Glucagon stimulates hepatic glucose production, in part by promoting the uptake and catabolism of amino acids. Inhibition of the liver glucagon receptor (GCGR) results in elevated plasma amino acids, which triggers the proliferation of pancreatic α-cells, forming a liver–α-cell loop. This study aims to delineate hepatic signaling molecules downstream of GCGR that mediate the liver–α-cell loop. We knocked down liver GCGR, its G-coupled protein GNAS, and two GNAS downstream effectors, PKA and EPAC2 (RAPGEF4). Mice with GCGR, GNAS, and PKA knockdown had similar suppression of hepatic amino acid catabolism genes, hyperaminoacidemia, and α-cell hyperplasia, but those with EPAC2 knockdown did not. We then demonstrated that activating liver PKA was sufficient to reverse hyperaminoacidemia and α-cell hyperplasia caused by GCGR blockade. These results suggest that liver GCGR signals through PKA to control amino acid metabolism and that hepatic PKA plays a critical role in the liver–α-cell loop.

Article Highlights

  • A liver–α-cell loop exists, where inhibition of the liver glucagon receptor (GCGR) causes hyperaminoacidemia and pancreatic α-cell hyperplasia, but the GCGR downstream factors responsible for these effects are not clear.

  • We silenced GCGR, its G-coupled protein GNAS, and two GNAS downstream effectors, PKA and EPAC2, to assess their effects on the liver–α-cell loop.

  • Inhibition of the GCGR-GNAS-PKA pathway suppresses amino acid catabolism and causes α-cell hyperplasia, whereas PKA activation promotes amino acid catabolism and reduces alpha cell mass even when GCGR is blocked.

  • Our study establishes hepatic PKA as the critical regulator of the liver–α-cell loop.

Glucagon is a peptide hormone secreted from α-cells in the pancreas and serves as a counter-regulatory hormone to insulin. Glucagon is secreted in response to a decline in blood glucose level and activates the glucagon receptor (GCGR) in the liver to stimulate glycogenolysis and gluconeogenesis (1). Glucagon levels are often elevated in patients with type 2 diabetes (T2D), exacerbating hyperglycemia (2). To investigate therapeutic potential of glucagon action blockade to lower blood glucose in patients with diabetes, GCGR inhibitors, including small molecules, antisense oligonucleotides, and antagonist antibodies, have been developed. In clinical trials, these molecules showed glucose-lowering efficacies without hypoglycemia but were often associated with potential safety signals, including increased circulating levels of ALT, LDL cholesterol, amino acids, and/or liver fat (3–7).

One of the major safety concerns associated with GCGR inhibition is hyperaminoacidemia, because it may lead to pancreatic α-cell hyperplasia and tumor formation (8). Glucagon controls whole-body amino acid homeostasis via regulation of hepatic amino acid use. Glucagon promotes amino acid uptake by hepatocytes and their breakdown into gluconeogenesis precursors via increased expression of hepatic genes involved in amino acid catabolism and transport (9,10). Gcgr-null mice present decreased hepatic expression of amino acid catabolism genes and hyperaminoacidemia (11–13), demonstrating a tight link between liver amino acid catabolism and plasma amino acid levels. The effects of glucagon on amino acid metabolism described in mice are conserved in humans. Patients with pancreatectomy develop hypoglucagonemia and hyperaminoacidemia (14), whereas patients with glucagonoma exhibit hyperglucagonemia and hypoaminoacidemia (15). Recent reports by us and others established that hyperaminoacidemia causes α-cell hyperplasia observed in whole-body and liver-specific GCGR-knockout mice (16,17). Mouse and zebrafish studies uncovered that amino acids promote α-cell proliferation in a calcium-sensing receptor and mTOR-dependent process (10,18) and that amino acid transporter SLC38A5 relays the signal of hyperaminoacidemia to induce α-cell proliferation (19,20).

These data highlight the role of glucagon in regulating hepatic and whole-body amino acid homeostasis and indicate that a lack of glucagon signaling results in hyperaminoacidemia and α-cell hyperplasia. However, the hepatic GCGR signaling pathway that controls amino acid metabolism remains unclear. GCGR is a G-protein–coupled receptor that signals through Gsα and possibly through Gqα (21). Gsα seems to be the key downstream mediator regulating amino acid metabolism, because Gsα-knockout mice develop α-cell hyperplasia (22). Activated Gsα stimulates adenyl cyclase to produce cAMP, the rise of which activates downstream effectors. Two such downstream effectors have been described in hepatocytes: protein kinase A (PKA) and exchange protein directly activated by cAMP 2 (EPAC2), also known as Rap guanine nucleotide exchange factor 4 (RAPGEF4). PKA has been described as a critical effector of hepatic GCGR signaling controlling glucose metabolism, because it phosphorylates key metabolic enzymes and activates transcriptional programs for gluconeogenesis through its downstream transcription factor cAMP response element-binding protein (CREB). EPAC2 is another hepatic cAMP effector, reported to regulate glucose metabolism, membrane current, cell survival, and cholesterol metabolism, independently of PKA (23–26). The roles of PKA, EPAC2, or other effectors downstream of Gsα in hepatic amino acid metabolism are not fully understood. In this study, we used AAV8-shRNA to knock down liver GCGR, Gsα, PKA, and EPAC2 in diet-induced obese (DIO) and lean mice and examined hepatic expression profiles, plasma levels of amino acids and glucagon, and pancreas morphology. We report here that PKA is the major GCGR downstream effector that regulates amino acid metabolism and α-cell mass.

Mouse Studies

All procedures were conducted in compliance with protocols approved by the Institutional Animal Care and Use Committee of Regeneron Pharmaceuticals. Male DIO mice (cat. no. 380050; The Jackson Laboratory) and lean mice (cat. no. 000664; The Jackson Laboratory) were housed with one to five mice per cage in a controlled environment (12-h light/dark cycle; 22 ± 1°C), with free access to food and water. DIO mice were fed a high-fat diet (cat. no. D12492i; Research Diet) starting at 6 weeks of age for 20–22 weeks at the start of all studies, except the study evaluating the off-tissue effect of AAV8, in which mice were fed a high-fat diet for 29 weeks. Lean mice were fed a regular chow diet (cat. no. 5053; LabDiet) and were 13 weeks of age at the start of the study. All mice were sacrificed in the fed condition. Mice were assigned to experimental groups based on baseline body weight and blood glucose levels. Experimenters were not blinded during the studies.

AAV8 expressing a control scrambled shRNA or shRNAs against Gcgr, Gnas, Prkaca/b, Epac2 (Rapgef4), and Prkar1a, under the U6 promoter, was designed and generated by Vector Biolabs. Mice were injected with AAV8 particles intravenously via the tail vein at a dose of 1 × 1012 (DIO) or 5 × 1011 (lean) genome copies per mouse on day 0. For antibody treatment, mice were injected subcutaneously with GCGR or control antibody (10 mg/kg; diluted with sterile PBS) weekly. Body weight was monitored weekly. Fed blood glucose was measured in the morning via the AlphaTrak glucose monitoring system (Zoetis). Blood samples were collected via submandibular bleeds weekly into lithium heparin microtainers (cat. no. 365985; Becton Dickinson) and mixed with DPP4 and protease inhibitors (cat. no. DPP4-010; EMD Millipore; cat. no. 11836170001; Sigma). The blood samples were centrifuged at 10,000 rpm for 10 min at 4°C, and the supernatant plasma was collected for storage at −80°C until analysis.

Blood Chemistry

Total plasma amino acid concentration, except for glycine, was measured with a colorimetric kit (cat. no. MAK002; Sigma). Individual amino acid concentration was measured with high-performance liquid chromatography by the Texas A&M University Protein Chemistry Laboratory. Plasma glucagon and insulin were measured by ELISA (cat. nos. 10-1281-01 and 10-1247-01; Mercodia).

Liver Glycogen Content Measurement

Liver glycogen content was measured with a colorimetric kit (cat. no. MAK016; Sigma) according to the manufacturer’s instructions. Glycogen was extracted from the liver by homogenizing 10–50 mg frozen liver tissue in water (20 μL/mg) using the FastPrep-24 instrument (MP Biomedicals). The homogenates were boiled for 10 min, and insoluble material was removed after centrifugation at 18,000g for 10 min. Samples were diluted 20-fold (for PKA activation) or 100-fold before assay. Glycogen content was normalized to the weight of the liver used for extraction.

Western Blot

Liver samples were lysed with ice-cold radioimmunoprecipitation assay lysis buffer in the presence of protease and phosphatase inhibitors (cat. nos. 78430 and 78420; Thermo Fisher Scientific) using the FastPrep-24 instrument. For GCGR Western blot, liver samples were extracted with a G-coupled protein receptor extraction reagent (cat. no. A43436; Thermo Fisher Scientific) using a tissue grinder (cat. no. 47732; VWR International). Lysates for GNAS and PRKACA blotting were diluted with sample loading buffer and incubated for 5 min at 90°C, whereas lysates for GCGR and EPAC2 blotting were incubated at room temperature for 30 min. Samples were separated by SDS-PAGE, transferred to nitrocellulose membranes, and probed with the following antibodies: GCGR (cat. no. ab75240; Abcam), GNAS (cat. no. PA5-19315; Invitrogen), PRKACA (cat. no. PA5-17626; Invitrogen), and EPAC2 (cat. no. 43239; Cell Signaling Technology).

Quantitative RT-PCR

All tissues were collected in RNALater (cat. no. AM7021; Thermo Fisher Scientific) and stored frozen. RNA was extracted from the tissue with the GeneJET RNA purification kit (cat. no. K0732; Thermo Fisher Scientific) or RNeasy Plus Mini Kit (cat. no. 74134; Qiagen). Genomic DNA was removed using the TURBO DNA-free kit (cat. no. AM1907; Invitrogen). mRNA was reverse transcribed into cDNA using the High-Capacity cDNA Reverse Transcription Kit (cat. no. 4374966; Thermo Fisher Scientific). TaqMan analysis was performed in triplicate with TaqMan Fast Advanced Master Mix (2×; cat. no. A44360; Thermo Fisher Scientific) using the QuantStudio 6 Flex Real-Time PCR System (Thermo Fisher Scientific). The following probes from Thermo Fisher Scientific were used: Gapdh (Mm99999915_g1), Gcgr (Mm00433546_m1), Gnas (Mm01242435_m1), Prkaca (Mm00660092_m1), Prkacb (Mm01312555_m1), Rapgef4 (Mm01327556_m1), and Prkar1a (Mm00660315_m1). Gapdh was used as the internal control gene for normalization. Expression of genes of interest was calculated relative to the Gapdh housekeeping gene and control group using the ΔΔ Ct method.

Immunohistochemistry and Immunofluorescence

Pancreata were fixed in 10% neutral buffered formalin solution for 48 h, dehydrated in 70% ethanol, and then embedded in paraffin. Tissue sections 6 mm in thickness were prepared from paraffin-embedded blocks. Analysis for α- and β-cell mass was carried out as previously described (27).

Immunofluorescence was carried out using another set of sections from the same pancreata. Slides were stained for glucagon with human α-glucagon antibody (REGN745) and for insulin with guinea pig α-insulin antibody (cat. no. A0564; Dako) or mouse α-insulin antibody conjugated to AF488 (cat. no. 53-9769-82; Invitrogen). Slides were scanned with a Zeiss Axio Scan.Z1 slide scanner (Zeiss) with a 20× objective, and the images were analyzed with HALO (version 3.6; Indica Labs). Islets from each section were identified using the classifier function in HALO based on glucagon, insulin, and DAPI staining. The islet selections were then manually checked and adjusted. All identified islets were used for subsequent analyses. Glucagon-positive cells were counted with the CytoNuclear (version 2.0) analysis module (Indica Labs). α-Cell size was estimated by dividing the glucagon-positive cell area in the islets by the number of glucagon-positive cells in the islets. Glucagon-positive cell area was measured by a classifier in HALO trained to identify glucagon-positive cells. Mean fluorescence intensity of glucagon-positive pixels in the islets was measured using Area Quantification FL (version 1.0) analysis module (Indica Labs).

RNA Sequencing

RNA extraction, library preparation, and analysis were performed as previously described (28). Differentially expressed genes were obtained using cutoffs Padj < 0.05 and fold-change >1.5 with the DESeq2 package (29). Fold-change for an individual sample was calculated by dividing the transcripts per million (TPM) for each gene of each sample by the average TPM for each gene of the control group. The combined differentially expressed genes from the four knockdown (KD) groups from gluconeogenesis (cat. no. MM15392; Reactome), amino acid transporters (30), and amino acid catabolism (Gene Ontology identifier 0009063) are displayed in the heat map. All urea cycle genes are displayed in the heat map. The RNA sequencing (RNA-seq) data have been deposited in the Gene Expression Omnibus (GEO) under accession no. GSE268864.

Statistical Analysis

Statistical and graphical data analyses were performed using Prism 9 (GraphPad Software, Inc.). Data are expressed as mean ± SEM. When two independent groups were present, mean values were compared with unpaired two-tailed t tests. When three or more independent groups were present, parameters were analyzed by one-way ANOVA when one independent variable was present or two-way ANOVA with post hoc Dunnett test when two independent variables were present; a threshold of P < 0.05 was considered statistically significant.

Data and Resource Availability

The RNAseq data have been deposited in GEO under accession no. GSE268864. This article does not report original code. Any additional information required to reanalyze the data reported in this article is available from the corresponding author on request.

Hepatic KD of Gcgr, Gnas, and Prkaca/b Downregulates Expression of Genes Involved in Amino Acid Catabolism

To identify the GCGR signaling molecules mediating amino acid regulation, we used AAV8-shRNA to knock down liver Gcgr, Gnas, Prkaca, and Prkacb (two catalytic subunits of PKA) and Epac2 (Rapgef4) in DIO mice, a model of T2D, for which therapeutic utility of GCGR antagonism has been considered. Liver TaqMan analysis 4 weeks after AAV injection showed >70% mRNA reduction for all target genes, including Epac2 (Fig. 1A). Reduction in the target protein levels was confirmed by Western blotting (Supplementary Fig. 1). GCGR inactivation in DIO mice led to 30–45% reduction in ad lib glucose levels (6,20). Therefore, changes in blood glucose levels at any point were expected to reflect changes in GCGR KD efficiency of the time point. Starting 2 weeks postinjection, ad lib fed glucose levels were 30% reduced in mice with liver Gcgr KD and remained at that level until the end of study, suggesting that maximal KD by the shRNA was likely achieved for the last 2 weeks of the study (Fig. 1B). Similar glucose lowering was observed in mice with Gnas and Prkaca/b KD, but not in mice with Epac2 KD. (Fig. 1B). Body weight of mice with Prkaca/b KD was slightly lower than in the control group at week 4 (Supplementary Fig. 2A). Terminal liver glycogen content was not different between the groups (Supplementary Fig. 2B).

Figure 1

Hepatic Gcgr, Gnas, and Prkaca/b KD downregulates genes involved in amino acid catabolism. A: TaqMan analysis of liver from DIO mice 4 weeks after AAV injections (1 × 1012 genome copies per mouse; n = 10). B: Fed blood glucose levels in DIO mice before and at multiple time points after injection of AAVs (n = 10). C: Heat map of combined differentially expressed genes from each AAV treatment. Genes from gluconeogenesis, amino acid catabolism, amino acid transporters, and urea cycle are displayed. Scale represents log2 fold-change of gene TPM of sample from treatment group over average gene TPM of control group (n = 10). All data are given as mean ± SEM. P values determined by one-way (A) or two-way (B) ANOVA in comparison with control group. **P ≤ 0.01, ***P ≤ 0.001, ****P ≤ 0.0001.

Figure 1

Hepatic Gcgr, Gnas, and Prkaca/b KD downregulates genes involved in amino acid catabolism. A: TaqMan analysis of liver from DIO mice 4 weeks after AAV injections (1 × 1012 genome copies per mouse; n = 10). B: Fed blood glucose levels in DIO mice before and at multiple time points after injection of AAVs (n = 10). C: Heat map of combined differentially expressed genes from each AAV treatment. Genes from gluconeogenesis, amino acid catabolism, amino acid transporters, and urea cycle are displayed. Scale represents log2 fold-change of gene TPM of sample from treatment group over average gene TPM of control group (n = 10). All data are given as mean ± SEM. P values determined by one-way (A) or two-way (B) ANOVA in comparison with control group. **P ≤ 0.01, ***P ≤ 0.001, ****P ≤ 0.0001.

Close modal

To examine the effect of target gene KD on the liver transcriptome, we conducted bulk RNA-seq analysis using the liver harvested at study termination. Consistent with the glucose lowering in mice with liver Gcgr, Gnas, and Prkaca/b KD, these mice showed reduced expression of key gluconeogenic genes, such as G6pc, Pck1, and Fbp1 (Fig. 1C). Genes involved in the catabolism and transport of amino acids and the urea cycle were largely downregulated in these mice, but not in mice with Epac2 KD (Fig. 1C). These results show that inactivation of GCGR, GNAS, and PKA limits hepatic amino acid use in mice, whereas EPAC2 does not affect liver amino acid metabolism.

Inhibition of Hepatic GCGR-GNAS-PKA Axis Induces Hyperaminoacidemia and Pancreatic α-Cell Hyperplasia

We next tested if the amino acid metabolism gene changes observed in the liver of mice with Gcgr, Gnas, and Prkaca/b KD accompanied alterations in circulating amino acid levels and α-cell mass. Starting 2 weeks after AAV injection, mice with liver Gcgr, Gnas, and Prkaca/b KD showed elevated plasma amino acids levels, which persisted throughout the study (Fig. 2A).

Figure 2

Hepatic Gcgr, Gnas, and Prkaca/b KD induces hyperaminoacidemia and α-cell hyperplasia. A: Plasma amino acid levels in DIO mice before and at multiple time points after injection of AAVs (n = 9–10). B: Individual plasma amino acid levels in DIO mice 4 weeks after AAV injections (n = 10). C: Pancreas weight collected from mice 4 weeks after AAV injections (n = 10). D: Glucagon and insulin immunohistochemistry of representative pancreas sections and single islets at ×10 magnification from mice injected with AAVs 4 weeks before pancreas collection. E and F: α-Cell (E) and β-cell (F) mass measured from pancreas staining as in panel D (n = 10). G and H: Plasma glucagon (G) and insulin (H) were measured from plasma collected at day 28 after AAV injections (n = 10). All data are given as mean ± SEM. P values determined by one-way (B, C, and EH) or two-way (A) ANOVA in comparison with control group. *P ≤ 0.05, **P ≤ 0.01, ***P ≤ 0.001, ****P ≤ 0.0001.

Figure 2

Hepatic Gcgr, Gnas, and Prkaca/b KD induces hyperaminoacidemia and α-cell hyperplasia. A: Plasma amino acid levels in DIO mice before and at multiple time points after injection of AAVs (n = 9–10). B: Individual plasma amino acid levels in DIO mice 4 weeks after AAV injections (n = 10). C: Pancreas weight collected from mice 4 weeks after AAV injections (n = 10). D: Glucagon and insulin immunohistochemistry of representative pancreas sections and single islets at ×10 magnification from mice injected with AAVs 4 weeks before pancreas collection. E and F: α-Cell (E) and β-cell (F) mass measured from pancreas staining as in panel D (n = 10). G and H: Plasma glucagon (G) and insulin (H) were measured from plasma collected at day 28 after AAV injections (n = 10). All data are given as mean ± SEM. P values determined by one-way (B, C, and EH) or two-way (A) ANOVA in comparison with control group. *P ≤ 0.05, **P ≤ 0.01, ***P ≤ 0.001, ****P ≤ 0.0001.

Close modal

Of the 19 measured amino acids, 10, seven, and six amino acids were increased significantly by liver Gcgr, Gnas, and Prkaca/b KD, respectively (Fig. 2B). α-Cell mass was increased in mice with liver Gcgr and Prkaca/b KD and had a trend toward increase in mice with Gnas KD (Fig. 2D and E). There were no differences in pancreas weight or β-cell mass between the groups (Fig. 2C and F). Circulating glucagon levels were more than 10-fold elevated in mice with liver Gcgr, Gnas, and Prkaca/b KD (Fig. 2G). Plasma insulin levels were not different between the groups (Fig. 2H). Changes in pancreas morphometry or circulating glucagon or insulin levels were not detected in mice with Epac2 KD compared with the control mice. These data demonstrate that PKA, not EPAC2, is the cAMP-responsive element downstream of GCGR-GNAS, controlling hepatic amino acid catabolism, plasma amino acid levels, and α-cell mass.

PKA Activation Reverses Effects of GCGR Blockade on Hepatic Amino Acid Metabolism Gene Expressions

To assess the contribution of hepatic PKA in mediating GCGR regulation of amino acid homeostasis and α-cell mass, we activated PKA in the liver of DIO mice administered with GCGR-blocking antibody, which promotes hyperaminoacidemia and α-cell hyperplasia (6,20). We activated PKA via AAV8-shRNA–mediated KD of Prkar1a, the major inhibitory PKA regulatory subunit (31–33). The Prkar1a mRNA level was reduced by 80% in the liver of DIO mice receiving AAV (Fig. 3A) and not in other tissues tested (Supplementary Fig. 3). Increased hepatic PKA activity by this approach was confirmed by Western blotting for phosphorylated PKA substrates. Band intensities for phosphorylated PKA substrates increased in mice with Prkar1a KD, irrespective of the antibody treatment, whereas the signal decreased in mice treated with control AAV and GCGR antibody (Fig. 3B). Terminal body weight was higher for the control group than for groups with liver PKA activation alone or GCGR inhibition alone (Supplementary Fig. 4A). Blood glucose levels were decreased by GCGR antibody treatment alone, consistent with our previous report (6). Mice with PKA activation alone showed transient hyperglycemia, reaching a peak of 330 mg/dL at day 14, before returning to baseline glucose levels at day 28 (Fig. 3C). Mice with GCGR inhibition and PKA reactivation showed glucose lowering at day 7, when the KD of Prkar1a may have been incomplete, followed by transient hyperglycemia, similar to mice with Prkar1a KD alone (Fig. 3C). Terminal liver glycogen content was significantly lower in mice with Prkar1a KD, with or without GCGR inhibition (Supplementary Fig. 4B).

Figure 3

Hepatic PKA activation reverses effects of GCGR inhibition on amino acid catabolism gene expression. A: TaqMan analysis of liver from DIO mice 4 weeks after AAV injections (1 × 1012 genome copies per mouse; n = 10). B: Western blot analysis of phosphorylated PKA (pPKA) substrate levels in mice liver (top). Ponceau S stain of membrane serves as loading control. Quantification of pPKA substrate/Ponceau signals (bottom; n = 5). C: Fed blood glucose levels in DIO mice before and at multiple time points after injection of AAVs (n = 10). D: Heat map showing expression of genes as shown in Fig. 1D from mouse liver. Scale represents log2 fold-change of gene TPM of each sample from treatment group over average gene TPM of control group (n = 10). All data are given as mean ± SEM. P values were determined by one-way (A and B) or two-way (C) ANOVA in comparison with control group. *P ≤ 0.05, **P ≤ 0.01, ***P ≤ 0.001, ****P ≤ 0.0001.

Figure 3

Hepatic PKA activation reverses effects of GCGR inhibition on amino acid catabolism gene expression. A: TaqMan analysis of liver from DIO mice 4 weeks after AAV injections (1 × 1012 genome copies per mouse; n = 10). B: Western blot analysis of phosphorylated PKA (pPKA) substrate levels in mice liver (top). Ponceau S stain of membrane serves as loading control. Quantification of pPKA substrate/Ponceau signals (bottom; n = 5). C: Fed blood glucose levels in DIO mice before and at multiple time points after injection of AAVs (n = 10). D: Heat map showing expression of genes as shown in Fig. 1D from mouse liver. Scale represents log2 fold-change of gene TPM of each sample from treatment group over average gene TPM of control group (n = 10). All data are given as mean ± SEM. P values were determined by one-way (A and B) or two-way (C) ANOVA in comparison with control group. *P ≤ 0.05, **P ≤ 0.01, ***P ≤ 0.001, ****P ≤ 0.0001.

Close modal

Terminal liver RNA-seq showed that gluconeogenic genes (Pck1, G6pc, Fbp1) were downregulated by GCGR antibody treatment alone and upregulated by PKA activation with or without GCGR antibody administration (Fig. 3D). These data confirm previously reported effects of GCGR inhibition or PKA activation on glucose homeostasis in mice (6,31–34). Next, effects of GCGR inhibition alone, PKA activation alone, or the combination of both on mRNA expression levels of hepatic amino acid metabolism genes were compared. Expression of genes involved in amino acid catabolism, amino acid transporters, and the urea cycle was decreased in mice with GCGR antibody treatment alone and increased in mice with liver PKA activation alone (Fig. 3D). Mice with both GCGR inhibition and PKA reactivation showed similar expression profiles to mice with PKA activation alone. These data demonstrate that PKA is the major signaling molecule mediating GCGR regulation of hepatic amino acid metabolism.

PKA Activation Reverses Effects of GCGR Blockade on Amino Acid Catabolism and Pancreatic α-Cell Mass

We assessed effects of GCGR inhibition alone, liver PKA activation alone, or the combination of both on plasma amino acid levels and pancreatic α-cells. Plasma amino acid levels were increased twofold by GCGR antibody treatment alone and 50% reduced by liver PKA activation alone (Fig. 4A). Mice with GCGR inhibition and PKA activation showed plasma amino acid levels similar to those in mice with PKA activation alone (Fig. 4A). GCGR antibody treatment increased the levels of all amino acids except aspartic acid and glutamic acid (cysteine was not measured), whereas PKA activation, alone or in combination with GCGR antibody, significantly reduced all amino acids except arginine, aspartic acid, glutamic acid, and lysine (Fig. 4B). Pancreas weight and α-cell mass was increased by GCGR antibody treatment alone and decreased by liver PKA activation alone or in combination with GCGR antibody treatment (Fig. 4C–E). β-Cell mass was unchanged across groups (Fig. 4F). Terminal plasma glucagon levels were increased by >25-fold in mice receiving GCGR antibody alone, whereas the levels were below the detection limit in mice with liver PKA activation alone or in combination with GCGR inhibition (Fig. 4G). Terminal plasma insulin levels were decreased in mice with GCGR antibody treatment, liver PKA activation, or the combination of both (Fig. 4H). Further pancreas analysis revealed that GCGR inhibition induced α-cell mass expansion accompanied by increases in the number and size of α-cells, as well as glucagon staining intensity (Fig. 5). Reduced α-cell mass observed with PKA activation alone, or in combination with GCGR inhibition, accompanied reductions in α-cell size and glucagon staining intensity without changes in α-cell number (Fig. 5). Collectively, these data demonstrate that PKA activation and GCGR inhibition have opposing effects on plasma amino acid levels and α-cell mass and that liver PKA activation fully reverses GCGR inhibition–induced hyperaminoacidemia and α-cell hyperplasia.

Figure 4

Hepatic PKA activation reduces α-cell mass. A: Plasma amino acid levels in DIO mice before and at multiple time points after injection of AAVs (n = 10). B: Individual plasma amino acid levels in DIO mice 4 weeks after AAV injections (n = 10). C: Pancreas weight collected from mice 4 weeks after AAV injections (n = 10). D: Glucagon and insulin immunohistochemistry of representative pancreas sections and single islets at ×10 magnification from mice injected with AAVs 4 weeks before pancreas collection. E and F: α-Cell (E) and β-cell (F) mass measured from pancreas staining as in panel D (n = 10). G and H: Plasma glucagon (G) and insulin (H) were measured from plasma collected at day 28 after AAV injections (n = 9–10). All data are given as mean ± SEM. P values determined by one-way (B, C, and EH) or two-way (A) ANOVA in comparison with control group. *P ≤ 0.05, **P ≤ 0.01, ***P ≤ 0.001, ****P ≤ 0.0001.

Figure 4

Hepatic PKA activation reduces α-cell mass. A: Plasma amino acid levels in DIO mice before and at multiple time points after injection of AAVs (n = 10). B: Individual plasma amino acid levels in DIO mice 4 weeks after AAV injections (n = 10). C: Pancreas weight collected from mice 4 weeks after AAV injections (n = 10). D: Glucagon and insulin immunohistochemistry of representative pancreas sections and single islets at ×10 magnification from mice injected with AAVs 4 weeks before pancreas collection. E and F: α-Cell (E) and β-cell (F) mass measured from pancreas staining as in panel D (n = 10). G and H: Plasma glucagon (G) and insulin (H) were measured from plasma collected at day 28 after AAV injections (n = 9–10). All data are given as mean ± SEM. P values determined by one-way (B, C, and EH) or two-way (A) ANOVA in comparison with control group. *P ≤ 0.05, **P ≤ 0.01, ***P ≤ 0.001, ****P ≤ 0.0001.

Close modal
Figure 5

Hepatic PKA activation reduces α-cell size. A: Glucagon (red), insulin (white), and DAPI nuclei (blue) immunofluorescence staining of representative pancreas sections from mice injected with AAVs 4 weeks before pancreas collection. B: Percentage of glucagon-positive cells in all identified islets from pancreas sections (n = 9–10). C: Average α-cell size, calculated by dividing glucagon-positive area in islets by number of glucagon-positive cells (n = 9–10). D: Mean intensity of glucagon-positive pixels in all identified islets from pancreas sections (n = 9–10). All data are given as mean ± SEM. P values determined by one-way ANOVA. **P ≤ 0.01, ***P ≤ 0.001, ****P ≤ 0.0001.

Figure 5

Hepatic PKA activation reduces α-cell size. A: Glucagon (red), insulin (white), and DAPI nuclei (blue) immunofluorescence staining of representative pancreas sections from mice injected with AAVs 4 weeks before pancreas collection. B: Percentage of glucagon-positive cells in all identified islets from pancreas sections (n = 9–10). C: Average α-cell size, calculated by dividing glucagon-positive area in islets by number of glucagon-positive cells (n = 9–10). D: Mean intensity of glucagon-positive pixels in all identified islets from pancreas sections (n = 9–10). All data are given as mean ± SEM. P values determined by one-way ANOVA. **P ≤ 0.01, ***P ≤ 0.001, ****P ≤ 0.0001.

Close modal

Hepatic PKA Mediates Amino Acid Metabolism in Lean Mice

To test if our DIO mouse findings were relevant to lean mice, we knocked down liver Gcgr alone, Prkaca/b alone, or Prkar1a with or without GCGR antibody treatment. TaqMan analysis 4 weeks after AAV injections confirmed that the target gene mRNAs were knocked down >70% (Supplementary Fig. 5A). Blood glucose levels were reduced by Gcgr KD, Prkaca/b KD, and GCGR antibody treatment and were increased by Prkar1a KD (Supplementary Fig. 5B). Terminal body weight was significantly reduced only by Prkaca/b KD (Supplementary Fig. 5C). Plasma levels of total amino acids and glucagon were increased by Gcgr KD, Prkaca/b KD, and GCGR antibody treatment and were reduced by PKA activation with or without GCGR antibody treatment (Supplementary Fig. 5D and E). These data demonstrate that the dominant role hepatic PKA plays in amino acid metabolism is applicable to lean mice.

Liver GCGR signaling is well recognized as a critical regulator of amino acid homeostasis; however, GCGR downstream mediators essential to convey the signal are not well characterized. Reduced expression of hepatic amino acid catabolism genes, hyperaminoacidemia, α-cell hyperplasia, and hyperglucagonemia are common features of mice with whole-body or liver-specific GCGR inhibition (6,16,17,19,20,35). We now extend the same observation to mice with hepatic PKA inactivation. PKA inactivation in the mouse liver was achieved by knocking down the catalytic subunits of PKA without affecting liver Gcgr expression. The mice showed changes in hepatic amino acid metabolism gene expression, hyperaminoacidemia, α-cell hyperplasia, and hyperglucagonemia, all to the same extent as mice with hepatic GCGR inactivation. In contrast, mice with hepatic Epac2 KD did not show changes in plasma levels of amino acids or glucagon or α-cell mass. These data indicate that PKA is the primary downstream effector of GCGR-GNAS, controlling hepatic amino acid metabolism, and that EPAC2 does not play a role in the pathway.

To further investigate the contribution of PKA in GCGR regulation of amino acids, PKA was activated via inhibition of the major inhibitory PKA regulatory subunit in mice, and simultaneously, GCGR signaling was inhibited by administration of GCGR-blocking antibody. Mice with hepatic PKA activation alone developed hypoaminoacidemia, hypoglucagonemia, and reduced α-cell mass, which are opposite phenotypes of mice with hepatic GCGR or PKA inactivation. Mice receiving GCGR-blocking antibody with hepatic PKA activation showed identical phenotypes to mice with hepatic PKA activation alone, suggesting that PKA activation fully overrides the effect of GCGR inhibition on amino acids, glucagon, and α-cells. These data highlight PKA as the dominant effector of GCGR-GNAS, controlling hepatic amino acid metabolism and α-cell mass.

Although PKA has been considered the major effector of GCGR signaling downstream of GNAS, the other effector, EPAC2, has been reported to regulate diverse liver functions in response to glucagon. A recent study found that liver EPAC2 and its downstream factor RAP1 regulate glucose homeostasis (26), and the inhibition of EPAC2 leads to increased gluconeogenesis. In our DIO mouse model, liver Epac2 KD resulted in a small yet persistent increase in fed blood glucose levels (Fig. 1B), which agrees with the previous report, although gluconeogenesis gene expressions were not significantly altered. Our results suggest that EPAC2 does not affect hepatic amino acid catabolism or plasma amino acid levels.

Several amino acids are known to play critical roles in the liver–α-cell axis. It has been reported that alanine, glutamine, and arginine stimulate α-cell proliferation (19,20,36), whereas alanine, arginine, cysteine, and proline stimulate glucagon secretion from α-cells (37). Among the five key amino acids in the liver–α-cell axis, cysteine was not measured in this study. Significant increases or a trend toward such increases in the circulating levels of alanine, glutamine, arginine, and proline were observed in mice with Gcgr, Gnas, and Prkaca/b KD (Fig. 2B). Mice receiving GCGR antibody showed further elevation in these amino acids compared with mice with the KDs, consistent with greater α-cell mass and plasma glucagon levels (Fig. 4B, E, and G). Liver PKA activation in mice with or without GCGR blockade lowered most amino acids, including alanine, glutamine, and proline, similar to reported changes in mice receiving a GCGR agonist treatment (35) (Fig. 4B). Heterozygosity in a GCGR variant that reduces cAMP signaling does not accompany changes in plasma amino acid levels in humans (38), similar to normal plasma amino acid levels detected in GCGR+/− mice (8). In this study, changes in plasma amino acids were observed in mice with 70–90% reductions in mRNA levels of Gcgr, Gnas, and Prkaca/b (Fig. 1A), suggesting the extent of GCGR-GNAS-PKA pathway inhibition required to induce detectable changes in plasma amino acids. Overall, the present mouse study demonstrates that the GCGR-GNAS-PKA pathway controls homeostasis of amino acids, including key amino acids in the liver–α-cell axis.

Mice with plasma amino acid changes in either direction showed broad hepatic expression changes in amino acid catabolism, transport, and the urea cycle genes. Certain plasma amino acid changes seemed to be a direct result of changes in genes that metabolize them; for example, mice with GCGR blockade and PKA activation/inactivation showed changes in plasma glutamine levels and expression levels of its catabolic enzyme Gls2 (Figs. 1C, 2,B, 3,D, and 4,B). In contrast, changes in the plasma alanine and proline levels of these mice did not accompany expression changes in their catabolic enzymes Gpt1/2, Prodh1/2, and Aldh4a1 (Figs. 2B and 4,B), suggesting additional mechanisms may influence their plasma levels. Posttranscriptional regulation of catabolic enzymes represents a potential mechanism, because acute and posttranscriptional regulation of amino acid metabolism by glucagon have been reported. Glucagon increases plasma amino acid clearance and ureagenesis within minutes in mice (12) and stimulates two urea cycle enzymes, CPS1 and OTC, by promoting their deacetylation (39). In another study, CPS1 activity was increased with GCGR activation and vice versa without changes in CPS1 mRNA levels (35). Whether PKA regulates amino acid metabolism posttranscriptionally and/or acutely remains to be tested.

Decreases in pancreatic α-cell mass and proglucagon transcription have been reported in mice receiving GCGR agonist and rats and mice with glucagonoma implants (35,40,41). Our data suggest that the observed α-cell phenotypes in these rodents were likely a result of PKA-mediated increases in amino acid catabolism and hypoaminoacidemia. α-Cell atrophy and reduced glucagon production in mice with liver PKA activation (Fig. 5A, C, and D) are consistent with prior observations in glucagonoma rodent models (40,41). A decrease in α-cell number observed in glucagonoma rodent models (40,41) was not observed in mice with liver PKA activation (Fig. 5B). The reduced pancreas mass observed in mice with PKA activation is reminiscent of the reported data in rats with glucagonoma implantation (40). In both studies, reductions in α-cell mass did not fully account for the extent of pancreas mass loss, indicative of diminished exocrine pancreas mass. Because cell contraction and increased apoptosis were observed in the exocrine pancreas of rats with glucagonoma implantation (40), similar abnormalities might have developed in the pancreas of mice with hepatic PKA activation.

Studies suggest that a liver–α-cell axis also exists in human. Hyperaminoacidemia, notably elevated glutamine, alanine, arginine, lysine, ornithine, threonine, and serine, has been documented in an individual with a GCGR defect resulting from biallelic mutations in Gcgr (42). Hyperglucagonemia and α-cell hyperplasia have been reported in multiple individuals with Gcgr-inactivating mutations (43–46). In contrast, patients with glucagonoma develop hypoaminoacidemia (15). Glucagon infusion in healthy individuals reduces plasma amino acid levels, and this effect is blunted in obese individuals with hepatic steatosis, likely because of glucagon resistance (47–50). GCGR agonism in combination with GLP-1R agonism has been considered a potential therapeutic option for obesity, T2D, and metabolic dysfunction–associated steatotic liver disease (51). Clinical studies evaluating GCGR/GLP-1R coagonists have documented decreases in key amino acids in the liver–α-cell axis, such as alanine, glutamine, arginine, and proline, in patients receiving such coagonists (52,53). Whether GCGR agonism affects the human pancreas in a similar fashion to hepatic PKA activation in mice remains to be seen.

In this study, reductions in blood glucose levels and hepatic expression of gluconeogenic genes were detected in mice with hepatic GCGR or PKA inactivation, whereas transient hyperglycemia and increased hepatic expression of gluconeogenic genes were observed in mice with hepatic PKA activation (Figs. 1B and C and 3,C and D). Because GCGR signaling promotes use of amino acids for hepatic glucose production, glycemic modulations in mice with GCGR-GNAS-PKA inactivation or activation may be at least partially explained by reduced/increased amino acid catabolism in the liver. Indeed, increased liver alanine catabolism has been shown to promote hyperglycemia in T2D mouse models (54). Hyperglycemia, accompanying hypoaminoacidemia, and increased hepatic amino acid catabolism have been reported in mice with hepatic liver kinase B1 (LKB1) deletion (55). LKB1 suppression of amino acid catabolism is posttranscriptional and associated with protein posttranslational modifications. Whether LKB1 lies downstream of GCGR-GNAS-PKA and serves as a brake on amino acid catabolism promoted by GCGR-GNAS-PKA signaling is of interest for future research.

It is unclear if known downstream regulators of GCGR-GNAS-PKA on glucose and/or lipid homeostasis have roles in the amino acid homeostasis control by hepatic PKA. CREB is a transcriptional factor activated by PKA phosphorylation and an important regulator of glucose and lipid metabolism genes in the liver (56). Inhibition of either PKA or CREB in primary mouse hepatocytes blunts glucagon-induced expression of amino acid catabolism genes, indicating the potential role CREB plays in hepatic amino acid metabolism control (9). Whether hepatic CREB inhibition or activation affects amino acid homeostasis and α-cell mass in an animal model remains to be tested. CREB-regulated transcription coactivator 2 (CRTC2) is another PKA downstream factor implicated in the regulation of hepatic amino acid metabolism (56). Rats with ASO-mediated silencing of hepatic CRTC2 show reduced expression of several hepatic amino acid catabolism genes and increased circulating levels of glucagon, but no changes in circulating levels of amino acids (57). Identification of hepatic of GCGR-GNAS-PKA downstream factors in the control of amino acid and α-cell homeostasis in vivo will require additional studies.

One limitation of the study is that we did not prove causality between hypoaminoacidemia and the reduction in α-cell mass in mice with hepatic PKA activation. Elevating circulating amino acids in mice with liver PKA activation in a future study may clarify this point. Another limitation is that only male mice were used in the study.

In conclusion, we demonstrate that GCGR signals through PKA to regulate amino acid catabolism in the liver and circulating amino acid levels, which is a potent growth factor on pancreatic α-cells. Hepatic PKA plays a dominant role in liver–α-cell cross talk, mediated by circulating glucagon and amino acids.

This article contains supplementary material online at https://doi.org/10.2337/figshare.28589564.

Acknowledgments. The authors thank Megan Binn and Lawrence Miloscio for help with tissue collection; Biin Sung, Jinrang Kim, Judith Altarejos, Megan Binn, Neha Shrestha, and Nhu Truong for critical evaluation of the manuscript (all from Regeneron Pharmaceuticals). The authors also thank Regeneron DNA Core/Molecular Profiling for assistance with RNA-seq.

Duality of Interest. This study was funded by Regeneron Pharmaceuticals. All authors are employees and shareholders of Regeneron Pharmaceuticals. No other potential conflicts of interest relevant to this article were reported.

Author Contributions. K.B., J.B., and E.N. performed investigations. K.B., J.B., E.N., and Q.S. performed formal analyses. K.B. and H.O. were responsible for conceptualization and wrote the original article draft. G.H., M.S., and H.O. supervised the study. All authors reviewed and edited the article. H.O. is the guarantor of this work and, as such, had full access to all the data in the study and takes responsibility for the integrity of the data and the accuracy of the data analysis.

Prior Presentation. Some of the data were presented as a poster at the Federation of American Societies for Experimental Biology Molecular Metabolism: Cells and Systems Science Research Conference, 14–18 July 2024.

1.
Capozzi
ME
,
D’Alessio
DA
,
Campbell
JE
.
The past, present, and future physiology and pharmacology of glucagon
.
Cell Metab
2022
;
34
:
1654
1674
2.
Unger
RH
,
Cherrington
AD
.
Glucagonocentric restructuring of diabetes: a pathophysiologic and therapeutic makeover
.
J Clin Invest
2012
;
122
:
4
12
3.
Bergman
A
,
Tan
B
,
Somayaji
VR
,
Calle
RA
,
Kazierad
DJ
.
A 4-week study assessing the pharmacokinetics, pharmacodynamics, safety, and tolerability of the glucagon receptor antagonist PF-06291874 administered as monotherapy in subjects with type 2 diabetes mellitus
.
Diabetes Res Clin Pract
2017
;
126
:
95
104
4.
Guzman
CB
,
Zhang
XM
,
Liu
R
, et al
.
Treatment with LY2409021, a glucagon receptor antagonist, increases liver fat in patients with type 2 diabetes
.
Diabetes Obes Metab
2017
;
19
:
1521
1528
5.
Morgan
ES
,
Tai
L-J
,
Pham
NC
, et al
.
Antisense inhibition of glucagon receptor by IONIS-GCGRRx improves type 2 diabetes without increase in hepatic glycogen content in patients with type 2 diabetes on stable metformin therapy
.
Diabetes Care
2019
;
42
:
585
593
6.
Okamoto
H
,
Kim
J
,
Aglione
J
, et al
.
Glucagon receptor blockade with a human antibody normalizes blood glucose in diabetic mice and monkeys
.
Endocrinology
2015
;
156
:
2781
2794
7.
Pettus
JH
,
D’Alessio
D
,
Frias
JP
, et al
.
Efficacy and safety of the glucagon receptor antagonist RVT-1502 in type 2 diabetes uncontrolled on metformin monotherapy: a 12-week dose-ranging study
.
Diabetes Care
2020
;
43
:
161
168
8.
Smith
DK
,
Kates
L
,
Durinck
S
, et al
.
Elevated serum amino acids induce a subpopulation of alpha cells to initiate pancreatic neuroendocrine tumor formation
.
Cell Rep Med
2020
;
1
:
100058
9.
Korenfeld
N
,
Finkel
M
,
Buchshtab
N
,
Bar-Shimon
M
,
Charni-Natan
M
,
Goldstein
I
.
Fasting hormones synergistically induce amino acid catabolism genes to promote gluconeogenesis
.
Cell Mol Gastroenterol Hepatol
2021
;
12
:
1021
1036
10.
Solloway
MJ
,
Madjidi
A
,
Gu
C
, et al
.
Glucagon couples hepatic amino acid catabolism to mTOR-dependent regulation of α-cell mass
.
Cell Rep
2015
;
12
:
495
510
11.
Watanabe
C
,
Seino
Y
,
Miyahira
H
, et al
.
Remodeling of hepatic metabolism and hyperaminoacidemia in mice deficient in proglucagon-derived peptides
.
Diabetes
2012
;
61
:
74
84
12.
Winther-Sørensen
M
,
Galsgaard
KD
,
Santos
A
, et al
.
Glucagon acutely regulates hepatic amino acid catabolism and the effect may be disturbed by steatosis
.
Mol Metab
2020
;
42
:
101080
13.
Yang
J
,
MacDougall
ML
,
McDowell
MT
, et al
.
Polyomic profiling reveals significant hepatic metabolic alterations in glucagon-receptor (GCGR) knockout mice: implications on anti-glucagon therapies for diabetes
.
BMC Genomics
2011
;
12
:
281
14.
Muller
WA
,
Berger
M
,
Suter
P
, et al
.
Glucagon immunoreactivities and amino acid profile in plasma of duodenopancreatectomized patients
.
J Clin Invest
1979
;
63
:
820
827
15.
Mallinson
CN
,
Bloom
SR
,
Warin
AP
,
Salmon
PR
,
Cox
B
.
A glucagonoma syndrome
.
Lancet
1974
;
2
:
1
5
16.
Gelling
RW
,
Du
XQ
,
Dichmann
DS
, et al
.
Lower blood glucose, hyperglucagonemia, and pancreatic alpha cell hyperplasia in glucagon receptor knockout mice
.
Proc Natl Acad Sci U S A
2003
;
100
:
1438
1443
17.
Longuet
C
,
Robledo
AM
,
Dean
ED
, et al
.
Liver-specific disruption of the murine glucagon receptor produces α-cell hyperplasia: evidence for a circulating α-cell growth factor
.
Diabetes
2013
;
62
:
1196
1205
18.
Gong
Y
,
Yang
B
,
Zhang
D
, et al
.
Hyperaminoacidemia induces pancreatic α cell proliferation via synergism between the mTORC1 and CaSR-Gq signaling pathways
.
Nat Commun
2023
;
14
:
235
19.
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.e1365
20.
Kim
J
,
Okamoto
H
,
Huang
Z
, et al
.
Amino acid transporter Slc38a5 controls glucagon receptor inhibition-induced pancreatic α cell hyperplasia in mice
.
Cell Metab
2017
;
25
:
1348
1361.e1348
21.
Jelinek
LJ
,
Lok
S
,
Rosenberg
GB
, et al
.
Expression cloning and signaling properties of the rat glucagon receptor
.
Science
1993
;
259
:
1614
1616
22.
Chen
M
,
Gavrilova
O
,
Zhao
W-Q
, et al
.
Increased glucose tolerance and reduced adiposity in the absence of fasting hypoglycemia in mice with liver-specific Gs alpha deficiency
.
J Clin Invest
2005
;
115
:
3217
3227
23.
Aromataris
EC
,
Roberts
ML
,
Barritt
GJ
,
Rychkov
GY
.
Glucagon activates Ca2+ and Cl- channels in rat hepatocytes
.
J Physiol
2006
;
573
:
611
625
24.
Sinclair
EM
,
Yusta
B
,
Streutker
C
, et al
.
Glucagon receptor signaling is essential for control of murine hepatocyte survival
.
Gastroenterology
2008
;
135
:
2096
2106
25.
Spolitu
S
,
Okamoto
H
,
Dai
W
, et al
.
Hepatic glucagon signaling regulates PCSK9 and low-density lipoprotein cholesterol
.
Circ Res
2019
;
124
:
38
51
26.
Wang
Y
,
Spolitu
S
,
Zadroga
JA
,
Sarecha
AK
,
Ozcan
L
.
Hepatocyte Rap1a contributes to obesity- and statin-associated hyperglycemia
.
Cell Rep
2022
;
40
:
111259
27.
Cavino
K
,
Sung
B
,
Su
Q
, et al
.
Glucagon receptor inhibition reduces hyperammonemia and lethality in male mice with urea cycle disorder
.
Endocrinology
2021
;
162
:
bqaa211
28.
Adam
RC
,
Pryce
DS
,
Lee
JS
, et al
.
Activin E-ACVR1C cross talk controls energy storage via suppression of adipose lipolysis in mice
.
Proc Natl Acad Sci U S A
2023
;
120
:
e2309967120
29.
Love
MI
,
Huber
W
,
Anders
S
.
Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2
.
Genome Biol
2014
;
15
:
550
30.
Bröer
S
,
Bröer
A
.
Amino acid homeostasis and signalling in mammalian cells and organisms
.
Biochem J
2017
;
474
:
1935
1963
31.
He
L
,
Chang
E
,
Peng
J
, et al
.
Activation of the cAMP-PKA pathway antagonizes metformin suppression of hepatic glucose production
.
J Biol Chem
2016
;
291
:
10562
10570
32.
Song
W-J
,
Mondal
P
,
Wolfe
A
, et al
.
Glucagon regulates hepatic kisspeptin to impair insulin secretion
.
Cell Metab
2014
;
19
:
667
681
33.
Soundarapandian
MM
,
Juliana
CA
,
Chai
J
,
Haslett
PA
,
Fitzgerald
K
,
De León
DD
.
Activation of protein kinase A (PKA) signaling mitigates congenital hyperinsulinism associated hypoglycemia in the Sur1-/- mouse model
.
PLoS One
2020
;
15
:
e0236892
34.
Niswender
CM
,
Willis
BS
,
Wallen
A
, et al
.
Cre recombinase-dependent expression of a constitutively active mutant allele of the catalytic subunit of protein kinase A
.
Genesis
2005
;
43
:
109
119
35.
Elmelund
E
,
Galsgaard
KD
,
Johansen
CD
, et al
.
Opposing effects of chronic glucagon receptor agonism and antagonism on amino acids, hepatic gene expression, and alpha cells
.
iScience
2022
;
25
:
105296
36.
Spears
E
,
Stanley
JE
,
Shou
M
, et al
.
Pancreatic islet α cell function and proliferation requires the arginine transporter SLC7A2
.
bioRxiv
2023
37.
Galsgaard
KD
,
Jepsen
SL
,
Kjeldsen
SAS
,
Pedersen
J
,
Wewer Albrechtsen
NJ
,
Holst
JJ
.
Alanine, arginine, cysteine, and proline, but not glutamine, are substrates for, and acute mediators of, the liver-α-cell axis in female mice
.
Am J Physiol Endocrinol Metab
2020
;
318
:
E920
E929
38.
Winther-Sørensen
M
,
Garcia
SL
,
Bartholdy
A
, et al
.
Determinants of plasma levels of proglucagon and the metabolic impact of glucagon receptor signalling: a UK Biobank study
.
Diabetologia
2024
;
67
:
1602
1615
39.
Li
L
,
Zhang
P
,
Bao
Z
,
Wang
T
,
Liu
S
,
Huang
F
.
PGC-1α promotes ureagenesis in mouse periportal hepatocytes through SIRT3 and SIRT5 in response to glucagon
.
Sci Rep
2016
;
6
:
24156
40.
Blume
N
,
Skouv
J
,
Larsson
LI
,
Holst
JJ
,
Madsen
OD
.
Potent inhibitory effects of transplantable rat glucagonomas and insulinomas on the respective endogenous islet cells are associated with pancreatic apoptosis
.
J Clin Invest
1995
;
96
:
2227
2235
41.
Drucker
DJ
,
Lee
YC
,
Asa
SL
,
Brubaker
PL
.
Inhibition of pancreatic glucagon gene expression in mice bearing a subcutaneous glucagon-producing GLUTag transplantable tumor
.
Mol Endocrinol
1992
;
6
:
2175
2184
42.
Li
H
,
Zhao
L
,
Singh
R
, et al
.
The first pediatric case of glucagon receptor defect due to biallelic mutations in GCGR is identified by newborn screening of elevated arginine
.
Mol Genet Metab Rep
2018
;
17
:
46
52
43.
Gild
ML
,
Tsang
V
,
Samra
J
,
Clifton-Bligh
RJ
,
Tacon
L
,
Gill
AJ
.
Hypercalcemia in glucagon cell hyperplasia and neoplasia (Mahvash syndrome): a new association
.
J Clin Endocrinol Metab
2018
;
103
:
3119
3123
44.
Miller
HC
,
Kidd
M
,
Modlin
IM
, et al
.
Glucagon receptor gene mutations with hyperglucagonemia but without the glucagonoma syndrome
.
World J Gastrointest Surg
2015
;
7
:
60
66
45.
Sipos
B
,
Sperveslage
J
,
Anlauf
M
, et al
.
Glucagon cell hyperplasia and neoplasia with and without glucagon receptor mutations
.
J Clin Endocrinol Metab
2015
;
100
:
E783
E788
46.
Zhou
C
,
Dhall
D
,
Nissen
NN
,
Chen
C-R
,
Yu
R
.
Homozygous P86S mutation of the human glucagon receptor is associated with hyperglucagonemia, alpha cell hyperplasia, and islet cell tumor
.
Pancreas
2009
;
38
:
941
946
47.
Vega
RB
,
Whytock
KL
,
Gassenhuber
J
, et al
.
A metabolomic signature of glucagon action in healthy individuals with overweight/obesity
.
J Endocr Soc
2021
;
5
:
bvab118
48.
Suppli
MP
,
Bagger
JI
,
Lund
A
, et al
.
Glucagon resistance at the level of amino acid turnover in obese subjects with hepatic steatosis
.
Diabetes
2020
;
69
:
1090
1099
49.
Suppli
MP
,
Høgedal
A
,
Bagger
JI
, et al
.
Signs of glucagon resistance after a 2-week hypercaloric diet intervention
.
J Clin Endocrinol Metab
2024
;
109
:
955
967
50.
Kjeldsen
SAS
,
Richter
MM
,
Jensen
NJ
, et al
.
Glucagon resistance in individuals with obesity and hepatic steatosis can be measured using the GLUSENTIC test and index
.
Diabetes
2024
;
73
:
1716
1727
51.
Tan
TM-M
.
Co-agonist therapeutics come of age for obesity
.
Nat Rev Endocrinol
2023
;
19
:
66
67
52.
Li
W
,
Kirchner
T
,
Ho
G
, et al
.
Amino acids are sensitive glucagon receptor-specific biomarkers for glucagon-like peptide-1 receptor/glucagon receptor dual agonists
.
Diabetes Obes Metab
2020
;
22
:
2437
2450
53.
Rosenstock
J
,
Frias
J
,
Jastreboff
AM
, et al
.
Retatrutide, a GIP, GLP-1 and glucagon receptor agonist, for people with type 2 diabetes: a randomised, double-blind, placebo and active-controlled, parallel-group, phase 2 trial conducted in the USA
.
Lancet
2023
;
402
:
529
544
54.
Okun
JG
,
Rusu
PM
,
Chan
AY
, et al
.
Liver alanine catabolism promotes skeletal muscle atrophy and hyperglycaemia in type 2 diabetes
.
Nat Metab
2021
;
3
:
394
409
55.
Just
P-A
,
Charawi
S
,
Denis
RGP
, et al
.
Lkb1 suppresses amino acid-driven gluconeogenesis in the liver
.
Nat Commun
2020
;
11
:
6127
56.
Altarejos
JY
,
Montminy
M
.
CREB and the CRTC co-activators: sensors for hormonal and metabolic signals
.
Nat Rev Mol Cell Biol
2011
;
12
:
141
151
57.
Erion
DM
,
Kotas
ME
,
McGlashon
J
, et al
.
cAMP-responsive element-binding protein (CREB)-regulated transcription coactivator 2 (CRTC2) promotes glucagon clearance and hepatic amino acid catabolism to regulate glucose homeostasis
.
J Biol Chem
2013
;
288
:
16167
16176
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.