Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 140
Filter
1.
Diabetes Obes Metab ; 16 Suppl 1: 33-40, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25200294

ABSTRACT

Dysregulation of hepatic glucose uptake (HGU) and inability of insulin to suppress hepatic glucose production (HGP) contribute to hyperglycaemia in patients with type 2 diabetes (T2D). Growing evidence suggests that insulin can inhibit HGP not only through a direct effect on the liver but also through a mechanism involving the brain. Yet, the notion that insulin action in the brain plays a physiological role in the control of HGP continues to be controversial. Although studies in dogs suggest that the direct hepatic effect of insulin is sufficient to explain day-to-day control of HGP, a surprising outcome has been revealed by recent studies in mice, investigating whether the direct hepatic action of insulin is necessary for normal HGP: when the hepatic insulin signalling pathway was genetically disrupted, HGP was maintained normally even in the absence of direct input from insulin. Here, we present evidence that points to a potentially important role of the brain in the physiological control of both HGU and HGP in response to input from insulin as well as other hormones and nutrients.


Subject(s)
Brain/metabolism , Feedback, Physiological , Gluconeogenesis , Glycolysis , Islets of Langerhans/metabolism , Liver/metabolism , Models, Biological , Animals , Glucose/metabolism , Humans , Insulin/metabolism , Insulin Receptor Substrate Proteins/agonists , Insulin Receptor Substrate Proteins/metabolism , Insulin Secretion , Islets of Langerhans/innervation , Liver/innervation , Neurons/metabolism , Signal Transduction
2.
Int J Obes (Lond) ; 35(12): 1455-65, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21386802

ABSTRACT

Energy homeostasis involves a complex network of hypothalamic and extra-hypothalamic neurons that transduce hormonal, nutrient and neuronal signals into responses that ultimately match caloric intake to energy expenditure and thereby promote stability of body fat stores. Growing evidence suggests that rather than reflecting a failure to regulate caloric intake, common forms of obesity involve fundamental changes to this homeostatic system that favor the defense of an elevated level of body adiposity. This article reviews emerging evidence that during high-fat feeding, obesity pathogenesis involves fundamental alteration of hypothalamic systems that regulate food intake and energy expenditure.


Subject(s)
Energy Intake , Energy Metabolism , Hypothalamus/physiopathology , Leptin/metabolism , Obesity/physiopathology , Animals , Homeostasis , Hypothalamus/metabolism , Mice , Obesity/metabolism , Signal Transduction
3.
Int J Obes (Lond) ; 33(6): 653-61, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19365394

ABSTRACT

OBJECTIVE: To develop a non-invasive method of studying brain mechanisms involved in energy homeostasis and appetite regulation in humans by using visual food cues that are relevant to individuals attempting weight loss. DESIGN: Functional magnetic resonance imaging (fMRI) was used to compare brain activation in regions of interest between groups of food photographs. PARTICIPANTS: Ten healthy, non-obese women who were not dieting for weight loss. MEASUREMENTS: Independent raters viewed food photographs and evaluated whether the foods depicted should be eaten by individuals attempting a calorically-restricted diet. Based on their responses, we categorized photographs into 'non-fattening' and 'fattening' food groups, the latter characterized by high-caloric content and usually also high-fat or high-sugar content. Blood oxygen level-dependent (BOLD) response was measured by fMRI while participants viewed photographs of 'fattening' food, 'non-fattening' food, and non-food objects. RESULTS: Viewing photographs of fattening food compared with non-food objects resulted in significantly greater activation in the brainstem; hypothalamus; left amygdala; left dorsolateral prefrontal cortex; left orbitofrontal cortex; right insular cortex; bilateral striatum, including the nucleus accumbens, caudate nucleus, and putamen; bilateral thalamus; and occipital lobe. By comparison, only the occipital region had greater activation by non-fattening food than by object photographs. Combining responses to all food types resulted in attenuation of activation in the brainstem, hypothalamus, and striatum. CONCLUSION: These findings suggest that, in non-obese women, neural circuits engaged in energy homeostasis and reward processing are selectively attuned to representations of high-calorie foods that are perceived as fattening. Studies to investigate hormonal action or manipulation of energy balance may benefit from fMRI protocols that contrast energy-rich food stimuli with non-food or low-calorie food stimuli.


Subject(s)
Appetite Regulation/physiology , Cerebral Cortex/physiology , Choice Behavior/physiology , Food Preferences/psychology , Hypothalamus/physiology , Adult , Cues , Female , Food , Humans , Magnetic Resonance Imaging , Middle Aged , Photic Stimulation/methods , Photography , Reward , Young Adult
4.
Diabetologia ; 52(5): 921-31, 2009 May.
Article in English | MEDLINE | ID: mdl-19219423

ABSTRACT

AIMS/HYPOTHESIS: Exposure of endothelial cells to high glucose levels suppresses responses to insulin, including induction of endothelial nitric oxide synthase activity, through pro-inflammatory signalling via the inhibitor of nuclear factor kappaB (IkappaB)alpha-nuclear factor kappaB (NF-kappaB) pathway. In the current study, we aimed to identify metabolic responses to glucose excess that mediate endothelial cell inflammation and insulin resistance. Since endothelial cells decrease their oxygen consumption rate (OCR) in response to glucose, we hypothesised that increased mitochondrial function would not mediate these cells' response to excess substrate. METHODS: The effects of glycolytic and mitochondrial fuels on metabolic intermediates and end-products of glycolytic and oxidative metabolism, including glucose 6-phosphate (G6P), lactate, CO(2), NAD(P)H and OCR, were measured in cultured human microvascular endothelial cells and correlated with IkappaBalpha phosphorylation. RESULTS: In response to increases in glucose concentration from low to physiological levels (0-5 mmol/l), production of G6P, lactate, NAD(P)H and CO(2) each increased as expected, while OCR was sharply reduced. IkappaBalpha activation was detected at glucose concentrations >5 mmol/l, which was associated with parallel increases of G6P levels, whereas downstream metabolic pathways were insensitive to excess substrate. CONCLUSIONS/INTERPRETATION: Phosphorylation of IkappaBalpha by excess glucose correlates with increased levels of the glycolytic intermediate G6P, but not with lactate generation or OCR, which are inhibited well below saturation levels at physiological glucose concentrations. These findings suggest that oxidative stress due to increased mitochondrial respiration is unlikely to mediate endothelial inflammation induced by excess glucose and suggests instead the involvement of G6P accumulation in the adverse effects of hyperglycaemia on endothelial cells.


Subject(s)
Endothelium, Vascular/physiopathology , Glucose-6-Phosphate/metabolism , Glucose/pharmacology , Inflammation/physiopathology , Mitochondria/metabolism , Cells, Cultured , Endothelium, Vascular/drug effects , Glucose/metabolism , Humans , Hyperglycemia/metabolism , Inflammation/chemically induced , Interleukin-6/metabolism , Kinetics , Microcirculation/physiology , Oxygen Consumption
5.
Exp Clin Endocrinol Diabetes ; 116(8): 475-80, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18465680

ABSTRACT

AIM: The BB rat model of type 1 diabetes exhibits altered body weight gain and body temperature regulation prior to hyperglycemia onset, implying the existence of as yet unidentified biomarkers of autoimmune processes that destroy pancreatic beta cells. To investigate this hypothesis, we compared the metabolic profile of diabetes-resistant DR.lyp/+ rats and their diabetes-prone, congenic DR.lyp/lyp littermates in the days leading up to diabetes onset. METHODS: Except for the Gimap5 mutation on chromosome 4, congenic DR.lyp/lyp rats are genetically identical to DR.lyp/+ littermates. They invariably develop hyperglycemia at 46-81 days of age, whereas DR.lyp/+ rats do not develop diabetes. In addition to daily food intake and body weight, indirect calorimetry was performed continuously on male DR.lyp/lyp and DR.lyp/+ rats (n=6/group) for 6-18 days to measure locomotor activity, VO (2), VCO (2) and RQ. RESULTS: DR.lyp/lyp rats exhibited a progressive decrease of RQ compared to DR.lyp/+ rats 0.005+/-0.001 units/day (p<0.005). Limiting the analysis to the six days prior to diabetes onset revealed a larger decrease of 0.007+/-0.002 units/day (p<0.001) in DR.lyp/lyp animals, whereas RQ of the DR.lyp/+ rats remained unchanged. This metabolic change occurred prior to hyperglycemia onset and was not associated with changes of any other parameter. CONCLUSIONS: Diabetes onset in DR.lyp/lyp rats is heralded by a progressive shift towards lipid oxidation relative to carbohydrate metabolism.


Subject(s)
Diabetes Mellitus, Type 1/metabolism , Lipid Peroxidation , Animals , Calorimetry , Drinking Behavior , Feeding Behavior , Genotype , Housing, Animal , Islets of Langerhans/physiopathology , Male , Motor Activity , Rats , Rats, Inbred BB , Rats, Inbred Strains , Water
6.
Nature ; 443(7109): 289-95, 2006 Sep 21.
Article in English | MEDLINE | ID: mdl-16988703

ABSTRACT

The capacity to adjust food intake in response to changing energy requirements is essential for survival. Recent progress has provided an insight into the molecular, cellular and behavioural mechanisms that link changes of body fat stores to adaptive adjustments of feeding behaviour. The physiological importance of this homeostatic control system is highlighted by the severe obesity that results from dysfunction of any of several of its key components. This new information provides a biological context within which to consider the global obesity epidemic and identifies numerous potential avenues for therapeutic intervention and future research.


Subject(s)
Body Weight/physiology , Central Nervous System/physiology , Feeding Behavior/physiology , Adipose Tissue/metabolism , Neuronal Plasticity/physiology , Satiety Response/physiology
7.
Neurology ; 60(12): 1899-903, 2003 Jun 24.
Article in English | MEDLINE | ID: mdl-12821730

ABSTRACT

BACKGROUND: Abnormal insulin metabolism may contribute to the clinical symptoms and pathophysiology of AD. In vitro studies show that insulin enhances the release of beta-amyloid protein (Abeta) or inhibits its degradation, either of which might increase amyloid burden. METHODS: On separate mornings, 16 healthy older adults (10 women, 6 men; mean age 68.7 years, SD 8.6 years) each underwent two infusions consisting of either saline (placebo) or insulin (1.0 mU x kg(-1) x min(-1)) plus dextrose to maintain euglycemia. After 120 minutes of infusion, blood, CSF, and cognitive measures were acquired. RESULTS: As expected, insulin infusion produced an increase in CSF insulin concentration. Insulin infusion also led to an increase in CSF Abeta42 levels, most notably in older subjects. As has been observed previously, insulin infusion facilitated declarative memory, but such facilitation was attenuated in the subjects with the greatest increase in CSF Abeta42 levels. CONCLUSIONS: These findings are consistent with recent in vitro studies of insulin effects on Abeta and support the notion that insulin may modulate Abeta42 levels acutely in humans.


Subject(s)
Amyloid beta-Peptides/cerebrospinal fluid , Insulin/pharmacology , Peptide Fragments/cerebrospinal fluid , Aged , Aged, 80 and over , Alzheimer Disease/etiology , Alzheimer Disease/metabolism , Attention/drug effects , Cognition/drug effects , Female , Humans , Male , Memory/drug effects , Middle Aged , Reference Values
9.
Exp Biol Med (Maywood) ; 226(11): 978-81, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11743132

ABSTRACT

Evidence has existed for more than 50 years in support of the hypothesis that body energy stored in the form of fat is homeostatically regulated. Implicit in this concept is the existence of a biological system that operates dynamically over time to match cumulative energy intake to energy expenditure. For example, to compensate for weight loss induced by energy restriction, animals must enter a period of positive energy balance (i.e., energy intake greater than energy expenditure) that is sustained for as long as it takes to correct the deficit in body fat stores. Having reached this point, the animal must return to a state of neutral energy balance if stable fat mass is to be maintained. The identification of neuronal circuits in the hypothalamus that, when activated, exert potent, unidirectional effects on energy balance provides a cornerstone of support for this model. The additional finding that these central effector pathways are regulated by humoral signals generated in proportion to body fat stores, including the hormones insulin and leptin, helps to round out the picture of how energy homeostasis is achieved. The goal of this overview is to highlight the evidence that specific subsets of hypothalamic neurons containing specific signaling molecules participate in this dynamic regulatory process, and to put these observations in the larger context of a biological system that controls body adiposity.


Subject(s)
Body Weight , Brain/metabolism , Brain/physiology , Eating , Energy Intake , Energy Metabolism , Feeding Behavior , Humans , Insulin/metabolism , Leptin/metabolism , Neurons , Obesity/physiopathology , Obesity/therapy , Weight Loss
13.
Endocrinology ; 142(8): 3292-301, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11459770

ABSTRACT

Anorexia is a debilitating manifestation of many malignancies. The etiology of cancer anorexia is poorly understood, and effective treatment options are limited. To investigate the role of central melanocortin receptor signaling in the pathogenesis of cancer anorexia, we assessed the effects on food intake of the melanocortin receptor antagonist SHU9119 administered into the third cerebral ventricle of Lobund-Wistar rats that were anorexic from prostate cancer. In anorexic tumor-bearing rats, daily treatment with SHU9119 (0.35 nmol, intracerebroventricularly) increased food intake from 71 +/- 3% to 110 +/- 6% of preanorectic baseline and caused significant weight gain (13 +/- 5 vs. 5 +/- 1 g/3 d, SHU9119 vs. baseline in tumor-bearing rats). In control rats pair-fed to the intake of tumor-bearing animals, SHU9119 was ineffective at increasing food intake. The specificity of the SHU9119 feeding response was assessed using two other orexigenic peptides, NPY and the novel hormone ghrelin. Treatment of tumor-bearing rats with intracerebroventricular ghrelin (10 microg) increased food intake, but the effect was blunted relative to that in controls. Intracerebroventricular injections of NPY (1 microg) also failed to reverse anorexia in tumor-bearing rats. Because SHU9119 completely reverses cancer anorexia in this model, whereas ghrelin and NPY do not, increased central nervous system melanocortin signaling is implicated in the pathogenesis of this disorder. This suggests that new targets for the treatment of cancer anorexia may be found in the melanocortin pathways.


Subject(s)
Adenocarcinoma/complications , Anorexia/drug therapy , Anorexia/etiology , Brain/metabolism , Melanocyte-Stimulating Hormones/therapeutic use , Peptide Hormones , Prostatic Neoplasms/complications , Receptors, Corticotropin/antagonists & inhibitors , Animals , Anorexia/pathology , Anorexia/physiopathology , Body Weight/drug effects , Eating/drug effects , Ghrelin , Injections, Intraventricular , Male , Neuropeptide Y/therapeutic use , Peptides/therapeutic use , Rats , Rats, Wistar , Receptors, Melanocortin , Reference Values , Third Ventricle
14.
15.
J Neurosci ; 21(10): 3639-45, 2001 May 15.
Article in English | MEDLINE | ID: mdl-11331393

ABSTRACT

The CNS melanocortin (MC) system is implicated as a mediator of the central effects of leptin, and reduced activity of the CNS MC system promotes obesity in both rodents and humans. Because activation of CNS MC receptors has direct effects on autonomic outflow and metabolism, we hypothesized that food intake-independent mechanisms contribute to development of obesity induced by pharmacological blockade of MC receptors in the brain and that changes in hypothalamic neuropeptidergic systems known to regulate weight gain [i.e., corticotropin-releasing hormone (CRH), cocaine-amphetamine-related transcript (CART), proopiomelanocortin (POMC), and neuropeptide Y (NPY)] would trigger this effect. Relative to vehicle-treated controls, third intracerebroventricular (i3vt) administration of the MC receptor antagonist SHU9119 to rats for 11 d doubled food and water intake (toward the end of treatment) and increased body weight ( approximately 14%) and fat content ( approximately 90%), hepatic glycogen content ( approximately 40%), and plasma levels of cholesterol ( approximately 48%), insulin ( approximately 259%), glucagon ( approximately 80%), and leptin ( approximately 490%), whereas spontaneous locomotor activity and body temperature were reduced. Pair-feeding of i3vt SHU9119-treated animals to i3vt vehicle-treated controls normalized plasma levels of insulin, glucagon, and hepatic glycogen content, but only partially reversed the elevations of plasma cholesterol ( approximately 31%) and leptin ( approximately 104%) and body fat content ( approximately 27%). Reductions in body temperature and locomotor activity induced by i3vt SHU9119 were not reversed by pair feeding, but rather were more pronounced. None of the effects found can be explained by peripheral action of the compound. The obesity effects occurred despite a lack in neuropeptide expression responses in the neuroanatomical range selected across the arcuate (i.e., CART, POMC, and NPY) and paraventricular (i.e., CRH) hypothalamus. The results indicate that reduced activity of the CNS MC pathway promotes fat deposition via both food intake-dependent and -independent mechanisms.


Subject(s)
Behavior, Animal/physiology , Hypothalamus/metabolism , Obesity/metabolism , Receptors, Corticotropin/metabolism , Signal Transduction/physiology , Animals , Behavior, Animal/drug effects , Body Composition/drug effects , Body Temperature/drug effects , Cholesterol/blood , Drinking/drug effects , Eating/drug effects , Glucagon/blood , Hypothalamus/drug effects , Injections, Intraventricular , Insulin/blood , Leptin/blood , Male , Melanocyte-Stimulating Hormones/administration & dosage , Motor Activity/drug effects , Neurotransmitter Agents/genetics , Neurotransmitter Agents/metabolism , Pro-Opiomelanocortin/genetics , Pro-Opiomelanocortin/metabolism , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptors, Corticotropin/antagonists & inhibitors , Receptors, Melanocortin , Signal Transduction/drug effects
16.
Am J Phys Anthropol ; 114(1): 4-17, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11150048

ABSTRACT

What biological traits distinguish taxa susceptible to extinction from less susceptible taxa? Substantiated island biogeographic theory suggests that after insularization, small islands lose more species than do large islands. Thus, susceptible taxa are those now found on only large islands. The traits of susceptible taxa can thus be found by comparing the biology of species found only on large islands with those also found on small islands. The islands examined here are those of the Sunda Shelf, created as a result of the Holocene rise in sea levels of 120 m. We use four statistical comparisons: comparative analysis by (phylogenetically) independent contrasts (N = 8 contrasts at the subgeneric or deeper level), Spearman correlations, stepwise regression, and principle components analysis (N = 9 subgenera/genera). The genera and one subgenus considered are: Hylobates, Macaca, Nasalis, Nycticebus, Pongo, Presbytis, Symphalangus, Tarsius, and Trachypithecus. Traits of risk appear to be large body mass, low density, large annual home range, and low maximum latitude. Expected traits that did not correlate with susceptibility were low interbirth interval, high percent frugivory, high group mass, low altitudinal range, and small geographic range. The risky traits also apply to just the anthropoids (i.e., prosimians excluded). The risky traits are explained if susceptibility is induced by requirements for a large extent of habitat, a small population size, and specialization. These findings, which indicate that efficiency and plasticity of use of the environment separate susceptible from successful primate taxa, might be relevant to an understanding of hominoid evolution.


Subject(s)
Biological Evolution , Primates , Altitude , Animals , Anthropology, Physical , Biometry , Classification , Diet , Female , Geography , Male , Movement , Reproduction
17.
Pediatr Case Rev ; 1(1): 1-2, 2001 Oct.
Article in English | MEDLINE | ID: mdl-12865698
18.
Int J Obes Relat Metab Disord ; 25 Suppl 5: S56-62, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11840217

ABSTRACT

Kennedy hypothesized nearly 50 y ago that negative feedback regulation of body fat stores involves hormones that circulate in proportion to adiposity and enter the brain, where they exert inhibitory effects on food intake and energy balance. Recent studies implicate leptin and insulin as 'adiposity signals' to the brain that promote negative energy balance in two ways: by inhibiting 'anabolic' hypothalamic neuronal circuits that stimulate food intake and promote weight gain, and by activating 'catabolic' pathways that reduce food intake and body weight. Chief among candidate 'anabolic' effector pathways is the NPY/AgRP neuron, found only in the hypothalamic arcuate nucleus. These neurons make peptides that potently stimulate food intake not only by increasing neuropeptide Y (NPY) signaling, but by reducing melanocortin signaling via the release of agouti-related peptide (AgRP), an endogenous melanocortin 3/4 receptor antagonist. Since NPY/AgRP neurons express receptors for leptin and insulin and are inhibited by these hormones, they are activated by a decrease of leptin or insulin signaling. Fasting, uncontrolled diabetes, and genetic leptin deficiency are examples of conditions in which food intake increases via a mechanism hypothesized to involve NPY/AgRP neurons. Data are reviewed which illustrate the role of these neurons in adaptive and maladaptive states characterized by hyperphagia and weight gain.


Subject(s)
Energy Metabolism , Homeostasis , Neurons/physiology , Neuropeptide Y/physiology , Proteins/physiology , Adipose Tissue/physiology , Agouti-Related Protein , Animals , Feedback , Humans , Intercellular Signaling Peptides and Proteins
19.
J Pediatr Endocrinol Metab ; 14 Suppl 6: 1417-29, 2001.
Article in English | MEDLINE | ID: mdl-11837495

ABSTRACT

The brain plays a key role in the regulation of energy homeostasis, balancing food intake and energy expenditure to maintain adipose tissue mass. A widely accepted model proposes that energy homeostasis is modulated by hormones that circulate in the blood in proportion to adipose tissue mass. A major candidate 'adiposity signal' to the brain is the adipocyte hormone, leptin; this inhibits neuropeptide circuits that promote anabolic metabolism, and stimulates those that promote catabolic metabolism. It is hypothesized that leptin-responsive circuits in the hypothalamus project to caudal brainstem neuronal groups that integrate satiety signals converging on the brain from the stomach and intestine following ingestion of food. Leptin signaling to the brainstem via hypothalamic pathways potentially increases the brain's motor and autonomic responses to satiety signals, leading to smaller individual meals, reduced cumulative food intake, and a lower body weight. This mechanism explains how leptin deficiency or defects in the brain's processing of leptin signaling can result in a sustained increase in food intake and obesity.


Subject(s)
Body Weight/physiology , Brain/physiology , Eating/physiology , Leptin/physiology , Adipose Tissue/physiology , Animals , Anorexia/etiology , Humans , Signal Transduction/physiology
20.
Endocrinology ; 141(12): 4442-8, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11108253

ABSTRACT

Leptin administration potentiates the satiety response to signals such as cholecystokinin (CCK), that are released from the gut during a meal. To investigate the physiological relevance of this observation, we hypothesized that leptin deficiency, induced by fasting, attenuates the satiety response to CCK. To test this hypothesis, 48-h-fasted or fed rats were injected with i.p. saline or CCK. Fasting blunted the satiety response to 3.0 microg/kg CCK, such that 30-min food intake was suppressed by 65.1% (relative to saline-treated controls) in fasted rats vs. 85.9% in the fed state (P < 0.05). In a subsequent experiment, rats were divided into three groups: 1) vehicle/fed; 2) vehicle/fasted; and 3) leptin-replaced/fasted; and each group received 3.0 microg/kg i.p. CCK. As expected, the satiety response to CCK was attenuated by fasting in vehicle-treated rats (30-min food intake: vehicle/fed, 0.3 +/- 0.1 g; vehicle/fasted, 1.7 +/- 0.4 g; P < 0.01), and this effect was prevented by leptin replacement (0.7 +/- 0.2 g, P < 0.05 vs. vehicle/fasted; P = not significant vs. vehicle/fed). To investigate whether elevated neuropeptide Y (NPY) signaling plays a role in the effect of leptin deficiency to impair the response to CCK, we measured the response to 3.0 microg/kg i.p. CCK after treatment with 7.5 microg intracerebroventricular NPY. We found that both CCK-induced satiety and its ability to increase c-Fos-like-immunoreactivity in key brainstem-feeding centers were attenuated by NPY pretreatment. We conclude that an attenuated response to meal-related satiety signals is triggered by leptin deficiency and may contribute to increased food intake.


Subject(s)
Cholecystokinin/pharmacology , Fasting/physiology , Leptin/deficiency , Satiation/drug effects , Animals , Brain Chemistry , Eating/drug effects , Injections, Intraventricular , Male , Neuropeptide Y/administration & dosage , Neuropeptide Y/pharmacology , Proto-Oncogene Proteins c-fos/analysis , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...