Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 383
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Dev Orig Health Dis ; 12(2): 203-219, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33349289

RESUMO

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.


Assuntos
Envelhecimento , Retardo do Crescimento Fetal/fisiopatologia , Coração/embriologia , Coração/fisiopatologia , Imageamento por Ressonância Magnética/métodos , Animais , Feminino , Humanos , Gravidez
2.
Br J Nutr ; 122(2): 141-151, 2019 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-31345278

RESUMO

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.


Assuntos
Dieta com Restrição de Proteínas/efeitos adversos , Lactação/fisiologia , Leite/química , Animais , Ácido Araquidônico/análise , Peso Corporal , Encéfalo/crescimento & desenvolvimento , Ácidos Docosa-Hexaenoicos/análise , Feminino , Hipocampo/química , Fígado/crescimento & desenvolvimento , Glândulas Mamárias Animais/anatomia & histologia , Glândulas Mamárias Animais/crescimento & desenvolvimento , Fenômenos Fisiológicos da Nutrição Materna , Tamanho do Órgão , Gravidez , Ratos , Ratos Wistar
3.
Hippocampus ; 28(1): 18-30, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28843045

RESUMO

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.


Assuntos
Dieta com Restrição de Proteínas/efeitos adversos , Hipocampo/crescimento & desenvolvimento , Hipocampo/fisiopatologia , Deficiências da Aprendizagem/fisiopatologia , Transtornos da Memória/fisiopatologia , Fenômenos Fisiológicos da Nutrição Pré-Natal , Animais , Modelos Animais de Doenças , Feminino , Hipocampo/patologia , Lactação , Deficiências da Aprendizagem/etiologia , Deficiências da Aprendizagem/patologia , Masculino , Desnutrição/patologia , Desnutrição/fisiopatologia , Desnutrição/psicologia , Transtornos da Memória/etiologia , Transtornos da Memória/patologia , Neurônios/patologia , Gravidez , Distribuição Aleatória , Ratos Wistar , Aprendizagem Espacial/fisiologia , Memória Espacial/fisiologia
4.
J Dev Orig Health Dis ; 9(2): 137-142, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29017630

RESUMO

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.


Assuntos
Aorta/diagnóstico por imagem , Pressão Sanguínea/fisiologia , Retardo do Crescimento Fetal/diagnóstico por imagem , Animais , Aorta/fisiopatologia , Feminino , Retardo do Crescimento Fetal/fisiopatologia , Masculino , Papio , Gravidez
5.
Domest Anim Endocrinol ; 60: 67-74, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28527530

RESUMO

We previously reported that maternal overnutrition and obesity (MO) throughout pregnancy and lactation in sheep (MOF0) decreases term fetal pancreatic ß-cell numbers and increases perirenal adiposity producing hyperphagia, increased adiposity and insulin resistance in adult female offspring (MOF1) fed ad libitum. Pregnant female MOF1 exhibited increased blood glucose from mid to late gestation vs control F1 (CTRF1) though both groups ate only to NRC recommendations. MOF1 ewes delivered female offspring (F2) who like their MOF1 mothers exhibited increased abdominal adiposity and absent neonatal leptin surge. In the current work, we determined if adult MOF2 exhibited metabolic syndrome components when fed ad libitum. After weaning, MOF2 males (n = 5), MOF2 females (n = 6), CTRF2 males (n = 5), and CTRF2 females (n = 6) were fed to NRC requirements until 19 mo followed by 12-wk ad libitum feeding. Body weight and % fat increased (P < 0.01) in all F2 during this feeding trial. MOF2 males were heavier (P < 0.01) than CTRF2 males and females, and MOF2 females throughout the trial. By wk 8, baseline blood glucose concentrations increased (P < 0.001) in MOF2 females, but not other groups, remaining elevated throughout the trial. Baseline insulin was similar through wk 6, increasing (P < 0.05) at wk 8 in MOF2 females only. MOF2 female insulin returned to CTRF2 female levels during wk 10 and 12. The progressive increase of plasma glucose on wk 8 in association with increased insulin in MOF2 females but not other groups demonstrated a diet-induced increase (P < 0.001) in MOF2 female insulin resistance. The subsequent decline in insulin during wk 10 and 12 despite elevated glucose in MOF2 females is consistent with a decrease in glucose-stimulated pancreatic ß-cell function. These data indicate that ad libitum feeding exceeds the pancreatic secretory response predisposing MOF2 females to hyperglycemia. Furthermore, there was a sex difference where MOF2 males increased body mass and MOF2 females displayed insulin/glucose dysregulation.


Assuntos
Fenômenos Fisiológicos da Nutrição Animal , Síndrome Metabólica/veterinária , Obesidade/veterinária , Hipernutrição , Fenômenos Fisiológicos da Nutrição Pré-Natal , Doenças dos Ovinos/etiologia , Ração Animal , Animais , Glicemia , Composição Corporal , Feminino , Insulina/sangue , Masculino , Síndrome Metabólica/etiologia , Gravidez , Efeitos Tardios da Exposição Pré-Natal , Ovinos , Aumento de Peso
6.
Int J Obes (Lond) ; 41(8): 1299-1302, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28337030

RESUMO

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.


Assuntos
Fígado Gorduroso/induzido quimicamente , Desenvolvimento Fetal/efeitos dos fármacos , Glucocorticoides/administração & dosagem , Glucocorticoides/efeitos adversos , Exposição Materna/efeitos adversos , Papio , Prenhez , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Animais , Betametasona/administração & dosagem , Betametasona/efeitos adversos , Betametasona/farmacocinética , Peso ao Nascer , Metilação de DNA , Modelos Animais de Doenças , Fígado Gorduroso/diagnóstico por imagem , Feminino , Glucocorticoides/farmacocinética , Metabolismo dos Lipídeos , Fígado/diagnóstico por imagem , Fígado/metabolismo , Espectroscopia de Ressonância Magnética , Masculino , Pericárdio/diagnóstico por imagem , Pericárdio/metabolismo , Gravidez
7.
Nutr Metab Cardiovasc Dis ; 26(9): 786-96, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27146364

RESUMO

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.


Assuntos
Restrição Calórica , Feto/metabolismo , Fígado/metabolismo , Desnutrição/metabolismo , Fenômenos Fisiológicos da Nutrição Materna , Metaboloma , Adaptação Fisiológica , Animais , Biomarcadores/sangue , Carnitina/análogos & derivados , Carnitina/sangue , Cromatografia Líquida , Modelos Animais de Doenças , Ácidos Graxos não Esterificados/sangue , Feminino , Idade Gestacional , Gluconeogênese , Humanos , Desnutrição/fisiopatologia , Espectrometria de Massas , Metabolômica/métodos , Papio , Fosfolipídeos/sangue , Gravidez
8.
Br J Nutr ; 115(3): 538-46, 2016 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-26608475

RESUMO

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.


Assuntos
Dieta Hiperlipídica , Leite/química , Obesidade/metabolismo , Acetiltransferases/metabolismo , Tecido Adiposo/metabolismo , Animais , Ácido Araquidônico/metabolismo , Glicemia/metabolismo , Dessaturase de Ácido Graxo Delta-5 , Ácidos Docosa-Hexaenoicos/metabolismo , Ácido Eicosapentaenoico/metabolismo , Ácidos Graxos Dessaturases/metabolismo , Elongases de Ácidos Graxos , Feminino , Lactação , Leptina/metabolismo , Fígado/metabolismo , Masculino , Glândulas Mamárias Animais/metabolismo , Fenômenos Fisiológicos da Nutrição Materna , Tamanho do Órgão , Gravidez , Efeitos Tardios da Exposição Pré-Natal , Ratos , Ratos Wistar , Estearoil-CoA Dessaturase/metabolismo
9.
Age (Dordr) ; 37(3): 9774, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25953670

RESUMO

Glucocorticoids are pleiotropic regulators of multiple cell types with critical roles in physiological systems that change across the life-course. Although glucocorticoids have been associated with aging, available data on the aging trajectory in basal circulating glucocorticoids are conflicting. A literature search reveals sparse life-course data. We evaluated (1) the profile of basal circulating corticosterone across the life-course from weaning (postnatal day-PND 21), young adult PND 110, adult PND 450, mature adult PND 650 to aged phase PND 850 in a well-characterized homogeneous rat colony to determine existence of significant changes in trajectory in the second half of life; (2) sex differences; and (3) whether developmental programming of offspring by exposure to maternal obesity during development alters the later-life circulating corticosterone trajectory. We identified (1) a fall in corticosterone between PND 450 and 650 in both males and females (p < 0.05) and (2) higher female than male concentrations (p < 0.05). (3) Using our five life-course time-point data set, corticosterone fell at a similar age but from higher levels in male and female offspring of obese mothers. In all four groups studied, there was a second half of life fall in corticosterone. Higher corticosterone levels in offspring of obese mothers may play a role in their shorter life-span, but the age-associated fall occurs at a similar time to control offspring. Although even more life-course time-points would be useful, a five life-course time-point analysis provides important new information on normative and programmed aging of circulating corticosterone.


Assuntos
Envelhecimento/metabolismo , Corticosterona/metabolismo , Obesidade/metabolismo , Animais , Feminino , Masculino , Ratos , Ratos Wistar , Caracteres Sexuais
10.
Int J Obes (Lond) ; 39(4): 695-701, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25354845

RESUMO

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.


Assuntos
Fenômenos Fisiológicos da Nutrição Animal/fisiologia , Glicemia/metabolismo , Leptina/metabolismo , Obesidade/patologia , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Ração Animal , Animais , Feminino , Teste de Tolerância a Glucose , Masculino , Desnutrição , Fenômenos Fisiológicos da Nutrição Materna/fisiologia , Hipernutrição , Gravidez , Ovinos
11.
Int J Obes (Lond) ; 39(4): 549-56, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25504042

RESUMO

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.


Assuntos
Fertilidade , Obesidade/metabolismo , Hipernutrição/metabolismo , Estresse Oxidativo , Complicações na Gravidez/metabolismo , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Fenômenos Fisiológicos da Nutrição Animal , Animais , Animais Recém-Nascidos , Dieta Hiperlipídica , Feminino , Infertilidade/etiologia , Lactação , Masculino , Fenômenos Fisiológicos da Nutrição Materna , Obesidade/complicações , Obesidade/etiologia , Hipernutrição/complicações , Gravidez , Ratos , Ratos Wistar , Fatores Sexuais
12.
Physiol Behav ; 140: 89-95, 2015 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-25496979

RESUMO

Maternal low protein (MLP) diets in pregnancy and lactation impair offspring brain development and modify offspring behavior. We hypothesized multigenerational passage of altered behavioral outcomes as has been demonstrated following other developmental programming challenges. We investigated potential multigenerational effects of MLP in rat pregnancy and/or lactation on offspring risk assessment behavior. Founder generation mothers (F0) ate 20% casein (C) or restricted (R) 10% casein diet, providing four groups: CC, RR, CR, and RC (first letter pregnancy, second letter lactation diet) to evaluate offspring (F1) effects influenced by MLP in F0. On postnatal day (PND 250), F1 males were mated to non-colony siblings producing F2. On PND 90, F2 females (in diestrous) and F2 males were tested in the elevated plus maze (EPM) and open field. Corticosterone was measured at PND 110. Female but not male CR and RC F2 made more entries and spent more time in EPM open arms than CC females. Overall activity was unchanged as observed in male F1 fathers. There were no open field differences in F2 of either sex, indicating that multigenerational MLP effects are due to altered risk assessment, not locomotion. MLP in pregnancy reduced F1 male and F2 female corticosterone. We conclude that MLP in pregnancy and/or lactation increases the innate tendency to explore novel environments in F2 females via the paternal linage, suggesting lower levels of caution and/or higher impulsiveness to explore unknown spaces. Further studies will be necessary to identify the epigenetic modifications in the germ line through the paternal linage.


Assuntos
Deficiências do Desenvolvimento/etiologia , Deficiências do Desenvolvimento/genética , Dieta com Restrição de Proteínas/efeitos adversos , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia , Caracteres Sexuais , Fatores Etários , Análise de Variância , Animais , Corticosterona , Comportamento Exploratório/fisiologia , Feminino , Lactação , Masculino , Aprendizagem em Labirinto/fisiologia , Gravidez , Ratos , Medição de Risco , Assunção de Riscos
13.
Int J Obes (Lond) ; 39(4): 712-9, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23949616

RESUMO

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.


Assuntos
Lactação/metabolismo , Leptina/sangue , Obesidade/metabolismo , Prenhez , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Adiposidade , Fenômenos Fisiológicos da Nutrição Animal , Animais , Glicemia/metabolismo , Dieta Hiperlipídica , Feminino , Resistência à Insulina/fisiologia , Masculino , Fenômenos Fisiológicos da Nutrição Materna , Condicionamento Físico Animal , Gravidez , Ratos , Ratos Wistar , Desmame
14.
Int J Obes (Lond) ; 38(2): 224-30, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23748190

RESUMO

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.


Assuntos
Adipócitos/metabolismo , Tecido Adiposo Marrom/patologia , Tecido Adiposo Branco/patologia , Fenômenos Fisiológicos da Nutrição Animal , Fenômenos Fisiológicos da Nutrição Materna , Fenômenos Fisiológicos da Nutrição Pré-Natal , Adipogenia , Animais , Diferenciação Celular , Proteínas de Ligação a Ácido Graxo/metabolismo , Feminino , Desenvolvimento Fetal , Feto , Regulação da Expressão Gênica no Desenvolvimento , Idade Gestacional , Humanos , Immunoblotting , Recém-Nascido , Masculino , PPAR alfa/metabolismo , Papio , Gravidez , Fatores Sexuais
15.
Br J Nutr ; 111(4): 616-24, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24124655

RESUMO

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.


Assuntos
Adiposidade/efeitos dos fármacos , Dieta com Restrição de Proteínas/efeitos adversos , Proteínas Alimentares/administração & dosagem , Sacarose Alimentar/efeitos adversos , Obesidade/etiologia , Efeitos Tardios da Exposição Pré-Natal , Fenômenos Fisiológicos da Nutrição Pré-Natal , Animais , Colesterol/sangue , Proteínas Alimentares/farmacologia , Feminino , Insulina/sangue , Leptina/sangue , Obesidade/sangue , Gravidez , Ratos , Fatores Sexuais , Triglicerídeos/sangue
16.
Growth Horm IGF Res ; 23(5): 187-92, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23911858

RESUMO

OBJECTIVE: The IGF system exerts systemic and local actions during development. We previously demonstrated that fetal cerebral cortical IGF1 is reduced at 0.5 gestation in our IUGR baboon nonhuman primate model. We hypothesized that by term protein expression of several key IGF system stimulatory peptide pathway components and downstream nutrient signaling effectors of IGF, mammalian target of rapamycin (mTOR) and S6, would decrease, indicating reduced cellular nutrient uptake and protein synthesis. DESIGN: We fed 7 control baboons ad libitum while 6 baboons ate a globally reduced diet (70% of feed eaten by controls) from 0.16 gestation through pregnancy that produces IUGR. Fetuses were removed at Cesarean section at 0.9 gestation. Frontal cortex sections were stained for IGFI, IGFII, IGFRI, IGFR2, IGFBP2, 3, 5 and 6, and mTOR and ribosomal protein S6 and double stained with NeuN a neuron-specific nuclear antigen. RESULTS: All proteins stained neuronal cytoplasm except IGFRI which showed only glial cell cytoplasmic and blood vessel staining. IUGR fetuses showed decreased frontal cortical immunoreactive IGFI, IGFII, IGFRI, IGFBP2, 5 and 6, and mTOR and S6 (p < 0.05). IGFBP3 increased (p < 0.05) and IGFR2 was unchanged (p > 0.05). There were no differences between male and female fetal brains. CONCLUSIONS: When fetal nutrient availability is decreased, IUGR down regulates the IGF system and its mTOR signaling pathway in the fetal frontal cortex coincident with slowed growth. These findings emphasize the importance of the local tissue IGF system in fetal primate brain development.


Assuntos
Retardo do Crescimento Fetal/metabolismo , Lobo Frontal/metabolismo , Proteínas de Ligação a Fator de Crescimento Semelhante a Insulina/metabolismo , Somatomedinas/metabolismo , Nascimento a Termo/metabolismo , Animais , Regulação para Baixo , Feminino , Retardo do Crescimento Fetal/patologia , Feto/metabolismo , Lobo Frontal/embriologia , Idade Gestacional , Masculino , Papio , Gravidez , Transdução de Sinais
17.
J Dev Orig Health Dis ; 4(1): 3-9, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23525085

RESUMO

Developmental programming can be defined as a response to a specific challenge to the mammalian organism during a critical developmental time window that alters the trajectory of development with persistent effects on offspring phenotype and predisposition to future illness. We focus on the need for studies in relevant, well-characterized animal models in the context of recent research discoveries on the challenges, mechanisms and outcomes of developmental programming. We discuss commonalities and differences in general principles of developmental programming as they apply to several species, including humans. The consequences of these differences are discussed. Obesity, metabolic disorders and cardiovascular diseases are associated with the highest percentage of morbidity and mortality worldwide. Although many of the causes are associated with lifestyle, high-energy diets and lack of physical activity, recent evidence has linked developmental programming to the epidemic of metabolic diseases. A better understanding of comparative systems physiology of mother, fetus and neonate using information provided by rapid advances in molecular biology has the potential to improve the lifetime health of future generations by providing better women's health, diagnostic tools and preventative and therapeutic interventions in individuals exposed during their development to programming influences.


Assuntos
Crescimento e Desenvolvimento/fisiologia , Doenças Metabólicas/epidemiologia , Doenças Metabólicas/etiologia , Modelos Animais , Efeitos Tardios da Exposição Pré-Natal/epidemiologia , Efeitos Tardios da Exposição Pré-Natal/etiologia , Animais , Feminino , Humanos , Doenças Metabólicas/tratamento farmacológico , Doenças Metabólicas/prevenção & controle , Camundongos , Gravidez , Especificidade da Espécie
18.
Int J Obes (Lond) ; 37(4): 568-75, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22614057

RESUMO

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.


Assuntos
Adipogenia/genética , Desenvolvimento Fetal/genética , MicroRNAs/metabolismo , Músculo Esquelético/metabolismo , Obesidade/metabolismo , Animais , Dieta , Regulação para Baixo , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Fenômenos Fisiológicos da Nutrição Materna , Músculo Esquelético/embriologia , Obesidade/genética , Gravidez , Reação em Cadeia da Polimerase em Tempo Real , Carneiro Doméstico
19.
J Anim Sci ; 90(7): 2201-10, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22266999

RESUMO

Maternal nutrient restriction leads to alteration in fetal adipose tissue, and offspring from obese mothers have an increased risk of developing obesity. We hypothesized that maternal obesity increases fetal adipogenesis. Multiparous ewes (Columbia/Rambouillet cross 3 to 5 yr of age) carrying twins were assigned to a diet of 100% (Control; CON; n = 4) or 150% (Obese; OB, n = 7) of NRC maintenance requirements from 60 d before conception until necropsy on d 135 of gestation. Maternal and fetal plasma were collected and stored at -80°C for glucose and hormone analyses. Fetal measurements were made at necropsy, and perirenal, pericardial, and subcutaneous adipose tissues were collected from 7 male twin fetuses per group and snap frozen at -80°C. Protein and mRNA expression of fatty acid translocase [cluster of differentiation (CD) 36], fatty acid transport proteins (FATP) 1 and 4, insulin-sensitive glucose transporter (GLUT-4), fatty acid synthase (FASN), and acetyl-coA carboxylase (ACC) was evaluated. Fetal weight was similar, but fetal carcass weight (FCW) was reduced (P < 0.05) in OB versus CON fetuses. Pericardial and perirenal adipose tissue weights were increased (P < 0.05) as a percentage of FCW in OB versus CON fetuses, as was subcutaneous fat thickness (P < 0.001). Average adipocyte diameter was greater (P < 0.01) in the perirenal fat and the pericardial fat (P = 0.06) in OB fetuses compared with CON fetuses. Maternal plasma showed no difference (P > 0.05) in glucose or other hormones, fetal plasma glucose was similar (P = 0.42), and cortisol, IGF-1, and thyroxine were reduced (P ≤ 0.05) in OB fetuses compared with CON fetuses. Protein and mRNA expression of CD 36, FATP 1 and 4, and GLUT-4 were increased (P ≤ 0.05) in all fetal adipose depots in OB versus CON fetuses. The mRNA expression of FASN and ACC was increased (P < 0.05) in OB vs. CON fetuses in all 3 fetal adipose tissue depots. Fatty acid concentrations were increased (P = 0.01) in the perirenal depot of OB versus CON fetuses, and specific fatty acid concentrations were altered (P < 0.05) in subcutaneous and pericardial adipose tissue because of maternal obesity. In conclusion, maternal obesity was associated with increased fetal adiposity, increased fatty acid and glucose transporters, and increased expression of enzymes mediating fatty acid biosynthesis in adipose depots. These alterations, if maintained into the postnatal period, could predispose the offspring to later obesity and metabolic disease.


Assuntos
Tecido Adiposo/metabolismo , Proteínas de Transporte de Ácido Graxo/metabolismo , Feto/metabolismo , Proteínas Facilitadoras de Transporte de Glucose/metabolismo , Obesidade/metabolismo , Doenças dos Ovinos/metabolismo , Tecido Adiposo/química , Alcaloides , Ração Animal , Animais , Dieta , Proteínas de Transporte de Ácido Graxo/genética , Ácidos Graxos/química , Ácidos Graxos/metabolismo , Feminino , Desenvolvimento Fetal , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteínas Facilitadoras de Transporte de Glucose/genética , Masculino , Gravidez , Reação em Cadeia da Polimerase em Tempo Real , Ovinos
20.
Int J Dev Neurosci ; 30(2): 75-81, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22239918

RESUMO

We studied the effects of maternal high fat diet (HFD, 25% calories from fat administered before and during pregnancy and lactation) and dietary intervention (switching dams from HFD to control diet) at different periconceptional periods on male offspring anxiety related behavior, exploration, learning, and motivation. From weaning at postnatal day (PND) 21, female subjects produced to be the mothers in the study received either control diet (CTR - 5% calories from fat), HFD through pregnancy and lactation (MO), HFD during PNDs 21-90 followed by CTR diet (pre-gestation (PG) intervention) or HFD from PND 21 to 120 followed by CTR diet (gestation and lactation (G) intervention) and bred at PND 120. At 19 days of gestation maternal serum corticosterone was increased in MO and the PG and G dams showed partial recovery with intermediate levels. In offspring, no effects were found in the elevated plus maze test. In the open field test, MO and G offspring showed increase zone entries, displaying less thigmotaxis; PG offspring showed partial recuperation of this behavior. During initial operant conditioning MO, PG and G offspring displayed decreased approach behavior with subsequent learning impairment during the acquisition of FR-1 and FR-5 operant conditioning for sucrose reinforcement. Motivation during the progressive ratio test increased in MO offspring; PG and G intervention recuperated this behavior. We conclude that dietary intervention can reverse negative effects of maternal HFD and offspring outcomes are potentially due to elevated maternal corticosterone.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Comportamento Exploratório/fisiologia , Deficiências da Aprendizagem/dietoterapia , Motivação/fisiologia , Obesidade/fisiopatologia , Efeitos Tardios da Exposição Pré-Natal/dietoterapia , Animais , Animais Recém-Nascidos , Modelos Animais de Doenças , Feminino , Deficiências da Aprendizagem/fisiopatologia , Masculino , Obesidade/complicações , Obesidade/dietoterapia , Gravidez , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia , Ratos , Ratos Wistar
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...