Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Sci Rep ; 7: 44392, 2017 03 10.
Article in English | MEDLINE | ID: mdl-28281681

ABSTRACT

Obstructive sleep apnea (OSA) is characterized by recurrent upper airway obstruction during sleep. OSA leads to high cardiovascular morbidity and mortality. The pathogenesis of OSA has been linked to a defect in neuromuscular control of the pharynx. There is no effective pharmacotherapy for OSA. The objective of this study was to determine whether upper airway patency can be improved using chemogenetic approach by deploying designer receptors exclusively activated by designer drug (DREADD) in the hypoglossal motorneurons. DREADD (rAAV5-hSyn-hM3(Gq)-mCherry) and control virus (rAAV5-hSyn-EGFP) were stereotactically administered to the hypoglossal nucleus of C57BL/6J mice. In 6-8 weeks genioglossus EMG and dynamic MRI of the upper airway were performed before and after administration of the DREADD ligand clozapine-N-oxide (CNO) or vehicle (saline). In DREADD-treated mice, CNO activated the genioglossus muscle and markedly dilated the pharynx, whereas saline had no effect. Control virus treated mice showed no effect of CNO. Our results suggest that chemogenetic approach can be considered as a treatment option for OSA and other motorneuron disorders.


Subject(s)
Antipsychotic Agents/pharmacology , Clozapine/analogs & derivatives , Genetic Vectors/administration & dosage , Hypoglossal Nerve/drug effects , Pharynx/drug effects , Sleep Apnea, Obstructive/therapy , Animals , Clozapine/pharmacology , Dependovirus/genetics , Dependovirus/metabolism , Disease Models, Animal , Electromyography , Genes, Reporter , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hypoglossal Nerve/metabolism , Hypoglossal Nerve/physiopathology , Injections, Intraventricular , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Magnetic Resonance Imaging , Male , Mice , Mice, Inbred C57BL , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Pharynx/diagnostic imaging , Pharynx/innervation , Pharynx/metabolism , Sleep Apnea, Obstructive/diagnostic imaging , Sleep Apnea, Obstructive/metabolism , Sleep Apnea, Obstructive/physiopathology , Stereotaxic Techniques , Red Fluorescent Protein
2.
Gastroenterol Clin North Am ; 45(4): 581-599, 2016 12.
Article in English | MEDLINE | ID: mdl-27837774

ABSTRACT

Multiple physiologic and neural systems contribute to the controls over what and how much we eat. These systems include signaling involved in the detection and signaling of nutrient availability, signals arising from consumed nutrients that provide feedback information during a meal to induce satiation, and signals related to the rewarding properties of eating. Each of these has a separate neural representation, but important interactions among these systems are critical to the overall controls of food intake.


Subject(s)
Appetite/physiology , Eating/physiology , Feeding Behavior/physiology , Nervous System Physiological Phenomena , Satiation/physiology , Eating/psychology , Feeding Behavior/psychology , Food , Humans , Reward , Signal Transduction
3.
Sleep ; 39(5): 1097-106, 2016 May 01.
Article in English | MEDLINE | ID: mdl-26951402

ABSTRACT

STUDY OBJECTIVES: Obesity hypoventilation and obstructive sleep apnea are common complications of obesity linked to defects in respiratory pump and upper airway neural control. Leptin-deficient ob/ob mice have impaired ventilatory control and inspiratory flow limitation during sleep, which are both reversed with leptin. We aimed to localize central nervous system (CNS) site(s) of leptin action on respiratory and upper airway neuroventilatory control. METHODS: We localized the effect of leptin to medulla versus hypothalamus by administering intracerbroventricular leptin (10 µg/2 µL) versus vehicle to the lateral (n = 14) versus fourth ventricle (n = 11) of ob/ob mice followed by polysomnographic recording. Analyses were stratified for effects on respiratory (nonflow-limited breaths) and upper airway (inspiratory flow limitation) functions. CNS loci were identified by (1) leptin-induced signal transducer and activator of transcription 3 (STAT3) phosphorylation and (2) projections of respiratory and upper airway motoneurons with a retrograde transsynaptic tracer (pseudorabies virus). RESULTS: Both routes of leptin administration increased minute ventilation during nonflow-limited breathing in sleep. Phrenic motoneurons were synaptically coupled to the nucleus of the solitary tract, which also showed STAT3 phosphorylation, but not to the hypothalamus. Inspiratory flow limitation and obstructive hypopneas were attenuated by leptin administration to the lateral but not to the fourth cerebral ventricle. Upper airway motoneurons were synaptically coupled with the dorsomedial hypothalamus, which exhibited STAT3 phosphorylation. CONCLUSIONS: Leptin relieves upper airway obstruction in sleep apnea by activating the forebrain, possibly in the dorsomedial hypothalamus. In contrast, leptin upregulates ventilatory control through hindbrain sites of action, possibly in the nucleus of the solitary tract.


Subject(s)
Leptin/pharmacology , Respiration/drug effects , Respiratory System/drug effects , Sleep/drug effects , Sleep/physiology , Animals , Hypothalamus/cytology , Hypothalamus/drug effects , Hypothalamus/physiology , Hypoventilation/complications , Hypoventilation/physiopathology , Leptin/administration & dosage , Leptin/deficiency , Male , Mice , Motor Neurons/drug effects , Obesity/complications , Obesity/physiopathology , Phosphorylation/drug effects , Polysomnography , Respiratory System/innervation , STAT3 Transcription Factor/metabolism , Sleep Apnea, Obstructive/complications , Sleep Apnea, Obstructive/physiopathology , Solitary Nucleus/cytology , Solitary Nucleus/drug effects , Solitary Nucleus/physiology
4.
Drug Des Devel Ther ; 9: 1867-75, 2015.
Article in English | MEDLINE | ID: mdl-25848222

ABSTRACT

The prevalence of obesity worldwide has nearly doubled since 1980 with current estimates of 2.1 billion in 2013. Overweight and obesity lead to numerous adverse conditions including type 2 diabetes, cardiovascular disease, stroke, and certain cancers. The worldwide spread of obesity and associated comorbidities not only threatens quality of life but also presents a significant economic burden. While bariatric surgery has proven to be a viable treatment option for the morbidly obese, there is clearly a need for less invasive alternatives. Recent research has suggested that long-acting analogs of the gut hormone, glucagon-like peptide 1 (GLP-1), may have potential as an antiobesity treatment. The GLP-1 receptor agonist, liraglutide (trade name Saxenda), was recently approved by the US Food and Drug Administration as an obesity treatment option and shown in clinical trials to be effective in reducing and sustaining body weight loss. This review presents the basis for GLP-1-based therapies with a specific focus on animal and human studies examining liraglutide's effects on food intake and body weight.


Subject(s)
Liraglutide/therapeutic use , Obesity/drug therapy , Animals , Glucagon-Like Peptide-1 Receptor/agonists , Humans , Liraglutide/administration & dosage
5.
Hum Mol Genet ; 23(23): 6302-17, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25035419

ABSTRACT

Huntington's disease (HD) is a neurodegenerative disorder caused by a CAG trinucleotide repeat expansion in the huntingtin (HTT) gene. Disease pathogenesis derives, at least in part, from the long polyglutamine tract encoded by mutant HTT. Therefore, considerable effort has been dedicated to the development of therapeutic strategies that significantly reduce the expression of the mutant HTT protein. Antisense oligonucleotides (ASOs) targeted to the CAG repeat region of HTT transcripts have been of particular interest due to their potential capacity to discriminate between normal and mutant HTT transcripts. Here, we focus on phosphorodiamidate morpholino oligomers (PMOs), ASOs that are especially stable, highly soluble and non-toxic. We designed three PMOs to selectively target expanded CAG repeat tracts (CTG22, CTG25 and CTG28), and two PMOs to selectively target sequences flanking the HTT CAG repeat (HTTex1a and HTTex1b). In HD patient-derived fibroblasts with expanded alleles containing 44, 77 or 109 CAG repeats, HTTex1a and HTTex1b were effective in suppressing the expression of mutant and non-mutant transcripts. CTGn PMOs also suppressed HTT expression, with the extent of suppression and the specificity for mutant transcripts dependent on the length of the targeted CAG repeat and on the CTG repeat length and concentration of the PMO. PMO CTG25 reduced HTT-induced cytotoxicity in vitro and suppressed mutant HTT expression in vivo in the N171-82Q transgenic mouse model. Finally, CTG28 reduced mutant HTT expression and improved the phenotype of Hdh(Q7/Q150) knock-in HD mice. These data demonstrate the potential of PMOs as an approach to suppressing the expression of mutant HTT.


Subject(s)
Morpholinos/pharmacology , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Oligonucleotides, Antisense/pharmacology , Animals , Base Sequence , Cells, Cultured , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression , Gene Knock-In Techniques , Humans , Huntingtin Protein , Huntington Disease/genetics , Mice , Mice, Transgenic , Morpholinos/chemistry , Mutation , Nerve Tissue Proteins/genetics , Neurons/metabolism , Oligonucleotides, Antisense/chemistry , RNA, Messenger/metabolism , Trinucleotide Repeat Expansion
6.
Physiol Behav ; 105(3): 720-6, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22005165

ABSTRACT

Endogenous cannabinoid signaling, mediated predominately by CB1 receptor activation, is involved in food intake control and body weight regulation. Despite advances in determining the role of the CB1 receptor in obesity, its involvement in the driven nature of eating pathologies has received little attention. The present study examined CB1 receptor alterations as a consequence of dietary-induced binge eating in female Sprague Dawley rats. Four control groups were used to control for calorie restriction and highly palatable food variables characterizing this behavioral model. All groups were kept on their respective feeding schedules for 6-weeks and were given a uniform 33% calorie restriction (~22 h food deprivation) prior to sacrifice. Our findings indicate that regional CB1 mRNA and density were influenced by dietary conditions, but were not specific to the dietary-induced binge eating paradigm used. An increase of approximately 50% (compared with naive controls) in CB1 receptor mRNA levels in the nucleus of the solitary tract as measured by in situ hybridization was found in animals receiving continuous access to a highly palatable food (i.e., vegetable shortening with 10% sucrose). This group also had a significant increase in body weight and adiposity. An approximate 20% reduction in CB1 mRNA was observed in the cingulate cortex (areas 1 and 2) in animals exposed to an intermittent schedule of feeding, compared with groups that had ad libitum feeding schedules (i.e., continuous access and naive controls). Receptor density as measured by [(3)H]CP55,940 autoradiography, was reduced by approximately 30% in the nucleus accumbens shell region in groups receiving repeated access to the highly palatable food. Taken together, these findings indicate that dietary conditions can differentially influence CB1 receptors in forebrain and hindbrain regions.


Subject(s)
Brain/metabolism , Diet , Food Preferences/physiology , Gene Expression Regulation/physiology , Receptor, Cannabinoid, CB1/metabolism , Analysis of Variance , Animals , Autoradiography , Body Weight/drug effects , Body Weight/physiology , Bulimia/etiology , Bulimia/metabolism , Cannabinoids/pharmacokinetics , Cyclohexanols/pharmacokinetics , Disease Models, Animal , Eating/drug effects , Eating/physiology , Female , Food Deprivation/physiology , Gene Expression Regulation/drug effects , Genes/drug effects , Nodose Ganglion/metabolism , Protein Binding/drug effects , Protein Binding/physiology , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Cannabinoid, CB1/genetics , Tritium/pharmacokinetics
7.
Curr Opin Endocrinol Diabetes Obes ; 19(1): 13-8, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22157396

ABSTRACT

PURPOSE OF REVIEW: This review focuses on recent advances in understanding the multiple roles of gastrointestinal peptides in the control of food intake and body weight with specific emphasis on ghrelin, amylin and glucagon-like peptide 1. RECENT FINDINGS: Recent studies support a role for ghrelin, amylin and glucagon-like peptide 1 in short-term and long-term effects on food intake and body weight. Apart from contributing to energy homeostasis, ghrelin's participation in reward and sensory processing has been the focus of much recent work. New findings on amylin's effects on food intake and energy balance provide further support for its role in meal-related food intake and suggest that it may also function as an adiposity signal. New investigations on the role of central and peripheral glucagon-like peptide 1 receptors in mediating the anorexic effects of glucagon-like peptide 1 have suggested that they differentially contribute to short-term and long term effects on food intake. SUMMARY: Gastrointestinal peptides can influence food intake through mechanisms that involve short-term meal-related effects or through activation of central pathways involved in energy balance. An appreciation of the multiple actions of gastrointestinal peptides on food intake will aid in developing new strategies for weight management.


Subject(s)
Appetite Depressants/pharmacology , Ghrelin/metabolism , Glucagon-Like Peptide 1/metabolism , Islet Amyloid Polypeptide/metabolism , Obesity/drug therapy , Animals , Appetite Regulation , Body Weight , Eating/drug effects , Energy Metabolism/drug effects , Feeding Behavior , Ghrelin/pharmacology , Glucagon-Like Peptide 1/pharmacology , Humans , Islet Amyloid Polypeptide/pharmacology , Mice , Rats
8.
Physiol Behav ; 103(1): 21-4, 2011 Apr 18.
Article in English | MEDLINE | ID: mdl-21110992

ABSTRACT

Signaling from energy stores provides feedback on overall nutrient availability to influence food intake. Beginning with seminal studies by Woods and colleagues identifying insulin as an adiposity signal, it has become clear that such factors affect food intake by modulating the efficacy of within meal feedback satiety signals. More recent work with leptin has revealed actions of the hormone in modulating the efficacy of multiple gut feedback signals, identified the dorsal hindbrain as a site of signal integration and suggested both local and descending hypothalamic to hindbrain actions in mediating these effects. The original work by Woods and colleagues provided the necessary experimental paradigms for these advances.


Subject(s)
Adiposity/physiology , Eating/physiology , Signal Transduction/physiology , Adiposity/drug effects , Animals , Eating/drug effects , Humans , Insulin/metabolism , Insulin/pharmacology , Leptin/metabolism , Leptin/pharmacology , Signal Transduction/drug effects
10.
J Pharmacol Exp Ther ; 334(3): 682-92, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20498254

ABSTRACT

Fusion proteins made up of glucagon-like peptide 1 (GLP-1) and exendin-4 (EX-4) fused to a nonglycosylated form of human transferrin (GLP-1-Tf or EX-4-Tf) were produced and characterized. GLP-1-Tf activated the GLP-1 receptor, was resistant to inactivation by peptidases, and had a half-life of approximately 2 days, compared with 1 to 2 min for native GLP-1. GLP-1-Tf retained the acute, glucose-dependent insulin-secretory properties of native GLP-1 in diabetic animals and had a profound effect on proliferation of pancreatic beta-cells. In addition, Tf and the fusion proteins did not cross the blood-brain-barrier but still reduced food intake after peripheral administration. EX-4-Tf proved to be as effective as EX-4 but had longer lived effects on blood glucose and food intake. This novel transferrin fusion technology could improve the pharmacology of various peptides.


Subject(s)
Hypoglycemic Agents/pharmacokinetics , Insulin/metabolism , Protein Engineering , Transferrin/genetics , Animals , Blood Glucose/metabolism , CHO Cells , Cell Proliferation/drug effects , Cricetinae , Cricetulus , Diabetes Mellitus/drug therapy , Diabetes Mellitus/genetics , Diabetes Mellitus/metabolism , Dipeptidyl Peptidase 4/metabolism , Eating/drug effects , Enzyme-Linked Immunosorbent Assay , Genes, fos/drug effects , Glucagon-Like Peptide 1/genetics , Glucagon-Like Peptide-1 Receptor , Half-Life , Humans , In Vitro Techniques , Insulin-Secreting Cells/drug effects , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Male , Mice , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley , Receptors, Glucagon/agonists , Recombinant Fusion Proteins , Saccharomyces cerevisiae/metabolism
11.
Endocrinology ; 150(2): 672-8, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18818295

ABSTRACT

Gastrin-releasing peptide (GRP) is a bombesin-like peptide widely distributed in the gastrointestinal tract and central nervous system. In the brain, GRP mRNA is located in the hypothalamic paraventricular nucleus (PVN), a region that receives neural input from the arcuate nucleus and plays a critical role in food intake and energy balance. Because GRP neurons are localized in the vicinity of projection sites in the PVN for peptides that participate in energy homeostasis, we investigated whether GRP mRNA expression in the PVN may be sensitive to challenges imposed by either 38 h food deprivation or stimulation of the melanocortin system by the melanocortin 3/4 receptor agonist, melanotan II (MTII). We found that food deprivation significantly decreased GRP mRNA expression, whereas lateral ventricular MTII administration increased GRP mRNA expression in ad libitum-fed rats 4 h after administration. Furthermore, administration of MTII at a dose that reduces 24 h food intake and body weight prevented the decrease in GRP mRNA expression observed in animals that were pair fed to the amount of food consumed by those injected with MTII. These results demonstrate that food deprivation and stimulation of the melanocortin system produce opposing changes in GRP gene expression in the PVN, suggesting that GRP-containing neurons in the PVN may be part of the hypothalamic signaling pathway controlling food intake and energy balance.


Subject(s)
Food Deprivation/physiology , Gastrin-Releasing Peptide/genetics , Gene Expression Regulation , Paraventricular Hypothalamic Nucleus/metabolism , Peptides, Cyclic/pharmacology , Receptors, Melanocortin/agonists , alpha-MSH/analogs & derivatives , Animals , Appetite Regulation/drug effects , Appetite Regulation/genetics , Energy Metabolism/drug effects , Energy Metabolism/genetics , Gastrin-Releasing Peptide/metabolism , Gene Expression Regulation/drug effects , Homeostasis/drug effects , Homeostasis/genetics , Male , Paraventricular Hypothalamic Nucleus/drug effects , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , alpha-MSH/pharmacology
12.
Endocrinology ; 149(3): 971-8, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18039774

ABSTRACT

Mice with a targeted disruption of bombesin receptor subtype-3 (BRS-3 KO) develop hyperphagia, obesity, hypertension, and impaired glucose metabolism. However, the factors contributing to their phenotype have not been clearly established. To determine whether their obesity is a result of increased food intake or a defect in energy regulation, we matched the caloric intake of BRS-3 KO mice to wild-type (WT) ad libitum (ad lib)-fed controls over 21 wk. Although BRS-3 KO ad lib-fed mice were 29% heavier, the body weights of BRS-3 KO pair-fed mice did not differ from WT ad lib-fed mice. Pair-feeding BRS-3 KO mice normalized plasma insulin but failed to completely reverse increased adiposity and leptin levels. Hyperphagia in ad lib-fed KO mice was due to an increase in meal size without a compensatory decrease in meal frequency resulting in an increase in total daily food intake. An examination of neuropeptide Y, proopiomelanocortin, and agouti-related peptide gene expression in the arcuate nucleus revealed that BRS-3 KO mice have some deficits in their response to energy regulatory signals. An evaluation of the satiety effects of cholecystokinin, bombesin, and gastrin-releasing peptide found no differences in feeding suppression by these peptides. We conclude that hyperphagia is a major factor leading to increased body weight and hyperinsulinemia in BRS-3 KO mice. However, our finding that pair-feeding did not completely normalize fat distribution and plasma leptin levels suggests there is also a metabolic dysregulation that may contribute to, or sustain, their obese phenotype.


Subject(s)
Hyperphagia/complications , Hyperphagia/metabolism , Obesity/etiology , Obesity/metabolism , Receptors, Bombesin/metabolism , Adiposity/drug effects , Adiposity/physiology , Animals , Body Weight/drug effects , Body Weight/physiology , Bombesin/pharmacology , Cholecystokinin/pharmacology , Eating/drug effects , Eating/physiology , Energy Metabolism/drug effects , Energy Metabolism/physiology , Gastrin-Releasing Peptide/pharmacology , Glucose/metabolism , Hyperinsulinism/etiology , Hypothalamus/drug effects , Hypothalamus/metabolism , Insulin/blood , Leptin/blood , Male , Mice , Mice, Knockout , Satiation/physiology , Weight Gain
13.
Proc Natl Acad Sci U S A ; 104(44): 17358-63, 2007 Oct 30.
Article in English | MEDLINE | ID: mdl-17956983

ABSTRACT

Hypothalamic fatty acid metabolism has recently been implicated in the controls of food intake and energy homeostasis. We report that intracerebroventricular (ICV) injection of leptin, concomitant with inhibiting AMP-activated kinase (AMPK), activates acetyl-CoA carboxylase (ACC), the key regulatory enzyme in fatty acid biosynthesis, in the arcuate nucleus (Arc) and paraventricular nucleus (PVN) in the hypothalamus. Arc overexpression of constitutively active AMPK prevents the Arc ACC activation in response to ICV leptin, supporting the hypothesis that AMPK lies upstream of ACC in leptin's Arc intracellular signaling pathway. Inhibiting hypothalamic ACC with 5-tetradecyloxy-2-furoic acid, a specific ACC inhibitor, blocks leptin-mediated decreases in food intake, body weight, and mRNA level of the orexigenic neuropeptide NPY. These results show that hypothalamic ACC activation makes an important contribution to leptin's anorectic effects. Furthermore, we find that ICV leptin up-regulates the level of malonyl-CoA (the intermediate of fatty acid biosynthesis) specifically in the Arc and increases the level of palmitoyl-CoA (a major product of fatty acid biosynthesis) specifically in the PVN. The rises of both levels are blocked by 5-tetradecyloxy-2-furoic acid along with the blockade of leptin-mediated hypophagia. These data suggest malonyl-CoA as a downstream mediator of ACC in leptin's signaling pathway in the Arc and imply that palmitoyl-CoA, instead of malonyl-CoA, could be an effector in relaying ACC signaling in the PVN. Together, these findings highlight site-specific impacts of hypothalamic ACC activation in leptin's anorectic signaling cascade.


Subject(s)
Acetyl-CoA Carboxylase/metabolism , Feeding Behavior/drug effects , Hypothalamus/drug effects , Hypothalamus/enzymology , Leptin/pharmacology , AMP-Activated Protein Kinases , Acetyl-CoA Carboxylase/antagonists & inhibitors , Animals , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Male , Multienzyme Complexes/antagonists & inhibitors , Multienzyme Complexes/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Rats , Rats, Sprague-Dawley
15.
Physiol Behav ; 91(1): 36-41, 2007 May 16.
Article in English | MEDLINE | ID: mdl-17343884

ABSTRACT

Peripheral administration of bombesin (BN) and the related mammalian peptides, gastrin-releasing peptide (GRP) and neuromedin B (NMB), suppress food intake in rats. To examine whether all BN-like peptides utilize the same neural pathways to reduce feeding, rats were treated on postnatal day 2 with the injection vehicle or capsaicin, a neurotoxin that damages a subset of visceral afferent fibers. When rats reached adulthood, we compared the ability of a dose range of systemically administered BN, GRP18-27 and NMB to reduce intake of a 0.5 kcal/ml glucose solution in a short-term feeding test. Our results demonstrate that capsaicin treatment abolished or attenuated the suppression of glucose intake produced by BN and NMB but had no effect on the ability of GRP to reduce feeding. These results suggest that different neural substrates underlie the anorexic effects of peripherally administered BN-like peptides.


Subject(s)
Bombesin/pharmacology , Capsaicin/pharmacology , Eating/drug effects , Animals , Animals, Newborn , Dose-Response Relationship, Drug , Glucose/pharmacology , Male , Neurokinin B/analogs & derivatives , Neurokinin B/pharmacology , Peptide Fragments/pharmacology , Rats , Rats, Sprague-Dawley , Reinforcement Schedule
16.
Physiol Behav ; 89(4): 511-6, 2006 Nov 30.
Article in English | MEDLINE | ID: mdl-16735044

ABSTRACT

Leptin reduces food intake through a specific effect on meal size. Investigations into how this within meal effect of leptin is mediated have demonstrated that leptin increases the ability of within meal inhibitory feedback signaling to limit intake and activate neurons within the nucleus of the solitary tract (NTS). Leptin's effects on neural activation can be demonstrated both as an increase in c-fos activation and as increase in electrophysiological activity in response to peripheral stimuli. Leptin can exert these effects through interactions at hypothalamic sites and activation of a descending pathway. NPY has opposite effect suggesting a role for reduced NPY signaling in the actions of leptin. Forebrain ventricular administration of a melanocortin agonist does not mimic the actions of leptin. As well as modulating within meal signaling through a descending pathway leptin, NPY and melanocortins could work directly at hindbrain integrative sites suggesting the possibility of distributed controls of meal size by anorexigenic and orexigenic signaling.


Subject(s)
Appetite Regulation/physiology , Feeding Behavior/physiology , Leptin/physiology , Satiation/physiology , Solitary Nucleus/physiology , Animals , Humans , Hypothalamus/physiology , Neural Pathways/physiology , Neurons/physiology , Solitary Nucleus/cytology
17.
Am J Physiol Regul Integr Comp Physiol ; 289(2): R473-R477, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15860644

ABSTRACT

Leptin amplifies feeding inhibition and neural activation produced by either cholecystokinin or intragastric preloads, suggesting that leptin may increase the efficacy of gastrointestinal meal-related signals. To determine whether leptin would similarly potentiate the feeding inhibitory actions of another putative satiety peptide, we evaluated the effects of third ventricular leptin administration on food intake and c-Fos activation in response to systemically administered bombesin (BN). Leptin (3.5 microg) was administered 1 h before either 0.9% saline or BN (0.32 and 1.0 nmol/kg) followed by 30-min access to Ensure liquid diet. Although neither leptin nor 0.32 nmol/kg BN alone suppressed Ensure intake, the combination reduced intake by 28%. The higher BN dose (1.0 nmol/kg) produced a significant suppression by itself but was further enhanced in the presence of leptin. Consistent with the behavioral results, c-Fos activation in the nucleus of the solitary tract was increased by combined dosages of leptin and 0.32 nmol/kg BN beyond the individual response to either peptide. In the presence of leptin, BN produced a 3.4- to 5.2-fold increase in the number of c-Fos-positive cells in the nucleus of the solitary tract compared with when BN was given alone. These data provide further support for the hypothesis that the effect of leptin on food intake may be mediated, in part, by modulating meal-related satiety signals.


Subject(s)
Bombesin/pharmacology , Eating/drug effects , Leptin/pharmacology , Proto-Oncogene Proteins c-fos/metabolism , Rhombencephalon/drug effects , Rhombencephalon/metabolism , Animals , Area Postrema/metabolism , Drug Synergism , Injections, Intraperitoneal , Injections, Intraventricular , Leptin/administration & dosage , Male , Rats , Rats, Sprague-Dawley , Satiety Response/drug effects , Solitary Nucleus/metabolism
18.
Discov Med ; 5(29): 467-71, 2005 Oct.
Article in English | MEDLINE | ID: mdl-20704844

ABSTRACT

Extract: During and following a meal, ingested nutrients come into contact with multiple sites in the gastrointestinal tract that have the potential to alter peptide and neural signaling. Such signals can serve as feedback mediators influencing current or subsequent food intake. Ingested nutrients accumulate within the stomach, activating gastric mechanoreceptors and providing a signal of gastric fullness. Even during a meal, some ingested nutrients pass from the stomach and contact intestinal receptors. Such contact results in gastrointestinal peptide release and the activation of neural fibers producing reflex alterations in gastrointestinal motor and secretory activity and providing feedback information about the volume and nature of ingested nutrients that could alter the size of the current meal or affect subsequent eating. Recent work has characterized the ability of multiple gut peptides to affect eating and, consistent with their different patterns of release around meals, various roles for these peptides in overall eating control have been suggested. With the current rapid increase in rates of obesity, peripheral peptides with the ability to affect food intake provide attractive targets for antiobesity drug development.

19.
Am J Physiol Regul Integr Comp Physiol ; 288(2): R384-8, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15388494

ABSTRACT

Peptide YY3-36 [PYY(3-36)], a gastrointestinal peptide that is released into the circulation in response to ingesting a meal, has recently been suggested to play a role in controlling food intake. PYY(3-36) has been reported to inhibit food intake following peripheral administration in rodents and in human subjects. To more fully characterize the potential feeding actions of PYY(3-36), we examined the ability of a dose range of PYY(3-36) (0.3-3.0 nmol/kg) to affect liquid gastric emptying and daily 6-h food intake in male rhesus monkeys. Intramuscular PYY(3-36) produced a dose-related inhibition of saline gastric emptying that was maximal at a dose of 3 nmol/kg. Intramuscular PYY(3-36) administered before daily 6-h food access produced significant feeding reductions at doses of 1 and 3 nmol/kg. Analyses of the patterns of food intake across the 6-h period of food access revealed that PYY(3-36) increased the latency to the first meal and reduced average meal size without altering meal number. Although single doses of PYY(3-36) reduced intake, a suppressive effect on food intake was not sustained over multiple administrations across successive days. Together, these data suggest that PYY(3-36) has the ability to reduce food intake in acute test situations in nonhuman primates. Whether this is a physiological action of the endogenous peptide remains to be determined.


Subject(s)
Appetite Regulation/physiology , Gastric Emptying/physiology , Peptide YY/physiology , Animals , Appetite Regulation/drug effects , Dose-Response Relationship, Drug , Gastric Emptying/drug effects , Macaca mulatta , Peptide Fragments , Peptide YY/pharmacology
20.
Endocrinology ; 146(4): 1676-85, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15625240

ABSTRACT

Otsuka Long-Evans Tokushima fatty (OLETF) rats lacking cholecystokinin-A receptors are hyperphagic, obese, and diabetic. Although exercise attenuates OLETF rats' obesity, the mechanisms underlying the effects of exercise are unclear. In this study, we determined the effects of running wheel activity on patterns of body weight gain, food intake, and hypothalamic gene expression. We demonstrate that voluntary running activity beginning at 8 wk of age normalized meal patterns, food intake, body weight, and plasma levels of glucose and leptin in OLETF rats. During the initial exercise period, corticotropin-releasing factor (CRF) mRNA expression was significantly elevated in the dorsomedial hypothalamus (DMH) but not in the paraventricular nucleus in both OLETF and control Long-Evans Tokushima rats. In response to long-term exercise, arcuate nucleus (Arc) neuropeptide Y (NPY), and proopiomelanocortin as well as DMH NPY and CRF mRNA expression were increased in Long-Evans Tokushima rats. In contrast, whereas exercising OLETF rats had increased Arc NPY and DMH CRF expression, Arc proopiomelanocortin and DMH NPY mRNA levels were not elevated. Finally, we demonstrate that the effects of exercise on body weight in OLETF rats were long lasting. Although food intake and body weight were increased in OLETF rats when running wheels were locked, weights did not return to those of sedentary OLETF rats. Together, these data suggest that the elevation of DMH CRF expression may mediate the short-term feeding inhibitory effects of exercise and that exercise limits the elevation of DMH NPY expression to account for the overall prevention of OLETF rats' obesity.


Subject(s)
Hyperphagia/prevention & control , Hypothalamus/physiology , Obesity/prevention & control , Physical Conditioning, Animal , Animals , Blood Glucose/analysis , Body Weight , Corticotropin-Releasing Hormone/genetics , Eating , Leptin/blood , Male , Neuropeptide Y/genetics , Pro-Opiomelanocortin/genetics , RNA, Messenger/analysis , Rats , Rats, Inbred OLETF , Rats, Long-Evans , Running , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...