Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Obes Rev ; 18(7): 765-775, 2017 07.
Article in English | MEDLINE | ID: mdl-28429582

ABSTRACT

Obesity is a multifactorial, chronic disease that has proven difficult to treat. An increased understanding of aetiological mechanisms is critical to the development of more effective obesity prevention and treatment strategies. A growing body of empirical evidence has demonstrated parallels between obesity, overeating and substance abuse, including shared behavioural, psychological and neurophysiological factors implicated in the excessive intake of both food and substances of abuse. Several different lines of research have recently emerged that hold the potential to shed light on the connection between obesity, food reward and addiction, with studies examining changes in alcohol use/misuse after weight loss surgery providing a particularly interesting perspective on these interrelationships. However, these lines of investigation have proceeded in relative isolation, and relevant research findings have yet to be integrated in a synthesized, comprehensive manner. To provide an opportunity to achieve such a synthesis, a scientific symposium was convened at the Radcliffe Institute in Cambridge, Massachusetts. Invited participants were researchers working in diverse domains related to the intersection between obesity and addiction. Extensive discussion was generated suggesting novel research directions. In this article, we summarize and synthesize the symposium participants' ongoing research in this area, incorporating additional relevant research holding potential clues regarding the connections between obesity, weight loss surgery and addiction.


Subject(s)
Alcoholism/epidemiology , Bariatric Surgery/adverse effects , Behavior, Addictive/psychology , Hyperphagia/psychology , Obesity/psychology , Obesity/surgery , Alcohol Drinking/epidemiology , Alcoholism/psychology , Animals , Bariatric Surgery/psychology , Ethanol/pharmacokinetics , Gastric Bypass/adverse effects , Gastric Bypass/psychology , Glucagon-Like Peptide 1/blood , Humans , Peptide YY/blood , Reward , Weight Loss
2.
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.

3.
Int J Obes (Lond) ; 38(3): 349-56, 2014 Mar.
Article in English | MEDLINE | ID: mdl-23736372

ABSTRACT

BACKGROUND: Much recent evidence suggest that obesity and related comorbidities contribute to cognitive decline, including the development of non age-related dementia and Alzheimer's disease. Obesity is a serious threat to public health, and few treatments offer proven long-term weight loss. In fact, bariatric surgery remains the most effective long-term therapy to reduce weight and alleviate other aspects of the metabolic syndrome (MetS). Unlike the demonstrated benefits of caloric restriction to prevent weight gain, few if any studies have compared various means of weight loss on central nervous system function and hippocampal-dependent cognitive processes. DESIGN AND RESULTS: Our studies comprise the first direct comparisons of caloric restriction to two bariatric surgeries (Roux-en-Y gastric bypass (RYGB) and vertical sleeve gastrectomy (VSG)) on cognitive function. Weight loss following caloric restriction, RYGB and VSG was associated with generalized improvements in metabolic health and hippocampal-dependent learning, as measured in the radial arm maze and spontaneous alternation tests. However, VSG-treated rats exhibited deficits on spatial learning tasks in the Morris water maze. In addition, whereas VSG animals had elevated hippocampal inflammation, comparable to that of obese controls, RYGB and calorie-restricted (pair-fed, PF) controls exhibited an amelioration of inflammation, as measured by the microglial protein ionized calcium binding adaptor molecule 1 (IBA1). We also assessed whether GHR (ghrelin) replacement would attenuate hippocampal inflammation in VSG, as post-surgical GHR levels are significantly reduced in VSG relative to RYGB and PF rats. However, GHR treatment did not attenuate the hippocampal inflammation. CONCLUSION: Although VSG was comparably effective at reducing body weight and improving glucose regulation as RYGB, VSG did not appear to confer an equal benefit on cognitive function and markers of inflammation.


Subject(s)
Caloric Restriction , Cognition Disorders/pathology , Gastrectomy , Gastric Bypass , Hippocampus/pathology , Inflammation/pathology , Weight Loss , Animals , Blood Glucose , Body Weight , Cognition Disorders/surgery , Disease Models, Animal , Gastrectomy/methods , Homeostasis , Inflammation/surgery , Male , Maze Learning , Rats , Rats, Long-Evans , Remission Induction
4.
Int J Obes (Lond) ; 37(2): 288-95, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22334194

ABSTRACT

OBJECTIVE: Diets high in fat are implicated in the development and maintenance of obesity, and obese individuals display greater preferences for high-fat foods than do their lean counterparts. Weight-reduction bariatric surgery is associated with changes in food choice. In particular, after Roux-en-Y gastric bypass (RYGB), humans and rodents select or prefer foods that are lower in fat content. We asked whether a bariatric surgical procedure limited to the stomach, vertical sleeve gastrectomy (VSG), causes a similar reduction of fat intake/preference. RESEARCH DESIGN AND METHODS: Rats received VSG or Sham surgery or remained surgically naïve, and were assessed for food preference using three diet-choice paradigms. Using progressive-ratio (PR) and conditioned taste aversion paradigms, we further asked whether surgically induced changes in food choice are secondary to changes in the reward value of food and/or to the formation of a food aversion. Finally, food choice was compared between VSG- and RYGB-operated rats. RESULTS: VSG rats decreased their intake of dietary fat, and shifted their preference toward lower caloric-density foods. This change in food choice was not associated with changes in motivated responding on a PR schedule for either a fat or a carbohydrate food reinforcer. When VSG and RYGB were compared directly, both procedures caused comparable changes in food choice. The conditioned taste aversion paradigm revealed that VSG rats form an aversion to an intra-gastric oil administration whereas RYGB rats do not. CONCLUSIONS: VSG and RYGB, two anatomically distinct bariatric procedures, produce similar changes in food choice.


Subject(s)
Dietary Fats/metabolism , Food Preferences , Gastric Bypass , Gastroplasty , Obesity/surgery , Animals , Body Weight , Choice Behavior , Energy Metabolism , Male , Physical Conditioning, Animal , Rats , Rats, Long-Evans , Reward , Taste
5.
Horm Behav ; 62(5): 598-604, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22982020

ABSTRACT

Ghrelin is an orexigenic hormone that regulates homeostatic and reward-related feeding behavior. Recent evidence indicates that acylation of ghrelin by the gut enzyme ghrelin O-acyl transferase (GOAT) is necessary to render ghrelin maximally active within its target tissues. Here we tested the hypothesis that GOAT activity modulates food motivation and food hedonics using behavioral pharmacology and mutant mice deficient for GOAT and the ghrelin receptor (GHSR). We evaluated operant responding following pharmacological administration of acyl-ghrelin and assessed the necessity of endogenous GOAT activity for operant responding in GOAT and GHSR-null mice. Hedonic-based feeding behavior also was examined in GOAT-KO and GHSR-null mice using a "Dessert Effect" protocol in which the intake of a palatable high fat diet "dessert" was assessed in calorically-sated mice. Pharmacological administration of acyl-ghrelin augmented operant responding; notably, this effect was dependent on intact GHSR signaling. GOAT-KO mice displayed attenuated operant responding and decreased hedonic feeding relative to controls. These behavioral results correlated with decreased expression of the orexin-1 receptor in reward-related brain regions in GOAT-KO mice. In summary, the ability of ghrelin to stimulate food motivation is dependent on intact GHSR signaling and modified by endogenous GOAT activity. Furthermore, GOAT activity is required for hedonic feeding behavior, an effect potentially mediated by forebrain orexin signaling. These data highlight the significance of the GOAT-ghrelin system for the mediation of food motivation and hedonic feeding.


Subject(s)
Acyltransferases/physiology , Feeding Behavior/physiology , Ghrelin/metabolism , Acylation/physiology , Acyltransferases/genetics , Acyltransferases/metabolism , Animals , Appetite Regulation/drug effects , Appetite Regulation/genetics , Appetite Regulation/physiology , Blood Glucose/drug effects , Blood Glucose/metabolism , Body Weight/drug effects , Body Weight/physiology , Feeding Behavior/drug effects , Ghrelin/blood , Ghrelin/pharmacology , Ghrelin/physiology , Intracellular Signaling Peptides and Proteins/blood , Intracellular Signaling Peptides and Proteins/metabolism , Male , Membrane Proteins , Mice , Mice, Inbred C57BL , Mice, Knockout , Motivation/drug effects , Motivation/genetics , Motivation/physiology , Neuropeptides/blood , Neuropeptides/metabolism , Orexins
6.
Neuroscience ; 210: 243-8, 2012 May 17.
Article in English | MEDLINE | ID: mdl-22433299

ABSTRACT

Data from our laboratory indicate that the orexin system is involved in the regulation of both conditioned and unconditioned responding for palatable foods. Anticipation of food rewards activates orexin receptor containing neurons within the paraventricular nucleus of the thalamus (PVT). The PVT regulates mesolimbic dopamine neurochemistry through direct connections with the nucleus accumbens and modulates the processing of cognitive-emotional information, suggesting that the PVT may represent a unique brain region with the capacity to mediate orexinergic effects on brain dopamine and behavior. Here, we tested the hypothesis that PVT orexin signaling mediates mesolimbic dopamine and reward-based feeding. To do this we used a behavioral pharmacological approach in tandem with central genetic manipulation of the orexin-1 receptor in the PVT. Data from these studies indicate that orexin-A action in the PVT increases dopamine levels in the nucleus accumbens. In addition, endogenous orexin signaling in the PVT mediates locomotor activity and hedonic feeding responses. Together these data highlight the PVT as a critical site capable of mediating orexin action on brain dopamine and reward-based feeding.


Subject(s)
Dopamine/metabolism , Feeding Behavior/physiology , Intracellular Signaling Peptides and Proteins/metabolism , Neuropeptides/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Signal Transduction/physiology , Animals , Immunohistochemistry , Male , Motor Activity/physiology , Orexins , RNA, Small Interfering , Rats , Rats, Long-Evans , Real-Time Polymerase Chain Reaction
7.
Neuroscience ; 171(4): 1032-40, 2010 Dec 29.
Article in English | MEDLINE | ID: mdl-20923696

ABSTRACT

Obesity is associated with increased risk of diabetes, cardiovascular disease and several types of cancers. The hypothalamus is a region of the brain critical in the regulation of body weight. One of the critical and best studied hypothalamic circuits is comprised of the melanocortinergic orexigenic agouti-related protein (AgRP) and anorexigenic α-melanocyte stimulating hormone (α-MSH) neurons. These neurons project axons to the same hypothalamic target neurons and balance each other's activity leading to body weight regulation. We previously showed that the brain proteoglycan syndecan-3 regulates feeding behavior and body weight, and syndecan-3 null (SDC-3(-/-)) mice are lean and obesity resistant. Here we show that the melanocortin agonist Melanotan II (MTII) potently suppresses food intake and activates the hypothalamic paraventricular nuclei (PVN) in SDC-3(-/-) mice based on c-fos immunoreactivity. Interestingly, we determined that the AgRP neuropeptide is reduced in the PVN of SDC-3(-/-) mice compared to wild type mice. In contrast, neuropeptide Y, coexpressed in the AgRP neuron, is not differentially expressed nor is the counteracting neuropeptide α-MSH. These findings are unprecedented and indicate that AgRP protein localization can be selectively regulated within the hypothalamus resulting in altered neuropeptide response and tone.


Subject(s)
Agouti-Related Protein/metabolism , Eating/genetics , Gene Expression Regulation/genetics , Signal Transduction/physiology , Syndecan-3/deficiency , Analysis of Variance , Animals , Body Weight/drug effects , Body Weight/genetics , Dose-Response Relationship, Drug , Eating/drug effects , Feeding Behavior/drug effects , Feeding Behavior/physiology , Food Deprivation/physiology , Gene Expression Regulation/drug effects , Male , Mice , Mice, Knockout , Neurons/drug effects , Neurons/metabolism , Neuropeptide Y/pharmacology , Paraventricular Hypothalamic Nucleus/cytology , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Time Factors , alpha-MSH/analogs & derivatives , alpha-MSH/pharmacology
8.
Neuroscience ; 167(1): 11-20, 2010 Apr 28.
Article in English | MEDLINE | ID: mdl-20149847

ABSTRACT

Consumption beyond homeostatic needs, referred to here as reward-based feeding behavior, is a central contributor to the current obesity epidemic worldwide. Importantly, reward-based feeding can be driven by palatability, the taste and texture of the food, as well as cues associated with the consumption of palatable foods. The hypothalamic orexin system regulates both diet preference and anticipation of food rewards making it a likely target to modulate reward-based feeding behavior. In the current manuscript we hypothesized that orexin signaling mediates food-motivated behaviors and reward-based feeding behavior. We further hypothesized that orexin neurons and targets of the orexin system become activated in response to cues associated with the consumption of palatable food. Data from these studies suggest that orexin signaling promotes progressive ratio responding for palatable foods while blockade of orexin signaling attenuates reward-based feeding of a high fat diet. In addition, cues linked to the consumption of chocolate, or the receipt of a daily meal, activate the orexin system and its target regions differentially. Collectively, these data suggest that orexin signaling mediates reward-based feeding behavior and, within specific target regions, may regulate cue-induced overconsumption of palatable foods.


Subject(s)
Feeding Behavior/physiology , Food , Intracellular Signaling Peptides and Proteins/metabolism , Motivation/physiology , Neurons/physiology , Neuropeptides/metabolism , Reward , Animals , Benzoxazoles/pharmacology , Brain/drug effects , Brain/physiology , Cacao , Central Nervous System Agents/pharmacology , Cues , Dietary Fats , Eating/drug effects , Eating/physiology , Feeding Behavior/drug effects , Male , Motivation/drug effects , Naphthyridines , Neurons/drug effects , Orexin Receptors , Orexins , Rats , Rats, Long-Evans , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/metabolism , Receptors, Neuropeptide/antagonists & inhibitors , Receptors, Neuropeptide/metabolism , Urea/analogs & derivatives , Urea/pharmacology
9.
Neuroscience ; 162(1): 23-30, 2009 Aug 04.
Article in English | MEDLINE | ID: mdl-19393296

ABSTRACT

Risk-taking behavior is a vital aspect mediating the formation of social structure in animals. Here, we utilized the visible burrow system (VBS), a model in which rats form dominance hierarchies, to test the hypothesis that dominant rats in the VBS are natural risk takers and display an increased motivational state after VBS exposure. In particular, we predicted that dominant rats would have attenuated anxiety-like behavior and augmented acquisition of operant responding for food reward relative to subordinate and controls. We further hypothesized that these behaviors would correlate with elevated mesocortical orexin signaling. Prior to burrow exposure, male Long-Evans rats were tested on the elevated plus maze (EPM), and subsequently exposed to the VBS for seven consecutive days. At the conclusion of burrow exposure body weight and plasma corticosterone were used to confirm social rank within each colony. Interestingly, rats that went on to become dominant in the VBS spent significantly more time in the open arms of the EPM prior to burrow exposure and displayed increased operant responding for food reward. This effect was present over a range of reinforcement schedules and also persisted for up to 1 month following VBS exposure. Moreover, dominant rats displayed increased orexin receptor mRNA in the medial prefrontal cortex (mPFC) relative to subordinate and control rats. These data support previous findings from our group and are consistent with the hypothesis that risk-taking behavior may precede dominance formation in social hierarchies.


Subject(s)
Feeding Behavior/psychology , Food , Motivation , Reward , Risk-Taking , Social Dominance , Animals , Body Weight , Conditioning, Operant/physiology , Corticosterone/blood , Exploratory Behavior/physiology , Feeding Behavior/physiology , Male , Maze Learning/physiology , Memory/physiology , Orexin Receptors , Prefrontal Cortex/metabolism , RNA, Messenger/metabolism , Random Allocation , Rats , Rats, Long-Evans , Receptors, G-Protein-Coupled/metabolism , Receptors, Neuropeptide/metabolism , Reinforcement Schedule
10.
J Clin Endocrinol Metab ; 92(2): 583-8, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17119001

ABSTRACT

BACKGROUND: Recent findings suggest that low plasma peptide YY (PYY) levels may contribute to diet-induced human obesity and justify PYY replacement therapy. Although the pharmacological value of PYY is controversial, further study of the secretion of the precursor PYY(1-36) and the pharmacologically active PYY(3-36) is indicated to determine the potential role in energy balance regulation. AIM: Our objective was to determine the effects of acute and chronic changes in human body weight on circulating levels of the putative satiety hormone peptide YY. DESIGN: Total plasma PYY levels (PYY(1-36) + PYY(3-36)) were measured in 66 lean, 18 anorectic, 63 obese, and 16 morbidly obese humans. In addition, total PYY was measured in 17 of the obese patients after weight loss and in the 18 anorectic patients after weight gain. Fasting PYY(3-36) levels were measured in 17 lean and 15 obese individuals. RESULTS: Fasting total plasma PYY levels were highest in patients with anorexia nervosa (80.9 +/- 12.9 pg/ml, P < 0.05) compared with lean (52.4 +/- 4.6 pg/ml), obese (43.9 +/- 3.8 pg/ml), or morbidly obese (45.6 +/- 11.2 pg/ml) subjects. In obese patients, weight loss of 5.4% was associated with a 30% decrease in fasting total PYY plasma levels. In anorectic patients, weight gain had no effect on fasting PYY. PYY(3-36) levels did not differ between lean (96.2 +/- 8.6 pg/ml) and obese (91.5 +/- 6.9 pg/ml) subjects. CONCLUSION: Our findings do not support a role for abnormal circulating PYY in human obesity. We conclude that circulating PYY levels in humans are significantly elevated in anorexia nervosa and, given the controversially discussed anorectic effect of PYY, could theoretically contribute to that syndrome.


Subject(s)
Anorexia/physiopathology , Body Weight/physiology , Obesity, Morbid/physiopathology , Peptide YY/blood , Satiety Response/physiology , Adult , Anorexia/metabolism , Energy Intake/physiology , Fasting/physiology , Female , Humans , Leptin/blood , Obesity, Morbid/metabolism , Peptide Fragments , Receptors, Cell Surface/blood , Receptors, Leptin , Weight Gain/physiology , Weight Loss/physiology
11.
Obes Rev ; 6(4): 307-22, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16246216

ABSTRACT

The neuropeptide Y (NPY)/peptide YY (PYY) system has been implicated in the physiology of obesity for several decades. More recently ignited enormous interest in PYY3-36, an endogenous Y2-receptor agonist, as a promising anti-obesity compound. Despite this interest, there have been remarkably few subsequent reports reproducing or extending the initial findings, while at the same time studies finding no anti-obesity effects have surfaced. Out of 41 different rodent studies conducted (in 16 independent labs worldwide), 33 (83%) were unable to reproduce the reported effects and obtained no change or sometimes increased food intake, despite use of the same experimental conditions (i.e. adaptation protocols, routes of drug administration and doses, rodent strains, diets, drug vendors, light cycles, room temperatures). Among studies by authors in the original study, procedural caveats are reported under which positive effects may be obtained. Currently, data speak against a sustained decrease in food intake, body fat, or body weight gain following PYY3-36 administration and make the previously suggested role of the hypothalamic melanocortin system unlikely as is the existence of PYY deficiency in human obesity. We review the studies that are in the public domain which support or challenge PYY3-36 as a potential anti-obesity target.


Subject(s)
Anti-Obesity Agents/pharmacology , Body Weight/drug effects , Eating/drug effects , Peptide YY/pharmacology , Animals , Behavior, Animal , Data Interpretation, Statistical , Dipeptidyl Peptidase 4/metabolism , Humans , Peptide Fragments , Peptide YY/administration & dosage , Receptors, Neuropeptide Y/agonists , Satiety Response/drug effects , Species Specificity , Stress, Physiological/physiopathology
12.
Nature ; 430(6996): 1 p following 165; discussion 2 p following 165, 2004 Jul 08.
Article in English | MEDLINE | ID: mdl-15243972

ABSTRACT

Batterham et al. report that the gut peptide hormone PYY3-36 decreases food intake and body-weight gain in rodents, a discovery that has been heralded as potentially offering a new therapy for obesity. However, we have been unable to replicate their results. Although the reasons for this discrepancy remain undetermined, an effective anti-obesity drug ultimately must produce its effects across a range of situations. The fact that the findings of Batterham et al. cannot easily be replicated calls into question the potential value of an anti-obesity approach that is based on administration of PYY3-36.


Subject(s)
Appetite Depressants/pharmacology , Appetite Regulation/drug effects , Feeding Behavior/drug effects , Peptide YY/pharmacology , Animals , Animals, Inbred Strains , Appetite/drug effects , Appetite/physiology , Appetite Depressants/therapeutic use , Behavior, Animal/drug effects , Body Weight/drug effects , Environment , Humans , Meta-Analysis as Topic , Mice , Obesity/drug therapy , Peptide Fragments , Peptide YY/administration & dosage , Peptide YY/blood , Peptide YY/therapeutic use , Rats , Reproducibility of Results , Stress, Physiological/complications , Stress, Physiological/physiopathology
13.
Int J Obes Relat Metab Disord ; 27(5): 550-6, 2003 May.
Article in English | MEDLINE | ID: mdl-12704398

ABSTRACT

BACKGROUND: The synthetic melanocortin (MC) agonist, melanotan-II (MTII), reduces food intake and body weight for hours to days after administration. One early report on the effect of MTII suggested that part of its anorexic action may be mediated by aversive consequences. In that experiment, MTII was found to support a mild conditioned taste aversion (CTA). OBJECTIVE: The present experiments replicate and extend those findings in two additional CTA paradigms to further characterize the aversive effects of MTII in rats. METHODS: Experiment 1 simultaneously assessed the ability of MTII to support CTA and reduce food intake, using a small oral infusion of a novel taste as the conditioned stimulus. Experiment 2 assessed the aversive consequences of chronic MTII administration. To accomplish this, we paired implantation of lithium chloride (LiCl)-, MTII- or saline-containing osmotic minipumps with a constantly available novel flavor. After 7 days, rats received a choice test between the minipump-paired flavor and a previously available neutral flavor. RESULTS: Rats with saline minipumps exhibited no preference for either flavor. By contrast, rats in both the LiCl and MTII minipump groups significantly preferred the neutral flavor, indicating the development of a CTA. Additionally, CTA produced by administration of MTII was found to be more resistant to extinction than that produced by LiCl. CONCLUSIONS: The reduction in food intake caused by MTII is accompanied by aversive consequences regardless of route of administration. These results present difficulties for the development of MCs-based therapies for obesity.


Subject(s)
Obesity/drug therapy , alpha-MSH/analogs & derivatives , alpha-MSH/adverse effects , Animals , Avoidance Learning , Dose-Response Relationship, Drug , Eating/drug effects , Male , Mice , Rats , Taste Disorders/chemically induced , alpha-MSH/administration & dosage
14.
Pharmacol Biochem Behav ; 69(3-4): 603-9, 2001.
Article in English | MEDLINE | ID: mdl-11509222

ABSTRACT

Much evidence suggests that the hypothalamic melanocortin (MC) system plays an important role in the control of food intake. However, investigations of the potential behavioral mechanisms have been limited to measures of aversion. The purpose of the present experiment was to assess whether other behavioral consequences of administration of MC peptides were similar to those produced by 0- or 24-h food deprivation, respectively. Rats were first trained while food deprived that a tone predicted the delivery of peanut oil. They then received exposure to oil under food deprivation, satiation, intra-third-cerebroventricular (i3vt) infusion of MTII (a potent MC agonist) or SHU-9119 (a potent MC antagonist). All rats were then tested during extinction for levels of responding to the tone under food satiation. Previous results demonstrated that sated exposure reduces subsequent test responding to the tone. During the present extinction test, rats that received sated exposure exhibited reduced responding to the tone, relative to rats that received deprived exposure. Unlike satiation, rats that received exposure after MTII exhibited continued high levels of responding to the tone. Further, rats that received SHU-9119 exhibited a small reduction in responding. These data suggest that MTII and SHU-9119 do not influence intake via the same mechanisms as hunger and food satiation, respectively.


Subject(s)
Appetitive Behavior/physiology , Hypothalamus/physiology , Receptors, Corticotropin/physiology , Animals , Appetitive Behavior/drug effects , Eating/drug effects , Eating/physiology , Hypothalamus/drug effects , Male , Melanocyte-Stimulating Hormones/pharmacology , Oligopeptides/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Corticotropin/agonists , Receptors, Corticotropin/antagonists & inhibitors , Receptors, Melanocortin , Third Ventricle/drug effects , Third Ventricle/physiology , alpha-MSH/analogs & derivatives
15.
Endocrinology ; 142(3): 1050-6, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11181518

ABSTRACT

Several lines of evidence substantiate the important role of the central nervous system melanocortin 3- and 4-receptor (MC3/4-R) system in the control of food intake and energy balance. Agouti-related peptide (AgRP), an endogenous antagonist of these receptors, produces a robust and unique pattern of increased food intake that lasts up to 7 days after a single injection. Little is known about brain regions that may mediate this powerful effect of AgRP on food intake. To this end we compared c-Fos-like immunoreactivity (c-FLI) in several brain sites of rats injected intracerebroventricularly with 1 nmol AgRP-(83--132) 2 and 24 h before death and compared c-FLI patterns to those induced by another potent orexigenic peptide, neuropeptide Y (NPY). Although both NPY and AgRP induced c-FLI in hypothalamic areas, AgRP also produced increased c-FLI in the accumbens shell and lateral septum. Although NPY elicited no changes in c-FLI 24 h after administration, AgRP induced c-FLI in the accumbens shell, nucleus of the solitary tract, central amygdala, and lateral hypothalamus. These results indicate that an NPY-like hypothalamic circuit mediates the short-term effects of AgRP, but that the unique sustained effect of AgRP on food intake involves a complex circuit of key extrahypothalamic reward and feeding regulatory nuclei.


Subject(s)
Brain/metabolism , Peptide Fragments/pharmacology , Proto-Oncogene Proteins c-fos/physiology , Agouti-Related Protein , Animals , Hypothalamus/metabolism , Immunohistochemistry , Injections, Intraventricular , Male , Neuropeptide Y/pharmacology , Rats , Rats, Long-Evans , Time Factors , Tissue Distribution/drug effects
16.
Am J Physiol Regul Integr Comp Physiol ; 280(3): R814-21, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11171662

ABSTRACT

Agouti-related peptide (AgRP) is a receptor antagonist of central nervous system (CNS) melanocortin receptors and appears to have an important role in the control of food intake since exogenous CNS administration in rats and overexpression in mice result in profound hyperphagia and weight gain. Given that AgRP is heavily colocalized with neuropeptide Y (NPY) and that orexigenic effects of NPY depend on activity at opioid receptors, we hypothesized that AgRP's food-intake effects are also mediated by opioid receptors. Subthreshold doses of the opioid receptor antagonist naloxone blocked AgRP-induced intake when given simultaneously but not 24 h after AgRP injection. Opioids not only influence food intake but food selection as well. Hence, we tested AgRP's effect to alter food choice between matched diets with differing dietary fat content. AgRP selectively enhanced intake of the high-fat but not the low-fat diet. Additionally, AgRP selectively increased chow intake in rats given ad libitum access to a 20% sucrose solution and standard rat chow. The current results indicate that AgRP influences not only caloric intake but food selection as well and that the early effects of AgRP depend critically on an interaction with opioid receptors.


Subject(s)
Eating/drug effects , Food Preferences/drug effects , Proteins/pharmacology , Receptors, Opioid/physiology , Agouti-Related Protein , Animals , Dietary Fats/administration & dosage , Dietary Sucrose/administration & dosage , Dose-Response Relationship, Drug , Energy Intake , Intercellular Signaling Peptides and Proteins , Male , Naloxone/administration & dosage , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Peptide Fragments/pharmacology , Rats , Rats, Long-Evans , Solutions
17.
Peptides ; 21(3): 345-51, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10793215

ABSTRACT

Corticotropin-releasing hormone (CRH) is a potent regulator of the hypothalamic-pituitary-adrenal axis, and reduces food intake when administered into the third cerebral ventricle (i3vt). However, CRH also promotes conditioned taste aversion (CTA) learning which indicates that its anorectic effects are accompanied by aversive consequences that would reduce food intake independently of energy regulation. Urocortin (Ucn) is a closely related mammalian peptide that binds to both identified CRH receptor subtypes and also reduces food intake when administered i3vt. The present experiments compared the aversive consequences of i3vt administration of CRH and Ucn at doses that produced comparable decrements in food intake. Experiment 1 found that 1.0 microg Ucn and 2.0 microg CRH produced similar reductions in food intake. Experiment 2 demonstrated that, at these doses, CRH but not Ucn promoted robust and reliable CTA learning. A third experiment showed comparable increased c-Fos-like immunoreactivity after Ucn and CRH in forebrain and hindbrain structures associated with food intake. It is concluded that Ucn, at doses that reduce food intake to levels like that observed after administration of CRH, do not produce similarly aversive consequences.


Subject(s)
Avoidance Learning/drug effects , Cerebral Ventricles/physiology , Corticotropin-Releasing Hormone/pharmacology , Energy Intake/drug effects , Genes, fos , Taste , Amygdala/drug effects , Amygdala/metabolism , Animals , Cerebral Ventricles/drug effects , Corticotropin-Releasing Hormone/administration & dosage , Feeding Behavior/drug effects , Gene Expression Regulation/drug effects , Injections, Intraventricular , Male , Rats , Rats, Long-Evans , Reaction Time , Saccharin , Solitary Nucleus/drug effects , Solitary Nucleus/metabolism , Urocortins
18.
J Neurosci ; 20(9): 3442-8, 2000 May 01.
Article in English | MEDLINE | ID: mdl-10777807

ABSTRACT

Studies using nonselective agonists and antagonists of melanocortin-3 receptor (MC3R) and MC4R point to the importance of the CNS melanocortin system in the control of food intake. We describe here a novel compound that is highly selective as an agonist at the MC4 receptor but has minimal activity at the MC3 receptor. When administered centrally to rats, this selective agonist increased Fos-like immunoreactivity in the paraventricular nucleus, central nucleus of the amygdala, nucleus of the solitary tract, and area postrema, a pattern of neuronal activation that is similar to that induced by a nonselective MC3/4R agonist. Additionally, it suppresses food intake when administered centrally to rats or peripherally to db/db mice that lack functional leptin receptors via a mechanism that is not accompanied by illness or other nonspecific effects. Conversely, a related compound that is a selective MC4R antagonist potently increased food intake when administered centrally in rats. These results support the hypothesis that the brain MC4R is intimately involved in the control of food intake and body weight and provide evidence that selective activation of MC4R causes anorexia that is not secondary to aversive effects.


Subject(s)
Eating/drug effects , Feeding Behavior/drug effects , Receptor, Melanocortin, Type 4/agonists , Animals , Eating/physiology , Feeding Behavior/physiology , Genes, fos/drug effects , Genes, fos/physiology , Male , Mice , Mice, Obese , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/metabolism , Rats , Rats, Long-Evans , Receptor, Melanocortin, Type 4/antagonists & inhibitors
19.
Behav Neurosci ; 113(6): 1233-41, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10636301

ABSTRACT

T. L. Davidson, A. M. Altizer, S. C. Benoit, E. K. Walls, and T. L. Powley (1997) reported that rats show facilitated responding to conditioned stimuli (CSs) that predict oil, after administration of the lipoprivic agent, Na-2-mercaptoacetate (MA). This facilitation was blocked by vagal deafferentation. The present article extends that investigation to another structure, the amygdala central nucleus (CN). The CN receives inputs from dorsal vagal nuclei, and neurotoxic lesions of this nucleus are reported to abolish feeding in response to lipoprivic challenges. In Experiment 1, rats with ibotenic acid (IBO) lesions of the CN failed to show enhanced appetitive responding during oil-predicting CSs after administration of MA. Experiment 2 used a conditioned taste-aversion procedure to establish that rats with IBO lesions of the CN were able to discriminate the tastes of sucrose and peanut oil and had intact CS-US representations. It is concluded that the amygdala CN is a necessary structure for the detection of lipoprivic challenges.


Subject(s)
Amygdala/physiology , Appetitive Behavior/physiology , Avoidance Learning/physiology , Conditioning, Classical/physiology , Amygdala/drug effects , Animals , Conditioning, Classical/drug effects , Excitatory Amino Acid Agonists/administration & dosage , Excitatory Amino Acid Agonists/pharmacology , Ibotenic Acid/administration & dosage , Ibotenic Acid/pharmacology , Male , Microinjections , Rats , Rats, Sprague-Dawley , Thioglycolates/administration & dosage , Thioglycolates/pharmacology
20.
Behav Neurosci ; 111(5): 1014-30, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9383521

ABSTRACT

Experiment 1 used Pavlovian conditioning procedures to show that rats formed distinct memorial representations of 2 (peanut oil and sucrose pellets) unconditioned stimuli (USs) that could be activated by 2 different conditioned stimuli (CSs). After training in Experiment 2, rats injected with the lipid antimetabolite Na-2-mercaptoacetate (MA) responded more to the CS for oil than to the CS for sucrose. This pattern was not shown by rats that received isotonic saline or systemic 2-deoxy-d-glucose (a glucose antimetabolite). By contrast, intracerebroventricular infusion of the glucose antimetabolite 5-thioglucose selectively promoted responding to the CS for sucrose (Experiment 4). Thus, lipoprivic and glucoprivic treatments selectively promoted the activation of the memories of fat and carbohydrate USs, respectively. In Experiment 3, the capacity of MA to augment responding to a CS for oil was abolished for rats that received subdiaphragmatic vagal deafferentation. This indicates that the capacity of lipoprivic signals to selectively activate the representations of fat USs may depend on vagal afferent fibers.


Subject(s)
Brain/physiology , Conditioning, Classical/physiology , Energy Metabolism/physiology , Feeding Behavior/physiology , Mental Recall/physiology , Animals , Antimetabolites/pharmacology , Association Learning/drug effects , Association Learning/physiology , Avoidance Learning/drug effects , Avoidance Learning/physiology , Blood Glucose/metabolism , Brain/drug effects , Conditioning, Classical/drug effects , Deoxyglucose/pharmacology , Energy Metabolism/drug effects , Fatty Acids, Nonesterified/blood , Feeding Behavior/drug effects , Food Preferences/drug effects , Food Preferences/physiology , Injections, Intraventricular , Male , Mental Recall/drug effects , Motivation , Peanut Oil , Plant Oils/administration & dosage , Rats , Rats, Sprague-Dawley , Sucrose/administration & dosage , Taste/drug effects , Taste/physiology , Thioglycolates/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...