Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Biosci Rep ; 44(4)2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38577975

ABSTRACT

Since 1975, the incidence of obesity has increased to epidemic proportions, and the number of patients with obesity has quadrupled. Obesity is a major risk factor for developing other serious diseases, such as type 2 diabetes mellitus, hypertension, and cardiovascular diseases. Recent epidemiologic studies have defined obesity as a risk factor for the development of neurodegenerative diseases, such as Alzheimer's disease (AD) and other types of dementia. Despite all these serious comorbidities associated with obesity, there is still a lack of effective antiobesity treatment. Promising candidates for the treatment of obesity are anorexigenic neuropeptides, which are peptides produced by neurons in brain areas implicated in food intake regulation, such as the hypothalamus or the brainstem. These peptides efficiently reduce food intake and body weight. Moreover, because of the proven interconnection between obesity and the risk of developing AD, the potential neuroprotective effects of these two agents in animal models of neurodegeneration have been examined. The objective of this review was to explore anorexigenic neuropeptides produced and acting within the brain, emphasizing their potential not only for the treatment of obesity but also for the treatment of neurodegenerative disorders.


Subject(s)
Anti-Obesity Agents , Neuropeptides , Neuroprotective Agents , Obesity , Humans , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Animals , Obesity/drug therapy , Obesity/metabolism , Neuropeptides/metabolism , Neuropeptides/pharmacology , Neuropeptides/therapeutic use , Anti-Obesity Agents/pharmacology , Anti-Obesity Agents/therapeutic use , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/prevention & control , Hypothalamus/drug effects , Hypothalamus/metabolism , Hypothalamus/pathology , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/prevention & control , Brain/drug effects , Brain/metabolism , Brain/pathology , Eating/drug effects
2.
Cell Mol Neurobiol ; 43(8): 4295-4307, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37828299

ABSTRACT

Disruption of leptin (LEP) signaling in the hypothalamus caused by type 2 diabetes (T2D) can impair appetite regulation. The aim of this study was to investigate whether the improvement in appetite regulation induced by high-intensity interval training (HIIT) in rats with T2D can be mediated by LEP signaling. In this study, 20 male Wister rats were randomly assigned to one of four groups: CO (non-type 2 diabetes control), T2D (type 2 diabetes), EX (non-type 2 diabetes exercise), and T2D + EX (type 2 diabetes + exercise).To induce T2D, a combination of a high-fat diet for 2 months and a single dose of streptozotocin (35 mg/kg) was administered. Rats in the EX and T2D + EX groups performed 4-10 intervals of treadmill running at 80-100% of their maximum velocity (Vmax). Homeostatic Model Assessment for Insulin Resistance (HOMA-IR), serum levels of insulin (INS) and LEP (LEPS) as well as hypothalamic expression of LEP receptors (LEP-R), Janus kinase 2 (JAK-2), signal transducer and activator of transcription 3 (STAT-3), neuropeptide Y (NPY), agouti-related protein (AGRP), pro-opiomelanocortin cocaine (POMC), amphetamine-related transcript (CART), suppressor of cytokine signaling (SOCS3), forkhead box protein O1 (FOXO1) were assessed. ANOVA and Tukey post hoc tests were used to compare the results between the groups. The levels of LEPS and INS, as well as the levels of LEP-R, JAK-2, STAT-3, POMC, and CART in the hypothalamus were found to be higher in the T2D + EX group compared to the T2D group. On the other hand, the levels of HOMA-IR, NPY, AGRP, SOCS3, and FOXO1 were lower in the T2D + EX group compared to the T2D group (P < 0.0001). The findings of this study suggest that HIIT may improve appetite regulation in rats with T2D, and LEP signaling may play a crucial role in this improvement. Graphical abstract (leptin signaling in the hypothalamus), Leptin (LEP), Leptin receptor (LEP-R), Janus kinase 2 (JAK2), Signal transducer and activator of transcription 3 (STAT3), expressing Neuropeptide Y (NPY), Agouti-related protein (AGRP), anorexigenic neurons (expressing pro-opiomelanocortin cocaine (POMC), Amphetamine-related transcript (CART), suppressor of cytokine signaling (SOCS3), forkhead box protein O1 (FOXO1).


Subject(s)
Cocaine , Diabetes Mellitus, Type 2 , High-Intensity Interval Training , Rats , Male , Animals , Agouti-Related Protein/metabolism , Neuropeptide Y/metabolism , Leptin/metabolism , Appetite Regulation/physiology , Pro-Opiomelanocortin/metabolism , STAT3 Transcription Factor/metabolism , Forkhead Box Protein O1/metabolism , Janus Kinase 2/metabolism , Diabetes Mellitus, Type 2/therapy , Diabetes Mellitus, Type 2/metabolism , Rats, Wistar , Hypothalamus/metabolism , Insulin/metabolism , Amphetamines/metabolism , Cocaine/metabolism , Cytokines/metabolism
3.
Endocr J ; 69(12): 1363-1372, 2022 Dec 28.
Article in English | MEDLINE | ID: mdl-36372440

ABSTRACT

It has been well established that undernutrition and low energy availability disturb female reproductive functions in humans and many animal species. These reproductive dysfunctions are mainly caused by alterations of some hypothalamic factors, and consequent reduction of gonadotrophin-releasing hormone (GnRH) secretion. Evidence from literature suggests that increased activity of orexigenic factors and decreased activity of anorexigenic/satiety-related factors in undernourished conditions attenuate GnRH secretion in an integrated manner. Likewise, the activity of kisspeptin neurons, which is a potent stimulator of GnRH, is also reduced in undernourished conditions. In addition, it has been suggested that gonadotrophin-inhibitory hormone, which has anti-GnRH and gonadotrophic effects, may be involved in reproductive dysfunctions under several kinds of stress conditions. It should be remembered that these alterations, i.e., promotion of feeding behavior and temporary suppression of reproductive functions, are induced to prioritize the survival of individual over that of species, and that improvements in metabolic and nutritional conditions should be considered with the highest priority.


Subject(s)
Gonadotropin-Releasing Hormone , Malnutrition , Animals , Female , Humans , Gonadotropins , Hypothalamus/metabolism , Kisspeptins/physiology
4.
J Obstet Gynaecol Res ; 48(3): 568-575, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34979587

ABSTRACT

It is well known that undernourished conditions disturb female reproductive functions in many species, including humans. These alterations are mainly caused by a reduction in gonadotrophin-releasing hormone (GnRH) secretion from the hypothalamus. Evidence from the literature suggests that some hypothalamic factors play pivotal roles in the coordination of reproductive functions and energy homeostasis in response to environmental cues and internal nutritional status. Generally, anorexigenic/satiety-related factors, such as leptin, alpha-melanocyte-stimulating hormone, and proopiomelanocortin, promote GnRH secretion, whereas orexigenic factors, such as neuropeptide Y, agouti-related protein, orexin, and ghrelin, attenuate GnRH secretion. Conversely, gonadotrophin-inhibitory hormone, which exerts anti-GnRH and gonadotrophic effects, promotes feeding behavior in many species. In addition, the activity of kisspeptin, which is a potent stimulator of GnRH, is reduced by undernourished conditions. Under normal nutritional conditions, these factors are coordinated to maintain both feeding behavior and reproductive functions. However, in undernourished conditions their activity levels are markedly altered to promote feeding behavior and temporarily suppress reproductive functions, in order to prioritize the survival of the individual over that of the species.


Subject(s)
Gonadotropin-Releasing Hormone , Kisspeptins , Female , Homeostasis/physiology , Humans , Hypothalamus/metabolism , Kisspeptins/physiology , Neuropeptide Y/metabolism
5.
Int J Mol Sci ; 22(9)2021 May 07.
Article in English | MEDLINE | ID: mdl-34067001

ABSTRACT

Investigations into the mechanisms regulating obesity are frantic and novel translational approaches are needed. The raccoon dog (Nyctereutes procyonoides) is a canid species representing a promising model to study metabolic regulation in a species undergoing cycles of seasonal obesity and fasting. To understand the molecular mechanisms of metabolic regulation in seasonal adaptation, we analyzed key central nervous system and peripheral signals regulating food intake and metabolism from raccoon dogs after autumnal fattening and winter fasting. Expressions of neuropeptide Y (NPY), orexin-2 receptor (OX2R), pro-opiomelanocortin (POMC) and leptin receptor (ObRb) were analyzed as examples of orexigenic and anorexigenic signals using qRT-PCR from raccoon dog hypothalamus samples. Plasma metabolic profiles were measured with 1H NMR-spectroscopy and LC-MS. Circulating hormones and cytokines were determined with canine specific antibody assays. Surprisingly, NPY and POMC were not affected by the winter fasting nor autumn fattening and the metabolic profiles showed a remarkable equilibrium, indicating conserved homeostasis. However, OX2R and ObRb expression changes suggested seasonal regulation. Circulating cytokine levels were not increased, demonstrating that the autumn fattening did not induce subacute inflammation. Thus, the raccoon dog developed seasonal regulatory mechanisms to accommodate the autumnal fattening and prolonged fasting making the species unique in coping with the extreme environmental challenges.


Subject(s)
Adiposity , Fasting/metabolism , Metabolome , Raccoon Dogs/metabolism , Seasons , Adipose Tissue/blood supply , Adipose Tissue/pathology , Animals , Biomarkers/metabolism , Body Weight , Discriminant Analysis , Female , Hormones/blood , Hypothalamus/metabolism , Inflammation/pathology , Least-Squares Analysis , Limit of Detection , Multivariate Analysis , Peptides/genetics , Peptides/metabolism , Proton Magnetic Resonance Spectroscopy , RNA, Messenger/genetics , RNA, Messenger/metabolism , Raccoon Dogs/blood , Receptors, Peptide/metabolism
6.
Article in English | MEDLINE | ID: mdl-34119636

ABSTRACT

Ferulic acid (FA) is a phenolic acid found within the plant cell wall that has physiological benefits as an antioxidant. Although metabolic benefits of FA supplementation are described, lacking are reports of effects on appetite regulation. Thus, our objective was to determine if FA affects food or water intake, using chicks as a model. At 4 days post-hatch, broiler chicks were intraperitoneally injected with 0 (vehicle), 12.5, 25, or 50 mg/kg of FA. Chicks treated with 50 mg/kg of FA consumed 70% less food than controls at 30 min post-injection, and the effect dissipated thereafter. Water intake was not affected at any time. In a behavior analysis, FA-treated chicks defecated fewer times than vehicle-injected chicks, while other behaviors were not affected. There was an increase in c-Fos immunoreactivity within the hypothalamic arcuate nucleus (ARC) of FA-treated chicks, and no differences were detected in other nuclei. mRNA abundance was measured in the whole hypothalamus and the ARC. There was decreased hypothalamic galanin, ghrelin, melanocortin receptor 3, and pro-opiomelanocortin (POMC) mRNA in FA-treated chicks. Within the ARC, there was an increase in c-Fos mRNA and a decrease in POMC mRNA in response to FA. It is likely that the mechanism responsible for mediating FA's transient effects on food intake originates within the ARC, possibly involving POMC. A greater understanding of the short-term, mild appetite-suppressive effects of FA may have applications to treating eating disorders and modulating food intake in animal models of obesity.


Subject(s)
Chickens/metabolism , Coumaric Acids/chemistry , Phytochemicals/chemistry , Animals , Animals, Newborn , Anorexia/chemically induced , Apoptosis , Appetite , Appetite Regulation , Arcuate Nucleus of Hypothalamus/metabolism , Behavior, Animal , Coumaric Acids/pharmacology , Disease Models, Animal , Drinking/drug effects , Galanin/metabolism , Ghrelin/metabolism , Hypothalamus/metabolism , Pro-Opiomelanocortin/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Receptor, Melanocortin, Type 3/metabolism , Signal Transduction
7.
Article in English | MEDLINE | ID: mdl-33785435

ABSTRACT

Cholecystokinin (CCK) and peptide YY (PYY) have been investigated as gut hormones that send satiation signals to the brain in mammals. There is evidence that chicken PYY mRNA expression was the highest in the pancreas compared to other tissues. We recently suggested that insulin-like growth factor (IGF)-1 and its binding proteins (IGFBPs) may be involved in the appetite regulation system in chicks. In the present study, in order to evaluate the possible roles of CCK, PYY, and IGF-related proteins in the appetite regulation system in chicks, we analyzed changes in the mRNA levels of these genes in response to fasting and re-feeding in layer and hyperphagic broiler chicks. In layer chicks, 12 h of fasting reduced the mRNA levels of intestinal CCK, PYY, Y2 receptor, and pancreatic PYY, and these changes were reversed by 12 h of re-feeding. On the other hand, in broiler chicks 12 h of fasting reduced the mRNA levels of intestinal PYY and Y2 receptor, but not intestinal CCK and pancreatic PYY, and these changes were reversed by 12 h of re-feeding. Hypothalamic NPY mRNA significantly increased by 12 h of fasting in both chicks, and these changes were reversed by re-feeding. Also, 12 h of fasting significantly increased the mRNA levels of hypothalamic agouti-related protein and reduced the mRNA levels of hepatic IGF-1 only in broiler chicks, and 12 h of re-feeding did not change these. IGFBP-1 and -2 mRNA levels were markedly increased by 12 h of fasting in both chicks, and these changes were reversed by re-feeding. IGFBP-3 mRNA levels were increased by 12 h of fasting only in layer chicks, while re-feeding reduced the mRNA levels of IGFBP-3 in both types of chicks. These results suggest that several peripheral hormones, such as pancreatic PYY and intestinal CCK, may not play important roles in the regulation of food intake in broiler chicks.


Subject(s)
Cholecystokinin/metabolism , Fasting/physiology , Pancreas/metabolism , Peptide YY/physiology , Animals , Appetite Regulation , Chickens , Eating/physiology , Feeding Behavior , Food Deprivation , Gene Expression Regulation , Hormones/metabolism , Hyperphagia , Hypothalamus/metabolism , Ileum/metabolism , Male , Neuropeptides/metabolism
8.
Neurosci Lett ; 736: 135282, 2020 09 25.
Article in English | MEDLINE | ID: mdl-32738351

ABSTRACT

Beta-melanocyte-stimulating hormone (ß-MSH), when centrally injected, induces anorexigenic effects in rodents and chickens but its mechanism remains unclear. Thus, the primary goal of this research was to elucidate the hypothalamic mechanism using chickens. Intracerebroventricular injection of 0.3, 1.0 and 3.0 nmol of ß-MSH decreased food intake for 540 min. Expression of hypothalamic mRNAs were affected by ß-MSH injection, including corticotrophin-releasing factor (CRF) and its receptor subtype 1 (CRFR1), mesotocin (MT) and its receptor (MTR), pro-opiomelanocortin, cocaine- and amphetamine-regulated transcript (CART), growth hormone secretagogue receptor (GHSR) and neuropeptide Y (NPY) receptor subtype 5 (NPYR5). Within the arcuate nucleus, expressions of NPY, agouti-related peptide, MT and MTR were increased by ß-MSH injection. ß-MSH-treated chicks had more CRF, CRFR1, CRF receptor subtype 2, GHSR, NPY receptor subtype 1 (NPYR1) and NPYR5 mRNA but lower levels of CART and ghrelin, in the paraventricular nucleus. Greater amounts of mRNA for MTR, GHSR, NPYR1 and NPYR5 and less CRF expression were observed in the ventromedial hypothalamus. In conclusion, central injection of ß-MSH potently reduced food intake and was associated with changes in mRNA expression of some anorexigenic factors in a hypothalamic nucleus-specific manner.


Subject(s)
Corticotropin-Releasing Hormone/metabolism , Eating/drug effects , Hypothalamus/drug effects , Oxytocin/analogs & derivatives , Receptors, Corticotropin-Releasing Hormone/metabolism , beta-MSH/pharmacology , Animals , Chickens , Hypothalamus/metabolism , Injections, Intraventricular , Oxytocin/metabolism
9.
Peptides ; 128: 170308, 2020 06.
Article in English | MEDLINE | ID: mdl-32229144

ABSTRACT

Since its discovery in 2006 by Oh-I and colleagues, NUCB2/nesfatin-1 encoded by nucleobindin-2 (NUCB2) has drawn sustained attention as reflected in over 500 publications. Among those, more than half focused on the alterations of food intake, body weight and metabolism (glucose, fat) induced by nesfatin-1 and/or NUCB2/nesfatin-1. In the current review we discuss the existing literature focusing on NUCB2/nesfatin-1's influence on food intake, body weight and glucose as well as fat metabolism and highlight gaps in knowledge.


Subject(s)
Body Weight/drug effects , Eating/drug effects , Nucleobindins/metabolism , Nucleobindins/pharmacology , Animals , Fats/metabolism , Glucose/metabolism , Humans , Hypothalamus/drug effects , Hypothalamus/metabolism
10.
Nutrients ; 12(3)2020 Mar 15.
Article in English | MEDLINE | ID: mdl-32183423

ABSTRACT

The satiating effect of whey proteins depends upon their unique amino acid composition because there is no difference when comparing whey proteins or a mix of amino acids mimicking the amino acid composition of whey proteins. The specific amino acids underlying the satiating effect of whey proteins have not been investigated to date. AIMS AND METHODS: The aim of the present study was to evaluate the appetite-suppressant effect of an isocaloric drink containing whey proteins or maltodextrins on appetite (satiety/hunger measured by a visual analogue scale or VAS), anorexigenic gastrointestinal peptides (circulating levels of glucagon-like peptide 1 (GLP-1) and peptide tyrosine tyrosine (PYY)) and amino acids (circulating levels of single, total [TAA] and branched-chain amino acids [BCAA]) in a cohort of obese female subjects (n = 8; age: 18.4 ± 3.1 years; body mass index, BMI: 39.2 ± 4.6 kg/m2). RESULTS: Each drink significantly increased satiety and decreased hunger, the effects being more evident with whey proteins than maltodextrins. Similarly, circulating levels of GLP-1, PYY and amino acids (TAA, BCAA and alanine, arginine, asparagine, citrulline, glutamine, hydroxyproline, isoleucine, histidine, leucine, lysine, methionine, ornithine, phenylalanine, proline, serine, threonine, tyrosine, and valine) were significantly higher with whey proteins than maltodextrins. In subjects administered whey proteins (but not maltodextrins), isoleucine, leucine, lysine, methionine, phenylalanine, proline, tyrosine, and valine were significantly correlated with hunger (negatively), satiety, and GLP-1 (positively). CONCLUSIONS: Eight specific amino acids (isoleucine, leucine, lysine, methionine, phenylalanine, proline, tyrosine, and valine) were implicated in the appetite-suppressant and GLP-1-stimulating effects of whey proteins, which may be mediated by their binding with nutrient-sensing receptors expressed by L cells within the gastrointestinal wall. The long-term satiating effect of whey proteins and the effectiveness of a supplementation with these amino acids (i.e., as a nutraceutical intervention) administered during body weight reduction programs need to be further investigated.


Subject(s)
Amino Acids/blood , Appetite Depressants/administration & dosage , Beverages , Glucagon-Like Peptide 1/drug effects , Obesity/physiopathology , Whey Proteins/administration & dosage , Adolescent , Appetite/drug effects , Cross-Over Studies , Dipeptides/drug effects , Enteroendocrine Cells/metabolism , Female , Humans , Isoleucine/blood , Leucine/blood , Lysine/blood , Methionine/blood , Obesity/therapy , Phenylalanine/blood , Polysaccharides/administration & dosage , Proline/blood , Tyrosine/blood , Valine/blood , Young Adult
11.
J Am Coll Nutr ; 39(4): 345-351, 2020.
Article in English | MEDLINE | ID: mdl-31369353

ABSTRACT

Nesfatin-1 is a peptide derived from nucleobindin-2 and involved in the regulation of food intake and hyperglycemia. Nesfatin-1 is a recently described anorexigenic peptide, which may be involved in weight loss, malnutrition, and the regulation of appetite. Nesfatin-1 has an effect on the regulation of glucose homeostasis as well as that of food intake. The aim of this article is to bring a different perspective to the readers on the effects of nesfatin-1 on food intake and hyperglycemia. The central injection of nesfatin-1 may produce anorexigenic effects. The circulating level of nesfatin-1 is thought to be regulated by nutritional status. Long-term changes in body weight can affect nesfatin-1 levels. In overweight and obese individuals, nesfatin-1 levels may increase. Nesfatin-1 is synthesized in the hypothalamic appetite control regions. Nesfatin-1 levels may decrease in individuals with diabetes but may increase in those with impaired glucose tolerance. Nesfatin-1 may have a reducing effect on glucose levels. In addition, an increase in glucose levels may lead to an increase in the release of nesfatin-1 from pancreatic cells. Injection of nesfatin-1 can prevent hepatic glucose formation and stimulate glucose uptake. Reduction of hypothalamic nesfatin-1 levels increases hepatic glucose flow and decreases glucose uptake from peripheral tissues. In the light of all this information, nesfatin-1 may be considered to be an important regulator in the metabolic process. Nesfatin-1 appears to be able to contribute to the treatment of obesity and diabetes because of its anorexigenic and antihyperglycemic effects. Key teaching pointsNesfatin-1 is a anorexigenic peptide.Nesfatin-1 is derived from Nucleobindin-2.Nucleobindin-2 mRNA is produced in different areas of the brain.Nesfatin-1 is an inhibitory factor on appetite and a regulator of energy balance that reduces the increase in body weight.


Subject(s)
Appetite Depressants/metabolism , Eating/drug effects , Hyperglycemia/metabolism , Nucleobindins/metabolism , Animals , Appetite/drug effects , Body Weight/drug effects , Energy Metabolism/drug effects , Glucose/metabolism , Glucose Intolerance/metabolism , Homeostasis/drug effects , Humans , Hypothalamus/metabolism , Nucleobindins/biosynthesis , Nutritional Status/drug effects
12.
Int J Mol Sci ; 20(20)2019 Oct 14.
Article in English | MEDLINE | ID: mdl-31615150

ABSTRACT

The micronutrients vitamins B9 and B12 act as methyl donors in the one-carbon metabolism involved in transmethylation reactions which critically influence epigenetic mechanisms and gene expression. Both vitamins are essential for proper development, and their deficiency during pregnancy has been associated with a wide range of disorders, including persisting growth retardation. Energy homeostasis and feeding are centrally regulated by the hypothalamus which integrates peripheral signals and acts through several orexigenic and anorexigenic mediators. We studied this regulating system in a rat model of methyl donor deficiency during gestation and lactation. At weaning, a predominance of the anorexigenic pathway was observed in deficient pups, with increased plasma peptide YY and increased hypothalamic pro-opiomelanocortin (POMC) mRNA, in line with abnormal leptin, ghrelin, and insulin secretion and/or signaling during critical periods of fetal and/or postnatal development of the hypothalamus. These results suggest that early methyl donor deficiency can affect the development and function of energy balance circuits, resulting in growth and weight deficits. Maternal administration of folic acid (3 mg/kg/day) during the perinatal period tended to rectify peripheral metabolic signaling and central neuropeptide and receptor expression, leading to reduced growth retardation.


Subject(s)
Energy Metabolism/genetics , Ghrelin/genetics , Peptide YY/genetics , Pro-Opiomelanocortin/genetics , Animals , Appetite Depressants/pharmacology , Energy Metabolism/drug effects , Feeding Behavior/drug effects , Female , Folic Acid/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Ghrelin/blood , Hypothalamus/metabolism , Insulin/blood , Insulin/genetics , Lactation , Leptin/blood , Leptin/genetics , Methylation/drug effects , Peptide YY/blood , Pregnancy , Pro-Opiomelanocortin/blood , RNA, Messenger/genetics , Rats , Vitamin B 12/genetics , Vitamin B 12/pharmacology
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.
Ecotoxicol Environ Saf ; 161: 459-466, 2018 10.
Article in English | MEDLINE | ID: mdl-29909315

ABSTRACT

Since 2010, Bisphenol A (BPA), an endocrine disruptor has been restricted and replaced by analogues like Bisphenol S (BPS). However, little is known about BPS effects and growing concern have suspected the "BPA-free" Label. Several recent studies suggest that BPS is associated with increased risk of diabetes and obesity. However, the underlying mechanisms remain unidentified. The current study investigates investigate BPS effects on hypothalamic neuropeptides regulating feeding behavior, either orexigenic or anorexigenic in Swiss Albino mice. We also studied the effect of BPS on the apelinergic system (apelin/apelin receptor (APJ)) as an original physiological system with pleiotropic actions. Bisphenol S at 25, 50, 100 µg/kg was administered to mice in water drink for 10 weeks started after weaning. Our results showed that BPS exposure alters orexigenic hypothalamic neuropeptide (AgRP) regulating feeding behavior but not anorexigenic neuropeptides (POMC, CART). Such orexigenic alterations may underlay appetite disorders leading to a concomitant food intake and body weight gain increase. In addition, data show that BPS affects the hypothalamic apelinergic system. We found a significant decrease in APJ mRNA but not in apelin expression. Based on hypothalamic APJ distribution, we suggested a potent specific physiological alteration of this receptor in mediating neuroendocrine responses in hypothalamus. Thus, our findings provide that BPS exposure could contribute to the development of obesity and metabolic disorders.


Subject(s)
Apelin/metabolism , Eating/drug effects , Hypothalamus/drug effects , Neuropeptides/metabolism , Phenols/toxicity , Sulfones/toxicity , Animals , Apelin Receptors/metabolism , Benzhydryl Compounds , Body Weight , Feeding Behavior/drug effects , Hypothalamus/metabolism , Male , Mice , Nerve Tissue Proteins/metabolism , Neuropeptides/genetics , Obesity/chemically induced , Pro-Opiomelanocortin/metabolism , RNA, Messenger/metabolism , Weight Gain
15.
Mol Neurobiol ; 55(6): 4718-4730, 2018 Jun.
Article in English | MEDLINE | ID: mdl-28717967

ABSTRACT

The hypothalamus is a key integrator of nutrient-seeking signals in the form of hormones and metabolites originated in both the central nervous system and the periphery. The main autocrine and paracrine target of orexinergic-related hormones such as leptin, orexin/hypocretin, and ghrelin are neuropeptide Y neurons located in the arcuate nucleus of the hypothalamus. The aim of this study was to investigate the expression and the molecular and functional relationships between leptin, orexin/hypocretin and ghrelin receptors. Biophysical studies in a heterologous system showed physical interactions between them, with potential formation of heterotrimeric complexes. Functional assays showed robust allosteric interactions particularly different when the three receptors are expressed together. Further biochemical and pharmacological assays provided evidence of heterotrimer functional expression in primary cultures of hypothalamic neurons. These findings constitute evidence of close relationships in the action of the three hormones already starting at the receptor level in hypothalamic cells.


Subject(s)
Ghrelin/metabolism , Hypothalamus/metabolism , Neurons/metabolism , Orexins/metabolism , Receptors, Ghrelin/metabolism , Receptors, Leptin/metabolism , Signal Transduction , Allosteric Regulation , Animals , HEK293 Cells , Humans , Protein Binding , Rats, Sprague-Dawley
16.
Adv Exp Med Biol ; 960: 381-397, 2017.
Article in English | MEDLINE | ID: mdl-28585208

ABSTRACT

Leptin signaling blockade by chronic overstimulation of the leptin receptor or hypothalamic pro-inflammatory responses due to elevated levels of saturated fatty acid can induce leptin resistance by activating negative feedback pathways. Although, long form leptin receptor (Ob-Rb) initiates leptin signaling through more than seven different signal transduction pathways, excessive suppressor of cytokine signaling-3 (SOCS-3) activity is a potential mechanism for the leptin resistance that characterizes human obesity. Because the leptin-responsive metabolic pathways broadly integrate with other neurons to control energy balance, the methods used to counteract the leptin resistance has extremely limited effect. In this chapter, besides the impairment of central and peripheral leptin signaling pathways, limited access of leptin to central nervous system (CNS) through blood-brain barrier, mismatch between high leptin and the amount of leptin receptor expression, contradictory effects of cellular and circulating molecules on leptin signaling, the connection between leptin signaling and endoplasmic reticulum (ER) stress and self-regulation of leptin signaling has been discussed in terms of leptin resistance.


Subject(s)
Diet/adverse effects , Leptin/metabolism , Obesity/physiopathology , Animals , Eating/physiology , Feeding Behavior/physiology , Humans , Hypothalamus/metabolism , Hypothalamus/physiopathology , Obesity/metabolism , Receptors, Leptin/metabolism , Signal Transduction/physiology
17.
J Exp Biol ; 220(Pt 14): 2563-2576, 2017 07 15.
Article in English | MEDLINE | ID: mdl-28495865

ABSTRACT

In mammals, ceramides are involved in the modulation of the orexigenic effects of ghrelin (GHRL). We previously demonstrated in rainbow trout that intracerebroventricular (ICV) treatment with ceramide (2.5 µg/100 g fish) resulted in an anorexigenic response, i.e. a response opposed to that described in mammals, where ceramide treatment is orexigenic. Therefore, we hypothesized that the putative interaction between GHRL and ceramide must be different in fish. Accordingly, in a first experiment, we observed that ceramide levels in the hypothalamus of rainbow trout did not change after ICV treatment with GHRL. In a second experiment, we assessed whether the effects of GHRL treatment on the regulation of food intake in rainbow trout changed in the presence of ceramide. Thus, we injected ICV GHRL and ceramide alone or in combination to evaluate in hypothalamus and hindbrain changes in parameters related to the metabolic control of food intake. The presence of ceramide generally counteracted the effects elicited by GHRL on fatty acid-sensing systems, the capacity of integrative sensors (AMPK, mTOR and SIRT-1), proteins involved in cellular signalling pathways (Akt and FoxO1) and neuropeptides involved in the regulation of food intake (AgRP, NPY, POMC and CART). The results are discussed in the context of regulation of food intake by metabolic and endocrine inputs.


Subject(s)
Ceramides/pharmacology , Eating/drug effects , Ghrelin/pharmacology , Oncorhynchus mykiss/physiology , Animals , Ceramides/analysis , Gene Expression , Hypothalamus/metabolism , Infusions, Intraventricular , RNA, Messenger , Rhombencephalon/metabolism
18.
Gen Comp Endocrinol ; 246: 116-119, 2017 05 15.
Article in English | MEDLINE | ID: mdl-27318277

ABSTRACT

LPLRFamide is a member of the RFamide peptide family that elicits an anorexigenic effect when centrally injected in chicks although the mechanism mediating this response is poorly understood. Therefore, the purpose of this experiment was to elucidate the hypothalamic mechanism of short-term anorexia after central administration of LPLRFamide in chicks. In Experiment 1 chicks centrally injected with LPLRFamide decreased food intake at 15min but not 30min following administration compared to vehicle-injected chicks. For Experiment 2, c-Fos immunoreactivity was quantified in several appetite-associated hypothalamic nuclei and in LPLRF-injected chicks, compared to vehicle-injected chicks, there was an increase in the number of reactive cells in the magnocellular division of the paraventricular nucleus. Lastly in Experiment 3, real time-PCR was performed and hypothalamic proopiomelanocortin (POMC) mRNA abundance was increased in LPRLFamide-injected chicks compared to vehicle-injected chicks. Thus, following central injection of LPLRFamide there is activation of the paraventricular nucleus of the hypothalamus and increased expression of hypothalamic POMC mRNA in chicks.


Subject(s)
Anorexia/metabolism , Eating/drug effects , Hypothalamus/metabolism , Oligopeptides/pharmacology , Paraventricular Hypothalamic Nucleus/drug effects , Pro-Opiomelanocortin/genetics , Animals , Anorexia/drug therapy , Chickens , Hypothalamus/drug effects , Immunoenzyme Techniques , Injections, Intraventricular , Oligopeptides/administration & dosage , Paraventricular Hypothalamic Nucleus/metabolism , Pro-Opiomelanocortin/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
19.
Mol Neurobiol ; 54(9): 7483-7496, 2017 11.
Article in English | MEDLINE | ID: mdl-27822713

ABSTRACT

Elevated levels of glucocorticoid, a steroid hormone released in response to stress, have been implicated in the pathophysiology of diabetes, which is now known to extend its effect on brain functions. Hence, we aimed to investigate the status of brain insulin signaling in response to dexamethasone (a synthetic glucocorticoid) treatment in female Charles Foster rat. This model exhibited pronounced hyperinsulinemia and glucose intolerance with loss in appetite and body weight. Immunoblotting of insulin receptor (INSR)-PI3kinase-AKT demonstrated reduced insulin signaling in hypothalamus but no change in hippocampus, cortex, and cerebellum in dexamethasone-treated rats as compared to vehicle-treated rats, signifying the diversity of distribution and function of insulin in different brain regions. These results also correlated with appetite change, a key function governed by hypothalamus. Hence, we further explored the hypothalamic feeding circuit and found altered levels of neuropeptide genes (Agrp, Npy, Pomc) and candidate nutrient sensors (GLUT1, SirT1, and PPARγ). There was also a considerable reduction in glycogen content and appetite-regulating neurotransmitters (GABA, glutamate, dopamine) in dexamethasone-treated rats. Thus, concluding that dexamethasone not only induces peripheral insulin resistance but also impairs hypothalamic function of appetite regulation via the interwoven cascade of insulin signaling, neurotransmitters, and neuropeptides. Graphical Abstract Reduced insulin signaling as well as elevated glucocorticoid levels in hypothalamus modulates the key appetite regulating neuropeptides, neurotransmitters, and nutrient sensors resulting into reduced appetite and bodyweight.


Subject(s)
Appetite Regulation/physiology , Dexamethasone/pharmacology , Feeding Behavior/physiology , Glucocorticoids/pharmacology , Hypothalamus/metabolism , Insulin Resistance/physiology , Animals , Appetite Regulation/drug effects , Body Weight/drug effects , Body Weight/physiology , Feeding Behavior/drug effects , Female , Hypothalamus/drug effects , Nerve Net/drug effects , Nerve Net/metabolism , Rats
20.
Neuroendocrinology ; 105(4): 372-383, 2017.
Article in English | MEDLINE | ID: mdl-28006784

ABSTRACT

Although the short-term effects of fasting or energy deficit on hypothalamic neuropeptide circuitries are now better understood, the effects of long-term energy deficit and refeeding remain to be elucidated. We showed that after a long-term energy deficit, mice exhibited persistent hypoleptinemia following the refeeding period despite restoration of fat mass, ovarian activity, and feeding behavior. We aimed to examine the hypothalamic adaptations after 10 weeks of energy deficit and after 10 further weeks of nutritional recovery. To do so, we assessed the mRNA levels of the leptin receptor and the main orexigenic and anorexigenic peptides, and their receptors regulated by leptin. Markers of hypothalamic inflammation were assessed as leptin can also participate in this phenomenon. Long-term time-restricted feeding and separation induced significant increase in mRNA levels of hypothalamic orexigenic peptides, while both Y1 and Y5 receptor mRNAs were downregulated. No changes occurred in the mRNA levels of orexin (OX), melanin-concentrating hormone, pro-opiomelanocortin, 26RFa (26-amino acid RF-amide peptide), and their receptors despite an increase in the expression of melanocortin receptors (MC3-R and MC4-R) and OXR1 (OX receptor 1). The refeeding period induced an overexpression of leptin receptor mRNA in the hypothalamus. The other assessed mRNA levels were normalized except for Y2, Y5, MC3-R, and MC4-R, which remained upregulated. No convincing changes were observed in neuroinflammatory markers, even if interleukin-1ß mRNA levels were increased in parallel with those of Iba1 (ionized calcium-binding adaptor molecule 1), a marker of microglial activation. Normalization of leptin-regulated functions and hypothalamic gene expressions in refed mice with low plasma leptin levels could be sustained by recalibration of hypothalamic sensitivity to leptin.


Subject(s)
Disease Models, Animal , Eating/physiology , Hypolipoproteinemias/pathology , Hypothalamus/metabolism , Leptin/metabolism , Agouti-Related Protein/metabolism , Animals , Body Weight/physiology , Cytokines/genetics , Cytokines/metabolism , Female , Hypolipoproteinemias/blood , Hypothalamic Hormones , Melanins , Mice , Mice, Inbred C57BL , Neuropeptide Y/metabolism , Neuropeptides/metabolism , Orexins/genetics , Orexins/metabolism , Pituitary Hormones , RNA, Messenger/metabolism , Receptors, Leptin/genetics , Receptors, Leptin/metabolism , Receptors, Neuropeptide/genetics , Receptors, Neuropeptide/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL