Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Nat Commun ; 11(1): 4458, 2020 09 07.
Article in English | MEDLINE | ID: mdl-32895383

ABSTRACT

In rodent models of type 2 diabetes (T2D), sustained remission of hyperglycemia can be induced by a single intracerebroventricular (icv) injection of fibroblast growth factor 1 (FGF1), and the mediobasal hypothalamus (MBH) was recently implicated as the brain area responsible for this effect. To better understand the cellular response to FGF1 in the MBH, we sequenced >79,000 single-cell transcriptomes from the hypothalamus of diabetic Lepob/ob mice obtained on Days 1 and 5 after icv injection of either FGF1 or vehicle. A wide range of transcriptional responses to FGF1 was observed across diverse hypothalamic cell types, with glial cell types responding much more robustly than neurons at both time points. Tanycytes and ependymal cells were the most FGF1-responsive cell type at Day 1, but astrocytes and oligodendrocyte lineage cells subsequently became more responsive. Based on histochemical and ultrastructural evidence of enhanced cell-cell interactions between astrocytes and Agrp neurons (key components of the melanocortin system), we performed a series of studies showing that intact melanocortin signaling is required for the sustained antidiabetic action of FGF1. These data collectively suggest that hypothalamic glial cells are leading targets for the effects of FGF1 and that sustained diabetes remission is dependent on intact melanocortin signaling.


Subject(s)
Diabetes Mellitus, Experimental/diet therapy , Diabetes Mellitus, Type 2/drug therapy , Fibroblast Growth Factor 1/administration & dosage , Hypoglycemic Agents/administration & dosage , Hypothalamus/drug effects , Recombinant Proteins/administration & dosage , Agouti-Related Protein/metabolism , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Blood Glucose/analysis , Cell Communication , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/etiology , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/etiology , Diabetes Mellitus, Type 2/pathology , Diet, High-Fat/adverse effects , Dietary Sucrose/administration & dosage , Dietary Sucrose/adverse effects , Humans , Hypothalamus/cytology , Hypothalamus/pathology , Injections, Intraventricular , Leptin/genetics , Male , Melanocortins/metabolism , Melanocyte-Stimulating Hormones/administration & dosage , Mice , Mice, Knockout , Neurons/drug effects , Neurons/metabolism , Oligodendroglia/drug effects , Oligodendroglia/metabolism , RNA-Seq , Receptor, Melanocortin, Type 4/genetics , Receptors, Melanocortin/antagonists & inhibitors , Receptors, Melanocortin/metabolism , Remission Induction/methods , Signal Transduction/drug effects , Single-Cell Analysis , Stereotaxic Techniques , Transcriptome/drug effects
2.
J Endocrinol ; 235(2): 111-122, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28851749

ABSTRACT

Nesfatin-1 is a bioactive polypeptide expressed both in the brain and peripheral tissues and involved in the control of energy balance by reducing food intake. Central administration of nesfatin-1 significantly increases energy expenditure, as demonstrated by a higher dry heat loss; yet, the mechanisms underlying the thermogenic effect of central nesfatin-1 remain unknown. Therefore, in this study, we sought to investigate whether the increase in energy expenditure induced by nesfatin-1 is mediated by the central melanocortin pathway, which was previously reported to mediate central nesfatin-1´s effects on feeding and numerous other physiological functions. With the application of direct calorimetry, we found that intracerebroventricular nesfatin-1 (25 pmol) treatment increased dry heat loss and that this effect was fully blocked by simultaneous administration of an equimolar dose of the melanocortin 3/4 receptor antagonist, SHU9119. Interestingly, the nesfatin-1-induced increase in dry heat loss was positively correlated with body weight loss. In addition, as assessed with thermal imaging, intracerebroventricular nesfatin-1 (100 pmol) increased interscapular brown adipose tissue (iBAT) as well as tail temperature, suggesting increased heat production in the iBAT and heat dissipation over the tail surface. Finally, nesfatin-1 upregulated pro-opiomelanocortin and melanocortin 3 receptor mRNA expression in the hypothalamus, accompanied by a significant increase in iodothyronine deiodinase 2 and by a nonsignificant increase in uncoupling protein 1 and peroxisome proliferator-activated receptor gamma coactivator-1 alpha mRNA in the iBAT. Overall, we clearly demonstrate that nesfatin-1 requires the activation of the central melanocortin system to increase iBAT thermogenesis and, in turn, overall energy expenditure.


Subject(s)
Calcium-Binding Proteins/metabolism , DNA-Binding Proteins/metabolism , Melanocortins/metabolism , Nerve Tissue Proteins/metabolism , Thermogenesis/physiology , Animals , Biomarkers , Calcium-Binding Proteins/genetics , DNA-Binding Proteins/genetics , Ear , Hypothalamus/metabolism , Male , Melanocyte-Stimulating Hormones/pharmacology , Nerve Tissue Proteins/genetics , Nucleobindins , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptors, Melanocortin/antagonists & inhibitors , Receptors, Melanocortin/genetics , Receptors, Melanocortin/metabolism , Tail , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism
3.
J Med Chem ; 59(7): 3112-28, 2016 Apr 14.
Article in English | MEDLINE | ID: mdl-26959173

ABSTRACT

Pharmacological probes for the melanocortin receptors have been utilized for studying various disease states including cancer, sexual function disorders, Alzheimer's disease, social disorders, cachexia, and obesity. This study focused on the design and synthesis of bivalent ligands to target melanocortin receptor homodimers. Lead ligands increased binding affinity by 14- to 25-fold and increased cAMP signaling potency by 3- to 5-fold compared to their monovalent counterparts. Unexpectedly, different bivalent ligands showed preferences for particular melanocortin receptor subtypes depending on the linker that connected the binding scaffolds, suggesting structural differences between the various dimer subtypes. Homobivalent compound 12 possessed a functional profile that was unique from its monovalent counterpart providing evidence of the discrete effects of bivalent ligands. Lead compound 7 significantly decreased feeding in mice after intracerebroventricular administration. To the best of our knowledge, this is the first report of a melanocortin bivalent ligand's in vivo physiological effects.


Subject(s)
Receptors, Melanocortin/agonists , Receptors, Melanocortin/antagonists & inhibitors , Animals , Binding, Competitive , Chemistry Techniques, Synthetic , Cyclic AMP/metabolism , Drug Design , Drug Evaluation, Preclinical/methods , Eating/drug effects , Female , Humans , Infusions, Intraventricular , Ligands , Male , Mice, Inbred C57BL , Peptides/chemistry , Peptides/metabolism , Peptides/pharmacology , Protein Multimerization , Receptor, Melanocortin, Type 1/metabolism , Receptor, Melanocortin, Type 3/metabolism , Receptor, Melanocortin, Type 4/metabolism , Receptors, Melanocortin/metabolism , Structure-Activity Relationship
4.
Diabetes ; 65(3): 660-72, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26467632

ABSTRACT

Hypothalamic proopiomelanocortin (POMC) is essential for the physiological regulation of energy balance; however, its role in glucose homeostasis remains less clear. We show that hypothalamic arcuate nucleus (Arc)POMC-deficient mice, which develop severe obesity and insulin resistance, unexpectedly exhibit improved glucose tolerance and remain protected from hyperglycemia. To explain these paradoxical phenotypes, we hypothesized that an insulin-independent pathway is responsible for the enhanced glucose tolerance. Indeed, the mutant mice demonstrated increased glucose effectiveness and exaggerated glycosuria relative to wild-type littermate controls at comparable blood glucose concentrations. Central administration of the melanocortin receptor agonist melanotan II in mutant mice reversed alterations in glucose tolerance and glycosuria, whereas, conversely, administration of the antagonist Agouti-related peptide (Agrp) to wild-type mice enhanced glucose tolerance. The glycosuria of ArcPOMC-deficient mice was due to decreased levels of renal GLUT 2 (rGLUT2) but not sodium-glucose cotransporter 2 and was associated with reduced renal catecholamine content. Epinephrine treatment abolished the genotype differences in glucose tolerance and rGLUT2 levels, suggesting that reduced renal sympathetic nervous system (SNS) activity is the underlying mechanism for the observed glycosuria and improved glucose tolerance in ArcPOMC-deficient mice. Therefore, the ArcPOMC-SNS-rGLUT2 axis is potentially an insulin-independent therapeutic target to control diabetes.


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Blood Glucose/metabolism , Glucose Transporter Type 2/genetics , Glycosuria, Renal/genetics , Insulin Resistance/genetics , Kidney/metabolism , Obesity/genetics , Pro-Opiomelanocortin/genetics , Sympathetic Nervous System/metabolism , Agouti-Related Protein/pharmacology , Animals , Blood Glucose/drug effects , Blotting, Western , Epinephrine/metabolism , Glucose Tolerance Test , Glucose Transporter Type 2/metabolism , Glycosuria, Renal/metabolism , Hypothalamus/metabolism , Injections, Intraventricular , Mice , Mice, Knockout , Norepinephrine/metabolism , Obesity/metabolism , Peptides, Cyclic/pharmacology , Pro-Opiomelanocortin/deficiency , Pro-Opiomelanocortin/metabolism , Receptors, Melanocortin/agonists , Receptors, Melanocortin/antagonists & inhibitors , alpha-MSH/analogs & derivatives , alpha-MSH/pharmacology
5.
Neuroscience ; 310: 252-67, 2015 Dec 03.
Article in English | MEDLINE | ID: mdl-26404873

ABSTRACT

Physical activity and non-exercise activity thermogenesis (NEAT) are crucial factors accounting for individual differences in body weight, interacting with genetic predisposition. In the brain, a number of neuroendocrine intermediates regulate food intake and energy expenditure (EE); this includes the brain melanocortin (MC) system, consisting of MC peptides as well as their receptors (MCR). MC3R and MC4R have emerged as critical modulators of EE and food intake. To determine how variance in MC signaling may underlie individual differences in physical activity levels, we examined behavioral response to MC receptor agonists and antagonists in rats that show high and low levels of physical activity and NEAT, that is, high- and low-capacity runners (HCR, LCR), developed by artificial selection for differential intrinsic aerobic running capacity. Focusing on the hypothalamus, we identified brain region-specific elevations in expression of MCR 3, 4, and also MC5R, in the highly active, lean HCR relative to the less active and obesity-prone LCR. Further, the differences in activity and associated EE as a result of MCR activation or suppression using specific agonists and antagonists were similarly region-specific and directly corresponded to the differential MCR expression patterns. The agonists and antagonists investigated here did not significantly impact food intake at the doses used, suggesting that the differential pattern of receptor expression may by more meaningful to physical activity than to other aspects of energy balance regulation. Thus, MCR-mediated physical activity may be a key neural mechanism in distinguishing the lean phenotype and a target for enhancing physical activity and NEAT.


Subject(s)
Energy Metabolism , Hypothalamus/metabolism , Motor Activity , Receptors, Melanocortin/metabolism , Animals , Body Weight , Eating , Female , Male , RNA, Messenger , Rats , Receptors, Melanocortin/agonists , Receptors, Melanocortin/antagonists & inhibitors
6.
Pharmacotherapy ; 33(4): 411-21, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23553810

ABSTRACT

Hypoactive sexual desire disorder (HSDD) affects nearly 1 in 10 women. Thus, it is essential for pharmacists and other health care providers to be comfortable when discussing a patient's sexual health to ensure appropriate triage so that the specific causes of HSDD can be identified and potential recommendations provided. HSDD is defined as the absence or deficiency of sexual interest and/or desire, leading to significant distress and interpersonal difficulties. As health care providers, pharmacists have a critical role in assessing the presence of HSDD and providing education on available treatment options. This article will review the potential causes of HSDD and low sexual desire, the screening tools available, and the significant role of health care professionals in communicating with patients about their sexual health. An overview of the importance of behavioral modifications, the current pharmacologic options being investigated, and the use of complementary and alternative therapies will also be explored. Currently, buproprion is the primary pharmacologic agent that has shown positive results in treating patients with HSDD. The use of testosterone therapy will not be addressed in this article, as this therapy is described in greater detail elsewhere.


Subject(s)
Health Communication , Reproductive Health , Sexual Dysfunctions, Psychological/drug therapy , Sexual Dysfunctions, Psychological/psychology , Antidepressive Agents, Second-Generation/therapeutic use , Benzimidazoles/therapeutic use , Bupropion/therapeutic use , Complementary Therapies/psychology , Female , Health Behavior , Humans , Peptides, Cyclic/therapeutic use , Piperazines/therapeutic use , Purines/therapeutic use , Receptors, Melanocortin/antagonists & inhibitors , Serotonin Antagonists/therapeutic use , Sexual Dysfunctions, Psychological/diagnosis , Sildenafil Citrate , Sulfones/therapeutic use , Vasodilator Agents/therapeutic use , alpha-MSH/therapeutic use
7.
Diabetes ; 62(5): 1500-4, 2013 May.
Article in English | MEDLINE | ID: mdl-23274904

ABSTRACT

C-type natriuretic peptide (CNP) and its receptor are abundantly distributed in the brain, especially in the arcuate nucleus (ARC) of the hypothalamus associated with regulating energy homeostasis. To elucidate the possible involvement of CNP in energy regulation, we examined the effects of intracerebroventricular administration of CNP on food intake in mice. The intracerebroventricular administration of CNP-22 and CNP-53 significantly suppressed food intake on 4-h refeeding after 48-h fasting. Next, intracerebroventricular administration of CNP-22 and CNP-53 significantly decreased nocturnal food intake. The increment of food intake induced by neuropeptide Y and ghrelin was markedly suppressed by intracerebroventricular administration of CNP-22 and CNP-53. When SHU9119, an antagonist for melanocortin-3 and melanocortin-4 receptors, was coadministered with CNP-53, the suppressive effect of CNP-53 on refeeding after 48-h fasting was significantly attenuated by SHU9119. Immunohistochemical analysis revealed that intracerebroventricular administration of CNP-53 markedly increased the number of c-Fos-positive cells in the ARC, paraventricular nucleus, dorsomedial hypothalamus, ventromedial hypothalamic nucleus, and lateral hypothalamus. In particular, c-Fos-positive cells in the ARC after intracerebroventricular administration of CNP-53 were coexpressed with α-melanocyte-stimulating hormone immunoreactivity. These results indicated that intracerebroventricular administration of CNP induces an anorexigenic action, in part, via activation of the melanocortin system.


Subject(s)
Appetite Regulation , Hypothalamus/metabolism , Melanocortins/agonists , Natriuretic Peptide, C-Type/metabolism , Neurons/metabolism , Receptors, Melanocortin/agonists , Signal Transduction , Animals , Appetite Regulation/drug effects , Behavior, Animal/drug effects , Feeding Behavior/drug effects , Ghrelin/antagonists & inhibitors , Ghrelin/metabolism , Hypothalamus/cytology , Hypothalamus/drug effects , Injections, Intraventricular , Male , Melanocortins/antagonists & inhibitors , Melanocortins/metabolism , Melanocyte-Stimulating Hormones/pharmacology , Mice , Mice, Inbred C57BL , Natriuretic Peptide, C-Type/administration & dosage , Natriuretic Peptide, C-Type/antagonists & inhibitors , Nerve Tissue Proteins/administration & dosage , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/metabolism , Neurons/cytology , Neurons/drug effects , Neuropeptide Y/antagonists & inhibitors , Neuropeptide Y/metabolism , Protein Isoforms/agonists , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/metabolism , Protein Precursors/administration & dosage , Protein Precursors/antagonists & inhibitors , Protein Precursors/metabolism , Receptors, Melanocortin/antagonists & inhibitors , Receptors, Melanocortin/metabolism , Signal Transduction/drug effects , Up-Regulation/drug effects , alpha-MSH/metabolism
8.
Diabetes ; 62(3): 801-10, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23160530

ABSTRACT

Hypothalamic glucose sensing is involved in the control of feeding behavior and peripheral glucose homeostasis, and glial cells are suggested to play an important role in this process. Diazepam-binding inhibitor (DBI) and its processing product the octadecaneuropeptide (ODN), collectively named endozepines, are secreted by astroglia, and ODN is a potent anorexigenic factor. Therefore, we investigated the involvement of endozepines in brain glucose sensing. First, we showed that intracerebroventricular administration of glucose in rats increases DBI expression in hypothalamic glial-like tanycytes. We then demonstrated that glucose stimulates endozepine secretion from hypothalamic explants. Feeding experiments indicate that the anorexigenic effect of central administration of glucose was blunted by coinjection of an ODN antagonist. Conversely, the hyperphagic response elicited by central glucoprivation was suppressed by an ODN agonist. The anorexigenic effects of centrally injected glucose or ODN agonist were suppressed by blockade of the melanocortin-3/4 receptors, suggesting that glucose sensing involves endozepinergic control of the melanocortin pathway. Finally, we found that brain endozepines modulate blood glucose levels, suggesting their involvement in a feedback loop controlling whole-body glucose homeostasis. Collectively, these data indicate that endozepines are a critical relay in brain glucose sensing and potentially new targets in treatment of metabolic disorders.


Subject(s)
Appetite Regulation , Diazepam Binding Inhibitor/metabolism , Feedback, Physiological , Glucose/metabolism , Hypothalamus/metabolism , Neuroglia/metabolism , Neuropeptides/metabolism , Peptide Fragments/metabolism , Animals , Appetite Depressants/administration & dosage , Appetite Depressants/pharmacology , Appetite Regulation/drug effects , Appetite Stimulants/administration & dosage , Appetite Stimulants/pharmacology , Appetitive Behavior/drug effects , Arcuate Nucleus of Hypothalamus/cytology , Arcuate Nucleus of Hypothalamus/drug effects , Arcuate Nucleus of Hypothalamus/metabolism , Diazepam Binding Inhibitor/agonists , Diazepam Binding Inhibitor/antagonists & inhibitors , Feedback, Physiological/drug effects , Gene Expression Regulation/drug effects , Glucose/administration & dosage , Hypothalamus/cytology , Hypothalamus/drug effects , Injections, Intraventricular , Male , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/metabolism , Neuroglia/drug effects , Neuropeptides/antagonists & inhibitors , Peptide Fragments/antagonists & inhibitors , Protein Processing, Post-Translational , Rats , Rats, Wistar , Receptors, Melanocortin/antagonists & inhibitors , Receptors, Melanocortin/metabolism , Synaptic Transmission/drug effects , Tissue Culture Techniques
9.
Neurosci Res ; 70(1): 55-61, 2011 May.
Article in English | MEDLINE | ID: mdl-21291921

ABSTRACT

Pituitary adenylate cyclase-activating polypeptide (PACAP) is a peptidergic neurotransmitter that is highly expressed in the nervous system. We have previously reported that a central injection of PACAP leads to changes in the autonomic nervous system tones including sympathetic excitation and parasympathetic inhibition. An anatomical study revealed that melanocortin and PACAP are colocalized in some hypothalamic nuclei. Here, we investigated the possible role of the melanocortin system in autonomic control by PACAP using SHU9119, an antagonist of the melanocortin receptors (MC3-R/MC4-R). Pretreatment with SHU-9119 did not affect the activating neural responses of adrenal, renal, and lumbar sympathetic nerves following a PACAP injection However, SHU9119 significantly eliminated the suppressing effect of a PACAP injection on gastric vagal nerve activity and excitation effects on liver and brown adipose tissue sympathetic nerve activities. These results suggest that the brain melanocortin system might play a key role in the control of thermogenic sympathetic outflows and digestive parasympathetic outflow by PACAP, but this system does not participate in the central effects of PACAP on cardiovascular function and neural activities of renal, adrenal, and lumbar sympathetic nerves.


Subject(s)
Autonomic Nervous System/physiology , Autonomic Pathways/physiology , Hypothalamus/physiology , Pituitary Adenylate Cyclase-Activating Polypeptide/physiology , Pro-Opiomelanocortin/physiology , Animals , Autonomic Nervous System/drug effects , Autonomic Pathways/drug effects , Digestive System/innervation , Hypothalamus/drug effects , Male , Melanocyte-Stimulating Hormones/pharmacology , Pituitary Adenylate Cyclase-Activating Polypeptide/pharmacology , Rats , Rats, Wistar , Receptors, Melanocortin/antagonists & inhibitors , Receptors, Melanocortin/physiology , Sympathetic Fibers, Postganglionic/drug effects , Sympathetic Fibers, Postganglionic/physiology , Thermogenesis/physiology , Vagus Nerve/drug effects , Vagus Nerve/physiology , Viscera/innervation , Viscera/physiology
10.
Int J Obes (Lond) ; 35(5): 629-41, 2011 May.
Article in English | MEDLINE | ID: mdl-20733584

ABSTRACT

OBJECTIVE: Reduction of melanocortin signaling in the brain results in obesity. However, where in the brain reduced melanocortin signaling mediates this effect is poorly understood. DESIGN: We determined the effects of long-term inhibition of melanocortin receptor activity in specific brain regions of the rat brain. Melanocortin signaling was inhibited by injection of a recombinant adeno-associated viral (rAAV) vector that overexpressed Agouti-related peptide (AgRP) into the paraventricular nucleus (PVN), the ventromedial hypothalamus (VMH), the lateral hypothalamus (LH) or the accumbens shell (Acc). RESULTS: Overexpression of AgRP in the rat PVN, VMH or LH increased bodyweight, the percentage of white adipose tissue, plasma leptin and insulin concentrations and food intake. Food intake was mainly increased because of an increase in meal size in the light and dark phases, after overexpression of AgRP in the PVN, LH or VMH. Overexpression of AgRP in the PVN or VMH reduced average body core temperature in the dark on day 40 post injection, whereas AgRP overexpression in the LH did not affect temperature. In addition, overexpression of AgRP in the PVN, LH or VMH did not significantly alter mRNA expression of AgRP, neuropeptide Y (NPY), pro-opiomelanocortin (POMC) or suppressor of cytokine signaling 3 (SOCS3) in the arcuate. Overexpression of AgRP in the Acc did not have any effect on the measured parameters. CONCLUSIONS: Reduction of melanocortin signaling in several hypothalamic regions increased meal size. However, there were brain area-specific effects on other parameters such as core temperature and plasma leptin concentrations. In a previous study, where NPY was overexpressed with an rAAV vector in the PVN and LH, meal frequency and meal size were increased respectively, whereas locomotor activity was reduced by NPY overexpression at both nuclei. Taken together, AgRP and NPY have complementary roles in energy balance.


Subject(s)
Agouti-Related Protein/metabolism , Body Weight/physiology , Energy Metabolism/physiology , Hypothalamus/metabolism , Obesity/metabolism , Receptors, Melanocortin/physiology , Animals , Cell Line , Eating/physiology , Hypothalamic Area, Lateral/metabolism , Hypothalamus/physiology , Male , Midline Thalamic Nuclei/metabolism , Nucleus Accumbens/metabolism , Obesity/physiopathology , Rats , Rats, Wistar , Receptors, Melanocortin/antagonists & inhibitors , Ventromedial Hypothalamic Nucleus/metabolism
11.
Diabetes ; 58(1): 87-94, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18984739

ABSTRACT

OBJECTIVE: Xenin, a 25-amino acid peptide, was initially isolated from human gastric mucosa. Plasma levels of xenin rise after a meal in humans, and administration of xenin inhibits feeding in rats and chicks. However, little is known about the mechanism by which xenin regulates food intake. Signaling pathways including leptin and melanocortins play a pivotal role in the regulation of energy balance. Therefore, we addressed the hypothesis that xenin functions as a satiety factor by acting through the melanocortin system or by interacting with leptin. RESEARCH DESIGN AND METHODS: The effect of intracerebroventricular and intraperitoneal administration of xenin on food intake was examined in wild-type, agouti, and ob/ob mice. The effect of intracerebroventricular injection of SHU9119, a melanocortin receptor antagonist, on xenin-induced anorexia was also examined in wild-type mice. To determine whether the hypothalamus mediates the anorectic effect of xenin, we examined the effect of intraperitoneal xenin on hypothalamic Fos expression. RESULTS: Both intracerebroventricular and intraperitoneal administration of xenin inhibited fasting-induced hyperphagia in wild-type mice in a dose-dependent manner. The intraperitoneal injection of xenin also reduced nocturnal intake in ad libitum-fed wild-type mice. The intraperitoneal injection of xenin increased Fos immunoreactivity in hypothalamic nuclei, including the paraventricular nucleus and the arcuate nucleus. Xenin reduced food intake in agouti and ob/ob mice. SHU9119 did not block xenin-induced anorexia. CONCLUSIONS: Our data suggest that xenin reduces food intake partly by acting through the hypothalamus but via signaling pathways that are independent of those used by leptin or melanocortins.


Subject(s)
Eating/drug effects , Melanocortins/metabolism , Peptides/pharmacology , Signal Transduction/drug effects , Animals , Fasting/physiology , Hypothalamus/drug effects , Hypothalamus/metabolism , Immunohistochemistry , Leptin/metabolism , Male , Melanocyte-Stimulating Hormones/pharmacology , Mice , Mice, Inbred C57BL , Mice, Obese , Neurotensin , Oncogene Proteins v-fos/metabolism , Peptides/administration & dosage , Receptors, Melanocortin/antagonists & inhibitors , Signal Transduction/physiology
12.
Brain Res ; 1047(2): 214-23, 2005 Jun 21.
Article in English | MEDLINE | ID: mdl-15907812

ABSTRACT

While there have been many studies in various species examining the mode of central leptin action on food intake, there is however a paucity of data in birds. We have, therefore, addressed this issue in broiler chickens because this strain was selected for high growth rate, hence high food intake. Continuous infusion of recombinant chicken leptin (8 microg/kg/h) during 6 h at a constant rate of 3 ml/h resulted in a significant reduction (49-57%) of food intake in 3-week-old broiler chickens (P < 0.05). The effect of leptin within the central nervous system (CNS) was mediated via selective hypothalamic neuropeptides. Leptin significantly decreased the expression of its receptor (Ob-R), neuropeptide Y (NPY), orexin (ORX), and orexin receptor (ORXR) (P < 0.05), but not that of agouti-related protein (AgRP) (anabolic/orexigenic effectors) in chicken hypothalamus. However, the catabolic/anorexigenic neuropeptides namely proopiomelanocortin (POMC) and corticotropin-releasing hormone (CRH) mRNA levels remained unchanged after leptin treatment. Despite the absence of leptin effect on AgRP (the antagonist of melanocortin receptor MCR) and POMC (the precursor of alpha-melanocyte stimulating hormone which is a potent agonist for MCR), leptin significantly decreased the expression of MCR-4/5 gene in chicken hypothalamus (P < 0.05) suggesting that leptin acts directly (as ligand) or indirectly (via other ligands) on MCRs to regulate food intake in birds. Additionally, leptin down-regulated the expression of fatty acid synthase (FAS) gene in chicken hypothalamus, indicating an additional pathway of leptin action on food intake such as described for FAS inhibitors. These findings provide new insight into the mechanism of leptin control of food intake in chickens.


Subject(s)
Chickens/physiology , Feeding Behavior/physiology , Hypothalamus/drug effects , Leptin/administration & dosage , Agouti-Related Protein , Animals , Blotting, Southern , Corticosterone/blood , Corticotropin-Releasing Hormone/biosynthesis , Corticotropin-Releasing Hormone/drug effects , DNA Primers , Fatty Acid Synthases/biosynthesis , Fatty Acid Synthases/drug effects , Feeding Behavior/drug effects , Gene Expression/drug effects , Infusions, Intravenous , Intercellular Signaling Peptides and Proteins , Intracellular Signaling Peptides and Proteins/drug effects , Leptin/blood , Neuropeptide Y/biosynthesis , Neuropeptide Y/drug effects , Neuropeptides/biosynthesis , Neuropeptides/drug effects , Orexin Receptors , Orexins , Pro-Opiomelanocortin/biosynthesis , Pro-Opiomelanocortin/drug effects , Proteins/drug effects , RNA, Messenger/analysis , Receptors, Cell Surface/biosynthesis , Receptors, Cell Surface/drug effects , Receptors, G-Protein-Coupled , Receptors, Leptin , Receptors, Melanocortin/agonists , Receptors, Melanocortin/antagonists & inhibitors , Receptors, Melanocortin/drug effects , Receptors, Neuropeptide/biosynthesis , Receptors, Neuropeptide/drug effects , Recombinant Proteins/administration & dosage , Reverse Transcriptase Polymerase Chain Reaction , Thyroid Hormones/blood
13.
Brain Res ; 1039(1-2): 137-45, 2005 Mar 28.
Article in English | MEDLINE | ID: mdl-15781055

ABSTRACT

Hypothalamic melanocortins are critical for the control of food intake, and alterations in POMC mRNA have been described in genetic models of obesity. However, the time course of changes in brain transmitters over the development of dietary obesity is less clear. Therefore, we examined the effect of diet-induced obesity on hypothalamic alpha-MSH content and feeding responsiveness to synthetic melanocortins. Male Sprague-Dawley rats fed a high-fat cafeteria diet (30% fat) or chow (5% fat) for 4 or 12 weeks were implanted with intracerebroventricular cannulae and feeding responses to the MC3/4R agonist MTII (0.5 nmol) and the selective MC4R antagonist HS014 (0.8 nmol) were determined. MTII had a long-lasting inhibitory effect on food intake. Chronically overfed animals had a significantly exaggerated inhibitory feeding response 15 and 24 h after MTII injection and lost more body weight (15 +/- 3 g) compared to control rats (4 +/- 4 g; P < 0.05). Daytime administration of HS014 significantly increased food intake in all rats to the same extent (P < 0.05). No change in hypothalamic alpha-MSH content was observed after 2 or 12 weeks of high-fat diet. The observation of increased responsiveness to the melanocortin agonist, in the face of a high-fat diet, suggests melanocortin analogues may have potential for the pharmacological treatment of obesity.


Subject(s)
Dietary Fats/metabolism , Obesity/metabolism , Peptides, Cyclic/administration & dosage , Receptors, Melanocortin/drug effects , alpha-MSH/analogs & derivatives , alpha-MSH/metabolism , Animals , Disease Models, Animal , Eating/drug effects , Eating/physiology , Hypothalamus/drug effects , Hypothalamus/metabolism , Injections, Intraventricular , Leptin/blood , Male , Obesity/blood , Overnutrition/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Melanocortin/agonists , Receptors, Melanocortin/antagonists & inhibitors , Time Factors , alpha-MSH/administration & dosage , alpha-MSH/drug effects
14.
Nature ; 428(6982): 569-74, 2004 Apr 01.
Article in English | MEDLINE | ID: mdl-15058305

ABSTRACT

Obesity is an epidemic in Western society, and causes rapidly accelerating rates of type 2 diabetes and cardiovascular disease. The evolutionarily conserved serine/threonine kinase, AMP-activated protein kinase (AMPK), functions as a 'fuel gauge' to monitor cellular energy status. We investigated the potential role of AMPK in the hypothalamus in the regulation of food intake. Here we report that AMPK activity is inhibited in arcuate and paraventricular hypothalamus (PVH) by the anorexigenic hormone leptin, and in multiple hypothalamic regions by insulin, high glucose and refeeding. A melanocortin receptor agonist, a potent anorexigen, decreases AMPK activity in PVH, whereas agouti-related protein, an orexigen, increases AMPK activity. Melanocortin receptor signalling is required for leptin and refeeding effects on AMPK in PVH. Dominant negative AMPK expression in the hypothalamus is sufficient to reduce food intake and body weight, whereas constitutively active AMPK increases both. Alterations of hypothalamic AMPK activity augment changes in arcuate neuropeptide expression induced by fasting and feeding. Furthermore, inhibition of hypothalamic AMPK is necessary for leptin's effects on food intake and body weight, as constitutively active AMPK blocks these effects. Thus, hypothalamic AMPK plays a critical role in hormonal and nutrient-derived anorexigenic and orexigenic signals and in energy balance.


Subject(s)
Adenylate Kinase/metabolism , Feeding Behavior/physiology , Hormones/metabolism , Hypothalamus/enzymology , Hypothalamus/physiology , Adenylate Kinase/antagonists & inhibitors , Adenylate Kinase/chemistry , Adenylate Kinase/genetics , Animals , Body Weight/drug effects , Energy Metabolism/drug effects , Feeding Behavior/drug effects , Glucose/metabolism , Glucose/pharmacology , Hormones/pharmacology , Hypothalamus/drug effects , Insulin/metabolism , Insulin/pharmacology , Leptin/metabolism , Leptin/pharmacology , Male , Mice , Models, Biological , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Melanocortin/antagonists & inhibitors , Receptors, Melanocortin/metabolism
15.
J Neuroendocrinol ; 15(11): 1046-53, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14622434

ABSTRACT

Ring doves (Streptopelia risoria) exhibit marked increases in food consumption and decreases in body weight when they are provisioning their young. This study examined changes in hypothalamic immunostaining for agouti-related peptide (AGRP), an endogenous melanocortin receptor antagonist and appetite stimulant, during the ring dove breeding cycle. Because prolactin is orexigenic in doves, and is elevated in blood at the onset of parental hyperphagia, we also explored the possibility that prolactin-induced hyperphagia is associated with AGRP changes. The numbers of AGRP-immunoreactive (ir) cells within the tuberal hypothalamus were maximal during the prelaying period of the breeding cycle but decreased dramatically during early incubation. AGRP-ir cell numbers began to increase again during late incubation and reached a peak during the early and late posthatching stages. Because posthatching elevations in AGRP-ir were temporally associated with marked elevations in parental food intake, and because AGRP is orexigenic in doves, these findings suggest that increased AGRP activity in the dove tuberal hypothalamus may contribute to parental hyperphagia. Rising prolactin secretion during late incubation and early posthatching may initiate this increase in AGRP-ir, since intracerebroventricular administration of prolactin significantly elevated food intake and AGRP-ir cell numbers in the tuberal hypothalamus of nonbreeding doves. Prolactin-induced elevations in AGRP-ir cell numbers persisted when the confounding effects of weight gain that accompany prolactin-induced hyperphagia in nonbreeding doves were eliminated by a food restriction procedure, thereby suggesting that prolactin can directly influence AGRP activity under neutral energy state conditions.


Subject(s)
Columbidae/physiology , Hyperphagia/metabolism , Hypothalamus/metabolism , Prolactin/metabolism , Proteins/metabolism , Receptors, Melanocortin/antagonists & inhibitors , Reproduction/physiology , Agouti-Related Protein , Animals , Feeding Behavior/physiology , Female , Hypothalamus/cytology , Immunohistochemistry , Intercellular Signaling Peptides and Proteins , Male , Maternal Behavior/physiology , Nesting Behavior/physiology , Neurons/metabolism , Paternal Behavior , Receptors, Melanocortin/metabolism
16.
Peptides ; 24(4): 603-9, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12860205

ABSTRACT

Agouti and agouti-related protein (AgRP) are endogenous antagonists of the melanocortin receptors (MCxR). Previous data showed that recombinant full-length agouti and a synthetic fragment of AgRP, AgRP (83-132), are inverse agonists at the MC1R and MC4R, respectively. This study demonstrates the smaller analogs AgRP (87-120) and ASIP [90-132 (L89Y)], and short peptides Yc[CRFFNAFC]Y and Qc[CRFFRSAC]S are also MC4R inverse agonists. Furthermore, the relative affinity of the series of MC4R ligands for displacement of radiolabeled antagonist 125I-AgRP (86-132) versus radiolabeled agonist 125I-NDP-MSH did not correlate with ligand efficacy, which is more consistent with an induced-fit model than a simple two-state model of MC4R activation. These data shed new light on the determinants and mechanism of inverse agonism at the MC4R.


Subject(s)
Intercellular Signaling Peptides and Proteins/physiology , Proteins/physiology , beta-MSH/analogs & derivatives , Agouti Signaling Protein , Agouti-Related Protein , Amino Acid Motifs , Cell Line , Cyclic AMP/metabolism , Dose-Response Relationship, Drug , Humans , Hypothalamus/pathology , Kinetics , Ligands , Melanocyte-Stimulating Hormones/pharmacology , Models, Molecular , Obesity , Peptides/chemistry , Peptides, Cyclic/pharmacology , Pigmentation , Protein Binding , Protein Structure, Tertiary , Receptors, Melanocortin/antagonists & inhibitors , Receptors, Melanocortin/chemistry , Recombinant Proteins/chemistry , alpha-MSH/metabolism , beta-MSH/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL