Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 376
Filtrar
1.
Br J Nutr ; 122(2): 141-151, 2019 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-31345278

RESUMEN

Before weaning, breast milk is the physiological form of neonatal nutrition, providing pups with all nutrient requirements. Maternal low-protein diet (LPD) during pregnancy and lactation induces adverse changes in key maternal organs, which have negative effects on pup development. We studied the effects of maternal LPD on liver weight, mammary gland (MG) cell differentiation, milk composition and production and pup development throughout lactation. We fed rats with control (C) or LPD (R) during pregnancy and lactation. At 7 d early, 14 d mid and 21 d late lactation stages, maternal biochemical parameters, body, liver and MG weights were analysed. MG cell differentiation was analysed by haematoxylin and eosin staining; milk nutrient composition and production were studied; pup body, liver and brain weights, hippocampal arachidonic acid (AA) and DHA were quantified. Results showed lower body and liver weights, minor MG cell differentiation and lower serum insulin and TAG in R compared with C. R milk contained less protein and higher AA at early and mid stages compared with C. R pup milk and fat intake were lower at all stages. R protein intake at early and mid stages and DHA intake at mid and late stages were lower compared with C. In R pups, lower body, liver and brain weights were associated with decreased hippocampal AA and DHA. We conclude that maternal LPD impairs liver and MG function and induces significant changes in maternal milk composition, pup milk intake and organ development.


Asunto(s)
Dieta con Restricción de Proteínas/efectos adversos , Lactancia/fisiología , Leche/química , Animales , Ácido Araquidónico/análisis , Peso Corporal , Encéfalo/crecimiento & desarrollo , Ácidos Docosahexaenoicos/análisis , Femenino , Hipocampo/química , Hígado/crecimiento & desarrollo , Glándulas Mamarias Animales/anatomía & histología , Glándulas Mamarias Animales/crecimiento & desarrollo , Fenómenos Fisiologicos Nutricionales Maternos , Tamaño de los Órganos , Embarazo , Ratas , Ratas Wistar
2.
Hippocampus ; 28(1): 18-30, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28843045

RESUMEN

Maternal nutritional challenges during fetal and neonatal development result in developmental programming of multiple offspring organ systems including brain maturation and function. A maternal low-protein diet during pregnancy and lactation impairs associative learning and motivation. We evaluated effects of a maternal low-protein diet during gestation and/or lactation on male offspring spatial learning and hippocampal neural structure. Control mothers (C) ate 20% casein and restricted mothers (R) 10% casein, providing four groups: CC, RR, CR, and RC (first letter pregnancy, second lactation diet). We evaluated the behavior of young adult male offspring around postnatal day 110. Corticosterone and ACTH were measured. Males were tested for 2 days in the Morris water maze (MWM). Stratum lucidum mossy fiber (MF) area, total and spine type in basal dendrites of stratum oriens in the hippocampal CA3 field were measured. Corticosterone and ACTH were higher in RR vs. CC. In the MWM acquisition test CC offspring required two, RC three, and CR seven sessions to learn the maze. RR did not learn in eight trials. In a retention test 24 h later, RR, CR, and RC spent more time locating the platform and performed fewer target zone entries than CC. RR and RC offspring spent less time in the target zone than CC. MF area, total, and thin spines were lower in RR, CR, and RC than CC. Mushroom spines were lower in RR and RC than CC. Stubby spines were higher in RR, CR, and RC than CC. We conclude that maternal low-protein diet impairs spatial acquisition and memory retention in male offspring, and that alterations in hippocampal presynaptic (MF), postsynaptic (spines) elements and higher glucocorticoid levels are potential mechanisms to explain these learning and memory deficits.


Asunto(s)
Dieta con Restricción de Proteínas/efectos adversos , Hipocampo/crecimiento & desarrollo , Hipocampo/fisiopatología , Discapacidades para el Aprendizaje/fisiopatología , Trastornos de la Memoria/fisiopatología , Fenómenos Fisiologicos de la Nutrición Prenatal , Animales , Modelos Animales de Enfermedad , Femenino , Hipocampo/patología , Lactancia , Discapacidades para el Aprendizaje/etiología , Discapacidades para el Aprendizaje/patología , Masculino , Desnutrición/patología , Desnutrición/fisiopatología , Desnutrición/psicología , Trastornos de la Memoria/etiología , Trastornos de la Memoria/patología , Neuronas/patología , Embarazo , Distribución Aleatoria , Ratas Wistar , Aprendizaje Espacial/fisiología , Memoria Espacial/fisiología
3.
Int J Obes (Lond) ; 41(8): 1299-1302, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28337030

RESUMEN

Developmental programming studies indicate that glucocorticoids modify fetal development. We hypothesized that administration of the synthetic glucocorticoid (sGC) betamethasone to pregnant baboons at doses and stages of fetal life equivalent to human obstetric practice to decrease premature offspring morbidity and mortality, programs lipid metabolism. In 10-year-old male baboons (human equivalent 40) exposed in fetal life to betamethasone or saline, we quantified pericardial fat and hepatic lipid content with magnetic resonance imaging and spectroscopy. sGC offspring delivered at term as do most sGC-exposed human neonates. Pericardial fat thickness (7.7±3.6 mm vs 3.1±1.1 mm, M±s.d.; P=0.022; n=5) and hepatic fatty acids (13.3±11.0% vs 2.5±2.2%; P=0.046; n=5) increased following sGC without birth weight or current body morphometric differences. Our results indicate that antenatal sGC therapy caused abnormal fat deposition and adult body composition in mid-life primate offspring. The concern raised is that this degree of pericardial and hepatic lipid accumulation can lead to harmful local lipotoxicity. In summary, developmental programing by sGC produces a mid-life metabolically obese but normal weight phenotype. Prior studies show sexually dimorphic responses to some programming challenges thus female studies are necessary.


Asunto(s)
Hígado Graso/inducido químicamente , Desarrollo Fetal/efectos de los fármacos , Glucocorticoides/administración & dosificación , Glucocorticoides/efectos adversos , Exposición Materna/efectos adversos , Papio , Preñez , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Animales , Betametasona/administración & dosificación , Betametasona/efectos adversos , Betametasona/farmacocinética , Peso al Nacer , Metilación de ADN , Modelos Animales de Enfermedad , Hígado Graso/diagnóstico por imagen , Femenino , Glucocorticoides/farmacocinética , Metabolismo de los Lípidos , Hígado/diagnóstico por imagen , Hígado/metabolismo , Espectroscopía de Resonancia Magnética , Masculino , Pericardio/diagnóstico por imagen , Pericardio/metabolismo , Embarazo
4.
Br J Nutr ; 115(3): 538-46, 2016 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-26608475

RESUMEN

Maternal obesity programmes offspring development. We addressed maternal obesity effects induced by high-fat diets on maternal mammary gland (MG) structure and function and offspring brain, liver and fat outcomes. Mothers were fed control (C, n 5) or obesogenic (MO, n 5) diet from the time they were weaned through pregnancy beginning at 120 d, through lactation. At offspring postnatal day (PND) 20, milk leptin and nutrients were determined. At the end of lactation, maternal liver and MG fatty acid profile were measured. Desaturase (Δ6D and Δ5D) and elongase (ELOVL 5 and ELOVL 2) protein was measured by immunohistochemistry and Western blotting (WB) in the liver and WB in the MG. In mothers, liver, MG and milk fat content were higher in MO than in C. Liver arachidonic acid (AA) and EPA and MG EPA were lower in MO than in C. Liver desaturases were higher in MO. The MG was heavier in MO than in C, with decreased Δ5D expression in MO. Desaturases and elongases were immunolocalised in parenchymal cells of both groups. Milk yield, water, carbohydrate content, EPA and DHA were lower, whereas milk leptin and AA were higher in MO than in C. At PND 21 and 36, brain weight was less and fat depots were greater in MO offspring than in C. MO decreased male absolute brain weight but not female absolute brain weight. In conclusion, maternal obesity induced by an obesogenic diet negatively affects maternal liver and MG function with the production of significant changes in milk composition. Maternal obesity adversely affects offspring metabolism and development.


Asunto(s)
Dieta Alta en Grasa , Leche/química , Obesidad/metabolismo , Acetiltransferasas/metabolismo , Tejido Adiposo/metabolismo , Animales , Ácido Araquidónico/metabolismo , Glucemia/metabolismo , delta-5 Desaturasa de Ácido Graso , Ácidos Docosahexaenoicos/metabolismo , Ácido Eicosapentaenoico/metabolismo , Ácido Graso Desaturasas/metabolismo , Elongasas de Ácidos Grasos , Femenino , Lactancia , Leptina/metabolismo , Hígado/metabolismo , Masculino , Glándulas Mamarias Animales/metabolismo , Fenómenos Fisiologicos Nutricionales Maternos , Tamaño de los Órganos , Embarazo , Efectos Tardíos de la Exposición Prenatal , Ratas , Ratas Wistar , Estearoil-CoA Desaturasa/metabolismo
5.
Nutr Metab Cardiovasc Dis ; 26(9): 786-96, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27146364

RESUMEN

BACKGROUND AND AIMS: Moderately reduced maternal nutrient availability during pregnancy has adverse effects on the fetuses' growth and metabolism during and after pregnancy. The aim of this study was to explore effects of maternal nutrition restriction (MNR) on key metabolites of the fetal energy metabolism, particularly amino acids (AA), nonesterified fatty acids (NEFA), acylcarnitines and phospholipids. These effects may reflect mechanisms relating MNR to later adverse outcomes. METHODS AND RESULTS: Plasma and liver samples of fetal baboons, whose mothers were fed ad libitum (CTR) or MNR (70% of CTR), were collected at 0.5 and 0.9 gestation (G - term 184 days). Metabolites were measured with liquid chromatography coupled to mass spectrometry. In both, CTR and MNR, fetal metabolic profiles changed markedly between 0.5G and 0.9G. Fetal liver glucose concentrations were strongly increased. Hepatic levels of NEFA, sphingomyelins, and alkyl-linked phospholipids increased while plasma NEFA and acyl-linked phospholipids levels decreased with progression of gestation. At 0.5G, MNR fetal plasma levels of short- and medium-chain acylcarnitines were elevated, but did no longer differ between groups at 0.9G. At 0.9G, plasma levels of methionine and threonine as well as hepatic threonine levels were lower in the MNR group. CONCLUSION: Small differences in the concentrations of plasma and liver metabolites between MNR and CTR fetuses reflect good adaptation to MNR. Fetal liver metabolic profiles changed markedly between the two gestation stages, reflecting enhanced liver glucose and lipid levels with advancing gestation. Decreased concentrations of AA suggest an up-regulation of gluconeogenesis in MNR.


Asunto(s)
Restricción Calórica , Feto/metabolismo , Hígado/metabolismo , Desnutrición/metabolismo , Fenómenos Fisiologicos Nutricionales Maternos , Metaboloma , Adaptación Fisiológica , Animales , Biomarcadores/sangre , Carnitina/análogos & derivados , Carnitina/sangre , Cromatografía Liquida , Modelos Animales de Enfermedad , Ácidos Grasos no Esterificados/sangre , Femenino , Edad Gestacional , Gluconeogénesis , Humanos , Desnutrición/fisiopatología , Espectrometría de Masas , Metabolómica/métodos , Papio , Fosfolípidos/sangre , Embarazo
6.
Int J Obes (Lond) ; 39(4): 695-701, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25354845

RESUMEN

BACKGROUND/OBJECTIVES: We have reported that maternal overnutrition/obesity (OB) in sheep resulting from feeding 150% of National Research Council (NRC) requirements throughout gestation leads to maternal hyperglycemia and hyperinsulinemia. Further, newborn lambs born to OB vs control-fed (CON, 100% of NRC) ewes exhibited greater adiposity, increased blood cortisol, insulin and glucose and the elimination of the postnatal leptin spike seen in lambs born to CON ewes. This early postnatal leptin peak is necessary for the development of hypothalamic circuits, which program appetite in later life. This study evaluated the multigenerational impact of OB on insulin:glucose dynamics of mature female F1 offspring fed only to requirements throughout gestation and on their lambs (F2 generation). DESIGN AND METHODS: Adult F1 female offspring born to OB (n=10) or CON (n=7) ewes were utilized. All F1 ewes were subjected to a glucose tolerance test at midgestation and late gestation. Jugular blood was obtained from F2 lambs at birth (day 1) through postnatal day 11, and plasma glucose, insulin, cortisol and leptin concentrations were determined. Dual-energy X-ray absorptiometry was utilized to determine bone mineral density, bone mineral content, lean tissue mass and fat tissue mass. RESULTS: Fasted blood glucose and insulin concentrations were greater (P<0.05) in OBF1 than CONF1 ewes at midgestation and late gestation. Further, after glucose infusion, both glucose and insulin concentrations remained higher in OBF1 ewes (P<0.05) than CONF1 ewes, demonstrating greater insulin resistance. Blood concentrations of glucose, insulin and cortisol and adiposity were higher (P<0.01) in OBF2 lambs than CONF2 lambs at birth. Importantly, OBF2 lambs failed to exhibit the early postnatal leptin peak exhibited by CONF2 lambs. CONCLUSIONS: These data suggest that these OBF2 lambs are predisposed to exhibit the same metabolic alterations as their mothers, suggesting a multigenerational programming effect.


Asunto(s)
Fenómenos Fisiológicos Nutricionales de los Animales/fisiología , Glucemia/metabolismo , Leptina/metabolismo , Obesidad/patología , Efectos Tardíos de la Exposición Prenatal/metabolismo , Alimentación Animal , Animales , Femenino , Prueba de Tolerancia a la Glucosa , Masculino , Desnutrición , Fenómenos Fisiologicos Nutricionales Maternos/fisiología , Hipernutrición , Embarazo , Ovinos
7.
Int J Obes (Lond) ; 39(4): 712-9, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23949616

RESUMEN

BACKGROUND: Maternal obesity (MO) impairs maternal and offspring health. Mechanisms and interventions to prevent adverse maternal and offspring outcomes need to be determined. Human studies are confounded by socio-economic status providing the rationale for controlled animal data on effects of maternal exercise (MEx) intervention on maternal (F0) and offspring (F1) outcomes in MO. HYPOTHESIS: MO produces metabolic and endocrine dysfunction, increases maternal and offspring glucocorticoid exposure, oxidative stress and adverse offspring outcomes by postnatal day (PND) 36. MEx in part prevents these outcomes. METHODS: F0 female rats ate either control or obesogenic diet from weaning through lactation. Half of each group wheel ran (from day 90 of life through pregnancy beginning day 120) providing four groups (n=8/group)--(i) controls, (ii) obese, (iii) exercised controls and (iv) exercised obese. After weaning, PND 21, F1 offspring ate a control diet. Metabolic parameters of F0 prepregnancy and end of lactation and F1 offspring at PND 36 were analyzed. RESULTS: Exercise did not change maternal weight. Before breeding, MO elevated F0 glucose, insulin, triglycerides, cholesterol, leptin, fat and oxidative stress. Exercise completely prevented the triglyceride rise and partially increases glucose, insulin, cholesterol and oxidative stress. MO decreased fertility, recovered by exercise. At the end of lactation, exercise returned all metabolic variables except leptin to control levels. Exercise partially prevented MO elevated corticosterone. F1 offspring weights were similar at birth. At PND 36, MO increased F1 male but not female offspring leptin, triglycerides and fat mass. In controls, exercise reduced male and female offspring glucose, prevented the offspring leptin increase and partially the triglyceride rise. CONCLUSIONS: MEx before and during pregnancy has beneficial effects on the maternal and offspring metabolism and endocrine function occurring with no weight change in mothers and offspring indicating the importance of body composition rather than weight in evaluations of metabolic status.


Asunto(s)
Lactancia/metabolismo , Leptina/sangre , Obesidad/metabolismo , Preñez , Efectos Tardíos de la Exposición Prenatal/metabolismo , Adiposidad , Fenómenos Fisiológicos Nutricionales de los Animales , Animales , Glucemia/metabolismo , Dieta Alta en Grasa , Femenino , Resistencia a la Insulina/fisiología , Masculino , Fenómenos Fisiologicos Nutricionales Maternos , Condicionamiento Físico Animal , Embarazo , Ratas , Ratas Wistar , Destete
8.
Int J Obes (Lond) ; 39(4): 549-56, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25504042

RESUMEN

PURPOSE: Increasing evidence exists that maternal obesity (MO) and overnutrition during pregnancy and lactation have long-lasting consequences for progeny metabolism, cardiovascular and endocrine function. Data on effects of MO on offspring reproduction are limited. We hypothesized that MO during pregnancy and lactation in founder F(0) rat mothers would increase testicular and sperm oxidative stress (OS) and adversely impact male fertility in their F(1) offspring. METHODS: We induced pre-pregnancy MO by feeding F(0) females a high-fat diet from weaning through pregnancy and lactation. After weaning, all F(1) rats ate control (C) diet. We determined serum testosterone, malondialdehyde (MDA), reactive oxygen species (ROS) and superoxide dismutase (SOD) and glutathione peroxidase (GPx) activity in F(1) testes and sperm at postnatal days (PNDs) 110, 450 and 650. RESULTS: At PNDs 450 and 650, MO offspring had lower luteinizing hormone while testosterone levels were lower at all ages. Testicular MDA and ROS concentrations and SOD and GPx activity were higher in MO F(1) at all ages. Nitrotyrosine immunostaining was higher at all ages in MO F(1) testes than C F(1). At PNDs 450 and 650, MO F(1) spermatozoa showed higher MDA concentrations and lower SOD and GPx activity with reduced sperm concentration, viability and motility, and more sperm abnormalities. Fertility rate was not affected at PND 110 but was lower in MO F(1) at PNDs 450 and 650. CONCLUSIONS: We conclude that MO during pregnancy and lactation increases F(1) testicular and sperm OS leading to premature aging of reproductive capacity.


Asunto(s)
Fertilidad , Obesidad/metabolismo , Hipernutrición/metabolismo , Estrés Oxidativo , Complicaciones del Embarazo/metabolismo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Fenómenos Fisiológicos Nutricionales de los Animales , Animales , Animales Recién Nacidos , Dieta Alta en Grasa , Femenino , Infertilidad/etiología , Lactancia , Masculino , Fenómenos Fisiologicos Nutricionales Maternos , Obesidad/complicaciones , Obesidad/etiología , Hipernutrición/complicaciones , Embarazo , Ratas , Ratas Wistar , Factores Sexuales
9.
Int J Obes (Lond) ; 38(2): 224-30, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23748190

RESUMEN

OBJECTIVE: To investigate in vitro adipocyte differentiation in baboon fetuses in response to reduced maternal nutrition. DESIGN: Cross-sectional comparison of adipocyte differentiation in normally grown fetuses and fetuses of pregnant baboons fed 70% of the control global diet from 30 days of pregnancy to term. SUBJECTS: The subjects comprised control (CTR) fetuses (five female and five male) of mothers fed ad libitum and fetuses of mothers fed 70% of the global diet consumed by CTR (maternal nutrient reduction (MNR), five female and five male fetuses). The expression of genes/proteins involved in adipogenesis (PPARγ, FABP4 and adiponectin) and brown adipose tissue development (UCP1, TBX15 and COXIV) were determined in in vitro-differentiated stromal-vascular cultures from subcutaneous abdominal, subcutaneous femoral and omental adipose tissue depots. Adipocyte number per area (mm(2)) was determined histologically to assist in the evaluation of adipocyte size. RESULTS: Maternal suboptimal nutrition suppressed growth of male but not female fetuses and led to adipocyte hypertrophy accompanied by increased markers of white- and, particularly, brown-type adipogenesis in male but not female fetuses. CONCLUSION: Adipose tissue responses to fetal nonhuman primate undernutrition are sexually dimorphic. While female fetuses adapt adequately, the male ones enhance pathways involved in white and brown adipose tissue development but are unable to compensate for a delayed development of adipose tissue associated with intrauterine growth restriction. These differences need to be considered when assessing developmental programming of adiposity in response to suboptimal maternal nutrition.


Asunto(s)
Adipocitos/metabolismo , Tejido Adiposo Pardo/patología , Tejido Adiposo Blanco/patología , Fenómenos Fisiológicos Nutricionales de los Animales , Fenómenos Fisiologicos Nutricionales Maternos , Fenómenos Fisiologicos de la Nutrición Prenatal , Adipogénesis , Animales , Diferenciación Celular , Proteínas de Unión a Ácidos Grasos/metabolismo , Femenino , Desarrollo Fetal , Feto , Regulación del Desarrollo de la Expresión Génica , Edad Gestacional , Humanos , Immunoblotting , Recién Nacido , Masculino , PPAR alfa/metabolismo , Papio , Embarazo , Factores Sexuales
10.
Br J Nutr ; 111(4): 616-24, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24124655

RESUMEN

Poor maternal nutrition predisposes offspring to metabolic disease. This predisposition is modified by various postnatal factors. We hypothesised that coupled to the initial effects of developmental programming due to a maternal low-protein diet, a second hit resulting from increased offspring postnatal sugar consumption would lead to additional changes in metabolism and adipose tissue function. The objective of the present study was to determine the effects of sugared water consumption (5% sucrose in the drinking-water) on adult offspring adiposity as a 'second hit' following exposure to maternal protein restriction during pregnancy. We studied four offspring groups: (1) offspring of mothers fed the control diet (C); (2) offspring of mothers fed the restricted protein diet (R); (3) offspring of control mothers that drank sugared water (C-S); (4) offspring of restricted mothers that drank sugared water (R-S). Maternal diet in pregnancy was considered the first factor and sugared water consumption as the second factor - the second hit. Body weight and total energy consumption, before and after sugared water consumption, were similar in all the groups. Sugared water consumption increased TAG, insulin and cholesterol concentrations in both the sexes of the C-S and R-S offspring. Sugared water consumption increased leptin concentrations in the R-S females and males but not in the R offspring. There was also an interaction between sugared water and maternal diet in males. Sugared water consumption increased adipocyte size and adiposity index in both females and males, but the interaction with maternal diet was observed only in females. Adiposity index and plasma leptin concentrations were positively correlated in both the sexes. The present study shows that a second hit during adulthood can amplify the effects of higher adiposity arising due to poor maternal pregnancy diet in an offspring sex dependent fashion.


Asunto(s)
Adiposidad/efectos de los fármacos , Dieta con Restricción de Proteínas/efectos adversos , Proteínas en la Dieta/administración & dosificación , Sacarosa en la Dieta/efectos adversos , Obesidad/etiología , Efectos Tardíos de la Exposición Prenatal , Fenómenos Fisiologicos de la Nutrición Prenatal , Animales , Colesterol/sangre , Proteínas en la Dieta/farmacología , Femenino , Insulina/sangre , Leptina/sangre , Obesidad/sangre , Embarazo , Ratas , Factores Sexuales , Triglicéridos/sangre
11.
Int J Obes (Lond) ; 37(4): 568-75, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22614057

RESUMEN

BACKGROUND: Obesity in women of childbearing age is increasing at an alarming rate. Growing evidence shows that maternal obesity induces detrimental effects on offspring health, including pre-disposition to obesity. We have shown that maternal obesity increases fetal intramuscular adipogenesis at mid-gestation. However, the mechanisms are poorly understood. MicroRNAs (miRNAs) regulate mRNA stability. We hypothesized that maternal obesity alters fetal muscle miRNA expression, thereby influencing intramuscular adipogenesis. METHODS: Non-pregnant ewes received a control diet (Con, fed 100% of National Research Council (NRC) recommendations, n=6) or obesogenic diet (OB; 150% NRC recommendations, n=6) from 60 days before to 75 days after conception when the fetal longissimus dorsi (LD) muscle was sampled and miRNA expression analyzed by miRNA microarray. One of miRNAs with differential expression between Con and OB fetal muscle, let-7g, was further tested for its role in adipogenesis and cell proliferation in C3H10T1/2 cells. RESULTS: A total of 155 miRNAs were found with a signal above 500, among which, three miRNAs, hsa-miR-381, hsa-let-7g and bta-miR-376d, were differentially expressed between Con and OB fetuses, and confirmed by quantitative real-time PCR (QRT-PCR) analyses. Reduced expression of miRNA let-7g, an abundantly expressed miRNA, in OB fetal muscle was correlated with higher expression of its target genes. Overexpression of let-7g in C3H10T1/2 cells reduced their proliferation rate. Expression of adipogenic markers decreased in cells overexpressing let-7g, and the formation of adipocytes was also reduced. Overexpression of let-7g decreased expression of inflammatory cytokines. CONCLUSION: Fetal muscle miRNA expression was altered due to maternal obesity, and let-7g downregulation may enhance intramuscular adipogenesis during fetal muscle development in the setting of maternal obesity.


Asunto(s)
Adipogénesis/genética , Desarrollo Fetal/genética , MicroARNs/metabolismo , Músculo Esquelético/metabolismo , Obesidad/metabolismo , Animales , Dieta , Regulación hacia Abajo , Femenino , Regulación del Desarrollo de la Expresión Génica , Fenómenos Fisiologicos Nutricionales Maternos , Músculo Esquelético/embriología , Obesidad/genética , Embarazo , Reacción en Cadena en Tiempo Real de la Polimerasa , Oveja Doméstica
12.
Br J Nutr ; 107(11): 1562-5, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21902873

RESUMEN

Maternal low-protein (LP) diets programme ß-cell secretion, potentially altering the emergence of ageing of offspring pancreatic function. We hypothesised that isolated pancreatic islet ß-cell secretory responses are blunted in offspring exposed to LP during development and age-related reduction is influenced by the developmental stage of exposure to decreased nutrition. We studied male offspring of rats fed control (C) or LP protein (R) diets in pregnancy, first letter and/or lactation second letter of CC, RR, CR or RC groups. Serum glucose, insulin and homeostatic model assessment (HOMA) were measured. Pancreatic islets were isolated and in vitro insulin secretion quantified in low (LG - 5 mM) or high glucose (HG - 11 mM). Body weight and serum values between groups were similar at all ages. Insulin and HOMA rose with age and were highest at postnatal day (PND) 450 in all groups. At PND 36, insulin secretion was greatest in RR and RC. Only CC increased insulin secretion to HG. By PND 110, restricted groups responded less to LG but increased secretion to HG. By PND 450, CC offspring alone increased secretion to HG. Despite minimal differences in circulating insulin and glucose, reduced maternal protein intake affected insulin secretion at all ages. In addition, ageing reduced function in all R groups compared with CC by PND 110 and further by PND 450 most markedly in RC. We conclude that maternal LP diet during pregnancy and/or lactation impairs offspring insulin secretory response to a glucose challenge and alters the trajectory of ageing of pancreatic insulin secretion.


Asunto(s)
Envejecimiento/fisiología , Dieta con Restricción de Proteínas/efectos adversos , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Fenómenos Fisiologicos de la Nutrición Prenatal , Envejecimiento/sangre , Animales , Glucemia/análisis , Femenino , Glucosa/metabolismo , Insulina/sangre , Resistencia a la Insulina , Secreción de Insulina , Islotes Pancreáticos/crecimiento & desarrollo , Lactancia , Masculino , Concentración Osmolar , Embarazo , Ratas , Ratas Wistar , Técnicas de Cultivo de Tejidos
13.
Nat Med ; 4(4): 456-9, 1998 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-9546793

RESUMEN

The central biochemical mechanisms involved in primate parturition are still unclear. Studies in both humans and nonhuman primates such as the baboon and rhesus monkey indicate that many factors play a part in the cascade of interactive positive feedforward loops that progressively promote parturition: changes in maternal endocrinology, a nocturnal switch in myometrial activity from low amplitude, infrequent contractures to high amplitude, high frequency contractions (see Fig. 1), dilation of the cervix and biochemical changes in the fetal membranes that lead to rupture. Here we demonstrate that infusion of the aromatase inhibitor 4-hydroxyandrostenedione (4OHA) inhibits conversion of androgen to estrogen and prevents premature delivery caused by administration of androgen to pregnant rhesus monkeys at 0.8 of pregnancy term. 4OHA also inhibited the androstenedione induced maternal endocrine and fetal membrane biochemical changes, and alteration of myometrial activity patterns. Secondly, peripheral estrogen infusions increased myometrial activity but did not produce preterm delivery or fetal membrane changes. We conclude that paracrine functions of estrogen at its site of production play critical and central roles in delivery in the non-human primate.


Asunto(s)
Androstenodiona/análogos & derivados , Androstenodiona/farmacología , Estradiol/farmacología , Trabajo de Parto/fisiología , Macaca mulatta/fisiología , Miometrio/fisiología , Contracción Uterina/fisiología , Androstenodiona/administración & dosificación , Animales , Inhibidores de la Aromatasa , Electromiografía/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Estradiol/administración & dosificación , Estradiol/sangre , Femenino , Humanos , Infusiones Intravenosas , Trabajo de Parto/sangre , Miometrio/efectos de los fármacos , Oxitocina/sangre , Embarazo , Progesterona/sangre , Contracción Uterina/efectos de los fármacos
14.
Nat Med ; 2(4): 443-8, 1996 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-8597955

RESUMEN

The endocrine mechanism involved in term and preterm delivery in primates, including pregnant women, are poorly understood. In the term monkey, fetal plasma androgen concentration rises to two hundred times the maternal concentration which remains unchanged. Placental conversion of androgen to estrogen results in increased maternal plasma estrogen concentration at term in both pregnant nonhuman primates and women. In the present study, continuous infusion of androstenedione to 0.8 gestation monkeys resulted in the premature occurrence of labor-type myometrial activity and increases in maternal plasma estrogen, oxytocin and amnion fibronectin concentrations similar to those measured at normal-term labor. Androstenedione induction of these normal-term biochemical and endocrine changes accompanied by fetal membrane rupture, cervical dilatation and live delivery provides a rich opportunity to study the molecular and physiological mechanisms of both term and preterm labor in primates.


Asunto(s)
Androstenodiona/administración & dosificación , Macaca mulatta , Enfermedades de los Monos/inducido químicamente , Miometrio/fisiopatología , Trabajo de Parto Prematuro/inducido químicamente , Trabajo de Parto Prematuro/veterinaria , Animales , Estrógenos/sangre , Femenino , Fibronectinas/metabolismo , Humanos , Infusiones Intravenosas , Trabajo de Parto Prematuro/fisiopatología , Oxitocina/sangre , Embarazo , Contracción Uterina/efectos de los fármacos
15.
J Dev Orig Health Dis ; 12(2): 203-219, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33349289

RESUMEN

Cardiovascular diseases (CVD) are important consequences of adverse perinatal conditions such as fetal hypoxia and maternal malnutrition. Cardiac magnetic resonance imaging (CMR) can produce a wealth of physiological information related to the development of the heart. This review outlines the current state of CMR technologies and describes the physiological biomarkers that can be measured. These phenotypes include impaired ventricular and atrial function, maladaptive ventricular remodeling, and the proliferation of myocardial steatosis and fibrosis. The discussion outlines the applications of CMR to understanding the developmental pathways leading to impaired cardiac function. The use of CMR, both in animal models of developmental programming and in human studies, is described. Specific examples are given in a baboon model of intrauterine growth restriction (IUGR). CMR offers great potential as a tool for understanding the sequence of dysfunctional adaptations of developmental origin that can affect the human cardiovascular system.


Asunto(s)
Envejecimiento , Retardo del Crecimiento Fetal/fisiopatología , Corazón/embriología , Corazón/fisiopatología , Imagen por Resonancia Magnética/métodos , Animales , Femenino , Humanos , Embarazo
16.
J Physiol ; 588(Pt 10): 1791-9, 2010 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-20351043

RESUMEN

Obesity involving women of reproductive years is increasing dramatically in both developing and developed nations. Maternal obesity and accompanying high energy obesogenic dietary (MO) intake prior to and throughout pregnancy and lactation program offspring physiological systems predisposing to altered carbohydrate and lipid metabolism. Whether maternal obesity-induced programming outcomes are reversible by altered dietary intake commencing before conception remains an unanswered question of physiological and clinical importance. We induced pre-pregnancy maternal obesity by feeding female rats with a high fat diet from weaning to breeding 90 days later and through pregnancy and lactation. A dietary intervention group (DINT) of MO females was transferred to normal chow 1 month before mating. Controls received normal chow throughout. Male offspring were studied. Offspring birth weights were similar. At postnatal day 21 fat mass, serum triglycerides, leptin and insulin were elevated in MO offspring and were normalized by DINT. At postnatal day 120 serum glucose, insulin and homeostasis model assessment (HOMA) were increased in MO offspring; glucose was restored, and HOMA partially reversed to normal by DINT. At postnatal day 150 fat mass was increased in MO and partially reversed in DINT. At postnatal day 150, fat cell size was increased by MO. DINT partially reversed these differences in fat cell size. We believe this is the first study showing reversibility of adverse metabolic effects of maternal obesity on offspring metabolic phenotype, and that outcomes and reversibility vary by tissue affected.


Asunto(s)
Dieta , Desarrollo Fetal/fisiología , Obesidad/metabolismo , Preñez/fisiología , Adipocitos/ultraestructura , Animales , Peso al Nacer/fisiología , Glucemia/metabolismo , Peso Corporal/fisiología , Tamaño de la Célula , Colesterol/sangre , Ingestión de Alimentos , Femenino , Insulina/sangre , Resistencia a la Insulina/fisiología , Lactancia/fisiología , Leptina/sangre , Tamaño de la Camada , Masculino , Fenotipo , Embarazo , Ratas , Ratas Wistar , Triglicéridos/sangre
17.
J Med Primatol ; 38(4): 219-27, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19602098

RESUMEN

BACKGROUND: A sub-optimal intrauterine environment alters the trajectory of fetal development with profound effects on life-time health. Altered methylation, a proposed epigenetic mechanism responsible for these changes, has been studied in non-primate species but not nonhuman primates. We tested the hypotheses that global methylation in fetal baboon demonstrates organ specificity, gestational age specificity, and changes with maternal nutritional status. METHODS: We measured global DNA methylation in fetuses of control fed (CTR) and nutrient restricted mothers fed 70% of controls (MNR) for brain, kidney, liver and heart at 0.5 and 0.9 gestation (G). RESULTS: We observed organ and gestation specific changes that were modified by maternal diet. Methylation in CTR fetuses was highest in frontal cortex and lowest in liver. MNR decreased methylation in 0.5G kidney and increased methylation in 0.9G kidney and frontal cortex. CONCLUSION: These results demonstrate a potential epigenetic mechanism whereby reduced maternal nutrition has long-term programming effects on fetal organ development.


Asunto(s)
Fenómenos Fisiológicos Nutricionales de los Animales , Metilación de ADN/fisiología , Feto/metabolismo , Fenómenos Fisiologicos Nutricionales Maternos , Papio/metabolismo , Animales , Encéfalo/anatomía & histología , Femenino , Feto/anatomía & histología , Edad Gestacional , Corazón/anatomía & histología , Riñón/anatomía & histología , Hígado/anatomía & histología , Tamaño de los Órganos , Embarazo
18.
J Dev Orig Health Dis ; 9(2): 137-142, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29017630

RESUMEN

The ability of the aorta to buffer blood flow and provide diastolic perfusion (Windkessel function) is a determinant of cardiovascular health. We have reported cardiac dysfunction indicating downstream vascular abnormalities in young adult baboons who were intrauterine growth restricted (IUGR) at birth as a result of moderate maternal nutrient reduction. Using 3 T MRI, we examined IUGR offspring (eight male, eight female; 5.7 years; human equivalent 25 years) and age-matched controls (eight male, eight female; 5.6 years) to quantify distal descending aortic cross-section (AC) and distensibility (AD). ANOVA showed decreased IUGR AC/body surface area (0.9±0.05 cm2/m2 v. 1.2±0.06 cm2/m2, M±s.e.m., P<0.005) and AD (1.7±0.2 v. 4.0±0.5×10-3/mmHg, P<0.005) without sex difference or group-sex interaction, suggesting intrinsic vascular pathology and impaired development persisting in adulthood. Future studies should evaluate potential consequences of these changes on coronary perfusion, afterload and blood pressure.


Asunto(s)
Aorta/diagnóstico por imagen , Presión Sanguínea/fisiología , Retardo del Crecimiento Fetal/diagnóstico por imagen , Animales , Aorta/fisiopatología , Femenino , Retardo del Crecimiento Fetal/fisiopatología , Masculino , Papio , Embarazo
19.
Placenta ; 28(11-12): 1192-9, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17586041

RESUMEN

This study evaluated the role of MAPK/ERK1/2 and/or PI3K/Akt signaling pathways in modulating ovine placentomal vascularity in response to periconceptional maternal nutrient restriction. Ewes were randomly assigned to be nutrient restricted (NR, 50% NRC recommendation, N=7) or control fed (CF, 100% NRC recommendation, N=7) from 60 +/- 2 days before to 30 days after conception (day 0). From day 31 of gestation, all ewes (CF and NR) were fed the control diet until necropsy on day 78. On day 78 of gestation, NR ewes exhibited greater vascularity in both caruncular (CAR) and cotyledon (COT) tissues than CF ewes. Akt or ERK1/2 content in CAR and COT arterial tissue did not differ across dietary treatment. The activated forms, phosphorylated Akt and phosphorylated ERK1/2, were significantly increased in COT but not CAR arterial tissues of NR ewes compared to those of CF ewes (P<0.05). For both CF and NR ewes, phosphorylated Akt and phosphorylated ERK1/2 content in COT are higher (P<0.05) than those in CAR arterial tissues. Immunohistochemical staining revealed cytoplasmic and nuclear localization of Akt, phosphorylated Akt, ERK1/2 and phosphorylated ERK1/2, with phosphorylated Akt and phosphorylated-ERK1/2 specifically localized in trophoblast cells, while binucleate cells remained unstained. In placentomal blood vessels, Akt, phosphorylated Akt, ERK1/2 and phosphorylated ERK1/2 were localized to both endothelium and smooth muscle cells. These findings demonstrate for the first time that periconceptional NR increases vascular density in both COT than CAR tissues of the ovine placentome, and that the MAPK/ERK1/2 and/or PI3K/Akt signaling pathways are increased in NR COT but not NR CAR arterial tissues.


Asunto(s)
Fenómenos Fisiológicos Nutricionales de los Animales/fisiología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Placenta/irrigación sanguínea , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Restricción Calórica , Femenino , Peso Fetal/fisiología , Inmunohistoquímica , Tamaño de los Órganos/fisiología , Placenta/química , Placentación , Embarazo , Ovinos , Transducción de Señal/fisiología
20.
Placenta ; 28(4): 361-8, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16822544

RESUMEN

This study evaluated the role of MAPK/ERK1/2 and/or PI3-K/Akt signaling pathways in modulating bovine placentomal vascularity in response to maternal nutrient restriction. Beef cows were randomly assigned to control fed (Control, n=15, 100% of requirements) or nutrient restricted (NR, n=15, 50% requirements) diets from day 30 to day 125 of gestation. Ten cows from each dietary group were necropsied on day 125 (approximately 45% gestation), and the remaining cows in each diet group were then fed control diets and necropsied on day 250 (approximately 90% gestation). At day 125 of gestation, NR cows exhibited increased (P=0.06) COT vascularity, improved (P<0.05) placentome efficiency (fetal weight/placentomal weight), and increased (P<0.05) phosphorylated Akt and ERK1/2 in COT arteries compared to Control cows. By day 250, however, treatment differences in COT vascularity and phosphorylated Akt and ERK1/2 in COT arteries were lost. On both gestational days, no treatment difference was observed in the levels of phosphorylated Akt or ERK1/2 in CAR arteries. CAR vascularity was similar across treatment on day 125, but tended to be greater (P<0.10) in NR than Control cows on day 250. These data suggest that conceptuses react to an early gestational nutrient restriction by up-regulating COT growth signaling pathways associated with angiogenesis, and that these compensations do not persist to term.


Asunto(s)
Fenómenos Fisiológicos Nutricionales de los Animales , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Privación de Alimentos/fisiología , Placenta/irrigación sanguínea , Circulación Placentaria/fisiología , Animales , Bovinos , Femenino , Peso Fetal , Edad Gestacional , Neovascularización Fisiológica/fisiología , Tamaño de los Órganos , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Placentación , Embarazo , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA