Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 20
1.
Cell Metab ; 36(5): 1076-1087.e4, 2024 May 07.
Article En | MEDLINE | ID: mdl-38653246

Approximately 1 in 4 people worldwide have non-alcoholic fatty liver disease (NAFLD); however, there are currently no medications to treat this condition. This study investigated the role of adiposity-associated orphan G protein-coupled receptor 75 (GPR75) in liver lipid accumulation. We profiled Gpr75 expression and report that it is most abundant in the brain. Next, we generated the first single-cell-level analysis of Gpr75 and identified a subpopulation co-expressed with key appetite-regulating hypothalamic neurons. CRISPR-Cas9-deleted Gpr75 mice fed a palatable western diet high in fat adjusted caloric intake to remain in energy balance, thereby preventing NAFLD. Consistent with mouse results, analysis of whole-exome sequencing data from 428,719 individuals (UK Biobank) revealed that variants in GPR75 are associated with a reduced likelihood of hepatic steatosis. Here, we provide a significant advance in understanding of the expression and function of GPR75, demonstrating that it is a promising pharmaceutical target for NAFLD treatment.


Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease , Receptors, G-Protein-Coupled , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/genetics , Animals , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/genetics , Mice , Humans , Male , Adipose Tissue/metabolism , Mice, Knockout , Liver/metabolism , Female , Adiposity
2.
Mol Metab ; 84: 101933, 2024 Jun.
Article En | MEDLINE | ID: mdl-38583571

OBJECTIVE: Alström Syndrome (AS), caused by biallelic ALMS1 mutations, includes obesity with disproportionately severe insulin resistant diabetes, dyslipidemia, and fatty liver. Prior studies suggest that hyperphagia is accounted for by loss of ALMS1 function in hypothalamic neurones, whereas disproportionate metabolic complications may be due to impaired adipose tissue expandability. We tested this by comparing the metabolic effects of global and mesenchymal stem cell (MSC)-specific Alms1 knockout. METHODS: Global Alms1 knockout (KO) mice were generated by crossing floxed Alms1 and CAG-Cre mice. A Pdgfrα-Cre driver was used to abrogate Alms1 function selectively in MSCs and their descendants, including preadipocytes. We combined metabolic phenotyping of global and Pdgfrα+ Alms1-KO mice on a 45% fat diet with measurements of body composition and food intake, and histological analysis of metabolic tissues. RESULTS: Assessed on 45% fat diet to promote adipose expansion, global Alms1 KO caused hyperphagia, obesity, insulin resistance, dyslipidaemia, and fatty liver. Pdgfrα-cre driven KO of Alms1 (MSC KO) recapitulated insulin resistance, fatty liver, and dyslipidaemia in both sexes. Other phenotypes were sexually dimorphic: increased fat mass was only present in female Alms1 MSC KO mice. Hyperphagia was not evident in male Alms1 MSC KO mice, but was found in MSC KO females, despite no neuronal Pdgfrα expression. CONCLUSIONS: Mesenchymal deletion of Alms1 recapitulates metabolic features of AS, including fatty liver. This confirms a key role for Alms1 in the adipose lineage, where its loss is sufficient to cause systemic metabolic effects and damage to remote organs. Hyperphagia in females may depend on Alms1 deficiency in oligodendrocyte precursor cells rather than neurones. AS should be regarded as a forme fruste of lipodystrophy.


Alstrom Syndrome , Mesenchymal Stem Cells , Mice, Knockout , Animals , Mice , Male , Female , Mesenchymal Stem Cells/metabolism , Alstrom Syndrome/metabolism , Alstrom Syndrome/genetics , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Insulin Resistance , Fatty Liver/metabolism , Fatty Liver/genetics , Obesity/metabolism , Obesity/genetics , Hyperphagia/metabolism , Hyperphagia/genetics , Adipose Tissue/metabolism , Mice, Inbred C57BL , Body Composition
3.
Curr Biol ; 34(8): 1646-1656.e4, 2024 04 22.
Article En | MEDLINE | ID: mdl-38518777

The obesity epidemic is principally driven by the consumption of more calories than the body requires. It is therefore essential that the mechanisms underpinning feeding behavior are defined. Neurons within the brainstem dorsal vagal complex (DVC) receive direct information from the digestive system and project to second-order regions in the brain to regulate food intake. Although γ-aminobutyric acid is expressed in the DVC (GABADVC), its function in this region has not been defined. In order to discover the unique gene expression signature of GABADVC cells, we used single-nucleus RNA sequencing (Nuc-seq), and this revealed 19 separate clusters. We next probed the function of GABADVC cells and discovered that the selective activation of GABADVC neurons significantly controls food intake and body weight. Optogenetic interrogation of GABADVC circuitry identified GABADVC → hypothalamic arcuate nucleus (ARC) projections as appetite suppressive without creating aversion. Electrophysiological analysis revealed that GABADVC → ARC stimulation inhibits hunger-promoting neuropeptide Y (NPY) neurons via GABA release. Adopting an intersectional genetics strategy, we clarify that the GABADVC → ARC circuit curbs food intake. These data identify GABADVC as a new modulator of feeding behavior and body weight and a controller of orexigenic NPY neuron activity, thereby providing insight into the neural underpinnings of obesity.


Arcuate Nucleus of Hypothalamus , Brain Stem , Feeding Behavior , Arcuate Nucleus of Hypothalamus/metabolism , Arcuate Nucleus of Hypothalamus/physiology , Animals , Brain Stem/physiology , Brain Stem/metabolism , Mice , Male , Feeding Behavior/physiology , GABAergic Neurons/physiology , GABAergic Neurons/metabolism , gamma-Aminobutyric Acid/metabolism , Eating/physiology , Mice, Inbred C57BL , Female
4.
bioRxiv ; 2023 Oct 13.
Article En | MEDLINE | ID: mdl-37873427

Background: Alström Syndrome (AS), a multi-system disease caused by mutations in the ALMS1 gene, includes obesity with disproportionately severe insulin resistant diabetes, dyslipidemia, and hepatosteatosis. How loss of ALMS1 causes this phenotype is poorly understood, but prior studies have circumstancially implicated impaired adipose tissue expandability. We set out to test this by comparing the metabolic effects of selective Alms1 knockout in mesenchymal cells including preadipocytes to those of global Alms1 knockout. Methods: Global Alms1 knockout (KO) mice were generated by crossing floxed Alms1 and CAG-Cre mice. A Pdgfrα -Cre driver was used to abrogate Alms1 function selectively in mesenchymal stem cells (MSCs) and their descendants, including preadipocytes. We combined metabolic phenotyping of global and Pdgfrα + Alms1 -KO mice on a 45% fat diet with measurements of body composition and food intake, and histological analysis of metabolic tissues. Results: Global Alms1 KO caused hyperphagia, obesity, insulin resistance, dyslipidaemia, and fatty liver. Pdgfrα - cre driven KO of Alms1 (MSC KO) recapitulated insulin resistance, fatty liver, and dyslipidaemia in both sexes. Other phenotypes were sexually dimorphic: increased fat mass was only present in female Alms1 MSC KO mice. Hyperphagia was not evident in male Alms1 MSC KO mice, but was found in MSC KO females, despite no neuronal Pdgfr α expression. Conclusions: Mesenchymal deletion of Alms1 recapitulates the metabolic features of AS, including severe fatty liver. This confirms a key role for Alms1 in the adipose lineage, where its loss is sufficient to cause systemic metabolic effects and damage to remote organs. AS should be regarded as a forme fruste of lipodystrophy. Therapies should prioritise targeting positive energy balance.

6.
Mol Metab ; 68: 101665, 2023 02.
Article En | MEDLINE | ID: mdl-36592795

OBJECTIVE: Overweight and obesity are endemic in developed countries, with a substantial negative impact on human health. Medications developed to treat obesity include agonists for the G-protein coupled receptors glucagon-like peptide-1 (GLP-1R; e.g. liraglutide), serotonin 2C (5-HT2CR; e.g, lorcaserin), and melanocortin4 (MC4R) which reduce body weight primarily by suppressing food intake. However, the mechanisms underlying the therapeutic food intake suppressive effects are still being defined and were investigated here. METHODS: We profiled PPG neurons in the nucleus of the solitary tract (PPGNTS) using single nucleus RNA sequencing (Nuc-Seq) and histochemistry. We next examined the requirement of PPGNTS neurons for obesity medication effects on food intake by virally ablating PPGNTS neurons. Finally, we assessed the effects on food intake of the combination of liraglutide and lorcaserin. RESULTS: We found that 5-HT2CRs, but not GLP-1Rs or MC4Rs, were widespread in PPGNTS clusters and that lorcaserin significantly activated PPGNTS neurons. Accordingly, ablation of PPGNTS neurons prevented the reduction of food intake by lorcaserin but not MC4R agonist melanotan-II, demonstrating the functional significance of PPGNTS 5-HT2CR expression. Finally, the combination of lorcaserin with GLP-1R agonists liraglutide or exendin-4 produced greater food intake reduction as compared to either monotherapy. CONCLUSIONS: These findings identify a necessary mechanism through which obesity medication lorcaserin produces its therapeutic benefit, namely brainstem PPGNTS neurons. Moreover, these data reveal a strategy to augment the therapeutic profile of the current frontline treatment for obesity, GLP-1R agonists, via coadministration with 5-HT2CR agonists.


Glucagon-Like Peptide 1 , Liraglutide , Humans , Glucagon-Like Peptide 1/pharmacology , Glucagon-Like Peptide 1/metabolism , Liraglutide/pharmacology , Liraglutide/therapeutic use , Glucagon-Like Peptide-1 Receptor/metabolism , Serotonin/metabolism , Appetite , Obesity/drug therapy , Obesity/metabolism , Solitary Nucleus/metabolism , Eating , Neurons/metabolism
7.
FASEB J ; 35(1): e21161, 2021 01.
Article En | MEDLINE | ID: mdl-33156577

Association of G protein-coupled receptors into heterodimeric complexes has been reported for over 50 receptor pairs in vitro but functional in vivo validation remains a challenge. Our recent in vitro studies defined the functional fingerprint of heteromers composed of Gi -coupled melatonin MT2 receptors and Gq -coupled serotonin 5-HT2C receptors, in which melatonin transactivates phospholipase C (PLC) through 5-HT2C . Here, we identified this functional fingerprint in the mouse brain. Gq protein activation was probed by [35 S]GTPγS incorporation followed by Gq immunoprecipitation, and PLC activation by determining the inositol phosphate levels in brain lysates of animals previously treated with melatonin. Melatonin concentration-dependently activated Gq proteins and PLC in the hypothalamus and cerebellum but not in cortex. These effects were inhibited by the 5-HT2C receptor-specific inverse agonist SB-243213, and were absent in MT2 and 5-HT2C knockout mice, fully recapitulating previous in vitro data and indicating the involvement of MT2 /5-HT2C heteromers. The antidepressant agomelatine had a similar effect than melatonin when applied alone but blocked the melatonin-promoted Gq activation due to its 5-HT2C antagonistic component. Collectively, we provide strong functional evidence for the existence of MT2 /5-HT2C heteromeric complexes in mouse brain. These heteromers might participate in the in vivo effects of agomelatine.


Brain/metabolism , Gene Expression Regulation, Enzymologic , Protein Multimerization , Receptor, Melatonin, MT2/metabolism , Receptor, Serotonin, 5-HT2C/metabolism , Transcriptional Activation , Type C Phospholipases/biosynthesis , Acetamides/pharmacology , Animals , Indoles/pharmacology , Male , Mice , Mice, Knockout , Pyridines/pharmacology , Receptor, Melatonin, MT2/genetics , Receptor, Serotonin, 5-HT2C/genetics , Type C Phospholipases/genetics
8.
Cell Rep ; 25(2): 413-423.e5, 2018 10 09.
Article En | MEDLINE | ID: mdl-30304681

Compelling evidence has shown that, besides its putative effect on the regulation of the gonadal axis, estradiol (E2) exerts a dichotomic effect on the hypothalamus to regulate food intake and energy expenditure. The anorectic effect of E2 is mainly mediated by its action on the arcuate nucleus (ARC), whereas its effects on brown adipose tissue (BAT) thermogenesis occur in the ventromedial nucleus (VMH). Here, we demonstrate that central E2 decreases hypothalamic ceramide levels and endoplasmic reticulum (ER) stress. Pharmacological or genetic blockade of ceramide synthesis and amelioration of ER stress selectively occurring in the VMH recapitulate the effect of E2, leading to increased BAT thermogenesis, weight loss, and metabolic improvement. These findings demonstrate that E2 regulation of ceramide-induced hypothalamic lipotoxicity and ER stress is an important determinant of energy balance, suggesting that dysregulation of this mechanism may underlie some changes in energy homeostasis seen in females.


Adipose Tissue, Brown/physiology , Ceramides/toxicity , Endoplasmic Reticulum Stress/drug effects , Energy Metabolism/drug effects , Estradiol/pharmacology , Hypothalamus/physiology , Thermogenesis/drug effects , Adipose Tissue, Brown/drug effects , Animals , Estrogens/pharmacology , Female , Homeostasis , Hypothalamus/drug effects , Rats
9.
J Endocrinol ; 238(3): 177-186, 2018 09.
Article En | MEDLINE | ID: mdl-29914932

Current evidence suggests that estradiol (E2), the main ovarian steroid, modulates energy balance by regulating both feeding and energy expenditure at the central level, through the energy sensor AMP-activated protein kinase (AMPK). We hypothesized that the hypothalamic mechanistic target of rapamycin (mTOR) pathway, a well-established nutrient sensor and modulator of appetite and puberty, could also mediate the anorectic effect of E2. Our data showed that ovariectomy (OVX) elicited a marked downregulation of the mTOR signaling in the arcuate nucleus of the hypothalamus (ARC), an effect that was reversed by either E2 replacement or central estrogen receptor alpha (ERα) agonism. The significance of this molecular signaling was given by the genetic inactivation of S6 kinase B1 (S6K1, a key downstream mTOR effector) in the ARC, which prevented the E2-induced hypophagia and weight loss. Overall, these data indicate that E2 induces hypophagia through modulation of mTOR pathway in the ARC.


Anorexia/chemically induced , Anorexia/metabolism , Arcuate Nucleus of Hypothalamus/drug effects , Estradiol/pharmacology , TOR Serine-Threonine Kinases/metabolism , Animals , Appetite Depressants/pharmacology , Arcuate Nucleus of Hypothalamus/metabolism , Body Weight/drug effects , Eating/drug effects , Eating/physiology , Female , Ovariectomy , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Signal Transduction/physiology , TOR Serine-Threonine Kinases/physiology
10.
Cell Metab ; 26(1): 212-229.e12, 2017 Jul 05.
Article En | MEDLINE | ID: mdl-28683288

Thyroid hormones (THs) act in the brain to modulate energy balance. We show that central triiodothyronine (T3) regulates de novo lipogenesis in liver and lipid oxidation in brown adipose tissue (BAT) through the parasympathetic (PSNS) and sympathetic nervous system (SNS), respectively. Central T3 promotes hepatic lipogenesis with parallel stimulation of the thermogenic program in BAT. The action of T3 depends on AMP-activated protein kinase (AMPK)-induced regulation of two signaling pathways in the ventromedial nucleus of the hypothalamus (VMH): decreased ceramide-induced endoplasmic reticulum (ER) stress, which promotes BAT thermogenesis, and increased c-Jun N-terminal kinase (JNK) activation, which controls hepatic lipid metabolism. Of note, ablation of AMPKα1 in steroidogenic factor 1 (SF1) neurons of the VMH fully recapitulated the effect of central T3, pointing to this population in mediating the effect of central THs on metabolism. Overall, these findings uncover the underlying pathways through which central T3 modulates peripheral metabolism.


Energy Metabolism , Hypothalamus/metabolism , Mitogen-Activated Protein Kinase 8/metabolism , Signal Transduction , Thyroid Hormones/metabolism , Adipose Tissue, Brown/metabolism , Animals , Lipid Metabolism , Liver/metabolism , Male , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley , Thermogenesis , Triiodothyronine/metabolism
11.
Clin Sci (Lond) ; 130(19): 1697-709, 2016 10 01.
Article En | MEDLINE | ID: mdl-27555613

Feeding behaviour and energy storage are both crucial aspects of survival. Thus, it is of fundamental importance to understand the molecular mechanisms regulating these basic processes. The AMP-activated protein kinase (AMPK) has been revealed as one of the key molecules modulating energy homoeostasis. Indeed, AMPK appears to be essential for translating nutritional and energy requirements into generation of an adequate neuronal response, particularly in two areas of the brain, the hypothalamus and the hindbrain. Failure of this physiological response can lead to energy imbalance, ultimately with extreme consequences, such as leanness or obesity. Here, we will review the data that put brain AMPK in the spotlight as a regulator of appetite.


Brain/enzymology , Obesity/enzymology , Protein Kinases/metabolism , AMP-Activated Protein Kinase Kinases , Animals , Appetite/drug effects , Energy Metabolism , Humans , Obesity/genetics , Obesity/psychology , Protein Kinases/genetics
12.
Cell Metab ; 23(5): 821-36, 2016 May 10.
Article En | MEDLINE | ID: mdl-27133129

Despite significant advances in our understanding of the biology determining systemic energy homeostasis, the treatment of obesity remains a medical challenge. Activation of AMP-activated protein kinase (AMPK) has been proposed as an attractive strategy for the treatment of obesity and its complications. AMPK is a conserved, ubiquitously expressed, heterotrimeric serine/threonine kinase whose short-term activation has multiple beneficial metabolic effects. Whether these translate into long-term benefits for obesity and its complications is unknown. Here, we observe that mice with chronic AMPK activation, resulting from mutation of the AMPK γ2 subunit, exhibit ghrelin signaling-dependent hyperphagia, obesity, and impaired pancreatic islet insulin secretion. Humans bearing the homologous mutation manifest a congruent phenotype. Our studies highlight that long-term AMPK activation throughout all tissues can have adverse metabolic consequences, with implications for pharmacological strategies seeking to chronically activate AMPK systemically to treat metabolic disease.


AMP-Activated Protein Kinases/metabolism , Insulin-Secreting Cells/enzymology , Insulin-Secreting Cells/pathology , Obesity/enzymology , Adiposity/genetics , Adult , Aging/pathology , Agouti-Related Protein/metabolism , Animals , Arcuate Nucleus of Hypothalamus/metabolism , Energy Metabolism/genetics , Enzyme Activation , Feeding Behavior , Female , Heterozygote , Humans , Hyperphagia/complications , Hyperphagia/enzymology , Hyperphagia/genetics , Hyperphagia/pathology , Hypothalamus/metabolism , Insulin/metabolism , Male , Mice , Mitochondria/metabolism , Mutation/genetics , Neurons/metabolism , Obesity/blood , Obesity/complications , Obesity/pathology , Oxidative Phosphorylation , Receptors, Ghrelin/metabolism , Ribosomes/metabolism , Signal Transduction/genetics , Transcriptome/genetics , Up-Regulation/genetics
13.
Mol Metab ; 5(3): 245-252, 2016 Mar.
Article En | MEDLINE | ID: mdl-26977396

OBJECTIVE: Obesity is one of the primary healthcare challenges of the 21st century. Signals relaying information regarding energy needs are integrated within the brain to influence body weight. Central among these integration nodes are the brain pro-opiomelanocortin (POMC) peptides, perturbations of which disrupt energy balance and promote severe obesity. However, POMC neurons are neurochemically diverse and the crucial source of POMC peptides that regulate energy homeostasis and body weight remains to be fully clarified. METHODS: Given that a 5-hydroxytryptamine 2c receptor (5-HT2CR) agonist is a current obesity medication and 5-HT2CR agonist's effects on appetite are primarily mediated via POMC neurons, we hypothesized that a critical source of POMC regulating food intake and body weight is specifically synthesized in cells containing 5-HT2CRs. To exclusively manipulate Pomc synthesis only within 5-HT2CR containing cells, we generated a novel 5-HT 2C R (CRE) mouse line and intercrossed it with Cre recombinase-dependent and hypothalamic specific reactivatable Pomc (NEO) mice to restrict Pomc synthesis to the subset of hypothalamic cells containing 5-HT2CRs. This provided a means to clarify the specific contribution of a defined subgroup of POMC peptides in energy balance and body weight. RESULTS: Here we transform genetically programed obese and hyperinsulinemic male mice lacking hypothalamic Pomc with increased appetite, reduced physical activity and compromised brown adipose tissue (BAT) into lean, healthy mice via targeted restoration of Pomc function only within 5-HT2CR expressing cells. Remarkably, the same metabolic transformation does not occur in females, who despite corrected feeding behavior and normalized insulin levels remain physically inactive, have lower energy expenditure, compromised BAT and develop obesity. CONCLUSIONS: These data provide support for the functional heterogeneity of hypothalamic POMC neurons, revealing that Pomc expression within 5-HT2CR expressing neurons is sufficient to regulate energy intake and insulin sensitivity in male and female mice. However, an unexpected sex difference in the function of this subset of POMC neurons was identified with regard to energy expenditure. We reveal that a large sex difference in physical activity, energy expenditure and the development of obesity is driven by this subpopulation, which constitutes approximately 40% of all POMC neurons in the hypothalamic arcuate nucleus. This may have broad implications for strategies utilized to combat obesity, which at present largely ignore the sex of the obese individual.

14.
Endocrinology ; 156(3): 947-60, 2015 Mar.
Article En | MEDLINE | ID: mdl-25535827

During gestation, hyperphagia is necessary to cope with the metabolic demands of embryonic development. There were three main aims of this study: Firstly, to investigate the effect of pregnancy on hypothalamic fatty acid metabolism, a key pathway for the regulation of energy balance; secondly, to study whether pregnancy induces resistance to the anorectic effect of fatty acid synthase (FAS) inhibition and accumulation of malonyl-coenzyme A (CoA) in the hypothalamus; and, thirdly, to study whether changes in hypothalamic AMPK signaling are associated with brown adipose tissue (BAT) thermogenesis during pregnancy. Our data suggest that in pregnant rats, the hypothalamic fatty acid pathway shows an overall state that should lead to anorexia and elevated BAT thermogenesis: decreased activities of AMP-activated protein kinase (AMPK), FAS, and carnitine palmitoyltransferase 1, coupled with increased acetyl-CoA carboxylase function with subsequent elevation of malonyl-CoA levels. This profile seems dependent of estradiol levels but not prolactin or progesterone. Despite the apparent anorexic and thermogenic signaling in the hypothalamus, pregnant rats remain hyperphagic and display reduced temperature and BAT function. Actually, pregnant rats develop resistance to the anorectic effects of central FAS inhibition, which is associated with a reduction of proopiomelanocortin (POMC) expression and its transcription factors phospho-signal transducer and activator of transcription 3, and phospho-forkhead box O1. This evidence demonstrates that pregnancy induces a state of resistance to the anorectic and thermogenic actions of hypothalamic cellular signals of energy surplus, which, in parallel to the already known refractoriness to leptin effects, likely contributes to gestational hyperphagia and adiposity.


AMP-Activated Protein Kinases/metabolism , Anorexia/chemically induced , Body Temperature Regulation/drug effects , Hypothalamus/metabolism , Malonyl Coenzyme A/pharmacology , Adipose Tissue, Brown/physiology , Animals , Fatty Acids/biosynthesis , Female , Gene Expression Regulation, Enzymologic , Lipid Metabolism/physiology , Malonyl Coenzyme A/metabolism , Ovariectomy , Pregnancy , Rats , Rats, Sprague-Dawley
15.
Cell Metab ; 20(1): 41-53, 2014 Jul 01.
Article En | MEDLINE | ID: mdl-24856932

Estrogens play a major role in the modulation of energy balance through central and peripheral actions. Here, we demonstrate that central action of estradiol (E2) inhibits AMP-activated protein kinase (AMPK) through estrogen receptor alpha (ERα) selectively in the ventromedial nucleus of the hypothalamus (VMH), leading to activation of thermogenesis in brown adipose tissue (BAT) through the sympathetic nervous system (SNS) in a feeding-independent manner. Genetic activation of AMPK in the VMH prevented E2-induced increase in BAT-mediated thermogenesis and weight loss. Notably, fluctuations in E2 levels during estrous cycle also modulate this integrated physiological network. Together, these findings demonstrate that E2 regulation of the VMH AMPK-SNS-BAT axis is an important determinant of energy balance and suggest that dysregulation in this axis may account for the common changes in energy homeostasis and obesity linked to dysfunction of the female gonadal axis.


AMP-Activated Protein Kinases/metabolism , Adipose Tissue, Brown/metabolism , Estradiol/pharmacology , Hypothalamus/drug effects , Thermogenesis/drug effects , AMP-Activated Protein Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases/chemistry , Animals , Energy Metabolism/drug effects , Estrogen Receptor alpha/metabolism , Female , Hypothalamus/enzymology , Hypothalamus/metabolism , Ovary/injuries , Rats , Rats, Sprague-Dawley , Rats, Wistar , Sympathetic Nervous System/metabolism
16.
Endocrinology ; 155(5): 1679-89, 2014 May.
Article En | MEDLINE | ID: mdl-24517227

Nicotine, the main addictive component of tobacco, promotes body weight reduction in humans and rodents. Recent evidence has suggested that nicotine acts in the central nervous system to modulate energy balance. Specifically, nicotine modulates hypothalamic AMP-activated protein kinase to decrease feeding and to increase brown adipose tissue thermogenesis through the sympathetic nervous system, leading to weight loss. Of note, most of this evidence has been obtained in animal models fed with normal diet or low-fat diet (LFD). However, its effectiveness in obese models remains elusive. Because obesity causes resistance towards many factors involved in energy homeostasis, the aim of this study has been to compare the effect of nicotine in a diet-induced obese (DIO) model, namely rats fed a high-fat diet, with rats fed a LFD. Our data show that chronic peripheral nicotine treatment reduced body weight by decreasing food intake and increasing brown adipose tissue thermogenesis in both LFD and DIO rats. This overall negative energy balance was associated to decreased activation of hypothalamic AMP-activated protein kinase in both models. Furthermore, nicotine improved serum lipid profile, decreased insulin serum levels, as well as reduced steatosis, inflammation, and endoplasmic reticulum stress in the liver of DIO rats but not in LFD rats. Overall, this evidence suggests that nicotine diminishes body weight and improves metabolic disorders linked to DIO and might offer a clear-cut strategy to develop new therapeutic approaches against obesity and its metabolic complications.


Appetite Depressants/therapeutic use , Appetite Regulation/drug effects , Endoplasmic Reticulum Stress/drug effects , Fatty Liver/prevention & control , Nicotine/therapeutic use , Nicotinic Agonists/therapeutic use , Obesity/drug therapy , AMP-Activated Protein Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases/metabolism , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/metabolism , Animals , Appetite Depressants/administration & dosage , Appetite Depressants/adverse effects , Diet, Fat-Restricted , Diet, High-Fat/adverse effects , Dyslipidemias/etiology , Dyslipidemias/prevention & control , Fatty Liver/etiology , Hyperinsulinism/etiology , Hyperinsulinism/prevention & control , Hypothalamus/drug effects , Hypothalamus/enzymology , Hypothalamus/metabolism , Injections, Subcutaneous , Liver/drug effects , Liver/enzymology , Liver/metabolism , Liver/pathology , Male , Nicotine/administration & dosage , Nicotine/adverse effects , Nicotinic Agonists/administration & dosage , Nicotinic Agonists/adverse effects , Non-alcoholic Fatty Liver Disease , Obesity/diet therapy , Obesity/etiology , Obesity/metabolism , Rats , Rats, Sprague-Dawley , Thermogenesis/drug effects , Weight Loss/drug effects
17.
Brain Res ; 1450: 40-8, 2012 Apr 23.
Article En | MEDLINE | ID: mdl-22425186

Thyroid hormones play an important role in the regulation of energy balance, sleep and emotional behaviors. Neuropeptide S (NPS) is a recently discovered neuropeptide, regulating feeding, sleep and anxiety. Here, we examined the effect of hyperthyroidism on the gene and protein expression of neuropeptide S and its receptor (NPS-R) in the hypothalamus, brainstem and amygdala of rats. Our results showed that the expression of NPS and NPS-R was differentially modulated by hyperthyroidism in the rat brain. NPS and NPS-R mRNA and protein levels were decreased in the hypothalamus of hyperthyroid rats. Conversely NPS-R expression was highly increased in the brainstem and NPS and NPS-R expression were unchanged in the amygdala of these rats. These data suggest that changes in anxiety and food intake patterns observed in hyperthyroidism could be associated with changes in the expression of NPS and NPS-R. Thus, the NPS/NPS-R system may be involved in several hyperthyroidism-associated comorbidities.


Amygdala/metabolism , Brain Stem/metabolism , Hyperthyroidism/metabolism , Hypothalamus/metabolism , Neuropeptides/metabolism , Animals , Eating/genetics , Gene Expression , Hyperthyroidism/genetics , Male , Neuropeptides/genetics , Rats , Rats, Sprague-Dawley , Sleep/genetics
18.
Diabetes ; 61(4): 807-17, 2012 Apr.
Article En | MEDLINE | ID: mdl-22315316

Smokers around the world commonly report increased body weight after smoking cessation as a major factor that interferes with their attempts to quit. Numerous controlled studies in both humans and rodents have reported that nicotine exerts a marked anorectic action. The effects of nicotine on energy homeostasis have been mostly pinpointed in the central nervous system, but the molecular mechanisms controlling its action are still not fully understood. The aim of this study was to investigate the effect of nicotine on hypothalamic AMP-activated protein kinase (AMPK) and its effect on energy balance. Here we demonstrate that nicotine-induced weight loss is associated with inactivation of hypothalamic AMPK, decreased orexigenic signaling in the hypothalamus, increased energy expenditure as a result of increased locomotor activity, increased thermogenesis in brown adipose tissue (BAT), and alterations in fuel substrate utilization. Conversely, nicotine withdrawal or genetic activation of hypothalamic AMPK in the ventromedial nucleus of the hypothalamus reversed nicotine-induced negative energy balance. Overall these data demonstrate that the effects of nicotine on energy balance involve specific modulation of the hypothalamic AMPK-BAT axis. These targets may be relevant for the development of new therapies for human obesity.


AMP-Activated Protein Kinases/metabolism , Eating/drug effects , Energy Metabolism/drug effects , Nicotine/pharmacology , AMP-Activated Protein Kinases/genetics , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/metabolism , Animals , Appetite/drug effects , Gene Expression Regulation/drug effects , Hypothalamus/metabolism , Male , Motor Activity/drug effects , Neuropeptide Y/metabolism , Pro-Opiomelanocortin/genetics , Pro-Opiomelanocortin/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Nicotinic/genetics , Receptors, Nicotinic/metabolism , Temazepam , Thermogenesis/drug effects
19.
Front Horm Res ; 38: 196-205, 2010.
Article En | MEDLINE | ID: mdl-20616512

Ghrelin, a peptide hormone first discovered as the endogenous ligand of the growth hormone secretagogue receptor (GHS-R), is predominantly produced and released into the circulation by ghrelin cells (X/A-like) of the stomach fundus cells. Ghrelin has multiple actions in multiple tissues. In particular, it is the most potent known endogenous orexigenic peptide, and plays a significant role in glucose homeostasis: deletion of the genes encoding ghrelin and/or its receptor prevents high-fat diet from inducing obesity, increases insulin levels, enhances glucose-stimulated insulin secretion and improves peripheral insulin sensitivity. In addition to its already mentioned roles, ghrelin has other activities including stimulation of pituitary hormones secretion, regulation of gastric and pancreatic activity, modulation of fatty acid metabolism via specific control of AMP-activated protein kinase (AMPK), and cardiovascular and hemodynamic activities. In addition, modulation of cartilage and bone homeostasis, sleep and behavioral influences, and modulation of the immune system, as well as effects on cell proliferation, are other relevant actions of ghrelin. In this review, we summarize several aspects of ghrelin effects at hypothalamic level and their implications in the control of food intake and energy balance.


Ghrelin/physiology , Agouti-Related Protein/physiology , Animals , Body Weight , Eating , Homeodomain Proteins/physiology , Humans , Hypothalamus/metabolism , Lipid Metabolism , Neuropeptide Y/physiology , Receptors, Ghrelin/physiology
20.
Biochim Biophys Acta ; 1801(3): 350-61, 2010 Mar.
Article En | MEDLINE | ID: mdl-19796707

Ectopic accumulation of lipids in peripheral tissues, such as pancreatic beta cells, liver, heart and skeletal muscle, leads to lipotoxicity, a process that contributes substantially to the pathophysiology of insulin resistance, type 2 diabetes, steatotic liver disease and heart failure. Current evidence has demonstrated that hypothalamic sensing of circulating lipids and modulation of hypothalamic endogenous fatty acid and lipid metabolism are two bona fide mechanisms modulating energy homeostasis at the whole body level. Key enzymes, such as AMP-activated protein kinase (AMPK) and fatty acid synthase (FAS), as well as intermediate metabolites, such as malonyl-CoA and long-chain fatty acids-CoA (LCFAs-CoA), play a major role in this neuronal network, integrating peripheral signals with classical neuropeptide-based mechanisms. However, one key question to be addressed is whether impairment of lipid metabolism and accumulation of specific lipid species in the hypothalamus, leading to lipotoxicity, have deleterious effects on hypothalamic neurons. In this review, we summarize what is known about hypothalamic lipid metabolism with focus on the events associated to lipotoxicity, such as endoplasmic reticulum (ER) stress in the hypothalamus. A better understanding of these molecular mechanisms will help to identify new drug targets for the treatment of obesity and metabolic syndrome.


Hypothalamus/metabolism , Lipid Metabolism , Metabolic Syndrome/metabolism , Animals , Endoplasmic Reticulum/metabolism , Energy Metabolism , Homeostasis , Humans , Models, Biological , Stress, Physiological
...