Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
J Endocrinol Invest ; 2024 Feb 09.
Article in English | MEDLINE | ID: mdl-38337094

ABSTRACT

PURPOSE: Liver-expressed antimicrobial peptide 2 (LEAP-2) has been recently identified as the endogenous non-competitive allosteric antagonist of the growth hormone secretagogue receptor 1a (GHSR1a). In rodents, LEAP-2 blunts ghrelin-induced feeding and its plasma levels are modulated in response to nutritional status, being decreased upon fasting and increased in high-fat diet (HFD) fed mice. Clinical data support the regulation of circulating LEAP-2 by nutrient availability in humans. In this work, our primary objective was to examine the chronic effects of ghrelin and LEAP-2 administration on food intake, adiposity, and energy expenditure in young mice subjected to standard and HFD at both room temperature and at thermoneutrality. Furthermore, we aimed to assess the impact of these two hormones on aging mice. RESULTS: Our results indicate that LEAP-2 produces a significant decrease of body weight and adiposity, an increase in energy expenditure, and activation of the thermogenic program in white and brown adipose tissue depots. However, this effect is not maintained under HFD or under thermoneutral conditions and is only partially observed in aging mice. CONCLUSION: In summary our studies describe the central effects of LEAP-2 within distinct experimental contexts, and contribute to the comprehension of LEAP-2's role in energy metabolism.

2.
J Endocrinol Invest ; 2023 Nov 13.
Article in English | MEDLINE | ID: mdl-37955834

ABSTRACT

AIM: Growth differentiation factor 15 (GDF15) is a stress response cytokine that has been proposed as a relevant metabolic hormone. Descriptive studies have shown that plasma GDF15 levels are regulated by short term changes in nutritional status, such as fasting, or in obesity. However, few data exist regarding how GDF15 levels are regulated in peripheral tissues. The aim of the present work was to study the variations on gastric levels of GDF15 and its precursor under different physiological conditions, such as short-term changes in nutritional status or overfeeding achieved by HFD. Moreover, we also address the sex- and age-dependent alterations in GDF15 physiology. METHODS: The levels of gastric and plasma GDF15 and its precursor were measured in lean and obese mice, rats and humans by western blot, RT-PCR, ELISA, immunohistochemistry and by an in vitro organ culture system. RESULTS: Our results show a robust regulation of gastric GDF15 production by fasting in rodents. In obesity an increase in GDF15 secretion from the stomach is reflected with an increase in circulating levels of GDF15 in rats and humans. Moreover, gastric GDF15 levels increase with age in both rats and humans. Finally, gastric GDF15 levels display sexual dimorphism, which could explain the difference in circulating GFD15 levels between males and females, observed in both humans and rodents. CONCLUSIONS: Our results provide clear evidence that gastric GDF15 is a critical contributor of circulating GDF15 levels and can explain some of the metabolic effects induced by GDF15.

3.
Nat Commun ; 9(1): 4194, 2018 10 10.
Article in English | MEDLINE | ID: mdl-30305620

ABSTRACT

Puberty is regulated by epigenetic mechanisms and is highly sensitive to metabolic and nutritional cues. However, the epigenetic pathways mediating the effects of nutrition and obesity on pubertal timing are unknown. Here, we identify Sirtuin 1 (SIRT1), a fuel-sensing deacetylase, as a molecule that restrains female puberty via epigenetic repression of the puberty-activating gene, Kiss1. SIRT1 is expressed in hypothalamic Kiss1 neurons and suppresses Kiss1 expression. SIRT1 interacts with the Polycomb silencing complex to decrease Kiss1 promoter activity. As puberty approaches, SIRT1 is evicted from the Kiss1 promoter facilitating a repressive-to-permissive switch in chromatin landscape. Early-onset overnutrition accelerates these changes, enhances Kiss1 expression and advances puberty. In contrast, undernutrition raises SIRT1 levels, protracts Kiss1 repression and delays puberty. This delay is mimicked by central pharmacological activation of SIRT1 or SIRT1 overexpression, achieved via transgenesis or virogenetic targeting to the ARC. Our results identify SIRT1-mediated inhibition of Kiss1 as key epigenetic mechanism by which nutritional cues and obesity influence mammalian puberty.


Subject(s)
Epigenesis, Genetic , Kisspeptins/genetics , Nutritional Physiological Phenomena , Obesity/metabolism , Sexual Maturation , Sirtuin 1/metabolism , Animals , Arcuate Nucleus of Hypothalamus/metabolism , Chromatin/metabolism , Female , Histones/metabolism , Hypothalamus/metabolism , Kisspeptins/metabolism , Mice, Transgenic , Models, Biological , Neurons/metabolism , Nutritional Status , Polycomb Repressive Complex 2/metabolism , Promoter Regions, Genetic , Rats , Rats, Wistar , Time Factors
4.
Acta Physiol (Oxf) ; 222(4): e13008, 2018 04.
Article in English | MEDLINE | ID: mdl-29193738

ABSTRACT

AIM: To explore the cooperation of GLP-1 receptor and ß3-adrenergic receptor (ß3-AR)-mediated signalling in the control of fat mass/feeding behaviour by studying the effects of a combined therapy composed of the GLP-1R agonist liraglutide and the ß3-AR agonist CL316243. METHODS: The study included the analysis of key mechanisms regulating lipid/cholesterol metabolism, and thermogenesis in brown (BAT) and epididymal white (eWAT) adipose tissues, abdominal muscle and liver of male rats. RESULTS: CL316243 (1 mg kg-1 ) and liraglutide (100 µg kg-1 ) co-administration over 6 days potentiated an overall negative energy balance (reduction in food intake, body weight gain, fat/non-fat mass ratio, liver fat content, and circulating levels of non-essential fatty acids, triglycerides, very low-density lipoprotein-cholesterol and leptin). These effects were accompanied by increased plasma levels of insulin and IL6. We also observed increased gene expression of uncoupling proteins regulating thermogenesis in BAT/eWAT (Ucp1) and muscle (Ucp2/3). Expression of transcription factor and enzymes involved either in de novo lipogenesis (Chrebp, Acaca, Fasn, Scd1, Insig1, Srebp1) or in fatty acid ß-oxidation (Cpt1b) was enhanced in eWAT and/or muscle but decreased in BAT. Pparα and Pparγ, essentials in lipid flux/storage, were decreased in BAT/eWAT but increased in the muscle and liver. Cholesterol synthesis regulators (Insig2, Srebp2, Hmgcr) were particularly over-expressed in muscle. These GLP-1R/ß3-AR-induced metabolic effects were associated with the downregulation of cAMP-dependent signalling pathways (PKA/AKT/AMPK). CONCLUSION: Combined activation of GLP-1 and ß3-ARs potentiate changes in peripheral pathways regulating lipid/cholesterol metabolism in a tissue-specific manner that favours a switch in energy availability/expenditure and may be useful for obesity treatment.


Subject(s)
Adipose Tissue/metabolism , Energy Metabolism/physiology , Glucagon-Like Peptide-1 Receptor/agonists , Muscle, Skeletal/metabolism , Receptors, Adrenergic, beta-3/metabolism , Signal Transduction/physiology , AMP-Activated Protein Kinases/metabolism , Adipose Tissue/drug effects , Adrenergic beta-3 Receptor Agonists/pharmacology , Animals , Body Composition/drug effects , Body Composition/physiology , Cyclic AMP-Dependent Protein Kinases/metabolism , Down-Regulation , Energy Metabolism/drug effects , Feeding Behavior/drug effects , Feeding Behavior/physiology , Lipid Metabolism/drug effects , Lipid Metabolism/physiology , Liraglutide/pharmacology , Male , Muscle, Skeletal/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Rats , Signal Transduction/drug effects
5.
J Mol Endocrinol ; 60(2): R31-R38, 2018 02.
Article in English | MEDLINE | ID: mdl-29203517

ABSTRACT

Uroguanylin (UGN) is a potential target in the fight against obesity. The mature protein is released after enzymatic cleavage from its natural precursor, proUGN. UGN is mostly produced in the gut, and its production is regulated by nutritional status. However, UGN is also produced in other tissues such as the kidneys. In the past, UGN has been widely studied as a natriuretic peptide owing to its involvement in several different pathologies such as heart failure, cancer and gastrointestinal diseases. However, recent studies have suggested that UGN also acts as a regulator of body weight homeostasis because it modulates both food intake and energy expenditure. This ultimately results in a decrease in body weight. This action is mediated by the sympathetic nervous system. Future studies should be directed at the potential effects of UGN agonists in regulating body weight in human obesity.


Subject(s)
Energy Metabolism , Natriuretic Peptides/metabolism , Animals , Energy Metabolism/drug effects , Homeostasis/drug effects , Humans , Intestines/drug effects , Models, Biological , Natriuretic Peptides/administration & dosage , Natriuretic Peptides/biosynthesis , Natriuretic Peptides/pharmacology
6.
Eur J Nutr ; 55(2): 529-536, 2016 Mar.
Article in English | MEDLINE | ID: mdl-25743370

ABSTRACT

PURPOSE: Uroguanylin (UGN) is a 16 amino acid peptide produced mainly by intestinal epithelial cells. Nutrients intake increases circulating levels of prouroguanylin that is processed and converted to UGN to activate the guanylyl cyclase 2C receptor (GUCY2C). Given that the UGN-GUCY2C system has been proposed as a novel gut-brain endocrine axis regulating energy balance, the aim of the present study was to investigate the regulation of UGN protein levels in duodenum and circulating levels in lean and obese mice under different nutritional conditions and its potential interaction with leptin. METHODS: Swiss, C57BL/6 wild-type and ob/ob male adult mice under different nutritional conditions were used: fed ad libitum standard diet (control); 48 h fasting (fasted); 48 h fasting followed by 24 h of feeding (refed); and fed high-fat diet (45 %) during 10 weeks. In addition, peripheral leptin administration was performed. Intestinal uroguanylin expression was studied by Western blot analysis; plasma levels were measured by ELISA. RESULTS: Food deprivation significantly reduced plasma UGN levels, which were correlated with the lower protein levels of UGN in duodenum. These effects were reverted after refeeding and leptin challenge. Consistently, in ob/ob mice UGN expression was decreased, whereas leptin treatment up-regulated UGN levels in duodenum in these genetically modified mice compared to WT. Diet-induced obese mice displayed increased UGN levels in intestine and plasma in comparison with lean mice. CONCLUSIONS: Our findings suggest that UGN levels are correlated with energy balance status and that the regulation of UGN by nutritional status is leptin-dependent.


Subject(s)
Intestinal Mucosa/metabolism , Leptin/pharmacology , Natriuretic Peptides/blood , Nutritional Status , Animals , Diet, High-Fat , Energy Metabolism , Leptin/blood , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Natriuretic Peptides/genetics , Up-Regulation
7.
Mol Metab ; 4(6): 437-60, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26042199

ABSTRACT

BACKGROUND: The gastrointestinal peptide hormone ghrelin was discovered in 1999 as the endogenous ligand of the growth hormone secretagogue receptor. Increasing evidence supports more complicated and nuanced roles for the hormone, which go beyond the regulation of systemic energy metabolism. SCOPE OF REVIEW: In this review, we discuss the diverse biological functions of ghrelin, the regulation of its secretion, and address questions that still remain 15 years after its discovery. MAJOR CONCLUSIONS: In recent years, ghrelin has been found to have a plethora of central and peripheral actions in distinct areas including learning and memory, gut motility and gastric acid secretion, sleep/wake rhythm, reward seeking behavior, taste sensation and glucose metabolism.

8.
Diabetes Obes Metab ; 17(8): 789-99, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25962313

ABSTRACT

AIM: To investigate the role of brain glucagon-like peptide-1 (GLP-1) in pancreatic ß-cell function. METHODS: To determine the role of brain GLP-1 receptor (GLP-1R) on ß-cell function, we administered intracerebroventricular (i.c.v.) infusions of GLP-1 or the specific GLP-1 antagonist exendin-9 (Ex-9), in both an acute and a chronic setting. RESULTS: We observed that acute i.c.v. GLP-1 infusion potentiates glucose-stimulated insulin secretion (GSIS) and improves glucose tolerance, whereas central GLP-1R blockade with Ex-9 impaired glucose excursion after a glucose load. Sustained activation of central nervous system GLP-1R, however, did not produce any effect on either GSIS or glucose tolerance. Similarly, ex vivo GSIS performed in islets from mice chronically infused with i.c.v. GLP-1 resulted in no differences compared with controls. In addition, in mice fed a high-fat diet we observed that acute i.c.v. GLP-1 infusion improved glucose tolerance without changes in GSIS, while chronic GLP-1R activation had no effect on glucose homeostasis. CONCLUSIONS: Our results indicate that, under non-clamped conditions, brain GLP-1 plays a functional neuroendocrine role in the acute regulation of glucose homeostasis in both lean and obese rodents.


Subject(s)
Brain/metabolism , Glucagon-Like Peptide 1/pharmacology , Glucagon-Like Peptide-1 Receptor/metabolism , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Animals , Blood Glucose/analysis , Blood Glucose/metabolism , Diet, High-Fat , Glucagon-Like Peptide 1/administration & dosage , Glucagon-Like Peptide-1 Receptor/antagonists & inhibitors , Glucose/administration & dosage , Homeostasis/drug effects , Incretins/administration & dosage , Incretins/pharmacology , Infusions, Intraventricular , Insulin Secretion , Insulin-Secreting Cells/drug effects , Male , Mice , Mice, Inbred C57BL
9.
Sci Rep ; 4: 4264, 2014 Mar 03.
Article in English | MEDLINE | ID: mdl-24584059

ABSTRACT

The NR4A is a subfamily of the orphan nuclear receptors (NR) superfamily constituted by three well characterized members: Nur77 (NR4A1), Nurr1 (NR4A2) and Nor 1 (NR4A3). They are implicated in numerous biological processes as DNA repair, arteriosclerosis, cell apoptosis, carcinogenesis and metabolism. Several studies have demonstrated the role of this subfamily on glucose metabolism, insulin sensitivity and energy balance. These studies have focused mainly in liver and skeletal muscle. However, its potential role in white adipose tissue (WAT), one of the most important tissues involved in the regulation of energy homeostasis, is not well-studied. The aim of this work was to elucidate the regulation of NR4A in WAT under different physiological and pathophysiological settings involved in energy balance such as fasting, postnatal development, gender, hormonal deficiency and pregnancy. We compared NR4A mRNA expression of Nur77, Nurr1 and Nor 1 and found a clear regulation by nutritional status, since the expression of the 3 isoforms is increased after fasting in a leptin-independent manner and sex steroid hormones also modulate NR4A expression in males and females. Our findings indicate that NR4A are regulated by different physiological and pathophysiological settings known to be associated with marked alterations in glucose metabolism and energy status.


Subject(s)
Adipose Tissue, White/metabolism , DNA-Binding Proteins/genetics , Gene Expression Regulation , Nerve Tissue Proteins/genetics , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Adipose Tissue, White/pathology , Animals , Caloric Restriction , DNA-Binding Proteins/metabolism , Energy Metabolism , Fasting , Female , Glucose/metabolism , Gonadal Steroid Hormones/genetics , Gonadal Steroid Hormones/metabolism , Growth Hormone/deficiency , Growth Hormone/genetics , Hypothyroidism/metabolism , Hypothyroidism/pathology , Injections, Intraventricular , Leptin/administration & dosage , Male , Mice, Inbred C57BL , Nerve Tissue Proteins/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Orchiectomy , Ovariectomy , Rats , Rats, Sprague-Dawley , Sex Characteristics , Signal Transduction
10.
Curr Mol Med ; 14(1): 3-21, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24236459

ABSTRACT

Optimal cellular function and therefore organism's survival is determined by the sensitive and accurate convergence of energy and nutrient abundance to cell growth and division. Among other factors, this integration is coupled by the target of rapamycin (TOR) pathway, which is able to sense nutrient, energy and oxygen availability and also growth factor signaling. Indeed, TOR signaling regulates cell energy homeostasis by coordinating anabolic and catabolic processes for survival. TOR, named mTOR in mammals, is a conserved serine/threonine kinase that exists in two different complexes, mTORC1 and mTORC2. Recently, studies are suggesting that alterations of those complexes promote disease and disrupted phenotypes, such as aging, obesity and related disorders and even cancer. The evidences linking mTOR to energy and metabolic homeostasis included the following. At central level mTOR regulates food intake and body weight being involved in the mechanism by which signals such as leptin and ghrelin exert its effects. At peripheral level it influences adipogenesis and lipogenesis in different tissues including the liver. Noteworthy chronic nutritional activation of mTOR signaling has been implicated in the development of beta cell mass expansion and on insulin resistance. Understanding of mTOR and other molecular switches, such as AMP-activated protein kinase (AMPK), as well as their interrelationship is crucial to know how organisms maintain optimal homeostasis. This review summarizes the role of hypothalamic TOR complex in cellular energy sensing, evidenced in the last years, focusing on the metabolic pathways where it is involved and the importance of this metabolic sensor in cellular and whole body energy management. Understanding the exact role of hypothalamic mTOR may provide new cues for therapeutic intervention in diseases.


Subject(s)
Hypothalamus/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Energy Metabolism , Hormones/metabolism , Hormones/pharmacology , Humans , Hypothalamus/drug effects , Mechanistic Target of Rapamycin Complex 1 , Mechanistic Target of Rapamycin Complex 2 , Models, Biological , Multiprotein Complexes/metabolism , Puberty/genetics , Puberty/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/genetics
11.
Horm Metab Res ; 45(13): 960-6, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23950036

ABSTRACT

The sirtuins are a family of highly conserved nicotine adenine dinucleotide (NAD+)-dependent deacetylases that act as cellular sensors to detect energy availability and regulate metabolic processes. Sirtuin 1 (SIRT1) is one of the family members that is activated in response to caloric restriction, acting on multiple targets in a wide range of tissues. Recent studies have shown that SIRT1 controls glucose and lipid metabolism in both liver and muscle, promotes fat mobilization, stimulates remodeling of white to brown fat, controls insulin secretion in the pancreas, and senses nutrient availability in the hypothalamus. SIRT1 is located in several areas of the brain and its central metabolic actions have attracted much attention in the last decade. In this short review, we summarize the main actions and molecular pathways triggered by SIRT1 that control feeding behavior, energy expenditure, glucose metabolism, and insulin sensitivity, with an emphasis on the emerging role of SIRT1 in the brain.


Subject(s)
Energy Metabolism/physiology , Feeding Behavior/physiology , Glucose/metabolism , Hypothalamus/metabolism , Nerve Tissue Proteins/metabolism , Sirtuin 1/metabolism , Animals , Humans
12.
Placenta ; 33(5): 373-8, 2012 May.
Article in English | MEDLINE | ID: mdl-22369992

ABSTRACT

BACKGROUND: Chemerin is an adipocytokine that is expressed in different fat deposits and has been shown to play an important role in adaptive and innate immunity due to its activity as a chemoattractant. Chemerin acts as a ligand for the G protein-coupled receptor chemokine-like receptor 1 (CMKLR1). Chemerin has been shown to regulate the development and metabolic function of adipocytes, liver and muscle tissue. OBJECTIVE: There is evidence indicating that several adipocytokines play an important role in placenta. This study aimed to investigate the regulation of chemerin in rat and human placentas throughout gestation. DESIGN AND SETTING: Chemerin was examined in rat and human placentas using immunohistochemistry. The chemerin expression pattern in the placenta and adipose tissue of female Sprague Dawley rats on days 12, 16, 19 and 21 of gestation (each of these days represents a group of 12 rats) was determined using TaqMan probe-based quantitative real-time PCR. Rat chemerin serum levels were analyzed with ELISA on days 8, 12, 16, 19 and 21 and compared to virgin controls. RESULTS: Chemerin expression was detected in the cytoplasm of rat placental trophoblastic cells and third trimester human placental cytotrophoblast and Hofbauer's cells. The serum chemerin levels of rats decreased significantly as gestation progressed. Furthermore, placental chemerin mRNA levels rose significantly at day 16 of gestation and decreased significantly towards the end of the gestation period. CONCLUSION: Taken together, this data suggests that chemerin may be an important regulator of maternal-fetal metabolism and metabolic homeostasis during pregnancy.


Subject(s)
Adipokines/metabolism , Chemokines/metabolism , Placenta/metabolism , Pregnancy, Animal/metabolism , Adipose Tissue, White/metabolism , Animals , Female , Food Deprivation , Humans , Intercellular Signaling Peptides and Proteins , Nutritional Status , Pregnancy , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley
13.
Diabetes Obes Metab ; 14(3): 234-43, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21981246

ABSTRACT

AIMS: Cannabinoids are known to control energy homeostasis. Atypical cannabinoids produce pharmacological effects via unidentified targets. We sought to investigate whether the atypical cannabinoid O-1602 controls food intake and body weight. METHODS: The rats were injected acutely or subchronically with O-1602, and the expression of several factors involved in adipocyte metabolism was assessed by real-time polymerase chain reaction. In vivo findings were corroborated with in vitro studies incubating 3T3-L1 adipocytes with O-1602, and measuring intracellular calcium and lipid accumulation. Finally, as some reports suggest that O-1602 is an agonist of the putative cannabinoid receptor GPR55, we tested it in mice lacking GPR55. RESULTS: Central and peripheral administration of O-1602 acutely stimulates food intake, and chronically increases adiposity. The hyperphagic action of O-1602 is mediated by the downregulation of mRNA and protein levels of the anorexigenic neuropeptide cocaine- and amphetamine-regulated transcript. The effects on fat mass are independent of food intake, and involve a decrease in the expression of lipolytic enzymes such as hormone sensitive lipase and adipose triglyceride lipase in white adipose tissue. Consistently, in vitro data showed that O-1602 increased the levels of intracellular calcium and lipid accumulation in adipocytes. Finally, we injected O-1602 in GPR55 -/- mice and found that O-1602 was able to induce feeding behaviour in GPR55-deficient mice. CONCLUSIONS: These findings show that O-1602 modulates food intake and adiposity independently of GPR55 receptor. Thus atypical cannabinoids may represent a novel class of molecules involved in energy balance.


Subject(s)
Adiposity/drug effects , Cannabinoid Receptor Agonists , Cannabinoids/pharmacology , Cyclohexanes/pharmacology , Eating/drug effects , Resorcinols/pharmacology , Adipocytes/metabolism , Animals , Body Weight , Cannabidiol/analogs & derivatives , Energy Metabolism , Male , Mice , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Receptors, Cannabinoid/deficiency
14.
Diabetologia ; 54(4): 900-9, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21181395

ABSTRACT

AIMS/HYPOTHESIS: Obesity and type 2 diabetes are among the most serious health pathologies worldwide. Stress has been proposed as a factor contributing to the development of these health risk factors; however, the underlying mechanisms that link stress to obesity and diabetes need to be further clarified. Here, we study in mice how chronic stress affects dietary consumption and how that relationship contributes to obesity and diabetes. METHODS: C57BL/6J mice were subjected to chronic variable stress (CVS) for 15 days and subsequently fed with a standard chow or high-fat diet. Food intake, body weight, respiratory quotient, energy expenditure and spontaneous physical activity were measured with a customised calorimetric system and body composition was measured with nuclear magnetic resonance. A glucose tolerance test was also applied and blood glucose levels were measured with a glucometer. Plasma levels of adiponectin and resistin were measured using Lincoplex kits. RESULTS: Mice under CVS and fed with a high-fat diet showed impaired glucose tolerance associated with low plasma adiponectin:resistin ratios. CONCLUSIONS/INTERPRETATION: This study demonstrates, in a novel mouse model, how post-traumatic stress disorder enhances vulnerability for impaired glucose metabolism in an energy-rich environment and proposes a potential adipokine-based mechanism.


Subject(s)
Stress, Physiological/physiology , Adiponectin/blood , Animals , Body Composition/physiology , Disease Models, Animal , Energy Metabolism/physiology , Glucose Tolerance Test , Male , Mice , Mice, Inbred C57BL , Resistin/blood , Stress, Physiological/genetics
15.
Obes Rev ; 11(3): 185-201, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19845870

ABSTRACT

The central nervous system (CNS) is crucial in the regulation of energy homeostasis. Many neuroanatomical studies have shown that the white adipose tissue (WAT) is innervated by the sympathetic nervous system, which plays a critical role in adipocyte lipid metabolism. Therefore, there are currently numerous reports indicating that signals from the CNS control the amount of fat by modulating the storage or oxidation of fatty acids. Importantly, some CNS pathways regulate adipocyte metabolism independently of food intake, suggesting that some signals possess alternative mechanisms to regulate energy homeostasis. In this review, we mainly focus on how neuronal circuits within the hypothalamus, such as leptin- ghrelin-and resistin-responsive neurons, as well as melanocortins, neuropeptide Y, and the cannabinoid system exert their actions on lipid metabolism in peripheral tissues such as WAT, liver or muscle. Dissecting the complicated interactions between peripheral signals and neuronal circuits regulating lipid metabolism might open new avenues for the development of new therapies preventing and treating obesity and its associated cardiometabolic sequelae.


Subject(s)
Adipocytes/metabolism , Energy Metabolism/physiology , Hypothalamus/metabolism , Lipid Metabolism/physiology , Peptide Hormones/physiology , Adipose Tissue, White/metabolism , Homeostasis , Humans , Neuropeptide Y/metabolism
16.
Acta Physiol (Oxf) ; 198(3): 325-34, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19769635

ABSTRACT

The secretion of growth hormone (GH) is regulated through a complex neuroendocrine control system that includes two major hypothalamic regulators, namely GH-releasing hormone (GHRH) and somatostatin (SST) that stimulate and inhibit, respectively, GH release. Classical experiments involving damage and electrical stimulation suggested that the lateral hypothalamic area (LHA) modulated the somatotropic axis, but the responsible molecular mechanisms were unclear. Evidence obtained during the last decade has demonstrated that orexins/hypocretins, a family of peptides expressed in the LHA controlling feeding and sleep, play an important regulatory role on GH, by inhibiting its secretion modulating GHRH and SST neurones. Considering that GH release is closely linked to the sleep-wake cycle and feeding state, understanding orexin/hypocretin physiology could open new therapeutic possibilities in the treatment of sleep, energy homeostasis and GH-related pathologies, such as GH deficiency.


Subject(s)
Growth Hormone-Releasing Hormone/metabolism , Growth Hormone/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Neuropeptides/metabolism , Neurotransmitter Agents/metabolism , Somatostatin/metabolism , Animals , Body Weight/physiology , Eating/physiology , Energy Metabolism/physiology , Homeostasis/physiology , Humans , Hypothalamus/metabolism , Neurons/metabolism , Neurosecretory Systems/metabolism , Orexins , Pituitary Gland/metabolism , Sleep/physiology , Wakefulness/physiology
17.
Diabetologia ; 52(10): 2159-68, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19644669

ABSTRACT

AIMS/HYPOTHESIS: High- vs low-glycaemic index (GI) diets unfavourably affect body fat mass and metabolic markers in rodents. Different effects of these diets could be age-dependent, as well as mediated, in part, by carbohydrate-induced stimulation of glucose-dependent insulinotrophic polypeptide (GIP) signalling. METHODS: Young-adult (16 weeks) and aged (44 weeks) male wild-type (C57BL/6J) and GIP-receptor knockout (Gipr ( -/- )) mice were exposed to otherwise identical high-carbohydrate diets differing only in GI (20-26 weeks of intervention, n = 8-10 per group). Diet-induced changes in body fat distribution, liver fat, locomotor activity, markers of insulin sensitivity and substrate oxidation were investigated, as well as changes in the gene expression of anorexigenic and orexigenic hypothalamic factors related to food intake. RESULTS: Body weight significantly increased in young-adult high- vs low-GI fed mice (two-way ANOVA, p < 0.001), regardless of the Gipr genotype. The high-GI diet in young-adult mice also led to significantly increased fat mass and changes in metabolic markers that indicate reduced insulin sensitivity. Even though body fat mass also slightly increased in high- vs low-GI fed aged wild-type mice (p < 0.05), there were no significant changes in body weight and estimated insulin sensitivity in these animals. However, aged Gipr ( -/- ) vs wild-type mice on high-GI diet showed significantly lower cumulative net energy intake, increased locomotor activity and improved markers of insulin sensitivity. CONCLUSIONS/INTERPRETATION: The metabolic benefits of a low-GI diet appear to be more pronounced in younger animals, regardless of the Gipr genotype. Inactivation of GIP signalling in aged animals on a high-GI diet, however, could be beneficial.


Subject(s)
Diet , Gastric Inhibitory Polypeptide/physiology , Glycemic Index , Age Factors , Animals , Blood Glucose/analysis , Body Composition , Calorimetry , Energy Intake/physiology , Glucose Tolerance Test , Insulin/blood , Male , Mice , Mice, Knockout , Receptors, Gastrointestinal Hormone/genetics , Receptors, Gastrointestinal Hormone/physiology , Reverse Transcriptase Polymerase Chain Reaction , Triglycerides/metabolism
18.
J Physiol ; 587(Pt 14): 3741-50, 2009 Jul 15.
Article in English | MEDLINE | ID: mdl-19470778

ABSTRACT

Visceral adipose tissue-derived serine protease inhibitor (vaspin) is a recently discovered adipocytokine mainly secreted from visceral adipose tissue, which plays a main role in insulin sensitivity. In this study, we have investigated the regulation of vaspin gene expression in rat white adipose tissue (WAT) in different physiological (nutritional status, pregnancy, age and gender) and pathophysiological (gonadectomy, thyroid status and growth hormone deficiency) settings known to be associated with energy homeostasis and alterations in insulin sensitivity. We have determined vaspin gene expression by real-time PCR. Vaspin was decreased after fasting and its levels were partially recovered after leptin treatment. Chronic treatment with metformin increased vaspin gene expression. Vaspin mRNA expression reached the highest peak at 45 days in both sexes after birth and its expression was higher in females than males, but its levels did not change throughout pregnancy. Finally, decreased levels of growth hormone and thyroid hormones suppressed vaspin expression. These findings suggest that WAT vaspin mRNA expression is regulated by nutritional status, and leptin seems to be the nutrient signal responsible for those changes. Vaspin is influenced by age and gender, and its expression is increased after treatment with insulin sensitizers. Finally, alterations in pituitary functions modify vaspin levels. Understanding the molecular mechanisms regulating vaspin will provide new insights into the pathogenesis of the metabolic syndrome.


Subject(s)
Aging/metabolism , Gene Expression Regulation/physiology , Intra-Abdominal Fat/enzymology , Metformin/metabolism , Nutritional Status , Pregnancy, Animal/metabolism , Serine Proteinase Inhibitors/metabolism , Animals , Female , Pregnancy , Rats , Rats, Sprague-Dawley , Sex Factors
19.
J Neuroendocrinol ; 20 Suppl 1: 147-51, 2008 May.
Article in English | MEDLINE | ID: mdl-18426514

ABSTRACT

Blockade of the CB(1) receptor is one of the promising strategies for the treatment of obesity. The first selective CB(1) receptor antagonist, rimonabant, which has already successfully completed phase III clinical trials, led to sustained weight loss and a reduction in waist circumference. Patients treated with rimonabant also demonstrated statistically significant improvement in high-density lipoprotein cholesterol levels, triglyceride levels and insulin resistance, as well as a reduced overall prevalence of metabolic syndrome. Currently, one of the most discussed aspects of endocannabinoid system function is to what extent the endocannabinoid system might affect metabolism independently of its control over body weight and food intake. Specifically, a food-intake- and body-weight-independent role in the regulation of glucose homeostasis and insulin sensitivity could have major impact on the potential of drug candidates targeting the endocannabinoid system for the prevention and treatment of metabolic syndrome. This review summarises the effects of the endocannabinoid system on glucose homeostasis and insulin sensitivity.


Subject(s)
Cannabinoid Receptor Modulators/physiology , Endocannabinoids , Glucose/metabolism , Receptors, Cannabinoid/physiology , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Animals , Cannabinoid Receptor Antagonists , Cannabinoid Receptor Modulators/antagonists & inhibitors , Cannabinoid Receptor Modulators/metabolism , Diabetes Mellitus, Type 2/metabolism , Energy Metabolism/drug effects , Homeostasis/drug effects , Homeostasis/physiology , Humans , Liver/drug effects , Liver/metabolism , Models, Biological , Muscles/drug effects , Muscles/metabolism , Obesity/metabolism , Pancreas/drug effects , Pancreas/metabolism , Piperidines/pharmacology , Pyrazoles/pharmacology , Receptors, Cannabinoid/metabolism , Rimonabant , Rodentia
20.
Endocrinology ; 149(7): 3390-402, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18403483

ABSTRACT

Adiponectin is an adipocyte hormone, with relevant roles in lipid metabolism and glucose homeostasis, recently involved in the control of different endocrine organs, such as the placenta, pituitary and, likely, the ovary. However, whether as described previously for other adipokines, such as leptin and resistin, adiponectin is expressed and/or conducts biological actions in the male gonad remains unexplored. In this study, we provide compelling evidence for the expression, putative hormonal regulation, and direct effects of adiponectin in the rat testis. Testicular expression of adiponectin was demonstrated along postnatal development, with a distinctive pattern of RNA transcripts and discernible protein levels that appeared mostly located at interstitial Leydig cells. Testicular levels of adiponectin mRNA were marginally regulated by pituitary gonadotropins but overtly modulated by metabolic signals, such as glucocorticoids, thyroxine, and peroxisome proliferator-activated receptor-gamma, whose effects were partially different from those on circulating levels of adiponectin. In addition, expression of the genes encoding adiponectin receptor (AdipoR)-1 and AdipoR2 was detected in the rat testis, with developmental changes and gonadotropin regulation for AdipoR2 mRNA, and prominent levels of AdipoR1 in seminiferous tubules. Moreover, recombinant adiponectin significantly inhibited basal and human choriogonadotropin-stimulated testosterone secretion ex vivo, whereas it failed to change relative levels of several Sertoli cell-expressed mRNAs, such as stem cell factor and anti-Müllerian hormone. In summary, our data are the first to document the expression, regulation and functional role of adiponectin in the rat testis. Taken together with its recently reported expression in the ovary and its effects on LH secretion and ovarian steroidogenesis, these results further substantiate a multifaceted role of adiponectin in the control of the reproductive axis, which might operate as endocrine integrator linking metabolism and gonadal function.


Subject(s)
Adiponectin/pharmacology , Leydig Cells/drug effects , Testis/drug effects , Adiponectin/genetics , Adiponectin/metabolism , Animals , Blotting, Western , Follicle Stimulating Hormone/pharmacology , Gene Expression/drug effects , Gonadotropins/pharmacology , Immunohistochemistry , Leydig Cells/metabolism , Male , Radioimmunoassay , Rats , Rats, Sprague-Dawley , Receptors, Adiponectin/genetics , Receptors, Adiponectin/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Rosiglitazone , Testis/metabolism , Thiazolidinediones/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...