Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Endocrinology ; 162(7)2021 07 01.
Article in English | MEDLINE | ID: mdl-33765118

ABSTRACT

Circulating branched chain amino acid (BCAA) levels are elevated in obese humans and genetically obese rodents. However, the relationship of BCAAs to insulin resistance in diet-induced obese mice, a commonly used model to study glucose homeostasis, is still ill-defined. Here we examined how high-fat high-sucrose (HFHS) or high-fat diet (HFD) feeding, with or without BCAA supplementation in water, alters the metabolome in serum/plasma and tissues in mice and whether raising circulating BCAA levels worsens insulin resistance and glucose intolerance. Neither HFHS nor HFD feeding raised circulating BCAA levels in insulin-resistant diet-induced obese mice. BCAA supplementation raised circulating BCAA and branched-chain α-keto acid levels and C5-OH/C3-DC acylcarnitines (AC) in muscle from mice fed an HFHS diet or HFD, but did not worsen insulin resistance. A set of short- and long-chain acyl CoAs were elevated by diet alone in muscle, liver, and white adipose tissue (WAT), but not increased further by BCAA supplementation. HFD feeding reduced valine and leucine oxidation in WAT but not in muscle. BCAA supplementation markedly increased valine oxidation in muscle from HFD-fed mice, while leucine oxidation was unaffected by diet or BCAA treatment. Here we establish an extensive metabolome database showing tissue-specific changes in mice on 2 different HFDs, with or without BCAA supplementation. We conclude that mildly elevating circulating BCAAs and a subset of ACs by BCAA supplementation does not worsen insulin resistance or glucose tolerance in mice. This work highlights major differences in the effects of BCAAs on glucose homeostasis in diet-induced obese mice versus data reported in obese rats and in humans.


Subject(s)
Amino Acids, Branched-Chain/administration & dosage , Blood Glucose/metabolism , Diet/adverse effects , Insulin Resistance/physiology , Metabolomics , Obesity/metabolism , Amino Acids, Branched-Chain/blood , Amino Acids, Branched-Chain/metabolism , Animals , Diet, High-Fat , Dietary Sucrose/administration & dosage , Dietary Supplements , Female , Glucose Intolerance/blood , Homeostasis/drug effects , Lipid Metabolism/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Muscle, Skeletal/metabolism , Obesity/etiology , Oxidation-Reduction
2.
Diabetes ; 66(3): 587-597, 2017 03.
Article in English | MEDLINE | ID: mdl-27797912

ABSTRACT

GLUT4 in muscle and adipose tissue is important in maintaining glucose homeostasis. However, the role of insulin-responsive GLUT4 in the central nervous system has not been well characterized. To assess its importance, a selective knockout of brain GLUT4 (BG4KO) was generated by crossing Nestin-Cre mice with GLUT4-floxed mice. BG4KO mice had a 99% reduction in GLUT4 protein expression throughout the brain. Despite normal feeding and fasting glycemia, BG4KO mice were glucose intolerant, demonstrated hepatic insulin resistance, and had reduced glucose uptake in the brain. In response to hypoglycemia, BG4KO mice had impaired glucose sensing, noted by impaired epinephrine and glucagon responses and impaired c-fos activation in the hypothalamic paraventricular nucleus. Moreover, in vitro glucose sensing of glucose-inhibitory neurons from the ventromedial hypothalamus was impaired in BG4KO mice. In summary, BG4KO mice are glucose intolerant, insulin resistant, and have impaired glucose sensing, indicating a critical role for brain GLUT4 in sensing and responding to changes in blood glucose.


Subject(s)
Blood Glucose/metabolism , Brain/metabolism , Glucose Intolerance/genetics , Hypoglycemia/genetics , Insulin Resistance/genetics , Animals , Blotting, Western , Diet, High-Fat , Epinephrine/metabolism , Glucagon/metabolism , Glucose/metabolism , Glucose Clamp Technique , Glucose Tolerance Test , Glucose Transporter Type 4 , Homeostasis/genetics , Hypothalamus/cytology , Hypothalamus/metabolism , In Vitro Techniques , Indinavir/pharmacology , Male , Mice , Mice, Knockout , Neurons/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Sprague-Dawley
3.
Cell Metab ; 18(3): 304-6, 2013 Sep 03.
Article in English | MEDLINE | ID: mdl-24011066

ABSTRACT

Leptin mimics many of the antidiabetic actions of insulin in insulin-deficient diabetes, but the mechanism is controversial. Fujikawa et al. (2013) reveal that leptin receptors in γ-aminobutyric acid (GABA)-ergic and pro-opiomelanocortin (POMC) neurons in the central nervous system are sufficient to mediate the lifesaving and antidiabetic actions of leptin in insulin-deficient mice.


Subject(s)
Hypothalamus/drug effects , Insulin/metabolism , Leptin/pharmacology , Neurons/drug effects , Animals
4.
Cell Metab ; 16(1): 104-12, 2012 Jul 03.
Article in English | MEDLINE | ID: mdl-22727014

ABSTRACT

The PI3K-AKT, mTOR-p70S6 kinase and AMPK pathways play distinct and critical roles in metabolic regulation. Each pathway is necessary for leptin's anorexigenic effects in the hypothalamus. Here we show that these pathways converge in an integrated phosphorylation cascade to mediate leptin action in the hypothalamus. We identify serine(491) on α2AMPK as the site of convergence and show that p70S6 kinase forms a complex with α2AMPK, resulting in phosphorylation on serine(491). Blocking α2AMPK-serine(491) phosphorylation increases hypothalamic AMPK activity, food intake, and body weight. Serine(491) phosphorylation is necessary for leptin's effects on hypothalamic α2AMPK activity, neuropeptide expression, food intake, and body weight. These results identify an inhibitory AMPK kinase, p70S6 kinase, and demonstrate that AMPK is a substrate for mTOR-p70S6 kinase. This discovery has broad biologic implications since mTOR-p70S6 kinase and AMPK have multiple, fundamental and generally opposing cellular effects that regulate metabolism, cell growth, and development.


Subject(s)
Adenylate Kinase/metabolism , Eating , Leptin/physiology , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Serine/metabolism , Agouti-Related Protein/metabolism , Animals , Body Weight , Cell Line , Hypothalamus/enzymology , Hypothalamus/metabolism , Hypothalamus/physiology , Male , Mice , Mice, Knockout , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Pro-Opiomelanocortin/metabolism , Protein Processing, Post-Translational , Protein Subunits/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/genetics , Signal Transduction
5.
PLoS One ; 7(5): e36335, 2012.
Article in English | MEDLINE | ID: mdl-22590531

ABSTRACT

Hypoglycemia and neuroglucopenia stimulate AMP-activated protein kinase (AMPK) activity in the hypothalamus and this plays an important role in the counterregulatory responses, i.e. increased food intake and secretion of glucagon, corticosterone and catecholamines. Several upstream kinases that activate AMPK have been identified including Ca(2+)/Calmodulin-dependent protein kinase kinase (CaMKK), which is highly expressed in neurons. However, the involvement of CaMKK in neuroglucopenia-induced activation of AMPK in the hypothalamus has not been tested. To determine whether neuroglucopenia-induced AMPK activation is mediated by CaMKK, we tested whether STO-609 (STO), a CaMKK inhibitor, would block the effects of 2-deoxy-D-glucose (2DG)-induced neuroglucopenia both ex vivo on brain sections and in vivo. Preincubation of rat brain sections with STO blocked KCl-induced α1 and α2-AMPK activation but did not affect AMPK activation by 2DG in the medio-basal hypothalamus. To confirm these findings in vivo, STO was pre-administrated intracerebroventricularly (ICV) in rats 30 min before 2DG ICV injection (40 µmol) to induce neuroglucopenia. 2DG-induced neuroglucopenia lead to a significant increase in glycemia and food intake compared to saline-injected control rats. ICV pre-administration of STO (5, 20 or 50 nmol) did not affect 2DG-induced hyperglycemia and food intake. Importantly, activation of hypothalamic α1 and α2-AMPK by 2DG was not affected by ICV pre-administration of STO. In conclusion, activation of hypothalamic AMPK by 2DG-induced neuroglucopenia is not mediated by CaMKK.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Kinase/metabolism , Hypoglycemia/enzymology , Hypothalamus/enzymology , Nerve Tissue Proteins/metabolism , AMP-Activated Protein Kinases/antagonists & inhibitors , Animals , Antimetabolites/adverse effects , Antimetabolites/pharmacology , Benzimidazoles/pharmacology , Deoxyglucose/adverse effects , Deoxyglucose/pharmacology , Hypoglycemia/chemically induced , Male , Naphthalimides/pharmacology , Nerve Tissue Proteins/antagonists & inhibitors , Neurons/metabolism , Rats , Rats, Sprague-Dawley
6.
Cell Metab ; 14(5): 684-99, 2011 Nov 02.
Article in English | MEDLINE | ID: mdl-22000926

ABSTRACT

In obesity, anorectic responses to leptin are diminished, giving rise to the concept of "leptin resistance." Increased expression of protein tyrosine phosphatase 1B (PTP1B) has been associated with the attenuation of leptin signaling and development of cellular leptin resistance. Here we report that hypothalamic levels of the tyrosine phosphatase TCPTP are also elevated in obesity to attenuate the leptin response. We show that mice that lack TCPTP in neuronal cells have enhanced leptin sensitivity and are resistant to high-fat-diet-induced weight gain and the development of leptin resistance. Also, intracerebroventricular administration of a TCPTP inhibitor enhances leptin signaling and responses in mice. Moreover, the combined deletion of TCPTP and PTP1B in neuronal cells has additive effects in the prevention of diet-induced obesity. Our results identify TCPTP as a critical negative regulator of hypothalamic leptin signaling and causally link elevated TCPTP to the development of cellular leptin resistance in obesity.


Subject(s)
Hypothalamus/metabolism , Leptin/metabolism , Neurons/metabolism , Obesity/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 1/deficiency , Protein Tyrosine Phosphatase, Non-Receptor Type 2/deficiency , Signal Transduction , Animals , Blood Glucose/analysis , Body Composition/drug effects , Diet, High-Fat , Enzyme Inhibitors/pharmacology , Female , Gene Expression , Hypothalamus/cytology , Infusions, Intraventricular , Insulin/blood , Male , Mice , Mice, Transgenic , Neurons/cytology , Obesity/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 1/antagonists & inhibitors , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 2/antagonists & inhibitors , Protein Tyrosine Phosphatase, Non-Receptor Type 2/genetics , Receptors, Leptin/metabolism , Tissue Culture Techniques
7.
Endocrinology ; 150(11): 4874-82, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19819947

ABSTRACT

Recent studies demonstrated a role for hypothalamic insulin and leptin action in the regulation of glucose homeostasis. This regulation involves proopiomelanocortin (POMC) neurons because suppression of phosphatidyl inositol 3-kinase (PI3K) signaling in these neurons blunts the acute effects of insulin and leptin on POMC neuronal activity. In the current study, we investigated whether disruption of PI3K signaling in POMC neurons alters normal glucose homeostasis using mouse models designed to both increase and decrease PI3K-mediated signaling in these neurons. We found that deleting p85alpha alone induced resistance to diet-induced obesity. In contrast, deletion of the p110alpha catalytic subunit of PI3K led to increased weight gain and adipose tissue along with reduced energy expenditure. Independent of these effects, increased PI3K activity in POMC neurons improved insulin sensitivity, whereas decreased PI3K signaling resulted in impaired glucose regulation. These studies show that activity of the PI3K pathway in POMC neurons is involved in not only normal energy regulation but also glucose homeostasis.


Subject(s)
Glucose/metabolism , Homeostasis , Hypothalamus/metabolism , Neurons/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Pro-Opiomelanocortin/metabolism , Signal Transduction , Animals , Energy Metabolism , Female , Hypothalamus/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/metabolism , Phosphatidylinositol 3-Kinases/genetics , Pro-Opiomelanocortin/genetics
8.
Nat Med ; 15(8): 940-5, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19633655

ABSTRACT

Although mast cell functions have classically been related to allergic responses, recent studies indicate that these cells contribute to other common diseases such as multiple sclerosis, rheumatoid arthritis, atherosclerosis, aortic aneurysm and cancer. This study presents evidence that mast cells also contribute to diet-induced obesity and diabetes. For example, white adipose tissue (WAT) from obese humans and mice contain more mast cells than WAT from their lean counterparts. Furthermore, in the context of mice on a Western diet, genetically induced deficiency of mast cells, or their pharmacological stabilization, reduces body weight gain and levels of inflammatory cytokines, chemokines and proteases in serum and WAT, in concert with improved glucose homeostasis and energy expenditure. Mechanistic studies reveal that mast cells contribute to WAT and muscle angiogenesis and associated cell apoptosis and cathepsin activity. Adoptive transfer experiments of cytokine-deficient mast cells show that these cells, by producing interleukin-6 (IL-6) and interferon-gamma (IFN-gamma), contribute to mouse adipose tissue cysteine protease cathepsin expression, apoptosis and angiogenesis, thereby promoting diet-induced obesity and glucose intolerance. Our results showing reduced obesity and diabetes in mice treated with clinically available mast cell-stabilizing agents suggest the potential of developing new therapies for these common human metabolic disorders.


Subject(s)
Cromolyn Sodium/therapeutic use , Diabetes Mellitus, Experimental/etiology , Mast Cells/drug effects , Obesity/drug therapy , Obesity/genetics , Proto-Oncogene Proteins c-kit/genetics , Animals , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/immunology , Diet, Atherogenic , Drug Evaluation, Preclinical , Female , Humans , Male , Mast Cells/metabolism , Mice , Mice, Transgenic , Obesity/complications , Obesity/etiology , Obesity/immunology , Organ Specificity/genetics , Proto-Oncogene Proteins c-kit/metabolism
9.
Mol Cell Biol ; 29(16): 4563-73, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19528236

ABSTRACT

PTP1B(-/-) mice are resistant to diet-induced obesity due to leptin hypersensitivity and consequent increased energy expenditure. We aimed to determine the cellular mechanisms underlying this metabolic state. AMPK is an important mediator of leptin's metabolic effects. We find that alpha1 and alpha2 AMPK activity are elevated and acetyl-coenzyme A carboxylase activity is decreased in the muscle and brown adipose tissue (BAT) of PTP1B(-/-) mice. The effects of PTP1B deficiency on alpha2, but not alpha1, AMPK activity in BAT and muscle are neuronally mediated, as they are present in neuron- but not muscle-specific PTP1B(-/-) mice. In addition, AMPK activity is decreased in the hypothalamic nuclei of neuronal and whole-body PTP1B(-/-) mice, accompanied by alterations in neuropeptide expression that are indicative of enhanced leptin sensitivity. Furthermore, AMPK target genes regulating mitochondrial biogenesis, fatty acid oxidation, and energy expenditure are induced with PTP1B inhibition, resulting in increased mitochondrial content in BAT and conversion to a more oxidative muscle fiber type. Thus, neuronal PTP1B inhibition results in decreased hypothalamic AMPK activity, isoform-specific AMPK activation in peripheral tissues, and downstream gene expression changes that promote leanness and increased energy expenditure. Therefore, the mechanism by which PTP1B regulates adiposity and leptin sensitivity likely involves the coordinated regulation of AMPK in hypothalamus and peripheral tissues.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Hypothalamus/enzymology , Isoenzymes/metabolism , Neurons/enzymology , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , AMP-Activated Protein Kinases/genetics , Adipose Tissue, Brown/cytology , Adipose Tissue, Brown/metabolism , Animals , Body Weight , Enzyme Activation , Isoenzymes/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Signal Transduction/physiology , Tissue Distribution
10.
J Biol Chem ; 283(21): 14230-41, 2008 May 23.
Article in English | MEDLINE | ID: mdl-18281274

ABSTRACT

Protein-tyrosine phosphatase 1B (PTP1B) is a major negative regulator of insulin and leptin sensitivity. PTP1B overexpression in adipose tissue and skeletal muscle of humans and rodents may contribute to insulin resistance and obesity. The mechanisms mediating PTP1B overexpression in obese and diabetic states have been unclear. We find that adipose tissue inflammation and the pro-inflammatory cytokine tumor necrosis factor alpha (TNFalpha) regulate PTP1B expression in vivo. High fat feeding of mice increased PTP1B expression 1.5- to 7-fold in adipose tissue, liver, skeletal muscle, and arcuate nucleus of hypothalamus. PTP1B overexpression in high fat-fed mice coincided with increased adipose tissue expression of the macrophage marker CD68 and TNFalpha, which is implicated in causing obesity-induced insulin resistance. TNFalpha increased PTP1B mRNA and protein levels by 2- to 5-fold in a dose- and time-dependent manner in adipocyte and hepatocyte cell lines. TNFalpha administration in mice increased PTP1B mRNA 1.4- to 4-fold in adipose tissue, liver, skeletal muscle, and hypothalamic arcuate nucleus and PTP1B protein 2-fold in liver. Actinomycin D treatment blocked, and high dose salicylate treatment inhibited by 80%, TNFalpha-induced PTP1B expression in adipocyte cell lines, suggesting TNFalpha may induce PTP1B transcription via nuclear factor kappaB (NFkappaB) activation. Chromatin immunoprecipitation from adipocyte cell lines and liver of mice demonstrated TNFalpha-induced recruitment of NFkappaB subunit p65 to the PTP1B promoter in vitro and in vivo. In mice with diet-induced obesity, TNFalpha deficiency also partly blocked PTP1B overexpression in adipose tissue. Our data suggest that PTP1B overexpression in multiple tissues in obesity is regulated by inflammation and that PTP1B may be a target of anti-inflammatory therapies.


Subject(s)
Gene Expression Regulation, Enzymologic , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Animal Feed , Animals , Base Sequence , Cells, Cultured , Diabetes Mellitus/enzymology , Gene Expression Regulation, Enzymologic/drug effects , Inflammation/enzymology , Insulin/metabolism , Insulin Resistance , Leptin/metabolism , Mice , Molecular Sequence Data , Obesity/enzymology , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Rats , Transcription, Genetic/drug effects , Transcription, Genetic/genetics , Tumor Necrosis Factor-alpha/deficiency , Tumor Necrosis Factor-alpha/pharmacology
11.
Endocrinology ; 148(3): 1367-75, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17185376

ABSTRACT

Antecedent hypoglycemia blunts counterregulatory responses that normally restore glycemia, a phenomenon known as hypoglycemia-associated autonomic failure (HAAF). The mechanisms leading to impaired counterregulatory responses are largely unknown. Hypothalamic AMP-activated protein kinase (AMPK) acts as a glucose sensor. To determine whether failure to activate AMPK could be involved in the etiology of HAAF, we developed a model of HAAF using repetitive intracerebroventricular (icv) injection of 2-deoxy-D-glucose (2DG) resulting in transient neuroglucopenia in normal rats. Ten minutes after a single icv injection of 2DG, both alpha1- and alpha2-AMPK activities were increased 30-50% in arcuate and ventromedial/dorsomedial hypothalamus but not in other hypothalamic regions, hindbrain, or cortex. Increased AMPK activity persisted in arcuate hypothalamus at 60 min after 2DG injection when serum glucagon and corticosterone levels were increased 2.5- to 3.4-fold. When 2DG was injected icv daily for 4 d, hypothalamic alpha1- and alpha2-AMPK responses were markedly blunted in arcuate hypothalamus, and alpha1-AMPK was also blunted in mediobasal hypothalamus 10 min after 2DG on d 4. Both AMPK isoforms were activated normally in arcuate hypothalamus at 60 min. Counterregulatory hormone responses were impaired by recurrent neuroglucopenia and were partially restored by icv injection of 5-aminoimidazole-4-carboxamide 1-beta-D-ribofuranoside, an AMPK activator, before 2DG. Glycogen content increased 2-fold in hypothalamus after recurrent neuroglucopenia, suggesting that glycogen supercompensation could be involved in down-regulating the AMPK glucose-sensing pathway in HAAF. Thus, activation of hypothalamic AMPK may be important for the full counterregulatory hormone response to neuroglucopenia. Furthermore, impaired or delayed AMPK activation in specific hypothalamic regions may play a critical role in the etiology of HAAF.


Subject(s)
Brain/metabolism , Hypothalamus/enzymology , Multienzyme Complexes/physiology , Protein Serine-Threonine Kinases/physiology , AMP-Activated Protein Kinases , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Animals , Brain/enzymology , Brain/pathology , Brain Chemistry/drug effects , Deoxyglucose/pharmacology , Eating/drug effects , Glucose/metabolism , Glycogen/analysis , Hypoglycemia/chemically induced , Hypoglycemia/enzymology , Hypoglycemia/physiopathology , Male , Multienzyme Complexes/metabolism , Neurosecretory Systems/physiology , Protein Serine-Threonine Kinases/metabolism , Rats , Rats, Sprague-Dawley , Recurrence , Ribonucleotides/pharmacology
13.
J Physiol ; 574(Pt 1): 73-83, 2006 Jul 01.
Article in English | MEDLINE | ID: mdl-16709629

ABSTRACT

The evolutionarily conserved serine/threonine kinase, AMP-activated protein kinase (AMPK), functions as a cellular fuel gauge that regulates metabolic pathways in glucose and fatty acid metabolism and protein synthesis. Recent data strongly implicate the AMPK-acetyl CoA carboxylase (ACC)-malonyl CoA pathway in the hypothalamus in the regulation of food intake, body weight and hepatic glucose production. Furthermore, data indicate that AMPK is a mediator of the effects of adipocyte-derived and gut-derived hormones and peptides on fatty acid oxidation and glucose uptake in peripheral tissues. Studies are now elucidating the potential role of kinases upstream of AMPK in these metabolic effects. In addition, recently, several novel downstream effectors of AMPK have been identified. The AMPK pathway in the hypothalamus and peripheral tissues coordinately integrates inputs from multiple hormones, peptides and nutrients to maintain energy homeostasis.


Subject(s)
Eating/physiology , Energy Metabolism/physiology , Glucose/metabolism , Hormones/metabolism , Hypothalamus/physiology , Liver/metabolism , Multienzyme Complexes/metabolism , Protein Serine-Threonine Kinases/metabolism , AMP-Activated Protein Kinases , Animals , Body Weight/physiology , Feedback/physiology , Homeostasis/physiology , Humans , Signal Transduction/physiology
14.
J Biol Chem ; 281(28): 18933-41, 2006 Jul 14.
Article in English | MEDLINE | ID: mdl-16687413

ABSTRACT

AMP-activated protein kinase (AMPK) is a key regulator of cellular energy balance and of the effects of leptin on food intake and fatty acid oxidation. Obesity is usually associated with resistance to the effects of leptin on food intake and body weight. To determine whether diet-induced obesity (DIO) impairs the AMPK response to leptin in muscle and/or hypothalamus, we fed FVB mice a high fat (55%) diet for 10-12 weeks. Leptin acutely decreased food intake by approximately 30% in chow-fed mice. DIO mice tended to eat less, and leptin had no effect on food intake. Leptin decreased respiratory exchange ratio in chow-fed mice indicating increased fatty acid oxidation. Respiratory exchange ratio was low basally in high fat-fed mice, and leptin had no further effect. Leptin (3 mg/kg intraperitoneally) increased alpha2-AMPK activity 2-fold in muscle in chow-fed mice but not in DIO mice. Leptin decreased acetyl-CoA carboxylase activity 40% in muscle from chow-fed mice. In muscle from DIO mice, acetyl-CoA carboxylase activity was basally low, and leptin had no further effect. In paraventricular, arcuate, and medial hypothalamus of chow-fed mice, leptin inhibited alpha2-AMPK activity but not in DIO mice. In addition, leptin increased STAT3 phosphorylation 2-fold in arcuate of chow-fed mice, but this effect was attenuated because of elevated basal STAT3 phosphorylation in DIO mice. Thus, DIO in FVB mice alters alpha2-AMPK in muscle and hypothalamus and STAT3 in hypothalamus and impairs further effects of leptin on these signaling pathways. Defective responses of AMPK to leptin may contribute to resistance to leptin action on food intake and energy expenditure in obese states.


Subject(s)
Animal Feed , Hypothalamus/enzymology , Multienzyme Complexes/physiology , Muscle, Skeletal/enzymology , Obesity , Protein Serine-Threonine Kinases/physiology , AMP-Activated Protein Kinases , Acetyl-CoA Carboxylase/metabolism , Animals , Body Weight , Energy Metabolism , Fatty Acids/metabolism , Hypothalamus/pathology , Leptin/metabolism , Male , Mice , Oxygen/metabolism
16.
Nature ; 428(6982): 569-74, 2004 Apr 01.
Article in English | MEDLINE | ID: mdl-15058305

ABSTRACT

Obesity is an epidemic in Western society, and causes rapidly accelerating rates of type 2 diabetes and cardiovascular disease. The evolutionarily conserved serine/threonine kinase, AMP-activated protein kinase (AMPK), functions as a 'fuel gauge' to monitor cellular energy status. We investigated the potential role of AMPK in the hypothalamus in the regulation of food intake. Here we report that AMPK activity is inhibited in arcuate and paraventricular hypothalamus (PVH) by the anorexigenic hormone leptin, and in multiple hypothalamic regions by insulin, high glucose and refeeding. A melanocortin receptor agonist, a potent anorexigen, decreases AMPK activity in PVH, whereas agouti-related protein, an orexigen, increases AMPK activity. Melanocortin receptor signalling is required for leptin and refeeding effects on AMPK in PVH. Dominant negative AMPK expression in the hypothalamus is sufficient to reduce food intake and body weight, whereas constitutively active AMPK increases both. Alterations of hypothalamic AMPK activity augment changes in arcuate neuropeptide expression induced by fasting and feeding. Furthermore, inhibition of hypothalamic AMPK is necessary for leptin's effects on food intake and body weight, as constitutively active AMPK blocks these effects. Thus, hypothalamic AMPK plays a critical role in hormonal and nutrient-derived anorexigenic and orexigenic signals and in energy balance.


Subject(s)
Adenylate Kinase/metabolism , Feeding Behavior/physiology , Hormones/metabolism , Hypothalamus/enzymology , Hypothalamus/physiology , Adenylate Kinase/antagonists & inhibitors , Adenylate Kinase/chemistry , Adenylate Kinase/genetics , Animals , Body Weight/drug effects , Energy Metabolism/drug effects , Feeding Behavior/drug effects , Glucose/metabolism , Glucose/pharmacology , Hormones/pharmacology , Hypothalamus/drug effects , Insulin/metabolism , Insulin/pharmacology , Leptin/metabolism , Leptin/pharmacology , Male , Mice , Models, Biological , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Melanocortin/antagonists & inhibitors , Receptors, Melanocortin/metabolism
17.
Dev Cell ; 2(4): 489-95, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11970898

ABSTRACT

Mice lacking the protein-tyrosine phosphatase PTP1B are hypersensitive to insulin and resistant to obesity. However, the molecular basis for resistance to obesity has been unclear. Here we show that PTP1B regulates leptin signaling. In transfection studies, PTP1B dephosphorylates the leptin receptor-associated kinase, Jak2. PTP1B is expressed in hypothalamic regions harboring leptin-responsive neurons. Compared to wild-type littermates, PTP1B(-/-) mice have decreased leptin/body fat ratios, leptin hypersensitivity, and enhanced leptin-induced hypothalamic Stat3 tyrosyl phosphorylation. Gold thioglucose treatment, which ablates leptin-responsive hypothalamic neurons, partially overcomes resistance to obesity in PTP1B(-/-) mice. Our data indicate that PTP1B regulates leptin signaling in vivo, likely by targeting Jak2. PTP1B may be a novel target to treat leptin resistance in obesity.


Subject(s)
Leptin/metabolism , Obesity/metabolism , Protein Tyrosine Phosphatases/metabolism , Proto-Oncogene Proteins , Signal Transduction/physiology , Animals , COS Cells , Gene Expression Regulation, Enzymologic , Hippocampus/physiology , Hypothalamus/physiology , Janus Kinase 2 , Mice , Mice, Knockout , Molecular Sequence Data , Protein Tyrosine Phosphatase, Non-Receptor Type 1 , Protein Tyrosine Phosphatases/genetics , Protein-Tyrosine Kinases/metabolism , Transfection , Weight Gain
SELECTION OF CITATIONS
SEARCH DETAIL