You are on page 1of 16

- -

15207560, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/dmrr.3609 by Cochrane Germany, Wiley Online Library on [30/01/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Received: 23 August 2022 Revised: 2 December 2022 Accepted: 30 December 2022

DOI: 10.1002/dmrr.3609

REVIEW ARTICLE

Glucagon in type 2 diabetes: Friend or foe?

Irene Caruso | Nicola Marrano | Giuseppina Biondi |


Valentina Annamaria Genchi | Rossella D'Oria | Gian Pio Sorice |
Sebastio Perrini | Angelo Cignarelli | Annalisa Natalicchio | Luigi Laviola |
Francesco Giorgino

Department of Precision and Regenerative


Medicine and Ionian Area, Section of Internal Abstract
Medicine, Endocrinology, Andrology and
Hyperglucagonemia is one of the ‘ominous’ eight factors underlying the pathogen-
Metabolic Diseases, University of Bari Aldo
Moro, Bari, Italy esis of type 2 diabetes (T2D). Glucagon is a peptide hormone involved in maintaining
glucose homoeostasis by increasing hepatic glucose output to counterbalance in-
Correspondence
Francesco Giorgino, Department of Precision
sulin action. Long neglected, the introduction of dual and triple agonists exploiting
and Regenerative Medicine and Ionian Area, glucagon signalling pathways has rekindled the interest in this hormone beyond its
Section of Internal Medicine, Endocrinology,
Andrology and Metabolic Diseases, University
classic effect on glycaemia. Glucagon can promote weight loss by regulating food
of Bari Aldo Moro, Bari, Italy. intake, energy expenditure, and brown and white adipose tissue functions through
Email: francesco.giorgino@uniba.it
mechanisms still to be fully elucidated, thus its role in T2D pathogenesis should be
further investigated. Moreover, the role of glucagon in the development of T2D
micro‐ and macro‐vascular complications is elusive. Mounting evidence suggests its
beneficial effect in non‐alcoholic fatty liver disease, while few studies postulated its
favourable role in peripheral neuropathy and retinopathy. Contrarily, glucagon re-
ceptor agonism might induce renal changes resembling diabetic nephropathy, and
data concerning glucagon actions on the cardiovascular system are conflicting. This
review aims to summarise the available findings on the role of glucagon in the
pathogenesis of T2D and its complications. Further experimental and clinical data
are warranted to better understand the implications of glucagon signalling modu-
lation with new antidiabetic drugs.

KEYWORDS
cotadutide, diabetic kidney disease, dual agonists, glucagon, NAFLD, type 2 diabetes

1 | INTRODUCTION are implicated in the pathogenesis of T2D and its micro‐ and mac-
rovascular complications have not been extensively studied. More-
Glucagon is a peptide hormone mainly produced by pancreatic α‐ over, little is known about the effect of the commonly used
cells.1 This hormone is involved not only in maintaining glucose antidiabetic drugs on fasting and postprandial glucagon levels,
homoeostasis and counterbalancing insulin actions but also in the especially following chronic treatment.1
regulation of lipids and amino acids metabolism, autophagy, food Neglected for several years, the recent introduction in the
intake, body weight, and cardiovascular (CV) functions.1 Its role in therapeutic arsenal against T2D of dual and triple agonists that take
the pathogenesis of type 2 diabetes (T2D) has first been postulated advantage of glucagon receptor (GCGR) activity has rekindled the
by Unger and Orci in the ‘bi‐hormonal theory’.2 To date, whether the

-
interest of the scientific community in the complex relationship be-
pleiotropic effects of glucagon, besides those on glucose metabolism, tween glucagon and T2D.

Diabetes Metab Res Rev. 2023;e3609. wileyonlinelibrary.com/journal/dmrr © 2023 John Wiley & Sons Ltd. 1 of 16
https://doi.org/10.1002/dmrr.3609
15207560, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/dmrr.3609 by Cochrane Germany, Wiley Online Library on [30/01/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
2 of 16
- CARUSO ET AL.

The aim of this narrative review is to summarise the available cell membrane is already depolarised via the glucose‐induced closure
evidence concerning the involvement of glucagon in the multifaceted of ATP‐sensitive K+ leak channels and the insulin secretory ma-
pathogenesis of T2D, addressing β‐cell function, peripheral insulin chinery is already in motion,7,19 whereas the activation of GCGRs and
sensitivity, food intake regulation, micro‐ and macro‐vascular com- GLP‐1Rs is unable to trigger insulin secretion on its own.18 Conse-
plications, and the therapeutic implications thereof. quently, glucagon stimulates insulin secretion in the post‐prandial
phase but not during fasting, when the counter‐regulatory and
hyperglycaemic effects of glucagon are required.
2 | THE ROLE OF GLUCAGON IN THE The importance of GCGRs and GLP‐1Rs in glucagon‐potentiated
PATHOGENESIS OF TYPE 2 DIABETES GSIS has been broadly demonstrated. The genetic deletion of the
GCGRs or GLP‐1Rs from β‐cells,13,20 as well as the blockade of GLP‐
2.1 | The role of glucagon in β‐cell function: A 1Rs by its antagonist exendin (9–39),13,20,21 resulted in decreased
somewhat neglected friendship insulin secretion in response to nutrient stimulation of mouse and
human islets.13,20 Interestingly, while in healthy mice the lack of
In an almost exclusively insulin‐centric vision, dysregulation of the β‐ GCGRs and GLP‐1Rs at β‐cellular level did not affect glucose toler-
cell functional mass has always been regarded as the key mechanistic ance, α‐cells‐secreted glucagon seemed to be essential to maintain
factor in the onset and progression of diabetes.3 However, in 1975, glucose tolerance during the metabolic stress induced by a high‐fat
Unger and Orci2 recognised that impaired glucagon secretion and α‐ diet, suggesting that the communication between α‐ and β‐cells is
cell function also contribute to the pathogenesis of T2D, proposing essential for metabolic adaptation to high‐fat feeding and obesity.13
the ‘bi‐hormonal theory’, which suggested the coexistence of both In line with this evidence, the overexpression of GCGRs in murine
relative or absolute hypoinsulinemia and relative hyperglucagonemia pancreatic β‐cells enhanced GSIS and increased β‐cell mass and
in T2D patients. pancreatic insulin content, protecting from high‐fat diet‐induced
impaired glucose tolerance and hyperglycaemia.22 Also, transgenic
mice overexpressing GCGRs were resistant to streptozotocin‐
2.1.1 | Physiology induced β‐cell injury partially due to enhanced intra‐islet action of
both glucagon and insulin on β‐cell function.23
In humans, α‐ and β‐cells account for ~30%–40% and ~50%–60% of Although several studies with mouse and human islets indicated
the endocrine islet cells, respectively, and are intermingled that intra‐islet glucagon stimulated insulin release primarily by acti-
throughout the islet.4 Under physiological conditions, α‐ and β‐cells vating β‐cell GLP‐1R,13,14 only the combined blockage of both re-
influence each other: while the ability of insulin, as well as of other β‐ ceptors significantly reduced insulin secretion.20
cells secretory products (e.g. zinc, gamma‐aminobutyric acid [GABA], Thus, the available evidence highlighted a paradoxical action of
serotonin), to inhibit glucagon secretion is widely recognised,5,6 the glucagon: under hyperglycaemic conditions, glucagon is able to
effects of glucagon to stimulate insulin secretion is a less‐known and potentiate insulin secretion and consequently glucose lowering, while
somewhat paradoxical event.7 Nevertheless, the insulinotropic effect it has also been associated with increased hepatic glucose production
of glucagon has been demonstrated in several studies with different concurring with hyperglycaemia. However, these two opposite ef-
model systems, such as pig pancreatic islets,8 rat islets cells,9 and fects do not occur simultaneously due to the tonic inhibition (without
10
human pancreatic islets. Islets with higher α‐cell content displayed full suppression) of glucagon secretion by a combination of somato-
improved glucose‐stimulated insulin secretion (GSIS) compared with statin, GABA and serotonin in the post‐prandial phase, keeping
islets with lower α‐cell content, a difference that could be eliminated glucagon levels sufficiently low to exert its paracrine effects on the β‐
by the addition of exogenous glucagon.11 Similarly, the response of cells, without increasing hepatic glucose output.7
isolated pancreatic β‐cells to a glucose stimulus is improved by the
presence of α‐cells or glucagon.12 Moreover, the genetic deletion of
glucagon from α‐cells,13 genetically engineered inhibition of glucagon 2.1.2 | Type 2 diabetes
secretion,14 and α‐cells ablation15 impaired insulin secretion in
mouse and human islets13 and resulted in hyperglycaemia and In T2D, both fasting and post‐prandial hyperglucagonemia may occur,
14,15
glucose intolerance in mice in vivo. maintaining an inadequately high rate of hepatic glucose production
Glucagon may act on β‐cells through the activation of glucagon in the fasting state, thus contributing to hyperglycaemia.5,24 Hyper-
and glucagon‐like peptide‐1 receptors (GCGRs and GLP‐1Rs, glucagonemia could result from the impaired regulation of α‐cell
respectively)7 on the β‐cell surface.16,17 These class B G protein‐ function by β‐cells5: the loss of β‐cell functional mass in T2D may
coupled receptors promote the generation of cyclic AMP (cAMP), cause decreased insulin‐mediated inhibition of glucagon secretion; in
and thus the activation of the cAMP/protein kinase A (PKA)/ex- addition, β‐cells might de‐differentiate to progenitor pluripotent cells
change protein directly activated by the cAMP (EPAC) system, which able to release glucagon, causing a further increase in glucagon
in turn potentiates GSIS.13,18 Importantly, this potentiation only oc- levels.25–27 However, the decrease in β‐cell mass in T2D might be too
curs in the presence of mildly hyperglycaemic conditions, when the β‐ small to explain the excessive glucagon secretion,28 and
15207560, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/dmrr.3609 by Cochrane Germany, Wiley Online Library on [30/01/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
CARUSO ET AL.
- 3 of 16

hyperglucagonemia may also result from α‐cell insulin resistance.29 hyperinsulinemia and that the lack of α‐cell mass expansion and
Also, the impaired function of insulin‐degrading enzyme, which has glucagon secretion may cause secondary inability of the β‐cell mass to
been associated with T2D in genome‐wide association and genetic adapt to peripheral insulin resistance in mice under conditions of
polymorphism studies, caused hyperplasia and dysregulated glucagon nutrient overload.34,35 Accordingly, mice with a complete knockout of
30
secretion in mice α‐cells. the glucagon gene displayed defective islet development and impaired
To date, whether hyperglucagonemia precedes β‐cell failure or β‐cell glucose competence.36,37 Hence, intra‐islet glucagon likely plays
31
vice versa in T2D pathogenesis is still under debate. Jamison et al. an important role in the maintenance of β‐cell mass homoeostasis and
demonstrated that hyperglucagonemia preceded the decline in insulin β‐cell competence.38
secretion and caused hyperglycaemia in chronically glucose‐infused Overall, these data suggest the possibility that at the pancreatic
rats by raising glucose hepatic production and increasing the ‘basal islet level, T2D is an example of ‘paracrinopathy’39 in which distur-
workload’ of β‐cells until their exhaustion. In turn, Solomon et al.32 bances in the physiological secretion of glucagon and insulin are both
showed that experimental hyperglycaemia acutely impaired β‐cell the cause and consequence of each other (Figure 1).
function but not α‐cell glucagon secretion in normal glucose‐tolerant
subjects. In type 1 diabetes, the total loss of β‐cell function was
accompanied by complete dysregulation of glucagon secretion, while 2.2 | The role of glucagon in peripheral tissues
in T2D with residual β‐cell function, hyperglucagonemia did not occur
in all patients.5 In fact, glucagon release from pure human α‐cells be- A growing body of evidence currently indicates that glucagon exerts
comes affected by glucose only if α‐cells are reaggregated with β‐ its effect in several organs, including the liver, the adipose tissue, and,
33
cells. Conversely, it has been demonstrated that α‐cell mass in- marginally, the skeletal muscle, thus affecting peripheral insulin
creases during the development of insulin resistance and sensitivity (Figure 2).

F I G U R E 1 The role of glucagon in β‐cell function under physiological (upper panels) and diabetic (lower panel) conditions. Upper panel,
left: under physiological fasting, the potentiation of insulin secretion from glucagon cannot occur due to the non‐activation of the triggering
pathway by the glucose. Upper panel, right: in the physiological post‐prandial phase, high glucose levels depolarise the β‐cell membrane by
closing the ATP‐sensitive K+ leak channels (KATP channel), thus activating the insulin secretory machinery; this triggering pathway allows the
glucagon to amplify insulin secretion by activating the GCGRs and GLP‐1Rs and favoring the generation of cAMP, and the activation of the
EPAC system; in this phase, however, glucagon levels are kept low by glucose and the counter‐regulatory action of insulin. Lower panel:
regardless of fasting or post‐prandial condition, in type 2 diabetes, hyperglucagonemia occurs as a result of both insulin resistance and
alteration of the glucose effect on the α‐cell; high glucagon levels together with high glucose levels abnormally stimulate insulin secretion in
the β‐cell; therefore, after a phase of compensatory hyperinsulinemia, the β‐cell becomes exhausted and no longer able to release insulin in
concentrations sufficient to maintain euglycemia. cAMP, cyclic AMP; EPAC, exchange protein directly activated by cAMP; GCGRs, glucagon
receptors; GLP‐1Rs, glucagon‐like peptide‐1 receptors.
15207560, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/dmrr.3609 by Cochrane Germany, Wiley Online Library on [30/01/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
4 of 16
- CARUSO ET AL.

2.2.1 | Liver glucagon enhances hepatic lipolysis, sustains cytosolic gluconeogen-


esis, and increases calcium‐dependent mitochondrial energy expen-
The first activity attributed to glucagon was its activation of diture (EE), reducing ectopic lipids.46,47 In vitro studies demonstrated
hepatic glycogenolysis and gluconeogenesis to counteract hypo- that glucagon is also able to stimulate ketogenesis by enhancing
glycaemia.40 In particular, glucagon binds to GCGR resulting in hepatic fatty acid oxidation especially under low circulating insulin
the activation of glycogen phosphorylase kinase and glycogen levels, increasing β‐hydroxybutyrate and acetoacetate production by
phosphorylase through the cAMP‐PKA pathway, allowing the about 150%.48
breakdown of glycogen and generation of glucose‐6‐phosphate, Hence, the hepatic functions of glucagon are activation of
the key substrate for glucose production.40 Moreover, glucagon glycogenolysis, gluconeogenesis and lipolysis, increasing mitochon-
fosters the activity of the glucose 6‐phosphatase (G6Pase) drial EE, and inhibition of glycogenosynthesis.
enzyme increasing glucose release and inhibits glycogen syn-
thase.41 Also, a transcriptome‐wide analysis of human hepatocytes
showed that glucagon might facilitate gluconeogenesis by acti- 2.2.2 | Muscle
vating a plethora of target genes of proliferator‐activated recep-
tor gamma coactivator 1‐alpha (PGC1α).42 In vivo experiments in rodents found that glucagon administration for
Hepatic intracellular signalling pathways of insulin and glucagon 6 h inhibited protein synthesis in muscles following a period of
converge on Foxo1, a mediator involved in glucose and lipid meta- feeding.49 Indeed, glucagon may promote the oxidation of L‐leucine
43 44
bolism, with opposite effects. Glucagon induces the phosphory- in human skeletal muscle under different metabolic conditions,
lation of Foxo1 on Ser276 determining its nuclear retention via a determining a whole‐body irreversible loss of this amino acid.50,51 L‐
cAMP and PKA‐dependent pathway, impairing the insulin‐induced leucine oxidation leads to reduced activation of target of rapamycin
Foxo1 ubiquitination and degradation.45 (TOR) protein and TOR complex 1 (TORC1),52 hindering muscle
New insights on glucagon biology in the liver showed that protein synthesis as observed in patients with severe muscle wasting
through the inositol 1,4,5‐triphosphate receptor type 1 (INSP3R1) affected by glucagonoma53 and hyperglucagonaemia.54

F I G U R E 2 Principal peripheral responses evoked by glucagon. Principal peripheral responses evoked by glucagon: (i) glycogenolytic/
gluconeogenic and anti‐glycogenic effects on hepatocytes; (ii) reduction of muscle mass; (iii) browning and increased BAT activity; (iv)
stimulation of brain areas controlling anorexigenic responses; (v) uncertain effect on ghrelin secretion and gut contractility. ↑, increase; ↓,
decrease. AgRP, agouti‐related protein; ARC, arcuate nucleus; cAMP, cyclic AMP; CaMKKβ/AMPK, Ca2+−calmodulin‐dependent protein
kinase kinase β/AMP‐activated protein kinase; FGF21, fibroblast growth factor 21; Foxo1, forkhead box protein O1; GCGR, glucagon receptor;
GP, glycogen phosphorylase; GPK, glycogen phosphorylase kinase; G6Pase, glucose 6‐phosphatase; GS, glycogen synthase; pACC,
phosphorylated acetyl‐CoA carboxylase; PKA, protein kinase a; TORC1, target of rapamycin complex 1.
15207560, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/dmrr.3609 by Cochrane Germany, Wiley Online Library on [30/01/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
CARUSO ET AL.
- 5 of 16

Beyond the effects on muscle mass, conflicting evidence hints at induced markers of browning (e.g. Ucp1, Prdm16, and Cidea), as
an indirect effect of glucagon on glucose metabolism in the skeletal well as lower circulating levels of FGF21.28 Similarly, Townsend
muscle. A study conducted in depancreatised dogs on nutritional et al. showed that mice with WAT‐specific GCGR knockout dis-
support reported that chronic exposure to high glucagon levels played attenuated induction of Ucp1, Cidea, and Fgf21 mRNA levels
impaired muscle glucose disposal, fuelling the development of insulin under cold exposure, despite a modest increase in browning indices
resistance as well as the aggravation of glucose tolerance.55 In at room temperature, providing evidence that glucagon signalling is
humans, intravenous infusion of glucagon has often been associated required for cold acclimation.67
with enhanced glucose uptake during positron emission tomography Finally, recent findings suggested that glucagon might play an
(PET), yet a retrospective study by Yasuda et al. failed to detect any anti‐adipogenic role, inhibiting adipose stem cell proliferation,
increase in glucose uptake in the gluteal muscle.56 increasing apoptosis and reducing accumulated lipids.68 Of note,
In light of these data, the regulation of protein synthesis appears supraphysiological concentrations of glucagon were used in these in
to be a definite effect of glucagon in skeletal muscle, whereas the role vitro experiments (10–100 nM), higher than those achieved in the
of this hormone in the control of glucose metabolism in this tissue presence of obesity and diabetes.69,70 However, the anti‐lipogenic
needs further elucidation. and catabolic actions of glucagon had already been observed in
mature adipocytes, in which the hormone led to increased phos-
phorylation of acetyl‐CoA carboxylase (ACC) which in turn dimin-
2.2.3 | Adipose tissue (WAT, BAT) ished lipid synthesis via the activation of the Ca2+−calmodulin‐
dependent protein kinase β/AMP‐activated protein kinase (CaMKKβ/
Long‐standing evidence supports the hypothesis that glucagon might AMPK) cascade.71
increase EE even though the underlying mechanism is still debated. Human studies in healthy volunteers confirmed that glucagon
One of the main contributors to EE is brown adipose tissue (BAT)‐ infusion may increase EE by enhancing carbohydrate and FFAs
mediated thermogenesis in which process glucagon may play a crit- oxidation.72 Yet, Salem et al. hypothesised the presence of other still
ical role.57 unidentified pathways through which glucagon might affect EE, as a
18
Early evidence indicated that glucagon increased the rate of F‐FDG PET/CT study in healthy male volunteers showed that
oxygen consumption and free fatty acids (FFAs) release from BAT58 glucagon increased EE to the same extent as cold exposure but
as well as inducing heat generation.59 In addition, glucagon appears without any changes in sympathetic activation, BAT thermogenesis
to be involved in the mechanisms of cold tolerance. In particular, and glucose uptake.73
glucagon secretion changes according to ambient temperature, as Taken together, these data highlight the weight‐lowering po-
observed in both rodents and humans, with increased circulating60 tential of glucagon, through increases in EE, enhancement of BAT
and within BAT61 glucagon levels under cold temperatures. Indeed, thermogenesis and probably other pathways yet to be discovered, as
mice with the abrogation of proglucagon‐derived peptides (GCGKO) well as anti‐adipogenic and lipolytic effects.
exhibited a greater decrease in rectal temperature as well as poor
response to β‐adrenergic stimuli and reduced expression of ther-
mogenic proteins (e.g. uncoupling protein 1 [UCP1], iodothyronine 2.3 | The role of glucagon in food intake regulation
deiodinase 2, and PGC1α) under cold exposure, all restored after
glucagon supplementation.62 2.3.1 | Central
The effect of glucagon on EE is mediated by multiple direct and
indirect GCGR‐dependent mechanisms.63 Glucagon actions on BAT Glucagon is also able to affect the brain regions involved in the
might be mediated via the central control of sympathetic activity, as control of satiety and hunger. Indeed, GCGR is widely expressed in
revealed by the fact that experimental denervation of BAT or phar- the central nervous system (CNS)74 and several studies have ascribed
macological inhibition of β‐adrenergic signalling, the main transducer an anorexigenic role to glucagon, supporting its pharmacological use
of BAT activity, led to a consistent reduction of glucagon‐induced in the treatment of obesity.75 In mice, intracerebroventricular infu-
64
augmentation of EE. Also, animal studies hinted at fibroblast sion of glucagon exerted a weight‐lowering effect by decreasing
growth factor 21 (FGF21) as a mediator of glucagon‐induced BAT appetite.76 Accordingly, rats treated with antibodies against glucagon
activation,65 though this finding was not replicated in humans.66 exhibited increased meal sizes.77 These effects appear to be medi-
Interestingly, results in mice with BAT‐specific GCGR knockdown ated by a hepatic‐neuronal loop: in a rat model, the activation of
indicated that the beneficial effects of glucagon on body weight (BW), hypothalamic glucagon signalling prevented hyperglycaemia through
EE, and metabolic adaptation to cold and high‐fat diet do not require the inhibition of hepatic glucose output via vagal nerve pulse control,
63
BAT GCGR and thermogenic mediators such as UCP‐1. suggesting that hypothalamic glucagon resistance might contribute
Moreover, several studies have demonstrated that glucagon to the disruption of glucose homoeostasis in diabetes.78 Similarly,
may activate browning within the white adipose tissue (WAT), even animals with hepatic vagotomy did not experience modifications in
though with a negligible clinical impact. In mice, WAT‐specific their feeding behaviour after central infusion of glucagon, suggesting
GCGR abrogation was associated with lower levels of cold‐ the essential role of the hepatic branch and the CNS areas dedicated
15207560, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/dmrr.3609 by Cochrane Germany, Wiley Online Library on [30/01/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
6 of 16
- CARUSO ET AL.

to the regulation of food consumption in promoting glucagon‐ 3 | THE ROLE OF GLUCAGON IN THE
induced satiety.79,80 PATHOGENESIS OF TYPE 2 DIABETES
Recent in vivo studies have also reported that glucagon displayed COMPLICATIONS
anorexigenic actions by regulating the PKA/Ca2+−calmodulin‐
dependent protein kinase kinase β/AMP‐activated protein kinase/ 3.1 | The role of glucagon in CV complications of
Agouti related protein (PKA/CaMKKβ/AMPK/AgRP) signalling type 2 diabetes
pathway within the hypothalamic arcuate nucleus (ARC).81 However,
the anorexigenic effects of glucagon are lost in an obesogenic envi- Expression of the GCGR has been detected in certain regions of the
ronment, as in rats fed a high fat diet, due to a disruption in its hy- heart57,90,91 and several animal and human experiments have
81
pothalamic signalling downstream of PKA. In the same model, demonstrated that glucagon exerts direct cardiac actions.25,26
glucagon action was restored after inactivation of CaMKKβ in the Glucagon facilitated the atrio‐ventricular conduction, exhibiting a
ARC, suggesting the critical role of this protein in mediating obesity‐ positive chronotropic and anti‐arrhythmogenic effect.25,92 In addi-
81
induced glucagon resistance. tion, glucagon also exerted a positive inotropic effect, which is
Considering these central effects, glucagon may reasonably play stronger in the ventricle rather than in the atrium,25 partially due to
a role in the management of obesity and prevention of T2D. site‐specific differential GCGR expression.93 These actions are
mediated by increased cAMP concentration and the inotropic effect
persists in the presence of β‐blockers.25,94 Being an inotropic and
2.3.2 | Peripheral (stomach and intestine) chronotropic agent, glucagon increases cardiac oxygen consumption,
lipolysis, and lipid β‐oxidation.91 In animal studies, cardiac infusion of
GCGRs have been detected throughout the gastrointestinal tract, glucagon‐promoted glycolysis and glucose oxidation, like insulin, and
where glucagon acts through the activation of a cAMP‐dependent these effects were mediated via phosphatidylinositol 3‐kinase‐
pathway. In particular, an in vivo analysis indicated that GCGRs are dependent and adenylate cyclase‐ and cAMP‐independent path-
predominantly expressed in the lamina propria mucosae of rat ways.25,95 Hence, cardiac glucagon and insulin actions may overlap in
stomach. Glucagon intravenous infusion or direct administration to regard to the regulation of fuel metabolism.25
the isolated stomach was associated with acute release and increased These findings suggest to consider glucagon as a useful resource
synthesis of ghrelin, an important orexigenic hormone involved in in the treatment of heart failure.96 Nonetheless, while glucagon
food intake regulation.82,83 Similarly, Gagnon et al. demonstrated infusion improved cardiac index and left ventricular ejection fraction
that glucagon induces the release of ghrelin from a dispersed gastric in patients without heart failure undergoing diagnostic coronary
cell culture model through a mechanism dependent on mitogen‐ catheterisation,97 evidence on a direct inotropic effect in patients
84
activated protein kinases and EPAC. Contrarywise, glucagon with low cardiac output is lacking.98 However, glucagon was associ-
administration was associated with the inhibition of ghrelin secretion ated with reduced peripheral and lung vascular resistances, reducing
65,85
in human studies. cardiac workload and indirectly improving cardiac performance.98
Also, glucagon appears to regulate the enteric activity. The Knowledge about the direct effects of glucagon on the heart under
spasmolytic action of glucagon has been well‐documented by pathological conditions is still limited and controversial, with heart‐
several early studies, demonstrating that its continuous intravenous specific activation of GCGR signalling reported to be either benefi-
infusion resulted in jejunal dilatation and increased mean transit cial or harmful, depending on the experimental or clinical context.99 Ali
86,87
time in humans, independent of its effect on carbohydrate et al. demonstrated that exogenous glucagon administration directly
metabolism.84 Nevertheless, Mochiki et al. observed that glucagon impaired the recovery of ventricular pressure in nondiabetic ischaemic
might also increase phase III‐like contractions in the duodenum and mouse hearts ex vivo and increased mortality from myocardial
jejunum, suggesting a potential side effect of glucagon‐based ther- infarction (MI) after left anterior descending coronary artery ligation
apy.84 The enteric effects of glucagon appeared to be related to the in mice in a p38 MAPK‐dependent manner.99 Accordingly,
dose used as well as to the route of administration. Indeed, Pascaud cardiomyocyte‐specific inactivation of GCGR significantly improved
et al. observed that low concentrations of glucagon (25 μg/kg) overall survival and reduced hypertrophy and infarct size following
stimulated contractile activity in the jejunum, while higher doses MI.99 These effects were associated with a marked reduction in long‐
(100 μg/kg) induced an immediate and significant decrease in chain acylcarnitines in both aerobic and ischaemic hearts following
motility.88 Also, clinical trials suggested that the anti‐peristaltic ef- high‐fat feeding, suggesting an essential role of GCGR signalling in the
fect of glucagon was more evident following intravenous rather control of cardiac fatty acid utilisation.99 Moreover, Gao et al.
than intramuscular administration.89 observed that treating mice with the anti‐GCGR antibody REMD 2.59
Overall, the effects of glucagon on the gastroenteric system following MI was able to blunt cardiac hypertrophy, fibrotic remod-
remain elusive. Expanding the knowledge regarding the effects of elling and contractile dysfunction after 4 weeks.100 In addition, the
glucagon on intestinal motility could be useful to understand the administration of REMD 2.59 at the onset of or after pressure over-
relative implication of glucagon‐based therapies on nutrient ab- load significantly reduced cardiac hypertrophy and chamber dilation
sorption and gastric fullness perception. with marked preservation of cardiac systolic and diastolic function.100
15207560, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/dmrr.3609 by Cochrane Germany, Wiley Online Library on [30/01/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
CARUSO ET AL.
- 7 of 16

Similarly, Sharma et al. demonstrated that treatment with REMD 2.59 the thick ascending tubule, distal tubule and collecting duct.108
preserved fractional shortening and ejection fraction in the Leprdb/db Glucagon induces glomerular hyperfiltration by modulating the
101
T2D mice model. Furthermore, GCGR antagonism was also able to characteristics of the tubular fluid at the macula densa by simulta-
prevent declines in the early diastolic filling peak velocity/late filling neously reducing the resorption of water, electrolytes and urea in the
peak velocity (E/A) ratio, cardiac output, as well as left ventricular proximal tubule and increasing NaCl resorption in the thick
developed pressure.101 REMD 2.59‐treated Leprdb/db mice were also ascending tubule, decreasing the tubulo‐glomerular feedback (TGF)
protected from the increase in isovolumic relaxation time (IVRT) and signal, and increasing glomerular filtration rate (GFR).109 While the
101
T2D‐associated hypertrophy. Also, REMD 2.59 lowered several effect of glucagon on the thick ascending tubule is directly mediated
toxic ceramide species, including C16:0 and C18:0 ceramides, and was by the GCGR, the effect on the proximal tubule is mediated by the
associated with a trend towards improved AMPK activation involved circulating cAMP produced by the liver under the influence of
101
in cardiac lipid metabolism. glucagon and insulin.109 Insulin facilitates the degradation of
Finally, it should be recalled that relative hyperglucagonemia is glucagon‐induced cAMP, hence, it is the insulin/glucagon ratio rather
involved in the pathogenesis of glycaemic variability, which repre- than the absolute concentration of glucagon to determine the
sents a predictor of all‐cause mortality, especially non‐cancer mor- amount of circulating cAMP and the extent of the indirect effects of
102–104
tality, in patients with T2D. In addition, hyperglucagonemia glucagon.108
also impairs lipid and amino acid metabolism, which in turn, along Of note, studies using mouse GCGR knockout models reported
with glycaemic disorders, fuel the pathogenesis of diabetes, obesity, no major impairments of kidney function.90 In non‐diabetic rodents,
non‐alcoholic fatty liver disease (NAFLD)105 and the related adverse infusion of glucagon was associated with moderately increased sys-
106
CV outcomes. tolic blood pressure, high fasting blood glucose and impaired glucose
These experimental data suggest that hyperglucagonemia might tolerance, glomerular hypertrophy and hyperfiltration, due to blunted
play a role in the CV complications of T2D triggering lipotoxic con- TGF,109 increased albuminuria, mesangial expansion and deposition
ditions and thus the development of insulin resistance, providing of extracellular matrix.110 These effects were mediated by the
proof‐of‐concept evidence that GCGR antagonism might be poten- phosphorylation of ERK1/2 and Akt, under euglycaemic conditions,
tially efficacious in preventing heart failure and ischaemic heart following the activation of PKA and the phospholipase C (PLC)
disease. pathways leading to angiotensin II synthesis.106,110 Angiotensin II is a
powerful contributor to the synthesis of extracellular matrix in
mesangial cells.111 Also, glucagon has been shown to exert a positive
3.2 | The role of glucagon in microvascular effect on renal blood flow.90 Interestingly, in rats with
complications of type 2 diabetes streptozotocin‐induced diabetes mellitus, the injection of glucagon
antibodies significantly reduced GFR within 2 h without effects on
In mice, the knockout of the GCGR gene was associated with a late‐ glycaemia, confirming the role of glucagon as a direct mediator of
onset loss of retinal function, visual acuity, and eventually retinal cell glomerular hyperfiltration also in the setting of diabetes.109
90
death. Likewise, deficiency of peptides derived from the glucagon Of note, glucagon relies almost entirely on renal clearance, rising
gene was associated with peripheral neuropathy in mice.107 Indeed, by 2‐to‐4‐fold in end‐stage renal disease.112 Accordingly, a Chinese
glucagon could exert a protective effect on the nervous system, as study conducted on 326 T2D individuals confirmed the linear in-
107
shown in the setting of post‐traumatic brain injury. In a model of crease in fasting plasma glucagon levels across the stages of chronic
diabetic neuropathy, represented by neuronal cells from the dorsal kidney disease (CKD).112 In 357 T2D patients, Wang et al. found that,
root ganglion (DRG) treated with methylglyoxal (MG), which causes after a 75 g glucose oral load, glucagon levels were significantly
intracellular accumulation of advanced glycated end products, reac- higher in patients with albuminuria and/or reduced eGFR, and the
tive oxygen species (ROS) and apoptosis, glucagon mitigated MG‐ glucagon peak was delayed compared to patients with preserved
induced mitochondrial ROS production, apoptosis and overall cyto- kidney function.113 In addition, the glucagon/insulin ratio was higher
107
toxicity. Additionally, treatment of DRG neurons with physiolog- in patients with eGFR <60 ml/min/1.73 m2 compared to those with
ical doses of glucagon, in the absence of MG, stimulated the cAMP/ >60 ml/min/1.73 m2.113 The degree of insulin resistance and eGFR
PKA pathway and promoted neurite outgrowth.107 explained 22.8% of the variance in fasting glucagon levels.112 Hence,
However, there are no clinical data on the role of glucagon in the higher levels of glucagon in nephropathic T2D patients might be
diabetic retinopathy and neuropathy, while a larger body of evidence explained by both the altered regulatory effects of insulin and
covers the relationship between glucagon and kidney. glucose on pancreatic α‐cells and the decreased renal clearance of
Glucagon is involved in the disposal of the nitrogen waste glucagon.113 This association was confirmed in a wider cohort of
products of protein metabolism, stimulating ureagenesis and gluco- 2436 T2D patients undergoing an oral glucose tolerance test: both
neogenesis from amino acids in the liver and urea excretion in the fasting and post‐challenge glucagon were mildly correlated with
kidney following a protein‐rich meal or amino acids infusion.108,109 eGFR and urinary albumin:creatinine ratio (UACR).106 Moreover,
The renal effects of glucagon are only partially mediated by patients in the higher quartile of fasting and post‐challenge plasma
GCGR, which is abundantly expressed in the kidney, specifically in glucagon exhibited a 1.52‐ and 2.06‐fold increase in the likelihood of
15207560, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/dmrr.3609 by Cochrane Germany, Wiley Online Library on [30/01/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
8 of 16
- CARUSO ET AL.

diabetic kidney disease compared to those in the lower quartile, oxidative stress and intracellular triglyceride accumulation, fuelling a
respectively.106 This association persisted even in the subgroup of vicious circle that sustains the progression of NAFLD.117
106
patients with optimal glycaemic control (HbA1c <7%). Some T2D patients have high fasting and post‐prandial levels of
In another study on 209 T2D patients, the increase in plasma glucagon, but this could be related to the degree of hepatic steatosis
glucose following glucagon injection (glucagon stimulation test) not rather than diabetes per se.118 Indeed, Junker et al. demonstrated
only was directly related to GFR at baseline, but was also indepen- that patients with biopsy‐proven NAFLD had hyperglucagonemia
dently associated with eGFR after 1 year at multiple regression regardless of the presence of T2D.123 In 4937 T2D patients, Wang
114
analysis. Therefore, responses to the glucagon stimulation test et al. found that higher glucagon levels were associated with prob-
might allow us to evaluate not only the hepatic glucose output but able inflammatory progression of NAFLD to NASH but not to wors-
also nephron functionality, as gluconeogenesis in the kidney also ening fibrosis; however, probable NASH was defined by the
114
contributes. simultaneous presence of NAFLD and metabolic syndrome, and
All in all, hyperglucagonemia might contribute to kidney disease by fibrosis was assessed with the NAFLD fibrosis score rather than
both glucose‐dependent and glucose‐independent mechanisms. providing histological confirmation.124 Accordingly, in a small study
Indeed, hyperglucagonemia is associated not only with fasting hyper- conducted on non‐obese Asian males with new‐onset T2D treated
glycaemia but also with GCGR‐dependent proliferation of glomerular with metformin, fasting glucagon levels were higher in patients with
mesangial cells and increased deposition of extracellular matrix.106 NAFLD and were independently associated with markers of trunkal
Further mechanistic studies are warranted to further understand the subcutaneous and visceral adiposity.125 Nonetheless, in a cross‐
role of glucagon in the pathogenesis of diabetic kidney disease. sectional study conducted on 172 hospitalised patients with T2D,
relative hyperinsulinemia, expressed by lower fasting and post-
prandial glucagon‐to‐insulin ratios, was significantly associated with
3.3 | The role of glucagon in NAFLD ultrasound‐detected NAFLD.126
Interestingly, in 33 obese individuals undergoing bariatric sur-
NAFLD is defined by the detection of hepatic steatosis either on gery, the liver‐α cell axis was similarly disrupted irrespective of
imaging or histology in the absence of secondary causes of hepatic NAFLD severity, whereas at 12 months after surgery only patients
lipid accumulation.115 T2D is a major risk factor for NAFLD and its with NASH or fibrosis at baseline exhibited persistent hyper-
progression to NASH (non‐alcoholic steatohepatitis).115 A recent glucagonemia despite significant improvements in BW and liver his-
meta‐analysis estimated 55.48% and 37.33% worldwide prevalence tology. This suggests a role of structural liver abnormalities and/or
115
of NAFLD and NASH in T2D patients, respectively. Conversely, mechanisms of metabolic memory in determining glucagon levels.127
the prevalence of T2D is 22.5% and 43.6% in patients with NAFLD As demonstrated in a pancreatic clamp study in patients with
and NASH, respectively, which is much higher than the 8.5% preva- biopsy‐proven NAFLD versus lean individuals, the presence of
lence in general population.116 NAFLD hampered the effects of glucagon on amino acid metabolism
Glucagon is involved in the liver‐α‐cell axis, regulating urea- but not on glucose metabolism,117,121 probably due to the fact that
genesis and hepatic amino acid metabolism, while in turn amino acids the glycogenolytic pathway is clearly separated from that of gluco-
stimulate pancreatic α‐cells proliferation and glucagon secretion.117 neogenesis and ureagenesis.118 The glucagon‐alanine index, obtained
Moreover, glucagon increases endogenous glucose production mainly by multiplying glucagon and alanine plasma concentrations, has been
by activating glycogenolysis rather than gluconeogenesis and induces suggested as a marker of glucagon resistance.118 In 79 young women,
117,118
lipolysis, fatty acid β‐oxidation, and autophagy. most of whom had preserved glucose tolerance, Gar et al. found that
Notably, in an animal model of high fat diet‐induced hepatic the glucagon‐alanine index was associated with MRI‐determined
steatosis, GCGR density and glucagon‐mediated intracellular signal- hepatic steatosis; this association was independent of the degree of
ling in hepatocytes were reduced, hampering liver response to insulin resistance and was already detectable at slightly increased
glucagon.117,119 Using several animal models, Winther‐Sørensen et al. liver fat content (>0.5%).128 As expected, the glucagon‐alanine index
showed that hepatic steatosis, GCGR antagonists and genetic was also significantly related to fasting and post‐prandial glucose
knockout of GCGR similarly increased glucagon levels and reduced (PPG) levels and insulin resistance (HOMA‐IR).128
ureagenesis through both transcriptional and rapid non‐ Impaired glucagon signalling may be held accountable of the
120
transcriptional mechanisms. Fat‐induced hepatic glucagon resis- bidirectional relationship between NAFLD and diabetes,117 contrib-
tance results in reduced ureagenesis, leading to hyperaminoacidemia uting to the pathogenesis and progression of NAFLD alongside
which in turn induces hyperglucagonemia.117,121 A weakened biolog- obesity, insulin resistance, and inflammation.129 Hence, restoration of
ical effect of glucagon is associated with reduced SIRT‐1 activity, glucagon signalling could be beneficial in NAFLD, as suggested by the
consequently reducing its anti‐inflammatory and anti‐fibrotic effects, weight loss and reversal of hepatic steatosis produced by chronic
down‐regulating NF‐κB and modulating TGF‐β signalling pathway.122 activation of the GCGR in diet‐induced obese mice130 and by the
Furthermore, hyperaminoacidemia and hepatic glucagon resistance encouraging evidence arising from clinical studies with dual or triple
contribute to impaired hepatocyte autophagy, resulting in increased agonists activating glucagon signalling.
15207560, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/dmrr.3609 by Cochrane Germany, Wiley Online Library on [30/01/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
CARUSO ET AL.
- 9 of 16

4 | USE OF GLUCAGON IN TYPE 2 DIABETES BW, food intake, HbA1c and lipids compared to GLP‐1RA.137 The
THERAPY similarities in primary and secondary structures between glucagon,
GLP‐1 and GIP allowed the synthesis of hybridised molecules with
Until recently, glucagon as a therapeutic agent had been recom- multiple epitope regions and a size comparable to that of the native
mended solely in the management of severe hypoglycaemia, now peptides.137 These compounds can be characterised by balanced or
available in a nasal, needle‐free formulation (Baqsimi®) for the preferential agonism in favour of one receptor over the others.137
131
management of severe hypoglycaemic episodes. Glucagon, GLP‐1 and GIP exert their effects by binding to different G
On the other hand, in agreement with the ‘bi‐hormonal theory’ protein‐coupled receptors; though cross‐reactivity has been
accounting for the dysregulation of glucagon secretion and function demonstrated, their differential tissue‐specific distribution facilitate
in T2D, GCGR antagonists (GRAs) have been proposed as glucose‐ a broader synergistic effect.137
lowering agents.2 Interestingly, short‐term use of several effica- Nowadays, both dual and triple agonists incorporating glucagon
cious anti‐diabetic drugs, such as metformin, DPP‐4 inhibitors and agonism are under development for clinical use.75,131,137 Experi-
GLP‐1 receptor agonists (GLP‐1RAs), has been associated with mental studies conducted in diet‐induced obese mice showed that
reduced glucagon levels132,133 without hindering the physiological dual GLP‐1/GCGR agonists induced weight loss, improved lipid and
1
response to hypoglycaemia. In experimental models, the inhibition glycaemic profile, increased EE, and reduced food intake.75,131 In
of GCGR has been associated with reduced plasma glucose and tri- humans, co‐infusion studies confirmed the favourable effects of this
glycerides and increased GLP‐1 levels.134 GRAs can be small mole- therapeutic strategy. In healthy overweight or obese volunteers, co‐
cules or antibodies interfering with the GCGR or antisense infusion of glucagon and GLP‐1 allowed glucagon‐dependent in-
131,134
oligonucleotides reducing the expression of the GCGR. Further crease in EE and the reduction of food intake without affecting
clinical development of GRAs was hampered by long‐term adverse plasma glucose excursion.72,76 Among the many GLP‐1/GCGR ago-
events such as increased BW, liver fat content, systolic blood pres- nists under development,131 the balanced agonist cotadutide
sure, LDL‐cholesterol, and transaminase levels, associated with α‐ (MEDI0382) is the most advanced. The phase 1 trial demonstrated a
cells hyperplasia and severe hypoglycaemia.131,134 Also, GRA dose‐dependent reduction in blood glucose and food intake in
135
LY2409021 did not improve PPG following a mixed meal test and healthy individuals, with gastrointestinal adverse events at the
paradoxically reduced glucose tolerance following an oral glucose highest dose.139 In phase 2a randomised, placebo‐controlled, double‐
tolerance test without restoring the incretin effect compared to blind trials conducted in overweight and obese T2D patients, cota-
placebo in 10 T2D patients.136 GCGR antagonism was associated dutide reduced HbA1c and both fasting and PPG following a mixed
with stable fasting insulin levels, despite the amelioration of glucose meal tolerance test at 41–49 days, as opposed to long‐acting GLP‐
control, and increased insulin levels during oral glucose tolerance 1RA reducing mainly fasting glucose.140,141 Furthermore, up to 40%
test with a subsequent greater post‐load decrease in glucose, sug- of patients lost ≥5% of their BW.140,141 Enhanced insulin secretion
gesting improved insulin sensitivity both in the fasting and fed and delayed gastric emptying have been proposed as the main
134
states. Further research is required to understand the mechanisms mechanisms behind short‐term amelioration of glucose control with
underlying GRA adverse events and their clinical relevance before cotadutide.140 In a phase 2b study, cotadutide was similar to lir-
approval for use. aglutide 1.8 mg in terms of HbA1c lowering both at 14 and 54 weeks,
Surprisingly, in the context of incretin‐based multiagonists, while only cotadutide 300 μg outperformed liraglutide 1.8 mg in
agonism of GCGR could also be beneficial for T2D management.137 terms of weight loss both at 14 (LS mean [95% CI] – 5.01 [−5.57,
GLP‐1RAs have been proven safe and effective in achieving glucose −4.45] vs. −3.44 [−4.20, −2.68], p = 0.001) and 54 weeks (LS mean
control and preventing some diabetes complications, such as CV dis- [95% CI] – 5.02 [−5.78, −4.26] vs. −3.33,142 p = 0.009).143 Inter-
eases. Their gastrointestinal side effects may limit the use of higher estingly, a recent phase 2a trial showed that treatment with cota-
137
doses to maximise weight loss and glucose control. The combi- dutide for 32 days was associated with a 51% decrease in albumin‐
nation of GLP‐1 with glucagon and/or glucose‐dependent insulino- to‐creatinin ratio compared to placebo in T2D patients with
tropic polypeptide (GIP) agonism opens thrilling possibilities in the albuminuria.144
field of obesity and diabetes management.75,137 Indeed, glucagon may Another dual GLP‐1/GCGR agonist, SAR425899, was associated
enhance the effects of GLP‐1RAs on satiety and further promote with a greater post‐prandial glycaemic control compared to liraglu-
weight loss by increasing EE, thermogenesis and lipolysis.137 In tide, due to enhanced β‐cell responsiveness.145
addition, glucagon and GLP‐1 both facilitate insulin secretion and the Moreover, in order to mimic the effects of GLP‐1, GIP, and
combination of the two agonisms may allow to counteract the glucagon contributing to the major metabolic advantage of bariatric
glucagon‐dependent increase in hepatic glucose output.137 The syn- surgery, the development of triple GLP‐1/GIP/GCGR agonists has
ergistic effects of glucagon and GLP‐1 are not surprising, being both been pursued.131 Adding both incretins to glucagon hampers its ef-
138
encoded by the Gcg gene. The addition of GIP to these multi- fect on hepatic glucose output, allowing to maximise its salutary ef-
agonists is corroborated by human studies highlighting that co‐ fects on liver and BW.75 Indeed, in diet‐induced obese rodents, the
infusion of GIP and GLP‐1 increased first‐phase insulin secretion in administration of a triple agonist was associated with a greater
diabetic patients, and animal studies showing a greater reduction of benefit on weight loss and glucose levels than liraglutide and a
15207560, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/dmrr.3609 by Cochrane Germany, Wiley Online Library on [30/01/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
10 of 16
- CARUSO ET AL.

GLP‐1/GIP coagonist, comparable to that observed following bar- to increased EE,57 anti‐adipogenic and lipolytic effects.68,71 Also,
iatric surgery in the same experimental setting. human studies suggested that glucagon may inhibit the secretion of
Besides their remarkable effects on glucose control and BW the orexigenic peptide hormone ghrelin,152 but this association and
reduction, the combination of GLP‐1 and GCGR monoagonists or its involvement in the modulation of gastrointestinal motility are still
GLP‐1, GIP and GCGR monoagonists has been associated with a controversial.
greater improvement than high dose liraglutide in NAFLD activity Accordingly, the role of glucagon in the development of T2D
146
score despite producing the same extent of weight loss. An complications is cryptic (Figures 3 and 4). Glucagon administration
exploratory MRI analysis of a phase 2a trial demonstrated that was able to induce glomerular hyperfiltration and hypertrophy,
administration of cotadutide reduced hepatic fat content in over- mesangial expansion, and albuminuria with both glucose‐dependent
weight or obese T2D patients compared to placebo.147 Nahra et al. and independent mechanisms,110 while the restoration of GCGR
showed that cotadutide 200 and 300 μg significantly lowered signalling has strongly been associated with the prevention of NAFLD
aspartate (AST) and alanine (ALT) transaminases compared to pla- worsening130 (Figure 4).
cebo, while only cotadutide 300 μg induced a greater reduction in The role of glucagon in the CV system is still elusive because
ALT levels compared to liraglutide 1.8 mg at 54 weeks.143,148 In the despite encouraging preliminary evidence regarding its anti‐
same study, liraglutide 1.8 mg and all investigated doses of cotadu- arrhythmic and inotropic effects, glucagon administration has not
tide produced a greater decrease in fatty liver index with respect to been proven beneficial in patients with low cardiac output.98
placebo. Instead, higher doses of cotadutide reduced FIB‐4 (fibrosis‐4 Interestingly, GCGR antagonism has been associated with improved
index), NFS (NAFLD fibrosis score) and PRO‐C3 (propeptide of type systolic and diastolic performance, and amelioration of fibrosis and
145
III collagen), while liraglutide had no effect on these endpoints. hypertrophy in several pathological settings.99 Additional studies
Indeed, in mice models of NASH, treatment with cotadutide using both GCGR agonism and antagonism, as well as GCGR
improved liver fibrosis to a greater extent than liraglutide despite ablation models, are needed to shed light on the biology of
inducing a similar weight loss.149 The beneficial effects of cotadutide glucagon, which still appears enigmatic not only from a patho-
in NAFLD/NASH are likely to be mediated by the GCGR‐dependent physiological perspective but also from a therapeutic point of view.
enhancement of mitochondrial turnover and reduced de novo Finally, the paucity of studies investigating the effect of glucagon
lipogenesis.145 signalling in the development of microvascular complications does
Finan et al. showed that a balanced GLP‐1/GIP/GCGR triple not allow to draw conclusions, but a beneficial role of GCGR
agonist induced amelioration of liver fat content and hepatocytes agonism in peripheral neuropathy and retinopathy could be
vacuolation to a greater extent than GLP‐1/GIP dual agonists.150 cautiously hypothesised.
More recently, in mice models of NASH, the triple agonist Most of the commonly used antidiabetic drugs, except SGLT‐
LY3437943 decreased circulating ALT levels and hepatic tri- 2i, are able to lower glucagon levels,1 and yet the development of
glycerides content. Accordingly, the triple agonist HM15211 out- targeted GRA has been halted by long‐term complications.134
performed both acylated GLP‐1 and GLP‐1/GIP on all the Instead, the development of dual and triple GLP‐1/GCGR and
subcomponents of the NAFLD activity score (hepatocytes ballooning, GLP‐1/GIP/GCGR agonists has been welcomed enthusiastically,
lobular inflammation, and steatosis).148 particularly due to significant weight‐ and glucose‐lowering po-
These results encourage the use of such dual and triple agonists tential and the amelioration of hepatic steatosis.137 Clinical studies
for the treatment of NAFLD/NASH. Further studies on patients with conducted so far failed to detect relevant adverse events other
biopsy‐proven NAFLD/NASH and fibrosis are awaited. than the expected mild‐to‐moderate nausea, but further evidence
is awaited.75
In the light of the future availability of drugs able to modulate
5 | CONCLUSIONS GCGR signalling, a better understanding of the involvement of
glucagon in the pathogenesis of T2D and particularly of its tissue‐
Increased glucagon secretion by α‐cells is one of the eight compo- specific effects and role in diabetes complications is eagerly awaited.
nents of the ‘ominous octet’ of factors underlying T2D pathogen-
esis.151 It is still unclear whether hyperglucagonemia results from β‐ A UT H O R CO N T R I B U T I O N S
cell dysfunction and its inability to suppress α‐cell secretion or from Conceptualisation: Irene Caruso. Writing – original draft preparation:
α‐cells insulin resistance. In this regard, T2D could likely be consid- Irene Caruso, Nicola Marrano, Giuseppina Biondi, Valentina Anna-
ered as a ‘paracrinopathy’, where α‐ and β‐cells influence each other maria Genchi and Rossella D'Oria. Writing – review and editing: Irene
in a vicious circle. Hyperglucagonemia sustains hyperglycaemia by Caruso, Angelo Cignarelli, Annalisa Natalicchio and Francesco Gior-
151
increasing hepatic glucose output, however its role in T2D path- gino. Supervision: Angelo Cignarelli, Gian Pio Sorice, Annalisa Nata-
ogenesis might not be as straightforward (Figure 3). Indeed, GCGR licchio, Sebastio Perrini, Luigi Laviola and Francesco Giorgino. All
agonism harbours a relevant weight‐lowering potential since it exerts authors have read and agreed to the published version of the
anorexigenic actions on the hypothalamic ARC81 and has been linked manuscript.
15207560, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/dmrr.3609 by Cochrane Germany, Wiley Online Library on [30/01/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
CARUSO ET AL.
- 11 of 16

F I G U R E 3 The role of glucagon in the pathogenesis of T2D and its complications: friend or foe? Glucagon has both favourable and
unfavourable effects on some of the mechanisms underlying the pathogenesis of T2D. Glucagon increases glucose‐dependent insulin secretion,
favourably affecting post‐prandial excursions. It might also contribute to controlling body weight, by regulating satiety through the reduction
of ghrelin levels and gastrointestinal motility in a dose‐dependent manner, and by increasing energy expenditure, likely via enhanced browning
and thermogenesis. Conversely, glucagon sustains hepatic glucose output contributing to fasting plasma glucose increase. It also reduces
peripheral insulin sensitivity by increasing lipolysis and probably reducing glucose uptake by skeletal muscles. The role of glucagon in T2D
complications is still vague. The reduction of peripheral vascular resistance is likely beneficial in patients with heart failure; however, clinical
evidence supporting its role in heart failure management is lacking and preclinical studies showed that glucagon might mediate cardiac fibrosis,
hypertrophy, and diastolic dysfunction. Glucagon might reduce the progression of NAFLD and improve peripheral diabetic neuropathy.
Conversely, glucagon has been associated with glomerular hyperfiltration and mesangial proliferation, while its effects on hard kidney
outcomes are still unknown. BW, body weight; DPN, diabetic peripheral neuropathy; FPG, fasting plasma glucose; GI, gastrointestinal; HF,
heart failure; MI, myocardial infarction; NAFLD, non‐alcoholic fatty liver disease; PPG, post‐prandial glucose; T2D, type 2 diabetes; ↑, increase;
↓, decrease.

F I G U R E 4 The putative role of glucagon in T2D complications. Green arrow, GCGR agonism; Red arrow, GCGR antagonism or knock‐out
or deficiency of glucagon gene‐derived peptides. GCGR, glucagon receptor; NAFLD, non‐alcoholic fatty liver disease; NASH, non‐alcoholic
steatohepatitis; NDRG, neurons from the dorsal root ganglion; T2D, type 2 diabetes.
15207560, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/dmrr.3609 by Cochrane Germany, Wiley Online Library on [30/01/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
12 of 16
- CARUSO ET AL.

A CK N O WL ED GE M EN T S 7. Huising MO. Paracrine regulation of insulin secretion. Diabetologia.


This research received no external funding. 2020;63(10):2057‐2063. https://doi.org/10.1007/s00125‐020‐05
213‐5
8. Bertuzzi F, Berra C, Socci C, et al. Glucagon improves insulin
C O N F L IC T S O F IN T ER E S T secretion from pig islets in vitro. J Endocrinol. 1995;147(1):87‐93.
I.C., N.M., G.B., V.A.G., R.D., S.P., A.C. and G.P.S. declare no relevant https://doi.org/10.1677/joe.0.1470087
conflict of interest. A.N. has received speaker fees from AstraZeneca, 9. Pipeleers D, In't Veld PI, Maes E, Van De Winkel M. Glucose‐
induced insulin release depends on functional cooperation be-
Novo Nordisk, and Sanofi. L.L. received speaker fees from Abbott,
tween islet cells. Proc Natl Acad Sci USA. 1982;79(23):7322‐7325.
AstraZeneca, Boehringer Ingelheim, Eli Lilly, Medtronic, Menarini, https://doi.org/10.1073/pnas.79.23.7322
Merck Sharp & Dohme, Novo Nordisk, Roche Diabetes Care, and 10. Huypens P, Ling Z, Pipeleers D, Schuit F. Glucagon receptors on
Sanofi and provided advisory services to Abbott, AstraZeneca, human islet cells contribute to glucose competence of insulin
release. Diabetologia. 2000;43(8):1012‐1019. https://doi.org/10.
Boehringer Ingelheim, Eli Lilly, Novo Nordisk, Roche Diabetes Care,
1007/s001250051484
Sanofi and Takeda. F.G. provided advisory services to AstraZeneca, Eli 11. Trimble ER, Halban PA, Wollheim CB, Renold AE. Functional differ-
Lilly, Novo Nordisk. Roche Diabetes Care, Sanofi, received speaker ences between rat islets of ventral and dorsal pancreatic origin. J Clin
fees, served as a consultant for Boehringer Ingelheim, Lifescan, Merck Invest. 1982;69(2):405‐413. https://doi.org/10.1172/jci110464
Sharp & Dohme, Sanofi, AstraZeneca, MedImmune, Roche Diabetes 12. Reissaus CA, Piston DW. Reestablishment of glucose inhibition of
glucagon secretion in small pseudoislets. Diabetes. 2017;66(4):
Care, Sanofi, Medtronic and received research support from Eli Lilly
960‐969. https://doi.org/10.2337/db16‐1291
and Roche Diabetes Care. 13. Capozzi ME, Svendsen B, Encisco SE, et al. β cell tone is defined by
proglucagon peptides through cAMP signaling. JCI Insight. 2019;4(5).
D A TA A V AIL AB IL I T Y S T A T EM E N T https://doi.org/10.1172/jci.insight.126742
14. Zhu L, Dattaroy D, Pham J, et al. Intra‐islet glucagon signaling is
Data sharing not applicable to this article as no datasets were
critical for maintaining glucose homeostasis. JCI Insight. 2019;5(10).
generated or analysed during the current study. https://doi.org/10.1172/jci.insight.127994
15. Thorel F, Damond N, Chera S, et al. Normal glucagon signaling and β‐
E T H IC S S T AT E M E N T cell function after near‐total α‐cell ablation in adult mice. Diabetes.
2011;60(11):2872‐2882. https://doi.org/10.2337/db11‐0876
Ethics approval is not applicable because the present study is based
16. DiGruccio MR, Mawla AM, Donaldson CJ, et al. Comprehensive
solely on published literature. alpha, beta and delta cell transcriptomes reveal that ghrelin selec-
tively activates delta cells and promotes somatostatin release from
O R C ID pancreatic islets. Mol Metab. 2016;5(7):449‐458. https://doi.org/10.
1016/j.molmet.2016.04.007
Valentina Annamaria Genchi https://orcid.org/0000-0001-8188-
17. Noguchi GM, Huising MO. Integrating the inputs that shape
9638 pancreatic islet hormone release. Nat Metab. 2019;1(12):1189‐1201.
Gian Pio Sorice https://orcid.org/0000-0001-9489-1534 https://doi.org/10.1038/s42255‐019‐0148‐2
Francesco Giorgino https://orcid.org/0000-0001-7372-2678 18. Moede T, Leibiger IB, Berggren PO. Alpha cell regulation of beta cell
function. Diabetologia. 2020;63(10):2064‐2075. https://doi.org/10.
1007/s00125‐020‐05196‐3
PE ER R E VI E W 19. Capozzi ME, Wait JB, Koech J, et al. Glucagon lowers glycemia
The peer review history for this article is available at https://publons. when β‐cells are active. JCI Insight. 2019;5(16). https://doi.org/10.
com/publon/10.1002/dmrr.3609. 1172/jci.insight.129954
20. Svendsen B, Larsen O, Gabe MBN, et al. Insulin secretion depends
REFERENCES on intra‐islet glucagon signaling. Cell Rep. 2018;25(5):1127‐1134.
e2. https://doi.org/10.1016/j.celrep.2018.10.018
1. Hædersdal S, Lund A, Knop FK, Vilsbøll T. The role of glucagon in
21. Chambers AP, Sorrell JE, Haller A, et al. The role of pancreatic
the pathophysiology and treatment of type 2 diabetes. Mayo Clin
preproglucagon in glucose homeostasis in mice. Cell Metab. 2017;
Proc. 2018;93(2):217‐239. https://doi.org/10.1016/j.mayocp.2017.
25(4):927‐934.e3. https://doi.org/10.1016/j.cmet.2017.02.008
12.003
22. Gelling RW, Vuguin PM, Du XQ, et al. Pancreatic beta‐cell over-
2. Unger RH, Orci L. The essential role of glucagon in the pathogenesis
expression of the glucagon receptor gene results in enhanced beta‐
of diabetes mellitus. Lancet. 1975;1(7897):14‐16. https://doi.org/10.
cell function and mass. Am J Physiol Endocrinol Metab. 2009;297(3):
1016/s0140‐6736(75)92375‐2
E695‐E707. https://doi.org/10.1152/ajpendo.00082.2009
3. Marrano N, Biondi G, Cignarelli A, et al. Functional loss of pancreatic
23. Maharaj A, Zhu L, Huang F, et al. Ectopic expression of glucagon
islets in type 2 diabetes: how can we halt it? Metabolism. 2020;
receptor in skeletal muscles improves glucose homeostasis in a
110:154304. https://doi.org/10.1016/j.metabol.2020.154304
mouse model of diabetes. Diabetologia. 2012;55(5):1458‐1468.
4. Cabrera O, Berman DM, Kenyon NS, Ricordi C, Berggren PO, Cai-
https://doi.org/10.1007/s00125‐012‐2464‐x
cedo A. The unique cytoarchitecture of human pancreatic islets has
24. Shah P, Vella A, Basu A, Basu R, Schwenk WF, Rizza RA. Lack of
implications for islet cell function. Proc Natl Acad Sci USA. 2006;
suppression of glucagon contributes to postprandial hyperglycemia
103(7):2334‐2339. https://doi.org/10.1073/pnas.0510790103
in subjects with type 2 diabetes mellitus. J Clin Endocrinol Metab.
5. Gilon P. The role of α‐cells in islet function and glucose homeostasis
2000;85(11):4053‐4059. https://doi.org/10.1210/jcem.85.11.6993
in health and type 2 diabetes. J Mol Biol. 2020;432(5):1367‐1394.
25. Ali S, Drucker DJ. Benefits and limitations of reducing glucagon
https://doi.org/10.1016/j.jmb.2020.01.004
action for the treatment of type 2 diabetes. Am J Physiol Endocrinol
6. Cooperberg BA, Cryer PE. Insulin reciprocally regulates glucagon
Metab. 2009;296(3):E415‐E421. https://doi.org/10.1152/ajpendo.
secretion in humans. Diabetes. 2010;59(11):2936‐2940. https://doi.
90887.2008
org/10.2337/db10‐0728
15207560, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/dmrr.3609 by Cochrane Germany, Wiley Online Library on [30/01/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
CARUSO ET AL.
- 13 of 16

26. Ahrén B. Glucagon – early breakthroughs and recent discoveries. 43. Lu M, Wan M, Leavens KF, et al. Insulin regulates liver metabolism in
Peptides. 2015;67:74‐81. https://doi.org/10.1016/j.peptides.2015. vivo in the absence of hepatic Akt and Foxo1. Nat Med. 2012;18(3):
03.011 388‐395. https://doi.org/10.1038/nm.2686
27. Menge BA, Grüber L, Jørgensen SM, et al. Loss of inverse rela- 44. Rena G, Guo S, Cichy SC, Unterman TG, Cohen P. Phosphorylation
tionship between pulsatile insulin and glucagon secretion in pa- of the transcription factor forkhead family member FKHR by
tients with type 2 diabetes. Diabetes. 2011;60(8):2160‐2168. protein kinase B. J Biol Chem. 1999;274(24):17179‐17183. https://
https://doi.org/10.2337/db11‐0251 doi.org/10.1074/jbc.274.24.17179
28. Henquin JC, Ibrahim MM, Rahier J. Insulin, glucagon and somato- 45. Wu Y, Pan Q, Yan H, et al. Novel mechanism of foxo1 phosphor-
statin stores in the pancreas of subjects with type‐2 diabetes and ylation in glucagon signaling in control of glucose homeostasis.
their lean and obese non‐diabetic controls. Sci Rep. 2017;7(1): Diabetes. 2018;67(11):2167‐2182. https://doi.org/10.2337/db18‐
11015. https://doi.org/10.1038/s41598‐017‐10296‐z 0674
29. Lee Y, Berglund ED, Yu X, et al. Hyperglycemia in rodent models of 46. Perry RJ. Regulation of hepatic lipid and glucose metabolism by
type 2 diabetes requires insulin‐resistant alpha cells. Proc Natl Acad INSP3R1. Diabetes. 2022;71(9):1834‐1841. https://doi.org/10.
Sci USA. 2014;111(36):13217‐13222. https://doi.org/10.1073/pnas. 2337/dbi22‐0003
1409638111 47. Perry RJ, Zhang D, Guerra MT, et al. Glucagon stimulates gluco-
30. Merino B, Casanueva‐Álvarez E, Quesada I, et al. Insulin‐degrading neogenesis by INSP3R1‐mediated hepatic lipolysis. Nature.
enzyme ablation in mouse pancreatic alpha cells triggers cell pro- 2020;579(7798):279‐283. https://doi.org/10.1038/s41586‐020‐
liferation, hyperplasia and glucagon secretion dysregulation. Dia- 2074‐6
betologia. 2022;65(8):1375‐1389. https://doi.org/10.1007/s00125‐ 48. Pégorier JP, Garcia‐Garcia MV, Prip‐Buus C, Duée PH, Kohl C,
022‐05729‐y Girard J. Induction of ketogenesis and fatty acid oxidation by
31. Jamison RA, Stark R, Dong J, et al. Hyperglucagonemia precedes a glucagon and cyclic AMP in cultured hepatocytes from rabbit fe-
decline in insulin secretion and causes hyperglycemia in chronically tuses. Evidence for a decreased sensitivity of carnitine palmitoyl-
glucose‐infused rats. Am J Physiol Endocrinol Metab. 2011;301(6): transferase I to malonyl‐CoA inhibition after glucagon or cyclic
E1174‐E1183. https://doi.org/10.1152/ajpendo.00175.2011 AMP treatment. Biochem J. 1989;264(1):93‐100. https://doi.org/10.
32. Solomon TP, Knudsen SH, Karstoft K, Winding K, Holst JJ, Ped- 1042/bj2640093
ersen BK. Examining the effects of hyperglycemia on pancreatic 49. Preedy VR, Garlick PJ. The effect of glucagon administration on
endocrine function in humans: evidence for in vivo glucotoxicity. J protein synthesis in skeletal muscles, heart and liver in vivo.
Clin Endocrinol Metab. 2012;97(12):4682‐4691. https://doi.org/10. Biochem J. 1985;228(3):575‐581. https://doi.org/10.1042/bj228
1210/jc.2012‐2097 0575
33. Liu W, Kin T, Ho S, et al. Abnormal regulation of glucagon secretion 50. Hartl WH, Miyoshi H, Jahoor F, Klein S, Elahi D, Wolfe RR. Bra-
by human islet alpha cells in the absence of beta cells. EBioMedicine. dykinin attenuates glucagon‐induced leucine oxidation in humans.
2019;50:306‐316. https://doi.org/10.1016/j.ebiom.2019.11.018 Am J Physiol. 1990;259(2 Pt 1):E239‐E245. https://doi.org/10.
34. Liu Z, Kim W, Chen Z, et al. Insulin and glucagon regulate pancreatic 1152/ajpendo.1990.259.2.E239
α‐cell proliferation. PLoS One. 2011;6(1):e16096. https://doi.org/10. 51. Pacy PJ, Cheng KN, Ford GC, Halliday D. Influence of glucagon on
1371/journal.pone.0016096 protein and leucine metabolism: a study in fasting man with
35. Ellingsgaard H, Ehses JA, Hammar EB, et al. Interleukin‐6 regulates induced insulin resistance. Br J Surg. 1990;77(7):791‐794. https://
pancreatic alpha‐cell mass expansion. Proc Natl Acad Sci USA. 2008; doi.org/10.1002/bjs.1800770723
105(35):13163‐13168. https://doi.org/10.1073/pnas.0801059105 52. Adeva‐Andany MM, Fernández‐Fernández C, López‐Pereiro Y,
36. Sørensen H, Winzell MS, Brand CL, et al. Glucagon receptor Castro‐Calvo I, Carneiro‐Freire N. The effects of glucagon and the
knockout mice display increased insulin sensitivity and impaired target of rapamycin (TOR) on skeletal muscle protein synthesis and
beta‐cell function. Diabetes. 2006;55(12):3463‐3469. https://doi. age‐dependent sarcopenia in humans. Clin Nutr ESPEN. 2021;44:
org/10.2337/db06‐0307 15‐25. https://doi.org/10.1016/j.clnesp.2021.06.025
37. Vuguin PM, Kedees MH, Cui L, et al. Ablation of the glucagon re- 53. Boden G, Tappy L, Jadali F, Hoeldtke RD, Rezvani I, Owen OE. Role
ceptor gene increases fetal lethality and produces alterations in of glucagon in disposal of an amino acid load. Am J Physiol. 1990;
islet development and maturation. Endocrinology. 2006;147(9): 259(2 Pt 1):E225‐E232. https://doi.org/10.1152/ajpendo.1990.259.
3995‐4006. https://doi.org/10.1210/en.2005‐1410 2.E225
38. Wang Q, Liang X, Wang S. Intra‐islet glucagon secretion and action 54. Thiessen SE, Gunst J, Van den Berghe G. Role of glucagon in pro-
in the regulation of glucose homeostasis. Front Physiol. 2012;3:485. tein catabolism. Curr Opin Crit Care. 2018;24(4):228‐234. https://
https://doi.org/10.3389/fphys.2012.00485 doi.org/10.1097/MCC.0000000000000509
39. Unger RH, Orci L. Paracrinology of islets and the paracrinopathy of 55. Chen SS, Zhang Y, Santomango TS, Williams PE, Lacy DB,
diabetes. Proc Natl Acad Sci USA. 2010;107(37):16009‐16012. McGuinness OP. Glucagon chronically impairs hepatic and muscle
https://doi.org/10.1073/pnas.1006639107 glucose disposal. Am J Physiol Endocrinol Metab. 2007;292(3):
40. Wewer Albrechtsen NJ, Kuhre RE, Pedersen J, Knop FK, Holst JJ. E928‐E935. https://doi.org/10.1152/ajpendo.00063.2006
The biology of glucagon and the consequences of hyper- 56. Yasuda S, Takechi M, Ono M, et al. The effect of glucagon on
glucagonemia. Biomark Med. 2016;10(11):1141‐1151. https://doi. FDG uptake in skeletal muscle. Tokai J Exp Clin Med. 2012;37(1):
org/10.2217/bmm‐2016‐0090 11‐13.
41. Ciudad C, Camici M, Ahmad Z, Wang Y, DePaoli‐Roach AA, Roach 57. Habegger KM, Heppner KM, Geary N, Bartness TJ, DiMarchi R,
PJ. Control of glycogen synthase phosphorylation in isolated rat Tschöp MH. The metabolic actions of glucagon revisited. Nat Rev
hepatocytes by epinephrine, vasopressin and glucagon. Eur J Bio- Endocrinol. 2010;6(12):689‐697. https://doi.org/10.1038/nrendo.
chem. 1984;142(3):511‐520. https://doi.org/10.1111/j.1432‐1033. 2010.187
1984.tb08315.x 58. Rudman D, Del Rio AE. Responsiveness to lipolytic hormones,
42. Tavares CDJ, Aigner S, Sharabi K, et al. Transcriptome‐wide anal- and inactivation of adrenocorticotropin, by adipose tissue slices
ysis of PGC‐1α‐binding RNAs identifies genes linked to glucagon and free fat cells from different mammalian species. Endocri-
metabolic action. Proc Natl Acad Sci USA. 2020;117(36):22204‐- nology. 1969;85(2):209‐213. https://doi.org/10.1210/endo‐85‐2‐
22213. https://doi.org/10.1073/pnas.2000643117 209
15207560, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/dmrr.3609 by Cochrane Germany, Wiley Online Library on [30/01/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
14 of 16
- CARUSO ET AL.

59. Kuroshima A, Yahata T. Thermogenic responses of brown adipo- 76. Tan TM, Field BC, McCullough KA, et al. Coadministration of
cytes to noradrenaline and glucagon in heat‐acclimated and cold‐ glucagon‐like peptide‐1 during glucagon infusion in humans results
acclimated rats. Jpn J Physiol. 1979;29(6):683‐690. https://doi. in increased energy expenditure and amelioration of hyperglycemia.
org/10.2170/jjphysiol.29.683 Diabetes. 2013;62(4):1131‐1138. https://doi.org/10.2337/db12‐
60. Seitz HJ, Krone W, Wilke H, et al. Rapid rise in plasma glucagon 0797
induced by acute cold exposure in man and rat. Pflugers Arch. 77. Le Sauter J, Noh U, Geary N. Hepatic portal infusion of glucagon
1981;389(2):115‐120. https://doi.org/10.1007/BF00582100 antibodies increases spontaneous meal size in rats. Am J Physiol.
61. Habara Y, Kuroshima A. Changes in glucagon and insulin contents 1991;261(1 Pt 2):R162‐R165. https://doi.org/10.1152/ajpregu.
of brown adipose tissue after temperature acclimation in rats. Jpn 1991.261.1.R162
J Physiol. 1983;33(4):661‐665. https://doi.org/10.2170/jjphysiol. 78. Mighiu PI, Yue JT, Filippi BM, et al. Hypothalamic glucagon
33.661 signaling inhibits hepatic glucose production. Nat Med. 2013;19(6):
62. Kinoshita K, Ozaki N, Takagi Y, Murata Y, Oshida Y, Hayashi Y. 766‐772. https://doi.org/10.1038/nm.3115
Glucagon is essential for adaptive thermogenesis in brown adipose 79. Geary N, Le Sauter J, Noh U. Glucagon acts in the liver to control
tissue. Endocrinology. 2014;155(9):3484‐3492. https://doi.org/10. spontaneous meal size in rats. Am J Physiol. 1993;264(1 Pt 2):
1210/en.2014‐1175 R116‐R122. https://doi.org/10.1152/ajpregu.1993.264.1.R116
63. Beaudry JL, Kaur KD, Varin EM, et al. The brown adipose tissue 80. Weatherford SC, Ritter S. Lesion of vagal afferent terminals impairs
glucagon receptor is functional but not essential for control of glucagon‐induced suppression of food intake. Physiol Behav. 1988;
energy homeostasis in mice. Mol Metab. 2019;22:37‐48. https://doi. 43(5):645‐650. https://doi.org/10.1016/0031‐9384(88)90220‐x
org/10.1016/j.molmet.2019.01.011 81. Quiñones M, Al‐Massadi O, Gallego R, et al. Hypothalamic
64. Billington CJ, Bartness TJ, Briggs J, Levine AS, Morley JE. Glucagon CaMKKβ mediates glucagon anorectic effect and its diet‐induced
stimulation of brown adipose tissue growth and thermogenesis. Am resistance. Mol Metab. 2015;4(12):961‐970. https://doi.org/10.
J Physiol. 1987;252(1 Pt 2):R160‐R165. https://doi.org/10.1152/ 1016/j.molmet.2015.09.014
ajpregu.1987.252.1.R160 82. Kamegai J, Tamura H, Shimizu T, Ishii S, Sugihara H, Oikawa S.
65. Habegger KM, Stemmer K, Cheng C, et al. Fibroblast growth factor Effects of insulin, leptin, and glucagon on ghrelin secretion from
21 mediates specific glucagon actions. Diabetes. 2013;62(5): isolated perfused rat stomach. Regul Pept. 2004;119(1–2):77‐81.
1453‐1463. https://doi.org/10.2337/db12‐1116 https://doi.org/10.1016/j.regpep.2004.01.012
66. Whytock KL, Carnero EA, Vega RB, et al. Prolonged glucagon infu- 83. Katayama T, Shimamoto S, Oda H, Nakahara K, Kangawa K, Mur-
sion does not affect energy expenditure in individuals with over- akami N. Glucagon receptor expression and glucagon stimulation
weight/obesity: a randomized trial. Obesity. 2021;29(6):1003‐1013. of ghrelin secretion in rat stomach. Biochem Biophys Res Commun.
https://doi.org/10.1002/oby.23141 2007;357(4):865‐870. https://doi.org/10.1016/j.bbrc.2007.04.001
67. Townsend LK, Medak KD, Knuth CM, Peppler WT, Charron MJ, 84. Mochiki E, Suzuki H, Takenoshita S, et al. Mechanism of inhibitory
Wright DC. Loss of glucagon signaling alters white adipose tissue effect of glucagon on gastrointestinal motility and cause of side
browning. FASEB J. 2019;33(4):4824‐4835. https://doi.org/10. effects of glucagon. J Gastroenterol. 1998;33(6):835‐841. https://
1096/fj.201802048RR doi.org/10.1007/s005350050184
68. Cantini G, Trabucco M, Di Franco A, Mannucci E, Luconi M. 85. Soule S, Pemberton C, Hunt P, Cole D, Raudsepp S, Inder W.
Glucagon modulates proliferation and differentiation of human Prandial regulation of ghrelin secretion in humans: does glucagon
adipose precursors. J Mol Endocrinol. 2019;63(4):249‐260. https:// contribute to the preprandial increase in circulating ghrelin? Clin
doi.org/10.1530/JME‐19‐0095 Endocrinol. 2005;63(4):412‐417. https://doi.org/10.1111/j.1365‐
69. Stern JH, Smith GI, Chen S, Unger RH, Klein S, Scherer PE. Obesity 2265.2005.02357.x
dysregulates fasting‐induced changes in glucagon secretion. J 86. Patel GK, Whalen GE, Soergel KH, Wu WC, Meade RC. Glucagon
Endocrinol. 2019;243(2):149‐160. https://doi.org/10.1530/JOE‐19‐ effects on the human small intestine. Dig Dis Sci. 1979;24(7):
0201 501‐508. https://doi.org/10.1007/BF01489316
70. Reaven GM, Chen YD, Golay A, Swislocki AL, Jaspan JB. Docu- 87. Ponce J, Garrigues V, Pertejo V, et al. Effect of intravenous
mentation of hyperglucagonemia throughout the day in nonobese glucagon and glucagon‐(1‐21)‐peptide on motor activity of
and obese patients with noninsulin‐dependent diabetes mellitus. J sphincter of Oddi in humans. Dig Dis Sci. 1989;34(1):61‐64. https://
Clin Endocrinol Metab. 1987;64(1):106‐110. https://doi.org/10. doi.org/10.1007/BF01536155
1210/jcem‐64‐1‐106 88. Pascaud X, Ferre JP, Genton M, Roger A, Ruckebusch M, Bueno L.
71. Peng IC, Chen Z, Sun W, et al. Glucagon regulates ACC activity in Intestinal motility responses to insulin and glucagon in streptozo-
adipocytes through the CAMKKβ/AMPK pathway. Am J Physiol tocin diabetic rats. Can J Physiol Pharmacol. 1982;60(7):960‐967.
Endocrinol Metab. 2012;302(12):E1560‐E1568. https://doi.org/10. https://doi.org/10.1139/y82‐135
1152/ajpendo.00504.2011 89. Gutzeit A, Binkert CA, Koh DM, et al. Evaluation of the anti‐
72. Cegla J, Troke RC, Jones B, et al. Coinfusion of low‐dose GLP‐1 and peristaltic effect of glucagon and hyoscine on the small bowel:
glucagon in man results in a reduction in food intake. Diabetes. comparison of intravenous and intramuscular drug administration.
2014;63(11):3711‐3720. https://doi.org/10.2337/db14‐0242 Eur Radiol. 2012;22(6):1186‐1194. https://doi.org/10.1007/s003
73. Salem V, Izzi‐Engbeaya C, Coello C, et al. Glucagon increases en- 30‐011‐2366‐1
ergy expenditure independently of brown adipose tissue activation 90. Charron MJ, Vuguin PM. Lack of glucagon receptor signaling and
in humans. Diabetes Obes Metab. 2016;18(1):72‐81. https://doi.org/ its implications beyond glucose homeostasis. J Endocrinol. 2015;
10.1111/dom.12585 224(3):R123‐R130. https://doi.org/10.1530/JOE‐14‐0614
74. Campos RV, Lee YC, Drucker DJ. Divergent tissue‐specific and 91. Godoy‐Matos AF. The role of glucagon on type 2 diabetes at a
developmental expression of receptors for glucagon and glucagon‐ glance. Diabetol Metab Syndr. 2014;6(1):91. https://doi.org/10.
like peptide‐1 in the mouse. Endocrinology. 1994;134(5): 1186/1758‐5996‐6‐91
2156‐2164. https://doi.org/10.1210/endo.134.5.8156917 92. Farah AE. Glucagon and the circulation. Pharmacol Rev. 1983;
75. Del Prato S, Gallwitz B, Holst JJ, Meier JJ. The incretin/glucagon 35(3):181‐217.
system as a target for pharmacotherapy of obesity. Obes Rev. 93. Gonzalez‐Muñoz C, Nieto‐Cerón S, Cabezas‐Herrera J, Hernán-
2022;23(2):e13372. https://doi.org/10.1111/obr.13372 dez‐Cascales J. Glucagon increases contractility in ventricle but
15207560, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/dmrr.3609 by Cochrane Germany, Wiley Online Library on [30/01/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
CARUSO ET AL.
- 15 of 16

not in atrium of the rat heart. Eur J Pharmacol. 2008;587(1‐3): 111. Li XC, Zhuo JL. Targeting glucagon receptor signalling in treating
243‐247. https://doi.org/10.1016/j.ejphar.2008.04.001 metabolic syndrome and renal injury in type 2 diabetes: theory
94. Ceriello A, Genovese S, Mannucci E, Gronda E. Glucagon and heart versus promise. Clin Sci. 2007;113(4):183‐193. https://doi.org/10.
in type 2 diabetes: new perspectives. Cardiovasc Diabetol. 2016; 1042/CS20070040
15(1):123. https://doi.org/10.1186/s12933‐016‐0440‐3 112. Liu JJ, Liu S, Gurung RL, et al. Relationship between fasting plasma
95. Harney JA, Rodgers RL. Insulin‐like stimulation of cardiac fuel glucagon level and renal function – a cross‐sectional study in in-
metabolism by physiological levels of glucagon: involvement of dividuals with type 2 diabetes. J Endocr Soc. 2019;3(1):273‐283.
PI3K but not cAMP. Am J Physiol Endocrinol Metab. 2008;295(1): https://doi.org/10.1210/js.2018‐00321
E155‐E161. https://doi.org/10.1152/ajpendo.90228.2008 113. Wang X, Yang J, Chang B, et al. Glucagon secretion is increased in
96. Müller TD, Finan B, Clemmensen C, DiMarchi RD, Tschöp MH. The patients with type 2 diabetic nephropathy. J Diabetes Complicat.
new biology and pharmacology of glucagon. Physiol Rev. 2017; 2016;30(3):488‐493. https://doi.org/10.1016/j.jdiacomp.2015.
97(2):721‐766. https://doi.org/10.1152/physrev.00025.2016 12.020
97. Linhart JW, Barold SS, Cohen LS, Hildner FJ, Samet P. Cardiovas- 114. Takeda Y, Fujita Y, Bessho R, et al. Increment of plasma glucose by
cular effects of glucagon in man. Am J Cardiol. 1968;22(5):706‐710. exogenous glucagon is associated with present and future renal
https://doi.org/10.1016/0002‐9149(68)90209‐9 function in type 2 diabetes: a retrospective study from glucagon
98. Hernández‐Cascales J. Does glucagon have a positive inotropic stimulation test. BMC Endocr Disord. 2019;19(1):99. https://doi.org/
effect in the human heart? Cardiovasc Diabetol. 2018;17(1):148. 10.1186/s12902‐019‐0428‐6
https://doi.org/10.1186/s12933‐018‐0791‐z 115. Younossi ZM, Golabi P, de Avila L, et al. The global epidemiology of
99. Ali S, Ussher JR, Baggio LL, et al. Cardiomyocyte glucagon receptor NAFLD and NASH in patients with type 2 diabetes: a systematic
signaling modulates outcomes in mice with experimental myocar- review and meta‐analysis. J Hepatol. 2019;71(4):793‐801. https://
dial infarction. Mol Metab. 2015;4(2):132‐143. https://doi.org/10. doi.org/10.1016/j.jhep.2019.06.021
1016/j.molmet.2014.11.005 116. Khandelwal R, Dassanayake AS, Conjeevaram HS, Singh SP. Non‐
100. Gao C, Ren SV, Yu J, et al. Glucagon receptor antagonism amelio- alcoholic fatty liver disease in diabetes: when to refer to the
rates progression of heart failure. JACC Basic Transl Sci. 2019; hepatologist? World J Diabetes. 2021;12(9):1479‐1493. https://doi.
4(2):161‐172. https://doi.org/10.1016/j.jacbts.2018.11.001 org/10.4239/wjd.v12.i9.1479
101. Sharma AX, Quittner‐Strom EB, Lee Y, et al. Glucagon receptor 117. Galsgaard KD. The vicious circle of hepatic glucagon resistance in
antagonism improves glucose metabolism and cardiac function by non‐alcoholic fatty liver disease. J Clin Med. 2020;9(12):4049.
promoting AMP‐mediated protein kinase in diabetic mice. Cell https://doi.org/10.3390/jcm9124049
Rep. 2018;22(7):1760‐1773. https://doi.org/10.1016/j.celrep.2018. 118. Wewer Albrechtsen NJ, Pedersen J, Galsgaard KD, et al. The liver‐
01.065 α‐cell axis and type 2 diabetes. Endocr Rev. 2019;40(5):1353‐1366.
102. Dunning BE, Gerich JE. The role of alpha‐cell dysregulation in https://doi.org/10.1210/er.2018‐00251
fasting and postprandial hyperglycemia in type 2 diabetes and 119. McGlone ER, Ansell TB, Dunsterville C, et al. Hepatocyte choles-
therapeutic implications. Endocr Rev. 2007;28(3):253‐283. https:// terol content modulates glucagon receptor signalling. Mol Metab.
doi.org/10.1210/er.2006‐0026 2022;63:101530. https://doi.org/10.1016/j.molmet.2022.101530
103. Takao T, Matsuyama Y, Yanagisawa H, Kikuchi M, Kawazu S. As- 120. Winther‐Sørensen M, Galsgaard KD, Santos A, et al. Glucagon
sociation between HbA1c variability and mortality in patients with acutely regulates hepatic amino acid catabolism and the effect may
type 2 diabetes. J Diabetes Complicat. 2014;28(4):494‐499. https:// be disturbed by steatosis. Mol Metab. 2020;42:101080. https://doi.
doi.org/10.1016/j.jdiacomp.2014.02.006 org/10.1016/j.molmet.2020.101080
104. Cryer PE. Minireview: glucagon in the pathogenesis of hypoglyce- 121. Suppli MP, Bagger JI, Lund A, et al. Glucagon resistance at the level
mia and hyperglycemia in diabetes. Endocrinology. 2012;153(3): of amino acid turnover in obese subjects with hepatic steatosis.
1039‐1048. https://doi.org/10.1210/en.2011‐1499 Diabetes. 2020;69(6):1090‐1099. https://doi.org/10.2337/db19‐
105. Janah L, Kjeldsen S, Galsgaard KD, et al. Glucagon receptor 0715
signaling and glucagon resistance. Int J Mol Sci. 2019;20(13):3314. 122. Zhang L, Yao W, Xia J, Wang T, Huang F. Glucagon‐induced acet-
https://doi.org/10.3390/ijms20133314 ylation of energy‐sensing factors in control of hepatic metabolism.
106. Huang HX, Shen LL, Huang HY, et al. Associations of plasma Int J Mol Sci. 2019;16(8):20. https://doi.org/10.3390/ijms20081885
glucagon levels with estimated glomerular filtration rate, albu- 123. Junker AE, Gluud L, Holst JJ, Knop FK, Vilsbøll T. Diabetic and
minuria and diabetic kidney disease in patients with type 2 dia- nondiabetic patients with nonalcoholic fatty liver disease have an
betes mellitus. Diabetes Metab J. 2021;45(6):868‐879. https://doi. impaired incretin effect and fasting hyperglucagonaemia. J Intern
org/10.4093/dmj.2020.0149 Med. 2016;279(5):485‐493. https://doi.org/10.1111/joim.12462
107. Mohiuddin MS, Himeno T, Yamada Y, et al. Glucagon prevents 124. Wang Y, Lin Z, Wan H, et al. Glucagon is associated with NAFLD
cytotoxicity induced by methylglyoxal in a rat neuronal cell line inflammatory progression in type 2 diabetes, not with NAFLD
model. Biomolecules. 2021;11(2):287. https://doi.org/10.3390/ fibrotic progression. Eur J Gastroenterol Hepatol. 2021;33(1S Suppl
biom11020287 1):e818‐e823. https://doi.org/10.1097/MEG.0000000000002269
108. Bankir L, Bouby N, Speth RC, Velho G, Crambert G. Glucagon 125. Anoop S, Misra A, Bhatt SP, Gulati S, Mahajan H, Prabakaran G. High
revisited: coordinated actions on the liver and kidney. Diabetes Res plasma glucagon levels correlate with waist‐to‐hip ratio, suprailiac
Clin Pract. 2018;146:119‐129. https://doi.org/10.1016/j.diabres. skinfold thickness, and deep subcutaneous abdominal and intra-
2018.10.004 peritoneal adipose tissue depots in nonobese Asian Indian males
109. Bankir L, Roussel R, Bouby N. Protein‐ and diabetes‐induced with type 2 diabetes in North India. J Diabetes Res. 2017;2017:
glomerular hyperfiltration: role of glucagon, vasopressin, and 2376016. https://doi.org/10.1155/2017/2376016
urea. Am J Physiol Ren Physiol. 2015;309(1):F2‐F23. https://doi.org/ 126. Moh Moh MA, Jung CH, Lee B, et al. Association of glucagon‐to‐
10.1152/ajprenal.00614.2014 insulin ratio and nonalcoholic fatty liver disease in patients with
110. Li XC, Liao TD, Zhuo JL. Long‐term hyperglucagonaemia induces type 2 diabetes mellitus. Diab Vasc Dis Res. 2019;16(2):186‐195.
early metabolic and renal phenotypes of type 2 diabetes in mice. https://doi.org/10.1177/1479164118810691
Clin Sci. 2008;114(9):591‐601. https://doi.org/10.1042/CS200 127. Pedersen JS, Rygg MO, Kristiansen VB, et al. Nonalcoholic fatty
70257 liver disease impairs the liver‐alpha cell axis independent of
15207560, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/dmrr.3609 by Cochrane Germany, Wiley Online Library on [30/01/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
16 of 16
- CARUSO ET AL.

hepatic inflammation and fibrosis. Hepatol Commun. 2020;4(11): 141. Ambery P, Parker VE, Stumvoll M, et al. MEDI0382, a GLP‐1 and
1610‐1623. https://doi.org/10.1002/hep4.1562 glucagon receptor dual agonist, in obese or overweight patients with
128. Gar C, Haschka SJ, Kern‐Matschilles S, et al. The liver‐alpha cell type 2 diabetes: a randomised, controlled, double‐blind, ascending
axis associates with liver fat and insulin resistance: a validation dose and phase 2a study. Lancet. 2018;391(10140):2607‐2618.
study in women with non‐steatotic liver fat levels. Dia- https://doi.org/10.1016/s0140‐6736(18)30726‐8
betologia. 2021;64(3):512‐520. https://doi.org/10.1007/s00125‐ 142. Visseren FLJ, Mach F, Smulders YM, et al. 2021 ESC Guidelines
020‐05334‐x on cardiovascular disease prevention in clinical practice. Eur
129. Tomah S, Alkhouri N, Hamdy O. Nonalcoholic fatty liver disease Heart J. 2021;42(34):3227‐3337. https://doi.org/10.1093/eurheartj/
and type 2 diabetes: where do Diabetologists stand? Clin Diabetes ehab484
Endocrinol. 2020;6(1):9. https://doi.org/10.1186/s40842‐020‐00 143. Nahra R, Wang T, Gadde KM, et al. Effects of cotadutide on
097‐1 metabolic and hepatic parameters in adults with overweight or
130. Nason SR, Kim T, Antipenko JP, et al. Glucagon‐receptor signaling obesity and type 2 diabetes: a 54‐week randomized phase 2b
reverses hepatic steatosis independent of leptin receptor expres- study. Diabetes Care. 2021;44(6):1433‐1442. https://doi.org/10.
sion. Endocrinology. 2020;161(1). https://doi.org/10.1210/endocr/ 2337/dc20‐2151
bqz013 144. Parker VER, Hoang T, Schlichthaar H, et al. Efficacy and safety of
131. Patil M, Deshmukh NJ, Patel M, Sangle GV. Glucagon‐based ther- cotadutide, a dual glucagon‐like peptide‐1 and glucagon receptor
apy: past, present and future. Peptides. 2020;127:170296. https:// agonist, in a randomized phase 2a study of patients with type 2
doi.org/10.1016/j.peptides.2020.170296 diabetes and chronic kidney disease. Diabetes Obes Metab. 2022;
132. Chai S, Zhang R, Zhang Y, et al. Effect of dipeptidyl peptidase‐4 in- 24(7):1360‐1369. https://doi.org/10.1111/dom.14712
hibitors on postprandial glucagon level in patients with type 2 dia- 145. Schiavon M, Visentin R, Göbel B, et al. Improved postprandial
betes mellitus: a systemic review and meta‐analysis. Front Endocrinol. glucose metabolism in type 2 diabetes by the dual glucagon‐like
2022;13:994944. https://doi.org/10.3389/fendo.2022.994944 peptide‐1/glucagon receptor agonist SAR425899 in comparison
133. Spiliotis II, Chalk R, Gough S, Rorsman P. Reducing hyper- with liraglutide. Diabetes Obes Metab. 2021;23(8):1795‐1805.
glucagonaemia in type 2 diabetes using low‐dose glibenclamide: https://doi.org/10.1111/dom.14394
results of the LEGEND‐A pilot study. Diabetes Obes Metab. 2022; 146. Kannt A, Madsen AN, Kammermeier C, et al. Incretin combination
24(8):1671‐1675. https://doi.org/10.1111/dom.14740 therapy for the treatment of non‐alcoholic steatohepatitis. Dia-
134. Pettus JH, D'Alessio D, Frias JP, et al. Efficacy and safety of the betes Obes Metab. 2020;22(8):1328‐1338. https://doi.org/10.1111/
glucagon receptor antagonist RVT‐1502 in type 2 diabetes uncon- dom.14035
trolled on metformin monotherapy: a 12‐week dose‐ranging study. 147. Jain M, Tsai L‐F, Robertson D, et al. MEDI0382, a GLP/glucagon
Diabetes Care. 2020;43(1):161‐168. https://doi.org/10.2337/dc19‐ receptor dual agonist, significantly reduces hepatic fat content in
1328 subjects with type 2 diabetes mellitus. Diabetes. 2018;67(1):78‐OR.
135. Hædersdal S, Lund A, Maagensen H, et al. The glucagon receptor https://doi.org/10.2337/db18‐78‐or
antagonist LY2409021 has no effect on postprandial glucose in 148. Choi JH, Lee JS, Kim JK, et al. Multi‐target engagement effect of a
type 2 diabetes. Eur J Endocrinol. 2022;186(2):207‐221. https://doi. novel long‐acting glucagon/GIP/GLP‐1 triple agonist (HM15211) in
org/10.1530/EJE‐21‐0865 animal model of NASH. Diabetes. 2021;70(Supplement_1):655‐P.
136. Hædersdal S, Lund A, Nielsen‐Hannerup E, et al. The glucagon 149. Boland ML, Laker RC, Mather K, et al. Resolution of NASH and
receptor antagonist LY2409021 does not affect gastrointestinal‐ hepatic fibrosis by the GLP‐1R/GcgR dual‐agonist cotadutide via
mediated glucose disposal or the incretin effect in individuals modulating mitochondrial function and lipogenesis. Nat Metab.
with and without type 2 diabetes. Eur J Endocrinol. 2022;187(4): 2020;2(5):413‐431. https://doi.org/10.1038/s42255‐020‐0209‐6
507‐518. https://doi.org/10.1530/EJE‐22‐0291 150. Finan B, Yang B, Ottaway N, et al. A rationally designed monomeric
137. Capozzi ME, DiMarchi RD, Tschöp MH, Finan B, Campbell JE. peptide triagonist corrects obesity and diabetes in rodents. Nat
Targeting the incretin/glucagon system with triagonists to treat Med. 2015;21(1):27‐36. https://doi.org/10.1038/nm.3761
diabetes. Endocr Rev. 2018;39(5):719‐738. https://doi.org/10.1210/ 151. Defronzo RA. Banting Lecture. From the triumvirate to the
er.2018‐00117 ominous octet: a new paradigm for the treatment of type 2 dia-
138. Irwin DM, Prentice KJ. Incretin hormones and the expanding betes mellitus. Diabetes. 2009;58(4):773‐795. https://doi.org/10.
families of glucagon‐like sequences and their receptors. Diabetes 2337/db09‐9028
Obes Metab. 2011;13(Suppl 1):69‐81. https://doi.org/10.1111/j. 152. Arafat MA, Otto B, Rochlitz H, et al. Glucagon inhibits ghrelin
1463‐1326.2011.01444.x secretion in humans. Eur J Endocrinol. 2005;153(3):397‐402.
139. Ambery PD, Klammt S, Posch MG, et al. MEDI0382, a GLP‐1/ https://doi.org/10.1530/eje.1.01981
glucagon receptor dual agonist, meets safety and tolerability end-
points in a single‐dose, healthy‐subject, randomized, phase 1 study.
Br J Clin Pharmacol. 2018;84(10):2325‐2335. https://doi.org/10.
1111/bcp.13688 How to cite this article: Caruso I, Marrano N, Biondi G, et al.
140. Parker VER, Robertson D, Wang T, et al. Efficacy, safety, and
Glucagon in type 2 diabetes: friend or foe? Diabetes Metab Res
mechanistic insights of cotadutide, a dual receptor glucagon‐like
peptide‐1 and glucagon agonist. J Clin Endocrinol Metab. 2020; Rev. 2023;e3609. https://doi.org/10.1002/dmrr.3609
105(3):803‐820. https://doi.org/10.1210/clinem/dgz047

You might also like