/fisiología , Enfermedades Fetales/metabolismo , Tiroidectomía , Glándulas Suprarrenales/patología , Médula Suprarrenal/metabolismo , Médula Suprarrenal/patología , Animales , Recuento de Células , Proliferación Celular , Hipotiroidismo Congénito/patología , Corticotrofos/patología , Enfermedades Fetales/patología , Madurez de los Órganos Fetales , Hidrocortisona/sangre , Sistema Hipotálamo-Hipofisario/metabolismo , Factor I del Crecimiento Similar a la Insulina/genética , Sistema Hipófiso-Suprarrenal/metabolismo , ARN Mensajero/metabolismo , Receptor IGF Tipo 1/genética , Ovinos , Tiroxina/deficiencia , Tiroxina/metabolismo , Triyodotironina/deficiencia , Triyodotironina/metabolismo , Zona Fascicular/metabolismo , Zona Fascicular/patología
17.
Thyroid ; 30(6): 794-805, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32070265

RESUMEN

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.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Hipotiroidismo Congénito/metabolismo , Termogénesis/fisiología , Animales , Modelos Animales de Enfermedad , Femenino , Insulina/sangre , Leptina/sangre , PPAR gamma/metabolismo , Ovinos , Transducción de Señal/fisiología , Transcriptoma , Proteína Desacopladora 1/metabolismo
18.
J Physiol ; 587(Pt 6): 1145-52, 2009 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-19188249

RESUMEN

In adult animals, leptin is an adipose-derived hormone that is important primarily in the regulation of energy balance during short- and long-term changes in nutritional state. Expression of leptin and its receptors is widespread in fetal and placental tissues, although the role of leptin as a nutritional signal in utero is unclear. Before birth, leptin concentration correlates with several indices of fetal growth, and may be an endocrine marker of fetal size and energy stores in the control of metabolism and maturation of fetal tissues. In addition, leptin synthesis and plasma concentration can be modified by insulin, glucocorticoids, thyroid hormones and oxygen availability in utero, and therefore, leptin may be part of the hormonal response to changes in the intrauterine environment. Evidence is emerging to show that leptin has actions before birth that are tissue-specific and may occur in critical periods of development. Some of these actions are involved in the growth and development of the fetus and others have long-term consequences for the control of energy balance in adult life.


Asunto(s)
Metabolismo Energético/fisiología , Desarrollo Fetal/fisiología , Feto/fisiología , Leptina/fisiología , Fenómenos Fisiológicos de la Nutrición , Animales , Femenino , Humanos , Placenta/metabolismo , Embarazo , Complicaciones del Embarazo/metabolismo
19.
Exp Physiol ; 94(6): 607-25, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19251980

RESUMEN

In mammals, including man, epidemiological and experimental studies have shown that a range of environmental factors acting during critical periods of early development can alter adult phenotype. Hormones have an important role in these epigenetic modifications and can signal the type, severity and duration of the environmental cue to the developing feto-placental tissues. They affect development of these tissues both directly and indirectly by changes in placental phenotype. They act to alter gene expression, hence the protein abundance in a wide range of different tissues, which has functional consequences for many physiological systems both before and after birth. By producing an epigenome specific to the prevailing condition in utero, hormones act as epigenetic signals in developmental programming, with important implications for adult health and disease. This review examines the role of hormones as epigenetic signals by considering their responses to environmental cues, their effects on phenotypical development and the molecular mechanisms by which they programme feto-placental development, with particular emphasis on the glucocorticoids.


Asunto(s)
Epigénesis Genética , Desarrollo Fetal/genética , Desarrollo Fetal/fisiología , Hormonas/fisiología , Animales , Femenino , Glucocorticoides/fisiología , Humanos , Masculino , Modelos Biológicos , Fenotipo , Embarazo , Transducción de Señal
20.
Exp Physiol ; 94(10): 1079-87, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19542188

RESUMEN

Developmental and glucocorticoid-induced changes in tissue glycogen content occur in the fetus near term coincident with an increase in plasma triiodothyronine (T(3)), although the role of thyroid hormones in mediating these changes is unknown. This study investigated glycogen content in the liver, heart and skeletal muscle of sheep fetuses after experimental manipulation of thyroid hormone concentration in utero by T(3) infusion and fetal thyroidectomy (TX). At 130 days of gestation (term 145 +/- 2 days), hepatic glycogen was greater, and muscle glycogen was lower, in the TX fetuses than in the intact fetuses. However, between 130 and 144 days of gestation the normal increment in hepatic glycogen, and decrement in cardiac glycogen, seen in intact fetuses was abolished when the prepartum rise in T(3), but not cortisol, was prevented by TX. At 144 days of gestation, hepatic glycogen was lower, and cardiac glycogen was higher, in the TX compared with intact fetuses. In intact fetuses at 130 days of gestation, 5 days of intravenous T(3) infusion (8-12 microg kg(-1) day(-1)) caused a small but significant increase in hepatic glycogen, although the concentration achieved was not as great as that observed in intact fetuses infused with cortisol (2-3 mg kg(-1) day(-1)) for 5 days. Infusion of T(3) reduced cardiac glycogen to the level observed in mature fetuses near term and immature fetuses infused with cortisol for 5 days. Glycogen content in fetal skeletal muscle increased between 100 and 115 days of gestation, but was unaffected by cortisol or T(3) infusion. Therefore, thyroid hormones are important in the developmental control of hepatic and cardiac glycogen content in the ovine fetus near term and may mediate, in part, the maturational effects of cortisol.


Asunto(s)
Feto/metabolismo , Glucógeno/metabolismo , Hormonas Tiroideas/fisiología , Animales , Femenino , Desarrollo Fetal/fisiología , Edad Gestacional , Hidrocortisona/sangre , Glucógeno Hepático/metabolismo , Músculo Esquelético/metabolismo , Miocardio/química , Miocardio/metabolismo , Embarazo , Ovinos , Hormonas Tiroideas/metabolismo , Hormonas Tiroideas/farmacología , Tiroidectomía , Tiroxina/sangre , Triyodotironina/sangre , Triyodotironina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA