Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Toxicology ; 505: 153839, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38782113

ABSTRACT

Neonicotinoid (NN) pesticides have been linked to increased brain dysfunction in mammals, such as anxiety-like behavior; this is thought to involve monoamines (MA), neurotransmitters that control behavior, memory, and learning. However, the mechanism by which NNs affect the central nervous system is not fully understood. In this study, we aimed to investigate whether MAs affect NNs-induced anxiety-like behavior. Mice were orally administered acetamiprid (ACE), an NN, at the no observed adverse effect level (NOAEL) of mouse (20 mg/kg body mass) set by the Food Safety Commission of Japan, and the elevated zero-maze (EZM) test was performed 30 min after administration. After behavioral analysis, levels of four MA (dopamine, 3-MT, serotonin, and histamine) in selected brain regions were determined by liquid chromatography mass spectrometry (LC/MS/MS). In the exposed group, a trend toward increased anxiety-like behavior was observed, and at least one MA concentration was significantly increased in each region. Further, significant correlations were found between behavioral test results and hippocampal serotonin and striatal dopamine concentrations, as well as between dopamine and serotonin concentrations, in the exposed group. As anxiety can influence activity in the behavioral tests, the activity of neurons in the raphe nuclei (RN), a brain region greatly involved in anxiety via the serotonergic system, was examined by staining with anti-serotonin antibodies, and increased serotonergic activity was observed. Taken together, these results suggest that ACE regulates MA levels, notably serotonin levels in the hippocampus and that RN plays an important role in ACE-induced anxiety-like behavior.


Subject(s)
Anxiety , Behavior, Animal , Biogenic Monoamines , Brain , Neonicotinoids , Animals , Anxiety/chemically induced , Anxiety/metabolism , Neonicotinoids/toxicity , Male , Biogenic Monoamines/metabolism , Mice , Brain/drug effects , Brain/metabolism , Behavior, Animal/drug effects , Insecticides/toxicity , Mice, Inbred ICR , Maze Learning/drug effects , Serotonin/metabolism , Dopamine/metabolism
2.
J Vis Exp ; (203)2024 Jan 26.
Article in English | MEDLINE | ID: mdl-38345259

ABSTRACT

Diabetes mellitus (DM) is caused by insufficient insulin release from the pancreatic ß-cells (Type1 DM) and insulin sensitivity in muscles, liver, and adipose tissues (Type2 DM). Insulin injection treats DM patients but leads to hypoglycemia as a side effect. Cortisol and catecholamines are released to activate glucose production from the liver to recover hypoglycemia, called counter-regulatory responses (CRR). In DM research using rodent models, glucose tolerance tests and 2-deoxy-glucose injection are used to measure insulin release and CRR, respectively. However, blood glucose concentrations change persistently during experiments, causing difficulties in assessing net insulin release and CRR. This article describes a method in which blood glucose is kept at 250 mg/dL or 50 mg/dL in conscious mice to compare the release of insulin and CRR hormones, respectively. Polyethylene tubing is implanted in the mice's carotid artery and jugular vein, and the mice are allowed to recover from the surgery. The jugular vein tubing is connected to a Hamilton syringe with a syringe pump to enable insulin or glucose infusion at a constant and variable rate. The carotid artery tubing is for blood collection. For the hyperglycemic clamp, 30% glucose is infused into the vein, and blood glucose levels are measured from the arterial blood every 5 min or 10 min. The infusion rate of 30% glucose is increased until the blood glucose level becomes 250 mg/dL. Blood is collected to measure insulin concentrations. For hypoglycemic clamp, 10 mU/kg/min insulin is infused together with 30% glucose, whose infusion rate is variable to maintain 50 mg/dL of blood glucose level. Blood is collected to measure counter-regulatory hormones when both glucose infusion and blood glucose reach a steady state. Both hyperglycemic and hypoglycemic clamps have the same surgical procedure and experimental setups. Thus, this method is useful for researchers of systemic glucose metabolism.


Subject(s)
Diabetes Mellitus , Hypoglycemia , Humans , Mice , Animals , Blood Glucose/metabolism , Hypoglycemic Agents/pharmacology , Glucose Clamp Technique , Hypoglycemia/metabolism , Insulin/metabolism , Glucose/metabolism
3.
J Neurosci ; 42(23): 4607-4618, 2022 06 08.
Article in English | MEDLINE | ID: mdl-35504726

ABSTRACT

Ubiquitin-specific protease 2 (USP2) participates in glucose metabolism in peripheral tissues such as the liver and skeletal muscles. However, the glucoregulatory role of USP2 in the CNS is not well known. In this study, we focus on USP2 in the ventromedial hypothalamus (VMH), which has dominant control over systemic glucose homeostasis. ISH, using a Usp2-specific probe, showed that Usp2 mRNA is present in VMH neurons, as well as other glucoregulatory nuclei, in the hypothalamus of male mice. Administration of a USP2-selective inhibitor ML364 (20 ng/head), into the VMH elicited a rapid increase in the circulating glucose level in male mice, suggesting USP2 has a suppressive role on glucose mobilization. ML364 treatment also increased serum norepinephrine concentration, whereas it negligibly affected serum levels of insulin and corticosterone. ML364 perturbated mitochondrial oxidative phosphorylation in neural SH-SY5Y cells and subsequently promoted the phosphorylation of AMP-activated protein kinase (AMPK). Consistent with these findings, hypothalamic ML364 treatment stimulated AMPKα phosphorylation in the VMH. Inhibition of hypothalamic AMPK prevented ML364 from increasing serum norepinephrine and blood glucose. Removal of ROS restored the ML364-evoked mitochondrial dysfunction in SH-SY5Y cells and impeded the ML364-induced hypothalamic AMPKα phosphorylation as well as prevented the elevation of serum norepinephrine and blood glucose levels in male mice. These results indicate hypothalamic USP2 attenuates perturbations in blood glucose levels by modifying the ROS-AMPK-sympathetic nerve axis.SIGNIFICANCE STATEMENT Under normal conditions (excluding hyperglycemia or hypoglycemia), blood glucose levels are maintained at a constant level. In this study, we used a mouse model to identify a hypothalamic protease controlling blood glucose levels. Pharmacological inhibition of USP2 in the VMH caused a deviation in blood glucose levels under a nonstressed condition, indicating that USP2 determines the set point of the blood glucose level. Modification of sympathetic nervous activity accounts for the USP2-mediated glucoregulation. Mechanistically, USP2 mitigates the accumulation of ROS in the VMH, resulting in attenuation of the phosphorylation of AMPK. Based on these findings, we uncovered a novel glucoregulatory axis consisting of hypothalamic USP2, ROS, AMPK, and the sympathetic nervous system.


Subject(s)
Blood Glucose , Neuroblastoma , Sympathetic Nervous System , Ubiquitin Thiolesterase , Ventromedial Hypothalamic Nucleus , AMP-Activated Protein Kinases/metabolism , Animals , Blood Glucose/metabolism , Glucose/metabolism , Humans , Male , Mice , Norepinephrine/metabolism , Oxidative Phosphorylation , Reactive Oxygen Species/metabolism , Sympathetic Nervous System/enzymology , Sympathetic Nervous System/metabolism , Ubiquitin Thiolesterase/metabolism , Ventromedial Hypothalamic Nucleus/enzymology , Ventromedial Hypothalamic Nucleus/metabolism
4.
Mol Metab ; 54: 101366, 2021 12.
Article in English | MEDLINE | ID: mdl-34728342

ABSTRACT

OBJECTIVE: The regulation of food intake is a major research area in the study of obesity, which plays a key role in the development of metabolic syndrome. Gene targeting studies have clarified the roles of hypothalamic neurons in feeding behavior, but the deletion of a gene has a long-term effect on neurophysiology. Our understanding of short-term changes such as appetite under physiological conditions is therefore still limited. METHODS: Targeted recombination in active populations (TRAP) is a newly developed method for labeling active neurons by using tamoxifen-inducible Cre recombination controlled by the promoter of activity-regulated cytoskeleton-associated protein (Arc/Arg3.1), a member of immediate early genes. Transgenic mice for TRAP were fasted overnight, re-fed with normal diet, and injected with 4-hydroxytamoxifen 1 h after the refeeding to label the active neurons. The role of labeled neurons was examined by expressing excitatory or inhibitory designer receptors exclusively activated by designer drugs (DREADDs). The labeled neurons were extracted and RNA sequencing was performed to identify genes that are specifically expressed in these neurons. RESULTS: Fasting-refeeding activated and labeled neurons in the compact part of the dorsomedial hypothalamus (DMH) that project to the paraventricular hypothalamic nucleus. Chemogenetic activation of the labeled DMH neurons decreased food intake and developed place preference, an indicator of positive valence. Chemogenetic activation or inhibition of these neurons had no influence on the whole-body glucose metabolism. The labeled DMH neurons expressed prodynorphin (pdyn), gastrin-releasing peptide (GRP), cholecystokinin (CCK), and thyrotropin-releasing hormone receptor (Trhr) genes. CONCLUSIONS: We identified a novel cell type of DMH neurons that can inhibit food intake and promote feeding-induced positive valence. Our study provides insight into the role of DMH and its molecular mechanism in the regulation of appetite and emotion.


Subject(s)
Dorsomedial Hypothalamic Nucleus/metabolism , Eating , Neurons/metabolism , Animals , Cholecystokinin/genetics , Enkephalins/genetics , Feeding Behavior , Male , Mice , Mice, Transgenic , Protein Precursors/genetics
5.
Nat Commun ; 12(1): 2330, 2021 04 20.
Article in English | MEDLINE | ID: mdl-33879780

ABSTRACT

The hypothalamus plays a central role in monitoring and regulating systemic glucose metabolism. The brain is enriched with phospholipids containing poly-unsaturated fatty acids, which are biologically active in physiological regulation. Here, we show that intraperitoneal glucose injection induces changes in hypothalamic distribution and amounts of phospholipids, especially arachidonic-acid-containing phospholipids, that are then metabolized to produce prostaglandins. Knockdown of cytosolic phospholipase A2 (cPLA2), a key enzyme for generating arachidonic acid from phospholipids, in the hypothalamic ventromedial nucleus (VMH), lowers insulin sensitivity in muscles during regular chow diet (RCD) feeding. Conversely, the down-regulation of glucose metabolism by high fat diet (HFD) feeding is improved by knockdown of cPLA2 in the VMH through changing hepatic insulin sensitivity and hypothalamic inflammation. Our data suggest that cPLA2-mediated hypothalamic phospholipid metabolism is critical for controlling systemic glucose metabolism during RCD, while continuous activation of the same pathway to produce prostaglandins during HFD deteriorates glucose metabolism.


Subject(s)
Glucose/metabolism , Phospholipases A2, Cytosolic/metabolism , Prostaglandins/biosynthesis , Ventromedial Hypothalamic Nucleus/metabolism , Animals , Arachidonic Acid/metabolism , Biosynthetic Pathways , Diet, High-Fat/adverse effects , Disease Models, Animal , Gene Knockdown Techniques , Group IV Phospholipases A2/antagonists & inhibitors , Group IV Phospholipases A2/genetics , Group IV Phospholipases A2/metabolism , Hyperglycemia/metabolism , Insulin Resistance , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phospholipases A2, Cytosolic/antagonists & inhibitors , Phospholipases A2, Cytosolic/genetics , Phospholipids/metabolism
7.
Cell Rep ; 22(3): 706-721, 2018 01 16.
Article in English | MEDLINE | ID: mdl-29346768

ABSTRACT

Food selection is essential for metabolic homeostasis and is influenced by nutritional state, food palatability, and social factors such as stress. However, the mechanism responsible for selection between a high-carbohydrate diet (HCD) and a high-fat diet (HFD) remains unknown. Here, we show that activation of a subset of corticotropin-releasing hormone (CRH)-positive neurons in the rostral region of the paraventricular hypothalamus (PVH) induces selection of an HCD over an HFD in mice during refeeding after fasting, resulting in a rapid recovery from the change in ketone metabolism. These neurons manifest activation of AMP-activated protein kinase (AMPK) during food deprivation, and this activation is necessary and sufficient for selection of an HCD over an HFD. Furthermore, this effect is mediated by carnitine palmitoyltransferase 1c (CPT1c). Thus, our results identify the specific neurons and intracellular signaling pathway responsible for regulation of the complex behavior of selection between an HCD and an HFD. VIDEO ABSTRACT.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Corticotropin-Releasing Hormone/metabolism , Neurons/physiology , Animals , Carbohydrates , Diet , Male , Mice
8.
Sci Rep ; 7(1): 15141, 2017 11 09.
Article in English | MEDLINE | ID: mdl-29123236

ABSTRACT

Leptin increases glucose uptake and fatty acid oxidation (FAO) in red-type skeletal muscle. However, the mechanism remains unknown. We have investigated the role of ß2-adrenergic receptor (AR), the major ß-AR isoform in skeletal muscle, and AMPK in leptin-induced muscle glucose uptake of mice. Leptin injection into the ventromedial hypothalamus (VMH) increased 2-deoxy-D-glucose (2DG) uptake in red-type skeletal muscle in wild-type (WT) mice accompanied with increased phosphorylation of the insulin receptor (IR) and Akt as well as of norepinephrine (NE) turnover in the muscle. Leptin-induced 2DG uptake was not observed in ß-AR-deficient (ß-less) mice despite that AMPK phosphorylation was increased in the muscle. Forced expression of ß2-AR in the unilateral hind limb of ß-less mice restored leptin-induced glucose uptake and enhancement of insulin signalling in red-type skeletal muscle. Leptin increased 2DG uptake and enhanced insulin signalling in red-type skeletal muscle of mice expressing a dominant negative form of AMPK (DN-AMPK) in skeletal muscle. Thus, leptin increases glucose uptake and enhances insulin signalling in red-type skeletal muscle via activation of sympathetic nerves and ß2-AR in muscle and in a manner independent of muscle AMPK.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Glucose/metabolism , Leptin/metabolism , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Mice , Receptors, Adrenergic, beta-2
9.
Biochem Biophys Rep ; 9: 322-329, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28956020

ABSTRACT

We previously reported that ubiquitin-specific protease (USP) 2 in macrophages down-regulates genes associated with metabolic diseases, suggesting a putative anti-diabetic role for USP2 in macrophages. In this study, we evaluate this role at both cellular and individual levels. Isolated macrophages forcibly expressing Usp2a, a longer splicing variant of USP2, failed to modulate the insulin sensitivity of 3T3-L1 adipocytes. Similarly, macrophage-selective overexpression of Usp2a in mice (Usp2a transgenic mice) had a negligible effect on insulin sensitivity relative to wild type littermates following a three-month high-fat diet. However, Usp2a transgenic mice exhibited fewer M1 macrophages in their mesenteric adipose tissue. Following a six-month high-fat diet, Usp2a transgenic mice exhibited a retarded progression of insulin resistance in their skeletal muscle and liver, and an improvement in insulin sensitivity at an individual level. Although conditioned media from Usp2a-overexpressing macrophages did not directly affect the insulin sensitivity of C2C12 myotubes compared to media from control macrophages, they did increase the insulin sensitivity of C2C12 cells after subsequent conditioning with 3T3-L1 cells. These results indicate that macrophage USP2A hampers obesity-elicited insulin resistance via an adipocyte-dependent mechanism.

10.
Diabetes ; 66(8): 2102-2111, 2017 08.
Article in English | MEDLINE | ID: mdl-28550108

ABSTRACT

The Lin28a/Let-7 axis has been studied in peripheral tissues for its role in metabolism regulation. However, its central function remains unclear. Here we found that Lin28a is highly expressed in the hypothalamus compared with peripheral tissues. Its expression is positively correlated with positive energy balance, suggesting a potential central role for Lin28a in metabolism regulation. Thus, we targeted the hypothalamic ventromedial nucleus (VMH) to selectively overexpress (Lin28aKIVMH ) or downregulate (Lin28aKDVMH ) Lin28a expression in mice. With mice on a standard chow diet, body weight and glucose homeostasis were not affected in Lin28aKIVMH or Lin28aKDVMH mice. On a high-fat diet, although no differences in body weight and composition were observed, Lin28aKIVMH mice showed improved glucose tolerance and insulin sensitivity compared with controls. Conversely, Lin28aKDVMH mice displayed glucose intolerance and insulin resistance. Changes in VMH AKT activation of diet-induced obese Lin28aKIVMH or Lin28aKDVMH mice were not associated with alterations in Let-7 levels or insulin receptor activation. Rather, we observed altered expression of TANK-binding kinase-1 (TBK-1), which was found to be a direct Lin28a target mRNA. VMH-specific inhibition of TBK-1 in mice with diet-induced obesity impaired glucose metabolism and AKT activation. Altogether, our data show a TBK-1-dependent role for central Lin28a in glucose homeostasis.


Subject(s)
Glucose/metabolism , Obesity/metabolism , RNA-Binding Proteins/physiology , Ventromedial Hypothalamic Nucleus/metabolism , Animals , Body Weight/physiology , Diet, High-Fat/adverse effects , Down-Regulation/genetics , Energy Metabolism/genetics , Gene Expression/physiology , Glucose Intolerance/genetics , Insulin Resistance/genetics , Mice , Mice, Inbred C57BL , Obesity/etiology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA-Binding Proteins/metabolism , Signal Transduction/genetics
11.
Annu Rev Physiol ; 79: 209-236, 2017 02 10.
Article in English | MEDLINE | ID: mdl-28192062

ABSTRACT

The hypothalamus is an evolutionarily conserved brain structure that regulates an organism's basic functions, such as homeostasis and reproduction. Several hypothalamic nuclei and neuronal circuits have been the focus of many studies seeking to understand their role in regulating these basic functions. Within the hypothalamic neuronal populations, the arcuate melanocortin system plays a major role in controlling homeostatic functions. The arcuate pro-opiomelanocortin (POMC) neurons in particular have been shown to be critical regulators of metabolism and reproduction because of their projections to several brain areas both in and outside of the hypothalamus, such as autonomic regions of the brain stem and spinal cord. Here, we review and discuss the current understanding of POMC neurons from their development and intracellular regulators to their physiological functions and pathological dysregulation.


Subject(s)
Hypothalamus/metabolism , Hypothalamus/physiology , Neurons/metabolism , Neurons/physiology , Pro-Opiomelanocortin/metabolism , Animals , Homeostasis/physiology , Humans
12.
Cell ; 164(5): 872-83, 2016 Feb 25.
Article in English | MEDLINE | ID: mdl-26919426

ABSTRACT

The ventromedial nucleus of the hypothalamus (VMH) plays a critical role in regulating systemic glucose homeostasis. How neurons in this brain area adapt to the changing metabolic environment to regulate circulating glucose levels is ill defined. Here, we show that glucose load results in mitochondrial fission and reduced reactive oxygen species in VMH neurons mediated by dynamin-related peptide 1 (DRP1) under the control of uncoupling protein 2 (UCP2). Probed by genetic manipulations and chemical-genetic control of VMH neuronal circuitry, we unmasked that this mitochondrial adaptation determines the size of the pool of glucose-excited neurons in the VMH and that this process regulates systemic glucose homeostasis. Thus, our data unmasked a critical cellular biological process controlled by mitochondrial dynamics in VMH regulation of systemic glucose homeostasis.


Subject(s)
Cell Nucleus/metabolism , Glucose/metabolism , Ion Channels/metabolism , Mitochondrial Dynamics , Mitochondrial Proteins/metabolism , Ventromedial Hypothalamic Nucleus/metabolism , Animals , Dynamins/metabolism , Gene Knock-In Techniques , Homeostasis , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/metabolism , Reactive Oxygen Species , Uncoupling Protein 2
13.
Sci Rep ; 5: 17565, 2015 Nov 30.
Article in English | MEDLINE | ID: mdl-26615883

ABSTRACT

High-fat diet (HFD) triggers insulin resistance and diabetes mellitus, but their link remains unclear. Characterization of overt hyperglycemia in insulin receptor mutant (Insr(P1195L/+)) mice exposed to HFD (Insr(P1195L/+)/HFD mice) revealed increased glucose-6-phosphatase (G6pc) expression in liver and increased gluconeogenesis from glycerol. Lipolysis in white adipose tissues (WAT) and lipolysis-induced blood glucose rise were increased in Insr(P1195L/+)/HFD mice, while wild-type WAT transplantation ameliorated the hyperglycemia and the increased G6pc expression. We found that the expressions of genes involved in bile acid (BA) metabolism were altered in Insr(P1195L/+)/HFD liver. Among these, the expression of Cyp7a1, a BA synthesis enzyme, was insulin-dependent and was markedly decreased in Insr(P1195L/+)/HFD liver. Reduced Cyp7a1 expression in Insr(P1195L/+)/HFD liver was rescued by WAT transplantation, and the expression of Cyp7a1 was suppressed by glycerol administration in wild-type liver. These findings suggest that unsuppressed lipolysis in adipocytes elicited by HFD feeding is linked with enhanced gluconeogenesis from glycerol and with alterations in BA physiology in Insr(P1195L/+)/HFD liver.


Subject(s)
Adipocytes/metabolism , Bile Acids and Salts/metabolism , Diet, High-Fat , Gluconeogenesis , Lipolysis , Receptor, Insulin/genetics , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Adipose Tissue, White/transplantation , Animals , Blood Glucose , Body Weight , Disease Models, Animal , Energy Metabolism , Fats/metabolism , Genotype , Glycerol/metabolism , Hyperglycemia/genetics , Hyperglycemia/metabolism , Insulin/metabolism , Insulin Resistance , Liver/metabolism , Mice , Mice, Transgenic , Models, Biological , Mutation , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Pyruvic Acid/metabolism
14.
Endocrinology ; 156(10): 3680-94, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26132918

ABSTRACT

Adipose tissue macrophages (ATMs) play an important role in the inflammatory response in obese animals. How ATMs are regulated in lean animals has remained elusive, however. We now show that the sympathetic nervous system (SNS) is necessary to maintain the abundance of the mRNA for the proinflammatory cytokine TNF-α at a low level in ATMs of lean mice. Intracerebroventricular injection of agouti-related neuropeptide increased the amount of TNF-α mRNA in epididymal (epi) white adipose tissue (WAT), but not in interscapular brown adipose tissue (BAT), through inhibition of sympathetic nerve activity in epiWAT. The surgical denervation and ß-adrenergic antagonist propranolol up-regulated TNF-α mRNA in both epiWAT and BAT in vivo. Signaling by the ß2-adrenergic receptor (AR) and protein kinase A down-regulated TNF-α mRNA in epiWAT explants and suppressed lipopolysaccharide-induced up-regulation of TNF-α mRNA in the stromal vascular fraction of this tissue. ß-AR-deficient (ß-less) mice manifested an increased plasma TNF-α concentration and increased TNF-α mRNA abundance in epiWAT and BAT. TNF-α mRNA abundance was greater in ATMs (CD11b(+) cells of the stromal vascular fraction) from epiWAT or BAT of wild-type mice than in corresponding CD11b(-) cells, and ß2-AR mRNA abundance was greater in ATMs than in CD11b(-) cells of epiWAT. Our results show that the SNS and ß2-AR-protein kinase A pathway maintain an anti-inflammatory state in ATMs of lean mice in vivo, and that the brain melanocortin pathway plays a role in maintaining this state in WAT of lean mice via the SNS.


Subject(s)
Adipose Tissue, White/metabolism , Inflammation Mediators/metabolism , Macrophages/metabolism , Sympathetic Nervous System/metabolism , Tumor Necrosis Factor-alpha/metabolism , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/innervation , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/drug effects , Adipose Tissue, White/innervation , Adrenergic beta-Antagonists/pharmacology , Agouti-Related Protein/administration & dosage , Animals , Cell Line , Epididymis/drug effects , Epididymis/metabolism , Gene Expression/drug effects , Immunoblotting , Injections, Intraventricular , Male , Mice, Inbred C57BL , Mice, Knockout , Peptide Fragments/administration & dosage , Propranolol/pharmacology , Receptors, Adrenergic, beta/genetics , Receptors, Adrenergic, beta/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sympathectomy , Tumor Necrosis Factor-alpha/genetics
15.
Best Pract Res Clin Endocrinol Metab ; 28(5): 757-64, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25256770

ABSTRACT

Uncoupling protein 2 (UCP2) is a mitochondrial anion carrier protein, which uncouples the oxidative phosphorylation from ATP production by dissipating the proton gradient generated across the mitochondrial inner membrane. UCP2 regulates not only mitochondrial ATP production, but also the generation of reactive oxygen species (ROS), considered important second-messenger signals within the cell. The importance of UCP2 was firstly reported in macrophages and pancreatic beta cells. However, several studies have revealed the important role of UCP2 in the Central Nervous System (CNS) in the regulation of homeostatic mechanisms including food intake, energy expenditure, glucose homeostasis and reward behaviors. The mechanisms by which central UCP2 affect these processes seem to be associated with synaptic and mitochondrial plasticity. In this review, we will describe recent findings on central UCP2 and discuss its role in CNS regulation of homeostasis.


Subject(s)
Energy Metabolism/physiology , Hypothalamus/metabolism , Ion Channels/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Animals , Humans , Reactive Oxygen Species/metabolism , Uncoupling Protein 2
16.
J Clin Invest ; 124(9): 4017-27, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25083994

ABSTRACT

Activation of central PPARγ promotes food intake and body weight gain; however, the identity of the neurons that express PPARγ and mediate the effect of this nuclear receptor on energy homeostasis is unknown. Here, we determined that selective ablation of PPARγ in murine proopiomelanocortin (POMC) neurons decreases peroxisome density, elevates reactive oxygen species, and induces leptin sensitivity in these neurons. Furthermore, ablation of PPARγ in POMC neurons preserved the interaction between mitochondria and the endoplasmic reticulum, which is dysregulated by HFD. Compared with control animals, mice lacking PPARγ in POMC neurons had increased energy expenditure and locomotor activity; reduced body weight, fat mass, and food intake; and improved glucose metabolism when exposed to high-fat diet (HFD). Finally, peripheral administration of either a PPARγ activator or inhibitor failed to affect food intake of mice with POMC-specific PPARγ ablation. Taken together, our data indicate that PPARγ mediates cellular, biological, and functional adaptations of POMC neurons to HFD, thereby regulating whole-body energy balance.


Subject(s)
Diet, High-Fat , Leptin/pharmacology , Neurons/metabolism , PPAR gamma/physiology , Pro-Opiomelanocortin/physiology , Anilides/pharmacology , Animals , Energy Metabolism , Female , Glucose/metabolism , Hyperphagia/prevention & control , Insulin Resistance , Male , Mice , Motor Activity , Neurons/drug effects , Reactive Oxygen Species/metabolism , Rosiglitazone , Thiazolidinediones/pharmacology
17.
Proc Natl Acad Sci U S A ; 111(32): 11876-81, 2014 Aug 12.
Article in English | MEDLINE | ID: mdl-25071172

ABSTRACT

Prolyl endopeptidase (PREP) has been implicated in neuronal functions. Here we report that hypothalamic PREP is predominantly expressed in the ventromedial nucleus (VMH), where it regulates glucose-induced neuronal activation. PREP knockdown mice (Prep(gt/gt)) exhibited glucose intolerance, decreased fasting insulin, increased fasting glucagon levels, and reduced glucose-induced insulin secretion compared with wild-type controls. Consistent with this, central infusion of a specific PREP inhibitor, S17092, impaired glucose tolerance and decreased insulin levels in wild-type mice. Arguing further for a central mode of action of PREP, isolated pancreatic islets showed no difference in glucose-induced insulin release between Prep(gt/gt) and wild-type mice. Furthermore, hyperinsulinemic euglycemic clamp studies showed no difference between Prep(gt/gt) and wild-type control mice. Central PREP regulation of insulin and glucagon secretion appears to be mediated by the autonomic nervous system because Prep(gt/gt) mice have elevated sympathetic outflow and norepinephrine levels in the pancreas, and propranolol treatment reversed glucose intolerance in these mice. Finally, re-expression of PREP by bilateral VMH injection of adeno-associated virus-PREP reversed the glucose-intolerant phenotype of the Prep(gt/gt) mice. Taken together, our results unmask a previously unknown player in central regulation of glucose metabolism and pancreatic function.


Subject(s)
Glucagon/metabolism , Hypothalamus/enzymology , Insulin/metabolism , Serine Endopeptidases/metabolism , Animals , Blood Glucose/metabolism , Gene Expression , Gene Knockdown Techniques , Glucose Clamp Technique , Glucose Intolerance/enzymology , Glucose Intolerance/etiology , Hypothalamus/physiology , Indoles/pharmacology , Insulin Secretion , Ion Channels/genetics , Male , Mice , Mice, Transgenic , Mitochondrial Proteins/genetics , Pancreas/metabolism , Phosphorylation , Prolyl Oligopeptidases , Receptor, Insulin/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Serine Endopeptidases/deficiency , Serine Endopeptidases/genetics , Serine Proteinase Inhibitors/pharmacology , Thiazolidines/pharmacology , Uncoupling Protein 1 , Ventromedial Hypothalamic Nucleus/enzymology , Ventromedial Hypothalamic Nucleus/physiology
18.
Adipocyte ; 2(4): 227-36, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-24052898

ABSTRACT

The anti-diabetic effects of Brazilian propolis were examined using ob/ob mice. Although repeated injection of an ethanol extract of Brazilian propolis (100 mg/kg, ip, twice a week for 12 weeks) did not affect body weight gain and food intake of ob/ob mice, blood glucose and plasma cholesterol levels were significantly attenuated. Moreover, the propolis extract partially restored glucose tolerance and insulin resistance, indicating anti-diabetic properties of the extract. The propolis-treated mice exhibited lower weight gain in mesenteric adipose tissue, while weight gains in inguinal and epididymal adipose tissues were not modulated. Flow cytometric and microscopic analyses suggested that the extract promoted accumulation of eosinophils into mesenteric and epididymal adipose tissues. Alternatively, the ratio of M1-like macrophages to M2-like macrophages in mesenteric adipose tissue was reduced by the propolis injection, coincident with the decrement of the number of interleukin-12A(+) cells. Levels of M1 macrophage markers, such as Itgax and Il12b transcripts, were decreased in the vascular stromal fraction of mesenteric adipose tissue, whereas those of pan-macrophage markers Emr1 and Cd68 were not influenced. Microarray and subsequent gene ontology term analyses suggested that propolis attenuated immune activation in mesenteric adipose tissues. Taken together, this indicates that Brazilian propolis improves diabetes in ob/ob mice, presumably through modification of immune cells in mesenteric adipose tissues.

19.
FASEB J ; 27(12): 4940-53, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24005904

ABSTRACT

Macrophages play a critical role in chronic inflammation and metabolic diseases. We identified a longer splice variant of ubiquitin specific protease (USP) 2-69 as a novel molecule that modulates pathways implicated in metabolic disorders. Expression levels of aP2/FABP4 and PAI-1/SERPINE1 genes were increased by 4- and 1.8-fold, respectively, after short hairpin RNA-mediated knockdown (KD) of the USP2 gene, and such expression was alleviated by overexpression of USP2-69 in human myeloid cell lines. Supernatants derived from USP2-KD cells induced IL6 (∼6-fold) and SAA3 (∼15-fold) in 3T3-L1 adipocytes to suggest the anti-inflammatory properties of USP2. In addition, we observed a 30% decrease in the number of macrophages in mesenteric adipose tissue derived from USP2-69 transgenic mice fed a high-fat diet for 14 wk compared with that in their C57BL/6 littermates (P<0.01), which was consistent with a ∼40% decrease in transcription of aP2 and PAI-1. The aP2 locus exhibited elevated chromatin accessibility (>2.1-fold), methylation of histone H3 lysine 4 (>4.5-fold), and acetylation of histone H4 (>2.5-fold) in USP2-KD cells. Transfection of isopeptidase-mutated USP2-69 did not alter chromatin conformation on the aP2 locus in USP2-KD cells. Our results suggest that USP2-69 suppresses meta-inflammatory molecules involved in the development of type-2 diabetes.


Subject(s)
Chromatin Assembly and Disassembly , Endopeptidases/genetics , Macrophages/metabolism , Transcription, Genetic , Ubiquitin-Specific Proteases/genetics , Adipocytes/metabolism , Animals , Cell Line , Chromatin/metabolism , Endopeptidases/metabolism , Epigenesis, Genetic , Histones/metabolism , Humans , Inflammation/genetics , Inflammation/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Mice , Mice, Inbred C57BL , Myeloid Cells/metabolism , Plasminogen Activator Inhibitor 1/genetics , Plasminogen Activator Inhibitor 1/metabolism , Serum Amyloid A Protein/genetics , Serum Amyloid A Protein/metabolism , Transcription Factor AP-2/genetics , Transcription Factor AP-2/metabolism , Ubiquitin Thiolesterase , Ubiquitin-Specific Proteases/metabolism
20.
Diabetes ; 62(7): 2295-307, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23530005

ABSTRACT

Leptin is a key regulator of glucose metabolism in mammals, but the mechanisms of its action have remained elusive. We now show that signaling by extracellular signal-regulated kinase (ERK) and its upstream kinase MEK in the ventromedial hypothalamus (VMH) mediates the leptin-induced increase in glucose utilization as well as its insulin sensitivity in the whole body and in red-type skeletal muscle of mice through activation of the melanocortin receptor (MCR) in the VMH. In contrast, activation of signal transducer and activator of transcription 3 (STAT3), but not the MEK-ERK pathway, in the VMH by leptin enhances the insulin-induced suppression of endogenous glucose production in an MCR-independent manner, with this effect of leptin occurring only in the presence of an increased plasma concentration of insulin. Given that leptin requires 6 h to increase muscle glucose uptake, the transient activation of the MEK-ERK pathway in the VMH by leptin may play a role in the induction of synaptic plasticity in the VMH, resulting in the enhancement of MCR signaling in the nucleus and leading to an increase in insulin sensitivity in red-type muscle.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Glucose/metabolism , Leptin/pharmacology , Muscle, Skeletal/drug effects , Ventromedial Hypothalamic Nucleus/drug effects , Animals , Body Weight/drug effects , Butadienes/pharmacology , Chromones/pharmacology , Enzyme Inhibitors/pharmacology , Insulin/pharmacology , Male , Melanocyte-Stimulating Hormones/pharmacology , Mice , Morpholines/pharmacology , Muscle, Skeletal/metabolism , Nitriles/pharmacology , Phosphorylation/drug effects , Receptors, Corticotropin/antagonists & inhibitors , Signal Transduction/drug effects , Signal Transduction/physiology , Ventromedial Hypothalamic Nucleus/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...