Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 166
Filtrar
1.
Neuropsychopharmacology ; 48(13): 1931-1940, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37474763

RESUMO

It is well-established that stress and negative affect trigger eating disorder symptoms and that the brains of men and women respond to stress in different ways. Indeed, women suffer disproportionately from emotional or stress-related eating, as well as associated eating disorders such as binge eating disorder. Nevertheless, our understanding of the precise neural circuits driving this maladaptive eating behavior, particularly in women, remains limited. We recently established a clinically relevant model of 'emotional' stress-induced binge eating whereby only female mice display binge eating in response to an acute "emotional" stressor. Here, we combined neuroanatomic, transgenic, immunohistochemical and pathway-specific chemogenetic approaches to investigate whole brain functional architecture associated with stress-induced binge eating in females, focusing on the role of Vglut2 projections from the paraventricular thalamus (PVTVglut2+) to the medial insular cortex in this behavior. Whole brain activation mapping and hierarchical clustering of Euclidean distances revealed distinct patterns of coactivation unique to stress-induced binge eating. At a pathway-specific level, PVTVglut2+ cells projecting to the medial insular cortex were specifically activated in response to stress-induced binge eating. Subsequent chemogenetic inhibition of this pathway suppressed stress-induced binge eating. We have identified a distinct PVTVglut2+ to insular cortex projection as a key driver of "emotional" stress-induced binge eating in female mice, highlighting a novel circuit underpinning this sex-specific behavior.


Assuntos
Transtorno da Compulsão Alimentar , Bulimia , Humanos , Masculino , Feminino , Camundongos , Animais , Córtex Insular , Bulimia/metabolismo , Encéfalo/metabolismo , Tálamo/metabolismo
2.
Am J Physiol Endocrinol Metab ; 322(6): E494-E507, 2022 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-35403437

RESUMO

Binge-eating disorder is the most prevalent eating disorder diagnosed, affecting three times more women than men. Ghrelin stimulates appetite and reward signaling, and loss of its receptor reduces binge-eating behavior in male mice. Here, we examined the influence of ghrelin itself on binge-eating behavior in both male and female mice. Five-wk-old wild-type (WT) and ghrelin-deficient (Ghrl-/-) mice were housed individually in indirect calorimetry cages for 9 wks. Binge-like eating was induced by giving mice ad libitum chow, but time-restricted access to a Western-style diet (WD; 2 h access, 3 days/wk) in the light phase (BE); control groups received ad libitum chow (CO), or ad libitum access to both diets (CW). All groups of BE mice showed binge-eating behavior, eating up to 60% of their 24-h intake during the WD access period. Subsequent dark phase chow intake was decreased in Ghrl-/- mice and remained decreased in Ghrl-/- females on nonbinge days. Also, nonbinge day locomotor activity was lower in Ghrl-/- than in WT BE females. Upon euthanasia, Ghrl-/- BE mice weighed less and had a lower lean body mass percentage than WT BE mice. In BE and CW groups, ghrelin and sex altered the expression of genes involved in lipid processing, thermogenesis, and aging in white adipose tissue and livers. We conclude that, although ghrelin deficiency does not hamper the development of binge-like eating, it sex-dependently alters food intake timing, locomotor activity, and metabolism. These results add to the growing body of evidence that ghrelin signaling is sexually dimorphic.NEW & NOTEWORTHY Ghrelin, a peptide hormone secreted from the gut, is involved in hunger and reward signaling, which are altered in binge-eating disorder. Although sex differences have been described in both binge-eating and ghrelin signaling, this interaction has not been fully elucidated. Here, we show that ghrelin deficiency affects the behavior and metabolism of mice in a binge-like eating paradigm, and that the sex of the mice impacts the magnitude and direction of these effects.


Assuntos
Transtorno da Compulsão Alimentar , Bulimia , Animais , Bulimia/genética , Bulimia/metabolismo , Modelos Animais de Doenças , Ingestão de Alimentos/genética , Comportamento Alimentar , Feminino , Expressão Gênica , Grelina/metabolismo , Fígado/metabolismo , Locomoção , Masculino , Camundongos
3.
Neuroreport ; 31(18): 1283-1288, 2020 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-33165198

RESUMO

Neuropeptides and peptide hormones affect food-directed motivation, in part, through actions on brain regions associated with reward processing. For instance, previous reports have shown that stimulating glucagon-like peptide-1 (GLP-1) receptors in the nucleus accumbens (NAc), an area that directs motivational processes towards food and drugs of abuse, has an anorectic effect. In contrast, µ-opioid receptor activation of the NAc increases feeding, particularly on highly palatable diets. While both neurotransmitters act within the NAc to impact food intake, it is not clear if and how they might interact to affect feeding. Therefore, these experiments tested the effects of NAc injections of the GLP-1 receptor agonist Exendin 4 (EX4) or antagonist Exendin 9 (EX9) on the consumption of a sweetened fat diet, with and without simultaneous µ-opioid receptor stimulation. Male Sprague-Dawley rats (n = 8/group, EX4 or EX9) underwent surgery to place bilateral cannula above the NAc core. After recovery, animals were tested following NAc injections of saline or the µ-opioid agonist [D-Ala, N-MePhe, Gly-ol]-enkephalin (DAMGO) (0.025 µg/side), combined with varying doses of EX4 (0, 0.05, or 0.10 µg/side) or EX9 (0, 2.5, 5.0 µg/side), counterbalanced across 6 testing days. Food and water intake, along with locomotor activity, was monitored for 2 h. Mu-opioid receptor stimulation significantly increased feeding, and this effect was reduced by GLP-1 receptor stimulation. In contrast, GLP-1 antagonism with EX9 altered the dynamics of DAMGO-induced binge-like feeding, extending µ-opioid-induced binging, and increasing food consumption. These findings are the first to demonstrate an interaction between NAc µ-opioid and GLP-1 receptors on palatable food intake.


Assuntos
Bulimia/fisiopatologia , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Comportamento Alimentar/fisiologia , Receptor do Peptídeo Semelhante ao Glucagon 1/fisiologia , Neurotransmissores/farmacologia , Núcleo Accumbens/efeitos dos fármacos , Receptores Opioides mu/agonistas , Animais , Bulimia/metabolismo , Exenatida/farmacologia , Comportamento Alimentar/efeitos dos fármacos , Receptor do Peptídeo Semelhante ao Glucagon 1/agonistas , Receptor do Peptídeo Semelhante ao Glucagon 1/antagonistas & inibidores , Masculino , Núcleo Accumbens/metabolismo , Fragmentos de Peptídeos/farmacologia , Ratos , Receptores Opioides mu/metabolismo
4.
Nutrients ; 12(6)2020 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-32486207

RESUMO

BACKGROUND: Binge eating disorder (BED) is characterized by recurrent binge eating episodes consisting of rapid consumption of excessive amounts of highly palatable, energy-dense food within discrete periods of time. The aim of this study was to test the consummatory, food microstructural, and metabolic effects of a one hour limited access to either a high-sucrose diet (HSD) or a high-fat diet (HFD) in an operant rat model of binge-like eating. METHODS: Female rats were subject to a binge-like eating procedure in which a HSD, a HFD, or a standard chow diet were provided in a fixed ratio 1 (FR1) operant schedule of reinforcement. RESULTS: Limiting access to either a HSD or a HFD promoted binge-like eating as compared to the control chow diet. However, binge-like eating of HSD, but not HFD, was based on a true increase in the amount of food consumed, an increased eating rate, and a decrease in the intake of the home-cage standard chow, altogether suggesting an increase in palatability. Moreover, while HSD rats consumed overall less energy than HFD rats, the former were more energy efficient and gained more body weight than the latter. CONCLUSIONS: These results provide information on how the quality of food can deeply influence the behavioral and metabolic outcomes of binge-like eating.


Assuntos
Bulimia/metabolismo , Dieta Hiperlipídica/efeitos adversos , Sacarose Alimentar/efeitos adversos , Sacarose Alimentar/metabolismo , Comportamento Alimentar/efeitos dos fármacos , Animais , Transtorno da Compulsão Alimentar , Peso Corporal , Modelos Animais de Doenças , Ingestão de Alimentos , Feminino , Ratos
5.
J Neurosci ; 40(28): 5362-5375, 2020 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-32532885

RESUMO

Binge-eating disorder is the most common eating disorder. Various neuropeptides play important roles in the regulation of feeding behavior, including relaxin-3 (RLN3), which stimulates food intake in rats through the activation of the relaxin-family peptide-3 receptor (RXFP3). Here we demonstrate that a likely mechanism underlying the orexigenic action of RLN3 is RXFP3-mediated inhibition of oxytocin- and arginine-vasopressin-synthesizing paraventricular nucleus (PVN) magnocellular neurosecretory cells. Moreover, we reveal that, in male and female rats, this action depends on M-like potassium conductance. Notably, higher intra- and peri-PVN RLN3 fiber densities were observed in females, which may constitute an anatomic substrate for observed sex differences in binge-eating disorder. Finally, in a model of binge-eating in female rats, RXFP3 blockade within the PVN prevented binge-eating behavior. These data demonstrate a direct RLN3/RXFP3 action in the PVN of male and female rats, identify the associated ionic mechanisms, and reveal that hypothalamic RLN3/RXFP3 signaling regulates binge-eating behavior.SIGNIFICANCE STATEMENT Binge-eating disorder is the most common eating disorder worldwide, affecting women twice as frequently as men. Various neuropeptides play important roles in the regulation of feeding behavior, including relaxin-3, which acts via the relaxin-family peptide-3 receptor (RXFP3). Using a model of binge-eating, we demonstrated that relaxin-3/RXFP3 signaling in the hypothalamic paraventricular nucleus (PVN) is necessary for the expression of binge-eating behavior in female rats. Moreover, we elucidated the neuronal mechanism of RLN3/RXFP3 signaling in PVN in male and female rats and characterized sex differences in the RLN3 innervation of the PVN. These findings increase our understanding of the brain circuits and neurotransmitters involved in binge-eating disorder pathology and identify RXFP3 as a therapeutic target for binge-like eating disorders.


Assuntos
Bulimia/metabolismo , Comportamento Alimentar/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Neurônios/fisiologia , Núcleo Hipotalâmico Paraventricular/metabolismo , Canais de Potássio/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Peptídeos/metabolismo , Relaxina/metabolismo , Transdução de Sinais/fisiologia , Animais , Comportamento Animal/fisiologia , Feminino , Masculino , Ratos , Caracteres Sexuais
6.
Alcohol ; 86: 45-56, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32224218

RESUMO

Alcoholism and high fat diet (HFD)-induced obesity individually promote insulin resistance and glucose intolerance in clinical populations, increasing risk for metabolic diseases. HFD can also stimulate alcohol intake in short-term clinical studies. Unfortunately, there is currently a disconnect between animal models and the clinical findings, as animal studies typically show that HFD decreases ethanol intake while ethanol intake mitigates HFD-induced effects on insulin and glucose dysfunction. However, most previous animal studies utilized forced or continuous HFD and/or ethanol. In three experiments we sought to determine whether HFD (HFD = 60% calories from fat) vs. control diet (chow = 16% fat) alters voluntary two-bottle choice ethanol intake in male C57Bl/6J mice given differing access schedules for 6-7 weeks, and we assessed the resultant impact on metabolic function via insulin and glucose tolerance tests. Experiment 1: Unlimited Access Ethanol + HFD (UAE + HFD; n = 15; 10% ethanol v/v, ad libitum diet and ethanol) or UAE + Chow (n = 15). Experiment 2: Limited Access Ethanol + HFD (LAE + HFD; n = 15; ethanol = 4 h/day; 3 days/week, ad libitum diet) or LAE + Chow (n = 15) with increasing ethanol concentrations (10%, 15%, 20%). Experiment 3: Intermittent HFD with limited access to ethanol (iHFD-E; HFD = single 24-h session/week; ethanol = 4 h/day; 4 days/week) (n = 10). UAE + HFD mice consumed significantly less ethanol and were insulin-resistant and hyperglycemic compared with UAE + Chow mice. LAE + HFD mice consumed ethanol similarly to LAE + Chow mice, but exhibited hyperglycemia, insulin resistance, and glucose intolerance. iHFD-E mice displayed binge eating-like behaviors and consumed significantly more ethanol than mice given ad libitum chow or HFD. iHFD-E mice did not have significantly altered body composition, but developed insulin insensitivity and glucose intolerance. These findings suggest that access schedules influence HFD effects on ethanol consumption and resultant metabolic dysfunction, ethanol intake does not improve HFD-induced metabolic dysfunction, and binge eating-like behaviors can transfer to binge drinking behaviors.


Assuntos
Consumo de Bebidas Alcoólicas/metabolismo , Dieta Hiperlipídica , Comportamento Alimentar/fisiologia , Glucose/metabolismo , Insulina/metabolismo , Animais , Bulimia/metabolismo , Ingestão de Alimentos , Ingestão de Energia , Etanol/metabolismo , Teste de Tolerância a Glucose , Masculino , Camundongos , Camundongos Endogâmicos C57BL
7.
Behav Neurosci ; 134(3): 267-281, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32150421

RESUMO

Binge eating behavior (BEB) is the most common condition among eating disorders. In animal models, binge eating behavior is defined as overconsumption in a brief time interval and it develops as a progressive increase in food intake along time. It is triggered by restricting food access to regular chow or to palatable food and is associated with dopamine release from the ventral tegmental area to the nucleus accumbens. The dopamine system, exhibits day-night patterns, suggesting regulation by the circadian system. This study explored in rats the differential contribution of restricted food access to chow and sucrose for developing BEB, it explored whether BEB exhibits a day-night pattern, and whether behavioral changes are associated with the number of tyrosine hydroxylase (TH) positive cells in the ventral tegmental area, with the expression of dopamine 1 receptors (D1) and glutamate receptor subunit 1 receptors (GLUR1) in the nucleus accumbens. Present data indicate that under conditions of restricted access binge eating is developed for chow or sucrose. Both types of binge eating were independent of each other and exhibited a day-night pattern with increased intensity during the active phase (night). Binge eating was preceded by anticipatory activation, except when restricted food access was given during the day. Increased optical density for D1 receptors was found after exposure to the combination of restricted food access and sucrose. No association was observed between binge eating and the number of positive cells to TH in the ventral tegmental area, nor for GLUR1 in the nucleus accumbens. Present results point out the importance of time schedules to eat, highlighting an increased vulnerability to develop binge eating during the active phase. (PsycInfo Database Record (c) 2020 APA, all rights reserved).


Assuntos
Bulimia/metabolismo , Ritmo Circadiano/fisiologia , Sacarose/metabolismo , Animais , Transtorno da Compulsão Alimentar/metabolismo , Transtorno da Compulsão Alimentar/fisiopatologia , Encéfalo/efeitos dos fármacos , Bulimia/fisiopatologia , Ingestão de Alimentos/fisiologia , Comportamento Alimentar/fisiologia , Alimentos , Preferências Alimentares/fisiologia , Masculino , Núcleo Accumbens/efeitos dos fármacos , Ratos , Ratos Wistar , Sacarose/farmacologia , Área Tegmentar Ventral/efeitos dos fármacos
8.
Behav Pharmacol ; 31(2&3): 249-255, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31503073

RESUMO

Binge eating in humans is driven by hedonic properties of food, suggesting that brain reward systems may contribute to this behaviour. We examined the role of mu opioid receptors (MOP) in binge eating by examining sweet solution intake in mice with genetic deletion of the MOP. Wildtype and MOP knockout mice had 4 hours access to food in the home cage combined with limited (4 hours) access to sucrose (17.1% w/v) or saccharin (0.09% w/v), or continuous (24 hours) access to sucrose. Only limited access groups exhibited binge intake, measured as increased solution consumption during the first hour. Knockout mice consumed less solution and food during the first hour as well as less food each day compared with wildtype mice. Limited access groups consumed more food and gained more weight than continuous access groups, and the effect was magnified in saccharin-consuming mice. Indeed, the increased food consumption in animals given limited access to saccharin was so excessive that caloric intake of this group was significantly higher than either of the sucrose groups (limited or continuous access). Within this group, females consumed more food per bodyweight than males, highlighting important sex differences in feeding behaviours under restricted access schedules.


Assuntos
Bulimia/fisiopatologia , Comportamento Alimentar/fisiologia , Receptores Opioides mu/metabolismo , Animais , Transtorno da Compulsão Alimentar , Peso Corporal , Bulimia/metabolismo , Modelos Animais de Doenças , Ingestão de Alimentos/psicologia , Ingestão de Energia/fisiologia , Feminino , Preferências Alimentares/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores Opioides mu/fisiologia , Recompensa , Sacarose/metabolismo
9.
Eat Behav ; 35: 101337, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31726420

RESUMO

OBJECTIVE: Eating disorder symptoms change in a predictable pattern over the menstrual cycle such that changes in symptoms are triggered by changes in the ovarian hormones estradiol (E2) and progesterone (P4). To date, work in this area has focused exclusively on young adult women. The objective of this pilot study was to examine the effect of E2 and P4 on eating disorder symptom change in midlife women during early perimenopause. METHOD: Participants included women aged 42-52 in early perimenopause (n=8). In-home self-assessments were completed for one menstrual cycle or 40-days, whichever occurred first. In-home self-assessments included collecting saliva samples each morning for E2 and P4 assays and completing online study questionnaires at the end of each day. Multilevel regression models examined the associations of E2 and P4 with daily symptoms of binge eating and body dissatisfaction. RESULTS: E2 was positively associated with binge eating when P4 was high, but not when P4 was low. E2 was inversely associated with body dissatisfaction when P4 was low, but positively associated with body dissatisfaction when P4 was high. However, the simple slopes for the effect of E2 at both high and low P4 were not significant for body dissatisfaction. CONCLUSIONS: Despite the pilot nature of this study, results are broadly consistent with the young adult literature indicating that P4 levels shape the impact of E2 on eating disorder symptoms. Larger studies with the inclusion of key moderators to account for individual heterogeneity are needed to confirm and extend these findings.


Assuntos
Bulimia/metabolismo , Estradiol/metabolismo , Menopausa/fisiologia , Ovário/metabolismo , Progesterona/metabolismo , Adulto , Feminino , Humanos , Pessoa de Meia-Idade , Projetos Piloto
10.
Obesity (Silver Spring) ; 27(11): 1874-1882, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31562706

RESUMO

OBJECTIVE: Amphetamine was formerly used as a treatment to combat obesity, but amphetamine's use as an appetite suppressant was discontinued because of its significant abuse potential. Most of the rewarding and reinforcing effects of amphetamine differ by sex, with females showing higher levels of drug intake and amphetamine-induced motivation, relapse, and locomotion, but it is unknown whether amphetamine's effects on feeding also differ by sex. Furthermore, previous research on the anorectic effects of amphetamine has been focused primarily on its effects on baseline homeostatic feeding, but it is unknown whether amphetamine also affects hedonic, reward-related feeding, which is an important factor driving the rise in obesity levels. METHODS: This study tested whether amphetamine alters food intake in a sex-dependent manner in two reward-related feeding paradigms: a sucrose two-bottle choice test and a high-fat/high-sugar binge intake model. RESULTS: Amphetamine altered food intake equally in males and females in both paradigms, with higher doses significantly inhibiting feeding and low doses of amphetamine increasing feeding at later time points. CONCLUSIONS: Amphetamine's effects on feeding and drug reward may be mediated by distinct mechanisms, which could allow for the development of new approaches to combat obesity with limited abuse and addiction-related side effects.


Assuntos
Anfetamina/farmacologia , Bulimia , Carboidratos da Dieta/administração & dosagem , Gorduras na Dieta/administração & dosagem , Ingestão de Energia/efeitos dos fármacos , Sacarose/administração & dosagem , Animais , Depressores do Apetite/farmacologia , Bulimia/induzido quimicamente , Bulimia/metabolismo , Bulimia/prevenção & controle , Relação Dose-Resposta a Droga , Ingestão de Alimentos/efeitos dos fármacos , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fatores Sexuais
11.
Nutrients ; 11(2)2019 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-30717427

RESUMO

Binge-eating disorder (BED) is the most common eating disorder, characterized by rapid, recurrent overconsumption of highly palatable food in a short time frame. BED shares an overlapping behavioral phenotype with obesity, which is also linked to the overconsumption of highly palatable foods. The reinforcing properties of highly palatable foods are mediated by the nucleus accumbens (NAc) and the ventral tegmental area (VTA), which have been implicated in the overconsumption behavior observed in BED and obesity. A potential regulator of binge-type eating behavior is the G protein-coupled receptor neuromedin U receptor 2 (NMUR2). Previous research demonstrated that NMUR2 knockdown potentiates binge-type consumption of high-fat food. We correlated binge-type consumption across a spectrum of fat and carbohydrate mixtures with synaptosomal NMUR2 protein expression in the NAc and VTA of rats. Synaptosomal NMUR2 protein in the NAc demonstrated a strong positive correlation with binge intake of a "lower"-fat (higher carbohydrate) mixture, whereas synaptosomal NMUR2 protein in the VTA demonstrated a strong negative correlation with binge intake of an "extreme" high-fat (0% carbohydrate) mixture. Taken together, these data suggest that NMUR2 may differentially regulate binge-type eating within the NAc and the VTA.


Assuntos
Transtorno da Compulsão Alimentar/metabolismo , Bulimia/metabolismo , Comportamento Alimentar/fisiologia , Neuropeptídeos/metabolismo , Núcleo Accumbens/metabolismo , Receptores de Neurotransmissores/metabolismo , Área Tegmentar Ventral/metabolismo , Animais , Transtorno da Compulsão Alimentar/psicologia , Bulimia/psicologia , Carboidratos da Dieta/administração & dosagem , Gorduras na Dieta/administração & dosagem , Ingestão de Alimentos/psicologia , Ingestão de Energia/fisiologia , Comportamento Alimentar/psicologia , Masculino , Obesidade/psicologia , Ratos Sprague-Dawley
12.
Pharmacol Biochem Behav ; 169: 35-47, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29654794

RESUMO

Binge eating disorder (BED) is a stress-related disorder characterized by acute episodes of excessive food intake. Piracetam, a nootropic agent has been reported to show several other neuropharmacological properties. The present study, evaluated the pharmacological effect of piracetam (200 mg/kg i.p.) on BED in female rats, induced by free access to palatable cookies for 2 h on alternate days. BED was confirmed by an increase in binge eating behavior and weight gain. BED leads to anxiety, cognitive and memory deficits, as evaluated by EPM (Elevated plus maze), OFT (open field test), and Y-maze tests. Increased levels of plasma corticosterone (CORT), glutamate in nucleus accumbens (NAC), hypothalamus (HYP) and prefrontal cortex (PFC) indicate stress and excitotoxicity. Moreover, it was observed that the levels of dopamine were higher in NAC and PFC, and less in HYP which may be responsible for motivational behavior for palatable feeding and cognitive deficits. More surprisingly, feeding behaviour regulating hormones namelyleptin was increased and ghrelin level was decreased in BED. Further, level of acetylcholine which regulates cognitive behaviour was compromised in BED. Piracetam significantly decreased binge eating behavior and associated body weight and regulated the levels of concerned neurotransmitters in respective regions. However, piracetam did not alter normal feeding behavior in the fast-refed model. Further, piracetam showed brain region-specific decrease in vascular endothelial growth factor expression. Piracetam showed anxiolytic activity and also alleviated cognitive deficit observed in BED. Hence, preclinical evidence indicates the potential use of piracetam for the treatment of BED.


Assuntos
Bulimia/prevenção & controle , Nootrópicos/farmacologia , Piracetam/farmacologia , Acetilcolina/metabolismo , Animais , Bulimia/metabolismo , Transtornos Cognitivos/prevenção & controle , Corticosterona/sangue , Dopamina/sangue , Comportamento Alimentar/efeitos dos fármacos , Feminino , Hipotálamo/metabolismo , Aprendizagem em Labirinto/efeitos dos fármacos , Memória/efeitos dos fármacos , Neurotransmissores/metabolismo , Núcleo Accumbens/metabolismo , Córtex Pré-Frontal/metabolismo , Ratos Wistar , Aumento de Peso/efeitos dos fármacos
13.
Physiol Behav ; 192: 3-16, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29654812

RESUMO

Compulsive binge eating is a hallmark of binge eating disorder and bulimia nervosa and is implicated in some obesity cases. Eating disorders are sexually dimorphic, with females more often affected than males. Animal models of binge-like eating based on intermittent access to palatable food exist; but, little is known regarding sex differences or individual vulnerability in these models with respect to the reinforcing efficacy of food, the development of compulsive- and binge-like eating, or associated changes in whole-body metabolism or body composition. Adolescent male (n = 24) and female (n = 32) Wistar rats were maintained on chow or a preferred, high-sucrose, chocolate-flavored diet in continuous or intermittent, extended access conditions. Body weight and composition, intake, fixed- and progressive-ratio operant self-administration, and whole body energy expenditure and respiratory exchange ratios were measured across an 11-week study period. Subgroup analyses were conducted to differentiate compulsive-like "high responder" intermittent access rats that escalated to extreme progressive-ratio self-administration performance vs. more resistant "low responders." Female rats had greater reinforcing efficacy of food than males in all diet conditions and were more often classified as "high responders". In both sexes, rats with intermittent access showed cycling of fuel substrate utilization and whole-body energy expenditure. Further, "high-responding" intermittent access female rats had especially elevated respiratory exchange ratios, indicating a fat-sparing phenotype. Future studies are needed to better understand the molecular and neurobiological basis of the sex and individual differences we have observed in rats and their translational impact for humans with compulsive, binge eating disorders.


Assuntos
Bulimia/metabolismo , Bulimia/psicologia , Suscetibilidade a Doenças/metabolismo , Suscetibilidade a Doenças/psicologia , Reforço Psicológico , Caracteres Sexuais , Animais , Composição Corporal/fisiologia , Peso Corporal/fisiologia , Modelos Animais de Doenças , Metabolismo Energético/fisiologia , Comportamento Alimentar/fisiologia , Comportamento Alimentar/psicologia , Feminino , Alimentos , Masculino , Ratos Wistar , Percepção Gustatória/fisiologia
14.
Nutrients ; 10(1)2018 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-29324652

RESUMO

The feeding process is required for basic life, influenced by environment cues and tightly regulated according to demands of the internal milieu by regulatory brain circuits. Although eating behaviour cannot be considered "addictive" under normal circumstances, people can become "addicted" to this behaviour, similarly to how some people are addicted to drugs. The symptoms, cravings and causes of "eating addiction" are remarkably similar to those experienced by drug addicts, and both drug-seeking behaviour as eating addiction share the same neural pathways. However, while the drug addiction process has been highly characterised, eating addiction is a nascent field. In fact, there is still a great controversy over the concept of "food addiction". This review aims to summarize the most relevant animal models of "eating addictive behaviour", emphasising binge eating disorder, that could help us to understand the neurobiological mechanisms hidden under this behaviour, and to improve the psychotherapy and pharmacological treatment in patients suffering from these pathologies.


Assuntos
Comportamento Animal , Bulimia/psicologia , Comportamento Alimentar , Dependência de Alimentos/psicologia , Animais , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Bulimia/genética , Bulimia/metabolismo , Bulimia/fisiopatologia , Sinais (Psicologia) , Modelos Animais de Doenças , Dopamina/metabolismo , Meio Ambiente , Dependência de Alimentos/genética , Dependência de Alimentos/metabolismo , Dependência de Alimentos/fisiopatologia , Predisposição Genética para Doença , Humanos , Vias Neurais/metabolismo , Vias Neurais/fisiopatologia , Peptídeos Opioides/metabolismo , Fatores de Risco , Transdução de Sinais
15.
Physiol Behav ; 183: 18-26, 2018 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-29050902

RESUMO

Binge eating is a specific form of overeating characterized by intermittent, excessive eating. To date, several studies have addressed the effects that bingeing on fat has on the rewarding effects of drugs of abuse, but they have found contradictory and highly variable results. Housing conditions could modulate these results, as most studies employ isolated animals to measure the exact amount of food that is ingested. The aim of this study was to evaluate the effects of housing conditions on the response of mice to cocaine, modulated by bingeing on a high-fat diet during adolescence. After 40days of binge-eating for 2h, three days a week (PND 29-69), the reinforcing effects of a non-effective dose of cocaine (1mg/kg) was evaluated using the conditioned place preference (CPP) paradigm. The anxiolytic profile using the Elevated Plus Maze and circulating leptin and corticosterone levels were also assessed. Our results show a significant escalation in the consumption of a high-fat diet between the first and the last week in both types of housed mice. Among the grouped mice, only those exposed to high-fat binge (HFB) developed CPP. Conversely, isolated mice fed with standard diet were more sensitive to the rewarding effects of a subthreshold dose of cocaine than those fed with HFB. Plasma leptin levels were elevated in both groups that developed CPP. Although isolated animals presented higher corticosterone levels with respect to the grouped ones, anxiety levels did not differ. Therefore, our results highlight the importance of housing conditions on the effects that a high-fat diet exerts on cocaine reward.


Assuntos
Bulimia/psicologia , Cocaína/farmacologia , Dieta Hiperlipídica/psicologia , Inibidores da Captação de Dopamina/farmacologia , Recompensa , Isolamento Social/psicologia , Animais , Animais não Endogâmicos , Ansiedade/metabolismo , Bulimia/metabolismo , Transtornos Relacionados ao Uso de Cocaína/metabolismo , Transtornos Relacionados ao Uso de Cocaína/psicologia , Condicionamento Psicológico/efeitos dos fármacos , Condicionamento Psicológico/fisiologia , Corticosterona/metabolismo , Modelos Animais de Doenças , Abrigo para Animais , Leptina/metabolismo , Masculino , Camundongos , Distribuição Aleatória , Comportamento Espacial/efeitos dos fármacos , Comportamento Espacial/fisiologia
16.
Neuropsychopharmacology ; 43(3): 617-626, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28895569

RESUMO

Binge-eating disorder (BED) is characterized by recurring episodes of excessive consumption of palatable food and an increased sensitivity to food cues. Patients with BED display an addiction-like symptomatology and the dopamine system might be a potential treatment target. The clinically safe monoamine stabilizer (-)-OSU6162 (OSU6162) restores dopaminergic dysfunction in long-term alcohol-drinking rats and shows promise as a novel treatment for alcohol use disorder. Here, the effects of OSU6162 on consummatory (binge-like eating) and appetitive (cue-controlled seeking) behavior motivated by chocolate-flavored sucrose pellets were evaluated in non-food-restricted male Lister Hooded rats. OSU6162 significantly reduced binge-like intake of chocolate-flavored sucrose pellets without affecting prior chow intake. Furthermore, OSU6162 significantly reduced the cue-controlled seeking of chocolate-flavored sucrose pellets under a second-order schedule of reinforcement before, but not after, the delivery and ingestion of reward, indicating a selective effect on incentive motivational processes. In contrast, the dopamine D2/D3 receptor antagonist raclopride reduced the seeking of chocolate-flavored sucrose pellets both pre- and post reward ingestion and also reduced responding under simpler schedules of seeking behavior. The D1/5 receptor antagonist SCH23390 had no effect on instrumental behavior under any reinforcement schedule tested. Finally, local administration of OSU6162 into the nucleus accumbens core, but not dorsolateral striatum, selectively reduced cue-controlled sucrose seeking. In conclusion, the present results show that OSU6162 reduces binge-like eating behavior and attenuates the impact of cues on seeking of palatable food. This indicates that OSU6162 might serve as a novel BED medication.


Assuntos
Transtorno da Compulsão Alimentar/tratamento farmacológico , Comportamento Alimentar/efeitos dos fármacos , Neurotransmissores/farmacologia , Piperidinas/farmacologia , Recompensa , Animais , Comportamento Apetitivo/efeitos dos fármacos , Comportamento Apetitivo/fisiologia , Benzazepinas/farmacologia , Transtorno da Compulsão Alimentar/metabolismo , Bulimia/tratamento farmacológico , Bulimia/metabolismo , Condicionamento Operante/efeitos dos fármacos , Condicionamento Operante/fisiologia , Sinais (Psicologia) , Sacarose Alimentar , Relação Dose-Resposta a Droga , Comportamento Alimentar/fisiologia , Masculino , Núcleo Accumbens/efeitos dos fármacos , Núcleo Accumbens/metabolismo , Racloprida/farmacologia , Ratos
18.
PLoS One ; 12(8): e0183063, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28813474

RESUMO

A compelling body of evidence suggests that the worldwide obesity epidemic is underpinned by excessive sugar consumption, typified by the modern western diet. Furthermore, evidence is beginning to emerge of maladaptive changes in the mesolimbic reward pathway of the brain in relation to excess sugar consumption that highlights the importance of examining this neural circuitry in an attempt to understand and subsequently mitigate the associated morbidities with obesity. While the basolateral amygdala (BLA) has been shown to mediate the reinforcing properties of drugs of abuse, it has also been shown to play an important role in affective and motivated behaviours and has been shown to undergo maladaptive changes in response to drugs of abuse and stress. Given the overlap in neural circuitry affected by drugs of abuse and sucrose, we sought to examine the effect of short- and long-term binge-like sucrose consumption on the morphology of the BLA principal neurons using an intermittent-access two-bottle choice paradigm. We used Golgi-Cox staining to impregnate principal neurons from the BLA of short- (4 week) and long-term (12 week) sucrose consuming adolescent rats and compared these to age-matched water controls. Our results indicate possibly maladaptive changes to the dendritic architecture of BLA principal neurons, particularly on apical dendrites following long-term sucrose consumption. Specifically, our results show reduced total dendritic arbor length of BLA principal neurons following short- and long-term sucrose consumption. Additionally, we found that long-term binge-like sucrose consumption caused a significant reduction in the length and complexity of apical dendrites. Taken together, our results highlight the differences between short- and long-term binge-like sucrose consumption on BLA principal neuron morphology and are suggestive of a perturbation in the diverse synaptic inputs to these neurons.


Assuntos
Tonsila do Cerebelo/metabolismo , Tonsila do Cerebelo/patologia , Bulimia/metabolismo , Bulimia/patologia , Dendritos/patologia , Neurônios/metabolismo , Neurônios/patologia , Sacarose , Fatores Etários , Animais , Masculino , Ratos
19.
eNeuro ; 4(3)2017.
Artigo em Inglês | MEDLINE | ID: mdl-28560316

RESUMO

Leptin receptors (LepRs) expressed in the midbrain contribute to the action of leptin on feeding regulation. The midbrain neurons release a variety of neurotransmitters including dopamine (DA), glutamate and GABA. However, which neurotransmitter mediates midbrain leptin action on feeding remains unclear. Here, we showed that midbrain LepR neurons overlap with a subset of dopaminergic, GABAergic and glutamatergic neurons. Specific removal of vesicular monoamine transporter 2 (VMAT2) in midbrain LepR neurons (KO mice) disrupted DA accumulation in vesicles, but failed to cause a significant change in the evoked release of either glutamate or GABA to downstream neurons. While KO mice showed no differences on chow, they presented a reduced high-fat diet (HFD) intake and resisted to HFD-induced obesity. Specific activation of midbrain LepR neurons promoted VMAT2-dependent feeding on chow and HFD. When tested with an intermittent access to HFD where first 2.5-h HFD eating (binge-like) and 24-h HFD feeding were measured, KO mice exhibited more binge-like, but less 24-h HFD feeding. Interestingly, leptin inhibited 24-h HFD feeding in controls but not in KO mice. Thus, VMAT2-mediated neurotransmission from midbrain LepR neurons contributes to both binge-like eating and HFD feeding regulation.


Assuntos
Comportamento Alimentar/fisiologia , Mesencéfalo/metabolismo , Neurônios/metabolismo , Receptores para Leptina/metabolismo , Transmissão Sináptica/fisiologia , Proteínas Vesiculares de Transporte de Monoamina/metabolismo , Animais , Bulimia/metabolismo , Bulimia/patologia , Dieta Hiperlipídica , Modelos Animais de Doenças , Suscetibilidade a Doenças/metabolismo , Dopamina/metabolismo , Feminino , Ácido Glutâmico/metabolismo , Leptina/administração & dosagem , Leptina/metabolismo , Masculino , Mesencéfalo/citologia , Mesencéfalo/patologia , Camundongos Transgênicos , Neurônios/citologia , Neurônios/patologia , Obesidade/metabolismo , Obesidade/patologia , Técnicas de Cultura de Tecidos , Proteínas Vesiculares de Transporte de Monoamina/genética , Ácido gama-Aminobutírico/metabolismo
20.
J Psychopharmacol ; 31(6): 770-783, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28376679

RESUMO

Adult, female rats given irregular, limited access to chocolate develop binge-eating behaviour with normal bodyweight and compulsive/perseverative and impulsive behaviours similar to those in binge-eating disorder. We investigated whether (a) dysregulated central nervous system dopaminergic and opioidergic systems are part of the psychopathology of binge-eating and (b) these neurotransmitter systems may mediate the actions of drugs ameliorating binge-eating disorder psychopathology. Binge-eating produced a 39% reduction of striatal D1 receptors with 22% and 23% reductions in medial and lateral caudate putamen and a 22% increase of striatal µ-opioid receptors. There was no change in D1 receptor density in nucleus accumbens, medial prefrontal cortex or dorsolateral frontal cortex, striatal D2 receptors and dopamine reuptake transporter sites, or µ-opioid receptors in frontal cortex. There were no changes in ligand affinities. The concentrations of monoamines, metabolites and estimates of dopamine (dopamine/dihydroxyphenylacetic acid ratio) and serotonin/5-hydroxyindolacetic acid ratio turnover rates were unchanged in striatum and frontal cortex. However, turnover of dopamine and serotonin in the hypothalamus was increased ~20% and ~15%, respectively. Striatal transmission via D1 receptors is decreased in binge-eating rats while µ-opioid receptor signalling may be increased. These changes are consistent with the attenuation of binge-eating by lisdexamfetamine, which increases catecholaminergic neurotransmission, and nalmefene, a µ-opioid antagonist.


Assuntos
Encéfalo/metabolismo , Dopamina/metabolismo , Receptores Opioides mu/metabolismo , Animais , Transtorno da Compulsão Alimentar/tratamento farmacológico , Transtorno da Compulsão Alimentar/metabolismo , Encéfalo/efeitos dos fármacos , Bulimia/tratamento farmacológico , Bulimia/metabolismo , Proteínas da Membrana Plasmática de Transporte de Dopamina/metabolismo , Feminino , Naltrexona/análogos & derivados , Naltrexona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Psicopatologia , Ratos , Ratos Wistar , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...