Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Cell ; 155(1): 172-87, 2013 Sep 26.
Article in English | MEDLINE | ID: mdl-24074867

ABSTRACT

Mitofusin 2 (MFN2) plays critical roles in both mitochondrial fusion and the establishment of mitochondria-endoplasmic reticulum (ER) interactions. Hypothalamic ER stress has emerged as a causative factor for the development of leptin resistance, but the underlying mechanisms are largely unknown. Here, we show that mitochondria-ER contacts in anorexigenic pro-opiomelanocortin (POMC) neurons in the hypothalamus are decreased in diet-induced obesity. POMC-specific ablation of Mfn2 resulted in loss of mitochondria-ER contacts, defective POMC processing, ER stress-induced leptin resistance, hyperphagia, reduced energy expenditure, and obesity. Pharmacological relieve of hypothalamic ER stress reversed these metabolic alterations. Our data establish MFN2 in POMC neurons as an essential regulator of systemic energy balance by fine-tuning the mitochondrial-ER axis homeostasis and function. This previously unrecognized role for MFN2 argues for a crucial involvement in mediating ER stress-induced leptin resistance.


Subject(s)
Endoplasmic Reticulum Stress , GTP Phosphohydrolases/metabolism , Neurons/metabolism , Obesity/metabolism , Animals , Hypothalamus/metabolism , Leptin/metabolism , Mice , Mice, Inbred C57BL , Neurons/cytology , Pro-Opiomelanocortin/metabolism
2.
Hepatology ; 64(4): 1086-104, 2016 10.
Article in English | MEDLINE | ID: mdl-27387967

ABSTRACT

UNLABELLED: The opioid system is widely known to modulate the brain reward system and thus affect the behavior of humans and other animals, including feeding. We hypothesized that the hypothalamic opioid system might also control energy metabolism in peripheral tissues. Mice lacking the kappa opioid receptor (κOR) and adenoviral vectors overexpressing or silencing κOR were stereotaxically delivered in the lateral hypothalamic area (LHA) of rats. Vagal denervation was performed to assess its effect on liver metabolism. Endoplasmic reticulum (ER) stress was inhibited by pharmacological (tauroursodeoxycholic acid) and genetic (overexpression of the chaperone glucose-regulated protein 78 kDa) approaches. The peripheral effects on lipid metabolism were assessed by histological techniques and western blot. We show that in the LHA κOR directly controls hepatic lipid metabolism through the parasympathetic nervous system, independent of changes in food intake and body weight. κOR colocalizes with melanin concentrating hormone receptor 1 (MCH-R1) in the LHA, and genetic disruption of κOR reduced melanin concentrating hormone-induced liver steatosis. The functional relevance of these findings was given by the fact that silencing of κOR in the LHA attenuated both methionine choline-deficient, diet-induced and choline-deficient, high-fat diet-induced ER stress, inflammation, steatohepatitis, and fibrosis, whereas overexpression of κOR in this area promoted liver steatosis. Overexpression of glucose-regulated protein 78 kDa in the liver abolished hypothalamic κOR-induced steatosis by reducing hepatic ER stress. CONCLUSIONS: This study reveals a novel hypothalamic-parasympathetic circuit modulating hepatic function through inflammation and ER stress independent of changes in food intake or body weight; these findings might have implications for the clinical use of opioid receptor antagonists. (Hepatology 2016;64:1086-1104).


Subject(s)
Diet , Endoplasmic Reticulum Stress , Hypothalamic Hormones/physiology , Hypothalamus/physiology , Liver Diseases/etiology , Melanins/physiology , Pituitary Hormones/physiology , Receptors, Opioid, kappa/physiology , Animals , Inflammation/complications , Inflammation/etiology , Mice , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley
3.
Gastroenterology ; 144(3): 636-649.e6, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23142626

ABSTRACT

BACKGROUND & AIMS: Specific neuronal circuits modulate autonomic outflow to liver and white adipose tissue. Melanin-concentrating hormone (MCH)-deficient mice are hypophagic, lean, and do not develop hepatosteatosis when fed a high-fat diet. Herein, we sought to investigate the role of MCH, an orexigenic neuropeptide specifically expressed in the lateral hypothalamic area, on hepatic and adipocyte metabolism. METHODS: Chronic central administration of MCH and adenoviral vectors increasing MCH signaling were performed in rats and mice. Vagal denervation was performed to assess its effect on liver metabolism. The peripheral effects on lipid metabolism were assessed by real-time polymerase chain reaction and Western blot. RESULTS: We showed that the activation of MCH receptors promotes nonalcoholic fatty liver disease through the parasympathetic nervous system, whereas it increases fat deposition in white adipose tissue via the suppression of sympathetic traffic. These metabolic actions are independent of parallel changes in food intake and energy expenditure. In the liver, MCH triggers lipid accumulation and lipid uptake, with c-Jun N-terminal kinase being an essential player, whereas in adipocytes MCH induces metabolic pathways that promote lipid storage and decreases lipid mobilization. Genetic activation of MCH receptors or infusion of MCH specifically in the lateral hypothalamic area modulated hepatic lipid metabolism, whereas the specific activation of this receptor in the arcuate nucleus affected adipocyte metabolism. CONCLUSIONS: Our findings show that central MCH directly controls hepatic and adipocyte metabolism through different pathways.


Subject(s)
Adipocytes/metabolism , Adipose Tissue/metabolism , Adiposity/physiology , Hypothalamic Area, Lateral/physiology , Hypothalamic Hormones/physiology , Liver/metabolism , Melanins/physiology , Mitogen-Activated Protein Kinase 8/metabolism , Pituitary Hormones/physiology , Adipocytes/drug effects , Adipose Tissue/drug effects , Animals , Eating , Fatty Acids/metabolism , Fatty Liver/metabolism , Fatty Liver/physiopathology , Hypothalamic Area, Lateral/drug effects , Hypothalamic Hormones/administration & dosage , Lipid Metabolism/drug effects , Lipid Metabolism/physiology , Lipogenesis/drug effects , Lipogenesis/physiology , Liver/drug effects , Male , Melanins/administration & dosage , Mice , Non-alcoholic Fatty Liver Disease , Pituitary Hormones/administration & dosage , Rats , Rats, Sprague-Dawley , Receptors, Pituitary Hormone/agonists , Receptors, Pituitary Hormone/physiology , Vagus Nerve/drug effects , Vagus Nerve/physiology , Vagus Nerve/physiopathology
4.
Mol Metab ; 87: 101996, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39047908

ABSTRACT

OBJECTIVES: In Western society, high-caloric diets rich in fats and sugars have fueled the obesity epidemic and its related disorders. Disruption of the body-brain communication, crucial for maintaining glucose and energy homeostasis, arises from both obesogenic and genetic factors, leading to metabolic disorders. Here, we investigate the role of hypothalamic tanycyte shuttles between the pituitary portal blood and the third ventricle cerebrospinal fluid in regulating energy balance. METHODS: We inhibited vesicle-associated membrane proteins (VAMP1-3)-mediated release in tanycytes by expressing the botulinum neurotoxin type B light chain (BoNT/B) in a Cre-dependent manner in tanycytes. This was achieved by injecting either TAT-Cre in the third ventricle or an AAV1/2 expressing Cre under the control of the tanycyte-specific promoter iodothyronine deiodinase 2 into the lateral ventricle of adult male mice. RESULTS: In male mice fed a standard diet, targeted expression of BoNT/B in adult tanycytes blocks leptin transport into the mediobasal hypothalamus and results in normal-weight central obesity, including increased food intake, abdominal fat deposition, and elevated leptin levels but no marked change in body weight. Furthermore, BoNT/B expression in adult tanycytes promotes fatty acid storage, leading to glucose intolerance and insulin resistance. Notably, these metabolic disturbances occur despite a compensatory increase in insulin secretion, observed both in response to exogenous glucose boluses in vivo and in isolated pancreatic islets. Intriguingly, these metabolic alterations are associated with impaired spatial memory in BoNT/B-expressing mice. CONCLUSIONS: These findings underscore the central role of tanycytes in brain-periphery communication and highlight their potential implication in the age-related development of type 2 diabetes and cognitive decline. Our tanycytic BoNT/B mouse model provides a robust platform for studying how these conditions progress over time, from prediabetic states to full-blown metabolic and cognitive disorders, and the mechanistic contribution of tanycytes to their development. The recognition of the impact of tanycytic transcytosis on hormone transport opens new avenues for developing targeted therapies that could address both metabolic disorders and their associated cognitive comorbidities, which often emerge or worsen with advancing age.


Subject(s)
Energy Metabolism , Ependymoglial Cells , Glucose , Homeostasis , Animals , Male , Mice , Glucose/metabolism , Ependymoglial Cells/metabolism , Cognition/drug effects , Leptin/metabolism , Mice, Inbred C57BL , Hypothalamus/metabolism , Obesity/metabolism
5.
Curr Biol ; 32(4): R173-R176, 2022 02 28.
Article in English | MEDLINE | ID: mdl-35231413

ABSTRACT

The suprachiasmatic nucleus (SCN) synchronizes physiology with the individual's environment to optimize bodily functions. A new study reveals that tanycytes follow the tempo set by the SCN to effect circadian changes in both brain entry of blood glucose and glycemia.


Subject(s)
Circadian Rhythm , Ependymoglial Cells , Blood Glucose , Circadian Rhythm/physiology , Suprachiasmatic Nucleus/physiology
6.
Cell Metab ; 34(7): 1054-1063.e7, 2022 07 05.
Article in English | MEDLINE | ID: mdl-35716660

ABSTRACT

Liraglutide, an anti-diabetic drug and agonist of the glucagon-like peptide one receptor (GLP1R), has recently been approved to treat obesity in individuals with or without type 2 diabetes. Despite its extensive metabolic benefits, the mechanism and site of action of liraglutide remain unclear. Here, we demonstrate that liraglutide is shuttled to target cells in the mouse hypothalamus by specialized ependymoglial cells called tanycytes, bypassing the blood-brain barrier. Selectively silencing GLP1R in tanycytes or inhibiting tanycytic transcytosis by botulinum neurotoxin expression not only hampers liraglutide transport into the brain and its activation of target hypothalamic neurons, but also blocks its anti-obesity effects on food intake, body weight and fat mass, and fatty acid oxidation. Collectively, these striking data indicate that the liraglutide-induced activation of hypothalamic neurons and its downstream metabolic effects are mediated by its tanycytic transport into the mediobasal hypothalamus, strengthening the notion of tanycytes as key regulators of metabolic homeostasis.


Subject(s)
Diabetes Mellitus, Type 2 , Liraglutide , Animals , Blood-Brain Barrier , Diabetes Mellitus, Type 2/metabolism , Ependymoglial Cells , Hypothalamus/metabolism , Liraglutide/pharmacology , Mice , Obesity/drug therapy , Obesity/metabolism
7.
Cell Rep ; 41(8): 111698, 2022 11 22.
Article in English | MEDLINE | ID: mdl-36417883

ABSTRACT

Therapies based on glucagon-like peptide-1 (GLP-1) long-acting analogs and insulin are often used in the treatment of metabolic diseases. Both insulin and GLP-1 receptors are expressed in metabolically relevant brain regions, suggesting a cooperative action. However, the mechanisms underlying the synergistic actions of insulin and GLP-1R agonists remain elusive. In this study, we show that insulin-induced hypoglycemia enhances GLP-1R agonists entry in hypothalamic and area, leading to enhanced whole-body fat oxidation. Mechanistically, this phenomenon relies on the release of tanycyctic vascular endothelial growth factor A, which is selectively impaired after calorie-rich diet exposure. In humans, low blood glucose also correlates with enhanced blood-to-brain passage of insulin, suggesting that blood glucose gates the passage other energy-related signals in the brain. This study implies that the preventing hyperglycemia is important to harnessing the full benefit of GLP-1R agonist entry in the brain and action onto lipid mobilization and body weight loss.


Subject(s)
Blood Glucose , Vascular Endothelial Growth Factor A , Humans , Blood Glucose/metabolism , Vascular Endothelial Growth Factor A/metabolism , Glucagon-Like Peptide 1/metabolism , Insulin/metabolism , Homeostasis , Brain/metabolism
8.
Science ; 377(6610): eabq4515, 2022 09 02.
Article in English | MEDLINE | ID: mdl-36048943

ABSTRACT

At the present time, no viable treatment exists for cognitive and olfactory deficits in Down syndrome (DS). We show in a DS model (Ts65Dn mice) that these progressive nonreproductive neurological symptoms closely parallel a postpubertal decrease in hypothalamic as well as extrahypothalamic expression of a master molecule that controls reproduction-gonadotropin-releasing hormone (GnRH)-and appear related to an imbalance in a microRNA-gene network known to regulate GnRH neuron maturation together with altered hippocampal synaptic transmission. Epigenetic, cellular, chemogenetic, and pharmacological interventions that restore physiological GnRH levels abolish olfactory and cognitive defects in Ts65Dn mice, whereas pulsatile GnRH therapy improves cognition and brain connectivity in adult DS patients. GnRH thus plays a crucial role in olfaction and cognition, and pulsatile GnRH therapy holds promise to improve cognitive deficits in DS.


Subject(s)
Cognition , Cognitive Dysfunction , Down Syndrome , Gonadotropin-Releasing Hormone , Olfaction Disorders , Adult , Animals , Cognition/drug effects , Cognition/physiology , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/etiology , Disease Models, Animal , Down Syndrome/complications , Down Syndrome/drug therapy , Down Syndrome/psychology , Female , Gonadotropin-Releasing Hormone/pharmacology , Gonadotropin-Releasing Hormone/physiology , Gonadotropin-Releasing Hormone/therapeutic use , Humans , Hypothalamus/drug effects , Hypothalamus/metabolism , Male , Mice , Middle Aged , Olfaction Disorders/drug therapy , Olfaction Disorders/etiology , Synaptic Transmission/drug effects , Young Adult
9.
Cell Metab ; 34(2): 269-284.e9, 2022 02 01.
Article in English | MEDLINE | ID: mdl-35108514

ABSTRACT

Obesity and type 2 diabetes are associated with cognitive dysfunction. Because the hypothalamus is implicated in energy balance control and memory disorders, we hypothesized that specific neurons in this brain region are at the interface of metabolism and cognition. Acute obesogenic diet administration in mice impaired recognition memory due to defective production of the neurosteroid precursor pregnenolone in the hypothalamus. Genetic interference with pregnenolone synthesis by Star deletion in hypothalamic POMC, but not AgRP neurons, deteriorated recognition memory independently of metabolic disturbances. Our data suggest that pregnenolone's effects on cognitive function were mediated via an autocrine mechanism on POMC neurons, influencing hippocampal long-term potentiation. The relevance of central pregnenolone on cognition was also confirmed in metabolically unhealthy patients with obesity. Our data reveal an unsuspected role for POMC neuron-derived neurosteroids in cognition. These results provide the basis for a framework to investigate new facets of POMC neuron biology with implications for cognitive disorders.


Subject(s)
Diabetes Mellitus, Type 2 , Metabolic Diseases , Animals , Diabetes Mellitus, Type 2/metabolism , Humans , Hypothalamus/metabolism , Metabolic Diseases/metabolism , Mice , Mice, Inbred C57BL , Pregnenolone/metabolism , Pro-Opiomelanocortin/metabolism
10.
Mol Cell Endocrinol ; 538: 111449, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34478806

ABSTRACT

Ghrelin is a peptide hormone mainly secreted from gastrointestinal tract that acts via the growth hormone secretagogue receptor (GHSR), which is highly expressed in the brain. Strikingly, the accessibility of ghrelin to the brain seems to be limited and restricted to few brain areas. Previous studies in mice have shown that ghrelin can access the brain via the blood-cerebrospinal fluid (CSF) barrier, an interface constituted by the choroid plexus and the hypothalamic tanycytes. Here, we performed a variety of in vivo and in vitro studies to test the hypothesis that the transport of ghrelin across the blood-CSF barrier occurs in a GHSR-dependent manner. In vivo, we found that the uptake of systemically administered fluorescent ghrelin in the choroid plexus epithelial (CPE) cells and in hypothalamic tanycytes depends on the presence of GHSR. Also, we detected lower levels of CSF ghrelin after a systemic ghrelin injection in GHSR-deficient mice, as compared to WT mice. In vitro, the internalization of fluorescent ghrelin was reduced in explants of choroid plexus from GHSR-deficient mice, and unaffected in primary cultures of hypothalamic tanycytes derived from GHSR-deficient mice. Finally, we found that the GHSR mRNA is detected in a pool of CPE cells, but is nearly undetectable in hypothalamic tanycytes with current approaches. Thus, our results suggest that circulating ghrelin crosses the blood-CSF barrier mainly by a mechanism that involves the GHSR, and also possibly via a GHSR-independent mechanism.


Subject(s)
Blood-Brain Barrier/metabolism , Ghrelin/blood , Ghrelin/cerebrospinal fluid , Receptors, Ghrelin/metabolism , Animals , Cells, Cultured , Choroid Plexus/metabolism , Ependymoglial Cells/cytology , Ependymoglial Cells/metabolism , Ghrelin/genetics , Mice , Primary Cell Culture , Signal Transduction
11.
J Clin Invest ; 131(18)2021 09 15.
Article in English | MEDLINE | ID: mdl-34324439

ABSTRACT

Hypothalamic glucose sensing enables an organism to match energy expenditure and food intake to circulating levels of glucose, the main energy source of the brain. Here, we established that tanycytes of the arcuate nucleus of the hypothalamus, specialized glia that line the wall of the third ventricle, convert brain glucose supplies into lactate that they transmit through monocarboxylate transporters to arcuate proopiomelanocortin neurons, which integrate this signal to drive their activity and to adapt the metabolic response to meet physiological demands. Furthermore, this transmission required the formation of extensive connexin-43 gap junction-mediated metabolic networks by arcuate tanycytes. Selective suppression of either tanycytic monocarboxylate transporters or gap junctions resulted in altered feeding behavior and energy metabolism. Tanycytic intercellular communication and lactate production are thus integral to the mechanism by which hypothalamic neurons that regulate energy and glucose homeostasis efficiently perceive alterations in systemic glucose levels as a function of the physiological state of the organism.


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Ependymoglial Cells/metabolism , Glucose/metabolism , Lactic Acid/metabolism , Pro-Opiomelanocortin/metabolism , Animals , Energy Metabolism , Feeding Behavior/physiology , Gap Junctions/metabolism , Gene Knockdown Techniques , Homeostasis , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Neurological , Monocarboxylic Acid Transporters/antagonists & inhibitors , Monocarboxylic Acid Transporters/genetics , Monocarboxylic Acid Transporters/metabolism , Muscle Proteins/antagonists & inhibitors , Muscle Proteins/genetics , Muscle Proteins/metabolism , Neurons/metabolism , Signal Transduction , Symporters/antagonists & inhibitors , Symporters/genetics , Symporters/metabolism
12.
Nat Metab ; 3(8): 1071-1090, 2021 08.
Article in English | MEDLINE | ID: mdl-34341568

ABSTRACT

Metabolic health depends on the brain's ability to control food intake and nutrient use versus storage, processes that require peripheral signals such as the adipocyte-derived hormone, leptin, to cross brain barriers and mobilize regulatory circuits. We have previously shown that hypothalamic tanycytes shuttle leptin into the brain to reach target neurons. Here, using multiple complementary models, we show that tanycytes express functional leptin receptor (LepR), respond to leptin by triggering Ca2+ waves and target protein phosphorylation, and that their transcytotic transport of leptin requires the activation of a LepR-EGFR complex by leptin and EGF sequentially. Selective deletion of LepR in tanycytes blocks leptin entry into the brain, inducing not only increased food intake and lipogenesis but also glucose intolerance through attenuated insulin secretion by pancreatic ß-cells, possibly via altered sympathetic nervous tone. Tanycytic LepRb-EGFR-mediated transport of leptin could thus be crucial to the pathophysiology of diabetes in addition to obesity, with therapeutic implications.


Subject(s)
Brain/metabolism , Ependymoglial Cells/metabolism , ErbB Receptors/metabolism , Leptin/metabolism , Lipid Metabolism , Pancreas/metabolism , Receptors, Leptin/metabolism , Diabetes Mellitus/etiology , Diabetes Mellitus/metabolism , Energy Metabolism , Insulin-Secreting Cells/metabolism , Phosphorylation
13.
Cell Metab ; 33(9): 1820-1835.e9, 2021 09 07.
Article in English | MEDLINE | ID: mdl-34343501

ABSTRACT

Appropriate cristae remodeling is a determinant of mitochondrial function and bioenergetics and thus represents a crucial process for cellular metabolic adaptations. Here, we show that mitochondrial cristae architecture and expression of the master cristae-remodeling protein OPA1 in proopiomelanocortin (POMC) neurons, which are key metabolic sensors implicated in energy balance control, is affected by fluctuations in nutrient availability. Genetic inactivation of OPA1 in POMC neurons causes dramatic alterations in cristae topology, mitochondrial Ca2+ handling, reduction in alpha-melanocyte stimulating hormone (α-MSH) in target areas, hyperphagia, and attenuated white adipose tissue (WAT) lipolysis resulting in obesity. Pharmacological blockade of mitochondrial Ca2+ influx restores α-MSH and the lipolytic program, while improving the metabolic defects of mutant mice. Chemogenetic manipulation of POMC neurons confirms a role in lipolysis control. Our results unveil a novel axis that connects OPA1 in POMC neurons with mitochondrial cristae, Ca2+ homeostasis, and WAT lipolysis in the regulation of energy balance.


Subject(s)
Lipolysis , Pro-Opiomelanocortin , Adipose Tissue/metabolism , Animals , GTP Phosphohydrolases , Homeostasis , Mice , Neurons/metabolism , Pro-Opiomelanocortin/metabolism
15.
Elife ; 82019 07 10.
Article in English | MEDLINE | ID: mdl-31291191

ABSTRACT

Congenital hypogonadotropic hypogonadism (CHH) is a condition characterized by absent puberty and infertility due to gonadotropin releasing hormone (GnRH) deficiency, which is often associated with anosmia (Kallmann syndrome, KS). We identified loss-of-function heterozygous mutations in anti-Müllerian hormone (AMH) and its receptor, AMHR2, in 3% of CHH probands using whole-exome sequencing. We showed that during embryonic development, AMH is expressed in migratory GnRH neurons in both mouse and human fetuses and unconvered a novel function of AMH as a pro-motility factor for GnRH neurons. Pathohistological analysis of Amhr2-deficient mice showed abnormal development of the peripheral olfactory system and defective embryonic migration of the neuroendocrine GnRH cells to the basal forebrain, which results in reduced fertility in adults. Our findings highlight a novel role for AMH in the development and function of GnRH neurons and indicate that AMH signaling insufficiency contributes to the pathogenesis of CHH in humans.


Subject(s)
Anti-Mullerian Hormone/metabolism , Gonadotropin-Releasing Hormone/metabolism , Hypogonadism/metabolism , Neurons/metabolism , Signal Transduction , Adolescent , Adult , Amino Acid Sequence , Animals , Anti-Mullerian Hormone/genetics , Axons/metabolism , Bone Morphogenetic Protein Receptors, Type I/metabolism , COS Cells , Cell Movement , Chlorocebus aethiops , Female , Fertility , Fetus/metabolism , Heterozygote , Humans , Loss of Function Mutation , Luteinizing Hormone/metabolism , Male , Mice, Inbred C57BL , Olfactory Bulb/metabolism , Pedigree , Receptors, Transforming Growth Factor beta/deficiency , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Young Adult
16.
Diabetes ; 68(12): 2210-2222, 2019 12.
Article in English | MEDLINE | ID: mdl-31530579

ABSTRACT

Melanin-concentrating hormone (MCH) is an important regulator of food intake, glucose metabolism, and adiposity. However, the mechanisms mediating these actions remain largely unknown. We used pharmacological and genetic approaches to show that the sirtuin 1 (SIRT1)/FoxO1 signaling pathway in the hypothalamic arcuate nucleus (ARC) mediates MCH-induced feeding, adiposity, and glucose intolerance. MCH reduces proopiomelanocortin (POMC) neuronal activity, and the SIRT1/FoxO1 pathway regulates the inhibitory effect of MCH on POMC expression. Remarkably, the metabolic actions of MCH are compromised in mice lacking SIRT1 specifically in POMC neurons. Of note, the actions of MCH are independent of agouti-related peptide (AgRP) neurons because inhibition of γ-aminobutyric acid receptor in the ARC did not prevent the orexigenic action of MCH, and the hypophagic effect of MCH silencing was maintained after chemogenetic stimulation of AgRP neurons. Central SIRT1 is required for MCH-induced weight gain through its actions on the sympathetic nervous system. The central MCH knockdown causes hypophagia and weight loss in diet-induced obese wild-type mice; however, these effects were abolished in mice overexpressing SIRT1 fed a high-fat diet. These data reveal the neuronal basis for the effects of MCH on food intake, body weight, and glucose metabolism and highlight the relevance of SIRT1/FoxO1 pathway in obesity.


Subject(s)
Adiposity/drug effects , Forkhead Box Protein O1/metabolism , Glucose Intolerance/metabolism , Hyperphagia/metabolism , Hypothalamic Hormones/pharmacology , Melanins/pharmacology , Neurons/drug effects , Pituitary Hormones/pharmacology , Pro-Opiomelanocortin/metabolism , Sirtuin 1/metabolism , Adiposity/physiology , Animals , Forkhead Box Protein O1/genetics , Glucose Intolerance/genetics , Hyperphagia/genetics , Hypothalamus/drug effects , Hypothalamus/metabolism , Male , Mice , Mice, Knockout , Neurons/metabolism , Patch-Clamp Techniques , Rats, Sprague-Dawley , Sirtuin 1/genetics
17.
Neuropharmacology ; 130: 62-70, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29191753

ABSTRACT

Melanin-Concentrating Hormone (MCH) is one of the most relevant orexigenic factors specifically located in the lateral hypothalamic area (LHA), with its physiological relevance demonstrated in studies using several genetically manipulated mice models. However, the central mechanisms controlling MCH-induced hyperphagia remain largely uncharacterized. Here, we show that central injection of MCH in mice deficient for kappa opoid receptor (k-OR) failed to stimulate feeding. To determine the hypothalamic area responsible for this MCH/k-OR interaction, we performed virogenetic studies and found that downregulation of k-OR by adeno-associated viruses (shOprk1-AAV) in LHA, but not in other hypothalamic nuclei, was sufficient to block MCH-induced food intake. Next, we sought to investigate the molecular signaling pathway within the LHA that mediates acute central MCH stimulation of food intake. We found that MCH activates k-OR and that increased levels of phosphorylated extracellular signal regulated kinase (ERK) are associated with downregulation of phospho-S6 Ribosomal Protein. This effect was prevented when a pharmacological inhibitor of k-OR was co-administered with MCH. Finally, the specific activation of the direct upstream regulator of S6 (p70S6K) in the LHA attenuated MCH-stimulated food consumption. Our results reveal that lateral hypothalamic k-OR system modulates the orexigenic action of MCH via the p70S6K/S6 pathway.


Subject(s)
Eating/drug effects , Hypothalamic Hormones/administration & dosage , Melanins/administration & dosage , Pituitary Hormones/administration & dosage , Receptors, Opioid, kappa/antagonists & inhibitors , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Animals , Appetite Depressants/administration & dosage , Appetite Depressants/metabolism , Dependovirus , Hypothalamic Area, Lateral/drug effects , Hypothalamic Area, Lateral/metabolism , Hypothalamic Hormones/metabolism , MAP Kinase Signaling System/drug effects , Male , Melanins/metabolism , Mice , Mice, Inbred C57BL , Pituitary Hormones/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Opioid, kappa/metabolism , Ribosomal Protein S6 Kinases/drug effects , Ribosomal Protein S6 Kinases/metabolism
18.
Eur J Pharmacol ; 815: 241-250, 2017 Nov 15.
Article in English | MEDLINE | ID: mdl-28943102

ABSTRACT

Behavioral studies have suggested that (p-ClPhSe)2 elicits an anorectic-like action in rats by inducing multiple effects such as satiety-enhancing effect, malaise and specific flavor; however, the molecular mechanisms underlying its anorexigenic action remain unclarified. Here, male Sprague-Dawley rats received acute and sub-chronic intraperitoneal treatments with (p-ClPhSe)2; thereafter, in vivo and ex vivo analyses were carried out. The present study reveals that the reduction of food intake resulting from a single treatment with (p-ClPhSe)2 (1mg/kg, i.p.) was associated with decreased hypothalamic levels of pro-melanin-concentrating hormone (pro-MCH) and orexin precursor. In addition, repeated administrations of (p-ClPhSe)2 (10mg/kg; i.p.) for 7 days induced sustained food intake suppression, body weight loss and white fat reduction. Measurements of brown adipose tissue content and temperature as well as data obtained from a pair-fed group indicated that the effects of (p-ClPhSe)2 on the body weight are closely related to its anorexigenic actions, ruling out the possibility of increased thermogenesis. Furthermore, (p-ClPhSe)2 reduced the hypothalamic orexin precursor levels when repeatedly administered to rats. Sub-chronic treatment with (p-ClPhSe)2 caused a decrease of serum triglyceride levels and down-regulation of hepatic cholesterol content. Therefore, the current study characterized the anorectic and reducing body weight actions of (p-ClPhSe)2 in Sprague-Dawley rats. Besides, the set of results suggests that food intake suppressant effects triggered after (p-ClPhSe)2 administration to rats are mainly related with the lower orexin levels in hypothalamus after acute and sub-chronic treatments.


Subject(s)
Anorexia/chemically induced , Anorexia/pathology , Hypothalamus/drug effects , Hypothalamus/pathology , Organoselenium Compounds/adverse effects , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Animals , Anorexia/blood , Anorexia/psychology , Body Composition/drug effects , Body Weight/drug effects , Eating/drug effects , Hypothalamus/metabolism , Liver/drug effects , Liver/metabolism , Male , Rats , Rats, Sprague-Dawley , Satiety Response/drug effects , Time Factors , Triglycerides/blood
19.
J Mol Endocrinol ; 58(3): R191-R202, 2017 04.
Article in English | MEDLINE | ID: mdl-28196832

ABSTRACT

GPR55 is a G-protein-coupled receptor (GPCR) that has been identified as a new cannabinoid receptor. Given the wide localization of GPR55 in brain and peripheral tissues, this receptor has emerged as a regulator of multiple biological actions. Lysophosphatidylinositol (LPI) is generally accepted as the endogenous ligand of GPR55. In this review, we will focus on the role of GPR55 in energy balance and glucose metabolism. We will summarize its actions on feeding, nutrient partitioning, gastrointestinal motility and insulin secretion in preclinical models and the scarce data available in humans. The potential of GPR55 to become a new pharmaceutical target to treat obesity and type 2 diabetes, as well as the foreseeing difficulties are also discussed.


Subject(s)
Energy Metabolism , Receptors, G-Protein-Coupled/metabolism , Adipose Tissue/metabolism , Animals , Cannabinoids/metabolism , Cannabinoids/pharmacology , Diabetes Mellitus, Type 2/metabolism , Drug Discovery , Energy Metabolism/drug effects , Gene Expression Regulation , Humans , Ligands , Molecular Targeted Therapy , Organ Specificity/genetics , Receptors, Cannabinoid/metabolism , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/genetics , Rodentia
20.
Cell Metab ; 25(6): 1390-1399.e6, 2017 Jun 06.
Article in English | MEDLINE | ID: mdl-28591639

ABSTRACT

Proopiomelanocortin (POMC) neurons are critical sensors of nutrient availability implicated in energy balance and glucose metabolism control. However, the precise mechanisms underlying nutrient sensing in POMC neurons remain incompletely understood. We show that mitochondrial dynamics mediated by Mitofusin 1 (MFN1) in POMC neurons couple nutrient sensing with systemic glucose metabolism. Mice lacking MFN1 in POMC neurons exhibited defective mitochondrial architecture remodeling and attenuated hypothalamic gene expression programs during the fast-to-fed transition. This loss of mitochondrial flexibility in POMC neurons bidirectionally altered glucose sensing, causing abnormal glucose homeostasis due to defective insulin secretion by pancreatic ß cells. Fed mice lacking MFN1 in POMC neurons displayed enhanced hypothalamic mitochondrial oxygen flux and reactive oxygen species generation. Central delivery of antioxidants was able to normalize the phenotype. Collectively, our data posit MFN1-mediated mitochondrial dynamics in POMC neurons as an intrinsic nutrient-sensing mechanism and unveil an unrecognized link between this subset of neurons and insulin release.


Subject(s)
GTP Phosphohydrolases/metabolism , Glucose/metabolism , Insulin-Secreting Cells/transplantation , Insulin/metabolism , Mitochondria/metabolism , Mitochondrial Dynamics , Neurons/metabolism , Pro-Opiomelanocortin , Animals , GTP Phosphohydrolases/genetics , Glucose/genetics , Insulin/genetics , Insulin Secretion , Mice , Mice, Knockout , Mitochondria/genetics
SELECTION OF CITATIONS
SEARCH DETAIL