Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Nutr Neurosci ; 26(2): 173-186, 2023 Feb.
Article in English | MEDLINE | ID: mdl-35125071

ABSTRACT

BACKGROUND: Development of obesity and its comorbidities is not only the result of excess energy intake, but also of dietary composition. Understanding how hypothalamic metabolic circuits interpret nutritional signals is fundamental to advance towards effective dietary interventions. OBJECTIVE: We aimed to determine the metabolic response to diets enriched in specific fatty acids. METHODS: Male mice received a diet enriched in unsaturated fatty acids (UOLF) or saturated fatty acids (SOLF) for 8 weeks. RESULTS: UOLF and SOLF mice gained more weight and adiposity, but with no difference between these two groups. Circulating leptin levels increased on both fatty acid-enriched diet, but were higher in UOLF mice, as were leptin mRNA levels in visceral adipose tissue. In contrast, serum non-esterified fatty acid levels only rose in SOLF mice. Hypothalamic mRNA levels of NPY decreased and of POMC increased in both UOLF and SOLF mice, but only SOLF mice showed signs of hypothalamic astrogliosis and affectation of central fatty acid metabolism. Exogenous leptin activated STAT3 in the hypothalamus of all groups, but the activation of AKT and mTOR and the decrease in AMPK activation in observed in controls and UOLF mice was not found in SOLF mice. CONCLUSIONS: Diets rich in fatty acids increase body weight and adiposity even if energy intake is not increased, while increased intake of saturated and unsaturated fatty acids differentially modify metabolic parameters that could underlie more long-term comorbidities. Thus, more understanding of how specific nutrients affect metabolism, weight gain, and obesity associated complications is necessary.


Subject(s)
Gliosis , Leptin , Mice , Male , Animals , Gliosis/metabolism , Dietary Fats , Fatty Acids, Unsaturated/pharmacology , Obesity/metabolism , Hypothalamus/metabolism , Fatty Acids/metabolism , RNA, Messenger/metabolism
2.
Neurosci Lett ; 793: 136996, 2023 01 10.
Article in English | MEDLINE | ID: mdl-36481371

ABSTRACT

Leptin receptors (LepR) are expressed in brain areas controlling food intake homeostasis, such as the hypothalamus, the hippocampus and the prefrontal cortex. In a previous study we reported that long-term intake of saturated and monounsaturated fat alters hypothalamic LepR signalling. The current study aims at investigating the effect of foods high in either saturated (SOLF) or monounsaturated fat (UOLF) on LepR functionality in the hippocampus and the prefrontal cortex. Male mice were placed on SOLF/UOLF (eight weeks), then treated with recombinant murine leptin (1 mg/kg). After 60 min, brain regions were dissected and processed for western blot of phosphorylated STAT3 (pSTAT3), Akt (pAkt) and AMPK (pAMPK). Levels of SOCS3 were also quantified. SOLF itself increased basal levels of pSTAT3, while UOLF impaired leptin-induced phosphorylation of both Akt and AMPK. SOCS3 levels were specifically increased by UOLF within the prefrontal cortex. Our results show that SOLF and UOLF differently affect LepR signalling within the hippocampus and the prefrontal cortex, which points to the complex effect of saturated and unsaturated fat on brain function, particularly in areas regulating food intake.


Subject(s)
Brain , Receptors, Leptin , Animals , Male , Mice , AMP-Activated Protein Kinases , Brain/metabolism , Fats, Unsaturated/administration & dosage , Hypothalamus/metabolism , Leptin/metabolism , Proto-Oncogene Proteins c-akt , Receptors, Leptin/metabolism , Suppressor of Cytokine Signaling Proteins/metabolism
3.
Nutr Neurosci ; 25(5): 931-944, 2022 May.
Article in English | MEDLINE | ID: mdl-32954972

ABSTRACT

Aim: We aimed to investigate whether maternal malnutrition during gestation/lactation induces long-lasting changes on inflammation, lipid metabolism and endocannabinoid signaling in the adult offspring hypothalamus and the role of hypothalamic astrocytes in these changes.Methods: We analyzed the effects of a free-choice hypercaloric palatable diet (P) during (pre)gestation, lactation and/or post-weaning on inflammation, lipid metabolism and endogenous cannabinoid signaling in the adult offspring hypothalamus. We also evaluated the response of primary hypothalamic astrocytes to palmitic acid and anandamide.Results: Postnatal exposure to a P diet induced factors involved in hypothalamic inflammation (Tnfa and Il6) and gliosis (Gfap, vimentin and Iba1) in adult offspring, being more significant in females. In contrast, maternal P diet reduced factors involved in astrogliosis (vimentin), fatty acid oxidation (Cpt1a) and monounsaturated fatty acid synthesis (Scd1). These changes were accompanied by an increase in the expression of the genes for the cannabinoid receptor (Cnr1) and Nape-pld, an enzyme involved in endocannabinoid synthesis, in females and a decrease in the endocannabinoid degradation enzyme Faah in males. These changes suggest that the maternal P diet results in sex-specific alterations in hypothalamic endocannabinoid signaling and lipid metabolism. This hypothesis was tested in hypothalamic astrocyte cultures, where palmitic acid (PA) and the polyunsaturated fatty acid N-arachidonoylethanolamine (anandamide or AEA) were found to induce similar changes in the endocannabinoid system (ECS) and lipid metabolism.Conclusion: These results stress the importance of both maternal diet and sex in long term metabolic programming and suggest a possible role of hypothalamic astrocytes in this process.


Subject(s)
Cannabinoids , Endocannabinoids , Adult Children , Arachidonic Acids , Astrocytes/metabolism , Cannabinoids/metabolism , Diet , Female , Gliosis/metabolism , Humans , Hypothalamus/metabolism , Inflammation/metabolism , Lipid Metabolism , Male , Palmitic Acid/metabolism , Polyunsaturated Alkamides , Vimentin/metabolism
4.
Int J Mol Sci ; 22(12)2021 Jun 11.
Article in English | MEDLINE | ID: mdl-34208173

ABSTRACT

Maternal malnutrition in critical periods of development increases the risk of developing short- and long-term diseases in the offspring. The alterations induced by this nutritional programming in the hypothalamus of the offspring are of special relevance due to its role in energy homeostasis, especially in the endocannabinoid system (ECS), which is involved in metabolic functions. Since astrocytes are essential for neuronal energy efficiency and are implicated in brain endocannabinoid signaling, here we have used a rat model to investigate whether a moderate caloric restriction (R) spanning from two weeks prior to the start of gestation to its end induced changes in offspring hypothalamic (a) ECS, (b) lipid metabolism (LM) and/or (c) hypothalamic astrocytes. Monitorization was performed by analyzing both the gene and protein expression of proteins involved in LM and ECS signaling. Offspring born from caloric-restricted mothers presented hypothalamic alterations in both the main enzymes involved in LM and endocannabinoids synthesis/degradation. Furthermore, most of these changes were similar to those observed in hypothalamic offspring astrocytes in culture. In conclusion, a maternal low caloric intake altered LM and ECS in both the hypothalamus and its astrocytes, pointing to these glial cells as responsible for a large part of the alterations seen in the total hypothalamus and suggesting a high degree of involvement of astrocytes in nutritional programming.


Subject(s)
Astrocytes/metabolism , Caloric Restriction , Endocannabinoids/metabolism , Hypothalamus/metabolism , Lipid Metabolism , Signal Transduction , Animals , Animals, Newborn , Body Weight , Brain/pathology , Female , Gene Expression Regulation , Gliosis/genetics , Gliosis/pathology , Inflammation/genetics , Inflammation/pathology , Lipid Metabolism/genetics , Pregnancy , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Signal Transduction/genetics
5.
Nutrients ; 13(3)2021 Mar 19.
Article in English | MEDLINE | ID: mdl-33808927

ABSTRACT

Vegetable oils such as palm oil (enriched in saturated fatty acids, SFA) and high-oleic-acid sunflower oil (HOSO, containing mainly monounsaturated fatty acids, MUFA) have emerged as the most common replacements for trans-fats in the food industry. The aim of this study is to analyze the impact of SFA and MUFA-enriched high-fat (HF) diets on endothelial function, vascular remodeling, and arterial stiffness compared to commercial HF diets. Five-week-old male C57BL6J mice were fed a standard (SD), a HF diet enriched with SFA (saturated oil-enriched Food, SOLF), a HF diet enriched with MUFA (unsaturated oil-enriched Food, UOLF), or a commercial HF diet for 8 weeks. Vascular function was analyzed in the thoracic aorta. Structural and mechanical parameters were assessed in mesenteric arteries by pressure myography. SOLF, UOLF, and HF diet reduced contractile responses to phenylephrine and induced endothelial dysfunction in the thoracic aorta. A significant increase in the ß-index, and thus in arterial stiffness, was also detected in mesenteric arteries from the three HF groups, due to enhanced deposition of collagen in the vascular wall. SOLF also induced hypotrophic inward remodeling. In conclusion, these data demonstrate a deleterious effect of HF feeding on obesity-related vascular alterations that is exacerbated by SFA.


Subject(s)
Arteries/drug effects , Dietary Fats/pharmacology , Fatty Acids, Monounsaturated/pharmacology , Vascular Stiffness/drug effects , Animals , Aorta, Thoracic/drug effects , Arteries/physiology , Body Weight , Collagen/metabolism , Diet, High-Fat , Dietary Fats, Unsaturated/pharmacology , Elastin , Fatty Acids/pharmacology , Fuchs' Endothelial Dystrophy , Glucose/metabolism , Male , Mice , Mice, Inbred C57BL , Nitric Oxide/metabolism , Oleic Acid , Plant Oils , Sunflower Oil , Vascular Remodeling/drug effects
6.
Cells ; 10(3)2021 03 06.
Article in English | MEDLINE | ID: mdl-33800837

ABSTRACT

Central actions of leptin and insulin on hepatic lipid metabolism can be opposing and the mechanism underlying this phenomenon remains unclear. Both hormones can modulate the central somatostatinergic system that has an inhibitory effect on growth hormone (GH) expression, which plays an important role in hepatic metabolism. Using a model of chronic central leptin infusion, we evaluated whether an increase in central leptin bioavailability modifies the serum lipid pattern through changes in hepatic lipid metabolism in male rats in response to an increase in central insulin and the possible involvement of the GH axis in these effects. We found a rise in serum GH in leptin plus insulin-treated rats, due to an increase in pituitary GH mRNA levels associated with lower hypothalamic somatostatin and pituitary somatostatin receptor-2 mRNA levels. An augment in hepatic lipolysis and a reduction in serum levels of non-esterified fatty acids (NEFA) and triglycerides were found in leptin-treated rats. These rats experienced a rise in lipogenic-related factors and normalization of serum levels of NEFA and triglycerides after insulin treatment. These results suggest that an increase in insulin in leptin-treated rats can act on the hepatic lipid metabolism through activation of the GH axis.


Subject(s)
Hypothalamus/drug effects , Insulin/pharmacology , Leptin/pharmacology , Lipid Metabolism/drug effects , Liver/drug effects , Pituitary Gland/drug effects , Animals , Fatty Acids, Nonesterified/blood , Gene Expression Regulation , Growth Hormone/genetics , Growth Hormone/metabolism , Hypothalamus/metabolism , Injections, Intravenous , Injections, Intraventricular , Insulin/metabolism , Leptin/metabolism , Lipid Metabolism/genetics , Liver/metabolism , Male , Pituitary Gland/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptors, Somatostatin/genetics , Receptors, Somatostatin/metabolism , Signal Transduction , Triglycerides/blood
7.
Int J Mol Sci ; 22(6)2021 Mar 11.
Article in English | MEDLINE | ID: mdl-33799501

ABSTRACT

The growth hormone (GH)/insulin-like growth factor I (IGF-I) axis is involved in metabolic control. Malnutrition reduces IGF-I and modifies the thermogenic capacity of brown adipose tissue (BAT). Leptin has effects on the GH/IGF-I axis and the function of BAT, but its interaction with IGF-I and the mechanisms involved in the regulation of thermogenesis remains unknown. We studied the GH/IGF-I axis and activation of IGF-I-related signaling and metabolism related to BAT thermogenesis in chronic central leptin infused (L), pair-fed (PF), and control rats. Hypothalamic somatostatin mRNA levels were increased in PF and decreased in L, while pituitary GH mRNA was reduced in PF. Serum GH and IGF-I concentrations were decreased only in PF. In BAT, the association between suppressor of cytokine signaling 3 and the IGF-I receptor was reduced, and phosphorylation of the IGF-I receptor increased in the L group. Phosphorylation of Akt and cyclic AMP response element binding protein and glucose transporter 4 mRNA levels were increased in L and mRNA levels of uncoupling protein-1 (UCP-1) and enzymes involved in lipid anabolism reduced in PF. These results suggest that modifications in UCP-1 in BAT and changes in the GH/IGF-I axis induced by negative energy balance are dependent upon leptin levels.


Subject(s)
Adipose Tissue, Brown/drug effects , Energy Metabolism/drug effects , Growth Hormone/genetics , Insulin-Like Growth Factor I/genetics , Leptin/pharmacology , Thermogenesis/drug effects , Adipose Tissue, Brown/metabolism , Animals , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Energy Metabolism/genetics , Glucose Transporter Type 4/genetics , Glucose Transporter Type 4/metabolism , Growth Hormone/metabolism , Hypothalamus/drug effects , Hypothalamus/metabolism , Injections, Intraventricular , Insulin-Like Growth Factor I/metabolism , Male , Phosphorylation/drug effects , Pituitary Gland/drug effects , Pituitary Gland/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism , Somatostatin/genetics , Somatostatin/metabolism , Suppressor of Cytokine Signaling 3 Protein/genetics , Suppressor of Cytokine Signaling 3 Protein/metabolism , Thermogenesis/genetics , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism
8.
Front Endocrinol (Lausanne) ; 12: 796661, 2021.
Article in English | MEDLINE | ID: mdl-34975768

ABSTRACT

Dietary intervention is a common tactic employed to curtail the current obesity epidemic. Changes in nutritional status alter metabolic hormones such as insulin or leptin, as well as the insulin-like growth factor (IGF) system, but little is known about restoration of these parameters after weight loss in obese subjects and if this differs between the sexes, especially regarding the IGF system. Here male and female mice received a high fat diet (HFD) or chow for 8 weeks, then half of the HFD mice were changed to chow (HFDCH) for 4 weeks. Both sexes gained weight (p < 0.001) and increased their energy intake (p < 0.001) and basal glycemia (p < 0.5) on the HFD, with these parameters normalizing after switching to chow but at different rates in males and females. In both sexes HFD decreased hypothalamic NPY and AgRP (p < 0.001) and increased POMC (p < 0.001) mRNA levels, with all normalizing in HFDCH mice, whereas the HFD-induced decrease in ObR did not normalize (p < 0.05). All HFD mice had abnormal glucose tolerance tests (p < 0.001), with males clearly more affected, that normalized when returned to chow. HFD increased insulin levels and HOMA index (p < 0.01) in both sexes, but only HFDCH males normalized this parameter. Returning to chow normalized the HFD-induced increase in circulating leptin (p < 0.001), total IGF1 (p < 0.001), IGF2 (p < 0.001, only in females) and IGFBP3 (p < 0.001), whereas free IGF1 levels remained elevated (p < 0.01). In males IGFBP2 decreased with HFD and normalized with chow (p < 0.001), with no changes in females. Although returning to a healthy diet improved of most metabolic parameters analyzed, fIGF1 levels remained elevated and hypothalamic ObR decreased in both sexes. Moreover, there was sex differences in both the response to HFD and the switch to chow including circulating levels of IGF2 and IGFBP2, factors previously reported to be involved in glucose metabolism. Indeed, glucose metabolism was also differentially modified in males and females, suggesting that these observations could be related.


Subject(s)
Diet, High-Fat , Obesity/diet therapy , Obesity/metabolism , Weight Loss/physiology , Animals , Blood Glucose/analysis , Blood Glucose/metabolism , Energy Intake , Female , Hypothalamus/metabolism , Insulin Resistance , Insulin-Like Growth Factor Binding Protein 2/blood , Insulin-Like Growth Factor Binding Protein 3/blood , Insulin-Like Growth Factor I/analysis , Insulin-Like Growth Factor II/analysis , Leptin/blood , Male , Mice , Mice, Inbred C57BL , Obesity/etiology , Sex Characteristics
9.
Neuroscience ; 426: 59-68, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31805254

ABSTRACT

Estradiol not only participates in the regulation of energy metabolism in adulthood, but also during the first stages of life as it modulates the alterations induced by under- and over-nutrition. The objectives of the present study were to determine: 1) If estradiol is involved in the normal programming of energy metabolism in rats; 2) If there is a specific window of time for this programming and 3) If males and females are differentially vulnerable to the action of this hormone. Estrogen receptors (ER) α, ERß and GPER were blocked by their specific antagonists MPP, PHTPP and G15, respectively, from postnatal day (P) 1 (the day of birth) to P5 or from P5 to P13. Physiological parameters such as body weight, fat depots and caloric intake were then analysed at P90. Hypothalamic AgRP, POMC, MC4R, ERα, ERß and GPER mRNA levels and plasma levels of estradiol, were also studied. We found that blocking ER receptors from P5 to P13 significantly decreases long-term body weight in males and hypothalamic POMC mRNA levels in females. The blocking of ERs from P1 to P5 only affected plasma estradiol levels in females. The present results indicate programming actions of estradiol from P5 to P13 on body weight in male and POMC expression in female rats and emphasize the importance of including both sexes in metabolic studies. It is necessary to unravel the mechanisms that underlie the actions of estradiol on food intake, both during development and in adulthood, and to determine how this programming differentially takes place in males and females.


Subject(s)
Energy Metabolism/drug effects , Estrogen Receptor beta/antagonists & inhibitors , Receptors, Estradiol/antagonists & inhibitors , Animals , Body Weight/drug effects , Body Weight/physiology , Energy Metabolism/physiology , Estradiol/pharmacology , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Female , Hypothalamus/metabolism , Male , Receptors, Estrogen/antagonists & inhibitors , Receptors, Estrogen/metabolism
10.
J Neuroendocrinol ; 32(1): e12756, 2020 01.
Article in English | MEDLINE | ID: mdl-31179596

ABSTRACT

Obesity is one of the most important health problems facing developed countries because being overweight is associated with a higher incidence of type 2 diabetes, cardiovascular disease and cancer, as well as other comorbidities. Although increased weight gain results from a combination of poor dietary habits and decreased energy expenditure, not all individuals have equal propensities to gain weight or to develop secondary complications of obesity. This is partially a result not only of genetics, including sex, but also the time during which an individual is exposed to an obesogenic environment. In the present study, we have compared the response of male and female mice to short-term exposure to a high-fat diet (HFD) or a low-fat diet during the peripubertal period (starting at 42 days of age) because this is a stage of dramatic hormonal and metabolic modifications. After 1 week on a HFD, there was no significant increase in body weight, although females significantly increased their energy intake. Serum leptin levels increased in both sexes, even though no change in fat mass was detected. Glyceamia and homeostasis model assessment increased in males, suggesting a rapid change in glucose metabolism. Hypothalamic pro-opiomelanocortin mRNA levels were significantly higher in females on a HFD compared to all other groups, which may be an attempt to reduce their increased energy intake. Hypothalamic inflammation and gliosis have been implicated in the development of secondary complications of obesity; however, no indication of activation of inflammatory processes or gliosis was found in response to 1 week of HFD in the hypothalamus, hippocampus or cerebellum of these young mice. These results indicate that there are both sex and age effects in the response to poor dietary intake because peripubertal male and female mice respond differently to short-term dietary changes and this response is different from that reported in adult rodents.


Subject(s)
Blood Glucose/metabolism , Body Weight/physiology , Diet, High-Fat , Hypothalamus/metabolism , Sexual Maturation/physiology , Adiposity/physiology , Animals , Energy Intake/physiology , Female , Insulin/blood , Leptin/blood , Male , Mice , Pro-Opiomelanocortin/genetics , Pro-Opiomelanocortin/metabolism , Sex Factors
11.
J Neuroendocrinol ; 31(5): e12671, 2019 05.
Article in English | MEDLINE | ID: mdl-30561077

ABSTRACT

The role of glial cells, including astrocytes, in metabolic control has received increasing attention in recent years. Although the original interest in these macroglial cells was a result of astrogliosis being observed in the hypothalamus of diet-induced obese subjects, studies have also focused on how they participate in the physiological control of appetite and energy expenditure. Astrocytes express receptors for numerous hormones, growth factors and neuropeptides. Some functions of astrocytes include transport of nutrients and hormones from the circulation to the brain, storage of glycogen, participation in glucose sensing, synaptic plasticity, uptake and metabolism of neurotransmitters, release of substances to modify neurotransmission, and cytokine production, amongst others. In the hypothalamus, these physiological glial functions impact on neuronal circuits that control systemic metabolism to modify their outputs. The initial response of astrocytes to poor dietary habits and obesity involves activation of neuroprotective mechanisms but, with chronic exposure to these situations, hypothalamic astrocytes participate in the development of some of the damaging secondary effects. The present review discusses not only some of the physiological functions of hypothalamic astrocytes in metabolism, but also their role in the secondary complications of obesity, such as insulin resistance and cardiovascular affectations.


Subject(s)
Astrocytes/metabolism , Hypothalamus/metabolism , Neurosecretory Systems/metabolism , Animals , Cardiovascular Diseases/complications , Cardiovascular Diseases/metabolism , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Energy Metabolism , Humans , Hypothalamus/physiopathology , Insulin Resistance/physiology , Neurons/metabolism , Neurosecretory Systems/physiopathology , Obesity/metabolism
12.
Acta Physiol (Oxf) ; 226(2): e13244, 2019 06.
Article in English | MEDLINE | ID: mdl-30589509

ABSTRACT

AIM: We aimed to investigate whether a dysregulated maternal diet during gestation and lactation induces long-lasting changes in the hypothalamic control of feeding behavior in the offspring and whether this effect is sex specific. METHODS: The study included an analysis of appetite-regulating metabolic hormones and hypothalamic signaling in male and female offspring in adulthood after exposure to a free-choice high-calorie palatable low-protein (P) diet or standard chow (C) during (pre)gestation/lactation (maternal) and/or postweaning (offspring). RESULTS: Maternal exposure to the P diet resulted in decreased protein intake and body weight gain in dams and decreased body weight gain in offspring during lactation. The maternal P diet (PC) specifically increased feed efficacy and decreased body weight and cholesterol levels in the female offspring in adulthood, but no changes in adiposity or leptin levels were found. In contrast, P diet exposure after weaning (CP and PP) increased caloric intake, adiposity and circulating levels of leptin in the male and female offspring in adulthood. The hypothalami of the female offspring exposed to the maternal P diet (PC and PP) expressed high levels of the phospho-leptin receptor and low levels of SOCS3, phospho-IRS1 and phospho-AMPK, regardless of the postweaning diet. The hypothalami of the female rats in the PC group also showed increased levels of STAT3 and the orexigenic neuropeptide Agrp. CONCLUSIONS: Maternal exposure to a free-choice high-calorie low-protein diet induces a long-term feed efficacy associated with changes in leptin signaling through IRS-1 and AMPK dephosphorylation in the hypothalami of female offspring in adulthood.


Subject(s)
Behavior, Animal/drug effects , Diet, Protein-Restricted/adverse effects , Leptin/pharmacology , Prenatal Exposure Delayed Effects , AMP-Activated Protein Kinases/drug effects , AMP-Activated Protein Kinases/metabolism , Animals , Behavior, Animal/physiology , Feeding Behavior/drug effects , Feeding Behavior/physiology , Female , Hypothalamus/drug effects , Hypothalamus/metabolism , Leptin/metabolism , Pregnancy , Prenatal Exposure Delayed Effects/metabolism , Rats, Wistar , Receptors, Leptin/drug effects , Signal Transduction/drug effects
13.
Nutr Neurosci ; 22(1): 29-39, 2019 Jan.
Article in English | MEDLINE | ID: mdl-28696162

ABSTRACT

BACKGROUND: Overnutrition due to a high-fat diet (HFD) can increase the vulnerability of the metabolic system to maladjustments. Estradiol has an inhibitory role on food intake and this hormone has demonstrated to be a crucial organizer during brain development. OBJECTIVE: Our aim was to determine whether increased levels of estradiol in the early postnatal period modulate the alterations in metabolism and brain metabolic circuits produced by overnutrition. METHODS: Twenty-four male and 24 female Wistar rats were submitted to a HFD (34.9% fat) or a control diet (5% fat) from gestational day 6. From postnatal (P) 6 to P13, both control and HFD groups were administered a s.c. injection of vehicle or estradiol benzoate (0.4 mg/kg), resulting in eight experimental groups (n = 6 in each group). Body weight, food intake and subcutaneous, visceral, and brown fat pads were measured. Agouti-related peptide, neuropeptide Y, orexin, and proopiomelanocortin (POMC) were analyzed by quantitative real-time polymerase chain reaction assay and plasma estradiol levels were measured by ELISA. RESULTS: Males fed a HFD showed an increase in body weight and the amount of visceral and subcutaneous fat, which was coincident with an increase in the number of kilocalories ingested. Neonatal estradiol treatment restored the body weight and subcutaneous fat of HFD males to control levels. Hypothalamic POMC mRNA levels in HFD females were increased with respect to control females. This increase was reverted with estradiol treatment during development. DISCUSSION: HFD and estradiol treatment have different effects on males and females. Overnutrition affects physiological parameters, such as body weight, visceral, and subcutaneous fat content, in males, while females present alterations in hypothalamic POMC mRNA levels. Hence, the increase in estradiol levels during a period that is critical for the programing of the feeding system can modulate some of the alterations produced by the continuous intake of high-fat content food.


Subject(s)
Diet, High-Fat/adverse effects , Estradiol/analogs & derivatives , Hypothalamus/pathology , Overnutrition/physiopathology , Adiposity , Animals , Body Weight , Diet , Disease Models, Animal , Estradiol/blood , Estradiol/pharmacology , Female , Hypothalamus/drug effects , Male , Neuropeptide Y/metabolism , Orexins/metabolism , Pro-Opiomelanocortin/metabolism , Rats , Rats, Wistar , Sex Factors
14.
Prog Neurobiol ; 176: 18-32, 2019 05.
Article in English | MEDLINE | ID: mdl-30194984

ABSTRACT

The hypothalamus is the main integrating center for metabolic control. Our understanding of how hypothalamic circuits function to control appetite and energy expenditure has increased dramatically in recent years, due to the rapid rise in the incidence of obesity and the search for effective treatments. Increasing evidence indicates that these treatments will most likely differ between males and females. Indeed, sex differences in metabolism have been demonstrated at various levels, including in two of the most studied neuronal populations involved in metabolic control: the anorexigenic proopiomelanocortin neurons and the orexigenic neuropeptide Y/Agouti-related protein neurons. Here we review what is known to date regarding the sex differences in these two neuronal populations, as well as other neuronal populations involved in metabolic control and glial cells.


Subject(s)
Energy Metabolism/physiology , Hypothalamus/metabolism , Sex Characteristics , Animals , Astrocytes/metabolism , Female , Humans , Male , Neurons/metabolism
15.
Front Neuroendocrinol ; 48: 3-12, 2018 01.
Article in English | MEDLINE | ID: mdl-28552663

ABSTRACT

Males and females have distinct propensities to develop obesity and its related comorbidities, partially due to gonadal steroids. There are sex differences in hypothalamic neuronal circuits, as well as in astrocytes, that participate in metabolic control and the development of obesity-associated complications. Astrocytes are involved in nutrient transport and metabolism, glucose sensing, synaptic remodeling and modulation of neuronal signaling. They express receptors for metabolic hormones and mediate effects of these metabolic signals on neurons, with astrogliosis occurring in response to high fat diet and excess weight gain. However, most studies of obesity have focused on males. Recent reports indicate that male and female astrocytes respond differently to metabolic signals and this could be involved in the differential response to high fat diet and the onset of obesity-associated pathologies. Here we focus on the sex differences in response to obesogenic paradigms and the possible role of hypothalamic astrocytes in this phenomenon.


Subject(s)
Astrocytes/metabolism , Gonadal Steroid Hormones/metabolism , Hypothalamus/metabolism , Obesity/metabolism , Sex Characteristics , Animals , Female , Humans , Male
16.
Endocrinology ; 159(1): 368-387, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29077836

ABSTRACT

Astrocytes participate in both physiological and pathophysiological responses to metabolic and nutrient signals. Although most studies have focused on the astrocytic response to weight gain due to high-fat/high-carbohydrate intake, surplus intake of a balanced diet also induces excess weight gain. We have accessed the effects of neonatal overnutrition, which has both age- and sex-dependent effects on weight gain, on hypothalamic inflammation/gliosis. Although both male and female Wistar rats accumulate excessive fat mass as early as postnatal day (PND) 10 with neonatal overnutrition, no increase in hypothalamic cytokine levels, markers of astrocytes or microglia, or inflammatory signaling pathways were observed. At PND 50, no effect of neonatal overnutriton was found in either sex, whereas at PND 150, males again weighed significantly more than their controls, and this was coincident with an increase in markers of inflammation and astrogliosis in the hypothalamus. Circulating triglycerides and free fatty acids were also elevated in these males, but not in females or in either sex at PND 10. Thus, the effects of fatty acids and estrogens on astrocytes in vitro were analyzed. Our results indicate that changes in circulating fatty acid levels may be involved in the induction of hypothalamic inflammation/gliosis in excess weight gain, even on a normal diet, and that estrogens could participate in the protection of females from these processes. In conclusion, the interaction of developmental influences, dietary composition, age, and sex determines the central inflammatory response and the associated long-term outcomes of excess weight gain.


Subject(s)
Astrocytes/metabolism , Gliosis/etiology , Hyperphagia/physiopathology , Hypothalamic Diseases/etiology , Hypothalamus/metabolism , Microglia/metabolism , Adiposity , Age Factors , Animals , Animals, Newborn , Astrocytes/immunology , Astrocytes/pathology , Biomarkers/metabolism , Cells, Cultured , Cytokines/metabolism , Female , Gene Expression Regulation, Developmental , Gliosis/immunology , Gliosis/metabolism , Gliosis/pathology , Hypothalamic Diseases/immunology , Hypothalamic Diseases/metabolism , Hypothalamic Diseases/pathology , Hypothalamus/immunology , Hypothalamus/pathology , Inflammation Mediators/metabolism , Male , Microglia/immunology , Microglia/pathology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Rats, Wistar , Sex Characteristics , Signal Transduction , Weight Gain
17.
Mol Cell Endocrinol ; 454: 125-134, 2017 10 15.
Article in English | MEDLINE | ID: mdl-28641938

ABSTRACT

The importance of the neonatal leptin surge in rodents in neurodevelopmental processes has aroused curiosity in its implication in other physiological systems. Given the role of leptin in neuro-immune interactions, we hypothesized that the neonatal leptin surge could have an effect on the oxidative and inflammatory stress situations of both systems. We blocked the neonatal leptin surge by a leptin antagonist and measured several parameters of oxidative and inflammatory stress in the spleen, hypothalamus and adipose tissue of peripubertal/adolescent rats. The treated rats showed lower activity of several antioxidant enzymes in the spleen and their leukocytes released lower levels of mitogen-stimulated IL-10 and IL-13 and higher levels of TNF-alpha. In conclusion, the neonatal leptin surge may have a key role in the establishment of adequate redox and inflammatory states in the immune system, which is important for the generation of adequate immune responses and to obtain and maintain good health.


Subject(s)
Inflammation/metabolism , Inflammation/pathology , Leptin/antagonists & inhibitors , Sexual Maturation , Adipose Tissue, White/metabolism , Animals , Animals, Newborn , Body Weight , Catalase/metabolism , Cytokines/metabolism , Female , Glutathione/metabolism , Glutathione Peroxidase/metabolism , Glutathione Reductase/metabolism , Hypothalamus/metabolism , Male , Organ Size , Oxidation-Reduction , Oxidative Stress , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Wistar , Superoxide Dismutase/metabolism
18.
J Mol Endocrinol ; 58(1): R59-R71, 2017 01.
Article in English | MEDLINE | ID: mdl-27864453

ABSTRACT

The search for new strategies and drugs to abate the current obesity epidemic has led to the intensification of research aimed at understanding the neuroendocrine control of appetite and energy expenditure. This intensified investigation of metabolic control has also included the study of how glial cells participate in this process. Glia, the most abundant cell type in the central nervous system, perform a wide spectrum of functions and are vital for the correct functioning of neurons and neuronal circuits. Current evidence indicates that hypothalamic glia, in particular astrocytes, tanycytes and microglia, are involved in both physiological and pathophysiological mechanisms of appetite and metabolic control, at least in part by regulating the signals reaching metabolic neuronal circuits. Glia transport nutrients, hormones and neurotransmitters; they secrete growth factors, hormones, cytokines and gliotransmitters and are a source of neuroprogenitor cells. These functions are regulated, as glia also respond to numerous hormones and nutrients, with the lack of specific hormonal signaling in hypothalamic astrocytes disrupting metabolic homeostasis. Here, we review some of the more recent advances in the role of glial cells in metabolic control, with a special emphasis on the differences between glial cell responses in males and females.


Subject(s)
Energy Metabolism , Neuroglia/metabolism , Animals , Astrocytes/metabolism , Ependymoglial Cells/metabolism , Female , Gliosis/metabolism , Gliosis/pathology , Humans , Hypothalamus/metabolism , Hypothalamus/pathology , Male , Microglia/metabolism , Neurons/metabolism , Sex Factors
19.
Peptides ; 86: 63-71, 2016 12.
Article in English | MEDLINE | ID: mdl-27751931

ABSTRACT

The neonatal leptin surge, occurring from postnatal day (PND) 5 to 13 and peaking at PND9 in rodents, is important for the development of neuroendocrine circuits involved in metabolic control and reproductive function. We previously demonstrated that treatment with a leptin antagonist from PND 5 to 9, coincident with peak leptin levels in the neonatal surge, modified trophic factors and markers of cell turnover and neuronal maturation in the hypothalamus of peri-pubertal rats. The kisspeptin system and metabolic neuropeptide and hormone levels were also modified. Here our aim was to investigate if the timing of pubertal onset is altered by neonatal leptin antagonism and if the previously observed peripubertal modifications in hormones and neuropeptides persist into adulthood and affect male sexual behavior. To this end, male Wistar rats were treated with a pegylated super leptin antagonist (5mg/kg, s.c.) from PND 5 to 9 and killed at PND102-103. The appearance of external signs of pubertal onset was delayed. Hypothalamic kiss1 mRNA levels were decreased in adult animals, but sexual behavior was not significantly modified. Although there was no effect on body weight or food intake, circulating leptin, insulin and triglyceride levels were increased, while hypothalamic leptin receptor, POMC and AgRP mRNA levels were decreased. In conclusion, alteration of the neonatal leptin surge can modify the timing of pubertal onset and have long-term effects on hypothalamic expression of reproductive and metabolic neuropeptides.


Subject(s)
Leptin/physiology , Sexual Maturation , Animals , Animals, Newborn , Gene Expression , Hypothalamus/metabolism , Kisspeptins/genetics , Kisspeptins/metabolism , Male , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neuropeptide Y/genetics , Neuropeptide Y/metabolism , Pro-Opiomelanocortin/genetics , Pro-Opiomelanocortin/metabolism , Rats, Wistar , Receptors, Leptin/metabolism , Sexual Behavior, Animal , Signal Transduction , Weight Gain
20.
Dis Model Mech ; 9(5): 573-83, 2016 05 01.
Article in English | MEDLINE | ID: mdl-27013528

ABSTRACT

Insulin receptor substrate-2-deficient (IRS2(-/-)) mice are considered a good model to study the development of diabetes because IRS proteins mediate the pleiotropic effects of insulin-like growth factor-I (IGF-I) and insulin on metabolism, mitogenesis and cell survival. The hypothalamus might play a key role in the early onset of diabetes, owing to its involvement in the control of glucose homeostasis and energy balance. Because some inflammatory markers are elevated in the hypothalamus of diabetic IRS2(-/-) mice, our aim was to analyze whether the diabetes associated with the absence of IRS2 results in hypothalamic injury and to analyze the intracellular mechanisms involved. Only diabetic IRS2(-/-) mice showed increased cell death and activation of caspase-8 and -3 in the hypothalamus. Regulators of apoptosis such as FADD, Bcl-2, Bcl-xL and p53 were also increased, whereas p-IκB and c-FLIPL were decreased. This was accompanied by increased levels of Nox-4 and catalase, enzymes involved in oxidative stress. In summary, the hypothalamus of diabetic IRS2(-/-) mice showed an increase in oxidative stress and inflammatory markers that finally resulted in cell death via substantial activation of the extrinsic apoptotic pathway. Conversely, non-diabetic IRS2(-/-) mice did not show cell death in the hypothalamus, possibly owing to an increase in the levels of circulating IGF-I and in the enhanced hypothalamic IGF-IR phosphorylation that would lead to the stimulation of survival pathways. In conclusion, diabetes in IRS2-deficient male mice is associated with increased oxidative stress and apoptosis in the hypothalamus.


Subject(s)
Apoptosis , Diabetes Mellitus, Experimental/pathology , Hypothalamus/pathology , Insulin Receptor Substrate Proteins/deficiency , Oxidative Stress , Animals , Biomarkers/metabolism , Caspases/metabolism , Cytokines/metabolism , Inflammation/pathology , Insulin Receptor Substrate Proteins/metabolism , Insulin-Like Growth Factor I/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptor, IGF Type 1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL