Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 136
Filtrar
1.
Neurochem Int ; 162: 105454, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36462683

RESUMO

It is well known that overnutrition, overweight, and obesity in children can modulate brain mechanisms of plasticity, monoaminergic systems, and mitochondrial function. The immediate effect of overnutrition during the developmental period has not been thoroughly examined in rats until the present. This study sought to evaluate the impact on adult rats of early life overfeeding and fluoxetine treatment from post-natal day 1 (PND1) to post-natal day 21 (PND21) relative to mitochondrial function, oxidative balance, and expression of specific monoaminergic genes in the hippocampus. The following were evaluated: mitochondrial function markers, oxidative stress biomarkers, dopamine-and serotonin-related genes, and BDNF mRNA levels. Overfeeding during the lactation period deregulates cellular metabolism and the monoaminergic systems in the hippocampus. Strikingly, serotonin modulation by fluoxetine treatment protected against some of the effects of early overnutrition. We conclude that overfeeding during brain development induce detrimental effects in mitochondria and in the genes that regulate homeostatic status that can be the molecular mechanisms related to neurological diseases.


Assuntos
Hipocampo , Hipernutrição , Animais , Feminino , Ratos , Fluoxetina/farmacologia , Fluoxetina/uso terapêutico , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Obesidade Pediátrica/metabolismo , Serotonina/metabolismo , Hipernutrição/metabolismo , Hipernutrição/fisiopatologia
2.
Cell Rep ; 37(10): 110075, 2021 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-34879284

RESUMO

The neuroendocrine system coordinates metabolic and behavioral adaptations to fasting, including reducing energy expenditure, promoting counterregulation, and suppressing satiation and anxiety to engage refeeding. Here, we show that steroid receptor coactivator-2 (SRC-2) in pro-opiomelanocortin (POMC) neurons is a key regulator of all these responses to fasting. POMC-specific deletion of SRC-2 enhances the basal excitability of POMC neurons; mutant mice fail to efficiently suppress energy expenditure during food deprivation. SRC-2 deficiency blunts electric responses of POMC neurons to glucose fluctuations, causing impaired counterregulation. When food becomes available, these mutant mice show insufficient refeeding associated with enhanced satiation and discoordination of anxiety and food-seeking behavior. SRC-2 coactivates Forkhead box protein O1 (FoxO1) to suppress POMC gene expression. POMC-specific deletion of SRC-2 protects mice from weight gain induced by an obesogenic diet feeding and/or FoxO1 overexpression. Collectively, we identify SRC-2 as a key molecule that coordinates multifaceted adaptive responses to food shortage.


Assuntos
Metabolismo Energético , Jejum/metabolismo , Comportamento Alimentar , Hipotálamo/metabolismo , Neurônios/metabolismo , Coativador 2 de Receptor Nuclear/metabolismo , Obesidade/metabolismo , Hipernutrição/metabolismo , Pró-Opiomelanocortina/metabolismo , Animais , Ansiedade/metabolismo , Ansiedade/fisiopatologia , Ansiedade/psicologia , Modelos Animais de Doenças , Jejum/psicologia , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Células HEK293 , Humanos , Hipotálamo/fisiopatologia , Masculino , Camundongos Knockout , Coativador 2 de Receptor Nuclear/genética , Obesidade/genética , Obesidade/fisiopatologia , Obesidade/psicologia , Hipernutrição/genética , Hipernutrição/fisiopatologia , Hipernutrição/psicologia , Pró-Opiomelanocortina/genética , Resposta de Saciedade , Transdução de Sinais , Aumento de Peso
3.
Life Sci ; 285: 119951, 2021 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-34516994

RESUMO

AIMS: We sought to evaluate the effects of overfeeding during lactation on the feeding behavior and expression of specific regulatory genes in brain areas associated with food intake in 22- and 60-day old male rats. METHODS: We evaluated body weight, food intake of standard and palatable diet, and mRNA expression of dopamine receptor D1 (DDR1), dopamine receptor (DDR2), melanocortin 4 receptor (MC4R), the µ-opioid receptor (MOR), neuropeptide Y (NPY), agouti-related protein (AGRP), proopiomelanocortin (POMC), cocaine-and amphetamine-regulated transcript (CART), serotonin (5-hydroxytryptamine; 5-HT) transporter (SERT), 5-hydroxytryptamine receptor 1B (5-HT1B), 5-hydroxytryptamine receptor 2C receptor (5-HT2C), Clock (CLOK), cryptochrome protein 1 (Cry1) and period circadian protein homolog 2 (Per2) in the striatum, hypothalamus and brainstem of male rats at post-natal days (PND) 22 and 60. KEY FINDINGS: Overfeeding resulted in significantly increased body weight through PND60, and a 2-fold increase in palatable food intake at PND22, but not at PND60. We observed significant increases in DDR1, DDR2, and MC4R gene expression in the striatum and brainstem and POMC/CART in the hypothalamus of the OF group at PND22 that were reversed by PND60. Hypothalamic levels of 5-HT1B, 5-HT2C and NPY/AGRP on the other hand were decreased at PND22 and increased at PND60 in OF animals. Clock genes were unaffected by OF at PND22, but were significantly elevated at PND60. SIGNIFICANCE: Overfeeding during early development of the rat brain results in obesity and altered feeding behavior in early adulthood. The altered behavior might be the consequence of the changes in food intake and reward gene expression.


Assuntos
Peso Corporal , Encéfalo/crescimento & desenvolvimento , Encéfalo/fisiopatologia , Comportamento Alimentar , Hipernutrição/fisiopatologia , Animais , Proteínas CLOCK/metabolismo , Criptocromos/metabolismo , Ingestão de Alimentos , Feminino , Lactação , Masculino , Proteínas de Ligação a RNA/metabolismo , Ratos , Ratos Wistar , Receptor 5-HT1B de Serotonina/metabolismo , Receptor 5-HT2C de Serotonina/metabolismo
4.
Neurosci Lett ; 765: 136261, 2021 11 20.
Artigo em Inglês | MEDLINE | ID: mdl-34562518

RESUMO

The ability to generate new hippocampal neurons throughout adulthood and successfully integrate them into existing neural networks is critical to cognitive function, while disordered regulation of this process results in neurodegenerative or psychiatric disease. Consequently, identifying the molecular mechanisms promoting homeostatic hippocampal neurogenesis in adults is essential to understanding the etiologies of these disorders and developing therapeutic interventions. For example, recent evidence identifies a strong association between metabolic function and adult hippocampal neurogenesis. Hippocampal neural stem cell (NSC) fate dynamically fluctuates with changes in substrate availability and energy status (AMP/ATP and NAD+/NADH ratios). Furthermore, many metabolic hormones, such as insulin, insulin-like growth factors, and leptin exhibit dual functions also modulating hippocampal neurogenesis and neuron survivability. These diverse metabolic inputs to NSC's from various tissues seemingly suggest the existence of a system in which energy status can finely modulate hippocampal neurogenesis. Supporting this hypothesis, interventions promoting energy balance, such as caloric restriction, intermittent fasting, and exercise, have shown encouraging potential enhancing hippocampal neurogenesis and cognitive function. Overall, there is a clear relationship between whole body energy status, adult hippocampal neurogenesis, and neuron survival; however, the molecular mechanisms underlying this phenomenon are multifaceted. Thus, the aim of this review is to analyze the literature investigating energy status-mediated regulation of adult neurogenesis in the hippocampus, highlight the neurocircuitry and intracellular signaling involved, and propose impactful future directions in the field.


Assuntos
Restrição Calórica , Hipocampo/crescimento & desenvolvimento , Células-Tronco Neurais/fisiologia , Neurogênese/fisiologia , Hipernutrição/fisiopatologia , Adulto , Ingestão de Energia/fisiologia , Exercício Físico/fisiologia , Jejum/metabolismo , Hipocampo/citologia , Humanos , Modelos Animais , Hipernutrição/metabolismo
5.
Diabetologia ; 64(10): 2237-2246, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34272965

RESUMO

AIMS/HYPOTHESIS: Our aim was to explore metabolic pathways linking overnutrition in utero to development of adiposity in normal-weight children. METHODS: We included 312 normal-weight youth exposed or unexposed to overnutrition in utero (maternal BMI ≥25 kg/m2 or gestational diabetes). Fasting insulin, glucose and body composition were measured at age ~10 years (baseline) and ~16 years (follow-up). We examined associations of overnutrition in utero with baseline fasting insulin, followed by associations of baseline fasting insulin with adiposity (BMI z score [BMIZ], subcutaneous adipose tissue [SAT], visceral adipose tissue [VAT]), insulin resistance (HOMA-IR) and fasting glucose during follow-up. RESULTS: >All participants were normal weight at baseline (BMIZ -0.32 ± 0.88), with no difference in BMIZ for exposed vs unexposed youth (p = 0.14). Of the study population, 47.8% were female sex and 47.4% were of white ethnicity. Overnutrition in utero corresponded with 14% higher baseline fasting insulin (geometric mean ratio 1.14 [95% CI 1.01, 1.29]), even after controlling for VAT/SAT ratio. Higher baseline fasting insulin corresponded with higher BMIZ (0.41 [95% CI 0.26, 0.55]), SAT (13.9 [95% CI 2.4, 25.4] mm2), VAT (2.0 [95% CI 0.1, 3.8] mm2), HOMA-IR (0.87 [95% CI 0.68, 1.07]) and fasting glucose (0.23 [95% CI 0.09, 0.38] SD). CONCLUSIONS/INTERPRETATION: Overnutrition in utero may result in hyperinsulinaemia during childhood, preceding development of adiposity. However, our study started at age 10 years, so earlier metabolic changes in response to overnutrition were not taken into account. Longitudinal studies in normal-weight youth starting earlier in life, and with repeated measurements of body weight, fat distribution, insulin sensitivity, beta cell function and blood glucose levels, are needed to clarify the sequence of metabolic changes linking early-life exposures to adiposity and dysglycaemia.


Assuntos
Adiposidade/fisiologia , Diabetes Mellitus Tipo 2/fisiopatologia , Hiperinsulinismo/fisiopatologia , Hipernutrição/fisiopatologia , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia , Adolescente , Glicemia/metabolismo , Composição Corporal , Índice de Massa Corporal , Criança , Feminino , Seguimentos , Humanos , Insulina/sangue , Resistência à Insulina/fisiologia , Gordura Intra-Abdominal/metabolismo , Masculino , Gravidez
6.
Nutrients ; 13(6)2021 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-34063868

RESUMO

The recent identification of brown adipose tissue in adult humans offers a new strategy to increase energy expenditure to treat obesity and associated metabolic disease. While white adipose tissue (WAT) is primarily for energy storage, brown adipose tissue (BAT) is a thermogenic organ that increases energy expenditure to generate heat. BAT is activated upon cold exposure and improves insulin sensitivity and lipid clearance, highlighting its beneficial role in metabolic health in humans. This review provides an overview of BAT physiology in conditions of overnutrition (obesity and associated metabolic disease), undernutrition and in conditions of altered fat distribution such as lipodystrophy. We review the impact of exercise, dietary macronutrients and bioactive compounds on BAT activity. Finally, we discuss the therapeutic potential of dietary manipulations or supplementation to increase energy expenditure and BAT thermogenesis. We conclude that chronic nutritional interventions may represent a useful nonpharmacological means to enhance BAT mass and activity to aid weight loss and/or improve metabolic health.


Assuntos
Tecido Adiposo Marrom/fisiopatologia , Metabolismo Energético/fisiologia , Desnutrição/fisiopatologia , Estado Nutricional/fisiologia , Hipernutrição/fisiopatologia , Exercício Físico/fisiologia , Humanos , Resistência à Insulina/fisiologia , Termogênese/fisiologia , Redução de Peso/fisiologia
7.
Int J Obes (Lond) ; 45(5): 1052-1060, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33594258

RESUMO

OBJECTIVE: Recent evidence indicates that levels of breast milk (BM) hormones such as leptin can fluctuate with maternal adiposity, suggesting that BM hormones may signal maternal metabolic and nutritional environments to offspring during postnatal development. The hormone apelin is highly abundant in BM but its regulation during lactation is completely unknown. Here, we evaluated whether maternal obesity and overnutrition impacted BM apelin and leptin levels in clinical cohorts and lactating rats. METHODS: BM and plasma samples were collected from normal-weight and obese breastfeeding women, and from lactating rats fed a control or a high fat (HF) diet during lactation. Apelin and leptin levels were assayed by ELISA. Mammary gland (MG) apelin expression and its cellular localization in lactating rats was measured by quantitative RT-PCR and immunofluorescence, respectively. RESULTS: BM apelin levels increased with maternal BMI, whereas plasma apelin levels decreased. BM apelin was also positively correlated with maternal insulin and C-peptide levels. In rats, maternal HF feeding exclusively during lactation was sufficient to increase BM apelin levels and decrease its plasma concentration without changing body weight. In contrast, BM leptin levels increased with maternal BMI in humans, but did not change with maternal HF feeding during lactation in rats. Apelin is highly expressed in the rat MG during lactation and was mainly localized to mammary myoepithelial cells. We found that MG apelin gene expression was up-regulated by maternal HF diet and positively correlated with BM apelin content and maternal insulinemia. CONCLUSIONS: Our study indicates that BM apelin levels increase with long- and short-term overnutrition, possibly via maternal hyperinsulinemia and transcriptional upregulation of MG apelin expression in myoepithelial cells. Apelin regulates many physiological processes, including energy metabolism, digestive function, and development. Further studies are needed to unravel the consequences of such changes in offspring development.


Assuntos
Apelina/análise , Leite Humano/química , Obesidade Materna/epidemiologia , Obesidade Materna/fisiopatologia , Hipernutrição/fisiopatologia , Animais , Aleitamento Materno , Dieta Hiperlipídica , Feminino , França , Humanos , Lactação , Leptina , Fenômenos Fisiológicos da Nutrição Materna , Gravidez , Ratos , Ratos Wistar
8.
Curr Opin Clin Nutr Metab Care ; 24(3): 229-235, 2021 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-33587365

RESUMO

PURPOSE OF REVIEW: This manuscript reviews evidence collected during COVID-19 pandemic and provides information on the impact of body composition on severity and outcomes of the disease, analysing methods used for body composition assessment. Malnutrition-screening tools will also be discussed to screen and diagnose the patients at higher risk of COVID-19 severity and related worse outcomes. RECENT FINDINGS: COVID-19 can occur in a wide range of presentation, from asymptomatic to severe forms. Among the major risk factors for worse severity, overnutrition, undernutrition and body composition play a role in the ability to respond to SARS-CoV-2 infection. Excess fat accumulation (i.e. obesity) or lean mass loss and functionality (i.e. sarcopenia) or a combination of both (i.e. sarcopenic obesity) can affect whole-body functioning. These body composition alterations in the short-term can influence susceptibility and immunological responses to the virus, inflammatory reaction, metabolic and respiratory distress, while in the long-term can modulate disease outcomes, namely length of stay, time required for recovery, risk of ICU-acquired weakness and long-term disabilities, and potentially increase the risk of death. SUMMARY: Individuals with malnutrition, sarcopenia, obesity, sarcopenic obesity and older adults with abnormal body composition or malnutrition risk may require tailored medical nutrition therapy to improve short and long-term COVID-19 outcomes.


Assuntos
Composição Corporal , COVID-19/fisiopatologia , Desnutrição/virologia , Estado Nutricional , SARS-CoV-2 , Adulto , Idoso , Idoso de 80 Anos ou mais , COVID-19/complicações , COVID-19/virologia , Feminino , Humanos , Masculino , Desnutrição/fisiopatologia , Pessoa de Meia-Idade , Terapia Nutricional , Obesidade/fisiopatologia , Obesidade/virologia , Hipernutrição/fisiopatologia , Hipernutrição/virologia , Sarcopenia/fisiopatologia , Sarcopenia/virologia , Índice de Gravidade de Doença
9.
Aging (Albany NY) ; 12(20): 19979-20000, 2020 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-33107844

RESUMO

Aging is associated with a decline in sex hormones, variable between sexes, that has an impact on many different body systems and might contribute to age-related disease progression. We aimed to characterize the sex differences in gut microbiota, and to explore the impact of depletion of gonadal hormones, alone or combined with postnatal overfeeding, in rats. Many of the differences in the gut microbiota between sexes persisted after gonadectomy, but removal of gonadal hormones shaped several gut microbiota features towards a more deleterious profile, the effect being greater in females than in males, mainly when animals were concurrently overfed. Moreover, we identified several intestinal miRNAs as potential mediators of the impact of changes in gut microbiota on host organism physiology. Our study points out that gonadal hormones contribute to defining sex-dependent differences of gut microbiota, and discloses a potential role of gonadal hormones in shaping gut microbiota, as consequence of the interaction between sex and nutrition. Our data suggest that the changes in gut microbiota, observed in conditions of sex hormone decline, as those caused by ageing in men and menopause in women, might exert different effects on the host organism, which are putatively mediated by gut microbiota-intestinal miRNA cross-talk.


Assuntos
Microbioma Gastrointestinal , Hormônios Gonadais/metabolismo , Intestinos/microbiologia , Hipernutrição/microbiologia , Fenômenos Fisiológicos da Nutrição Animal , Animais , Disbiose , Feminino , Interações Hospedeiro-Patógeno , Masculino , MicroRNAs/genética , MicroRNAs/metabolismo , Estado Nutricional , Orquiectomia , Ovariectomia , Hipernutrição/metabolismo , Hipernutrição/fisiopatologia , Ratos Wistar , Fatores Sexuais
10.
Int J Obes (Lond) ; 44(12): 2405-2418, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32999409

RESUMO

BACKGROUND: The prevalence of adolescent obesity has increased dramatically, becoming a serious public health concern. While previous evidence suggests that in utero- and early postnatal overnutrition increases adult-onset obesity risk, the neurobiological mechanisms underlying this outcome are not well understood. Non-neuronal cells play an underestimated role in the physiological responses to metabolic/nutrient signals. Hypothalamic glial-mediated inflammation is now considered a contributing factor in the development and perpetuation of obesity; however, attention on the role of gliosis and microglia activation in other nuclei is still needed. METHODS/RESULTS: Here, we demonstrate that early life consumption of high-fat/sucrose diet (HFSD) is sufficient to increase offspring body weight, hyperleptinemia and potentially maladaptive cytoarchitectural changes in the brainstem dorsal-vagal-complex (DVC), an essential energy balance processing hub, across postnatal development. Our data demonstrate that pre- and postnatal consumption of HFSD result in increased body weight, hyperleptinemia and dramatically affects the non-neuronal landscape, and therefore the plasticity of the DVC in the developing offspring. CONCLUSIONS: Current findings are very provocative, considering the importance of the DVC in appetite regulation, suggesting that HFSD-consumption during early life may contribute to subsequent obesity risk via DVC cytoarchitectural changes.


Assuntos
Tronco Encefálico/fisiopatologia , Plasticidade Neuronal , Obesidade/fisiopatologia , Hipernutrição/fisiopatologia , Animais , Peso Corporal , Dieta Hiperlipídica , Sacarose na Dieta/administração & dosagem , Feminino , Peptídeo 1 Semelhante ao Glucagon/sangue , Insulina/sangue , Leptina/sangue , Masculino , Fenômenos Fisiológicos da Nutrição Materna , Ratos , Ratos Sprague-Dawley , Aumento de Peso
11.
Am J Physiol Endocrinol Metab ; 319(6): E1008-E1018, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-32954829

RESUMO

Skeletal muscle is sensitive to environmental cues that are first present in utero. Maternal overnutrition is a model of impaired muscle development leading to structural and metabolic dysfunction in adult life. In this study, we investigated the effect of an obesogenic maternal environment on growth and postnatal myogenesis in the offspring. Male C57BL/6J mice born to chow- or high-fat-diet-fed mothers were allocated to four different groups at the end of weaning. For the following 10 wk, half of the pups were maintained on the same diet as their mother and half of the pups were switched to the other diet (chow or high-fat). At 12 wk of age, muscle injury was induced using an intramuscular injection of barium chloride. Seven days later, mice were humanely killed and muscle tissue was harvested. A high-fat maternal diet impaired offspring growth patterns and downregulated satellite cell activation and markers of postnatal myogenesis 7 days after injury without altering the number of newly synthetized fibers over the whole 7-day period. Importantly, a healthy postnatal diet could not reverse any of these effects. In addition, we demonstrated that postnatal myogenesis was associated with a diet-independent upregulation of three miRNAs, mmu-miR-31-5p, mmu-miR-136-5p, and mmu-miR-296-5p. Furthermore, in vitro analysis confirmed the role of these miRNAs in myocyte proliferation. Our findings are the first to demonstrate that maternal overnutrition impairs markers of postnatal myogenesis in the offspring and are particularly relevant to today's society where the incidence of overweight/obesity in women of childbearing age is increasing.


Assuntos
Dieta Hiperlipídica , Crescimento e Desenvolvimento/fisiologia , Desenvolvimento Muscular/fisiologia , Efeitos Tardios da Exposição Pré-Natal , Células Satélites de Músculo Esquelético/fisiologia , Animais , Biomarcadores/análise , Biomarcadores/metabolismo , Dieta Hiperlipídica/efeitos adversos , Feminino , Masculino , Fenômenos Fisiológicos da Nutrição Materna , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/genética , MicroRNAs/metabolismo , Obesidade/etiologia , Obesidade/fisiopatologia , Hipernutrição/complicações , Hipernutrição/fisiopatologia , Gravidez , Complicações na Gravidez/etiologia , Complicações na Gravidez/fisiopatologia , Efeitos Tardios da Exposição Pré-Natal/etiologia , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia
12.
J Nutr Biochem ; 86: 108494, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32920089

RESUMO

Overfeeding and rapid weight gain during early life are risk factors for the development of obesity in adulthood. This metabolic malprogramming may be mediated by endocrine disturbances during critical periods of development. Cholecystokinin (CCK) acts on the central nervous system by elevating thermogenesis and the activity of anorectic neurons, modulating overall energy balance. Therefore, we tested the hypothesis that postnatal overfeeding impaired CCK effects. Pups were raised in either a litter of three (neonatal overnutrition/small litter group) or 12 (controls/normal litter group) pups per dam to study the effects of postnatal overfeeding on the central and peripheral CCK systems in adulthood. Rats raised in small litters became overweight during lactation and remained overweight as adults, with increased adiposity and plasma levels of lipids, glucose, insulin, and leptin. Neonatally over-nourished rats showed attenuation of gastric emptying and anorexigenic response to CCK, suggesting that offspring from the SL group may present CCK resistance as adult male rats. Consistent with this idea, overweight rats displayed impaired central response in c-Fos immunoreactivity on the nucleus tractus solitarius, area postrema, paraventricular nucleus, central amygdala, arcuate nucleus, and dorsomedial hypothalamus in response to peripheral CCK at adulthood. The small litter group of adult male rats also exhibited reduced norepinephrine- and CCK-stimulated thermogenesis. Unresponsiveness to the effects of CCK may contribute to overweight and metabolic dysfunctions observed in postnatally over-nourished adult rats. Thus, the involvement of an impaired CCK system, among other neurohormonal failures, may contribute to the development of obesity.


Assuntos
Adiposidade , Sistema Nervoso Central/fisiopatologia , Colecistocinina/metabolismo , Sistema Endócrino/fisiopatologia , Hipernutrição/fisiopatologia , Tecido Adiposo/metabolismo , Animais , Animais Recém-Nascidos , Mapeamento Encefálico , Metabolismo Energético , Feminino , Esvaziamento Gástrico , Glucose/metabolismo , Homeostase , Hipotálamo , Leptina/sangue , Lipídeos/química , Masculino , Obesidade/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Ratos Wistar , Termogênese , Aumento de Peso
13.
J Dev Orig Health Dis ; 11(5): 492-498, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32524941

RESUMO

Nutritional disorders during the perinatal period cause cardiometabolic dysfunction, which is observable in the early overfeeding (EO) experimental model. Therefore, severe caloric restriction has the potential of affecting homeostasis through the same epigenetic mechanisms, and its effects need elucidation. This work aims to determine the impact of food restriction (FR) during puberty in early overfed obese and non-obese animals in adult life. Three days after delivery (PN3), Wistar rats were separated into two groups: normal litter (NL; 9 pups) and small litter (SL; 3 pups). At PN30, some offspring were subjected to FR (50%) until PN60, or maintained with free access to standard chow. NL and SL animals submitted to food restriction (NLFR and SLFR groups) were kept in recovery with free access to standard chow from PN60 until PN120. Body weight and food intake were monitored throughout the experimental period. At PN120 cardiovascular parameters were analyzed and the animals were euthanized for sample collection. SLNF and SLFR offspring were overweight and had increased adiposity. Differences in blood pressure were observed only between obese and non-obese animals. Obese and FR animals have cardiac remodeling showing cardiomyocyte hypertrophy and the presence of interstitial and perivascular fibrosis. FR animals also show increased expression of AT1 and AT2 receptors and of total ERK and p-ERK. The present study showed that EO leads to the obese phenotype and cardiovascular disruptions. Interestingly, we demonstrated that severe FR during puberty leads to cardiac remodeling.


Assuntos
Fenômenos Fisiológicos da Nutrição do Lactente/fisiologia , Desnutrição/complicações , Obesidade/fisiopatologia , Hipernutrição/complicações , Remodelação Ventricular/fisiologia , Animais , Animais Recém-Nascidos , Peso Corporal , Restrição Calórica/efeitos adversos , Modelos Animais de Doenças , Feminino , Ventrículos do Coração/crescimento & desenvolvimento , Humanos , Lactente , Recém-Nascido , Masculino , Desnutrição/diagnóstico , Desnutrição/fisiopatologia , Obesidade/etiologia , Hipernutrição/fisiopatologia , Ratos , Ratos Wistar , Índice de Gravidade de Doença
14.
Physiol Rep ; 8(12): e14498, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32597039

RESUMO

The aim was to test the hypothesis that prenatal under- and overnutrition in late gestation can program small intestinal (SI) growth, angiogenesis, and endocrine function to predispose for a hyperabsorptive state, thereby increasing the susceptibility to the adverse effects of an early postnatal obesogenic diet. Twin-pregnant ewes were exposed to adequate (NORM), LOW (50% of NORM), or HIGH (150% energy and 110% protein of NORM) diets through the last trimester (term ~147 days). From 3 days to 6 months of age, their lambs were fed either a moderate (CONV) or a high-carbohydrate high-fat (HCHF) diet. At 6 months of age, responses in plasma metabolites and insulin to refeeding after fasting were determined and then different segments of the SI were sampled at autopsy. Prenatal overnutrition impacts were most abundant in the duodenum where HIGH had increased villus amplification factor and lowered villi thickness with increased IRS-1 and reduced GH-R expressions. In jejunum, HIGH lambs had an increased expression of Lactate gene and amplified when exposed to HCHF postnatally. Specifically, in LOW, sensitivity to HCHF was affected in ileum. Thus, the mismatching LOW-HCHF nutrition increased expressions of angiogenic genes (VEGF, VEGF-R1, ANGPT1, RTK) and increased mucosa layer (tunica mucosa) thickness but reduced muscle layer (Tunica muscularis) thickness. The SI is a target of prenatal nutritional programming, where late gestation overnutrition increased and shifted digestive capacity for carbohydrates toward the jejunum, whereas late gestation undernutrition predisposed for ileal angiogenesis and carbohydrate and fat hyperabsorptive capacity upon subsequent exposure to postnatal obesogenic diet.


Assuntos
Sistema Endócrino/fisiopatologia , Intestino Delgado/crescimento & desenvolvimento , Desnutrição/fisiopatologia , Hipernutrição/fisiopatologia , Animais , Modelos Animais de Doenças , Sistema Endócrino/enzimologia , Sistema Endócrino/crescimento & desenvolvimento , Jejum/metabolismo , Feminino , Insulina/sangue , Absorção Intestinal , Intestino Delgado/irrigação sanguínea , Intestino Delgado/embriologia , Intestino Delgado/patologia , Neovascularização Fisiológica , Gravidez , Efeitos Tardios da Exposição Pré-Natal , Ovinos
15.
PLoS One ; 15(5): e0232400, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32384084

RESUMO

Metabolic parameters ranging from circulating nutrient levels and substrate utilization to energy expenditure and thermogenesis are temporally modulated by the circadian timing system. During critical embryonic developmental periods, maternal over-nutrition could alter key elements in different tissues associated with the generation of circadian rhythmicity, compromising normal rhythmicity development. To address this issue, we determine whether maternal over-nutrition leads to alterations in the development of circadian rhythmicity at physiological and behavioral levels in the offspring. For this, female rabbits were fed a standard diet (SD) or high-fat and carbohydrate diet (HFCD) before mating and during gestation. Core body temperature and gross locomotor activity were continuously recorded in newborn rabbits, daily measurements of body weight and the amount of milk ingested was carried out. At the end of lactation, tissue samples, including brown adipose tissue (BAT) and white adipose tissue (WAT), were obtained for determining the expression of uncoupling protein-1 (UCP1) and cell death-inducing DNA fragmentation factor-like effector A (CIDEA) genes. HFCD pups exhibited conspicuous differences in the development of the daily rhythm of temperature and locomotor activity compared to the SD pups, including a significant increase in the daily mean core temperature, changes in the time when temperature or activity remains above the average, shifts in the acrophase, decrease in the duration and intensity of the anticipatory rise previous to nursing, and changes in frequency of the rhythms. HFCD pups exhibited a significant increase in BAT thermogenesis markers, and a decrease of these markers in WAT, indicating more heat generation by brown adipocytes and alterations in the browning process. These results indicate that maternal over-nutrition alters offspring homeostatic and chronostatic regulation at the physiological and behavioral levels. Further studies are needed to determine whether these alterations are associated with the changes in the organization of the circadian system of the progeny.


Assuntos
Regulação da Temperatura Corporal/fisiologia , Ritmo Circadiano/fisiologia , Lactação/fisiologia , Locomoção/fisiologia , Tecido Adiposo Marrom/fisiopatologia , Tecido Adiposo Branco/fisiopatologia , Animais , Proteínas Reguladoras de Apoptose/genética , Regulação da Temperatura Corporal/genética , Ritmo Circadiano/genética , Modelos Animais de Doenças , Feminino , Expressão Gênica , Lactação/genética , Locomoção/genética , Fenômenos Fisiológicos da Nutrição Materna , Hipernutrição/complicações , Hipernutrição/genética , Hipernutrição/fisiopatologia , Gravidez , Complicações na Gravidez/genética , Complicações na Gravidez/fisiopatologia , Coelhos , Proteína Desacopladora 1/genética
16.
Eur J Pharmacol ; 881: 173200, 2020 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-32445706

RESUMO

Nutritional imbalance in early life may disrupt the hypothalamic control of energy homeostasis and increase the risk of metabolic disease. The hypothalamic serotonin (5-hydroxytryptamine; 5-HT) system based in the hypothalamus plays an important role in the homeostatic control of energy balance, however the mechanisms underlying the regulation of energy metabolism by 5-HT remain poorly described. Several crucial mitochondrial functions are altered by mitochondrial stress. Adaptations to this stress include changes in mitochondrial multiplication (i.e, mitochondrial biogenesis). Due to the scarcity of evidence regarding the effects of serotonin reuptake inhibitors (SSRI) such as fluoxetine (FLX) on mitochondrial function, we sought to investigate the potential contribution of FLX on changes in mitochondrial function and biogenesis occurring in overfed rats. Using a neonatal overfeeding model, male Wistar rats were divided into 4 groups between 39 and 59 days of age based on nutrition and FLX administration: normofed + vehicle (NV), normofed + FLX (NF), overfed + vehicle (OV) and overfed + FLX (OF). We found that neonatal overfeeding impaired mitochondrial respiration and increased oxidative stress biomarkers in the hypothalamus. FLX administration in overfed rats reestablished mitochondrial oxygen consumption, increased mitochondrial uncoupling protein 2 (Ucp2) expression, reduced total reactive species (RS) production and oxidative stress biomarkers, and up-regulated mitochondrial biogenesis-related genes. Taken together our results suggest that FLX administration in overfed rats improves mitochondrial respiratory chain activity and oxidative balance and increases the transcription of genes employed in mitochondrial biogenesis favoring mitochondrial energy efficiency in response to early nutritional imbalance.


Assuntos
Fármacos Antiobesidade/farmacologia , Metabolismo Energético/efeitos dos fármacos , Fluoxetina/farmacologia , Hipotálamo/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Biogênese de Organelas , Hipernutrição/tratamento farmacológico , Estresse Oxidativo/efeitos dos fármacos , Fatores Etários , Fenômenos Fisiológicos da Nutrição Animal , Animais , Animais Recém-Nascidos , Animais Lactentes , Hipotálamo/metabolismo , Hipotálamo/patologia , Hipotálamo/fisiopatologia , Masculino , Mitocôndrias/genética , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Estado Nutricional , Hipernutrição/metabolismo , Hipernutrição/patologia , Hipernutrição/fisiopatologia , Oxirredução , Consumo de Oxigênio , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo , Transcrição Gênica , Proteína Desacopladora 2/genética , Proteína Desacopladora 2/metabolismo
17.
Nutrients ; 12(2)2020 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-32093229

RESUMO

Childhood obesity is associated with metabolic and cardiovascular comorbidities. The development of these alterations may have its origin in early life stages such as the lactation period through metabolic programming. Insulin resistance is a common complication in obese patients and may be responsible for the cardiovascular alterations associated with this condition. This study analyzed the development of cardiovascular insulin resistance in a rat model of childhood overweight induced by overfeeding during the lactation period. On birth day, litters were divided into twelve (L12) or three pups per mother (L3). Overfed rats showed a lower increase in myocardial contractility in response to insulin perfusion and a reduced insulin-induced vasodilation, suggesting a state of cardiovascular insulin resistance. Vascular insulin resistance was due to decreased activation of phosphoinositide 3-kinase (PI3K)/Akt pathway, whereas cardiac insulin resistance was associated with mitogen-activated protein kinase (MAPK) hyperactivity. Early overfeeding was also associated with a proinflammatory and pro-oxidant state; endothelial dysfunction; decreased release of nitrites and nitrates; and decreased gene expression of insulin receptor (IR), glucose transporter-4 (GLUT-4), and endothelial nitric oxide synthase (eNOS) in response to insulin. In conclusion, overweight induced by lactational overnutrition in rat pups is associated with cardiovascular insulin resistance that could be related to the cardiovascular alterations associated with this condition.


Assuntos
Doenças Cardiovasculares/etiologia , Resistência à Insulina/fisiologia , Insulina/metabolismo , Hipernutrição/fisiopatologia , Obesidade Pediátrica/fisiopatologia , Animais , Modelos Animais de Doenças , Feminino , Coração/fisiopatologia , Lactação/fisiologia , Masculino , Contração Miocárdica , Miocárdio/metabolismo , Hipernutrição/complicações , Obesidade Pediátrica/complicações , Ratos , Transdução de Sinais , Vasodilatação
18.
Front Neuroendocrinol ; 57: 100834, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32084515

RESUMO

The maternal environment during pregnancy is critical for fetal development and perinatal perturbations can prime offspring disease risk. Here, we briefly review evidence linking two well-characterized maternal stressors - psychosocial stress and infection - to increased neuropsychiatric risk in offspring. In the current climate of increasing obesity and globalization of the Western-style diet, maternal overnutrition emerges as a pressing public health concern. We focus our attention on recent epidemiological and animal model evidence showing that, like psychosocial stress and infection, maternal overnutrition can also increase offspring neuropsychiatric risk. Using lessons learned from the psychosocial stress and infection literature, we discuss how altered maternal and placental physiology in the setting of overnutrition may contribute to abnormal fetal development and resulting neuropsychiatric outcomes. A better understanding of converging pathophysiological pathways shared between stressors may enable development of interventions against neuropsychiatric illnesses that may be beneficial across stressors.


Assuntos
Sintomas Afetivos/etiologia , Transtornos Mentais/etiologia , Complicações na Gravidez/fisiopatologia , Efeitos Tardios da Exposição Pré-Natal/psicologia , Estresse Fisiológico/fisiologia , Estresse Psicológico/fisiopatologia , Animais , Disfunção Cognitiva/etiologia , Meio Ambiente , Feminino , Desenvolvimento Fetal , Humanos , Hipernutrição/complicações , Hipernutrição/fisiopatologia , Placenta/fisiopatologia , Gravidez , Complicações na Gravidez/imunologia , Complicações na Gravidez/psicologia , Efeitos Tardios da Exposição Pré-Natal/imunologia , Fatores de Risco , Estresse Psicológico/imunologia , Estresse Psicológico/psicologia
19.
Nutr Neurosci ; 23(2): 161-169, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29855223

RESUMO

Objectives: This investigation studied whether physical exercise could modulate cortical spreading depression (CSD) propagation velocity in adult rat offspring from dams that had received a high-fat (HF) diet during lactation. Methods: Wistar male rats suckled by dams fed either control (C) or HF diet ad libitum. After weaning, pups received standard laboratory chow. From 40 to 60 days of life, half of the animals exercised on a treadmill (group E); the other half remained sedentary (group S). Two additional HF groups (E and S) received fluoxetine (F; 10 mg/kg/day, orogastrically) from 40 to 60 days of life (groups HF/EF and HF/F). Results: At 40 days of life, rats from the maternal HF diet presented higher weight, thoracic circumference, and Lee Index than C animals and remained heavier at 60 days of life. Physical exercise decreased abdominal circumference. HF diet increased CSD propagation velocity (mean ± SD; mm/min) in sedentaries (HF/S 3.47 ± 0.31 versus C/S 3.24 ± 0.26). Treadmill exercise decelerated CSD propagation in both groups C/E (2.94 ± 0.28) and HF/E (2.97 ± 0.40). Fluoxetine alone decreased CSD propagation (HF/F 2.88 ± 0.45) compared with HF/S group. The combination of fluoxetine + exercise under HF condition (2.98 ± 0.27) was similar to HF/E group. Discussion: Physical exercise is able to reduce CSD propagation velocity in rat adult brains even when they have suffered over-nourishing during lactation. The effects of exercise alone or fluoxetine alone on CSD were similar to the effects of fluoxetine + exercise, under the HF condition. Data reinforce malnutrition during lactation modifies cortical electrophysiology even when the HF condition no longer exists.


Assuntos
Depressão Alastrante da Atividade Elétrica Cortical/fisiologia , Lactação/fisiologia , Hipernutrição/fisiopatologia , Condicionamento Físico Animal/fisiologia , Animais , Animais Recém-Nascidos/fisiologia , Peso Corporal , Encéfalo/fisiopatologia , Depressão Alastrante da Atividade Elétrica Cortical/efeitos dos fármacos , Dieta Hiperlipídica/efeitos adversos , Feminino , Fluoxetina/administração & dosagem , Masculino , Ratos , Inibidores Seletivos de Recaptação de Serotonina/administração & dosagem
20.
Sci Rep ; 9(1): 17322, 2019 11 21.
Artigo em Inglês | MEDLINE | ID: mdl-31754139

RESUMO

Maternal overnutrition during sensitive periods of early development increases the risk for obesity and neuropsychiatric disorders later in life. However, it still remains unclear during which phases of early development the offspring is more vulnerable. Here, we investigate the effects of maternal high-fat diet (MHFD) at different stages of pre- or postnatal development and characterize the behavioral, neurochemical and metabolic phenotypes. We observe that MHFD exposure at pre-conception has no deleterious effects on the behavioral and metabolic state of the offspring. Late gestational HFD exposure leads to more prominent addictive-like behaviors with reduced striatal dopamine levels compared to early gestational HFD. Conversely, offspring exposed to MHFD during lactation display the metabolic syndrome and schizophrenia-like phenotype. The latter, is manifested by impaired sensory motor gating, and latent inhibition as well as enhanced sensitivity to amphetamine. These effects are accompanied by higher striatal dopamine levels. Together, our data suggest that MHFD exposure during specific stages of development leads to distinct neuropathological alterations that determine the severity and nature of poor health outcome in adulthood, which may provide insight in identifying effective strategies for early intervention.


Assuntos
Desenvolvimento Fetal/fisiologia , Fenômenos Fisiológicos da Nutrição Materna/fisiologia , Hipernutrição/complicações , Efeitos Tardios da Exposição Pré-Natal/epidemiologia , Animais , Comportamento Aditivo/epidemiologia , Comportamento Aditivo/etiologia , Comportamento Aditivo/fisiopatologia , Dieta Hiperlipídica/efeitos adversos , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Camundongos , Obesidade/epidemiologia , Obesidade/etiologia , Obesidade/fisiopatologia , Hipernutrição/fisiopatologia , Gravidez , Efeitos Tardios da Exposição Pré-Natal/etiologia , Esquizofrenia/epidemiologia , Esquizofrenia/etiologia , Esquizofrenia/fisiopatologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...