Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 133
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Crit Rev Food Sci Nutr ; 56(1): 82-91, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-24628089

RESUMO

Pregnancy is a complex period of human growth, development, and imprinting. Nutrition and metabolism play a crucial role for the health and well-being of both mother and fetus, as well as for the long-term health of the offspring. Nevertheless, several biological and physiological mechanisms related to nutritive requirements together with their transfer and utilization across the placenta are still poorly understood. In February 2009, the Child Health Foundation invited leading experts of this field to a workshop to critically review and discuss current knowledge, with the aim to highlight priorities for future research. This paper summarizes our main conclusions with regards to maternal preconceptional body mass index, gestational weight gain, placental and fetal requirements in relation to adverse pregnancy and long-term outcomes of the fetus (nutritional programming). We conclude that there is an urgent need to develop further human investigations aimed at better understanding of the basis of biochemical mechanisms and pathophysiological events related to maternal-fetal nutrition and offspring health. An improved knowledge would help to optimize nutritional recommendations for pregnancy.


Assuntos
Saúde Global , Transtornos da Nutrição do Lactente/prevenção & controle , Fenômenos Fisiológicos da Nutrição Materna , Modelos Biológicos , Política Nutricional , Cooperação do Paciente , Complicações na Gravidez/prevenção & controle , Adulto , Desenvolvimento Infantil , Feminino , Desenvolvimento Fetal , Humanos , Transtornos da Nutrição do Lactente/epidemiologia , Recém-Nascido , Estado Nutricional , Gravidez , Complicações na Gravidez/epidemiologia , Resultado da Gravidez , Risco , Aumento de Peso
2.
Diabetes Obes Metab ; 18(1): 72-81, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26434748

RESUMO

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


Assuntos
Tecido Adiposo Marrom/metabolismo , Metabolismo Energético/efeitos dos fármacos , Glucagon/farmacocinética , Adulto , Temperatura Baixa , Estudos Controlados Antes e Depois , Fluordesoxiglucose F18 , Voluntários Saudáveis , Humanos , Masculino , Tomografia por Emissão de Pósitrons/métodos , Distribuição Aleatória , Termogênese/efeitos dos fármacos , Tomografia Computadorizada por Raios X , Adulto Jovem
3.
Reprod Fertil Dev ; 27(7): 1057-64, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24717142

RESUMO

Piglet neonatal mortality rates are high (~20%), so nutritional strategies to reduce this are highly desirable. Maternal fat substitution (FS) may promote the preweaning survival of piglets by improving their energy status. Therefore, the aim of the present study was to investigate the effects of FS throughout pregnancy on offspring viability, together with the gene expression of stress-related markers in the liver. Sixteen pregnant sows were randomly allocated to one of two isocaloric diets, control (C) or FS in the form of palm oil, fed from 0 to 110 days gestation. Glucose tolerance was examined on Day 108. Median and low birthweight offspring were allocated to tissue sampling at either 7 days or 6 months postnatal age. In response to a glucose tolerance test, FS sows exhibited a raised glucose area under the curve with no change in basal glucose. Average piglet mortality (up to Day 28) was increased fourfold in the FS group, with surviving median-sized piglets exhibiting significantly lower fatty acid binding protein 1 (FABP1) expression at 7 days. There were no effects on the abundance of any other stress- or metabolic-related genes examined. Thus, this study demonstrates that maternal FS throughout gestation causes maternal glucose intolerance that may be linked to the observed increase in piglet mortality. However, the surviving offspring do not exhibit any detectable differences in postnatal growth or hepatic gene profile in later life.


Assuntos
Fenômenos Fisiológicos da Nutrição Animal/fisiologia , Dieta , Expressão Gênica , Fígado/metabolismo , Fenômenos Fisiológicos da Nutrição Materna/fisiologia , Óleos de Plantas/administração & dosagem , Ração Animal/análise , Animais , Animais Recém-Nascidos , Proteínas de Ligação a Ácido Graxo/genética , Proteínas de Ligação a Ácido Graxo/metabolismo , Feminino , Intolerância à Glucose/genética , Intolerância à Glucose/metabolismo , Óleo de Palmeira , Gravidez , Suínos
4.
Int J Obes (Lond) ; 38(7): 887-905, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24662696

RESUMO

The 2013 Pennington Biomedical Research Center's Scientific Symposium focused on the treatment and management of pediatric obesity and was designed to (i) review recent scientific advances in the prevention, clinical treatment and management of pediatric obesity, (ii) integrate the latest published and unpublished findings and (iii) explore how these advances can be integrated into clinical and public health approaches. The symposium provided an overview of important new advances in the field, which led to several recommendations for incorporating the scientific evidence into practice. The science presented covered a range of topics related to pediatric obesity, including the role of genetic differences, epigenetic events influenced by in utero development, pre-pregnancy maternal obesity status, maternal nutrition and maternal weight gain on developmental programming of adiposity in offspring. Finally, the relative merits of a range of various behavioral approaches targeted at pediatric obesity were covered, together with the specific roles of pharmacotherapy and bariatric surgery in pediatric populations. In summary, pediatric obesity is a very challenging problem that is unprecedented in evolutionary terms; one which has the capacity to negate many of the health benefits that have contributed to the increased longevity observed in the developed world.


Assuntos
Adiposidade , Pesquisa Biomédica , Obesidade Infantil/prevenção & controle , Saúde Pública , Aumento de Peso , Adolescente , Adulto , Criança , Pré-Escolar , Dieta , Epigenômica , Medicina Baseada em Evidências , Exercício Físico , Conhecimentos, Atitudes e Prática em Saúde , Promoção da Saúde , Humanos , Obesidade Infantil/epidemiologia , Obesidade Infantil/genética , Vigilância da População , Prevalência , Fatores de Risco , Aumento de Peso/genética
5.
Reprod Fertil Dev ; 25(5): 728-36, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-22951182

RESUMO

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


Assuntos
Desenvolvimento Fetal/fisiologia , Resistência à Insulina/fisiologia , Rim/embriologia , Lipídeos/análise , Fenômenos Fisiológicos da Nutrição Materna/fisiologia , Obesidade/fisiopatologia , Animais , Western Blotting , Primers do DNA/genética , Feminino , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Imuno-Histoquímica , Rim/química , Gravidez , Reação em Cadeia da Polimerase em Tempo Real , Ovinos , Estatísticas não Paramétricas , Triglicerídeos/análise , Canal de Ânion 1 Dependente de Voltagem/metabolismo
6.
Ann Nutr Metab ; 63(3): 208-15, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24107818

RESUMO

Offspring of obese and diabetic mothers are at increased risk of being born with excess adiposity as a consequence of their intrauterine environment. Excessive fetal fat accretion reflects additional placental nutrient transfer, suggesting an effect of the maternal environment on placental function. High plasma levels of particular nutrients in obese and diabetic mothers are likely to be the important drivers of nutrient transfer to the fetus, resulting in excess fat accretion. However, not all offspring of obese and diabetic mothers are born large for gestational age and the explanation may involve the regulation of placental nutrient transfer required for fetal growth. The placenta integrates maternal and fetal signals across gestation in order to determine nutrient transfer rate. Understanding the nature of these signals and placental responses to them is key to understanding the pathology of both fetal growth restriction and macrosomia. The overall effects of the maternal environment on the placenta are the product of its exposures throughout gestation, the 'placental exposome'. Understanding these environmental influences is important as exposures early in gestation, for instance causing changes in the function of genes involved in nutrient transfer, may determine how the placenta will respond to exposures later in gestation, such as to raised maternal plasma glucose or lipid concentrations. Longitudinal studies are required which allow investigation of the influences on the placenta across gestation. These studies need to make full use of developing technologies characterising placental function, fetal growth and body composition. Understanding these processes will assist in the development of preventive strategies and treatments to optimise prenatal growth in those pregnancies at risk of either excess or insufficient nutrient supply and could also reduce the risk of chronic disease in later life.


Assuntos
Adiposidade , Composição Corporal/fisiologia , Feto/metabolismo , Placentação , Peso ao Nascer , Epigênese Genética , Feminino , Desenvolvimento Fetal , Humanos , Troca Materno-Fetal , Obesidade/metabolismo , Gravidez , Resultado da Gravidez , Fatores de Risco
7.
Diabetologia ; 55(6): 1597-606, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22402988

RESUMO

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


Assuntos
Tecido Adiposo Marrom/metabolismo , Tecido Adiposo Branco/metabolismo , Desenvolvimento Fetal/fisiologia , Tecido Adiposo Marrom/embriologia , Tecido Adiposo Branco/embriologia , Animais , Feminino , Desenvolvimento Fetal/genética , Humanos , Resistência à Insulina , Modelos Biológicos , Gravidez
8.
Reproduction ; 141(1): 119-26, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21045167

RESUMO

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


Assuntos
Fenômenos Fisiológicos da Nutrição Animal , Fígado Gorduroso/metabolismo , Fígado/metabolismo , Desnutrição/metabolismo , Fenômenos Fisiológicos da Nutrição Materna , Obesidade/metabolismo , Efeitos Tardios da Exposição Pré-Natal , Fatores Etários , Animais , Modelos Animais de Doenças , Fígado Gorduroso/embriologia , Fígado Gorduroso/genética , Fígado Gorduroso/patologia , Fígado Gorduroso/fisiopatologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Idade Gestacional , Gluconeogênese/genética , Metabolismo dos Lipídeos/genética , Fígado/embriologia , Fígado/patologia , Fígado/fisiopatologia , Desnutrição/embriologia , Desnutrição/genética , Desnutrição/fisiopatologia , Mitocôndrias Hepáticas/metabolismo , Obesidade/embriologia , Obesidade/genética , Obesidade/patologia , Obesidade/fisiopatologia , PPAR gama/genética , Gravidez , Ovinos , Triglicerídeos/metabolismo
9.
Reproduction ; 139(1): 265-74, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19786398

RESUMO

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


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Fenômenos Fisiológicos da Nutrição Materna , Sobrepeso/metabolismo , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Proteínas/genética , Envelhecimento/metabolismo , Animais , DNA Complementar/química , Feminino , Peso Fetal , Hipotálamo/metabolismo , Masculino , Obesidade/prevenção & controle , Tamanho do Órgão , Especificidade de Órgãos , Placenta/metabolismo , Gravidez , Proteínas/química , Proteínas/metabolismo , RNA Mensageiro/metabolismo , Alinhamento de Sequência , Carneiro Doméstico , Magreza/metabolismo
10.
Reproduction ; 138(3): 601-8, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19525364

RESUMO

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


Assuntos
Restrição Calórica/veterinária , Proliferação de Células , Glucocorticoides/farmacologia , Placenta/efeitos dos fármacos , Prenhez , Ovinos , Ração Animal , Animais , Restrição Calórica/efeitos adversos , Proliferação de Células/efeitos dos fármacos , Resistência a Medicamentos/efeitos dos fármacos , Feminino , Alimentos , Canais Iônicos/genética , Canais Iônicos/metabolismo , Metabolismo dos Lipídeos/genética , Fenômenos Fisiológicos da Nutrição Materna , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , PPAR gama/genética , PPAR gama/metabolismo , Placenta/metabolismo , Gravidez , Prenhez/genética , Prenhez/fisiologia , Ovinos/embriologia , Ovinos/genética , Ovinos/metabolismo , Ovinos/fisiologia , Fatores de Tempo , Proteína Desacopladora 2
11.
Reproduction ; 138(3): 609-17, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19502453

RESUMO

Epidemiological studies suggest that low-birth weight infants show poor neonatal growth and increased susceptibility to metabolic syndrome, in particular, obesity and diabetes. Adipose tissue development is regulated by many genes, including members of the peroxisome proliferator-activated receptor (PPAR) and the fatty acid-binding protein (FABP) families. The aim of this study was to determine the influence of birth weight on key adipose and skeletal muscle tissue regulating genes. Piglets from 11 litters were ranked according to birth weight and 3 from each litter assigned to small, normal, or large-birth weight groups. Tissue samples were collected on day 7 or 14. Plasma metabolite concentrations and the expression of PPARG2, PPARA, FABP3, and FABP4 genes were determined in subcutaneous adipose tissue and skeletal muscle. Adipocyte number and area were determined histologically. Expression of FABP3 and 4 was significantly reduced in small and large, compared with normal, piglets in adipose tissue on day 7 and in skeletal muscle on day 14. On day 7, PPARA and PPARG2 were significantly reduced in adipose tissue from small and large piglets. Adipose tissue from small piglets contained more adipocytes than normal or large piglets. Birth weight had no effect on adipose tissue and skeletal muscle lipid content. Low-birth weight is associated with tissue-specific and time-dependent effects on lipid-regulating genes as well as morphological changes in adipose tissue. It remains to be seen whether these developmental changes alter an individual's susceptibility to metabolic syndrome.


Assuntos
Peso ao Nascer/fisiologia , Metabolismo dos Lipídeos/genética , Músculo Esquelético/metabolismo , Gordura Subcutânea/metabolismo , Suínos , Adipócitos/citologia , Adipócitos/metabolismo , Adipogenia/genética , Animais , Animais Recém-Nascidos/sangue , Animais Recém-Nascidos/genética , Animais Recém-Nascidos/crescimento & desenvolvimento , Animais Recém-Nascidos/metabolismo , Peso ao Nascer/genética , Proteínas de Ligação a Ácido Graxo/genética , Proteínas de Ligação a Ácido Graxo/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Lipólise/genética , Lipólise/fisiologia , PPAR alfa/genética , PPAR alfa/metabolismo , PPAR gama/genética , PPAR gama/metabolismo , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/metabolismo , Suínos/sangue , Suínos/genética , Suínos/metabolismo , Suínos/fisiologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
12.
Reproduction ; 135(5): 723-32, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18304988

RESUMO

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


Assuntos
Temperatura Baixa , Privação de Alimentos , Fígado/metabolismo , Exposição Materna , Carneiro Doméstico/metabolismo , Somatomedinas/metabolismo , Animais , Animais Recém-Nascidos/crescimento & desenvolvimento , Feminino , Glucose-6-Fosfatase/metabolismo , Glicogênio/metabolismo , Hormônio do Crescimento/metabolismo , Fator de Crescimento Insulin-Like I/genética , Fator de Crescimento Insulin-Like I/metabolismo , Fator de Crescimento Insulin-Like II/metabolismo , PPAR alfa/metabolismo , Gravidez , RNA Mensageiro/análise
13.
J Endocrinol ; 192(1): 87-97, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17210746

RESUMO

The liver is a major metabolic and endocrine organ of critical importance in the regulation of growth and metabolism. Its function is determined by a complex interaction of nutritionally regulated counter-regulatory hormones. The extent to which hepatic endocrine sensitivity can be programed in utero and whether the resultant adaptations persist into adulthood is unknown and was therefore the subject of this study. Young adult male sheep born to mothers that were fed either a control diet (i.e.100% of total live weight-maintenance requirements) throughout gestation or 50% of that intake (i.e. nutrient restricted (NR)) from 0 to 95 days gestation and thereafter 100% of requirements (taking into account increasing fetal mass) were entered into the study. All mothers gave birth normally at term, the singleton offspring were weaned at 16 weeks, and then reared at pasture until 3 years of age when their livers were sampled. NR offspring were of similar birth and body weights at 3 years of age when they had disproportionately smaller livers than controls. The abundance of mRNA for GH, prolactin, and IGF-II receptors, plus hepatocyte growth factor and suppressor of cytokine signaling-3 were all lower in livers of NR offspring. In contrast, the abundance of the mitochondrial protein voltage-dependent anion channel and the pro-apoptotic factor Bax were up regulated relative to controls. In conclusion, maternal nutrient restriction in early gestation results in adult offspring with smaller livers. This may be mediated by alterations in both hepatic mitogenic and apoptotic factors.


Assuntos
Fenômenos Fisiológicos da Nutrição Animal , Privação de Alimentos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Fígado/embriologia , Fenômenos Fisiológicos da Nutrição Pré-Natal , RNA Mensageiro/análise , Animais , Primers do DNA/genética , Feminino , Idade Gestacional , Fator de Crescimento de Hepatócito/genética , Fígado/anatomia & histologia , Fígado/metabolismo , Masculino , Tamanho do Órgão , Gravidez , Receptor IGF Tipo 2/genética , Receptores da Prolactina/genética , Receptores da Somatotropina/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ovinos , Proteínas Supressoras da Sinalização de Citocina/genética
14.
J Endocrinol ; 188(3): 375-86, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16522718

RESUMO

Glucocorticoid action has a major role in regulating fetal and postnatal lung development, although its impact on mitochondrial development is less well understood. Critically, the consequences of any change in glucocorticoid action and mitochondrial function in early life may not be limited to the postnatal period, but may extend into later life. This paper focuses on more recent findings on the impact of ontogeny, fetal cortisol status, maternal nutrient restriction and postnatal leptin administration on mitochondrial uncoupling protein (UCP)-2, glucocorticoid receptor (GR) and 11 beta-hydroxysteroid dehydrogenase type 1 (11betaHSD1) isoform abundance in the lung. For example, in sheep, GR and 11betaHSD1 mRNA are maximal at 140 days' gestation (term approximately 147 days), while UCP2 mRNA peaks at 1 day after birth and then decreases with advancing age. In the fetus, chronic umbilical cord compression enhances the abundance of these genes, an outcome that can also be produced after birth following chronic, but not acute, leptin administration. Irrespective of the timing of maternal nutrient restriction in pregnancy, glucocorticoid sensitivity and UCP2 abundance are both upregulated in the lungs of the resulting offspring. In conclusion, prenatal and postnatal endocrine challenges have distinct effects on mitochondrial development in the lung resulting from changes in glucocorticoid action, which can persist into later life. As a consequence, changes in glucocorticoid sensitivity and mitochondrial protein abundance have the potential to be used to identify those at greatest risk of developing later lung disease.


Assuntos
Desenvolvimento Fetal/fisiologia , Glucocorticoides/metabolismo , Pulmão/embriologia , Proteínas de Membrana Transportadoras/metabolismo , Proteínas Mitocondriais/metabolismo , Fenômenos Fisiológicos da Nutrição Pré-Natal , Animais , Animais Recém-Nascidos , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Idade Gestacional , Canais Iônicos , Leptina/metabolismo , Leptina/farmacologia , Pulmão/crescimento & desenvolvimento , Pulmão/metabolismo , Mitocôndrias/metabolismo , Gravidez , Ovinos , Proteína Desacopladora 2
15.
J Endocrinol ; 190(2): 203-12, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16899555

RESUMO

Glucocorticoids are proposed to act as intermediary factors that transcribe the developmental programming sequelae of maternal nutrient restriction (NR). Periconceptional under-nutrition of sheep markedly activates fetal hypothalamic-pituitary-adrenal (HPA) axis activity leading to preterm birth, while transient undernutrition during late gestation in sheep programs adult HPA axis function. To date, no study has examined resting or stimulated HPA axis function in young adult offspring following a periconceptional nutritional challenge. In the present study, 20 ewes were either periconceptionally undernourished (50% metabolisable energy requirements from days 1 to 30 gestation; NR, n = 8) or fed to control levels (100% requirement; controls, n = 12) to term (147 days gestation). Ewes were blood sampled remotely at 2 and 30 days using automated blood sampling equipment. Thereafter, offspring (controls, n = 6/6 males/females; NR, n = 4/4 males/females) were reared to 1 year of age and on separate days received either an i.v. corticotrophin-releasing hormone (CRH; 0.5 microg/kg) and vasopressin (AVP; 0.1 microg/kg) challenge or a synthetic ACTH i.v. bolus (Synacthen; 1.25 microg/kg), and blood samples were taken (manually and remotely) at appropriate intervals for measurement of plasma ACTH and cortisol accordingly. Resting plasma cortisol, assessed remotely, was similar in ewes during undernutrition (control 18.3 +/- 1.4 vs NR 23.4 +/- 1.9 nmol/l) and in offspring at 4 months of age (control male 17.6 +/- 2.9; control female 17.2 +/- 0.4, NR male 16.5 +/- 3.1, NR female 21.7 +/- 4.0 nmol/l). At 12 months of age, however, resting plasma cortisol was significantly increased in NR females (control male 28.0 +/- 1.5, control female 32.9 +/- 9, NR male 32 +/- 7, NR female 53 +/- 10 nmol/l, F 5.7, P = 0.02) despite no difference in plasma ACTH concentration. There was an interaction between nutritional group and gender for both the pituitary and adrenal responses to CRH and AVP, i.e. for controls, females exhibited increased plasma ACTH or cortisol relative to males but for NR this trend was either not present or reversed. The adrenocortical response to synthetic ACTH was gender-dependent only, being greater in female offspring. Combined CRH and AVP provoked a transient hypertension and marked bradycardia in all animals, irrespective of dietary group or gender and could be effectively reproduced by an AVP bolus alone. In conclusion, the present study has shown that periconceptional undernutrition of sheep has only a minor influence on HPA axis function in their young adult offspring when considered alongside the effect of gender per se.


Assuntos
Sistema Hipotálamo-Hipofisário/fisiologia , Desnutrição/metabolismo , Fenômenos Fisiológicos da Nutrição Materna , Sistema Hipófise-Suprarrenal/fisiologia , Sexo , Ovinos/metabolismo , Hormônio Adrenocorticotrópico/sangue , Animais , Arginina Vasopressina , Pressão Sanguínea/efeitos dos fármacos , Coleta de Amostras Sanguíneas/métodos , Hormônio Liberador da Corticotropina , Feminino , Idade Gestacional , Frequência Cardíaca/efeitos dos fármacos , Hidrocortisona/sangue , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Masculino , Sistema Hipófise-Suprarrenal/efeitos dos fármacos , Gravidez , Ovinos/crescimento & desenvolvimento
16.
Theriogenology ; 86(1): 120-9, 2016 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-27173959

RESUMO

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


Assuntos
Adiposidade/fisiologia , Reprodução/fisiologia , Ovinos/fisiologia , Animais , Feminino , Desenvolvimento Fetal , Masculino , Gravidez
17.
J Clin Endocrinol Metab ; 101(1): 59-68, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26513002

RESUMO

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


Assuntos
Peso Corporal , Diabetes Gestacional/fisiopatologia , Placenta/fisiopatologia , Resultado da Gravidez , Adolescente , Adulto , Antropometria , Peso ao Nascer/genética , Índice de Massa Corporal , Diabetes Gestacional/genética , Ingestão de Energia/genética , Feminino , Expressão Gênica/genética , Intolerância à Glucose/complicações , Intolerância à Glucose/genética , Humanos , Recém-Nascido , Inflamação/genética , Inflamação/patologia , Estudos Longitudinais , Redes e Vias Metabólicas/genética , Pessoa de Meia-Idade , Obesidade/complicações , Obesidade/genética , Estresse Oxidativo , Placenta/metabolismo , Gravidez , Espanha/epidemiologia , Adulto Jovem
18.
Endocrinology ; 146(9): 3943-9, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15961559

RESUMO

Maternal nutrient restriction at specific stages of gestation has differential effects on fetal development such that the offspring are programmed to be at increased risk of a range of adult diseases, including obesity. We investigated the effect of maternal nutritional manipulation through gestation on fetal adipose tissue deposition in conjunction with mRNA abundance for uncoupling protein (UCP)1 and 2, peroxisome proliferator-activated receptors (PPAR)alpha and gamma, together with long and short forms of the prolactin receptor (PRLR). Singleton-bearing ewes were either nutrient restricted (3.2-3.8 MJ day(-1) metabolizable energy) or fed to appetite (8.7-9.9 MJ day(-1)) over the period of maximal placental growth, i.e. between 28 and 80 d gestation. After 80 d gestation, ewes were either fed to calculated requirements, (6.7-7.5 MJ day(-1)), or to appetite (8.0-10.9 MJ day(-1)). At term, offspring of nutrient-restricted ewes possessed more adipose tissue, an adaptation that was greatest in those born to mothers that fed to requirements in late gestation. This was accompanied by an increased mRNA abundance for UCP2 and PPARalpha, an adaptation not seen in mothers re-fed to appetite. Maternal nutrition had no effect on mRNA abundance for UCP1, PPARgamma, or PRLR. Irrespective of maternal nutrition, mRNA abundance for UCP1 was positively correlated with PPARgamma and the long and short forms of PRLR, indicating that these factors may act together to ensure that UCP1 abundance is maximized in the newborn. In conclusion, we have shown, for the first time, differential effects of maternal nutrition on key regulatory components of fetal fat metabolism.


Assuntos
Tecido Adiposo/fisiologia , Proteínas de Transporte/genética , Transtornos da Nutrição Fetal/fisiopatologia , Proteínas de Membrana/genética , PPAR alfa/genética , PPAR gama/genética , Receptores da Prolactina/genética , Tecido Adiposo/embriologia , Animais , Feminino , Feto/fisiopatologia , Canais Iônicos , Proteínas de Membrana Transportadoras/genética , Proteínas Mitocondriais/genética , RNA Mensageiro/metabolismo , Ovinos , Proteína Desacopladora 1 , Proteína Desacopladora 2
19.
J Endocrinol ; 184(2): 351-9, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15684343

RESUMO

A primary role of the prolactin receptor (PRLR) during fetal and postnatal development has been suggested to be the regulation of uncoupling protein (UCP) expression. We, therefore, determined whether: (1) the rate of loss of UCP1 from brown adipose tissue after birth was paralleled by the disappearance of PRLR; and (2) administration of either pituitary extract prolactin (PRL) containing a mixture of posttranslationally modified forms or its pseudophosphorylated form (S179D PRL) improved thermoregulation and UCP1 function over the first week of neonatal life. PRLR abundance was greatest in adipose tissue 6 h after birth before declining up to 30 days of age, a trend mirrored by first a gain and then a loss of UCP1. In contrast, in the liver--which does not possess UCPs--a postnatal decline in PRLR was not observed. Administration of PRL resulted in an acute increase in colonic temperature in conjunction with increased plasma concentrations of non-esterified fatty acids and, as a result, the normal postnatal decline in body temperature was delayed. S179D PRL at lower concentrations resulted in a transient rise in colonic temperature at both 2 and 6 days of age. In conclusion, we have demonstrated a close relationship between the ontogeny of UCP1 and the PRLR. Exogenous PRL administration elicits a thermogenic effect suggesting an important role for the PRLR in regulating UCP1 function.


Assuntos
Tecido Adiposo/metabolismo , Proteínas de Transporte/metabolismo , Proteínas de Membrana/metabolismo , Prolactina/farmacologia , Receptores da Prolactina/metabolismo , Ovinos/crescimento & desenvolvimento , Ovinos/metabolismo , Animais , Animais Recém-Nascidos , Regulação da Temperatura Corporal/efeitos dos fármacos , Colo/fisiologia , Ácidos Graxos não Esterificados/sangue , Feminino , Canais Iônicos , Fígado/metabolismo , Proteínas Mitocondriais , Prolactina/metabolismo , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/farmacologia , Proteína Desacopladora 1
20.
J Endocrinol ; 187(1): 81-8, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16214943

RESUMO

Many tissues undergo a rapid transition after birth, accompanied by dramatic changes in mitochondrial protein function. In particular, uncoupling protein (UCP) abundance increases at birth in the lung and adipose tissue, to then gradually decline, an adaptation that is important in enabling normal tissue function. Leptin potentially mediates some of these changes and is known to promote the loss of UCP1 from brown fat but its effects on UCP2 and related mitochondrial proteins (i.e. voltage-dependent anion channel (VDAC) and cytochrome c) in other tissues are unknown. We therefore determined the effects of once-daily jugular venous administration of ovine recombinant leptin on mitochondrial protein abundance as determined by immunoblotting in tissues that do (i.e. the brain and pancreas) and do not (i.e. liver and skeletal muscle) express UCP2. Eight pairs of 1-day-old lambs received either 100 mug leptin or vehicle daily for 6 days, before tissue sampling on day 7. Administration of leptin diminished UCP2 abundance in the pancreas, but not the brain. Leptin administration had no affect on the abundance of VDAC or cytochrome c in any tissue examined. In leptin-administered animals, but not controls, UCP2 abundance in the pancreas was positively correlated with VDAC and cytochrome c content, and UCP2 abundance in the brain with colonic temperature. In conclusion, leptin administration to neonatal lambs causes a tissue-specific loss of UCP2 from the pancreas. These effects may be important in the regulation of neonatal tissue development and potentially for optimising metabolic control mechanisms in later life.


Assuntos
Leptina/farmacologia , Proteínas Mitocondriais/metabolismo , Pâncreas/metabolismo , Animais , Animais Recém-Nascidos , Temperatura Corporal , Córtex Cerebral/química , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Colo/fisiologia , Citocromos c/análise , Citocromos c/metabolismo , Ácidos Graxos não Esterificados/sangue , Immunoblotting , Infusões Intravenosas , Canais Iônicos , Leptina/sangue , Fígado/química , Fígado/efeitos dos fármacos , Fígado/metabolismo , Proteínas de Membrana Transportadoras/análise , Proteínas de Membrana Transportadoras/metabolismo , Proteínas Mitocondriais/análise , Músculo Esquelético/química , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/metabolismo , Pâncreas/química , Pâncreas/efeitos dos fármacos , Proteínas Recombinantes/farmacologia , Ovinos , Estatísticas não Paramétricas , Proteína Desacopladora 2 , Canais de Ânion Dependentes de Voltagem/análise , Canais de Ânion Dependentes de Voltagem/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA