Skip to main content
Top
Published in: Diabetologia 10/2014

01-10-2014 | Article

Glucagon regulates orexin A secretion in humans and rodents

Authors: Ayman M. Arafat, Przemysław Kaczmarek, Marek Skrzypski, Ewa Pruszyńska-Oszmałek, Paweł Kołodziejski, Aikaterini Adamidou, Stephan Ruhla, Dawid Szczepankiewicz, Maciej Sassek, Maria Billert, Bertram Wiedenmann, Andreas F. H. Pfeiffer, Krzysztof W. Nowak, Mathias Z. Strowski

Published in: Diabetologia | Issue 10/2014

Login to get access

Abstract

Aims/hypothesis

Orexin A (OXA) modulates food intake, energy expenditure, and lipid and glucose metabolism. OXA regulates the secretion of insulin and glucagon, while glucose regulates OXA release. Here, we evaluate the role of glucagon in regulating OXA release both in vivo and in vitro.

Methods

In a double-blind crossover study, healthy volunteers and type 1 diabetic patients received either intramuscular glucagon or placebo. Patients newly diagnosed with type 2 diabetes underwent hyperinsulinaemic–euglycaemic clamp experiments, and insulin–hypoglycaemia tests were performed on healthy volunteers. The primary endpoint was a change in OXA levels after intramuscular glucagon or placebo administration in healthy participants and patients with type 1 diabetes. Secondary endpoints included changes in OXA in healthy participants during insulin tolerance tests and in patients with type 2 diabetes under hyperinsulinaemic–euglycaemic conditions. Participants and staff conducting examinations and taking measurements were blinded to group assignment. OXA secretion in response to glucagon treatment was assessed in healthy and obese mice, the streptozotocin-induced mouse model of type 1 diabetes, and isolated rat pancreatic islets.

Results

Plasma OXA levels declined in lean volunteers and in type 1 diabetic patients injected with glucagon. OXA levels increased during hyperinsulinaemic hypoglycaemia testing in healthy volunteers and during hyperinsulinaemic euglycaemic conditions in type 2 diabetic patients. Plasma OXA concentrations in healthy lean and obese mice and in a mouse model of type 1 diabetes were lower after glucagon treatment, compared with vehicle control. Glucagon decreased OXA secretion from isolated rat pancreatic islets at both low and high glucose levels. OXA secretion declined in pancreatic islets exposed to diazoxide at high and low glucose levels, and after exposure to an anti-insulin antibody. Glucagon further reduced OXA secretion in islets pretreated with diazoxide or an anti-insulin antibody.

Conclusions/interpretation

Glucagon inhibits OXA secretion in humans and animals, irrespective of changes in glucose or insulin levels. Through modifying OXA secretion, glucagon may influence energy expenditure, body weight, food intake and glucose metabolism.
Appendix
Available only for authorised users
Literature
1.
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
2.
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
3.
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
4.
go back to reference Funato H, Tsai AL, Willie JT et al (2009) Enhanced orexin receptor-2 signaling prevents diet-induced obesity and improves leptin sensitivity. Cell Metab 9:64–76PubMedCentralPubMedCrossRef Funato H, Tsai AL, Willie JT et al (2009) Enhanced orexin receptor-2 signaling prevents diet-induced obesity and improves leptin sensitivity. Cell Metab 9:64–76PubMedCentralPubMedCrossRef
5.
go back to reference Sellayah D, Bharaj P, Sikder D (2011) Orexin is required for brown adipose tissue development, differentiation, and function. Cell Metab 14:478–490PubMedCrossRef Sellayah D, Bharaj P, Sikder D (2011) Orexin is required for brown adipose tissue development, differentiation, and function. Cell Metab 14:478–490PubMedCrossRef
6.
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 95:322–327PubMedCentralPubMedCrossRef 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 95:322–327PubMedCentralPubMedCrossRef
7.
go back to reference Marcus JN, Aschkenasi CJ, Lee CE et al (2001) Differential expression of orexin receptors 1 and 2 in the rat brain. J Comp Neurol 435:6–25PubMedCrossRef Marcus JN, Aschkenasi CJ, Lee CE et al (2001) Differential expression of orexin receptors 1 and 2 in the rat brain. J Comp Neurol 435:6–25PubMedCrossRef
8.
go back to reference Nakabayashi M, Suzuki T, Takahashi K et al (2003) Orexin-A expression in human peripheral tissues. Mol Cell Endocrinol 205:43–50PubMedCrossRef Nakabayashi M, Suzuki T, Takahashi K et al (2003) Orexin-A expression in human peripheral tissues. Mol Cell Endocrinol 205:43–50PubMedCrossRef
9.
go back to reference Blanco M, Lopez M, Garcia-Caballero T et al (2001) Cellular localization of orexin receptors in human pituitary. J Clin Endocrinol Metab 86:1616–1619 Blanco M, Lopez M, Garcia-Caballero T et al (2001) Cellular localization of orexin receptors in human pituitary. J Clin Endocrinol Metab 86:1616–1619
10.
go back to reference Johren O, Bruggemann N, Dendorfer A, Dominiak P (2003) Gonadal steroids differentially regulate the messenger ribonucleic acid expression of pituitary orexin type 1 receptors and adrenal orexin type 2 receptors. Endocrinology 144:1219–1225PubMedCrossRef Johren O, Bruggemann N, Dendorfer A, Dominiak P (2003) Gonadal steroids differentially regulate the messenger ribonucleic acid expression of pituitary orexin type 1 receptors and adrenal orexin type 2 receptors. Endocrinology 144:1219–1225PubMedCrossRef
11.
go back to reference Rodgers RJ, Halford JC, Nunes de Souza RL et al (2001) SB-334867, a selective orexin-1 receptor antagonist, enhances behavioural satiety and blocks the hyperphagic effect of orexin-A in rats. Eur J Neurosci 13:1444–1452PubMedCrossRef Rodgers RJ, Halford JC, Nunes de Souza RL et al (2001) SB-334867, a selective orexin-1 receptor antagonist, enhances behavioural satiety and blocks the hyperphagic effect of orexin-A in rats. Eur J Neurosci 13:1444–1452PubMedCrossRef
12.
go back to reference Nishino S, Ripley B, Overeem S, Lammers GJ, Mignot E (2000) Hypocretin (orexin) deficiency in human narcolepsy. Lancet 355:39–40PubMedCrossRef Nishino S, Ripley B, Overeem S, Lammers GJ, Mignot E (2000) Hypocretin (orexin) deficiency in human narcolepsy. Lancet 355:39–40PubMedCrossRef
13.
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
14.
go back to reference Peyron C, Faraco J, Rogers W et al (2000) A mutation in a case of early onset narcolepsy and a generalized absence of hypocretin peptides in human narcoleptic brains. Nat Med 6:991–997PubMedCrossRef Peyron C, Faraco J, Rogers W et al (2000) A mutation in a case of early onset narcolepsy and a generalized absence of hypocretin peptides in human narcoleptic brains. Nat Med 6:991–997PubMedCrossRef
15.
go back to reference Siegel JM, Moore R, Thannickal T, Nienhuis R (2001) A brief history of hypocretin/orexin and narcolepsy. Neuropsychopharmacology 25:S14–S20PubMedCrossRef Siegel JM, Moore R, Thannickal T, Nienhuis R (2001) A brief history of hypocretin/orexin and narcolepsy. Neuropsychopharmacology 25:S14–S20PubMedCrossRef
16.
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
17.
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
18.
go back to reference Tsuneki H, Murata S, Anzawa Y et al (2008) Age-related insulin resistance in hypothalamus and peripheral tissues of orexin knockout mice. Diabetologia 51:657–667PubMedCrossRef Tsuneki H, Murata S, Anzawa Y et al (2008) Age-related insulin resistance in hypothalamus and peripheral tissues of orexin knockout mice. Diabetologia 51:657–667PubMedCrossRef
19.
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
20.
go back to reference Harada S, Yamazaki Y, Tokuyama S (2013) Orexin-A suppresses postischemic glucose intolerance and neuronal damage through hypothalamic brain-derived neurotrophic factor. J Pharmacol Exp Ther 344:276–285PubMedCrossRef Harada S, Yamazaki Y, Tokuyama S (2013) Orexin-A suppresses postischemic glucose intolerance and neuronal damage through hypothalamic brain-derived neurotrophic factor. J Pharmacol Exp Ther 344:276–285PubMedCrossRef
21.
go back to reference Shiuchi T, Haque MS, Okamoto S et al (2009) Hypothalamic orexin stimulates feeding-associated glucose utilization in skeletal muscle via sympathetic nervous system. Cell Metab 10:466–480PubMedCrossRef Shiuchi T, Haque MS, Okamoto S et al (2009) Hypothalamic orexin stimulates feeding-associated glucose utilization in skeletal muscle via sympathetic nervous system. Cell Metab 10:466–480PubMedCrossRef
22.
go back to reference Ducroc R, Voisin T, El Firar A, Laburthe M (2007) Orexins control intestinal glucose transport by distinct neuronal, endocrine, and direct epithelial pathways. Diabetes 56:2494–2500PubMedCentralPubMedCrossRef Ducroc R, Voisin T, El Firar A, Laburthe M (2007) Orexins control intestinal glucose transport by distinct neuronal, endocrine, and direct epithelial pathways. Diabetes 56:2494–2500PubMedCentralPubMedCrossRef
23.
go back to reference Skrzypski M, Le TT, Kaczmarek P et al (2011) Orexin A stimulates glucose uptake, lipid accumulation and adiponectin secretion from 3T3-L1 adipocytes and isolated primary rat adipocytes. Diabetologia 54:1841–1852PubMedCrossRef Skrzypski M, Le TT, Kaczmarek P et al (2011) Orexin A stimulates glucose uptake, lipid accumulation and adiponectin secretion from 3T3-L1 adipocytes and isolated primary rat adipocytes. Diabetologia 54:1841–1852PubMedCrossRef
24.
go back to reference Switonska MM, Kaczmarek P, Malendowicz LK, Nowak KW (2002) Orexins and adipoinsular axis function in the rat. Regul Pept 104:69–73PubMedCrossRef Switonska MM, Kaczmarek P, Malendowicz LK, Nowak KW (2002) Orexins and adipoinsular axis function in the rat. Regul Pept 104:69–73PubMedCrossRef
25.
go back to reference Nowak KW, Strowski MZ, Switonska MM et al (2005) Evidence that orexins A and B stimulate insulin secretion from rat pancreatic islets via both receptor subtypes. Int J Mol Med 15:969–972PubMed Nowak KW, Strowski MZ, Switonska MM et al (2005) Evidence that orexins A and B stimulate insulin secretion from rat pancreatic islets via both receptor subtypes. Int J Mol Med 15:969–972PubMed
26.
go back to reference Goncz E, Strowski MZ, Grotzinger C et al (2008) Orexin-A inhibits glucagon secretion and gene expression through a Fox01-dependent pathway. Endocrinology 149:1618–1626PubMedCrossRef Goncz E, Strowski MZ, Grotzinger C et al (2008) Orexin-A inhibits glucagon secretion and gene expression through a Fox01-dependent pathway. Endocrinology 149:1618–1626PubMedCrossRef
27.
go back to reference Ehrstrom M, Naslund E, Levin F et al (2004) Pharmacokinetic profile of orexin A and effects on plasma insulin and glucagon in the rat. Regul Pept 119:209–212PubMedCrossRef Ehrstrom M, Naslund E, Levin F et al (2004) Pharmacokinetic profile of orexin A and effects on plasma insulin and glucagon in the rat. Regul Pept 119:209–212PubMedCrossRef
28.
go back to reference Ouedraogo R, Naslund E, Kirchgessner AL (2003) Glucose regulates the release of orexin-a from the endocrine pancreas. Diabetes 52:111–117PubMedCrossRef Ouedraogo R, Naslund E, Kirchgessner AL (2003) Glucose regulates the release of orexin-a from the endocrine pancreas. Diabetes 52:111–117PubMedCrossRef
30.
go back to reference Komaki G, Matsumoto Y, Nishikata H et al (2001) Orexin-A and leptin change inversely in fasting non-obese subjects. Eur J Endocrinol 144:645–651PubMedCrossRef Komaki G, Matsumoto Y, Nishikata H et al (2001) Orexin-A and leptin change inversely in fasting non-obese subjects. Eur J Endocrinol 144:645–651PubMedCrossRef
31.
go back to reference Arafat AM, Kaczmarek P, Skrzypski M et al (2013) Glucagon increases circulating fibroblast growth factor 21 independently of endogenous insulin levels: a novel mechanism of glucagon-stimulated lipolysis? Diabetologia 56:588–597PubMedCrossRef Arafat AM, Kaczmarek P, Skrzypski M et al (2013) Glucagon increases circulating fibroblast growth factor 21 independently of endogenous insulin levels: a novel mechanism of glucagon-stimulated lipolysis? Diabetologia 56:588–597PubMedCrossRef
32.
go back to reference Arafat MA, Otto B, Rochlitz H et al (2005) Glucagon inhibits ghrelin secretion in humans. Eur J Endocrinol 153:397–402PubMedCrossRef Arafat MA, Otto B, Rochlitz H et al (2005) Glucagon inhibits ghrelin secretion in humans. Eur J Endocrinol 153:397–402PubMedCrossRef
33.
go back to reference Arafat AM, Perschel FH, Otto B et al (2006) Glucagon suppression of ghrelin secretion is exerted at hypothalamus-pituitary level. J Clin Endocrinol Metab 91:3528–3533PubMedCrossRef Arafat AM, Perschel FH, Otto B et al (2006) Glucagon suppression of ghrelin secretion is exerted at hypothalamus-pituitary level. J Clin Endocrinol Metab 91:3528–3533PubMedCrossRef
34.
go back to reference Lee Y, Wang MY, Du XQ, Charron MJ, Unger RH (2011) Glucagon receptor knockout prevents insulin-deficient type 1 diabetes in mice. Diabetes 60:391–397PubMedCentralPubMedCrossRef Lee Y, Wang MY, Du XQ, Charron MJ, Unger RH (2011) Glucagon receptor knockout prevents insulin-deficient type 1 diabetes in mice. Diabetes 60:391–397PubMedCentralPubMedCrossRef
35.
go back to reference Skrzypski M, Kakkassery M, Mergler S et al (2013) Activation of TRPV4 channel in pancreatic INS-1E beta cells enhances glucose-stimulated insulin secretion via calcium-dependent mechanisms. FEBS Lett 587:3281–3287PubMedCrossRef Skrzypski M, Kakkassery M, Mergler S et al (2013) Activation of TRPV4 channel in pancreatic INS-1E beta cells enhances glucose-stimulated insulin secretion via calcium-dependent mechanisms. FEBS Lett 587:3281–3287PubMedCrossRef
36.
go back to reference Dostmann WR (1995) (RP)-cAMPS inhibits the cAMP-dependent protein kinase by blocking the cAMP-induced conformational transition. FEBS Lett 375:231–234PubMedCrossRef Dostmann WR (1995) (RP)-cAMPS inhibits the cAMP-dependent protein kinase by blocking the cAMP-induced conformational transition. FEBS Lett 375:231–234PubMedCrossRef
37.
go back to reference Goode PN, Farndon JR, Anderson J, Johnston ID, Morte JA (1986) Diazoxide in the management of patients with insulinoma. World J Surg 10:586–592PubMedCrossRef Goode PN, Farndon JR, Anderson J, Johnston ID, Morte JA (1986) Diazoxide in the management of patients with insulinoma. World J Surg 10:586–592PubMedCrossRef
38.
go back to reference Trube G, Rorsman P (1986) Calcium and potassium currents recorded from pancreatic beta-cells under voltage clamp control. Adv Exp Med Biol 211:167–175PubMedCrossRef Trube G, Rorsman P (1986) Calcium and potassium currents recorded from pancreatic beta-cells under voltage clamp control. Adv Exp Med Biol 211:167–175PubMedCrossRef
39.
go back to reference Gilon P, Henquin JC (1992) Influence of membrane potential changes on cytoplasmic Ca2+ concentration in an electrically excitable cell, the insulin-secreting pancreatic B cell. J Biol Chem 267:20713–20720PubMed Gilon P, Henquin JC (1992) Influence of membrane potential changes on cytoplasmic Ca2+ concentration in an electrically excitable cell, the insulin-secreting pancreatic B cell. J Biol Chem 267:20713–20720PubMed
40.
go back to reference Urdanivia E, Pek S, Santiago JC (1979) Inhibition of glucagon secretion by diazoxide in vitro. Diabetes 28:26–31PubMedCrossRef Urdanivia E, Pek S, Santiago JC (1979) Inhibition of glucagon secretion by diazoxide in vitro. Diabetes 28:26–31PubMedCrossRef
41.
go back to reference Cherrington AD (1997) The metabolic actions of glucagon. In: Draznin B, Rizza E (eds) Clinical research in diabetes and obesity, volume 1: methods, assessment, and metabolic regulation (contemporary biomedicine), 1st edn. Humana Press, Totowa, p 222 Cherrington AD (1997) The metabolic actions of glucagon. In: Draznin B, Rizza E (eds) Clinical research in diabetes and obesity, volume 1: methods, assessment, and metabolic regulation (contemporary biomedicine), 1st edn. Humana Press, Totowa, p 222
42.
go back to reference Geary N, Kissileff HR, Pi-Sunyer FX, Hinton V (1992) Individual, but not simultaneous, glucagon and cholecystokinin infusions inhibit feeding in men. Am J Physiol 262:R975–R980PubMed Geary N, Kissileff HR, Pi-Sunyer FX, Hinton V (1992) Individual, but not simultaneous, glucagon and cholecystokinin infusions inhibit feeding in men. Am J Physiol 262:R975–R980PubMed
43.
go back to reference Geary N (1990) Pancreatic glucagon signals postprandial satiety. Neurosci Biobehav Rev 14:323–338PubMedCrossRef Geary N (1990) Pancreatic glucagon signals postprandial satiety. Neurosci Biobehav Rev 14:323–338PubMedCrossRef
44.
go back to reference Vanderweele DA, Macrum BL, Oetting RL (1986) Glucagon, satiety from feeding and liver/pancreatic interactions. Brain Res Bull 17:539–543PubMedCrossRef Vanderweele DA, Macrum BL, Oetting RL (1986) Glucagon, satiety from feeding and liver/pancreatic interactions. Brain Res Bull 17:539–543PubMedCrossRef
Metadata
Title
Glucagon regulates orexin A secretion in humans and rodents
Authors
Ayman M. Arafat
Przemysław Kaczmarek
Marek Skrzypski
Ewa Pruszyńska-Oszmałek
Paweł Kołodziejski
Aikaterini Adamidou
Stephan Ruhla
Dawid Szczepankiewicz
Maciej Sassek
Maria Billert
Bertram Wiedenmann
Andreas F. H. Pfeiffer
Krzysztof W. Nowak
Mathias Z. Strowski
Publication date
01-10-2014
Publisher
Springer Berlin Heidelberg
Published in
Diabetologia / Issue 10/2014
Print ISSN: 0012-186X
Electronic ISSN: 1432-0428
DOI
https://doi.org/10.1007/s00125-014-3335-4

Other articles of this Issue 10/2014

Diabetologia 10/2014 Go to the issue
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 discuss last year's major advances in heart failure and cardiomyopathies.