Skip to main content

Sensing of Glucose in the Brain

  • Chapter
  • First Online:
Appetite Control

Part of the book series: Handbook of Experimental Pharmacology ((HEP,volume 209))

Abstract

The brain, and in particular the hypothalamus and brainstem, have been recognized for decades as important centers for the homeostatic control of feeding, energy expenditure, and glucose homeostasis. These structures contain neurons and neuronal circuits that may be directly or indirectly activated or inhibited by glucose, lipids, or amino acids. The detection by neurons of these nutrient cues may become deregulated, and possibly cause metabolic diseases such as obesity and diabetes. Thus, there is a major interest in identifying these neurons, how they respond to nutrients, the neuronal circuits they form, and the physiological function they control. Here I will review some aspects of glucose sensing by the brain. The brain is responsive to both hyperglycemia and hypoglycemia, and the glucose sensing cells involved are distributed in several anatomical sites that are connected to each other. These eventually control the activity of the sympathetic or parasympathetic nervous system, which regulates the function of peripheral organs such as liver, white and brown fat, muscle, and pancreatic islets alpha and beta cells. There is now evidence for an extreme diversity in the sensing mechanisms used, and these will be reviewed.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 259.00
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 329.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 329.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

References

  • Adachi A, Shimizu N, Oomura Y, Kobashi M (1984) Convergence of hepatoportal glucose-sensitive afferents signal to glucose sensitive units within the nucleus of the solitary tract. Neurosci Lett 46:215–218

    Article  PubMed  CAS  Google Scholar 

  • Adamantidis A, de Lecea L (2008) Sleep and metabolism: shared circuits, new connections. Trends Endocrinol Metab 19:362–370

    Article  PubMed  CAS  Google Scholar 

  • 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 544:429–445

    Article  PubMed  CAS  Google Scholar 

  • Akmayev IG, Fidelina OV (1974) Morphological aspects of the hypothalamic-hypophyseal system. V. The tanycytes: their relation to the hypophyseal adrenocorticotrophic function. An enzyme-histochemical study. Cell Tissue Res 152:403–410

    Article  PubMed  CAS  Google Scholar 

  • Anand BK, Chhina GS, Sharma KN, Dua S, Singh B (1964) Activity of single neurons in the hypothalamic feeding centers: effect of glucose. Am J Physiol 207:1146–1154

    PubMed  CAS  Google Scholar 

  • Archer SL, Wu XC, Thebaud B, Moudgil R, Hashimoto K, Michelakis ED (2004) O2 sensing in the human ductus arteriosus: redox-sensitive K+ channels are regulated by mitochondria-derived hydrogen peroxide. Biol Chem 385:205–216

    Article  PubMed  CAS  Google Scholar 

  • Arluison M, Quignon M, Nguyen P, Thorens B, Leloup C, Penicaud L (2004a) Distribution and anatomical localization of the glucose transporter 2 (GLUT2) in the adult rat brain–an immunohistochemical study. J Chem Neuroanat 28:117–136

    Article  PubMed  CAS  Google Scholar 

  • Arluison M, Quignon M, Thorens B, Leloup C, Penicaud L (2004b) Immunocytochemical localization of the glucose transporter 2 (GLUT2) in the adult rat brain. II. Electron microscopic study. J Chem Neuroanat 28:137–146

    Article  PubMed  CAS  Google Scholar 

  • Bady I, Marty N, Dallaporta M, Emery M, Gyger J, Tarussio D, Foretz M, Thorens B (2006) Evidence from glut2-null mice that glucose is a critical physiological regulator of feeding. Diabetes 55:988–995

    Article  PubMed  CAS  Google Scholar 

  • Balfour RH, Hansen AM, Trapp S (2006) Neuronal responses to transient hypoglycaemia in the dorsal vagal complex of the rat brainstem. J Physiol 570:469–484

    Article  PubMed  CAS  Google Scholar 

  • Balfour RH, Trapp S (2007) Ionic currents underlying the response of rat dorsal vagal neurones to hypoglycaemia and chemical anoxia. J Physiol 579:691–702

    Article  PubMed  CAS  Google Scholar 

  • Bernard C (1849) Chiens rendus diabétiques. CR Soc Biol 1:60

    Google Scholar 

  • Berthoud H-R (2002) Multiple neural systems controlling food intake and body weight. Neurosci Biobehav Rev 26:393–428

    Article  PubMed  Google Scholar 

  • Berthoud HR, Mogenson GJ (1977) Ingestive behavior after intracerebral and intracerebroventricular infusions of glucose and 2-deoxy-D-glucose. Am J Physiol 233:R127–R133

    PubMed  CAS  Google Scholar 

  • Biggers DW, Myers SR, Neal D, Stinson R, Cooper NB, Jaspan JB, Williams PE, Cherrington AD, Frizzell RT (1989) Role of brain in counterregulation of insulin-induced hypoglycemia in dogs. Diabetes 38:7–16

    Article  PubMed  CAS  Google Scholar 

  • Borg MA, Sherwin RS, Borg WP, Tamborlane WV, Shulman GI (1997) Local ventromedial hypothalamus glucose perfusion blocks counterregulation during systemic hypoglycemia in awake rats. J Clin Invest 99:361–365

    Article  PubMed  CAS  Google Scholar 

  • Borg WP, Sherwin RS, During MJ, Borg MA, Shulman GI (1995) Local ventromedial hypothalamus glucopenia triggers counterregulatory hormone release. Diabetes 44:180–184

    Article  PubMed  CAS  Google Scholar 

  • Bray GA (1985) Autonomic and endocrine factors in the regulation of food intake. Brain Res Bull 14:505–510

    Article  PubMed  CAS  Google Scholar 

  • Briski KP, Patil GD (2005) Induction of Fos immunoreactivity labeling in rat forebrain metabolic loci by caudal fourth ventricular infusion of the monocarboxylate transporter inhibitor, alpha-cyano-4-hydroxycinnamic acid. Neuroendocrinology 82:49–57

    Article  PubMed  CAS  Google Scholar 

  • Burcelin R, Thorens B (2001) Evidence that extrapancreatic GLUT2-dependent glucose sensors control glucagon secretion. Diabetes 50:1282–1289

    Article  PubMed  CAS  Google Scholar 

  • Burdakov D, Jensen LT, Alexopoulos H, Williams RH, Fearon IM, O’Kelly I, Gerasimenko O, Fugger L, Verkhratsky A (2006) Tandem-pore K+ channels mediate inhibition of orexin neurons by glucose. Neuron 50:711–722

    Article  PubMed  CAS  Google Scholar 

  • Canabal DD, Song Z, Potian JG, Beuve A, McArdle JJ, Routh VH (2007) Glucose, insulin, and leptin signaling pathways modulate nitric oxide synthesis in glucose-inhibited neurons in the ventromedial hypothalamus. Am J Physiol Regul Integr Comp Physiol 292:R1418–R1428

    Article  PubMed  CAS  Google Scholar 

  • Canteras NS, Simerly RB, Swanson LW (1994) Organization of projections from the ventromedial nucleus of the hypothalamus: a Phaseolus vulgaris-leucoagglutinin study in the rat. J Comp Neurol 348:41–79

    Article  PubMed  CAS  Google Scholar 

  • Castillo J, Crespo D, Capilla E, Diaz M, Chauvigne F, Cerda J, Planas JV (2009) Evolutionary structural and functional conservation of an ortholog of the GLUT2 glucose transporter gene (SLC2A2) in zebrafish. Am J Physiol Regul Integr Comp Physiol 297:R1570–R1581

    Article  PubMed  CAS  Google Scholar 

  • Claret M, Smith MA, Batterham RL, Selman C, Choudhury AI, Fryer LG, Clements M, Al-Qassab H, Heffron H, Xu AW, Speakman JR, Barsh GS, Viollet B, Vaulont S, Ashford ML, Carling D, Withers DJ (2007) AMPK is essential for energy homeostasis regulation and glucose sensing by POMC and AgRP neurons. J Clin Invest 117:2325–2336

    Article  PubMed  CAS  Google Scholar 

  • Coppola A, Liu ZW, Andrews ZB, Paradis E, Roy MC, Friedman JM, Ricquier D, Richard D, Horvath TL, Gao XB, Diano S (2007) A central thermogenic-like mechanism in feeding regulation: an interplay between arcuate nucleus T3 and UCP2. Cell Metab 5:21–33

    Article  PubMed  CAS  Google Scholar 

  • Cummings DE, Schwartz MW (2000) Melanocortins and body weight: a tale of two receptors. Nat Genet 26:8–9

    Article  PubMed  CAS  Google Scholar 

  • Dallaporta M, Himmi T, Perrin J, Orsini J-C (1999) A solitary tract nucleus sensitivity to moderate changes in glucose level. Neuroreport 10:1–4

    Article  Google Scholar 

  • Dhillon H, Zigman JM, Ye C, Lee CE, McGovern RA, Tang V, Kenny CD, Christiansen LM, White RD, Edelstein EA, Coppari R, Balthasar N, Cowley MA, Chua S Jr, Elmquist JK, Lowell BB (2006) Leptin directly activates SF1 neurons in the VMH, and this action by leptin is required for normal body-weight homeostasis. Neuron 49:191–203

    Article  PubMed  CAS  Google Scholar 

  • Diez-Sampedro A, Hirayama BA, Osswald C, Gorboulev V, Baumgarten K, Volk C, Wright EM, Koepsell H (2003) A glucose sensor hiding in a family of transporters. Proc Natl Acad Sci USA 100:11753–11758

    Article  PubMed  CAS  Google Scholar 

  • Dunn-Meynell AA, Rawson NE, Levin BE (1998) Distribution and phenotype of neurons containing the ATP-sensitive K+ channel in rat brain. Brain Res 814:41–54

    Article  PubMed  CAS  Google Scholar 

  • 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 51:2056–2065

    Article  PubMed  CAS  Google Scholar 

  • 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 53:2767–2775

    Article  PubMed  CAS  Google Scholar 

  • Fioramonti X, Marsollier N, Song Z, Fakira KA, Patel RM, Brown S, Duparc T, Pica-Mendez A, Sanders NM, Knauf C, Valet P, McCrimmon RJ, Beuve A, Magnan C, Routh VH (2010) Ventromedial hypothalamic nitric oxide production is necessary for hypoglycemia detection and counterregulation. Diabetes 59:519–528

    Article  PubMed  CAS  Google Scholar 

  • Flament-Durand J, Brion JP (1985) Tanycytes: morphology and functions: a review. Int Rev Cytol 96:121–155

    Article  PubMed  CAS  Google Scholar 

  • Fraley GS, Ritter S (2003) Immunolesion of norepinephrine and epinephrine afferents to medial hypothalamus alters basal and 2-deoxy-D-glucose-induced neuropeptide Y and agouti-gene-related protein messenger ribonucleic acid expression in the arcuate nucleus. Endocrinology 411:75–83

    Article  CAS  Google Scholar 

  • Freinkel N, Metzger BE, Harris E, Robinson S, Mager M (1972) The hypothermia of hypoglycemia. Studies with 2-deoxy-D-glucose in normal human subjects and mice. N Engl J Med 287:841–845

    Article  PubMed  CAS  Google Scholar 

  • Frizzell RT, Jones EM, Davis SN, Biggers DW, Myers SR, Connolly CC, Neal DW, Jaspan JB, Cherrington AD (1993) Counterregulation during hypoglycemia is directed by widespread brain regions. Diabetes 42:1253–1261

    Article  PubMed  CAS  Google Scholar 

  • Gale SM, Castracane VD, Mantzoros CS (2004) Energy homeostasis, obesity and eating disorders: recent advances in endocrinology. J Nutr 134:295–298

    PubMed  CAS  Google Scholar 

  • Garcia Mde L, Millan C, Balmaceda-Aguilera C, Castro T, Pastor P, Montecinos H, Reinicke K, Zuniga F, Vera JC, Onate SA, Nualart F (2003) Hypothalamic ependymal-glial cells express the glucose transporter GLUT2, a protein involved in glucose sensing. J Neurochem 86:709–724

    Article  CAS  Google Scholar 

  • Gautron L, Elmquist JK (2011) Sixteen years and counting: an update on leptin in energy balance. J Clin Invest 121:2087–2093

    Article  PubMed  CAS  Google Scholar 

  • Gonzalez JA, Jensen LT, Doyle SE, Miranda-Anaya M, Menaker M, Fugger L, Bayliss DA, Burdakov D (2009) Deletion of TASK1 and TASK3 channels disrupts intrinsic excitability but does not abolish glucose or pH responses of orexin/hypocretin neurons. Eur J Neurosci 30:57–64

    Article  PubMed  CAS  Google Scholar 

  • Gonzalez JA, Jensen LT, Fugger L, Burdakov D (2008) Metabolism-independent sugar sensing in central orexin neurons. Diabetes 57:2569–2576

    Article  PubMed  CAS  Google Scholar 

  • Guillod-Maximin E, Lorsignol A, Alquier T, Penicaud L (2004) Acute intracarotid glucose injection towards the brain induces specific c-fos activation in hypothalamic nuclei: involvement of astrocytes in cerebral glucose-sensing in rats. J Neuroendocrinol 16:464–471

    Article  PubMed  CAS  Google Scholar 

  • Hoebel BG (1965) Hypothalamic lesions by electrocauterization: disinhibition of feeding and self-stimulation. Science 149:452–453

    Article  PubMed  CAS  Google Scholar 

  • Ibrahim N, Bosch MA, Smart JL, Qiu J, Rubinstein M, Ronnekleiv OK, Low MJ, Kelly MJ (2003) Hypothalamic proopiomelanocortin neurons are glucose responsive and express K(ATP) channels. Endocrinology 144:1331–1340

    Article  PubMed  CAS  Google Scholar 

  • Jang HJ, Kokrashvili Z, Theodorakis MJ, Carlson OD, Kim BJ, Zhou J, Kim HH, Xu X, Chan SL, Juhaszova M, Bernier M, Mosinger B, Margolskee RF, Egan JM (2007) Gut-expressed gustducin and taste receptors regulate secretion of glucagon-like peptide-1. Proc Natl Acad Sci USA 104:15069–15074

    Article  PubMed  CAS  Google Scholar 

  • Kang L, Dunn-Meynell AA, Routh VH, Gaspers LD, Nagata Y, Nishimura T, Eiki J, Zhang BB, Levin BE (2006) Glucokinase is a critical regulator of ventromedial hypothalamic neuronal glucosensing. Diabetes 55:412–420

    Article  PubMed  CAS  Google Scholar 

  • Kang L, Routh VH, Kuzhikandathil EV, Gaspers LD, Levin BE (2004) Physiological and molecular characteristics of rat hypothalamic ventromedial nucleus glucosensing neurons. Diabetes 53:549–559

    Article  PubMed  CAS  Google Scholar 

  • Karnani M, Burdakov D (2011) Multiple hypothalamic circuits sense and regulate glucose levels. Am J Physiol Regul Integr Comp Physiol 300:R47–R55

    Article  PubMed  CAS  Google Scholar 

  • King BM (2006) The rise, fall, and resurrection of the ventromedial hypothalamus in the regulation of feeding behavior and body weight. Physiol Behav 87:221–244

    Article  PubMed  CAS  Google Scholar 

  • Kong D, Vong L, Parton LE, Ye C, Tong Q, Hu X, Choi B, Bruning JC, Lowell BB (2010) Glucose stimulation of hypothalamic MCH neurons involves K(ATP) channels, is modulated by UCP2, and regulates peripheral glucose homeostasis. Cell Metab 12:545–552

    Article  PubMed  CAS  Google Scholar 

  • Kozlowski GP, Coates PW (1985) Ependymoneuronal specializations between LHRH fibers and cells of the cerebroventricular system. Cell Tissue Res 242:301–311

    Article  PubMed  CAS  Google Scholar 

  • Kraft R, Grimm C, Grosse K, Hoffmann A, Sauerbruch S, Kettenmann H, Schultz G, Harteneck C (2004) Hydrogen peroxide and ADP-ribose induce TRPM2-mediated calcium influx and cation currents in microglia. Am J Physiol Cell Physiol 286:C129–C137

    Article  PubMed  CAS  Google Scholar 

  • Lam TK, Gutierrez-Juarez R, Pocai A, Rossetti L (2005) Regulation of blood glucose by hypothalamic pyruvate metabolism. Science 309:943–947

    Article  PubMed  CAS  Google Scholar 

  • Lee K, Li B, Xi X, Suh Y, Martin RJ (2005) Role of neuronal energy status in the regulation of adenosine 5′-monophosphate-activated protein kinase, orexigenic neuropeptides expression, and feeding behavior. Endocrinology 146:3–10

    Article  PubMed  CAS  Google Scholar 

  • Leloup C, Magnan C, Benani A, Bonnet E, Alquier T, Offer G, Carriere A, Periquet A, Fernandez Y, Ktorza A, Casteilla L, Penicaud L (2006) Mitochondrial reactive oxygen species are required for hypothalamic glucose sensing. Diabetes 55:2084–2090

    Article  PubMed  CAS  Google Scholar 

  • Lemon CH, Margolskee RF (2009) Contribution of the T1r3 taste receptor to the response properties of central gustatory neurons. J Neurophysiol 101:2459–2471

    Article  PubMed  CAS  Google Scholar 

  • Li B, Xi X, Roane DS, Ryan DH, Martin RJ (2003) Distribution of glucokinase, glucose transporter GLUT2, sulfonylurea receptor-1, glucagon-like peptide-1 receptor and neuropeptide Y messenger RNAs in rat brain by quantitative real time RT-PCR. Brain Res Mol Brain Res 113:139–142

    Article  PubMed  CAS  Google Scholar 

  • Magistretti PJ, Pellerin L, Rothman DL, Shulman RG (1999) Energy on demand. Science 283:496–497

    Article  PubMed  CAS  Google Scholar 

  • Marshall NB, Mayer J (1956) Specificity of gold thioglucose for ventromedial hypothalamic lesions and hyperphagia. Nature 178:1399–1400

    Article  PubMed  CAS  Google Scholar 

  • Marty N, Dallaporta M, Foretz M, Emery M, Tarussio D, Bady I, Binnert C, Beermann F, Thorens B (2005) Regulation of glucagon secretion by glucose transporter type 2 (glut2) and astrocyte-dependent glucose sensors. J Clin Invest 115:3545–3553

    Article  PubMed  CAS  Google Scholar 

  • Marty N, Dallaporta M, Thorens B (2007a) Brain glucose sensing, counterregulation and feeding behavior. Physiology (Bethesda) 22:241–251

    Article  CAS  Google Scholar 

  • Marty N, Dallaporta M, Thorens B (2007b) Brain glucose sensing, counterregulation, and energy homeostasis. Physiology (Bethesda) 22:241–251

    Article  CAS  Google Scholar 

  • Mayer J (1953) Glucostatic mechanism of regulation of food intake. N Engl J Med 249:13–16

    Article  PubMed  CAS  Google Scholar 

  • Mayer J, Thomas DW (1967) Regulation of food intake and obesity. Science 156:328–337

    Article  PubMed  CAS  Google Scholar 

  • Millan C, Martinez F, Cortes-Campos C, Lizama I, Yanez MJ, Llanos P, Reinicke K, Rodriguez F, Peruzzo B, Nualart F, Garcia MA (2010) Glial glucokinase expression in adult and post-natal development of the hypothalamic region. ASN Neuro 2:e00035

    Article  PubMed  CAS  Google Scholar 

  • Minokoshi Y, Alquier T, Furukawa N, Kim YB, Lee A, Xue B, Mu J, Foufelle F, Ferre P, Birnbaum MJ, Stuck BJ, Kahn BB (2004) AMP-kinase regulates food intake by responding to hormonal and nutrient signals in the hypothalamus. Nature 428:569–574

    Article  PubMed  CAS  Google Scholar 

  • Miselis RR, Epstein AN (1975) Feeding induced by intracerebroventricular 2-deoxy-D-glucose in the rat. Am J Physiol 229:1438–1447

    PubMed  CAS  Google Scholar 

  • Mizuno Y, Oomura Y (1984) Glucose responding neurons in the nucleus tractus solitarius of the rat: in vitro studies. Brain Res 307:109–116

    Article  PubMed  CAS  Google Scholar 

  • Mounien L, Marty N, Tarussio D, Metref S, Genoux D, Preitner F, Foretz M, Thorens B (2010) Glut2-dependent glucose-sensing controls thermoregulation by enhancing the leptin sensitivity of NPY and POMC neurons. FASEB J 24:1747–1758

    Article  PubMed  CAS  Google Scholar 

  • Murphy BA, Fakira KA, Song Z, Beuve A, Routh VH (2009) AMP-activated protein kinase and nitric oxide regulate the glucose sensitivity of ventromedial hypothalamic glucose-inhibited neurons. Am J Physiol Cell Physiol 297:C750–C758

    Article  PubMed  CAS  Google Scholar 

  • O’Malley D, Reimann F, Simpson AK, Gribble FM (2006) Sodium-coupled glucose cotransporters contribute to hypothalamic glucose sensing. Diabetes 55:3381–3386

    Article  PubMed  CAS  Google Scholar 

  • Oomura Y, Yoshimatsu H (1984) Neural network of glucose monitoring system. J Auton Nerv Syst 10:359–372

    Article  PubMed  CAS  Google Scholar 

  • Pan Y, Weng J, Cao Y, Bhosle RC, Zhou M (2008) Functional coupling between the Kv1.1 channel and aldoketoreductase Kvbeta1. J Biol Chem 283:8634–8642

    Article  PubMed  CAS  Google Scholar 

  • Parton LE, Ye CP, Coppari R, Enriori PJ, Choi B, Zhang CY, Xu C, Vianna CR, Balthasar N, Lee CE, Elmquist JK, Cowley MA, Lowell BB (2007) Glucose sensing by POMC neurons regulates glucose homeostasis and is impaired in obesity. Nature 449:228–232

    Article  PubMed  CAS  Google Scholar 

  • Patil GD, Briski KP (2005a) Lactate is a critical “sensed” variable in caudal hindbrain monitoring of CNS metabolic stasis. Am J Physiol Regul Integr Comp Physiol 289:R1777–R1786

    Article  PubMed  CAS  Google Scholar 

  • Patil GD, Briski KP (2005b) Transcriptional activation of nucleus tractus solitarii/area postrema catecholaminergic neurons by pharmacological inhibition of caudal hindbrain monocarboxylate transporter function. Neuroendocrinology 81:96–102

    Article  PubMed  CAS  Google Scholar 

  • Pellerin L, Bouzier-Sore AK, Aubert A, Serres S, Merle M, Costalat R, Magistretti PJ (2007) Activity-dependent regulation of energy metabolism by astrocytes: an update. Glia 55:1251–1262

    Article  PubMed  Google Scholar 

  • Penicaud L, Leloup C, Fioramonti X, Lorsignol A, Benani A (2006) Brain glucose sensing: a subtle mechanism. Curr Opin Clin Nutr Metab Care 9:458–462

    Article  PubMed  CAS  Google Scholar 

  • Pi J, Bai Y, Daniel KW, Liu D, Lyght O, Edelstein D, Brownlee M, Corkey BE, Collins S (2009) Persistent oxidative stress due to absence of uncoupling protein 2 associated with impaired pancreatic beta-cell function. Endocrinology 150:3040–3048

    Article  PubMed  CAS  Google Scholar 

  • Pi J, Bai Y, Zhang Q, Wong V, Floering LM, Daniel K, Reece JM, Deeney JT, Andersen ME, Corkey BE, Collins S (2007) Reactive oxygen species as a signal in glucose-stimulated insulin secretion. Diabetes 56:1783–1791

    Article  PubMed  CAS  Google Scholar 

  • Polakof S, Miguez JM, Moon TW, Soengas JL (2007) Evidence for the presence of a glucosensor in hypothalamus, hindbrain, and Brockmann bodies of rainbow trout. Am J Physiol Regul Integr Comp Physiol 292:R1657–R1666

    Article  PubMed  CAS  Google Scholar 

  • Ren X, Zhou L, Terwilliger R, Newton SS, de Araujo IE (2009) Sweet taste signaling functions as a hypothalamic glucose sensor. Front Integr Neurosci 3:12

    Article  PubMed  CAS  Google Scholar 

  • Rhee SG (2006) Cell signaling. H2O2, a necessary evil for cell signaling. Science 312:1882–1883

    Article  PubMed  Google Scholar 

  • Ritter RC, Slusser PG, Stone S (1981) Glucoreceptors controlling feeding and blood glucose: location in the hindbrain. Science 213:451–453

    Article  PubMed  CAS  Google Scholar 

  • Ritter S, Bugarith K, Dinh TT (2001) Immunotoxic destruction of distinct catecholamine subgroups produces selective impairment of glucoregulatory responses and neuronal activation. J Comp Neurol 43:197–216

    Article  Google Scholar 

  • Ritter S, Dinh TT, Zhang Y (2000) Localization of hindbrain glucoreceptive sites controlling food intake and blood glucose. Brain Res 856:37–47

    Article  PubMed  CAS  Google Scholar 

  • Roncero I, Alvarez E, Chowen JA, Sanz C, Rabano A, Vazquez P, Blazquez E (2004) Expression of glucose transporter isoform GLUT-2 and glucokinase genes in human brain. J Neurochem 88:1203–1210

    Article  PubMed  CAS  Google Scholar 

  • Routh VH (2002) Glucose-sensing neurons: are they physiologically relevant ? Physiol Behav 76:403–413

    Article  PubMed  CAS  Google Scholar 

  • Sakurai T (2007) The neural circuit of orexin (hypocretin): maintaining sleep and wakefulness. Nat Rev Neurosci 8:171–181

    Article  PubMed  CAS  Google Scholar 

  • Schwartz GJ (2000) The role of gastrointestinal vagal afferents in the control of food intake: current prospects. Nutrition 16:866–873

    Article  PubMed  CAS  Google Scholar 

  • Schwartz MW, Woods SC, Porte D, Seeley RJ, Baskin DG (2000) Central nervous system control of food intake. Nature 404:661–671

    PubMed  CAS  Google Scholar 

  • Silver IA, Erecinska M (1998) Glucose-induced intracellular ion changes in sugar-sensitive hypothalamic neurons. J Neurophysiol 79:1733–1745

    PubMed  CAS  Google Scholar 

  • Steinert RE, Gerspach AC, Gutmann H, Asarian L, Drewe J, Beglinger C (2011) The functional involvement of gut-expressed sweet taste receptors in glucose-stimulated secretion of glucagon-like peptide-1 (GLP-1) and peptide YY (PYY). Clin Nutr 30:524–532

    Article  PubMed  CAS  Google Scholar 

  • Tabet F, Savoia C, Schiffrin EL, Touyz RM (2004) Differential calcium regulation by hydrogen peroxide and superoxide in vascular smooth muscle cells from spontaneously hypertensive rats. J Cardiovasc Pharmacol 44:200–208

    Article  PubMed  CAS  Google Scholar 

  • Thorens B (2003) A gene knockout approach in mice to identify glucose sensors controlling glucose homeostasis. Pflügers Arch – Eur J Physiol 445:482–490

    CAS  Google Scholar 

  • Thorens B, Larsen PJ (2004) Gut-derived signaling molecules and vagal afferents in the control of glucose and energy homeostasis. Curr Opin Clin Nutr Metab Care 7:471–478

    Article  PubMed  CAS  Google Scholar 

  • Todt I, Ngezahayo A, Ernst A, Kolb HA (2001) Hydrogen peroxide inhibits gap junctional coupling and modulates intracellular free calcium in cochlear Hensen cells. J Membr Biol 181:107–114

    PubMed  CAS  Google Scholar 

  • Tong Q, Ye C, McCrimmon RJ, Dhillon H, Choi B, Kramer MD, Yu J, Yang Z, Christiansen LM, Lee CE, Choi CS, Zigman JM, Shulman GI, Sherwin RS, Elmquist JK, Lowell BB (2007) Synaptic glutamate release by ventromedial hypothalamic neurons is part of the neurocircuitry that prevents hypoglycemia. Cell Metab 5:383–393

    Article  PubMed  CAS  Google Scholar 

  • Tsujii S, Bray GA (1990) Effects of glucose, 2-deoxyglucose, phlorizin, and insulin on food intake of lean and fatty rats. Am J Physiol 258:E476–E481

    PubMed  CAS  Google Scholar 

  • Voets T, Talavera K, Owsianik G, Nilius B (2005) Sensing with TRP channels. Nat Chem Biol 1:85–92

    Article  PubMed  CAS  Google Scholar 

  • Wang R, Liu X, Hentges ST, Dunn-Meynell AA, Levin BE, Wang W, Routh VH (2004) The regulation of glucose-excited neurons in the hypothalamic arcuate nucleus by glucose and feeding-relevant peptides. Diabetes 53:1959–1965

    Article  PubMed  CAS  Google Scholar 

  • Woods SC, Seeley RJ, Porte D, Schwartz MW (1998) Signals that regulate food intake and energy homeostasis. Science 280:1378–1383

    Article  PubMed  CAS  Google Scholar 

  • Yang X-J, Kow L-M, Funabashi T, Mobbs CV (1999) Hypothalamic glucose sensor. Similarities to and differences from pancreatic b cell mechanisms. Diabetes 48:1763–1772

    Article  PubMed  CAS  Google Scholar 

  • Yang XJ, Low LM, Pfaff DW, Mobbs CV (2004) Metabolic pathways that mediate inhibition of hypothalamic neurons by glucose. Diabetes 53:67–73

    Article  PubMed  CAS  Google Scholar 

  • Yettefti K, Orsini J-C, Perrin J (1997) Characteristics of glycemia-sensitive neurons in the nucleus tractus solitarii: possible involvement in nutritional regulation. Physiol Behav 61:93–100

    Article  PubMed  CAS  Google Scholar 

  • Yettefti K, Orsini JC, El Ouazzani T, Himmi T, Boyer A, Perrin J (1995) Sensitivity of nucleus tractus solitarius neurons to induced moderate hyperglycemia, with special reference to catecholaminergic regions. J Auton Nerv Syst 51:191–197

    Article  PubMed  CAS  Google Scholar 

  • Young JK, Baker JH, Montes MI (2000) The brain response to 2-deoxy glucose is blocked by a glial drug. Pharmacol Biochem Behav 67:233–239

    Article  PubMed  CAS  Google Scholar 

  • Zhang C-Y, Baffy G, Perret P, Krauss S, Peroni O, Grujic D, Hagen T, Vidal-Puig AJ, Boss O, Kim Y-B, Zheng XX, Wheeler MB, Shulman GI, Chan CB, Lowell BB (2001) Uncoupling protein-2 negatively regulates insulin secretion and is a major link between obesity, b cell dysfunction, and type 2 diabetes. Cell 105:745–755

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgements

Work in the author laboratory has been supported by grants from the Swiss National Science Foundation No. 3100A0-113525 and by the National Competence Center in Research “Frontiers in Genetics.”

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Bernard Thorens .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2012 Springer-Verlag Berlin Heidelberg

About this chapter

Cite this chapter

Thorens, B. (2012). Sensing of Glucose in the Brain. In: Joost, HG. (eds) Appetite Control. Handbook of Experimental Pharmacology, vol 209. Springer, Berlin, Heidelberg. https://doi.org/10.1007/978-3-642-24716-3_12

Download citation

Publish with us

Policies and ethics