Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 83
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Int J Eat Disord ; 56(6): 1098-1113, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36840536

RESUMO

OBJECTIVE: Consumption of energy-dense palatable "comfort" food can alleviate stress and negative emotions, while abrupt withdrawal from a palatable diet can worsen these symptoms, causing difficulties with adherence to weight-loss diets. Currently, no pharmacological treatment is effective for obesity-related anxiety, so we investigated the endocannabinoid system (ECS), and specifically the fatty acid amide hydrolase (FAAH), as an interesting emerging target in this context because of its key role in the regulation of both energy homeostasis and emotional behavior. METHODS: Rats were subjected to exposure and subsequent abstinence from a palatable cafeteria diet. During abstinence period, rats were treated with the selective FAAH inhibitor PF-3845 (10 mg/kg; intraperitoneal administration every other day). RESULTS: Abstinent rats displayed an anxiogenic-like behavior and changes in the proteins of ECS signaling machinery in brain areas involved both in anxiety and food intake regulation. In particular, withdrawal caused a reduction of the expression of cannabinoid receptors in the nucleus accumbens and of enzymes diacylglycerol lipase alpha and monoacylglycerol lipase (MAGL) in the amygdala. Pharmacological inhibition of FAAH exerted an anxiolytic-like effect in abstinent animals and increased both MAGL expression in amygdala and CB2 expression in prefrontal cortex. DISCUSSION: Overall, our results suggest that emotional disturbances associated with dieting are coupled with region-specific alterations in the cerebral expression of the ECS and that the enhancement of the endocannabinoid signaling by FAAH inhibition might represent a novel pharmacological strategy for the treatment of anxiety related to abstinence from palatable food. PUBLIC SIGNIFICANCE: The present study focused on evaluating the role of the endocannabinoid system in modulating withdrawal from naturally rewarding activities that have an impact on mood, such as feeding. The variations observed in the emotional behavior of abstinent rats was linked to neuroadaptations of the ECS in specific brain areas.


Assuntos
Amidoidrolases , Endocanabinoides , Ratos , Humanos , Animais , Amidoidrolases/metabolismo , Ansiedade/tratamento farmacológico
2.
Int J Mol Sci ; 24(3)2023 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-36769334

RESUMO

In the present study, we used a mouse model of Alzheimer's disease (AD) (3×Tg-AD mice) to longitudinally analyse the expression level of PDIA3, a protein disulfide isomerase and endoplasmic reticulum (ER) chaperone, in selected brain limbic areas strongly affected by AD-pathology (amygdala, entorhinal cortex, dorsal and ventral hippocampus). Our results suggest that, while in Non-Tg mice PDIA3 levels gradually reduce with aging in all brain regions analyzed, 3×Tg-AD mice showed an age-dependent increase in PDIA3 levels in the amygdala, entorhinal cortex, and ventral hippocampus. A significant reduction of PDIA3 was observed in 3×Tg-AD mice already at 6 months of age, as compared to age-matched Non-Tg mice. A comparative immunohistochemistry analysis performed on 3×Tg-AD mice at 6 (mild AD-like pathology) and 18 (severe AD-like pathology) months of age showed a direct correlation between the cellular level of Aß and PDIA3 proteins in all the brain regions analysed, even if with different magnitudes. Additionally, an immunohistochemistry analysis showed the presence of PDIA3 in all post-mitotic neurons and astrocytes. Overall, altered PDIA3 levels appear to be age- and/or pathology-dependent, corroborating the ER chaperone's involvement in AD pathology, and supporting the PDIA3 protein as a potential novel therapeutic target for the treatment of AD.


Assuntos
Doença de Alzheimer , Camundongos , Animais , Doença de Alzheimer/metabolismo , Camundongos Transgênicos , Isomerases de Dissulfetos de Proteínas/genética , Isomerases de Dissulfetos de Proteínas/metabolismo , Encéfalo/metabolismo , Camundongos Endogâmicos , Modelos Animais de Doenças , Peptídeos beta-Amiloides/metabolismo , Proteínas tau/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo
3.
Pharmacol Res ; 185: 106521, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36272641

RESUMO

The melanocortinergic neural circuit, known for its influence on energy expenditure and feeding behavior, also plays a role in stress and stress-induced psychiatric disorders, including anxiety and depression. The major contribution is given by the melanocortin-4 receptor (MC4R) subtype, highly expressed in brain regions involved in the control of stress responses. Furthermore, the MC4R appears to profoundly affect the activity of the hypothalamic-pituitary-adrenal (HPA) axis, and it has been also highlighted a functional and anatomical interaction with the corticotropin-releasing factor (CRF), an important mediator of stress and stress-related behaviors. The MC4R agonists seem to exacerbate stress-inducing anxiety- and depressive-like behavior, while MC4R antagonists have been demonstrated to mitigate such disorders, as shown in several preclinical behavioral tests. The evidence collected in the present review suggests that the melanocortin system, through the MC4R, could possibly modulate behavioral responses to stress, suggesting the use of MC4R antagonists as a possible novel treatment for anxiety and depression induced by stress.


Assuntos
Melanocortinas , Sistema Hipófise-Suprarrenal , Humanos , Ansiedade/tratamento farmacológico , Sistema Hipotálamo-Hipofisário , Estresse Fisiológico
4.
FASEB J ; 34(7): 9358-9371, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32463138

RESUMO

Numerous nutritional approaches aimed at reducing body weight have been developed as a strategy to reduce obesity. Most of these interventions rely on reducing caloric intake or limiting calories access to a few hours per day. In this work, we analyzed the effects of the extended (24 hours/day) or restricted (1 hour/day) access to a cafeteria-style (CAF) diet, on rat body weight and hepatic lipid metabolism, with respect to control rats (CTR) fed with a standard chow diet. The body weight gain of restricted-fed rats was not different from CTR, despite the slightly higher total caloric intake, but resulted significantly lower than extended-fed rats, which showed a CAF diet-induced obesity and a dramatically higher total caloric intake. However, both CAF-fed groups of rats showed, compared to CTR, unhealthy serum and hepatic parameters such as higher serum glucose level, lower HDL values, and increased hepatic triacylglycerol and cholesterol amount. The hepatic expression and activity of key enzymes of fatty acid synthesis, acetyl-CoA carboxylase (ACC), and fatty acid synthase (FAS), was similarly reduced in both CAF-fed groups of rats with respect to CTR. Anyway, while in extended-fed rats this reduction was associated to a long-term mechanism involving sterol regulatory element-binding protein-1 (SREBP-1), in restricted-fed animals a short-term mechanism based on PKA and AMPK activation occurred in the liver. Furthermore, hepatic fatty acid oxidation (FAO) and oxidative stress resulted significantly increased in extended, but not in restricted-fed rats, as compared to CTR. Overall, these results demonstrate that although limiting the total caloric intake might successfully fight obesity development, the nutritional content of the diet is the major determinant for the health status.


Assuntos
Peso Corporal , Dieta Hiperlipídica/efeitos adversos , Lipogênese , Fígado/metabolismo , Fígado/patologia , Aumento de Peso , Animais , Ingestão de Energia , Lipídeos/sangue , Masculino , Ratos , Ratos Wistar
5.
J Neurochem ; 152(6): 623-626, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31917872

RESUMO

The dorsal striatum coordinates input-output processing of numerous functions including those related to motor activity, motivation, and learning. Considerable anatomical and biochemical heterogeneity across striatal subregions has long been known to result in distinct functional outcomes, and for imbalances in these pathways to contribute to many complex disorders. Here we highlight the study of Hörtnagl et al. (2019) who utilize precision dissection of human caudate nucleus and putamen for detailed measurement of major neurochemical markers to address the question of anatomical heterogeneity of neurotransmitter distribution and turnover in these regions. The findings identify gradients of neurotransmitter markers in rostro-caudal, dorso-lateral, and anterior-posterior directions with a precision that has not been previously determined in humans. Correlative analyses of the results also suggest tentative links between content of various neurotransmitters in specific subregions, raising the intriguing possibility that neurotransmitter quantity in one territory may correlate with the quantity of the same or different transmitter from another territory. This suggests the presence of a functional anatomy over extensive brain regions and networks that can be studied through multiple correlative analyses, and identify a possible basis for a new approach for postmortem analysis of neurotransmitter distribution and function.


Assuntos
Biomarcadores/análise , Núcleo Caudado/química , Neurotransmissores/análise , Putamen/química , Idoso , Feminino , Humanos , Masculino , Mudanças Depois da Morte
6.
FASEB J ; 33(1): 1428-1439, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30133327

RESUMO

It is widely accepted that chronic stress may alter the homeostatic mechanisms of body weight control. In this study, we followed the metabolic changes occurring in mice when chronic stress caused by psychosocial defeat (CPD) is associated with ad libitum exposure to a palatable high-fat diet (HFD). In this model, CPD mice consumed more HFD than unstressed (Un) mice without gaining body weight. We focused on metabolic processes involved in weight control, such as de novo lipogenesis (DNL), fatty acid ß-oxidation (FAO), and thermogenesis. The activity and expression of DNL enzymes were reduced in the liver and white adipose tissue of mice consuming the HFD. Such effects were particularly evident in stressed mice. In both CPD and Un mice, HFD consumption increased the hepatic expression of the mitochondrial FAO enzyme carnitine palmitoyltransferase-1. In the liver of mice consuming the HFD, stress exposure prevented accumulation of triacylglycerols; however, accumulation of triacylglycerols was observed in Un mice under the same dietary regimen. In brown adipose tissue, stress increased the expression of uncoupling protein-1, which is involved in energy dissipation, both in HFD and control diet-fed mice. We consider increased FAO and energy dissipation responsible for the antiobesity effect seen in CPD/HFD mice. However, CPD associated with HFD induced hepatic oxidative stress.-Giudetti, A. M., Testini, M., Vergara, D., Priore, P., Damiano, F., Gallelli, C. A., Romano, A., Villani, R., Cassano, T., Siculella, L., Gnoni, G. V., Moles, A., Coccurello, R., Gaetani, S. Chronic psychosocial defeat differently affects lipid metabolism in liver and white adipose tissue and induces hepatic oxidative stress in mice fed a high-fat diet.


Assuntos
Tecido Adiposo Branco/metabolismo , Dieta Hiperlipídica , Metabolismo dos Lipídeos , Fígado/metabolismo , Estresse Oxidativo , Estresse Psicológico , Acetil-CoA Carboxilase/metabolismo , Tecido Adiposo Marrom/enzimologia , Tecido Adiposo Marrom/metabolismo , Tecido Adiposo Branco/enzimologia , Animais , Peso Corporal , Carnitina O-Palmitoiltransferase/genética , Carnitina O-Palmitoiltransferase/metabolismo , Modelos Animais de Doenças , Ingestão de Energia , Ácido Graxo Sintases/metabolismo , Ácidos Graxos/metabolismo , Glutationa/metabolismo , Fígado/enzimologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , RNA Mensageiro/genética , Proteína Desacopladora 1/metabolismo
7.
Eur J Nutr ; 57(4): 1485-1498, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28314964

RESUMO

PURPOSE: Chronic exposure to stress may represent a risk factor for developing metabolic and eating disorders, mostly driven by the overconsumption of easily accessible energy-dense palatable food, although the mechanisms involved remain still unclear. In this study, we used an ethologically oriented murine model of chronic stress caused by chronic psychosocial defeat (CPD) to investigate the effects of unrestricted access to a palatable high fat diet (HFD) on food intake, body weight, energy homeostasis, and expression of different brain neuropeptides. Our aim was to shed light on the mechanisms responsible for body weight and body composition changes due to chronic social stress. METHODS: In our model of subordinate (defeated), mice (CPD) cohabitated in constant sensory contact with dominants, being forced to interact on daily basis, and were offered ad libitum access either to an HFD or to a control diet (CD). Control mice (of the same strain as CPD mice) were housed in pairs and left unstressed in their home cage (UN). In all these mice, we evaluated body weight, different adipose depots, energy metabolism, caloric intake, and neuropeptide expression. RESULTS: CPD mice increased the intake of HFD and reduced body weight in the presence of enhanced lipid oxidation. Resting energy expenditure and interscapular brown adipose tissue (iBAT) were increased in CPD mice, whereas epididymal adipose tissue increased only in HFD-fed unstressed mice. Propiomelanocortin mRNA levels in hypothalamic arcuate nucleus increased only in HFD-fed unstressed mice. Oxytocin mRNA levels in the paraventricular nucleus and neuropeptide Y mRNA levels within the arcuate were increased only in CD-fed CPD mice. In the arcuate, CART was increased in HFD-fed UN mice and in CD-fed CPD mice, while HFD intake suppressed CART increase in defeated animals. In the basolateral amygdala, CART expression was increased only in CPD animals on HFD. CONCLUSIONS: CPD appears to uncouple the intake of HFD from energy homeostasis causing higher HFD intake, larger iBAT accumulation, increased energy expenditure and lipid oxidation, and lower body weight. Overall, the present study confirms the notion that the chronic activation of the stress response can be associated with metabolic disorders, altered energy homeostasis, and changes of orexigenic and anorexigenic signaling. These changes might be relevant to better understand the etiology of stress-induced obesity and eating disorders and might represent a valid therapeutic approach for the development of new therapies in this field.


Assuntos
Dieta Hiperlipídica , Ingestão de Energia/fisiologia , Metabolismo Energético/fisiologia , Animais , Peso Corporal , Itália , Leptina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Obesidade
8.
Pharmacol Res ; 122: 20-34, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28535974

RESUMO

The satiety-promoting action of oleoylethanolamide (OEA) has been associated to the indirect activation of selected brain areas, such as the nucleus of the solitary tract (NST) in the brainstem and the tuberomammillary (TMN) and paraventricular (PVN) nuclei in the hypothalamus, where noradrenergic, histaminergic and oxytocinergic neurons play a necessary role. Visceral ascending fibers were hypothesized to mediate such effects. However, our previous findings demonstrated that the hypophagic action of peripherally administered OEA does not require intact vagal afferents and is associated to a strong activation of the area postrema (AP). Therefore, we hypothesized that OEA may exert its central effects through the direct activation of this circumventricular organ. To test this hypothesis, we subjected rats to the surgical ablation of the AP (APX rats) and evaluated the effects of OEA (10mgkg-1 i.p.) on food intake, Fos expression, hypothalamic oxytocin (OXY) immunoreactivity and on the expression of dopamine beta hydroxylase (DBH) in the brainstem and hypothalamus. We found that the AP lesion completely prevented OEA's behavioral and neurochemical effects in the brainstem and the hypothalamus. Moreover OEA increased DBH expression in AP and NST neurons of SHAM rats while the effect in the NST was absent in APX rats, thus suggesting the possible involvement of noradrenergic AP neurons. These results support the hypothesis of a necessary role of the AP in mediating OEA's central effects that sustain its pro-satiety action.


Assuntos
Área Postrema/efeitos dos fármacos , Tronco Encefálico/efeitos dos fármacos , Ingestão de Alimentos/efeitos dos fármacos , Endocanabinoides/farmacologia , Hipotálamo/efeitos dos fármacos , Ácidos Oleicos/farmacologia , Animais , Área Postrema/fisiologia , Tronco Encefálico/fisiologia , Dopamina beta-Hidroxilase/análise , Dopamina beta-Hidroxilase/metabolismo , Hipotálamo/fisiologia , Masculino , Ocitocina/análise , Ocitocina/metabolismo , PPAR alfa/análise , PPAR alfa/metabolismo , Proteínas Proto-Oncogênicas c-fos/análise , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos Wistar
9.
Proc Natl Acad Sci U S A ; 111(31): 11527-32, 2014 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-25049422

RESUMO

Key factors driving eating behavior are hunger and satiety, which are controlled by a complex interplay of central neurotransmitter systems and peripheral stimuli. The lipid-derived messenger oleoylethanolamide (OEA) is released by enterocytes in response to fat intake and indirectly signals satiety to hypothalamic nuclei. Brain histamine is released during the appetitive phase to provide a high level of arousal in anticipation of feeding, and mediates satiety. However, despite the possible functional overlap of satiety signals, it is not known whether histamine participates in OEA-induced hypophagia. Using different experimental settings and diets, we report that the anorexiant effect of OEA is significantly attenuated in mice deficient in the histamine-synthesizing enzyme histidine decarboxylase (HDC-KO) or acutely depleted of histamine via interocerebroventricular infusion of the HDC blocker α-fluoromethylhistidine (α-FMH). α-FMH abolished OEA-induced early occurrence of satiety onset while increasing histamine release in the CNS with an H3 receptor antagonist-increased hypophagia. OEA augmented histamine release in the cortex of fasted mice within a time window compatible to its anorexic effects. OEA also increased c-Fos expression in the oxytocin neurons of the paraventricular nuclei of WT but not HDC-KO mice. The density of c-Fos immunoreactive neurons in other brain regions that receive histaminergic innervation and participate in the expression of feeding behavior was comparable in OEA-treated WT and HDC-KO mice. Our results demonstrate that OEA requires the integrity of the brain histamine system to fully exert its hypophagic effect and that the oxytocin neuron-rich nuclei are the likely hypothalamic area where brain histamine influences the central effects of OEA.


Assuntos
Encéfalo/metabolismo , Ingestão de Alimentos/efeitos dos fármacos , Histamina/metabolismo , Ácidos Oleicos/farmacologia , Resposta de Saciedade/efeitos dos fármacos , Animais , Comportamento Animal/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Endocanabinoides , Comportamento Alimentar/efeitos dos fármacos , Histidina Descarboxilase/metabolismo , Masculino , Camundongos , Camundongos Knockout , Modelos Biológicos , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Núcleo Hipotalâmico Paraventricular/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Fatores de Tempo
10.
J Neurochem ; 139(5): 691-699, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27649625

RESUMO

The central nervous system and viscera constitute a functional ensemble, the gut-brain axis, that allows bidirectional information flow that contributes to the control of feeding behavior based not only on the homeostatic, but also on the hedonic aspects of food intake. The prevalence of eating disorders, such as anorexia nervosa, binge eating and obesity, poses an enormous clinical burden, and involves an ever-growing percentage of the population worldwide. Clinical and preclinical research is constantly adding new information to the field and orienting further studies with the aim of providing a foundation for developing more specific and effective treatment approaches to pathological conditions. A recent symposium at the XVI Congress of the Societá Italiana di Neuroscienze (SINS, 2015) 'Eating disorders: from bench to bedside and back' brought together basic scientists and clinicians with the objective of presenting novel perspectives in the neurobiology of eating disorders. Clinical studies presented by V. Ricca illustrated some genetic aspects of the psychopathology of anorexia nervosa. Preclinical studies addressed different issues ranging from the description of animal models that mimic human pathologies such as anorexia nervosa, diet-induced obesity, and binge eating disorders (T. Lutz), to novel interactions between peripheral signals and central circuits that govern food intake, mood and stress (A. Romano and G. Provensi). The gut-brain axis has received increasing attention in the recent years as preclinical studies are demonstrating that the brain and visceral organs such as the liver and guts, but also the microbiota are constantly engaged in processes of reciprocal communication, with unexpected physiological and pathological implications. Eating is controlled by a plethora of factors; genetic predisposition, early life adverse conditions, peripheral gastrointestinal hormones that act directly or indirectly on the central nervous system, all are receiving attention as they presumably contribute to the development of eating disorders.


Assuntos
Comportamento Alimentar/fisiologia , Transtornos da Alimentação e da Ingestão de Alimentos/metabolismo , Hormônios Gastrointestinais/metabolismo , Sistema Hipotálamo-Hipofisário/metabolismo , Sistema Hipófise-Suprarrenal/metabolismo , Animais , Ensaios Clínicos como Assunto/métodos , Metabolismo Energético/fisiologia , Comportamento Alimentar/psicologia , Transtornos da Alimentação e da Ingestão de Alimentos/psicologia , Transtornos da Alimentação e da Ingestão de Alimentos/terapia , Humanos , Obesidade/metabolismo , Obesidade/psicologia
11.
Pharmacol Res ; 105: 186-97, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26816085

RESUMO

Intracellular accumulation of hyperphosphorylated tau protein is linked to neuronal degeneration in Alzheimer's disease (AD). Mounting evidence suggests that tau phosphorylation and O-N-acetylglucosamine glycosylation (O-GlcNAcylation) are mutually exclusive post-translational modifications. O-GlcNAcylation depends on 3-5% of intracellular glucose that enters the hexosamine biosynthetic pathway. To our knowledge, the existence of an imbalance between tau phosphorylation and O-GlcNAcylation has not been reported in animal models of AD, as yet. Here, we used triple transgenic (3xTg-AD) mice at 12 months, an age at which hyperphosphorylated tau is already detected and associated with cognitive decline. In these mice, we showed that tau was hyperphosphorylated on both Ser396 and Thr205 in the hippocampus, and to a lower extent and exclusively on Thr205 in the frontal cortex. Tau O-GlcNAcylation, assessed in tau immunoprecipitates, was substantially reduced in the hippocampus of 3xTg-AD mice, with no changes in the frontal cortex or in the cerebellum. No changes in the expression of the three major enzymes involved in O-GlcNAcylation, i.e., glutamine fructose-6-phosphate amidotransferase, O-linked ß-N-acetylglucosamine transferase, and O-GlcNAc hydrolase were found in the hippocampus of 3xTg-AD mice. These data demonstrate that an imbalance between tau phosphorylation and O-GlcNAcylation exists in AD mice, and strengthens the hypothesis that O-GlcNAcylation might be targeted by disease modifying drugs in AD.


Assuntos
Acetilglucosamina/metabolismo , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Hipocampo/metabolismo , Hipocampo/patologia , Proteínas tau/metabolismo , Acilação , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Modelos Animais de Doenças , Glicosilação , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Fosforilação
12.
J Neurosci ; 34(34): 11316-24, 2014 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-25143612

RESUMO

We developed recently a binge-eating model in which female rats with a history of intermittent food restriction show binge-like palatable food consumption after 15 min exposure to the sight of the palatable food. This "frustration stress" manipulation also activates the hypothalamic-pituitary-adrenal stress axis. Here, we determined the role of the stress neurohormone corticotropin-releasing factor (CRF) in stress-induced binge eating in our model. We also assessed the role of CRF receptors in the bed nucleus of the stria terminalis (BNST), a brain region implicated in stress responses and stress-induced drug seeking, in stress-induced binge eating. We used four groups that were first exposed or not exposed to repeated intermittent cycles of regular chow food restriction during which they were also given intermittent access to high-caloric palatable food. On the test day, we either exposed or did not expose the rats to the sight of the palatable food for 15 min (frustration stress) before assessing food consumption for 2 h. We found that systemic injections of the CRF1 receptor antagonist R121919 (2,5-dimethyl-3-(6-dimethyl-4-methylpyridin-3-yl)-7 dipropylamino pyrazolo[1,5-a]pyrimidine) (10-20 mg/kg) and BNST (25-50 ng/side) or ventricular (1000 ng) injections of the nonselective CRF receptor antagonist D-Phe-CRF(12-41) decreased frustration stress-induced binge eating in rats with a history of food restriction. Frustration stress also increased Fos (a neuronal activity marker) expression in ventral and dorsal BNST. Results demonstrate a critical role of CRF receptors in BNST in stress-induced binge eating in our rat model. CRF1 receptor antagonists may represent a novel pharmacological treatment for bingeing-related eating disorders.


Assuntos
Bulimia/etiologia , Privação de Alimentos , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Núcleos Septais/metabolismo , Estresse Psicológico/complicações , Animais , Comportamento Consumatório/efeitos dos fármacos , Comportamento Consumatório/fisiologia , Hormônio Liberador da Corticotropina/análogos & derivados , Hormônio Liberador da Corticotropina/farmacologia , Feminino , Injeções Intraventriculares , Proteínas Oncogênicas v-fos/metabolismo , Pirimidinas/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores de Hormônio Liberador da Corticotropina/agonistas , Receptores de Hormônio Liberador da Corticotropina/antagonistas & inibidores , Núcleos Septais/efeitos dos fármacos , Fatores de Tempo
13.
J Neurosci Res ; 93(5): 777-87, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25581607

RESUMO

Limbic forebrain endocannabinoid (eCB) signaling is critically involved in stress integration by modulating neurotransmitters release. The purpose of this study was to examine, by brain microdialysis, the effects of fatty acid amide hydrolase (FAAH) inhibition on noradrenergic and γ-aminobutyric acid (GABA)-ergic neurotransmission in the prefrontal cortex (PFC) and basolateral amygdala (BLA) of rats subjected to a 20-min swim stress. Microdialysis started on stress- and drug-naïve rats that were treated with the FAAH inhibitor URB597 (0.1 or 0.3 mg/kg) 30 min before undergoing the stress procedure. Dialysate samples were collected every 20 min from the beginning of the experiment. Concentrations of noradrenaline (NA) and GABA were determined by HPLC coupled to electrochemical and fluorescence detection, respectively. We found that neither URB597 treatment nor 20 min of swim stress exposure per se altered NA and GABA extracellular levels in PFC or BLA. Interestingly, rats treated with 0.1 mg/kg of URB597 followed by 20 min of stress showed significantly higher NA and GABA levels in PFC and BLA. These effects were absent in rats treated with 0.3 mg/kg URB597, indicating a dose-specific effect. Moreover, we found that the pretreatment with the CB1 receptor antagonist rimonabant blocked the URB597 effects on NA and GABA release in PFC and BLA of animals subjected to forced swimming. The present study might provide an important first step toward understanding the mechanisms through which URB597 modulates stress-induced neuroendocrine secretion and behavioral coping strategies.


Assuntos
Ácidos Araquidônicos/metabolismo , Complexo Nuclear Basolateral da Amígdala/metabolismo , Endocanabinoides/metabolismo , Norepinefrina/metabolismo , Alcamidas Poli-Insaturadas/metabolismo , Estresse Psicológico/patologia , Natação/psicologia , Ácido gama-Aminobutírico/metabolismo , Análise de Variância , Animais , Complexo Nuclear Basolateral da Amígdala/efeitos dos fármacos , Benzamidas/farmacologia , Carbamatos/farmacologia , Cromatografia Líquida de Alta Pressão , Modelos Animais de Doenças , Líquido Extracelular/efeitos dos fármacos , Líquido Extracelular/metabolismo , Masculino , Microdiálise , Córtex Pré-Frontal , Ratos , Ratos Wistar
14.
J Neurosci Res ; 92(3): 347-58, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24273027

RESUMO

Parkinson's disease (PD) is a progressive neurodegenerative disorder whose etiology is still unclear in spite of extensive investigations. It has been hypothesized that 5-S-cysteinyldopamine (CysDA), a catechol-thioether metabolite of dopamine (DA), could be an endogenous parkinsonian neurotoxin. To gain further insight into its role in the neurodegenerative process, both CD1 mice and SH-SY5Y neuroblastoma cells were treated with CysDA, and the data were compared with those obtained by the use of 6-hydroxydopamine, a well-known parkinsonian mimetic. Intrastriatal injection of CysDA in CD1 mice caused a long-lasting depletion of DA, providing evidence of in vivo neurotoxicity of CysDA. Both in mice and in SH-SY5Y cells, CysDA treatment induced extensive oxidative stress, as evidenced by protein carbonylation and glutathione depletion, and affected the expression of two proteins, α-synuclein (α-Syn) and ERp57, whose levels are modulated by oxidative insult. Real-time PCR experiments support these findings, indicating an upregulation of both ERp57 and α-Syn expression. α-Syn aggregation was also found to be modulated by CysDA treatment. The present work provides a solid background sustaining the hypothesis that CysDA is involved in parkinsonian neurodegeneration by inducing extensive oxidative stress and protein aggregation.


Assuntos
Encéfalo/metabolismo , Dopaminérgicos/toxicidade , Dopamina/análogos & derivados , Doença de Parkinson/etiologia , Isomerases de Dissulfetos de Proteínas/metabolismo , alfa-Sinucleína/metabolismo , Animais , Monoaminas Biogênicas/metabolismo , Encéfalo/efeitos dos fármacos , Linhagem Celular Tumoral , Modelos Animais de Doenças , Dopamina/toxicidade , Relação Dose-Resposta a Droga , Glutationa/metabolismo , Dissulfeto de Glutationa/metabolismo , Humanos , Masculino , Camundongos , Neuroblastoma/patologia , Estresse Oxidativo/efeitos dos fármacos , Oxidopamina/toxicidade , Carbonilação Proteica/efeitos dos fármacos , Isomerases de Dissulfetos de Proteínas/genética , alfa-Sinucleína/genética
15.
Am J Physiol Regul Integr Comp Physiol ; 307(2): R167-78, 2014 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-24829501

RESUMO

The endogenous lipid messenger oleoylethanolamide (OEA) inhibits eating and modulates fat metabolism supposedly through the activation of peroxisome proliferator-activated receptor-α (PPARα) and vagal sensory fibers. We tested in adult male rats whether OEA stimulates fatty acid oxidation (FAO) and ketogenesis and whether it increases plasma levels of the satiating gut peptides glucagon-like peptide-1 (GLP-1) and peptide YY (PYY). We also explored whether OEA still inhibits eating after subdiaphragmatic vagal deafferentation (SDA). We found that intraperitoneally injected OEA (10 mg/kg body wt) reduced (P < 0.05) food intake mainly by increasing meal latency and that this effect was stronger in rats fed a 60% high-fat diet (HFD) than in chow-fed rats. OEA increased (P < 0.05) postprandial plasma nonesterified fatty acids and ß-hydroxybutyrate (BHB) in the hepatic portal vein (HPV) and vena cava (VC) 30 min after injection, which was more pronounced in HFD- than in chow-fed rats. OEA also increased the protein expression of the key ketogenetic enzyme, mitochondrial 3-hydroxy-3-methylglutaryl-CoA synthase, in the jejunum of HFD-fed rats, but not in the liver or duodenum of either diet group. Furthermore, OEA decreased GLP-1 and PYY concentrations (P < 0.05) in the HPV and VC 30 min after administration. Finally, OEA reduced food intake in SDA and sham-operated rats similarly. Our findings indicate that neither intact abdominal vagal afferents nor prandial increases in GLP-1 or


Assuntos
Ingestão de Alimentos/efeitos dos fármacos , Trato Gastrointestinal/efeitos dos fármacos , Metabolismo dos Lipídeos , Ácidos Oleicos/farmacologia , Saciação/efeitos dos fármacos , Animais , Dieta Hiperlipídica/efeitos adversos , Ingestão de Alimentos/fisiologia , Endocanabinoides , Trato Gastrointestinal/inervação , Trato Gastrointestinal/metabolismo , Peptídeo 1 Semelhante ao Glucagon/efeitos dos fármacos , Metabolismo dos Lipídeos/efeitos dos fármacos , Metabolismo dos Lipídeos/fisiologia , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Ácidos Oleicos/administração & dosagem , Ratos , Ratos Sprague-Dawley , Saciação/fisiologia , Nervo Vago/cirurgia
16.
Int J Neuropsychopharmacol ; 18(4)2014 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-25609597

RESUMO

BACKGROUND: Neuropsychiatric signs are critical in primary caregiving of Alzheimer patients and have not yet been fully investigated in murine models. METHODS: 18-month-old 3×Tg-AD male mice and their wild-type male littermates (non-Tg) were used. The open field test and the elevated plus maze test were used to evaluate anxiety-like behaviors, whereas the Porsolt forced swim test, the tail suspension test, and the sucrose preference test for antidepressant/depression-coping behaviors. Neurochemical study was conducted by microdialysis in freely-moving mice, analyzing the basal and K(+)-stimulated monoamine output in the frontal cortex and ventral hippocampus. Moreover by immunohistochemistry, we analysed the expression of Tyrosin hydroxylase and Tryptophan hydroxylase, which play a key role in the synthesis of monoamines. RESULTS: Aged 3×Tg-AD mice exhibited a higher duration of immobility in the forced swim and tail suspension tests (predictors of depression-like behavior) which was not attenuated by a noradrenaline reuptake inhibitor, desipramine. In the sucrose preference test, 3×Tg-AD mice showed a significantly lower sucrose preference compared to the non-Tg group, without any difference in total fluid intake. In contrast, the motor functions and anxiety-related emotional responses of 3×Tg-AD mice were normal, as detected by the open-field and elevated plus-maze tests. To strengthen these results, we then evaluated the monoaminergic neurotransmissions by in vivo microdialysis and immunohistochemistry. In particular, with the exception of the basal hippocampal dopamine levels, 3×Tg-AD mice exhibited a lower basal extracellular output of amines in the frontal cortex and ventral hippocampus and also a decreased extracellular response to K(+) stimulation. Such alterations occur with obvious local amyloid-ß and tau pathologies and without gross alterations in the expression of Tyrosin and Tryptophan hydroxylase. CONCLUSIONS: These results suggest that 3×Tg-AD mice exhibit changes in depression-related behavior involving aminergic neurotrasmitters and provide an animal model for investigating AD with depression.


Assuntos
Doença de Alzheimer/metabolismo , Monoaminas Biogênicas/metabolismo , Encéfalo/metabolismo , Depressão/metabolismo , Doença de Alzheimer/patologia , Doença de Alzheimer/psicologia , Peptídeos beta-Amiloides/metabolismo , Animais , Ansiedade/tratamento farmacológico , Ansiedade/metabolismo , Ansiedade/patologia , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Fármacos do Sistema Nervoso Central/farmacologia , Depressão/tratamento farmacológico , Depressão/patologia , Modelos Animais de Doenças , Espaço Extracelular/efeitos dos fármacos , Espaço Extracelular/metabolismo , Masculino , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Atividade Motora/efeitos dos fármacos , Atividade Motora/fisiologia , Potássio/farmacologia , Triptofano Hidroxilase/metabolismo , Tirosina 3-Mono-Oxigenase/metabolismo , Proteínas tau/metabolismo
17.
Transl Psychiatry ; 14(1): 193, 2024 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-38632257

RESUMO

Autism Spectrum Disorders (ASD) are principally diagnosed by three core behavioural symptoms, such as stereotyped repertoire, communication impairments and social dysfunctions. This complex pathology has been linked to abnormalities of corticostriatal and limbic circuits. Despite experimental efforts in elucidating the molecular mechanisms behind these abnormalities, a clear etiopathogenic hypothesis is still lacking. To this aim, preclinical studies can be really helpful to longitudinally study behavioural alterations resembling human symptoms and to investigate the underlying neurobiological correlates. In this regard, the BTBR T+ Itpr3tf/J (BTBR) mice are an inbred mouse strain that exhibits a pattern of behaviours well resembling human ASD-like behavioural features. In this study, the BTBR mice model was used to investigate neurochemical and biomolecular alterations, regarding Nerve Growth Factor (NGF) and Brain-Derived Neurotrophic Factor (BDNF), together with GABAergic, glutamatergic, cholinergic, dopaminergic and noradrenergic neurotransmissions and their metabolites in four different brain areas, i.e. prefrontal cortex, hippocampus, amygdala and hypothalamus. In our results, BTBR strain reported decreased noradrenaline, acetylcholine and GABA levels in prefrontal cortex, while hippocampal measurements showed reduced NGF and BDNF expression levels, together with GABA levels. Concerning hypothalamus, no differences were retrieved. As regarding amygdala, we found reduced dopamine levels, accompanied by increased dopamine metabolites in BTBR mice, together with decreased acetylcholine, NGF and GABA levels and enhanced glutamate content. Taken together, our data showed that the BTBR ASD model, beyond its face validity, is a useful tool to untangle neurotransmission alterations that could be underpinned to the heterogeneous ASD-like behaviours, highlighting the crucial role played by amygdala.


Assuntos
Transtorno do Espectro Autista , Transtorno Autístico , Camundongos , Animais , Humanos , Transtorno Autístico/metabolismo , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Acetilcolina , Dopamina , Fator de Crescimento Neural/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Transmissão Sináptica/fisiologia , Transtorno do Espectro Autista/metabolismo , Tonsila do Cerebelo/metabolismo , Ácido gama-Aminobutírico , Modelos Animais de Doenças
18.
Am J Physiol Endocrinol Metab ; 305(10): E1266-73, 2013 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-24064338

RESUMO

Oleoylethanolamide (OEA) is a gut-derived endogenous lipid that stimulates vagal fibers to induce satiety. Our previous work has shown that peripherally administered OEA activates c-fos transcription in the nucleus of the solitary tract (NST) and in the paraventricular nucleus (PVN), where it enhances oxytocin (OXY) expression. The anorexigenic action of OEA is prevented by the intracerebroventricular administration of a selective OXY receptor antagonist, suggesting a necessary role of OXYergic mediation of OEA's effect. The NST is the source of direct noradrenergic afferent input to hypothalamic OXY neurons, and therefore, we hypothesized that the activation of this pathway might mediate OEA effects on PVN neurons. To test this hypothesis, we subjected rats to intra-PVN administration of the toxin saporin (DSAP) conjugated to an antibody against dopamine-ß-hydroxylase (DBH) to destroy hindbrain noradrenergic neurons. In these rats we evaluated the effects of OEA (10 mg/kg, ip) on feeding behavior, on c-Fos and OXY immunoreactivity in the PVN, and on OXY immunoreactivity in the posterior pituitary gland. We found that the DSAP lesion completely prevented OEA's effects on food intake, on Fos and OXY expression in the PVN, and on OXY immunoreactivity of the posterior pituitary gland; all effects were maintained in sham-operated rats. These results support the hypothesis that noradrenergic NST-PVN projections are involved in the activation of the hypothalamic OXY system, which mediates OEA's prosatiety action.


Assuntos
Neurônios Adrenérgicos/efeitos dos fármacos , Ácidos Oleicos/farmacologia , Ocitocina/metabolismo , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Rombencéfalo/efeitos dos fármacos , Saciação/efeitos dos fármacos , Neurônios Adrenérgicos/fisiologia , Animais , Dopamina beta-Hidroxilase/imunologia , Endocanabinoides , Imunotoxinas/farmacologia , Masculino , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Núcleo Hipotalâmico Paraventricular/fisiologia , Ratos , Ratos Wistar , Rombencéfalo/fisiologia , Proteínas Inativadoras de Ribossomos Tipo 1/farmacologia , Saporinas
19.
Int J Neuropsychopharmacol ; 16(2): 323-38, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22310059

RESUMO

Agomelatine is a novel antidepressant acting as an MT1/MT2 melatonin receptor agonist/5-HT2C serotonin receptor antagonist. Because of its peculiar pharmacological profile, this drug caters the potential to correct the abnormalities of circadian rhythms associated with mood disorders, including abnormalities of the sleep/wake cycle. Here, we examined the effect of chronic agomelatine treatment on sleep architecture and circadian rhythms of motor activity using the rat model of prenatal restraint stress (PRS) as a putative 'aetiological' model of depression. PRS was delivered to the mothers during the last 10 d of pregnancy. The adult progeny ('PRS rats') showed a reduced duration of slow wave sleep, an increased duration of rapid eye movement (REM) sleep, an increased number of REM sleep events and an increase in motor activity before the beginning of the dark phase of the light/dark cycle. In addition, adult PRS rats showed an increased expression of the transcript of the primary response gene, c-Fos, in the hippocampus just prior to the beginning of the dark phase. All these changes were reversed by a chronic oral treatment with agomelatine (2000 ppm in the diet). The effect of agomelatine on sleep was largely attenuated by treatment with the MT1/MT2 melatonin receptor antagonist, S22153, which caused PRS-like sleep disturbances on its own. These data provide the first evidence that agomelatine corrects sleep architecture and restores circadian homeostasis in a preclinical model of depression and supports the value of agomelatine as a novel antidepressant that resynchronizes circadian rhythms under pathological conditions.


Assuntos
Acetamidas/uso terapêutico , Transtornos Cronobiológicos/tratamento farmacológico , Hipnóticos e Sedativos/uso terapêutico , Transtornos dos Movimentos/tratamento farmacológico , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia , Transtornos do Sono-Vigília/tratamento farmacológico , Análise de Variância , Animais , Animais Recém-Nascidos , Nível de Alerta/efeitos dos fármacos , Autorradiografia , Transtornos Cronobiológicos/etiologia , Modelos Animais de Doenças , Esquema de Medicação , Eletroencefalografia , Eletromiografia , Feminino , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Masculino , Transtornos dos Movimentos/etiologia , Gravidez , Efeitos Tardios da Exposição Pré-Natal/tratamento farmacológico , Efeitos Tardios da Exposição Pré-Natal/patologia , Proteínas Proto-Oncogênicas c-fos/genética , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Melatonina/antagonistas & inibidores , Restrição Física/efeitos adversos , Transtornos do Sono-Vigília/etiologia , Tiofenos/farmacologia
20.
Front Endocrinol (Lausanne) ; 14: 1158287, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37234803

RESUMO

Introduction: Oleoylethanolamide (OEA), an endogenous N-acylethanolamine acting as a gut-to-brain signal to control food intake and metabolism, has been attracting attention as a target for novel therapies against obesity and eating disorders. Numerous observations suggested that the OEA effects might be peripherally mediated, although they involve central pathways including noradrenergic, histaminergic and oxytocinergic systems of the brainstem and the hypothalamus. Whether these pathways are activated directly by OEA or whether they are downstream of afferent nerves is still highly debated. Some early studies suggested vagal afferent fibers as the main route, but our previous observations have contradicted this idea and led us to consider the blood circulation as an alternative way for OEA's central actions. Methods: To test this hypothesis, we first investigated the impact of subdiaphragmatic vagal deafferentation (SDA) on the OEA-induced activation of selected brain nuclei. Then, we analyzed the pattern of OEA distribution in plasma and brain at different time points after intraperitoneal administration in addition to measuring food intake. Results: Confirming and extending our previous findings that subdiaphragmatic vagal afferents are not necessary for the eating-inhibitory effect of exogenous OEA, our present results demonstrate that vagal sensory fibers are also not necessary for the neurochemical effects of OEA. Rather, within a few minutes after intraperitoneal administration, we found an increased concentration of intact OEA in different brain areas, associated with the inhibition of food intake. Conclusion: Our results support that systemic OEA rapidly reaches the brain via the circulation and inhibits eating by acting directly on selected brain nuclei.


Assuntos
Encéfalo , Ingestão de Alimentos , Ingestão de Alimentos/fisiologia , Encéfalo/metabolismo , Endocanabinoides/farmacologia , Endocanabinoides/metabolismo , Ácidos Oleicos/farmacologia , Ácidos Oleicos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA