Skip to main content
Top
Published in: Diabetologia 1/2007

01-01-2007 | Article

Inhibition by glucose or leptin of hypothalamic neurons expressing neuropeptide Y requires changes in AMP-activated protein kinase activity

Authors: P. D. Mountjoy, S. J. Bailey, G. A. Rutter

Published in: Diabetologia | Issue 1/2007

Login to get access

Abstract

Aims/hypothesis

Changes in the activity of glucose-excited and glucose-inhibited neurons within the basomedial hypothalamus are key to the central regulation of satiety. However, the molecular mechanisms through which these cells respond to extracellular stimuli remain poorly understood. Here, we investigate the role of 5′-AMP-activated protein kinase (AMPK), a trimeric complex encoded by seven distinct genes of the PRKA family, in the responses to glucose and leptin of each cell type.

Methods

The activity of isolated rat basomedial hypothalamic neurons was assessed by: (1) recording cellular voltage responses under current clamp; (2) measuring intracellular free Ca2+ with fluo-3 or fura-2; and (3) developing a neuropeptide Y (NPY) promoter-driven adenovirally produced ratiometric ‘pericam’ (a green fluorescent protein-based Ca2+ sensor) to monitor [Ca2+] changes selectively in NPY-positive neurons.

Results

The stimulatory effects of decreased (0 or 1.0 vs 15 mmol/l) glucose on glucose-inhibited neurons were mimicked by the AMPK activator, 5-amino-imidazole-4-carboxamide riboside (AICAR) and blocked by the inhibitor Compound C. Similarly, AICAR reversed the inhibitory effects of leptin in the majority of glucose-inhibited neurons. The responses to glucose of Npy-expressing cells, which represented ∼40 % of all glucose-inhibited neurons, were also sensitive to Compound C or AICAR. Forced changes in AMPK activity had no effect on glucose-excited and non-glucose-responsive neurons.

Conclusions/interpretation

Changes in AMPK activity are involved in the responses of glucose-inhibited neurons to large fluctuations in glucose concentration, and possibly also to leptin. This mechanism may contribute to the acute reduction of electrical activity and Ca2+ oscillation frequency in these, but not other neurons, in the basomedial hypothalamus.
Appendix
Available only for authorised users
Literature
2.
go back to reference Grundy SM, Hansen B, Smith SC Jr et al (2004) Clinical management of metabolic syndrome: report of the American Heart Association/National Heart, Lung, and Blood Institute/American Diabetes Association conference on scientific issues related to management. Circulation 109:551–556PubMedCrossRef Grundy SM, Hansen B, Smith SC Jr et al (2004) Clinical management of metabolic syndrome: report of the American Heart Association/National Heart, Lung, and Blood Institute/American Diabetes Association conference on scientific issues related to management. Circulation 109:551–556PubMedCrossRef
3.
go back to reference Elias CF, Aschkenasi C, Lee C et al (1999) Leptin differentially regulates NPY and POMC neurons projecting to the lateral hypothalamic area. Neuron 23:775–786PubMedCrossRef Elias CF, Aschkenasi C, Lee C et al (1999) Leptin differentially regulates NPY and POMC neurons projecting to the lateral hypothalamic area. Neuron 23:775–786PubMedCrossRef
4.
go back to reference Wang Q, Bing C, Al-Barazanji K et al (1997) Interactions between leptin and hypothalamic neuropeptide Y neurons in the control of food intake and energy homeostasis in the rat. Diabetes 46:335–341PubMed Wang Q, Bing C, Al-Barazanji K et al (1997) Interactions between leptin and hypothalamic neuropeptide Y neurons in the control of food intake and energy homeostasis in the rat. Diabetes 46:335–341PubMed
5.
go back to reference Jobst EE, Enriori PJ, Cowley MA (2004) The electrophysiology of feeding circuits. Trends Endocrinol Metab 15:488–499PubMedCrossRef Jobst EE, Enriori PJ, Cowley MA (2004) The electrophysiology of feeding circuits. Trends Endocrinol Metab 15:488–499PubMedCrossRef
6.
go back to reference Cowley MA, Smart JL, Rubinstein M et al (2001) Leptin activates anorexigenic POMC neurons through a neural network in the arcuate nucleus. Nature 411:480–484PubMedCrossRef Cowley MA, Smart JL, Rubinstein M et al (2001) Leptin activates anorexigenic POMC neurons through a neural network in the arcuate nucleus. Nature 411:480–484PubMedCrossRef
7.
go back to reference Cowley MA, Smith RG, Diano S et al (2003) The distribution and mechanism of action of ghrelin in the CNS demonstrates a novel hypothalamic circuit regulating energy homeostasis. Neuron 37:649–661PubMedCrossRef Cowley MA, Smith RG, Diano S et al (2003) The distribution and mechanism of action of ghrelin in the CNS demonstrates a novel hypothalamic circuit regulating energy homeostasis. Neuron 37:649–661PubMedCrossRef
8.
go back to reference Shintani M, Ogawa Y, Ebihara K et al (2001) Ghrelin, an endogenous growth hormone secretagogue, is a novel orexigenic peptide that antagonizes leptin action through the activation of hypothalamic neuropeptide Y/Y1 receptor pathway. Diabetes 50:227–232PubMed Shintani M, Ogawa Y, Ebihara K et al (2001) Ghrelin, an endogenous growth hormone secretagogue, is a novel orexigenic peptide that antagonizes leptin action through the activation of hypothalamic neuropeptide Y/Y1 receptor pathway. Diabetes 50:227–232PubMed
9.
go back to reference Oomura Y, Ono T, Ooyama H, Wayner MJ (1969) Glucose and osmosensitive neurones of the rat hypothalamus. Nature 222:282–284PubMedCrossRef Oomura Y, Ono T, Ooyama H, Wayner MJ (1969) Glucose and osmosensitive neurones of the rat hypothalamus. Nature 222:282–284PubMedCrossRef
10.
go back to reference Oomura Y, Ooyama H, Sugimori M, Nakamura T, Yamada Y (1974) Glucose inhibition of the glucose-sensitive neurone in the rat lateral hypothalamus. Nature 247:284–286PubMedCrossRef Oomura Y, Ooyama H, Sugimori M, Nakamura T, Yamada Y (1974) Glucose inhibition of the glucose-sensitive neurone in the rat lateral hypothalamus. Nature 247:284–286PubMedCrossRef
11.
go back to reference Ashford ML, Boden PR, Treherne JM (1990) Glucose-induced excitation of hypothalamic neurones is mediated by ATP-sensitive K+ channels. Pflugers Arch 415:479–483PubMedCrossRef Ashford ML, Boden PR, Treherne JM (1990) Glucose-induced excitation of hypothalamic neurones is mediated by ATP-sensitive K+ channels. Pflugers Arch 415:479–483PubMedCrossRef
12.
go back to reference Spanswick D, Smith MA, Groppi VE, Logan SD, Ashford ML (1997) Leptin inhibits hypothalamic neurons by activation of ATP-sensitive potassium channels. Nature 390:521–525PubMedCrossRef Spanswick D, Smith MA, Groppi VE, Logan SD, Ashford ML (1997) Leptin inhibits hypothalamic neurons by activation of ATP-sensitive potassium channels. Nature 390:521–525PubMedCrossRef
13.
go back to reference Song Z, Levin BE, McArdle JJ, Bakhos N, Routh VH (2001) Convergence of pre- and postsynaptic influences on glucosensing neurons in the ventromedial hypothalamic nucleus. Diabetes 50:2673–2681PubMed Song Z, Levin BE, McArdle JJ, Bakhos N, Routh VH (2001) Convergence of pre- and postsynaptic influences on glucosensing neurons in the ventromedial hypothalamic nucleus. Diabetes 50:2673–2681PubMed
14.
go back to reference Dunn-Meynell AA, Routh VH, Kang L, Gaspers L, Levin BE (2002) Glucokinase is the likely mediator of glucosensing in both glucose-excited and glucose-inhibited central neurons. Diabetes 51:2056–2065PubMed Dunn-Meynell AA, Routh VH, Kang L, Gaspers L, Levin BE (2002) Glucokinase is the likely mediator of glucosensing in both glucose-excited and glucose-inhibited central neurons. Diabetes 51:2056–2065PubMed
15.
go back to reference Kang L, Routh VH, Kuzhikandathil EV, Gaspers LD, Levin BE (2004) Physiological and molecular characteristics of rat hypothalamic ventromedial nucleus glucosensing neurons. Diabetes 53:549–559PubMed Kang L, Routh VH, Kuzhikandathil EV, Gaspers LD, Levin BE (2004) Physiological and molecular characteristics of rat hypothalamic ventromedial nucleus glucosensing neurons. Diabetes 53:549–559PubMed
16.
go back to reference Rutter GA (2004) Visualising insulin secretion. The Minkowski Lecture 2004. Diabetologia 47:1861–1872PubMedCrossRef Rutter GA (2004) Visualising insulin secretion. The Minkowski Lecture 2004. Diabetologia 47:1861–1872PubMedCrossRef
17.
go back to reference Ainscow EK, Mirshamsi S, Tang T, Ashford ML, Rutter GA (2002) Dynamic imaging of free cytosolic ATP concentration during fuel sensing by rat hypothalamic neurones: evidence for ATP-independent control of ATP-sensitive K(+) channels. J Physiol 544:429–445PubMedCrossRef Ainscow EK, Mirshamsi S, Tang T, Ashford ML, Rutter GA (2002) Dynamic imaging of free cytosolic ATP concentration during fuel sensing by rat hypothalamic neurones: evidence for ATP-independent control of ATP-sensitive K(+) channels. J Physiol 544:429–445PubMedCrossRef
18.
go back to reference Hardie DG, Carling D, Carlson M (1998) The AMP-activated/SNF1 protein kinase subfamily: metabolic sensors of the eukaryotic cell? Annu Rev Biochem 67:821–855PubMedCrossRef Hardie DG, Carling D, Carlson M (1998) The AMP-activated/SNF1 protein kinase subfamily: metabolic sensors of the eukaryotic cell? Annu Rev Biochem 67:821–855PubMedCrossRef
19.
go back to reference Rutter GA, da Silva Xavier G, Leclerc I (2003) Roles of 5′-AMP-activated protein kinase (AMPK) in mammalian glucose homoeostasis. Biochem J 375:1–16PubMedCrossRef Rutter GA, da Silva Xavier G, Leclerc I (2003) Roles of 5′-AMP-activated protein kinase (AMPK) in mammalian glucose homoeostasis. Biochem J 375:1–16PubMedCrossRef
20.
go back to reference Minokoshi Y, Alquier T, Furukawa N et al (2004) AMP-kinase regulates food intake by responding to hormonal and nutrient signals in the hypothalamus. Nature 428:569–574PubMedCrossRef Minokoshi Y, Alquier T, Furukawa N et al (2004) AMP-kinase regulates food intake by responding to hormonal and nutrient signals in the hypothalamus. Nature 428:569–574PubMedCrossRef
21.
go back to reference Andersson U, Filipsson K, Abbott CR et al (2004) AMP-activated protein kinase plays a role in the control of food intake. J Biol Chem 279:12005–12008PubMedCrossRef Andersson U, Filipsson K, Abbott CR et al (2004) AMP-activated protein kinase plays a role in the control of food intake. J Biol Chem 279:12005–12008PubMedCrossRef
22.
go back to reference Nagai T, Sawano A, Park ES, Miyawaki A (2001) Circularly permuted green fluorescent proteins engineered to sense Ca2+. Proc Natl Acad Sci USA 98:3197–3202PubMedCrossRef Nagai T, Sawano A, Park ES, Miyawaki A (2001) Circularly permuted green fluorescent proteins engineered to sense Ca2+. Proc Natl Acad Sci USA 98:3197–3202PubMedCrossRef
23.
go back to reference Glover CPJ, Bienemann AS, Heywood DJ, Cosgrave AS, Uney JB (2002) Adenoviral-mediated, high-level, cell-specific transgene expression: a SYN1-WPRE cassette mediates increased transgene expression with no loss of neuron specificity. Mol Ther 5:509–516PubMedCrossRef Glover CPJ, Bienemann AS, Heywood DJ, Cosgrave AS, Uney JB (2002) Adenoviral-mediated, high-level, cell-specific transgene expression: a SYN1-WPRE cassette mediates increased transgene expression with no loss of neuron specificity. Mol Ther 5:509–516PubMedCrossRef
24.
go back to reference He TC, Zhou S, da Costa LT, Yu J, Kinzler KW, Vogelstein B (1998) A simplified system for generating recombinant adenoviruses. Proc Natl Acad Sci USA 95:2509–2514PubMedCrossRef He TC, Zhou S, da Costa LT, Yu J, Kinzler KW, Vogelstein B (1998) A simplified system for generating recombinant adenoviruses. Proc Natl Acad Sci USA 95:2509–2514PubMedCrossRef
25.
go back to reference Williams AG, Hargreaves AC, Gunn-Moore FJ, Tavare JM (1998) Stimulation of neuropeptide Y gene expression by brain-derived neurotrophic factor requires both the phospholipase Cgamma and Shc binding sites on its receptor, TrkB. Biochem J 333:505–509PubMed Williams AG, Hargreaves AC, Gunn-Moore FJ, Tavare JM (1998) Stimulation of neuropeptide Y gene expression by brain-derived neurotrophic factor requires both the phospholipase Cgamma and Shc binding sites on its receptor, TrkB. Biochem J 333:505–509PubMed
26.
go back to reference Ainscow EK, Rutter GA (2001) Mitochondrial priming modifies Ca2+ oscillations and insulin secretion in pancreatic islets. Biochem J 353:175–180PubMedCrossRef Ainscow EK, Rutter GA (2001) Mitochondrial priming modifies Ca2+ oscillations and insulin secretion in pancreatic islets. Biochem J 353:175–180PubMedCrossRef
27.
go back to reference da Silva Xavier G, Leclerc I, Varadi A, Tsuboi T, Moule SK, Rutter GA (2003) Role for AMP-activated protein kinase in glucose-stimulated insulin secretion and preproinsulin gene expression. Biochem J 371:761–774PubMedCrossRef da Silva Xavier G, Leclerc I, Varadi A, Tsuboi T, Moule SK, Rutter GA (2003) Role for AMP-activated protein kinase in glucose-stimulated insulin secretion and preproinsulin gene expression. Biochem J 371:761–774PubMedCrossRef
28.
go back to reference Varadi A, Rutter GA (2002) Dynamic imaging of endoplasmic reticulum Ca2+ concentration in insulin-secreting MIN6 cells using recombinant targeted cameleons: roles of sarco(endo)plasmic reticulum Ca2+-ATPase (SERCA)-2 and ryanodine receptors. Diabetes 51:S190–S201PubMed Varadi A, Rutter GA (2002) Dynamic imaging of endoplasmic reticulum Ca2+ concentration in insulin-secreting MIN6 cells using recombinant targeted cameleons: roles of sarco(endo)plasmic reticulum Ca2+-ATPase (SERCA)-2 and ryanodine receptors. Diabetes 51:S190–S201PubMed
29.
go back to reference Silver I, Erecinska M (1994) Extracellular glucose concentration in mammalian brain: continuous monitoring of changes during increased neuronal activity and upon limitation in oxygen supply in normo-, hypo-, and hyperglycemic animals. J Neurosci 14:5068–5076PubMed Silver I, Erecinska M (1994) Extracellular glucose concentration in mammalian brain: continuous monitoring of changes during increased neuronal activity and upon limitation in oxygen supply in normo-, hypo-, and hyperglycemic animals. J Neurosci 14:5068–5076PubMed
30.
go back to reference de Vries MG, Arseneau LM, Lawson ME, Beverly JL (2003) Extracellular glucose in rat ventromedial hypothalamus during acute and recurrent hypoglycemia. Diabetes 52:2767–2773PubMed de Vries MG, Arseneau LM, Lawson ME, Beverly JL (2003) Extracellular glucose in rat ventromedial hypothalamus during acute and recurrent hypoglycemia. Diabetes 52:2767–2773PubMed
31.
go back to reference Fioramonti X, Lorsignol A, Taupignon A, Penicaud L (2004) A new ATP-sensitive K+ channel-independent mechanism is involved in glucose-excited neurons of mouse arcuate nucleus. Diabetes 53:2767–2775PubMed Fioramonti X, Lorsignol A, Taupignon A, Penicaud L (2004) A new ATP-sensitive K+ channel-independent mechanism is involved in glucose-excited neurons of mouse arcuate nucleus. Diabetes 53:2767–2775PubMed
32.
go back to reference Mobbs CV, Kow LM, Yang XJ (2001) Brain glucose-sensing mechanisms: ubiquitous silencing by aglycemia vs. hypothalamic neuroendocrine responses. Am J Physiol Endocrinol Metab 281:E649–E654PubMed Mobbs CV, Kow LM, Yang XJ (2001) Brain glucose-sensing mechanisms: ubiquitous silencing by aglycemia vs. hypothalamic neuroendocrine responses. Am J Physiol Endocrinol Metab 281:E649–E654PubMed
33.
go back to reference Muroya S, Yada T, Shioda S, Takigawa M (1999) Glucose-sensitive neurons in the rat arcuate nucleus contain neuropeptide Y. Neurosci Lett 264:113–116PubMedCrossRef Muroya S, Yada T, Shioda S, Takigawa M (1999) Glucose-sensitive neurons in the rat arcuate nucleus contain neuropeptide Y. Neurosci Lett 264:113–116PubMedCrossRef
34.
go back to reference Levin BE, Dunn-Meynell AA, Routh VH (1999) Brain glucose sensing and body energy homeostasis: role in obesity and diabetes. Am J Physiol 276:R1223–R1231PubMed Levin BE, Dunn-Meynell AA, Routh VH (1999) Brain glucose sensing and body energy homeostasis: role in obesity and diabetes. Am J Physiol 276:R1223–R1231PubMed
35.
go back to reference Zhou G, Myers R, Li Y et al (2001) Role of AMP-activated protein kinase in mechanism of metformin action. J Clin Invest 108:1167–1174PubMedCrossRef Zhou G, Myers R, Li Y et al (2001) Role of AMP-activated protein kinase in mechanism of metformin action. J Clin Invest 108:1167–1174PubMedCrossRef
36.
go back to reference Kim EK, Miller I, Aja S et al (2004) C75, a fatty acid synthase inhibitor, reduces food intake via hypothalamic AMP-activated protein kinase. J Biol Chem 279:19970–19976PubMedCrossRef Kim EK, Miller I, Aja S et al (2004) C75, a fatty acid synthase inhibitor, reduces food intake via hypothalamic AMP-activated protein kinase. J Biol Chem 279:19970–19976PubMedCrossRef
37.
go back to reference Muroya S, Funahashi H, Yamanaka A et al (2004) Orexins (hypocretins) directly interact with neuropeptide Y, POMC and glucose-responsive neurons to regulate Ca2+ signaling in a reciprocal manner to leptin: orexigenic neuronal pathways in the mediobasal hypothalamus. Eur J Neurosci 19:1524–1534PubMedCrossRef Muroya S, Funahashi H, Yamanaka A et al (2004) Orexins (hypocretins) directly interact with neuropeptide Y, POMC and glucose-responsive neurons to regulate Ca2+ signaling in a reciprocal manner to leptin: orexigenic neuronal pathways in the mediobasal hypothalamus. Eur J Neurosci 19:1524–1534PubMedCrossRef
38.
go back to reference Wang R, Liu X, Hentges ST et al (2004) The regulation of glucose-excited neurons in the hypothalamic arcuate nucleus by glucose and feeding-relevant peptides. Diabetes 53:1959–1965PubMed Wang R, Liu X, Hentges ST et al (2004) The regulation of glucose-excited neurons in the hypothalamic arcuate nucleus by glucose and feeding-relevant peptides. Diabetes 53:1959–1965PubMed
39.
go back to reference Muroya S, Uramura K, Sakurai T, Takigawa M, Yada T (2001) Lowering glucose concentrations increases cytosolic Ca2+ in orexin neurons of the rat lateral hypothalamus. Neurosci Lett 309:165–168PubMedCrossRef Muroya S, Uramura K, Sakurai T, Takigawa M, Yada T (2001) Lowering glucose concentrations increases cytosolic Ca2+ in orexin neurons of the rat lateral hypothalamus. Neurosci Lett 309:165–168PubMedCrossRef
40.
go back to reference Burdakov D, Gerasimenko O, Verkhratsky A (2005) Physiological changes in glucose differentially modulate the excitability of hypothalamic melanin-concentrating hormone and orexin neurons in situ. J Neurosci 25:2429–2433PubMedCrossRef Burdakov D, Gerasimenko O, Verkhratsky A (2005) Physiological changes in glucose differentially modulate the excitability of hypothalamic melanin-concentrating hormone and orexin neurons in situ. J Neurosci 25:2429–2433PubMedCrossRef
41.
go back to reference Kang L, Dunn-Meynell AA, Routh VH et al (2006) Glucokinase is a critical regulator of ventromedial hypothalamic neuronal glucosensing. Diabetes 55:412–420PubMedCrossRef Kang L, Dunn-Meynell AA, Routh VH et al (2006) Glucokinase is a critical regulator of ventromedial hypothalamic neuronal glucosensing. Diabetes 55:412–420PubMedCrossRef
42.
go back to reference Ganong WF (2000) Circumventricular organs: definition and role in the regulation of endocrine and autonomic function. Clin Exp Pharmacol Physiol 27:422–427PubMedCrossRef Ganong WF (2000) Circumventricular organs: definition and role in the regulation of endocrine and autonomic function. Clin Exp Pharmacol Physiol 27:422–427PubMedCrossRef
43.
go back to reference Kim M-S, Park J-Y, Namkoong C et al (2004) Anti-obesity effects of [alpha]-lipoic acid mediated by suppression of hypothalamic AMP-activated protein kinase. Nat Med 10:727–733PubMedCrossRef Kim M-S, Park J-Y, Namkoong C et al (2004) Anti-obesity effects of [alpha]-lipoic acid mediated by suppression of hypothalamic AMP-activated protein kinase. Nat Med 10:727–733PubMedCrossRef
44.
go back to reference McCrimmon RJ, Fan X, Ding Y, Zhu W, Jacob RJ, Sherwin RS (2004) Potential role for AMP-activated protein kinase in hypoglycemia sensing in the ventromedial hypothalamus. Diabetes 53:1953–1958PubMed McCrimmon RJ, Fan X, Ding Y, Zhu W, Jacob RJ, Sherwin RS (2004) Potential role for AMP-activated protein kinase in hypoglycemia sensing in the ventromedial hypothalamus. Diabetes 53:1953–1958PubMed
45.
go back to reference Namkoong C, Kim MS, Jang PG et al (2005) Enhanced hypothalamic AMP-activated protein kinase activity contributes to hyperphagia in diabetic rats. Diabetes 54:63–68PubMed Namkoong C, Kim MS, Jang PG et al (2005) Enhanced hypothalamic AMP-activated protein kinase activity contributes to hyperphagia in diabetic rats. Diabetes 54:63–68PubMed
46.
go back to reference Gadalla AE, Pearson T, Currie AJ et al (2004) AICA riboside both activates AMP-activated protein kinase and competes with adenosine for the nucleoside transporter in the CA1 region of the rat hippocampus. J Neurochem 88:1272–1282PubMedCrossRef Gadalla AE, Pearson T, Currie AJ et al (2004) AICA riboside both activates AMP-activated protein kinase and competes with adenosine for the nucleoside transporter in the CA1 region of the rat hippocampus. J Neurochem 88:1272–1282PubMedCrossRef
47.
go back to reference Chen ZP, Levy A, Lightman SL (1994) Activation of specific ATP receptors induces a rapid increase in intracellular calcium ions in rat hypothalamic neurons. Brain Res 641:249–256PubMedCrossRef Chen ZP, Levy A, Lightman SL (1994) Activation of specific ATP receptors induces a rapid increase in intracellular calcium ions in rat hypothalamic neurons. Brain Res 641:249–256PubMedCrossRef
48.
go back to reference Hardingham GE, Chawla S, Johnson CM, Bading H (1997) Distinct functions of nuclear and cytoplasmic calcium in the control of gene expression. Nature 385:260–265PubMedCrossRef Hardingham GE, Chawla S, Johnson CM, Bading H (1997) Distinct functions of nuclear and cytoplasmic calcium in the control of gene expression. Nature 385:260–265PubMedCrossRef
49.
go back to reference Koo S-H, Flechner L, Qi L et al (2005) The CREB coactivator TORC2 is a key regulator of fasting glucose metabolism. Nature 437:1109–1111PubMedCrossRef Koo S-H, Flechner L, Qi L et al (2005) The CREB coactivator TORC2 is a key regulator of fasting glucose metabolism. Nature 437:1109–1111PubMedCrossRef
50.
go back to reference Gribble FM, Proks P, Corkey BE, Ashcroft FM (1998) Mechanism of cloned ATP-sensitive potassium channel activation by oleoyl-CoA. J Biol Chem 273:26383–26387PubMedCrossRef Gribble FM, Proks P, Corkey BE, Ashcroft FM (1998) Mechanism of cloned ATP-sensitive potassium channel activation by oleoyl-CoA. J Biol Chem 273:26383–26387PubMedCrossRef
51.
go back to reference Fulceri R, Nori A, Gamberucci A, Volpe P, Giunti R, Benedetti A (1994) Fatty acyl-CoA esters induce calcium release from terminal cisternae of skeletal muscle. Cell Calcium 15:109–116PubMedCrossRef Fulceri R, Nori A, Gamberucci A, Volpe P, Giunti R, Benedetti A (1994) Fatty acyl-CoA esters induce calcium release from terminal cisternae of skeletal muscle. Cell Calcium 15:109–116PubMedCrossRef
52.
go back to reference Loftus TM, Jaworsky DE, Frehywot GL et al (2000) Reduced food intake and body weight in mice treated with fatty acid synthase inhibitors. Science 288:2379–2381PubMedCrossRef Loftus TM, Jaworsky DE, Frehywot GL et al (2000) Reduced food intake and body weight in mice treated with fatty acid synthase inhibitors. Science 288:2379–2381PubMedCrossRef
Metadata
Title
Inhibition by glucose or leptin of hypothalamic neurons expressing neuropeptide Y requires changes in AMP-activated protein kinase activity
Authors
P. D. Mountjoy
S. J. Bailey
G. A. Rutter
Publication date
01-01-2007
Publisher
Springer-Verlag
Published in
Diabetologia / Issue 1/2007
Print ISSN: 0012-186X
Electronic ISSN: 1432-0428
DOI
https://doi.org/10.1007/s00125-006-0473-3

Other articles of this Issue 1/2007

Diabetologia 1/2007 Go to the issue
Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

Live: Thursday 27th June 2024, 18:00-19:30 (CEST)

WHO estimates that half of all patients worldwide are non-adherent to their prescribed medication. The consequences of poor adherence can be catastrophic, on both the individual and population level.

Join our expert panel to discover why you need to understand the drivers of non-adherence in your patients, and how you can optimize medication adherence in your clinics to drastically improve patient outcomes.

Prof. Kevin Dolgin
Prof. Florian Limbourg
Prof. Anoop Chauhan
Developed by: Springer Medicine
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine