Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
Add more filters










Publication year range
1.
J Neuroinflammation ; 20(1): 302, 2023 Dec 19.
Article in English | MEDLINE | ID: mdl-38111048

ABSTRACT

G protein-coupled receptor 120 (GPR120, Ffar4) is a sensor for long-chain fatty acids including omega-3 polyunsaturated fatty acids (n-3 PUFAs) known for beneficial effects on inflammation, metabolism, and mood. GPR120 mediates the anti-inflammatory and insulin-sensitizing effects of n-3 PUFAs in peripheral tissues. The aim of this study was to determine the impact of GPR120 stimulation on microglial reactivity, neuroinflammation and sickness- and anxiety-like behaviors by acute proinflammatory insults. We found GPR120 mRNA to be enriched in  both murine and human microglia, and in situ hybridization revealed GPR120 expression in microglia of the nucleus accumbens (NAc) in mice. In a manner similar to or exceeding n-3 PUFAs, GPR120 agonism (Compound A, CpdA) strongly attenuated lipopolysaccharide (LPS)-induced proinflammatory marker expression in primary mouse microglia, including tumor necrosis factor-α (TNF-α) and interleukin-1ß (IL-1ß), and inhibited nuclear factor-ĸB translocation to the nucleus. Central administration of CpdA to adult mice blunted LPS-induced hypolocomotion and anxiety-like behavior and reduced TNF-α, IL-1ß and IBA-1 (microglia marker) mRNA in the NAc, a brain region modulating anxiety and motivation and implicated in neuroinflammation-induced mood deficits. GPR120 agonist pre-treatment attenuated NAc microglia reactivity and alleviated sickness-like behaviors elicited by central injection TNF-α and IL-1ß. These findings suggest that microglial GPR120 contributes to neuroimmune regulation and behavioral changes in response to acute infection and elevated brain cytokines. GPR120 may participate in the protective action of n-3 PUFAs at the neural and behavioral level and offers potential as treatment target for neuroinflammatory conditions.


Subject(s)
Fatty Acids, Omega-3 , Microglia , Receptors, G-Protein-Coupled , Adult , Animals , Humans , Mice , Anxiety/chemically induced , Anxiety/drug therapy , Fatty Acids/metabolism , Fatty Acids, Omega-3/pharmacology , Inflammation/metabolism , Lipopolysaccharides/toxicity , Microglia/metabolism , Neuroinflammatory Diseases , RNA, Messenger/metabolism , Tumor Necrosis Factor-alpha/metabolism , Receptors, G-Protein-Coupled/metabolism
2.
Cell Rep ; 42(5): 112485, 2023 05 30.
Article in English | MEDLINE | ID: mdl-37149866

ABSTRACT

Neurovascular abnormalities in mouse models of 16p11.2 deletion autism syndrome are reminiscent of alterations reported in murine models of glucose transporter deficiency, including reduced brain angiogenesis and behavioral alterations. Yet, whether cerebrovascular alterations in 16p11.2df/+ mice affect brain metabolism is unknown. Here, we report that anesthetized 16p11.2df/+ mice display elevated brain glucose uptake, a phenomenon recapitulated in mice with endothelial-specific 16p11.2 haplodeficiency. Awake 16p11.2df/+ mice display attenuated relative fluctuations of extracellular brain glucose following systemic glucose administration. Targeted metabolomics on cerebral cortex extracts reveals enhanced metabolic responses to systemic glucose in 16p11.2df/+ mice that also display reduced mitochondria number in brain endothelial cells. This is not associated with changes in mitochondria fusion or fission proteins, but 16p11.2df/+ brain endothelial cells lack the splice variant NT-PGC-1α, suggesting defective mitochondrial biogenesis. We propose that altered brain metabolism in 16p11.2df/+ mice is compensatory to endothelial dysfunction, shedding light on previously unknown adaptative responses.


Subject(s)
Endothelial Cells , Haploinsufficiency , Mice , Animals , Endothelial Cells/metabolism , Organelle Biogenesis , Chromosome Deletion , Brain
3.
J Neuroendocrinol ; 34(12): e13218, 2022 12.
Article in English | MEDLINE | ID: mdl-36471907

ABSTRACT

Acyl-CoA binding protein (ACBP), also known as diazepam binding inhibitor (DBI), has recently emerged as a hypothalamic and brainstem gliopeptide regulating energy balance. Previous work has shown that the ACBP-derived octadecaneuropeptide exerts strong anorectic action via proopiomelanocortin (POMC) neuron activation and the melanocortin-4 receptor. Importantly, targeted ACBP loss-of-function in astrocytes promotes hyperphagia and diet-induced obesity while its overexpression in arcuate astrocytes reduces feeding and body weight. Despite this knowledge, the role of astroglial ACBP in adaptive feeding and metabolic responses to acute metabolic challenges has not been investigated. Using different paradigms, we found that ACBP deletion in glial fibrillary acidic protein (GFAP)-positive astrocytes does not affect weight loss when obese male mice are transitioned from a high fat diet to a chow diet, nor metabolic parameters in mice fed with a normal chow diet (e.g., energy expenditure, body temperature) during fasting, cold exposure and at thermoneutrality. In contrast, astroglial ACBP deletion impairs meal pattern and feeding responses during refeeding after a fast and during cold exposure, thereby showing that ACBP is required to stimulate feeding in states of increased energy demand. These findings challenge the general view that astroglial ACBP exerts anorectic effects and suggest that regulation of feeding by ACBP is dependent on metabolic status.


Subject(s)
Appetite Depressants , Diazepam Binding Inhibitor , Energy Metabolism , Animals , Male , Mice , Astrocytes/metabolism , Diazepam Binding Inhibitor/metabolism , Energy Metabolism/physiology , Hyperphagia/metabolism
4.
Diabetes ; 70(12): 2850-2859, 2021 12.
Article in English | MEDLINE | ID: mdl-34610983

ABSTRACT

Controllable genetic manipulation is an indispensable tool in research, greatly advancing our understanding of cell biology and physiology. However in ß-cells, transgene silencing, low inducibility, ectopic expression, and off-targets effects are persistent challenges. In this study, we investigated whether an inducible Tetracycline (Tet)-Off system with ß-cell-specific mouse insulin promoter (MIP)-itTA-driven expression of tetracycline operon (TetO)-CreJaw/J could circumvent previous issues of specificity and efficacy. Following assessment of tissue-specific gene recombination, ß-cell architecture, in vitro and in vivo glucose-stimulated insulin secretion, and whole-body glucose homeostasis, we discovered that expression of any tetracycline-controlled transactivator (e.g., improved itTA, reverse rtTA, or tTA) in ß-cells significantly reduced Insulin gene expression and decreased insulin content. This translated into lower pancreatic insulin levels and reduced insulin secretion in mice carrying any tTA transgene, independent of Cre recombinase expression or doxycycline exposure. Our study echoes ongoing challenges faced by fundamental researchers working with ß-cells and highlights the need for consistent and comprehensive controls when using the tetracycline-controlled transactivator systems (Tet-On or Tet-Off) for genome editing.


Subject(s)
Insulin-Secreting Cells/metabolism , Insulin/genetics , Insulin/metabolism , Animals , Cells, Cultured , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Integrases/genetics , Integrases/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/genetics , Tetracycline/pharmacology , Trans-Activators/drug effects , Trans-Activators/genetics , Transgenes/drug effects
5.
Trends Endocrinol Metab ; 32(11): 890-903, 2021 11.
Article in English | MEDLINE | ID: mdl-34565656

ABSTRACT

Four decades ago Costa and colleagues identified a small, secreted polypeptide in the brain that can displace the benzodiazepine diazepam from the GABAA receptor, and was thus termed diazepam binding inhibitor (DBI). Shortly after, an identical polypeptide was identified in liver by its ability to induce termination of fatty acid synthesis, and was named acyl-CoA binding protein (ACBP). Since then, ACBP/DBI has been studied in parallel without a clear and integrated understanding of its dual roles. The first genetic loss-of-function models have revived the field, allowing targeted approaches to better understand the physiological roles of ACBP/DBI in vivo. We discuss the roles of ACBP/DBI in central and tissue-specific functions in mammals, with an emphasis on metabolism and mechanisms of action.


Subject(s)
Benzodiazepines , Fatty Acids , Animals , Humans , Benzodiazepines/pharmacology , Diazepam Binding Inhibitor/genetics , Diazepam Binding Inhibitor/metabolism , Fatty Acids/metabolism , Mammals/metabolism
6.
Sci Rep ; 11(1): 15767, 2021 08 03.
Article in English | MEDLINE | ID: mdl-34344941

ABSTRACT

The beneficial effects of brown adipose tissue (BAT) on obesity and associated metabolic diseases are mediated through its capacity to dissipate energy as heat. While immune cells, such as tissue-resident macrophages, are known to influence adipose tissue homeostasis, relatively little is known about their contribution to BAT function. Here we report that neuropilin-1 (NRP1), a multiligand single-pass transmembrane receptor, is highly expressed in BAT-resident macrophages. During diet-induced obesity (DIO), myeloid-resident NRP1 influences interscapular BAT mass, and consequently vascular morphology, innervation density and ultimately core body temperature during cold exposure. Thus, NRP1-expressing myeloid cells contribute to the BAT homeostasis and potentially its thermogenic function in DIO.


Subject(s)
Adipose Tissue, Brown/physiology , Homeostasis , Myeloid Cells/metabolism , Neuropilin-1/physiology , Obesity/prevention & control , Thermogenesis , Animals , Diet/adverse effects , Energy Metabolism , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/etiology , Obesity/metabolism , Obesity/pathology
7.
eNeuro ; 8(2)2021.
Article in English | MEDLINE | ID: mdl-33820803

ABSTRACT

Prevalence of metabolic disturbances is higher among individuals with neurodevelopmental disorders (NDDs), yet this association has been largely overlooked. Investigation into human disease remains challenging, as a complete pathophysiological understanding relies on accurate modeling and highly controlled variables. Genetically engineered mouse models are widely used to gain insight into the biology of human NDDs, but research focus has been on behavioral and neurophysiological abnormalities. Such models not only allow for evaluating usefulness in reproducing human features, including similarities and discrepancies with rodent phenotypes, but they also represent a unique opportunity to observe and quantify novel anomalies. Here, we present the first characterization and comparison of basal metabolism in three mouse models of NDDs, namely, Down syndrome (DS; Dp(16)Yey/+ mice), 16p11.2 deletion syndrome (16pDel; 16p11.2df/+ mice), and fragile X syndrome [FXS; Fmr1 knock-out (KO) mice] and their wild-type (WT) counterparts. Using the Comprehensive Lab Animal Monitoring System (CLAMS) coupled to EchoMRI, as well as quantification of key plasma metabolites by liquid chromatography mass spectrometry (LC-MS), our in vivo study reveals that each mouse model expresses a unique metabolic signature that is sex-specific, independent of the amount of food consumed and minimally influenced by physical activity. In particular, we identify striking differences in body composition, respiratory exchange ratio (RER), caloric expenditure (CE), and concentrations of circulating plasma metabolites related to mitochondrial function. Providing novel insight into NDD-associated metabolic alterations is an essential prerequisite for future preclinical and clinical interventions.


Subject(s)
Fragile X Syndrome , Neurodevelopmental Disorders , Animals , Basal Metabolism , Disease Models, Animal , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/genetics , Humans , Mice , Mice, Knockout
8.
Front Immunol ; 11: 1953, 2020.
Article in English | MEDLINE | ID: mdl-32973799

ABSTRACT

Obesity is a major risk factor for the development of insulin resistance and type 2 diabetes. However, the mechanisms that trigger the underlying adipose tissues inflammation are not completely understood. Here, we show that the E3 ubiquitin ligase March1 controls the phenotypic and functional properties of CD8+ T cells in mice white adipose tissue. In a diet-induced obesity model, mice lacking March1 [March1 knockout (KO)] show increased insulin resistance compared to their WT counterparts. Also, in obese March1 KO mice, the proportions of effector/memory (Tem) and resident/memory (Trm) CD8+ T cells were higher in the visceral adipose tissue, but not in the spleen. The effect of March1 on insulin resistance and on the phenotype of adipose tissue CD8+ T cells was independent of major histocompatibility complex class II ubiquitination. Interestingly, we adoptively transferred either WT or March1 KO splenic CD8+ T cells into obese WT chimeras that had been reconstituted with Rag1-deficient bone marrow. We observed an enrichment of Tem and Trm cells and exacerbated insulin resistance in mice that received March1 KO CD8 T cells. Mechanistically, we found that March1 deficiency alters the metabolic activity of CD8+ T cells. Our results provide additional evidence of the involvement of CD8+ T cells in adipose tissue inflammation and suggest that March1 controls the metabolic reprogramming of these cells.


Subject(s)
Adipose Tissue, White/enzymology , CD8-Positive T-Lymphocytes/metabolism , Immunologic Memory , Insulin Resistance , Obesity/enzymology , Ubiquitin-Protein Ligases/deficiency , Adipose Tissue, White/immunology , Adoptive Transfer , Animals , Blood Glucose/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/transplantation , Diet, High-Fat , Disease Models, Animal , Energy Metabolism , Lymphocyte Activation , Mice, Inbred C57BL , Mice, Knockout , Obesity/genetics , Obesity/immunology , Phenotype , Spleen/enzymology , Spleen/immunology , Ubiquitin-Protein Ligases/genetics
9.
Int J Obes (Lond) ; 44(9): 1936-1945, 2020 09.
Article in English | MEDLINE | ID: mdl-32546855

ABSTRACT

OBJECTIVE: Obesity significantly elevates the odds of developing mood disorders. Chronic consumption of a saturated high-fat diet (HFD) elicits anxiodepressive behavior in a manner linked to metabolic dysfunction and neuroinflammation in mice. Dietary omega-3 polyunsaturated fatty acids (n-3 PUFA) can improve both metabolic and mood impairments by relieving inflammation. Despite these findings, the effects of n-3 PUFA supplementation on energy homeostasis, anxiodepressive behavior, brain lipid composition, and gliosis in the diet-induced obese state are unclear. METHODS: Male C57Bl/6J mice were fed a saturated high-fat diet (HFD) or chow for 20 weeks. During the last 5 weeks mice received daily gavage ("supplementation") of fish oil (FO) enriched with equal amounts of docosahexaenoic (DHA) and eicosapentaenoic acid (EPA) or control corn oil. Food intake and body weight were measured throughout while additional metabolic parameters and anxiety- and despair-like behavior (elevated-plus maze, light-dark box, and forced swim tasks) were evaluated during the final week of supplementation. Forebrain lipid composition and markers of microglia activation and astrogliosis were assessed by gas chromatography-mass spectrometry and real-time PCR, respectively. RESULTS: Five weeks of FO supplementation corrected glucose intolerance and attenuated hyperphagia in HFD-induced obese mice without affecting adipose mass. FO supplementation also defended against the anxiogenic and depressive-like effects of HFD. Brain lipids, particularly anti-inflammatory PUFA, were diminished by HFD, whereas FO restored levels beyond control values. Gene expression markers of brain reactive gliosis were supressed by FO. CONCLUSIONS: Supplementing a saturated HFD with FO rich in EPA and DHA corrects glucose intolerance, inhibits food intake, suppresses anxiodepressive behaviors, enhances anti-inflammatory brain lipids, and dampens indices of brain gliosis in obese mice. Together, these findings support increasing dietary n-3 PUFA for the treatment of metabolic and mood disturbances associated with excess fat intake and obesity.


Subject(s)
Behavior, Animal/drug effects , Brain , Diet, High-Fat/adverse effects , Fish Oils/pharmacology , Obesity , Adipose Tissue/drug effects , Animals , Anxiety , Brain/drug effects , Brain/metabolism , Brain Chemistry/drug effects , Depression , Dietary Supplements , Male , Mice , Mice, Inbred C57BL , Obesity/metabolism , Obesity/psychology
10.
Ultrasound Med Biol ; 46(7): 1715-1726, 2020 07.
Article in English | MEDLINE | ID: mdl-32381381

ABSTRACT

Shear wave elastography (speed and dispersion), local attenuation coefficient slope and homodyned-K parametric imaging were used for liver steatosis grading. These ultrasound biomarkers rely on physical interactions between shear and compression waves with tissues at both macroscopic and microscopic scales. These techniques were applied in a context not yet studied with ultrasound imaging, that is, monitoring steatosis of force-fed duck livers from pre-force-fed to foie gras stages. Each estimated feature presented a statistically significant trend along the feeding process (p values <10-3). However, whereas a monotonic increase in the shear wave speed was observed along the process, most quantitative ultrasound features exhibited an absolute maximum value halfway through the process. As the liver fat fraction in foie gras is much higher than that seen clinically, we hypothesized that a change in the ultrasound scattering regime is encountered for high-fat fractions, and consequently, care has to be taken when applying ultrasound biomarkers to grading of severe states of steatosis.


Subject(s)
Ducks , Elasticity Imaging Techniques , Liver/diagnostic imaging , Ultrasonography , Animals , Elasticity Imaging Techniques/methods , Elasticity Imaging Techniques/veterinary , Enteral Nutrition/veterinary , Fatty Liver/diagnostic imaging , Fatty Liver/veterinary , Liver/anatomy & histology , Ultrasonography/methods , Ultrasonography/veterinary
11.
Diabetologia ; 63(4): 673-682, 2020 04.
Article in English | MEDLINE | ID: mdl-32030470

ABSTRACT

The appropriate utilisation, storage and conversion of nutrients in peripheral tissues, referred to as nutrient partitioning, is a fundamental process to adapt to nutritional and metabolic challenges and is thus critical for the maintenance of a healthy energy balance. Alterations in this process during nutrient excess can have deleterious effects on glucose and lipid homeostasis and contribute to the development of obesity and type 2 diabetes. Nutrient partitioning is a complex integrated process under the control of hormonal and neural signals. Neural control relies on the capacity of the brain to sense circulating metabolic signals and mount adaptive neuroendocrine and autonomic responses. This review aims to discuss the hypothalamic neurocircuits and molecular mechanisms controlling nutrient partitioning and their potential contribution to metabolic maladaptation and disease.


Subject(s)
Energy Metabolism/physiology , Neurons/physiology , Nutrients/metabolism , Animals , Carbohydrate Metabolism/physiology , Diabetes Mellitus, Type 2/etiology , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/physiopathology , Glucose/metabolism , Homeostasis/physiology , Humans , Hypothalamus/physiology , Lipid Metabolism/physiology , Nerve Net/physiology , Nutrients/chemistry , Obesity/etiology , Obesity/metabolism , Obesity/physiopathology
12.
Am J Physiol Endocrinol Metab ; 317(2): E234-E243, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31013146

ABSTRACT

The pancreatic ß-cell responds to changes in the nutrient environment to maintain glucose homeostasis by adapting its function and mass. Nutrients can act directly on the ß-cell and also indirectly through the brain via autonomic nerves innervating islets. Despite the importance of the brain-islet axis in insulin secretion, relatively little is known regarding its involvement in ß-cell proliferation. We previously demonstrated that prolonged infusions of nutrients in rats provoke a dramatic increase in ß-cell proliferation in part because of the direct action of nutrients. Here, we addressed the contribution of the autonomic nervous system. In isolated islets, muscarinic stimulation increased, whereas adrenergic stimulation decreased, glucose-induced ß-cell proliferation. Blocking α-adrenergic receptors reversed the effect of epinephrine on glucose + nonesterified fatty acids (NEFA)-induced ß-cell proliferation, whereas activation of ß-adrenergic receptors was without effect. Infusion of glucose + NEFA toward the brain stimulated ß-cell proliferation, and this effect was abrogated following celiac vagotomy. The increase in ß-cell proliferation following peripheral infusions of glucose + NEFA was not inhibited by vagotomy or atropine treatment but was blocked by coinfusion of epinephrine. We conclude that ß-cell proliferation is stimulated by parasympathetic and inhibited by sympathetic signals. Whereas glucose + NEFA in the brain stimulates ß-cell proliferation through the vagus nerve, ß-cell proliferation in response to systemic nutrient excess does not involve parasympathetic signals but may be associated with decreased sympathetic tone.


Subject(s)
Autonomic Nervous System/physiology , Cell Proliferation , Insulin-Secreting Cells/physiology , Aging/physiology , Animals , Autonomic Nervous System/drug effects , Blood Glucose/metabolism , Carbachol/pharmacology , Cell Proliferation/drug effects , Cells, Cultured , Epinephrine/pharmacology , Fatty Acids, Nonesterified/pharmacology , Glucose/pharmacology , Insulin Secretion/drug effects , Insulin-Secreting Cells/drug effects , Male , Rats , Rats, Inbred Lew
13.
J Clin Invest ; 129(6): 2417-2430, 2019 04 02.
Article in English | MEDLINE | ID: mdl-30938715

ABSTRACT

Glial cells have emerged as key players in the central control of energy balance and etiology of obesity. Astrocytes play a central role in neural communication via the release of gliotransmitters. Acyl-CoA binding protein (ACBP)-derived endozepines are secreted peptides that modulate the GABAA receptor. In the hypothalamus, ACBP is enriched in arcuate nucleus (ARC) astrocytes, ependymocytes and tanycytes. Central administration of the endozepine octadecaneuropeptide (ODN) reduces feeding and improves glucose tolerance, yet the contribution of endogenous ACBP in energy homeostasis is unknown. We demonstrated that ACBP deletion in GFAP+ astrocytes, but not in Nkx2.1-lineage neural cells, promoted diet-induced hyperphagia and obesity in both male and female mice, an effect prevented by viral rescue of ACBP in ARC astrocytes. ACBP-astrocytes were observed in apposition with proopiomelanocortin (POMC) neurons and ODN selectively activated POMC neurons through the ODN-GPCR but not GABAA, and supressed feeding while increasing carbohydrate utilization via the melanocortin system. Similarly, ACBP overexpression in ARC astrocytes reduced feeding and weight gain. Finally, the ODN-GPCR agonist decreased feeding and promoted weight loss in ob/ob mice. These findings uncover ACBP as an ARC gliopeptide playing a key role in energy balance control and exerting strong anorectic effects via the central melanocortin system.


Subject(s)
Astrocytes/metabolism , Diazepam Binding Inhibitor/metabolism , Eating , Energy Metabolism , Hyperphagia/metabolism , Obesity/metabolism , Pro-Opiomelanocortin/metabolism , Animals , Astrocytes/pathology , Cell Line , Diazepam Binding Inhibitor/genetics , Female , Hyperphagia/genetics , Hyperphagia/pathology , Male , Mice , Mice, Knockout , Neurons/metabolism , Neurons/pathology , Obesity/genetics , Obesity/pathology , Pro-Opiomelanocortin/genetics
15.
Mol Metab ; 10: 1-13, 2018 04.
Article in English | MEDLINE | ID: mdl-29454579

ABSTRACT

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 IKKß, 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 IKKß 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.


Subject(s)
Anxiety Disorders/metabolism , Depressive Disorder/metabolism , Food Addiction/metabolism , Nucleus Accumbens/metabolism , Animals , Anxiety Disorders/etiology , Anxiety Disorders/physiopathology , Depressive Disorder/etiology , Depressive Disorder/physiopathology , Diet, High-Fat/adverse effects , Dietary Sucrose/adverse effects , Food Addiction/etiology , Food Addiction/physiopathology , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Inflammation/metabolism , Male , Mice , Mice, Inbred C57BL , Nucleus Accumbens/pathology
16.
Neuropsychopharmacology ; 43(3): 607-616, 2018 02.
Article in English | MEDLINE | ID: mdl-28857071

ABSTRACT

Long-chain fatty acids (FAs) act centrally to decrease food intake and hepatic glucose production and alter hypothalamic neuronal activity in a manner that depends on FA type and cellular transport proteins. However, it is not known whether FAs are sensed by ventral tegmental area (VTA) dopamine (DA) neurons to control food-motivated behavior and DA neurotransmission. We investigated the impact of the monounsaturated FA oleate in the VTA on feeding, locomotion, food reward, and DA neuronal activity and DA neuron expression of FA-handling proteins and FA uptake. A single intra-VTA injection of oleate, but not of the saturated FA palmitate, decreased food intake and increased locomotor activity. Furthermore, intra-VTA oleate blunted the rewarding effects of high-fat/sugar food in an operant task and inhibited DA neuronal firing. Using sorted DA neuron preparations from TH-eGFP mice we found that DA neurons express FA transporter and binding proteins, and are capable of intracellular transport of long-chain FA. Finally, we demonstrate that a transporter blocker attenuates FA uptake into DA neurons and blocks the effects of intra-VTA oleate to decrease food-seeking and DA neuronal activity. Together, these results suggest that DA neurons detect FA and that oleate has actions in the VTA to suppress DA neuronal activity and food seeking following cellular incorporation. These findings highlight the capacity of DA neurons to act as metabolic sensors by responding not only to hormones but also to FA nutrient signals to modulate food-directed behavior.


Subject(s)
Dopamine/metabolism , Eating/physiology , Feeding Behavior/physiology , Oleic Acid/metabolism , Reward , Ventral Tegmental Area/metabolism , Animals , Appetitive Behavior/physiology , Cells, Cultured , Conditioning, Operant/physiology , Dopaminergic Neurons/metabolism , Eating/psychology , Feeding Behavior/psychology , Male , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity/physiology , Rats, Wistar
17.
Diabetologia ; 61(3): 526-538, 2018 03.
Article in English | MEDLINE | ID: mdl-29143855

ABSTRACT

Mice are the most commonly used species in preclinical research on the pathophysiology of metabolic diseases. Although they are extremely useful for identifying pathways, mechanisms and genes regulating glucose and energy homeostasis, the specificities of the various mouse models and methodologies used to investigate a metabolic phenotype can have a profound impact on experimental results and their interpretation. This review aims to: (1) describe the most commonly used experimental tests to assess glucose and energy homeostasis in mice; (2) provide some guidelines regarding the design, analysis and interpretation of these tests, as well as for studies using genetic models; and (3) identify important caveats and confounding factors that must be taken into account in the interpretation of findings.


Subject(s)
Diabetes Mellitus/metabolism , Animals , Diabetes Mellitus/pathology , Disease Models, Animal , Energy Metabolism/physiology , Glucose/metabolism , Mice
18.
Psychoneuroendocrinology ; 83: 142-149, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28623763

ABSTRACT

Overconsumption of dietary fat can elicit impairments in emotional processes and the response to stress. While excess dietary lipids have been shown to alter hypothalamus-pituitary-adrenal (HPA) axis function and promote anxiety-like behaviour, it is not known if such changes rely on elevated body weight and if these effects are specific to the type of dietary fat. The objective of this study was to investigate the effect of a saturated and a monounsaturated high-fat diet (HFD) on HPA axis function and anxiety-like behaviour in rats. Biochemical, metabolic and behavioural responses were evaluated following eight weeks on one of three diets: (1) a monounsaturated HFD (50%kcal olive oil), (2) a saturated HFD (50%kcal palm oil), or (3) a control low-fat diet. Weight gain was similar across the three diets while visceral fat mass was elevated by the two HFDs. The saturated HFD had specific actions to increase peak plasma levels of corticosterone and tumour-necrosis-factor-alpha and suppress mRNA expression of glucocorticoid and mineralocorticoid receptors, corticotropin-releasing hormone and 11ß-hydroxysteroid dehydrogenase-1 in the paraventricular nucleus of the hypothalamus. Both HFDs enhanced the corticosterone-suppressing response to dexamethasone administration without affecting the physiological response to a restraint stress and failed to increase anxiety-like behaviour as measured in the elevated-plus maze and open field tests. These findings demonstrate that prolonged intake of saturated fat, without added weight gain, increases CORT and modulates central HPA feedback processes. That saturated HFD failed to affect anxiety-like behaviour can suggest that the anxiogenic effects of prolonged high-fat feeding may rely on more pronounced metabolic dysfunction.


Subject(s)
Anxiety/metabolism , Diet, High-Fat/adverse effects , Diet, High-Fat/psychology , Animals , Anxiety Disorders/metabolism , Body Weight , Corticosterone/analysis , Corticosterone/blood , Corticotropin-Releasing Hormone/metabolism , Dexamethasone/pharmacology , Dietary Fats/metabolism , Disease Models, Animal , Fatty Acids/adverse effects , Fatty Acids/metabolism , Fatty Acids, Monounsaturated/metabolism , Glucocorticoids/pharmacology , Hypothalamo-Hypophyseal System/metabolism , Male , Obesity/metabolism , Obesity/psychology , Paraventricular Hypothalamic Nucleus/metabolism , Pituitary-Adrenal System/metabolism , Rats , Rats, Wistar , Receptors, Corticotropin-Releasing Hormone/metabolism , Receptors, Glucocorticoid/metabolism , Tumor Necrosis Factor-alpha/analysis , Tumor Necrosis Factor-alpha/blood , Weight Gain
19.
Endocrinology ; 158(4): 903-919, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28324005

ABSTRACT

Oxidative stress induces endogenous antioxidants via nuclear factor erythroid 2-related factor 2 (Nrf2), potentially preventing tissue injury. We investigated whether insulin affects renal Nrf2 expression in type 1 diabetes (T1D) and studied its underlying mechanism. Insulin normalized hyperglycemia, hypertension, oxidative stress, and renal injury; inhibited renal Nrf2 and angiotensinogen (Agt) gene expression; and upregulated heterogeneous nuclear ribonucleoprotein F and K (hnRNP F and hnRNP K) expression in Akita mice with T1D. In immortalized rat renal proximal tubular cells, insulin suppressed Nrf2 and Agt but stimulated hnRNP F and hnRNP K gene transcription in high glucose via p44/42 mitogen-activated protein kinase signaling. Transfection with small interfering RNAs of p44/42 MAPK, hnRNP F, or hnRNP K blocked insulin inhibition of Nrf2 gene transcription. Insulin curbed Nrf2 promoter activity via a specific DNA-responsive element that binds hnRNP F/K, and hnRNP F/K overexpression curtailed Nrf2 promoter activity. In hyperinsulinemic-euglycemic mice, renal Nrf2 and Agt expression was downregulated, whereas hnRNP F/K expression was upregulated. Thus, the beneficial actions of insulin in diabetic nephropathy appear to be mediated, in part, by suppressing renal Nrf2 and Agt gene transcription and preventing Nrf2 stimulation of Agt expression via hnRNP F/K. These findings identify hnRNP F/K and Nrf2 as potential therapeutic targets in diabetes.


Subject(s)
Angiotensinogen/genetics , Diabetes Mellitus, Type 1/genetics , Heterogeneous-Nuclear Ribonucleoprotein Group F-H/genetics , Heterogeneous-Nuclear Ribonucleoprotein K/genetics , Insulin/pharmacology , NF-E2-Related Factor 2/genetics , Transcription, Genetic/drug effects , Angiotensinogen/metabolism , Animals , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/metabolism , Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/genetics , Diabetic Nephropathies/metabolism , Gene Expression/drug effects , Heterogeneous-Nuclear Ribonucleoprotein Group F-H/metabolism , Heterogeneous-Nuclear Ribonucleoprotein K/metabolism , Insulin/therapeutic use , Kidney/drug effects , Kidney/metabolism , Male , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinases/metabolism , NF-E2-Related Factor 2/metabolism , Promoter Regions, Genetic/drug effects , Signal Transduction/drug effects
20.
Cell Rep ; 17(5): 1217-1226, 2016 10 25.
Article in English | MEDLINE | ID: mdl-27783937

ABSTRACT

α/ß-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.


Subject(s)
Energy Metabolism , Hypothalamus/metabolism , Monoacylglycerol Lipases/metabolism , Animals , Arachidonic Acids/pharmacology , Cold Temperature , Diet, High-Fat , Endocannabinoids/pharmacology , Energy Metabolism/drug effects , Gene Deletion , Glycerides/pharmacology , Hypothalamus/drug effects , Mice, Inbred C57BL , Mice, Knockout , Neurons/drug effects , Neurons/metabolism , Obesity/metabolism , Obesity/pathology , Reproducibility of Results , Thermogenesis/drug effects , Weight Loss/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL