Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Front Behav Neurosci ; 16: 910461, 2022.
Article in English | MEDLINE | ID: mdl-35722192

ABSTRACT

In addition to the well-known motor control, the cerebellum has recently been implicated in memory, cognition, addiction, and social behavior. Given that the cerebellum contains more neurons than the cerebral cortex and has tight connections to the thalamus and brainstem nuclei, it is possible that the cerebellum also regulates sleep/wakefulness. However, the role of the cerebellum in sleep was unclear, since cerebellar lesion studies inevitably involved massive inflammation in the adjacent brainstem, and sleep changes in lesion studies were not consistent with each other. Here, we examine the role of the cerebellum in sleep and wakefulness using mesencephalon- and rhombomere 1-specific Ptf1a conditional knockout (Ptf1a cKO) mice, which lack the cerebellar cortex and its related structures, and exhibit ataxic gait. Ptf1a cKO mice had similar wake and non-rapid eye movement sleep (NREMS) time as control mice and showed reduced slow wave activity during wakefulness, NREMS and REMS. Ptf1a cKO mice showed a decrease in REMS time during the light phase and had increased NREMS delta power in response to 6 h of sleep deprivation, as did control mice. Ptf1a cKO mice also had similar numbers of sleep spindles and fear memories as control mice. Thus, the cerebellum does not appear to play a major role in sleep-wake control, but may be involved in the generation of slow waves.

2.
Mol Metab ; 59: 101465, 2022 05.
Article in English | MEDLINE | ID: mdl-35218946

ABSTRACT

OBJECTIVE: Bone morphogenetic protein 8B (BMP8B) plays a major role in the regulation of energy homeostasis by modulating brown adipose tissue (BAT) thermogenesis and white adipose tissue (WAT) browning. Here, we investigated whether BMP8B's role in metabolism is affected by obesity and the possible molecular mechanisms underlying that action. METHODS: Central treatments with BMP8B were performed in rats fed a standard (SD) and high-fat diet (HFD), as well as in genetically modified mice. Energy balance studies, infrared thermographic analysis of BAT and molecular analysis of the hypothalamus, BAT and WAT were carried out. RESULTS: We show for the first time that HFD-induced obesity elicits resistance to the central actions of BMP8B on energy balance. This obesity-induced BMP8B resistance is explained by i) lack of effects on AMP-activated protein kinase (AMPK) signaling, ii) decreased BMP receptors signaling and iii) reduced expression of Bardet-Biedl Syndrome 1 (BBS1) protein, a key component of the protein complex BBSome in the ventromedial nucleus of the hypothalamus (VMH). The possible mechanistic involvement of BBS1 in this process is demonstrated by lack of a central response to BMP8B in mice carrying a single missense disease-causing mutation in the Bbs1 gene. CONCLUSIONS: Overall, our data uncover a new mechanism of central resistance to hormonal action that may be of relevance in the pathophysiology of obesity.


Subject(s)
Adipose Tissue, Brown , Bone Morphogenetic Proteins , Thermogenesis , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Animals , Bone Morphogenetic Proteins/metabolism , Mice , Obesity/metabolism , Rats , Thermogenesis/physiology
3.
Arthritis Rheumatol ; 74(2): 212-222, 2022 02.
Article in English | MEDLINE | ID: mdl-34398520

ABSTRACT

OBJECTIVE: To investigate whether thermogenesis and the hypothalamus may be involved in the physiopathology of experimental arthritis (EA). METHODS: EA was induced in male Lewis rats by intradermal injection of Freund's complete adjuvant (CFA). Food intake, body weight, plasma cytokines, thermographic analysis, gene and protein expression of thermogenic markers in brown adipose tissue (BAT) and white adipose tissue (WAT), and hypothalamic AMP-activated protein kinase (AMPK) were analyzed. Virogenetic activation of hypothalamic AMPK was performed. RESULTS: We first demonstrated that EA was associated with increased BAT thermogenesis and browning of subcutaneous WAT leading to elevated energy expenditure. Moreover, rats experiencing EA showed inhibition of hypothalamic AMPK, a canonical energy sensor modulating energy homeostasis at the central level. Notably, specific genetic activation of AMPK in the ventromedial nucleus of the hypothalamus (a key site modulating energy metabolism) reversed the effect of EA on energy balance, brown fat, and browning, as well as promoting amelioration of synovial inflammation in experimental arthritis. CONCLUSION: Overall, these data indicate that EA promotes a central catabolic state that can be targeted and reversed by the activation of hypothalamic AMPK. This might provide new therapeutic alternatives to treat rheumatoid arthritis (RA)-associated metabolic comorbidities, improving the overall prognosis in patients with RA.


Subject(s)
AMP-Activated Protein Kinases/physiology , Arthritis/metabolism , Arthritis/physiopathology , Hypothalamus/enzymology , Thermogenesis , Animals , Arthritis/complications , Male , Rats , Rats, Inbred Lew
4.
Int J Mol Sci ; 22(4)2021 Feb 03.
Article in English | MEDLINE | ID: mdl-33546289

ABSTRACT

Several studies have reported that nicotine, the main bioactive component of tobacco, exerts a marked negative energy balance. Apart from its anorectic action, nicotine also modulates energy expenditure, by regulating brown adipose tissue (BAT) thermogenesis and white adipose tissue (WAT) browning. These effects are mainly controlled at the central level by modulation of hypothalamic neuropeptide systems and energy sensors, such as AMP-activated protein kinase (AMPK). In this study, we aimed to investigate the kappa opioid receptor (κOR)/dynorphin signaling in the modulation of nicotine's effects on energy balance. We found that body weight loss after nicotine treatment is associated with a down-regulation of the κOR endogenous ligand dynorphin precursor and with a marked reduction in κOR signaling and the p70 S6 kinase/ribosomal protein S6 (S6K/rpS6) pathway in the lateral hypothalamic area (LHA). The inhibition of these pathways by nicotine was completely blunted in κOR deficient mice, after central pharmacological blockade of κOR, and in rodents where κOR was genetically knocked down specifically in the LHA. Moreover, κOR-mediated nicotine effects on body weight do not depend on orexin. These data unravel a new central regulatory pathway modulating nicotine's effects on energy balance.


Subject(s)
Hypothalamic Area, Lateral/metabolism , Nicotine/pharmacology , Receptors, Opioid, kappa/metabolism , Signal Transduction , AMP-Activated Protein Kinases/metabolism , Animals , Body Weight , Dynorphins/metabolism , Energy Metabolism , Hypothalamic Area, Lateral/drug effects , Male , Mice , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley
5.
Pharmacol Ther ; 219: 107693, 2021 03.
Article in English | MEDLINE | ID: mdl-32987056

ABSTRACT

Obesity has reached pandemic proportions and is associated with severe comorbidities, such as type 2 diabetes mellitus, hepatic and cardiovascular diseases, and certain cancer types. However, the therapeutic options to treat obesity are limited. Extensive epidemiological studies have shown a strong relationship between smoking and body weight, with non-smokers weighing more than smokers at any age. Increased body weight after smoking cessation is a major factor that interferes with their attempts to quit smoking. Numerous controlled studies in both humans and rodents have reported that nicotine, the main bioactive component of tobacco, exerts a marked anorectic action. Furthermore, nicotine is also known to modulate energy expenditure, by regulating the thermogenic activity of brown adipose tissue (BAT) and the browning of white adipose tissue (WAT), as well as glucose homeostasis. Many of these actions occur at central level, by controlling the activity of hypothalamic neuropeptide systems such as proopiomelanocortin (POMC), or energy sensors such as AMP-activated protein kinase (AMPK). However, direct impact of nicotine on metabolic tissues, such as BAT, WAT, liver and pancreas has also been described. Here, we review the actions of nicotine on energy balance. The relevance of this interaction is interesting, because considering the restricted efficiency of obesity treatments, a possible complementary approach may focus on compounds with known pharmacokinetic profile and pharmacological actions, such as nicotine or nicotinic acetylcholine receptors signaling.


Subject(s)
Diabetes Mellitus, Type 2 , Nicotine , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Body Weight , Diabetes Mellitus, Type 2/metabolism , Energy Metabolism , Humans , Nicotine/metabolism , Obesity/drug therapy , Obesity/metabolism , Thermogenesis
6.
EMBO Rep ; 21(9): e49807, 2020 09 03.
Article in English | MEDLINE | ID: mdl-32657019

ABSTRACT

This study investigated the role of CDK4 in the oxidative metabolism of brown adipose tissue (BAT). BAT from Cdk4-/- mice exhibited fewer lipids and increased mitochondrial volume and expression of canonical thermogenic genes, rendering these mice more resistant to cold exposure. Interestingly, these effects were not BAT cell-autonomous but rather driven by increased sympathetic innervation. In particular, the ventromedial hypothalamus (VMH) is known to modulate BAT activation via the sympathetic nervous system. We thus examined the effects of VMH neuron-specific Cdk4 deletion. These mice display increased sympathetic innervation and enhanced cold tolerance, similar to Cdk4-/- mice, in addition to browning of scWAT. Overall, we provide evidence showing that CDK4 modulates thermogenesis by regulating sympathetic innervation of adipose tissue depots through hypothalamic nuclei, including the VMH. This demonstrates that CDK4 not only negatively regulates oxidative pathways, but also modulates the central regulation of metabolism through its action in the brain.


Subject(s)
Adipose Tissue, White , Thermogenesis , Adipocytes, Brown , Adipose Tissue, Brown , Animals , Hypothalamus , Mice , Thermogenesis/genetics
7.
Nutrients ; 12(2)2020 Feb 13.
Article in English | MEDLINE | ID: mdl-32069871

ABSTRACT

Leptin is a hormone released by adipose tissue that plays a key role in the control of energy homeostasis through its binding to leptin receptors (LepR), mainly expressed in the hypothalamus. Most scientific evidence points to leptin's satiating effect being due to its dual capacity to promote the expression of anorexigenic neuropeptides and to reduce orexigenic expression in the hypothalamus. However, it has also been demonstrated that leptin can stimulate (i) thermogenesis in brown adipose tissue (BAT) and (ii) the browning of white adipose tissue (WAT). Since the demonstration of the importance of BAT in humans 10 years ago, its study has aroused great interest, mainly in the improvement of obesity-associated metabolic disorders through the induction of thermogenesis. Consequently, several strategies targeting BAT activation (mainly in rodent models) have demonstrated great potential to improve hyperlipidemias, hepatic steatosis, insulin resistance and weight gain, leading to an overall healthier metabolic profile. Here, we review the potential therapeutic ability of leptin to correct obesity and other metabolic disorders, not only through its satiating effect, but by also utilizing its thermogenic properties.


Subject(s)
Leptin/metabolism , Obesity/metabolism , Satiation/drug effects , Thermogenesis/drug effects , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Animals , Energy Metabolism , Humans , Hypothalamus/metabolism , Receptors, Leptin/metabolism
8.
Nat Commun ; 10(1): 4037, 2019 09 06.
Article in English | MEDLINE | ID: mdl-31492869

ABSTRACT

Increased body weight is a major factor that interferes with smoking cessation. Nicotine, the main bioactive compound in tobacco, has been demonstrated to have an impact on energy balance, since it affects both feeding and energy expenditure at the central level. Among the central actions of nicotine on body weight, much attention has been focused on its effect on brown adipose tissue (BAT) thermogenesis, though its effect on browning of white adipose tissue (WAT) is unclear. Here, we show that nicotine induces the browning of WAT through a central mechanism and that this effect is dependent on the κ opioid receptor (KOR), specifically in the lateral hypothalamic area (LHA). Consistent with these findings, smokers show higher levels of uncoupling protein 1 (UCP1) expression in WAT, which correlates with smoking status. These data demonstrate that central nicotine-induced modulation of WAT browning may be a target against human obesity.


Subject(s)
Adipose Tissue, Brown/drug effects , Hypothalamus/drug effects , Nicotine/pharmacology , Receptors, Opioid, kappa/metabolism , Thermogenesis/drug effects , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Adult , Animals , Body Weight/drug effects , Female , Ganglionic Stimulants/administration & dosage , Ganglionic Stimulants/pharmacology , Humans , Hypothalamus/metabolism , Male , Mice, Knockout , Middle Aged , Nicotine/administration & dosage , Rats, Sprague-Dawley , Receptors, Opioid, kappa/genetics , Uncoupling Protein 1/metabolism
9.
Aging Cell ; 18(3): e12948, 2019 06.
Article in English | MEDLINE | ID: mdl-30920127

ABSTRACT

Age-related increased adiposity is an important contributory factor in the development of insulin resistance (IR) and is associated with metabolic defects. Caloric restriction (CR) is known to induce weight loss and to decrease adiposity while preventing metabolic risk factors. Here, we show that moderate 20% CR delays early deleterious effects of aging on white and brown adipose tissue (WAT and BAT, respectively) function and improves peripheral IR. To elucidate the role of CR in delaying early signs of aging, young (3 months), middle-aged (12 months), and old (20 months) mice fed al libitum and middle-aged and old mice subjected to early-onset CR were used. We show that impaired plasticity of subcutaneous WAT (scWAT) contributes to IR, which is already evident in middle-aged mice. Moreover, alteration of thyroid axis status with age is an important factor contributing to BAT dysfunction in middle-aged animals. Both defects in WAT and BAT/beige cells are ameliorated by CR. Accordingly, CR attenuated the age-related decline in scWAT function and decreased the extent of fibro-inflammation. Furthermore, CR promoted scWAT browning. In brief, our study identifies the contribution of scWAT impairment to age-associated metabolic dysfunction and identifies browning in response to food restriction, as a potential therapeutic strategy to prevent the adverse metabolic effects in middle-aged animals.


Subject(s)
Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Aging/metabolism , Caloric Restriction , Animals , Glucose Tolerance Test , Male , Mice , Mice, 129 Strain , Positron Emission Tomography Computed Tomography
10.
Proc Natl Acad Sci U S A ; 115(45): E10758-E10767, 2018 11 06.
Article in English | MEDLINE | ID: mdl-30348767

ABSTRACT

Conditions of metabolic distress, from malnutrition to obesity, impact, via as yet ill-defined mechanisms, the timing of puberty, whose alterations can hamper later cardiometabolic health and even life expectancy. AMP-activated protein kinase (AMPK), the master cellular energy sensor activated in conditions of energy insufficiency, has a major central role in whole-body energy homeostasis. However, whether brain AMPK metabolically modulates puberty onset remains unknown. We report here that central AMPK interplays with the puberty-activating gene, Kiss1, to control puberty onset. Pubertal subnutrition, which delayed puberty, enhanced hypothalamic pAMPK levels, while activation of brain AMPK in immature female rats substantially deferred puberty. Virogenetic overexpression of a constitutively active form of AMPK, selectively in the hypothalamic arcuate nucleus (ARC), which holds a key population of Kiss1 neurons, partially delayed puberty onset and reduced luteinizing hormone levels. ARC Kiss1 neurons were found to express pAMPK, and activation of AMPK reduced ARC Kiss1 expression. The physiological relevance of this pathway was attested by conditional ablation of the AMPKα1 subunit in Kiss1 cells, which largely prevented the delay in puberty onset caused by chronic subnutrition. Our data demonstrate that hypothalamic AMPK signaling plays a key role in the metabolic control of puberty, acting via a repressive modulation of ARC Kiss1 neurons in conditions of negative energy balance.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Arcuate Nucleus of Hypothalamus/metabolism , Kisspeptins/metabolism , Malnutrition/metabolism , Neurons/metabolism , Sexual Maturation/genetics , AMP-Activated Protein Kinases/genetics , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Animals , Animals, Genetically Modified , Arcuate Nucleus of Hypothalamus/cytology , Arcuate Nucleus of Hypothalamus/drug effects , Caloric Restriction/adverse effects , Estradiol/pharmacology , Female , Gene Expression Regulation, Developmental , Kisspeptins/genetics , Luteinizing Hormone/blood , Malnutrition/genetics , Malnutrition/physiopathology , Mice , Mice, Inbred C57BL , Neurons/cytology , Neurons/drug effects , Rats , Rats, Wistar , Ribonucleotides/pharmacology , Signal Transduction , Time Factors
11.
Diabetes ; 67(11): 2213-2226, 2018 11.
Article in English | MEDLINE | ID: mdl-30104247

ABSTRACT

AMPK is a cellular gauge that is activated under conditions of low energy, increasing energy production and reducing energy waste. Current evidence links hypothalamic AMPK with the central regulation of energy balance. However, it is unclear whether targeting hypothalamic AMPK has beneficial effects in obesity. Here, we show that genetic inhibition of AMPK in the ventromedial nucleus of the hypothalamus (VMH) protects against high-fat diet (HFD)-induced obesity by increasing brown adipose tissue (BAT) thermogenesis and subsequently energy expenditure. Notably, this effect depends upon the AMPKα1 isoform in steroidogenic factor 1 (SF1) neurons of the VMH, since mice bearing selective ablation of AMPKα1 in SF1 neurons display resistance to diet-induced obesity, increased BAT thermogenesis, browning of white adipose tissue, and improved glucose and lipid homeostasis. Overall, our findings point to hypothalamic AMPK in specific neuronal populations as a potential druggable target for the treatment of obesity and associated metabolic disorders.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Diet, High-Fat/adverse effects , Neurons/metabolism , Obesity/metabolism , RNA Splicing Factors/metabolism , Ventromedial Hypothalamic Nucleus/metabolism , AMP-Activated Protein Kinases/genetics , Adipose Tissue, Brown/metabolism , Animals , Body Composition/physiology , Energy Metabolism/physiology , Male , Obesity/etiology , Obesity/genetics , Oxygen Consumption/physiology , Rats , Rats, Sprague-Dawley
12.
Genes (Basel) ; 9(7)2018 Jul 17.
Article in English | MEDLINE | ID: mdl-30018241

ABSTRACT

Recent data have demonstrated that the hypothalamic GRP78/BiP (glucose regulated protein 78 kDa/binding immunoglobulin protein) modulates brown adipose tissue (BAT) thermogenesis by acting downstream on AMP-activated protein kinase (AMPK). Herein, we aimed to investigate whether genetic over-expression of GRP78 in the ventromedial nucleus of the hypothalamus (VMH: a key site regulating thermogenesis) could ameliorate very high fat diet (vHFD)-induced obesity. Our data showed that stereotaxic treatment with adenoviruses harboring GRP78 in the VMH reduced hypothalamic endoplasmic reticulum ER stress and reversed vHFD-induced obesity. Herein, we also demonstrated that this body weight decrease was more likely associated with an increased BAT thermogenesis and browning of white adipose tissue (WAT) than to anorexia. Overall, these results indicate that the modulation of GRP78 in the VMH may be a target against obesity.

13.
Methods Mol Biol ; 1732: 433-448, 2018.
Article in English | MEDLINE | ID: mdl-29480491

ABSTRACT

Hypothalamic AMPK plays a key role in the control of energy homeostasis by regulating energy intake and energy expenditure, particularly modulating brown adipose tissue (BAT) thermogenesis. The function of AMPK can be assayed by analyzing its phosphorylated protein levels in tissues, since AMPK is activated when it is phosphorylated at Thr-172. Here, we describe a method to obtain hypothalamic (nuclei-specific) protein extracts and the suitable conditions to assay AMPK phosphorylation by Western blotting.


Subject(s)
AMP-Activated Protein Kinases/analysis , Enzyme Activation/drug effects , Enzyme Assays/methods , Hypothalamus/metabolism , Isoenzymes/metabolism , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/immunology , AMP-Activated Protein Kinases/metabolism , Adenoviridae/genetics , Animals , Antibodies, Phospho-Specific/immunology , Enzyme Activation/genetics , Enzyme Activators/pharmacology , Enzyme Assays/instrumentation , Enzyme Inhibitors/pharmacology , Genetic Vectors/genetics , Isoenzymes/genetics , Isoenzymes/immunology , Mice , Phosphorylation/drug effects , Phosphorylation/genetics , Phosphorylation/immunology , Rats , Stereotaxic Techniques/instrumentation , Threonine/immunology , Threonine/metabolism
14.
Cell Metab ; 26(1): 212-229.e12, 2017 Jul 05.
Article in English | MEDLINE | ID: mdl-28683288

ABSTRACT

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.


Subject(s)
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
15.
Endocrinology ; 158(7): 2226-2238, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28472467

ABSTRACT

Carnitine palmitoyltransferase (CPT) 1C, a brain-specific protein localized in the endoplasmic reticulum of neurons, is expressed in almost all brain regions. Based on global knockout (KO) models, CPT1C has demonstrated relevance in hippocampus-dependent spatial learning and in hypothalamic regulation of energy balance. Specifically, it has been shown that CPT1C is protective against high-fat diet-induced obesity (DIO), and that CPT1C KO mice show reduced peripheral fatty acid oxidation (FAO) during both fasting and DIO. However, the mechanisms mediating CPT1C-dependent regulation of energy homeostasis remain unclear. Here, we focus on the mechanistic understanding of hypothalamic CPT1C on the regulation of fuel selection in liver and muscle of male mice during energy deprivation situations, such as fasting. In CPT1C-deficient mice, modulation of the main hypothalamic energy sensors (5' adenosine monophosphate-activated protein kinase, Sirtuin 1, and mammalian target of rapamycin) was impaired and plasma catecholamine levels were decreased. Consequently, CPT1C-deficient mice presented defective fasting-induced FAO in liver, leading to higher triacylglycerol accumulation and lower glycogen levels. Moreover, muscle pyruvate dehydrogenase activity was increased, which was indicative of glycolysis enhancement. The respiratory quotient did not decrease in CPT1C KO mice after 48 hours of fasting, confirming a defective switch on fuel substrate selection under hypoglycemia. Phenotype reversion studies identified the mediobasal hypothalamus (MBH) as the main area mediating CPT1C effects on fuel selection. Overall, our data demonstrate that CPT1C in the MBH is necessary for proper hypothalamic sensing of a negative energy balance and fuel partitioning in liver and muscle.


Subject(s)
Carnitine O-Palmitoyltransferase/physiology , Energy Metabolism/genetics , Hypothalamus/physiology , Liver/metabolism , Muscles/metabolism , Animals , Brain/metabolism , Carnitine O-Palmitoyltransferase/genetics , Homeostasis , Hypothalamus/metabolism , Lipid Metabolism/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Specificity/genetics
16.
Diabetes ; 66(1): 87-99, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27634226

ABSTRACT

The chaperone GRP78/BiP (glucose-regulated protein 78 kDa/binding immunoglobulin protein) modulates protein folding in reply to cellular insults that lead to endoplasmic reticulum (ER) stress. This study investigated the role of hypothalamic GRP78 on energy balance, with particular interest in thermogenesis and browning of white adipose tissue (WAT). For this purpose, we used diet-induced obese rats and rats administered thapsigargin, and by combining metabolic, histologic, physiologic, pharmacologic, thermographic, and molecular techniques, we studied the effect of genetic manipulation of hypothalamic GRP78. Our data showed that rats fed a high-fat diet or that were centrally administered thapsigargin displayed hypothalamic ER stress, whereas genetic overexpression of GRP78 specifically in the ventromedial nucleus of the hypothalamus was sufficient to alleviate ER stress and to revert the obese and metabolic phenotype. Those effects were independent of feeding and leptin but were related to increased thermogenic activation of brown adipose tissue and induction of browning in WAT and could be reversed by antagonism of ß3 adrenergic receptors. This evidence indicates that modulation of hypothalamic GRP78 activity may be a potential strategy against obesity and associated comorbidities.


Subject(s)
Adipose Tissue, White/drug effects , Endoplasmic Reticulum Stress/drug effects , Hypothalamus/drug effects , Hypothalamus/metabolism , Obesity/drug therapy , Obesity/metabolism , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Animals , Blotting, Western , Diet, High-Fat , Fatty Acids, Nonesterified/blood , Immunohistochemistry , Male , Obesity/blood , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Taurochenodeoxycholic Acid/therapeutic use , Temperature , Thermogenesis/drug effects
17.
Cell Rep ; 16(8): 2231-2242, 2016 08 23.
Article in English | MEDLINE | ID: mdl-27524625

ABSTRACT

AMP-activated protein kinase (AMPK) in the ventromedial nucleus of the hypothalamus (VMH) and orexin (OX) in the lateral hypothalamic area (LHA) modulate brown adipose tissue (BAT) thermogenesis. However, whether these two molecular mechanisms act jointly or independently is unclear. Here, we show that the thermogenic effect of bone morphogenetic protein 8B (BMP8B) is mediated by the inhibition of AMPK in the VMH and the subsequent increase in OX signaling via the OX receptor 1 (OX1R). Accordingly, the thermogenic effect of BMP8B is totally absent in ox-null mice. BMP8B also induces browning of white adipose tissue (WAT), its thermogenic effect is sexually dimorphic (only observed in females), and its impact on OX expression and thermogenesis is abolished by the knockdown of glutamate vesicular transporter 2 (VGLUT2), implicating glutamatergic signaling. Overall, our data uncover a central network controlling energy homeostasis that may be of considerable relevance for obesity and metabolic disorders.


Subject(s)
AMP-Activated Protein Kinases/genetics , Adipose Tissue, Brown/metabolism , Bone Morphogenetic Proteins/genetics , Hypothalamic Area, Lateral/metabolism , Orexins/genetics , Thermogenesis/genetics , Ventromedial Hypothalamic Nucleus/metabolism , AMP-Activated Protein Kinases/metabolism , Adipose Tissue, Brown/drug effects , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Animals , Body Weight , Bone Morphogenetic Proteins/metabolism , Bone Morphogenetic Proteins/pharmacology , Energy Metabolism/genetics , Female , Gene Expression Regulation , Glutamic Acid/metabolism , Male , Mice, Knockout , Orexin Receptors/genetics , Orexin Receptors/metabolism , Orexins/metabolism , Rats , Rats, Sprague-Dawley , Sex Factors , Signal Transduction , Vesicular Glutamate Transport Protein 2/antagonists & inhibitors , Vesicular Glutamate Transport Protein 2/genetics , Vesicular Glutamate Transport Protein 2/metabolism
18.
Endocrine ; 50(2): 276-91, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26089260

ABSTRACT

Regulation of energy homeostasis is tightly controlled by the central nervous system (CNS). Several key areas such as the hypothalamus and brainstem receive and integrate signals conveying energy status from the periphery, such as leptin, thyroid hormones, and insulin, ultimately leading to modulation of food intake, energy expenditure (EE), and peripheral metabolism. The autonomic nervous system (ANS) plays a key role in the response to such signals, innervating peripheral metabolic tissues, including brown and white adipose tissue (BAT and WAT), liver, pancreas, and skeletal muscle. The ANS consists of two parts, the sympathetic and parasympathetic nervous systems (SNS and PSNS). The SNS regulates BAT thermogenesis and EE, controlled by central areas such as the preoptic area (POA) and the ventromedial, dorsomedial, and arcuate hypothalamic nuclei (VMH, DMH, and ARC). The SNS also regulates lipid metabolism in WAT, controlled by the lateral hypothalamic area (LHA), VMH, and ARC. Control of hepatic glucose production and pancreatic insulin secretion also involves the LHA, VMH, and ARC as well as the dorsal vagal complex (DVC), via splanchnic sympathetic and the vagal parasympathetic nerves. Muscle glucose uptake is also controlled by the SNS via hypothalamic nuclei such as the VMH. There is recent evidence of novel pathways connecting the CNS and ANS. These include the hypothalamic AMP-activated protein kinase-SNS-BAT axis which has been demonstrated to be a key modulator of thermogenesis. In this review, we summarize current knowledge of the role of the ANS in the modulation of energy balance.


Subject(s)
Autonomic Nervous System/metabolism , Homeostasis/physiology , Hypothalamus/metabolism , Humans
19.
Int J Neuropsychopharmacol ; 18(9)2015 Feb 05.
Article in English | MEDLINE | ID: mdl-25655433

ABSTRACT

BACKGROUND: Cumulative data indicate that the endocannabinoid system plays a major role in feeding behavior and energy balance. Genetic silencing of cannabinoid receptor type 1 (CB1) reduces body weight gain, independently of food intake. METHODS: In this work, we investigated whether the hypothalamic neuropeptide expression pattern supports the absence of the anorexigenic response observed under constitutive CB1 ablation, by using neuronal CB1 conditional null mice (CamK-CB1-KO) and whole body CB1 null mice (CB1-KO). RESULTS: Our data showed that both CB1 null models display a marked decrease in proopiomelanocortin (POMC) and cocaine-amphetamine-regulated transcript (CART) expression in the arcuate nucleus of the hypothalamus (ARC). CONCLUSIONS: This evidence suggests that a lack of hypophagia is associated with the suppression of ARC anorexigenic neuropeptides and that behavioral changes in food intake (or lack thereof) after constitutive CB1 ablation are likely mediated by impaired melanocortin and CART signaling in the hypothalamus.


Subject(s)
Anorexia/metabolism , Arcuate Nucleus of Hypothalamus/metabolism , Feeding Behavior/physiology , Nerve Tissue Proteins/metabolism , Pro-Opiomelanocortin/metabolism , Receptor, Cannabinoid, CB1/physiology , Animals , Behavior, Animal , Body Weight , Cannabinoid Receptor Antagonists/pharmacology , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB1/deficiency , Receptor, Cannabinoid, CB1/genetics
20.
Cell Rep ; 9(1): 366-377, 2014 Oct 09.
Article in English | MEDLINE | ID: mdl-25284795

ABSTRACT

Hypothalamic endoplasmic reticulum (ER) stress is a key mechanism leading to obesity. Here, we demonstrate that ceramides induce lipotoxicity and hypothalamic ER stress, leading to sympathetic inhibition, reduced brown adipose tissue (BAT) thermogenesis, and weight gain. Genetic overexpression of the chaperone GRP78/BiP (glucose-regulated protein 78 kDa/binding immunoglobulin protein) in the ventromedial nucleus of the hypothalamus (VMH) abolishes ceramide action by reducing hypothalamic ER stress and increasing BAT thermogenesis, which leads to weight loss and improved glucose homeostasis. The pathophysiological relevance of this mechanism is demonstrated in obese Zucker rats, which show increased hypothalamic ceramide levels and ER stress. Overexpression of GRP78 in the VMH of these animals reduced body weight by increasing BAT thermogenesis as well as decreasing leptin and insulin resistance and hepatic steatosis. Overall, these data identify a triangulated signaling network involving central ceramides, hypothalamic lipotoxicity/ER stress, and BAT thermogenesis as a pathophysiological mechanism of obesity.


Subject(s)
Adipose Tissue, Brown/metabolism , Ceramides/metabolism , Hypothalamus/metabolism , Obesity/etiology , Thermogenesis/physiology , Animals , Endoplasmic Reticulum Stress , Insulin Resistance/physiology , Male , Obesity/metabolism , Rats , Rats, Sprague-Dawley , Rats, Zucker , Weight Loss
SELECTION OF CITATIONS
SEARCH DETAIL
...