Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Adv Sci (Weinh) ; 10(35): e2305472, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37867217

RESUMO

Early life nutrition can reprogram development and exert long-term consequences on body weight regulation. In mice, maternal high-fat diet (HFD) during lactation predisposed male but not female offspring to diet-induced obesity when adult. Molecular and cellular changes in the hypothalamus at important time points are examined in the early postnatal life in relation to maternal diet and demonstrated sex-differential hypothalamic reprogramming. Maternal HFD in lactation decreased the neurotropic development of neurons formed at the embryo stage (e12.5) and impaired early postnatal neurogenesis in the hypothalamic regions of both males and females. Males show a larger increased ratio of Neuropeptide Y (NPY) to Pro-opiomelanocortin (POMC) neurons in early postnatal neurogenesis, in response to maternal HFD, setting an obese tone for male offspring. These data provide insights into the mechanisms by which hypothalamic reprograming by early life overnutrition contributes to the sex-dependent susceptibility to obesity in adult life in mice.


Assuntos
Dieta Hiperlipídica , Obesidade , Feminino , Camundongos , Animais , Masculino , Dieta Hiperlipídica/efeitos adversos , Obesidade/etiologia , Hipotálamo , Peso Corporal , Lactação
2.
Biomed Pharmacother ; 139: 111627, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33965728

RESUMO

Lipids excess from an uterine environment can increase free radicals production of and thus induce oxidative status imbalance, a key factor for progression of non-alcoholic fatty liver disease (NAFLD) in offspring. Food antioxidant components in maternal diet may play an important role in preventing offspring metabolic disorders. The objective of the study was to evaluate the effects of açaí pulp supplementation on maternal high-fat diet, by assessing activity and expression of antioxidant enzymes and biomarkers of oxidative stress in the liver. Female Fisher rats were divided into four groups and fed a control diet (C), a high-fat diet (HF), a control diet supplemented with açaí (CA) and a high-fat diet supplemented with açaí (HFA) before mating, during gestation and lactation. The effects of açaí supplementation on oxidative stress biomarkers and antioxidant enzymes expression were evaluated in dams and male offspring after weaning. HFA diet increased body weight in dams, however reduced absolute and relative liver weight. There was a reduction in liver biomarkers of oxidative stress, malondialdehyde and carbonyl protein, as well as in catalase, glutathione peroxidase and superoxide dismutase activity. In offspring, HFA diet reduced liver weight and increased Gpx1, Gpx4 and Sod1 mRNA expression. These results suggest that açaí is able to restore redox status, preventing oxidative damage in dams by a direct mechanism and to promote beneficial effects on expression of antioxidant defences related genes in offspring.


Assuntos
Antioxidantes/metabolismo , Euterpe/química , Fígado/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Animais , Biomarcadores/metabolismo , Peso Corporal/efeitos dos fármacos , Dieta Hiperlipídica , Suplementos Nutricionais , Feminino , Expressão Gênica/efeitos dos fármacos , Glutationa Peroxidase/metabolismo , Lactação , Fígado/efeitos dos fármacos , Fígado/enzimologia , Camundongos , Hepatopatia Gordurosa não Alcoólica , Tamanho do Órgão/efeitos dos fármacos , Fosfolipídeo Hidroperóxido Glutationa Peroxidase/metabolismo , Gravidez , Ratos , Ratos Endogâmicos F344 , Superóxido Dismutase-1/metabolismo , Glutationa Peroxidase GPX1
3.
Mol Metab ; 44: 101135, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33279727

RESUMO

OBJECTIVE: Amylin was found to regulate glucose and lipid metabolism by acting on the arcuate nucleus of the hypothalamus (ARC). Maternal high-fat diet (HFD) induces sex-specific metabolic diseases mediated by the ARC in offspring. This study was performed to explore 1) the effect of maternal HFD-induced alterations in amylin on the differentiation of hypothalamic neurons and metabolic disorders in male offspring and 2) the specific molecular mechanism underlying the regulation of amylin and its receptor in response to maternal HFD. METHODS: Maternal HFD and gestational hyper-amylin mice models were established to explore the role of hypothalamic amylin and receptor activity-modifying protein 3 (Ramp3) in regulating offspring metabolism. RNA pull-down, mass spectrometry, RNA immunoprecipitation, and RNA decay assays were performed to investigate the mechanism underlying the influence of maternal HFD on Ramp3 deficiency in the fetal hypothalamus. RESULTS: Male offspring with maternal HFD grew heavier and developed metabolic disorders, whereas female offspring with maternal HFD showed a slight increase in body weight and did not develop metabolic disorders compared to those exposed to maternal normal chow diet (NCD). Male offspring exposed to a maternal HFD had hyperamylinemia from birth until adulthood, which was inconsistent with offspring exposed to maternal NCD. Hyperamylinemia in the maternal HFD-exposed male offspring might be attributed to amylin accumulation following Ramp3 deficiency in the fetal hypothalamus. After Ramp3 knockdown in hypothalamic neural stem cells (htNSCs), amylin was found to fail to promote the differentiation of anorexigenic alpha-melanocyte-stimulating hormone-proopiomelanocortin (α-MSH-POMC) neurons but not orexigenic agouti-related protein-neuropeptide Y (AgRP-Npy) neurons. An investigation of the mechanism involved showed that IGF2BP1 could specifically bind to Ramp3 in htNSCs and maintain its mRNA stability. Downregulation of IGF2BP1 in htNSCs in the HFD group could decrease Ramp3 expression and lead to an impairment of α-MSH-POMC neuron differentiation. CONCLUSIONS: These findings suggest that gestational exposure to HFD decreases the expression of IGF2BP1 in the hypothalami of male offspring and destabilizes Ramp3 mRNA, which leads to amylin resistance. The subsequent impairment of POMC neuron differentiation induces sex-specific metabolic disorders in adulthood.


Assuntos
Diferenciação Celular , Dieta Hiperlipídica/efeitos adversos , Hipotálamo/metabolismo , Neurônios/metabolismo , Pró-Opiomelanocortina/metabolismo , Receptores de Polipeptídeo Amiloide de Ilhotas Pancreáticas/metabolismo , Proteína Relacionada com Agouti/metabolismo , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Peso Corporal , Feminino , Células HEK293 , Humanos , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurogênese , Neuropeptídeo Y/metabolismo , Gravidez , Proteínas de Ligação a RNA/metabolismo , Proteína 3 Modificadora da Atividade de Receptores/genética , Proteína 3 Modificadora da Atividade de Receptores/metabolismo , Células-Tronco , alfa-MSH/metabolismo
4.
Front Endocrinol (Lausanne) ; 11: 591559, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33324346

RESUMO

Excessive dietary fat intake has extensive impacts on several physiological systems and can lead to metabolic and nonmetabolic disease. In animal models of ingestion, exposure to a high fat diet during pregnancy predisposes offspring to increase intake of dietary fat and causes increase in weight gain that can lead to obesity, and without intervention, these physiological and behavioral consequences can persist for several generations. The hypothalamus is a region of the brain that responds to physiological hunger and fullness and contains orexigenic neuropeptide systems that have long been associated with dietary fat intake. The past fifteen years of research show that prenatal exposure to a high fat diet increases neurogenesis of these neuropeptide systems in offspring brain and are correlated to behavioral changes that induce a pro-consummatory and obesogenic phenotype. Current research has uncovered several potential molecular mechanisms by which excessive dietary fat alters the hypothalamus and involve dietary fatty acids, the immune system, gut microbiota, and transcriptional and epigenetic changes. This review will examine the current knowledge of dietary fat-associated changes in the hypothalamus and the potential pathways involved in modifying the development of orexigenic peptide neurons that lead to changes in ingestive behavior, with a special emphasis on inflammation by chemokines.


Assuntos
Gorduras na Dieta/efeitos adversos , Ingestão de Alimentos , Transtornos da Nutrição Fetal/patologia , Hipotálamo/patologia , Mediadores da Inflamação/metabolismo , Inflamação/patologia , Efeitos Tardios da Exposição Pré-Natal/patologia , Animais , Feminino , Transtornos da Nutrição Fetal/etiologia , Transtornos da Nutrição Fetal/metabolismo , Humanos , Hipotálamo/metabolismo , Inflamação/etiologia , Inflamação/metabolismo , Neuropeptídeos/metabolismo , Gravidez , Efeitos Tardios da Exposição Pré-Natal/etiologia , Efeitos Tardios da Exposição Pré-Natal/metabolismo
5.
J Nutr Biochem ; 75: 108257, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31710935

RESUMO

Maternal high-fat diet (HFD) overfeeding pre- and during pregnancy and lactation may 'program' a 'diabesity' predisposition in the offspring, for inconclusive reasons. Acquired alterations of the hypothalamic promoter methylation and mRNA expression of the satiety neurohormone Pomc are possibly of critical importance here. We investigated within one developmental approach, including male and female rats, the sex-specific DNA methylation pattern and corresponding mRNA expression of both Pomc and its endogenous functional antagonist Agrp in the hypothalamus of adult HFD offspring. Obesity and diabetic disturbances occurred in both male and female HFD offspring, accompanied by altered Pomc promoter methylation pattern. However, this was not related to significant Pomc mRNA expression alterations. In contrast, male-specific alterations of Agrp promoter methylation were found, even associated with reduced mRNA expression of this orexigenic/anabolic Pomc antagonist. In conclusion, acquired epigenetic alterations of the hypothalamic Agrp-Pomc system hardly explain the 'diabesity' phenotype in HFD offspring, while distinct vulnerability and functionality of Agrp promoter and related genomic regions methylation should be further investigated.


Assuntos
Proteína Relacionada com Agouti/genética , Diabetes Mellitus/genética , Epigênese Genética , Hipotálamo/metabolismo , Obesidade/genética , Pró-Opiomelanocortina/genética , Animais , Glicemia/análise , Composição Corporal , Metilação de DNA , Complicações do Diabetes , Dieta Hiperlipídica , Feminino , Masculino , Fenômenos Fisiológicos da Nutrição Materna , Neuropeptídeos/química , Hipernutrição/genética , Fenótipo , Gravidez , Prenhez , Efeitos Tardios da Exposição Pré-Natal/genética , Regiões Promotoras Genéticas , Ratos , Ratos Wistar , Fatores Sexuais
6.
Br J Nutr ; 121(12): 1345-1356, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30940241

RESUMO

Perinatal maternal high-fat diet (HFD) increases susceptibility to obesity and fatty liver diseases in adult offspring, which can be attenuated by the potent hypolipidaemic action of fish oil (FO), an n-3 PUFA source, during adult life. Previously, we described that adolescent HFD offspring showed resistance to FO hypolipidaemic effects, although FO promoted hepatic molecular changes suggestive of reduced lipid accumulation. Here, we investigated whether this FO intervention only during the adolescence period could affect offspring metabolism in adulthood. Then, female Wistar rats received isoenergetic, standard (STD: 9 % fat) or high-fat (HFD: 28·6 % fat) diet before mating, and throughout pregnancy and lactation. After weaning, male offspring received the standard diet; and from 25 to 45 d old they received oral administration of soyabean oil or FO. At 150 d old, serum and hepatic metabolic parameters were evaluated. Maternal HFD adult offspring showed increased body weight, visceral adiposity, hyperleptinaemia and decreased hepatic pSTAT3/STAT3 ratio, suggestive of hepatic leptin resistance. FO intake only during the adolescence period reduced visceral adiposity and serum leptin, regardless of maternal diet. Maternal HFD promoted dyslipidaemia and hepatic TAG accumulation, which was correlated with reduced hepatic carnitine palmitoyl transferase-1a content, suggesting lipid oxidation impairment. FO intake did not change serum lipids; however, it restored hepatic TAG content and hepatic markers of lipid oxidation to STD offspring levels. Therefore, we concluded that FO intake exclusively during adolescence programmed STD offspring and reprogrammed HFD offspring male rats to a healthier metabolic phenotype in adult life, reducing visceral adiposity, serum leptin and hepatic TAG content in offspring adulthood.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Suplementos Nutricionais , Dislipidemias/prevenção & controle , Óleos de Peixe/administração & dosagem , Efeitos Tardios da Exposição Pré-Natal/prevenção & controle , Animais , Dislipidemias/etiologia , Ácidos Graxos Ômega-3/metabolismo , Feminino , Gordura Intra-Abdominal/metabolismo , Leptina/sangue , Fígado/metabolismo , Masculino , Fenômenos Fisiológicos da Nutrição Materna , Gravidez , Efeitos Tardios da Exposição Pré-Natal/etiologia , Ratos , Ratos Wistar , Triglicerídeos/metabolismo
7.
J Nutr Biochem ; 67: 28-35, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30849557

RESUMO

Maternal overnutrition around reproduction has been shown to increase the offspring's risk for "diabesity," mediated by altered hypothalamic neuropeptide expression. In this report, a possible contribution of altered hypothalamic sensing capacity for the peripheral satiety signals glucose, insulin and leptin will be addressed, taking into account potential sex differences. Specifically, we evaluated the effects a maternal high-fat diet (HFD) overfeeding has in rats pre- and during pregnancy and lactation on the hypothalamic gene expression patterns of insulin and leptin receptors (InsR, ObRb) and glucose transporter 3 (Glut3) as well as DNA methylation in the offspring at adult age (day 200 of life). Maternal HFD consumption resulted in a metabolic syndrome phenotype, i.e., obesity, hyperleptinemia, hyperinsulinemia, impaired glucose tolerance and increased homeostatic model assessment of insulin resistance. Interestingly, in turn, insulin resistance was more pronounced in male offspring, accompanied by decreased hypothalamic InsR-mRNA. This was linked with hypermethylation of an activating transcription factor binding site within the hypothalamic InsR promoter. The degree of methylation correlated inversely with respective InsR expression, while InsR expression itself was inversely related to phenotypic "diabesity." Expression of ObRb and Glut3 mRNA was not significantly changed. In conclusion, sex-specific alterations of hypothalamic InsR expression and DNA promoter methylation in adult offspring of HFD-overfed dams may lead to hypothalamic insulin resistance and "diabesity," with males predisposed to this epigenetic malprogramming.


Assuntos
Metilação de DNA , Dieta Hiperlipídica/efeitos adversos , Hipotálamo/fisiologia , Receptor de Insulina/genética , Adiposidade , Animais , Feminino , Regulação da Expressão Gênica , Intolerância à Glucose , Transportador de Glucose Tipo 3/genética , Masculino , Fenômenos Fisiológicos da Nutrição Materna , Obesidade/etiologia , Gravidez , Efeitos Tardios da Exposição Pré-Natal , Regiões Promotoras Genéticas , Receptor de Insulina/metabolismo , Receptores para Leptina/genética , Fatores Sexuais , Aumento de Peso/efeitos dos fármacos
8.
J Basic Clin Physiol Pharmacol ; 29(2): 185-194, 2018 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-28988221

RESUMO

BACKGROUND: Maternal high fat diet has been implicated in the aetiology of metabolic diseases in their offspring. The hypolipidaemic actions of Cocos nucifera water improve metabolic indices of dams consuming a high fat diet during gestation. This study investigated the effects of C. nucifera water on metabolism of offspring of dams exposed to high fat diet during gestation. METHODS: Four groups of pregnant Wistar rat dams (n=6) were treated orally from Gestation Day (GD) 1 to GD 21 as follows: standard rodent feed+10 mL/kg distilled water (Control), standard rodent feed+10 mL/kg C. nucifera water, high fat feed+10 mL/kg distilled water (high fat diet), and high fat feed+10 mL/kg C. nucifera water (high fat diet+C. nucifera water). The feeds were given ad libitum and all dams received standard rodent feed after parturition. Fasting blood glucose was measured in offspring before being euthanized on Postnatal Day (PND) 120. Serum insulin, leptin, lipid profile and liver enzymes were measured. RESULTS: Serum total cholesterol (TC), insulin, alanine transaminase (ALT) and alkaline phosphatase levels were significantly increased (p<0.05) in high fat diet offspring compared with controls. Similar changes were not observed in high fat diet+C. nucifera water offspring. CONCLUSIONS: Results suggest that the adverse effects of maternal high fat diet on offspring's metabolism can be ameliorated by C. nucifera water.


Assuntos
Cocos/química , Dieta Hiperlipídica/efeitos adversos , Metabolismo/efeitos dos fármacos , Extratos Vegetais/farmacologia , Água/farmacologia , Animais , Glicemia/efeitos dos fármacos , Feminino , Insulina/metabolismo , Leptina/metabolismo , Metabolismo dos Lipídeos/efeitos dos fármacos , Obesidade/metabolismo , Gravidez , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Ratos , Ratos Wistar
9.
Mol Nutr Food Res ; 60(11): 2493-2504, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27342757

RESUMO

SCOPE: Maternal high-fat diet (HFD) promotes obesity and metabolic disturbances in offspring at weaning and adult life. We investigated metabolic consequences of maternal HFD in adolescent rat offspring and the potential benefic effects of fish oil (FO) (n-3 polyunsaturated fatty acid source). METHODS AND RESULTS: Female rats received isocaloric, standard diet (STD: 9% fat) or HFD (28.6%) before mating, and throughout pregnancy and lactation. After weaning, male offspring received standard diet and, from 25th to 45th day, received oral administration of soybean oil (SO) or FO. HFD offspring showed higher body weight and adiposity, which was not attenuated by FO. In STD offspring, FO reduced serum triglyceride and cholesterol, as expected, but not in HFD offspring. Liver of HFD offspring groups showed increased free cholesterol and FO-treated HFD group showed lower expression of Abcg8, suggesting decreased cholesterol biliary excretion. HFD offspring presented higher hepatic expression of lipogenic markers, Srebf1 mRNA and acetyl CoA carboxylase (ACC). Serum n-3 PUFA were decreased in FO-treated HFD compared to FO-treated STD offspring, which may explain the reduced hypolipidemic FO effect. CONCLUSION: Maternal HFD impaired the ability of FO to reduce adiposity and serum lipids in adolescent offspring, suggesting a potential predisposition to future development of metabolic disorders.


Assuntos
Óleos de Peixe/farmacologia , Hipolipemiantes/farmacologia , Metabolismo dos Lipídeos/efeitos dos fármacos , Adiposidade/efeitos dos fármacos , Adolescente , Animais , Colesterol/sangue , Dieta Hiperlipídica , Gorduras Insaturadas na Dieta/metabolismo , Ácidos Graxos Ômega-3/farmacologia , Feminino , Óleos de Peixe/administração & dosagem , Humanos , Lactação/efeitos dos fármacos , Fígado/metabolismo , Obesidade/metabolismo , Gravidez , Efeitos Tardios da Exposição Pré-Natal , Ratos , Triglicerídeos/sangue , Desmame
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA