Contact
Header_1077x260px_About_Profil.jpg

Glucagon - the rising star in diabetes research (Part 2)

Dr.
Posted by Dr. Ulrike Hövelmann on Oct 21, 2016 5:00:00 PM

Promising developments in new treatments for diabetes involving glucagon

Role of glucagon action

Glucagon is secreted by the pancreatic alpha cells mainly stimulated by changes in local concentration of glucose, amino acids and insulin and through the autonomic (vegetative) nervous system. Recently, it has been demonstrated that sodium-glucose co-transporter 2 (SGLT-2) is expressed in pancreatic alpha cells, designated for an inhibitory effect on glucagon release in the range of physiological glucose concentrations [1]. The endocrine effects on the liver include the activation on the glucagon receptor (GCGR), a G-protein-coupled receptor (GPCR), and engagement of the GαS and β-arrestin pathways (signalling cascades). Glucagon initiates the increase in the export of glucose from the liver as a result of enhanced glycogenolysis and gluconeogenesis [2, 3, 4, 5].

Controversy remains regarding the relative importance of the acute metabolic and more long-term transcriptional actions of glucagon. There is little doubt of the robust increase in the transcription of ‘cAMP response element binding protein’ (CREB) target genes (intracellular production of cAMP and subsequent activation of protein kinase A) whose protein products are at potentially rate-controlling steps in the gluconeogenic pathway [6]. However, rapidity of glucagon response provides arguments against a transcriptional mechanism for gluconeogenic fluxes [7]. However, it remains likely that after prolonged fasting or in a setting of chronic hyperglucagonemia, such as diabetes, transcriptional regulation drives meaningful changes in hepatic metabolism.

Further scientific research showed also direct post-translational effects of hepatic metabolism (such as phosphorylation) to cover rapid glucagon related changes in systematic and hepatic metabolism [8]. Additional metabolic actions of glucagon have been identified, which are parts of its conserved role as a regulator of fasting metabolism. These actions include the ability to promote the amino acid catabolism to provide gluconeogenic precursors [9, 10, 11] and to promote an increase of mitochondrial oxygen consumption through enhanced mitochondrial calcium uptake which is required for glucagon`s stimulation of gluconeogenesis. [12, 13, 14].

The central role of glucagon action in the brain

The brain is increasingly being recognised as an important glucagon-sensitive organ. It has been demonstrated that in contrast to the systemic effect on glucose response, glucagon action in the mediobasal hypothalamus (MBH) site linked to the nervous system and endocrine system and the dorsal vagal complex (DVC) site located in the brainstem, known to regulate several autonomic functions actually reduces hepatic glucose production [15]. The central sites of the brain are also sensitive to circulating glucagon induced either by i.v. glucagon or high-protein meals to antagonise the hepatic action of glucagon. However, this central negative autoregulation for glucose homeostasis was lost following a three day high fat diet in rats [16]. In addition, the role of hypothalamic glucagon is also implicated in reducing food intake via differential signalling cascades [17, 18]. The body weight changes induced by glucagon may be partly explained by its effect on energy expenditure; it induces thermogenesis through the sympathetic nervous innervation of brown adipose tissue [19].

 

Glucagon Antagonism

The recent renewed interest in glucagon antagonism as a viable therapeutic approach mainly derived from preclinical results in rodents suggesting that reducing glucagon action or secretion by potent glucagon suppressor leptin and glucagon receptor antibodies suppressed all catabolic manifestations of diabetes during insulin deficiency [20, 21, 22,23, 24, 25, 26]. Glucagon antagonism as a sustainable therapeutic approach has shown impressive glucose lowering with small molecule glucagon receptor antagonists in diabetic type 2 individuals [27]. In addition, significantly lowered HbA1c and glucose levels with a low risk for hypoglycemia have been demonstrated. However, modest, reversible increases in serum aminotransferases were observed [28]. The latter had also been noted with other glucagon receptor antagonists, including a human glucagon receptor monoclonal antibody [21], other small molecules [29, 30,31], and an antisense glucagon receptor antagonist [32]. Further contribution of distinct glucagon effects on clinical efficacy and safety will be an important area of research.

Glucagon Polyagonists

Novel findings indicate that activation of glucagon receptors in conjunction with other G-protein-coupled receptors showed metabolical improvements in diabetes and obesity. Therefore, maintaining a sufficient glucagon action to activate the central glucagon system may be therapeutically beneficial. A series of single-molecule glucagon receptor (GCGR)/glucagon-like peptide-1 receptor (GLP-1R) dual agonists were generated using glucagon as a template sequence to which chemical modifications were introduced. The resulting peptides were of comparable structure to glucagon and GLP-1, but of balanced agonism at each receptor and comparable inherent potency to native hormones. This dual activation of GCGR and GLP-1R normalised glucose tolerance and reduced food intake in mice with diet-induced obesity [33, 34]. It had also been suggested that oxyntomodulin, another gut peptide involved in metabolic control, would potentially represent a therapeutic approach [35].

Oxyntomodulin is a dual agonist, acting at both the GLP-1R (enhanced glucose-stimulated insulin secretion and suppression of food intake) and the GCGR (suppression of food intake, increased energy expenditure and improved lipid metabolism) with similar potency. The metabolic improvements resulting from  high body fat burning and weight loss, together with the known beneficial metabolic actions of GLP-1, have a stronger impact than the inherent diabetogenic property of glucagon agonism. The exact molecular action that governs the action profile of the dual agonist is still being investigated, as well as the relative advantage in accomplishing the mixed pharmacology in a single peptide.

One important component has already been discovered: glucagon activates the secretion of fibroblast growth factor 21 (FGF21), an endogenous protein acting at the level of the brain, liver, and adipose tissue to decrease body weight and improve dyslipidemia, which itself has demonstrated translational benefits as an anti-obesity therapy [36, 37].

Aiming to enhance the glycemic benefits of GLP-1, the structurally related second member of the two principle incretin hormones, glucose-dependent insulinotropic peptide (GIP), was selected as the second component in a new series of dual agonists. These dual GIP/GLP co-agonists exhibit superior in vivo efficacy in mice, rats, nonhuman primates, and humans when compared to equimolar monoagonists. In particular, insulin secretion and glucose tolerance improved and neither chronic hyperinsulinemia nor hypoglycemia was observed [38].

A triagonist aimed at simultaneous activation of GCG, GLP-1 and GIP receptors improved metabolic and glycemic profiles in obese and diabetic rodents. When specifically compared to the dual incretin co-agonists, these novel triple-acting peptides were superior in correcting the excess of adipose tissue mass, liver fat, food intake, and plasma cholesterol while demonstrating increased energy expenditure, improved glucose tolerance, and protection from glucolipotoxic pancreatic islet destruction [39].

Work on safety and efficacy clinical data will help to expand the knowledge about increased energy expenditure and decreased food intake and its hyperglycemic effects to be countered by the actions of GLP-1 and/or GIP.

However, the brain penetrance of these polyagonists and whether activation of central glucagon signalling plays a role in counteracting the diabetogenic effects of peripheral glucagon remain to be investigated.

Bihormonal Bionic Pancreas

A promising approach treating diabetes is the development of fully automated artificial/bionic pancreas systems that use both insulin and glucagon as intermittent mini-boluses to maintain euglycemia. Compared with an insulin pump, an automated, bihormonal, pancreas showed improved mean glycemic levels, with less frequent hypoglycemic episodes, among both adults and adolescents with type 1 diabetes mellitus [40]. In dual-hormone artificial pancreas short-term studies glucagon concentrations are considered to be nearly within the physiologic fasting ranges [41]. Furthermore, glucagon’s effects on reducing caloric intake and increasing energy expenditure could even be beneficial within the context of the growing prevalence of overweight and obesity in patients with diabetes. So far, a stable liquid glucagon formulation for longer use in the bihormonal pancreas is still under development [42]. Therefore, comprehensive long-term artificial pancreas studies using repeated micro glucagon doses are still lacking. Further research is needed to characterize the potential benefits and safety profiles of future chronic glucagon use.

 

If you enjoyed this article, then check our part 1 and make sure to register for our blog newsletter (on the right of this page) so you don't miss similar articles in the future. 

 

[1] Bonner C, Kerr-Conte J, Gmyr V, et al. Inhibition of the glucose transporter SGLT2 with dapagliflozin in pancreatic alpha cells triggers glucagon secretion. Nat Med 2015; 21. doi:10.1038/nm.3828.

[2] Cohen P. Activation and phosphorylation of the subunits of phosphorylase kinase. Biochem J.1972; 130:5P–6P.

[3] Titanji VP, Zetterqvist O, Engstroom L (1976) Regulation in vitro of rat liver pyruvate kinase by phosphorylation-dephosphorylation reactions, catalyzed by cyclic-AMP dependent protein kinases and a histone phosphatase. Biochim Biophys Acta. 1976; 422:98–108.

[4] El-Maghrabi MR, Claus TH, Pilkis J, Fox E, Pilkis SJ (1982) Regulation of rat liver fructose 2,6-bisphosphatase. J Biol Chem. 1982; 257:7603–7607.

[5] Exton JH. Mechanisms of hormonal regulation of hepatic glucose metabolism. Diabetes Metab Rev. 1987; 3:163–183.

[6] Lin HV, Accili D. Hormonal regulation of hepatic glucose production in health and disease. Cell Metab. 2011; 14:9–19.

[[7]] Burgess SC, He T, Yan Z et al. Cytosolic phosphoenolpyruvate carboxykinase does not solely control the rate of hepatic gluconeogenesis in the intact mouse liver. Cell Metab. 2007; 5:313–320.

[[8]] Exton JH, Park CR. Control of gluconeogenesis in liver. 3. Effects of L-lactate, pyruvate, fructose, glucagon, epinephrine, and adenosine 3 ,5 -monophosphate on gluconeogenic intermediates in the perfused rat liver. J Biol Chem. 1969; 244:1424–1433.

[9] Fukita Y, Gotto AM, Unger RH. Basal and postprotein insulin and glucagon levels during a high and low carbohydrate intake and their relationships to plasma triglycerides. Diabetes. 1975; 24:552– 558.

[10] .Flakoll PJ, Borel MJ, Wentzel LS, Williams PE, Lacy DB, Abumrad NN (1994) The role of glucagon in the control of protein and amino acid metabolismin vivo.Metab Clin Exp. 1994; 43:1509–1516.

[11] Charlton MR, Adey DB, Nair KS (1996) Evidence for a catabolic role of glucagon during an amino acid load. J Clin Invest 98:90–99.

[12] Bartlett PJ, Gaspers LD, Pierobon N, Thomas AP. Calcium dependent regulation of glucose homeostasis in the liver. Cell 2014; Calcium. 2014;55:306–316.

[13] Cardenas C, Miller RA, Smith I et al. Essential regulation of cell bioenergetics by constitutive InsP3 receptor Ca2+ transfer to mitochondria. Cell. 2019; 142:270–283.

[14] Amigo I, Traba J, Gonzalez-Barroso MM et al (2013) Glucagon regulation of oxidative phosphorylation requires an increase in matrix adenine nucleotide content through Ca2+ activation of the mitochondrial ATP-Mg/Pi carrier SCaMC-3. J Biol Chem 288:7791–7802.

[15] Abraham MA, Filippi BM, Kang GM, Kim MS, Lam TK. Insulin action in the hypothalamus and dorsal vagal complex. Exp Physiol. 2014; 99:1104–1109.

[16] Mighiu PI, Yue JT, Filippi BM et al. Hypothalamic glucagon signaling inhibits hepatic glucose production. NatMed. 2013; 19:766–772.

[17] Quiñones M, Al-Massadi O, Gallego R et al. Hypothalamic CaMKK mediates glucagon anorectic effect and its diet-induced resistance. Mol Metab. 2015; 4:961–970.

[18] Kurose Y, Kamisoyama H, Honda K et al. Effects of central administration of glucagon on feed intake and endocrine responses in sheep. Anim Sci J. 2009; 80:686–690.

[19] Heppner KM, Habegger KM, Day J, et al. Glucagon regulation of energy metabolism. Physiol Behav 2010;100:545–8.

[20] Kelly RP, Garhyan P, Raddad E et al. Short-term administration of the glucagon receptor antagonist LY2409021 lowers blood glucose in healthy people and in those with type 2 diabetes. Diabetes Obes Metab. 2015; 17:414–422.

[21] Mu J, Jiang G, Brady E et al. Chronic treatment with a glucagon receptor antagonist lowers glucose and moderately raises circulating glucagon and glucagon-like peptide 1 without severe alpha cell hypertrophy in diet-induced obese mice. Diabetologia. 2011; 54:2381–2391.

[22] Mu J, Qureshi SA, Brady EJ et al. Anti-diabetic efficacy and impact on amino acid metabolism of GRA1, a novel small molecule glucagon receptor antagonist. PLoS One. 2012; 7:e49572.

[23] Petersen KF, Sullivan JT Effects of a novel glucagon receptor antagonist (Bay 27-9955) on glucagon-stimulated glucose production in humans. Diabetologia. 2001; 44:2018–2024.

[24] Sammons MF, Lee EC. Recent progress in the development of small-molecule glucagon receptor antagonists. Bioorg Med Chem Lett. 2015; 25:4057–4064.

[25] Wang MY, Chen L, Clark GO et al. Leptin therapy in insulin deficient type 1 diabetes. Proc Natl Acad Sci U S A. 2010; 107:4813–4819.

[26] Yu X, Park BH, Wang MY, Wang ZW, Unger RH. Making insulin-deficient type 1 diabetic rodents thrive without insulin. Proc Natl Acad Sci U S A. 2008; 105:14070–14075.

[27] Kazda CM, Garhyan P, Kelly RP et al. A randomized, double- blind, placebo-controlled phase 2 study of the glucagon receptor antagonist LY2409021 in patients with type 2 diabetes. Diabetes Care. 2015; doi:10.2337/dc15-1643.

[28] Kazda CM, Ding Y, Kelly RP et al. Evaluation of Efficacy and Safety of the Glucagon Receptor Antagonist LY2409021 in Patients With Type 2 Diabetes: 12- and 24-Week Phase 2 Studies. Diabetes Care. 2016; 39:1241-1249. doi:10.2337/dc15-1643.

[29] Kazierad DJ, Bergman A, Tan B, Erion DM, Somayaji V, Lee DS, Rolph T. Effects of multiple ascending doses of the glucagon receptor antagonist PF-06291874 in patients with type 2 diabetes mellitus. Diabetes, Obesity and Metabolism. 2016; 18: 795–802., 2016.pharmacodynamics (PD) of PF-06291874 (PF).

[30] Vajda EG, Logan D, Lasseter K, et al. Pharmacokinetics and pharmacodynamics of single and multiple doses of the glucagon receptor antagonist LGD-6972 in healthy subjects and subjects with type 2 diabetes mellitus. Diabetes, Obesity and Metabolism.2016; doi 10.1111/dom.12752.

[31] Engel SS, Xu L, Andryuk PJ, et al. Efficacy and tolerability of MK-0893, a glucagon receptor antagonist (GRA), in patients with type 2 diabetes (T2DM) (Abstract 309-OR). Diabetes 2011;60 (Suppl. 1):A85.

[32] Morgan E, SmithA,Watts L, et al. ISIS-GCGRRX, an antisense glucagon receptor antagonist, caused rapid, robust, and sustained improvements in glycemic control without changes in BW, BP, lipids, or hypoglycemia in T2DM patients on stable metformin therapy (Abstract 109-LB). Diabetes 2014; 63(Suppl. 1A):LB28.

[33] Pocai A, Carrington PE, Adams JR et al.  Glucagon-like peptide 1/glucagon receptor dual agonism reverses obesity in mice. Diabetes. 2009; 58:2258–2266.

[34] Day JW, Ottaway N, Patterson JT et al (2009) A new glucagon and GLP-1 co-agonist eliminates obesity in rodents. Nat Chem Biol. 2009; 5:749–757.

[35] Dakin, C.L., Gunn, I., Small, C.J., Edwards, C.M., Hay, D.L., Smith, D.M., Ghatei, M.A., and Bloom, S.R. (2001). Oxyntomodulin inhibits food intake in the rat. Endocrinology. 2001; 142, 4244–4250.

[36] Habegger, K.M., Stemmer, K., Cheng, C., Mu¨ ller, T.D., Heppner, K.M., Ottaway, N., Holland, J., Hembree, J.L., Smiley, D., Gelfanov, V., et al. (2013b). Fibroblast growth factor 21 mediates specific glucagon actions. Diabetes 62, 1453–1463.

[37] Talukdar, S., Zhou, Y., Li, D., Rossulek, M., Dong, J., Somayaji, V., Weng, Y., Clark, R., Lanba, A., Owen, B.M., et al. A long-acting FGF21 molecule, PF-05231023, decreases body weight and improves lipid profile in non-human primates and type 2 diabetic subjects. Cell Metab.  2016;23, 427–440.

[38] Tschöp MH, Finan B, Clemmensen C. Unimolecular Polypharmacy for Treatment of Diabetes and Obesity. Cell metabolism.2016; 24:51-62.

[39] Finan, B., Yang, B., Ottaway, N., Smiley, D.L., Ma, T., Clemmensen, C., Chabenne, J., Zhang, L., Habegger, K.M., Fischer, K., et al. A rationally designed monomeric peptide triagonist corrects obesity and diabetes in rodents. Nat. Med. 2015b; 21, 27–36.

[40] Russell SJ, El-Khatib FH Sinha M, et al. Outpatient Glycemic Control with a Bionic Pancreas in Type 1 Diabetes. New England Journal of Medicine. 2014: 1-13.doi: 10.1056/NEJMoa1314474.

[41] Taleb N, Haidar A, Messier V et al. Glucagon in the artificial pancreas systems; potential benefits and safety profile of future chronic use. Diabetes Obesity and Metabolism 2016; doi: 10.1111/dom.12789.

[42] Pohl R, Li M, Krasner A, De Souza E. Development of stable liquid glucagon formulations for use in artificial pancreas. J Diabetes Sci Technol 2015;9:8–16.

Topics: The Science behind Diabetes