Glucose-sensing neurons of the hypothalamus

Specialized subgroups of hypothalamic neurons exhibit specific excitatory or inhibitory electrical responses to changes in extracellular levels of glucose. Glucose-excited neurons were traditionally assumed to employ a ‘β-cell’ glucose-sensing strategy, where glucose elevates cytosolic ATP, which closes KATP channels containing Kir6.2 subunits, causing depolarization and increased excitability. Recent findings indicate that although elements of this canonical model are functional in some hypothalamic cells, this pathway is not universally essential for excitation of glucose-sensing neurons by glucose. Thus glucose-induced excitation of arcuate nucleus neurons was recently reported in mice lacking Kir6.2, and no significant increases in cytosolic ATP levels could be detected in hypothalamic neurons after changes in extracellular glucose. Possible alternative glucose-sensing strategies include electrogenic glucose entry, glucose-induced release of glial lactate, and extracellular glucose receptors. Glucose-induced electrical inhibition is much less understood than excitation, and has been proposed to involve reduction in the depolarizing activity of the Na+/K+ pump, or activation of a hyperpolarizing Cl− current. Investigations of neurotransmitter identities of glucose-sensing neurons are beginning to provide detailed information about their physiological roles. In the mouse lateral hypothalamus, orexin/hypocretin neurons (which promote wakefulness, locomotor activity and foraging) are glucose-inhibited, whereas melanin-concentrating hormone neurons (which promote sleep and energy conservation) are glucose-excited. In the hypothalamic arcuate nucleus, excitatory actions of glucose on anorexigenic POMC neurons in mice have been reported, while the appetite-promoting NPY neurons may be directly inhibited by glucose. These results stress the fundamental importance of hypothalamic glucose-sensing neurons in orchestrating sleep-wake cycles, energy expenditure and feeding behaviour.

[1]  Shedding new light on brain metabolism and glial function , 2002, The Journal of physiology.

[2]  S. Goldman,et al.  New roles for astrocytes: Redefining the functional architecture of the brain , 2003, Trends in Neurosciences.

[3]  A. Blanks,et al.  Orexigen-sensitive NPY/AgRP pacemaker neurons in the hypothalamic arcuate nucleus , 2004, Nature Neuroscience.

[4]  C. Saper,et al.  From Lesions to Leptin Hypothalamic Control of Food Intake and Body Weight , 1999, Neuron.

[5]  Robert M Sears,et al.  The Hypothalamic Neuropeptide Melanin-Concentrating Hormone Acts in the Nucleus Accumbens to Modulate Feeding Behavior and Forced-Swim Performance , 2005, The Journal of Neuroscience.

[6]  E. Wright,et al.  A glucose sensor hiding in a family of transporters , 2003, Proceedings of the National Academy of Sciences of the United States of America.

[7]  A. N. van den Pol,et al.  Neurons Containing Hypocretin (Orexin) Project to Multiple Neuronal Systems , 1998, The Journal of Neuroscience.

[8]  F. Ashcroft,et al.  Type 2 diabetes mellitus: not quite exciting enough? , 2004, Human molecular genetics.

[9]  B. Anand,et al.  ACTIVITY OF SINGLE NEURONS IN THE HYPOTHALAMIC FEEDING CENTERS: EFFECT OF GLUCOSE. , 1964, The American journal of physiology.

[10]  V. Routh,et al.  The role of glucosensing neurons in the detection of hypoglycemia. , 2004, Diabetes technology & therapeutics.

[11]  Nurhadi Ibrahim,et al.  Hypothalamic proopiomelanocortin neurons are glucose responsive and express K(ATP) channels. , 2003, Endocrinology.

[12]  V. Routh,et al.  Physiological and molecular characteristics of rat hypothalamic ventromedial nucleus glucosensing neurons. , 2004, Diabetes.

[13]  J. Thevelein,et al.  The eukaryotic plasma membrane as a nutrient-sensing device. , 2004, Trends in biochemical sciences.

[14]  J. Sutcliffe,et al.  The hypocretins: Setting the arousal threshold , 2002, Nature Reviews Neuroscience.

[15]  B. Lowell,et al.  Mice lacking melanin-concentrating hormone are hypophagic and lean , 1998, Nature.

[16]  A. Verkhratsky,et al.  Glial calcium: homeostasis and signaling function. , 1998, Physiological reviews.

[17]  L. Kow,et al.  Brain glucose-sensing mechanisms: ubiquitous silencing by aglycemia vs. hypothalamic neuroendocrine responses. , 2001, American journal of physiology. Endocrinology and metabolism.

[18]  I. Silver,et al.  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 , 1994, The Journal of neuroscience : the official journal of the Society for Neuroscience.

[19]  Pierre J Magistretti,et al.  Neuroenergetics: Calling Upon Astrocytes to Satisfy Hungry Neurons , 2004, The Neuroscientist : a review journal bringing neurobiology, neurology and psychiatry.

[20]  S. Carr,et al.  Orexins and Orexin Receptors: A Family of Hypothalamic Neuropeptides and G Protein-Coupled Receptors that Regulate Feeding Behavior , 1998, Cell.

[21]  V. Routh Glucosensing neurons in the ventromedial hypothalamic nucleus (VMN) and hypoglycemia‐associated autonomic failure (HAAF) , 2003, Diabetes/metabolism research and reviews.

[22]  V. Routh,et al.  Neuronal glucosensing: what do we know after 50 years? , 2004, Diabetes.

[23]  S. Carr,et al.  Orexins and orexin receptors: a family of hypothalamic neuropeptides and G protein-coupled receptors that regulate feeding behavior. , 1998, Cell.

[24]  H. Kennedy,et al.  Glucose generates sub-plasma membrane ATP microdomains in single islet beta-cells. Potential role for strategically located mitochondria. , 1999, The Journal of biological chemistry.

[25]  H. Brooks,et al.  Localization of glucokinase gene expression in the rat brain. , 2000, Diabetes.

[26]  V. Routh,et al.  The regulation of glucose-excited neurons in the hypothalamic arcuate nucleus by glucose and feeding-relevant peptides. , 2004, Diabetes.

[27]  T. Alquier,et al.  Acute Intracarotid Glucose Injection Towards the Brain Induces Specific c‐fos Activation in Hypothalamic Nuclei: Involvement of Astrocytes in Cerebral Glucose‐Sensing in Rats , 2004, Journal of neuroendocrinology.

[28]  W. Samson,et al.  The other side of the orexins: endocrine and metabolic actions. , 2003, American journal of physiology. Endocrinology and metabolism.

[29]  M. Takigawa,et al.  Lowering glucose concentrations increases cytosolic Ca2+ in orexin neurons of the rat lateral hypothalamus , 2001, Neuroscience Letters.

[30]  L. Kow,et al.  Hypothalamic glucose sensor: similarities to and differences from pancreatic beta-cell mechanisms. , 1999, Diabetes.

[31]  D. Spiller,et al.  Orexin a preferentially excites glucose-sensitive neurons in the lateral hypothalamus of the rat in vitro. , 2001, Diabetes.

[32]  E. Maratos-Flier Melanin-Concentrating Hormone , 2006 .

[33]  Takeshi Sakurai,et al.  Hypothalamic Orexin Neurons Regulate Arousal According to Energy Balance in Mice , 2003, Neuron.

[34]  Jochen Roeper,et al.  ATP-sensitive K+ channels in the hypothalamus are essential for the maintenance of glucose homeostasis , 2001, Nature Neuroscience.

[35]  H. Hsiung,et al.  Targeted disruption of the melanin-concentrating hormone receptor-1 results in hyperphagia and resistance to diet-induced obesity. , 2002, Endocrinology.

[36]  C. Saper,et al.  Fos Expression in Orexin Neurons Varies with Behavioral State , 2001, The Journal of Neuroscience.

[37]  室屋 真二 Glucose-sensitive neurons in the rat arcuate nucleus contain neuropeptide Y , 2000 .

[38]  Ming-Fung Wu,et al.  Release of Hypocretin (Orexin) during Waking and Sleep States , 2002, The Journal of Neuroscience.

[39]  J. Vaughan,et al.  The melanin‐concentrating hormone system of the rat brain: An immuno‐ and hybridization histochemical characterization , 1992, The Journal of comparative neurology.

[40]  T. Miyoshi,et al.  Printed in U.S.A. Copyright © 1997 by The Endocrine Society Melanin-Concentrating Hormone Acutely Stimulates Feeding, But Chronic Administration Has No Effect on , 2022 .

[41]  G. Rutter,et al.  Dynamic imaging of free cytosolic ATP concentration during fuel sensing by rat hypothalamic neurones: evidence for ATP‐independent control of ATP‐sensitive K+ channels , 2002, The Journal of physiology.

[42]  M. Low,et al.  The arcuate nucleus as a conduit for diverse signals relevant to energy homeostasis , 2001, International Journal of Obesity.

[43]  M. Sugimori,et al.  Glucose Inhibition of the Glucose-sensitive Neurone in the Rat Lateral Hypothalamus , 1974, Nature.

[44]  P S Kalra,et al.  Interacting appetite-regulating pathways in the hypothalamic regulation of body weight. , 1999, Endocrine reviews.

[45]  D. Burdakov,et al.  Physiological Changes in Glucose Differentially Modulate the Excitability of Hypothalamic Melanin-Concentrating Hormone and Orexin Neurons In Situ , 2005, The Journal of Neuroscience.

[46]  Jon T. Willie,et al.  Narcolepsy in orexin Knockout Mice Molecular Genetics of Sleep Regulation , 1999, Cell.

[47]  S. Gammeltoft,et al.  A role for melanin-concentrating hormone in the central regulation of feeding behaviour , 1996, Nature.

[48]  F E Bloom,et al.  The hypocretins: hypothalamus-specific peptides with neuroexcitatory activity. , 1998, Proceedings of the National Academy of Sciences of the United States of America.

[49]  T. Ono,et al.  Glucose and Osmosensitive Neurones of the Rat Hypothalamus , 1969, Nature.

[50]  Emmanuel Mignot,et al.  The role of hypocretins (orexins) in sleep regulation and narcolepsy. , 2002, Annual review of neuroscience.

[51]  A. Araque,et al.  Dynamic signaling between astrocytes and neurons. , 2001, Annual review of physiology.

[52]  S. Woods,et al.  Central nervous system control of food intake , 2000, Nature.

[53]  V. Routh,et al.  Glucose-sensing neurons Are they physiologically relevant? , 2002, Physiology & Behavior.

[54]  J. Mark Treherne,et al.  Glucose-induced excitation of hypothalamic neurones is mediated by ATP-sensitive K+ channels , 2004, Pflügers Archiv.

[55]  J. McArdle,et al.  Convergence of pre- and postsynaptic influences on glucosensing neurons in the ventromedial hypothalamic nucleus. , 2001, Diabetes.

[56]  V. Routh,et al.  Differential effects of glucose and lactate on glucosensing neurons in the ventromedial hypothalamic nucleus. , 2005, Diabetes.

[57]  G. Rutter,et al.  Glucose-stimulated oscillations in free cytosolic ATP concentration imaged in single islet beta-cells: evidence for a Ca2+-dependent mechanism. , 2002, Diabetes.

[58]  F. Reimann,et al.  A novel glucose-sensing mechanism contributing to glucagon-like peptide-1 secretion from the GLUTag cell line. , 2003, Diabetes.

[59]  L. Elsas,et al.  Glucose transporters. , 1992, Annual review of medicine.

[60]  Jon T. Willie,et al.  Genetic Ablation of Orexin Neurons in Mice Results in Narcolepsy, Hypophagia, and Obesity , 2001, Neuron.

[61]  Michael Esterman,et al.  The Distribution and Mechanism of Action of Ghrelin in the CNS Demonstrates a Novel Hypothalamic Circuit Regulating Energy Homeostasis , 2003, Neuron.

[62]  S. Nicolaidis,et al.  Melanin-concentrating hormone is a potent anorectic peptide regulated by food-deprivation and glucopenia in the rat , 1996, Neuroscience.

[63]  G. Bray,et al.  Effects of glucose, 2-deoxyglucose, phlorizin, and insulin on food intake of lean and fatty rats. , 1990, The American journal of physiology.

[64]  Jon T. Willie,et al.  To eat or to sleep? Orexin in the regulation of feeding and wakefulness. , 2001, Annual review of neuroscience.

[65]  M. Funahashi,et al.  Glucose-responsive neurons in the brainstem. , 1995, Obesity research.

[66]  Linda M Arseneau,et al.  Extracellular glucose in rat ventromedial hypothalamus during acute and recurrent hypoglycemia. , 2003, Diabetes.

[67]  Yue Feng,et al.  Melanin-concentrating hormone 1 receptor-deficient mice are lean, hyperactive, and hyperphagic and have altered metabolism , 2002, Proceedings of the National Academy of Sciences of the United States of America.

[68]  M. Blaustein,et al.  Sodium/calcium exchange: its physiological implications. , 1999, Physiological reviews.

[69]  J. Siegel Hypocretin (orexin): role in normal behavior and neuropathology. , 2004, Annual review of psychology.

[70]  Dan Zhou,et al.  Enhanced running wheel activity of both Mch1r- and Pmch-deficient mice , 2005, Regulatory Peptides.

[71]  M. Low,et al.  Leptin activates anorexigenic POMC neurons through a neural network in the arcuate nucleus , 2001, Nature.

[72]  W F Ganong,et al.  Circumventricular Organs: Definition And Role In The Regulation Of Endocrine And Autonomic Function , 2000, Clinical and experimental pharmacology & physiology.

[73]  Paul Antoine Salin,et al.  A role of melanin-concentrating hormone producing neurons in the central regulation of paradoxical sleep , 2003, BMC Neuroscience.

[74]  L. Pénicaud,et al.  A new ATP-sensitive K+ channel-independent mechanism is involved in glucose-excited neurons of mouse arcuate nucleus. , 2004, Diabetes.

[75]  J. Mayer,et al.  Hyperphagia caused by Cerebral Ventricular Infusion of Phloridzin , 1968, Nature.