While the consumption of external energy (i.e., feeding) is essential to life, this action induces a temporary disturbance of homeostasis in an animal. A primary example of this effect is found in the regulation of glycemia. In the fasted state, stored energy is released to maintain physiological glycemic levels. Liver glycogen is liberated to glucose, glycerol and (glucogenic) amino acids are used to build new glucose molecules (i.e., gluconeogenesis), and fatty acids are oxidized to fuel long-term energetic demands. This regulation is driven primarily by the counterregulatory hormones epinephrine, growth hormone, cortisol, and glucagon. Conversely, feeding induces a rapid influx of diverse nutrients, including glucose, that disrupt homeostasis. Consistently, a host of hormonal and neural systems under the coordination of insulin are engaged in the transition from fasting to prandial states to reduce this disruption. The ultimate action of these systems is to appropriately store the newly acquired energy and to return to the homeostatic norm. Thus, at first glance it is tempting to assume that glucagon is solely antagonistic regarding the anabolic effects of insulin. We have been intrigued by the role of glucagon in the prandial transition and have attempted to delineate its role as beneficial or inhibitory to glycemic control. The following review highlights this long-known yet poorly understood hormone.

In 1921 Banting and Best (1) identified insulin, a life-saving therapeutic for millions of individuals with diabetes, which set a new course for our understanding of glucose metabolism. Two years later Kimball and Murlin (2) described the second hormone, glucagon, which appeared to oppose insulin and elevate blood glucose (3). Subsequent work by Burger, Brandt, and Kramer (46) identified the liver as the primary target of glucagon-stimulated hyperglycemia. Finally, in 1948 Sutherland and de Duve (7) published the first evidence that glucagon was produced from the pancreatic α-cells, closing the loop between its initial discovery as a pancreatic hormone and its primary target tissue, the liver.

Since this early codiscovery, the contrasting roles of insulin and glucagon have been studied in detail, often with an emphasis on the pathophysiological role of unopposed glucagon action in diabetes (812). However, emerging preclinical studies have highlighted potential insulin-sensitizing effects of glucagon receptor (GCGR) agonism, both alone and in combination with other incretin signals (i.e., glucagon-like peptide 1 [GLP-1] and glucose-dependent insulinotropic polypeptide [GIP]) (1320). Consistently, clinical studies of a single-molecule GCGR/GLP-1R coagonist uncovered reduced glucose excursion during a mixed-meal challenge (21). Although individual receptor contributions to this effect were not specifically investigated, similar findings have also been reported for single-molecule GCGR/GLP-1R/GIPR triagonists (22). Hence, a new emphasis has emerged on understanding the mechanisms and applications of GCGR agonism, especially in metabolic diseases.

Five main cell types (i.e., α-, β-, δ-, γ-, and ε-cells) make up the endocrine pancreas and are clustered into island-like structures called islets of Langerhans (23). Like insulin, glucagon is produced by the endocrine pancreas and secreted in response to changing nutritional demands (23). Glucagon is encoded by the proglucagon gene, which also encodes GLP-1, GLP-2, oxyntomodulin, glicentin, and the metabolically inert cleavage products glucagon-reactive polypeptide and major proglucagon fragment (24). Pancreatic α-cells preferentially express prohormone convertase-2, which is essential in processing the proglucagon peptide to produce the 29-amino-acid (AA) native glucagon peptide (2527). Glucagon is secreted from the α-cells, which make up 15–20% of total rodent islet cells (23) but 30–45% of the human islet (28). Thus, in human islets there is far greater interaction (i.e., more contact) between α- and β-cells than in rodent islets. These compositional differences in islet morphology suggest that glucagon plays a greater physiological role in humans than in rodents.

Glucagon secretion is influenced by nutritional state and is best known in the context of fasting and hypoglycemia (29,30). α-Cells preferentially express the low-Km glucose transporter 1 (GLUT1) (31) and ATP-sensitive potassium (KATP) channels (32). Glucose-dependent increases in cellular ATP levels close KATP channels, depolarizing the cell and inhibiting glucagon secretion (33,34). Intriguingly, the regulation of glucagon secretion is not restricted to glucose alone.

Free fatty acids (FFA) may stimulate glucagon secretion. However, this regulation appears to be dependent on the FFA characteristics and if the FFA source was exogenous or endogenous (30). AAs, excluding the branched-chain AAs, stimulate glucagon secretion in dogs (35). This was consistent with the observation that high-protein meals (3638), arginine (39,40), and alanine (41,42) stimulate glucagon secretion in humans. Importantly, the stimulatory effects of these AAs on glucagon secretion are far greater than those observed during hypoglycemia (20) yet are attenuated (43,44) or abolished (44) in the presence of hyperglycemia. Reciprocally, glucagon increases ureagenesis in hepatocytes to regulate AA metabolism (45). Insulin-resistant and steatotic individuals exhibit hyperaminoacidemia, leading to hyperglucagonemia and disruption of the liver–α-cell axis in humans (45). Likewise, inhibition of hepatic GCGR signaling results in increased circulating AAs and α-cell hyperplasia of both endogenous mouse islets and human islet transplants (46). Importantly, α-cell hyperplasia can be mimicked by culturing islets in high concentrations of AAs, especially l-glutamine (46). By extension, lipid-induced disruption of hepatic glucagon sensitivity has been postulated to contribute to impaired AA homeostasis, hyperglucagonemia, and eventually to type 2 diabetes (T2D) (47).

Glucagon secretion is also regulated via endocrine/paracrine factors, including insulin, amylin, zinc, GABA, GLP-1, GIP, and somatostatin. α-Cells express both insulin receptors (INSR) and GABA receptors (48,49). Consistently, insulin and GABA from neighboring β-cells both inhibit glucagon secretion (5052). However, work in rat islets supports that the key inhibitory factor from β-cells may be zinc bound to the insulin protein (53). Similarly, somatostatin of the δ-cells inhibits glucagon secretion (54). Only a minority (∼20%) of mouse, rat, and human α-cells express GLP-1R (55,56). Thus, inhibition via GLP-1 (5759) is likely secondary to GLP-1R–stimulated release of zinc-insulin, GABA, and amylin. Conversely, in healthy individuals GIP stimulates glucagon secretion in a glucose-dependent manner (i.e., during hypoglycemia) (60,61). Reciprocally, glucagon acts in a paracrine manner to increase insulin secretion through activation of both β-cell GCGR and GLP-1R (19).

Finally, glucagon secretion is directly mediated by the autonomic nervous system. Via their effects on insulin secretion, vagal stimulation (parasympathetic) inhibits (62), whereas splanchnic (sympathetic) stimulation increases, glucagon secretion (6366). Together, these findings clearly support the idea that glucagon secretion is regulated in response to multiple stimuli and systems. Among them is a potential cosecretion with insulin in the early prandial state. Together these observations support a more complex role for glucagon beyond simple counterregulation of insulin in glucose homeostasis.

GCGR is a member of the class B family of G protein–coupled receptors (67). Gcgr mRNA is primarily expressed in the liver, with low-level expression in the kidney, adipose tissue, pancreas, spleen, lymphoblasts, brain, gastrointestinal tract, and adrenal gland (68). Hepatic Gcgr expression and subsequent metabolic actions are restricted to the periportal area (69), where they overlap with INSR (Insr) expression (70). Hepatic GCGR signaling stimulates two intracellular cascades (Fig. 1), a cAMP stimulatory G protein, Gs, and a Gq protein that signals via Ca2+ (29,30). Canonical Gs signaling activates adenylate cyclase to produce cAMP. This second messenger stimulates both protein kinase A (PKA) and Rap guanine nucleotide exchange factor 3 (RAPGEF3; also known as EPAC1). EPAC1 activation stimulates the small GTPase Rap1 and the AMP-dependent protein kinase (AMPK) (71). Concomitantly, PKA phosphorylates the cAMP response element-binding protein (CREB) and stimulates protein phosphatase 2B-dependent dephosphorylation of the CREB-regulated transcription coactivator 2 (Crtc2) (72). CREB/CRTC2 signaling is associated with gluconeogenic and glycogenolytic gene expression (e.g., glucose-6-phosphatase [G6pc], phosphoenolpyruvate kinase [Pck1], and peroxisome proliferator-activated receptor γ coactivator 1-α [Ppargc1a]) (30). GCGR-stimulated Ca2+ signaling occurs downstream of Gq activation and is associated with hepatic glycogen phosphorylase activation, bile acid homeostasis, and liver regeneration (73).

Figure 1

Overview of GCGR signaling pathways in the regulation of hepatic glucose homeostasis. Figure created with BioRender.com.

Figure 1

Overview of GCGR signaling pathways in the regulation of hepatic glucose homeostasis. Figure created with BioRender.com.

Close modal

Termination of signaling is equally important to metabolic regulation. GCGR signaling is terminated by internalization of the ligand–receptor complex and occurs primarily via clathrin- and arrestin-facilitated endocytosis. Intriguingly, sustained GCGR signaling has been described after internalization, suggesting a second wave of signaling from this receptor (30). However, the biological relevance of this intracellular signaling has yet to be fully elucidated. Intracellular GCGR palmitoylation and ubiquitination have been observed and may also contribute to signal termination (30). Intriguingly, glucagon stimulates both GCGR internalization and deubiquitination, facilitating rapid recycling of the receptor (74).

As introduced above, the best-known actions of GCGR signaling involve its counterregulatory effect on insulin action. In the context of glucose metabolism, GCGR signaling stimulates hepatic glycogenolysis and gluconeogenesis (GNG) with concomitant inhibition of glycogen synthesis (29). GCGR signaling rapidly increases hepatic glycogenolysis via a signaling cascade involving the canonical cAMP–PKA pathway. This signaling activates glycogen phosphorylase kinase and subsequent activation of glycogen phosphorylase. GCGR signaling (via PKA) likewise inhibits glycogen synthase, preventing hepatic glycogen synthesis (75).

GCGR regulation of hepatic GNG occurs via both transcriptional induction and allosteric modulation of GNG enzymes. PKA-dependent phosphorylation of phosphofructokinase 2 and pyruvate kinase shifts metabolic flux from glycolysis to GNG. GCGR signaling stimulates CREBSer133 phosphorylation coupled with dephosphorylation and nuclear translocation of its coactivator, Creb-regulated transcription coactivator 2 (Crtc2). These actions not only stimulate the induction of target GNG genes G6pc, Pck1, Ppargc1a and hepatocyte nuclear factor 4 (Hnf4a) but also regulate GNG-associated transcription factors FOXO1 and PGC-1-α via modulation of their acetylation states (30). Additionally, GCGR-stimulated Ca2+ signaling activates glycogenolysis and GNG via p38 kinase (76). Consistent with these signaling events, exogenous glucagon elevates glycemia (77). Moreover, genetic Gcgr deficiency and neutralizing antibodies targeting glucagon are sufficient to reduce glycemia (7880). In contrast, the antidiabetic effects of Gcgr knockout in streptozotocin (STZ)-treated mice are lost when STZ is administered prior to Gcgr ablation (81). These rodent data must be interpreted with some caution, as GCGR antagonists clearly lower glycemia in individuals with T1D (82). Together, these findings highlight the complex and context-dependent relationship between glucagon and insulin in glucose homeostasis.

In addition to its effects on glucose metabolism, mounting evidence suggests hepatic glucagon is a potent regulator of energy balance, lipid homeostasis, and fat mass mobilization (30). In the context of energy balance, glucagon both stimulates energy expenditure and suppresses food intake, as highlighted by the negative energy balance observed in glucagonoma patients (83). This stimulation of energy expenditure and thermogenesis is conserved across a range of species (29). However, the conservation of this system in humans is still controversial, with reports observing both increased and unchanged energy expenditure (84,85). Energy expenditure regulation in mice is dependent upon hepatic GCGR signaling and is mechanistically associated with hepatic FXR activity and endocrine FGF21 action (14,15,86). Glucose futile cycling may also contribute to the upregulation of energy expenditure following GCGR agonism (87,88). Intriguingly, glucagon administration also decreases hunger and food intake in both rats (89) and human subjects (90,91). Consistently, GCGR agonism in diet-induced obese mice suppressed food intake; however, this effect was preserved in mice lacking hepatic Gcgr expression, suggesting that the liver is not the tissue of origin for this regulation (14).

Glucagon also regulates multiple components of lipid metabolism (29). Gcgr is expressed by rodent adipocytes (92). Consistently, glucagon mediates rodent white adipose tissue lipolysis (93). Conversely, evidence of Gcgr expression in human adipocytes is lacking (94), as is that for glucagon-induced lipolysis at physiological levels in patients (95). In rodents, glucagon-mediated white adipose tissue lipolysis (96,97) via hormone-sensitive lipase results in the liberation of nonesterified fatty acids (NEFA) (98). The majority of these NEFAs are catabolized. However, in the liver, NEFAs may be alternatively converted to ketone bodies to provide energy during times of glucose deficiency (99,100). Consistent with this shift to lipid energy substrates, glucagon exposure inhibits hepatic lipogenesis while stimulating FA transport and oxidation (101). Inhibition of hepatic lipogenesis occurs via two potential mechanisms: 1) CREB-mediated induction of insulin-induced gene 2 (Insig2) and sequestration of the lipogenic sterol regulatory element binding protein (SREBP) transcription factor (102) and 2) Ca2+-dependent activation of p38 kinase and subsequent inhibition of SREBP (76). GCGR agonism is also a potent regulator of bile acid metabolism, stimulating robust changes in the expression of bile acid enzymes and the composition of circulating bile acids (14). As introduced above, emerging data support that hepatic GCGR signaling is a crucial regulator of AA metabolism. GCGR agonism stimulates hepatic AA uptake and urea production and subsequently induces hypoaminoacidemia (103). Together these pieces of evidence point to glucagon as a potent regulator of AA and lipid homeostasis, energy balance, and fat mass mobilization.

Insulin is a powerful anabolic factor, stimulating growth and energy accrual throughout the organism. This pleiotropic hormone is essential to glucose metabolism and crucial to lipid and AA metabolism. Insulin action in the liver stimulates lipogenesis and glycogen synthesis while concomitantly inhibiting glycogenolysis, GNG, and liver fatty acid oxidation (104).

Insulin signals via the INSR, a member of the receptor tyrosine kinase family, and, to a lesser extent, the insulin-like growth factor 1 receptor. These receptors are endogenously inhibited by the recently discovered Inceptor protein in mouse β-cells (105). Insr is expressed in the central nervous system and a wide range of peripheral tissues. Unlike Gcgr, hepatic Insr expression is found in both periportal and perivenous zones (70). The role of this essential hormone and INSR signaling (summarized in Fig. 2) has been extensively covered, including the following review (104). Therefore, this Perspective will focus on hepatic signaling and biological functions arising from INSR activation. INSR signaling is initiated when insulin binds to the receptor, derepressing the receptor’s intrinsic kinase activity. INSR then phosphorylates intracellular substrates, including members of the insulin/insulin-like growth factor 1 receptor substrate (IRS) protein family, Gab-1, DOK1, Cbl, SH2B2 (APS), SHP2, and isoforms of Shc (104). Canonical insulin regulation of hepatic glucose and lipid metabolism involves subsequent IRS-dependent activation of phosphatidylinositol-3-kinase, 3′-phosphoinositide–dependent kinase 1 (PDK1), and AKT/PKB (104). AKT is a central node of hepatic insulin signaling and is crucial for both glucose and lipid metabolism. This serine/threonine kinase is activated by phosphorylation on two residues, Thr308 and Ser473. Thr308 phosphorylation occurs in a PDK1-dependent manner and is essential for AKT kinase activity. Ser473 is phosphorylated by the rapamycin-insensitive mTOR complex (mTORC2) and is permissive for full kinase activity (104). Importantly, the mechanisms of mTORC2 regulation remain uncertain. AKT activation leads to subsequent phosphorylation of forkhead box–containing protein, O subfamily (FOXO). FOXO proteins (especially members 1 and 6) are transcription factors that induce GNG. AKT-dependent phosphorylation triggers nuclear exclusion and, thus, is inhibitory to this action (106).

Figure 2

Overview of INSR signaling pathways in the regulation of hepatic glucose homeostasis. MAPK, mitogen-activated protein kinase; PI3K, phosphatidylinositol 3-kinase; PLCγ, phospholipase Cγ. Figure created with BioRender.com.

Figure 2

Overview of INSR signaling pathways in the regulation of hepatic glucose homeostasis. MAPK, mitogen-activated protein kinase; PI3K, phosphatidylinositol 3-kinase; PLCγ, phospholipase Cγ. Figure created with BioRender.com.

Close modal

Insulin also regulates hepatic Ca2+ signaling. INSR activation stimulates phospholipase Cγ, generating inositol-1,4,5-triphosphate (InsP3). Increased InsP3 levels stimulate InsP3 ligand–gated Ca2+ channels of the endoplasmic reticulum and thus increase intracellular Ca2+ levels. Increased hepatic Ca2+ levels further stimulate INSR-dependent activation of the mitogen-activated protein kinase signaling cascade and activation of transcription factors (e.g., MYC, FOS, and JUN) in this mitogenic pathway (107).

Diabetes, whether type 1 (T1D) or type 2 (T2D), is defined by hyperglycemia and is ultimately the result of insufficient insulin action. In the case of T1D, this deficiency is caused by destruction of the pancreatic β-cell and therefore a lack of the insulin hormone. In T2D, insulin resistance accumulates to a point where β-cell compensatory hypersecretion is insufficient to counteract the resistance (108). In the liver, this insufficiency is manifested as a failure to suppress hepatic glucose output (i.e., GNG and glycogenolysis). Intriguingly, in T2D this resistance is often incomplete, resulting in a preservation of insulin-stimulated lipogenesis (108). Consistent with its counterregulatory role, both fasting and postprandial plasma glucagon levels are elevated in diabetes (109). However, these observations have been made in individuals with established cases of diabetes, and thus the causality of hyperglucagonemia is difficult to assign.

As a counterregulatory hormone with a role in maintaining fasting blood glucose, it is tempting to assume that glucagon opposes all actions of insulin. Consistent with this hypothesis, circulating glucagon levels are elevated in all known instances of T1D or T2D, including animal models of the disease (77). Likewise, preclinical GCGR ablation or pharmacological GCGR inhibition (including neutralizing antibodies against glucagon) in individuals with diabetes is sufficient to reduce glycemia and HbA1c. However, many of these strategies have been slowed due to adverse effects on liver transaminases, liver fat, and dyslipidemia (30).

Conversely, the increased concentrations and action of glucagon in the fasting state are well suited to potentiate subsequent insulin-mediated glucose control. To this point, glucagon acts in a paracrine manner to increase insulin secretion through activation of both β-cell GCGR and GLP-1R (19). Likewise, postprandial elevations of glucagon and GLP-1 contribute to the improved postprandial glucose profile observed in Roux-en-Y gastric bypass patients (110) and rodent models of this powerful intervention (111). Importantly, these physiological conditions are all characterized by their heightened insulin sensitivity. Regarding glucagon enhancement of insulin action, the use of the bionic pancreas (glucagon and insulin) must be mentioned (112). This technology was hypothesized to prevent life-threatening hypoglycemic episodes in people with diabetes. Beyond reducing hypoglycemic episodes, the bihormonal (glucagon and insulin) pump reduced average glycemia while requiring a similar total daily insulin dose in adolescents (112). Likewise, 13-h glucagon infusion increased both glucose appearance and disappearance in patients, suggesting that its regulation of human glucose metabolism is not restricted to increasing hepatic glucose output (113). Together, these observations support the hypothesis that glucagon, released during fasting and the prandial response, acts to prime metabolic tissues for the subsequent nutrient challenge of feeding. Moreover, it positions cooperative actions of glucagon and insulin as crucial to this physiology.

INSR and GCGR signaling also converge at the hepatocyte. Our group described the unexpected enhancement of insulin action in db/db mice following chronic (7-day) treatment with the long-acting GCGR agonist IUB288 (86). This initial observation was followed by more detailed investigation of acute (i.e., 60-min) GCGR agonism and its beneficial effect on insulin sensitivity (114). This work identified enhanced insulin-dependent signaling in the phosphorylation of AKTSer473 in mice treated with IUB288 60 min prior to insulin and was exclusive of PDK1-dependent phosphorylation (Thr308) (114). This single, acute IUB288 treatment increased insulin sensitivity, as defined by increased glucose infusion rate and improved insulin-stimulated suppression of hepatic glucose output during hyperinsulinemic-euglycemic clamps (114). These observations suggest GCGR and INSR signaling intersect via a TORC2-dependent phosphorylation of AKTSer473. Our observation was quickly followed by work by Besse-Patin et al. (115). This elegant study confirmed glucagon-enhanced AKTSer473 phosphorylation and identified glucagon-dependent induction of Ppargc1a as a transcriptional regulator of relative levels of hepatocyte IRS1:IRS2 ratios (115). This shift toward IRS2 favors insulin-dependent suppression of hepatic glucose output (115) and is consistent with our observations in hyperinsulinemic-euglycemic clamps (114). Congruous with our study and interpretation, Besse-Patin et al. concluded that glucagon (via PGC-1-α) primes the liver for subsequent insulin action.

However, an importation caveat to these studies is that the observations of Besse-Patin et al. were made 4 h after glucagon treatment. Subsequent observations in cultured hepatocytes suggest GCGR signaling transiently stimulates protein synthesis via an mTORC1-dependent action (116). This effect was also observed to be convergent with insulin signaling and dependent on EPAC activity (116). Additionally, work by Perry et al. (117) identified enhanced glucose tolerance and insulin sensitivity in rats infused with glucagon for 3.5 weeks. This work supported a role for inositol triphosphate receptor 1 (INSP3R1)-mediated calcium signaling downstream of GCGR activation. In this model, the benefits of GCGR signaling on glucose metabolism are related to hepatic mitochondrial oxidation (117). In summary, emerging data support a beneficial role for GCGR signaling in hepatic insulin glucose metabolism. While the precise mechanisms have yet to be elucidated, data support roles for mTORC1, mTORC2, and PCG1a-IRS2 as potential points for cross talk with hepatic insulin signaling (Fig. 3). INSP3R1 may also represent a mechanism by which hepatic GCGR signaling benefits glucose metabolism secondary to its regulation of mitochondrial oxidation.

Figure 3

Potential and reported cross talk in hepatic glucagon (GCG) and INSR signaling. PI3K, phosphatidylinositol 3-kinase. Figure created with BioRender.com.

Figure 3

Potential and reported cross talk in hepatic glucagon (GCG) and INSR signaling. PI3K, phosphatidylinositol 3-kinase. Figure created with BioRender.com.

Close modal

As introduced above, GCGR ablation/antagonism is beneficial for glucose metabolism (78,79). Of note, treating mice with the INSR antagonist S961 induces severe insulin resistance, hyperglycemia, and ketonemia, yet the GCGR-blocking antibody REGN1193 was sufficient to normalize blood glucose and β-hydroxybutyrate levels in these mice (118). Subsequent clinical investigation uncovered reductions in fasting plasma glucose and HbA1c in REGN1193-treated T2D patients (119). Similar benefits in mice have been reported for the monoclonal antibody and competitive GCGR antagonist REMD 2.59 (120). Moreover, GCGR antagonism, when combined with GLP-1R agonism, stimulates cell regeneration in STZ-treated mice (121). However, enthusiasm for GCGR antagonism is offset by observations of dose-dependent increases in hepatic aminotransferases (122) and induction of profound dyslipidemia (79). Conversely, the benefits of GCGR agonism on energy expenditure, hepatic steatosis, and lipid homeostasis are of great therapeutic interest. Intriguingly, coupling of the antidiabetic properties of GLP-1R agonism with GCGR agonism profoundly enhances the therapeutic action of both receptors (17,18,123). The mechanisms underlying these benefits are still the focus of intense investigation. GLP-1/GCGR dual agonism drives weight loss in a synergistic manner. This weight loss is likely due to GCGR stimulation of energy expenditure and GLP-1R inhibition of gastric emptying (124), the latter also contributing to slower glucose uptake into the circulation. It is also likely that these compounds increase glucose-stimulated insulin secretion via activation of GCGR and GLP-1R at the β-cell while concomitantly enhancing insulin action via GCGR agonism at the liver. Based on this hypothesis, coupling GCGR agonism with other known insulin secretagogues should have similar effects. This hypothesis is supported by the observation in mice that tolbutamide enhanced glucagon-stimulated decreases in glycemia (19). It should be noted that while GLP-1/GCGR dual agonism drives weight loss and improves glucose homeostasis in both preclinical and clinical studies (21,125), clinical application of these molecules has targeted treatment of nonalcoholic steatohepatitis and nonalcoholic fatty liver disease (e.g., cotadutide) (125).

In summary, the glucagon peptide was discovered a century ago, yet our understanding of its metabolic actions is still evolving. The original view that GCGR signaling is antagonistic to insulin action is certainly true in some contexts yet is clearly incomplete. Studies currently underway will continue to refine the role of this long-known hormone and its therapeutic utility in metabolic diseases.

See accompanying articles, pp. 1834 and 1852.

Acknowledgments. I thank Dr. Teayoun Kim, Dr. Shelly Nason, and Jessica Antipenko (Comprehensive Diabetes Center and Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL) for helpful discussion.

Funding. The project described in this work was supported by National Institutes of Health grant 1R01DK112934 (K.M.H.).

Duality of Interest. No potential conflicts of interest relevant to this article were reported.

Prior Presentation. Parts of this work were presented at the 82nd Scientific Sessions of the American Diabetes Association, New Orleans, LA, 3–7 June 2022.

1.
Banting
FG
,
Best
CH
.
The internal secretions of the pancreas
.
J Lab Clin Med
1922
;
5
:
251
266
2.
Kimball
CP
,
Murlin
JR
.
Aqueous extracts of pancreas. III. Some precipitation reactions of insulin
.
J Biol Chem
1923
;
58
:
337
346
3.
Murlin
JR
,
Clough
HD
,
Gibbs
CBF
,
Stokes
AM
.
Aqueous extracts of the pancreas. I. Influence on the carbohydrate metabolism of depancreatized animals
.
J Biol Chem
1923
;
56
:
253
296
4.
Bürger
M
,
Brandt
W
.
Über das glukagon (die hyperglykämisierende substanz des pankreas)
.
Z Gesamte Exp Med
1935
;
96
:
375
5.
Bürger
M
,
Kramer
H
.
Über den hepatischen angriffspunkt des insulins
.
Z Gesamte Exp Med
1929
;
65
:
487
497
6.
Bürger
M
,
Kramer
H
.
Primäre hyperglykämie und glykogenverarmung der leber als folge intraportaler insulininjektion nach untersuchungen am hund
.
Z Gesamte Exp Med
1929
;
67
:
441
450
7.
Sutherland
EW
,
de Duve
C
.
Origin and distribution of the hyperglycemic-glycogenolytic factor of the pancreas
.
J Biol Chem
1948
;
175
:
663
674
8.
Gu
W
,
Yan
H
,
Winters
KA
, et al
.
Long-term inhibition of the glucagon receptor with a monoclonal antibody in mice causes sustained improvement in glycemic control, with reversible alpha-cell hyperplasia and hyperglucagonemia
.
J Pharmacol Exp Ther
2009
;
331
:
871
881
9.
Wang
MY
,
Chen
L
,
Clark
GO
, et al
.
Leptin therapy in insulin-deficient type I diabetes
.
Proc Natl Acad Sci U S A
2010
;
107
:
4813
4819
10.
Brown
RJ
,
Sinaii
N
,
Rother
KI
.
Too much glucagon, too little insulin: time course of pancreatic islet dysfunction in new-onset type 1 diabetes
.
Diabetes Care
2008
;
31
:
1403
1404
11.
Dunning
BE
,
Gerich
JE
.
The role of alpha-cell dysregulation in fasting and postprandial hyperglycemia in type 2 diabetes and therapeutic implications
.
Endocr Rev
2007
;
28
:
253
283
12.
Gromada
J
,
Franklin
I
,
Wollheim
CB
.
Alpha-cells of the endocrine pancreas: 35 years of research but the enigma remains
.
Endocr Rev
2007
;
28
:
84
116
13.
Kim
T
,
Holleman
CL
,
Nason
S
, et al
.
Hepatic glucagon receptor signaling enhances insulin-stimulated glucose disposal in rodents
.
Diabetes
2018
;
67
:
2157
2166
14.
Kim
T
,
Nason
S
,
Holleman
C
, et al
.
Glucagon receptor signaling regulates energy metabolism via hepatic farnesoid X receptor and fibroblast growth factor 21
.
Diabetes
2018
;
67
:
1773
1782
15.
Nason
SR
,
Antipenko
J
,
Presedo
N
, et al
.
Glucagon receptor signaling regulates weight loss via central KLB receptor complexes
.
JCI Insight
2021
;
6
:
e141323
16.
Day
JW
,
Gelfanov
V
,
Smiley
D
, et al
.
Optimization of co-agonism at GLP-1 and glucagon receptors to safely maximize weight reduction in DIO-rodents
.
Biopolymers
2012
;
98
:
443
450
17.
Day
JW
,
Ottaway
N
,
Patterson
JT
, et al
.
A new glucagon and GLP-1 co-agonist eliminates obesity in rodents
.
Nat Chem Biol
2009
;
5
:
749
757
18.
Finan
B
,
Yang
B
,
Ottaway
N
, et al
.
A rationally designed monomeric peptide triagonist corrects obesity and diabetes in rodents
.
Nat Med
2015
;
21
:
27
36
19.
Capozzi
MEWJ
,
Wait
JB
,
Koech
J
, et al
.
Glucagon lowers glycemia when β-cells are active
.
JCI Insight
2019
;
5
:
e129954
20.
Finan
B
,
Capozzi
ME
,
Campbell
JE
.
Repositioning glucagon action in the physiology and pharmacology of diabetes
.
Diabetes
2020
;
69
:
532
541
21.
Ambery
P
,
Parker
VE
,
Stumvoll
M
, et al
.
MEDI0382, a GLP-1 and glucagon receptor dual agonist, in obese or overweight patients with type 2 diabetes: a randomised, controlled, double-blind, ascending dose and phase 2a study
.
Lancet
2018
;
391
:
2607
2618
22.
Bossart
M
,
Wagner
M
,
Elvert
R
, et al
.
Effects on weight loss and glycemic control with SAR441255, a potent unimolecular peptide GLP-1/GIP/GCG receptor triagonist
.
Cell Metab
2022
;
34
:
59
74.e10
23.
Röder
PVWB
,
Wu
B
,
Liu
Y
,
Han
W
.
Pancreatic regulation of glucose homeostasis
.
Exp Mol Med
2016
;
48
:
e219
24.
Wewer Albrechtsen
NJ
,
Kuhre
RE
,
Pedersen
J
,
Knop
FK
,
Holst
JJ
.
The biology of glucagon and the consequences of hyperglucagonemia
.
Biomarkers Med
2016
;
10
:
1141
1151
25.
Drucker
DJ
.
Glucagon and the glucagon-like peptides
.
Pancreas
1990
;
5
:
484
488
26.
White
JW
,
Saunders
GF
.
Structure of the human glucagon gene
.
Nucleic Acids Res
1986
;
14
:
4719
4730
27.
Ramzy
A
,
Kieffer
TJ
.
Altered islet prohormone processing: a cause or consequence of diabetes?
Physiol Rev
2022
;
102
:
155
208
28.
Cabrera
O
,
Berman
DM
,
Kenyon
NS
,
Ricordi
C
,
Berggren
PO
,
Caicedo
A
.
The unique cytoarchitecture of human pancreatic islets has implications for islet cell function
.
Proc Natl Acad Sci U S A
2006
;
103
:
2334
2339
29.
Habegger
KM
,
Heppner
KM
,
Geary
N
,
Bartness
TJ
,
DiMarchi
R
,
Tschöp
MH
.
The metabolic actions of glucagon revisited
.
Nat Rev Endocrinol
2010
;
6
:
689
697
30.
Zeigerer
A
,
Sekar
R
,
Kleinert
M
,
Nason
S
,
Habegger
KM
,
Müller
TD
.
Glucagon’s metabolic action in health and disease
.
Compr Physiol
2021
;
11
:
1759
1783
31.
Heimberg
H
,
De Vos
A
,
Pipeleers
D
,
Thorens
B
,
Schuit
F
.
Differences in glucose transporter gene expression between rat pancreatic alpha- and beta-cells are correlated to differences in glucose transport but not in glucose utilization
.
J Biol Chem
1995
;
270
:
8971
8975
32.
Bokvist
K
,
Olsen
HL
,
Høy
M
, et al
.
Characterisation of sulphonylurea and ATP-regulated K+ channels in rat pancreatic A-cells
.
Pflugers Arch
1999
;
438
:
428
436
33.
Rorsman
P
,
Braun
M
,
Zhang
Q
.
Regulation of calcium in pancreatic α- and β-cells in health and disease
.
Cell Calcium
2012
;
51
:
300
308
34.
Rorsman
P
,
Salehi
SA
,
Abdulkader
F
,
Braun
M
,
MacDonald
PE
.
K(ATP)-channels and glucose-regulated glucagon secretion
.
Trends Endocrinol Metab
2008
;
19
:
277
284
35.
Rocha
DMFG
,
Faloona
GR
,
Unger
RH
.
Glucagon-stimulating activity of 20 amino acids in dogs
.
J Clin Invest
1972
;
51
:
2346
2351
36.
Gannon
MCNF
,
Nuttall
FQ
.
Effect of a high-protein, low-carbohydrate diet on blood glucose control in people with type 2 diabetes
.
Diabetes
2004
;
53
:
2375
2382
37.
Linn
T
,
Santosa
B
,
Grönemeyer
D
, et al
.
Effect of long-term dietary protein intake on glucose metabolism in humans
.
Diabetologia
2000
;
43
:
1257
1265
38.
Markova
M
,
Hornemann
S
,
Sucher
S
, et al
.
Rate of appearance of amino acids after a meal regulates insulin and glucagon secretion in patients with type 2 diabetes: a randomized clinical trial
.
Am J Clin Nutr
2018
;
108
:
279
291
39.
Blackard
WGNN
,
Nelson
NC
,
Andrews
SS
.
Portal and peripheral vein immunoreactive glucagon concentrations after arginine or glucose infusions
.
Diabetes
1974
;
23
:
199
202
40.
Palmer
JPBJ
,
Benson
JW
,
Walter
RM
,
Ensinck
JW
.
Arginine-stimulated acute phase of insulin and glucagon secretion in diabetic subjects
.
J Clin Invest
1976
;
58
:
565
570
41.
Williams
PRSM
,
Sperling
MA
,
Racasa
Z
.
Blunting of spontaneous and alanine-stimulated glucagon secretion in newborn infants of diabetic mothers
.
Am J Obstet Gynecol
1979
;
133
:
51
56
42.
Porcellati
F
,
Pampanelli
S
,
Rossetti
P
, et al
.
Effect of the amino acid alanine on glucagon secretion in non-diabetic and type 1 diabetic subjects during hyperinsulinaemic euglycaemia, hypoglycaemia and post-hypoglycaemic hyperglycaemia
.
Diabetologia
2007
;
50
:
422
430
43.
Unger
RHA-PE
,
Aguilar-Parada
E
,
Müller
WA
,
Eisentraut
AM
.
Studies of pancreatic alpha cell function in normal and diabetic subjects
.
J Clin Invest
1970
;
49
:
837
848
44.
Raskin
P
,
Aydin
I
,
Yamamoto
T
,
Unger
RH
.
Abnormal alpha cell function in human diabetes: the response to oral protein
.
Am J Med
1978
;
64
:
988
997
45.
Wewer Albrechtsen
NJ
,
Færch
K
,
Jensen
TM
, et al
.
Evidence of a liver-alpha cell axis in humans: hepatic insulin resistance attenuates relationship between fasting plasma glucagon and glucagonotropic amino acids
.
Diabetologia
2018
;
61
:
671
680
46.
Dean
ED
,
Li
M
,
Prasad
N
, et al
.
Interrupted glucagon signaling reveals hepatic α cell axis and role for l-glutamine in α cell proliferation
.
Cell Metab
2017
;
25
:
1362
1373.e5
47.
Wewer Albrechtsen
NJ
,
Pedersen
J
,
Galsgaard
KD
, et al
.
The liver-α-cell axis and type 2 diabetes
.
Endocr Rev
2019
;
40
:
1353
1366
48.
Diao
J
,
Asghar
Z
,
Chan
CB
,
Wheeler
MB
.
Glucose-regulated glucagon secretion requires insulin receptor expression in pancreatic alpha-cells
.
J Biol Chem
2005
;
280
:
33487
33496
49.
Wendt
A
,
Birnir
B
,
Buschard
K
, et al
.
Glucose inhibition of glucagon secretion from rat alpha-cells is mediated by GABA released from neighboring beta-cells
.
Diabetes
2004
;
53
:
1038
1045
50.
Cooperberg
BACP
,
Cryer
PE
.
Insulin reciprocally regulates glucagon secretion in humans
.
Diabetes
2010
;
59
:
2936
2940
51.
Franklin
IKWC
,
Wollheim
CB
.
GABA in the endocrine pancreas: its putative role as an islet cell paracrine-signalling molecule
.
J Gen Physiol
2004
;
123
:
185
190
52.
Taneera
J
,
Jin
Z
,
Jin
Y
, et al
.
γ-Aminobutyric acid (GABA) signalling in human pancreatic islets is altered in type 2 diabetes
.
Diabetologia
2012
;
55
:
1985
1994
53.
Zhou
H
,
Zhang
T
,
Harmon
JS
,
Bryan
J
,
Robertson
RP
.
Zinc, not insulin, regulates the rat alpha-cell response to hypoglycemia in vivo
.
Diabetes
2007
;
56
:
1107
1112
54.
Hauge-Evans
AC
,
King
AJ
,
Carmignac
D
, et al
.
Somatostatin secreted by islet delta-cells fulfills multiple roles as a paracrine regulator of islet function
.
Diabetes
2009
;
58
:
403
411
55.
Heller
RS
,
Kieffer
TJ
,
Habener
JF
.
Insulinotropic glucagon-like peptide I receptor expression in glucagon-producing alpha-cells of the rat endocrine pancreas
.
Diabetes
1997
;
46
:
785
791
56.
Tornehave
D
,
Kristensen
P
,
Rømer
J
,
Knudsen
LB
,
Heller
RS
.
Expression of the GLP-1 receptor in mouse, rat, and human pancreas
.
J Histochem Cytochem
2008
;
56
:
841
851
57.
Junker
AEGL
,
Gluud
LL
,
van Hall
G
,
Holst
JJ
,
Knop
FK
,
Vilsbøll
T
.
Effects of glucagon-like peptide-1 on glucagon secretion in patients with non-alcoholic fatty liver disease
.
J Hepatol
2016
;
64
:
908
915
58.
Hare
KJVT
,
Vilsbøll
T
,
Asmar
M
,
Deacon
CF
,
Knop
FK
,
Holst
JJ
.
The glucagonostatic and insulinotropic effects of glucagon-like peptide 1 contribute equally to its glucose-lowering action
.
Diabetes
2010
;
59
:
1765
1770
59.
Creutzfeldt
WOKN
,
Kleine
N
,
Willms
B
,
Orskov
C
,
Holst
JJ
,
Nauck
MA
.
Glucagonostatic actions and reduction of fasting hyperglycemia by exogenous glucagon-like peptide I(7-36) amide in type I diabetic patients
.
Diabetes Care
1996
;
19
:
580
586
60.
El
K
,
Campbell
JE
.
The role of GIP in α-cells and glucagon secretion
.
Peptides
2020
;
125
:
170213
61.
Chia
CWCO
,
Carlson
OD
,
Kim
W
, et al
.
Exogenous glucose-dependent insulinotropic polypeptide worsens post prandial hyperglycemia in type 2 diabetes
.
Diabetes
2009
;
58
:
1342
1349
62.
Frohman
LAEE
,
Ezdinli
EZ
,
Javid
R
.
Effect of vagotomy and vagal stimulation on insulin secretion
.
Diabetes
1967
;
16
:
443
448
63.
Osundiji
MAEM
,
Evans
ML
.
Brain control of insulin and glucagon secretion
.
Endocrinol Metab Clin North Am
2013
;
42
:
1
14
64.
Bloom
SR
Sr
,
Edwards
AV
,
Hardy
RN
.
The role of the autonomic nervous system in the control of glucagon, insulin and pancreatic polypeptide release from the pancreas
.
J Physiol
1978
;
280
:
9
23
65.
Taborsky
GJ
Jr
.
The physiology of glucagon
.
J Diabetes Sci Technol
2010
;
4
:
1338
1344
66.
Kurose
T
,
Seino
Y
,
Nishi
S
, et al
.
Mechanism of sympathetic neural regulation of insulin, somatostatin, and glucagon secretion
.
Am J Physiol
1990
;
258
:
E220
E227
67.
de Graaf
C
,
Song
G
,
Cao
C
, et al
.
Extending the structural view of class B GPCRs
.
Trends Biochem Sci
2017
;
42
:
946
960
68.
Svoboda
M
,
Tastenoy
M
,
Vertongen
P
,
Robberecht
P
.
Relative quantitative analysis of glucagon receptor mRNA in rat tissues
.
Mol Cell Endocrinol
1994
;
105
:
131
137
69.
Krones
A
,
Kietzmann
T
,
Jungermann
K
.
Periportal localization of glucagon receptor mRNA in rat liver and regulation of its expression by glucose and oxygen in hepatocyte cultures
.
FEBS Lett
1998
;
421
:
136
140
70.
Krones
A
,
Kietzmann
T
,
Jungermann
K
.
Perivenous localization of insulin receptor protein in rat liver, and regulation of its expression by glucose and oxygen in hepatocyte cultures
.
Biochem J
2000
;
348
:
433
438
71.
Cyphert
HA
,
Alonge
KM
,
Ippagunta
SM
,
Hillgartner
FB
.
Glucagon stimulates hepatic FGF21 secretion through a PKA- and EPAC-dependent posttranscriptional mechanism
.
PLoS One
2014
;
9
:
e94996
72.
Oh
KJ
,
Han
HS
,
Kim
MJ
,
Koo
SH
.
Transcriptional regulators of hepatic gluconeogenesis
.
Arch Pharm Res
2013
;
36
:
189
200
73.
Amaya
MJ
,
Nathanson
MH
.
Calcium signaling in the liver
.
Compr Physiol
2013
;
3
:
515
539
74.
Kaur
S
,
Chen
Y
,
Shenoy
SK
.
Agonist-activated glucagon receptors are deubiquitinated at early endosomes by two distinct deubiquitinases to facilitate Rab4a-dependent recycling
.
J Biol Chem
2020
;
295
:
16630
16642
75.
Jiang
G
,
Zhang
BB
.
Glucagon and regulation of glucose metabolism
.
Am J Physiol Endocrinol Metab
2003
;
284
:
E671
E678
76.
Barella
LF
,
Jain
S
,
Kimura
T
,
Pydi
SP
.
Metabolic roles of G protein-coupled receptor signaling in obesity and type 2 diabetes
.
FEBS J
2021
;
288
:
2622
2644
77.
Unger
RH
,
Cherrington
AD
.
Glucagonocentric restructuring of diabetes: a pathophysiologic and therapeutic makeover
.
J Clin Invest
2012
;
122
:
4
12
78.
Sørensen
H
,
Winzell
MS
,
Brand
CL
, et al
.
Glucagon receptor knockout mice display increased insulin sensitivity and impaired beta-cell function
.
Diabetes
2006
;
55
:
3463
3469
79.
Guan
HP
,
Yang
X
,
Lu
K
, et al
.
Glucagon receptor antagonism induces increased cholesterol absorption
.
J Lipid Res
2015
;
56
:
2183
2195
80.
Longuet
C
,
Sinclair
EM
,
Maida
A
, et al
.
The glucagon receptor is required for the adaptive metabolic response to fasting
.
Cell Metab
2008
;
8
:
359
371
81.
Rivero-Gutierrez
B
,
Haller
A
,
Holland
J
, et al
.
Deletion of the glucagon receptor gene before and after experimental diabetes reveals differential protection from hyperglycemia
.
Mol Metab
2018
;
17
:
28
38
82.
Pettus
J
,
Reeds
D
,
Cavaiola
TS
, et al
.
Effect of a glucagon receptor antibody (REMD-477) in type 1 diabetes: a randomized controlled trial
.
Diabetes Obes Metab
2018
;
20
:
1302
1305
83.
Ro
C
,
Chai
W
,
Yu
VE
,
Yu
R
.
Pancreatic neuroendocrine tumors: biology, diagnosis, and treatment
.
Chin J Cancer
2013
;
32
:
312
324
84.
Whytock
KL
,
Carnero
EA
,
Vega
RB
, et al
.
Prolonged glucagon infusion does not affect energy expenditure in individuals with overweight/obesity: a randomized trial
.
Obesity (Silver Spring)
2021
;
29
:
1003
1013
85.
Tan
TM
,
Field
BC
,
McCullough
KA
, et al
.
Coadministration of glucagon-like peptide-1 during glucagon infusion in humans results in increased energy expenditure and amelioration of hyperglycemia
.
Diabetes
2013
;
62
:
1131
1138
86.
Habegger
KM
,
Stemmer
K
,
Cheng
C
, et al
.
Fibroblast growth factor 21 mediates specific glucagon actions
.
Diabetes
2013
;
62
:
1453
1463
87.
Hinds
CE
,
Owen
BM
,
Hope
DCD
, et al
.
A glucagon analogue decreases body weight in mice via signalling in the liver
.
Sci Rep
2021
;
11
:
22577
88.
Miyoshi
H
,
Shulman
GI
,
Peters
EJ
,
Wolfe
MH
,
Elahi
D
,
Wolfe
RR
.
Hormonal control of substrate cycling in humans
.
J Clin Invest
1988
;
81
:
1545
1555
89.
Martin
JR
,
Novin
D
.
Decreased feeding in rats following hepatic-portal infusion of glucagon
.
Physiol Behav
1977
;
19
:
461
466
90.
Penick
SB
,
Hinkle
LE
Jr
.
Depression of food intake induced in healthy subjects by glucagon
.
N Engl J Med
1961
;
264
:
893
897
91.
Geary
N
,
Kissileff
HR
,
Pi-Sunyer
FX
,
Hinton
V
.
Individual, but not simultaneous, glucagon and cholecystokinin infusions inhibit feeding in men
.
Am J Physiol
1992
;
262
:
R975
R980
92.
Burcelin
R
,
Li
J
,
Charron
MJ
.
Cloning and sequence analysis of the murine glucagon receptor-encoding gene
.
Gene
1995
;
164
:
305
310
93.
Heckemeyer
CM
,
Barker
J
,
Duckworth
WC
,
Solomon
SS
.
Studies of the biological effect and degradation of glucagon in the rat perifused isolated adipose cell
.
Endocrinology
1983
;
113
:
270
276
94.
Wu
MS
,
Jeng
CY
,
Hollenbeck
CB
,
Chen
YD
,
Jaspan
J
,
Reaven
GM
.
Does glucagon increase plasma free fatty acid concentration in humans with normal glucose tolerance?
J Clin Endocrinol Metab
1990
;
70
:
410
416
95.
Gerich
JE
,
Lorenzi
M
,
Bier
DM
, et al
.
Effects of physiologic levels of glucagon and growth hormone on human carbohydrate and lipid metabolism. Studies involving administration of exogenous hormone during suppression of endogenous hormone secretion with somatostatin
.
J Clin Invest
1976
;
57
:
875
884
96.
Richter
WO
,
Robl
H
,
Schwandt
P
.
Human glucagon and vasoactive intestinal polypeptide (VIP) stimulate free fatty acid release from human adipose tissue in vitro
.
Peptides
1989
;
10
:
333
335
97.
Lefebvre
P
,
Luyckx
A
,
Bacq
ZM
.
Effects of denervation on the metabolism and the response to glucagon of white adipose tissue of rats
.
Horm Metab Res
1973
;
5
:
245
250
98.
Perea
A
,
Clemente
F
,
Martinell
J
,
Villanueva-Peñacarrillo
ML
,
Valverde
I
.
Physiological effect of glucagon in human isolated adipocytes
.
Horm Metab Res
1995
;
27
:
372
375
99.
Nair
KS
,
Welle
SL
,
Halliday
D
,
Campbell
RG
.
Effect of beta-hydroxybutyrate on whole-body leucine kinetics and fractional mixed skeletal muscle protein synthesis in humans
.
J Clin Invest
1988
;
82
:
198
205
100.
Gerich
JE
,
Lorenzi
M
,
Bier
DM
, et al
.
Prevention of human diabetic ketoacidosis by somatostatin. Evidence for an essential role of glucagon
.
N Engl J Med
1975
;
292
:
985
989
101.
Prip-Buus
C
,
Pegorier
JP
,
Duee
PH
,
Kohl
C
,
Girard
J
.
Evidence that the sensitivity of carnitine palmitoyltransferase I to inhibition by malonyl-CoA is an important site of regulation of hepatic fatty acid oxidation in the fetal and newborn rabbit. Perinatal development and effects of pancreatic hormones in cultured rabbit hepatocytes
.
Biochem J
1990
;
269
:
409
415
102.
Wang
H
,
Zhao
M
,
Sud
N
, et al
.
Glucagon regulates hepatic lipid metabolism via cAMP and Insig-2 signaling: implication for the pathogenesis of hypertriglyceridemia and hepatic steatosis
.
Sci Rep
2016
;
6
:
32246
103.
Scott
RV
,
Bloom
SR
.
Problem or solution: the strange story of glucagon
.
Peptides
2018
;
100
:
36
41
104.
Saltiel
AR
.
Insulin signaling in health and disease
.
J Clin Invest
2021
;
131
:
e142241
105.
Ansarullah
JC
,
Jain
C
,
Far
FF
, et al
.
Inceptor counteracts insulin signalling in β-cells to control glycaemia
.
Nature
2021
;
590
:
326
331
106.
Lee
S
,
Dong
HH
.
FoxO integration of insulin signaling with glucose and lipid metabolism
.
J Endocrinol
2017
;
233
:
R67
R79
107.
Oliva-Vilarnau
N
,
Hankeova
S
,
Vorrink
SU
,
Mkrtchian
S
,
Andersson
ER
,
Lauschke
VM
.
Calcium signaling in liver injury and regeneration
.
Front Med (Lausanne)
2018
;
5
:
192
108.
James
DE
,
Stöckli
J
,
Birnbaum
MJ
.
The aetiology and molecular landscape of insulin resistance
.
Nat Rev Mol Cell Biol
2021
;
22
:
751
771
109.
Lund
A
,
Bagger
JI
,
Christensen
M
,
Knop
FK
,
Vilsbøll
T
.
Glucagon and type 2 diabetes: the return of the alpha cell
.
Curr Diab Rep
2014
;
14
:
555
110.
Campos
GM
,
Rabl
C
,
Havel
PJ
,
Rao
M
,
Schwarz
JM
,
Schambelan
M
,
Mulligan
K
.
Changes in post-prandial glucose and pancreatic hormones, and steady-state insulin and free fatty acids after gastric bypass surgery
.
Surg Obes Relat Dis
2014
;
10
:
1
8
111.
Habegger
KM
,
Heppner
KM
,
Amburgy
SE
, et al
.
GLP-1R responsiveness predicts individual gastric bypass efficacy on glucose tolerance in rats
.
Diabetes
2014
;
63
:
505
513
112.
Russell
SJ
,
El-Khatib
FH
,
Sinha
M
, et al
.
Outpatient glycemic control with a bionic pancreas in type 1 diabetes
.
N Engl J Med
2014
;
371
:
313
325
113.
Chakravarthy
M
,
Parsons
S
,
Lassman
ME
, et al
.
Effects of 13-hour hyperglucagonemia on energy expenditure and hepatic glucose production in humans
.
Diabetes
2017
;
66
:
36
44
114.
Kim
T
,
Holleman
CL
,
Nason
S
, et al
.
Hepatic glucagon receptor signaling enhances insulin-stimulated glucose disposal in rodents
.
Diabetes
2018
;
67
:
2157
2166
115.
Besse-Patin
A
,
Jeromson
S
,
Levesque-Damphousse
P
,
Secco
B
,
Laplante
M
,
Estall
JL
.
PGC1A regulates the IRS1:IRS2 ratio during fasting to influence hepatic metabolism downstream of insulin
.
Proc Natl Acad Sci U S A
2019
;
116
:
4285
4290
116.
Sunilkumar
S
,
Kimball
SR
,
Dennis
MD
.
Glucagon transiently stimulates mTORC1 by activation of an EPAC/Rap1 signaling axis
.
Cell Signal
2021
;
84
:
110010
117.
Perry
RJ
,
Zhang
D
,
Guerra
MT
, et al
.
Glucagon stimulates gluconeogenesis by INSP3R1-mediated hepatic lipolysis
.
Nature
2020
;
579
:
279
283
118.
Okamoto
H
,
Cavino
K
,
Na
E
, et al
.
Glucagon receptor inhibition normalizes blood glucose in severe insulin-resistant mice
.
Proc Natl Acad Sci U S A
2017
;
114
:
2753
2758
119.
Gumbiner
B
,
Esteves
B
,
Dell
V
, et al
.
Single and multiple ascending-dose study of glucagon-receptor antagonist RN909 in type 2 diabetes: a phase 1, randomized, double-blind, placebo-controlled trial
.
Endocrine
2018
;
62
:
371
380
120.
Sharma
AX
,
Quittner-Strom
EB
,
Lee
Y
, et al
.
Glucagon receptor antagonism improves glucose metabolism and cardiac function by promoting AMP-mediated protein kinase in diabetic mice
.
Cell Rep
2018
;
22
:
1760
1773
121.
Gu
L
,
Wang
D
,
Cui
X
, et al
.
Combination of GLP-1 receptor activation and glucagon blockage promotes pancreatic β-cell regeneration in situ in type 1 diabetic mice
.
J Diabetes Res
2021
;
2021
:
7765623
122.
Kostic
A
,
King
TA
,
Yang
F
, et al
.
A first-in-human pharmacodynamic and pharmacokinetic study of a fully human anti-glucagon receptor monoclonal antibody in normal healthy volunteers
.
Diabetes Obes Metab
2018
;
20
:
283
291
123.
Clemmensen
C
,
Chabenne
J
,
Finan
B
, et al
.
GLP-1/glucagon coagonism restores leptin responsiveness in obese mice chronically maintained on an obesogenic diet
.
Diabetes
2014
;
63
:
1422
1427
124.
Varin
EM
,
Mulvihill
EE
,
Baggio
LL
, et al
.
Distinct neural sites of GLP-1R expression mediate physiological versus pharmacological control of incretin action
.
Cell Rep
2019
;
27
:
3371
3384.e3
125.
Nahra
R
,
Wang
T
,
Gadde
KM
, et al
.
Effects of cotadutide on metabolic and hepatic parameters in adults with overweight or obesity and type 2 diabetes: a 54-week randomized phase 2b study
.
Diabetes Care
2021
;
44
:
1433
1442
Readers may use this article as long as the work is properly cited, the use is educational and not for profit, and the work is not altered. More information is available at https://www.diabetesjournals.org/journals/pages/license.