neuropeptide-y and Hyperphagia

neuropeptide-y has been researched along with Hyperphagia* in 149 studies

Reviews

4 review(s) available for neuropeptide-y and Hyperphagia

ArticleYear
Gut Hormones in Health and Obesity: The Upcoming Role of Short Chain Fatty Acids.
    Nutrients, 2021, Jan-31, Volume: 13, Issue:2

    We are currently facing an obesity pandemic, with worldwide obesity rates having tripled since 1975. Obesity is one of the main risk factors for the development of non-communicable diseases, which are now the leading cause of death worldwide. This calls for urgent action towards understanding the underlying mechanisms behind the development of obesity as well as developing more effective treatments and interventions. Appetite is carefully regulated in humans via the interaction between the central nervous system and peripheral hormones. This involves a delicate balance in external stimuli, circulating satiating and appetite stimulating hormones, and correct functioning of neuronal signals. Any changes in this equilibrium can lead to an imbalance in energy intake versus expenditure, which often leads to overeating, and potentially weight gain resulting in overweight or obesity. Several lines of research have shown imbalances in gut hormones are found in those who are overweight or obese, which may be contributing to their condition. Therefore, this review examines the evidence for targeting gut hormones in the treatment of obesity by discussing how their dysregulation influences food intake, the potential possibility of altering the circulating levels of these hormones for treating obesity, as well as the role of short chain fatty acids and protein as novel treatments.

    Topics: Acetic Acid; Animals; Appetite; Appetite Regulation; Butyrates; Central Nervous System; Cholecystokinin; Dipeptides; Energy Intake; Energy Metabolism; Fatty Acids, Volatile; Gastrointestinal Hormones; Gastrointestinal Tract; Ghrelin; Glucagon-Like Peptide 1; Humans; Hyperphagia; Mice; Neuropeptide Y; Obesity; Overweight; Oxyntomodulin; Pancreatic Polypeptide; Propionates; Satiation

2021
Detection of amino acid deprivation in the central nervous system.
    Current opinion in clinical nutrition and metabolic care, 2013, Volume: 16, Issue:1

    To understand the principles of amino acid deprivation sensing in the brain and its behavioral and metabolic outcomes with an emphasis on the current literature.. Sensing essential amino acid (EAA) depletion occurs in the anterior piriform cortex (APC) via general control nonderepressible 2 (GCN2) binding to deacylated tRNA and subsequent glutamatergic signaling to influence behavior. Mapping of the APC output during EAA insufficiency shows axons projecting to the hypothalamus as well as other regions that are involved in feeding and locomotion. Whereas these neurocircuits are clearly important in regulating anorectic responses to an EAA-devoid diet, the propagating events and regulatory factors are still unclear. Recently, several groups examined signaling and gene expression in the arcuate nucleus and lateral hypothalamus during EAA deficiency. In these efforts, several gene products, including somatostatin, corticotrophin-releasing hormone, neuropeptide Y, agouti-related protein, and several novel targets were identified as factors involved in regulating the aversion to EAA-deficient diets. On a different note, marginal EAA deficiency in the form of methionine restriction promotes hyperphagia similar to low-protein diets, yet animals are leaner and live longer. The central mechanisms are unclear but involve sympathetic nervous signaling. How and why different degrees of EAA deficiency cause opposite changes in behavior and body composition require further study.. Scientific inquiry into the central mechanism by which EAA insufficiency is sensed has identified the APC as the brain's initial EAA chemosensor. Beyond this, much remains uncertain. Future investigation into the signaling and gene expression events occurring in the hypothalamus and other brain regions is warranted.

    Topics: Agouti-Related Protein; Amino Acids, Essential; Animals; Diet, Protein-Restricted; Humans; Hyperphagia; Hypothalamus; Models, Animal; Neuropeptide Y; Signal Transduction

2013
Low abundance of NPY in the hypothalamus can produce hyperphagia and obesity.
    Peptides, 2007, Volume: 28, Issue:2

    States of increased metabolic demand are associated with up-regulation of NPY and hyperphagia. However, we present some instances of hyperphagia in which NPY is not up-regulated. Ablation or functional disruption of specific sites in the hypothalamus, such as the ventromedial or paraventricular nuclei, or transection of inputs to the hypothalamus from the hindbrain results in hyperphagia and excess body weight gain. However, NPY expression and concentration in these experimental models is either decreased or unchanged. While there is no up-regulation of NPY in these models, there is increased sensitivity to the orexigenic effects of NPY. This enhanced responsiveness to NPY may more than compensate for the reduced levels of NPY and result in hyperphagia and excess body weight gain. The hyper-responsiveness may be due either to an increase in NPY receptors or to other changes in target cells and response pathways that may result from the treatments used in these models.

    Topics: Humans; Hyperphagia; Hypothalamus; Neuropeptide Y; Obesity

2007
Peptides and feeding.
    Peptides, 1985, Volume: 6 Suppl 2

    This report reviews the hypothesis that peptides play a role in appetite modulation, stressing that the available evidence is predominantly pharmacological and thus caution needs to be taken in assigning physiological significance at this time. Two peptide systems have been postulated--a peripheral satiety system, typified by the gastrointestinal hormone cholecystokinin and a central feeding system driven by the opioid peptides and neuropeptide Y. This review also discusses the putative role of peptides in the anorexia of aging, drinking elicited by feeding and as mediators of the autonomic effects seen in association with ventromedial hypothalamic lesions.

    Topics: Aging; Animals; Appetite Depressants; Autonomic Nervous System; Cats; Cholecystokinin; Corticotropin-Releasing Hormone; Dogs; Drinking Behavior; Feeding Behavior; Hyperphagia; Hypothalamus, Middle; Male; Nerve Tissue Proteins; Neuropeptide Y; Neurotransmitter Agents; Peptides; Rats; Satiety Response

1985

Trials

2 trial(s) available for neuropeptide-y and Hyperphagia

ArticleYear
Endocrine and metabolic aspects of adult Prader-Willi syndrome with special emphasis on the effect of growth hormone treatment.
    Growth hormone & IGF research : official journal of the Growth Hormone Research Society and the International IGF Research Society, 2004, Volume: 14, Issue:1

    Prader-Willi syndrome (PWS) is a genetic disorder characterized by mild mental retardation, short stature, abnormal body composition, muscular hypotonia and distinctive behavioural features. Excessive eating causes progressive obesity with increased cardiovascular morbidity and mortality. In the PWS genotype loss of one or more normally active paternal genes in region q11-13 on chromosome 15 is seen. It is supposed that the genetic alteration leads to dysfunction of several hypothalamic centres and growth hormone (GH) deficiency (GHD) is common. PWS is well described in children, in whom GH treatment improves body composition, linear growth, physical strength and agility. Few studies have focused on adults. We examined a cohort of 19 young adults with clinical PWS (13 with positive genotype) and mean BMI of 35 kg/m2. At baseline the activity of the GH-insulin-like growth factor-I (IGF-I) system was impaired with low GH values, low total IGF-I and in relation to the obesity low levels of free IGF-I and non-suppressed IGF-binding-protein-1 (IGFBP-1). 2/3 were hypogonadal. Bone mineral density (BMD) was low. Four patients had impaired glucose tolerance and nine patients high homeostasis model assessment (HOMA) index, indicating insulin resistance. Seven patients had a moderate dyslipidemia. The 13 patients with the PWS genotype were shorter and had significantly lower IGF-I. Seventeen (9 men and 8 women), subsequently completed a 12 months GH treatment trial, and GH had beneficial effects on body composition without significant adverse effects. The effects were more pronounced in the patients with the PWS genotype. Analysis of peptides involved in appetite regulation showed that leptin levels were high reflecting obesity and as a consequence NPY levels were low. In relation to the patients obesity circulating oxytocin levels were abnormally low and ghrelin levels abnormally high. Thus, oxytocin and ghrelin might be involved in the hyperphagia. NPY, leptin and ghrelin did not change during GH treatment. In conclusion this pilot study showed that adults with PWS have a partial GH deficiency, and GH treatment has beneficial effects on body composition in adult PWS without significant side-effects. Larger and longer term studies on the effect of GH replacement in adult PWS are encouraged.

    Topics: Adolescent; Adult; Appetite; Body Composition; Bone Density; Carbohydrate Metabolism; DNA Methylation; Endocrine System; Female; Ghrelin; Growth Hormone; Humans; Hyperphagia; Insulin-Like Growth Factor Binding Protein 1; Insulin-Like Growth Factor Binding Protein 2; Insulin-Like Growth Factor Binding Protein 3; Insulin-Like Growth Factor I; Leptin; Lipid Metabolism; Male; Neuropeptide Y; Obesity; Oxytocin; Peptide Hormones; Prader-Willi Syndrome

2004
Peptides associated with hyperphagia in adults with Prader-Willi syndrome before and during GH treatment.
    Growth hormone & IGF research : official journal of the Growth Hormone Research Society and the International IGF Research Society, 2003, Volume: 13, Issue:6

    Prader-Willi syndrome (PWS) is a complex genetic disorder characterised by mild mental retardation and distinct physical, behavioural, and psychiatric features. One of the cardinal symptoms is excessive eating, which--if left untreated--leads to extreme obesity. In the present study we have examined circulating levels of peptides with documented association to hyperphagia in young adults with PWS. Since growth hormone (GH) is often used nowadays to correct GH insufficiency during childhood PWS, we also studied the impact of GH administration on the peptides. Seventeen adults, 9 men and 8 women, 17-32 years of age with a mean BMI of 35+/-3.2 kg/m(2) participated. All had clinical PWS (Holm's criteria). Genetic testing was performed in all patients and in 11 the diagnosis was confirmed. They were randomized to treatment with either placebo or GH (Genotropin, Pharmacia Corporation) for 6 months. Subsequently all received open label treatment to provide all subjects with 12 months active GH treatment. Doses were individually titrated. Weight, BMI, oxytocin (baseline only), leptin, Neuropeptide Y (NPY), and ghrelin were evaluated at baseline and after 6 and 12 months. At baseline plasma mean oxytocin was within and serum ghrelin just above the normal range (14.7+/-1.2 pmol/L and 0.87+/-0.12 microg/L, respectively). Serum leptin levels were high above and plasma NPY levels within the lower normal range (47.8+/-29.1 microg/L and 13+/-1 pmol/L, respectively). Results were independent of genotype. No changes in mean BMI, ghrelin, leptin or NPY were seen following GH treatment.. Leptin levels were in general high reflecting obesity and as a consequence NPY levels were low. In simple obesity oxytocin levels are high, while ghrelin levels are suppressed. In view of the adiposity oxytocin circulated in abnormally low and ghrelin in abnormally high concentrations in our patients. GH treatment of PWS patients did not change ghrelin, leptin or NPY. We suggest that both oxytocin and ghrelin are involved in the pathogenesis of hyperphagia seen in PWS.

    Topics: Adolescent; Adult; Body Mass Index; Eating; Fasting; Female; Ghrelin; Human Growth Hormone; Humans; Hyperphagia; Leptin; Male; Neuropeptide Y; Obesity; Oxytocin; Peptide Hormones; Prader-Willi Syndrome

2003

Other Studies

143 other study(ies) available for neuropeptide-y and Hyperphagia

ArticleYear
d-Allulose Inhibits Ghrelin-Responsive, Glucose-Sensitive and Neuropeptide Y Neurons in the Arcuate Nucleus and Central Injection Suppresses Appetite-Associated Food Intake in Mice.
    Nutrients, 2022, Jul-29, Volume: 14, Issue:15

    d-allulose, a rare sugar, has sweetness with few calories. d-allulose regulates feeding and glycemia, and ameliorates hyperphagia, obesity and diabetes. All these functions involve the central nervous system. However, central mechanisms underlying these effects of d-allulose remain unknown. We recently reported that d-allulose activates the anorexigenic neurons in the hypothalamic arcuate nucleus (ARC), the neurons that respond to glucagon-like peptide-1 and that express proopiomelanocortin. However, its action on the orexigenic neurons remains unknown. This study investigated the effects of d-allulose on the ARC neurons implicated in hunger, by measuring cytosolic Ca

    Topics: Animals; Appetite; Arcuate Nucleus of Hypothalamus; Eating; Fructose; Ghrelin; Glucose; Hyperphagia; Mice; Neurons; Neuropeptide Y; Obesity; Rats; Rats, Sprague-Dawley

2022
Hypothalamic hormone-sensitive lipase regulates appetite and energy homeostasis.
    Molecular metabolism, 2021, Volume: 47

    The goal of this study was to investigate the importance of central hormone-sensitive lipase (HSL) expression in the regulation of food intake and body weight in mice to clarify whether intracellular lipolysis in the mammalian hypothalamus plays a role in regulating appetite.. Using pharmacological and genetic approaches, we investigated the role of HSL in the rodent brain in the regulation of feeding and energy homeostasis under basal conditions during acute stress and high-fat diet feeding.. We found that HSL, a key enzyme in the catabolism of cellular lipid stores, is expressed in the appetite-regulating centers in the hypothalamus and is activated by acute stress through a mechanism similar to that observed in adipose tissue and skeletal muscle. Inhibition of HSL in rodent models by a synthetic ligand, global knockout, or brain-specific deletion of HSL prevents a decrease in food intake normally seen in response to acute stress and is associated with the increased expression of orexigenic peptides neuropeptide Y (NPY) and agouti-related peptide (AgRP). Increased food intake can be reversed by adeno-associated virus-mediated reintroduction of HSL in neurons of the mediobasal hypothalamus. Importantly, metabolic stress induced by a high-fat diet also enhances the hyperphagic phenotype of HSL-deficient mice. Specific deletion of HSL in the ventromedial hypothalamic nucleus (VMH) or AgRP neurons reveals that HSL in the VMH plays a role in both acute stress-induced food intake and high-fat diet-induced obesity.. Our results indicate that HSL activity in the mediobasal hypothalamus is involved in the acute reduction in food intake during the acute stress response and sensing of a high-fat diet.

    Topics: Agouti-Related Protein; Animals; Appetite; Body Weight; Diet, High-Fat; Eating; Energy Metabolism; Female; Homeostasis; Hyperphagia; Hypothalamus; Male; Mice; Mice, Inbred C57BL; Mice, Knockout; Neurons; Neuropeptide Y; Obesity; RNA Splicing Factors; Sterol Esterase; Stress, Physiological; Transcriptome

2021
Hypothalamic tanycytes generate acute hyperphagia through activation of the arcuate neuronal network.
    Proceedings of the National Academy of Sciences of the United States of America, 2020, 06-23, Volume: 117, Issue:25

    Topics: Agouti-Related Protein; Animals; Appetite; Arcuate Nucleus of Hypothalamus; Channelrhodopsins; Energy Metabolism; Ependymoglial Cells; Feeding Behavior; Female; Genes, Reporter; Hyperphagia; Injections, Intraventricular; Male; Mice; Mice, Transgenic; Models, Animal; Nerve Net; Neurons; Neuropeptide Y; Optical Imaging; Optogenetics; Patch-Clamp Techniques; Pro-Opiomelanocortin; Stereotaxic Techniques

2020
Glycyl-l-glutamine attenuates NPY-induced hyperphagia via the melanocortin system.
    Neuroscience letters, 2020, 09-25, Volume: 736

    This study aimed to determine whether glycyl-l-glutamine (Gly-Gln; β-endorphin (30-31)), a non-opioid peptide derived from β-endorphin processing, modulates neuropeptide Y (NPY)-induced feeding and hypothalamic mRNA expression of peptide hormones in male broiler chicks. Intracerebroventricular injection of NPY (235 pmol) generated a hyperphagic response in ad libitum chicks within 30 min. Co-administration of Gly-Gln (100 nmol) attenuated this response, inducing a 30 % decrease. This was not attributable to Gly-Gln hydrolysis because co-administration of glycine (Gly) and glutamine (Gln) had no effect on NPY-induced hyperphagia. Gly-Gln injected alone also showed no effect. The hypothalamic pro-opiomelanocortin mRNA expression in the co-injection group was significantly higher than that in the NPY alone group. These data indicate that endogenous Gly-Gln may contribute to regulate feeding behavior via the central melanocortin system in chicks and acts as a counter regulator of the neural activity in energy metabolism.

    Topics: Animals; Chickens; Dipeptides; Eating; Hyperphagia; Hypothalamus; Male; Neuropeptide Y; Pro-Opiomelanocortin

2020
Expression of the genes encoding hypothalamic feeding-related neuropeptides in the streptozotocin-induced diabetic rats with variable hyperglycemia and hyperphagia.
    Neuropeptides, 2019, Volume: 75

    Topics: Agouti-Related Protein; Animals; Blood Glucose; Diabetes Mellitus, Experimental; Eating; Hyperglycemia; Hyperphagia; Hypothalamus; Leptin; Male; Nerve Tissue Proteins; Neuropeptide Y; Pro-Opiomelanocortin; Rats; Rats, Wistar; Thyrotropin-Releasing Hormone

2019
Exogenous nicotinamide adenine dinucleotide regulates energy metabolism via hypothalamic connexin 43.
    Metabolism: clinical and experimental, 2018, Volume: 88

    Nicotinamide adenine dinucleotide (NAD)-dependent deacetylase SIRT1 is an important regulator of hypothalamic neuronal function. Thus, an adequate hypothalamic NAD content is critical for maintaining normal energy homeostasis.. We investigated whether NAD supplementation increases hypothalamic NAD levels and affects energy metabolism in mice. Furthermore, we investigated the mechanisms underlying the effects of exogenous NAD on central metabolism upon entering the hypothalamus.. Central and peripheral NAD administration suppressed fasting-induced hyperphagia and weight gain in mice. Extracellular NAD was imported into N1 hypothalamic neuronal cells in a connexin 43-dependent and CD73-independent manner. Consistent with the in vitro data, inhibition of hypothalamic connexin 43 blocked hypothalamic NAD uptake and NAD-induced anorexia. Exogenous NAD suppressed NPY and AgRP transcriptional activity, which was mediated by SIRT1 and FOXO1.. Exogenous NAD is effectively transported to the hypothalamus via a connexin 43-dependent mechanism and increases hypothalamic NAD content. Therefore, NAD supplementation is a potential therapeutic method for metabolic disorders characterized by hypothalamic NAD depletion.

    Topics: Agouti-Related Protein; Animals; Biological Transport; Connexin 43; Energy Metabolism; Hyperphagia; Hypothalamus; Injections, Intraperitoneal; Injections, Intraventricular; Male; Mice, Inbred C57BL; NAD; Neurons; Neuropeptide Y; Sirtuin 1; Transcription, Genetic; Weight Gain

2018
Early life disruption to the ghrelin system with over-eating is resolved in adulthood in male rats.
    Neuropharmacology, 2017, Volume: 113, Issue:Pt A

    Early life overweight is a significant risk factor for developmental programming of adult obesity due to changes in the availability of metabolic factors crucial for the maturation of brain appetite-regulatory circuitry. The appetite-stimulating hormone, ghrelin, has been recently identified as a major regulator of the establishment of hypothalamic feeding pathways. Ghrelin exists in circulation in two major forms, as acylated and des-acylated ghrelin. While most research has focused on acyl ghrelin, the role of neonatal des-acyl ghrelin in metabolic programming is currently unknown. Here we assessed the influences of early life overfeeding on the ghrelin system, including acyl and des-acyl ghrelin's ability to access the hypothalamus in male rats. Our data show that early life overfeeding influences the ghrelin system short-term, leading to an acute reduction in circulating des-acyl ghrelin and increased expression of the growth hormone secretagogue receptor (GHSR) in the arcuate nucleus of the hypothalamus (ARC). These changes are associated with increased neuronal activation in response to exogenous acyl, but not des-acyl, ghrelin in the ARC and the paraventricular nucleus of the hypothalamus (PVN). Interestingly, while we observed no differences in the accessibility of the ARC to acyl or des-acyl ghrelin, less exogenous acyl ghrelin reaches the PVN in the neonatally overfed. Importantly, the influences of neonatal overfeeding on the ghrelin system were not maintained into adulthood. Therefore, while early life overfeeding results in excess body weight and stimulates acute changes in the brain's sensitivity to metabolic signals, this developmental mal-programming is at least partially alleviated in adulthood.

    Topics: Acyltransferases; Agouti-Related Protein; Animals; Animals, Newborn; Arcuate Nucleus of Hypothalamus; Body Weight; Feeding Behavior; Ghrelin; Hyperphagia; Male; Neuropeptide Y; Paraventricular Hypothalamic Nucleus; Pro-Opiomelanocortin; Rats; Rats, Wistar; Receptors, Ghrelin

2017
Combined parental obesity augments single-parent obesity effects on hypothalamus inflammation, leptin signaling (JAK/STAT), hyperphagia, and obesity in the adult mice offspring.
    Physiology & behavior, 2016, Jan-01, Volume: 153

    We aimed to evaluate the effects of maternal and/or paternal obesity on offspring body mass, leptin signaling, appetite-regulating neurotransmitters and local inflammatory markers. C57BL/6 mice received standard chow (SC, lean groups) or high-fat diet (HF, obese groups) starting from one month of age. At three months, HF mice became obese relative to SC mice. They were then mated as follows: lean mother and lean father, lean mother and obese father, obese mother and lean father, and obese mother and obese father. The offspring received the SC diet from weaning until three months of age, when they were sacrificed. In the offspring, paternal obesity did not lead to changes in the Janus kinase (JAK)/signal transducer and activation of the transcription (STAT) pathway or feeding behavior but did induce hypothalamic inflammation. On the other hand, maternal obesity resulted in increased weight gain, hyperleptinemia, decreased leptin OBRb receptor expression, JAK/STAT pathway impairment, and increased SOCS3 signaling in the offspring. In addition, maternal obesity elevated inflammatory markers and altered NPY and POMC expression in the hypothalamus. Interestingly, combined parental obesity exacerbated the deleterious outcomes compared to single-parent obesity. In conclusion, while maternal obesity is known to program metabolic changes and obesity in offspring, the current study demonstrated that obese fathers induce hypothalamus inflammation in offspring, which may contribute to the development of metabolic syndromes in adulthood.

    Topics: Animals; Body Weight; Diet, High-Fat; Energy Intake; Fathers; Female; Hyperphagia; Hypothalamus; Inflammation Mediators; Janus Kinase 1; Leptin; Male; Mice; Mothers; Neuropeptide Y; Obesity; Parents; Pro-Opiomelanocortin; Receptors, Leptin; Signal Transduction; STAT1 Transcription Factor; Suppressor of Cytokine Signaling 3 Protein; Suppressor of Cytokine Signaling Proteins

2016
Leptin regulates energy intake but fails to facilitate hibernation in fattening Daurian ground squirrels (Spermophilus dauricus).
    Journal of thermal biology, 2016, Volume: 57

    Body fat storage before hibernation affects the timing of immergence in Daurian ground squirrels (Spermophilus dauricus). Leptin is an adipose signal and plays vital role in energy homeostasis mainly by action in brain. To test the hypothesis that leptin plays a role in facilitating the process of hibernation, squirrels were administrated with recombinant murine leptin (1μg/day) through intracerebroventricular (ICV) injection for 12 days during fattening. From day 7 to 12, animals were moved into a cold room (5±1°C) with constant darkness which functioned as hibernaculum. Energy intake, body mass and core body temperature (Tb) were continuously monitored throughout the course of experiment. Resting metabolic rate (RMR) was measured under both warm and cold conditions. At the end of leptin administration, we measured the serum concentration of hormones related to energy regulation, mRNA expression of hypothalamic neuropeptides and uncoupling protein 1 (UCP1) levels in brown adipose tissue (BAT). Our results showed that during leptin administration, the cumulative food intake and increase of body mass were suppressed while Tb and RMR were unaltered. The proportion of torpid squirrels was not different between two groups. At the end of leptin administration, the expressions of hypothalamic neuropeptide Y and agouti gene-related protein were suppressed. There were no differences in UCP1 mRNA expression or protein content in BAT between groups. Our data suggest that leptin can affect energy intake via hypothalamic neuropeptides, but is not involved in the initiation of hibernation in fattening Daurian ground squirrels.

    Topics: Adipose Tissue, Brown; Animals; Body Weight; Energy Intake; Hibernation; Hyperphagia; Hypothalamus; Leptin; Neuropeptide Y; Sciuridae; Thermogenesis; Uncoupling Protein 1

2016
Central transthyretin acts to decrease food intake and body weight.
    Scientific reports, 2016, Apr-07, Volume: 6

    Transthyretin (TTR) is a blood and cerebrospinal fluid transporter of thyroxine and retinol. Gene expression profiling revealed an elevation of Ttr expression in the dorsomedial hypothalamus (DMH) of rats with exercise-induced anorexia, implying that central TTR may also play a functional role in modulating food intake and energy balance. To test this hypothesis, we have examined the effects of brain TTR on food intake and body weight and have further determined hypothalamic signaling that may underlie its feeding effect in rats. We found that intracerebroventricular (icv) administration of TTR in normal growing rats decreased food intake and body weight. This effect was not due to sickness as icv TTR did not cause a conditioned taste aversion. ICV TTR decreased neuropeptide Y (NPY) levels in the DMH and the paraventricular nucleus (P < 0.05). Chronic icv infusion of TTR in Otsuka Long-Evans Tokushima Fatty rats reversed hyperphagia and obesity and reduced DMH NPY levels. Overall, these results demonstrate a previously unknown anorectic action of central TTR in the control of energy balance, providing a potential novel target for treating obesity and its comorbidities.

    Topics: Animals; Arcuate Nucleus of Hypothalamus; Blotting, Western; Body Weight; Cells, Cultured; Eating; Gene Expression Profiling; Hyperphagia; Hypothalamus; Infusions, Intraventricular; Male; Neuropeptide Y; Obesity; Oligonucleotide Array Sequence Analysis; Prealbumin; Rats, Inbred OLETF; Rats, Sprague-Dawley; Reverse Transcriptase Polymerase Chain Reaction

2016
Enhanced Ghrelin Levels and Hypothalamic Orexigenic AgRP and NPY Neuropeptide Expression in Models of Jejuno-Colonic Short Bowel Syndrome.
    Scientific reports, 2016, 06-21, Volume: 6

    Short bowel syndrome (SBS) patients developing hyperphagia have a better outcome. Gastrointestinal endocrine adaptations help to improve intestinal functions and food behaviour. We investigated neuroendocrine adaptations in SBS patients and rat models with jejuno-ileal (IR-JI) or jejuno-colonic (IR-JC) anastomosis with and without parenteral nutrition. Circulating levels of ghrelin, PYY, GLP-1, and GLP-2 were determined in SBS rat models and patients. Levels of mRNA for proglucagon, PYY and for hypothalamic neuropeptides were quantified by qRT-PCR in SBS rat models. Histology and immunostaining for Ki67, GLP-1 and PYY were performed in SBS rats. IR-JC rats, but not IR-JI, exhibited significantly higher crypt depths and number of Ki67-positive cells than sham. Fasting and/or postprandial plasma ghrelin and PYY concentrations were higher, or tend to be higher, in IR-JC rats and SBS-JC patients than in controls. Proglucagon and Pyy mRNA levels were significantly enhanced in IR-JC rats. Levels of mRNA coding hypothalamic orexigenic NPY and AgRP peptides were significantly higher in IR-JC than in sham rats. We demonstrate an increase of plasma ghrelin concentrations, major changes in hypothalamic neuropeptides levels and greater induction of PYY in SBS-JC rats and patients suggesting that jejuno-colonic continuity creates a peculiar environment promoting further gut-brain adaptations.

    Topics: Adult; Aged; Agouti-Related Protein; Anastomosis, Surgical; Animals; Colon; Disease Models, Animal; Feeding Behavior; Female; Ghrelin; Glucagon-Like Peptide 1; Glucagon-Like Peptide 2; Humans; Hyperphagia; Hypothalamus; Intestinal Mucosa; Jejunum; Ki-67 Antigen; Male; Middle Aged; Neuropeptide Y; Peptide YY; Proglucagon; Rats; Rats, Wistar; Real-Time Polymerase Chain Reaction; RNA, Messenger; Short Bowel Syndrome

2016
Hypothalamic AMPK-induced autophagy increases food intake by regulating NPY and POMC expression.
    Autophagy, 2016, Volume: 12, Issue:11

    Hypothalamic AMP-activated protein kinase (AMPK) plays important roles in the regulation of food intake by altering the expression of orexigenic or anorexigenic neuropeptides. However, little is known about the mechanisms of this regulation. Here, we report that hypothalamic AMPK modulates the expression of NPY (neuropeptide Y), an orexigenic neuropeptide, and POMC (pro-opiomelanocortin-α), an anorexigenic neuropeptide, by regulating autophagic activity in vitro and in vivo. In hypothalamic cell lines subjected to low glucose availability such as 2-deoxy-d-glucose (2DG)-induced glucoprivation or glucose deprivation, autophagy was induced via the activation of AMPK, which regulates ULK1 and MTOR complex 1 followed by increased Npy and decreased Pomc expression. Pharmacological or genetic inhibition of autophagy diminished the effect of AMPK on neuropeptide expression in hypothalamic cell lines. Moreover, AMPK knockdown in the arcuate nucleus of the hypothalamus decreased autophagic activity and changed Npy and Pomc expression, leading to a reduction in food intake and body weight. AMPK knockdown abolished the orexigenic effects of intraperitoneal 2DG injection by decreasing autophagy and changing Npy and Pomc expression in mice fed a high-fat diet. We suggest that the induction of autophagy is a possible mechanism of AMPK-mediated regulation of neuropeptide expression and control of feeding in response to low glucose availability.

    Topics: AMP-Activated Protein Kinases; Animals; Arcuate Nucleus of Hypothalamus; Autophagy; Autophagy-Related Protein 5; Autophagy-Related Protein-1 Homolog; Body Weight; Cell Line; Deoxyglucose; Down-Regulation; Eating; Enzyme Activation; Gene Expression Regulation; Gene Knockdown Techniques; Hyperphagia; Hypothalamus; Male; Mechanistic Target of Rapamycin Complex 1; Mice, Inbred C57BL; Multiprotein Complexes; Neuropeptide Y; Pro-Opiomelanocortin; RNA, Messenger; Signal Transduction; Sirolimus; TOR Serine-Threonine Kinases; Up-Regulation

2016
A Small Potassium Current in AgRP/NPY Neurons Regulates Feeding Behavior and Energy Metabolism.
    Cell reports, 2016, 11-08, Volume: 17, Issue:7

    Neurons that co-express agouti-related peptide (AgRP) and neuropeptide Y (NPY) are indispensable for normal feeding behavior. Firing activities of AgRP/NPY neurons are dynamically regulated by energy status and coordinate appropriate feeding behavior to meet nutritional demands. However, intrinsic mechanisms that regulate AgRP/NPY neural activities during the fed-to-fasted transition are not fully understood. We found that AgRP/NPY neurons in satiated mice express high levels of the small-conductance calcium-activated potassium channel 3 (SK3) and are inhibited by SK3-mediated potassium currents; on the other hand, food deprivation suppresses SK3 expression in AgRP/NPY neurons, and the decreased SK3-mediated currents contribute to fasting-induced activation of these neurons. Genetic mutation of SK3 specifically in AgRP/NPY neurons leads to increased sensitivity to diet-induced obesity, associated with chronic hyperphagia and decreased energy expenditure. Our results identify SK3 as a key intrinsic mediator that coordinates nutritional status with AgRP/NPY neural activities and animals' feeding behavior and energy metabolism.

    Topics: Action Potentials; Agouti-Related Protein; Animals; Circadian Rhythm; Diet, High-Fat; Energy Metabolism; Fasting; Feeding Behavior; Hyperphagia; Ion Channel Gating; Mice, Inbred C57BL; Mutation; N-Methylaspartate; Neurons; Neuropeptide Y; Obesity; Small-Conductance Calcium-Activated Potassium Channels

2016
Early and Long-term Undernutrition in Female Rats Exacerbates the Metabolic Risk Associated with Nutritional Rehabilitation.
    The Journal of biological chemistry, 2015, Jul-31, Volume: 290, Issue:31

    Human studies have suggested that early undernutrition increases the risk of obesity, thereby explaining the increase in overweight among individuals from developing countries who have been undernourished as children. However, this conclusion is controversial, given that other studies do not concur. This study sought to determine whether rehabilitation after undernutrition increases the risk of obesity and metabolic disorders. We employed a published experimental food-restriction model. Wistar female rats subjected to severe food restriction since fetal stage and controls were transferred to a moderately high-fat diet (cafeteria) provided at 70 days of life to 6.5 months. Another group of undernourished rats were rehabilitated with chow. The energy intake of undernourished animals transferred to cafeteria formula exceeded that of the controls under this regime and was probably driven by hypothalamic disorders in insulin and leptin signal transduction. The cafeteria diet resulted in greater relative increases in both fat and lean body mass in the undernourished rats when compared with controls, enabling the former group to completely catch up in length and body mass index. White adipose tissues of undernourished rats transferred to the high-lipid regime developed a browning which, probably, contributed to avoid the obesigenic effect observed in controls. Nevertheless, the restricted group rehabilitated with cafeteria formula had greater accretion of visceral than subcutaneous fat, showed increased signs of macrophage infiltration and inflammation in visceral pad, dyslipidemia, and ectopic fat accumulation. The data indicate that early long-term undernutrition is associated with increased susceptibility to the harmful effects of nutritional rehabilitation, without causing obesity.

    Topics: Adipose Tissue, White; Adiposity; Animals; Diet, High-Fat; Energy Intake; Female; Hyperphagia; Hypothalamus; Insulin Resistance; Leptin; Liver; Male; Malnutrition; Muscle, Skeletal; Neuropeptide Y; Obesity; Oxidation-Reduction; Pregnancy; Prenatal Exposure Delayed Effects; Pro-Opiomelanocortin; Rats, Wistar; Risk Factors

2015
Hypothalamic Non-AgRP, Non-POMC GABAergic Neurons Are Required for Postweaning Feeding and NPY Hyperphagia.
    The Journal of neuroscience : the official journal of the Society for Neuroscience, 2015, Jul-22, Volume: 35, Issue:29

    The hypothalamus is critical for feeding and body weight regulation. Prevailing studies focus on hypothalamic neurons that are defined by selectively expressing transcription factors or neuropeptides including those expressing proopiomelanocortin (POMC) and agouti-related peptides (AgRP). The Cre expression driven by the pancreas-duodenum homeobox 1 promoter is abundant in several hypothalamic nuclei but not in AgRP or POMC neurons. Using this line, we generated mice with disruption of GABA release from a major subset of non-POMC, non-AgRP GABAergic neurons in the hypothalamus. These mice exhibited a reduction in postweaning feeding and growth, and disrupted hyperphagic responses to NPY. Disruption of GABA release severely diminished GABAergic input to the paraventricular hypothalamic nucleus (PVH). Furthermore, disruption of GABA-A receptor function in the PVH also reduced postweaning feeding and blunted NPY-induced hyperphagia. Given the limited knowledge on postweaning feeding, our results are significant in identifying GABA release from a major subset of less appreciated hypothalamic neurons as a key mediator for postweaning feeding and NPY hyperphagia, and the PVH as one major downstream site that contributes significantly to the GABA action. Significance statement: Prevalent studies on feeding in the hypothalamus focus on well characterized, selective groups neurons [e.g., proopiomelanocortin (POMC) and agouti-related peptide (AgRP) neurons], and as a result, the role of the majority of other hypothalamic neurons is largely neglected. Here, we demonstrated an important role for GABAergic projections from non-POMC non-AgRP neurons to the paraventricular hypothalamic nucleus in promoting postweaning (mainly nocturnal) feeding and mediating NPY-induced hyperphagia. Thus, these results signify an importance to study those yet to be defined hypothalamic neurons in the regulation of energy balance and reveal a neural basis for postweaning (nocturnal) feeding and NPY-mediated hyperphagia.

    Topics: Agouti-Related Protein; Animals; Feeding Behavior; GABAergic Neurons; Hyperphagia; Hypothalamus; In Situ Hybridization; Mice; Mice, Mutant Strains; Neuropeptide Y; Organ Culture Techniques; Paraventricular Hypothalamic Nucleus; Patch-Clamp Techniques; Pro-Opiomelanocortin

2015
Down-regulation of hypothalamic pro-opiomelanocortin (POMC) expression after weaning is associated with hyperphagia-induced obesity in JCR rats overexpressing neuropeptide Y.
    The British journal of nutrition, 2014, Mar-14, Volume: 111, Issue:5

    We hypothesised that hypothalamic feeding-related neuropeptides are differentially expressed in obese-prone and lean-prone rats and trigger overeating-induced obesity. To test this hypothesis, in the present study, we measured energy balance and hypothalamic neuropeptide Y (NPY) and pro-opiomelanocortin (POMC) mRNA expressions in male JCR:LA-cp rats. We compared, in independent cohorts, free-feeding obese-prone (Obese-FF) and lean-prone (Lean-FF) rats at pre-weaning (10 d old), weaning (21-25 d old) and early adulthood (8-12 weeks). A group of Obese-pair-feeding (PF) rats pair-fed to the Lean-FF rats was included in the adult cohort. The body weights of 10-d-old Obese-FF and Lean-FF pups were not significantly different. However, when the pups were shifted from dams' milk to solid food (weaning), the obese-prone rats exhibited more energy intake over the days than the lean-prone rats and higher body and fat pad weights and fasting plasma glucose, leptin, insulin and lipid levels. These differences were consistent with higher energy consumption and lower energy expenditure. In the young adult cohort, the differences between the Obese-FF and Lean-FF rats became more pronounced, yielding significant age effects on most of the parameters of the metabolic syndrome, which were reduced in the Obese-PF rats. The obese-prone rats displayed higher NPY expression than the lean-prone rats at pre-weaning and weaning, and the expression levels did not differ by age. In contrast, POMC expression exhibited significant age-by-genotype differences. At pre-weaning, there was no genotype difference in POMC expression, but in the weanling cohort, obese-prone pups exhibited lower POMC expression than the lean-prone rats. This genotype difference became more pronounced at adulthood. Overall, the development of hyperphagia-induced obesity in obese-prone JCR rats is related to POMC expression down-regulation in the presence of established NPY overexpression.

    Topics: Adiposity; Animals; Arcuate Nucleus of Hypothalamus; Behavior, Animal; Caloric Restriction; Down-Regulation; Energy Intake; Energy Metabolism; Gene Expression Regulation, Developmental; Hyperphagia; Male; Metabolic Syndrome; Neurons; Neuropeptide Y; Obesity; Pro-Opiomelanocortin; Random Allocation; Rats; Rats, Mutant Strains; Receptors, Leptin; RNA, Messenger; Weaning

2014
Intermittent fasting induces hypothalamic modifications resulting in low feeding efficiency, low body mass and overeating.
    Endocrinology, 2014, Volume: 155, Issue:7

    Intermittent fasting (IF) is an often-used intervention to decrease body mass. In male Sprague-Dawley rats, 24 hour cycles of IF result in light caloric restriction, reduced body mass gain, and significant decreases in the efficiency of energy conversion. Here, we study the metabolic effects of IF in order to uncover mechanisms involved in this lower energy conversion efficiency. After 3 weeks, IF animals displayed overeating during fed periods and lower body mass, accompanied by alterations in energy-related tissue mass. The lower efficiency of energy use was not due to uncoupling of muscle mitochondria. Enhanced lipid oxidation was observed during fasting days, whereas fed days were accompanied by higher metabolic rates. Furthermore, an increased expression of orexigenic neurotransmitters AGRP and NPY in the hypothalamus of IF animals was found, even on feeding days, which could explain the overeating pattern. Together, these effects provide a mechanistic explanation for the lower efficiency of energy conversion observed. Overall, we find that IF promotes changes in hypothalamic function that explain differences in body mass and caloric intake.

    Topics: Agouti-Related Protein; Animals; Body Weight; Eating; Energy Metabolism; Fasting; Feeding Behavior; Gene Expression; Hyperphagia; Hypothalamus; Immunoblotting; Leptin; Lipid Metabolism; Male; Mitochondria, Muscle; Neuropeptide Y; Oxidation-Reduction; Rats; Rats, Sprague-Dawley; Reverse Transcriptase Polymerase Chain Reaction; Thyrotropin-Releasing Hormone; Time Factors

2014
Hypothalamic miR-103 protects from hyperphagic obesity in mice.
    The Journal of neuroscience : the official journal of the Society for Neuroscience, 2014, Aug-06, Volume: 34, Issue:32

    The role of neuronal noncoding RNAs in energy control of the body is not fully understood. The arcuate nucleus (ARC) of the hypothalamus comprises neurons regulating food intake and body weight. Here we show that Dicer-dependent loss of microRNAs in these neurons of adult (DicerCKO) mice causes chronic overactivation of the signaling pathways involving phosphatidylinositol-3-kinase (PI3K), Akt, and mammalian target of rapamycin (mTOR) and an imbalance in the levels of neuropeptides, resulting in severe hyperphagic obesity. Similarly, the activation of PI3K-Akt-mTOR pathway due to Pten deletion in the adult forebrain leads to comparable weight increase. Conversely, the mTORC1 inhibitor rapamycin normalizes obesity in mice with an inactivated Dicer1 or Pten gene. Importantly, the continuous delivery of oligonucleotides mimicking microRNAs, which are predicted to target PI3K-Akt-mTOR pathway components, to the hypothalamus attenuates adiposity in DicerCKO mice. Furthermore, loss of miR-103 causes strong upregulation of the PI3K-Akt-mTOR pathway in vitro and its application into the ARC of the Dicer-deficient mice both reverses upregulation of Pik3cg, the mRNA encoding the catalytic subunit p110γ of the PI3K complex, and attenuates the hyperphagic obesity. Our data demonstrate in vivo the crucial role of neuronal microRNAs in the control of energy homeostasis.

    Topics: Absorptiometry, Photon; Agouti-Related Protein; Animals; DEAD-box RNA Helicases; HeLa Cells; Humans; Hyperphagia; Hypothalamus; Luminescent Proteins; Mice; Mice, Inbred C57BL; MicroRNAs; Neuropeptide Y; Obesity; Oncogene Protein v-akt; Phosphatidylinositol 3-Kinases; PTEN Phosphohydrolase; Ribonuclease III; TOR Serine-Threonine Kinases; Transduction, Genetic

2014
Programmed hyperphagia secondary to increased hypothalamic SIRT1.
    Brain research, 2014, Nov-17, Volume: 1589

    Small for gestational age (SGA) offspring exhibit reduced hypothalamic neural satiety pathways leading to programmed hyperphagia and adult obesity. Appetite regulatory site, the hypothalamic arcuate nucleus (ARC) contains appetite (NPY/AgRP) and satiety (POMC) neurons. Using in vitro culture of hypothalamic neuroprogenitor cells (NPC) which form the ARC, we demonstrated that SGA offspring exhibit reduced NPC proliferation and neuronal differentiation. bHLH protein Hes1 promotes NPC self-renewal and inhibits differentiation by repressing neuronal differentiation genes (Mash1, neurogenin3). We hypothesized that Hes1/Mash1 and ultimately ARC neuronal differentiation and expression of NPY/POMC neurons are influenced by SIRT1 which is a nutrient sensor and a histone deacetylase. Control dams received ad libitum food, whereas study dams were 50% food-restricted from pregnancy day 10 to 21 (SGA). In vivo studies showed that SGA newborns and adult offspring had increased protein expression of hypothalamic/ARC SIRT1 and AgRP with decreased POMC. Additionally, SGA newborns had decreased expression of hypothalamic neurogenic factors with reduced in vivo NPC proliferation. In vitro culture of hypothalamic NPCs showed similar changes with elevated SIRT1 binding to Hes1 in SGA newborn. Silencing SIRT1 increased NPC proliferation and Hes1 and Tuj1expression in both Control and SGA NPCs. Although SGA NPC proliferation remained below that of Controls, it was higher than Control NPCs in the absence of SIRT1 siRNA. The direct impact of SIRT1 on NPC proliferation and differentiation were further confirmed with pharmacologic SIRT1 inhibitor and activator. Thus, in SGA newborns elevated SIRT1 induces premature differentiation of NPCs, reducing the NPC pool and cell proliferation.

    Topics: Agouti-Related Protein; Animals; Arcuate Nucleus of Hypothalamus; Basic Helix-Loop-Helix Transcription Factors; Cell Differentiation; Cell Proliferation; Cells, Cultured; Female; Homeodomain Proteins; Hyperphagia; Male; Neurons; Neuropeptide Y; Pregnancy; Prenatal Nutritional Physiological Phenomena; Pro-Opiomelanocortin; Rats; Rats, Sprague-Dawley; Sirtuin 1; Stem Cells; Transcription Factor HES-1

2014
Overexpression of neuropeptide Y in the dorsomedial hypothalamus causes hyperphagia and obesity in rats.
    Obesity (Silver Spring, Md.), 2013, Volume: 21, Issue:6

    We sought to determine a role for NPY overexpression in the dorsomedial hypothalamus (DMH) in obesity etiology using the rat model of adeno-associated virus (AAV)-mediated expression of NPY (AAVNPY) in the DMH. Rats received bilateral DMH injections of AAVNPY or control vector and were fed on regular chow. Five-week postviral injection, half the rats from each group were switched to access to a high-fat diet for another 11 weeks. We examined variables including body weight, food intake, energy efficiency, meal patterns, glucose tolerance, fat mass, plasma insulin, plasma leptin, and hypothalamic gene expression. Rats with DMH NPY overexpression had increased food intake and body weight and lowered metabolic efficiency. The hyperphagia was mediated through increased meal size during the dark. Although these rats had normal blood glucose, their plasma insulin levels were increased in both basal and glucose challenge conditions. While high-fat diet induced hyperphagia, obesity, and hyperinsulinemia, these effects were amplified in rats with DMH NPY overexpression. Arcuate Npy, agouti-related protein and proopiomelanocortin expression was appropriately regulated in response to positive energy balance. These results indicate that DMH NPY overexpression can cause hyperphagia and obesity and DMH NPY may have actions in glucose homeostasis.

    Topics: Agouti-Related Protein; Animals; Body Weight; Dependovirus; Diet, High-Fat; Disease Models, Animal; Energy Metabolism; Gene Expression Regulation; Glucose; Glucose Tolerance Test; Homeostasis; Hyperphagia; Hypothalamus; Leptin; Male; Neuropeptide Y; Obesity; Pro-Opiomelanocortin; Rats; Rats, Sprague-Dawley

2013
Contributions of endocrinology to the migration life history of birds.
    General and comparative endocrinology, 2013, Sep-01, Volume: 190

    Migration is a key life cycle stage in nearly 2000 species of birds and is a greatly appreciated phenomenon in both cultural and academic arenas. Despite a long research tradition concerning many aspects of migration, investigations of hormonal contributions to migratory physiology and behavior are more limited and represent a comparatively young research field. We review advances in our understanding of the hormonal mechanisms of migration with particular emphasis on the sub-stages of the migration life history: development, departure, flight and arrival. These sub-stages vary widely in their behavioral, ecological and physiological contexts and, as such, should be given appropriate individual consideration.

    Topics: Animal Migration; Animals; Birds; Endocrinology; Hyperphagia; Leptin; Neuropeptide Y

2013
Deficiency of p62/Sequestosome 1 causes hyperphagia due to leptin resistance in the brain.
    The Journal of neuroscience : the official journal of the Society for Neuroscience, 2013, Sep-11, Volume: 33, Issue:37

    The cytoplasmic regulatory protein p62 (Sequestosome 1/A170) is known to modulate various receptor-mediated intracellular signaling pathways. p62 deficiency was shown to result in mature-onset obesity in mice, but the mechanisms underlying this abnormality remained unclear. Here we report that hyperphagia due to central leptin resistance is the cause of obesity in p62(-/-) mice. We found that these mice show hyperphagia. Restriction of food to the amount eaten by wild-type mice prevented excess body weight gain and fat accumulation, suggesting that overfeeding is the primary cause of obesity in p62(-/-) mice. Brain-specific p62 deficiency caused mature-onset obesity to the same extent as in p62(-/-) mice, further supporting a neuronal mechanism as the major cause of obesity in these mice. Immunohistochemical analysis revealed that p62 is highly expressed in hypothalamic neurons, including POMC neurons in the arcuate nucleus. Central leptin resistance was observed even in young preobese p62(-/-) mice. We found a defect in intracellular distribution of the transcription factor Stat3, which is essential for the action of leptin, in p62(-/-) mice. These results indicate that brain p62 plays an important role in bodyweight control by modulating the central leptin-signaling pathway and that lack of p62 in the brain causes leptin resistance, leading to hyperphagia. Thus, p62 could be a clinical target for treating obesity and metabolic syndrome.

    Topics: Animals; Body Weight; Brain; Eating; Embryo, Mammalian; Food Deprivation; Gene Expression Regulation; Hyperphagia; In Vitro Techniques; Leptin; Male; Mice; Mice, Inbred C57BL; Mice, Transgenic; Nestin; Neuropeptide Y; Oxygen Consumption; Pro-Opiomelanocortin; Receptors, Leptin; STAT3 Transcription Factor; Suppressor of Cytokine Signaling 3 Protein; Suppressor of Cytokine Signaling Proteins; Transcription Factor TFIIH; Transcription Factors

2013
Efferent projections of neuropeptide Y-expressing neurons of the dorsomedial hypothalamus in chronic hyperphagic models.
    The Journal of comparative neurology, 2013, Jun-01, Volume: 521, Issue:8

    The dorsomedial hypothalamus (DMH) has long been implicated in feeding behavior and thermogenesis. The DMH contains orexigenic neuropeptide Y (NPY) neurons, but the role of these neurons in the control of energy homeostasis is not well understood. NPY expression in the DMH is low under normal conditions in adult rodents but is significantly increased during chronic hyperphagic conditions such as lactation and diet-induced obesity (DIO). To understand better the role of DMH-NPY neurons, we characterized the efferent projections of DMH-NPY neurons using the anterograde tracer biotinylated dextran amine (BDA) in lactating rats and DIO mice. In both models, BDA- and NPY-colabeled fibers were limited mainly to the hypothalamus, including the paraventricular nucleus of the hypothalamus (PVH), lateral hypothalamus/perifornical area (LH/PFA), and anteroventral periventricular nucleus (AVPV). Specifically in lactating rats, BDA-and NPY-colabeled axonal swellings were in close apposition to cocaine- and amphetamine-regulated transcript (CART)-expressing neurons in the PVH and AVPV. Although the DMH neurons project to the rostral raphe pallidus (rRPa), these projections did not contain NPY immunoreactivity in either the lactating rat or the DIO mouse. Instead, the majority of BDA-labeled fibers in the rRPa were orexin positive. Furthermore, DMH-NPY projections were not observed within the nucleus of the solitary tract (NTS), another brainstem site critical for the regulation of sympathetic outflow. The present data suggest that NPY expression in the DMH during chronic hyperphagic conditions plays important roles in feeding behavior and thermogenesis by modulating neuronal functions within the hypothalamus, but not in the brainstem.

    Topics: Age Factors; Animals; Animals, Newborn; Biotin; Chronic Disease; Dextrans; Disease Models, Animal; Efferent Pathways; Female; Gonadotropin-Releasing Hormone; Hyperphagia; Hypothalamic Hormones; Hypothalamus; Intracellular Signaling Peptides and Proteins; Lactic Acid; Male; Melanins; Mice; Mice, Inbred C57BL; Nerve Tissue Proteins; Neurons; Neuropeptide Y; Neuropeptides; Obesity; Orexins; Peptide Fragments; Pituitary Hormones; Pregnancy; Rats; Rats, Wistar; Tryptophan Hydroxylase

2013
Repeated gastric distension alters food intake and neuroendocrine profiles in rats.
    Physiology & behavior, 2012, Feb-28, Volume: 105, Issue:4

    The consumption of a large food bolus leads to stomach distension. Gastric distension potently signals the termination of a meal by stimulating gastric mechanoreceptors and activating neuroendocrine circuitry. The ability to terminate a meal is altered in disorders such as bulimia nervosa (BN), binge-eating disorder (BED) and certain subtypes of obesity in which large quantities of food are frequently ingested. When a large meal is consumed, the stomach is rapidly stretched. We modeled this rapid distension of the stomach in order to determine if the neuroendocrine abnormalities present in these disorders, including increased gastric capacit3y, leptin dysregulation, and alterations in neuropeptide Y (NPY), and proopiomelanocortin (POMC) expression, were influenced by the rapid stretch aspect of repeatedly consuming a large meal. To test the effects of repeated gastric distension (RGD) on neuroendocrine factors involved in energy homeostasis, a permanent intra-gastric balloon was implanted in rats, and briefly inflated daily for 4 weeks. Though body weights and daily food intakes remained equivalent in RGD and control rats, a significant delay in the onset of feeding was present during the first and second, but not the third and fourth weeks of inflations. Despite equivalent body weights and daily caloric consumption, RGD animals had significantly decreased leptin levels (p<0.05), and tended to have increased fasting arcuate NPY levels (p=0.08), which were suppressed more than control animals following food intake (control and RGD decreases from baseline were 184.95% and 257.42%, respectively). NPY expression in the nucleus of the solitary tract followed a similar pattern. These data demonstrate that the act of regularly distending the stomach can have effects on the regulation of energy balance that are independent from those related to caloric consumption, and may be related to disorders such as BN, BED, and certain types of obesity in which meal termination is impaired.

    Topics: Animals; Arcuate Nucleus of Hypothalamus; Body Weight; Bulimia; Eating; Energy Metabolism; Feeding Behavior; Gastric Balloon; Hyperphagia; Leptin; Male; Neuropeptide Y; Pro-Opiomelanocortin; Rats; Rats, Long-Evans; Solitary Nucleus; Stomach

2012
Hypothalamic mTOR pathway mediates thyroid hormone-induced hyperphagia in hyperthyroidism.
    The Journal of pathology, 2012, Volume: 227, Issue:2

    Hyperthyroidism is characterized in rats by increased energy expenditure and marked hyperphagia. Alterations of thermogenesis linked to hyperthyroidism are associated with dysregulation of hypothalamic AMPK and fatty acid metabolism; however, the central mechanisms mediating hyperthyroidism-induced hyperphagia remain largely unclear. Here, we demonstrate that hyperthyroid rats exhibit marked up-regulation of the hypothalamic mammalian target of rapamycin (mTOR) signalling pathway associated with increased mRNA levels of agouti-related protein (AgRP) and neuropeptide Y (NPY), and decreased mRNA levels of pro-opiomelanocortin (POMC) in the arcuate nucleus of the hypothalamus (ARC), an area where mTOR co-localizes with thyroid hormone receptor-α (TRα). Central administration of thyroid hormone (T3) or genetic activation of thyroid hormone signalling in the ARC recapitulated hyperthyroidism effects on feeding and the mTOR pathway. In turn, central inhibition of mTOR signalling with rapamycin in hyperthyroid rats reversed hyperphagia and normalized the expression of ARC-derived neuropeptides, resulting in substantial body weight loss. The data indicate that in the hyperthyroid state, increased feeding is associated with thyroid hormone-induced up-regulation of mTOR signalling. Furthermore, our findings that different neuronal modulations influence food intake and energy expenditure in hyperthyroidism pave the way for a more rational design of specific and selective therapeutic compounds aimed at reversing the metabolic consequences of this disease.

    Topics: Agouti-Related Protein; AMP-Activated Protein Kinases; Animals; Disease Models, Animal; Eating; Feeding Behavior; Hyperphagia; Hyperthyroidism; Hypothalamus; Male; Neural Pathways; Neuropeptide Y; Phosphorylation; Pro-Opiomelanocortin; Protein Kinase Inhibitors; Rats; Rats, Sprague-Dawley; RNA, Messenger; Signal Transduction; Sirolimus; Thyroid Hormone Receptors alpha; Time Factors; TOR Serine-Threonine Kinases; Triiodothyronine; Weight Loss

2012
Lesion of area postrema attenuated hyperphagic responses to glucoprivation, but not transcriptional activation of the neuropeptide Y gene in rats.
    Neuroreport, 2012, Aug-01, Volume: 23, Issue:11

    The area postrema (AP) is a circumventricular organ that lacks a blood-brain barrier. Previous studies have shown that the lesion of AP (APX) attenuated hyperphagic responses to glucoprivation. As the orexigenic neuropeptide Y (NPY) neurons have been implicated in the regulation of food intake, we examined whether the activation of NPY neurons by glucoprivation is mediated through the AP as well. In agreement with previous studies, hyperphagic responses to an injection of 2-deoxy-D-glucose that blocks glucose utilization were significantly attenuated in the APX group compared with the sham-operated (Sham) group. However, the expression levels of NPY heteronuclear RNA, a sensitive indicator for the gene transcription, were significantly increased in the arcuate nucleus by a 2-deoxy-D-glucose injection in both the APX and the Sham groups, and there were no significant differences in the values between groups. These data suggest that the hyperphagic response to glucoprivation, but not the activation of NPY gene transcription in the arcuate nucleus, was mediated through the AP in the hindbrain.

    Topics: Animals; Area Postrema; Deoxyglucose; Eating; Glucose; Hyperphagia; Male; Neuropeptide Y; Rats; Rats, Sprague-Dawley; RNA, Messenger; Transcriptional Activation

2012
Improvement of hyperphagia by activation of cerebral I(1)-imidazoline receptors in streptozotocin-induced diabetic mice.
    Hormone and metabolic research = Hormon- und Stoffwechselforschung = Hormones et metabolisme, 2012, Volume: 44, Issue:9

    Imidazoline I1-receptors (I1R) are known to regulate blood pressure and rilmenidine, an agonist, is widely used as antihypertensive agent in clinic. However, the role of I1R in feeding behavior is still unclear. In the present study, we used the agonist of I1R to investigate the effect on hyperphagia in streptozotocin (STZ)-induced diabetic mice. Rilmenidine decreased the food intake of STZ-diabetic mice in a dose-dependent manner. The reduction of food intake was abolished by pretreatment with efaroxan at the dose sufficient to block I1R. Intracerebroventricular (icv) administration of rilmenidine into STZ-diabetic mice also significantly reduced hyperphagia, which was reversed by icv administration of efaroxan. In addition, similar results were observed in STZ-diabetic mice, which received chronic treatment with rilmenidine 3 times daily (t.i.d.) for 7 days. Moreover, the hypothalamic neuropeptide Y (NPY) level was reduced by rilmenidine that was also reversed by pretreatment with efaroxan. In conclusion, the obtained results suggest that rilmenidine can decrease food intake in STZ-diabetic mice through an activation of I1R to lower hypothalamic NPY level.

    Topics: Animals; Benzofurans; Cerebrum; Diabetes Mellitus, Type 1; Disease Models, Animal; Eating; Female; Humans; Hyperphagia; Hypothalamus; Imidazoles; Imidazoline Receptors; Male; Mice; Mice, Inbred BALB C; Neuropeptide Y; Oxazoles; Rilmenidine; Streptozocin

2012
Amylin blunts hyperphagia and reduces weight and fat gain during recovery in socially stressed rats.
    American journal of physiology. Regulatory, integrative and comparative physiology, 2012, Sep-15, Volume: 303, Issue:6

    During recovery from social stress in a visible burrow system (VBS), during which a dominance hierarchy is formed among the males, rats display hyperphagia and gain weight preferentially as visceral adipose tissue. By proportionally increasing visceral adiposity, social stress may contribute to the establishment of metabolic disorder. Amylin was administered to rats fed ad libitum during recovery from VBS stress in an attempt to prevent hyperphagia and the resultant gain in body weight and fat mass. Amylin treatment reduced food intake, weight gain, and accumulation of fat mass in male burrow rats, but not in male controls that spent time housed with a single female rather than in the VBS. Amylin did not alter neuropeptide Y (NPY), agouti-related peptide (AgRP), or proopiomelanocortin (POMC) mRNA expression in the arcuate nucleus of the hypothalamus as measured at the end of the recovery period, nor did it affect plasma corticosterone or leptin. Amylin exerted most of its effect on food intake during the first few days of recovery, possibly through antagonism of NPY and/or increasing leptin sensitivity. The potential for chronic social stress to contribute to metabolic disorder is diminished by amylin treatment, though the neuroendocrine mechanisms behind this effect remain elusive.

    Topics: Adipose Tissue; Agouti-Related Protein; Animals; Arcuate Nucleus of Hypothalamus; Eating; Female; Gene Expression Regulation; Hyperphagia; Islet Amyloid Polypeptide; Male; Neuropeptide Y; Pro-Opiomelanocortin; Rats; Social Behavior; Stress, Psychological; Time Factors; Weight Gain

2012
Central nervous system neuropeptide Y signaling via the Y1 receptor partially dissociates feeding behavior from lipoprotein metabolism in lean rats.
    American journal of physiology. Endocrinology and metabolism, 2012, Dec-15, Volume: 303, Issue:12

    Elevated plasma triglyceride (TG) levels contribute to an atherogenic dyslipidemia that is associated with obesity, diabetes, and metabolic syndrome. Numerous models of obesity are characterized by increased central nervous system (CNS) neuropeptide Y (NPY) tone that contributes to excess food intake and obesity. Previously, we demonstrated that intracerebroventricular (icv) administration of NPY in lean fasted rats also elevates hepatic production of very low-density lipoprotein (VLDL)-TG. Thus, we hypothesize that elevated CNS NPY action contributes to not only the pathogenesis of obesity but also dyslipidemia. Here, we sought to determine whether the effects of NPY on feeding and/or obesity are dissociable from effects on hepatic VLDL-TG secretion. Pair-fed, icv NPY-treated, chow-fed Long-Evans rats develop hypertriglyceridemia in the absence of increased food intake and body fat accumulation compared with vehicle-treated controls. We then modulated CNS NPY signaling by icv injection of selective NPY receptor agonists and found that Y1, Y2, Y4, and Y5 receptor agonists all induced hyperphagia in lean, ad libitum chow-fed Long-Evans rats, with the Y2 receptor agonist having the most pronounced effect. Next, we found that at equipotent doses for food intake NPY Y1 receptor agonist had the most robust effect on VLDL-TG secretion, a Y2 receptor agonist had a modest effect, and no effect was observed for Y4 and Y5 receptor agonists. These findings, using selective agonists, suggest the possibility that the effect of CNS NPY signaling on hepatic VLDL-TG secretion may be relatively dissociable from effects on feeding behavior via the Y1 receptor.

    Topics: Animals; Appetite Regulation; Behavior, Animal; Central Nervous System; Humans; Hyperphagia; Infusions, Intraventricular; Lipoproteins, VLDL; Liver; Male; Nerve Tissue Proteins; Neurons; Neuropeptide Y; Obesity; Protein Isoforms; Rats; Rats, Long-Evans; Receptors, Neuropeptide Y; Recombinant Proteins; Signal Transduction; Triglycerides

2012
Repeated glucoprivation delayed hyperphagic responses while activating neuropeptide Y neurons in rats.
    Peptides, 2011, Volume: 32, Issue:4

    It is well known that glucoprivation induces the release of counterregulatory hormones such as glucagon, and that the response is attenuated when the stimuli are repeated. Glucoprivation also activates orexigenic neurons and induces hyperphagic responses, although it remains unclear whether these responses are attenuated in repeated glucoprivation. In this study, we examined time course changes in feeding as well as activities of orexigenic neuropeptide Y (NPY) neurons in repeated glucoprivation in rats. Either 2-deoxy-d-glucose (2DG), which blocks glucose utilization, or isotonic saline (control) was injected subcutaneously to rats for 14 days, and food consumption for 1 and 2h after injection was monitored throughout the experiment. While 2DG injection induced robust feeding responses during the first 1h after injection, the response was gradually attenuated and the food consumption was significantly less on days 12-14 compared to that on day 1. On the other hand, food consumption during 2h after 2DG injection was not changed significantly for 14 days. The transcriptional activities of NPY neurons in the arcuate nucleus and C1/A1 region of the hindbrain, measured by intronic in situ hybridization, were significantly enhanced after repeated 2DG injection for 14 days, while the feeding responses to intracerebroventricular injection of NPY were significantly less in the 2DG-repeated group compared to the saline-repeated group. It is thus demonstrated that repeated glucoprivation delayed hyperphagic responses while activating NPY neurons in rats. Our data also suggest that decreased feeding responses to NPY might be at least partially responsible for the delayed response.

    Topics: Animals; Blood Glucose; Deoxyglucose; Hyperphagia; Insulin; Male; Neurons; Neuropeptide Y; Rats; Rats, Sprague-Dawley

2011
Neuropeptide Y and agouti-related peptide mediate complementary functions of hyperphagia and reduced energy expenditure in leptin receptor deficiency.
    Endocrinology, 2011, Volume: 152, Issue:3

    Neuropeptide Y (NPY) and agouti-related peptide (AGRP) can produce hyperphagia, reduce energy expenditure, and promote triglyceride deposition in adipose depots. As these two neuropeptides are coexpressed within the hypothalamic arcuate nucleus and mediate a major portion of the obesity caused by leptin signaling deficiency, we sought to determine whether the two neuropeptides mediated identical or complementary actions. Because of separate neuropeptide receptors and signal transduction mechanisms, there is a possibility of distinct encoding systems for the feeding and energy expenditure aspects of leptin-regulated metabolism. We have genetically added NPY deficiency and/or AGRP deficiency to LEPR deficiency isolated to AGRP cells. Our results indicate that the obesity of LEPR deficiency in AGRP/NPY neurons can produce obesity with either AGRP or NPY alone with AGRP producing hyperphagia while NPY promotes reduced energy expenditure. The absence of both NPY and AGRP prevents the development of obesity attributable to isolated LEPR deficiency in AGRP/NPY neurons. Operant behavioral testing indicated that there were no alterations in the reward for a food pellet from the AGRP-specific LEPR deficiency.

    Topics: Adiposity; Agouti-Related Protein; Animals; Body Composition; Energy Metabolism; Female; Hyperphagia; Male; Mice; Mice, Knockout; Neurons; Neuropeptide Y; Receptors, Leptin

2011
Hyperphagia and central mechanisms for leptin resistance during pregnancy.
    Endocrinology, 2011, Volume: 152, Issue:4

    The purpose of this work was to study the central mechanisms involved in food intake regulation and leptin resistance during gestation in the rat. Sprague Dawley rats of 7, 13, and 18 d of pregnancy [days of gestation (G) 7, G13, and G18] were used and compared with nonpregnant animals in diestrus-1. Food intake was already increased in G7, before hyperleptinemia and central leptin resistance was established in midpregnancy. Leptin resistance was due to a reduction in leptin transport through the blood-brain barrier (BBB) and to alterations in leptin signaling within the hypothalamus based on an increase in suppressor of cytokine signaling 3 levels and a blockade of signal transducer and activator of transcription-3 phosphorylation (G13), followed by a decrease in LepRb and of Akt phosphorylation (G18). In early gestation (G7), no change in hypothalamic neuropeptide Y (NPY), agouti-related peptide (AgRP), or proopiomelanocortin (POMC) expression was shown. Nevertheless, an increase in NPY and AgRP and a decrease in POMC mRNA were observed in G13 and G18 rats, probably reflecting the leptin resistance. To investigate the effect of maternal vs. placental hormones on these mechanisms, we used a model of pseudogestation. Rats of 9 d of pseudogestation were hyperphagic, showing an increase in body and adipose tissue weight, normoleptinemia, and normal responses to iv/intracerebroventricular leptin on hypothalamic leptin signaling, food intake, and body weight. Leptin transport through the BBB, and hypothalamic NPY, AgRP and POMC expression were unchanged. Finally, the transport of leptin through the BBB was assessed using a double-chamber culture system of choroid plexus epithelial cells or brain microvascular endothelial cells. We found that sustained high levels of prolactin significantly reduced leptin translocation through the barrier, whereas progesterone and β-estradiol did not show any effect. Our data demonstrate a dual mechanism of leptin resistance during mid/late-pregnancy, which is not due to maternal hormones and which allows the maintenance of hyperphagia in the presence of hyperleptinemia driven by an increase in NPY and AgRP and a decrease in POMC mRNA. By contrast, in early pregnancy maternal hormones induce hyperphagia without the regulation of hypothalamic NPY, AgRP, or POMC and in the absence of leptin resistance.

    Topics: Agouti-Related Protein; Animals; Animals, Newborn; Blood-Brain Barrier; Blotting, Western; Body Weight; Cells, Cultured; Eating; Enzyme-Linked Immunosorbent Assay; Female; Hyperlipidemias; Hyperphagia; Hypothalamus; In Situ Hybridization; Infusions, Intraventricular; Injections, Intravenous; Leptin; Neuropeptide Y; Pregnancy; Pro-Opiomelanocortin; Progesterone; Rats; Rats, Sprague-Dawley

2011
Corticotropin-releasing hormone (CRH) transgenic mice display hyperphagia with increased Agouti-related protein mRNA in the hypothalamic arcuate nucleus.
    Endocrine journal, 2011, Volume: 58, Issue:4

    Although glucocorticoid-induced hyperphagia is observed in the patients with glucocorticoid treatment or Cushing's syndrome, its molecular mechanism is not clear. We thus explored the expression of neuropeptide mRNAs in the hypothalamus related to appetite regulation in CRH over-expressing transgenic mice (CRH-Tg), a model of Cushing's syndrome. We measured food intake, body weight (including body fat weight) and plasma corticosterone levels in CRH-Tg and their wild-type littermates (WT) at 6 and 14 weeks old. We also examined neuropeptide Y (NPY), proopiomelanocortin (POMC) and Agouti-related protein (AgRP) mRNAs in the arcuate nucleus (ARC) using in situ hybridization. Circulating corticosterone levels in CRH-Tg were markedly elevated at both 6 and 14 weeks old. Body fat weight in CRH-Tg was significantly increased at 14 weeks old, which is considered as an effect of chronic glucocorticoid excess. At both 6 and 14 weeks old, CRH-Tg mice showed significant hyperphagia compared with WT (14w old: WT 3.9±0.1, CRH-Tg 5.1±0.7 g/day, p<0.05). Unexpectedly, NPY mRNA levels in CRH-Tg were significantly decreased at 14 weeks old (WT: 1571.5±111.2, CRH-Tg: 949.1±139.3 dpm/mg, p<0.05), and there were no differences in POMC mRNA levels between CRH-Tg and WT. On the other hand, AgRP mRNA levels in CRH-Tg were significantly increased compared with WT at both ages (14w old: WT 365.6±88.6, CRH-Tg 660.1±87.2 dpm/ mg, p<0.05). These results suggest that glucocorticoid-induced hyperphagia is associated with increased hypothalamic AgRP. Our results also indicate that hypothalamic NPY does not have an essential role in the increased food intake during glucocorticoid excess.

    Topics: Adipose Tissue; Agouti-Related Protein; Animals; Arcuate Nucleus of Hypothalamus; Body Weight; Corticosterone; Corticotropin-Releasing Hormone; Cushing Syndrome; Disease Models, Animal; Eating; Glucocorticoids; Hyperphagia; Male; Mice; Mice, Transgenic; Neuropeptide Y; Pro-Opiomelanocortin; RNA, Messenger

2011
Diet-induced obesity attenuates fasting-induced hyperphagia.
    Journal of neuroendocrinology, 2011, Volume: 23, Issue:7

    Obesity impairs arcuate (ARC) neuropeptide Y (NPY)/agouti-releated peptide (AgRP) neuronal function and renders these homeostatic neurones unresponsive to the orexigenic hormone ghrelin. In the present study, we investigated the effect of diet-induced obesity (DIO) on feeding behaviour, ARC neuronal activation and mRNA expression following another orexigenic stimulus, an overnight fast. We show that 9 weeks of high-fat feeding attenuates fasting-induced hyperphagia by suppressing ARC neuronal activation and hypothalamic NPY/AgRP mRNA expression. Thus, the lack of appropriate feeding responses in DIO mice to a fast is caused by failure ARC neurones to recognise and/or respond to orexigenic cues. We propose that fasting-induced hyperphagia is regulated not by homeostatic control of appetite in DIO mice, but rather by changes in the reward circuitry.

    Topics: Agouti-Related Protein; Animals; Arcuate Nucleus of Hypothalamus; Diet; Diet, Atherogenic; Down-Regulation; Fasting; Feeding Behavior; Hyperphagia; Male; Mice; Mice, Inbred C57BL; Mice, Obese; Neuropeptide Y; Obesity

2011
Agmatine in the hypothalamic paraventricular nucleus stimulates feeding in rats: involvement of neuropeptide Y.
    British journal of pharmacology, 2011, Volume: 164, Issue:2b

    Agmatine, a multifaceted neurotransmitter, is abundantly expressed in the hypothalamic paraventricular nucleus (PVN). Our aim was to assess (i) the effect of agmatine on feeding behaviour and (ii) its association, if any, with neuropeptide Y (NPY).. Satiated rats fitted with intra-PVN cannulae were administered agmatine, alone or jointly with (i) α₂-adrenoceptor agonist, clonidine, or antagonist, yohimbine; (ii) NPY, NPY Y₁ receptor agonist, [Leu³¹, Pro³⁴]-NPY, or antagonist, BIBP3226; or (iii) yohimbine and NPY. Cumulative food intake was monitored at different post-injection time points. Furthermore, the expression of hypothalamic NPY following i.p. treatment with agmatine, alone or in combination with yohimbine (i.p.), was evaluated by immunocytochemistry.. Agmatine robustly increased feeding in a dose-dependent manner. While pretreatment with clonidine augmented, yohimbine attenuated the orexigenic response to agmatine. Similarly, NPY and [Leu³¹, Pro³⁴]-NPY potentiated the agmatine-induced hyperphagia, whereas BIBP3226 inhibited it. Moreover, yohimbine attenuated the synergistic orexigenic effect induced by the combination of NPY and agmatine. Agmatine increased NPY immunoreactivity in the PVN fibres and in the cells of the hypothalamic arcuate nucleus (ARC) and this effect was prevented by pretreatment with yohimbine. NPY immunoreactivity in the fibres of the ARC, dorsomedial, ventromedial and lateral nuclei of the hypothalamus was not affected by any of the above treatments.. The orexigenic effect of agmatine is coupled to increased NPY activity mediated by stimulation of α₂-adrenoceptors within the PVN. This signifies the importance of agmatine or α₂-adrenoceptor modulators in the development of novel therapeutic agents to treat feeding-related disorders.

    Topics: Adrenergic alpha-2 Receptor Agonists; Adrenergic alpha-2 Receptor Antagonists; Agmatine; Animals; Arcuate Nucleus of Hypothalamus; Arginine; Clonidine; Drug Synergism; Eating; Feeding Behavior; Hyperphagia; Male; Neuropeptide Y; Paraventricular Hypothalamic Nucleus; Rats; Rats, Sprague-Dawley; Receptors, Adrenergic, alpha-2; Receptors, Neuropeptide Y; Yohimbine

2011
Olanzapine-induced hyperphagia and weight gain associate with orexigenic hypothalamic neuropeptide signaling without concomitant AMPK phosphorylation.
    PloS one, 2011, Volume: 6, Issue:6

    The success of antipsychotic drug treatment in patients with schizophrenia is limited by the propensity of these drugs to induce hyperphagia, weight gain and other metabolic disturbances, particularly evident for olanzapine and clozapine. However, the molecular mechanisms involved in antipsychotic-induced hyperphagia remain unclear. Here, we investigate the effect of olanzapine administration on the regulation of hypothalamic mechanisms controlling food intake, namely neuropeptide expression and AMP-activated protein kinase (AMPK) phosphorylation in rats. Our results show that subchronic exposure to olanzapine upregulates neuropeptide Y (NPY) and agouti related protein (AgRP) and downregulates proopiomelanocortin (POMC) in the arcuate nucleus of the hypothalamus (ARC). This effect was evident both in rats fed ad libitum and in pair-fed rats. Of note, despite weight gain and increased expression of orexigenic neuropeptides, subchronic administration of olanzapine decreased AMPK phosphorylation levels. This reduction in AMPK was not observed after acute administration of either olanzapine or clozapine. Overall, our data suggest that olanzapine-induced hyperphagia is mediated through appropriate changes in hypothalamic neuropeptides, and that this effect does not require concomitant AMPK activation. Our data shed new light on the hypothalamic mechanism underlying antipsychotic-induced hyperphagia and weight gain, and provide the basis for alternative targets to control energy balance.

    Topics: Agouti-Related Protein; Aminoimidazole Carboxamide; AMP-Activated Protein Kinases; Animals; Arcuate Nucleus of Hypothalamus; Benzodiazepines; Clozapine; Feeding Behavior; Female; Gene Expression Regulation; Hyperphagia; Hypothalamus; Injections, Intraventricular; Insulin; Intracellular Signaling Peptides and Proteins; Leptin; Neuropeptide Y; Neuropeptides; Olanzapine; Orexins; Phosphorylation; Pro-Opiomelanocortin; Rats; Rats, Sprague-Dawley; Ribonucleotides; Signal Transduction; Weight Gain

2011
Moderate long-term modulation of neuropeptide Y in hypothalamic arcuate nucleus induces energy balance alterations in adult rats.
    PloS one, 2011, Volume: 6, Issue:7

    Neuropeptide Y (NPY) produced by arcuate nucleus (ARC) neurons has a strong orexigenic effect on target neurons. Hypothalamic NPY levels undergo wide-ranging oscillations during the circadian cycle and in response to fasting and peripheral hormones (from 0.25 to 10-fold change). The aim of the present study was to evaluate the impact of a moderate long-term modulation of NPY within the ARC neurons on food consumption, body weight gain and hypothalamic neuropeptides. We achieved a physiological overexpression (3.6-fold increase) and down-regulation (0.5-fold decrease) of NPY in the rat ARC by injection of AAV vectors expressing NPY and synthetic microRNA that target the NPY, respectively. Our work shows that a moderate overexpression of NPY was sufficient to induce diurnal over-feeding, sustained body weight gain and severe obesity in adult rats. Additionally, the circulating levels of leptin were elevated but the immunoreactivity (ir) of ARC neuropeptides was not in accordance (POMC-ir was unchanged and AGRP-ir increased), suggesting a disruption in the ability of ARC neurons to response to peripheral metabolic alterations. Furthermore, a dysfunction in adipocytes phenotype was observed in these obese rats. In addition, moderate down-regulation of NPY did not affect basal feeding or normal body weight gain but the response to food deprivation was compromised since fasting-induced hyperphagia was inhibited and fasting-induced decrease in locomotor activity was absent.These results highlight the importance of the physiological ARC NPY levels oscillations on feeding regulation, fasting response and body weight preservation, and are important for the design of therapeutic interventions for obesity that include the NPY.

    Topics: Adipocytes; Adipogenesis; Animals; Arcuate Nucleus of Hypothalamus; Cell Size; Dependovirus; Down-Regulation; Eating; Energy Metabolism; Fasting; Feedback, Physiological; Genetic Vectors; HEK293 Cells; Humans; Hyperphagia; Male; MicroRNAs; Neurons; Neuropeptide Y; Obesity; PPAR gamma; Rats; Rats, Wistar; Time Factors; Weight Gain

2011
Central neuropeptide Y infusion and melanocortin 4 receptor antagonism inhibit thyrotropic function by divergent pathways.
    Neuropeptides, 2011, Volume: 45, Issue:6

    Weight loss inhibits thyrotropic function and reduces metabolic rate, thereby contributing to weight regain. Under negative energy balance there is an increase in the hypothalamic expression of both neuropeptide Y (NPY) and agouti related peptide (AgRP), the endogenous antagonist of melanocortin 4 (MC4) receptors. Both NPY and MC4 receptor antagonism reduce thyrotropic function centrally, but it is not known whether these pathways operate by similar or distinct mechanisms. We compared the time-course of effects of acute or chronic intracerebroventricular (ICV) administration of NPY (1.2 nmol acute bolus, or 3.5 nmol/day for 6 days) or the MC4 receptor antagonist HS014 (1.5 nmol bolus, or 4.8 nmol/day) on plasma concentrations of thyroid stimulating hormone (TSH) or free thyroxine (T4) in male rats pair-fed with vehicle-infused controls. These doses equipotently induced hyperphagia in acute studies, reduced latency to feed, and increased white adipose tissue mass after 6 days of infusion. Acute central NPY but not HS014 administration significantly reduced plasma TSH concentrations within 30-60 min and plasma free T4 levels within 90-120 min. These inhibitory effects were sustained for up to 5-6 days of continuous NPY infusion. HS014 induced a transient decrease in plasma free T4 levels that was observed only after 1-2 days of continuous ICV infusion. While both NPY and HS014 significantly increased corticosteronemia within an hour after ICV injection, the effect of NPY was significantly more pronounced and was sustained for up to 4 days of administration. Both NPY and HS014 significantly decreased the brown adipose tissue protein levels of uncoupling protein-3. We conclude that central NPY and MC4 antagonism decrease thyrotropic function via partially distinct mechanisms with different time courses, possibly involving glucocorticoid effects of NPY. MC4 receptor antagonism increases adiposity via pathways independent of increased food intake or changes in circulating concentrations of TSH, free T4 or corticosterone.

    Topics: Adipose Tissue, Brown; Agouti-Related Protein; Animals; Body Weight; Corticosterone; Eating; Energy Metabolism; Humans; Hyperphagia; Male; Neuropeptide Y; Peptides, Cyclic; Rats; Receptor, Melanocortin, Type 4; Thyrotrophs; Thyrotropin; Thyroxine

2011
Circadian integration of sleep-wake and feeding requires NPY receptor-expressing neurons in the mediobasal hypothalamus.
    American journal of physiology. Regulatory, integrative and comparative physiology, 2011, Volume: 301, Issue:5

    Sleep and feeding rhythms are highly coordinated across the circadian cycle, but the brain sites responsible for this coordination are unknown. We examined the role of neuropeptide Y (NPY) receptor-expressing neurons in the mediobasal hypothalamus (MBH) in this process by injecting the targeted toxin, NPY-saporin (NPY-SAP), into the arcuate nucleus (Arc). NPY-SAP-lesioned rats were initially hyperphagic, became obese, exhibited sustained disruption of circadian feeding patterns, and had abnormal circadian distribution of sleep-wake patterns. Total amounts of rapid eye movement sleep (REMS) and non-REMS (NREMS) were not altered by NPY-SAP lesions, but a peak amount of REMS was permanently displaced to the dark period, and circadian variation in NREMS was eliminated. The phase reversal of REMS to the dark period by the lesion suggests that REMS timing is independently linked to the function of MBH NPY receptor-expressing neurons and is not dependent on NREMS pattern, which was altered but not phase reversed by the lesion. Sleep-wake patterns were altered in controls by restricting feeding to the light period, but were not altered in NPY-SAP rats by restricting feeding to either the light or dark period, indicating that disturbed sleep-wake patterns in lesioned rats were not secondary to changes in food intake. Sleep abnormalities persisted even after hyperphagia abated during the static phase of the lesion. Results suggest that the MBH is required for the essential task of integrating sleep-wake and feeding rhythms, a function that allows animals to accommodate changeable patterns of food availability. NPY receptor-expressing neurons are key components of this integrative function.

    Topics: Animals; Arcuate Nucleus of Hypothalamus; Caloric Restriction; Circadian Rhythm; Corticosterone; Eating; Feeding Behavior; Hyperphagia; In Situ Hybridization; Male; Neurons; Neuropeptide Y; Obesity; Photoperiod; Rats; Rats, Sprague-Dawley; Receptors, Neuropeptide Y; Saponins; Sleep; Sleep Wake Disorders; Sleep, REM; Time Factors; Wakefulness

2011
Changes in NPY and POMC, but not serotonin transporter, following a restricted feeding/repletion protocol in rats.
    Brain research, 2010, Feb-08, Volume: 1313

    Serotonin (5-HT) plays a key role in controlling food intake and feeding behaviour and drugs targeting the 5-HT transporter (SERT) at the synaptic cleft have been used to treat feeding related disorders. To test the hypothesis that SERT might be one of the etiologic factors in the rebound hyperphagia that frequently follows the abandoning of calorie restriction diets, brain SERT content and gene expression were assessed in a restricted feeding/repletion (RFR) protocol in female rats. Animals were food-restricted (2 h access to food per day) for 7 consecutive days and then allowed constant free access to food (FAF). This intermittent fasting protocol resulted in rebound hyperphagia. Higher levels of plasma corticosterone during fasting in food-deprived rats were used as an index of hypothalamic-pituitary-adrenal axis activation. Neither brain SERT density nor expression was modified following the RFR protocol. Nevertheless, with respect to other messengers involved in eating behaviour, in the presence of low plasma leptin levels, an increase in NPY expression and a parallel decrease in POMC expression were observed in the hypothalamic arcuate nucleus of rats killed just before rebound hyperphagia. Food-restricted animals provide a tool for the further study of neurochemical alterations and for the development of new drugs to treat alterations that may occur in humans when dieting is abandoned.

    Topics: Animals; Arcuate Nucleus of Hypothalamus; Brain; Corticosterone; Diet, Reducing; Fasting; Feeding Behavior; Female; Gene Expression; Hyperphagia; Leptin; Neuropeptide Y; Pro-Opiomelanocortin; Rats; Rats, Sprague-Dawley; Serotonin Plasma Membrane Transport Proteins; Time Factors

2010
A free-choice high-fat high-sugar diet induces changes in arcuate neuropeptide expression that support hyperphagia.
    International journal of obesity (2005), 2010, Volume: 34, Issue:3

    The mechanisms for how saturated fat and sugar-based beverages contribute to human obesity are poorly understood. This paper describes a series of experiments developed to examine the response of hypothalamic neuropeptides to diets rich in sugar and fat, using three different diets: a high-fat high-sugar (HFHS) choice diet with access to chow, saturated fat and a 30% sugar solution; a high-fat (HF) choice diet with access to chow and saturated fat; or to a high-sugar (HS) choice diet with access to chow and a sugar solution.. We first studied caloric intake, body weight gain, hormonal alterations and hypothalamic neuropeptide expression when male Wistar rats were subjected to an HFHS choice, an HF choice or an HS choice diet for 1 week. Next, we studied caloric intake and body weight gain when rats were subjected to the choice diets for 5 weeks. Finally, we measured neuropeptide expression in hepatic vagotomized rats subjected to an HFHS choice, an HF choice or an HS choice diet for 1 week.. In rats on an HF choice diet, plasma leptin concentrations and proopiomelanocortin (POMC) mRNA increased and neuropeptide Y (NPY) mRNA decreased. Rats on an HFHS choice diet showed identical plasma leptin concentrations as rats on an HF choice diet. However, NPY mRNA increased and POMC mRNA decreased. An HS choice diet for 1 week did not alter hypothalamic neuropeptide expression or plasma leptin concentrations. As hormonal changes did not explain the differences in hypothalamic neuropeptide expression between rats on the choice diets, we addressed whether neuronal feedback signals mediated the hypothalamic neuropeptide response. The POMC mRNA response to different diets depended on an intact innervation of liver and upper intestinal tract.. Our data suggest that the specific combination of saturated fat and a 30% sugar solution results in hyperphagia-induced obesity and alters hypothalamic neuropeptide expression, and that the response of the melanocortin system is mediated by the hepatic vagus.

    Topics: Animals; Body Weight; Dietary Fats; Dietary Sucrose; Energy Intake; Gene Expression; Hyperphagia; Leptin; Liver; Male; Neuropeptide Y; Obesity; Pro-Opiomelanocortin; Rats; Rats, Wistar

2010
Orexin activation precedes increased NPY expression, hyperphagia, and metabolic changes in response to sleep deprivation.
    American journal of physiology. Endocrinology and metabolism, 2010, Volume: 298, Issue:3

    Several pieces of evidence support that sleep duration plays a role in body weight control. Nevertheless, it has been assumed that, after the identification of orexins (hypocretins), the molecular basis of the interaction between sleep and energy homeostasis has been provided. However, no study has verified the relationship between neuropeptide Y (NPY) and orexin changes during hyperphagia induced by sleep deprivation. In the current study we aimed to establish the time course of changes in metabolite, endocrine, and hypothalamic neuropeptide expression of Wistar rats sleep deprived by the platform method for a distinct period (from 24 to 96 h) or sleep restricted for 21 days (SR-21d). Despite changes in the stress hormones, we found no changes in food intake and body weight in the SR-21d group. However, sleep-deprived rats had a 25-35% increase in their food intake from 72 h accompanied by slight weight loss. Such changes were associated with increased hypothalamus mRNA levels of prepro-orexin (PPO) at 24 h followed by NPY at 48 h of sleep deprivation. Conversely, sleep recovery reduced the expression of both PPO and NPY, which rapidly brought the animals to a hypophagic condition. Our data also support that sleep deprivation rapidly increases energy expenditure and therefore leads to a negative energy balance and a reduction in liver glycogen and serum triacylglycerol levels despite the hyperphagia. Interestingly, such changes were associated with increased serum levels of glucagon, corticosterone, and norepinephrine, but no effects on leptin, insulin, or ghrelin were observed. In conclusion, orexin activation accounts for the myriad changes induced by sleep deprivation, especially the hyperphagia induced under stress and a negative energy balance.

    Topics: Animals; Body Weight; Eating; Gene Expression Regulation; Hyperphagia; Hypothalamus; Male; Neuropeptide Y; Rats; Rats, Wistar; Sleep Deprivation

2010
Regulation of food consumption during pregnancy and lactation in mice.
    Neuroscience and behavioral physiology, 2010, Volume: 40, Issue:3

    The aim of the present work was to assess the expression of agouti-like protein and neuropeptide Y in pregnant and lactating mice and to compare this with the leptin level and food consumption. Food consumption, blood leptin levels, and agouti-like protein and neuropeptide Y mRNA levels in the hypothalamus of C57Bl/6J mice were assessed on days 7, 13, and 18 of pregnancy and on days 10 and 21 of lactation, and in virgin females of the same ages. During pregnancy, food consumption and leptin levels decreased on day 7 and increased in day 18 of pregnancy, while neuropeptide Y mRNA levels increased on day 13 and then remained unaltered, and the agouti-like protein level increased on day 18. After parturition, food consumption continued to increase, while leptin levels and neuropeptide Y mRNA levels decreased to normal. Thus, hyperphagia in pregnancy was due to sequential activation of the expression of neuropeptide Y and agouti-like protein, while in lactation hyperphagia resulted from mechanisms not associated with changes in the expression of these neuropeptides.

    Topics: Agouti-Related Protein; Animals; Eating; Female; Gene Expression Regulation; Hyperphagia; Hypothalamus; Lactation; Leptin; Mice; Neuropeptide Y; Pregnancy; Time Factors

2010
Ghrelin acts on rat dorsal vagal complex to stimulate feeding via arcuate neuropeptide Y/agouti-related peptide neurons activation.
    Sheng li xue bao : [Acta physiologica Sinica], 2010, Aug-25, Volume: 62, Issue:4

    Ghrelin, an endogenous ligand for the growth hormone secretagogue (GHS) receptor, stimulates feeding and increases body weight. The primary action site of ghrelin has been reported to be the neuropeptide Y (NPY)/agouti-related peptide (AgRP) neurons in the hypothalamic arcuate nucleus (ARC). In addition to the hypothalamus, the caudal brainstem also appears to be an important mediator for the orexigenic activity of ghrelin. However, it is not clear whether ghrelin applied directly to the caudal brainstem activates forebrain structures. The aim of this study was to determine whether recruitment of forebrain structures was required for hyperphagic responses stimulated by ghrelin delivery within the caudal brainstem. In our experiment, all rats were surgically implanted with indwelling cannulas in the dorsal vagal complex (DVC), and ghrelin (20 pmol in 0.5 μL) was delivered to the DVC. After the injection, the orexigenic response to ghrelin was recorded by Feeding and Activity Analyser, and NPY/AgRP mRNA expressions in rat hypothalamus were detected by real-time PCR. In addition, the NPY immunoreactive neurons in the ARC were assayed by immunohistochemistry. The results showed that ghrelin significantly increased cumulative food intake at 1, 2 and 3 h after ghrelin injection, maximal response occurring at 2 h after injection. NPY/AgRP mRNA levels in ARC treated with ghrelin increased significantly compared with those in control group (injected with saline). The highest levels of NPY and AgRP mRNA were detected at 2 h after injection. The total number and mean optical density of NPY-positive neurons increased in ghrelin treated rats compared with those in control group. Consistently, ghrelin's effect was most pronounced at 2 h after injection. Taken together, we conclude that the activation of NPY/AgRP neurons in the ARC is involved in the mediation of the hyperphagic response to brainstem ghrelin administration in neurologically intact rats.

    Topics: Agouti-Related Protein; Animals; Arcuate Nucleus of Hypothalamus; Brain Stem; Feeding Behavior; Ghrelin; Hyperphagia; Hypothalamus; Male; Neurons; Neuropeptide Y; Peptide Fragments; Rats; Rats, Sprague-Dawley; RNA, Messenger

2010
Sex-associated differences in the leptin and ghrelin systems related with the induction of hyperphagia under high-fat diet exposure in rats.
    Hormones and behavior, 2009, Volume: 55, Issue:1

    Leptin and ghrelin are known to be main hormones involved in the control of food intake, with opposing effects. Here we have explored whether changes in the leptin and ghrelin system are involved in the long-term effects of high-fat (HF) diet feeding in rats and whether sex-associated differences exist. Male and female Wistar rats were fed until the age of 6 months with a normal-fat (NF) or an HF-diet. Food intake and body weight were followed. Gastric and serum levels of leptin and ghrelin, and mRNA levels of leptin (in stomach and adipose tissue), ghrelin (in stomach), and NPY, POMC, and leptin and ghrelin receptors (OB-Rb and GHS-R) (in the hypothalamus) were measured. In both males and females, total caloric intake and body weight were greater under the HF-diet feeding. In females, circulating ghrelin levels and leptin mRNA expression in the stomach were higher under HF-diet. HF-diet feeding also resulted in higher hypothalamic NPY/POMC mRNA levels, more marked in females, and in lower OB-Rb mRNA levels, more marked in males. In addition, in females, serum ghrelin levels correlated positively with hypothalamic NPY mRNA levels, and these with caloric intake. In males, hypothalamic OB-Rb mRNA levels correlated positively with POMC mRNA levels and these correlated negatively with caloric intake and with body weight. These data reflect differences between sexes in the effects of HF-diet feeding on food intake control systems, suggesting an impairment of the anorexigenic leptin-POMC system in males and an over-stimulation of the orexigenic ghrelin-NPY system in females.

    Topics: Adipose Tissue; Animals; Body Weight; Diet; Dietary Fats; Feeding Behavior; Female; Gastric Mucosa; Ghrelin; Hyperphagia; Hypothalamus; Leptin; Male; Neuropeptide Y; Pro-Opiomelanocortin; Rats; Rats, Wistar; Receptors, Ghrelin; Receptors, Leptin; RNA, Messenger; Sex Characteristics

2009
Complexity of neural mechanisms underlying overconsumption of sugar in scheduled feeding: involvement of opioids, orexin, oxytocin and NPY.
    Peptides, 2009, Volume: 30, Issue:2

    A regular daily meal regimen, as opposed to ad libitum consumption, enforces eating at a predefined time and within a short timeframe. Hence, it is important to study food intake regulation in animal feeding models that somewhat reflect this pattern. We investigated the effect of scheduled feeding on the intake of a palatable, high-sugar diet in rats and attempted to define central mechanisms - especially those related to opioid signaling--responsible for overeating sweet foods under such conditions. We found that scheduled access to food, even as challenging as 20 min per day, does not prevent overconsumption of a high-sucrose diet compared to a standard one. An opioid receptor antagonist, naloxone, at 0.3-1 mg/kg b. wt., decreased the intake of the sweet diet, whereas higher doses were required to reduce bland food consumption. Real-time PCR analysis revealed that expression of hypothalamic and brainstem genes encoding opioid peptides and receptors did not differ in sucrose versus regular diet-fed rats, which suggests that scheduled intake of sweet food produces only a transient change in the opioid tone. Intake of sugar was also associated with upregulation of orexin and oxytocin genes in the hypothalamus and NPY in the brainstem. We conclude that scheduled consumption of sugar diets is associated with activity of a complex network of neuroregulators involving opioids, orexin, oxytocin and NPY.

    Topics: Animals; Appetite Regulation; Dietary Sucrose; Eating; Food Preferences; Hyperphagia; Intracellular Signaling Peptides and Proteins; Naloxone; Neurons; Neuropeptide Y; Neuropeptides; Neurotransmitter Agents; Opioid Peptides; Orexins; Oxytocin; Rats; Rats, Sprague-Dawley

2009
Hyperphagia and increased meal size are responsible for weight gain in rats treated sub-chronically with olanzapine.
    Psychopharmacology, 2009, Volume: 203, Issue:4

    Atypical antipsychotic-induced weight gain is a significant impediment in the treatment of schizophrenia.. In a putative model of antipsychotic drug-induced weight gain, we investigated the effects of sub-chronic olanzapine on body weight, meal patterns, the expression of genes encoding for hypothalamic feeding-related neuropeptides and the contribution of hyperphagia to olanzapine-induced weight gain in rats.. In experiment 1, female rats received either olanzapine (1 mg/kg, p.o.) or vehicle, twice daily for 7 days, while meal patterns were recorded. At the end of the treatment regimen, we measured the levels of hypothalamic messenger RNAs (mRNAs) encoding neuropeptide-Y (NPY), hypocretin/orexin (HCRT), melanin concentrating hormone and pro-opiomelanocortin. NPY and HCRT mRNA levels were also assessed in a separate cohort of female rats treated acutely with olanzapine (1 mg/kg, p.o.). In experiment 2, we investigated the effect of a pair-feeding paradigm on sub-chronic (1 mg/kg, p.o.) olanzapine-induced weight gain.. In experiment 1, sub-chronic olanzapine increased body weight, food intake and meal size. Hypothalamic neuropeptide mRNA levels were unchanged after both acute and sub-chronic olanzapine treatment. In experiment 2, the restriction of food intake to the level of vehicle-treated controls abolished the sub-chronic olanzapine-induced increase in body weight.. Hyperphagia mediated by drug-induced impairments in satiety (as evidenced by increased meal size) is a key requirement for olanzapine-induced weight gain in this paradigm. However, olanzapine-induced hyperphagia and weight gain may not be mediated via alterations in the expression of the feeding-related hypothalamic neuropeptides examined in this study.

    Topics: Animals; Antipsychotic Agents; Benzodiazepines; Eating; Female; Hyperphagia; Intracellular Signaling Peptides and Proteins; Neuropeptide Y; Neuropeptides; Olanzapine; Orexins; Pro-Opiomelanocortin; Rats; Rats, Sprague-Dawley; RNA, Messenger; Weight Gain

2009
Possible common central pathway for resistin and insulin in regulating food intake.
    Acta physiologica (Oxford, England), 2009, Volume: 196, Issue:4

    Adipose tissue has been the object of intense research in the field of obesity and diabetes diseases in the last decade. Examination of adipocyte-secreted peptides led to the identification of a unique polypeptide, resistin (RSTN), which has been suggested as a link between obesity and diabetes. RSTN plays a clearly documented role in blocking insulin (INS)-induced hypoglycaemia. As brain injection of INS affects feeding behaviour, we studied the possible interaction between INS and RSTN in food-deprived rats, measuring effects on food intake. In addition, we examined how RSTN might affect neuropeptide Y (NPY)-induced feeding, as studies have shown that rat RSTN can interfere with the NPY system.. Overnight food-deprived rats were injected into the third brain ventricle (3V) with either INS (10 or 20 mUI), RSTN (0.1-0.4 nmol/rat), or saline before access to food. Another group of rats was injected into the 3V with RSTN alone, NPY alone or RSTN plus NPY. Their food intake and body weight were measured.. Our results confirm the hypophagic effect of RSTN on food deprivation-induced food intake, and more importantly, show that RSTN neither potentiates nor blocks the effects of INS on food intake, but does reduce the hyperphagic effect of NPY.. The observation that RSTN does not modify feeding INS-induced hypophagia, but does influence NPY-induced feeding, points to the possibility that RSTN may be involved in control of food intake through an NPY-ergic mechanism as INS.

    Topics: Adipose Tissue; Animals; Appetite Regulation; Body Weight; Brain; Diabetes Mellitus, Type 2; Dose-Response Relationship, Drug; Humans; Hyperphagia; Injections, Intraventricular; Insulin; Male; Metabolic Networks and Pathways; Neuropeptide Y; Obesity; Rats; Rats, Wistar; Resistin

2009
Differential responses of orexigenic neuropeptides to fasting in offspring of obese mothers.
    Obesity (Silver Spring, Md.), 2009, Volume: 17, Issue:7

    Maternal obesity due to long-term high-fat diet (HFD) consumption leads to faster growth in offspring during suckling, and increased adiposity at 20 days of age. Decreased expression of the orexigenic neuropeptide Y (NPY) and increased anorexigenic proopiomelanocortin (POMC) mRNA expression were observed in the fed state. However, hunger is the major drive to eat and hypothalamic appetite regulators change in response to meals. Therefore, it is important to compare both satiated and fasting states. Female Sprague-Dawley rats (8 weeks old) were fed a cafeteria-style HFD (15.33 kJ/g) or chow for 5 weeks before mating, with the same diet continuing throughout gestation and lactation. At postnatal day 20, male pups were killed either after overnight fasting or in the fed state. Pups from obese dams were hyperphagic during both pre- and postweaning periods. Pups from obese dams had higher hypothalamic mRNA expression of POMC and NPY Y1 receptor, but lower hypothalamic melanocortin-4 receptor (MC4R) and its downstream target single-minded gene 1 (Sim1), in the fed state. Overnight fasting reduced circulating glucose, insulin, and leptin and increased hypothalamic NPY Y1 receptor mRNA in pups from both lean and obese dams. Hypothalamic NPY and agouti-related protein (AgRP) were only increased by fasting in pups from obese dams; reductions in MC4R and Sim1 were only seen in pups from lean dams. At weaning, the suppressed orexigenic signals in offspring from obese dams were normalized after overnight fasting, although anorexigenic signaling appeared impaired in these animals. This may contribute to their hyperphagia and faster growth.

    Topics: Agouti-Related Protein; Animals; Animals, Newborn; Appetite Regulation; Basic Helix-Loop-Helix Transcription Factors; Blood Glucose; Disease Models, Animal; Fasting; Female; Hyperphagia; Hypothalamus; Insulin; Leptin; Male; Maternal Nutritional Physiological Phenomena; Neuropeptide Y; Obesity; Pregnancy; Prenatal Exposure Delayed Effects; Pro-Opiomelanocortin; Rats; Rats, Sprague-Dawley; Receptor, Melanocortin, Type 4; Repressor Proteins; RNA, Messenger

2009
[Food intake regulation in pregnant and lactating mice].
    Rossiiskii fiziologicheskii zhurnal imeni I.M. Sechenova, 2009, Volume: 95, Issue:1

    Hormone leptin inhibits appetite. It decreases hypothalamic expression of orexigenic neuropeptides, agouti-related peptide (AgRP), and neuropeptide Y (NPY). Pregnancy and lactation are characterized by hyperphagia. Food intake regulation during pregnancy and lactation is poorly understood. The aim of the work was to estimate hypothalamic expression of NPY and AgRP in pregnant and suckling mice and correlate it with food intake and blood leptin level.. Daily food intake, and levels of leptin in blood and mRNA of AgRP and NPY in hypothalamus on day 7, 13, 18 of pregnancy, 10, 21 of lactation in C57B1/6J female mice and in age-matched virgin females.. In the course of pregnancy, food intake increased, leptin level decreased on day 7 and increased on day 18, NPY expression was increasing until day 13 and then remained unchanged, AgRP expression increased on day 18 of pregnancy. In suckling females, food intake continued to grow, but levels ofleptin and NPY and AgRP expression decresed and became equal with that in virgin females.. In pregnant mice, hyperphagia results from NPY followed by AgRP expression activation, in suckling mice--from other mechanisms which are not connected with changes in expression of these neuropeptides.

    Topics: Agouti-Related Protein; Animals; Eating; Female; Gene Expression Regulation; Hyperphagia; Hypothalamus; Lactation; Leptin; Mice; Neuropeptide Y; Pregnancy; Time Factors

2009
Sustained NPY overexpression in the PVN results in obesity via temporarily increasing food intake.
    Obesity (Silver Spring, Md.), 2009, Volume: 17, Issue:7

    Increasing neuropeptide Y (NPY) signaling in the paraventricular nucleus (PVN) by recombinant adeno-associated virus (rAAV)-mediated overexpression of NPY in rats, results in hyperphagia and obesity in rats. To determine the importance of hyperphagia in the observed obesity phenotype, we pair-fed a group of AAV-NPY-injected rats to AAV-control-injected rats and compared parameters of energy balance to ad libitum fed AAV-NPY-injected rats. For 3 weeks, AAV-NPY-injected rats, received the same amount of food as ad libitum-fed rats injected with control rAAV They did not gain more body weight than these controls. When allowed access to food ad libitum, these AAV-NPY-injected rats increased food intake, which subsequently decreased when rats reached the same body weight as AAV-NPY-injected rats that were fed ad libitum for the entire study. These data indicate that overexpression of NPY in the PVN results in obesity by increasing food intake until a certain body weight is achieved.

    Topics: Adenoviridae; Animals; Body Temperature; Body Weight; Disease Models, Animal; Eating; Hyperphagia; Injections; Male; Neuropeptide Y; Obesity; Paraventricular Hypothalamic Nucleus; Rats; Rats, Wistar

2009
Hypothalamic proopiomelanocortin promoter methylation becomes altered by early overfeeding: an epigenetic model of obesity and the metabolic syndrome.
    The Journal of physiology, 2009, Oct-15, Volume: 587, Issue:Pt 20

    Pre- and neonatal overfeeding programmes a permanent obesity disposition and accompanying diabetic and cardiovascular disorders, by unknown mechanisms. We proposed that early overfeeding may alter DNA methylation patterns of hypothalamic promoter regions of genes critically involved in the lifelong regulation of food intake and body weight. We induced neonatal overfeeding by rearing Wistar rats in small litters (SL) and thereafter mapped the DNA methylation status of CpG dinucleotides of gene promoters from hypothalamic tissue, using bisulfite sequencing. Neonatal overfeeding led to rapid early weight gain, resulting in a metabolic syndrome phenotype, i.e. obesity, hyperleptinaemia, hyperglycaemia, hyperinsulinaemia, and an increased insulin/glucose ratio. Accompanying, without group difference to controls, the promoter of the main orexigenic neurohormone, neuropeptide Y, was methylated at low levels (i.e. < 5%). In contrast, in SL rats the hypothalamic gene promoter of the main anorexigenic neurohormone, proopiomelanocortin (POMC), showed hypermethylation (P < 0.05) of CpG dinucleotides within the two Sp1-related binding sequences (Sp1, NF-kappaB) which are essential for the mediation of leptin and insulin effects on POMC expression. Consequently, POMC expression lacked upregulation, despite hyperleptinaemia and hyperinsulinaemia. Accordingly, the extent of DNA methylation within Sp1-related binding sequences was inversely correlated to the quotients of POMC expression/leptin (P = 0.02) and POMC expression/insulin (P < 0.001), indicating functionality of acquired epigenomic alterations. These data for the first time demonstrate a nutritionally acquired alteration of the methylation pattern and, consequently, the regulatory 'set point' of a gene promoter that is critical for body weight regulation. Our findings reveal overfeeding as an epigenetic risk factor of obesity programming and consecutive diabetic and cardiovascular disorders and diseases, in terms of the metabolic syndrome.

    Topics: Animals; DNA; DNA Methylation; Epigenesis, Genetic; Gene Expression Regulation; Humans; Hyperphagia; Hypothalamus; Metabolic Diseases; Neuropeptide Y; Obesity; Pro-Opiomelanocortin; Promoter Regions, Genetic; Rats; Rats, Wistar

2009
In vivo characterization of high Basal signaling from the ghrelin receptor.
    Endocrinology, 2009, Volume: 150, Issue:11

    The receptor for the orexigenic peptide, ghrelin, is one of the most constitutively active 7TM receptors known, as demonstrated under in vitro conditions. Change in expression of a constitutively active receptor is associated with change in signaling independent of the endogenous ligand. In the following study, we found that the expression of the ghrelin receptor in the hypothalamus was up-regulated approximately 2-fold in rats both during 48-h fasting and by streptozotocin-induced hyperphagia. In a separate experiment, to probe for the effect of the high basal signaling of the ghrelin receptor in vivo, we used intracerebroventricular administration by osmotic pumps of a peptide [D-Arg(1), D-Phe(5), D-Trp(7,9), Leu(11)]-substance P. This peptide selectively displays inverse agonism at the ghrelin receptor as compared with an inactive control peptide with just a single amino acid substitution. Food intake and body weight were significantly decreased in the group of rats treated with the inverse agonist, as compared with the groups treated with the control peptide or the vehicle. In the hypothalamus, the expression of neuropeptide Y and uncoupling protein 2 was decreased by the inverse agonist. In a hypothalamic cell line that endogenously expresses the ghrelin receptor, we observed high basal activity of the cAMP response element binding protein, an important signaling transduction pathway for appetite regulation. The activation was further increased by ghrelin administration and decreased by administration of the inverse agonist. It is suggested that the high constitutive signaling activity is important for the in vivo function of the ghrelin receptor in the control of food intake and body weight.

    Topics: Animals; Body Weight; Eating; Gene Expression; Hyperphagia; Hypothalamus; Ion Channels; Male; Mitochondrial Proteins; Neuropeptide Y; Rats; Rats, Wistar; Receptors, Ghrelin; Signal Transduction; Streptozocin; Uncoupling Protein 2; Up-Regulation

2009
Blunted fasting-induced hypothalamic activation and refeeding hyperphagia in late-onset obesity.
    Neuroendocrinology, 2009, Volume: 90, Issue:4

    Hormonal and metabolic factors signal the status of energy balance to hypothalamic nuclei. Obesity is characterized by neuronal, metabolic and hormonal alterations. We therefore hypothesized that hypothalamic responses to challenges of energy balance may differ between lean and obese animals. To test this, we compared c-Fos expression in the hypothalamic arcuate (ARC) and paraventricular nuclei (PVN) and the lateral hypothalamic area (LHA) of mice (1-year-old) with late-onset obesity (LOO) and of lean controls under different feeding conditions. Fourteen hours of fasting induced high c-Fos expression in neuropeptide-Y-positive ARC neurons, in the PVN and in the rostral LHA in lean but not in LOO mice. c-Fos expression in melanin-concentrating hormone (MCH) and orexin-containing neurons in the caudal LHA was not affected by fasting. LOO mice showed fasting hyperinsulinemia, hyperleptinemia, elevated fasting blood glucose and an attenuated hyperphagic response during refeeding. Moreover, the anorectic response to leptin and hypoglycemic response to insulin were reduced in LOO mice. We conclude that adiposity blunts the neuronal responses to metabolic challenges in hypothalamic centers which control feeding behavior and energy balance. Elevated blood glucose may be one factor that suppresses hypothalamic responsiveness in obese mice. A similar impact of hyperinsulinemia and hyperleptinemia in LOO mice is also likely although under the current experimental conditions responsiveness to some effects of these hormones appeared to be reduced.

    Topics: Age of Onset; Animals; Arcuate Nucleus of Hypothalamus; Blood Glucose; Fasting; Green Fluorescent Proteins; Hyperinsulinism; Hyperphagia; Hypothalamic Area, Lateral; Intracellular Signaling Peptides and Proteins; Leptin; Male; Melanins; Mice; Mice, Inbred C57BL; Mice, Transgenic; Neurons; Neuropeptide Y; Neuropeptides; Obesity; Orexins; Paraventricular Hypothalamic Nucleus; Proto-Oncogene Proteins c-fos

2009
Hyperphagia and obesity produced by arcuate injection of NPY-saporin do not require upregulation of lateral hypothalamic orexigenic peptide genes.
    Peptides, 2008, Volume: 29, Issue:10

    Neuropeptide Y (NPY) conjugated with a ribosomal inactivating toxin, saporin (SAP), is a toxin that targets NPY receptor-expressing cells. Injection of NPY-SAP into the rat arcuate nucleus (Arc) and basomedial hypothalamus (BMH) destroys two populations of NPY-receptor-expressing neurons important for the control of food intake and body weight, NPY and pro-opiomelanocortin (POMC) and cocaine and amphetamine related transcript (CART) neurons, and produces profound hyperphagia and obesity. Here, we investigated the contribution of lateral hypothalamus (LHA) orexigenic peptides, orexins and melanocortin concentrating hormone (MCH), to these lesion effects. We microinjected NPY-SAP into two sites on each side of the Arc, causing a loss of NPY and POMC/CART neurons that was limited to the Arc. Lesioned rats rapidly became hyperphagic and obese. However, MCH and prepro-orexin mRNA expression were not increased in the LHA in the lesioned rats, but were decreased at some levels of the LHA or were unchanged. NPY-SAP-induced obesity therefore differs from dietary obesity and from obesity associated with leptin or leptin receptor deficiency in which MCH gene expression is increased. The Arc NPY-SAP lesion produces obesity and hyperphagia that does not require overexpression of hypothalamic neuropeptides currently considered to provide major stimulatory drive for food intake: NPY, agouti gene-related protein, MCH or orexins. The source of the seemingly unregulated stimulatory drive for feeding in these animals has not been identified, but may be associated with hindbrain or endocrine mechanisms.

    Topics: Animals; Arcuate Nucleus of Hypothalamus; Body Weight; Eating; Female; Hyperphagia; Hypothalamic Area, Lateral; Hypothalamic Hormones; Intracellular Signaling Peptides and Proteins; Male; Melanins; Microinjections; Nerve Tissue Proteins; Neurons; Neuropeptide Y; Neuropeptides; Obesity; Orexins; Pituitary Hormones; Protein Precursors; Rats; Rats, Sprague-Dawley; Up-Regulation

2008
Sustained hyperphagia in adolescent rats that experienced neonatal maternal separation.
    International journal of obesity (2005), 2008, Volume: 32, Issue:9

    To examine the neurobiological basis of bingeing-related eating disorders using an animal model system.. Sprague-Dawley pups were separated from dam for 3 h daily during the first two weeks of birth (maternal separation (MS)), or left undisturbed (non-handled (NH)). Pups were subjected to repeated fasting/refeeding (RF) cycles; that is, 24 h food deprivation and 24 h RF (NH/RF or MS/RF), or had free access to food and water (NH/fed control (FC) or MS/FC) from postnatal day (PND) 28-40.. Body weight gain and food intake were recorded. The arcuate expression of neuropeptide Y (NPY) and plasma corticosterone levels were analyzed on PND 29 and 40.. Decrease in weight gain by repeated fasting/RF cycles was smaller in MS pups than in NH. Interestingly, weight changes responding to fasting or RF increased in MS/RF compared with NH/RF. Compensatory hyperphagia was diminished in NH/RF after the third fasting trial, but persisted in MS/RF throughout the experimental period. The arcuate expression of NPY mRNA responding to food deprivation was blunted, but elevation of plasma corticosterone exaggerated, in the MS group, compared to the NH group, on PND 29 after the first fasting session. However, both the arcuate NPY mRNA and plasma corticosterone levels were increased in MS/RF, but not in NH/RF, on PND 40 after the six sets of fasting/RF cycles, compared to the free FC groups.. Experience of neonatal MS may lead to an exaggerated feeding response to repeated fasting/RF challenges at adolescence, perhaps, due to increased responsiveness of the hypothalamic-pituitary-adrenal gland axis. Additionally, the results suggested that an increased action of the hypothalamic NPY may not be necessary to induce compensatory hyperphagia following food deprivation.

    Topics: Animals; Animals, Newborn; Corticosterone; Eating; Fasting; Female; Hyperphagia; Hypothalamo-Hypophyseal System; Male; Maternal Deprivation; Models, Animal; Neuropeptide Y; Pituitary-Adrenal System; Rats; Rats, Sprague-Dawley; Weight Gain

2008
Sleep deprivation of rats: the hyperphagic response is real.
    Sleep, 2008, Volume: 31, Issue:7

    Chronic sleep deprivation of rats causes hyperphagia without body weight gain. Sleep deprivation hyperphagia is prompted by changes in pathways governing food intake; hyperphagia may be adaptive to sleep deprivation hypermetabolism. A recent paper suggested that sleep deprivation might inhibit ability of rats to increase food intake and that hyperphagia may be an artifact of uncorrected chow spillage. To resolve this, a palatable liquid diet (Ensure) was used where spillage is insignificant.. Sleep deprivation of male Sprague Dawley rats was enforced for 10 days by the flowerpot/platform paradigm. Daily food intake and body weight were measured. On day 10, rats were transcardially perfused for analysis of hypothalamic mRNA expression of the orexigen, neuropeptide Y (NPY).. Morgan State University, sleep deprivation and transcardial perfusion; University of Maryland, NPY in situ hybridization and analysis.. Using a liquid diet for accurate daily measurements, there was no change in food intake in the first 5 days of sleep deprivation. Importantly, from days 6-10 it increased significantly, peaking at 29% above baseline. Control rats steadily gained weight but sleep-deprived rats did not. Hypothalamic NPY mRNA levels were positively correlated to stimulation of food intake and negatively correlated with changes in body weight.. Sleep deprivation hyperphagia may not be apparent over the short term (i.e., < or = 5 days), but when extended beyond 6 days, it is readily observed. The timing of changes in body weight and food intake suggests that the negative energy balance induced by sleep deprivation prompts the neural changes that evoke hyperphagia.

    Topics: Animals; Body Weight; Energy Metabolism; Gene Expression Regulation; Hyperphagia; Hypothalamus; Male; Neuropeptide Y; Rats; Rats, Sprague-Dawley; RNA, Messenger; Sleep Deprivation

2008
Role of ghrelin in the relationship between hyperphagia and accelerated gastric emptying in diabetic mice.
    Gastroenterology, 2008, Volume: 135, Issue:4

    Ghrelin is an orexigenic peptide with gastroprokinetic effects. Mice with streptozotocin (STZ)-induced diabetes exhibit hyperphagia, altered gastric emptying, and increased plasma ghrelin levels. We investigated the causative role of ghrelin herein by comparing changes in ghrelin receptor knockout (growth hormone secretagogue receptor [GHS-R](-/-)) and wild-type (GHS-R(+/+)) mice with STZ-induced diabetes.. Gastric emptying was measured with the [(13)C]octanoic acid breath test. The messenger RNA (mRNA) expression of neuropeptide Y (NPY), agouti-related peptide (AgRP), and proopiomelanocortin was quantified by real-time reverse-transcription polymerase chain reaction. Neural contractions were elicited by electrical field stimulation in fundic smooth muscle strips.. Diabetes increased plasma ghrelin levels to a similar extent in both genotypes. Hyperphagia was more pronounced in GHS-R(+/+) than in GHS-R(-/-) mice between days 12 and 21. Increases in NPY and AgRP mRNA expression were less pronounced in diabetic GHS-R(-/-) than in GHS-R(+/+) mice from day 15 on, whereas decreases in proopiomelanocortin mRNA levels were similar in both genotypes. Gastric emptying was accelerated to a similar extent in both genotypes, starting on day 16. In fundic smooth muscle strips of diabetic GHS-R(+/+) and GHS-R(-/-) mice, neuronal relaxations were reduced, whereas contractions were increased; this increase was related to an increased affinity of muscarinic and tachykinergic receptors.. Diabetic hyperphagia is regulated by central mechanisms in which the ghrelin-signaling pathway affects the expression of NPY and AgRP in the hypothalamus. The acceleration of gastric emptying, which is not affected by ghrelin signaling, is not the cause of diabetic hyperphagia and probably involves local contractility changes in the fundus.

    Topics: Acetylcholine; Agouti-Related Protein; Animals; Blood Glucose; Body Weight; Cholinergic Agents; Diabetes Mellitus, Experimental; Eating; Gastric Emptying; Gastric Fundus; Ghrelin; Hyperphagia; Hypothalamus; Male; Mice; Mice, Knockout; Muscle Contraction; Neuropeptide Y; Neurotransmitter Agents; Pro-Opiomelanocortin; Receptors, Ghrelin; RNA, Messenger; Substance P

2008
Early undernutrition leads to long-lasting reductions in body weight and adiposity whereas increased intake increases cardiac fibrosis in male rats.
    The Journal of nutrition, 2008, Volume: 138, Issue:9

    Previous studies suggest that both overfeeding and undernutrition during development increase the risk of obesity and hypertension in adulthood. In this study, we examined both short- (24 d) and long- (16 wk) term effects of early postnatal over- and underfeeding in rats on body weight, body composition, plasma hormones, adiposity markers, and hypothalamic neuropeptide Y content. Cardiovascular changes were also examined by measuring blood pressure and cardiac fibrosis. Rats raised in litters of 3, 12, or 18 pups per mother were used to model early onset overfeeding, control, and underfeeding, respectively. At 24 d of age, pups raised in small litters (SL) were 10% heavier than pups from normal litters, accompanied by increased organ mass and fat mass, elevated plasma leptin, corticosterone, and uncoupling protein-1 mRNA in brown adipose tissue. On the other hand, pups raised in large litters were 17% lighter with no significant changes in plasma leptin. Overfeeding during the first 3 wk of life led to increased plasma leptin concentration in adulthood, whereas underfed rats remained significantly lighter throughout the study, with no evidence of catch-up growth. Rats raised in SL were more susceptible to developing cardiac fibrosis with a 22% increase in collagen deposition compared with control rats at 16 wk of age (P < 0.05). This was independent of any changes in blood pressure. This study demonstrates that nutritional changes early in postnatal development can have long-lasting effects on body weight, adiposity, and some mediators involved in energy homeostasis and can also lead to structural changes in the heart in adulthood. This highlights the importance of identifying potential early life risk factors involved in the modulation of childhood nutrition.

    Topics: Adiposity; Aging; Animals; Animals, Newborn; Blood Pressure; Body Weight; Corticosterone; Energy Intake; Female; Fibrosis; Food Deprivation; Heart Diseases; Hyperphagia; Hypothalamus; Insulin; Leptin; Litter Size; Longitudinal Studies; Male; Neuropeptide Y; Obesity; Random Allocation; Rats; Rats, Sprague-Dawley

2008
Fluvoxamine inhibits weight gain and food intake in food restricted hyperphagic Wistar rats.
    Biological & pharmaceutical bulletin, 2008, Volume: 31, Issue:12

    The effects of fluvoxamine, a selective serotonin reuptake inhibitor (SSRI), were studied in normophagic and food-restriction-induced hyperphagic middle-aged Wistar rats. Normophagic intact Wistar rats were given fluvoxamine (100 mg/kg/d, per os (p.o.)) or vehicle for 10 d. Hyperphagic middle-aged Wistar rats were subjected to 10 d of food restriction; they were allowed to refeed for 10 d, with ad libitum food access and administered fluvoxamine (100 mg/kg/d, p.o.) or vehicle during the 10-d refeeding period. Fluvoxamine administration to normophagic middle-aged Wistar rats affected neither their weight nor food intake. However, administration to food-restricted rats showed inhibitory effects of weight gain and food intake during 10 d of refeeding. Fluvoxamine-treated rats showed significantly lower neuropeptide Y (NPY) immunostaining levels in the paraventricular nucleus (PVN) and dorsomedial hypothalamic nucleus (DMH) than untreated controls. Hypophagic and weight-inhibiting effects of fluvoxamine might be mediated via decreased NPY in PVN and DMH. These results suggest that the appetite-controlling effect of fluvoxamine might be responsive to the rats' appetite condition.

    Topics: Animals; Antidepressive Agents, Second-Generation; Body Weight; Eating; Fluvoxamine; Hyperphagia; Hypothalamus; Male; Neuropeptide Y; Rats; Rats, Wistar; Weight Gain

2008
Brain neuropeptide Y and CCK and peripheral adipokine receptors: temporal response in obesity induced by palatable diet.
    International journal of obesity (2005), 2008, Volume: 32, Issue:2

    Palatable food disrupts normal appetite regulation, which may contribute to the etiology of obesity. Neuropeptide Y (NPY) and cholecystokinin play critical roles in the regulation of food intake and energy homeostasis, while adiponectin and carnitine palmitoyltransferase (CPT) are important for insulin sensitivity and fatty acid oxidation. This study examined the impact of short- and long-term consumption of palatable high-fat diet (HFD) on these critical metabolic regulators.. Male C57BL/6 mice were exposed to laboratory chow (12% fat), or cafeteria-style palatable HFD (32% fat) for 2 or 10 weeks. Body weight and food intake were monitored throughout. Plasma leptin, hypothalamic NPY and cholecystokinin, and mRNA expression of leptin, adiponectin, their receptors and CPT-1, in fat and muscles were measured.. Caloric intake of the palatable HFD group was 2-3 times greater than control, resulting in a 37% higher body weight. Fat mass was already increased at 2 weeks; plasma leptin concentrations were 2.4 and 9 times higher than control at 2 and 10 weeks, respectively. Plasma adiponectin was increased at 10 weeks. Muscle adiponectin receptor 1 was increased at 2 weeks, while CPT-1 mRNA was markedly upregulated by HFD at both time points. Hypothalamic NPY and cholecystokinin content were significantly decreased at 10 weeks.. Palatable HFD induced hyperphagia, fat accumulation, increased adiponectin, leptin and muscle fatty acid oxidation, and reduced hypothalamic NPY and cholecystokinin. Our data suggest that the adaptive changes in hypothalamic NPY and muscle fatty acid oxidation are insufficient to reverse the progress of obesity and metabolic consequences induced by a palatable HFD.

    Topics: Adiponectin; Animals; Body Weight; Cholecystokinin; Dietary Fats; Eating; Energy Metabolism; Hyperphagia; Leptin; Male; Mice; Mice, Inbred C57BL; Neuropeptide Y; Obesity; Receptors, Adiponectin; RNA, Messenger

2008
Distinctive role of central histamine H3 receptor in various orexigenic pathways.
    European journal of pharmacology, 2008, Jan-28, Volume: 579, Issue:1-3

    Despite the well-established role of histamine as an anorexigenic neurotransmitter, the role of histamine H(3) receptors in feeding behavior is controversial. Herein we investigated the role of histamine H(3) receptor on several orexigenic agents in mice. Thioperamide (histamine H(3) receptor inverse agonist) inhibited neuropeptide Y- and nociceptin-induced hyperphagia but had no effect on U-50488 (opioid kappa-receptor agonist)-induced hyperphagia. In contrast, imetit (histamine H(3) receptor agonist) inhibited U-50488-induced hyperphagia but augmented neuropeptide Y-induced hyperphagia while it did not alter nociceptin-induced hyperphagia. These results indicate distinctive roles of histamine H(3) receptors in various orexigenic pathways.

    Topics: 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer; Animals; Appetite; Histamine Agonists; Histamine H3 Antagonists; Hyperphagia; Imidazoles; Male; Mice; Mice, Inbred C57BL; Neuropeptide Y; Nociceptin; Opioid Peptides; Piperidines; Receptors, Histamine H3; Thiourea

2008
Collective and individual functions of leptin receptor modulated neurons controlling metabolism and ingestion.
    Endocrinology, 2008, Volume: 149, Issue:4

    Two known types of leptin-responsive neurons reside within the arcuate nucleus: the agouti gene-related peptide (AgRP)/neuropeptide Y (NPY) neuron and the proopiomelanocortin (POMC) neuron. By deleting the leptin receptor gene (Lepr) specifically in AgRP/NPY and/or POMC neurons of mice, we examined the several and combined contributions of these neurons to leptin action. Body weight and adiposity were increased by Lepr deletion from AgRP and POMC neurons individually, and simultaneous deletion in both neurons (A+P LEPR-KO mice) further increased these measures. Young (periweaning) A+P LEPR-KO mice exhibit hyperphagia and decreased energy expenditure, with increased weight gain, oxidative sparing of triglycerides, and increased fat accumulation. Interestingly, however, many of these abnormalities were attenuated in adult animals, and high doses of leptin partially suppress food intake in the A+P LEPR-KO mice. Although mildly hyperinsulinemic, the A+P LEPR-KO mice displayed normal glucose tolerance and fertility. Thus, AgRP/NPY and POMC neurons each play mandatory roles in aspects of leptin-regulated energy homeostasis, high leptin levels in adult mice mitigate the importance of leptin-responsiveness in these neurons for components of energy balance, suggesting the presence of other leptin-regulated pathways that partially compensate for the lack of leptin action on the POMC and AgRP/NPY neurons.

    Topics: Agouti-Related Protein; Animals; Body Composition; Eating; Energy Metabolism; Fertility; Hyperinsulinism; Hyperphagia; Lactation; Male; Mice; Neuropeptide Y; Pro-Opiomelanocortin; Receptors, Leptin

2008
Feeding and stress interact through the serotonin 2C receptor in developing mice.
    Physiology & behavior, 2008, Jul-05, Volume: 94, Issue:4

    Feeding and stress neurocircuits are intertwined. Among the neurotransmitters and receptors common to both circuits, the serotonin 2C receptor is particularly intriguing because its distribution is limited to the central nervous system. Hence, deficits in energy balance and stress responses in mice lacking this gene are likely due to defects in central regulation. The phenotype of the serotonin 2C receptor null (KO) mouse is adult-onset hyperphagia, depressed metabolic rate, and disruption in satiety, with a progression to midlife obesity. A provocative feature of this obese model is our recent finding of a childhood component where the KO mouse is heavier at weaning, a distinction that only returns in adulthood. To determine when the KO mouse becomes heavier, longitudinal and cross-sectional timecourse studies followed weight gain and found significantly heavier body weight, higher plasma leptin, and rectal temperature, only in unhandled KO compared to sibling wildtype controls. To map what metabolic compensations cause the KO weight increase, we launched thermal and behavioral studies in 10 day old mice before there was any genotype difference in body weight, corticosterone levels, or the levels leptin during the developmental leptin peak. The heavier KO weanling is, in part, explained by hyperphagia, lower metabolic rate and activity, and behavioral thermogenesis measured at 10 days of age. However, the infant KO mouse is stress-sensitive and growth is impaired with handling. The serotonin 2C receptor has a role in fine-tuning energetic and stress demands even as neurocircuits are developing, and unbalanced compensations in infancy may program responses in adulthood that are "off target" from optimal function.

    Topics: Adiponectin; Age Factors; Animals; Animals, Newborn; Appetite Regulation; Body Temperature Regulation; Corticosterone; Cross-Sectional Studies; Feeding Behavior; Female; Hyperphagia; Insulin; Leptin; Longitudinal Studies; Male; Mice; Mice, Knockout; Neuropeptide Y; Phenotype; Receptor, Serotonin, 5-HT2C; Stress, Psychological; Weaning; Weight Gain

2008
Evidence for increased neuropeptide Y synthesis in mediobasal hypothalamus in relation to parental hyperphagia and gonadal activation in breeding ring doves.
    Journal of neuroendocrinology, 2007, Volume: 19, Issue:3

    Like lactating mammals, male and female ring dove parents increase their food consumption to meet the energetic challenges of provisioning their young. To clarify the neurochemical mechanisms involved, the present study investigated the relationship between parental hyperphagia and changes in activity of the potent orexigen neuropeptide Y (NPY) in the hypothalamus of breeding doves. Changes in NPY-immunoreactive (NPY-ir) cell numbers in the tuberal hypothalamus of male and female doves were examined by immunocytochemistry at six stages of the breeding cycle. Parallel NPY mRNA measurements were recorded in mediobasal hypothalamus (which includes the tuberal hypothalamus) by semiquantitative reverse transcription-polymerase chain reaction using 18S rRNA as the internal standard. NPY mRNA changes were also measured in the mediobasal hypothalamus of nonbreeding doves following intracranial administration of prolactin, an orexigenic hormone that is elevated in the plasma of parent doves, and in response to food deprivation, which mimics the negative energy state that develops in parents as they provision their growing young. NPY-ir cell numbers in the tuberal hypothalamus and NPY mRNA levels in the mediobasal hypothalamus were significantly higher in breeding males and females during the period of parental hyperphagia after hatching than during the late incubation period when food intake remains unchanged. In nonbreeding doves, food deprivation and prolactin treatment increased NPY mRNA in this region by two- to three-fold, which suggests that NPY expression is sensitive to hormonal and metabolic signals associated with parenting. We conclude that NPY synthesis is increased in the mediobasal hypothalamus during the posthatching period, which presumably supports increased NPY release and resulting parental hyperphagia. NPY-ir and mRNA were also high in the mediobasal hypothalamus prior to egg laying when food intake remained unchanged. Several lines of evidence suggest that this elevation in NPY supports the increased gonadal activity that accompanies intense courtship and nest building interactions in breeding doves.

    Topics: Animals; Columbidae; Energy Metabolism; Feeding Behavior; Female; Gene Expression Regulation; Gonads; Hyperphagia; Hypothalamo-Hypophyseal System; Hypothalamus; Male; Maternal Behavior; Neuropeptide Y; Paternal Behavior; Prolactin; Reproduction; RNA, Messenger; Statistics, Nonparametric

2007
Prenatal influences on susceptibility to diet-induced obesity are mediated by altered neuroendocrine gene expression.
    The Journal of endocrinology, 2007, Volume: 193, Issue:1

    The escalating rates of obesity and type 2 diabetes have reached pandemic proportions. It has been proposed that the risk of developing metabolic disorders in adult life is influenced by environmental factors, which operate during the early periods of development. We have previously shown that an interaction between the prenatal and the postnatal dietary environment amplifies the propensity towards diet-induced obesity, although the mechanisms are unclear. In the present study, we investigated the interaction between prenatal undernutrition and postnatal high-fat nutrition on key genes of the hypothalamic appetite regulatory network. Pregnant Wistar rats were fed a standard chow diet either ad libitum (AD) or at 30% of AD intake throughout gestation (UN). From weaning, female AD and UN offspring were fed either a standard chow (ADC n = 8, UNC n = 8) or a high-fat diet (45% kcal as fat; ADHF n = 8, UNHF n = 8) ad libitum for the remainder of the study. At 24 weeks of age, body composition was assessed by dual energy X-ray absorptiometry analysis and total RNA was extracted from whole rat hypothalami. Real-time PCR was performed to characterise pro-opiomelanocortin (POMC), neuropeptide Y (NPY), agouti-related protein (AgRP) and OBRb gene expression at the mRNA level. Our results demonstrate that the amplification of postnatal obesity develops as a consequence of an interaction between prenatal under-nutrition and postnatal high-fat nutrition. This phenotype also shows significant alterations in POMC, NPY, AgRP and OBRb gene expression together with elevations in circulating levels of both plasma leptin and insulin. These findings are consistent with the predictive adaptive response hypothesis that neuroendocrine development during fetal life may be based on predictions about postnatal environmental conditions. Increased susceptibility to diet-induced obesity develops if a mismatch between the anticipated and the actual conditions are encountered.

    Topics: Agouti Signaling Protein; Agouti-Related Protein; Animals; Appetite Regulation; Body Composition; Dietary Fats; Disease Susceptibility; Female; Gene Expression; Hyperphagia; Hypothalamus; Insulin; Intercellular Signaling Peptides and Proteins; Leptin; Malnutrition; Maternal Nutritional Physiological Phenomena; Neuropeptide Y; Obesity; Pregnancy; Prenatal Exposure Delayed Effects; Pro-Opiomelanocortin; Rats; Rats, Wistar; RNA, Messenger

2007
A role for brain-specific homeobox factor Bsx in the control of hyperphagia and locomotory behavior.
    Cell metabolism, 2007, Volume: 5, Issue:6

    Food intake and activity-induced thermogenesis are important components of energy balance regulation. The molecular mechanism underlying the coordination of food intake with locomotory behavior to maintain energy homeostasis is unclear. We report that the brain-specific homeobox transcription factor Bsx is required for locomotory behavior, hyperphagia, and expression of the hypothalamic neuropeptides Npy and Agrp, which regulate feeding behavior and body weight. Mice lacking Bsx exhibit reduced locomotor activity and lower expression of Npy and Agrp. They also exhibit attenuated physiological responses to fasting, including reduced increase of Npy/Agrp expression, lack of food-seeking behavior, and reduced rebound hyperphagia. Furthermore, Bsx gene disruption rescues the obese phenotype of leptin-deficient ob/ob mice by reducing their hyperphagia without increasing their locomotor activity. Thus, Bsx represents an essential factor for NPY/AgRP neuronal function and locomotory behavior in the control of energy balance.

    Topics: Agouti-Related Protein; Animals; Behavior, Animal; Body Weight; Brain; Cells, Cultured; Chromatin Immunoprecipitation; Energy Intake; Energy Metabolism; Fasting; Homeodomain Proteins; Hyperphagia; Immunoenzyme Techniques; In Situ Hybridization; Locomotion; Luciferases; Mice; Mice, Inbred C57BL; Mice, Obese; Nerve Tissue Proteins; Neuropeptide Y; Phenotype; Reverse Transcriptase Polymerase Chain Reaction; RNA, Messenger

2007
Sympathetic nervous system activity may link hyperphagia and fat deposition in human obesity.
    American journal of physiology. Endocrinology and metabolism, 2007, Volume: 293, Issue:4

    Topics: Adipose Tissue; Adipose Tissue, Brown; Humans; Hyperphagia; Neuropeptide Y; Obesity; Receptors, Neuropeptide Y; Sympathetic Nervous System

2007
Effects of neuropeptide Y antagonists on food intake in rats: differences with cold-adaptation.
    Peptides, 2006, Volume: 27, Issue:1

    Hyperphagia followed both central neuropeptide Y (NPY) administration and the presumed increase of endogenous NPY activity after food deprivation. NPY induced greater hyperphagia in cold-adapted than non-adapted rats; fasting of comparable severity caused similar hyperphagia in the two groups. NPY-receptor-antagonist D-Tyr(27,36), D-Thr32-NPY(27,36) or functional NPY-antagonist D-myo-inositol-1,2,6-trisphosphate attenuated the hyperphagic effect of both NPY and fasting in non-adapted rats. However, while completely preventing the NPY-hyperphagia, they did not influence the fasting-induced hyperphagia in cold-adapted rats. With cold-adaptation the sensitivity to NPY and to its antagonists increases, but the hypothalamic NPY loses from its fundamental role in the regulation of food intake, and the hyperphagia seen in cold-adaptation may need some other explanation.

    Topics: Animals; Body Weight; Cold Temperature; Feeding Behavior; Female; Food Deprivation; Hyperphagia; Injections, Intraventricular; Inositol Phosphates; Neuropeptide Y; Peptide Fragments; Rats; Rats, Wistar

2006
Increased infarct size and lack of hyperphagic response after focal cerebral ischemia in peroxisome proliferator-activated receptor beta-deficient mice.
    Journal of cerebral blood flow and metabolism : official journal of the International Society of Cerebral Blood Flow and Metabolism, 2006, Volume: 26, Issue:3

    Peroxisome proliferator-activated receptors (PPARs) are involved in energy expenditure, regulation of inflammatory processes, and cellular protection in peripheral tissues. Among the different types of PPARs, PPARbeta is the only one to be widely expressed in cortical neurons. Using PPARbeta knockout (KO) mice, we report here a detailed investigation of the role of PPARbeta in cerebral ischemic damage, associated inflammatory and antioxidant processes as well as food intake regulation after middle cerebral artery occlusion (MCAO). The PPARbeta KO mice had a two-fold increase in infarct size compared with wild-type (WT) mice. Brain oxidative stress was dramatically enhanced in these KO mice, as documented by an increased content of malondialdehyde, decreased levels of glutathione and manganese superoxide dismutase, and no induction of uncoupling protein 2 (UCP2) mRNA. Unlike WT mice, PPARbeta KO mice showed a marked increase of prooxidant interferon-gamma but no induction of nerve growth factor and tumor necrosis factor alpha after MCAO. In WT mice, MCAO resulted in inflammation-specific transient hyperphagia from day 3 to day 5 after ischemia, which was associated with an increase in neuropeptide Y (NPY) mRNA. This hyperphagic phase and NPY mRNA induction were not observed in PPARbeta KO mice. Furthermore, our study also suggests for the first time that UCP2 is involved in MCAO food intake response. These data indicate that PPARbeta plays an important role in integrating and regulating central inflammation, antioxidant mechanisms, and food intake after MCAO, and suggest that the use of PPARbeta agonists may be of interest for the prevention of central ischemic damage.

    Topics: Animals; Brain Ischemia; Cerebral Infarction; Disease Models, Animal; Gene Expression Profiling; Glutathione; Hyperphagia; Infarction, Middle Cerebral Artery; Interferon-gamma; Ion Channels; Lipid Peroxidation; Male; Membrane Transport Proteins; Mice; Mice, Inbred C57BL; Mice, Knockout; Mitochondrial Proteins; Nerve Growth Factor; Neuropeptide Y; PPAR-beta; RNA, Messenger; Superoxide Dismutase; Uncoupling Protein 2

2006
Inhibitory effect of 5-hydroxytryptamine on hyperphagia in mice with genetic overexpression of neuropeptide Y.
    Neuroscience letters, 2006, Feb-20, Volume: 394, Issue:3

    The present study examined the effect of 5-hydroxytraptamine (5-HT) on the feeding behavior of transgenic mice with neuropeptide Y (NPY) overexpression. Solution of 5-HT (1, 2.5 or 5 mg/kg) was administered intraperitoneally into (1) NPY-overexpressing mice, and (2) wild-type mice with 2-deoxy-d-glucose (2-DG) induced hyperphagia. The NPY-overexpressing mice were further divided into five groups: (1) control mice, (2) mice treated with 5-HT (5 mg/kg), (3) mice treated with 5-HT (5 mg/kg) and ketanserin (0.5 or 1 mg/kg), a 5-HT2A receptor antagonist, (4) mice treated with insulin (1 IU/kg), and (5) mice treated with insulin (1 IU/kg) and 5-HT (5 mg/kg). Food intake and plasma glucose levels were measured. The results showed that 5-HT reduced hyperphagia in both NPY-overexpressing mice and 2-DG-treated mice in dose-dependent manner. Hyperglycemia was induced by 5-HT administration. Ketanserin antagonized the 5-HT induced hypophagia and hyperglycemia. Insulin, on the other hand, prevented 5-HT induced hyperglycemia but not the hypophagic effect. In conclusion, 5-HT reduces hyperphagia in the NPY-overexpressing rat through action on 5-HT2A receptors and this hypophagic effect of 5-HT does not depend on the hyperglycemia.

    Topics: Animals; Antimetabolites; Blood Glucose; Deoxyglucose; Dose-Response Relationship, Drug; Eating; Hyperphagia; Hypoglycemic Agents; Insulin; Ketanserin; Male; Mice; Mice, Transgenic; Neuropeptide Y; Serotonin; Serotonin Antagonists

2006
Role of endogenous ghrelin in the hyperphagia of mice with streptozotocin-induced diabetes.
    Endocrinology, 2006, Volume: 147, Issue:6

    Ghrelin is an orexigenic peptide involved in the regulation of energy homeostasis. To investigate the role of ghrelin in the hyperphagia associated with uncontrolled streptozotocin-induced diabetes, food intake was followed in diabetic ghrelin knockout (ghrelin(-/-)) and control wild-type (ghrelin(+/+)) mice and diabetic Naval Medical Research Institute noninbred Swiss mice treated with either saline or the ghrelin receptor antagonist, D-Lys3-GH-releasing peptide-6 (D-Lys3-GHRP-6) for 5 d. In diabetic ghrelin(-/-) mice, hyperphagia was attenuated, and the maximal increase in food intake was 50% lower in mutant than in wild-type mice. The increased food intake observed during the light period (1000-1200 h) in ghrelin(+/+) mice was abolished in mutant mice. Diabetic ghrelin(-/-) mice lost 12.4% more body weight than ghrelin(+/+) mice. In diabetic ghrelin(+/+) mice, but not in ghrelin(-/-) mice, the number of neuropeptide Y (NPY)-immunoreactive neurons was significantly increased. Diabetic Naval Medical Research Institute noninbred Swiss mice were hyperphagic and had increased plasma ghrelin levels. Treatment with D-Lys3-GHRP-6 reduced daily food intake by 23% and reversed the increased food intake observed during the light period. The change in the number of NPY- (2.4-fold increase) and alpha-MSH (1.7-fold decrease)-immunoreactive hypothalamic neurons induced by diabetes was normalized by D-Lys3-GHRP-6 treatment. Our results suggest that enhanced NPY and reduced alpha-MSH expression are secondary to the release of ghrelin, which should be considered the underlying trigger of hyperphagia associated with uncontrolled diabetes.

    Topics: alpha-MSH; Animals; Arcuate Nucleus of Hypothalamus; Blood Glucose; Body Weight; Diabetes Mellitus, Experimental; Ghrelin; Glucagon; Hyperphagia; Male; Mice; Mice, Inbred C57BL; Neuropeptide Y; Oligopeptides; Peptide Hormones; Streptozocin

2006
Orosensory stimulation is sufficient and postingestive negative feedback is not necessary for neuropeptide Y to increase sucrose intake.
    Physiology & behavior, 2006, Apr-15, Volume: 87, Issue:4

    Although central administration of neuropeptide Y (NPY) has a potent orexic effect, it is not clear how NPY changes the potency of peripheral feedbacks from the gut to prolong eating and increase meal size. It has been suggested that NPY increases the stimulating effect of orosensory sweet stimuli or that it decreases the inhibitory effect of postingestive stimuli. To clarify this issue, we compared the orexic effect of NPY (2 microg) injected into the third ventricle of the brain on the volume and microstructure of intake of 0.8M sucrose during sham feeding (SF) and real feeding (RF) in male Sprague Dawley rats. The rationale for this comparison is that orosensory stimulation occurs in SF and RF, but postingestive negative feedback is present only in RF. NPY increased the volume ingested and the rate and number of clusters of licking significantly more in SF than in RF. This demonstrates that orosensory sucrose stimulation is sufficient and postingestive negative feedback is not necessary for the orexic effect of NPY under these experimental conditions.

    Topics: Analysis of Variance; Animals; Appetite; Appetite Regulation; Eating; Feedback, Psychological; Feeding Behavior; Hyperphagia; Injections, Intraventricular; Male; Neuropeptide Y; Postprandial Period; Rats; Rats, Sprague-Dawley; Sucrose; Taste

2006
Hypothalamic neuropeptide Y (NPY) and the attenuation of hyperphagia in streptozotocin diabetic rats treated with dopamine D1/D2 agonists.
    British journal of pharmacology, 2006, Volume: 148, Issue:5

    1. Dopamine is an appetite suppressant, while neuropeptide Y (NPY), an appetite stimulant in the brain, is reported to be involved in anorectic action induced by a combined administration of D1/D2 agonists in normal rats. In diabetic rats, however, these factors have not been studied. 2. Rats (including normal, diabetic and insulin-treated diabetic rats) were given daily injections of saline or D1/D2 agonists for 6 days. Changes in food intake and hypothalamic NPY content of these rats were assessed and compared. 3. The D1/D2 agonist-induced anorectic responses were altered in diabetic rats compared to normal rats treated similarly. Both the anorectic response on the first day of dosing and the tolerant response on the subsequent days were attenuated. 4. This alteration was independent of the neuroendocrine disturbance on feeding behavior since the basic pattern of food intake during the time course of a 24-h day/night cycle was similar in normal and diabetic rats; the decrease of food intake following drug treatment was only shown at the initial interval of 0-6 h in both groups of rats. 5. However, this alteration coincided with changes in NPY content following D1/D2 coadministration. The replacement of insulin in diabetic rats could normalize both NPY content and D1/D2 agonist-induced anorexia. 6. It is demonstrated that the response of D1/D2 agonist-induced appetite suppression is attenuated in diabetic rats compared to normal rats and that elevated hypothalamic NPY content may contribute to this alteration.

    Topics: 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine; Animals; Appetite Depressants; Diabetes Mellitus, Experimental; Dopamine Agonists; Drug Administration Routes; Drug Therapy, Combination; Eating; Feeding Behavior; Hormone Replacement Therapy; Hyperphagia; Hypothalamus; Injections; Insulin; Male; Neuropeptide Y; Quinpirole; Rats; Rats, Wistar; Receptors, Dopamine D1; Receptors, Dopamine D2; Time Factors

2006
Diabetic hyperphagia--ghrelin in the driver's seat.
    Endocrinology, 2006, Volume: 147, Issue:6

    Topics: Animals; Arcuate Nucleus of Hypothalamus; Diabetes Mellitus, Experimental; Ghrelin; Hyperphagia; Mice; Neuropeptide Y; Peptide Hormones; Receptors, G-Protein-Coupled; Receptors, Ghrelin; Streptozocin

2006
11 beta-hydroxysteroid dehydrogenase type 1 induction in the arcuate nucleus by high-fat feeding: A novel constraint to hyperphagia?
    Endocrinology, 2006, Volume: 147, Issue:9

    11 beta-Hydroxysteroid dehydrogenase type 1 (11 beta-HSD1) catalyzes regeneration of active intracellular glucocorticoids in fat, liver, and discrete brain regions. Although overexpression of 11 beta-HSD1 in adipose tissue causes hyperphagia and the metabolic syndrome, male 11 beta-HSD1 null (11 beta-HSD1-/-) mice resist metabolic disease on high-fat (HF) diet, but also show hyperphagia. This suggests 11 beta-HSD1 may influence the central actions of glucocorticoids on appetite and perhaps energy balance. We show that 11 beta-HSD1-/- mice express lower hypothalamic mRNA levels of the anorexigenic cocaine and amphetamine-regulated transcript and melanocortin-4 receptor, but higher levels of the orexigenic melanin-concentrating hormone mRNAs than controls (C57BL/6J) on a low-fat diet (11% fat). HF (58% fat) diet promoted transient ( approximately 8 wk) hyperphagia and decreased food efficiency in 11 beta-HSD1-/- mice and decreased melanocortin-4 receptor mRNA expression in control but not 11 beta-HSD1-/- mice. 11 beta-HSD1-/- mice showed a HF-mediated up-regulation of the orexigenic agouti-related peptide (AGRP) mRNA in the arcuate nucleus which paralleled the transient HF hyperphagia. Conversely, control mice showed a rapid (48 h) HF-mediated increase in arcuate 11 beta-HSD1 associated with subsequent down-regulation of AGRP. This regulatory pattern was unexpected because glucocorticoids increase AGRP, suggesting an alternate hyperphagic mechanism despite partial colocalization of 11 beta-HSD1 and AGRP in arcuate nucleus cells. One major alternate mechanism governing selective fat ingestion and the AGRP system is endogenous opioids. Treatment of HF-fed mice with the mu opioid agonist DAMGO recapitulated the HF-induced dissociation of arcuate AGRP expression between control and 11 beta-HSD1-/- mice, whereas the opioid antagonist naloxone given with HF induced a rise in arcuate AGRP and blocked HF-diet induction of 11 beta-HSD1. These data suggest that 11 beta-HSD1 in brain plays a role in the adaptive restraint of excess fat intake, in part by increasing inhibitory opioid tone on AGRP expression in the arcuate nucleus.

    Topics: 11-beta-Hydroxysteroid Dehydrogenase Type 1; Agouti-Related Protein; Animals; Arcuate Nucleus of Hypothalamus; Corticosterone; Dietary Fats; Enkephalin, Ala(2)-MePhe(4)-Gly(5)-; Enzyme Induction; Female; Gene Expression; Hyperphagia; Insulin; Intercellular Signaling Peptides and Proteins; Leptin; Male; Mice; Mice, Inbred C57BL; Mice, Knockout; Nerve Tissue Proteins; Neurons; Neuropeptide Y; Pro-Opiomelanocortin; Proteins; Receptor, Melanocortin, Type 4; Receptors, Opioid, mu; RNA, Messenger; Thyroid Hormones; Weight Gain

2006
Combined deletion of Y1, Y2, and Y4 receptors prevents hypothalamic neuropeptide Y overexpression-induced hyperinsulinemia despite persistence of hyperphagia and obesity.
    Endocrinology, 2006, Volume: 147, Issue:11

    Neuropeptide Y (NPY) is a key regulator of energy homeostasis and is implicated in the development of obesity and type 2 diabetes. Whereas it is known that hypothalamic administration of exogenous NPY peptides leads to increased body weight gain, hyperphagia, and many hormonal and metabolic changes characteristic of an obesity syndrome, the Y receptor(s) mediating these effects is disputed and unclear. To investigate the role of different Y receptors in the NPY-induced obesity syndrome, we used recombinant adeno-associated viral vector to overexpress NPY in mice deficient of selective single or multiple Y receptors (including Y1, Y2, and Y4). Results from this study demonstrated that long-term hypothalamic overexpression of NPY lead to marked hyperphagia, hypogonadism, body weight gain, enhanced adipose tissue accumulation, hyperinsulinemia, and other hormonal changes characteristic of an obesity syndrome. NPY-induced hyperphagia, hypogonadism, and obesity syndrome persisted in all genotypes studied (Y1(-/-), Y2(-/-), Y2Y4(-/-), and Y1Y2Y4(-/-) mice). However, triple deletion of Y1, Y2, and Y4 receptors prevented NPY-induced hyperinsulinemia. These findings suggest that Y1, Y2, and Y4 receptors under this condition are not crucially involved in NPY's hyperphagic, hypogonadal, and obesogenic effects, but they are responsible for the central regulation of circulating insulin levels by NPY.

    Topics: Adipose Tissue; Animals; Blood Glucose; Body Weight; Female; Humans; Hyperinsulinism; Hyperphagia; Hypothalamus; Mice; Mice, Inbred C57BL; Neuropeptide Y; Obesity; Receptors, Neuropeptide Y

2006
Young adult-specific hyperphagia in diabetic Goto-kakizaki rats is associated with leptin resistance and elevation of neuropeptide Y mRNA in the arcuate nucleus.
    Journal of neuroendocrinology, 2006, Volume: 18, Issue:10

    The present study aimed to examine whether hyperphagia, which is frequently observed in type 1 diabetic patients and model animals, also occurs in type 2 diabetic Goto-Kakizaki (GK) rats and, if so, to explore underlying abnormalities in the hypothalamus. GK rats at postnatal weeks 6-12, compared to control Wistar rats, exhibited hyperphagia, hyperglycaemia, hyperleptinemia and increased visceral fat accumulation, whereas body weight was unaltered. The ability of leptin to suppress feeding was reduced in GK rats compared to Wistar rats of these ages. In GK rats, leptin-induced phosphorylation of signal transducer and activator of transcription 3 was significantly reduced in the cells of the hypothalamic arcuate nucleus (ARC), but not of the ventromedial hypothalamus, whereas the mRNA level of functional leptin receptor was unaltered. By real-time polymerase chain reaction and in situ hybridisation, mRNA levels of neuropeptide Y, but not pro-opiomelanocortin and galanin-like peptide, were significantly increased in the ARC of GK rats at 11 weeks, but not 26 weeks. Following i.c.v. injection of a NPY Y1 antagonist, 1229U91, the amount of food intake in GK rats was indistinguishable from that in Wistar rats, thus eliminating the hyperphagia of GK rats. These results demonstrate that young adult GK rats display hyperphagia in association with leptin resistance and increased NPY mRNA level in the ARC.

    Topics: Animals; Arcuate Nucleus of Hypothalamus; Blood Glucose; Diabetes Mellitus; Eating; Hyperphagia; Hypothalamus; Immunohistochemistry; In Situ Hybridization; Injections, Intraventricular; Leptin; Neuropeptide Y; Peptides, Cyclic; Rats; Rats, Wistar; Reverse Transcriptase Polymerase Chain Reaction; RNA, Messenger; STAT3 Transcription Factor

2006
Involvement of specific orexigenic neuropeptides in sweetener-induced overconsumption in rats.
    Behavioural brain research, 2006, Dec-15, Volume: 175, Issue:2

    Palatability is one of the factors that regulates food and fluid intake and contributes to overconsumption in turn contributing to obesity. To elucidate the brain mechanisms of the palatability-induced ingestion, we explored the roles of six hypothalamic orexigenic neuropeptides, orexin, melanin-concentrating hormone (MCH), neuropeptide Y (NPY), agouti-related protein (AgRP), ghrelin and dynorphin, in the intake of a palatable solution, saccharin. Of the six peptides, intracerebroventricular (i.c.v.) administrations of orexin, MCH and NPY increased the intake of saccharin. Drinking of saccharin in turn elevated the mRNA levels of orexin and NPY, but not MCH. Pre-treatments of naloxone, an opioid antagonist, blocked the orexigenic effects of orexin and NPY. Specific gastric motor responses induced by central orexin-A and NPY are well known, however, MCH did not induce such responses. The i.c.v. administration of orexin-A facilitated gastric emptying. These results suggest that the overconsumption promoted by sweet and palatable tastes is attributed to the activation of orexigenic neuropeptides, such as orexin and NPY, and a downstream opioid system together with enhanced digestive functions.

    Topics: Agouti-Related Protein; Animals; Appetite Regulation; Dynorphins; Feeding Behavior; Gastrointestinal Motility; Ghrelin; Hyperphagia; Hypothalamic Hormones; Injections, Intraventricular; Intercellular Signaling Peptides and Proteins; Intracellular Signaling Peptides and Proteins; Male; Melanins; Neuropeptide Y; Neuropeptides; Orexins; Peptide Hormones; Pituitary Hormones; Rats; Rats, Wistar; RNA, Messenger; Sweetening Agents; Taste

2006
Involvement of neuropeptide Y in hyperphagia in human growth hormone transgenic rats.
    The Journal of veterinary medical science, 2006, Volume: 68, Issue:9

    We have previously produced human growth hormone (hGH) transgenic (TG) rats that show low circulating levels of both hGH and endogenous rat GH. Although body length of the TG rats is normal, they develop hyperphagia and severe obesity. The present study was undertaken to elucidate the causes of hyperphagia in the TG rats by focusing on temporal changes in plasma ghrelin levels and hypothalamic neuropeptide Y (NPY) contents. In both wild-type (WT) and TG rats, the highest value of plasma ghrelin levels was observed just before the dark phase, and thereafter plasma ghrelin levels were maintained higher in the TG than WT rats. Although NPY contents also showed the peak level just before the dark phase in both the arcuate (ARC) and paraventricular nuclei (PVN) of the hypothalamus, the values in the ARC, but not the PVN, of the TG rats was always lower than those of the WT rats, suggesting increased transport of NPY from the ARC to PVN in the TG rats. In addition, treatment with antagonists for Y1 and Y5 receptors for NPY reduced food intake much more effectively in the TG than WT rats. Intermittent treatment with recombinant hGH for a week significantly decreased food consumption, adipose tissue weight and plasma triglyceride concentrations in the TG rats. These results suggest that, in the TG rats, insufficiency in circulating GH stimulates the ghrelin-NPY system with a resultant increase in food intake.

    Topics: Aging; Animals; Animals, Genetically Modified; Female; Human Growth Hormone; Humans; Hyperphagia; Hypothalamus; Neuropeptide Y; Photoperiod; Rats; Time Factors

2006
Running wheel activity prevents hyperphagia and obesity in Otsuka long-evans Tokushima Fatty rats: role of hypothalamic signaling.
    Endocrinology, 2005, Volume: 146, Issue:4

    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.

    Topics: Animals; Blood Glucose; Body Weight; Corticotropin-Releasing Hormone; Eating; Hyperphagia; Hypothalamus; Leptin; Male; Neuropeptide Y; Obesity; Physical Conditioning, Animal; Pro-Opiomelanocortin; Rats; Rats, Inbred OLETF; Rats, Long-Evans; RNA, Messenger; Running; Signal Transduction

2005
Central relaxin-3 administration causes hyperphagia in male Wistar rats.
    Endocrinology, 2005, Volume: 146, Issue:8

    Relaxin-3 (INSL-7) is a recently discovered member of the insulin superfamily. Relaxin-3 mRNA is expressed in the nucleus incertus of the brainstem, which has projections to the hypothalamus. Relaxin-3 binds with high affinity to the LGR7 receptor and to the previously orphan G protein-coupled receptor GPCR135. GPCR135 mRNA is expressed predominantly in the central nervous system, particularly in the paraventricular nucleus (PVN). The presence of relaxin-3 and these receptors in the PVN led us to investigate the effect of central administration of relaxin-3 on food intake in male Wistar rats. The receptor involved in mediating these effects was also investigated. Intracerebroventricular injections of human relaxin-3 (H3) to satiated rats significantly increased food intake 1 h post administration in the early light phase [0.96 +/- 0.16 g (vehicle) vs. 1.81 +/- 0.21 g (180 pmol H3), P < 0.05] and the early dark phase [2.95 +/- 0.45 g (vehicle) vs. 4.39 +/- 0.39 g (180 pmol H3), P < 0.05]. Intra-PVN H3 administration significantly increased 1-h food intake in satiated rats in the early light phase [0.34 +/- 0.16 g (vehicle) vs. 1.23 +/- 0.30 g (18 pmol H3), P < 0.05] and the early dark phase [4.43 +/- 0.32 g (vehicle) vs. 6.57 +/- 0.42 g (18 pmol H3), P < 0.05]. Feeding behavior increased after intra-PVN H3. Equimolar doses of human relaxin-2, which binds the LGR7 receptor but not GPCR135, did not increase feeding. Hypothalamic neuropeptide Y, proopiomelanocortin, or agouti-related peptide mRNA expression did not change after acute intracerebroventricular H3. These results suggest a novel role for relaxin-3 in appetite regulation.

    Topics: Animals; Cerebral Ventricles; Gene Expression Regulation; Hyperphagia; Hypothalamus; Injections, Intraventricular; Male; Midline Thalamic Nuclei; Neuropeptide Y; Pro-Opiomelanocortin; Rats; Rats, Wistar; Relaxin; RNA, Messenger

2005
Distinct forebrain and caudal brainstem contributions to the neuropeptide Y mediation of ghrelin hyperphagia.
    Diabetes, 2005, Volume: 54, Issue:7

    Neuropeptide Y (NPY) has been implicated in the downstream mediation of ghrelin hyperphagia, with the site of action for both peptides considered to be intrinsic to the hypothalamus. Here, however, we observed robust hyperphagia with caudal brainstem (CBS) (fourth intracerebroventricular) ghrelin delivery and, moreover, that this response was reversed with coadministration of either of two NPY receptor antagonists (1229U91 and D-Tyr27,36, D-Thr32 NPY27-36) with contrasting NPY receptor subtype-binding properties. The same results were obtained after forebrain (third intracerebroventricular) administration, but the sites for both ghrelin and antagonist action were open to question, given the caudal flow of cerebrospinal fluid (CSF) through the ventricular system. To control for this, we occluded the cerebral aqueduct to restrict CSF flow between the forebrain and CBS ventricles and tested all combinations (same and cross ventricle) of ghrelin (150 pmol/1 microl) and NPY receptor antagonist delivery. With fourth intracerebroventricular ghrelin delivery after aqueduct occlusion, preadministration of either of the two antagonists through the same cannula reversed the hyperphagic response but neither was effective when delivered to the third ventricle. With third intracerebroventricular ghrelin administration, however, 1229U91 reversed the ingestive response only when delivered to the fourth ventricle, whereas D-Tyr27,36) D-Thr32 NPY27-36 was effective only when delivered to the forebrain. These results demonstrate distinct mediating pathways (due to location and subtypes of relevant NPY receptor) for the hyperphagic response driven separately by forebrain and CBS ghrelin administration.

    Topics: Animals; Brain Stem; Cerebral Ventricles; Disease Models, Animal; Ghrelin; Hyperphagia; Injections, Intraventricular; Male; Neuropeptide Y; Peptide Hormones; Peptides, Cyclic; Prosencephalon; Rats; Rats, Sprague-Dawley; Time Factors

2005
Hyperphagia of hyperthyroidism: is neuropeptide Y involved?
    Regulatory peptides, 2005, Volume: 131, Issue:1-3

    The possible role of neuropeptide Y (NPY) was studied in rats with hypermetabolism and hyperphagia induced by thyroxine (50-100-200 microg/day s.c. for 3-4 weeks). Both metabolic rate and body temperature increased quickly with thyroxine treatment, while hyperphagia started to develop only after 2 weeks of treatment. The weight gain rate progressively decreased or stopped. The NPY-induced hyperphagia was not altered significantly during thyroxine treatment (in severe thyrotoxicosis it was rather suppressed); the fasting-induced hyperphagia was smaller than in controls following 1 week of treatment, and it became enhanced only after 3 weeks, when the deficit in body weight indicated a certain level of starvation already prior to the food deprivation. The NPY-antagonist D-Tyr27,36,D-Thr32-NPY27,36 suppressed this fasting-induced hyperphagia, suggesting that endogenous NPY is involved in this late phase. In conclusion, hyperthyroidism per se does not increase the NPY activity, instead the quickly developing hyperthermia may inhibit the NPY actions; NPY may, however, be activated by a concurrent hypermetabolism-induced starvation.

    Topics: Animals; Body Temperature; Body Weight; Eating; Energy Metabolism; Female; Food Deprivation; Hyperphagia; Hyperthyroidism; Male; Neuropeptide Y; Rats; Rats, Wistar; Thyroxine

2005
Chronic neuropeptide Y infusion into the lateral ventricle induces sustained feeding and obesity in mice lacking either Npy1r or Npy5r expression.
    Endocrinology, 2004, Volume: 145, Issue:1

    Neuropeptide Y (NPY) is a powerful orexigenic neurotransmitter. The NPY Y1 and Y5 receptors have been implicated in mediating the appetite-stimulating activity of NPY. To further investigate the importance of these two receptors in NPY-induced hyperphagia after chronic central administration, we used mice lacking either Npy1r or Npy5r expression. NPY infusion into the lateral ventricle of wild-type mice stimulated food intake and induced obesity over a 7-d period. Fat pad weight as well as plasma insulin, leptin, and corticosterone levels were strongly increased in NPY-treated mice. In addition, NPY infusion resulted in a significant decrease in hypothalamic NPY and proopiomelanocortin expression. Interestingly, the lack of either Npy1r or Npy5r expression in knockout mice did not affect such feeding response to chronic NPY infusion. Moreover, the obesity syndrome that developed in these animals was similar to that in wild-type animals. Taken together, these data strongly suggest biological redundancies between Y1 and Y5 receptor signaling in the NPY-mediated control of food intake.

    Topics: Adipose Tissue; Animals; Body Weight; Corticosterone; Drinking; Eating; Feeding Behavior; Hyperphagia; Injections, Intraventricular; Insulin; Lateral Ventricles; Leptin; Male; Mice; Mice, Inbred C57BL; Mice, Knockout; Neuropeptide Y; Obesity; Pro-Opiomelanocortin; Receptors, Neuropeptide Y; RNA, Messenger

2004
Adaptive responses in hypothalamic neuropeptide Y in the face of prolonged high-fat feeding in the rat.
    Journal of neurochemistry, 2004, Volume: 88, Issue:4

    While a dysregulation in neuropeptide Y (NPY) signaling has been described in rodent models of obesity, few studies have investigated the time-course of changes in NPY content and responsiveness during development of diet-induced obesity. Therefore we investigated the effect of differing lengths (2-17 weeks) of high-fat diet on hypothalamic NPY peptide content, release and NPY-induced hyperphagia. Male Sprague-Dawley rats (211 +/- 3 g) were fed either a high-fat diet (30% fat) or laboratory chow (5% fat). Animals were implanted with intracerebroventricular cannulae to investigate feeding responses to NPY (0.5 nmol, 1 nmol) after 4 or 12 weeks of diet. At the earlier stage of obesity, NPY-induced hyperphagia was not altered; however, animals maintained on the high-fat diet for the longer duration were hyper-responsive to NPY, compared to chow-fed control rats (p < 0.05). Overall, hypothalamic NPY peptide content tended to be decreased from 9 to 17 weeks of diet (p < 0.05). Total hypothalamic NPY content was negatively correlated with plasma leptin concentration (p < 0.05), suggesting the hypothalamic NPY system remains responsive to leptin's inhibitory signal. In addition, hypothalamic NPY overflow was significantly reduced in high-fat fed animals (p < 0.05). Together these results suggest a reduction in hypothalamic NPY activity in high-fat fed animals, perhaps in an attempt to restore energy balance.

    Topics: Adipose Tissue; Animals; Body Weight; Brain; Brain Chemistry; Dietary Fats; Eating; Hyperphagia; Hypothalamus; Injections, Intraventricular; Male; Neuropeptide Y; Random Allocation; Rats; Rats, Sprague-Dawley; Time Factors

2004
Neuropeptide Y is required for hyperphagic feeding in response to neuroglucopenia.
    Endocrinology, 2004, Volume: 145, Issue:7

    To investigate the role played by the orexigenic peptide, neuropeptide Y (NPY), in adaptive responses to insulin-induced hypoglycemia, we measured hypothalamic, feeding, and hormonal responses to this stimulus in both wild-type (Npy+/+) and NPY-deficient (Npy-/-) mice. After administration of insulin at a dose (60 mU ip) sufficient to cause moderate hypoglycemia (plasma glucose levels, 40 +/- 3 and 37 +/- 2 mg/dl for Npy+/+ and Npy-/- mice, respectively; P = not significant), 4-h food intake was increased 2.5-fold in Npy+/+ mice relative to saline-injected controls. By comparison, the increase of intake in Npy-/- mice was far smaller (45%) and did not achieve statistical significance (P = 0.08). Hyperphagic feeding in response to insulin-induced hypoglycemia was therefore markedly attenuated in mice lacking NPY, and a similar feeding deficit was detected in these animals after neuroglucopenia induced by 2-deoxyglucose (500 mg/kg ip). A role for NPY in glucoprivic feeding is further supported by our finding that Npy mRNA content (measured by real-time PCR) increased 2.4-fold in the hypothalamus of Npy+/+ mice by 7 h after insulin injection. Unlike the feeding deficits observed in mice lacking NPY, the effect of hypoglycemia to increase plasma glucagon and corticosterone levels was fully intact in these animals, as were both the nadir glucose value and time to recovery of euglycemia after insulin injection (P = not significant). We conclude that NPY signaling is required for hyperphagic feeding, but not neuroendocrine responses to moderate hypoglycemia.

    Topics: Animals; Antimetabolites; Blood Glucose; Corticosterone; Deoxyglucose; Feeding Behavior; Female; Glucagon; Hyperphagia; Hypoglycemia; Hypothalamus; Male; Mice; Mice, Inbred Strains; Mice, Knockout; Neuropeptide Y; Signal Transduction

2004
Melanocortin 4 receptor-mediated hyperphagia and activation of neuropeptide Y expression in the dorsomedial hypothalamus during lactation.
    The Journal of neuroscience : the official journal of the Society for Neuroscience, 2004, Jun-02, Volume: 24, Issue:22

    In several hyperphagic models, including lactation, in which hypothalamic melanocortin signaling is reduced, a novel expression of NPY mRNA in the dorsomedial hypothalamus (DMH) has been observed, suggesting that melanocortin signaling and the induced NPY in the DMH may constitute unique neurocircuitry in mediating energy balance. Using lactating rats as a model, the present study first showed that in the DMH abundant alpha-MSH and agouti-related protein fibers are in close apposition to NPY-positive cells. However, no NPY and MC4R (a melanocortin receptor) double-labeled neurons were observed. These data suggested that melanocortin input may synapse on presynaptic terminals that then synapse on DMH NPY cells. To study the function of DMH MC4Rs in energy balance, an MC3/4R-selective agonist, melanotan II (MTII), was injected bilaterally into the DMH. MTII injection significantly suppressed feeding induced by 24 hr fasting or suckling-induced hyperphagia. Furthermore, MTII treatment greatly attenuated suckling-induced NPY expression in the DMH. MTII treatment also stimulated uncoupling protein 1 activity in the brown adipose tissue of suckling female rats, indicative of increased sympathetic outflow. In summary, the present study demonstrated that the melanocortin system in the DMH not only plays an important role in inducing NPY expression in the DMH of lactating rats but also in regulating energy homeostasis, at least in part, by modulating appetite and energy expenditure.

    Topics: Adipose Tissue, Brown; Agouti-Related Protein; alpha-MSH; Animals; Appetite Regulation; Carrier Proteins; Eating; Energy Metabolism; Female; Gene Expression; Glutamate Decarboxylase; Homeostasis; Hyperphagia; Hypothalamus; Intercellular Signaling Peptides and Proteins; Ion Channels; Isoenzymes; Lactation; Leptin; Membrane Proteins; Mitochondrial Proteins; Models, Animal; Nerve Fibers; Neurons; Neuropeptide Y; Peptides, Cyclic; Proteins; Rats; Rats, Sprague-Dawley; Receptor, Melanocortin, Type 4; RNA, Messenger; Uncoupling Protein 1

2004
Adipogenic and orexigenic effects of the ghrelin-receptor ligand tabimorelin are diminished in leptin-signalling-deficient ZDF rats.
    European journal of endocrinology, 2004, Volume: 150, Issue:6

    The aim was to investigate the possible interactions of the two peripheral hormones, leptin and ghrelin, that regulate the energy balance in opposite directions.. Leptin-receptor mutated Zucker diabetic fatty (ZDF) and lean control rats were treated with the ghrelin-receptor ligand, tabimorelin (50 mg/kg p.o.) for 18 days, and the effects on body weight, food intake and body composition were investigated. The level of expression of anabolic and catabolic neuropeptides and their receptors in the hypothalamic area were analysed by in situ hybridization.. Tabimorelin treatment induced hyperphagia and adiposity (increased total fat mass and gain in body weight) in lean control rats, while these parameters were not increased in ZDF rats. Treatment with tabimorelin of lean control rats increased hypothalamic mRNA expression of the anabolic neuropeptide Y (NPY) mRNA and decreased hypothalamic expression of the catabolic peptide pro-opiomelanocortin (POMC) mRNA. In ZDF rats, the expression of POMC mRNA was not affected by treatment with tabimorelin, whereas NPY mRNA expression was increased in the hypothalamic arcuate nucleus.. This shows that tabimorelin-induced adiposity and hyperphagia in lean control rats are correlated with increased hypothalamic NPY mRNA and decreased POMC mRNA expression. The elimination of tabimorelin-induced adiposity and hyperphagia in ZDF rats may be due to lack of POMC mRNA downregulation. In conclusion, we suggest that ghrelin-receptor ligands exert their adipogenic and orexigenic effects via hypothalamic mechanisms that are dependent on intact leptin-receptor signalling.

    Topics: Adipose Tissue; Animals; Body Composition; Body Weight; Dipeptides; Eating; Gene Expression; Hyperphagia; Hypothalamus; In Situ Hybridization; Mutation; Neuropeptide Y; Pro-Opiomelanocortin; Rats; Rats, Zucker; Receptors, Cell Surface; Receptors, G-Protein-Coupled; Receptors, Ghrelin; Receptors, Leptin; RNA, Messenger; Signal Transduction

2004
Induction of brain-region-specific forms of obesity by agouti.
    The Journal of neuroscience : the official journal of the Society for Neuroscience, 2004, Nov-10, Volume: 24, Issue:45

    Disruption of melanocortin (MC) signaling, such as by ectopic Agouti overexpression, leads to an obesity syndrome with hyperphagia, obesity, and accelerated body weight gain during high-fat diet. To investigate where in the brain disruption of MC signaling results in obesity, long-term Agouti expression was induced after local injections of recombinant adeno-associated viral particles in selected brain nuclei of adult rats. Agouti expression in the paraventricular nucleus, a hypothalamic region with a high density of MC receptors, induced acute onset hyperphagia and rapid weight gain that persisted for at least 6 weeks. In contrast, obesity and hyperphagia developed with a 3 week delay when Agouti was expressed in the dorsal medial hypothalamus. Agouti expression in the lateral hypothalamus (LH) did not affect food intake and body weight during regular diet, despite the presence of MC receptors in this region. However, during exposure to a high-fat diet, animals with Agouti expression in the LH exhibited a marked increase in body weight. Here we show that the LH is important for the protection against diet-induced obesity by controlling caloric intake during consumption of a high-fat diet. Together, this study provides evidence that different aspects of the Agouti-induced obesity syndrome, such as hyperphagia and diet responsiveness, are mediated by distinct brain regions and opens challenging opportunities for further understanding of pathophysiological processes in the development of the obesity syndrome.

    Topics: Agouti Signaling Protein; Agouti-Related Protein; Animals; Cell Line; Dietary Fats; Energy Intake; Humans; Hyperphagia; Hypothalamus; Hypothalamus, Middle; Intercellular Signaling Peptides and Proteins; Male; Neuropeptide Y; Obesity; Organ Specificity; Paraventricular Hypothalamic Nucleus; Proteins; Rats; Rats, Wistar; Receptors, Melanocortin; Recombinant Fusion Proteins; RNA, Messenger; Weight Gain

2004
Evidence for involvement of neuropeptide Y and melanocortin systems in the hyperphagia of lactation in rats.
    Pharmacology, biochemistry, and behavior, 2003, Volume: 74, Issue:2

    Hypothalamic neuropeptide Y (NPY) systems are upregulated during lactation in rats. Because NPY is central to the hypothalamic control of energy balance, the present studies tested the hypothesis that NPY contributes to the marked hyperphagia during lactation. A 4-day infusion of [D-tyr (27,36), D-thr (32)] NPY (27-36) (D-NPY(27-36)), a peptide analogue of NPY that antagonizes NPY-induced feeding, into the third ventricle at 1 microg/h transiently inhibited nocturnal feeding in nonlactating female rats. However, this antagonist had no effect on nocturnal feeding, but did transiently reduce food intake during the light hours, when infused into the third ventricle at the same dose in lactating females. An essentially similar pattern of results was obtained with chronic infusion into the third ventricle of the anorexigenic peptide alpha-melanocyte-stimulating hormone (alpha-MSH, 1 microg/h), in nonlactating and lactating rats. Both D-NPY(27-36) and alpha-MSH transiently reduced nocturnal food intake in lactating rats by approximately 10% when infused at the higher dose of 5 microg/h, and a marked inhibition of approximately 40% of both nocturnal and diurnal feeding was produced by a combined infusion of both at 5 microg/h. These results provide the first pharmacological evidence implicating specific neuromessengers in mediating the hyperphagia of lactation, and suggest that, while an action of NPY may contribute to the increased food intake seen in lactating animals, other systems are also involved. In particular, a reduction in melanocortin signaling during lactation may allow for an increased orexigenic influence of the agouti-related protein (AgRP), which is co-expressed with NPY.

    Topics: alpha-MSH; Animals; Body Weight; Darkness; Eating; Female; Hyperphagia; Injections, Intraventricular; Lactation; Light; Neuropeptide Y; Peptide Fragments; Rats; Rats, Sprague-Dawley; Stereotaxic Techniques

2003
Hyperphagia, not hypometabolism, causes early onset obesity in melanocortin-4 receptor knockout mice.
    Physiological genomics, 2003, Mar-18, Volume: 13, Issue:1

    Previous studies on mice with melanocortin-4 receptor gene (MC4r) knockout have focused on obese adults. Because humans with functional MC4r mutations show early-onset obesity, we determined the onset of excessive fat deposition in 10- to 56-day-old mice, taking into account sex and litter influences. Total body fat content of MC4r-/- on day 35 and MC4r+/- on day 56 significantly exceeds that of MC4r+/+. Plasma leptin levels increase in proportion to fat mass. According to cumulative food intake and energy expenditure measurements from day 21 to 35, onset of excessive fat deposition in MC4r-/- is fueled by hyperphagia and counteracted partially by hypermetabolism. In 35- to 56-day-old mice, arcuate nucleus neuropeptide Y (NPY) mRNA decreases and pro-opiomelanocortin (POMC) mRNA increases with fat content and plasma leptin levels independently of genotype. Taking into account fat content by ANCOVA reveals, however, increases in both NPY mRNA and POMC mRNA due to melanocortin-4 receptor (MC4R) deficiency. We conclude that hyperphagia, not hypometabolism, is the primary disturbance initiating excessive fat deposition in MC4R-deficient mice at weaning and that the overall changes in NPY and POMC expression tend to antagonize the onset of excessive fat deposition.

    Topics: Adipose Tissue; Animals; Body Composition; Eating; Energy Intake; Energy Metabolism; Female; Genotype; Hyperphagia; Leptin; Male; Mice; Mice, Knockout; Neuropeptide Y; Obesity; Peptide Fragments; Pro-Opiomelanocortin; Receptor, Melanocortin, Type 4; Receptors, Corticotropin

2003
A role for the endogenous opioid beta-endorphin in energy homeostasis.
    Endocrinology, 2003, Volume: 144, Issue:5

    Proopiomelanocortin (POMC) neurons in the hypothalamus are direct targets of the adipostatic hormone leptin and contribute to energy homeostasis by integrating peripheral and central information. The melanocortin and beta-endorphin neuropeptides are processed from POMC and putatively coreleased at axon terminals. Melanocortins have been shown by a combination of pharmacological and genetic methods to have inhibitory effects on appetite and body weight. In contrast, pharmacological studies have generally indicated that opioids stimulate food intake. Here we report that male mice engineered to selectively lack beta-endorphin, but that retained normal melanocortin signaling, were hyperphagic and obese. Furthermore, beta-endorphin mutant and wild-type mice had identical orexigenic responses to exogenous opioids and identical anorectic responses to the nonselective opioid antagonist naloxone, implicating an alternative endogenous opioid tone to beta-endorphin that physiologically stimulates feeding. These genetic data indicate that beta-endorphin is required for normal regulation of feeding, but, in contrast to earlier reports suggesting opposing actions of beta-endorphin and melanocortins on appetite, our results suggest a more complementary interaction between the endogenously released POMC-derived peptides in the regulation of energy homeostasis.

    Topics: Animals; beta-Endorphin; Eating; Energy Metabolism; Glucose; Homeostasis; Hyperinsulinism; Hyperphagia; Leptin; Male; Mice; Mice, Knockout; Naloxone; Narcotic Antagonists; Neuropeptide Y; Obesity; Reference Values

2003
Characterization of neuropeptide Y (NPY) Y5 receptor-mediated obesity in mice: chronic intracerebroventricular infusion of D-Trp(34)NPY.
    Endocrinology, 2003, Volume: 144, Issue:5

    To clarify the role of the neuropeptide Y (NPY) Y5 receptor subtype in energy homeostasis, the effect of the intracerebroventricular infusion of a selective Y5 agonist, D-Trp(34)NPY, was investigated in C57BL/6J mice. Intracerebroventricular infusion of D-Trp(34)NPY (5 and 10 microg/d) produced hyperphagia and body weight gain, accompanied by increased adipose tissue weight, hypercholesterolemia, hyperinsulinemia, and hyperleptinemia. Oral administration of a selective Y5 antagonist at a dose of 100 mg/kg twice a day completely suppressed all of these D-Trp(34)NPY-induced changes, indicating that chronic activation of the Y5 receptor produces hyperphagia and obesity. In addition, D-Trp(34)NPY still resulted in an increase in adipose tissue weight accompanied by hyperleptinemia and hypercholesterolemia, although D-Trp(34)NPY-induced food intake was restricted by pair-feeding. Under the pair-fed condition, D-Trp(34)NPY decreased hormone-sensitive lipase activity in white adipose tissue and uncoupling protein-1 mRNA expression in brown adipose tissue. These findings indicate that Y5-mediated obesity may involve metabolic changes, such as decreased lipolysis and thermogenesis, as well as hyperphagia. Therefore, the Y5 receptor can play a key role in regulating energy homeostasis.

    Topics: Animals; Binding, Competitive; Carrier Proteins; CCAAT-Enhancer-Binding Proteins; DNA-Binding Proteins; Drug Administration Schedule; Energy Metabolism; Glycogen; Homeostasis; Hyperphagia; Injections, Intraventricular; Ligands; Lipoprotein Lipase; Male; Mice; Mice, Inbred C57BL; Neuropeptide Y; Obesity; Receptors, Neuropeptide Y; RNA, Messenger; Sterol Esterase; Sterol Regulatory Element Binding Protein 1; Transcription Factors; Triglycerides

2003
Decreased hypothalamic concentration of neuropeptide Y correlates with onset of hyperphagia in fa/fa rats on postnatal day 12.
    Physiology & behavior, 2003, Volume: 78, Issue:4-5

    An increased action of hypothalamic neuropeptide Y (NPY) has been proposed as a major factor in the pathophysiology of the obesity syndrome in Zucker (fa/fa) rats. Using a developmental strategy to test this hypothesis, we showed previously that significantly more arcuate NPY was expressed in fa/fa pups than in lean littermates on postnatal day (P) 2 and throughout the preweaning period [Physiol. Behav. 67 (1999) 521], and that hyperphagia first appeared on P12 [Am. J. Physiol. 275 (1998) R1106]. To test the hypothesis further, we used a specific radioimmunoassay to measure the concentration of hypothalamic NPY peptide in lean (+/+ and +/fa) and obese fa/fa Zucker rat pups on P9, P10, and P12. The concentration of NPY in fa/fa pups was not significantly different from that of the other genotypes. There was, however, a significant decrease in NPY concentration from P9 to P12 in fa/fa pups, but not in lean pups. The combination of increased NPY message and decreasing concentration of NPY peptide in fa/fa pups with age is consistent with, but does not prove, increased release of hypothalamic NPY in fa/fa pups just before and on P12 when hyperphagia emerges. These results provide further support for the importance of hypothalamic NPY in the phenotypic expression of hyperphagia in the fa/fa pups during the second postnatal week.

    Topics: Aging; Animals; Female; Genotype; Hyperphagia; Hypothalamus; Male; Neuropeptide Y; Obesity; Phenotype; Rats; Rats, Zucker

2003
Hypothalamic levels of NPY, MCH, and prepro-orexin mRNA during pregnancy and lactation in the rat: role of prolactin.
    FASEB journal : official publication of the Federation of American Societies for Experimental Biology, 2003, Volume: 17, Issue:11

    Pregnancy and lactation provide excellent models of physiological hyperphagia and hyperprolactinemia. To identify possible factors associated with the increased feeding in these situations, we measured hypothalamic mRNA levels of three orexigenic neuropeptides--NPY, MCH, and orexins--in nonpregnant, pregnant, and lactating rats by in situ hybridization. NPY mRNA content in the arcuate nucleus was significantly increased during pregnancy and lactation. However, MCH and prepro-orexin expression was decreased in both states. 48 or 72 h of fasting in pregnant and lactating rats further elevated NPY mRNA levels and increased the low MCH mRNA content. Surprisingly, no effect was observed in prepro-orexin mRNA levels. Finally, we investigated the possible effect of high PRL levels on these orexigenic signals using a model of hyperprolactinemia induced by pituitary graft. NPY mRNA content was unchanged, but MCH and prepro-orexin mRNA levels were significantly decreased. Our results suggest that the increased NPY expression might be partly responsible for the hyperphagia observed during pregnancy and lactation. MCH and prepro-orexin may be involved in the adaptation of other homeostatic mechanisms and their decreased levels in these physiological settings could be mediated by the elevated circulating PRL levels.

    Topics: Animals; Female; Gene Expression Regulation; Hyperphagia; Hyperprolactinemia; Hypothalamic Hormones; Hypothalamus; In Situ Hybridization; Intracellular Signaling Peptides and Proteins; Lactation; Melanins; Neuropeptide Y; Neuropeptides; Orexins; Pituitary Hormones; Pregnancy; Prolactin; Protein Precursors; Rats; Rats, Sprague-Dawley; RNA, Messenger

2003
Hypothalamic neuropeptide Y/Y1 receptor pathway activated by a reduction in circulating leptin, but not by an increase in circulating ghrelin, contributes to hyperphagia associated with triiodothyronine-induced thyrotoxicosis.
    Neuroendocrinology, 2003, Volume: 78, Issue:6

    Food intake is regulated by hypothalamic neuropeptides which respond to peripheral signals. Plasma ghrelin and leptin levels reflect peripheral energy balance and regulate hypothalamic neuropeptides such as neuropeptide Y (NPY), pro-opiomelanocortin (POMC), cocaine- and amphetamine-regulated transcript (CART), melanin-concentrating hormone (MCH), and orexins. Thyroid hormone stimulates food intake in humans and rodents. However, the mechanisms responsible for this stimulation have not been fully elucidated. To investigate the hyperphagic response to triiodothyronine (T(3))-induced thyrotoxicosis, adult male rats were studied 7 days after daily intraperitoneal injections of T(3) or vehicle. T(3)-treated rats were markedly hyperphagic. During this hyperphagia, plasma leptin levels were markedly decreased. However, the expression of the ghrelin gene in the stomach and the plasma ghrelin concentrations did not differ between the 2 groups. Hypothalamic NPY mRNA levels were significantly increased and associated with a marked decreased in both hypothalamic POMC and CART mRNA levels in the T(3)-treated rats. Hypothalamic MCH and orexin mRNA levels did not differ between the 2 groups. In addition, hyperphagia was partially reversed by intracerebroventricular administration of the NPY Y1 receptor antagonist BIBO3304. Therefore, the decreased plasma leptin levels could contribute to hyperphagia in T(3)-induced thyrotoxicosis. However, plasma ghrelin levels did not contribute to this hyperphagia.

    Topics: Animals; Arginine; Body Weight; Eating; Energy Metabolism; Gastric Mucosa; Gene Expression; Ghrelin; Hyperphagia; Hypothalamus; Leptin; Male; Nerve Tissue Proteins; Neuropeptide Y; Peptide Hormones; Pro-Opiomelanocortin; Rats; Rats, Sprague-Dawley; Receptors, Neuropeptide Y; Thyrotoxicosis; Triiodothyronine

2003
Differential response to NPY of PVH and dopamine-responsive VMH neurons in overweight rats.
    Neuroreport, 2002, Aug-27, Volume: 13, Issue:12

    Neuronal responses to neuropeptide Y and dopamine were recorded in brain slices of hypothalamic paraventricular (PVH) and ventromedial (VMH) nuclei in normal and hyperphagic overweight rats reared in small litters of three pups. NPY significantly activated PVH neurons of normal rats, but inhibited neurons of overweight rats. In the VMH, a significantly higher coincidence of inhibition induced by NPY and dopamine was found in overweight rats. Similar neuronal responses were evoked by a NPY Y5 receptor agonist. Effects of NPY could be blocked by a Y1 receptor antagonist. The altered response of PVH neurons to the feeding-inducing NPY and the increased inhibition by NPY and dopamine in the VMH might contribute to the persisting hyperphagia and overweight of postnatally overnourished rats.

    Topics: Action Potentials; Animals; Animals, Outbred Strains; Dopamine; Hyperphagia; Male; Neuropeptide Y; Obesity; Organ Culture Techniques; Paraventricular Hypothalamic Nucleus; Rats; Rats, Wistar; Ventromedial Hypothalamic Nucleus

2002
Role of ghrelin in streptozotocin-induced diabetic hyperphagia.
    Endocrinology, 2002, Volume: 143, Issue:12

    Ghrelin, an endogenous ligand for the growth hormone (GH) secretagogue receptor, was originally purified from the rat stomach. We have previously reported that central administration of ghrelin increases food intake and body weight. To investigate the role of ghrelin in the hyperphagic response to uncontrolled diabetes, adult male rats were studied 14 days after administration of streptozotocin (STZ) or vehicle. STZ-treated diabetic rats were markedly hyperphagic. This hyperphagia was accompanied by hyperglycemia, hypoinsulinemia, and reduced plasma GH levels. Treatment of diabetic rats with insulin reversed these changes. Plasma ghrelin concentrations in untreated diabetic rats were significantly higher than in control rats and were normalized by insulin treatment. The ghrelin gene expression in the stomach was also higher in STZ diabetic rats than in control rats, but this difference was not significant. In contrast, plasma leptin was markedly reduced in STZ diabetic rats. This reduction in plasma leptin levels was reversed by insulin treatment. In addition, hypothalamic NPY mRNA levels were increased in STZ-treated diabetic rats and were reversed by insulin treatment. Furthermore, the hyperphagia was partially reversed by the administration of a ghrelin-receptor antagonist. Therefore, we conclude that the elevated plasma ghrelin levels, along with decreased plasma leptin levels, could contribute to the diabetic hyperphagia in part by increasing hypothalamic NPY. This is the first report to show the pathophysiological significance of ghrelin in diabetes.

    Topics: Animals; Blood Glucose; Diabetes Mellitus, Experimental; Gastric Mucosa; Gene Expression; Ghrelin; Growth Hormone; Hyperphagia; Hypothalamus; Insulin; Leptin; Male; Neuropeptide Y; Peptide Hormones; Rats; Rats, Sprague-Dawley; Receptors, Cell Surface; Receptors, G-Protein-Coupled; Receptors, Ghrelin; RNA, Messenger

2002
Melanin-concentrating hormone 1 receptor-deficient mice are lean, hyperactive, and hyperphagic and have altered metabolism.
    Proceedings of the National Academy of Sciences of the United States of America, 2002, Mar-05, Volume: 99, Issue:5

    Melanin-concentrating hormone (MCH) is a cyclic 19-aa hypothalamic neuropeptide derived from a larger prohormone precursor of MCH (Pmch), which also encodes neuropeptide EI (NEI) and neuropeptide GE (NGE). Pmch-deficient (Pmch-/-) mice are lean, hypophagic, and have an increased metabolic rate. Transgenic mice overexpressing Pmch are hyperphagic and develop mild obesity. Consequently, MCH has been implicated in the regulation of energy homeostasis. The MCH 1 receptor (MCH1R) is one of two recently identified G protein-coupled receptors believed to be responsible for the actions of MCH. We evaluated the physiological role of MCH1R by generating MCH1R-deficient (Mch1r-/-) mice. Mch1r-/- mice have normal body weights, yet are lean and have reduced fat mass. Surprisingly, Mch1r-/- mice are hyperphagic when maintained on regular chow, and their leanness is a consequence of hyperactivity and altered metabolism. Consistent with the hyperactivity, Mch1r-/- mice are less susceptible to diet-induced obesity. Importantly, chronic central infusions of MCH induce hyperphagia and mild obesity in wild-type mice, but not in Mch1r-/- mice. We conclude that MCH1R is a physiologically relevant MCH receptor in mice that plays a role in energy homeostasis through multiple actions on locomotor activity, metabolism, appetite, and neuroendocrine function.

    Topics: Agouti-Related Protein; Animals; Appetite Stimulants; Body Composition; Corticotropin-Releasing Hormone; Dietary Fats; Eating; Energy Metabolism; Female; Gene Expression; Growth; Hyperkinesis; Hyperphagia; Male; Mice; Mice, Inbred C57BL; Mice, Knockout; Motor Activity; Neuropeptide Y; Neurosecretory Systems; Obesity; Peptide Fragments; Receptors, Pituitary Hormone

2002
Attenuation of diabetic hyperphagia in neuropeptide Y--deficient mice.
    Diabetes, 2002, Volume: 51, Issue:3

    The combined effects of increased hypothalamic signaling by neuropeptide Y (NPY) and decreased signaling by melanocortins are hypothesized to stimulate food intake when body fat stores are depleted. To investigate NPY's role in the hyperphagic response to uncontrolled diabetes, streptozotocin (STZ) (200 mg/kg intraperitoneally) or saline vehicle was given to NPY-deficient (Npy(--/--)) and wild-type (Npy(+/+)) mice. In Npy(+/+) mice, STZ-induced diabetes increased mean daily food intake to plateau values 50% above baseline intake (+2.0 +/- 0.6 g/day; P < or = 0.05), an effect that was not seen in STZ-treated Npy(--/--) mice (+0.8 +/- 0.1 g/day; NS), despite comparably elevated levels of plasma glucose and comparably decreased levels of body weight, fat content, and plasma leptin. Unlike the impaired feeding response to uncontrolled diabetes, Npy(--/--) mice exhibit intact hyperphagic responses to fasting (Erickson et al. [1], Nature 381:415-418, 1996). To investigate whether differences in hypothalamic melanocortin signaling can explain this discrepancy, we used in situ hybridization to compare the effects of STZ-diabetes and fasting on pro-opiomelanocortin (POMC) and agouti-related peptide (AgRP) mRNA levels in the hypothalamic arcuate nucleus (ARC) of Npy(--/--) and Npy(+/+) mice. AgRP mRNA levels were increased by both fasting and STZ-diabetes, but the increase in STZ-diabetes was small (50-80%) compared with the effect of fasting (approximately 20-fold increase of AgRP mRNA). STZ-diabetes also lowered POMC mRNA levels by 65% in the ARC of Npy(+/+) mice (P less-than-or-equal 0.05) but by only 11% in Npy(--/--) mice (NS); fasting significantly lowered POMC mRNA levels in both genotypes. We conclude that NPY is required for both the increase of food intake and the decrease of hypothalamic POMC gene expression induced by uncontrolled diabetes. In contrast, NPY is not required for either of these responses when the stimulus is food deprivation. Moreover, fasting is a more potent stimulus to hypothalamic AgRP gene expression than is STZ-diabetes. Therefore, central nervous system melanocortin signaling appears to be suppressed more effectively by fasting than by uncontrolled diabetes, which provides a plausible explanation for differences in the feeding response to these two stimuli in mice lacking NPY.

    Topics: Adipose Tissue; Agouti-Related Protein; alpha-MSH; Animals; Arcuate Nucleus of Hypothalamus; Blood Glucose; Body Composition; Body Weight; Diabetes Mellitus, Experimental; Fasting; Hyperphagia; Hypothalamus; In Situ Hybridization; Intercellular Signaling Peptides and Proteins; Leptin; Mice; Mice, Inbred C57BL; Neuropeptide Y; Pro-Opiomelanocortin; Proteins; RNA, Messenger; Signal Transduction

2002
Decreasing hypothalamic insulin receptors causes hyperphagia and insulin resistance in rats.
    Nature neuroscience, 2002, Volume: 5, Issue:6

    We investigated the role of hypothalamic insulin signaling in the regulation of energy balance and insulin action in rats through selective decreases in insulin receptor expression in discrete hypothalamic nuclei. We generated an antisense oligodeoxynucleotide directed against the insulin receptor precursor protein and administered this directly into the third cerebral ventricle. Immunostaining of rat brains after 7-day administration of the oligodeoxynucleotide showed a selective decrease of insulin receptor protein within cells in the medial portion of the arcuate nucleus (decreased by approximately 80% as compared to rats treated with a control oligodeoxynucleotide). Insulin receptors in other hypothalamic and extra-hypothalamic areas were not affected. This selective decrease in hypothalamic insulin receptor protein was accompanied by rapid onset of hyperphagia and increased fat mass. During insulin-clamp studies, physiological hyperinsulinemia decreased glucose production by 55% in rats treated with control oligodeoxynucleotides but by only 25% in rats treated with insulin receptor antisense oligodeoxynucleotides. Thus, insulin receptors in discrete areas of the hypothalamus have a physiological role in the control of food intake, fat mass and hepatic action of insulin.

    Topics: Adipose Tissue; Agouti-Related Protein; Animals; Arcuate Nucleus of Hypothalamus; Eating; Glucose; Hyperphagia; Hypothalamus; Injections, Intraventricular; Insulin; Insulin Resistance; Intercellular Signaling Peptides and Proteins; Liver; Male; Neuropeptide Y; Oligonucleotides, Antisense; Proteins; Rats; Rats, Sprague-Dawley; Receptor, Insulin

2002
Agouti-related protein is a mediator of diabetic hyperphagia.
    Regulatory peptides, 2001, Apr-02, Volume: 98, Issue:1-2

    To explore the role of agouti-related protein (AGRP) in diabetic hyperphagia changes in hypothalamic AGRP mRNA levels were examined in diabetic rats. Rats rendered diabetic by streptozotocin displayed marked hyperglycemia (blood glucose 456.0+/-8.4 mg/dl versus 71.8+/-1.9 mg/dl) and hyperphagia (36.9+/-1.0 g/day versus 22.0+/-0.4 g/day), that was associated with a 286.6+/-4.4% increase in hypothalamic AGRP mRNA and a 178.9+/-13.5% increase in hypothalamic NPY mRNA. Insulin treatment of diabetic rats partially corrected blood glucose (147.4+/-13.1 mg/dl) and ameliorated hyperphagia (26.6+/-2.0 g/day). Insulin replacement was also associated with a return of hypothalamic AGRP mRNA (111.7+/-8.3% of controls) and NPY mRNA (125.0+/-8.9% of controls) from the elevated levels that were observed in untreated diabetic rats. In contrast to insulin treated rats, sodium orthovanadate treated diabetic rats remained significantly hyperglycemic (361.5+/-12.5 mg/dl). However, despite their persistent hyperglycemia, orthovanadate treated diabetic rats were still observed to have a significant reduction of hypothalamic AGRP mRNA (138.7+/-11.4%) and NPY mRNA (129.9+/-9.8%). Simultaneous measurement of serum leptin revealed suppressed levels in both untreated diabetic (0.5+/-0.1 ng/ml) and sodium orthovanadate treated rats (0.5+/-0.1 ng/ml) compared to non-diabetic controls (2.1+/-0.1 ng/ml). These data indicate that AGRP is a mediator of diabetic hyperhpagia and suggest that insulin can directly influence hypothalamic AGRP and NPY mRNA expression.

    Topics: Agouti-Related Protein; Animals; Blood Glucose; Body Weight; Diabetes Mellitus, Experimental; Drinking; Eating; Enzyme Inhibitors; Hyperphagia; Hypothalamus; Insulin; Intercellular Signaling Peptides and Proteins; Leptin; Male; Neuropeptide Y; Protein Tyrosine Phosphatases; Proteins; Rats; Rats, Sprague-Dawley; RNA, Messenger; Vanadates

2001
Hypoleptinemia, but not hypoinsulinemia, induces hyperphagia in streptozotocin-induced diabetic rats.
    Journal of neurochemistry, 2001, Volume: 77, Issue:4

    To assess the dominance between hypoinsulinemia and hypoleptinemia as factors in the development of hyperphagia in streptozotocin (STZ)-induced diabetes mellitus (STZ-DM) rodents with respect to hormone-neuropeptide interactions, changes in gene expression of agouti gene-related protein (AGRP) in the arcuate nucleus of the hypothalamus were investigated using STZ-DM rats, fasting Zucker fa/fa rats and STZ-DM agouti (STZ-DM A(y)/a) mice. AGRP mRNA and neuropeptide Y mRNA were both significantly up-regulated in STZ-DM rats, which are associated with body weight loss, hyperglycemia, hypoinsulinemia and hypoleptinemia. We proceeded to analyze whether insulin or leptin played the greater role in the regulation of AGRP using Zucker fa/fa rats. The AGRP mRNA did not differ significantly between fasted fa/fa rats, which have both leptin-insensitivity and hypoinsulinemia, and fed Zuckers, which have leptin-insensitivity and hyperinsulinemia. We further found that up-regulation of AGRP expression was normalized by infusion of leptin into the third cerebroventricle (i3vt), but not by i3vt infusion of insulin, although up-regulation of AGRP was partially corrected by systemic insulin infusion. The latter finding supports hypoleptinemia as a key-modulator of STZ-DM-induced hyperphagia because systemic insulin infusion, at least partially, restored hypoleptinemia through its acceleration of fat deposition, as demonstrated by the partial recovery of lost body weight. After STZ-DM induction, A(y)/a mice whose melanocortin-4 receptor (MC4-R) was blocked by ectopic expression of agouti protein additionally accelerated hyperphagia and up-regulated AGRP mRNA, implying that the mechanism is triggered by a leptin deficit rather than by the main action of the message through MC4-R. Hypoleptinemia, but not hypoinsulinemia per se, thus develops hyperphagia in STZ-DM rodents. These results are very much in line with evidence that hypothalamic neuropeptides are potently regulated by leptin as downstream targets of its actions.

    Topics: Agouti Signaling Protein; Animals; Blood Glucose; Cerebral Ventricles; Diabetes Mellitus, Experimental; Feeding Behavior; Food Deprivation; Gene Expression Regulation; Hyperphagia; Hypothalamus; Infusions, Parenteral; Insulin; Intercellular Signaling Peptides and Proteins; Leptin; Male; Neuropeptide Y; Protein Precursors; Proteins; Rats; Rats, Wistar; Rats, Zucker; RNA, Messenger; Transcription, Genetic; Weight Loss

2001
A role for NPY overexpression in the dorsomedial hypothalamus in hyperphagia and obesity of OLETF rats.
    American journal of physiology. Regulatory, integrative and comparative physiology, 2001, Volume: 281, Issue:1

    Otsuka Long-Evans Tokushima Fatty (OLETF) rats lacking CCK-A receptors are hyperphagic, obese, and diabetic. We have previously demonstrated that these rats have a peripheral satiety deficit resulting in increased meal size. To examine the potential role of hypothalamic pathways in the hyperphagia and obesity of OLETF rats, we compared patterns of hypothalamic neuropeptide Y (NPY), proopiomelanocortin (POMC), and leptin receptor mRNA expression in ad libitum-fed Long-Evans Tokushima (LETO) and OLETF rats and food-restricted OLETF rats that were pair-fed to the intake of LETO controls. Pair feeding OLETF rats prevented their increased body weight and elevated levels of plasma insulin and leptin and normalized their elevated POMC and decreased NPY mRNA expression in the arcuate nucleus. In contrast, NPY expression was upregulated in the dorsomedial hypothalamus (DMH) in pair-fed OLETF rats. A similar DMH NPY overexpression was evident in 5-wk-old preobese OLETF rats. These findings suggest a role for DMH NPY upregulation in the etiology of OLETF hyperphagia and obesity.

    Topics: Age Factors; Animals; Body Composition; Carrier Proteins; Dorsomedial Hypothalamic Nucleus; Eating; Energy Metabolism; Gene Expression; Hyperphagia; In Situ Hybridization; Insulin; Leptin; Male; Neuropeptide Y; Obesity; Pro-Opiomelanocortin; Rats; Rats, Inbred OLETF; Receptor, Cholecystokinin A; Receptors, Cell Surface; Receptors, Cholecystokinin; Receptors, Leptin; RNA, Messenger; Satiation

2001
Insulin-sensitizing action of rosiglitazone is enhanced by preventing hyperphagia.
    Diabetes, obesity & metabolism, 2001, Volume: 3, Issue:3

    We investigated whether pair-feeding to prevent hyperphagia would potentiate the insulin-sensitizing effect of rosiglitazone in chow-fed and insulin-resistant dietary obese rats, and studied the role of leptin and hypothalamic neuropeptide Y as mediators of weight gain during treatment.. Dietary obese and chow-fed rats (575 +/- 10 vs. 536 +/- 7 g; p < 0.01) were given rosiglitazone (30 mg/kg p.o.) or vehicle daily for 14 days.. Energy intake and weight gain were greater in rosiglitazone-treated ad-lib-fed rats (body weight: chow + 24 +/- 2 g, rosiglitazone-treated + 55 +/- 2 g, p < 0.001; dietary obese + 34 +/- 2 g, rosiglitazone-treated + 74 +/- 7 g, p < 0.001). Half of each rosiglitazone-treated group were pair-fed to vehicle-treated controls. Rosiglitazone normalized circulating free fatty acids (FFAs) and insulin sensitivity in dietary obese rats (homeostasis model assessment (HOMA): chow-fed controls, 3.9 +/- 0.3; dietary obese controls, 6.7 +/- 0.7; rosiglitazone-treated, ad lib-fed dietary obese, 4.2 +/- 0.5; both p < 0.01). Insulin sensitivity improved further with pair-feeding (HOMA: 2.9 +/- 0.4; p < 0.05 vs. rosiglitazone-treated, ad lib-fed dietary obese), despite unchanged FFAs. Qualitatively similar findings were made in chow-fed rats. Pair-feeding prevented rosiglitazone-related weight gain in chow-fed, but not dietary obese rats (body weight: + 49 +/- 5 g, p < 0.001 vs. untreated dietary obese controls). Adipose tissue OB mRNA was elevated in dietary obese rats, reduced 49% (p < 0.01) by rosiglitazone treatment, and further (by 16%) with pair-feeding (p < 0.0001). Plasma leptin, however, only fell in the pair-fed group. Hypothalamic neuropeptide Y mRNA was unchanged throughout, suggesting that weight gain associated with high-dose rosiglitazone treatment is independent of hypothalamic neuropeptide Y.. Food restriction potentiates the insulin-sensitizing effect of rosiglitazone in rats, and this effect is independent of a fall in FFAs.

    Topics: Animals; Body Weight; Carrier Proteins; Eating; Food Deprivation; Hyperphagia; Hypoglycemic Agents; Hypothalamus; Insulin Resistance; Ion Channels; Leptin; Male; Mitochondrial Proteins; Neuropeptide Y; Obesity; Rats; Rats, Wistar; RNA, Messenger; Rosiglitazone; Thiazoles; Thiazolidinediones; Uncoupling Protein 3

2001
Hypothalamic neuropeptide Y mRNA in pregnant, lactating and suckling rats.
    The Nigerian postgraduate medical journal, 2001, Volume: 8, Issue:1

    Blood glucose, plasma insulin and luteinizing hormone levels were studied in pregnant wistra rats and those in early and late stages of lactation. NPY mRNA was also measured in whole hypothalamic tissue of these rats which were either fed ad libitum or food deprived to 80% of the relative controls. When fed ad libitum, hypothalamic NPY mRNA was not significantly elevated in the pregnant rats (111 +/- 2.1%). By the 5th and 4th days of lactation the mRNA had increased progressively (141 +/- 4.7% of control, p<0.01; 186 +/- 9%, p<0.001) respectively. Blood glucose levels were unchanged in pregnancy and lactation, however, insulin levels dropped significantly by the ]4th day of lactation (control 322.3 +/- 3.2; lactating 298.6 +/- 4.8 pmol/l; p<0.05). Luteinizing hormone was significantly reduced in the lactating rats (control 2.2 +/- 0.21, lactating 0.81 +/- 0.2 ng/ml;p<0.05). In food restriction, NPY mRNA was increased moderately in the non-pregnant state and enormously in late lactation (non-pregnant 157 +/- 21%, lactating 333 +/- 35%, p<0.001). In a lactation, blood glucose was unchanged while plasma insulin and LH were reduced to 20% and 50% of controls respectively (insulin: control 110.3 +/- 2.0; lactating 18.3 pmol/l; LH. control 1.3 +/- 0.1; lactating 0.59 +/- 0.4 ng/ml p<0.01). Orexigenic effect of hypothalamic NPY is possibly responsible for the hyperphagia in lactating. Food restriction and lactation had additive lowering effect on plasma insulin but an additive increase on hypothalamic NPYmRNA. NPY message may be partially responsible for the anovulatory effect of lactation.

    Topics: Analysis of Variance; Animals; Animals, Suckling; Appetite; Female; Hyperphagia; Hypothalamus; Insulin; Lactation; Luteinizing Hormone; Neuropeptide Y; Pregnancy; Rats; Rats, Wistar; RNA, Messenger

2001
Chronic administration of neuropeptide Y into the lateral ventricle of C57BL/6J male mice produces an obesity syndrome including hyperphagia, hyperleptinemia, insulin resistance, and hypogonadism.
    Molecular and cellular endocrinology, 2001, Dec-20, Volume: 185, Issue:1-2

    Neuropeptide Y (NPY) is involved in the central regulation of appetite, sexual behavior, and reproductive function. We have previously shown that chronic infusion of NPY into the lateral ventricle of normal rats produced an obesity syndrome characterized by hyperphagia, hyperinsulinism and collapse of reproductive function. We further demonstrated that acute inhibition of LH secretion in castrated rats was preferentially mediated by the NPY receptor subtype 5 (Y(5)). In the present study, the effects of chronic, central infusion of NPY, or the mixed Y2-Y5 agonist PYY(3-36), were evaluated both in normal male C57BL/6J mice and Sprague-Dawley rats. After a 7-day infusion to male mice, both NPY and PYY(3-36) at 5 nmol per day, induced marked hyperphagia leading to significant increases in body and fat pad weights. Furthermore, both compounds markedly reduced several markers of the reproductive axis. In the rat study, PYY(3-36) was more active than NPY to inhibit the pituitary-testicular axis, confirming the importance of the Y5 subtype for such effects. In the mouse, chronic NPY infusion induced a sustained increase in corticosterone and insulin secretion. Plasma leptin levels were also markedly increased possibly explaining the observed reduction in gene expression for hypothalamic NPY. Gene expression for hypothalamic POMC was reduced in the NPY- or PYY(3-36)-infused mice, suggesting that NPY exacerbated food intake by both acting through its own receptor(s), and reducing the satiety signal driven by the POMC-derived alpha-MSH. The present study in the mouse suggests in analogy with available rat data, that constant exposure to elevated NPY in the hypothalamic area unabatedly enhances food intake leading to an obesity syndrome including increased adiposity, insulin resistance, hypercorticism, and hypogonadism, reminiscent of the phenotype of the ob/ob mouse, that displays elevated hypothalamic NPY secondary to lack of leptin negative feedback action.

    Topics: Animals; Hyperphagia; Hypogonadism; Insulin Resistance; Lateral Ventricles; Leptin; Male; Mice; Mice, Inbred C57BL; Neuropeptide Y; Obesity; Peptide Fragments; Peptide YY; Rats; Rats, Sprague-Dawley; Syndrome

2001
Hypothalamic galanin is up-regulated during hyperphagia and increased body weight gain induced by disruption of signaling in the ventromedial nucleus.
    Peptides, 2000, Volume: 21, Issue:4

    Disruption of signaling in the ventromedial nucleus (VMN) by colchicine (COL) produces transient (4 days) hyperphagia and weight gain. Microinjection of galanin into various hypothalamic sites stimulates feeding, so we tested the hypothesis that galanin is up-regulated in COL-treated rats by analyzing galanin concentrations in micropunched hypothalamic sites. Galanin was increased in the paraventricular nucleus on Days 1 through 4 after COL-injection. Galanin was also elevated in three other hypothalamic sites, the dorsomedial nucleus, lateral hypothalamic area, and perifornical hypothalamus, on Days 2-4 and in the lateral preoptic area, on Day 1 only. In the median eminence-arcuate nucleus and amygdala an initial decrease on Day 1 was followed by a then progressive increase through Day 4. These increases occurred despite marked elevations in blood insulin and leptin, hormones known to suppress hypothalamic galanin. When COL- or saline-treated rats were injected intracerebroventricularly with galanin, it stimulated feeding further in the hyperphagic COL-treated rats, but the relative response over basal consumption was similar in both COL-treated and control rats. These results in VMN disrupted rats suggest that neurochemical rearrangements, including increased availability of galanin, may contribute to the hyperphagia and increased weight gain; additionally, it seems that neurons in the VMN normally exert a restraint on galanin signaling.

    Topics: Animals; Colchicine; Eating; Galanin; Hyperphagia; Hypothalamus; Insulin; Leptin; Male; Microinjections; Neuropeptide Y; Rats; Rats, Sprague-Dawley; Time Factors; Up-Regulation; Ventromedial Hypothalamic Nucleus; Weight Gain

2000
Pharmacological characterization of the nociceptin receptor mediating hyperphagia: identification of a selective antagonist.
    Psychopharmacology, 2000, Volume: 148, Issue:4

    Central injections of nociceptin (NC) stimulate feeding in rats.. The present study evaluated the effect of N-terminal partial sequences or analogues of NC on food intake in male Wistar rats, to characterize pharmacologically the NC receptor mediating the hyperphagic effect.. NC and related peptides were injected into the lateral (LV) or third (3V) cerebroventricle in freely feeding rats.. In the LV, NC stimulated feeding. The N-terminal fragment NC(1-13)NH2 proved to be the least active sequence with hyperphagic activity; NC(1-12)NH2 and NC(1-9)NH2 were inactive. [Phe(1)psi(CH2-NH)Gly(2)]NC(1-13)NH2 ([F/G)]NC(1-13)NH2), an analogue of NC(1-13)NH2, markedly stimulated feeding and, coadministered in the LV with NC, never reduced the hyperphagic effect of the natural sequence. These findings suggest that [F/G)]NC(1-13)NH2, which has been reported to act as a NC receptor antagonist in peripheral tissues, behaves as a full agonist at the central NC receptors controlling feeding. The hyperphagic potencies of NC and [F/G)]NC(1-13)NH2 were much higher following injection into the 3V than in the LV. Another analogue of NC(1-13)NH2, namely [Nphe(1)]NC(1-13)NH2, injected into the 3V did not stimulate feeding, but reduced the effect of NC. [Nphe(1)]NC(1-13)NH2 at a dose of 16.8 nmol/rat significantly reduced, and at 168 nmol/rat almost completely abolished the effect of NC (1.68 nmol/rat). The latter dose of [Nphe(1)]NC(1-13)NH2 significantly reduced also feeding induced by food deprivation, but did not modify the hyperphagic effect of neuropeptide Y (0.3 nmol/rat).. The present results confirm the orexigenic effect of NC in freely feeding rats and indicate that [Nphe(1)]NC(1-13)NH2 may represent a selective NC receptor antagonist to study the physiological and pathophysiological role of NC in feeding behaviour.

    Topics: Animals; Appetite Stimulants; Eating; Food Deprivation; Hyperphagia; Injections, Intraventricular; Male; Narcotic Antagonists; Neuropeptide Y; Nociceptin; Nociceptin Receptor; Opioid Peptides; Peptide Fragments; Rats; Rats, Wistar; Receptors, Opioid

2000
Ontogeny of hyperphagia in the Zucker (fa/fa) rat: role of neuropeptide Y.
    Appetite, 1999, Volume: 32, Issue:2

    Topics: Animals; Animals, Newborn; Feeding Behavior; Gene Expression; Hyperphagia; In Situ Hybridization; Neuropeptide Y; Obesity; Rats; Rats, Zucker

1999
Hyperphagia in cold-adapted rats: a possible role for neuropeptide Y.
    Acta physiologica Hungarica, 1999, Volume: 86, Issue:3-4

    The feeding response to intracerebroventricular injection of neuropeptide Y or to starvation is greater in cold-adapted than in non-adapted rats, suggesting that with cold-adaptation the central sensitivity to this peptide is increased. Hypometabolism and hypothermia (which usually follow the administration of neuropeptide Y) cannot, however, be demonstrated in the course of cold-adaptation per se.

    Topics: Adaptation, Physiological; Animals; Body Weight; Cold Temperature; Eating; Fasting; Female; Hyperphagia; Injections, Intraventricular; Neuropeptide Y; Rats; Rats, Wistar

1999
Hypothalamic neuropeptide Y and galanin in overweight rats fed a cafeteria diet.
    Peptides, 1998, Volume: 19, Issue:1

    We evaluated neuropeptide Y (NPY) and galanin (GAL) immunoreactivity (IR) and mRNA in the paraventricular and arcuate nucleus, respectively, in rats that became overweight (Ov) or not (NOv) when fed a cafeteria diet. After 2 months of diet, NOv rats showed a significant increase in NPY IR, whereas Ov rats showed a significant increase in GAL mRNA levels. None of these changes was present in rats overfed for 6.5 months. These differential changes in hypothalamic GAL and NPY transmissions may contribute to the different susceptibility of the two rat subpopulations to the weight-promoting effects of the hypercaloric diet.

    Topics: Animals; Arcuate Nucleus of Hypothalamus; Blood Glucose; Body Weight; Diet; Eating; Fatty Acids; Galanin; Hyperphagia; Immunohistochemistry; In Situ Hybridization; Insulin; Male; Neuropeptide Y; Obesity; Oligodeoxyribonucleotides; Paraventricular Hypothalamic Nucleus; Rats; Rats, Sprague-Dawley; RNA, Messenger

1998
Hyperphagia in cold-exposed rats is accompanied by decreased plasma leptin but unchanged hypothalamic NPY.
    The American journal of physiology, 1998, Volume: 274, Issue:1

    Chronic cold exposure stimulates sympathetically driven thermogenesis in brown adipose tissue (BAT), resulting in fat mobilization, weight loss, and compensatory hyperphagia. Hypothalamic neuropeptide Y (NPY) neurons are implicated in stimulating food intake in starvation, but may also suppress sympathetic outflow to BAT. This study investigated whether the NPY neurons drive hyperphagia in rats that have lost weight through cold exposure. Rats exposed to 4 degrees C for 21 days weighed 14% less than controls maintained at 22 degrees C (P < 0.001). Food intake increased after 3 days and remained 10% higher thereafter (P < 0.001). Increase BAT activity was confirmed by 64, 96, and 335% increases in uncoupling protein-1 mRNA at 2, 8, and 21 days. Plasma leptin decreased during prolonged cold exposure. Cold-exposed rats showed no significant changes in NPY concentrations in any hypothalamic regions or in hypothalamic NPY mRNA at any time. We conclude that the NPY neurons are not activated during cold exposure. This is in contrast with starvation-induced hyperphagia, but is biologically appropriate since enhanced NPY release would inhibit thermogenesis causing potentially lethal hypothermia. Other neuronal pathways must therefore mediate hyperphagia in chronic cold exposure.

    Topics: Acclimatization; Animals; Body Temperature Regulation; Body Weight; Cold Temperature; Feeding Behavior; Hyperphagia; Hypothalamus; Leptin; Male; Neurons; Neuropeptide Y; Obesity; Proteins; Rats; Rats, Wistar; RNA, Messenger; Time Factors; Transcription, Genetic

1998
Effects of palatability-induced hyperphagia and food restriction on mRNA levels of neuropeptide-Y in the arcuate nucleus.
    Brain research, 1998, Sep-21, Volume: 806, Issue:1

    This study examined the effect of feeding either a bland cornstarch-based diet (BCD) or a highly palatable, high fat diet containing sucrose (HPD) on hypothalamic arcuate nucleus (ARC) gene expression for neuropeptide-Y (NPY). Male Sprague-Dawley rats received either BCD ad libitum, HPD ad libitum, HPD pair-fed to the caloric intake of the BCD, or the HPD at 60% of ad libitum HPD intake for 7 days. Animals receiving the HPD ad libitum consumed more calories and gained more weight than animals receiving the BCD (P<0.001). The HPD did not affect ARC NPY mRNA levels, whether the subjects were allowed to overeat or pair-fed to the BCD (P>0.05). However, feeding the HPD at 60% of ad libitum intake of the HPD, increased NPY mRNA levels in the ARC relative to the other treatments (P<0.01). The present data are consistent with the view that NPY in ARC responds to energy deficits rather than to hyperphagia stimuli related to palatability.

    Topics: Animals; Arcuate Nucleus of Hypothalamus; Diet; Dietary Carbohydrates; Dietary Fats; Dietary Sucrose; Food Deprivation; Hyperphagia; Male; Neuropeptide Y; Rats; Rats, Sprague-Dawley; RNA, Messenger; Starch; Sucrose; Taste

1998
Hyperphagia and weight gain after gold-thioglucose: relation to hypothalamic neuropeptide Y and proopiomelanocortin.
    Endocrinology, 1998, Volume: 139, Issue:11

    Genetic obesity is associated with increased neuropeptide Y (NPY) messenger RNA (mRNA) and decreased POMC mRNA in the hypothalamus of ob/ob and db/db mice, or impaired sensitivity to alphaMSH (derived from POMC) in the yellow agouti mouse. Acquired obesity can be produced by chemically lesioning the hypothalamus with either monosodium glutamate (MSG) in neonates or gold thioglucose (GTG) in adult mice. The present study examined whether elevated NPY mRNA and/or decreased POMC mRNA in the hypothalamus are associated with obesity due to hypothalamic lesions. GTG injection into adult mice produced a profound obese phenotype, including hyperphagia, increased body weight, and increased leptin mRNA and peptide, in association with reduced hypothalamic NPY mRNA and POMC mRNA. MSG treatment produced virtual elimination of NPY mRNA in the arcuate nucleus and a reduction of hypothalamic POMC mRNA, and led to elevated leptin. MSG pretreatment did not attenuate GTG-induced hyperphagia and obese phenotype. These results do not support a role for NPY-synthesizing neurons in the arcuate nucleus in mediating hypothalamic acquired obesity, but are consistent with the hypothesis that decreased activity of hypothalamic neurons synthesizing POMC play a role in mediating hypothalamic obesity.

    Topics: Animals; Aurothioglucose; Blotting, Northern; Body Weight; Hyperphagia; Hypothalamus; In Situ Hybridization; Leptin; Mice; Mice, Inbred C57BL; Neuropeptide Y; Obesity; Pro-Opiomelanocortin; Protein Biosynthesis; RNA, Messenger; Sodium Glutamate; Weight Gain

1998
NPY-induced overfeeding suppresses hypothalamic NPY mRNA expression: potential roles of plasma insulin and leptin.
    Regulatory peptides, 1998, Sep-25, Volume: 75-76

    To test the hypothesis that NPY-induced overfeeding activates compensatory responses that inhibit hypothalamic NPY gene expression, we investigated the effect of chronically administered neuropeptide Y (NPY) on plasma hormones involved in energy balance and on the level of mRNA for hypothalamic neuropeptides. After cannulation of the third cerebral ventricle, male Long-Evans rats received a 4.5-day intracerebroventricular (i.c.v.) infusion of either human NPY (12 microg per day), or synthetic cerebrospinal fluid (CSF). NPY-treated animals were either allowed ad libitum access to food or were pairfed to the intake of CSF-treated controls. In rats fed ad libitum, i.c.v. NPY induced significant increases in food intake (75%), body weight (9%), plasma insulin (150%) and plasma leptin levels (300%) as compared to the i.c.v. CSF group. Levels of plasma leptin, but not insulin, remained elevated in NPY-treated rats that were pairfed to the intake of the CSF group. NPY mRNA levels in the midregion of the arcuate nucleus (ARC) were reduced by 50% in NPY-treated rats that were allowed to overeat, but not in the pairfed group, as determined by in situ hybridization. In contrast, mRNA for corticotropin-releasing hormone (CRH) in the paraventricular nucleus (PVN) and proopiomelanocortin (POMC) in the rostral ARC were not significantly different among groups. These findings indicate that NPY-induced overfeeding suppresses ARC NPY mRNA expression, and that this effect unlikely to be mediated by a direct action of NPY, since it was abolished by limiting food intake in NPY-treated animals to that observed in controls. NPY-induced overfeeding was also associated with elevated plasma levels of leptin and insulin. The effect of these hormones to inhibit NPY gene expression may therefore have contributed to the decrease of NPY mRNA.

    Topics: Animals; Arcuate Nucleus of Hypothalamus; Eating; Gene Expression; Humans; Hyperphagia; Hypothalamus; In Situ Hybridization; Injections, Intraventricular; Insulin; Leptin; Male; Neuropeptide Y; Obesity; Paraventricular Hypothalamic Nucleus; Proteins; Rats; RNA, Messenger; Weight Gain

1998
Neuropeptide Y release in the paraventricular nucleus is decreased during transient hyperphagia induced by microinjection of colchicine into the ventromedial nucleus of rats.
    Neuroscience letters, 1998, Oct-30, Volume: 256, Issue:1

    Disruption of neural signaling in the ventromedial nucleus (VMN) of rats by microinjection of the neurotoxin colchicine (COL) results in transient hyperphagia accompanied by enhanced weight gain. We tested the hypothesis that release of neuropeptide Y (NPY), a potent orexigenic signal is augmented within the paraventricular nucleus (PVN) of COL-treated hyperphagic rats. Adult male rats were microinjected bilaterally with either COL (4 microg/0.5 microl in saline) or saline in the VMN and a push-pull guide cannula aimed at the PVN was implanted for analysis of extra-cellular NPY. COL-injected rats gained 37.8+/-6.1 g while the saline-injected rats lost 9.3+/-3.4 g during the 4 days following surgery. On day 4, post-injection, the PVN of these rats was perfused with artificial cerebrospinal fluid via the push-pull cannula. NPY levels in perfusates collected at 10 min intervals from hyperphagic, COL-injected rats were markedly diminished. Cumulative NPY efflux over the 180 min sampling period was significantly less in COL-treated (27.7+/-6.0 pg) versus saline-injected control rats (110.6+/-32.2 pg; P < 0.05). These results show that impairment of neural signaling in the VMN by COL suppressed NPY release in the PVN. These observations taken together with previous studies showing diminution in preproNPY mRNA in the arcuate nucleus (ARC) and NPY levels in the PVN are in accordance with the thesis that the VMN normally exerts a facilitatory influence on NPYergic signaling in the ARC-PVN axis.

    Topics: Animals; Colchicine; Hyperphagia; Male; Microinjections; Neuropeptide Y; Neurotoxins; Paraventricular Hypothalamic Nucleus; Rats; Rats, Sprague-Dawley; Weight Gain

1998
Hyperphagia induced by hypoglycemia in rats is independent of leptin and hypothalamic neuropeptide Y (NPY).
    Peptides, 1998, Volume: 19, Issue:9

    Hypoglycemia causes hyperphagia and weight gain, through unknown peripheral and central signals. We investigated the effect of hypoglycemia on NPY and leptin expression and the ability of leptin to inhibit hypoglycemia-induced hyperphagia. Acute hypoglycemia (60 U/kg SC insulin; n = 8) increased food intake (p < 0.01) compared with controls (n = 8). Insulin- and leptin-treated rats (300 microg/kg IP leptin; n = 8) had reduced hyperphagia (p < 0.05 vs. controls; p < 0.05 vs. insulin alone) and a 15% fall in NPY mRNA levels compared with controls (p < 0.01). Chronic hypoglycemia, (20-60 U/kg/day insulin; n = 8) increased food intake compared with vehicle-treated controls (p < 0.01). Leptin and insulin administration (300 microg/kg/day IP leptin; n = 8) reduced hyperphagia (p < 0.01 vs. controls, p < 0.05 vs. insulin alone), and NPY mRNA fell by 18% vs. controls (p < 0.01). We conclude that hypoglycemia-induced hyperphagia is not mediated by either a fall in leptin or an increase in hypothalamic NPY mRNA. Leptin can inhibit feeding in hyperphagic hypoglycemic rats, and this may partly be attributable to its inhibition of the NPY neurons.

    Topics: Acute Disease; Analysis of Variance; Animals; Arcuate Nucleus of Hypothalamus; Blood Glucose; Body Weight; Chronic Disease; Eating; Hyperphagia; Hypoglycemia; Insulin; Leptin; Male; Models, Biological; Neuropeptide Y; Protein Biosynthesis; Proteins; Rats; Rats, Wistar; RNA, Messenger

1998
Interactions between leptin and hypothalamic neuropeptide Y neurons in the control of food intake and energy homeostasis in the rat.
    Diabetes, 1997, Volume: 46, Issue:3

    Leptin acts on the brain to inhibit feeding, increase thermogenesis, and decrease body weight. Neuropeptide Y (NPY)-ergic neurons of the hypothalamic arcuate nucleus (ARC) that project to the paraventricular nuclei (PVN) and dorsomedial nuclei (DMH) are postulated to control energy balance by stimulating feeding and inhibiting thermogenesis, especially under conditions of energy deficit. We investigated whether leptin's short-term effects on energy balance are mediated by inhibition of the NPY neurons. Recombinant murine leptin (11 microg) injected into the lateral ventricle of fasted adult Wistar rats inhibited food intake by 20-25% between 2 and 6 h after administration, compared with saline-treated controls (P < 0.05). Uncoupling protein mRNA levels in brown adipose tissue (BAT) rose by 70% (P < 0.01). Leptin treatment significantly reduced NPY concentrations by 20-50% (P < 0.05) in the ARC, PVN, and DMH and significantly decreased hypothalamic NPY mRNA levels (0.61 +/- 0.02 vs. 0.78 +/- 0.03 arbitrary units; P < 0.01). A second study examined changes in leptin during 5 days' intracerebroventricular NPY administration (10 microg/day), which induced sustained hyperphagia and excessive weight gain. In NPY-treated rats, leptin mRNA levels in epididymal fat were comparable to those in saline-treated controls (0.94 +/- 0.17 vs. 1.0 +/- 0.28 arbitrary units; P > 0.1), but plasma leptin levels were significantly higher (4.88 +/- 0.66 vs. 2.85 +/- 0.20 ng/ml; P < 0.01). Leptin therefore acts centrally to decrease NPY synthesis and NPY levels in the ARC-PVN projection; reduced NPY release in the PVN may mediate leptin's hypophagic and thermogenic actions. Conversely, NPY-induced obesity results in raised circulating leptin concentrations. Leptin and the NPY-ergic ARC-PVN neurons may interact in a homeostatic loop to regulate body fat mass and energy balance.

    Topics: Adipose Tissue, Brown; Animals; Carrier Proteins; Cerebral Ventricles; Feeding Behavior; Hyperphagia; Hypothalamus; Infusions, Parenteral; Ion Channels; Leptin; Male; Membrane Proteins; Mitochondria; Mitochondrial Proteins; Neurons; Neuropeptide Y; Obesity; Oligonucleotide Probes; Protein Biosynthesis; Proteins; Rats; Rats, Wistar; Recombinant Proteins; RNA, Messenger; Transcription, Genetic; Uncoupling Protein 1; Weight Gain

1997
Evidence that hypothalamic neuropeptide Y gene expression and NPY levels in the paraventricular nucleus increase before the onset of hyperphagia in experimental diabetes.
    Brain research, 1997, May-02, Volume: 755, Issue:2

    Neuropeptide Y (NPY) is the most potent endogenous orexigenic signal. Several lines of evidence indicate that the site of NPY action in transducing feeding signal may reside in the paraventricular nucleus (PVN) and neighboring sites in the hypothalamus. To test the hypothesis that an increase in NPY activity in the ARC-PVN pathway precedes the onset of diabetic hyperphagia, we evaluated NPY levels in seven hypothalamic nuclei and NPY gene expression in the hypothalamus at 48, 72 or 96 h after streptozotocin (STZ) treatment in rat. In STZ-treated diabetic rats, NPY gene expression in the hypothalamus and NPY levels only in the PVN significantly elevated at 48 h, while hyperphagia occurred sometimes after 48 h post-injection. These results show that augmentation in NPY neuronal activity in the ARC-PVN axis precedes the onset of increased food intake produced by STZ-induced insulinopenia. These findings affirm the hypothesis that increased NPY neurosecretion in the PVN may underlie the diabetes-induced hyperphagia.

    Topics: Animals; Diabetes Mellitus, Experimental; Gene Expression; Hyperphagia; Hypothalamus; Male; Neuropeptide Y; Paraventricular Hypothalamic Nucleus; Rats; Rats, Sprague-Dawley

1997
Increased feeding in fatty Zucker rats by the thiazolidinedione BRL 49653 (rosiglitazone) and the possible involvement of leptin and hypothalamic neuropeptide Y.
    British journal of pharmacology, 1997, Volume: 122, Issue:7

    1. The thiazolidinedione BRL 49653 (rosiglitazone) induces hyperphagia and weight gain in obese, insulin-resistant fatty Zucker rats but not in lean insulin-sensitive rats. We investigated whether these responses might involve neuropeptide Y (NPY), leptin and insulin. 2. BRL 49653 (1 mg kg(-1) day(-1), orally) was given for 7 or 20 days to fatty and lean Zucker and Wistar rats. 3. In lean rats of either strain, BRL 49653 had no effect on food intake, body weight, plasma insulin and corticosterone, NPY or NPY mRNA levels. 4. Fatty rats given BRL 49653 showed a 30% increase in food intake and accelerated body weight gain (both P<0.01) after 7 and 20 days, but without significant changes in regional hypothalamic NPY or NPY mRNA levels. 5. Plasma leptin levels were twice as high in untreated fatty Zucker rats as in lean rats (P<0.01), but were unaffected by BRL 49653 given for 20 days. However, BRL 49653 reduced insulin levels by 42% and increased corticosterone levels by 124% in fatty rats (both P<0.01). 6. Hyperphagia induced in fatty Zucker rats by BRL 49653 does not appear to be mediated by either a fall in circulating leptin levels or increased activity of hypothalamic NPYergic neurones. The fall in plasma insulin and/or rise in corticosterone levels during BRL 49653 treatment may be involved, consistent with the postulated role of these hormones in the control of food intake.

    Topics: Animals; Appetite Stimulants; Body Weight; Eating; Hyperphagia; Hypoglycemic Agents; Hypothalamus; Leptin; Male; Neuropeptide Y; Proteins; Rats; Rats, Wistar; Rats, Zucker; Rosiglitazone; Thiazoles; Thiazolidinediones

1997
Increased hypothalamic neuropeptide Y concentration or hyperphagia in streptozotocin-diabetic rats are not mediated by glucocorticoids.
    European journal of pharmacology, 1997, Dec-11, Volume: 340, Issue:2-3

    Hypothalamic neuropeptide Y containing neurones are overactive and may mediate hyperphagia in insulin-deficient diabetic rats, but the factors stimulating them remain uncertain. To determine the possible role of glucocorticoids, we investigated the effects of the glucocorticoid receptor blocker mifepristone (RU486) on food intake and regional hypothalamic neuropeptide Y concentrations in streptozotocin-diabetic rats. RU486 (30 mg/kg) or corn oil vehicle control was given orally for 3 weeks to diabetic rats. Food intake and neuropeptide Y levels in the hypothalamic arcuate and paraventricular nuclei were increased in untreated diabetic rat groups (P < 0.01), and though RU486 did increase plasma corticosterone levels (P < 0.01) it did not have any effect on either feeding or neuropeptide Y levels (P = NS). These negative findings suggest that glucocorticoids may not be responsible for increasing hypothalamic neuropeptide Y or for hyperphagia in insulin-deficient diabetes.

    Topics: Animals; Blood Glucose; Body Weight; Diabetes Mellitus, Experimental; Drinking; Eating; Hormone Antagonists; Hyperphagia; Hypothalamus; Insulin; Male; Mifepristone; Neuropeptide Y; Rats; Rats, Wistar; Receptors, Glucocorticoid

1997
Increased receptor sensitivity to neuropeptide Y in the hypothalamus may underlie transient hyperphagia and body weight gain.
    Regulatory peptides, 1997, Oct-31, Volume: 72, Issue:2-3

    Disruption of neural signaling by microinjection of a neurotoxin, colchicine (COL), in the ventromedial hypothalamus (VMH) of rats results in rapid and transient hyperphagia and body weight gain. Since neuropeptide Y (NPY) is a potent hypothalamic orexigenic signal and continuous NPY receptor activation by intracerebroventricular (icv) NPY infusion results in hyperphagia and obesity, we tested the hypothesis that altered NPYergic signaling may underlie the transient hyperphagia in COL-injected rats. Immediately following COL (4 microg) microinjections in the ventromedial nucleus (VMN) rats displayed hyperphagia both during the lights-on and lights-off periods. Concomitant with hyperphagia, preproNPY mRNA levels in the arcuate nucleus and NPY levels in the paraventricular nucleus decreased in a time-dependent manner. However, food intake in response to intracerebroventricular injections of NPY (29, 117 and 470 pmole) was significantly higher in COL-injected rats and the latency to initiation of feeding was markedly reduced as compared to controls. The smallest dose of NPY which was virtually ineffective in control rats, evoked near maximal intake in COL-injected rats. This enhanced response lasted for only 4 days paralleling the transient hyperphagia. The NPY Y1 receptor antagonist 1229U91 (5 or 30 microg/rat, icv) significantly suppressed feeding in COL-treated rats thereby indicating that hyperphagia in these rats was dependent upon endogenous NPY. Overall, these studies demonstrate that not only high levels, but low levels of NPY may also result in hyperphagia and increased body weight and this hyperphagia may be attributed to the rapid development of NPY Y1 receptor hypersensitivity.

    Topics: Animals; Appetite Stimulants; Behavior, Animal; Binding, Competitive; Colchicine; Darkness; Dose-Response Relationship, Drug; Eating; Hyperphagia; Hypothalamus; Injections, Intraventricular; Light; Male; Neuropeptide Y; Obesity; Peptides, Cyclic; Rats; Rats, Sprague-Dawley; Receptors, Neuropeptide Y; RNA, Messenger; Signal Transduction; Ventromedial Hypothalamic Nucleus; Weight Gain

1997
Sensitivity to leptin and susceptibility to seizures of mice lacking neuropeptide Y.
    Nature, 1996, May-30, Volume: 381, Issue:6581

    Neuropeptide Y (NPY), a 36-amino-acid transmitter distributed throughout the nervous system, is thought to function as a central stimulator of feeding behaviour. NPY has also been implicated in the modulation of mood, cerebrocortical excitability, hypothalamic-pituitary signalling, cardiovascular physiology and sympathetic function. However, the biological significance of NPY has been difficult to establish owing to a lack of pharmacological antagonists. We report here that mice deficient for NPY have normal food intake and body weight, and become hyperphagic following food deprivation. Mutant mice decrease their food intake and lose weight, initially to a greater extent than controls, when treated with recombinant leptin. Occasional, mild seizures occur in NPY-deficient mice and mutants are more susceptible to seizures induced by a GABA (gamma-aminobutyric acid) antagonist. These results indicate that NPY is not essential for certain feeding responses or leptin actions but is an important modulator of excitability in the central nervous system.

    Topics: Animals; Body Weight; Eating; Feeding Behavior; Female; Food Deprivation; GABA Antagonists; Gene Targeting; Humans; Hyperphagia; Leptin; Male; Mice; Neuropeptide Y; Pentylenetetrazole; Proteins; Seizures

1996
Stimulation of 5-HT2A receptors in the paraventricular hypothalamus attenuates neuropeptide Y-induced hyperphagia through activation of corticotropin releasing factor.
    Brain research, 1996, Feb-05, Volume: 708, Issue:1-2

    The effect of 5-HT1 and 5-HT2 receptor agonists administered into the paraventricular hypothalamus was studied on the hyperphagia caused by neuropeptide Y (NPY) injected into the same area. The 5-HT2A/2C receptor agonist DOI (10-20 nmol/0.5 microliter) significantly reduced NPY overeating while the 5-HT1A/1B receptor agonist RU 24969 (3.5-14 nmol/0.5 microliter) and the 5-HT1B/2C receptor agonist mCPP (5-20 nmol/0.5 microliter) had no such effect. The 5-HT2A receptor antagonist spiperone (5 microgram/0.5 microliter) and the corticotropin releasing factor antagonist alpha-helical-CRF9-41 (0.5-1 micrograms/0.5 microliter) completely antagonized the effect of 10 nmol DOI.

    Topics: Amphetamines; Animals; Corticotropin-Releasing Hormone; Feeding Behavior; Hyperphagia; Indoles; Male; Neuropeptide Y; Paraventricular Hypothalamic Nucleus; Piperazines; Rats; Rats, Sprague-Dawley; Receptor, Serotonin, 5-HT2A; Receptors, Serotonin; Serotonin Receptor Agonists; Spiperone

1996
Intracerebroventricular administration of neuropeptide Y to normal rats increases obese gene expression in white adipose tissue.
    Diabetologia, 1996, Volume: 39, Issue:3

    The aim of this work was to determine the possible inter-relationship between neuropeptide Y (NPY, a hypothalamic stimulator of feeding) and adipose tissue expression of the ob protein (a novel potent inhibitor of feeding). Such a relationship could be of importance in the maintenance of normal body weight. To this end, normal rats were intracerebroventricularly (i.c.v.) infused for 6 days with NPY. NPY infusion resulted in hyperphagia and a marked increase in adipose tissue ob mRNA levels. The effect of NPY on ob expression persisted when hyperphagia was prevented by pair-feeding, and was reversed following cessation of NPY infusion. Basal and glucose-stimulated insulinaemia were increased by i.c.v. NPY infusion compared to control values, regardless of whether animals were ad libitum-fed or pair-fed. Cessation of NPY infusion was accompanied by normalisation of insulinaemia. These changes in insulinaemia produced by i.c.v. NPY infusion paralleled the observed changes in ob expression. When normal rats were made hyperinsulinaemic-euglycaemic for 24 h, such hyperinsulinaemia also resulted in increased ob mRNA levels in white adipose tissue. This suggested that NPY-induced hyperinsulinaemia could be responsible for the upregulation of ob mRNA levels of NPY-infused rats. It is concluded that central (i.c.v.) NPY infusion increases adipose tissue ob expression, a functional relationship that is linked, at least in part, via NPY-induced hyperinsulinaemia.

    Topics: Adipose Tissue; Animals; Blood Glucose; Cerebral Ventricles; Female; Gene Expression; Glucose Clamp Technique; Hyperinsulinism; Hyperphagia; Infusions, Parenteral; Insulin; Leptin; Neuropeptide Y; Protein Biosynthesis; Rats; Rats, Zucker; Reference Values

1996
Effects of gold thioglucose on neuropeptide Y messenger RNA levels in the mouse hypothalamus.
    The American journal of physiology, 1996, Volume: 270, Issue:6 Pt 2

    Elevated hypothalamic neuropeptide Y (NPY) expression is found in several rodent genetic models of obesity, but any association in nongenetic models of obesity is unclear. Consequently, we have measured NPY mRNA levels in the ventromedial hypothalamus of a well-characterized model of obesity, the gold thioglucose (GTG)-injected mouse. Fourteen days after injection (early stage), animals were hyperphagic but not obese, hyperglycemic, or overtly hyperinsulinemic. Ten weeks after treatment (late stage), animals were obese, markedly hyperinsulinemic, and hyperglycemic. In both the early and late stages, NPY mRNA levels were reduced in the arcuate nucleus of GTG-injected animals. Although overnight fasting doubled NPY mRNA levels in control animals, there was no change at either stage in GTG-injected animals. NPY mRNA levels in the deep layers of the cerebral cortex and in the dentate gyrus were not affected by GTG treatment or overnight fasting. We conclude that GTG treatment reduces the expression of NPY mRNA in the arcuate nucleus and that, therefore, increased hypothalamic NPY expression is unlikely to be an important factor causing the obesity and other metabolic changes found in this model.

    Topics: Animals; Arcuate Nucleus of Hypothalamus; Aurothioglucose; Hyperglycemia; Hyperinsulinism; Hyperphagia; In Situ Hybridization; Male; Mice; Mice, Inbred CBA; Neuropeptide Y; Obesity; RNA, Messenger; Ventromedial Hypothalamic Nucleus

1996
Behavioral, endocrine, and hypothalamic responses to involuntary overfeeding.
    The American journal of physiology, 1996, Volume: 271, Issue:3 Pt 2

    The suppression of food intake after a period of forced overfeeding is potent and long lasting, yet little is known of the underlying mechanisms for this regulatory response. Rats were overfed via a surgically implanted gastrostomy tube. During overfeeding, plasma insulin and corticotropin-releasing hormone (CRH) mRNA in the paraventricular nucleus of the hypothalamus were elevated compared with controls and with overfed rats allowed 3 days to recover from the overfeeding regimen such that body weight returned to the level of controls. In contrast, rats that were not overfed but were pair-fed to the low spontaneous food intake of previously overfed rats lost weight and had significantly reduced plasma insulin and elevated mRNA for neuropeptide Y (NPY) in the arcuate nucleus of the hypothalamus. The results indicate that overfeeding produces an activation of hypothalamic CRH system that may contribute to the hypophagia that accompanies involuntary overfeeding. Furthermore, the hypothalamic NPY response to food restriction is not tied to low food intake per se, but rather to negative energy balance.

    Topics: Animals; Behavior, Animal; Body Weight; Brain; Eating; Endocrine Glands; Hormones; Hyperphagia; Hypothalamus; In Situ Hybridization; Intubation, Gastrointestinal; Male; Neuropeptide Y; Rats; Rats, Inbred Strains; RNA, Messenger

1996
Intracerebroventricular neuropeptide Y produces hyperinsulinemia in the presence and absence of food.
    Physiology & behavior, 1996, Volume: 60, Issue:3

    Acute administration of neuropeptide Y into the hypothalamus or cerebral ventricles produces hyperphagia and hyperinsulinemia. However, it is not known to what extent the hyperinsulinemia depends on the food intake. Consequently, serum insulin and glucose, as well as food and water consumption, were measured over 3 h, following injection of 1-20 micrograms neuropeptide Y into the third ventricle of adult female rats. In the presence of food, 1-10 micrograms neuropeptide Y produced a dose-dependent increase in food and water intake and serum insulin. Insulin levels were closely correlated with the quantity of food ingested. In the absence of food, 1-20 micrograms neuropeptide Y produced a dose-dependent increase in water intake, whereas 1-5 micrograms produced a does-dependent increase in serum insulin. We concluded that ICV neuropeptide Y can stimulate insulin secretion even at low doses and this response does not completely depend on food intake.

    Topics: Animals; Dose-Response Relationship, Drug; Eating; Female; Hyperinsulinism; Hyperphagia; Injections, Spinal; Neuropeptide Y; Rats; Rats, Wistar

1996
Withdrawal of [corrected] estrogen increases hypothalamic neuropeptide Y (NPY) mRNA expression in ovariectomized obese rat .
    Neuroscience letters, 1996, Feb-02, Volume: 204, Issue:1-2

    We determined the changes in neuropeptide Y (NPY) mRNA expression of the arcuate nucleus (ARC) in sham-operated (SHAM) and bilaterally ovariectomized (OVX) rats with estradiol (E2) supplement. Ovariectomy increases body weight gain for 3 weeks, accompanied by an increase of daily food intake. Ovariectomy significantly reduced serum corticosterone levels. E2 supplement reversed the effects of ovariectomy on body weight gain, food intake and serum corticosterone levels. Ovariectomy significantly increased NPY mRNA expression in the ARC. E2 supplement decreased NPY mRNA expression in the ARC of OVX rats. The present findings indicated that hypothalamic NPY mRNA expression, which involves the regulation of feeding behavior, are in parallel with circulating estrogen levels. Hypothalamic NPY mRNA expression may be important in the induction of hyperphagia after the withdrawal of estrogen by bilateral ovariectomy.

    Topics: Animals; Arcuate Nucleus of Hypothalamus; Base Sequence; DNA Primers; Estradiol; Female; Hyperphagia; Hypothalamus; Molecular Sequence Data; Neuropeptide Y; Obesity; Ovariectomy; Rats; Rats, Wistar; RNA, Messenger; Stimulation, Chemical

1996
The 5-HT1B receptor mediates the effect of d-fenfluramine on eating caused by intra-hypothalamic injection of neuropeptide Y.
    European journal of pharmacology, 1995, Feb-14, Volume: 274, Issue:1-3

    d-Fenfluramine (0.63 mg/kg i.p.), a serotonin (5-hydroxytryptamine, 5-HT) releaser and re-uptake inhibitor, reduced the eating caused by neuropeptide Y (235 pmol) injected into the paraventricular nucleus of the hypothalamus. The 5-HT1 and 5-HT2 receptor antagonist metergoline (1.0 and 2.0 mg/kg i.p.) and the 5-HT1A and 5-HT1B receptor antagonist (+/-)-cyanopindolol (3.0 and 8.0 mg/kg s.c.) significantly antagonized the effect of d-fenfluramine. The 5-HT2A and 5-HT2C receptor antagonist mesulergine (0.1 and 0.3 mg/kg s.c.) and the 5-HT2A receptor antagonist ketanserin (2.5 and 5.0 mg/kg i.p.) did not significantly modify the effect, nor did the 5-HT1A and 5-HT1B receptor antagonist (-)-propranolol (20-40 nmol), injected bilaterally into the paraventricular nucleus of the hypothalamus. The results suggest that d-fenfluramine reduces neuropeptide Y's hyperphagia by indirectly stimulating 5-HT1B receptors outside the paraventricular nucleus of the hypothalamus.

    Topics: Analysis of Variance; Animals; Antiparkinson Agents; Drug Interactions; Eating; Ergolines; Fenfluramine; Hyperphagia; Hypothalamus; Injections, Intraperitoneal; Injections, Subcutaneous; Ketanserin; Male; Metergoline; Neuropeptide Y; Paraventricular Hypothalamic Nucleus; Pindolol; Rats; Rats, Sprague-Dawley; Receptor, Serotonin, 5-HT1B; Receptors, Serotonin; Serotonin Antagonists

1995
Effect of intracerebroventricular insulin infusion on diabetic hyperphagia and hypothalamic neuropeptide gene expression.
    Diabetes, 1995, Volume: 44, Issue:2

    To test the hypothesis that diabetic hyperphagia results from insulin deficiency in the brain, diabetic rats (streptozotocin-induced) were given an intracerebroventricular (ICV) infusion of saline or insulin (at a dose that did not affect plasma glucose levels) for 6 days. Food and water intake were significantly increased in diabetic rats, but only food intake was affected by ICV insulin. Diabetic hyperphagia was reduced 58% by ICV insulin compared with ICV saline (P < 0.05) and was accompanied by a 69% increase in diabetes-induced weight loss (P < 0.05). To evaluate whether central nervous system (CNS) insulin deficiency affects expression of neuropeptides involved in food intake, in situ hybridization was done for neuropeptide Y (NPY), which stimulates feeding, in the hypothalamic arcuate nucleus and for cholecystokinin (CCK) and corticotropin-releasing hormone (CRH), which inhibit feeding, in the hypothalamic paraventricular nucleus. In diabetic rats, NPY mRNA hybridization increased 280% (P < 0.05), an effect reduced 40% by ICV insulin (P < 0.05). CCK mRNA hybridization increased 50% in diabetic rats (P < 0.05), a response reduced slightly by ICV insulin (P < 0.05), whereas CRH mRNA hybridization decreased 33% in diabetic rats (P < 0.05) and was unchanged by ICV insulin. The results demonstrate that CNS infusion of insulin to diabetic rats reduces both hyperphagia and overexpression of hypothalamic NPY mRNA. This observation supports the hypothesis that a deficiency of insulin in the brain is an important cause of diabetic hyperphagia and that increased hypothalamic NPY gene expression contributes to this phenomenon.

    Topics: Animals; Base Sequence; Brain; Cholecystokinin; Corticotropin-Releasing Hormone; Diabetes Mellitus, Experimental; Gene Expression; Hyperphagia; Hypothalamus; In Situ Hybridization; Insulin; Male; Molecular Sequence Data; Neuropeptide Y; Neuropeptides; Rats; Rats, Wistar; RNA, Messenger

1995
Evidence of a physiological role for neuropeptide Y in ventromedial hypothalamic lesion-induced hyperphagia.
    Brain research, 1995, Sep-04, Volume: 690, Issue:2

    We evaluated the role of neuropeptide Y (NPY), a potent endogenous orexigenic signal, in the ventromedial hypothalamic (VMH) lesion-induced hyperphagia in rats. To produce hyperphagia and excessive weight gain, adult female rats received bilateral electrolytic or sham lesions in the VMH. Concurrently, a permanent intracerebroventricular cannula was implanted in the third ventricle of the brain. After a recovery period, these rats were passively immunized against NPY to evaluate the role of endogenous NPY on hyperphagia. The results showed that intraventricular administration of NPY antibodies abolished the hyperphagia in VMH-lesioned rats. These revelations are in agreement with the notion that altered hypothalamic NPY release or action may underlie the hyperphagia and excessive weight gain seen in response to structural damage in the VMH.

    Topics: Animals; Electric Stimulation; Female; Hyperphagia; Hypothalamus, Middle; Immunization, Passive; Injections, Intraventricular; Neuropeptide Y; Rats; Rats, Sprague-Dawley; Weight Gain

1995
Chronic cerebroventricular galanin does not induce sustained hyperphagia or obesity.
    Peptides, 1994, Volume: 15, Issue:7

    Acute central administration of galanin has been reported to increase fat consumption. These experiments were designed to test the hypothesis that repeated injections of galanin would elicit hyperphagia and weight gain and that this response would depend on the available diet. Male Sprague-Dawley rats were fed high (56% energy) or low (10% energy) fat diets. Galanin (300 pmol) or saline vehicle was injected into the third ventricle twice daily for 7 days and three times daily for another 6 days. On both the high-carbohydrate and high-fat diets, twice daily galanin increased daytime food intake, but there was a compensatory decrease in nighttime intake. The addition of a third, nighttime injection of galanin was ineffective in producing an increase in total 24-h intake. There was no significant increase in body weight during galanin treatment in rats eating either diet although animals eating the high-fat diet gained more weight as reflected by a significant increase in epididymal fat pad weight. Galanin treatment had no effects on serum insulin, glucose or corticosterone concentrations, measured at the end of the experiment. However, animals fed the high-fat diet had significantly higher insulin concentrations at the time of sacrifice. Although repeated central infusions of galanin reliably stimulated daytime intake of both diets, they failed to increase total daily energy intake or body weight.

    Topics: Animals; Blood Glucose; Body Weight; Corticosterone; Dietary Carbohydrates; Dietary Fats; Eating; Galanin; Hyperphagia; Injections, Intraventricular; Insulin; Male; Neuropeptide Y; Obesity; Peptides; Rats; Rats, Sprague-Dawley

1994
Putative neuropeptide Y antagonist failed to decrease overeating in obese Zucker rats.
    Neuroscience letters, 1994, Nov-07, Volume: 181, Issue:1-2

    A central dysregulation of several neuropeptides could be at the origin of the marked hyperphagia of the obese Zucker rat, a well-known animal model used for the study of obesity. Neuropeptide Y (NPY), which stimulates food intake and increases early in life in obese rats, plays a major role in the development of this hyperphagia. The aim of our experiment was to test a proposed NPY antagonist namely PYX-2 in obese hyperphagic Zucker rats in order to know if it could be an interesting drug for limiting their food intakes. Four doses of PYX-2 (50-1000 pmol) were injected in a counterbalanced order in the lateral brain ventricles of 10 adult male Zucker rats. Food intake was recorded 0.5, 1, 2, 3, 6, and 23 h after PYX-2 injection and compared either to the rat's spontaneous food intake or to the food intake following injection of artificial CSF (vehicle) only. It was not modified by any dose of PYX-2 whatever the time considered (1 h after injection: 4.3 +/- 0.5 (1000 pmol) vs 4.6 +/- 0.8 (CSF) g; 23 h period: 27.0 +/- 1.9 (1000 pmol) vs 26.6 +/- 2.9 (CSF) g; N.S.). Thus, PYX-2, the putative NPY antagonist, totally failed to inhibit food intake in the obese rats. The absence of effect of PYX-2 on food intake can be explained by the structure of PYX-2, a modified 27-36 amino acid sequence that may not be recognized by the Y1-type NPY receptors which are involved in the regulation of feeding behavior.

    Topics: Amino Acid Sequence; Animals; Eating; Hyperphagia; Injections, Intraventricular; Male; Neuropeptide Y; Obesity; Peptide Fragments; Rats; Rats, Zucker

1994
Increased neuropeptide Y concentrations in specific hypothalamic regions of lactating rats: possible relationship to hyperphagia and adaptive changes in energy balance.
    Peptides, 1994, Volume: 15, Issue:1

    Lactation is accompanied by hyperphagia and a reduction in brown adipose tissue (BAT) thermogenesis, which are unexplained. Neuropeptide Y (NPY) powerfully stimulates feeding and inhibits BAT thermogenesis when injected into the paraventricular nucleus and other specific regions of the rat hypothalamus. We have tested the hypothesis that hypothalamic NPY activity is increased in lactating rats. Lactating rats consumed over four times as much food as nonlactating controls (n = 10; p < 0.001). Final plasma insulin concentrations in lactating rats were lower than in controls (6.8 +/- 0.8 vs. 11.7 +/- 2.1 pmol/l; p < 0.05) although plasma glucose and corticosterone concentrations were comparable (p > 0.05). Lactating rats showed significantly higher NPY levels than controls in specific hypothalamic regions, namely the arcuate nucleus-median eminence complex (a 41% rise; p < 0.001), paraventricular nucleus (35%; p < 0.001), ventromedial nucleus (66%; p = 0.003), and dorsomedial nucleus (78%; p < 0.001). Other hypothalamic regions showed no significant differences between groups. Increased NPY concentrations in specific hypothalamic regions, particularly the arcuate nucleus where NPY is synthesized, suggest increased activity of the hypothalamic NPYergic system in lactation. Neuropeptide Y may mediate hyperphagia and reduced BAT thermogenesis in lactation. Hypoinsulinemia may be a stimulus to hypothalamic NPY in lactation, as has been postulated in other conditions of negative energy balance.

    Topics: Adaptation, Physiological; Animals; Body Weight; Energy Metabolism; Female; Hyperphagia; Hypothalamus; Lactation; Neuropeptide Y; Rats; Rats, Wistar

1994
Neuropeptide Y release from the paraventricular nucleus increases in association with hyperphagia in streptozotocin-induced diabetic rats.
    Endocrinology, 1992, Volume: 131, Issue:6

    We tested the hypothesis that the hyperphagia observed in streptozotocin (STZ)-induced diabetic rats is due to increased release of neuropeptide Y (NPY) in the paraventricular nucleus (PVN) of the hypothalamus. In the first experiment, male rats were injected with STZ or vehicle (control) via the tail vein and 18-20 days later, NPY levels in seven hypothalamic sites and release in vitro from selected hypothalamic sites were evaluated. The results showed that in association with STZ-produced marked hyperglycemia and hyperphagia, NPY concentrations were increased in four hypothalamic sites, including the PVN. Evaluation of NPY release in vitro showed that both basal and KCl-induced release was significantly higher from the micro-dissected PVN of STZ-treated than control rats. A similar augmentation in the NPY efflux in vitro was detected from the median eminence arcuate nucleus, but not from the neighboring ventromedial nucleus of STZ-treated rats. In the second experiment, rats were treated with STZ or vehicle and received permanent push-pull cannula (PPC) in the PVN for evaluation of NPY release in vivo 18-21 days after STZ treatment. The results showed that mean NPY levels in the perfusates collected from the PVN of diabetic rats were significantly higher as compared to control rats. Since NPY is the most potent naturally occurring orexigenic signal and the PVN is an important initial site of NPY action in the stimulatory pathway regulating feeding, our findings of augmented PVN NPY release in vivo and in vitro are in accord with the hypothesis that increased NPY secretion in the PVN may be responsible for hyperphagia in diabetic rats.

    Topics: Animals; Diabetes Mellitus, Experimental; Hyperphagia; Male; Neuropeptide Y; Paraventricular Hypothalamic Nucleus; Potassium Chloride; Rats; Rats, Sprague-Dawley

1992
Unexpected regulation of hypothalamic neuropeptide Y by food deprivation and refeeding in the Zucker rat.
    Life sciences, 1992, Volume: 50, Issue:13

    Neuropeptide Y strongly stimulates food intake when it is injected in the hypothalamic paraventricular (PVN) and ventromedian (VMN) nuclei. In Sprague-Dawley (SD) rats, NPY synthesis in the arcuate nucleus (ARC) is increased by food deprivation and is normalized by refeeding. We have previously shown that the obese hyperphagic Zucker rat is characterized by higher NPY concentrations in this nucleus. NPY might therefore play an important role in the development of hyperphagia. The aim of the present study was to determine if the regulation by the feeding state works in the obese Zucker rat. For this purpose, 10 weeks-old male lean (n = 30) and obese (n = 30) Zucker rats were either fed ad libitum, either food-deprived (FD) for 48 hours or food-deprived for 48 h and refed (RF) for 6 hours. NPY was measured in several microdissected brain areas involved in the regulation of feeding behavior. NPY concentrations in the ARC was about 50% greater in obese rats than in lean rats (p less than 0.02) whatever the feeding state. In the VMN, NPY concentrations were higher in the lean FD rats than in the obese FD rat (p less than 0.001). Food deprivation or refeeding did not modify NPY in the ARC, in the VMN or in the dorsomedian nucleus whatever the genotype considered. On the other hand, food deprivation induced a significant decrease in NPY concentrations in the PVN of lean rats. This decrease was localized in the parvocellular part of this nucleus (43.0 +/- 1.9 (FD) vs 54.2 +/- 2.1 (Ad lib) ng/mg protein; p less than 0.005). Ad lib levels were restored by 6 hours of refeeding. These variations were not observed in the obese rat. The regulation of NPY by the feeding state in the Zucker rat was therefore very different from that described in the SD rats. Strain or age of the animals used might explain these differences. High NPY levels and absence of regulation in obese Zucker rats could contribute to the abnormal feeding behavior of these rats.

    Topics: Animals; Arcuate Nucleus of Hypothalamus; Body Weight; Dorsomedial Hypothalamic Nucleus; Eating; Feeding Behavior; Food Deprivation; Hyperphagia; Hypothalamus; Male; Neuropeptide Y; Obesity; Paraventricular Hypothalamic Nucleus; Radioimmunoassay; Random Allocation; Rats; Rats, Zucker; Ventromedial Hypothalamic Nucleus

1992
Effects of intracerebroventricular D-myo-inositol-1,2,6-trisphosphate (PP56), a proposed neuropeptide Y (NPY) antagonist, on locomotor activity, food intake, central effects of NPY and NPY-receptor binding.
    European journal of pharmacology, 1991, Dec-10, Volume: 209, Issue:1-2

    D-myo-Inositol-1,2,6-trisphosphate (PP56) is a novel experimental drug which is structurally related to the intracellular second messenger IP3. Among other pharmacological effects, PP56 has been shown to antagonize neuropeptide Y (NPY) induced vasoconstriction with a high degree of specificity. We examined the effects of i.c.v. PP56 on locomotor activity and food intake in rats, and on the hypoactivity and hyperphagia induced by NPY. In the open field, PP56 given alone increased locomotor activity by up to 85%. It did not prevent NPY induced hypoactivity to any extent. PP56 given alone did not affect food intake except for a small increase after the highest dose tested (200 nmol). When NPY was given after pretreatment with PP56, NPY induced hyperphagia was significantly reduced. A similar effect, however, was seen with regard to the hyperphagia produced by another orexigenic peptide, galanin. PP56 did not affect the binding of 125I-NPY to brain membranes in vitro, or to cells of two different neuroblastoma cell lines which selectively express NPY Y1 or Y2 receptors. In summary, PP56 acted as a locomotor stimulant per se. Only one of the two tested central effects of NPY could be antagonized by PP56, and then only partially and in a non-specific manner. The central effects of PP56 do not seem to be produced at the level of NPY receptors.

    Topics: Animals; Behavior, Animal; Binding, Competitive; Brain; Eating; Humans; Hyperphagia; Immobilization; Injections, Intraventricular; Inositol Phosphates; Locomotion; Male; Membranes; Neuroblastoma; Neuropeptide Y; Rats; Rats, Inbred Strains; Receptors, Neuropeptide Y; Receptors, Neurotransmitter; Tumor Cells, Cultured

1991
Hypothalamic neuropeptide Y (NPY) in obese Zucker rats: implications in feeding and sexual behaviors.
    Physiology & behavior, 1990, Volume: 47, Issue:3

    Neuropeptide Y (NPY), a peptide of the pancreatic polypeptide family, is actually considered to be the most potent stimulator of food intake in rats when centrally injected. It has also suppressive effects on several components of sexual behavior. It was measured in discrete microdissected brain nuclei in obese hyperphagic Zucker fa/fa rats also characterized by a deficient reproductive function, as well as in their lean homozygous (Fa/Fa) and heterozygous (Fa/fa) counterparts. When compared with the lean (Fa/Fa) rats, NPY concentrations were significantly increased in the obese rats in the arcuate nucleus-median eminence (ARCME, +300%), in the paraventricular (PVN, +60%), suprachiasmatic (SCH, +90%), accumbens (+100%) and supraoptic (+40%) nuclei, as well as in the median preoptic area (MPOA, +70%). As PVN is one of the most important nuclei involved in the control of food intake and one site of NPY action, the high levels found in this nucleus might be a major component at the origin of hyperphagia in the obese animals. Food intake might be overstimulated by a sustained production of NPY as shown by the high concentrations found in the ARCME. NPY might also intervene in the pattern of food intake, for NPY contents were also largely modified in the SCH, the nucleus regulating feeding periodicity and in the MPOA, which is possibly involved in the regulation of energy balance. Finally, as the MPOA is the only site of action of NPY on sexual behavior, the higher levels measured in this area might contribute to the defective reproductive function of the obese Zucker fa/fa rat.

    Topics: Animals; Brain; Feeding Behavior; Hyperphagia; Hypothalamus; Male; Neuropeptide Y; Obesity; Rats; Rats, Zucker; Sexual Behavior, Animal; Species Specificity

1990
Hyperphagia in obesity is associated with a central peptidergic dysregulation in rats.
    The Journal of nutrition, 1990, Volume: 120, Issue:7

    Hyperphagia and obesity are often associated, and the origins of the biochemical modifications leading to these syndromes might be in the hypothalamus. Indeed, food intake is regulated by numerous neuropeptides in various hypothalamic nuclei, including the paraventricular (PVN), arcuate (ARC), ventromedian (VMN) and suprachiasmatic (SCH) nuclei. Among these peptides, neuropeptide Y (NPY) is the most potent inducer of food intake whereas neurotensin (NT) decreases food intake. We measured these two peptides in microdissected hypothalamic nuclei in obese Zucker rats that ate 30% more food than their lean counterparts. Neuropeptide Y and neurotensin levels varied in opposite directions: In the hyperphagic obese Zucker rats, the NPY concentrations were significantly greater than those in the lean normophagic rats in the ARC (+30%), PVN (+60%) and SCH (+94%) nuclei, whereas the NT levels were significantly lower in the ARC (-40%), PVN (-31%) VMN (-66%) and SCH (-47%) nuclei. Both these variations tend to increase food intake. Feeding periodicity might also be modified because large variations of the two peptides have been measured in the supra-chiasmatic nucleus, which is considered the most important regulator of feeding rhythm. The results reinforce the hypothesis that hyperphagia in obesity is associated with a biochemical modification in the central nervous system because the peripheral status of NT and NPY was not modified in the obese rats. Because levels of other hypothalamic peptides, such as opioid peptides and somatostatin, are also slightly modified, it can be concluded that hyperphagia in obesity is associated with a central peptidergic dysregulation. Research on drugs reacting specifically with the receptor of these peptides might have interesting implications for the treatment of hyperphagia and, therefore, of obesity.

    Topics: Animals; Arcuate Nucleus of Hypothalamus; Disease Models, Animal; Hyperphagia; Hypothalamus; Neuropeptide Y; Neurotensin; Obesity; Paraventricular Hypothalamic Nucleus; Rats; Rats, Zucker; Ventromedial Hypothalamic Nucleus

1990
Peptide YY (PYY), a potent orexigenic agent.
    Brain research, 1985, Aug-19, Volume: 341, Issue:1

    Peptide YY (PYY) enhances feeding and drinking more potently than does neuropeptide Y after central administration. Chronic administration of PYY every 6 h for 48 h causes massive food ingestion. Tolerance to this effect of PYY does not appear to develop. This data suggests that PYY is one of the most potent orexigenic substances yet to be identified. PYY may play a role in the pathogenesis of bulimic syndromes.

    Topics: Animals; Appetite; Drinking; Eating; Hyperphagia; Male; Nerve Tissue Proteins; Neuropeptide Y; Peptide YY; Peptides; Rats; Rats, Inbred Strains

1985