Skip to main content
Top
Published in: Diabetologia 4/2008

01-04-2008 | Article

Age-related insulin resistance in hypothalamus and peripheral tissues of orexin knockout mice

Authors: H. Tsuneki, S. Murata, Y. Anzawa, Y. Soeda, E. Tokai, T. Wada, I. Kimura, M. Yanagisawa, T. Sakurai, T. Sasaoka

Published in: Diabetologia | Issue 4/2008

Login to get access

Abstract

Aims/hypothesis

Orexin/hypocretin is a hypothalamic neuropeptide that regulates motivated behaviours, such as feeding and arousal, and, importantly, is also involved in energy homeostasis. The aim of this study was to reveal the role of orexin in the regulation of insulin sensitivity for glucose metabolism.

Methods

Orexin knockout mice fasted overnight underwent oral glucose tolerance testing and insulin tolerance testing. The impact of orexin deficiency on insulin signalling was studied by Western blotting to measure levels of Akt phosphorylation and its upstream and downstream molecules in the hypothalamus, muscle and liver in orexin knockout mice.

Results

We found that orexin deficiency caused the age-related development of impaired glucose tolerance and insulin resistance in both male mice without obesity and female mice with mild obesity, fed a normal chow diet. When maintained on a high-fat diet, these abnormalities became more pronounced exclusively in female orexin knockout mice that developed severe obesity. Insulin signalling through Akt was disrupted in peripheral tissues of middle-aged (9-month-old) but not young adult (2-to-3-month-old) orexin knockout mice fed a normal chow diet. Moreover, basal levels of hypothalamic Akt phosphorylation were abnormally elevated in orexin knockout mice at every age studied, and insulin stimulation failed to increase the level of phosphorylation. Similar abnormalities were observed with respect to GSK3β phosphorylation in the hypothalamus and peripheral tissues of middle-aged orexin knockout mice.

Conclusions/interpretation

Our results demonstrate a novel role for orexin in hypothalamic insulin signalling, which is likely to be responsible for preventing the development of peripheral insulin resistance with age.
Appendix
Available only for authorised users
Literature
1.
go back to reference Sakurai T (2007) The neural circuit of orexin (hypocretin): maintaining sleep and wakefulness. Nat Rev Neurosci 8:171–181PubMedCrossRef Sakurai T (2007) The neural circuit of orexin (hypocretin): maintaining sleep and wakefulness. Nat Rev Neurosci 8:171–181PubMedCrossRef
2.
go back to reference de Lecea L, Kilduff TS, Peyron C et al (1998) The hypocretins: hypothalamus-specific peptides with neuroexcitatory activity. Proc Natl Acad Sci U S A 95:322–327PubMedCrossRef de Lecea L, Kilduff TS, Peyron C et al (1998) The hypocretins: hypothalamus-specific peptides with neuroexcitatory activity. Proc Natl Acad Sci U S A 95:322–327PubMedCrossRef
3.
go back to reference Sakurai T, Amemiya A, Ishii M et al (1998) Orexins and orexin receptors: a family of hypothalamic neuropeptides and G protein-coupled receptors that regulate feeding behavior. Cell 92:573–585PubMedCrossRef Sakurai T, Amemiya A, Ishii M et al (1998) Orexins and orexin receptors: a family of hypothalamic neuropeptides and G protein-coupled receptors that regulate feeding behavior. Cell 92:573–585PubMedCrossRef
4.
go back to reference Yamanaka A, Beuckmann CT, Willie JT et al (2003) Hypothalamic orexin neurons regulate arousal according to energy balance in mice. Neuron 38:701–713PubMedCrossRef Yamanaka A, Beuckmann CT, Willie JT et al (2003) Hypothalamic orexin neurons regulate arousal according to energy balance in mice. Neuron 38:701–713PubMedCrossRef
5.
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
6.
go back to reference Burdakov D, Jensen LT, Alexopoulos H et al (2006) Tandem-pore K+ channels mediate inhibition of orexin neurons by glucose. Neuron 50:711–722PubMedCrossRef Burdakov D, Jensen LT, Alexopoulos H et al (2006) Tandem-pore K+ channels mediate inhibition of orexin neurons by glucose. Neuron 50:711–722PubMedCrossRef
7.
go back to reference Moriguchi T, Sakurai T, Nambu T, Yanagisawa M, Goto K (1999) Neurons containing orexin in the lateral hypothalamic area of the adult rat brain are activated by insulin-induced acute hypoglycemia. Neurosci Lett 264:101–104PubMedCrossRef Moriguchi T, Sakurai T, Nambu T, Yanagisawa M, Goto K (1999) Neurons containing orexin in the lateral hypothalamic area of the adult rat brain are activated by insulin-induced acute hypoglycemia. Neurosci Lett 264:101–104PubMedCrossRef
8.
go back to reference Griffond B, Risold PY, Jacquemard C, Colard C, Fellmann D (1999) Insulin-induced hypoglycemia increases preprohypocretin (orexin) mRNA in the rat lateral hypothalamic area. Neurosci Lett 262:77–80PubMedCrossRef Griffond B, Risold PY, Jacquemard C, Colard C, Fellmann D (1999) Insulin-induced hypoglycemia increases preprohypocretin (orexin) mRNA in the rat lateral hypothalamic area. Neurosci Lett 262:77–80PubMedCrossRef
9.
go back to reference VanItallie TB (2006) Sleep and energy balance: interactive homeostatic systems. Metabolism 55:S30–S35PubMedCrossRef VanItallie TB (2006) Sleep and energy balance: interactive homeostatic systems. Metabolism 55:S30–S35PubMedCrossRef
10.
go back to reference Thannickal TC, Moore RY, Nienhuis R et al (2000) Reduced number of hypocretin neurons in human narcolepsy. Neuron 27:469–474PubMedCrossRef Thannickal TC, Moore RY, Nienhuis R et al (2000) Reduced number of hypocretin neurons in human narcolepsy. Neuron 27:469–474PubMedCrossRef
11.
go back to reference Sakurai T (2005) Roles of orexin/hypocretin in regulation of sleep/wakefulness and energy homeostasis. Sleep Med Rev 9:231–241PubMedCrossRef Sakurai T (2005) Roles of orexin/hypocretin in regulation of sleep/wakefulness and energy homeostasis. Sleep Med Rev 9:231–241PubMedCrossRef
12.
go back to reference Honda Y, Doi Y, Ninomiya R, Ninomiya C (1986) Increased frequency of non-insulin-dependent diabetes mellitus among narcoleptic patients. Sleep 9:254–259PubMed Honda Y, Doi Y, Ninomiya R, Ninomiya C (1986) Increased frequency of non-insulin-dependent diabetes mellitus among narcoleptic patients. Sleep 9:254–259PubMed
13.
go back to reference Lammers GJ, Pijl H, Iestra J, Langius JA, Buunk G, Meinders AE (1996) Spontaneous food choice in narcolepsy. Sleep 19:75–76PubMed Lammers GJ, Pijl H, Iestra J, Langius JA, Buunk G, Meinders AE (1996) Spontaneous food choice in narcolepsy. Sleep 19:75–76PubMed
14.
go back to reference Schuld A, Hebebrand J, Geller F, Pollmacher T (2000) Increased body-mass index in patients with narcolepsy. Lancet 355:1274–1275PubMedCrossRef Schuld A, Hebebrand J, Geller F, Pollmacher T (2000) Increased body-mass index in patients with narcolepsy. Lancet 355:1274–1275PubMedCrossRef
15.
go back to reference Nishino S, Ripley B, Overeem S et al (2001) Low cerebrospinal fluid hypocretin (orexin) and altered energy homeostasis in human narcolepsy. Ann Neurol 50:381–388PubMedCrossRef Nishino S, Ripley B, Overeem S et al (2001) Low cerebrospinal fluid hypocretin (orexin) and altered energy homeostasis in human narcolepsy. Ann Neurol 50:381–388PubMedCrossRef
16.
go back to reference Hara J, Beuckmann CT, Nambu T et al (2001) Genetic ablation of orexin neurons in mice results in narcolepsy, hypophagia, and obesity. Neuron 30:345–354PubMedCrossRef Hara J, Beuckmann CT, Nambu T et al (2001) Genetic ablation of orexin neurons in mice results in narcolepsy, hypophagia, and obesity. Neuron 30:345–354PubMedCrossRef
17.
go back to reference Hara J, Yanagisawa M, Sakurai T (2005) Difference in obesity phenotype between orexin-knockout mice and orexin neuron-deficient mice with same genetic background and environmental conditions. Neurosci Lett 380:239–242PubMedCrossRef Hara J, Yanagisawa M, Sakurai T (2005) Difference in obesity phenotype between orexin-knockout mice and orexin neuron-deficient mice with same genetic background and environmental conditions. Neurosci Lett 380:239–242PubMedCrossRef
18.
go back to reference Burdakov D, Alexopoulos H (2005) Metabolic state signalling through central hypocretin/orexin neurons. J Cell Mol Med 9:795–803PubMedCrossRef Burdakov D, Alexopoulos H (2005) Metabolic state signalling through central hypocretin/orexin neurons. J Cell Mol Med 9:795–803PubMedCrossRef
19.
go back to reference Saltiel AR, Kahn CR (2001) Insulin signalling and the regulation of glucose and lipid metabolism. Nature 414:799–806PubMedCrossRef Saltiel AR, Kahn CR (2001) Insulin signalling and the regulation of glucose and lipid metabolism. Nature 414:799–806PubMedCrossRef
20.
go back to reference Taniguchi CM, Emanuelli B, Kahn CR (2006) Critical nodes in signalling pathways: insights into insulin action. Nat Rev Mol Cell Biol 7:85–96PubMedCrossRef Taniguchi CM, Emanuelli B, Kahn CR (2006) Critical nodes in signalling pathways: insights into insulin action. Nat Rev Mol Cell Biol 7:85–96PubMedCrossRef
21.
go back to reference Gerozissis K (2004) Brain insulin and feeding: a bi-directional communication. Eur J Pharmacol 490:59–70PubMedCrossRef Gerozissis K (2004) Brain insulin and feeding: a bi-directional communication. Eur J Pharmacol 490:59–70PubMedCrossRef
22.
go back to reference Plum L, Belgardt BF, Brüning JC (2006) Central insulin action in energy and glucose homeostasis. J Clin Invest 116:1761–1766PubMedCrossRef Plum L, Belgardt BF, Brüning JC (2006) Central insulin action in energy and glucose homeostasis. J Clin Invest 116:1761–1766PubMedCrossRef
23.
go back to reference Prodi E, Obici S (2006) Minireview: the brain as a molecular target for diabetic therapy. Endocrinology 147:2664–2669PubMedCrossRef Prodi E, Obici S (2006) Minireview: the brain as a molecular target for diabetic therapy. Endocrinology 147:2664–2669PubMedCrossRef
24.
go back to reference Tsuneki H, Sugihara Y, Honda R, Wada T, Sasaoka T, Kimura I (2002) Reduction of blood glucose level by orexins in fasting normal and streptozotocin-diabetic mice. Eur J Pharmacol 448:245–252PubMedCrossRef Tsuneki H, Sugihara Y, Honda R, Wada T, Sasaoka T, Kimura I (2002) Reduction of blood glucose level by orexins in fasting normal and streptozotocin-diabetic mice. Eur J Pharmacol 448:245–252PubMedCrossRef
25.
go back to reference Chemelli RM, Willie JT, Sinton CM et al (1999) Narcolepsy in orexin knockout mice: molecular genetics of sleep regulation. Cell 98:437–451PubMedCrossRef Chemelli RM, Willie JT, Sinton CM et al (1999) Narcolepsy in orexin knockout mice: molecular genetics of sleep regulation. Cell 98:437–451PubMedCrossRef
26.
go back to reference Wada T, Naito M, Kenmochi H, Tsuneki H, Sasaoka T (2007) Chronic nicotine exposure enhances insulin-induced mitogenic signaling via up-regulation of α7 nicotinic receptors in isolated rat aortic smooth muscle cells. Endocrinology 148:790–799PubMedCrossRef Wada T, Naito M, Kenmochi H, Tsuneki H, Sasaoka T (2007) Chronic nicotine exposure enhances insulin-induced mitogenic signaling via up-regulation of α7 nicotinic receptors in isolated rat aortic smooth muscle cells. Endocrinology 148:790–799PubMedCrossRef
27.
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
28.
go back to reference Willie JT, Chemelli RM, Sinton CM, Yanagisawa M (2001) To eat or to sleep? Orexin in the regulation of feeding and wakefulness. Annu Rev Neurosci 24:429–458PubMedCrossRef Willie JT, Chemelli RM, Sinton CM, Yanagisawa M (2001) To eat or to sleep? Orexin in the regulation of feeding and wakefulness. Annu Rev Neurosci 24:429–458PubMedCrossRef
30.
go back to reference Spinazzi R, Andreis PG, Rossi GP, Nussdorfer GG (2006) Orexins in the regulation of the hypothalamic–pituitary–adrenal axis. Pharmacol Rev 58:46–57PubMedCrossRef Spinazzi R, Andreis PG, Rossi GP, Nussdorfer GG (2006) Orexins in the regulation of the hypothalamic–pituitary–adrenal axis. Pharmacol Rev 58:46–57PubMedCrossRef
31.
go back to reference Wang J, Osaka T, Inoue S (2001) Energy expenditure by intracerebroventricular administration of orexin to anesthetized rats. Neurosci Lett 315:49–52PubMedCrossRef Wang J, Osaka T, Inoue S (2001) Energy expenditure by intracerebroventricular administration of orexin to anesthetized rats. Neurosci Lett 315:49–52PubMedCrossRef
32.
go back to reference Zhang S, Zeitzer JM, Sakurai T, Nishino S, Mignot E (2007) Sleep/wake fragmentation disrupts metabolism in a mouse model of narcolepsy. J Physiol 581:649–663PubMedCrossRef Zhang S, Zeitzer JM, Sakurai T, Nishino S, Mignot E (2007) Sleep/wake fragmentation disrupts metabolism in a mouse model of narcolepsy. J Physiol 581:649–663PubMedCrossRef
33.
go back to reference Pessin JE, Saltiel AR (2000) Signaling pathways in insulin action: molecular targets of insulin resistance. J Clin Invest 106:165–169PubMedCrossRef Pessin JE, Saltiel AR (2000) Signaling pathways in insulin action: molecular targets of insulin resistance. J Clin Invest 106:165–169PubMedCrossRef
34.
go back to reference Cook SA, Aitman T, Naoumova RP (2005) Therapy insight: heart disease and the insulin-resistant patient. Nat Clin Pract Cardiovasc Med 2:252–260PubMedCrossRef Cook SA, Aitman T, Naoumova RP (2005) Therapy insight: heart disease and the insulin-resistant patient. Nat Clin Pract Cardiovasc Med 2:252–260PubMedCrossRef
35.
go back to reference Fujiki N, Yoshida Y, Zhang S, Sakurai T, Yanagisawa M, Nishino S (2006) Sex difference in body weight gain and leptin signaling in hypocretin/orexin deficient mouse models. Peptides 27:2326–2331PubMedCrossRef Fujiki N, Yoshida Y, Zhang S, Sakurai T, Yanagisawa M, Nishino S (2006) Sex difference in body weight gain and leptin signaling in hypocretin/orexin deficient mouse models. Peptides 27:2326–2331PubMedCrossRef
36.
go back to reference de la Monte SM, Wands JR (2005) Review of insulin and insulin-like growth factor expression, signaling, and malfunction in the central nervous system: relevance to Alzheimer’s disease. J Alzheimers Dis 7:45–61PubMed de la Monte SM, Wands JR (2005) Review of insulin and insulin-like growth factor expression, signaling, and malfunction in the central nervous system: relevance to Alzheimer’s disease. J Alzheimers Dis 7:45–61PubMed
37.
go back to reference Sakurai T (2006) Roles of orexins and orexin receptors in central regulation of feeding behavior and energy homeostasis. CNS Neurol Disord Drug Targets 5:313–325PubMed Sakurai T (2006) Roles of orexins and orexin receptors in central regulation of feeding behavior and energy homeostasis. CNS Neurol Disord Drug Targets 5:313–325PubMed
38.
go back to reference Brüning JC, Gautam D, Burks DJ et al (2000) Role of brain insulin receptor in control of body weight and reproduction. Science 289:2122–2125PubMedCrossRef Brüning JC, Gautam D, Burks DJ et al (2000) Role of brain insulin receptor in control of body weight and reproduction. Science 289:2122–2125PubMedCrossRef
39.
go back to reference Zhao WQ, Chen H, Quon MJ, Alkon DL (2004) Insulin and the insulin receptor in experimental models of learning and memory. Eur J Pharmacol 490:71–81PubMedCrossRef Zhao WQ, Chen H, Quon MJ, Alkon DL (2004) Insulin and the insulin receptor in experimental models of learning and memory. Eur J Pharmacol 490:71–81PubMedCrossRef
40.
go back to reference Kim MS, Pak YK, Jang PG et al (2006) Role of hypothalamic Foxo1 in the regulation of food intake and energy homeostasis. Nat Neurosci 9:901–906PubMedCrossRef Kim MS, Pak YK, Jang PG et al (2006) Role of hypothalamic Foxo1 in the regulation of food intake and energy homeostasis. Nat Neurosci 9:901–906PubMedCrossRef
41.
go back to reference Lee MG, Hassani OK, Jones BE (2005) Discharge of identified orexin/hypocretin neurons across the sleep–waking cycle. J Neurosci 25:6716–6720PubMedCrossRef Lee MG, Hassani OK, Jones BE (2005) Discharge of identified orexin/hypocretin neurons across the sleep–waking cycle. J Neurosci 25:6716–6720PubMedCrossRef
42.
go back to reference Clodfelder-Miller B, De Sarno P, Zmijewska AA, Song L, Jope RS (2005) Physiological and pathological changes in glucose regulate brain Akt and glycogen synthase kinase-3. J Biol Chem 280:39723–39731PubMedCrossRef Clodfelder-Miller B, De Sarno P, Zmijewska AA, Song L, Jope RS (2005) Physiological and pathological changes in glucose regulate brain Akt and glycogen synthase kinase-3. J Biol Chem 280:39723–39731PubMedCrossRef
43.
go back to reference Dummler B, Tschopp O, Hynx D et al (2006) Life with a single isoform of Akt: mice lacking Akt2 and Akt3 are viable but display impaired glucose homeostasis and growth deficiencies. Mol Cell Biol 26:8042–8051PubMedCrossRef Dummler B, Tschopp O, Hynx D et al (2006) Life with a single isoform of Akt: mice lacking Akt2 and Akt3 are viable but display impaired glucose homeostasis and growth deficiencies. Mol Cell Biol 26:8042–8051PubMedCrossRef
44.
go back to reference El-Haschimi K, Pierroz DD, Hileman SM, Bjorbaek C, Flier JS (2000) Two defects contribute to hypothalamic leptin resistance in mice with diet-induced obesity. J Clin Invest 105:1827–1832PubMedCrossRef El-Haschimi K, Pierroz DD, Hileman SM, Bjorbaek C, Flier JS (2000) Two defects contribute to hypothalamic leptin resistance in mice with diet-induced obesity. J Clin Invest 105:1827–1832PubMedCrossRef
45.
46.
go back to reference Arnulf I, Lin L, Zhang J et al (2006) CSF vs serum leptin in narcolepsy: is there an effect of hypocretin deficiency? Sleep 29:1017–1024PubMed Arnulf I, Lin L, Zhang J et al (2006) CSF vs serum leptin in narcolepsy: is there an effect of hypocretin deficiency? Sleep 29:1017–1024PubMed
47.
go back to reference Yamamoto Y, Ueta Y, Date Y et al (1999) Down regulation of the prepro-orexin gene expression in genetically obese mice. Mol Brain Res 65:14–22PubMedCrossRef Yamamoto Y, Ueta Y, Date Y et al (1999) Down regulation of the prepro-orexin gene expression in genetically obese mice. Mol Brain Res 65:14–22PubMedCrossRef
Metadata
Title
Age-related insulin resistance in hypothalamus and peripheral tissues of orexin knockout mice
Authors
H. Tsuneki
S. Murata
Y. Anzawa
Y. Soeda
E. Tokai
T. Wada
I. Kimura
M. Yanagisawa
T. Sakurai
T. Sasaoka
Publication date
01-04-2008
Publisher
Springer-Verlag
Published in
Diabetologia / Issue 4/2008
Print ISSN: 0012-186X
Electronic ISSN: 1432-0428
DOI
https://doi.org/10.1007/s00125-008-0929-8

Other articles of this Issue 4/2008

Diabetologia 4/2008 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

Highlights from the ACC 2024 Congress

Year in Review: Pediatric cardiology

Watch Dr. Anne Marie Valente present the last year's highlights in pediatric and congenital heart disease in the official ACC.24 Year in Review session.

Year in Review: Pulmonary vascular disease

The last year's highlights in pulmonary vascular disease are presented by Dr. Jane Leopold in this official video from ACC.24.

Year in Review: Valvular heart disease

Watch Prof. William Zoghbi present the last year's highlights in valvular heart disease from the official ACC.24 Year in Review session.

Year in Review: Heart failure and cardiomyopathies

Watch this official video from ACC.24. Dr. Biykem Bozkurt discusses last year's major advances in heart failure and cardiomyopathies.