Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Int J Mol Sci ; 23(7)2022 Apr 02.
Article in English | MEDLINE | ID: mdl-35409318

ABSTRACT

A sedentary lifestyle and excessive nutrient intake resulting from the consumption of high-fat and calorie-rich diets are environmental factors contributing to the rapid growth of the current pandemic of type 2 diabetes mellitus (DM2). Fasting hyperglycemia, an established hallmark of DM2, is caused by excessive production of glucose by the liver, resulting in the inability of insulin to suppress endogenous glucose production. To prevent inappropriate elevations of circulating glucose resulting from changes in nutrient availability, mammals rely on complex mechanisms for continuously detecting these changes and to respond to them with metabolic adaptations designed to modulate glucose output. The mediobasal hypothalamus (MBH) is the key center where nutritional cues are detected and appropriate modulatory responses are integrated. However, certain environmental factors may have a negative impact on these adaptive responses. For example, consumption of a diet enriched in saturated fat in rodents resulted in the development of a metabolic defect that attenuated these nutrient sensing mechanisms, rendering the animals prone to developing hyperglycemia. Thus, high-fat feeding leads to a state of "metabolic disability" in which animals' glucoregulatory responses fail. We postulate that the chronic faltering of the hypothalamic glucoregulatory mechanisms contributes to the development of metabolic disease.


Subject(s)
Diabetes Mellitus, Type 2 , Hyperglycemia , Animals , Diabetes Mellitus, Type 2/metabolism , Diet , Glucose/metabolism , Hyperglycemia/metabolism , Hypothalamus/metabolism , Nutrients , Rodentia/metabolism
2.
Cell Tissue Res ; 381(3): 451-460, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32710274

ABSTRACT

The content of gonadotropin-releasing hormone (GnRH), its mRNA, and estrogen receptor alpha (ERα) and beta (ERß) in the hypothalamus varies throughout the estrous cycle. Furthermore, the abundance of these molecules displays asymmetry between the right and left side. In the present study, we investigated the changes in the content of ERα, ERß, kisspeptin, and GnRH by western blot in the left and right anteromedial hypothalamus, at four different times during each stage of the rat estrous cycle. The serum levels of the follicle-stimulating hormone (FSH) and luteinizing hormone (LH) were also measured. ERα and ERß levels changed depending on the stage of the estrous cycle, meanwhile that of kisspeptin was modified according to both the hour of the day and the stage of the cycle. Except in estrus day, ERß was higher in the right hypothalamus, while ERα was similar in both sides. During both proestrus and estrus, the content of kisspeptin and GnRH was higher in the right hypothalamus. The highest levels of FSH and LH occurred at 17:00 h of proestrus. But at estrus, the highest FSH levels were observed at 08:00 h and the lowest at 17:00 h. Thus, the current results show that the content of ERα, ERß, kisspeptin, and GnRH in the anteromedial hypothalamus are regulated as a function of the stage of the estrous cycle and the hour of the day. Furthermore, the content of these proteins is regularly higher in the right anteromedial hypothalamus, regardless of the stage of the cycle or time of the day.


Subject(s)
Estrous Cycle/metabolism , Gonadotropin-Releasing Hormone/metabolism , Hypothalamus/metabolism , Kisspeptins/metabolism , Receptors, Estrogen/metabolism , Animals , Female , Rats
3.
Molecules ; 23(12)2018 12 19.
Article in English | MEDLINE | ID: mdl-30572603

ABSTRACT

Cacalolides are a kind of sesquiterpenoids natural compounds synthesized by Psacalium decompositum (A. Gray) H. Rob. & Brettell or Psacalium peltatum (Kunth) Cass. Antioxidant and hypoglycemic effects have been found for cacalolides such as cacalol, cacalone or maturine, however, their effects on inflammatory processes are still largely unclear. The main aim of this study was to investigate the biological activities of secondary metabolites from P. decompositum and P. peltatum through two approaches: (1) chemoinformatic and toxicoinformatic analysis based on ethnopharmacologic background; and (2) the evaluation of their potential anti-inflammatory/anti-allergic effects in bone marrow-derived mast cells by IgE/antigen complexes. The bioinformatics properties of the compounds: cacalol; cacalone; cacalol acetate and maturin acetate were evaluated through Osiris DataWarrior software and Molinspiration and PROTOX server. In vitro studies were performed to test the ability of these four compounds to inhibit antigen-dependent degranulation and intracellular calcium mobilization, as well as the production of reactive oxygen species in bone marrow-derived mast cells. Our findings showed that cacalol displayed better bioinformatics properties, also exhibited a potent inhibitory activity on IgE/antigen-dependent degranulation and significantly reduced the intracellular calcium mobilization on mast cells. These data suggested that cacalol could reduce the negative effects of the mast cell-dependent inflammatory process.


Subject(s)
Mast Cells/metabolism , Psacalium/chemistry , Receptors, IgE/metabolism , Animals , Calcium/metabolism , Calcium Channels/metabolism , Inflammation/metabolism , Male , Mast Cells/drug effects , Mice , Mice, Inbred C57BL , Reactive Oxygen Species/metabolism , Sesquiterpenes/metabolism , Sesquiterpenes/pharmacology
4.
Adv Nutr ; 7(4): 790S-7S, 2016 07.
Article in English | MEDLINE | ID: mdl-27422516

ABSTRACT

A pandemic of diabetes and obesity has been developing worldwide in close association with excessive nutrient intake and a sedentary lifestyle. Variations in the protein content of the diet have a direct impact on glucose homeostasis because amino acids (AAs) are powerful modulators of insulin action. In this work we review our recent findings on how elevations in the concentration of the circulating AAs leucine and proline activate a metabolic mechanism located in the mediobasal hypothalamus of the brain that sends a signal to the liver via the vagus nerve, which curtails glucose output. This neurogenic signal is strictly dependent on the metabolism of leucine and proline to acetyl-coenzyme A (CoA) and the subsequent production of malonyl-CoA; the signal also requires functional neuronal ATP-sensitive potassium channels. The liver then responds by lowering the rate of gluconeogenesis and glycogenolysis, ultimately leading to a net decrease in glucose production and in concentrations of circulating glucose. Furthermore, we review here how our work with proline suggests a new role of astrocytes in the central regulation of glycemia. Last, we outline how factors such as the consumption of fat-rich diets can interfere with glucoregulatory mechanisms and, in the long term, may contribute to the development of hyperglycemia, a hallmark of type 2 diabetes.


Subject(s)
Amino Acids/blood , Glucose/metabolism , Hypothalamus/physiology , Liver/metabolism , Animals , Astrocytes/physiology , Blood Glucose/analysis , Dietary Fats/administration & dosage , Gluconeogenesis/physiology , Glycogenolysis/physiology , Homeostasis/physiology , Humans , Leucine , Liver/innervation , Neurons/physiology , Proline/blood , Vagus Nerve/physiology
5.
Nutrients ; 8(2): 79, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26891318

ABSTRACT

Leucine has been shown to acutely inhibit hepatic glucose production in rodents by a mechanism requiring its metabolism to acetyl-CoA in the mediobasal hypothalamus (MBH). In the early stages, all branched-chain amino acids (BCAA) are metabolized by a shared set of enzymes to produce a ketoacid, which is later metabolized to acetyl-CoA. Consequently, isoleucine and valine may also modulate glucose metabolism. To examine this possibility we performed intrahypothalamic infusions of isoleucine or valine in rats and assessed whole body glucose kinetics under basal conditions and during euglycemic pancreatic clamps. Furthermore, because high fat diet (HFD) consumption is known to interfere with central glucoregulation, we also asked whether the action of BCAAs was affected by HFD. We fed rats a lard-rich diet for a short interval and examined their response to central leucine. The results showed that both isoleucine and valine individually lowered blood glucose by decreasing liver glucose production. Furthermore, the action of the BCAA leucine was markedly attenuated by HFD feeding. We conclude that all three BCAAs centrally modulate glucose metabolism in the liver and that their action is disrupted by HFD-induced insulin resistance.


Subject(s)
Blood Glucose/metabolism , Diet , Dietary Proteins/chemistry , Gluconeogenesis/drug effects , Isoleucine/pharmacology , Liver/drug effects , Valine/pharmacology , Animals , Diet, High-Fat , Dietary Fats/pharmacology , Dietary Proteins/pharmacology , Hypothalamus/metabolism , Insulin Resistance , Liver/metabolism , Male , Rats, Sprague-Dawley
6.
Metabolism ; 64(2): 315-22, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25467844

ABSTRACT

OBJECTIVE: Free fatty acids (FFAs) cause insulin resistance and are often elevated in obesity. Chronic ingestion of diets rich in saturated fat induces more insulin resistance than diets rich in unsaturated fat, however, it remains unclear whether different FFAs cause distinct levels of insulin resistance in the short-term, which is relevant to the feeding and fasting cycle. Protein kinase C (PKC)-δ is implicated in hepatic insulin resistance. Therefore, we investigated the effects of short-term elevation of fatty acids with different degrees of unsaturation on hepatic insulin action and liver PKC-δ membrane translocation, a marker of activation. MATERIALS/METHODS: Triglyceride emulsions of Soybean Oil+Heparin (polyunsaturated (POLY)), Olive Oil+Heparin (monounsaturated (MONO)), Lard Oil+Heparin (saturated (SATU)), or saline (SAL) were infused intravenously for 7h to elevate plasma FFA concentrations ~3-4 fold in rats. During the last 2h of infusion, a hyperinsulinemic-euglycemic clamp with tritiated glucose methodology was performed to examine hepatic and peripheral insulin sensitivity. RESULTS: Surprisingly, SATU, MONO, and POLY impaired peripheral insulin sensitivity (glucose utilization divided by insulin) to a similar extent. Furthermore, all lipids induced a similar degree of hepatic insulin resistance compared to SAL. Although there were changes in hepatic content of lipid metabolites, there were no significant differences in liver PKC-δ membrane translocation across fat groups. CONCLUSIONS: In summary, in the short-term, FFAs with different degrees of unsaturation impair peripheral insulin sensitivity and induce hepatic insulin resistance as well as hepatic PKC-δ translocation to the same extent.


Subject(s)
Dietary Fats, Unsaturated/adverse effects , Dietary Fats/adverse effects , Fatty Acids, Nonesterified/blood , Insulin Resistance , Liver/metabolism , Up-Regulation , Animals , Cell Membrane/enzymology , Dietary Fats/administration & dosage , Dietary Fats/analysis , Dietary Fats/metabolism , Dietary Fats, Unsaturated/administration & dosage , Dietary Fats, Unsaturated/analysis , Dietary Fats, Unsaturated/metabolism , Enzyme Activation , Fat Emulsions, Intravenous , Fatty Acids/adverse effects , Fatty Acids/analysis , Fatty Acids/blood , Fatty Acids/metabolism , Fatty Acids, Monounsaturated/adverse effects , Fatty Acids, Monounsaturated/analysis , Fatty Acids, Monounsaturated/blood , Fatty Acids, Monounsaturated/metabolism , Fatty Acids, Nonesterified/metabolism , Fatty Acids, Unsaturated/adverse effects , Fatty Acids, Unsaturated/analysis , Fatty Acids, Unsaturated/blood , Fatty Acids, Unsaturated/metabolism , Female , Glucose Clamp Technique , Liver/enzymology , Olive Oil , Plant Oils/administration & dosage , Plant Oils/adverse effects , Plant Oils/chemistry , Plant Oils/metabolism , Protein Kinase C-delta/chemistry , Protein Kinase C-delta/metabolism , Protein Transport , Rats, Wistar , Soybean Oil/administration & dosage , Soybean Oil/adverse effects , Soybean Oil/chemistry , Soybean Oil/metabolism
7.
Diabetes ; 62(10): 3373-83, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23835335

ABSTRACT

Successful development of antiobesity agents requires detailed knowledge of neural pathways controlling body weight, eating behavior, and peripheral metabolism. Genetic ablation of FoxO1 in selected hypothalamic neurons decreases food intake, increases energy expenditure, and improves glucose homeostasis, highlighting the role of this gene in insulin and leptin signaling. However, little is known about potential effects of FoxO1 in other neurons. To address this question, we executed a broad-based neuronal ablation of FoxO1 using Synapsin promoter-driven Cre to delete floxed Foxo1 alleles. Lineage-tracing experiments showed that NPY/AgRP and POMC neurons were minimally affected by the knockout. Nonetheless, Syn-Cre-Foxo1 knockouts demonstrated a catabolic energy homeostatic phenotype with a blunted refeeding response, increased sensitivity to leptin and amino acid signaling, and increased locomotor activity, likely attributable to increased melanocortinergic tone. We confirmed these data in mice lacking the three Foxo genes. The effects on locomotor activity could be reversed by direct delivery of constitutively active FoxO1 to the mediobasal hypothalamus, but not to the suprachiasmatic nucleus. The data reveal that the integrative function of FoxO1 extends beyond the arcuate nucleus, suggesting that central nervous system inhibition of FoxO1 function can be leveraged to promote hormone sensitivity and prevent a positive energy balance.


Subject(s)
Eating , Forkhead Transcription Factors/antagonists & inhibitors , Hypothalamus/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Locomotion/drug effects , Neurons/drug effects , Animals , Anti-Obesity Agents/pharmacology , Drug Design , Eating/drug effects , Energy Metabolism/drug effects , Forkhead Box Protein O1 , Forkhead Transcription Factors/deficiency , Gene Expression Regulation/drug effects , Genotype , Hypothalamus/drug effects , Immunohistochemistry , Male , Mice , Mice, Knockout , Signal Transduction/drug effects
8.
Diabetes ; 62(4): 1152-8, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23274895

ABSTRACT

The metabolism of lactate to pyruvate in the mediobasal hypothalamus (MBH) regulates hepatic glucose production. Because astrocytes and neurons are functionally linked by metabolic coupling through lactate transfer via the astrocyte-neuron lactate shuttle (ANLS), we reasoned that astrocytes might be involved in the hypothalamic regulation of glucose metabolism. To examine this possibility, we used the gluconeogenic amino acid proline, which is metabolized to pyruvate in astrocytes. Our results showed that increasing the availability of proline in rats either centrally (MBH) or systemically acutely lowered blood glucose. Pancreatic clamp studies revealed that this hypoglycemic effect was due to a decrease of hepatic glucose production secondary to an inhibition of glycogenolysis, gluconeogenesis, and glucose-6-phosphatase flux. The effect of proline was mimicked by glutamate, an intermediary of proline metabolism. Interestingly, proline's action was markedly blunted by pharmacological inhibition of hypothalamic lactate dehydrogenase (LDH) suggesting that metabolic flux through LDH was required. Furthermore, short hairpin RNA-mediated knockdown of hypothalamic LDH-A, an astrocytic component of the ANLS, also blunted the glucoregulatory action of proline. Thus our studies suggest not only a new role for proline in the regulation of hepatic glucose production but also indicate that hypothalamic astrocytes are involved in the regulatory mechanism as well.


Subject(s)
Astrocytes/metabolism , Glucose/metabolism , Hypothalamus/cytology , Proline/metabolism , Animals , Blood Glucose , Gene Expression Regulation, Enzymologic/drug effects , Glucose/administration & dosage , Glucose/pharmacology , Glucose-6-Phosphatase/genetics , Glucose-6-Phosphatase/metabolism , Insulin/pharmacology , Liver/drug effects , Liver/enzymology , Male , Phosphoenolpyruvate Carboxykinase (GTP)/genetics , Phosphoenolpyruvate Carboxykinase (GTP)/metabolism , Rats , Rats, Sprague-Dawley , Somatostatin/pharmacology
9.
Cell ; 149(6): 1314-26, 2012 Jun 08.
Article in English | MEDLINE | ID: mdl-22682251

ABSTRACT

Hypothalamic neurons expressing Agouti-related peptide (AgRP) are critical for initiating food intake, but druggable biochemical pathways that control this response remain elusive. Thus, genetic ablation of insulin or leptin signaling in AgRP neurons is predicted to reduce satiety but fails to do so. FoxO1 is a shared mediator of both pathways, and its inhibition is required to induce satiety. Accordingly, FoxO1 ablation in AgRP neurons of mice results in reduced food intake, leanness, improved glucose homeostasis, and increased sensitivity to insulin and leptin. Expression profiling of flow-sorted FoxO1-deficient AgRP neurons identifies G-protein-coupled receptor Gpr17 as a FoxO1 target whose expression is regulated by nutritional status. Intracerebroventricular injection of Gpr17 agonists induces food intake, whereas Gpr17 antagonist cangrelor curtails it. These effects are absent in Agrp-Foxo1 knockouts, suggesting that pharmacological modulation of this pathway has therapeutic potential to treat obesity.


Subject(s)
Agouti-Related Protein/metabolism , Eating , Forkhead Transcription Factors/metabolism , Hypothalamus/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Receptors, G-Protein-Coupled/metabolism , Agouti-Related Protein/genetics , Animals , Energy Metabolism , Forkhead Box Protein O1 , Forkhead Transcription Factors/genetics , Glucose/metabolism , Leptin/metabolism , Mice
10.
Diabetes ; 61(1): 85-93, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22187376

ABSTRACT

Amino acids profoundly affect insulin action and glucose metabolism in mammals. Here, we investigated the role of the mediobasal hypothalamus (MBH), a key center involved in nutrient-dependent metabolic regulation. Specifically, we tested the novel hypothesis that the metabolism of leucine within the MBH couples the central sensing of leucine with the control of glucose production by the liver. We performed either central (MBH) or systemic infusions of leucine in Sprague-Dawley male rats during basal pancreatic insulin clamps in combination with various pharmacological and molecular interventions designed to modulate leucine metabolism in the MBH. We also examined the role of hypothalamic ATP-sensitive K(+) channels (K(ATP) channels) in the effects of leucine. Enhancing the metabolism of leucine acutely in the MBH lowered blood glucose through a biochemical network that was insensitive to rapamycin but strictly dependent on the hypothalamic metabolism of leucine to α-ketoisocaproic acid and, further, insensitive to acetyl- and malonyl-CoA. Functional K(ATP) channels were also required. Importantly, molecular attenuation of this central sensing mechanism in rats conferred susceptibility to developing hyperglycemia. We postulate that the metabolic sensing of leucine in the MBH is a previously unrecognized mechanism for the regulation of hepatic glucose production required to maintain glucose homeostasis.


Subject(s)
Glucose/metabolism , Hypothalamus/metabolism , Leucine/metabolism , Liver/metabolism , Animals , Cells, Cultured , Gluconeogenesis/drug effects , Gluconeogenesis/physiology , Humans , Leucine/pharmacology , Leucine/physiology , Liver/drug effects , Male , Metabolic Networks and Pathways/drug effects , Metabolic Networks and Pathways/physiology , Mice , Mice, Knockout , Models, Biological , Rats , Rats, Sprague-Dawley
11.
J Clin Invest ; 121(12): 4916-20, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22056385

ABSTRACT

Increased endogenous glucose production (EGP) is a hallmark of type 2 diabetes mellitus. While there is evidence for central regulation of EGP by activation of hypothalamic ATP-sensitive potassium (K(ATP)) channels in rodents, whether these central pathways contribute to regulation of EGP in humans remains to be determined. Here we present evidence for central nervous system regulation of EGP in humans that is consistent with complementary rodent studies. Oral administration of the K(ATP) channel activator diazoxide under fixed hormonal conditions substantially decreased EGP in nondiabetic humans and Sprague Dawley rats. In rats, comparable doses of oral diazoxide attained appreciable concentrations in the cerebrospinal fluid, and the effects of oral diazoxide were abolished by i.c.v. administration of the K(ATP) channel blocker glibenclamide. These results suggest that activation of hypothalamic K(ATP) channels may be an important regulator of EGP in humans and that this pathway could be a target for treatment of hyperglycemia in type 2 diabetes mellitus.


Subject(s)
Diazoxide/pharmacology , Gluconeogenesis/drug effects , Hypothalamus/metabolism , Potassium Channels/physiology , Adult , Animals , Blood Glucose/analysis , Blood-Brain Barrier , Calcium Channel Blockers/administration & dosage , Calcium Channel Blockers/pharmacology , Diazoxide/administration & dosage , Diazoxide/cerebrospinal fluid , Diazoxide/pharmacokinetics , Diazoxide/therapeutic use , Double-Blind Method , Enzyme Induction/drug effects , Female , Gluconeogenesis/physiology , Glucose Clamp Technique , Glucose-6-Phosphatase/antagonists & inhibitors , Glucose-6-Phosphatase/biosynthesis , Glucose-6-Phosphatase/genetics , Glyburide/administration & dosage , Glyburide/pharmacology , Humans , Hypothalamus/physiopathology , Injections, Intraventricular , Insulin/blood , Ion Channel Gating/drug effects , Liver/drug effects , Liver/enzymology , Male , Phosphorylation/drug effects , Potassium Channels/agonists , Protein Processing, Post-Translational/drug effects , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/biosynthesis , Protein Serine-Threonine Kinases/genetics , Rats , Rats, Sprague-Dawley , STAT3 Transcription Factor/metabolism
12.
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
13.
Diabetes ; 57(8): 2061-5, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18511848

ABSTRACT

OBJECTIVE: A selective rise in hypothalamic lipid metabolism and the subsequent activation of SUR1/Kir6.2 ATP-sensitive K(+) (K(ATP)) channels inhibit hepatic glucose production. The mechanisms that link the ability of hypothalamic lipid metabolism to the activation of K(ATP) channels remain unknown. RESEARCH DESIGN AND METHODS: To examine whether hypothalamic protein kinase C (PKC) mediates the ability of central nervous system lipids to activate K(ATP) channels and regulate glucose production in normal rodents, we first activated hypothalamic PKC in the absence or presence of K(ATP) channel inhibition. We then inhibited hypothalamic PKC in the presence of lipids. Tracer-dilution methodology in combination with the pancreatic clamp technique was used to assess the effect of hypothalamic administrations on glucose metabolism in vivo. RESULTS: We first reported that direct activation of hypothalamic PKC via direct hypothalamic delivery of PKC activator 1-oleoyl-2-acetyl-sn-glycerol (OAG) suppressed glucose production. Coadministration of hypothalamic PKC-delta inhibitor rottlerin with OAG prevented the ability of OAG to activate PKC-delta and lower glucose production. Furthermore, hypothalamic dominant-negative Kir6.2 expression or the delivery of the K(ATP) channel blocker glibenclamide abolished the glucose production-lowering effects of OAG. Finally, inhibition of hypothalamic PKC eliminated the ability of lipids to lower glucose production. CONCLUSIONS: These studies indicate that hypothalamic PKC activation is sufficient and necessary for lowering glucose production.


Subject(s)
Glucose/biosynthesis , Hypothalamus/metabolism , Protein Kinase C/metabolism , Acetophenones/administration & dosage , Acetophenones/pharmacology , Animals , Benzopyrans/administration & dosage , Benzopyrans/pharmacology , Diglycerides/administration & dosage , Diglycerides/pharmacology , Enzyme Activation/drug effects , Glyburide/administration & dosage , Glyburide/pharmacology , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/pharmacology , Hypothalamus/drug effects , Hypothalamus/enzymology , KATP Channels/antagonists & inhibitors , Male , Protein Kinase C/antagonists & inhibitors , Protein Kinase C-delta/antagonists & inhibitors , Protein Kinase C-delta/metabolism , Protein Transport/drug effects , Rats , Rats, Sprague-Dawley
14.
J Clin Invest ; 116(4): 1081-91, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16528412

ABSTRACT

Short-term overfeeding blunts the central effects of fatty acids on food intake and glucose production. This acquired defect in nutrient sensing could contribute to the rapid onset of hyperphagia and insulin resistance in this model. Here we examined whether central inhibition of lipid oxidation is sufficient to restore the hypothalamic levels of long-chain fatty acyl-CoAs (LCFA-CoAs) and to normalize food intake and glucose homeostasis in overfed rats. To this end, we targeted the liver isoform of carnitine palmitoyltransferase-1 (encoded by the CPT1A gene) by infusing either a sequence-specific ribozyme against CPT1A or an isoform-selective inhibitor of CPT1A activity in the third cerebral ventricle or in the mediobasal hypothalamus (MBH). Inhibition of CPT1A activity normalized the hypothalamic levels of LCFA-CoAs and markedly inhibited feeding behavior and hepatic glucose fluxes in overfed rats. Thus central inhibition of lipid oxidation is sufficient to restore hypothalamic lipid sensing as well as glucose and energy homeostasis in this model and may be an effective approach to the treatment of diet-induced obesity and insulin resistance.


Subject(s)
Energy Metabolism , Glucose/biosynthesis , Hypothalamus/physiology , Lipid Metabolism , Animals , Carnitine O-Palmitoyltransferase/antagonists & inhibitors , Carnitine O-Palmitoyltransferase/metabolism , Energy Metabolism/physiology , Homeostasis , Hyperphagia/metabolism , Hypothalamus/metabolism , Lipid Metabolism/physiology , Liver/cytology , Liver/metabolism , Male , Models, Biological , Oxidation-Reduction , Rats , Rats, Sprague-Dawley
15.
Diabetes ; 54(11): 3182-9, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16249443

ABSTRACT

Voluntary overfeeding rapidly induces resistance to the effects of systemic insulin and leptin on liver glucose metabolism. To examine whether central administration of recombinant leptin can restore leptin and insulin action on liver glucose fluxes, we infused leptin in the third cerebral ventricle of conscious overfed rats during pancreatic-insulin clamp studies. The effect of leptin on the phosphorylation of the signal transducer and activator of transcription-3 in the arcuate nuclei of the hypothalamus was similar in animals fed a regular diet or a high-fat diet for 3 days. The infusion of leptin in the third cerebral ventricle markedly inhibited glucose production in rats fed a high-fat diet mainly by decreasing glycogenolysis. The inhibition of glycogenolysis was sufficient to normalize glucose production and was accompanied by leptin-induced decreases in the hepatic expression of glucose-6-phosphatase and phosphoenolpyruvate carboxykinase. Thus central administration of leptin rescues the hepatic insulin resistance induced by short-term hyperphagia.


Subject(s)
Diet , Insulin Resistance , Leptin/administration & dosage , Leptin/pharmacology , Liver/drug effects , Liver/metabolism , Animals , Glucose/metabolism , Hypothalamus/metabolism , Insulin/metabolism , Insulin/pharmacology , Male , Rats , Rats, Sprague-Dawley , STAT3 Transcription Factor/metabolism
16.
Science ; 309(5736): 943-7, 2005 Aug 05.
Article in English | MEDLINE | ID: mdl-16081739

ABSTRACT

The brain keenly depends on glucose for energy, and mammalians have redundant systems to control glucose production. An increase in circulating glucose inhibits glucose production in the liver, but this negative feedback is impaired in type 2 diabetes. Here we report that a primary increase in hypothalamic glucose levels lowers blood glucose through inhibition of glucose production in rats. The effect of glucose requires its conversion to lactate followed by stimulation of pyruvate metabolism, which leads to activation of adenosine triphosphate (ATP)-sensitive potassium channels. Thus, interventions designed to enhance the hypothalamic sensing of glucose may improve glucose homeostasis in diabetes.


Subject(s)
Blood Glucose/metabolism , Glucose/metabolism , Hypothalamus/metabolism , Liver/metabolism , Pyruvates/metabolism , Animals , Astrocytes/metabolism , Citric Acid Cycle , Feedback, Physiological , Glucose/administration & dosage , Glucose-6-Phosphatase/metabolism , Injections, Intraventricular , Lactic Acid/metabolism , Male , Neurons/metabolism , Potassium Channels/metabolism , Rats , Rats, Sprague-Dawley
17.
Nature ; 434(7036): 1026-31, 2005 Apr 21.
Article in English | MEDLINE | ID: mdl-15846348

ABSTRACT

Obesity is the driving force behind the worldwide increase in the prevalence of type 2 diabetes mellitus. Hyperglycaemia is a hallmark of diabetes and is largely due to increased hepatic gluconeogenesis. The medial hypothalamus is a major integrator of nutritional and hormonal signals, which play pivotal roles not only in the regulation of energy balance but also in the modulation of liver glucose output. Bidirectional changes in hypothalamic insulin signalling therefore result in parallel changes in both energy balance and glucose metabolism. Here we show that activation of ATP-sensitive potassium (K(ATP)) channels in the mediobasal hypothalamus is sufficient to lower blood glucose levels through inhibition of hepatic gluconeogenesis. Finally, the infusion of a K(ATP) blocker within the mediobasal hypothalamus, or the surgical resection of the hepatic branch of the vagus nerve, negates the effects of central insulin and halves the effects of systemic insulin on hepatic glucose production. Consistent with these results, mice lacking the SUR1 subunit of the K(ATP) channel are resistant to the inhibitory action of insulin on gluconeogenesis. These findings suggest that activation of hypothalamic K(ATP) channels normally restrains hepatic gluconeogenesis, and that any alteration within this central nervous system/liver circuit can contribute to diabetic hyperglycaemia.


Subject(s)
Adenosine Triphosphate/metabolism , Gluconeogenesis , Glucose/biosynthesis , Hypothalamus/metabolism , Liver/metabolism , Potassium Channels/metabolism , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Animals , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/physiopathology , Glucose/metabolism , Hyperinsulinism/metabolism , Hyperinsulinism/physiopathology , Insulin/metabolism , Liver/innervation , Male , Mice , Multidrug Resistance-Associated Proteins/deficiency , Multidrug Resistance-Associated Proteins/genetics , Multidrug Resistance-Associated Proteins/metabolism , Potassium Channels/chemistry , Potassium Channels/genetics , Potassium Channels, Inwardly Rectifying/genetics , Potassium Channels, Inwardly Rectifying/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Drug , Sulfonylurea Receptors , Vagus Nerve/physiology
18.
Nat Med ; 11(3): 320-7, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15735652

ABSTRACT

Increased glucose production is a hallmark of type 2 diabetes and alterations in lipid metabolism have a causative role in its pathophysiology. Here we postulate that physiological increments in plasma fatty acids can be sensed within the hypothalamus and that this sensing is required to balance their direct stimulatory action on hepatic gluconeogenesis. In the presence of physiologically-relevant increases in the levels of plasma fatty acids, negating their central action on hepatic glucose fluxes through (i) inhibition of the hypothalamic esterification of fatty acids, (ii) genetic deletion (Sur1-deficient mice) of hypothalamic K(ATP) channels or pharmacological blockade (K(ATP) blocker) of their activation by fatty acids, or (iii) surgical resection of the hepatic branch of the vagus nerve led to a marked increase in liver glucose production. These findings indicate that a physiological elevation in circulating lipids can be sensed within the hypothalamus and that a defect in hypothalamic lipid sensing disrupts glucose homeostasis.


Subject(s)
Diabetes Mellitus, Type 2/physiopathology , Fatty Acids, Nonesterified/blood , Glucose/metabolism , Hypothalamus/physiology , Liver/metabolism , Animals , Coenzyme A Ligases/antagonists & inhibitors , Dietary Fats/administration & dosage , Fat Emulsions, Intravenous/administration & dosage , Fat Emulsions, Intravenous/pharmacology , Glucose-6-Phosphatase/antagonists & inhibitors , Glucose-6-Phosphatase/metabolism , Glyburide/pharmacology , Homeostasis/physiology , Injections, Intraventricular , Liver/drug effects , Male , Potassium Channel Blockers/pharmacology , Potassium Channels, Inwardly Rectifying/physiology , Rats , Rats, Sprague-Dawley , Triazenes/pharmacology , Vagotomy
SELECTION OF CITATIONS
SEARCH DETAIL