Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
1.
Mol Psychiatry ; 28(5): 1857-1867, 2023 05.
Article in English | MEDLINE | ID: mdl-36765131

ABSTRACT

Antipsychotic (AP) drugs are efficacious treatments for various psychiatric disorders, but excessive weight gain and subsequent development of metabolic disease remain serious side effects of their use. Increased food intake leads to AP-induced weight gain, but the underlying molecular mechanisms remain unknown. In previous studies, we identified the neuropeptide Agrp and the transcription factor nuclear receptor subfamily 5 group A member 2 (Nr5a2) as significantly upregulated genes in the hypothalamus following AP-induced hyperphagia. While Agrp is expressed specifically in the arcuate nucleus of the hypothalamus and plays a critical role in appetite stimulation, Nr5a2 is expressed in both the CNS and periphery, but its role in food intake behaviors remains unknown. In this study, we investigated the role of hypothalamic Nr5a2 in AP-induced hyperphagia and weight gain. In hypothalamic cell lines, olanzapine treatment resulted in a dose-dependent increase in gene expression of Nr5a2 and Agrp. In mice, the pharmacological inhibition of NR5A2 decreased olanzapine-induced hyperphagia and weight gain, while the knockdown of Nr5a2 in the arcuate nucleus partially reversed olanzapine-induced hyperphagia. Chromatin-immunoprecipitation studies showed for the first time that NR5A2 directly binds to the Agrp promoter region. Lastly, the analysis of single-cell RNA seq data confirms that Nr5a2 and Agrp are co-expressed in a subset of neurons in the arcuate nucleus. In summary, we identify Nr5a2 as a key mechanistic driver of AP-induced food intake. These findings can inform future clinical development of APs that do not activate hyperphagia and weight gain.


Subject(s)
Hyperphagia , Animals , Humans , Mice , Agouti-Related Protein/genetics , Agouti-Related Protein/metabolism , Agouti-Related Protein/pharmacology , Antipsychotic Agents/adverse effects , Eating , Hyperphagia/chemically induced , Hyperphagia/genetics , Hyperphagia/metabolism , Hypothalamus/metabolism , Olanzapine/adverse effects , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Cytoplasmic and Nuclear/pharmacology , Receptors, Cytoplasmic and Nuclear/therapeutic use , Weight Gain
2.
Sci Rep ; 10(1): 18581, 2020 10 29.
Article in English | MEDLINE | ID: mdl-33122657

ABSTRACT

Antipsychotic drugs (AP) are used to treat a multitude of psychiatric conditions including schizophrenia and bipolar disorder. However, APs also have metabolic side effects including increased food intake and body weight, but the underlying mechanisms remain unknown. We previously reported that minocycline (MINO) co-treatment abrogates olanzapine (OLZ)-induced hyperphagia and weight gain in mice. Using this model, we investigated the changes in the pharmacometabolome in the plasma and hypothalamus associated with OLZ-induced hyperphagia and weight gain. Female C57BL/6 mice were divided into groups and fed either i) control, CON (45% fat diet) ii) CON + MINO, iii) OLZ (45% fat diet with OLZ), iv) OLZ + MINO. We identified one hypothalamic metabolite indoxylsulfuric acid and 389 plasma metabolites (including 19 known metabolites) that were specifically associated with AP-induced hyperphagia and weight gain in mice. We found that plasma citrulline, tricosenoic acid, docosadienoic acid and palmitoleic acid were increased while serine, asparagine and arachidonic acid and its derivatives were decreased in response to OLZ. These changes were specifically blocked by co-treatment with MINO. These pharmacometabolomic profiles associated with AP-induced hyperphagia and weight gain provide candidate biomarkers and mechanistic insights related to the metabolic side effects of these widely used drugs.


Subject(s)
Eating/drug effects , Hyperphagia/metabolism , Metabolome/drug effects , Minocycline/pharmacology , Olanzapine/toxicity , Weight Gain , Animals , Anti-Bacterial Agents/pharmacology , Antipsychotic Agents/toxicity , Female , Hyperphagia/chemically induced , Hyperphagia/drug therapy , Hyperphagia/pathology , Hypothalamus/drug effects , Hypothalamus/metabolism , Mice , Mice, Inbred C57BL
3.
Nat Commun ; 9(1): 5272, 2018 12 10.
Article in English | MEDLINE | ID: mdl-30532051

ABSTRACT

Antipsychotic (AP) drugs are used to treat psychiatric disorders but are associated with significant weight gain and metabolic disease. Increased food intake (hyperphagia) appears to be a driving force by which APs induce weight gain but the mechanisms are poorly understood. Here we report that administration of APs to C. elegans induces hyperphagia by a mechanism that is genetically distinct from basal food intake. We exploit this finding to screen for adjuvant drugs that suppress AP-induced hyperphagia in C. elegans and mice. In mice AP-induced hyperphagia is associated with a unique hypothalamic gene expression signature that is abrogated by adjuvant drug treatment. Genetic analysis of this signature using C. elegans identifies two transcription factors, nhr-25/Nr5a2 and nfyb-1/NFYB to be required for AP-induced hyperphagia. Our study reveals that AP-induced hyperphagia can be selectively suppressed without affecting basal food intake allowing for novel drug discovery strategies to combat AP-induced metabolic side effects.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans/genetics , Eating/genetics , Hyperphagia/genetics , Animals , Antipsychotic Agents/toxicity , CCAAT-Binding Factor/genetics , Chemotherapy, Adjuvant , DNA-Binding Proteins/genetics , Eating/drug effects , Gene Expression/drug effects , Gene Expression Profiling , Hyperphagia/chemically induced , Hyperphagia/drug therapy , Hypothalamus/metabolism , Mice , Phenotype , Transcription Factors/genetics , Vemurafenib/pharmacology
4.
Brain Res ; 1695: 45-52, 2018 09 15.
Article in English | MEDLINE | ID: mdl-29775565

ABSTRACT

Although olanzapine is highly efficacious and most widely used second generation antipsychotic drug, the success of treatment has been hampered by its propensity to induce weight gain. While the underlying neuronal mechanisms are unclear, their elucidation may help to target alternative pathways regulating energy balance. The present study was undertaken to define the role of cocaine- and amphetamine-regulated transcript (CART), a well-known anorexic peptide, in olanzapine-induced hyperphagia and body weight gain in female rats. Olanzapine was administered daily by intraperitoneal route, alone or in combination with CART (intracerebroventricular) for a period of two weeks. Immediately after drug administrations, preweighed food was offered to the animals at the commencement of the dark phase. The food intake and body weight were measured daily just prior to next injection. Furthermore, the brains of olanzapine-treated rats were processed for the immunohistochemical analysis of CART-containing elements in the hypothalamus. Treatment with olanzapine (0.5 mg/kg) for the duration of 14 days produced a significant increase in food intake and body weight as compared to control. However, concomitant administration of CART (0.5 µg) attenuated the olanzapine-induced hyperphagia and weight gain. Olanzapine administration resulted in a significant reduction in CART immunoreactivity in the hypothalamic arcuate, paraventricular, dorsomedial and ventromedial nuclei. We suggest that decreased CART contents in the hypothalamus may be causally linked with the hyperphagia and weight gain induced by olanzapine.


Subject(s)
Body Weight/drug effects , Hyperphagia/drug therapy , Neuropeptides/pharmacology , Weight Gain/drug effects , Animals , Antipsychotic Agents/pharmacology , Eating/drug effects , Feeding Behavior/drug effects , Female , Hyperphagia/chemically induced , Hypothalamus/drug effects , Hypothalamus/metabolism , Neuropeptides/metabolism , Olanzapine/pharmacology , Rats, Sprague-Dawley
5.
J Biochem Mol Toxicol ; 31(6)2017 Jun.
Article in English | MEDLINE | ID: mdl-28186389

ABSTRACT

Corticosterone plays an important role in feeding behavior. However, its mechanism remains unclear. Therefore, the present study aimed to investigate the effect of corticosterone on feeding behavior. In this study, cumulative food intake was increased by acute corticosterone administration in a dose-dependent manner. Administration of the 5-HT2c receptor agonist m-chlorophenylpiperazin (mCPP) reversed the effect of corticosterone on food intake. The anorectic effects of mCPP were also blocked by the 5-HT2c receptor antagonist RS102221 in corticosterone-treated mice. Both corticosterone and mCPP increased c-Fos expression in hypothalamic nuclei, but not the nucleus of the solitary tract. RS102221 inhibited c-Fos expression induced by mCPP, but not corticosterone. In addition, mCPP had little effect on TH and POMC levels in the hypothalamus. Furthermore, mCPP antagonized decreasing effect of the leptin produced by corticosterone. Taken together, our findings suggest that 5-HT2c receptors and leptin may be involved in the effects of corticosterone-induced hyperphagia.


Subject(s)
Appetite Regulation/drug effects , Corticosterone/pharmacology , Hypothalamus/drug effects , Leptin/agonists , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Receptor, Serotonin, 5-HT2C/metabolism , Animals , Appetite Depressants/chemistry , Appetite Depressants/pharmacology , Appetite Stimulants/administration & dosage , Appetite Stimulants/agonists , Appetite Stimulants/antagonists & inhibitors , Appetite Stimulants/pharmacology , Behavior, Animal/drug effects , Corticosterone/administration & dosage , Corticosterone/agonists , Corticosterone/antagonists & inhibitors , Dose-Response Relationship, Drug , Energy Intake/drug effects , Hyperphagia/blood , Hyperphagia/chemically induced , Hyperphagia/metabolism , Hyperphagia/pathology , Hypothalamus/metabolism , Hypothalamus/pathology , Leptin/antagonists & inhibitors , Leptin/blood , Leptin/metabolism , Mice, Inbred ICR , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Neurons/metabolism , Neurons/pathology , Organ Specificity , Piperazines/antagonists & inhibitors , Piperazines/pharmacology , Proto-Oncogene Proteins c-fos/agonists , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Receptor, Serotonin, 5-HT2C/chemistry , Serotonin 5-HT2 Receptor Agonists/pharmacology , Serotonin 5-HT2 Receptor Antagonists/pharmacology , Spiro Compounds/pharmacology , Sulfonamides/pharmacology , Up-Regulation/drug effects
6.
Behav Pharmacol ; 28(4): 280-284, 2017 06.
Article in English | MEDLINE | ID: mdl-28125508

ABSTRACT

Nonpsychoactive phytocannabinoids (pCBs) from Cannabis sativa may represent novel therapeutic options for cachexia because of their pleiotropic pharmacological activities, including appetite stimulation. We have recently shown that purified cannabigerol (CBG) is a novel appetite stimulant in rats. As standardized extracts from Cannabis chemotypes dominant in one pCB [botanical drug substances (BDSs)] often show greater efficacy and/or potency than purified pCBs, we investigated the effects of a CBG-rich BDS, devoid of psychoactive [INCREMENT]-tetrahydrocannabinol, on feeding behaviour. Following a 2 h prefeed satiation procedure, 16 male Lister-hooded rats were administered CBG-BDS (at 30-240 mg/kg) or vehicle. Food intake, meal pattern microstructure and locomotor activity were recorded over 2 h. The total food intake was increased by 120 and 240 mg/kg CBG-BDS (1.53 and 1.36 g, respectively, vs. 0.56 g in vehicle-treated animals). Latency to feeding onset was dose dependently decreased at all doses, and 120 and 240 mg/kg doses increased both the number of meals consumed and the cumulative size of the first two meals. No significant effect was observed on ambulatory activity or rearing behaviour. CBG-BDS is a novel appetite stimulant, which may have greater potency than purified CBG, despite the absence of [INCREMENT]-tetrahydrocannabinol in the extract.


Subject(s)
Cannabinoids/pharmacology , Cannabis/chemistry , Hyperphagia/chemically induced , Plant Extracts/pharmacology , Animals , Appetite Stimulants/administration & dosage , Appetite Stimulants/pharmacology , Cachexia/drug therapy , Cannabinoids/administration & dosage , Dose-Response Relationship, Drug , Feeding Behavior/drug effects , Locomotion , Male , Plant Extracts/administration & dosage , Rats
7.
Nat Commun ; 8: 14014, 2017 01 10.
Article in English | MEDLINE | ID: mdl-28072397

ABSTRACT

Alcohol intake associates with overeating in humans. This overeating is a clinical concern, but its causes are puzzling, because alcohol (ethanol) is a calorie-dense nutrient, and calorie intake usually suppresses brain appetite signals. The biological factors necessary for ethanol-induced overeating remain unclear, and societal causes have been proposed. Here we show that core elements of the brain's feeding circuits-the hypothalamic Agrp neurons that are normally activated by starvation and evoke intense hunger-display electrical and biochemical hyperactivity on exposure to dietary doses of ethanol in brain slices. Furthermore, by circuit-specific chemogenetic interference in vivo, we find that the Agrp cell activity is essential for ethanol-induced overeating in the absence of societal factors, in single-housed mice. These data reveal how a widely consumed nutrient can paradoxically sustain brain starvation signals, and identify a biological factor required for appetite evoked by alcohol.


Subject(s)
Agouti-Related Protein/metabolism , Ethanol/adverse effects , Hyperphagia/chemically induced , Neurons/metabolism , Agouti-Related Protein/genetics , Animals , Eating/drug effects , Electrophysiology/methods , Female , Hyperphagia/pathology , Hypothalamus/cytology , Hypothalamus/drug effects , Male , Mice, Inbred C57BL , Mice, Transgenic , Neurons/drug effects , Single-Cell Analysis/methods
8.
J Cell Sci ; 129(9): 1855-65, 2016 05 01.
Article in English | MEDLINE | ID: mdl-26985063

ABSTRACT

Sustained endoplasmic reticulum (ER) stress disrupts normal cellular homeostasis and leads to the development of many types of human diseases, including metabolic disorders. TAK1 (also known as MAP3K7) is a member of the mitogen-activated protein kinase kinase kinase (MAP3K) family and is activated by a diverse set of inflammatory stimuli. Here, we demonstrate that TAK1 regulates ER stress and metabolic signaling through modulation of lipid biogenesis. We found that deletion of Tak1 increased ER volume and facilitated ER-stress tolerance in cultured cells, which was mediated by upregulation of sterol-regulatory-element-binding protein (SREBP)-dependent lipogenesis. In the in vivo setting, central nervous system (CNS)-specific Tak1 deletion upregulated SREBP-target lipogenic genes and blocked ER stress in the hypothalamus. Furthermore, CNS-specific Tak1 deletion prevented ER-stress-induced hypothalamic leptin resistance and hyperphagic obesity under a high-fat diet (HFD). Thus, TAK1 is a crucial regulator of ER stress in vivo, which could be a target for alleviation of ER stress and its associated disease conditions.


Subject(s)
Endoplasmic Reticulum Stress , Hypothalamus/metabolism , Leptin/metabolism , MAP Kinase Kinase Kinases/metabolism , Animals , Dietary Fats/adverse effects , Dietary Fats/pharmacology , Hyperphagia/chemically induced , Hyperphagia/genetics , Hyperphagia/metabolism , Hyperphagia/pathology , Hypothalamus/pathology , Leptin/genetics , MAP Kinase Kinase Kinases/genetics , Mice , Mice, Knockout , Obesity/chemically induced , Obesity/genetics , Obesity/metabolism , Obesity/pathology , Sterol Regulatory Element Binding Proteins/genetics , Sterol Regulatory Element Binding Proteins/metabolism
9.
PLoS One ; 11(3): e0151511, 2016.
Article in English | MEDLINE | ID: mdl-26977813

ABSTRACT

Type 2 diabetes mellitus (T2DM) is associated with a high incidence of non-alcoholic fatty liver disease (NAFLD) related to obesity and insulin resistance. Currently, medical interventions for NAFLD have focused on diet control and exercise to reduce body weight, and there is a requirement for effective pharmacological therapies. Sodium-glucose cotransporter 2 (SGLT2) inhibitors are oral antidiabetic drugs that promote the urinary excretion of glucose by blocking its reabsorption in renal proximal tubules. SGLT2 inhibitors lower blood glucose independent of insulin action and are expected to reduce body weight because of urinary calorie loss. Here we show that an SGLT2 inhibitor ipragliflozin improves hepatic steatosis in high-fat diet-induced and leptin-deficient (ob/ob) obese mice irrespective of body weight reduction. In the obese mice, ipragliflozin-induced hyperphagia occurred to increase energy intake, attenuating body weight reduction with increased epididymal fat mass. There is an inverse correlation between weights of liver and epididymal fat in ipragliflozin-treated obese mice, suggesting that ipragliflozin treatment promotes normotopic fat accumulation in the epididymal fat and prevents ectopic fat accumulation in the liver. Despite increased adiposity, ipragliflozin ameliorates obesity-associated inflammation and insulin resistance in epididymal fat. Clinically, ipragliflozin improves liver dysfunction in patients with T2DM irrespective of body weight reduction. These findings provide new insight into the effects of SGLT2 inhibitors on energy homeostasis and fat accumulation and indicate their potential therapeutic efficacy in T2DM-associated hepatic steatosis.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Glucosides/therapeutic use , Hypoglycemic Agents/therapeutic use , Non-alcoholic Fatty Liver Disease/drug therapy , Obesity/complications , Thiophenes/therapeutic use , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Adult , Animals , Body Weight/drug effects , Diabetes Mellitus, Type 2/complications , Diet, High-Fat/adverse effects , Drug Evaluation, Preclinical , Energy Intake/drug effects , Epididymis/drug effects , Epididymis/metabolism , Glucose/metabolism , Glucosides/pharmacology , Humans , Hyperphagia/chemically induced , Hypoglycemic Agents/pharmacology , Insulin Resistance , Leptin/deficiency , Lipids/analysis , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/prevention & control , Organ Size/drug effects , Sodium-Glucose Transporter 2 , Sodium-Glucose Transporter 2 Inhibitors , Thiophenes/pharmacology , Weight Loss
10.
Physiol Behav ; 154: 175-83, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26596703

ABSTRACT

Consumption of sugar-sweetened beverages is associated with overweight and obesity. In this study, we hypothesized that obesity-prone (OP) mice fed a high-fat high-sucrose diet (HFHS) are more sensitive to consumption of sucrose-sweetened water (SSW) than obesity-resistant (OR) mice. After 3weeks of ad libitum access to the HFHS diet (7.5h/day), 180 male mice were classified as either OP (upper quartile of body weight gain, 5.2±0.1g, n=45) or OR (lower quartile, 3.2±0.1g, n=45). OP and OR mice were subsequently divided into 3 subgroups that had access to HFHS (7.5h/day) for 16weeks, supplemented with: i) water (OP/water and OR/water); ii) water and SSW (12.6% w/v), available for 2h/day randomly when access to HFHS was available and for 5 randomly-chosen days/week (OP/SSW and OR/SSW); or iii) water and SSW for 8weeks, then only water for 8weeks (OP/SSW-water and OR/SSW-water). OR/SSW mice decreased their food intake compared to OR/water mice, while OP/SSW mice exhibited an increase in food and total energy intake compared to OP/water mice. OP/SSW mice also gained more body weight and fat mass than OP/water mice, showed an increase in liver triglycerides and developed insulin resistance. These effects were fully reversed in OP/SSW-water mice. In the gut, OR/SSW mice, but not OP/SSW mice, had an increase GLP-1 and CCK response to a liquid meal compared to mice drinking only water. OP/SSW mice had a decreased expression of melanocortin receptor 4 in the hypothalamus and increased expression of delta opioid receptor in the nucleus accumbens compared to OP/water mice when fasted that could explain the hyperphagia in these mice. When access to the sucrose solution was removed for 8weeks, OP mice had increased dopaminergic and opioidergic response to a sucrose solution. Thus, intermittent access to a sucrose solution in mice fed a HFHS diet induces changes in the gut and brain signaling, leading to increased energy intake and adverse metabolic consequences only in mice prone to HFHS-induced obesity.


Subject(s)
Diet, High-Fat/adverse effects , Hyperphagia/chemically induced , Obesity/physiopathology , Sucrose/adverse effects , Sweetening Agents/adverse effects , Animals , Body Composition , Body Weight , Cholecystokinin/genetics , Cholecystokinin/metabolism , Disease Models, Animal , Drinking , Eating , Energy Metabolism , Gene Expression Regulation/physiology , Glucagon-Like Peptide 1/genetics , Glucagon-Like Peptide 1/metabolism , Glucose Tolerance Test , Lipid Peroxidation/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Sucrose/metabolism , Sweetening Agents/metabolism
11.
J Endocrinol ; 224(3): 245-59, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25663705

ABSTRACT

The aim of this study was to understand whether high folic acid (HFA) exposure during the perigestational period induces metabolic dysfunction in the offspring, later in life. To do this, female Sprague-Dawley rats (G0) were administered a dose of folic acid (FA) recommended for pregnancy (control, C, 2 mg FA/kg of diet, n=5) or a high dose of FA (HFA, 40 mg FA/kg of diet, n=5). Supplementation began at mating and lasted throughout pregnancy and lactation. Body weight and food and fluid intake were monitored in G0 and their offspring (G1) till G1 were 13 months of age. Metabolic blood profiles were assessed in G1 at 3 and 13 months of age (3M and 13M respectively). Both G0 and G1 HFA females had increased body weight gain when compared with controls, particularly 22 (G0) and 10 (G1) weeks after FA supplementation had been stopped. G1 female offspring of HFA mothers had increased glycemia at 3M, and both female and male G1 offspring of HFA mothers had decreased glucose tolerance at 13M, when compared with matched controls. At 13M, G1 female offspring of HFA mothers had increased insulin and decreased adiponectin levels, and G1 male offspring of HFA mothers had increased levels of leptin, when compared with matched controls. In addition, feeding of fructose to adult offspring revealed that perigestational exposure to HFA renders female progeny more susceptible to developing metabolic unbalance upon such a challenge. The results of this work indicate that perigestational HFA exposure the affects long-term metabolic phenotype of the offspring, predisposing them to an insulin-resistant state.


Subject(s)
Folic Acid/adverse effects , Metabolic Diseases/chemically induced , Prenatal Exposure Delayed Effects/metabolism , Animals , Animals, Newborn , Dietary Supplements , Dose-Response Relationship, Drug , Female , Folic Acid/administration & dosage , Hyperphagia/chemically induced , Hyperphagia/metabolism , Male , Metabolic Diseases/metabolism , Pregnancy , Rats , Rats, Sprague-Dawley , Weight Gain/drug effects
12.
Int J Neuropsychopharmacol ; 17(5): 807-18, 2014 May.
Article in English | MEDLINE | ID: mdl-24468236

ABSTRACT

Excessive weight gain is a major metabolic side effect of second-generation antipsychotics (SGAs) in the treatment of schizophrenia. Ghrelin is an orexigenic hormone secreted mainly from the stomach, which can induce weight gain and hyperphagia through regulating neuropeptides at the hypothalamus. Accumulating evidence implicates a relationship between ghrelin signalling and SGA-induced hyperphagia and weight gain. We report that olanzapine (a SGA with high weight gain liability) potently and time-dependently up-regulate ghrelin and ghrelin signalling, leading to hyperphagia and weight gain in female Sprague-Dawley rats, an action reversed by i.c.v. injection of a ghrelin receptor (GHS-R1a) antagonist. These findings indicate a crucial role of ghrelin signalling in hyperphagia induced by olanzapine, supporting the notion that GHS-R1a antagonist may be useful for pharmacological treatment of SGA-induced weight gain resulted from hyperphagia.


Subject(s)
Antipsychotic Agents/adverse effects , Benzodiazepines/adverse effects , Ghrelin/metabolism , Hyperphagia/chemically induced , Hypothalamus/drug effects , Weight Gain/drug effects , Animals , Central Nervous System Agents/pharmacology , Eating/drug effects , Eating/physiology , Female , Ghrelin/blood , Hyperphagia/metabolism , Hypothalamus/metabolism , Neuropeptides/metabolism , Olanzapine , Oligopeptides/pharmacology , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Ghrelin/antagonists & inhibitors , Receptors, Ghrelin/metabolism , Signal Transduction/drug effects , Time Factors , Transcription Factors/metabolism , Up-Regulation/drug effects , Weight Gain/physiology
13.
Brain Res ; 1532: 44-55, 2013 Sep 26.
Article in English | MEDLINE | ID: mdl-23933097

ABSTRACT

Allopregnanolone (ALLO), a gamma-aminobutyric acid (GABA) type A receptor active neurosteroid, elicits hyperphagic response in rodents. Since GABA-A receptors are present on the peptidergic neurons in the hypothalamus, we were interested in finding out if ALLO and neuropeptide cocaine- and amphetamine-regulated transcript (CART) interact and influence feeding behavior. While subcutaneous ALLO treatment, for a period of 7 days, produced a significant increase in food intake and body weight, pretreatment with subthreshold dose of CART (intracerebroventricular) attenuated both the effects. On the other hand, subcutaneous administration of dehydroepiandrosterone sulfate (DHEAS; GABA-A inhibitor neurosteroid) for a period of 7 days resulted in a significant reduction in food intake and body weight. These effects of DHEAS were potentiated by intracerebroventricular pretreatment with subeffective dose of CART. The brains of ALLO-treated rats were processed for the immunohistochemical analysis of CART immunoreactive elements. ALLO treatment resulted in a significant reduction in CART immunoreactivity in the hypothalamic arcuate, paraventricular and lateral nuclei, and nucleus accumbens shell. The results of the present study suggest that ALLO and CART might interact in the brain, and influence food intake and body weight. However, further investigations are needed to clarify the precise mechanisms by which ALLO modulate feeding behavior.


Subject(s)
Eating/drug effects , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/pharmacology , Pregnanolone/pharmacology , Weight Gain/drug effects , Animals , Hyperphagia/chemically induced , Hypothalamus/metabolism , Male , Nerve Tissue Proteins/drug effects , Nucleus Accumbens/metabolism , Rats , Rats, Sprague-Dawley
14.
Obesity (Silver Spring) ; 21(9): 1850-7, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23408466

ABSTRACT

OBJECTIVE: To investigate the effect of risperidone on energy expenditure and weight gain in female C57BL/6J mice. DESIGN AND METHODS: Body weight and composition, food intake, energy expenditure, and activity were determined weekly. mRNA expression of uncoupling protein 1 in brown adipose tissue, orexin, and brain-derived neurotrophic factor in the hypothalamus were quantified using real-time PCR. RESULTS: Risperidone tended to induce a greater body weight gain (P = 0.052) and significantly higher food intake (P = 0.038) relative to the placebo-treated group. Risperidone-treated mice had a higher resting energy expenditure (P = 0.001) and total energy expenditure (TEE) (P = 0.005) than the placebo group. There were no effects of treatment, time, and treatment by time on non-resting (or activity-related) energy expenditure between groups. Risperidone-treated mice showed a significantly lesser locomotor activity than placebo-treated mice over 3 weeks (P < 0.001). Risperidone induced a higher UCP1 mRNA (P = 0.003) and a lower orexin mRNA (P = 0.001) than placebo. CONCLUSION: Risperidone-induced weight gain is associated with hyperphagia and a reduction in locomotor activity in C57BL/6J mice. Additionally, higher total and resting energy expenditure were accompanied by higher levels of UCP1 mRNA in BAT. The increased TEE could not offset the total intake of energy through risperidone-induced hyperphagia, therefore resulting in weight gain in female C57BL/6J mice.


Subject(s)
Antipsychotic Agents/adverse effects , Energy Intake/drug effects , Energy Metabolism/drug effects , Hyperphagia/chemically induced , Motor Activity/drug effects , Risperidone/adverse effects , Weight Gain/drug effects , Adipose Tissue, Brown/metabolism , Animals , Basal Metabolism/drug effects , Brain-Derived Neurotrophic Factor/metabolism , Female , Hyperphagia/metabolism , Hypothalamus/drug effects , Hypothalamus/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Ion Channels/genetics , Ion Channels/metabolism , Mice , Mice, Inbred C57BL , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Neuropeptides/genetics , Neuropeptides/metabolism , Obesity/chemically induced , Obesity/genetics , Obesity/metabolism , Orexins , Uncoupling Protein 1
15.
PLoS One ; 7(4): e36453, 2012.
Article in English | MEDLINE | ID: mdl-22558467

ABSTRACT

Single-minded 1 (Sim1) is a transcription factor necessary for development of the paraventricular nucleus of the hypothalamus (PVH). This nucleus is a critical regulator of appetite, energy expenditure and body weight. Previously we showed that Sim1(+/-) mice and conditional postnatal Sim1(-/-) mice exhibit hyperphagia, obesity, increased linear growth and susceptibility to diet-induced obesity, but no decrease in energy expenditure. Bilateral ablation of the PVH causes obesity due to hyperphagia and reduced energy expenditure. It remains unknown whether Sim1 neurons regulate energy expenditure. In this study, Sim1cre mice were bred to homozygous inducible diphtheria toxin receptor (iDTR) mice to generate mice expressing the simian DTR in Sim1 cells. In these mice, Sim1 neuron ablation was performed by intracerebroventricular (ICV) injection of diphtheria toxin. Compared to controls, mice with Sim1 neuron ablation became obese (with increased fat mass) on a chow diet due to increased food intake and reduced energy expenditure. In post-injection mice, we observed a strong inverse correlation between the degree of obesity and hypothalamic Sim1 expression. The reduction in baseline energy expenditure observed in these mice was accompanied by a reduction in activity. This reduction in activity did not fully account for the reduced energy expenditure as these mice exhibited decreased resting energy expenditure, decreased body temperature, decreased brown adipose tissue temperature, and decreased UCP1 expression suggesting an impairment of thermogenesis. In injected mice, hypothalamic gene expression of Sim1, oxytocin (OXT) and thyrotropin releasing hormone (TRH) was reduced by about 50%. These results demonstrate that Sim1 neurons in adult mice regulate both food intake and energy expenditure. Based on the body of work in the field, feeding regulation by Sim1 neurons likely occurs in both the PVH and medial amygdala, in contrast to energy expenditure regulation by Sim1 neurons, which likely is localized to the PVH.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Energy Metabolism , Hyperphagia/metabolism , Hyperphagia/pathology , Neurons/metabolism , Obesity/metabolism , Obesity/pathology , Repressor Proteins/metabolism , Animals , Body Weight/drug effects , Diphtheria Toxin/toxicity , Energy Metabolism/drug effects , Feeding Behavior/drug effects , Female , Gene Expression Regulation/drug effects , Heparin-binding EGF-like Growth Factor , Hyperphagia/chemically induced , Hyperphagia/genetics , Hypothalamus/drug effects , Hypothalamus/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Mice , Mice, Transgenic , Neurons/drug effects , Neuropeptides/metabolism , Obesity/chemically induced , Obesity/genetics , Thermogenesis/drug effects
16.
Mol Cell Biochem ; 368(1-2): 37-45, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22638648

ABSTRACT

The metabolic syndrome is recognized as a cluster of disturbances associated with obesity, type 2 diabetes and hypertension. Over the past two decades, the number of people with the metabolic syndrome has increased at an alarming rate. This increase is associated with the global epidemic of both obesity and diabetes. Cardiovascular mortality is increased among diabetics and obesity-related insulin-resistant patients, and obesity is currently recognized as independent risk factor for cardiovascular disease. We aimed to establish the effects of a short period of an altered diet on the heart using a rat model of hyperphagia-induced obesity (diet supplemented with sucrose and condensed milk for 8 weeks = DIO) compared to age-matched controls. Isolated, perfused hearts were subjected to global or regional ischaemia/reperfusion. Function on reperfusion was recorded and infarct size determined. A plasma lipid profile was established via HPLC-based methods and proteins involved in metabolic signalling determined either by western blotting or RT-PCR. 8 weeks of diet resulted in whole-body but not myocardial insulin resistance, increased plasma triglyceride and phospholipid levels as well as increased lipid peroxidation. Despite the similar baseline function, hearts from DIO animals showed significantly poorer postischaemic recovery than controls (41.9 % RPP recovery vs 57.9 %, P < 0.05, n = 7-11/group) but surprisingly, smaller infarct size (24.95 ± 1.97 vs 47.26 ± 4.05 % of the area at risk, P < 0.005, n = 8/group). Basal phosphorylation of PKB/Akt was elevated but IRS-1 and SERCA-2 expression severely downregulated. In conclusion, after only 8 weeks of a slight change in diet, the rat heart shows signs of metabolic remodelling. Some of these changes may be protective but others may be detrimental and eventually lead to maladaptation.


Subject(s)
Diet/adverse effects , Insulin Resistance , Myocardial Infarction/metabolism , Myocardial Infarction/physiopathology , Obesity/metabolism , Obesity/physiopathology , Animals , Hyperphagia/chemically induced , Hyperphagia/metabolism , Hyperphagia/pathology , Hyperphagia/physiopathology , Insulin Receptor Substrate Proteins/metabolism , Muscle Proteins/metabolism , Myocardial Infarction/chemically induced , Myocardial Infarction/mortality , Myocardium/metabolism , Myocardium/pathology , Obesity/chemically induced , Obesity/pathology , Phospholipids/blood , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Triglycerides/blood
17.
Amino Acids ; 43(3): 1265-75, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22200933

ABSTRACT

L-Arginine (L-Arg) is a conditionally essential amino acid and a natural constituent of dietary proteins. Studies in obese rats and type 2 diabetic humans have indicated that dietary supplementation with L-Arg can diminish gain in white adipose tissue (WAT) and improve insulin sensitivity. However, the effects of L-Arg on glucose homeostasis, body composition and energy metabolism remain unclear. In addition, no studies have, to our knowledge, examined whether L-Arg has beneficial effects as a dietary supplement in the mouse model. In the present study, we investigated the effects of L-Arg supplementation to male C57BL/6 mice on an array of physiological parameters. L-Arg supplemented mice were maintained on a low-protein diet and body composition, appetite regulation, glucose tolerance, insulin sensitivity and energy expenditure were evaluated. A significant reduction in epididymal WAT was observed in L-Arg supplemented mice compared with mice fed an isocaloric control diet. Surprisingly, the L-Arg supplemented animals were hyperphagic corresponding to a highly significant decrease in feed efficiency, as body weight developed in a similar pattern in both experimental groups. Glucose homeostasis experiments revealed a major effect of L-Arg supplementation on glucose tolerance and insulin sensitivity, interestingly, independent of a parallel regulation in whole-body adiposity. Increased L-Arg ingestion also raised energy expenditure; however, no concurrent effect on locomotor activity, substrate metabolism or expression of uncoupling proteins (UCP1 and UCP2) in adipose tissues was displayed. In conclusion, dietary L-Arg supplementation substantially affects an array of metabolic-associated parameters including a reduction in WAT, hyperphagia, improved insulin sensitivity and increased energy expenditure in mice fed a low-protein diet.


Subject(s)
Arginine/administration & dosage , Hypoglycemic Agents/administration & dosage , Adipose Tissue, White/drug effects , Adipose Tissue, White/pathology , Adiposity/drug effects , Animals , Arginine/adverse effects , Blood Glucose , Diet, Protein-Restricted/adverse effects , Dietary Supplements , Energy Intake/drug effects , Energy Metabolism/drug effects , Gene Expression/drug effects , Genes, Mitochondrial , Glucose/metabolism , Homeostasis , Hyperphagia/chemically induced , Hypoglycemic Agents/adverse effects , Insulin/blood , Insulin Resistance , Male , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Motor Activity/drug effects , Oxygen Consumption/drug effects
18.
PLoS One ; 6(6): e20571, 2011.
Article in English | MEDLINE | ID: mdl-21695181

ABSTRACT

The success of antipsychotic drug treatment in patients with schizophrenia is limited by the propensity of these drugs to induce hyperphagia, weight gain and other metabolic disturbances, particularly evident for olanzapine and clozapine. However, the molecular mechanisms involved in antipsychotic-induced hyperphagia remain unclear. Here, we investigate the effect of olanzapine administration on the regulation of hypothalamic mechanisms controlling food intake, namely neuropeptide expression and AMP-activated protein kinase (AMPK) phosphorylation in rats. Our results show that subchronic exposure to olanzapine upregulates neuropeptide Y (NPY) and agouti related protein (AgRP) and downregulates proopiomelanocortin (POMC) in the arcuate nucleus of the hypothalamus (ARC). This effect was evident both in rats fed ad libitum and in pair-fed rats. Of note, despite weight gain and increased expression of orexigenic neuropeptides, subchronic administration of olanzapine decreased AMPK phosphorylation levels. This reduction in AMPK was not observed after acute administration of either olanzapine or clozapine. Overall, our data suggest that olanzapine-induced hyperphagia is mediated through appropriate changes in hypothalamic neuropeptides, and that this effect does not require concomitant AMPK activation. Our data shed new light on the hypothalamic mechanism underlying antipsychotic-induced hyperphagia and weight gain, and provide the basis for alternative targets to control energy balance.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Benzodiazepines/pharmacology , Hyperphagia/chemically induced , Hypothalamus/enzymology , Intracellular Signaling Peptides and Proteins/metabolism , Neuropeptides/metabolism , Signal Transduction/drug effects , Weight Gain/drug effects , Agouti-Related Protein/genetics , Agouti-Related Protein/metabolism , Aminoimidazole Carboxamide/administration & dosage , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Animals , Arcuate Nucleus of Hypothalamus/drug effects , Arcuate Nucleus of Hypothalamus/metabolism , Benzodiazepines/administration & dosage , Clozapine/administration & dosage , Clozapine/pharmacology , Feeding Behavior/drug effects , Female , Gene Expression Regulation/drug effects , Hyperphagia/blood , Hypothalamus/drug effects , Injections, Intraventricular , Insulin/blood , Leptin/blood , Neuropeptide Y/genetics , Neuropeptide Y/metabolism , Olanzapine , Orexins , Phosphorylation/drug effects , Pro-Opiomelanocortin/genetics , Pro-Opiomelanocortin/metabolism , Rats , Rats, Sprague-Dawley , Ribonucleotides/administration & dosage , Ribonucleotides/pharmacology
19.
Cell Mol Neurobiol ; 31(7): 985-9, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21681559

ABSTRACT

Olanzapine is known to be advantageous with respect to outcome and drug compliance in patients with schizophrenia. However, olanzapine has adverse effects, including a higher incidence of weight gain and metabolic disturbances, when compared with those of other antipsychotic agents. The mechanisms underlying these adverse events remain obscure. Female rats were orally administered olanzapine (2 mg/kg) or vehicle once a day for 2 weeks to ascertain if hypothalamic AMP-activated protein kinase (AMPK) mediates olanzapine-induced weight gain and hyperphagia. Body weight and food intake in each rat were evaluated every day and every two days, respectively. After the termination of drug treatment, we measured the protein levels of AMPK and phosphorylated AMPK in the hypothalamus using western blot analyses. Olanzapine significantly increased body weight and food intake. The phosphorylation levels of AMPK were significantly elevated by olanzapine. These results suggest that activation of hypothalamic AMPK may mediate hyperphagia and weight gain induced by chronic treatment with olanzapine.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Antipsychotic Agents/pharmacology , Benzodiazepines/pharmacology , Hyperphagia/chemically induced , Hypothalamus/drug effects , Hypothalamus/enzymology , Weight Gain/drug effects , Animals , Body Weight/drug effects , Eating/drug effects , Female , Hyperphagia/metabolism , Olanzapine , Rats , Rats, Sprague-Dawley
20.
Exp Physiol ; 96(4): 439-50, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21239462

ABSTRACT

Cholecystokinin (CCK) provides a meal-related signal that activates brainstem neurons, which have reciprocal interconnections with the hypothalamic paraventricular nucleus. Neurons that express corticotrophin-releasing factor (CRF) in the hypothalamus possess anorexigenic effects and are activated during endotoxaemia. This study investigated the effects of CCK(1) receptor blockade on lipopolysaccharide (LPS)-induced hypophagia and hypothalamic CRF neuronal activation. Male Wistar rats were pretreated with a specific CCK(1) receptor antagonist (devazepide; 1 mg kg(-1); i.p.) or vehicle; 30 min later they received LPS (100 µg kg(-1); i.p.) or saline injection. Food intake, corticosterone responses and Fos-CRF and Fos-α-melanocyte-stimulating hormone (α-MSH) immunoreactivity in the hypothalamus and Fos-tyrosine hydroxylase immunoreactivity in the nucleus of the solitary tract (NTS) were evaluated. In comparison with saline treatment, LPS administration decreased food intake and increased plasma corticosterone levels, as well as the number of Fos-CRF and Fos- tyrosine hydroxylase double-labelled neurons in vehicle-pretreated rats; no change in Fos-α-MSH immunoreactivity was observed after LPS injection. In saline-treated animals, devazepide pretreatment increased food intake, but it did not modify other parameters compared with vehicle-pretreated rats. Devazepide pretreatment partly reversed LPS-induced hypophagia and Fos-CRF and brainstem neuronal activation. Devazepide did not modify the corticosterone and Fos-α-MSH responses in rats treated with LPS. In conclusion, the present data suggest that LPS-induced hypophagia is mediated at least in part by CCK effects, via CCK(1) receptor, on NTS and hypothalamic CRF neurons.


Subject(s)
Cholecystokinin/metabolism , Corticotropin-Releasing Hormone/metabolism , Endotoxins/pharmacology , Hyperphagia/metabolism , Hypothalamus/metabolism , Pituitary Hormone-Releasing Hormones/metabolism , Animals , Brain Stem/metabolism , Corticosterone/blood , Devazepide/pharmacology , Eating/drug effects , Endotoxemia/chemically induced , Endotoxemia/metabolism , Hyperphagia/chemically induced , Lipopolysaccharides , Male , Neurons/enzymology , Neurons/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Wistar , Receptor, Cholecystokinin A/antagonists & inhibitors , Receptor, Cholecystokinin A/metabolism , Solitary Nucleus/metabolism , Tyrosine 3-Monooxygenase/metabolism , alpha-MSH/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL