5 research outputs found

    Comportements anxiodépressifs et motivation alimentaire en contexte d'obésité : impacts des acides gras saturés sur le noyau accumbens

    Full text link
    L'obésité augmente la susceptibilité aux troubles anxieux et de l’humeur et, à son tour, un affect négatif influence les comportements alimentaires. Faisant partie du circuit de la récompense, le noyau accumbens intègre la signalisation dopaminergique avec des signaux corticaux et traduit ces informations en comportements motivés. Chez le rongeur, une alimentation riche en gras favorise des adaptations dopaminergiques au noyau accumbens et des études d'imagerie révèlent un recrutement amoindri de cette région du cerveau lors de la consommation d’aliments palatables en obésité humaine. Les altérations dopaminergiques au noyau accumbens perturbent les fonctions de récompense et contribuent fortement aux déficits motivationnels couramment observés en dépression. L'obésité induite par la diète résulte d'une surconsommation chronique d'aliments à forte densité énergétique, tels que ceux riches en gras, mais des acides gras alimentaires distincts influencent différemment la santé métabolique et l'humeur. Alors que les acides gras monoinsaturés, prédominants dans le régime méditerranéen, améliorent les paramètres du syndrome métabolique, les acides gras saturés, enrichis en produits d'origine animale et en aliments transformés, ont des actions pro-inflammatoires et leur consommation est corrélée aux symptômes dépressifs chez l’humain. Ainsi, nous avons émis l'hypothèse que l'obésité induite par une diète riche en gras saturés, et non monoinsaturés, favorise l’expression de comportements anxieux et dépressifs chez la souris via des adaptations moléculaires au noyau accumbens. Afin de tester cette hypothèse, nous avons exposé des souris mâles et femelles à une diète riche en gras saturés ou monoinsaturés ou à une diète faible en gras et avons évalué le rôle de l'inflammation au noyau accumbens dans l'expression d'un phénotype anxiodépressif. Dans notre première étude, impliquant uniquement des souris mâles, nous avons constaté que, malgré une prise de poids et une adiposité similaires, seules les souris nourries avec une diète riche en gras saturés présentaient des altérations métaboliques et des comportements anxiodépressifs. De plus, ces effets étaient accompagnés d'une augmentation des niveaux de marqueurs inflammatoires dans le noyau accumbens et, par approche virale, l’inhibition spécifique de la voie pro-inflammatoire du facteur nucléaire kappa-b dans cette région était suffisante pour empêcher l'expression de comportements anxiodépressifs ainsi que de recherche compulsive de sucrose. Dans notre deuxième étude, des souris femelles ont été placées sur les mêmes 3 régimes et nous avons également observé un phénotype anxiodépressif spécifique à la diète riche en gras saturés. Contrairement aux mâles, l'inflammation n'était pas associée à l’expression de comportements anxiodépressifs. À la place, des niveaux d'œstrogènes circulants élevés et une expression diminuée du récepteur à l’estrogène bêta dans le noyau accumbens ont distingué les souris sous diète riche en gras saturés des autres. Dans notre troisième étude, nous avons induit l'obésité et des comportements anxiodépressifs chez des souris mâles en les exposant à la diète riche en gras saturés. Par une approche chimiogénétique, nous avons renversé le phénotype dépressif induit par la diète riche en gras saturés en activant les neurones du noyau accumbens exprimant le récepteur dopaminergique de type 1. De plus, nous avons mis en évidence des effets différentiels de ces neurones, en condition de diète riche en gras saturés, avec une hausse des comportements anxieux par cette même manipulation. Dans notre quatrième étude, nous évaluons le potentiel d'un nouveau co-agoniste glucagon-like peptide-1/dexaméthasone à réduire la motivation pour les aliments riches en gras et en sucre chez les souris mâles. Nous avons constaté que ce co-agoniste pouvait inhiber, de manière aiguë, la motivation à obtenir des récompenses alimentaires à la fois sous diète contrôle et suite au sevrage d’une diète riche en gras et en sucre. Le traitement prolongé avec ce composé chez des souris obèses a permis de réduire le poids corporel et l'apport alimentaire, sans favoriser de comportements anxiodépressifs ou de déficit cognitif. Dans l'ensemble, notre travail supporte le rôle du noyau accumbens dans la susceptibilité accrue à l'anxiété et à la dépression chez les personnes obèses. Nous démontrons que la composition du régime alimentaire en différents acides gras, et non seulement la teneur, influence les altérations métaboliques et de l'humeur en obésité. Nos données suggèrent que cette relation est gouvernée par des mécanismes moléculaires distincts en fonction du sexe. Enfin, ces études pourraient orienter le développement de nouvelles approches thérapeutiques traitant à la fois les composantes métaboliques et motivationnelles de l'obésité.Obesity increases the odds for mood disorders such as depression and anxiety and, in turn, negative affect influences feeding behaviours. Part of the reward circuitry, the nucleus accumbens integrates dopaminergic signaling with cortical inputs and translates this information into goal-oriented behaviours. In rodents, high-fat feeding promotes dopaminergic adaptations in the nucleus accumbens and imaging studies reveal blunted recruitment of this brain region during palatable food consumption in human obesity. Alterations in nucleus accumbens dopamine signaling disrupt reward function and heavily contribute to the motivational deficits commonly observed in depression. Diet-induced obesity results from chronic overconsumption of energy-dense foods, such as those with a high fat content, yet distinct dietary fatty acids influence metabolic health and mood differently. While monounsaturated fatty acids, predominant in the Mediterranean diet, have overall benefits for features of the metabolic syndrome, saturated fatty acids, enriched in animal-derived products and processed foods, have pro-inflammatory actions and their consumption correlates with depressive symptoms. Thus, we hypothesized that obesity induced by a saturated, but not monounsaturated, high-fat diet promotes anxiety and depressive behaviours in mice via molecular adaptations in the nucleus accumbens. In order to test this hypothesis, we placed male and female mice on either a saturated or monounsaturated high-fat diet or a control low-fat diet and assessed the role of nucleus accumbens inflammation in mediating the expression of an anxiodepressive phenotype. In our first study, only involving male mice, we found that, despite similar weight gain and overall adiposity, only mice fed the saturated high-fat diet displayed metabolic impairments and anxiodepressive behaviours. In addition, these impairments were accompanied by enhanced levels of inflammatory markers in the nucleus accumbens and region-specific viral mediated inhibition of the pro-inflammatory nuclear factor kappa-b pathway was sufficient to prevent the expression of anxiodepressive behaviours as well as compulsive sucrose-seeking. In our second study, female mice were placed on the same 3 diets and we also observed an anxiodepressive phenotype specific to the saturated high-fat diet. In contrast to the males, inflammation was not associated to the increase in anxiodepressive behaviours. Instead, elevated circulating levels of estrogen and diminished expression of estrogen receptor beta in the nucleus accumbens distinguished mice on the saturated high-fat diet from the others. In our third study, we induced obesity and anxiodepressive behaviours in male mice by exposing them to the saturated high-fat diet. Via a chemogenetic approach, we blocked the depressive phenotype induced by saturated high-fat feeding by activating neurons of the nucleus accumbens expressing the type 1 dopamine receptor. In addition, we evidenced differential effects of these neurons, under conditions of saturated high-fat feeding, as anxiety behaviours were enhanced by this same manipulation. In our fourth study, we assess the potential of a novel glucagon-like peptide-1/dexamethasone co-agonist to reduce motivation for high-fat/high-sugar rewards in male mice. We found that this co-agonist could acutely inhibit operant response for palatable foods under both chow and withdrawal from high-fat/high-sucrose diet conditions. Prolonged treatment with this compound in diet-induced obese mice successfully reduced body weight and food intake, without promoting anxiodepressive behaviours as well as cognitive impairment. Overall, our work supports a role for the nucleus accumbens in the greater susceptibility to anxiety and depression in obese individuals. We demonstrate that diet composition in fatty acids, and not just fat content, influences metabolic and mood impairments in obesity. Our data suggests this relationship to be mediating by distinct molecular mechanisms in a sex-specific manner. Finally, these studies may orient the development of novel therapeutic approaches addressing both the metabolic and motivational components of obesity

    Nucleus accumbens inflammation mediates anxiodepressive behavior and compulsive sucrose seeking elicited by saturated dietary fat

    No full text
    Objective: The incidence of depression is significantly compounded by obesity. Obesity arising from excessive intake of high-fat food provokes anxiodepressive behavior and elicits molecular adaptations in the nucleus accumbens (NAc), a region well-implicated in the hedonic deficits associated with depression and in the control of food-motivated behavior. To determine the etiology of diet-induced depression, we studied the impact of different dietary lipids on anxiodepressive behavior and metabolic and immune outcomes and the contribution of NAc immune activity. Methods: Adult C57Bl/6 mice were subjected to isocaloric high-fat/high-sucrose diets (HFD), enriched in either saturated or monounsaturated fat, or a control low-fat diet (LFD). Metabolic responses, anxiodepressive behavior, and plasma and NAc inflammatory markers were assessed after 12 weeks. In separate experiments, an adenoviral construct inhibiting IKKβ, an upstream component of the nuclear factor kappa-b (NFkB) pathway, was a priori injected into the NAc. Results: Both HFDs resulted in obesity and hyperleptinemia; however, the saturated HFD uniquely triggered anxiety-like behavior, behavioral despair, hyperinsulinemia, glucose intolerance, peripheral inflammation, and multiple pro-inflammatory signs in the NAc, including reactive gliosis, increased expression of cytokines, antigen-presenting markers and NFкB transcriptional activity. Selective NAc IKKβ inhibition reversed the upregulated expression of inflammatory markers, prevented anxiodepressive behavior and blunted compulsive sucrose-seeking in mice fed the saturated HFD. Conclusions: Metabolic inflammation and NFкB-mediated neuroinflammatory responses in the NAc contribute to the expression of anxiodepressive behavior and heightened food cravings caused by a diet high in saturated fat and sugar. Keywords: Diet-induced obesity, Dietary fatty acids, Nuclear factor kappa-b, Neuroinflammation, Depression, Anxiety, Food rewar

    α/β-Hydrolase Domain 6 in the Ventromedial Hypothalamus Controls Energy Metabolism Flexibility

    Get PDF
    Summary: α/β-Hydrolase domain 6 (ABHD6) is a monoacylglycerol hydrolase that degrades the endocannabinoid 2-arachidonoylglycerol (2-AG). Although complete or peripheral ABHD6 loss of function is protective against diet-induced obesity and insulin resistance, the role of ABHD6 in the central control of energy balance is unknown. Using a viral-mediated knockout approach, targeted endocannabinoid measures, and pharmacology, we discovered that mice lacking ABHD6 from neurons of the ventromedial hypothalamus (VMHKO) have higher VMH 2-AG levels in conditions of endocannabinoid recruitment and fail to physiologically adapt to key metabolic challenges. VMHKO mice exhibited blunted fasting-induced feeding and reduced food intake, energy expenditure, and adaptive thermogenesis in response to cold exposure, high-fat feeding, and dieting (transition to a low-fat diet). Our findings identify ABHD6 as a regulator of the counter-regulatory responses to major metabolic shifts, including fasting, nutrient excess, cold, and dieting, thereby highlighting the importance of ABHD6 in the VMH in mediating energy metabolism flexibility. : Fisette et al. demonstrate that the endocannabinoid-degrading enzyme ABHD6 in ventromedial hypothalamus neurons is a regulator of VMH 2-AG accumulation and energy metabolism flexibility in response to homeostatic challenges. ABHD6 knockout in VMH neurons of adult mice impairs the feeding response to fasting and increases susceptibility to cold-induced hypothermia, diet-induced obesity, and resistance to diet-induced weight loss. Keywords: endocannabinoids, 2-arachidonoylglycerol, cannabinoid receptor 1, α/β-hydrolase-domain 6, ventromedial hypothalamus, energy homeostasis, thermogenesis, obesit

    Early Life Low-Calorie Sweetener Consumption Impacts Energy Balance during Adulthood

    No full text
    Children frequently consume beverages that are either sweetened with sugars (sugar-sweetened beverages; SSB) or low-calorie sweeteners (LCS). Here, we evaluated the effects of habitual early life consumption of either SSB or LCS on energy balance later during adulthood. Male and female rats were provided with chow, water, and a solution containing either SSB (sucrose), LCS (acesulfame potassium (ACE-K) or stevia), or control (no solution) during the juvenile and adolescent periods (postnatal days 26–70). SSB or LCS consumption was voluntary and restricted within the recommended federal daily limits. When subsequently maintained on a cafeteria-style junk food diet (CAF; various high-fat, high-sugar foods) during adulthood, ACE-K-exposed rats demonstrated reduced caloric consumption vs. the controls, which contributed to lower body weights in female, but not male, ACE-K rats. These discrepant intakes and body weight effects in male ACE-K rats are likely to be based on reduced gene expression of thermogenic indicators (UCP1, BMP8B) in brown adipose tissue. Female stevia-exposed rats did not differ from the controls in terms of caloric intake or body weight, yet they consumed more SSB during CAF exposure in adulthood. None of the SSB-exposed rats, neither male nor female, differed from the controls in terms of total adult caloric consumption or body weight measures. The collective results reveal that early life LCS consumption alters sugar preference, body weight, and gene expression for markers of thermogenesis during adulthood, with both sex- and sweetener-dependent effects

    Hypothalamic melanin-concentrating hormone neurons integrate food-motivated appetitive and consummatory processes in rats

    No full text
    Food intake is determined by learned appetitive responses and physiological “appetition” signals after eating begins. Here, authors show melanin-concentrating hormone (MCH)-producing neurons integrate these processes to promote caloric intake
    corecore