Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Front Endocrinol (Lausanne) ; 14: 1202089, 2023.
Article in English | MEDLINE | ID: mdl-37448468

ABSTRACT

Histamine is a biogenic amine that acts as a neuromodulator within the brain. In the hypothalamus, histaminergic signaling contributes to the regulation of numerous physiological and homeostatic processes, including the regulation of energy balance. Histaminergic neurons project extensively throughout the hypothalamus and two histamine receptors (H1R, H3R) are strongly expressed in key hypothalamic nuclei known to regulate energy homeostasis, including the paraventricular (PVH), ventromedial (VMH), dorsomedial (DMH), and arcuate (ARC) nuclei. The activation of different histamine receptors is associated with differential effects on neuronal activity, mediated by their different G protein-coupling. Consequently, activation of H1R has opposing effects on food intake to that of H3R: H1R activation suppresses food intake, while H3R activation mediates an orexigenic response. The central histaminergic system has been implicated in atypical antipsychotic-induced weight gain and has been proposed as a potential therapeutic target for the treatment of obesity. It has also been demonstrated to interact with other major regulators of energy homeostasis, including the central melanocortin system and the adipose-derived hormone leptin. However, the exact mechanisms by which the histaminergic system contributes to the modification of these satiety signals remain underexplored. The present review focuses on recent advances in our understanding of the central histaminergic system's role in regulating feeding and highlights unanswered questions remaining in our knowledge of the functionality of this system.


Subject(s)
Hypothalamus , Obesity , Humans , Hypothalamus/physiology , Arcuate Nucleus of Hypothalamus , Brain , Eating
2.
Obesity (Silver Spring) ; 29(10): 1635-1649, 2021 10.
Article in English | MEDLINE | ID: mdl-34449134

ABSTRACT

OBJECTIVE: This study investigated the effects of a low-dose salmon peptide fraction (SPF) and vitamin D3 (VitD3 ) in obese and VitD3 -deficient mice at risk of metabolic syndrome (MetS). METHODS: Obese and VitD3 -deficient low-density lipoprotein receptor (LDLr)-/- /apolipoprotein B100 (ApoB)100/100 mice were treated with high-fat high-sucrose diets, with 25% of dietary proteins replaced by SPF or a nonfish protein mix (MP). The SPF and MP groups received a VitD3 -deficient diet or a supplementation of 15,000 IU of VitD3 per kilogram of diet. Glucose homeostasis, atherosclerosis, nonalcoholic fatty liver disease, and gut health were assessed. RESULTS: VitD3 supplementation increased plasma 25-hydroxyvitamin D to optimal status whereas the VitD3 -deficient diet maintained moderate deficiency. SPF-treated groups spent more energy and accumulated less visceral fat in association with an improved adipokine profile. SPF lowered homeostatic model assessment of insulin resistance compared with MP, suggesting that SPF can improve insulin sensitivity. SPF alone blunted hepatic and colonic inflammation, whereas VitD3 supplementation attenuated ileal inflammation. These effects were associated with changes in gut microbiota such as increased Mogibacterium and Muribaculaceae. CONCLUSIONS: SPF treatment improves MetS by modulating hepatic and gut inflammation along with gut microbiota, suggesting that SPF operates through a gut-liver axis. VitD3 supplementation has limited influence on MetS in this model.


Subject(s)
Insulin Resistance , Salmon , Animals , Diet, High-Fat/adverse effects , Liver , Mice , Mice, Inbred C57BL , Obesity , Peptides , Vitamin D/pharmacology
3.
J Nutr ; 151(5): 1175-1189, 2021 05 11.
Article in English | MEDLINE | ID: mdl-33851198

ABSTRACT

BACKGROUND: Cholecalciferol (D3) may improve inflammation, and thus provide protection from cardiometabolic diseases (CMD), although controversy remains. Omega-3 fatty acids (ω-3FA) may also prevent the development of CMD, but the combined effects of ω-3FA and D3 are not fully understood. OBJECTIVES: We determined the chronic independent and combined effects of D3 and ω-3FA on body weight, glucose homeostasis, and markers of inflammation in obese mice. METHODS: We gave 8-week-old male C57BL/6J mice, which had been fed a high-fat, high-sucrose (HF) diet (65.5% kcal fat, 19.8% kcal carbohydrate, and 14% kcal protein) for 12 weeks, either a standard D3 dose (+SD3; 1400 IU D3/kg diet) or a high D3 dose (+HD3; 15,000 IU D3/kg diet). We fed 1 +SD3 group and 1 +HD3 group with 4.36% (w/w) fish oil (+ω-3FA; 44% eicosapentaenoic acid, 25% docosahexaenoic acid), and fed the other 2 groups with corn oil [+omega-6 fatty acids (ω-6FA)]. A fifth group was fed a low-fat (LF; 15.5% kcal) diet. LF and HF+ω-6+SD3 differences were tested by a Student's t-test and HF treatment differences were tested by a 2-way ANOVA. RESULTS: D3 supplementation in the +HD3 groups did not significantly increase plasma total 25-hydroxyvitamin D and 25-hydroxyvitamin D3 [25(OH)D3] versus the +SD3 groups, but it increased 3-epi-25-hydroxyvitamin D3 levels by 3.4 ng/mL in the HF+ω-6+HD3 group and 4.0 ng/mL in the HF+ω-3+HD3 group, representing 30% and 70%, respectively, of the total 25(OH)D3 increase. Energy expenditure increased in those mice fed diets +ω-3FA, by 3.9% in the HF+ω-3+SD3 group and 7.4% in the HF+ω-3+HD3 group, but it did not translate into lower body weight. The glucose tolerance curves of the HF+ω-3+SD3 and HF+ω-3+HD3 groups were improved by 11% and 17%, respectively, as compared to the respective +ω-6FA groups. D3 supplementation, within the ω-3FA groups, altered the gut microbiota by increasing the abundance of S24-7 and Lachnospiraceae taxa compared to the standard dose, while within the ω-6FA groups, D3 supplementation did not modulate specific taxa. CONCLUSIONS: Overall, D3 supplementation does not prevent CMD or enhance the beneficial effects of ω-3FA in vitamin D-sufficient obese mice.


Subject(s)
Cholecalciferol/administration & dosage , Cholecalciferol/pharmacology , Fatty Acids, Omega-3/pharmacology , Metabolic Syndrome/prevention & control , Obesity/chemically induced , Animals , Diet, High-Fat , Dietary Sucrose/administration & dosage , Dietary Sucrose/adverse effects , Dietary Supplements , Drug Synergism , Fatty Acids, Omega-3/administration & dosage , Glucose Intolerance , Humans , Leptin/blood , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/complications , Random Allocation
4.
Chem Biol Interact ; 258: 115-25, 2016 Oct 25.
Article in English | MEDLINE | ID: mdl-27475863

ABSTRACT

Piceatannol is a hydroxylated derivative of resveratrol. While both dietary polyphenols coexist in edible plants and fruits, and share equivalent concentrations in several wines, the influence of piceatannol on adiposity has been less studied than that of resveratrol. Though resveratrol is now recognized to limit fat deposition in various obesity models, the benefit of its dietary supplementation remains under debate regarding human obesity treatment or prevention. The research for more potent resveratrol analogs is therefore still undergoing. This prompted us to compare various effects of piceatannol and resveratrol directly on human adipose tissue (hAT). Hydrogen peroxide release was measured by Amplex Red-based fluorescence in subcutaneous hAT samples from obese patients. Interactions of stilbenes with human amine oxidases and quinone reductase were assessed by radiometric methods, computational docking and electron paramagnetic resonance. Influences on lipogenic and lipolytic activities were compared in mouse adipocytes. Resveratrol and piceatannol inhibited monoamine oxidase (MAO) with respective IC50 of 18.5 and 133.7 µM, but not semicarbazide-sensitive amine oxidase (SSAO) in hAT. For both stilbenes, the docking scores were better for MAO than for SSAO. Piceatannol and resveratrol similarly hampered hydrogen peroxide detection in assays with and without hAT, while they shared pro-oxidant activities when incubated with purified quinone reductase. They exhibited similar dose-dependent inhibition of adipocyte lipogenic activity. Only piceatannol inhibited basal and stimulated lipolysis when incubated at a dose ≥100 µM. Thus, piceatannol exerted on fat cells dose-dependent effects similar to those of resveratrol, except for a stronger antilipolytic action. In this regard, piceatannol should be useful in limiting the lipotoxicity related to obesity when ingested or administered alone - or might hamper the fat mobilization induced by resveratrol when simultaneously administered with it.


Subject(s)
Hydrogen Peroxide/metabolism , Lipogenesis/drug effects , Lipolysis/drug effects , Monoamine Oxidase/metabolism , Stilbenes/pharmacology , Subcutaneous Fat/metabolism , Adipocytes/drug effects , Adipocytes/metabolism , Adult , Animals , Benzylamines/metabolism , Biocatalysis/drug effects , Catalase/metabolism , Electron Spin Resonance Spectroscopy , Female , Humans , Mice, Inbred C57BL , Molecular Docking Simulation , Oxidants/pharmacology , Resveratrol , Stilbenes/chemistry , Subcutaneous Fat/drug effects , Tyramine/metabolism
5.
Obesity (Silver Spring) ; 21(7): 1389-95, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23754443

ABSTRACT

INTRODUCTION: Trefoil factor family member 2 (Tff2) is a small gut peptide, mainly known for its protective and healing functions. As previously demonstrated, high-fat (HF) feeding can rapidly and specifically modulate Tff2 transcription in key tissues of mice, including the duodenum and mesenteric adipose tissue, therefore suggesting a novel role for this gene in energy balance. DESIGN AND METHODS: To explore whether and how Tff2 can influence feeding behavior and energy metabolism, Tff2 knock-out (KO) mice were challenged with HF diet for 12 weeks, hence food and energy intakes, body composition, as well as energy excretion and serum lipid and hormonal levels were analyzed. Finally, energy efficiency was estimated. RESULTS: Tff2 KO mice showed a greater appetite and higher energy intake compared to wild-type (WT). Consistently, they presented lower levels of serum leptin, and increased transcription of agouti-related protein (Agrp) in the hypothalamus. Though energy and triglyceride fecal excretion were augmented in Tff2 KO mice, digestible energy intake was superior. However, KO mice were finally protected from HF diet-induced obesity, and accumulated less weight and fat depots than WT animals, while keeping a normal lean mass. Energy efficiency was lower in HF-KO mice, while energy expenditure and locomotor activity were globally increased. CONCLUSIONS: The present work demonstrates previously unsuspected roles for Tff2 and suggests it to be a mastermind in the control of energy balance and a promising therapeutic target for obesity.


Subject(s)
Diet, High-Fat , Mucins/genetics , Muscle Proteins/genetics , Obesity/genetics , Peptides/genetics , Adipose Tissue/metabolism , Agouti-Related Protein/metabolism , Animals , Appetite , Body Composition , Energy Intake , Energy Metabolism , Gene Expression Regulation , Hypothalamus/metabolism , Leptin/blood , Lipids/blood , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mucins/metabolism , Muscle Proteins/metabolism , Obesity/blood , Peptides/metabolism , Satiation , Trefoil Factor-2
6.
Diabetes ; 62(3): 801-10, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23160530

ABSTRACT

Hypothalamic glucose sensing is involved in the control of feeding behavior and peripheral glucose homeostasis, and glial cells are suggested to play an important role in this process. Diazepam-binding inhibitor (DBI) and its processing product the octadecaneuropeptide (ODN), collectively named endozepines, are secreted by astroglia, and ODN is a potent anorexigenic factor. Therefore, we investigated the involvement of endozepines in brain glucose sensing. First, we showed that intracerebroventricular administration of glucose in rats increases DBI expression in hypothalamic glial-like tanycytes. We then demonstrated that glucose stimulates endozepine secretion from hypothalamic explants. Feeding experiments indicate that the anorexigenic effect of central administration of glucose was blunted by coinjection of an ODN antagonist. Conversely, the hyperphagic response elicited by central glucoprivation was suppressed by an ODN agonist. The anorexigenic effects of centrally injected glucose or ODN agonist were suppressed by blockade of the melanocortin-3/4 receptors, suggesting that glucose sensing involves endozepinergic control of the melanocortin pathway. Finally, we found that brain endozepines modulate blood glucose levels, suggesting their involvement in a feedback loop controlling whole-body glucose homeostasis. Collectively, these data indicate that endozepines are a critical relay in brain glucose sensing and potentially new targets in treatment of metabolic disorders.


Subject(s)
Appetite Regulation , Diazepam Binding Inhibitor/metabolism , Feedback, Physiological , Glucose/metabolism , Hypothalamus/metabolism , Neuroglia/metabolism , Neuropeptides/metabolism , Peptide Fragments/metabolism , Animals , Appetite Depressants/administration & dosage , Appetite Depressants/pharmacology , Appetite Regulation/drug effects , Appetite Stimulants/administration & dosage , Appetite Stimulants/pharmacology , Appetitive Behavior/drug effects , Arcuate Nucleus of Hypothalamus/cytology , Arcuate Nucleus of Hypothalamus/drug effects , Arcuate Nucleus of Hypothalamus/metabolism , Diazepam Binding Inhibitor/agonists , Diazepam Binding Inhibitor/antagonists & inhibitors , Feedback, Physiological/drug effects , Gene Expression Regulation/drug effects , Glucose/administration & dosage , Hypothalamus/cytology , Hypothalamus/drug effects , Injections, Intraventricular , Male , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/metabolism , Neuroglia/drug effects , Neuropeptides/antagonists & inhibitors , Peptide Fragments/antagonists & inhibitors , Protein Processing, Post-Translational , Rats , Rats, Wistar , Receptors, Melanocortin/antagonists & inhibitors , Receptors, Melanocortin/metabolism , Synaptic Transmission/drug effects , Tissue Culture Techniques
7.
Am J Physiol Regul Integr Comp Physiol ; 301(4): R1011-24, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21775649

ABSTRACT

The aim of our study was to investigate the anorectic and brain stimulatory effects of various doses of exendin-4 (Ex-4) and to investigate the role of the vagus nerve in Ex-4-induced brain activation. A dose-related increase in c-fos mRNA expression was observed following Ex-4 administration (0.155-15.5 µg/kg). Doses of Ex-4 that caused anorexia without aversive effects (0.155, 0.775 µg/kg) induced c-fos expression in the hypothalamic arcuate and paraventricular (PVH; parvocellular) nuclei as well as in the limbic and brainstem structures. Doses of Ex-4 that caused aversion (1.55, 15.5 µg/kg) stimulated the same regions (in a more intense way) and additionally activated the magnocellular hypothalamic structures (supraoptic nucleus and PVH magnocellular). The brain c-fos pattern induced by Ex-4 showed both similarities and differences with that induced by refeeding. Subdiaphragmatic vagotomy significantly blunted the stimulation of c-fos mRNA expression induced by Ex-4 in the nodose ganglion, the medial part of nucleus of the solitary tract, and the parvocellular division of the PVH. Pretreatment with Ex-9-39 (330 µg/kg ip) impaired the neuronal activation evoked by Ex-4 in all brain regions and in the nodose ganglion. Effects of Ex-4 on hypothalamic-pituitary-adrenal axis activity were not altered by vagotomy. Results of this study demonstrate and relate the anorectic and brain stimulatory effects of aversive and nonaversive doses of Ex-4 and indicate that the activation of specific central regions induced by the peripheral administration of Ex-4 is, at least in part, dependent on the integrity of the vagus nerve.


Subject(s)
Brain/drug effects , Brain/metabolism , Peptides/pharmacology , Proto-Oncogene Proteins c-fos/metabolism , Receptors, Glucagon/agonists , Venoms/pharmacology , Animals , Corticotropin-Releasing Hormone/metabolism , Dose-Response Relationship, Drug , Exenatide , Glucagon-Like Peptide-1 Receptor , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/physiology , Hypothalamus/drug effects , Hypothalamus/metabolism , Male , Models, Animal , Nodose Ganglion/drug effects , Nodose Ganglion/metabolism , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/metabolism , Pituitary-Adrenal System/drug effects , Pituitary-Adrenal System/physiology , Rats , Rats, Wistar , Vagus Nerve/drug effects , Vagus Nerve/physiology , Vagus Nerve/surgery
8.
BMC Physiol ; 10: 4, 2010 Apr 23.
Article in English | MEDLINE | ID: mdl-20416070

ABSTRACT

BACKGROUND: Acylation stimulating protein (ASP) is an adipogenic hormone that stimulates triglyceride (TG) synthesis and glucose transport in adipocytes. Previous studies have shown that ASP-deficient C3 knockout mice are hyperphagic yet lean, as they display increased oxygen consumption and fatty acid oxidation compared to wildtype mice. In the present study, antibodies against ASP (Anti-ASP) and human recombinant ASP (rASP) were tested in vitro and in vivo. Continuous administration for 4 weeks via osmotic mini-pump of Anti-ASP or rASP was evaluated in wildtype mice on a high-fat diet (HFD) to examine their effects on body weight, food intake and energy expenditure. RESULTS: In mature murine adipocytes, rASP significantly stimulated fatty acid uptake (+243% vs PBS, P < 0.05) while Anti-ASP neutralized the rASP response. Mice treated with Anti-ASP showed elevated energy expenditure (P < 0.0001), increased skeletal muscle glucose oxidation (+141%, P < 0.001), reduced liver glycogen (-34%, P < 0.05) and glucose-6-phosphate content (-64%, P = 0.08) compared to control mice. There was no change in body weight, food intake, fasting insulin, adiponectin, CRP or TG levels compared to controls. Interestingly, HFD mice treated with rASP showed the opposite phenotype with reduced energy expenditure (P < 0.0001) and increased body weight (P < 0.05), cumulative food intake (P < 0.0001) and liver glycogen content (+59%, P < 0.05). Again, there was no change in circulating insulin, adiponectin, CRP or TG levels, however, plasma free fatty acids were reduced (-48%, P < 0.05). CONCLUSION: In vitro, Anti-ASP effectively neutralized ASP stimulated fatty acid uptake. In vivo, Anti-ASP treatment increased whole body energy utilization while rASP increased energy storage. Therefore, ASP is a potent anabolic hormone that may also be a mediator of energy expenditure.


Subject(s)
Adipocytes/metabolism , Antibodies, Neutralizing/pharmacology , Energy Metabolism/physiology , Intercellular Signaling Peptides and Proteins , 3T3-L1 Cells , Acylation/physiology , Adipocytes/drug effects , Animals , Antibodies, Neutralizing/blood , Blood Glucose/metabolism , Body Weight/drug effects , Body Weight/physiology , Calorimetry, Indirect , Complement C3 , Dietary Fats/pharmacokinetics , Eating/drug effects , Eating/physiology , Energy Metabolism/drug effects , Fatty Acids, Nonesterified/blood , Fatty Acids, Nonesterified/pharmacokinetics , Hormones/blood , Humans , Infusion Pumps , Intercellular Signaling Peptides and Proteins/immunology , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/pharmacology , Male , Mice , Mice, Inbred C57BL , Oxidation-Reduction , Recombinant Proteins/pharmacology
9.
Neurosci Lett ; 480(1): 1-3, 2010 Aug 09.
Article in English | MEDLINE | ID: mdl-20074616

ABSTRACT

Sirt1 is a NAD-dependent deacetylase that has been shown as a link between energy metabolism and aging. Its putative role as a target for neurodegenerative disorders has recently been suggested; yet, little is known about the changes that occur in Sirt1 levels in the aging brain. Here we show by in situ hybridization that Sirt1 expression is modified in specific areas of the brain in mice upon aging, and that gender also impacts on this regulation. Mice aged 12 and 24 months had a lower Sirt1 expression specifically in the antero ventral thalamic nucleus (AV) and in the arcuate nucleus (ARC) than their young (4mo) counterparts, whereas changes were either not noticeable or not significantly modulated in other parts of the brain. Regulation of Sirt1 mRNA levels in the subfornical organ (SFO) and in the substancia nigra part compacta (SNC) depended on gender. These findings suggest that reduced Sirt1 levels upon aging could contribute to a lower Sirt1 activity, and that specific nuclei might be particularly affected.


Subject(s)
Hypothalamus/metabolism , RNA, Messenger/biosynthesis , Sirtuin 1/biosynthesis , Age Factors , Animals , Female , Male , Mice , Mice, Inbred C57BL , Sex Factors , Sirtuin 1/genetics
10.
Endocrinology ; 149(5): 2121-30, 2008 May.
Article in English | MEDLINE | ID: mdl-18218698

ABSTRACT

Peroxisome proliferator-activated receptor-gamma (PPARgamma) activation up-regulates thermogenesis-related genes in rodent white and brown adipose tissues (WAT and BAT) without increasing whole-body energy expenditure. We tested here whether such dissociation is the result of a negative modulation of sympathetic activity to WAT and BAT and thyroid axis components by PPARgamma activation. Administration of the PPARgamma agonist rosiglitazone (15 mg/kg.d) for 7 d to male Sprague Dawley rats increased food intake (10%), feed efficiency (31%), weight gain (45%), spontaneous motor activity (60%), and BAT and WAT mass and reduced whole-body oxygen consumption. Consistent with an anabolic setting, rosiglitazone markedly reduced sympathetic activity to BAT and WAT (>50%) and thyroid status as evidenced by reduced levels of plasma thyroid hormones (T(4) and T(3)) and mRNA levels of BAT and liver T(3)-generating enzymes iodothyronine type 2 (-40%) and type 1 (-32%) deiodinases, respectively. Rosiglitazone also decreased mRNA levels of the thyroid hormone receptor (THR) isoforms alpha1 (-34%) and beta (-66%) in BAT and isoforms alpha1 (-20%) and alpha2 (-47%) in retroperitoneal WAT. These metabolic effects were associated with a reduction in mRNA levels of the pro-energy expenditure peptides CRH and CART in specific hypothalamic nuclei. A direct central action of rosiglitazone is, however, unlikely based on its low brain uptake and lack of metabolic effects of intracerebroventricular administration. In conclusion, a reduction in BAT sympathetic activity and thyroid status appears to, at least partly, explain the PPARgamma-induced reduction in energy expenditure and the fact that up-regulation of thermogenic gene expression does not translate into functional stimulation of whole-body thermogenesis in vivo.


Subject(s)
Adipose Tissue/innervation , Adrenergic Fibers/drug effects , Energy Metabolism/drug effects , PPAR gamma/agonists , Thiazolidinediones/pharmacology , Thyroid Gland/physiology , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Adrenergic Fibers/physiology , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Body Weight/drug effects , Eating/drug effects , Energy Metabolism/physiology , Hypothalamus/drug effects , Hypothalamus/metabolism , Injections, Intraventricular , Ion Channels/metabolism , Male , Mice , Mitochondrial Proteins/metabolism , Nerve Tissue Proteins/metabolism , PPAR gamma/physiology , Rats , Rats, Sprague-Dawley , Rosiglitazone , Thermogenesis/drug effects , Thermogenesis/genetics , Thiazolidinediones/administration & dosage , Thiazolidinediones/pharmacokinetics , Thyroid Gland/drug effects , Uncoupling Protein 1 , Up-Regulation/drug effects
11.
Am J Physiol Regul Integr Comp Physiol ; 293(3): R1076-85, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17581834

ABSTRACT

We previously reported an exaggerated endocrine and weight loss response to stress in rats fed a high-fat (HF) diet for 5 days. Others report blunted stress-induced anxiety in rats made obese on a HF diet. Experiments described here tested whether sensitivity to stress-related peptides was changed in obese and nonobese HF-fed rats. Third ventricle infusion of corticotropin-releasing factor (CRF) in rats made obese on HF diet (40% kcal fat) produced an exaggerated hypophagia, which is thought to be mediated by CRF(2) receptors. Obese rats responded to a lower dose of CRF for a longer time than rats fed a low-fat (LF) diet (12% kcal fat). CRF-induced release of corticosterone, which is thought to be mediated by CRF(1) receptors, was not exaggerated in obese HF-fed rats. In contrast, rats fed HF diet for 5 days showed the same food intake and corticosterone response to CRF as LF-fed rats. CRF mRNA expression in the paraventricular nucleus of the hypothalamus was stimulated by mild stress (ip saline injection and placement in a novel cage) in LF-fed rats but not in rats fed HF diet for 5 days because of a nonsignificant increase in expression in nonstressed HF-fed rats. In addition, nonstressed levels of urocortin (UCN) I mRNA expression in the Edinger-Westphal nucleus were significantly inhibited in HF-fed rats. These data suggest that rats that have become obese on a HF diet show a change in responsiveness to stress peptides, whereas the increased stress response in nonobese HF-fed rats may be associated with changes in basal CRF and UCN I mRNA expression.


Subject(s)
Corticotropin-Releasing Hormone/pharmacology , Dietary Fats/pharmacology , Adipose Tissue/physiology , Adiposity/physiology , Animals , Area Under Curve , Body Composition/drug effects , Body Composition/physiology , Body Weight/physiology , Brain Chemistry/drug effects , Brain Chemistry/physiology , Corticosterone/metabolism , Corticotropin-Releasing Hormone/administration & dosage , Corticotropin-Releasing Hormone/biosynthesis , Corticotropin-Releasing Hormone/genetics , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Diet , Diet, Fat-Restricted , Dose-Response Relationship, Drug , Eating/drug effects , Energy Intake/drug effects , Male , Organ Size/physiology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Stress, Psychological/psychology , Time Factors , Urocortins
12.
J Endocrinol ; 193(1): 11-9, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17400798

ABSTRACT

The corticotropin-releasing factor (CRF) system is involved in numerous physiological and behavioral actions, including the regulation of energy balance. We examined the effects of the CRF(1) receptor antagonist, SSR125543, on energy balance and food deprivation-induced neuronal activation in obese rats. Lean (Fa/?) and obese (fa/fa) Zucker rats were treated orally with SSR125543 at a daily dose of 30 mg/kg for 21 days. Rats were killed either fed ad libitum or food deprived for 6 h in order to induce a mild stress response in obese rats. SSR125543 reduced plasma corticosterone levels in lean rats, prevented corticosterone response to fasting in obese rats, and increased CRF mRNA levels in the paraventricular hypothalamic nucleus (PVN) of both lean and obese rats, further confirming that the antagonist partially blocked CRF(1) receptors. SSR125543 increased protein gain in obese rats. Whole carcass analyses showed reduced energy and fat gains in lean rats. Consistent with reduced fat gain, circulating triglyceride and leptin levels were reduced in SSR125543-treated lean rats. In obese rats, circulating glucose levels and the homeostasis model assessment of insulin resistance index of insulin resistance were reduced by SSR125543 treatment. CRF(1) receptor blockade increased uncoupling protein-1 mRNA levels in interscapular brown adipose tissue of obese rats. The antagonist partly blocked the fasting-induced changes in c-fos mRNA levels in the PVN and arcuate nucleus of obese rats. Overall, these results suggest that although SSR125543 had relatively mild effects on energy balance, CRF(1) receptor blockade attenuated several metabolic effects of short-term fasting and improved plasma variables related to the metabolic syndrome and diabetes.


Subject(s)
Energy Metabolism , Food Deprivation , Hydrocarbons, Halogenated/therapeutic use , Obesity/drug therapy , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Thiazines/therapeutic use , Animals , Blood Glucose/analysis , Corticosterone/blood , Corticotropin-Releasing Hormone/genetics , Corticotropin-Releasing Hormone/metabolism , Fatty Acids, Nonesterified/blood , Gene Expression , Hypothalamus/metabolism , In Situ Hybridization/methods , Insulin/blood , Insulin Resistance , Male , Obesity/metabolism , Proteins/metabolism , Proto-Oncogene Proteins c-fos/genetics , Rats , Rats, Zucker , Receptors, Corticotropin-Releasing Hormone/genetics , Receptors, Corticotropin-Releasing Hormone/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Triglycerides/blood
13.
Cell Metab ; 5(1): 21-33, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17189204

ABSTRACT

The active thyroid hormone, triiodothyronine (T3), regulates mitochondrial uncoupling protein activity and related thermogenesis in peripheral tissues. Type 2 deiodinase (DII), an enzyme that catalyzes active thyroid hormone production, and mitochondrial uncoupling protein 2 (UCP2) are also present in the hypothalamic arcuate nucleus, where their interaction and physiological significance have not been explored. Here, we report that DII-producing glial cells are in direct apposition to neurons coexpressing neuropeptide Y (NPY), agouti-related protein (AgRP), and UCP2. Fasting increased DII activity and local thyroid hormone production in the arcuate nucleus in parallel with increased GDP-regulated UCP2-dependent mitochondrial uncoupling. Fasting-induced T3-mediated UCP2 activation resulted in mitochondrial proliferation in NPY/AgRP neurons, an event that was critical for increased excitability of these orexigenic neurons and consequent rebound feeding following food deprivation. These results reveal a physiological role for a thyroid-hormone-regulated mitochondrial uncoupling in hypothalamic neuronal networks.


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Fasting , Feeding Behavior , Ion Channels/metabolism , Mitochondrial Proteins/metabolism , Neurons/metabolism , Thermogenesis , Triiodothyronine/metabolism , Agouti-Related Protein , Animals , Arcuate Nucleus of Hypothalamus/cytology , Eating , Green Fluorescent Proteins , Guanosine Diphosphate/metabolism , Hypothalamus/cytology , Intercellular Signaling Peptides and Proteins/metabolism , Iodide Peroxidase/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Neuroglia/metabolism , Neuropeptide Y/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Uncoupling Protein 2 , Iodothyronine Deiodinase Type II
14.
Horm Behav ; 49(5): 615-25, 2006 May.
Article in English | MEDLINE | ID: mdl-16423352

ABSTRACT

Rats exposed to restraint stress for 3 h on each of 3 days lose weight and do not return to the weight of their non-stressed controls for extended periods of time. Studies described here demonstrate that the initial weight loss is associated with increased energy expenditure and reduced food intake on the days of restraint but that there is no difference between stressed and control rats once stress ends. The failure to compensate for this energy deficit accounts for the sustained reduction in weight which lasts for up to 80 days after the end of restraint. In an additional experiment, in situ hybridization was used to measure mRNA expression of corticotrophin releasing factor (CRF) and CRF receptors in hypothalamic nuclei, of urocortin (UCN) in the Edinger Westphal nucleus and of UCN III in the rostral perifornical area and medial amygdaloidal nucleus. Immediately after the second 3 h bout of restraint stress, there was a significant increase in expression of UCN in the Edinger Westphal nucleus and of CRF-R1 in the paraventricular nucleus of the hypothalamus and a less pronounced decrease in CRF-R2 expression in the ventromedial nucleus of the hypothalamus. There were no differences in expression of stress-related peptides or their receptors 40 days after the end of repeated restraint. These results suggest that the sustained reduction in body weight and increased responsiveness to subsequent stressors in rats that have been exposed to repeated restraint are not associated with prolonged changes in mRNA expression of CRF receptors or their ligands.


Subject(s)
Body Weight/physiology , Corticotropin-Releasing Hormone/genetics , Receptors, Corticotropin-Releasing Hormone/genetics , Stress, Physiological/physiopathology , Amygdala/physiology , Animals , Energy Metabolism/physiology , Gene Expression/physiology , Hypothalamus/physiology , In Situ Hybridization , Male , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Restraint, Physical , Stress, Physiological/metabolism , Tegmentum Mesencephali/physiology , Urocortins
15.
Eur J Neurosci ; 22(6): 1489-501, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16190902

ABSTRACT

Following refeeding, c-fos expression is induced in a particular set of brain regions that include the nucleus of the solitary tract (NTS), parabrachial nucleus (PB), central amygdala (CeA), paraventricular hypothalamic nucleus (PVH), supraoptic nucleus (SON) and the circumventricular organs. Within the PVH, the expression is particularly intense in the magnocellular division of the nucleus and it is as yet not clear how this activation occurs. The respective contribution of the vagus afferents and lamina terminalis, which conveys signals entering the brain through the forebrain circumventricular organs, has been investigated in rats subjected to a unilateral cervical vagotomy (UCV) or a unilateral lesion of the fibres running within the lamina terminalis (ULT) and projecting to the neuroendocrine hypothalamus. UCV significantly decreased postprandial c-fos expression in the NTS, PB, CeA and parvocellular division of the PVH. In contrast, ULT impaired postprandial activation of the magnocellular neurons in the PVH and SON. The present study also characterized the types of neurons activated in the PVH and SON during refeeding. In the magnocellular regions, arginine-vasopressin (AVP) neurons were activated upon refeeding whereas there was no apparent induction of Fos expression in oxytocin cells. In the parvocellular PVH, postprandial Fos was induced only in 30% of the corticotrophin-releasing factor (CRF) and AVP neurons. The results of the present study suggest that the postprandial activation of the brain requires the integrity of both the vagal- and lamina terminalis-associated pathways.


Subject(s)
Brain/physiology , Eating/physiology , Fasting/physiology , Hypothalamus/physiology , Vagus Nerve/physiology , Animals , Antisense Elements (Genetics) , Gene Expression/physiology , Genes, fos/genetics , In Situ Hybridization , In Vitro Techniques , Male , Nerve Fibers/physiology , Neural Pathways/physiology , Neurons, Afferent/physiology , Neurosecretory Systems/physiology , Paraventricular Hypothalamic Nucleus/physiology , RNA, Messenger/biosynthesis , Rats , Sulfur Radioisotopes , Vagotomy
16.
J Biol Chem ; 280(23): 22036-43, 2005 Jun 10.
Article in English | MEDLINE | ID: mdl-15809292

ABSTRACT

The mitochondrial carrier family transports a variety of metabolites across the inner mitochondrial membrane. We identified and cloned a new member of this family, KMCP1 (kidney mitochondrial carrier protein-1), that is highly homologous to the previously identified protein BMCP1 (brain mitochondrial carrier protein-1). Western blotting and in situ experiments showed that this carrier is expressed predominantly within the kidney cortex in the proximal and distal tubules. KMCP1 was increased during fasting and during the regenerative phase of glycerol-induced renal failure. We show that both situations are associated with transiently increased expression of superoxide-generating enzymes, followed by increased mitochondrial metabolism and antioxidant defenses. Given that KMCP1 expression occurs simultaneously with these latter events, we propose that KMCP1 is involved in situations in which mitochondrial metabolism is increased, in particular when the cellular redox balance tends toward a pro-oxidant status.


Subject(s)
Antioxidants/pharmacology , Carrier Proteins/biosynthesis , Carrier Proteins/chemistry , Carrier Proteins/physiology , Kidney Tubules/physiology , Kidney/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/biosynthesis , Mitochondrial Proteins/physiology , Regeneration , Up-Regulation , Amino Acid Sequence , Animals , Blotting, Northern , Blotting, Western , Brain/metabolism , COS Cells , Carrier Proteins/metabolism , Cloning, Molecular , DNA, Complementary/metabolism , Glutamine/chemistry , Glycerol/chemistry , Glycerol/metabolism , Immunoprecipitation , Ion Channels , Membrane Potentials , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Oxidants/pharmacology , Oxidation-Reduction , Oxidative Stress , Oxygen/metabolism , Oxygen Consumption , Phylogeny , RNA/chemistry , RNA/metabolism , Superoxide Dismutase/metabolism , Time Factors , Tissue Distribution , Uncoupling Protein 1
17.
Endocrinology ; 143(12): 4683-92, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12446596

ABSTRACT

Leptin regulates food intake in adult mammals by stimulating hypothalamic anorexigenic pathways and inhibiting orexigenic ones. In developing rodents, fat stores are low, yet circulating leptin levels are high and do not appear to regulate food intake. We determined whether two appetite-related neuropeptides [neuropeptide Y (NPY) and proopiomelanocortin (POMC)] and food intake behavior are sensitive to leptin [3 mg/kg body weight (BW), ip] in neonates. We measured the effects of 1) acute leptin administration (3 mg/kg BW, ip, 3 h before testing) on food intake on postnatal day (PND) 5, 8, and 10; and 2) chronic leptin treatment (3 mg/kg BW, ip, daily PND3-PND10) on BW gain and fat pads weight on PND10. In addition to hypothalamic POMC and NPY expression, we determined the expression of suppressor of cytokine signaling-3, all subtypes of leptin receptors, and corticotropin-releasing factor receptor-2 mRNA in PND10 pups receiving either an acute (PND10) or a chronic (PND 3-10) leptin (3 mg/kg BW, ip) or vehicle treatment. Brains were removed 30 or 120 min after the last injection. Acute leptin administration did not affect food intake at any age tested. Chronic leptin treatment did not change BW but decreased fat pad weight significantly. In the arcuate nucleus (ARC), acute leptin increased SOCS-3 and POMC mRNA levels, but decreased NPY mRNA levels in the rostral part of ARC. Chronic leptin down-regulated all subtypes of leptin receptors mRNA and decreased NPY mRNA levels in the caudal ARC but had no further effect on POMC expression. Chronic leptin increased corticotropin-releasing factor receptor-2 mRNA levels in the ventromedial hypothalamus. We conclude that despite adult-like effects of leptin on POMC, NPY, and CRFR-2 expression in neonates, leptin does not regulate food intake during early development.


Subject(s)
Animals, Newborn/growth & development , Appetite/physiology , Eating/drug effects , Hypothalamus/metabolism , Leptin/pharmacology , Neuropeptides/genetics , Repressor Proteins , Transcription Factors , Adipose Tissue/growth & development , Animals , Arcuate Nucleus of Hypothalamus/metabolism , Gene Expression Regulation/drug effects , Hypothalamus, Middle/metabolism , Leptin/administration & dosage , Neuropeptide Y/genetics , Organ Size/drug effects , Pro-Opiomelanocortin/genetics , Proteins/genetics , Rats , Rats, Sprague-Dawley , Receptors, Cell Surface/genetics , Receptors, Corticotropin-Releasing Hormone/genetics , Receptors, Leptin , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins , Weight Gain/drug effects
18.
Behav Neurosci ; 116(4): 651-9, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12148932

ABSTRACT

The rewarding effect produced by electrical stimulation of some lateral hypothalamic sites is modulated by chronic food restriction and weight loss. The sensitivity of the rewarding effect to restriction predicts the modulation of brain stimulation reward (BSR) by the adiposity hormone, leptin. The present study examined the effect of corticotropin-releasing hormone (CRH) on the rewarding effect of stimulating restriction-sensitive and restriction-insensitive sites. Chronic food restriction reduced frequency thresholds for BSR in half of the subjects but had no effect in the others. CRH increased thresholds only in subjects in which the rewarding effect was insensitive to restriction. In contrast, frequency thresholds remained stable in nearly all rats with restriction-sensitive stimulation sites. These findings provide further evidence that sensitivity to food restriction is an important factor in determining the influence of hormones and neuropeptides on brain reward circuitry.


Subject(s)
Corticotropin-Releasing Hormone/pharmacology , Food Deprivation , Hypothalamus/physiology , Reinforcement, Psychology , Animals , Electric Stimulation , Energy Metabolism , Male , Rats , Weight Loss
19.
Eur J Neurosci ; 15(6): 1013-29, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11918662

ABSTRACT

The present study was conducted to investigate the pattern of neuronal activation and corticotropin-releasing hormone (CRH) expression in fed, food deprived and refed lean (Fa/?) and obese (fa/fa) Zucker rats. The pattern of neuronal activation was studied by measuring the expression of the immediate-early gene c-fos. Expression of c-fos and CRH mRNA was determined by in situ hybridization histochemistry. In both lean and obese rats, one hour of refeeding led to a transient increase in c-fos mRNA levels which was detected in the paraventricular hypothalamic nucleus (PVH), the dorsomedial hypothalamic nucleus, the supraoptic nucleus, the paraventricular thalamic nucleus, the central nucleus of amygdala (CeA), the lateral and medial parabrachial nuclei, the nucleus of the solitary tract, and the area postrema. In addition, refeeding led to strong activation of the arginine-vasopressin neurons located in the magnocellular part of the PVH. Following 24 h of food deprivation, CRH expression in the parvocellular division of the PVH was significantly higher in obese rats compared to lean animals. During refeeding, PVH CRH mRNA levels in obese rats decreased to reach control values. The decrease in CRH expression in obese rats was accompanied by the alleviation of the hypercorticosteronemia that characterized obese Zucker rats. CRH mRNA levels in the central nucleus of the amygdala were significantly higher in lean rats than in obese animals, when the rats were fed ad libitum During food deprivation, CeA CRH mRNA levels decreased in lean rats and gradually returned to predeprivation values during refeeding. In refed obese rats, CeA levels of CRH mRNA were higher than those of ad libitum fed or food-deprived obese mutants. In the perifornical region of the lateral hypothalamic area (LHA), the expression of CRH mRNA rose significantly in response to refeeding in lean rats, but not in obese animals. Following the first hour of refeeding, the number of neurons expressing CRH mRNA in the LHA in lean rats almost doubled. The present results demonstrate that refeeding has a stimulating effect in obese Zucker rats in a pattern of activation similar to that seen in lean Fa/? rats. They also demonstrate differences in CRH expression between Fa/? and fa/fa rats after refeeding. The most apparent of these differences was seen in the lateral hypothalamus in which refeeding failed to up-regulate CRH expression in obese rats.


Subject(s)
Appetite Regulation/physiology , Body Weight/physiology , Brain/metabolism , Corticotropin-Releasing Hormone/genetics , Food Deprivation/physiology , Neurons/metabolism , Obesity/metabolism , Animals , Brain/cytology , Brain Stem/cytology , Brain Stem/metabolism , Corticosterone/blood , Disease Models, Animal , Hypothalamus/cytology , Hypothalamus/metabolism , Male , Neurons/cytology , Obesity/genetics , Obesity/physiopathology , Proto-Oncogene Proteins c-fos/genetics , RNA, Messenger/metabolism , Rats , Rats, Zucker , Up-Regulation/physiology
SELECTION OF CITATIONS
SEARCH DETAIL