Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
J Clin Invest ; 133(7)2023 04 03.
Article in English | MEDLINE | ID: mdl-36787185

ABSTRACT

Glucose homeostasis can be improved after bariatric surgery, which alters bile flow and stimulates gut hormone secretion, particularly FGF15/19. FGFR1 expression in AGRP-expressing cells is required for bile acids' ability to improve glucose control. We show that the mouse Agrp gene has 3 promoter/enhancer regions that direct transcription of each of their own AGRP transcripts. One of these Agrp promoters/enhancers, Agrp-B, is regulated by bile acids. We generated an Agrp-B knockin FLP/knockout allele. AGRP-B-expressing cells are found in endocrine cells of the pars tuberalis and coexpress diacylglycerol lipase B - an endocannabinoid biosynthetic enzyme - distinct from pars tuberalis thyrotropes. AGRP-B expression is also found in the folliculostellate cells of the pituitary's anterior lobe. Mice without AGRP-B were protected from glucose intolerance induced by high-fat feeding but not from excess weight gain. Chemogenetic inhibition of AGRP-B cells improved glucose tolerance by enhancing glucose-stimulated insulin secretion. Inhibition of the AGRP-B cells also caused weight loss. The improved glucose tolerance and reduced body weight persisted up to 6 weeks after cessation of the DREADD-mediated inhibition, suggesting the presence of a biological switch for glucose homeostasis that is regulated by long-term stability of food availability.


Subject(s)
Hypothalamus , Neurons , Mice , Animals , Agouti-Related Protein/metabolism , Hypothalamus/metabolism , Neurons/metabolism , Homeostasis , Glucose/metabolism , Bile Acids and Salts/metabolism , Eating
2.
Trends Endocrinol Metab ; 31(1): 46-52, 2020 01.
Article in English | MEDLINE | ID: mdl-31629614

ABSTRACT

Obesity is a neurological disorder that operates by favoring energy storage within adipose depots and increased caloric intake. Most cases of human obesity are acquired without any underlying genetic basis. Here, we suggest that obesity can impair the function of some hypothalamic neurons critical to body weight regulation. Genetic ablation of the retinoblastoma (Rb) gene within pro-opiomelanocortin (POMC) neurons leads to death of the neurons and subsequent obesity. The Rb protein (pRb), a key inhibitor of the cell cycle, can also be inactivated by cyclin dependent kinase (CDK)-mediated phosphorylation. Extensive development led to the production of FDA-approved CDK4/6 inhibitors. Based on our own results, we propose that maintaining or re-instating pRb function using CDK4/6 inhibitors are potentially effective treatments of diet-induced obesity (DIO).


Subject(s)
Obesity/metabolism , Animals , Cell Cycle/genetics , Cell Cycle/physiology , Humans , Hypothalamus/metabolism , Pro-Opiomelanocortin/metabolism , Retinoblastoma/metabolism
3.
JCI Insight ; 3(17)2018 09 06.
Article in English | MEDLINE | ID: mdl-30185666

ABSTRACT

When obesity is caused by consumption of a high-fat diet, the tumor suppressor pRb is phosphoinactivated in the neurons of the mediobasal hypothalamus, a brain area critical for energy-balance regulation. However, the functional relevance of pRb phosphoinactivation in the mediobasal hypothalamus to diet-induced obesity remains unknown. Here, we show that inhibiting pRb phosphorylation in the mediobasal hypothalamus can prevent and treat diet-induced obesity in mice. Expressing an unphosphorylable pRb nonselectively in the mediobasal hypothalamus or conditionally in anorexigenic POMC neurons inhibits diet-induced obesity. Intracerebroventricular delivery of US Food and Drug Administration-approved (FDA-approved) cyclin-dependent kinase 4 (CDK4) inhibitor abemaciclib inhibits pRb phosphorylation in the mediobasal hypothalamus and prevents diet-induced obesity. Oral administration of abemaciclib at doses approved for human use reduces fat mass in diet-induced obese mice by increasing lipid oxidation without significantly reducing lean mass. With analysis of recent literature identifying CDK4 as the most abundantly expressed neuronal CDK in the mediobasal hypothalamus, our work uncovers CDK4 as the major kinase for hypothalamic pRb phosphoinactivation and a highly effective central antiobesity target. As three CDK4/6 inhibitors have recently received FDA approval for life-long breast cancer therapy, our study provides a preclinical basis for their expedient repurposing for obesity management.


Subject(s)
Aminopyridines/pharmacology , Benzimidazoles/pharmacology , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 4/metabolism , Diet, High-Fat/adverse effects , Obesity/metabolism , Obesity/prevention & control , Animals , Disease Models, Animal , Drug Approval , Energy Metabolism , Hypothalamus/metabolism , Lipid Metabolism , Male , Mice , Mice, Inbred C57BL , Phosphorylation/drug effects , Retinoblastoma Protein/metabolism , United States , United States Food and Drug Administration
4.
Mol Metab ; 8: 37-50, 2018 02.
Article in English | MEDLINE | ID: mdl-29290621

ABSTRACT

OBJECTIVE: Bile acids have been implicated as important regulators of glucose metabolism via activation of FXR and GPBAR1. We have previously shown that FGF19 can modulate glucose handling by suppressing the activity of hypothalamic AGRP/NPY neurons. As bile acids stimulate the release of FGF19/FGF15 into the circulation, we pursued the potential of bile acids to improve glucose tolerance via a gut-brain axis involving FXR and FGF15/FGF19 within enterocytes and FGF receptors on hypothalamic AGRP/NPY neurons. METHODS: A 5-day gavage of taurocholic acid, mirroring our previous protocol of a 5-day FGF19 treatment, was performed. Oral glucose tolerance tests in mice with genetic manipulations of FGF signaling and melanocortin signaling were used to define a gut-brain axis responsive to bile acids. RESULTS: The taurocholic acid gavage led to increased serum concentrations of taurocholic acid as well as increases of FGF15 mRNA in the ileum and improved oral glucose tolerance in obese (ob/ob) mice. In contrast, lithocholic acid, an FXR antagonist but a potent agonist for GPBAR1, did not improve glucose tolerance. The positive response to taurocholic acid is dependent upon an intact melanocortinergic system as obese MC4R-null mice or ob/ob mice without AGRP did not show improvements in glucose tolerance after taurocholate gavage. We also tested the FGF receptor isoform necessary for the bile acid response, using AGRP:Fgfr1-/- and AGRP:Fgfr2-/- mice. While the absence of FGFR1 in AGRP/NPY neurons did not alter glucose tolerance after taurocholate gavage, manipulations of Fgfr2 caused bidirectional changes depending upon the experimental model. We hypothesized the existence of an endogenous hypothalamic FGF, most likely FGF17, that acted as a chronic activator of AGRP/NPY neurons. We developed two short peptides based on FGF8 and FGF17 that should antagonize FGF17 action. Both of these peptides improved glucose homeostasis after a 4-day course of central and peripheral injections. Significantly, daily average blood glucose from continuous glucose monitoring was reduced in all tested animals but glucose concentrations remained in the euglycemia range. CONCLUSIONS: We have defined a gut-brain axis that regulates glucose metabolism mediated by antagonistic fibroblast growth factors. From the intestine, bile acids stimulate FGF15 secretion, leading to activation of the FGF receptors in hypothalamic AGRP/NPY neurons. FGF receptor intracellular signaling subsequently silences AGRP/NPY neurons, leading to improvements of glucose tolerance that are likely mediated by the autonomic nervous system. Finally, short peptides that antagonize homodimeric FGF receptor signaling within the hypothalamus have beneficial effects on glucose homeostasis without inducing hypoglycemia. These peptides could provide a new mode of regulating glucose metabolism.


Subject(s)
Bile Acids and Salts/metabolism , Blood Glucose/metabolism , Fibroblast Growth Factors/metabolism , Glucose Intolerance/metabolism , Hypothalamus/metabolism , Animals , Hypothalamus/physiology , Mice , Mice, Obese , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Receptors, G-Protein-Coupled/metabolism
5.
J Neurosci ; 33(29): 11972-85, 2013 Jul 17.
Article in English | MEDLINE | ID: mdl-23864684

ABSTRACT

Like obesity, prolonged food deprivation induces severe hepatic steatosis; however, the functional significance of this phenomenon is not well understood. In this study, we show that the fall in plasma leptin concentration during fasting is required for the development of hepatic steatosis in mice. Removal of leptin receptors from AGRP neurons diminishes fasting-induced hepatic steatosis. Furthermore, the suppressive effects of leptin on fasting-induced hepatic steatosis are absent in mice lacking the gene encoding agouti-related protein (Agrp), suggesting that this function of leptin is mediated by AGRP. Prolonged fasting leads to suppression of hepatic sympathetic activity, increased expression of acyl CoA:diacylglycerol acyltransferase-2 in the liver, and elevation of hepatic triglyceride content and all of these effects are blunted in the absence of AGRP. AGRP deficiency, despite having no effects on feeding or body adiposity in the free-fed state, impairs triglyceride and ketone body release from the liver during prolonged fasting. Furthermore, reducing CNS Agrp expression in wild-type mice by RNAi protected against the development of hepatic steatosis not only during starvation, but also in response to consumption of a high-fat diet. These findings identify the leptin-AGRP circuit as a critical modulator of hepatic triglyceride stores in starvation and suggest a vital role for this circuit in sustaining the supply of energy from the liver to extrahepatic tissues during periods of prolonged food deprivation.


Subject(s)
Agouti-Related Protein/genetics , Energy Metabolism/physiology , Hypothalamus/metabolism , Leptin/metabolism , Liver/metabolism , Receptors, Leptin/genetics , Acyl Coenzyme A/genetics , Acyl Coenzyme A/metabolism , Agouti-Related Protein/metabolism , Animals , Body Composition/drug effects , Body Composition/physiology , Eating/drug effects , Eating/physiology , Energy Metabolism/drug effects , Fatty Liver/genetics , Fatty Liver/metabolism , Food Deprivation/physiology , Hypothalamus/drug effects , Leptin/pharmacology , Liver/drug effects , Male , Mice , Mice, Inbred C57BL , Neurons/drug effects , Neurons/metabolism , Norepinephrine/metabolism , Obesity/genetics , Obesity/metabolism , Receptors, Leptin/metabolism
6.
EMBO J ; 32(6): 844-57, 2013 Mar 20.
Article in English | MEDLINE | ID: mdl-23403926

ABSTRACT

pRb is frequently inactivated in tumours by mutations or phosphorylation. Here, we investigated whether pRb plays a role in obesity. The Arcuate nucleus (ARC) in hypothalamus contains antagonizing POMC and AGRP/NPY neurons for negative and positive energy balance, respectively. Various aspects of ARC neurons are affected in high-fat diet (HFD)-induced obesity mouse model. Using this model, we show that HFD, as well as pharmacological activation of AMPK, induces pRb phosphorylation and E2F target gene de-repression in ARC neurons. Some affected neurons express POMC; and deleting Rb1 in POMC neurons induces E2F target gene de-repression, cell-cycle re-entry, apoptosis, and a hyperphagia-obesity-diabetes syndrome. These defects can be corrected by combined deletion of E2f1. In contrast, deleting Rb1 in the antagonizing AGRP/NPY neurons shows no effects. Thus, pRb-E2F1 is an obesity suppression mechanism in ARC POMC neurons and HFD-AMPK inhibits this mechanism by phosphorylating pRb in this location.


Subject(s)
Diet, High-Fat , Dietary Fats/pharmacology , Hypothalamus , Obesity/genetics , Retinoblastoma Protein/antagonists & inhibitors , Retinoblastoma Protein/physiology , Adenylate Kinase/metabolism , Adenylate Kinase/physiology , Animals , Arcuate Nucleus of Hypothalamus/cytology , Arcuate Nucleus of Hypothalamus/drug effects , Arcuate Nucleus of Hypothalamus/metabolism , Arcuate Nucleus of Hypothalamus/physiology , Diet, High-Fat/adverse effects , Down-Regulation/genetics , E2F1 Transcription Factor/metabolism , E2F1 Transcription Factor/physiology , Female , Hypothalamus/drug effects , Hypothalamus/metabolism , Hypothalamus/pathology , Ideal Body Weight/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/drug effects , Neurons/metabolism , Neurons/physiology , Obesity/metabolism , Obesity/pathology , Phosphorylation/drug effects , Pro-Opiomelanocortin/metabolism , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism
7.
J Neurosci ; 32(29): 9870-7, 2012 Jul 18.
Article in English | MEDLINE | ID: mdl-22815502

ABSTRACT

Thioredoxin interacting protein (TXNIP) has recently been described as a key regulator of energy metabolism through pleiotropic actions that include nutrient sensing in the mediobasal hypothalamus (MBH). However, the role of TXNIP in neurochemically specific hypothalamic subpopulations and the circuits downstream from MBH TXNIP engaged to regulate energy homeostasis remain unexplored. To evaluate the metabolic role of TXNIP activity specifically within arcuate Agrp neurons, we generated Agrp-specific TXNIP gain-of-function and loss-of-function mouse models using Agrp-Ires-cre mice, TXNIP (flox/flox) mice, and a lentivector expressing the human TXNIP isoform conditionally in the presence of Cre recombinase. Overexpression of TXNIP in Agrp neurons predisposed to diet-induced obesity and adipose tissue storage by decreasing energy expenditure and spontaneous locomotion, without affecting food intake. Conversely, Agrp neuronal TXNIP deletion protected against diet-induced obesity and adipose tissue storage by increasing energy expenditure and spontaneous locomotion, also without affecting food intake. TXNIP overexpression in Agrp neurons did not primarily affect glycemic control, whereas deletion of TXNIP in Agrp neurons improved fasting glucose levels and glucose tolerance independently of its effects on body weight and adiposity. Bidirectional manipulation of TXNIP expression induced reciprocal changes in central leptin sensitivity and the neural regulation of lipolysis. Together, these results identify a critical role for TXNIP in Agrp neurons in mediating diet-induced obesity through the regulation of energy expenditure and adipose tissue metabolism, independently of food intake. They also reveal a previously unidentified role for Agrp neurons in the brain-adipose axis.


Subject(s)
Adiposity/physiology , Agouti-Related Protein/metabolism , Carrier Proteins/metabolism , Energy Metabolism/physiology , Hypothalamus/metabolism , Leptin/pharmacology , Neurons/metabolism , Thioredoxins/metabolism , Agouti-Related Protein/genetics , Animals , Blood Glucose/metabolism , Carrier Proteins/genetics , Diet , Eating/physiology , Homeostasis/physiology , Hypothalamus/drug effects , Mice , Mice, Transgenic , Neurons/drug effects , Obesity/genetics , Obesity/metabolism , Thioredoxins/genetics
8.
Peptides ; 37(1): 6-12, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22800691

ABSTRACT

This study used two mouse models with genetic manipulation of the melanocortin system to investigate prolactin regulation. Mice with overexpression of the melanocortin receptor (MC-R) agonist, α-melanocyte-stimulating hormone (Tg-MSH) or deletion of the MC-R antagonist agouti-related protein (AgRP KO) were studied. Male Tg-MSH mice had lower blood prolactin levels at baseline (2.9±0.3 vs. 4.7±0.7ng/ml) and after restraint stress (68±6.5 vs. 117±22ng/ml) vs. WT (p<0.05); however, pituitary prolactin content was not different. Blood prolactin was also decreased in male AgRP KO mice at baseline (4.2±0.5 vs. 7.6±1.3ng/ml) and after stress (60±4.5 vs. 86.1±5.7ng/ml) vs. WT (p<0.001). Pituitary prolactin content was lower in male AgRP KO mice (4.3±0.3 vs. 6.7±0.5µg/pituitary, p<0.001) vs. WT. No differences in blood or pituitary prolactin levels were observed in female AgRP KO mice vs. WT. Hypothalamic dopamine activity was assessed as the potential mechanism responsible for changes in prolactin levels. Hypothalamic tyrosine hydroxylase mRNA was measured in both genetic models vs. WT mice and hypothalamic dopamine and 3,4-dihydroxyphenylacetic acid (DOPAC) content were measured in male AgRP KO and WT mice but neither were significantly different. However, these results do not preclude changes in dopamine activity as dopamine turnover was not directly investigated. This is the first study to show that baseline and stress-induced prolactin release and pituitary prolactin content are reduced in mice with genetic alterations of the melanocortin system and suggests that changes in hypothalamic melanocortin activity may be reflected in measurements of serum prolactin levels.


Subject(s)
Hypothalamus/metabolism , Melanocortins/physiology , Prolactin/blood , 3,4-Dihydroxyphenylacetic Acid/metabolism , Agouti-Related Protein/genetics , Agouti-Related Protein/metabolism , Animals , Corticosterone/blood , Dopamine/metabolism , Dopamine Antagonists/pharmacology , Female , Gene Expression , Gene Knockout Techniques , Hypothalamus/enzymology , Male , Melanocortins/metabolism , Metoclopramide/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Pituitary Gland/metabolism , Tyrosine 3-Monooxygenase/genetics , Tyrosine 3-Monooxygenase/metabolism , alpha-MSH/genetics , alpha-MSH/metabolism
9.
Endocrinology ; 153(5): 2408-19, 2012 May.
Article in English | MEDLINE | ID: mdl-22408174

ABSTRACT

Leptin and melanocortin signaling control ingestive behavior, energy balance, and substrate utilization, but only leptin signaling defects cause hypothalamic hypogonadism and infertility. Although GnRH neurons do not express leptin receptors, leptin influences GnRH neuron activity via regulation of immediate downstream mediators including the neuropeptides neuropeptide Y and the melanocortin agonist and antagonist, α-MSH, agouti-related peptide, respectively. Here we show that modulation of melanocortin signaling in female db/db mice through ablation of agouti-related peptide, or heterozygosity of melanocortin 4 receptor, restores the timing of pubertal onset, fertility, and lactation. Additionally, melanocortin 4 receptor activation increases action potential firing and induces c-Fos expression in GnRH neurons, providing further evidence that melanocortin signaling influences GnRH neuron activity. These studies thus establish melanocortin signaling as an important component in the leptin-mediated regulation of GnRH neuron activity, initiation of puberty and fertility.


Subject(s)
Agouti-Related Protein/metabolism , Hypothalamus/metabolism , Leptin/metabolism , Neurons/metabolism , Receptors, Leptin/metabolism , Sexual Maturation/physiology , Signal Transduction/physiology , Agouti-Related Protein/genetics , Animals , Blood Glucose/metabolism , Cell Count , Estradiol/blood , Female , Insulin/blood , Leptin/genetics , Mice , Mice, Knockout , Ovary/metabolism , Progesterone/blood , Receptors, Leptin/genetics
10.
Transl Psychiatry ; 2: e83, 2012.
Article in English | MEDLINE | ID: mdl-22408745

ABSTRACT

The glutamatergic system has been implicated in the pathophysiology of depression and the mechanism of action of antidepressants. Leptin, an adipocyte-derived hormone, has antidepressant-like properties. However, the functional role of leptin receptor (Lepr) signaling in glutamatergic neurons remains to be elucidated. In this study, we generated conditional knockout mice in which the long form of Lepr was ablated selectively in glutamatergic neurons located in the forebrain structures, including the hippocampus and prefrontal cortex (Lepr cKO). Lepr cKO mice exhibit normal growth and body weight. Behavioral characterization of Lepr cKO mice reveals depression-like behavioral deficits, including anhedonia, behavioral despair, enhanced learned helplessness and social withdrawal, with no evident signs of anxiety. In addition, loss of Lepr in forebrain glutamatergic neurons facilitates NMDA-induced hippocampal long-term synaptic depression (LTD), whereas conventional LTD or long-term potentiation (LTP) was not affected. The facilitated LTD induction requires activation of the GluN2B subunit as it was completely blocked by a selective GluN2B antagonist. Moreover, Lepr cKO mice are highly sensitive to the antidepressant-like behavioral effects of the GluN2B antagonist but resistant to leptin. These results support important roles for Lepr signaling in glutamatergic neurons in regulating depression-related behaviors and modulating excitatory synaptic strength, suggesting a possible association between synaptic depression and behavioral manifestations of depression.


Subject(s)
Depression/physiopathology , Glutamine/physiology , Leptin/physiology , Long-Term Synaptic Depression/physiology , Prosencephalon/physiopathology , Receptors, Leptin/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Animals , Arousal/physiology , Cerebral Cortex/physiopathology , Corticosterone/blood , Dominance-Subordination , Exploratory Behavior/physiology , Helplessness, Learned , Hippocampus/physiopathology , Homeodomain Proteins/genetics , Homeodomain Proteins/physiology , Hypothalamus/physiopathology , Insulin/blood , Leptin/genetics , Mice , Mice, Knockout , Motivation/physiology , Motor Activity/physiology , Neurons/physiology , Orientation/physiology , Real-Time Polymerase Chain Reaction , Receptors, Leptin/genetics , Receptors, N-Methyl-D-Aspartate/genetics , Signal Transduction/genetics , Signal Transduction/physiology , Social Environment , Transcription Factors/genetics , Transcription Factors/physiology
11.
J Clin Invest ; 121(1): 355-68, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21183787

ABSTRACT

Studies in humans and rodents indicate that a minimum amount of stored energy is required for normal pubertal development. The adipocyte-derived hormone leptin is a key metabolic signal to the neuroendocrine reproductive axis. Humans and mice lacking leptin or the leptin receptor (LepR) (ob/ob and db/db mice, respectively) are infertile and fail to enter puberty. Leptin administration to leptin-deficient subjects and ob/ob mice induces puberty and restores fertility, but the exact site or sites of leptin action are unclear. Here, we found that genetic deletion of LepR selectively from hypothalamic Kiss1 neurons in mice had no effect on puberty or fertility, indicating that direct leptin signaling in Kiss1 neurons is not required for these processes. However, bilateral lesions of the ventral premammillary nucleus (PMV) of ob/ob mice blunted the ability of exogenous leptin to induce sexual maturation. Moreover, unilateral reexpression of endogenous LepR in PMV neurons was sufficient to induce puberty and improve fertility in female LepR-null mice. This LepR reexpression also normalized the increased hypothalamic GnRH content characteristic of leptin-signaling deficiency. These data suggest that the PMV is a key site for leptin's permissive action at the onset of puberty and support the hypothesis that the multiple actions of leptin to control metabolism and reproduction are anatomically dissociated.


Subject(s)
Hypothalamus/metabolism , Leptin/metabolism , Proteins/metabolism , Sexual Maturation/physiology , Animals , Base Sequence , Female , Fertility/genetics , Fertility/physiology , Gene Expression , Humans , Kisspeptins , Leptin/deficiency , Leptin/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Mice, Transgenic , Neurons/metabolism , Pregnancy , Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Leptin/deficiency , Receptors, Leptin/genetics , Receptors, Leptin/metabolism , Sexual Maturation/genetics , Signal Transduction
12.
Cell Metab ; 11(4): 286-97, 2010 Apr 07.
Article in English | MEDLINE | ID: mdl-20374961

ABSTRACT

Circulating leptin and insulin convey information regarding energy stores to the central nervous system, particularly the hypothalamus. Hypothalamic pro-opiomelanocortin (POMC) neurons regulate energy balance and glucose homeostasis and express leptin and insulin receptors. However, the physiological significance of concomitant leptin and insulin action on POMC neurons remains to be established. Here, we show that mice lacking both leptin and insulin receptors in POMC neurons (Pomc-Cre, Lepr(flox/flox) IR(flox/flox) mice) display systemic insulin resistance, which is distinct from the single deletion of either receptor. In addition, Pomc-Cre, Lepr(flox/flox) IR(flox/flox) female mice display elevated serum testosterone levels and ovarian abnormalities, resulting in reduced fertility. We conclude that direct action of insulin and leptin on POMC neurons is required to maintain normal glucose homeostasis and reproductive function.


Subject(s)
Blood Glucose/metabolism , Fertility/physiology , Homeostasis/physiology , Hypothalamus/metabolism , Insulin Resistance/physiology , Insulin/metabolism , Leptin/metabolism , Neurons/metabolism , Analysis of Variance , Animals , Female , Hypothalamus/cytology , Insulin Secretion , Islets of Langerhans/metabolism , Male , Mice , Pro-Opiomelanocortin/metabolism , Testosterone/blood
13.
J Neurophysiol ; 101(5): 2305-16, 2009 May.
Article in English | MEDLINE | ID: mdl-19261705

ABSTRACT

The mammalian CNS relies on a constant supply of external glucose for its undisturbed operation. However, neurons can readily switch to using fatty acids and ketones as alternative fuels. Here, we show that oleic acid (OA) excites pro-opiomelanocortin (POMC) neurons by inhibition of ATP-activated potassium (K(ATP)) channels. The involvement of K(ATP) channels is further supported by experiments in SUR1 KO animals. Inhibition of beta-oxidation using carnitine palmitoyltransferase-1 inhibitors blocks OA-induced depolarization. The depolarizing effect of OA is specific because it is not mimicked by octanoic acid. Furthermore, OA does not regulate the excitability of agouti-related peptide neurons. High-fat feeding alters POMC neuron excitability, but not its response to OA. Thus beta-oxidation in POMC neurons may mediate the appetite-suppressing (anorexigenic) effects of OA.


Subject(s)
Hypothalamus/cytology , Neurons/drug effects , Oleic Acid/pharmacology , Pro-Opiomelanocortin/metabolism , ATP-Binding Cassette Transporters , Action Potentials/drug effects , Action Potentials/genetics , Adenosine Triphosphate/pharmacology , Analysis of Variance , Animals , Biophysics , Diazoxide/pharmacology , Dietary Fats/administration & dosage , Dose-Response Relationship, Drug , Electric Stimulation/methods , Glucose/metabolism , Glyburide/pharmacology , Green Fluorescent Proteins/genetics , Hypoglycemic Agents/pharmacology , In Vitro Techniques , Mice , Mice, Transgenic , Models, Biological , Neural Inhibition/drug effects , Neural Inhibition/physiology , Neurons/metabolism , Patch-Clamp Techniques/methods , Potassium Channel Blockers , Potassium Channels, Inwardly Rectifying/deficiency , Potassium Channels, Inwardly Rectifying/drug effects , Potassium Channels, Inwardly Rectifying/physiology , Pro-Opiomelanocortin/genetics , Receptors, Drug/deficiency , Sulfonylurea Receptors
14.
J Biol Chem ; 283(19): 13087-99, 2008 May 09.
Article in English | MEDLINE | ID: mdl-18337240

ABSTRACT

Hepatic steatosis is often associated with insulin resistance and obesity and can lead to steatohepatitis and cirrhosis. In this study, we have demonstrated that hormone-sensitive lipase (HSL) and adipose triglyceride lipase (ATGL), two enzymes critical for lipolysis in adipose tissues, also contribute to lipolysis in the liver and can mobilize hepatic triglycerides in vivo and in vitro. Adenoviral overexpression of HSL and/or ATGL reduced liver triglycerides by 40-60% in both ob/ob mice and mice with high fat diet-induced obesity. However, these enzymes did not affect fasting plasma triglyceride and free fatty acid levels or triglyceride and apolipoprotein B secretion rates. Plasma 3-beta-hydroxybutyrate levels were increased 3-5 days after infection in both HSL- and ATGL-overexpressing male mice, suggesting an increase in beta-oxidation. Expression of genes involved in fatty acid transport and synthesis, lipid storage, and mitochondrial bioenergetics was unchanged. Mechanistic studies in oleate-supplemented McA-RH7777 cells with adenoviral overexpression of HSL or ATGL showed that reduced cellular triglycerides could be attributed to increases in beta-oxidation as well as direct release of free fatty acids into the medium. In summary, hepatic overexpression of HSL or ATGL can promote fatty acid oxidation, stimulate direct release of free fatty acid, and ameliorate hepatic steatosis. This study suggests a direct functional role for both HSL and ATGL in hepatic lipid homeostasis and identifies these enzymes as potential therapeutic targets for ameliorating hepatic steatosis associated with insulin resistance and obesity.


Subject(s)
Adipose Tissue/enzymology , Carboxylic Ester Hydrolases/metabolism , Fatty Acids, Nonesterified/metabolism , Fatty Liver/enzymology , Fatty Liver/pathology , Gene Expression Regulation, Enzymologic , Sterol Esterase/metabolism , 3-Hydroxybutyric Acid/metabolism , Animals , Apolipoproteins B/metabolism , Cell Line , Fasting , Fatty Liver/genetics , Female , Lipase , Lipogenesis , Male , Mice , Mice, Inbred C57BL , Oxidation-Reduction , Phenotype , Sterol Esterase/genetics , Triglycerides/metabolism , Up-Regulation
15.
Neuron ; 48(6): 1055-66, 2005 Dec 22.
Article in English | MEDLINE | ID: mdl-16364907

ABSTRACT

Recently developed therapeutics for obesity, targeted against cannabinoid receptors, result in decreased appetite and sustained weight loss. Prior studies have demonstrated CB1 receptors (CB1Rs) and leptin modulation of cannabinoid synthesis in hypothalamic neurons. Here, we show that depolarization of perifornical lateral hypothalamus (LH) neurons elicits a CB1R-mediated suppression of inhibition in local circuits thought to be involved in appetite and "natural reward." The depolarization-induced decrease in inhibitory tone to LH neurons is blocked by leptin. Leptin inhibits voltage-gated calcium channels in LH neurons via the activation of janus kinase 2 (JAK2) and of mitogen-activated protein kinase (MAPK). Leptin-deficient mice are characterized by both an increase in steady-state voltage-gated calcium currents in LH neurons and a CB1R-mediated depolarization-induced suppression of inhibition that is 6-fold longer than that in littermate controls. Our data provide direct electrophysiological support for the involvement of endocannabinoids and leptin as modulators of hypothalamic circuits underlying motivational aspects of feeding behavior.


Subject(s)
Appetite Regulation/physiology , Cannabinoid Receptor Modulators/metabolism , Endocannabinoids , Hypothalamic Area, Lateral/metabolism , Leptin/metabolism , Neural Pathways/metabolism , Signal Transduction/physiology , Animals , Appetite Regulation/drug effects , Calcium Channels/drug effects , Calcium Channels/metabolism , Calcium Signaling/drug effects , Calcium Signaling/genetics , Cannabinoid Receptor Modulators/pharmacology , Hypothalamic Area, Lateral/drug effects , Hypothalamus/drug effects , Hypothalamus/physiology , Janus Kinase 2 , Leptin/genetics , Leptin/pharmacology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motivation , Neural Inhibition/drug effects , Neural Inhibition/physiology , Neural Pathways/drug effects , Neurons/drug effects , Neurons/metabolism , Organ Culture Techniques , Patch-Clamp Techniques , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB1/metabolism , Receptors, Leptin , Reward , Signal Transduction/drug effects , Time Factors
16.
J Clin Invest ; 115(12): 3484-93, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16284652

ABSTRACT

We have generated mice that carry a neuron-specific leptin receptor (LEPR) transgene whose expression is driven by the rat synapsin I promoter synapsin-LEPR B (SYN-LEPR-B). We have also generated mice that are compound hemizygotes for the transgenes SYN-LEPR-B and neuron-specific enolase-LEPR B (NSE-LEPR-B). We observed a degree of correction in db/db mice that are hemizygous (Syn db/db) and homozygous (Syn/Syn db/db) for the SYN-LEPR-B transgene similar to that previously reported for the NSE-LEPR-B transgene. We also show complete correction of the obesity and related phenotypes of db/db mice that are hemizygous for both NSE-LEPR-B and SYN-LEPR-B transgenes (Nse+Syn db/db). Body composition, insulin sensitivity, and cold tolerance were completely normalized in Nse+Syn db/db mice at 12 weeks of age compared with lean controls. In situ hybridization for LEPR B isoform expression in Nse+Syn db/db mice showed robust expression in the energy homeostasis-relevant regions of the hypothalamus. Expression of 3 neuropeptide genes, agouti-related peptide (Agrp), neuropeptide Y (Npy), and proopiomelanocortin (Pomc), was fully normalized in dual transgenic db/db mice. The 2 transgenes in concert conferred normal fertility to male and female db/db mice. Male mice with partial peripheral deletion of Lepr, induced in the periweaning phase, did not show alterations in body composition or mass. In summary, we show that brain-specific leptin signaling is sufficient to reverse the obesity, diabetes, and infertility of db/db mice.


Subject(s)
Diabetes Mellitus/therapy , Genetic Therapy/methods , Infertility/therapy , Neurons/metabolism , Obesity/therapy , Receptors, Cell Surface/genetics , Agouti-Related Protein , Alleles , Animals , Blood Glucose/metabolism , Body Composition , Body Weight , Cold Temperature , DNA, Complementary/metabolism , Diabetes Mellitus/genetics , Female , Fertility , Gene Expression Regulation , Genotype , Glucose/metabolism , Homeostasis , Homozygote , Hypothalamus/pathology , In Situ Hybridization , Infertility/genetics , Infertility, Female/therapy , Infertility, Male/therapy , Insulin/metabolism , Intercellular Signaling Peptides and Proteins , Male , Mice , Mice, Transgenic , Neuropeptide Y/genetics , Obesity/genetics , Peptides/chemistry , Phenotype , Phosphopyruvate Hydratase/genetics , Polymerase Chain Reaction , Pro-Opiomelanocortin/genetics , Promoter Regions, Genetic , Protein Isoforms , Proteins/genetics , Rats , Receptors, Leptin , Signal Transduction , Synapsins/genetics , Time Factors , Tissue Distribution , Transgenes
17.
Cell Metab ; 1(1): 63-72, 2005 Jan.
Article in English | MEDLINE | ID: mdl-16054045

ABSTRACT

Leptin is required for normal energy and glucose homeostasis. The hypothalamic arcuate nucleus (ARH) has been proposed as an important site of leptin action. To assess the physiological significance of leptin signaling in the ARH, we used mice homozygous for a FLPe-reactivatable, leptin receptor null allele (Lepr(neo/neo) mice). Similar to Lepr(db/db) mice, these mice are obese, hyperglycemic, hyperinsulinemic, infertile, and hypoactive. To selectively restore leptin signaling in the ARH, we generated an adeno-associated virus expressing FLPe-recombinase, which was delivered unilaterally into the hypothalamus using stereotaxic injections. We found that unilateral restoration of leptin signaling in the ARH of Lepr(neo/neo) mice leads to a modest decrease in body weight and food intake. In contrast, unilateral reactivation markedly improved hyperinsulinemia and normalized blood glucose levels and locomotor activity. These data demonstrate that leptin signaling in the ARH is sufficient for mediating leptin's effects on glucose homeostasis and locomotor activity.


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Glucose/metabolism , Hypothalamus/metabolism , Leptin/metabolism , Alleles , Animals , Body Composition , Body Weight , Cell Nucleus/metabolism , DNA Nucleotidyltransferases/metabolism , Fertility , Green Fluorescent Proteins/metabolism , Homeostasis , Homozygote , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Genetic , Motor Activity , Neurons/metabolism , Oxygen Consumption , Receptors, Cell Surface/metabolism , Receptors, Leptin , Signal Transduction , Time Factors
18.
Am J Physiol Endocrinol Metab ; 289(3): E403-11, 2005 Sep.
Article in English | MEDLINE | ID: mdl-15870101

ABSTRACT

Leptin signaling in the brain regulates energy intake and expenditure. To test the degree of functional neuronal leptin signaling required for the maintenance of body composition, fertility, and cold tolerance, transgenic mice expressing Cre in neurons (CaMKIIalpha-Cre) were crossed to mice carrying a floxed leptin receptor (Lepr) allele to generate mice with neuron-specific deletion of Lepr in approximately 50% (C F/F mice) and approximately 75% (C Delta17/F mice) of hypothalamic neurons. Leptin receptor (LEPR)-deficient mice (Delta17/Delta17) with heat-shock-Cre-mediated global Lepr deletion served as obese controls. At 16 wk, male C F/F, C Delta17/F, and Delta17/Delta17 mice were 13.2 (P < 0.05), 45.0, and 55.9% (P < 0.001) heavier, respectively, than lean controls, whereas females showed 31.6, 68.8, and 160.7% increases in body mass (P < 0.001). Significant increases in total fat mass (C F/F: P < 0.01; C Delta17/F and Delta17/Delta17:P < 0.001 vs. sex-matched, lean controls), and serum leptin concentrations (P < 0.001 vs. controls) were present in proportion to Lepr deletion. Male C Delta17/F mice had significant elevations in basal serum insulin concentrations (P < 0.001 vs. controls) and were glucose intolerant, as measured by glucose tolerance test (AUC P < 0.01 vs. controls). In contrast with previous observations in mice null for LEPR signaling, C F/F and C Delta17/F mice were fertile and cold tolerant. These findings support the hypothesis that body weight, adiposity, serum leptin concentrations, and glucose intolerance are proportional to hypothalamic LEPR deficiency. However, fertility and cold tolerance remain intact unless hypothalamic LEPR deficiency is complete.


Subject(s)
Adaptation, Physiological/physiology , Cold Temperature , Diabetes Mellitus, Experimental/physiopathology , Fertility/physiology , Receptors, Cell Surface/genetics , Adipose Tissue, Brown/physiology , Animals , Arginine Vasopressin/genetics , Body Weight , DNA, Complementary , Diabetes Mellitus, Experimental/genetics , Eating , Female , Hypothalamus/cytology , Hypothalamus/physiology , Male , Mice , Mice, Knockout , Mice, Transgenic , Neurons/physiology , Neuropeptide Y/genetics , Obesity/genetics , Obesity/physiopathology , Pro-Opiomelanocortin/genetics , Receptors, Leptin
19.
Physiol Behav ; 82(1): 145-7, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15234603

ABSTRACT

Postnatal development in most mammals is accompanied by the acquisition of controls of ingestion. In rodents, the initial and default controller appears to be gastric stretch. In the second week of life, rat pups acquire the ability to sense the presence of nutrients within the gut and appropriately modulate ingestion. In the third week of life, rat pups start to become weaned from the dam's milk and begin independent ingestion. There have been strong indications that neuropeptide Y is a stimulator of ingestion in adults, although there was very little information in pups. Dr. Gerard Smith initiated a series of studies that provide strong evidence to indicate that hypothalamic neuropeptide Y (NPY) neurons are strong candidates for providing the ability of preweaning rat pups to modulate ingestion according to caloric intake. Moreover, the studies also suggest that the overactivity of hypothalamic NPY neurons presage the onset of hyperphagia in syndromes associated with defects in leptin signaling.


Subject(s)
Aging/physiology , Feeding Behavior/physiology , Gene Expression Regulation, Developmental/physiology , Hypothalamus/cytology , Neurons/physiology , Neuropeptide Y/genetics , Animals , Disease Models, Animal , Hypothalamus/growth & development , Mice , Neuropeptide Y/metabolism , Obesity/genetics , Rats
20.
Neuron ; 42(6): 983-91, 2004 Jun 24.
Article in English | MEDLINE | ID: mdl-15207242

ABSTRACT

Neuroanatomical and electrophysiological studies have shown that hypothalamic POMC neurons are targets of the adipostatic hormone leptin. However, the physiological relevance of leptin signaling in these neurons has not yet been directly tested. Here, using the Cre/loxP system, we critically test the functional importance of leptin action on POMC neurons by deleting leptin receptors specifically from these cells in mice. Mice lacking leptin signaling in POMC neurons are mildly obese, hyperleptinemic, and have altered expression of hypothalamic neuropeptides. In summary, leptin receptors on POMC neurons are required but not solely responsible for leptin's regulation of body weight homeostasis.


Subject(s)
Body Weight/physiology , Homeostasis/physiology , Neurons/metabolism , Pro-Opiomelanocortin/metabolism , Receptors, Cell Surface/physiology , Signal Transduction/physiology , Age Factors , Animals , Body Composition/genetics , Eating/genetics , Female , Gene Expression/genetics , Green Fluorescent Proteins , Hypothalamus/cytology , Hypothalamus/metabolism , Immunohistochemistry/methods , In Situ Hybridization/methods , Leptin/blood , Leptin/pharmacology , Luminescent Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neuropeptides/metabolism , Oxygen Consumption/genetics , Pro-Opiomelanocortin/genetics , RNA, Messenger/metabolism , Receptors, Leptin , Repressor Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods , Sex Factors , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins , Transcription Factors/metabolism , alpha-MSH/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL