Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 100
Filter
2.
Mol Metab ; 48: 101206, 2021 06.
Article in English | MEDLINE | ID: mdl-33684608

ABSTRACT

BACKGROUND: Over the past 20 years, insights from human and mouse genetics have illuminated the central role of the brain leptin-melanocortin pathway in controlling mammalian food intake, with genetic disruption resulting in extreme obesity, and more subtle polymorphic variations influencing the population distribution of body weight. At the end of 2020, the U.S. Food and Drug Administration (FDA) approved setmelanotide, a melanocortin 4 receptor agonist, for use in individuals with severe obesity due to either pro-opiomelanocortin (POMC), proprotein convertase subtilisin/kexin type 1 (PCSK1), or leptin receptor (LEPR) deficiency. SCOPE OF REVIEW: Herein, we chart the melanocortin pathway's history, explore its pharmacology, genetics, and physiology, and describe how a neuropeptidergic circuit became an important druggable obesity target. MAJOR CONCLUSIONS: Unravelling the genetics of the subset of severe obesity has revealed the importance of the melanocortin pathway in appetitive control; coupling this with studying the molecular pharmacology of compounds that bind melanocortin receptors has brought a new obesity drug to the market. This process provides a drug discovery template for complex disorders, which for setmelanotide took 25 years to transform from a single gene into an approved drug.


Subject(s)
Anti-Obesity Agents/therapeutic use , Energy Metabolism/drug effects , Homeostasis/drug effects , Melanocortins/metabolism , Obesity/drug therapy , Obesity/metabolism , Receptor, Melanocortin, Type 4/agonists , Signal Transduction/drug effects , alpha-MSH/analogs & derivatives , Animals , Anti-Obesity Agents/pharmacology , Drug Approval/history , Drug Discovery/history , History, 20th Century , History, 21st Century , Humans , Mice , Obesity/epidemiology , Receptor, Melanocortin, Type 4/metabolism , United States/epidemiology , alpha-MSH/pharmacology , alpha-MSH/therapeutic use
3.
FASEB J ; 35(1): e21269, 2021 01.
Article in English | MEDLINE | ID: mdl-33368660

ABSTRACT

Obese individuals often show low growth hormone (GH) secretion, which leads to reduced lipid mobilization and further fat accumulation. Pharmacological approaches to increase GH levels in obese individuals by GH injection or GH-releasing hormone receptor agonist showed promising effects on fat reduction. However, side effects on glucose metabolism and the heavy costs on making large peptides hindered their clinical application. Here, we tested whether stimulation of endogenous GH secretion by a synthetic GH secretagogue receptor (GHSR) agonist, hexarelin, improved the metabolism in a hyperphagic obese mouse model. Male melanocortin 4 receptor knockout mice (MC4RKO) were pair-fed and received continuous hexarelin (10.56 µg/day) or vehicle infusion by an osmotic pump for 3-4 weeks. Hexarelin treatment significantly increased the pulsatile GH secretion without detectable alteration on basal GH secretion in MC4RKO mice. The treated mice showed increased lipolysis and lipid oxidation in the adipose tissue, and reduced de novo lipogenesis in the liver, leading to reduced visceral fat mass, reduced triglyceride content in liver, and unchanged circulating free fatty acid levels. Importantly, hexarelin treatment improved the whole-body insulin sensitivity but did not alter glucose tolerance, insulin levels, or insulin-like growth factor 1 (IGF-1) levels. The metabolic effects of hexarelin were likely through the direct action of GH, as indicated by the increased expression level of genes involved in GH signaling pathways in visceral adipose tissues and liver. In conclusion, hexarelin treatment stimulated the pulsatile GH secretion and reduced the fat accumulation in visceral depots and liver in obese MC4RKO mice with improved insulin sensitivity without altered levels of insulin or IGF-1. It provides evidence for managing obesity by enhancing pulsatile GH secretion through activation of GHSR in the pituitary gland.


Subject(s)
Growth Hormone-Releasing Hormone/metabolism , Growth Hormone/metabolism , Lipid Metabolism/drug effects , Obesity/metabolism , Oligopeptides/pharmacology , Receptors, Ghrelin/metabolism , Animals , Disease Models, Animal , Intra-Abdominal Fat/metabolism , Liver/metabolism , Mice , Mice, Obese , Receptors, Ghrelin/agonists
4.
J Endocrinol ; 245(1): 1-12, 2020 04.
Article in English | MEDLINE | ID: mdl-31977312

ABSTRACT

The well-documented hormonal disturbance in a general obese population is characterised by an increase in insulin secretion and a decrease in growth hormone (GH) secretion. Such hormonal disturbance promotes an increase in fat mass, which deteriorates obesity and accelerates the development of insulin resistance and type 2 diabetes. While the pathological consequence is alarming, the pharmaceutical approach attempting to correct such hormonal disturbance remains limited. By applying an emerging anti-diabetic drug, the sodium-glucose cotransporter 2 inhibitor, dapagliflozin (1 mg/kg/day for 10 weeks), to a hyperphagic obese mouse model, we observed a significant improvement in insulin and GH secretion as early as 4 weeks after the initiation of the treatment. Restoration of pathological disturbance of insulin and GH secretion reduced fat accumulation and preserved lean body mass in the obese animal model. Such phenotypic improvement followed with concurrent improvements in glucose and lipid metabolism, insulin sensitivity, as well as the expression of metabolic genes that were regulated by insulin and GH. In conclusion, 10 weeks of treatment with dapagliflozin effectively reduces hyperinsulinemia and restores pulsatile GH secretion in the hyperphagic obese mice with considerable improvement in lipid and glucose metabolism. Promising outcomes from this study may provide insights into drug intervention to correct hormonal disturbance in obesity to delay the diabetes progression.


Subject(s)
Benzhydryl Compounds/pharmacology , Glucosides/pharmacology , Growth Hormone/metabolism , Insulin Secretion/drug effects , Insulin/metabolism , Obesity/metabolism , Animals , Body Composition/drug effects , Diabetes Mellitus, Type 2/metabolism , Energy Metabolism/drug effects , Energy Metabolism/genetics , Gene Expression/drug effects , Humans , Insulin Resistance , Mice, Inbred C57BL , Mice, Knockout , Obesity/genetics , Sodium-Glucose Transporter 2 Inhibitors/pharmacology
5.
Nature ; 574(7776): 63-68, 2019 10.
Article in English | MEDLINE | ID: mdl-31554967

ABSTRACT

The gp130 receptor cytokines IL-6 and CNTF improve metabolic homeostasis but have limited therapeutic use for the treatment of type 2 diabetes. Accordingly, we engineered the gp130 ligand IC7Fc, in which one gp130-binding site is removed from IL-6 and replaced with the LIF-receptor-binding site from CNTF, fused with the Fc domain of immunoglobulin G, creating a cytokine with CNTF-like, but IL-6-receptor-dependent, signalling. Here we show that IC7Fc improves glucose tolerance and hyperglycaemia and prevents weight gain and liver steatosis in mice. In addition, IC7Fc either increases, or prevents the loss of, skeletal muscle mass by activation of the transcriptional regulator YAP1. In human-cell-based assays, and in non-human primates, IC7Fc treatment results in no signs of inflammation or immunogenicity. Thus, IC7Fc is a realistic next-generation biological agent for the treatment of type 2 diabetes and muscle atrophy, disorders that are currently pandemic.


Subject(s)
Cytokine Receptor gp130/metabolism , Cytokines/chemical synthesis , Cytokines/therapeutic use , Diabetes Mellitus, Type 2/drug therapy , Immunoglobulin G/therapeutic use , Recombinant Fusion Proteins/therapeutic use , Adaptor Proteins, Signal Transducing/metabolism , Animals , Binding, Competitive , Cytokines/chemistry , Diabetes Mellitus, Type 2/metabolism , Drug Design , Fatty Liver/prevention & control , Glucose Tolerance Test , Humans , Hyperglycemia/drug therapy , Hyperglycemia/metabolism , Incretins/metabolism , Interleukin-6/antagonists & inhibitors , Interleukin-6/metabolism , Male , Mice , Muscle, Skeletal/drug effects , Obesity/metabolism , Pancreas/metabolism , Phosphoproteins/metabolism , Protein Engineering , Receptors, Interleukin-6/metabolism , Signal Transduction , Transcription Factors , Weight Gain/drug effects , YAP-Signaling Proteins
6.
Cell Rep ; 28(11): 2905-2922.e5, 2019 Sep 10.
Article in English | MEDLINE | ID: mdl-31509751

ABSTRACT

The importance of hypothalamic leptin and insulin resistance in the development and maintenance of obesity remains unclear. The tyrosine phosphatases protein tyrosine phosphatase 1B (PTP1B) and T cell protein tyrosine phosphatase (TCPTP) attenuate leptin and insulin signaling and are elevated in the hypothalami of obese mice. We report that elevated PTP1B and TCPTP antagonize hypothalamic leptin and insulin signaling and contribute to the maintenance of obesity. Deletion of PTP1B and TCPTP in the hypothalami of obese mice enhances CNS leptin and insulin sensitivity, represses feeding, and increases browning, to decrease adiposity and improve glucose metabolism. The daily intranasal administration of a PTP1B inhibitor, plus the glucocorticoid antagonist RU486 that decreases TCPTP expression, represses feeding, increases browning, promotes weight loss, and improves glucose metabolism in obese mice. Our findings causally link heightened hypothalamic PTP1B and TCPTP with leptin and insulin resistance and the maintenance of obesity and define a viable pharmacological approach by which to promote weight loss in obesity.


Subject(s)
Hypothalamus/metabolism , Insulin Resistance/genetics , Leptin/metabolism , Obesity/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 2/metabolism , Weight Loss/genetics , Adipose Tissue, White/metabolism , Administration, Intranasal , Animals , Blood-Brain Barrier/metabolism , Cholestanes/administration & dosage , Diet, High-Fat , Feeding Behavior/drug effects , Gliosis/genetics , Gliosis/metabolism , Glucocorticoids/pharmacology , Hypothalamus/drug effects , Leptin/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mifepristone/administration & dosage , Obesity/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 1/antagonists & inhibitors , Spermine/administration & dosage , Spermine/analogs & derivatives
7.
Mol Metab ; 26: 18-29, 2019 08.
Article in English | MEDLINE | ID: mdl-31230943

ABSTRACT

OBJECTIVE: Reelin (RELN) is a large glycoprotein involved in synapse maturation and neuronal organization throughout development. Deficits in RELN signaling contribute to multiple psychological disorders, such as autism spectrum disorder, schizophrenia, and bipolar disorder. Nutritional stress alters RELN expression in brain regions associated with these disorders; however, the involvement of RELN in the neural circuits involved in energy metabolism is unknown. The RELN receptors apolipoprotein E receptor 2 (ApoER2) and very low-density lipoprotein receptor (VLDLR) are involved in lipid metabolism and expressed in the hypothalamus. Here we explored the involvement of RELN in hypothalamic signaling and the impact of diet-induced obesity (DIO) on this system. METHODS: Adult male mice were fed a chow diet or maintained on a high-fat diet (HFD) for 12-16 weeks. HFD-fed DIO mice exhibited decreased ApoER2 and VLDLR expression and increased RELN protein in the hypothalamus. Electrophysiology was used to determine the mechanism by which the central fragment of RELN (CF-RELN) acts on arcuate nucleus (ARH) satiety-promoting proopiomelanocortin (POMC) neurons and the impact of DIO on this circuitry. RESULTS: CF-RELN exhibited heterogeneous presynaptic actions on inhibitory inputs onto ARH-POMC-EGFP neurons and consistent postsynaptic actions. Additionally, central administration of CF-RELN caused a significant increase in ARH c-Fos expression and an acute decrease in food intake and body weight. CONCLUSIONS: We conclude that RELN signaling is modulated by diet, that RELN is involved in synaptic signaling onto ARH-POMC neurons, and that altering central CF-RELN levels can impact food intake and body weight.


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Extracellular Matrix Proteins/metabolism , Nerve Tissue Proteins/metabolism , Obesity/metabolism , Pro-Opiomelanocortin/metabolism , Serine Endopeptidases/metabolism , Animals , Diet, High-Fat/adverse effects , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Obesity/chemically induced , Reelin Protein
8.
PLoS One ; 14(4): e0214621, 2019.
Article in English | MEDLINE | ID: mdl-30947272

ABSTRACT

Experimental non-human primate models of obesity are induced through the introduction of atypically calorically rich diets. Studies in captive-bred macaques show the development of obesity and diabetes with similar complications to humans including eye and kidney diseases, nerve damage associated with pain and blood vessel damage. Diets differ in outcomes and here we document inflammation of the gastrointestinal tract that can be exacerbated through these dietary interventions. Following baseline physiological evaluation of body composition, Southern pigtail macaques were given a high-fat diet (HFD) for three months. This HFD consisted of lard, grains (including gluten), dairy and fructose that was otherwise omitted from a standard macaque diet (Chow). Physiological parameters were then reassessed before animals were reverted back to standard Chow for a further three months (remission). Consumption of the HFD resulted in food-mediated hypersensitivity marked by chronic weight loss, alopecia, malabsorption, protein-losing enteropathy and gross diffuse intestinal villi atrophy and lamina propria hypertrophy. Physiological changes were more highly pronounced in female macaques suggesting sex-specific differences but could be fully reversed through change of diet. Care should be taken in choosing non-human primate HFD diets for creating experimental models of obesity because they can induce severe food-driven chronic inflammation of the gastrointestinal tract that can eventuate to diet-induced chronic wasting and mortality.


Subject(s)
Diet, High-Fat , Food Hypersensitivity/pathology , Gastrointestinal Diseases/pathology , Macaca nemestrina , Animals , Body Composition , Chronic Disease , Female , Inflammation/pathology , Male , Sex Factors
9.
Neuroendocrinology ; 109(4): 374-390, 2019.
Article in English | MEDLINE | ID: mdl-30995667

ABSTRACT

BACKGROUND: Obesity is associated with reduced physiological responses to leptin and insulin, leading to the concept of obesity-associated hormonal resistance. OBJECTIVES: Here, we demonstrate that contrary to expectations, leptin signaling not only remains functional but also is constantly activated in the arcuate nucleus of the hypothalamus (ARH) neurons of obese mice. This state of persistent response to endogenous leptin underpins the lack of response to exogenous leptin. METHODS AND RESULTS: The study of combined leptin and insulin signaling demonstrates that there is a common pool of ARH neurons responding to both hormones. More importantly, we show that the constant activation of leptin receptor neurons in the ARH prevents insulin signaling in these neurons, leading to impaired glucose tolerance. Accordingly, antagonising leptin signaling in diet-induced obese (DIO) mice restores insulin signaling in the ARH and improves glucose homeostasis. Direct inhibition of PTP1B in the CNS restores arcuate insulin signaling similarly to leptin inhibition; this effect is likely to be mediated by AgRP neurons since PTP1B deletion specifically in AgRP neurons restores glucose and insulin tolerance in DIO mice. CONCLUSIONS: Finally, our results suggest that the constant activation of arcuate leptin signaling in DIO mice increases PTP1B expression, which exerts an inhibitory effect on insulin signaling leading to impaired glucose homeostasis.


Subject(s)
Arcuate Nucleus of Hypothalamus/physiopathology , Glucose/metabolism , Homeostasis , Hypothalamus/physiopathology , Insulin , Leptin , Obesity/physiopathology , Animals , Diet , Diet, High-Fat , Genotype , Glucose Tolerance Test , Mice , Mice, Inbred C57BL , Neurons , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , STAT3 Transcription Factor/biosynthesis , Signal Transduction
10.
Diabetes ; 68(4): 683-695, 2019 04.
Article in English | MEDLINE | ID: mdl-30674622

ABSTRACT

Liraglutide, a glucagon-like peptide 1 (GLP-1) receptor agonist, and phentermine, a psychostimulant structurally related to amphetamine, are drugs approved for the treatment of obesity and hyperphagia. There is significant interest in combination use of liraglutide and phentermine for weight loss; however, both drugs have been reported to induce systemic hemodynamic changes, and as such the therapeutic window for this drug combination needs to be determined. To understand their impact on metabolic and cardiovascular physiology, we tested the effects of these drugs alone and in combination for 21 days in lean and obese male mice. The combination of liraglutide and phentermine, at 100 µg/kg/day and 10 mg/kg/day, respectively, produced the largest reduction in body weight in both lean and diet-induced obese (DIO) mice, when compared with both vehicle and monotherapy-treated mice. In lean mice, combination treatment at the aforementioned doses significantly increased heart rate and reduced blood pressure, whereas in DIO mice, combination therapy induced a transient increase in heart rate and decreased blood pressure. These studies demonstrate that in obese mice, the combination of liraglutide and phentermine may reduce body weight but only induce modest improvements in cardiovascular functions. Conversely, in lean mice, the additional weight loss from combination therapy does not improve cardiovascular parameters.


Subject(s)
Anti-Obesity Agents/pharmacology , Blood Pressure/drug effects , Body Weight/drug effects , Heart Rate/drug effects , Liraglutide/pharmacology , Phentermine/pharmacology , Animals , Anti-Obesity Agents/therapeutic use , Drug Therapy, Combination , Liraglutide/therapeutic use , Male , Mice , Obesity/drug therapy , Phentermine/therapeutic use , Treatment Outcome , Weight Loss/drug effects
12.
Cell Rep ; 26(2): 346-355.e3, 2019 01 08.
Article in English | MEDLINE | ID: mdl-30625317

ABSTRACT

Insulin action in the hypothalamus results in the suppression of hepatic glucose production (HGP). Obesity is often associated with a diminished response to insulin, leading to impaired suppression of HGP in obese mice. Here, we demonstrate that blocking central leptin signaling in diet-induced obese (DIO) mice restores the liver's ability to suppress glucose production. Leptin increases the expression of the insulin receptor phosphatase PTP1B, which is highly expressed in the hypothalamus of DIO mice. We demonstrate that the central pharmacological inhibition or ARH-targeted deletion of PTP1B restores the suppression of HGP in obese mice. Additionally, mice that lack PTP1B in AgRP neurons exhibit enhanced ARH insulin signaling and have improved glucose tolerance and insulin sensitivity. Overall, our findings indicate that obesity-induced increases in PTP1B diminish insulin action in the hypothalamus, resulting in unconstrained HGP and contributing to hyperglycemia in obesity.


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Gluconeogenesis , Insulin/metabolism , Leptin/metabolism , Liver/metabolism , Obesity/metabolism , Animals , Diet, High-Fat/adverse effects , Male , Mice , Mice, Inbred C57BL , Obesity/etiology , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Signal Transduction
13.
Nat Metab ; 1(9): 851-852, 2019 09.
Article in English | MEDLINE | ID: mdl-32694744
14.
J Med Chem ; 61(24): 11144-11157, 2018 12 27.
Article in English | MEDLINE | ID: mdl-30525586

ABSTRACT

Celastrol is a natural pentacyclic triterpene used in traditional Chinese medicine with significant weight-lowering effects. Celastrol-administered mice at 100 µg/kg decrease food consumption and body weight via a leptin-dependent mechanism, yet its molecular targets in this pathway remain elusive. Here, we demonstrate in vivo that celastrol-induced weight loss is largely mediated by the inhibition of leptin negative regulators protein tyrosine phosphatase (PTP) 1B (PTP1B) and T-cell PTP (TCPTP) in the arcuate nucleus (ARC) of the hypothalamus. We show in vitro that celastrol binds reversibly and inhibits noncompetitively PTP1B and TCPTP. NMR data map the binding site to an allosteric site in the catalytic domain that is in proximity of the active site. By using a panel of PTPs implicated in hypothalamic leptin signaling, we show that celastrol additionally inhibited PTEN and SHP2 but had no activity toward other phosphatases of the PTP family. These results suggest that PTP1B and TCPTP in the ARC are essential for celastrol's weight lowering effects in adult obese mice.


Subject(s)
Anti-Obesity Agents/pharmacology , Obesity/drug therapy , Protein Tyrosine Phosphatase, Non-Receptor Type 1/antagonists & inhibitors , Protein Tyrosine Phosphatase, Non-Receptor Type 2/antagonists & inhibitors , Triterpenes/pharmacology , Allosteric Site , Animals , Anti-Obesity Agents/metabolism , Catalytic Domain , Diet, High-Fat/adverse effects , Hypothalamus/drug effects , Hypothalamus/metabolism , Magnetic Resonance Spectroscopy , Male , Mice, Transgenic , Obesity/etiology , Pentacyclic Triterpenes , Protein Tyrosine Phosphatase, Non-Receptor Type 1/chemistry , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 2/chemistry , Protein Tyrosine Phosphatase, Non-Receptor Type 2/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 2/metabolism , Structure-Activity Relationship , Triterpenes/chemistry , Triterpenes/metabolism , Weight Loss/drug effects
15.
Nat Commun ; 9(1): 4975, 2018 11 20.
Article in English | MEDLINE | ID: mdl-30459311

ABSTRACT

In the original PDF version of this article, affiliation 1, 'Institute for Diabetes and Obesity, Helmholtz Diabetes Center (HDC), Helmholtz Zentrum Muenchen & German Center for Diabetes Research (DZD), Neuherberg, Germany', was incorrectly given as 'Institute of Diabetes and Regeneration Research, Helmholtz Zentrum Muenchen, German Research Center for Environmental Health (GmbH), Neuherberg, Germany '. This has now been corrected in the PDF version of the article; the HTML version was correct at the time of publication.

16.
Nat Commun ; 9(1): 4304, 2018 10 23.
Article in English | MEDLINE | ID: mdl-30353008

ABSTRACT

Pharmacological stimulation of brown adipose tissue (BAT) thermogenesis to increase energy expenditure is progressively being pursued as a viable anti-obesity strategy. Here, we report that pharmacological activation of the cold receptor transient receptor potential cation channel subfamily M member 8 (TRPM8) with agonist icilin mimics the metabolic benefits of cold exposure. In diet-induced obese (DIO) mice, treatment with icilin enhances energy expenditure, and decreases body weight, without affecting food intake. To further potentiate the thermogenic action profile of icilin and add complementary anorexigenic mechanisms, we set out to identify pharmacological partners next to icilin. To that end, we specifically targeted nicotinic acetylcholine receptor (nAChR) subtype alpha3beta4 (α3ß4), which we had recognized as a potential regulator of energy homeostasis and glucose metabolism. Combinatorial targeting of TRPM8 and nAChR α3ß4 by icilin and dimethylphenylpiperazinium (DMPP) orchestrates synergistic anorexic and thermogenic pathways to reverse diet-induced obesity, dyslipidemia, and glucose intolerance in DIO mice.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Obesity/metabolism , Receptors, Nicotinic/metabolism , TRPM Cation Channels/antagonists & inhibitors , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/metabolism , Animals , Body Weight/drug effects , Cold Temperature , Diabetes Mellitus, Type 2/drug therapy , Diet , Dimethylphenylpiperazinium Iodide/pharmacology , Dimethylphenylpiperazinium Iodide/therapeutic use , Energy Metabolism/drug effects , Fatty Liver/pathology , Glucose Intolerance/pathology , Insulin Resistance , Male , Melanocortins/metabolism , Mice, Inbred C57BL , Mice, Obese , Obesity/drug therapy , Pyrimidinones/pharmacology , Pyrimidinones/therapeutic use , Receptor, Melanocortin, Type 4/metabolism , TRPM Cation Channels/metabolism , Thermogenesis/drug effects
17.
Elife ; 72018 09 19.
Article in English | MEDLINE | ID: mdl-30230471

ABSTRACT

Hypothalamic neurons respond to nutritional cues by altering gene expression and neuronal excitability. The mechanisms that control such adaptive processes remain unclear. Here we define populations of POMC neurons in mice that are activated or inhibited by insulin and thereby repress or inhibit hepatic glucose production (HGP). The proportion of POMC neurons activated by insulin was dependent on the regulation of insulin receptor signaling by the phosphatase TCPTP, which is increased by fasting, degraded after feeding and elevated in diet-induced obesity. TCPTP-deficiency enhanced insulin signaling and the proportion of POMC neurons activated by insulin to repress HGP. Elevated TCPTP in POMC neurons in obesity and/or after fasting repressed insulin signaling, the activation of POMC neurons by insulin and the insulin-induced and POMC-mediated repression of HGP. Our findings define a molecular mechanism for integrating POMC neural responses with feeding to control glucose metabolism.


Subject(s)
Glucose/metabolism , Insulin/pharmacology , Neuronal Plasticity/drug effects , Neurons/metabolism , Pro-Opiomelanocortin/metabolism , Animals , Humans , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/pharmacology , Hypothalamus/cytology , Insulin/administration & dosage , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neuronal Plasticity/genetics , Pro-Opiomelanocortin/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 2/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 2/metabolism , Receptor, Insulin/genetics , Receptor, Insulin/metabolism
18.
Diabetes ; 67(11): 2456-2465, 2018 11.
Article in English | MEDLINE | ID: mdl-30158241

ABSTRACT

Celastrol, a plant-derived constituent of traditional Chinese medicine, has been proposed to offer significant potential as an antiobesity drug. However, the molecular mechanism for this activity is unknown. We show that the weight-lowering effects of celastrol are driven by decreased food consumption. Although young Lep ob mice respond with a decrease in food intake and body weight, adult Lep db and Lep ob mice are unresponsive to celastrol, suggesting that functional leptin signaling in adult mice is required to elicit celastrol's catabolic actions. Protein tyrosine phosphatase 1 (PTP1B), a leptin negative-feedback regulator, has been previously reported to be one of celastrol's targets. However, we found that global PTP1B knockout (KO) and wild-type (WT) mice have comparable weight loss and hypophagia when treated with celastrol. Increased levels of uncoupling protein 1 (UCP1) in subcutaneous white and brown adipose tissue suggest celastrol-induced thermogenesis as a further mechanism. However, diet-induced obese UCP1 WT and KO mice have comparable weight loss upon celastrol treatment, and celastrol treatment has no effect on energy expenditure under ambient housing or thermoneutral conditions. Overall, our results suggest that celastrol-induced weight loss is hypophagia driven and age-dependently mediated by functional leptin signaling. Our data encourage reconsideration of therapeutic antiobesity strategies built on leptin sensitization.


Subject(s)
Eating/drug effects , Obesity/metabolism , Plant Extracts/pharmacology , Triterpenes/pharmacology , Uncoupling Protein 1/metabolism , Weight Loss/drug effects , Animals , Diet, High-Fat , Energy Metabolism/drug effects , Mice, Knockout , Obesity/genetics , Pentacyclic Triterpenes , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Uncoupling Protein 1/genetics
19.
Physiol Behav ; 194: 184-190, 2018 10 01.
Article in English | MEDLINE | ID: mdl-29842854

ABSTRACT

Obesity is an ongoing global public health problem. For many people dieting is the preferred method of combating elevated body fat. Weight lost during caloric restriction is often soon regained and so a pattern of recurrent dieting develops. Here an individual's food intake fluctuates up and down with intermittent periods of normal eating and restrained eating. The metabolic consequences of 'yoyo dieting' or 'weight cycling' are not well understood. Here we monitor the effects of multiple, repeated dieting periods on body composition and metabolic health in overweight mice. Compared to mice that were continuously fed a high fat diet, the energy expenditure of diet-cycled mice was reduced. This resulted in mice rapidly regaining body weight upon the reintroduction of high fat chow diet subsequent to periods of caloric restriction. Diet cycling also increased the appetite for high fat chow and diminished glucose tolerance. These data demonstrate the detrimental effects of diet cycling upon metabolic health.


Subject(s)
Appetite/physiology , Caloric Restriction , Glucose Intolerance/physiopathology , Motor Activity/physiology , Obesity/diet therapy , Weight Loss/physiology , Animals , Body Composition , Body Weight , Diet, High-Fat , Disease Models, Animal , Eating , Feeding Behavior , Male , Mice, Inbred C57BL , Obesity/physiopathology
20.
Trends Endocrinol Metab ; 29(4): 218-237, 2018 04.
Article in English | MEDLINE | ID: mdl-29475578

ABSTRACT

Metabolic surgical procedures, such as Roux-en-Y gastric bypass (RYGB), uniquely reprogram feeding behavior and body weight in obese subjects. Clinical neuroimaging and animal studies are only now beginning to shed light on some of the underlying central mechanisms. We present here the roles of key brain neurotransmitter/neuromodulator systems in food choice, value, and intake at various stages after RYGB. In doing so, we elaborate on how known signals emanating from the reorganized gut, including peptide hormones and microbiota products, impinge on newly mapped homeostatic and hedonic brain feeding circuits. Continued progress in the rapidly evolving field of metabolic surgery will inform the design of more effective weight-loss compounds.


Subject(s)
Brain/diagnostic imaging , Gastric Bypass , Nerve Net/diagnostic imaging , Obesity/surgery , Animals , Body Weight/physiology , Humans , Neuroimaging , Obesity/diagnostic imaging
SELECTION OF CITATIONS
SEARCH DETAIL
...